72 results on '"J., Fahrer"'
Search Results
2. KRAS-mutated iCCA display distinct molecular alterations and a preferential sensitivity towards PARP-1 inhibition
- Author
-
FL Keggenhoff, D Castven, D Becker, SP Pereira, M Hartmann, B Straub, J Fahrer, JB Andersen, MS Matter, S Rössler, B Kaina, PR Galle, and JU Marquardt
- Subjects
Chemistry ,Poly ADP ribose polymerase ,Cancer research ,medicine ,Sensitivity (control systems) ,KRAS ,medicine.disease_cause - Published
- 2021
- Full Text
- View/download PDF
3. Chemische Kanzerogenese
- Author
-
J. Fahrer and B. Kaina
- Published
- 2017
- Full Text
- View/download PDF
4. KRAS-mutated intrahepatic cholangiocarcinoma shows preferential sensitivity towards PARP-1-based interventions
- Author
-
FL Mahn, D Castven, SP Pereira, M Hartmann, J Fahrer, JB Andersen, MS Matter, T Gaiser, S Rössler, PR Galle, and JU Marquardt
- Subjects
business.industry ,Poly ADP ribose polymerase ,Cancer research ,medicine ,Sensitivity (control systems) ,KRAS ,medicine.disease_cause ,business ,Intrahepatic Cholangiocarcinoma - Published
- 2020
- Full Text
- View/download PDF
5. DEK Is a Poly(ADP-Ribose) Acceptor in Apoptosis and Mediates Resistance to Genotoxic Stress
- Author
-
Christine Strasser, Anja Tabbert, Elisa Ferrando-May, Alexander Bürkle, Maria Moreno-Villanueva, J. Fahrer, Nirit Mor-Vaknin, David M. Markovitz, Michael S. Khodadoust, and Ferdinand Kappes
- Subjects
Poly Adenosine Diphosphate Ribose ,Chromosomal Proteins, Non-Histone ,Apoptosis ,Biology ,medicine.disease_cause ,Jurkat cells ,Cell Line ,Jurkat Cells ,Mice ,03 medical and health sciences ,0302 clinical medicine ,ddc:570 ,medicine ,Animals ,Humans ,Poly-ADP-Ribose Binding Proteins ,Phosphorylation ,Molecular Biology ,Autoantibodies ,030304 developmental biology ,Oncogene Proteins ,0303 health sciences ,Antibodies, Monoclonal ,Articles ,Cell Biology ,Molecular biology ,Arthritis, Juvenile ,Chromatin ,Recombinant Proteins ,3. Good health ,Cell biology ,stomatognathic diseases ,Cell culture ,030220 oncology & carcinogenesis ,Phosphoprotein ,Carcinogenesis ,Protein Processing, Post-Translational ,HeLa Cells ,Mutagens - Abstract
DEK is a nuclear phosphoprotein implicated in oncogenesis and autoimmunity and a major component of metazoan chromatin. The intracellular cues that control the binding of DEK to DNA and its pleiotropic functions in DNA- and RNA-dependent processes have remained mainly elusive so far. Our recent finding that the phosphorylation status of DEK is altered during death receptor-mediated apoptosis suggested a potential involvement of DEK in stress signaling. In this study, we show that in cells committed to die, a portion of the cellular DEK pool is extensively posttranslationally modified by phosphorylation and poly(ADP-ribosyl)ation. Through interference with DEK expression, we further show that DEK promotes the repair of DNA lesions and protects cells from genotoxic agents that typically trigger poly(ADP-ribose) polymerase activation. The posttranslational modification of DEK during apoptosis is accompanied by the removal of the protein from chromatin and its release into the extracellular space. Released modified DEK is recognized by autoantibodies present in the synovial fluids of patients affected by juvenile rheumatoid arthritis/juvenile idiopathic arthritis. These findings point to a crucial role of poly(ADP-ribosyl)ation in shaping DEK's autoantigenic properties and in its function as a promoter of cell survival.
- Published
- 2008
6. High-affinity interaction of poly(ADP-ribose) and the human DEK oncoprotein depends upon chain length
- Author
-
David M. Markovitz, Ferdinand Kappes, Oliver Popp, Sascha Beneke, Maria Malanga, Jörg Fahrer, Elisa Ferrando-May, Alexander Bürkle, J., Fahrer, O., Popp, Malanga, Maria, S., Beneke, D. M., Markovitz, E., Ferrando May, A., Bürkle, and F., Kappes
- Subjects
Poly Adenosine Diphosphate Ribose ,Chromosomal Proteins, Non-Histone ,DNA repair ,DNA damage ,Molecular Sequence Data ,Biology ,Biochemistry ,DNA-binding protein ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,ddc:570 ,Humans ,Electrophoretic mobility shift assay ,Amino Acid Sequence ,Poly-ADP-Ribose Binding Proteins ,Binding site ,Poly(ADP-ribose) Polymerase ,Oncogene ,030304 developmental biology ,Oncogene Proteins ,0303 health sciences ,Binding Sites ,DNA-repair ,DNA ,Post-translational Modification ,Chromatin ,Protein Structure, Tertiary ,Cell biology ,stomatognathic diseases ,chemistry ,030220 oncology & carcinogenesis ,Protein Multimerization ,Sequence Alignment - Abstract
Poly(ADP-ribose) polymerase-1 (PARP-1) is a molecular DNA damage sensor that catalyzes the synthesis of the complex biopolymer poly(ADP-ribose) (PAR) under consumption of NAD(+). PAR engages in fundamental cellular processes such as DNA metabolism and transcription and interacts noncovalently with specific binding proteins involved in DNA repair and regulation of chromatin structure. A factor implicated in DNA repair and chromatin organization is the DEK oncoprotein, an abundant and conserved constituent of metazoan chromatin, and the only member of its protein class. We have recently demonstrated that DEK, under stress conditions, is covalently modified with PAR by PARP-1, leading to a partial release of DEK into the cytoplasm. Additionally, we have also observed a noncovalent interaction between DEK and PAR, which we detail here. Using sequence alignment, we identify three functional PAR-binding sites in the DEK primary sequence and confirm their functionality in PAR binding studies. Furthermore, we show that the noncovalent binding to DEK is dependent on PAR chain length as revealed by an overlay blot technique and a PAR electrophoretic mobility shift assay. Intriguingly, DEK promotes the formation of a defined complex with a 54mer PAR (K(D) = 6 x 10(-8) M), whereas no specific interaction is detected with a short PAR chain (18mer). In stark contrast to covalent poly(ADP-ribosyl)ation of DEK, the noncovalent interaction does not affect the overall ability of DEK to bind to DNA. Instead the noncovalent interaction interferes with subsequent DNA-dependent multimerization activities of DEK, as seen in South-Western, electrophoretic mobility shift, topology, and aggregation assays. In particular, noncovalent attachment of PAR to DEK promotes the formation of DEK-DEK complexes by competing with DNA binding. This was seen by the reduced affinity of PAR-bound DEK for DNA templates in solution. Taken together, our findings deepen the molecular understanding of the DEK-PAR interplay and support the existence of a cellular "PAR code" represented by PAR chain length.
- Published
- 2010
7. Targeting PARP-1 and DNA Damage Response Defects in Colorectal Cancer Chemotherapy with Established and Novel PARP Inhibitors.
- Author
-
Demuth P, Thibol L, Lemsch A, Potlitz F, Schulig L, Grathwol C, Manolikakes G, Schade D, Roukos V, Link A, and Fahrer J
- Abstract
The DNA repair protein PARP-1 emerged as a valuable target in the treatment of tumor entities with deficiencies of BRCA1/2 , such as breast cancer. More recently, the application of PARP inhibitors (PARPi) such as olaparib has been expanded to other cancer entities including colorectal cancer (CRC). We previously demonstrated that PARP-1 is overexpressed in human CRC and promotes CRC progression in a mouse model. However, acquired resistance to PARPi and cytotoxicity-mediated adverse effects limit their clinical applicability. Here, we detailed the role of PARP-1 as a therapeutic target in CRC and studied the efficacy of novel PARPi compounds in wildtype (WT) and DNA repair-deficient CRC cell lines together with the chemotherapeutics irinotecan (IT), 5-fluorouracil (5-FU), and oxaliplatin (OXA). Based on the ComPlat molecule archive, we identified novel PARPi candidates by molecular docking experiments in silico, which were then confirmed by in vitro PARP activity measurements. Two promising candidates (X17613 and X17618) also showed potent PARP-1 inhibition in a CRC cell-based assay. In contrast to olaparib, the PARPi candidates caused no PARP-1 trapping and, consistently, were not or only weakly cytotoxic in WT CRC cells and their BRCA2- or ATR-deficient counterparts. Importantly, both PARPi candidates did not affect the viability of nonmalignant human colonic epithelial cells. While both olaparib and veliparib increased the sensitivity of WT CRC cells towards IT, no synergism was observed for X17613 and X17618. Finally, we provided evidence that all PARPi (olaparib > veliparib > X17613 > X17618) synergize with chemotherapeutic drugs (IT > OXA) in a BRCA2-dependent manner in CRC cells, whereas ATR deficiency had only a minor impact. Collectively, our study identified novel lead structures with potent PARP-1 inhibitory activity in CRC cells but low cytotoxicity due to the lack of PARP-1 trapping, which synergized with IT in homologous recombination deficiency.
- Published
- 2024
- Full Text
- View/download PDF
8. PARP-1 selectively impairs KRAS -driven phenotypic and molecular features in intrahepatic cholangiocarcinoma.
- Author
-
Keggenhoff FL, Castven D, Becker D, Stojkovic S, Castven J, Zimpel C, Straub BK, Gerber T, Langer H, Hähnel P, Kindler T, Fahrer J, O'Rourke CJ, Ehmer U, Saborowski A, Ma L, Wang XW, Gaiser T, Matter MS, Sina C, Derer S, Lee JS, Roessler S, Kaina B, Andersen JB, Galle PR, and Marquardt JU
- Subjects
- Humans, Animals, Mice, Mice, Knockout, Cell Line, Tumor, Mutation, Cholangiocarcinoma genetics, Cholangiocarcinoma pathology, Cholangiocarcinoma metabolism, Bile Duct Neoplasms genetics, Bile Duct Neoplasms pathology, Bile Duct Neoplasms metabolism, Proto-Oncogene Proteins p21(ras) genetics, Poly (ADP-Ribose) Polymerase-1 genetics, Poly (ADP-Ribose) Polymerase-1 metabolism, Poly (ADP-Ribose) Polymerase-1 antagonists & inhibitors, Phenotype
- Abstract
Objective: Intrahepatic cholangiocarcinoma (iCCA) is the second most common primary liver cancer with limited therapeutic options. KRAS mutations are among the most abundant genetic alterations in iCCA associated with poor clinical outcome and treatment response. Recent findings indicate that Poly(ADP-ribose)polymerase1 (PARP-1) is implicated in KRAS -driven cancers, but its exact role in cholangiocarcinogenesis remains undefined., Design: PARP-1 inhibition was performed in patient-derived and established iCCA cells using RNAi, CRISPR/Cas9 and pharmacological inhibition in KRAS -mutant, non-mutant cells. In addition, Parp-1 knockout mice were combined with iCCA induction by hydrodynamic tail vein injection to evaluate an impact on phenotypic and molecular features of Kras -driven and Kras -wildtype iCCA. Clinical implications were confirmed in authentic human iCCA., Results: PARP-1 was significantly enhanced in KRAS -mutant human iCCA. PARP-1-based interventions preferentially impaired cell viability and tumourigenicity in human KRAS -mutant cell lines. Consistently, loss of Parp-1 provoked distinct phenotype in Kras/Tp53- induced versus Akt/Nicd- induced iCCA and abolished Kras -dependent cholangiocarcinogenesis. Transcriptome analyses confirmed preferential impairment of DNA damage response pathways and replicative stress response mediated by CHK1. Consistently, inhibition of CHK1 effectively reversed PARP-1 mediated effects. Finally, Parp-1 depletion induced molecular switch of KRAS -mutant iCCA recapitulating good prognostic human iCCA patients., Conclusion: Our findings identify the novel prognostic and therapeutic role of PARP-1 in iCCA patients with activation of oncogenic KRAS signalling., Competing Interests: Competing interests: None declared., (© Author(s) (or their employer(s)) 2024. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2024
- Full Text
- View/download PDF
9. Decoding the genetic program of micronucleus formation: linking chromosomal instability to human disease.
- Author
-
Fahrer J
- Subjects
- Humans, Micronuclei, Chromosome-Defective, Chromosomal Instability genetics
- Published
- 2024
- Full Text
- View/download PDF
10. Influence of Bilberry Extract on Neuronal Cell Toxicity.
- Author
-
König S, Bakuradze T, Jesser S, Sreeja HA, Carlsson MJ, Fahrer J, Kins S, and Richling E
- Abstract
Increased intake of dietary antioxidants such as anthocyanins, which are enriched in colourful fruits, is a promising alternative to reduce the risk of degenerative diseases such as Alzheimer's Disease (AD). Since Amyloid β (Aβ) is one of the key components contributing to AD pathology, probably by reactive oxygen species (ROS) induction, this study investigated the preventive effect of anthocyanin-rich bilberry extract (BE) and its anthocyanin fraction (ACN) on ROS generation and cell toxicity. The results showed a significant and concentration-dependent decrease in neuroblastoma cell (SH-SY5Y) viability by BE or ACN, whereas no cell toxicity was observed in HeLa cells. Incubation with BE and ACN for 24 h diminished the generation of induced ROS levels in SH-SY5Y and HeLa cells. In addition, low concentrations of BE (1-5 µg/mL) showed protective effects against Aβ-induced cytotoxicity in SH-SY5Y cells. In conclusion, our results suggest antioxidant and protective effects of BE and ACN, which could potentially be used to delay the course of neurodegenerative diseases such as AD. Further studies are needed to clarify the high potential of anthocyanins and their in vivo metabolites on neuronal function.
