10 results on '"Annette Seibt"'
Search Results
2. Mesenchymal stem cells improve redox homeostasis and mitochondrial respiration in fibroblast cell lines with pathogenic MT-ND3 and MT-ND6 variants
- Author
-
Tharsini Navaratnarajah, Marlen Bellmann, Annette Seibt, Ruchika Anand, Özer Degistirici, Roland Meisel, Ertan Mayatepek, Andreas Reichert, Fabian Baertling, and Felix Distelmaier
- Subjects
Mitochondrial DNA ,Mesenchymal stem cells ,Complex I ,Gene therapy ,Mitochondrial transfer ,ND3 ,Medicine (General) ,R5-920 ,Biochemistry ,QD415-436 - Abstract
Abstract The most frequent biochemical defect of inherited mitochondrial disease is isolated complex I deficiency. There is no cure for this disorder, and treatment is mainly supportive. In this study, we investigated the effects of human mesenchymal stem cells (MSCs) on skin fibroblast derived from three individuals with complex I deficiency carrying different pathogenic variants in mitochondrial DNA-encoded subunits (MT-ND3, MT-ND6). Complex I-deficient fibroblasts were transiently co-cultured with bone marrow-derived MSCs. Mitochondrial transfer was analysed by fluorescence labelling and validated by Sanger sequencing. Levels of reactive oxygen species (ROS) were measured using MitoSOX Red. Moreover, mitochondrial respiration was analysed by Seahorse XFe96 Extracellular Flux Analyzer. Levels of antioxidant proteins were investigated via immunoblotting. Co-culturing of complex I-deficient fibroblast with MSCs lowered cellular ROS levels. The effect on ROS production was more sustained compared to treatment of patient fibroblasts with culture medium derived from MSC cultures. Investigation of cellular antioxidant defence systems revealed an upregulation of SOD2 (superoxide dismutase 2, mitochondrial) and HO-1 (heme oxygenase 1) in patient-derived cell lines. This adaptive response was normalised upon MSC treatment. Moreover, Seahorse experiments revealed a significant improvement of mitochondrial respiration, indicating a mitigation of the oxidative phosphorylation defect. Experiments with repetitive MSC co-culture at two consecutive time points enhanced this effect. Our study indicates that MSC-based treatment approaches might constitute an interesting option for patients with mitochondrial DNA-encoded mitochondrial diseases. We suggest that this strategy may prove more promising for defects caused by mitochondrial DNA variants compared to nuclear-encoded defects.
- Published
- 2022
- Full Text
- View/download PDF
3. The long non-coding RNA HOTAIRM1 promotes tumor aggressiveness and radiotherapy resistance in glioblastoma
- Author
-
Ulvi Ahmadov, Daniel Picard, Jasmin Bartl, Manuela Silginer, Marija Trajkovic-Arsic, Nan Qin, Lena Blümel, Marietta Wolter, Jonathan K. M. Lim, David Pauck, Alina Marie Winkelkotte, Marlen Melcher, Maike Langini, Viktoria Marquardt, Felix Sander, Anja Stefanski, Sascha Steltgens, Christina Hassiepen, Anna Kaufhold, Frauke-Dorothee Meyer, Annette Seibt, Lara Kleinesudeik, Anika Hain, Carsten Münk, Christiane Brigitte Knobbe-Thomsen, Alexander Schramm, Ute Fischer, Gabriel Leprivier, Kai Stühler, Simone Fulda, Jens T. Siveke, Felix Distelmaier, Arndt Borkhardt, Michael Weller, Patrick Roth, Guido Reifenberger, and Marc Remke
- Subjects
Cytology ,QH573-671 - Abstract
Abstract Glioblastoma is the most common malignant primary brain tumor. To date, clinically relevant biomarkers are restricted to isocitrate dehydrogenase (IDH) gene 1 or 2 mutations and O6-methylguanine DNA methyltransferase (MGMT) promoter methylation. Long non-coding RNAs (lncRNAs) have been shown to contribute to glioblastoma pathogenesis and could potentially serve as novel biomarkers. The clinical significance of HOXA Transcript Antisense RNA, Myeloid-Specific 1 (HOTAIRM1) was determined by analyzing HOTAIRM1 in multiple glioblastoma gene expression data sets for associations with prognosis, as well as, IDH mutation and MGMT promoter methylation status. Finally, the role of HOTAIRM1 in glioblastoma biology and radiotherapy resistance was characterized in vitro and in vivo. We identified HOTAIRM1 as a candidate lncRNA whose up-regulation is significantly associated with shorter survival of glioblastoma patients, independent from IDH mutation and MGMT promoter methylation. Glioblastoma cell line models uniformly showed reduced cell viability, decreased invasive growth and diminished colony formation capacity upon HOTAIRM1 down-regulation. Integrated proteogenomic analyses revealed impaired mitochondrial function and determination of reactive oxygen species (ROS) levels confirmed increased ROS levels upon HOTAIRM1 knock-down. HOTAIRM1 knock-down decreased expression of transglutaminase 2 (TGM2), a candidate protein implicated in mitochondrial function, and knock-down of TGM2 mimicked the phenotype of HOTAIRM1 down-regulation in glioblastoma cells. Moreover, HOTAIRM1 modulates radiosensitivity of glioblastoma cells both in vitro and in vivo. Our data support a role for HOTAIRM1 as a driver of biological aggressiveness, radioresistance and poor outcome in glioblastoma. Targeting HOTAIRM1 may be a promising new therapeutic approach.