- Published
- 2024
- Full Text
- View/download PDF
11. Parent-child interactive behavior in a German sample of parents with and without a mental illness: model replication and adaption of the Coding Interactive Behavior system.
- Author
-
Fahrer J, Doebler P, Hagelweide K, Kern P, Nonnenmacher N, Seipp V, Reck C, Schwenck C, Weigelt S, Zietlow AL, and Christiansen H
- Abstract
Studies using observational measures often fail to meet statistical standards for both reliability and validity. The present study examined the psychometric properties of the Coding Interactive Behavior (CIB) System within a German sample of parent-child dyads. The sample consisted of 149 parents with and without a mental illness and their children [n experimental group (EG) = 75, n control group (CG) = 74] who participated in the larger Children of Mentally Ill Parents at Risk Evaluation (COMPARE) study. The age of the children ranged from 3 to 12 years (M = 7.99, SD = 2.5). Exploratory factor analysis supported a five-factor model of the CIB with items describing 1) parental sensitivity/reciprocity, 2) parental intrusiveness, 3) child withdrawal, 4) child involvement, and 5) parent limit setting/child compliance. Compared to international samples, the model was reduced by two independent dyadic factors. Testing for predictive validity identified seven items with predictive power to differentiate parental group membership. The CIB factors did not seem to be sufficiently sensitive to illustrate differences in interaction within a sample of parents with various mental illnesses. To apply the CIB to the described sample or similar ones in the future, additional measurement instruments may be necessary., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest. The reviewer AD declared a past co-authorship with one of the authors HC to the handling editor. The reviewer AT declared a past co-authorship with the author(s) to the handling editor., (Copyright © 2024 Fahrer, Doebler, Hagelweide, Kern, Nonnenmacher, Seipp, Reck, Schwenck, Weigelt, Zietlow and Christiansen.)
- Published
- 2024
- Full Text
- View/download PDF
12. Toxins in Botanical Drugs and Plant-derived Food and Feed - from Science to Regulation: A Workshop Review.
- Author
-
Schrenk D, Allemang A, Fahrer J, Harms H, Li X, Lin G, Mahony C, Mulder P, Peijnenburg A, Pfuhler S, Punt A, Sievers H, Troutman J, and Widjaja F
- Subjects
- Pyrrolizidine Alkaloids toxicity, Plants, Medicinal
- Abstract
In September 2022, the 3rd International Workshop on pyrrolizidine alkaloids (PAs) and related phytotoxins was held on-line, entitled 'Toxins in botanical drugs and plant-derived food and feed - from science to regulation'. The workshop focused on new findings about the occurrence, exposure, toxicity, and risk assessment of PAs. In addition, new scientific results related to the risk assessment of alkenylbenzenes, a distinct class of herbal constituents, were presented. The presence of PAs and alkenylbenzenes in plant-derived food, feed, and herbal medicines has raised health concerns with respect to their acute and chronic toxicity but mainly related to the genotoxic and carcinogenic properties of several congeners. The compounds are natural constituents of a variety of plant families and species widely used in medicinal, food, and feed products. Their individual occurrence, levels, and toxic properties, together with the broad range of congeners present in nature, represent a striking challenge to modern toxicology. This review tries to provide an overview of the current knowledge on these compounds and indicates needs and perspectives for future research., Competing Interests: The authors declare that they have no conflict of interest., (Thieme. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
13. New Insights into Wine Color Analysis: A Comparison of Analytical Methods to Sensory Perception for Red and White Varietal Wines.
- Author
-
Hensel M, Scheiermann M, Fahrer J, and Durner D
- Subjects
- Humans, Color, Sensation, Perception, Wine analysis, Vitis
- Abstract
The Glories method and CIE L * a * b * color space are poorly correlated in the light red high-saturation color area. Non-uniformities in the CIE L * a * b * color space have led to the CIEDE2000 formula, while the Euclidean color distance is still used in wine research. In this study, 112 white and red wines were examined to compare the Glories method, CIE L * a * b , and human perception using monovarietal wines from different grape varieties. The objective of this study was to investigate which of the two methods and which parameter of each method better aligned with human perception. The visual color threshold was re-evaluated with the CIEDE2000 formula and triangle testing. CIE L * a * b * more closely matched the human perception, elevating the use of CIE L * a * b * over the use of the Glories method. Visual color thresholds were better expressed with CIEDE2000 but still varied depending upon the color area in the CIE L * a * b * color space.
- Published
- 2024
- Full Text
- View/download PDF
14. OCT1-dependent uptake of structurally diverse pyrrolizidine alkaloids in human liver cells is crucial for their genotoxic and cytotoxic effects.
- Author
-
Haas M, Ackermann G, Küpper JH, Glatt H, Schrenk D, and Fahrer J
- Subjects
- Humans, Cytochrome P-450 CYP3A metabolism, Liver, Hepatocytes, DNA Damage, Pyrrolizidine Alkaloids metabolism, Antineoplastic Agents pharmacology
- Abstract
Pyrrolizidine alkaloids (PAs) are important plant hepatotoxins, which occur as contaminants in plant-based foods, feeds and phytomedicines. Numerous studies demonstrated that the genotoxicity and cytotoxicity of PAs depend on their chemical structure, allowing for potency ranking and grouping. Organic cation transporter-1 (OCT1) was previously shown to be involved in the cellular uptake of the cyclic PA diesters monocrotaline, retrorsine and senescionine. However, little is known about the structure-dependent transport of PAs. Therefore, we investigated the impact of OCT1 on the uptake and toxicity of three structurally diverse PAs (heliotrine, lasiocarpine and riddelliine) differing in their degree and type of esterification in metabolically competent human liver cell models and hamster fibroblasts. Human HepG2-CYP3A4 liver cells were exposed to the respective PA in the presence or absence of the OCT1-inhibitors D-THP and quinidine, revealing a strongly attenuated cytotoxicity upon OCT1 inhibition. The same experiments were repeated in V79-CYP3A4 hamster fibroblasts, confirming that OCT1 inhibition prevents the cytotoxic effects of all tested PAs. Interestingly, OCT1 protein levels were much lower in V79-CYP3A4 than in HepG2-CYP3A4 cells, which correlated with their lower susceptibility to PA-induced cytotoxicity. The cytoprotective effect of OCT1 inhibiton was also demonstrated in primary human hepatocytes following PA exposure. Our experiments further showed that the genotoxic effects triggered by the three PAs are blocked by OCT1 inhibition as evidenced by strongly reduced γH2AX and p53 levels. Consistently, inhibition of OCT1-mediated uptake suppressed the activation of the DNA damage response (DDR) as revealed by decreased phosphorylation of checkpoint kinases upon PA treatment. In conclusion, we demonstrated that PAs, independent of their degree of esterification, are substrates for OCT1-mediated uptake into human liver cells. We further provided evidence that OCT1 inhibition prevents PA-triggered genotoxicity, DDR activation and subsequent cytotoxicity. These findings highlight the crucial role of OCT1 together with CYP3A4-dependent metabolic activation for PA toxicity., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
15. Heme Oxygenase-1 and Its Role in Colorectal Cancer.
- Author
-
Fahrer J, Wittmann S, Wolf AC, and Kostka T
- Abstract
Heme oxygenase-1 (HO-1) is an enzyme located at the endoplasmic reticulum, which is responsible for the degradation of cellular heme into ferrous iron, carbon monoxide and biliverdin-IXa. In addition to this main function, the enzyme is involved in many other homeostatic, toxic and cancer-related mechanisms. In this review, we first summarize the importance of HO-1 in physiology and pathophysiology with a focus on the digestive system. We then detail its structure and function, followed by a section on the regulatory mechanisms that control HO-1 expression and activity. Moreover, HO-2 as important further HO isoform is discussed, highlighting the similarities and differences with regard to HO-1. Subsequently, we describe the direct and indirect cytoprotective functions of HO-1 and its breakdown products carbon monoxide and biliverdin-IXa, but also highlight possible pro-inflammatory effects. Finally, we address the role of HO-1 in cancer with a particular focus on colorectal cancer. Here, relevant pathways and mechanisms are presented, through which HO-1 impacts tumor induction and tumor progression. These include oxidative stress and DNA damage, ferroptosis, cell cycle progression and apoptosis as well as migration, proliferation, and epithelial-mesenchymal transition.
- Published
- 2023
- Full Text
- View/download PDF
16. Endogenous anti-tumorigenic nitro-fatty acids inhibit the ubiquitin-proteasome system by directly targeting the 26S proteasome.
- Author
-
Brat C, Huynh Phuoc HP, Awad O, Parmar BS, Hellmuth N, Heinicke U, Amr S, Grimmer J, Sürün D, Husnjak K, Carlsson M, Fahrer J, Bauer T, Krieg SC, Manolikakes G, Zacharowski K, Steinhilber D, Münch C, Maier TJ, and Roos J
- Subjects
- Animals, Fatty Acids pharmacology, Proteasome Inhibitors pharmacology, Proteasome Endopeptidase Complex metabolism, Ubiquitin metabolism
- Abstract
Nitro-fatty acids (NFAs) are endogenous lipid mediators causing a spectrum of anti-inflammatory effects by covalent modification of key proteins within inflammatory signaling pathways. Recent animal models of solid tumors have helped demonstrate their potential as anti-tumorigenic therapeutics. This study evaluated the anti-tumorigenic effects of NFAs in colon carcinoma cells and other solid and leukemic tumor cell lines. NFAs inhibited the ubiquitin-proteasome system (UPS) by directly targeting the 26S proteasome, leading to polyubiquitination and inhibition of the proteasome activities. UPS suppression induced the unfolded protein response, resulting in tumor cell death. The NFA-mediated effects were substantial, specific, and enduring, representing a unique mode of action for UPS suppression. This study provides mechanistic insights into the biological actions of NFAs as possible endogenous tumor-suppressive factors, indicating that NFAs might be key structures for designing a novel class of direct proteasome inhibitors., Competing Interests: Declaration of interests The authors declare no potential conflicts of interest., (Copyright © 2023 Elsevier Ltd. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
17. New approach methodologies to facilitate and improve the hazard assessment of non-genotoxic carcinogens-a PARC project.
- Author
-
Audebert M, Assmann AS, Azqueta A, Babica P, Benfenati E, Bortoli S, Bouwman P, Braeuning A, Burgdorf T, Coumoul X, Debizet K, Dusinska M, Ertych N, Fahrer J, Fetz V, Le Hégarat L, López de Cerain A, Heusinkveld HJ, Hogeveen K, Jacobs MN, Luijten M, Raitano G, Recoules C, Rundén-Pran E, Saleh M, Sovadinová I, Stampar M, Thibol L, Tomkiewicz C, Vettorazzi A, Van de Water B, El Yamani N, Zegura B, and Oelgeschläger M
- Abstract
Carcinogenic chemicals, or their metabolites, can be classified as genotoxic or non-genotoxic carcinogens (NGTxCs). Genotoxic compounds induce DNA damage, which can be detected by an established in vitro and in vivo battery of genotoxicity assays. For NGTxCs, DNA is not the primary target, and the possible modes of action (MoA) of NGTxCs are much more diverse than those of genotoxic compounds, and there is no specific in vitro assay for detecting NGTxCs. Therefore, the evaluation of the carcinogenic potential is still dependent on long-term studies in rodents. This 2-year bioassay, mainly applied for testing agrochemicals and pharmaceuticals, is time-consuming, costly and requires very high numbers of animals. More importantly, its relevance for human risk assessment is questionable due to the limited predictivity for human cancer risk, especially with regard to NGTxCs. Thus, there is an urgent need for a transition to new approach methodologies (NAMs), integrating human-relevant in vitro assays and in silico tools that better exploit the current knowledge of the multiple processes involved in carcinogenesis into a modern safety assessment toolbox. Here, we describe an integrative project that aims to use a variety of novel approaches to detect the carcinogenic potential of NGTxCs based on different mechanisms and pathways involved in carcinogenesis. The aim of this project is to contribute suitable assays for the safety assessment toolbox for an efficient and improved, internationally recognized hazard assessment of NGTxCs, and ultimately to contribute to reliable mechanism-based next-generation risk assessment for chemical carcinogens., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Audebert, Assmann, Azqueta, Babica, Benfenati, Bortoli, Bouwman, Braeuning, Burgdorf, Coumoul, Debizet, Dusinska, Ertych, Fahrer, Fetz, Le Hégarat, López de Cerain, Heusinkveld, Hogeveen, Jacobs, Luijten, Raitano, Recoules, Rundén-Pran, Saleh, Sovadinová, Stampar, Thibol, Tomkiewicz, Vettorazzi, Van de Water, El Yamani, Zegura and Oelgeschläger.)
- Published
- 2023
- Full Text
- View/download PDF
18. Potency ranking of pyrrolizidine alkaloids in metabolically competent human liver cancer cells and primary human hepatocytes using a genotoxicity test battery.
- Author
-
Haas M, Wirachowski K, Thibol L, Küpper JH, Schrenk D, and Fahrer J
- Subjects
- Humans, Cytochrome P-450 CYP3A genetics, Cytochrome P-450 CYP3A metabolism, Hepatocytes, Mutagenicity Tests, Pyrrolizidine Alkaloids metabolism, Liver Neoplasms metabolism
- Abstract
Pyrrolizidine alkaloids (PAs) occur as contaminants in plant-based foods and herbal medicines. Following metabolic activation by cytochrome P450 (CYP) enzymes, PAs induce DNA damage, hepatotoxicity and can cause liver cancer in rodents. There is ample evidence that the chemical structure of PAs determines their toxicity. However, more quantitative genotoxicity data are required, particularly in primary human hepatocytes (PHH). Here, the genotoxicity of eleven structurally different PAs was investigated in human HepG2 liver cells with CYP3A4 overexpression and PHH using an in vitro test battery. Furthermore, the data were subject to benchmark dose (BMD) modeling to derive the genotoxic potency of individual PAs. The cytotoxicity was initially determined in HepG2-CYP3A4 cells, revealing a clear structure-toxicity relationship for the PAs. Importantly, experiments in PHH confirmed the structure-dependent toxicity and cytotoxic potency ranking of the tested PAs. The genotoxicity markers γH2AX and p53 as well as the alkaline Comet assay consistently demonstrated a structure-dependent genotoxicity of PAs in HepG2-CYP3A4 cells, correlating well with their cytotoxic potency. BMD modeling yielded BMD values in the range of 0.1-10 µM for most cyclic and open diesters, followed by the monoesters. While retrorsine showed the highest genotoxic potency, monocrotaline and lycopsamine displayed the lowest genotoxicity. Finally, experiments in PHH corroborated the genotoxic potency ranking, and revealed genotoxic effects even in the absence of detectable cytotoxicity. In conclusion, our findings strongly support the concept of grouping PAs into potency classes and help to pave the way for a broader acceptance of relative potency factors in risk assessment., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
19. DNA Alkylation Damage by Nitrosamines and Relevant DNA Repair Pathways.
- Author
-
Fahrer J and Christmann M
- Subjects
- DNA Damage, Alkylation, DNA Repair, Alkylating Agents pharmacology, DNA Adducts, Nitrosamines
- Abstract
Nitrosamines occur widespread in food, drinking water, cosmetics, as well as tobacco smoke and can arise endogenously. More recently, nitrosamines have been detected as impurities in various drugs. This is of particular concern as nitrosamines are alkylating agents that are genotoxic and carcinogenic. We first summarize the current knowledge on the different sources and chemical nature of alkylating agents with a focus on relevant nitrosamines. Subsequently, we present the major DNA alkylation adducts induced by nitrosamines upon their metabolic activation by CYP450 monooxygenases. We then describe the DNA repair pathways engaged by the various DNA alkylation adducts, which include base excision repair, direct damage reversal by MGMT and ALKBH, as well as nucleotide excision repair. Their roles in the protection against the genotoxic and carcinogenic effects of nitrosamines are highlighted. Finally, we address DNA translesion synthesis as a DNA damage tolerance mechanism relevant to DNA alkylation adducts.
- Published
- 2023
- Full Text
- View/download PDF
20. Analyzing the Effects of HDAC Inhibitors on DNA Damage and Associated Cytotoxicity in Primary Hepatocytes.
- Author
-
Carlsson MJ and Fahrer J
- Subjects
- Mice, Animals, DNA Damage, Liver, Cell Survival, Histone Deacetylase Inhibitors pharmacology, Hepatocytes metabolism
- Abstract
Primary hepatocytes are the gold standard in pharmaco- and toxicokinetic studies during preclinical development of drug candidates. Such cells are a valuable tool to identify potential hepatotoxicity, an important adverse drug reaction. Primary hepatocytes can be obtained not only from wild-type mice but also from genetically engineered knockout mouse strains. Liver perfusion yields murine primary hepatocytes (mpH) with high vitality, expressing an array of metabolic enzymes and transporters that are impaired or even absent in established liver cell lines. Furthermore, mpH display no genetic alterations and are proficient in the DNA damage response pathway. This makes mpH a suitable model to analyze the effects of histone deacetylase inhibitors on DNA damage and cell viability. Here, we report an efficient and fast protocol for the isolation of mpH by liver perfusion. These mpH can be used for downstream applications such as the detection of the DNA damage marker γH2AX by confocal laser scanning microscopy., (© 2023. The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2023
- Full Text
- View/download PDF
21. p53 triggers mitochondrial apoptosis following DNA damage-dependent replication stress by the hepatotoxin methyleugenol.
- Author
-
Carlsson MJ, Vollmer AS, Demuth P, Heylmann D, Reich D, Quarz C, Rasenberger B, Nikolova T, Hofmann TG, Christmann M, Fuhlbrueck JA, Stegmüller S, Richling E, Cartus AT, and Fahrer J
- Subjects
- Humans, bcl-2-Associated X Protein, DNA Damage, Apoptosis, Carcinogens, Tumor Suppressor Protein p53 genetics, DNA Adducts
- Abstract
Liver cancer is one of the most frequent tumor entities worldwide, which is causally linked to viral infection, fatty liver disease, life-style factors and food-borne carcinogens, particularly aflatoxins. Moreover, genotoxic plant toxins including phenylpropenes are suspected human liver carcinogens. The phenylpropene methyleugenol (ME) is a constituent of essential oils in many plants and occurs in herbal medicines, food, and cosmetics. Following its uptake, ME undergoes Cytochrome P450 (CYP) and sulfotransferase 1A1 (SULT1A1)-dependent metabolic activation, giving rise to DNA damage. However, little is known about the cellular response to the induced DNA adducts. Here, we made use of different SULT1A1-proficient cell models including primary hepatocytes that were treated with 1'-hydroxymethyleugenol (OH-ME) as main phase I metabolite. Firstly, mass spectrometry showed a concentration-dependent formation of N
2 -MIE-dG as major DNA adduct, strongly correlating with SULT1A1 expression as attested in cells with and without human SULT1A1. ME-derived DNA damage activated mainly the ATR-mediated DNA damage response as shown by phosphorylation of CHK1 and histone 2AX, followed by p53 accumulation and CHK2 phosphorylation. Consistent with these findings, the DNA adducts decreased replication speed and caused replication fork stalling. OH-ME treatment reduced viability particularly in cell lines with wild-type p53 and triggered apoptotic cell death, which was rescued by pan-caspase-inhibition. Further experiments demonstrated mitochondrial apoptosis as major cell death pathway. ME-derived DNA damage caused upregulation of the p53-responsive genes NOXA and PUMA, Bax activation, and cytochrome c release followed by caspase-9 and caspase-3 cleavage. We finally demonstrated the crucial role of p53 for OH-ME triggered cell death as evidenced by reduced pro-apoptotic gene expression, strongly attenuated Bax activation and cell death inhibition upon genetic knockdown or pharmacological inhibition of p53. Taken together, our study demonstrates for the first time that ME-derived DNA damage causes replication stress and triggers mitochondrial apoptosis via the p53-Bax pathway., (© 2022. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
22. The Essential Role of Rac1 Glucosylation in Clostridioides difficile Toxin B-Induced Arrest of G1-S Transition.
- Author
-
Petersen L, Stroh S, Schöttelndreier D, Grassl GA, Rottner K, Brakebusch C, Fahrer J, and Genth H
- Abstract
Clostridioides difficile infection (CDI) in humans causes pseudomembranous colitis (PMC), which is a severe pathology characterized by a loss of epithelial barrier function and massive colonic inflammation. PMC has been attributed to the action of two large protein toxins, Toxin A (TcdA) and Toxin B (TcdB). TcdA and TcdB mono-O-glucosylate and thereby inactivate a broad spectrum of Rho GTPases and (in the case of TcdA) also some Ras GTPases. Rho/Ras GTPases promote G1-S transition through the activation of components of the ERK, AKT, and WNT signaling pathways. With regard to CDI pathology, TcdB is regarded of being capable of inhibiting colonic stem cell proliferation and colonic regeneration, which is likely causative for PMC. In particular, it is still unclear, the glucosylation of which substrate Rho-GTPase is critical for TcdB-induced arrest of G1-S transition. Exploiting SV40-immortalized mouse embryonic fibroblasts (MEFs) with deleted Rho subtype GTPases, evidence is provided that Rac1 (not Cdc42) positively regulates Cyclin D1, an essential factor of G1-S transition. TcdB-catalyzed Rac1 glucosylation results in Cyclin D1 suppression and arrested G1-S transition in MEFs and in human colonic epithelial cells (HCEC), Remarkably, Rac1
-/- MEFs are insensitive to TcdB-induced arrest of G1-S transition, suggesting that TcdB arrests G1-S transition in a Rac1 glucosylation-dependent manner. Human intestinal organoids (HIOs) specifically expressed Cyclin D1 (neither Cyclin D2 nor Cyclin D3), which expression was suppressed upon TcdB treatment. In sum, Cyclin D1 expression in colonic cells seems to be regulated by Rho GTPases (most likely Rac1) and in turn seems to be susceptible to TcdB-induced suppression. With regard to PMC, toxin-catalyzed Rac1 glucosylation and subsequent G1-S arrest of colonic stem cells seems to be causative for decreased repair capacity of the colonic epithelium and delayed epithelial renewal., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Petersen, Stroh, Schöttelndreier, Grassl, Rottner, Brakebusch, Fahrer and Genth.)- Published
- 2022
- Full Text
- View/download PDF
23. How Do Children of Parents With Mental Illness Experience Stigma? A Systematic Mixed Studies Review.
- Author
-
Dobener LM, Fahrer J, Purtscheller D, Bauer A, Paul JL, and Christiansen H
- Abstract
Stigma can have devastating health and wellbeing impacts, not just on people with mental health problems, but on people associated with the stigmatized person. This is called stigma-by-association. Children whose parents have mental health problems are a particularly vulnerable group, and stigma acts as a mechanism, contributing to the transgenerational transmission of mental disorders. The current study is a systematic mixed studies review, synthesizing knowledge about how this group of children experience stigma-by-association. Overall, 32 studies were included, after a systematic search including quantitative, qualitatative, and mixed methods studies. The methodological quality was assessed and qualitative content analysis undertaken. We grouped children's stigma experiences into four dimensions, i.e., experienced stigma, anticipated stigma, internalized stigma, and structural discrimination. Results show that stigma is an important factor in those children's lives, and needs further investigation in qualitative and quantitative research. The current study emphasizes the importance of anti-stigma interventions and campaigns., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Dobener, Fahrer, Purtscheller, Bauer, Paul and Christiansen.)
- Published
- 2022
- Full Text
- View/download PDF
24. Expressed Emotion in the Family: A Meta-Analytic Review of Expressed Emotion as a Mechanism of the Transgenerational Transmission of Mental Disorders.