- Published
- 2021
- Full Text
- View/download PDF
4. Modulation of oxidative phosphorylation and redox homeostasis in mitochondrial NDUFS4 deficiency via mesenchymal stem cells
- Author
-
Marlen Melcher, Katharina Danhauser, Annette Seibt, Özer Degistirici, Fabian Baertling, Arun Kumar Kondadi, Andreas S. Reichert, Werner J. H. Koopman, Peter H. G. M. Willems, Richard J. Rodenburg, Ertan Mayatepek, Roland Meisel, and Felix Distelmaier
- Subjects
Medicine (General) ,R5-920 ,Biochemistry ,QD415-436 - Abstract
Abstract Background Disorders of the oxidative phosphorylation (OXPHOS) system represent a large group among the inborn errors of metabolism. The most frequently observed biochemical defect is isolated deficiency of mitochondrial complex I (CI). No effective treatment strategies for CI deficiency are so far available. The purpose of this study was to investigate whether and how mesenchymal stem cells (MSCs) are able to modulate metabolic function in fibroblast cell models of CI deficiency. Methods We used human and murine fibroblasts with a defect in the nuclear DNA encoded NDUFS4 subunit of CI. Fibroblasts were co-cultured with MSCs under different stress conditions and intercellular mitochondrial transfer was assessed by flow cytometry and fluorescence microscopy. Reactive oxygen species (ROS) levels were measured using MitoSOX-Red. Protein levels of CI were analysed by blue native polyacrylamide gel electrophoresis (BN-PAGE). Results Direct cellular interactions and mitochondrial transfer between MSCs and human as well as mouse fibroblast cell lines were demonstrated. Mitochondrial transfer was visible in 13.2% and 6% of fibroblasts (e.g. fibroblasts containing MSC mitochondria) for human and mouse cell lines, respectively. The transfer rate could be further stimulated via treatment of cells with TNF-α. MSCs effectively lowered cellular ROS production in NDUFS4-deficient fibroblast cell lines (either directly via co-culture or indirectly via incubation of cell lines with cell-free MSC supernatant). However, CI protein expression and activity were not rescued by MSC treatment. Conclusion This study demonstrates the interplay between MSCs and fibroblast cell models of isolated CI deficiency including transfer of mitochondria as well as modulation of cellular ROS levels. Further exploration of these cellular interactions might help to develop MSC-based treatment strategies for human CI deficiency.
- Published
- 2017
- Full Text
- View/download PDF
5. Tissue-specific strategies of the very-long chain acyl-CoA dehydrogenase-deficient (VLCAD-/-) mouse to compensate a defective fatty acid β-oxidation.