- Author
-
Fahrer J, Brill N, Dobener LM, Asbrand J, and Christiansen H
- Abstract
Background: High Expressed Emotion (HEE) has been identified as a risk factor for the exacerbation and course of mental illness. EE has been investigated as a caregiver's response to an offspring's problem behavior and pathology. The present meta-analysis regards EE from a transgenerational perspective and as one mechanism that might explain the transgenerational transmission of mental disorders., Method: We identified a total of 13 studies relying on 16 independent samples of parent-child dyads of parents with a mental illness and healthy controls; these were included in our analysis. Results were synthesized into one effect size per sample; meta-regression on additional effects of parental diagnostic category, child mental illness, and child age were also applied., Results: Parents with a mental illness are classified as HEE significantly more often. Effects were established for high criticism, albeit of small size (OR = 1.45), although they become stronger whenever offspring exhibit mental illness themselves (OR = 2.82)., Conclusion: The current study highlights the dearth of studies on EE in families in which a parent has a mental illness and its effects on their children. Our findings highlight EE as a potential mechanism for attributing the transgenerational transmission of mental disorders, especially for the EE-variable criticism, indicating dysfunctional parent-child interactions., Systematic Review Registration: http://www.crd.york.ac.uk/PROSPERO/display_record.php?ID=CRD42019117609, identifier: CRD42019117609., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Fahrer, Brill, Dobener, Asbrand and Christiansen.)
- Published
- 2022
- Full Text
- View/download PDF
25. Novel Insights into Pyrrolizidine Alkaloid Toxicity and Implications for Risk Assessment: Occurrence, Genotoxicity, Toxicokinetics, Risk Assessment-A Workshop Report.
- Author
-
Schrenk D, Fahrer J, Allemang A, Fu P, Lin G, Mahony C, Mulder PPJ, Peijnenburg A, Pfuhler S, Rietjens IMCM, Sachse B, Steinhoff B, These A, Troutman J, and Wiesner J
- Subjects
- Animals, Food Contamination analysis, Risk Assessment, Toxicokinetics, Pyrrolizidine Alkaloids toxicity, Teas, Herbal
- Abstract
This paper reports on the major contributions and results of the 2nd International Workshop of Pyrrolizidine Alkaloids held in September 2020 in Kaiserslautern, Germany. Pyrrolizidine alkaloids are among the most relevant plant toxins contaminating food, feed, and medicinal products of plant origin. Hundreds of PA congeners with widespread occurrence are known, and thousands of plants are assumed to contain PAs. Due to certain PAs' pronounced liver toxicity and carcinogenicity, their occurrence in food, feed, and phytomedicines has raised serious human health concerns. This is particularly true for herbal teas, certain food supplements, honey, and certain phytomedicinal drugs. Due to the limited availability of animal data, broader use of in vitro data appears warranted to improve the risk assessment of a large number of relevant, 1,2-unsaturated PAs. This is true, for example, for the derivation of both toxicokinetic and toxicodynamic data. These efforts aim to understand better the modes of action, uptake, metabolism, elimination, toxicity, and genotoxicity of PAs to enable a detailed dose-response analysis and ultimately quantify differing toxic potencies between relevant PAs. Accordingly, risk-limiting measures comprising production, marketing, and regulation of food, feed, and medicinal products are discussed., Competing Interests: The authors declare that they have no conflict of interest., (Thieme. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
26. The Mitochondrial Disruptor Devimistat (CPI-613) Synergizes with Genotoxic Anticancer Drugs in Colorectal Cancer Therapy in a Bim-Dependent Manner.
- Author
-
Arnold C, Demuth P, Seiwert N, Wittmann S, Boengler K, Rasenberger B, Christmann M, Huber M, Brunner T, Linnebacher M, and Fahrer J
- Subjects
- Animals, Antineoplastic Agents pharmacology, Caprylates pharmacology, Cell Line, Tumor, Colorectal Neoplasms mortality, Colorectal Neoplasms pathology, Humans, Male, Mice, Sulfides pharmacology, Survival Analysis, Antineoplastic Agents therapeutic use, Caprylates therapeutic use, Colorectal Neoplasms drug therapy, Sulfides therapeutic use
- Abstract
Colorectal cancer is one of the most frequent tumor entities, with an increasing incidence and mortality in younger adults in Europe and the United States. Five-year survival rates for advanced colorectal cancer are still low, highlighting the need for novel targets in colorectal cancer therapy. Here, we investigated the therapeutic potential of the compound devimistat (CPI-613) that targets altered mitochondrial cancer cell metabolism and its synergism with the antineoplastic drugs 5-fluorouracil (5-FU) and irinotecan (IT) in colorectal cancer. Devimistat exerted a comparable cytotoxicity in a panel of established colorectal cancer cell lines and patient-derived short-term cultures independent of their genetic and epigenetic status, whereas human colonic epithelial cells were more resistant, indicating tumor selectivity. These findings were corroborated in intestinal organoid and tumoroid models. Mechanistically, devimistat disrupted mitochondrial membrane potential and severely impaired mitochondrial respiration, resulting in colorectal cancer cell death induction independent of p53. Combination treatment of devimistat with 5-FU or IT demonstrated synergistic cell killing in colorectal cancer cells as shown by Combenefit modeling and Chou-Talalay analysis. Increased cell death induction was revealed as a major mechanism involving downregulation of antiapoptotic genes and accumulation of proapoptotic Bim, which was confirmed by its genetic knockdown. In human colorectal cancer xenograft mouse models, devimistat showed antitumor activity and synergized with IT, resulting in prolonged survival and enhanced therapeutic efficacy. In human tumor xenografts, devimistat prevented IT-triggered p53 stabilization and caused synergistic Bim induction. Taken together, our study revealed devimistat as a promising candidate in colorectal cancer therapy by synergizing with established antineoplastic drugs in vitro and in vivo ., (©2021 American Association for Cancer Research.)
- Published
- 2022
- Full Text
- View/download PDF
27. Repair of O6-carboxymethylguanine adducts by O6-methylguanine-DNA methyltransferase in human colon epithelial cells.
- Author
-
Kostka T, Empl MT, Seiwert N, Geisen SM, Hoffmann P, Adam J, Seeger B, Shay JW, Christmann M, Sturla SJ, Fahrer J, and Steinberg P
- Subjects
- Cell Line, Colon cytology, DNA Damage, DNA Repair, Guanine metabolism, Humans, Intestinal Mucosa cytology, Colon metabolism, Guanine analogs & derivatives, Intestinal Mucosa metabolism, O(6)-Methylguanine-DNA Methyltransferase metabolism
- Abstract
The protein O6-methylguanine-DNA methyltransferase (MGMT) is able to repair the mutagenic O6-methylguanine (O6-MeG) adduct back to guanine. In this context, it may protect against colorectal cancer formation associated with N-nitroso compounds. Such compounds may be endogenously formed by nitrosylation of amino acids, which can give rise to mutagenic O6-MeG and O6-carboxymethylguanine (O6-CMG) adducts. It is well established that O6-MeG is repaired by MGMT. However, up to now, whether O6-CMG is repaired by this enzyme remains unresolved. Therefore, the aim of the present study was to analyze the fate of both types of O6-guanine adducts in the presence and absence of MGMT activity. To this end, MGMT activity was efficiently blocked by its chemical inhibitor O6-benzylguanine in human colon epithelial cells (HCECs). Exposure of cells to azaserine (AZA) caused significantly higher levels of both O6-MeG and O6-CMG adducts in MGMT-inhibited cells, with O6-CMG as the more abundant DNA lesion. Interestingly, MGMT inhibition did not result in higher levels of AZA-induced DNA strand breaks in spite of elevated DNA adduct levels. In contrast, MGMT inhibition significantly increased DNA strand break formation after exposure to temozolomide (TMZ), a drug that exclusively generates O6-MeG adducts. In line with this finding, the viability of the cells was moderately reduced by TMZ upon MGMT inhibition, whereas no clear effect was observed in cells treated with AZA. In conclusion, our study clearly shows that O6-CMG is repaired by MGMT in HCEC, thereby suggesting that MGMT might play an important role as a tumor suppressor in diet-mediated colorectal cancer., (© The Author(s) 2021. Published by Oxford University Press. All rights reserved. For Permissions, please email: journals.permissions@oup.com.)
- Published
- 2021
- Full Text
- View/download PDF
28. STAT1 N-terminal domain discriminatively controls type I and type II IFN signaling.
- Author
-
Göder A, Ginter T, Heinzel T, Stroh S, Fahrer J, Henke A, and Krämer OH
- Subjects
- Antiviral Agents metabolism, Cell Line, Gene Expression Regulation physiology, HEK293 Cells, Humans, Phosphorylation physiology, Protein Transport physiology, STAT2 Transcription Factor metabolism, Interferon Type I metabolism, Interferon-gamma metabolism, STAT1 Transcription Factor metabolism, Signal Transduction physiology
- Abstract
The seven signal transducers of transcription (STATs) are cytokine-inducible modular transcription factors. They transmit the stimulation of cells with type I interferons (IFN-α/IFN-β) and type II interferon (IFN-ɣ) into altered gene expression patterns. The N-terminal domain (NTD) of STAT1 is a surface for STAT1/STAT1 homodimer and STAT1/STAT2 heterodimer formation and allows the cooperative DNA binding of STAT1. We investigated whether the STAT1 NTD-mediated dimerization affected the IFN-induced tyrosine phosphorylation of STAT1, its nuclear translocation, STAT1-dependent gene expression, and IFN-dependent antiviral defense. We reconstituted human STAT1-negative and STAT2-negative fibrosarcoma cells with STAT1, NTD-mutated STAT1 (STAT1AA), STAT1 with a mutated DNA-binding domain (DBD), or STAT2. We treated these cells with IFN-α and IFN-ɣ to assess differences between IFN-α-induced STAT1 homo- and heterodimers and IFN-ɣ-induced STAT1 homodimers. Our data demonstrate that IFNs induce the phosphorylation of STAT1 and STAT1AA at Y701 and their nuclear accumulation. We further reveal that STAT1AA can be phosphorylated in response to IFN-α in the absence of STAT2 and that IFN-ɣ-induced STAT1AA can activate gene expression directly. However, STAT1AA largely fails to bind STAT2 and to activate IFN-α-induced expression of endogenous antiviral STAT1/STAT2 target proteins. Congruent herewith, both an intact STAT1 NTD and STAT2 are indispensable to establish an antiviral state with IFN-α. These data provide new insights into the biological importance of the STAT1 NTD., (Copyright © 2021 Elsevier Ltd. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
29. Chronic intestinal inflammation drives colorectal tumor formation triggered by dietary heme iron in vivo.
- Author
-
Seiwert N, Adam J, Steinberg P, Wirtz S, Schwerdtle T, Adams-Quack P, Hövelmeyer N, Kaina B, Foersch S, and Fahrer J
- Subjects
- Animals, Diet, Inflammation pathology, Intestinal Mucosa pathology, Iron, Mice, RNA, Ribosomal, 16S, Colorectal Neoplasms chemically induced, Colorectal Neoplasms pathology, Heme toxicity
- Abstract
The consumption of red meat is associated with an increased risk for colorectal cancer (CRC). Multiple lines of evidence suggest that heme iron as abundant constituent of red meat is responsible for its carcinogenic potential. However, the underlying mechanisms are not fully understood and particularly the role of intestinal inflammation has not been investigated. To address this important issue, we analyzed the impact of heme iron (0.25 µmol/g diet) on the intestinal microbiota, gut inflammation and colorectal tumor formation in mice. An iron-balanced diet with ferric citrate (0.25 µmol/g diet) was used as reference. 16S rRNA sequencing revealed that dietary heme reduced α-diversity and caused a persistent intestinal dysbiosis, with a continuous increase in gram-negative Proteobacteria. This was linked to chronic gut inflammation and hyperproliferation of the intestinal epithelium as attested by mini-endoscopy, histopathology and immunohistochemistry. Dietary heme triggered the infiltration of myeloid cells into colorectal mucosa with an increased level of COX-2 positive cells. Furthermore, flow cytometry-based phenotyping demonstrated an increased number of T cells and B cells in the lamina propria following heme intake, while γδ-T cells were reduced in the intraepithelial compartment. Dietary heme iron catalyzed formation of fecal N-nitroso compounds and was genotoxic in intestinal epithelial cells, yet suppressed intestinal apoptosis as evidenced by confocal microscopy and western blot analysis. Finally, a chemically induced CRC mouse model showed persistent intestinal dysbiosis, chronic gut inflammation and increased colorectal tumorigenesis following heme iron intake. Altogether, this study unveiled intestinal inflammation as important driver in heme iron-associated colorectal carcinogenesis.