- Author
-
Sara Tucci, Diran Herebian, Marga Sturm, Annette Seibt, and Ute Spiekerkoetter
- Subjects
Medicine ,Science - Abstract
Very long-chain acyl-CoA dehydrogenase (VLCAD)-deficiency is the most common long-chain fatty acid oxidation disorder presenting with heterogeneous phenotypes. Similar to many patients with VLCADD, VLCAD-deficient mice (VLCAD(-/-)) remain asymptomatic over a long period of time. In order to identify the involved compensatory mechanisms, wild-type and VLCAD(-/-) mice were fed one year either with a normal diet or with a diet in which medium-chain triglycerides (MCT) replaced long-chain triglycerides, as approved intervention in VLCADD. The expression of the mitochondrial long-chain acyl-CoA dehydrogenase (LCAD) and medium-chain acyl-CoA dehydrogenase (MCAD) was quantified at mRNA and protein level in heart, liver and skeletal muscle. The oxidation capacity of the different tissues was measured by LC-MS/MS using acyl-CoA substrates with a chain length of 8 to 20 carbons. Moreover, in white skeletal muscle the role of glycolysis and concomitant muscle fibre adaptation was investigated. In one year old VLCAD(-/-) mice MCAD and LCAD play an important role in order to compensate deficiency of VLCAD especially in the heart and in the liver. However, the white gastrocnemius muscle develops alternative compensatory mechanism based on a different substrate selection and increased glucose oxidation. Finally, the application of an MCT diet over one year has no effects on LCAD or MCAD expression. MCT results in the VLCAD(-/-) mice only in a very modest improvement of medium-chain acyl-CoA oxidation capacity restricted to cardiac tissue. In conclusion, VLCAD(-/-) mice develop tissue-specific strategies to compensate deficiency of VLCAD either by induction of other mitochondrial acyl-CoA dehydrogenases or by enhancement of glucose oxidation. In the muscle, there is evidence of a muscle fibre type adaptation with a predominance of glycolytic muscle fibres. Dietary modification as represented by an MCT-diet does not improve these strategies long-term.
- Published
- 2012
- Full Text
- View/download PDF
6. Estrogen receptor alpha expression in podocytes mediates protection against apoptosis in-vitro and in-vivo.
- Author
-
Sebastian Kummer, Stefanie Jeruschke, Lara Vanessa Wegerich, Andrea Peters, Petra Lehmann, Annette Seibt, Friederike Mueller, Nadezda Koleganova, Elisabeth Halbenz, Claus Peter Schmitt, Markus Bettendorf, Ertan Mayatepek, Marie-Luise Gross-Weissmann, and Jun Oh
- Subjects
Medicine ,Science - Abstract
CONTEXT/OBJECTIVE: Epidemiological studies have demonstrated that women have a significantly better prognosis in chronic renal diseases compared to men. This suggests critical influences of gender hormones on glomerular structure and function. We examined potential direct protective effects of estradiol on podocytes. METHODS: Expression of estrogen receptor alpha (ERα) was examined in podocytes in vitro and in vivo. Receptor localization was shown using Western blot of separated nuclear and cytoplasmatic protein fractions. Podocytes were treated with Puromycin aminonucleoside (PAN, apoptosis induction), estradiol, or both in combination. Apoptotic cells were detected with Hoechst nuclear staining and Annexin-FITC flow cytometry. To visualize mitochondrial membrane potential depolarization as an indicator for apoptosis, cells were stained with tetramethyl rhodamine methylester (TMRM). Estradiol-induced phosphorylation of ERK1/2 and p38 MAPK was examined by Western blot. Glomeruli of ERα knock-out mice and wild-type controls were analysed by histomorphometry and immunohistochemistry. RESULTS: ERα was consistently expressed in human and murine podocytes. Estradiol stimulated ERα protein expression, reduced PAN-induced apoptosis in vitro by 26.5±24.6% or 56.6±5.9% (flow cytometry or Hoechst-staining, respectively; both p
- Published
- 2011
- Full Text
- View/download PDF
7. Modulation of oxidative phosphorylation and redox homeostasis in mitochondrial NDUFS4 deficiency via mesenchymal stem cells
- Author
-
Richard J. Rodenburg, Andreas S. Reichert, Fabian Baertling, Marlen Melcher, Werner J.H. Koopman, Özer Degistirici, Arun Kumar Kondadi, Katharina Danhauser, Peter H.G.M. Willems, Felix Distelmaier, Roland Meisel, Ertan Mayatepek, and Annette Seibt
- Subjects