- Published
- 2021
- Full Text
- View/download PDF
30. Natural Merosesquiterpenes Activate the DNA Damage Response via DNA Strand Break Formation and Trigger Apoptotic Cell Death in p53-Wild-type and Mutant Colorectal Cancer.
- Author
-
Jiso A, Demuth P, Bachowsky M, Haas M, Seiwert N, Heylmann D, Rasenberger B, Christmann M, Dietrich L, Brunner T, Riyanti, Schäberle TF, Plubrukarn A, and Fahrer J
- Abstract
Colorectal cancer (CRC) is a frequently occurring malignant disease with still low survival rates, highlighting the need for novel therapeutics. Merosesquiterpenes are secondary metabolites from marine sponges, which might be useful as antitumor agents. To address this issue, we made use of a compound library comprising 11 isolated merosesquiterpenes. The most cytotoxic compounds were smenospongine > ilimaquinone ≈ dactylospontriol, as shown in different human CRC cell lines. Alkaline Comet assays and γH2AX immunofluorescence microscopy demonstrated DNA strand break formation in CRC cells. Western blot analysis revealed an activation of the DNA damage response with CHK1 phosphorylation, stabilization of p53 and p21, which occurred both in CRC cells with p53 knockout and in p53-mutated CRC cells. This resulted in cell cycle arrest followed by a strong increase in the subG1 population, indicative of apoptosis, and typical morphological alterations. In consistency, cell death measurements showed apoptosis following exposure to merosesquiterpenes. Gene expression studies and analysis of caspase cleavage revealed mitochondrial apoptosis via BAX , BIM , and caspase-9 as the main cell death pathway. Interestingly, the compounds were equally effective in p53-wild-type and p53-mutant CRC cells. Finally, the cytotoxic activity of the merosesquiterpenes was corroborated in intestinal tumor organoids, emphasizing their potential for CRC chemotherapy.
- Published
- 2021
- Full Text
- View/download PDF
31. Curcumin Administered as Micellar Solution Suppresses Intestinal Inflammation and Colorectal Carcinogenesis.
- Author
-
Seiwert N, Fahrer J, Nagel G, Frank J, Behnam D, and Kaina B
- Subjects
- Animals, Azoxymethane toxicity, Carcinogenesis, Dextran Sulfate toxicity, Disease Models, Animal, Inflammation drug therapy, Mice, Mice, Inbred C57BL, Micelles, Colitis chemically induced, Colitis drug therapy, Colorectal Neoplasms drug therapy, Colorectal Neoplasms prevention & control, Curcumin pharmacology
- Abstract
Colorectal cancer (CRC) is one of the most common cancers and preventive strategies based on natural compounds are highly desirable. Curcumin, the principal bioactive compound in Curcuma longa , was described to have multiple beneficial health effects. A drawback, however, is the low bioavailability due to its insolubility in water. Here, we studied whether nanoscaled micellar curcumin with improved bioavailability administered in drinking water reduces inflammation and CRC formation in a mouse model. C57BL6 wild-type (WT) mice and a strain defective in the DNA repair enzyme O
6 -methylguanine-DNA methyltransferase (MGMT) were used, in which tumors were induced by azoxymethane (AOM) followed by dextran sodium sulfate (DSS). Inflammation and tumor formation were determined by mini-colonoscopy. Micellar curcumin (mCur) administered in drinking water significantly reduced AOM/DSS-induced colorectal inflammation in both WT and MGMT-deficient mice as compared to animals receiving drinking water with micelles not containing curcumin. In line with this, the tumor yield and tumor score were significantly lower in mCur-treated mice compared to the control group. No adverse effects were observed in animals receiving mCur daily for at least three months. Overall, our data show that chronic oral administered micellar curcumin is well tolerated and reduces chemical-induced gut inflammation and CRC formation in mice. Impact: The study shows that micellar curcumin with high bioavailability chronically administered at low and physiologically relevant concentration suppresses inflammation and carcinogenesis in a mouse colorectal tumor model.- Published
- 2021
- Full Text
- View/download PDF
32. Synthesis and in vitro characterization of the genotoxic, mutagenic and cell-transforming potential of nitrosylated heme.
- Author
-
Kostka T, Fohrer J, Guigas C, Briviba K, Seiwert N, Fahrer J, Steinberg P, and Empl MT
- Subjects
- Animals, BALB 3T3 Cells, Caco-2 Cells, Carcinogenesis chemically induced, Cell Line, Comet Assay, Cricetinae, Heme chemistry, Humans, Mice, Mutagenesis, Mutation, Nitric Oxide chemistry, Red Meat toxicity, Risk Factors, Single-Cell Analysis, Cell Transformation, Neoplastic drug effects, DNA Damage drug effects, Heme toxicity, Intestinal Neoplasms chemically induced, Nitric Oxide toxicity
- Abstract
Data from epidemiological studies suggest that consumption of red and processed meat is a factor contributing to colorectal carcinogenesis. Red meat contains high amounts of heme, which in turn can be converted to its nitrosylated form, NO-heme, when adding nitrite-containing curing salt to meat. NO-heme might contribute to colorectal cancer formation by causing gene mutations and could thereby be responsible for the association of (processed) red meat consumption with intestinal cancer. Up to now, neither in vitro nor in vivo studies characterizing the mutagenic and cell transforming potential of NO-heme have been published due to the fact that the pure compound is not readily available. Therefore, in the present study, an already existing synthesis protocol was modified to yield, for the first time, purified NO-heme. Thereafter, newly synthesized NO-heme was chemically characterized and used in various in vitro approaches at dietary concentrations to determine whether it can lead to DNA damage and malignant cell transformation. While NO-heme led to a significant dose-dependent increase in the number of DNA strand breaks in the comet assay and was mutagenic in the HPRT assay, this compound tested negative in the Ames test and failed to induce malignant cell transformation in the BALB/c 3T3 cell transformation assay. Interestingly, the non-nitrosylated heme control showed similar effects, but was additionally able to induce malignant transformation in BALB/c 3T3 murine fibroblasts. Taken together, these results suggest that it is the heme molecule rather than the NO moiety which is involved in driving red meat-associated carcinogenesis.
- Published
- 2020
- Full Text
- View/download PDF
33. Heme oxygenase 1 protects human colonocytes against ROS formation, oxidative DNA damage and cytotoxicity induced by heme iron, but not inorganic iron.
- Author
-
Seiwert N, Wecklein S, Demuth P, Hasselwander S, Kemper TA, Schwerdtle T, Brunner T, and Fahrer J
- Subjects
- Cell Survival drug effects, Colon drug effects, Colon metabolism, Heme Oxygenase-1 metabolism, Hemin metabolism, Humans, Iron metabolism, Protective Agents pharmacology, DNA Damage drug effects, Heme Oxygenase-1 pharmacology, Iron pharmacology, Oxidative Stress drug effects, Reactive Oxygen Species metabolism
- Abstract
The consumption of red meat is probably carcinogenic to humans and is associated with an increased risk to develop colorectal cancer (CRC). Red meat contains high amounts of heme iron, which is thought to play a causal role in tumor formation. In this study, we investigated the genotoxic and cytotoxic effects of heme iron (i.e., hemin) versus inorganic iron in human colonic epithelial cells (HCEC), human CRC cell lines and murine intestinal organoids. Hemin catalyzed the formation of reactive oxygen species (ROS) and induced oxidative DNA damage as well as DNA strand breaks in both HCEC and CRC cells. In contrast, inorganic iron hardly affected ROS levels and only slightly increased DNA damage. Hemin, but not inorganic iron, caused cell death and reduced cell viability. This occurred preferentially in non-malignant HCEC, which was corroborated in intestinal organoids. Both hemin and inorganic iron were taken up into HCEC and CRC cells, however with differential kinetics and efficiency. Hemin caused stabilization and nuclear translocation of Nrf2, which induced heme oxygenase-1 (HO-1) and ferritin heavy chain (FtH). This was not observed after inorganic iron treatment. Chemical inhibition or genetic knockdown of HO-1 potentiated hemin-triggered ROS generation and oxidative DNA damage preferentially in HCEC. Furthermore, HO-1 abrogation strongly augmented the cytotoxic effects of hemin in HCEC, revealing its pivotal function in colonocytes and highlighting the toxicity of free intracellular heme iron. Taken together, this study demonstrated that hemin, but not inorganic iron, induces ROS and DNA damage, resulting in a preferential cytotoxicity in non-malignant intestinal epithelial cells. Importantly, HO-1 conferred protection against the detrimental effects of hemin.
- Published
- 2020
- Full Text
- View/download PDF
34. Targeting Altered Energy Metabolism in Colorectal Cancer: Oncogenic Reprogramming, the Central Role of the TCA Cycle and Therapeutic Opportunities.
- Author
-
Neitzel C, Demuth P, Wittmann S, and Fahrer J
- Abstract
Colorectal cancer (CRC) is among the most frequent cancer entities worldwide. Multiple factors are causally associated with CRC development, such as genetic and epigenetic alterations, inflammatory bowel disease, lifestyle and dietary factors. During malignant transformation, the cellular energy metabolism is reprogrammed in order to promote cancer cell growth and proliferation. In this review, we first describe the main alterations of the energy metabolism found in CRC, revealing the critical impact of oncogenic signaling and driver mutations in key metabolic enzymes. Then, the central role of mitochondria and the tricarboxylic acid (TCA) cycle in this process is highlighted, also considering the metabolic crosstalk between tumor and stromal cells in the tumor microenvironment. The identified cancer-specific metabolic transformations provided new therapeutic targets for the development of small molecule inhibitors. Promising agents are in clinical trials and are directed against enzymes of the TCA cycle, including isocitrate dehydrogenase, pyruvate dehydrogenase kinase, pyruvate dehydrogenase complex (PDC) and α-ketoglutarate dehydrogenase (KGDH). Finally, we focus on the α-lipoic acid derivative CPI-613, an inhibitor of both PDC and KGDH, and delineate its anti-tumor effects for targeted therapy.
- Published
- 2020
- Full Text
- View/download PDF
35. Switching off DNA repair-how colorectal cancer evades targeted therapies through adaptive mutability.
- Author
-
Fahrer J
- Subjects
- Colorectal Neoplasms pathology, Humans, Mutation genetics, Colorectal Neoplasms genetics, DNA Repair genetics, Molecular Targeted Therapy
- Published
- 2020
- Full Text
- View/download PDF
36. Mechanism of colorectal carcinogenesis triggered by heme iron from red meat.
- Author
-
Seiwert N, Heylmann D, Hasselwander S, and Fahrer J
- Subjects
- Carcinogenesis genetics, Carcinogenesis pathology, Colorectal Neoplasms genetics, Colorectal Neoplasms pathology, DNA Damage, Diet, Humans, Risk Factors, Carcinogenesis metabolism, Colorectal Neoplasms metabolism, Heme metabolism, Iron metabolism, Red Meat
- Abstract
Colorectal cancer (CRC) is one of the major tumor entities worldwide, with an increasing incidence in younger people. CRC formation is causally linked to various genetic, life-style and dietary risk factors. Among the ladder, the consumption of red meat has emerged as important risk factor contributing to CRC. A large body of evidence shows that heme iron is the critical component of red meat, which promotes colorectal carcinogenesis. In this review, we describe the uptake and cellular fate of both heme and inorganic iron in intestinal epithelial cells. Next, an overview on the DNA damaging properties of heme iron is provided, highlighting the DNA adducts relevant for CRC etiology. Moreover, heme triggered mechanisms leading to colonic hyperproliferation are presented, which are intimately linked to changes in the intestinal microbiota induced by heme. A special focus was set on the impact of heme iron on innate and adaptive immune cells, which could be relevant in the context of CRC. Finally, we recapitulate in vivo studies providing evidence for the tumor-promoting potential of dietary heme iron. Altogether, heme iron affects numerous key pathways involved in the pathogenesis of CRC., (Copyright © 2019 Elsevier B.V. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
37. Formation of trans-epoxy fatty acids correlates with formation of isoprostanes and could serve as biomarker of oxidative stress.
- Author
-
Rund KM, Heylmann D, Seiwert N, Wecklein S, Oger C, Galano JM, Durand T, Chen R, Gueler F, Fahrer J, Bornhorst J, and Schebb NH
- Subjects
- Animals, Caenorhabditis elegans, Kidney injuries, Male, Mice, Reperfusion Injury metabolism, Biomarkers metabolism, Isoprostanes biosynthesis, Oxidative Stress, Trans Fatty Acids biosynthesis
- Abstract
In mammals, epoxy-polyunsaturated fatty acids (epoxy-PUFA) are enzymatically formed from naturally occurring all-cis PUFA by cytochrome P450 monooxygenases leading to the generation of cis-epoxy-PUFA (mixture of R,S- and S,R-enantiomers). In addition, also non-enzymatic chemical peroxidation gives rise to epoxy-PUFA leading to both, cis- and trans-epoxy-PUFA (mixture of R,R- and S,S-enantiomers). Here, we investigated for the first time trans-epoxy-PUFA and the trans/cis-epoxy-PUFA ratio as potential new biomarker of lipid peroxidation. Their formation was analyzed in correlation with the formation of isoprostanes (IsoP), which are commonly used as biomarkers of oxidative stress. Five oxidative stress models were investigated including incubations of three human cell lines as well as the in vivo model Caenorhabditis elegans with tert-butyl hydroperoxide (t-BOOH) and analysis of murine kidney tissue after renal ischemia reperfusion injury (IRI). A comprehensive set of IsoP and epoxy-PUFA derived from biologically relevant PUFA (ARA, EPA and DHA) was simultaneously quantified by LC-ESI(-)-MS/MS. Following renal IRI only a moderate increase in the kidney levels of IsoP and no relevant change in the trans/cis-epoxy-PUFA ratio was observed. In all investigated cell lines (HCT-116, HepG2 and Caki-2) as well as C. elegans a dose dependent increase of both, IsoP and the trans/cis-epoxy-PUFA ratio in response to the applied t-BOOH was observed. The different cell lines showed a distinct time dependent pattern consistent for both classes of autoxidatively formed oxylipins. Clear and highly significant correlations of the trans/cis-epoxy-PUFA ratios with the IsoP levels were found in all investigated cell lines and C. elegans. Based on this, we suggest the trans/cis-epoxy-PUFA ratio as potential new biomarker of oxidative stress, which warrants further investigation., (Copyright © 2019 Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
38. Lipoic Acid Synergizes with Antineoplastic Drugs in Colorectal Cancer by Targeting p53 for Proteasomal Degradation.
- Author
-
Neitzel C, Seiwert N, Göder A, Diehl E, Weber C, Nagel G, Stroh S, Rasenberger B, Christmann M, and Fahrer J
- Subjects
- Cell Line, Tumor, Cell Proliferation drug effects, Cell Survival drug effects, Colorectal Neoplasms drug therapy, Colorectal Neoplasms genetics, Doxorubicin pharmacology, Drug Synergism, Energy Metabolism drug effects, Fluorouracil pharmacology, HCT116 Cells, Humans, Mutation, Proteolysis, Tumor Suppressor Protein p53 genetics, Ubiquitination, Antineoplastic Agents pharmacology, Colorectal Neoplasms metabolism, Proteasome Endopeptidase Complex metabolism, Thioctic Acid pharmacology, Tumor Suppressor Protein p53 metabolism
- Abstract
Lipoic acid (LA) is a redox-active disulphide compound, which functions as a pivotal co-factor for mitochondrial oxidative decarboxylation. LA and chemical derivatives were shown to target mitochondria in cancer cells with altered energy metabolism, thereby inducing cell death. In this study, the impact of LA on the tumor suppressor protein p53 was analyzed in various colorectal cancer (CRC) cell lines, with a focus on the mechanisms driving p53 degradation. First, LA was demonstrated to trigger the depletion of both wildtype and mutant p53 protein in all CRC cells tested without influencing its gene expression and preceded LA-triggered cytotoxicity. Depletion of p53 coincided with a moderate, LA-dependent ROS production, but was not rescued by antioxidant treatment. LA induced the autophagy receptor p62 and differentially modulated autophagosome formation in CRC cells. However, p53 degradation was not mediated via autophagy as shown by chemical inhibition and genetic abrogation of autophagy. LA treatment also stabilized and activated the transcription factor Nrf2 in CRC cells, which was however dispensable for p53 degradation. Mechanistically, p53 was found to be readily ubiquitinylated and degraded by the proteasomal machinery following LA treatment, which did not involve the E3 ubiquitin ligase MDM2. Intriguingly, the combination of LA and anticancer drugs (doxorubicin, 5-fluorouracil) attenuated p53-mediated stabilization of p21 and resulted in synergistic killing in CRC cells in a p53-dependant manner.
- Published
- 2019
- Full Text
- View/download PDF
39. Immunological and mass spectrometry-based approaches to determine thresholds of the mutagenic DNA adduct O 6 -methylguanine in vivo.
- Author
-
Kraus A, McKeague M, Seiwert N, Nagel G, Geisen SM, Ziegler N, Trantakis IA, Kaina B, Thomas AD, Sturla SJ, and Fahrer J
- Subjects
- Animals, Antineoplastic Agents, Alkylating administration & dosage, Azoxymethane administration & dosage, DNA Adducts immunology, DNA Modification Methylases genetics, DNA Modification Methylases metabolism, DNA Repair Enzymes genetics, DNA Repair Enzymes metabolism, Dose-Response Relationship, Drug, Guanine analysis, Guanine immunology, HCT116 Cells, Humans, Immunoblotting methods, Leukocytes, Mononuclear drug effects, Mice, Inbred C57BL, Mice, Mutant Strains, Microscopy, Fluorescence methods, Sensitivity and Specificity, Temozolomide administration & dosage, Tumor Suppressor Proteins genetics, Tumor Suppressor Proteins metabolism, Chromatography, Liquid methods, DNA Adducts analysis, Guanine analogs & derivatives, Tandem Mass Spectrometry methods
- Abstract
N-nitroso compounds are alkylating agents, which are widespread in our diet and the environment. They induce DNA alkylation adducts such as O
6 -methylguanine (O6 -MeG), which is repaired by O6 -methylguanine-DNA methyltransferase (MGMT). Persistent O6 -MeG lesions have detrimental biological consequences like mutagenicity and cytotoxicity. Due to its pivotal role in the etiology of cancer and in cytotoxic cancer therapy, it is important to detect and quantify O6 -MeG in biological specimens in a sensitive and accurate manner. Here, we used immunological approaches and established an ultra performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS) to monitor O6 -MeG adducts. First, colorectal cancer (CRC) cells were treated with the methylating anticancer drug temozolomide (TMZ). Immunofluorescence microscopy and an immuno-slot blot assay, both based on an adduct-specific antibody, allowed for the semi-quantitative, dose-dependent assessment of O6 -MeG in CRC cells. Using the highly sensitive and specific UPLC-MS/MS, TMZ-induced O6 -MeG adducts were quantified in CRC cells and even in peripheral blood mononuclear cells exposed to clinically relevant TMZ doses. Furthermore, all methodologies were used to detect O6 -MeG in wildtype (WT) and MGMT-deficient mice challenged with the carcinogen azoxymethane. UPLC-MS/MS measurements and dose-response modeling revealed a non-linear formation of hepatic and colonic O6 -MeG adducts in WT, whereas linear O6 -MeG formation without a threshold was observed in MGMT-deficient mice. Collectively, the UPLC-MS/MS analysis is highly sensitive and specific for O6 -MeG, thereby allowing for the first time for the determination of a genotoxic threshold upon exposure to O6 -methylating agents. We envision that this method will be instrumental to monitor the efficacy of methylating chemotherapy and to assess dietary exposures.- Published
- 2019
- Full Text
- View/download PDF
40. Endothelial Leptin Receptor Deletion Promotes Cardiac Autophagy and Angiogenesis Following Pressure Overload by Suppressing Akt/mTOR Signaling.
- Author
-
Gogiraju R, Hubert A, Fahrer J, Straub BK, Brandt M, Wenzel P, Münzel T, Konstantinides S, Hasenfuss G, and Schäfer K
- Subjects
- Animals, Cardiomegaly genetics, Cardiomegaly pathology, Cardiomegaly physiopathology, Cells, Cultured, Disease Models, Animal, Endothelial Cells pathology, Female, Fibrosis, Gene Deletion, Genetic Predisposition to Disease, Heart Failure genetics, Heart Failure pathology, Heart Failure physiopathology, Humans, Mice, Knockout, Myocardium pathology, Phenotype, Receptors, Leptin genetics, Receptors, Leptin metabolism, Signal Transduction, Ventricular Function, Left, Autophagy, Cardiomegaly enzymology, Endothelial Cells enzymology, Heart Failure enzymology, Myocardium enzymology, Neovascularization, Pathologic, Proto-Oncogene Proteins c-akt metabolism, Receptors, Leptin deficiency, TOR Serine-Threonine Kinases metabolism
- Abstract
Background: Cardiac remodeling is modulated by overnutrition or starvation. The adipokine leptin mediates energy balance between adipose tissue and brain. Leptin and its receptors are expressed in the heart., Methods and Results: To examine the importance of endothelial leptin signaling in cardiac hypertrophy, transverse aortic constriction was used in mice with inducible endothelium-specific deletion of leptin receptors (End.LepR-KO) or littermate controls (End.LepR-WT). End.LepR-KO was associated with improved left ventricular function (fractional shortening, 28.4% versus 18.8%; P=0.0114), reduced left ventricular dilation (end-systolic inner left ventricular diameter, 3.59 versus 4.08 mm; P=0.0188) and lower heart weight (133 versus 173 mg; P<0.0001) 20 weeks after transverse aortic constriction. Histology and quantitative polymerase chain reaction analysis confirmed reduced cardiomyocyte hypertrophy. STAT3 (signal transducer and activator of transcription) activation was reduced, and Akt (protein kinase B) and mTOR (mammalian target of rapamycin) phosphorylation after transverse aortic constriction were blunted in End.LepR-KO hearts. Elevated LC3 (microtubule associated protein 1 light chain 3)-I/-II conversion ( P=0.0041) and increased (LC3II-positive) endothelial cells ( P=0.0042) in banded hearts of End.LepR-KO mice suggested improved cardiac angiogenesis because of activated autophagy. Microscopy confirmed autophagosome accumulation after genetic or small interfering RNA-mediated LepR downregulation. Enhanced sprouting angiogenesis was observed in endothelial cells ( P<0.0001) and aortic rings ( P=0.0060) from End.LepR-KO mice, and murine and human endothelial sprouting angiogenesis was reduced after mTOR inhibition using rapamycin or autophagy inhibition using 3-methyladenine. Banded End.LepR-KO mouse hearts exhibited less apoptosis ( P=0.0218), inflammation ( P=0.0251), and fibrosis ( P=0.0256). Reduced endothelial autophagy was also observed in myocardial biopsies of heart failure patients with cardiac fibrosis., Conclusions: Our findings suggest that endothelial leptin signaling contributes to cardiac fibrosis and functional deterioration by suppressing endothelial autophagy and promoting endothelial dysfunction in a chronic pressure overload model.
- Published
- 2019
- Full Text
- View/download PDF
41. Sensitivity of CD3/CD28-stimulated versus non-stimulated lymphocytes to ionizing radiation and genotoxic anticancer drugs: key role of ATM in the differential radiation response.
- Author
-
Heylmann D, Badura J, Becker H, Fahrer J, and Kaina B
- Subjects
- Amino Acid Chloromethyl Ketones pharmacology, Antibodies pharmacology, Ataxia Telangiectasia Mutated Proteins antagonists & inhibitors, Ataxia Telangiectasia Mutated Proteins genetics, CD28 Antigens antagonists & inhibitors, CD28 Antigens genetics, CD3 Complex antagonists & inhibitors, CD3 Complex genetics, Caspases genetics, Caspases immunology, Cell Proliferation drug effects, Cell Proliferation radiation effects, Chromones pharmacology, DNA-Activated Protein Kinase antagonists & inhibitors, DNA-Activated Protein Kinase genetics, DNA-Activated Protein Kinase immunology, Drug Resistance genetics, Drug Resistance immunology, Gene Expression Regulation, Humans, Isoxazoles pharmacology, Lymphocyte Activation drug effects, Lymphocyte Activation radiation effects, MRE11 Homologue Protein antagonists & inhibitors, MRE11 Homologue Protein genetics, MRE11 Homologue Protein immunology, Morpholines pharmacology, Primary Cell Culture, Pyrazines pharmacology, Pyrones pharmacology, Radiation Tolerance genetics, Radiation Tolerance immunology, Signal Transduction, T-Lymphocytes, Cytotoxic cytology, T-Lymphocytes, Cytotoxic drug effects, T-Lymphocytes, Cytotoxic radiation effects, T-Lymphocytes, Regulatory cytology, T-Lymphocytes, Regulatory drug effects, T-Lymphocytes, Regulatory radiation effects, Thiophenes pharmacology, Thioxanthenes pharmacology, Ataxia Telangiectasia Mutated Proteins immunology, CD28 Antigens immunology, CD3 Complex immunology, Gamma Rays, T-Lymphocytes, Cytotoxic immunology, T-Lymphocytes, Regulatory immunology
- Abstract
Activation of T cells, a major fraction of peripheral blood lymphocytes (PBLCS), is essential for the immune response. Genotoxic stress resulting from ionizing radiation (IR) and chemical agents, including anticancer drugs, has serious impact on T cells and, therefore, on the immune status. Here we compared the sensitivity of non-stimulated (non-proliferating) vs. CD3/CD28-stimulated (proliferating) PBLC to IR. PBLCs were highly sensitive to IR and, surprisingly, stimulation to proliferation resulted in resistance to IR. Radioprotection following CD3/CD28 activation was observed in different T-cell subsets, whereas stimulated CD34+ progenitor cells did not become resistant to IR. Following stimulation, PBLCs showed no significant differences in the repair of IR-induced DNA damage compared with unstimulated cells. Interestingly, ATM is expressed at high level in resting PBLCs and CD3/CD28 stimulation leads to transcriptional downregulation and reduced ATM phosphorylation following IR, indicating ATM to be key regulator of the high radiosensitivity of resting PBLCs. In line with this, pharmacological inhibition of ATM caused radioresistance of unstimulated, but not stimulated, PBLCs. Radioprotection was also achieved by inhibition of MRE11 and CHK1/CHK2, supporting the notion that downregulation of the MRN-ATM-CHK pathway following CD3/CD28 activation results in radioprotection of proliferating PBLCs. Interestingly, the crosslinking anticancer drug mafosfamide induced, like IR, more death in unstimulated than in stimulated PBLCs. In contrast, the bacterial toxin CDT, damaging DNA through inherent DNase activity, and the DNA methylating anticancer drug temozolomide induced more death in CD3/CD28-stimulated than in unstimulated PBLCs. Thus, the sensitivity of stimulated vs. non-stimulated lymphocytes to genotoxins strongly depends on the kind of DNA damage induced. This is the first study in which the killing response of non-proliferating vs. proliferating T cells was comparatively determined. The data provide insights on how immunotherapeutic strategies resting on T-cell activation can be impacted by differential cytotoxic effects resulting from radiation and chemotherapy.
- Published
- 2018
- Full Text
- View/download PDF
42. PARP-1 protects against colorectal tumor induction, but promotes inflammation-driven colorectal tumor progression.
- Author
-
Dörsam B, Seiwert N, Foersch S, Stroh S, Nagel G, Begaliew D, Diehl E, Kraus A, McKeague M, Minneker V, Roukos V, Reißig S, Waisman A, Moehler M, Stier A, Mangerich A, Dantzer F, Kaina B, and Fahrer J
- Subjects
- Animals, Cell Line, Tumor, Colorectal Neoplasms genetics, Colorectal Neoplasms pathology, Colorectal Neoplasms prevention & control, Guanine analogs & derivatives, Guanine metabolism, Humans, Mice, Mice, Knockout, Poly (ADP-Ribose) Polymerase-1 genetics, Tumor Suppressor Proteins genetics, Colorectal Neoplasms enzymology, Poly (ADP-Ribose) Polymerase-1 metabolism, Tumor Suppressor Proteins metabolism
- Abstract
Colorectal cancer (CRC) is one of the most common tumor entities, which is causally linked to DNA repair defects and inflammatory bowel disease (IBD). Here, we studied the role of the DNA repair protein poly(ADP-ribose) polymerase-1 (PARP-1) in CRC. Tissue microarray analysis revealed PARP-1 overexpression in human CRC, correlating with disease progression. To elucidate its function in CRC, PARP-1 deficient (PARP-1
-/- ) and wild-type animals (WT) were subjected to azoxymethane (AOM)/ dextran sodium sulfate (DSS)-induced colorectal carcinogenesis. Miniendoscopy showed significantly more tumors in WT than in PARP-1-/- mice. Although the lack of PARP-1 moderately increased DNA damage, both genotypes exhibited comparable levels of AOM-induced autophagy and cell death. Interestingly, miniendoscopy revealed a higher AOM/DSS-triggered intestinal inflammation in WT animals, which was associated with increased levels of innate immune cells and proinflammatory cytokines. Tumors in WT animals were more aggressive, showing higher levels of STAT3 activation and cyclin D1 up-regulation. PARP-1-/- animals were then crossed with O6 -methylguanine-DNA methyltransferase (MGMT)-deficient animals hypersensitive to AOM. Intriguingly, PARP-1-/- /MGMT-/- double knockout (DKO) mice developed more, but much smaller tumors than MGMT-/- animals. In contrast to MGMT-deficient mice, DKO animals showed strongly reduced AOM-dependent colonic cell death despite similar O6 -methylguanine levels. Studies with PARP-1-/- cells provided evidence for increased alkylation-induced DNA strand break formation when MGMT was inhibited, suggesting a role of PARP-1 in the response to O6 -methylguanine adducts. Our findings reveal PARP-1 as a double-edged sword in colorectal carcinogenesis, which suppresses tumor initiation following DNA alkylation in a MGMT-dependent manner, but promotes inflammation-driven tumor progression., Competing Interests: The authors declare no conflict of interest.- Published
- 2018
- Full Text
- View/download PDF
43. AKT2 suppresses pro-survival autophagy triggered by DNA double-strand breaks in colorectal cancer cells.
- Author
-
Seiwert N, Neitzel C, Stroh S, Frisan T, Audebert M, Toulany M, Kaina B, and Fahrer J
- Subjects
- Ataxia Telangiectasia Mutated Proteins genetics, Ataxia Telangiectasia Mutated Proteins metabolism, Autophagy drug effects, Autophagy genetics, Bacterial Toxins pharmacology, Cell Line, Tumor, Colorectal Neoplasms metabolism, Colorectal Neoplasms pathology, Gene Knockdown Techniques, HCT116 Cells, Humans, Proto-Oncogene Proteins c-akt metabolism, Signal Transduction, Transfection, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Protein p53 metabolism, Colorectal Neoplasms genetics, DNA Breaks, Double-Stranded, Proto-Oncogene Proteins c-akt genetics
- Abstract
DNA double-strand breaks (DSBs) are critical DNA lesions, which threaten genome stability and cell survival. DSBs are directly induced by ionizing radiation (IR) and radiomimetic agents, including the cytolethal distending toxin (CDT). This bacterial genotoxin harbors a unique DNase-I-like endonuclease activity. Here we studied the role of DSBs induced by CDT and IR as a trigger of autophagy, which is a cellular degradation process involved in cell homeostasis, genome protection and cancer. The regulatory mechanisms of DSB-induced autophagy were analyzed, focusing on the ATM-p53-mediated DNA damage response and AKT signaling in colorectal cancer cells. We show that treatment of cells with CDT or IR increased the levels of the autophagy marker LC3B-II. Consistently, an enhanced formation of autophagosomes and a decrease of the autophagy substrate p62 were observed. Both CDT and IR concomitantly suppressed mTOR signaling and stimulated the autophagic flux. DSBs were demonstrated as the primary trigger of autophagy using a DNase I-defective CDT mutant, which neither induced DSBs nor autophagy. Genetic abrogation of p53 and inhibition of ATM signaling impaired the autophagic flux as revealed by LC3B-II accumulation and reduced formation of autophagic vesicles. Blocking of DSB-induced apoptotic cell death by the pan-caspase inhibitor Z-VAD stimulated autophagy. In line with this, pharmacological inhibition of autophagy increased cell death, while ATG5 knockdown did not affect cell death after DSB induction. Interestingly, both IR and CDT caused AKT activation, which repressed DSB-triggered autophagy independent of the cellular DNA-PK status. Further knockdown and pharmacological inhibitor experiments provided evidence that the negative autophagy regulation was largely attributable to AKT2. Finally, we show that upregulation of CDT-induced autophagy upon AKT inhibition resulted in lower apoptosis and increased cell viability. Collectively, the findings demonstrate that DSBs trigger pro-survival autophagy in an ATM- and p53-dependent manner, which is curtailed by AKT2 signaling.
- Published
- 2017
- Full Text
- View/download PDF
44. Impact of DNA repair on the dose-response of colorectal cancer formation induced by dietary carcinogens.
- Author
-
Fahrer J and Kaina B
- Subjects
- Animals, DNA Adducts genetics, DNA Damage, Humans, Meat analysis, Carcinogens toxicity, Colorectal Neoplasms etiology, Colorectal Neoplasms genetics, DNA Repair, Food Contamination analysis
- Abstract
Colorectal cancer (CRC) is one of the most frequently diagnosed cancers, which is causally linked to dietary habits, notably the intake of processed and red meat. Processed and red meat contain dietary carcinogens, including heterocyclic aromatic amines (HCAs) and N-nitroso compounds (NOC). NOC are agents that induce various N-methylated DNA adducts and O
6 -methylguanine (O6 -MeG), which are removed by base excision repair (BER) and O6 -methylguanine-DNA methyltransferase (MGMT), respectively. HCAs such as the highly mutagenic 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP) cause bulky DNA adducts, which are removed from DNA by nucleotide excision repair (NER). Both O6 -MeG and HCA-induced DNA adducts are linked to the occurrence of KRAS and APC mutations in colorectal tumors of rodents and humans, thereby driving CRC initiation and progression. In this review, we focus on DNA repair pathways removing DNA lesions induced by NOC and HCA and assess their role in protecting against mutagenicity and carcinogenicity in the large intestine. We further discuss the impact of DNA repair on the dose-response relationship in colorectal carcinogenesis in view of recent studies, demonstrating the existence of 'no effect' point of departures (PoDs), i.e. thresholds for genotoxicity and carcinogenicity. The available data support the threshold concept for NOC with DNA repair being causally involved., (Copyright © 2016 Elsevier Ltd. All rights reserved.)- Published
- 2017
- Full Text
- View/download PDF
45. DNA damage response curtails detrimental replication stress and chromosomal instability induced by the dietary carcinogen PhIP.
- Author
-
Mimmler M, Peter S, Kraus A, Stroh S, Nikolova T, Seiwert N, Hasselwander S, Neitzel C, Haub J, Monien BH, Nicken P, Steinberg P, Shay JW, Kaina B, and Fahrer J
- Subjects
- Animals, Ataxia Telangiectasia Mutated Proteins metabolism, Cattle, Cell Line, Cell Survival drug effects, Cell Survival genetics, Checkpoint Kinase 1 metabolism, Chromosome Aberrations, Cricetinae, DNA Adducts, DNA Breaks, Double-Stranded, Discoidin Domain Receptors metabolism, Fibroblasts drug effects, Fibroblasts metabolism, Humans, Phosphorylation, Signal Transduction drug effects, Carcinogens toxicity, Chromosomal Instability drug effects, DNA Damage drug effects, DNA Replication drug effects, Imidazoles toxicity, Stress, Physiological drug effects, Stress, Physiological genetics
- Abstract
PhIP is an abundant heterocyclic aromatic amine (HCA) and important dietary carcinogen. Following metabolic activation, PhIP causes bulky DNA lesions at the C8-position of guanine. Although C8-PhIP-dG adducts are mutagenic, their interference with the DNA replication machinery and the elicited DNA damage response (DDR) have not yet been studied. Here, we analyzed PhIP-triggered replicative stress and elucidated the role of the apical DDR kinases ATR, ATM and DNA-PK
cs in the cellular defense response. First, we demonstrate that PhIP induced C8-PhIP-dG adducts and DNA strand breaks. This stimulated ATR-CHK1 signaling, phosphorylation of histone 2AX and the formation of RPA foci. In proliferating cells, PhIP treatment increased the frequency of stalled replication forks and reduced fork speed. Inhibition of ATR in the presence of PhIP-induced DNA damage strongly promoted the formation of DNA double-strand breaks, activation of the ATM-CHK2 pathway and hyperphosphorylation of RPA. The abrogation of ATR signaling potentiated the cell death response and enhanced chromosomal aberrations after PhIP treatment, while ATM and DNA-PK inhibition had only marginal effects. These results strongly support the notion that ATR plays a key role in the defense against cancer formation induced by PhIP and related HCAs., (© The Author(s) 2016. Published by Oxford University Press on behalf of Nucleic Acids Research.)- Published
- 2016
- Full Text
- View/download PDF
46. Nitroglycerin induces DNA damage and vascular cell death in the setting of nitrate tolerance.
- Author
-
Mikhed Y, Fahrer J, Oelze M, Kröller-Schön S, Steven S, Welschof P, Zinßius E, Stamm P, Kashani F, Roohani S, Kress JM, Ullmann E, Tran LP, Schulz E, Epe B, Kaina B, Münzel T, and Daiber A
- Subjects
- Animals, Blotting, Western, Comet Assay, Disease Models, Animal, Immunoblotting, Immunohistochemistry, Mice, Oxidative Stress, Rats, Rats, Wistar, DNA Damage, Drug Tolerance physiology, Endothelium, Vascular drug effects, Nitroglycerin toxicity, Vasodilator Agents toxicity
- Abstract
Nitroglycerin (GTN) and other organic nitrates are widely used vasodilators. Their side effects are development of nitrate tolerance and endothelial dysfunction. Given the potential of GTN to induce nitro-oxidative stress, we investigated the interaction between nitro-oxidative DNA damage and vascular dysfunction in experimental nitrate tolerance. Cultured endothelial hybridoma cells (EA.hy 926) and Wistar rats were treated with GTN (ex vivo: 10-1000 µM; in vivo: 10, 20 and 50 mg/kg/day for 3 days, s.c.). The level of DNA strand breaks, 8-oxoguanine and O (6)-methylguanine DNA adducts was determined by Comet assay, dot blot and immunohistochemistry. Vascular function was determined by isometric tension recording. DNA adducts and strand breaks were induced by GTN in cells in vitro in a concentration-dependent manner. GTN in vivo administration leads to endothelial dysfunction, nitrate tolerance, aortic and cardiac oxidative stress, formation of DNA adducts, stabilization of p53 and apoptotic death of vascular cells in a dose-dependent fashion. Mice lacking O (6)-methylguanine-DNA methyltransferase displayed more vascular O (6)-methylguanine adducts and oxidative stress under GTN therapy than wild-type mice. Although we were not able to prove a causal role of DNA damage in the etiology of nitrate tolerance, the finding of GTN-induced DNA damage such as the mutagenic and toxic adduct O (6)-methylguanine, and cell death supports the notion that GTN based therapy may provoke adverse side effects, including endothelial function. Further studies are warranted to clarify whether GTN pro-apoptotic effects are related to an impaired recovery of patients upon myocardial infarction.
- Published
- 2016
- Full Text
- View/download PDF
47. Enhanced Histone Deacetylase Activity in Malignant Melanoma Provokes RAD51 and FANCD2-Triggered Drug Resistance.
- Author
-
Krumm A, Barckhausen C, Kücük P, Tomaszowski KH, Loquai C, Fahrer J, Krämer OH, Kaina B, and Roos WP
- Subjects
- Animals, Apoptosis drug effects, Blotting, Western, Cell Proliferation drug effects, DNA Breaks, Double-Stranded drug effects, DNA Repair drug effects, Fanconi Anemia Complementation Group D2 Protein antagonists & inhibitors, Fanconi Anemia Complementation Group D2 Protein genetics, Fluorescent Antibody Technique, Histone Deacetylases chemistry, Histone Deacetylases genetics, Homologous Recombination drug effects, Humans, Immunoenzyme Techniques, Melanoma drug therapy, Melanoma metabolism, Mice, Mice, Inbred NOD, Mice, SCID, RNA, Messenger genetics, Rad51 Recombinase antagonists & inhibitors, Rad51 Recombinase genetics, Real-Time Polymerase Chain Reaction, Reverse Transcriptase Polymerase Chain Reaction, Skin drug effects, Skin metabolism, Skin pathology, Skin Neoplasms drug therapy, Skin Neoplasms metabolism, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Melanoma, Cutaneous Malignant, Drug Resistance, Neoplasm, Fanconi Anemia Complementation Group D2 Protein metabolism, Histone Deacetylase Inhibitors pharmacology, Histone Deacetylases metabolism, Melanoma pathology, Rad51 Recombinase metabolism, Skin Neoplasms pathology
- Abstract
DNA-damaging anticancer drugs remain a part of metastatic melanoma therapy. Epigenetic reprogramming caused by increased histone deacetylase (HDAC) activity arising during tumor formation may contribute to resistance of melanomas to the alkylating drugs temozolomide, dacarbazine, and fotemustine. Here, we report on the impact of class I HDACs on the response of malignant melanoma cells treated with alkylating agents. The data show that malignant melanomas in situ contain a high level of HDAC1/2 and malignant melanoma cells overexpress HDAC1/2/3 compared with noncancer cells. Furthermore, pharmacologic inhibition of class I HDACs sensitizes malignant melanoma cells to apoptosis following exposure to alkylating agents, while not affecting primary melanocytes. Inhibition of HDAC1/2/3 caused sensitization of melanoma cells to temozolomide in vitro and in melanoma xenografts in vivo HDAC1/2/3 inhibition resulted in suppression of DNA double-strand break (DSB) repair by homologous recombination because of downregulation of RAD51 and FANCD2. This sensitized cells to the cytotoxic DNA lesion O(6)-methylguanine and caused a synthetic lethal interaction with the PARP-1 inhibitor olaparib. Furthermore, knockdown experiments identified HDAC2 as being responsible for the regulation of RAD51. The influence of class I HDACs on DSB repair by homologous recombination and the possible clinical implication on malignant melanoma therapy with temozolomide and other alkylating drugs suggests a combination approach where class I HDAC inhibitors such as valproic acid or MS-275 (entinostat) appear to counteract HDAC- and RAD51/FANCD2-mediated melanoma cell resistance. Cancer Res; 76(10); 3067-77. ©2016 AACR., (©2016 American Association for Cancer Research.)
- Published
- 2016
- Full Text
- View/download PDF
48. The disulfide compound α-lipoic acid and its derivatives: A novel class of anticancer agents targeting mitochondria.
- Author
-
Dörsam B and Fahrer J
- Subjects
- Animals, Antineoplastic Agents chemistry, Antineoplastic Agents pharmacokinetics, Apoptosis drug effects, Caprylates chemistry, Caprylates pharmacokinetics, Drug Discovery, Energy Metabolism drug effects, Humans, Mitochondria metabolism, Mitochondria pathology, Molecular Structure, Neoplasms metabolism, Neoplasms pathology, Signal Transduction drug effects, Structure-Activity Relationship, Sulfides chemistry, Sulfides pharmacokinetics, Thioctic Acid analogs & derivatives, Thioctic Acid chemistry, Thioctic Acid pharmacokinetics, Antineoplastic Agents therapeutic use, Caprylates therapeutic use, Mitochondria drug effects, Neoplasms drug therapy, Sulfides therapeutic use, Thioctic Acid therapeutic use
- Abstract
The endogenous disulfide α-lipoic acid (LA) is an essential mitochondrial co-factor. In addition, LA and its reduced counterpart dihydro lipoic acid form a potent redox couple with antioxidative functions, for which it is used as dietary supplement and therapeutic. Recently, it has gained attention due to its cytotoxic effects in cancer cells, which is the key aspect of this review. We initially recapitulate the dietary occurrence, gastrointestinal absorption and pharmacokinetics of LA, illustrating its diverse antioxidative mechanisms. We then focus on its mode of action in cancer cells, in which it triggers primarily the mitochondrial pathway of apoptosis, whereas non-transformed primary cells are hardly affected. Furthermore, LA impairs oncogenic signaling and displays anti-metastatic potential. Novel LA derivatives such as CPI-613, which target mitochondrial energy metabolism, are described and recent pre-clinical studies are presented, which demonstrate that LA and its derivatives exert antitumor activity in vivo. Finally, we highlight clinical studies currently performed with the LA analog CPI-613. In summary, LA and its derivatives are promising candidates to complement the arsenal of established anticancer drugs due to their mitochondria-targeted mode of action and non-genotoxic properties., (Copyright © 2015 Elsevier Ireland Ltd. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
49. Lipoic acid induces p53-independent cell death in colorectal cancer cells and potentiates the cytotoxicity of 5-fluorouracil.
- Author
-
Dörsam B, Göder A, Seiwert N, Kaina B, and Fahrer J
- Subjects
- Antimetabolites, Antineoplastic administration & dosage, Caco-2 Cells, Caspases metabolism, Cell Death drug effects, Cell Survival drug effects, Colorectal Neoplasms pathology, Dose-Response Relationship, Drug, Drug Synergism, Fluorouracil administration & dosage, HCT116 Cells, Humans, Tumor Suppressor Protein p53 metabolism, Antimetabolites, Antineoplastic pharmacology, Colorectal Neoplasms drug therapy, Fluorouracil pharmacology, Thioctic Acid pharmacology
- Abstract
Alpha-lipoic acid (LA), which plays a pivotal role in mitochondrial energy metabolism, is an endogenous dithiol compound with an array of antioxidative functions. It has been shown that LA triggers cell death in tumor cell lines, whereas non-transformed cells are hardly affected. In the present study, we analyzed the cytotoxicity of LA on colorectal cancer (CRC) cells differing in their p53 status and investigated a putative synergistic effect with the anticancer drug 5-fluorouracil (5-FU). We show that LA induces a dose-dependent decrease in cell viability, which was independent of the p53 status as attested in isogenic p53-proficient and p53-deficient cell lines. This effect was largely attributable to cell death induction as revealed by Annexin-V/PI staining. LA-treated HCT116 cells underwent caspase-dependent and caspase-independent cell death, which was blocked by the pan-caspase inhibitor zVAD and the RIP-kinase inhibitor Necrostatin-1, respectively. In CaCO-2 and HT29 cells, LA induced caspase-dependent cell demise via activation of caspase-9, caspase-3 and caspase-7 with subsequent PARP-1 cleavage as demonstrated by immunoblot analysis, activity assays and pan-caspase inhibition. Interestingly, LA treatment did neither activate p53 nor induced genotoxic effects as shown by lack of DNA strand breaks and phosphorylation of histone 2AX. Finally, we provide evidence that LA increases the cytotoxic effect induced by the anticancer drug 5-FU as revealed by significantly enhanced cell death rates in HCT116 and CaCO-2 cells. Collectively, these findings demonstrate that LA induces CRC cell death independent of their p53 status and potentiates the cytotoxicity of 5-FU without causing DNA damage on its own, which makes it a candidate for tumor therapy.
- Published
- 2015
- Full Text
- View/download PDF
50. DNA repair by MGMT, but not AAG, causes a threshold in alkylation-induced colorectal carcinogenesis.
- Author
-
Fahrer J, Frisch J, Nagel G, Kraus A, Dörsam B, Thomas AD, Reißig S, Waisman A, and Kaina B
- Subjects
- Animals, Carcinogenesis chemically induced, Carcinogenesis genetics, Colorectal Neoplasms chemically induced, Colorectal Neoplasms pathology, DNA Repair drug effects, Disease Models, Animal, Humans, Mice, Mice, Transgenic, Nitroso Compounds toxicity, Colorectal Neoplasms genetics, DNA Glycosylases genetics, DNA Modification Methylases genetics, DNA Repair genetics, DNA Repair Enzymes genetics, Tumor Suppressor Proteins genetics
- Abstract
Epidemiological studies indicate that N-nitroso compounds (NOC) are causally linked to colorectal cancer (CRC). NOC induce DNA alkylations, including O (6)-methylguanine (O (6)-MeG) and N-methylated purines, which are repaired by O (6)-MeG-DNA methyltransferase (MGMT) and N-alkyladenine-DNA glycosylase (AAG)-initiated base excision repair, respectively. In view of recent evidence of nonlinear mutagenicity for NOC-like compounds, the question arises as to the existence of threshold doses in CRC formation. Here, we set out to determine the impact of DNA repair on the dose-response of alkylation-induced CRC. DNA repair proficient (WT) and deficient (Mgmt (-/-), Aag (-/-) and Mgmt (-/-)/Aag (-/-)) mice were treated with azoxymethane (AOM) and dextran sodium sulfate to trigger CRC. Tumors were quantified by non-invasive mini-endoscopy. A non-linear increase in CRC formation was observed in WT and Aag (-/-) mice. In contrast, a linear dose-dependent increase in tumor frequency was found in Mgmt (-/-) and Mgmt (-/-)/Aag (-/-) mice. The data were corroborated by hockey stick modeling, yielding similar carcinogenic thresholds for WT and Aag (-/-) and no threshold for MGMT lacking mice. O (6)-MeG levels and depletion of MGMT correlated well with the observed dose-response in CRC formation. AOM induced dose-dependently DNA double-strand breaks in colon crypts including Lgr5-positive colon stem cells, which coincided with ATR-Chk1-p53 signaling. Intriguingly, Mgmt (-/-) mice displayed significantly enhanced levels of γ-H2AX, suggesting the usefulness of γ-H2AX as an early genotoxicity marker in the colorectum. This study demonstrates for the first time a non-linear dose-response for alkylation-induced colorectal carcinogenesis and reveals DNA repair by MGMT, but not AAG, as a key node in determining a carcinogenic threshold., (© The Author 2015. Published by Oxford University Press. All rights reserved. For Permissions, please email: journals.permissions@oup.com.)
- Published
- 2015
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.