0301 basic medicine ,Cell ,Medicine (miscellaneous) ,Mitochondrion ,Biology ,Biochemistry, Genetics and Molecular Biology (miscellaneous) ,Oxidative Phosphorylation ,lcsh:Biochemistry ,03 medical and health sciences ,Mice ,medicine ,Animals ,Humans ,lcsh:QD415-436 ,Fibroblast ,lcsh:R5-920 ,Electron Transport Complex I ,Research ,Mesenchymal stem cell ,NDUFS4 ,Metabolic Disorders Radboud Institute for Molecular Life Sciences [Radboudumc 6] ,Mesenchymal Stem Cells ,NADH Dehydrogenase ,Cell Biology ,Ci protein ,Fibroblasts ,Coculture Techniques ,Cell biology ,Mitochondria ,030104 developmental biology ,medicine.anatomical_structure ,Cell culture ,Molecular Medicine ,Stem cell ,lcsh:Medicine (General) - Abstract
Background Disorders of the oxidative phosphorylation (OXPHOS) system represent a large group among the inborn errors of metabolism. The most frequently observed biochemical defect is isolated deficiency of mitochondrial complex I (CI). No effective treatment strategies for CI deficiency are so far available. The purpose of this study was to investigate whether and how mesenchymal stem cells (MSCs) are able to modulate metabolic function in fibroblast cell models of CI deficiency. Methods We used human and murine fibroblasts with a defect in the nuclear DNA encoded NDUFS4 subunit of CI. Fibroblasts were co-cultured with MSCs under different stress conditions and intercellular mitochondrial transfer was assessed by flow cytometry and fluorescence microscopy. Reactive oxygen species (ROS) levels were measured using MitoSOX-Red. Protein levels of CI were analysed by blue native polyacrylamide gel electrophoresis (BN-PAGE). Results Direct cellular interactions and mitochondrial transfer between MSCs and human as well as mouse fibroblast cell lines were demonstrated. Mitochondrial transfer was visible in 13.2% and 6% of fibroblasts (e.g. fibroblasts containing MSC mitochondria) for human and mouse cell lines, respectively. The transfer rate could be further stimulated via treatment of cells with TNF-α. MSCs effectively lowered cellular ROS production in NDUFS4-deficient fibroblast cell lines (either directly via co-culture or indirectly via incubation of cell lines with cell-free MSC supernatant). However, CI protein expression and activity were not rescued by MSC treatment. Conclusion This study demonstrates the interplay between MSCs and fibroblast cell models of isolated CI deficiency including transfer of mitochondria as well as modulation of cellular ROS levels. Further exploration of these cellular interactions might help to develop MSC-based treatment strategies for human CI deficiency. Electronic supplementary material The online version of this article (doi:10.1186/s13287-017-0601-7) contains supplementary material, which is available to authorized users.
- Published
- 2017
8. NAXE mutations disrupt the cellular NAD(P)HX repair system and cause a lethal neurometabolic disorder of early childhood
- Author
-
Felix Distelmaier, Klaus Lohmeier, Diran Herebian, Dorota Piekutowska-Abramczuk, Holger Prokisch, Dirk Klee, Danijela Petković Ramadža, Tobias B. Haack, Ivo Barić, Ewa Pronicka, Thomas Meitinger, Wolfgang Müller-Felber, Tim M. Strom, Annette Seibt, Dariusz Rokicki, Katharina Danhauser, Rafał Płoski, Thomas Klopstock, Laura S. Kremer, Ertan Mayatepek, Johannes A. Mayr, and Dominik T. Schneider
- Subjects
0301 basic medicine ,Male ,analogs & derivatives [NAD] ,NAXE protein, human ,Metabolite ,Cell ,genetics [Metabolic Diseases] ,genetics [Carrier Proteins] ,chemistry.chemical_compound ,0302 clinical medicine ,Cerebrospinal fluid ,Fatal Outcome ,metabolism [Nervous System Diseases] ,genetics [Skin Abnormalities] ,Genetics (clinical) ,Exome sequencing ,chemistry.chemical_classification ,genetics [Nervous System Diseases] ,Nad(p)hx ,Energy Metabolism ,Metabolite Repair ,Mitochondrial ,medicine.anatomical_structure ,Nicotinic agonist ,Biochemistry ,Child, Preschool ,metabolism [NAD] ,Female ,medicine.symptom ,medicine.medical_specialty ,Ataxia ,metabolism [Metabolic Diseases] ,Racemases and Epimerases ,Neuroimaging ,6-hydroxy-1,4,5,6-tetrahydronicotinamide adenine dinucleotide ,Biology ,pathology [Metabolic Diseases] ,genetics [Racemases and Epimerases] ,Cell Line ,pathology [Nervous System Diseases] ,03 medical and health sciences ,Metabolic Diseases ,ddc:570 ,Internal medicine ,Report ,Genetics ,medicine ,Humans ,Infant ,Fibroblasts ,NAD ,030104 developmental biology ,Endocrinology ,Enzyme ,chemistry ,Mutation ,Skin Abnormalities ,pathology [Skin Abnormalities] ,NAD+ kinase ,Nervous System Diseases ,Carrier Proteins ,030217 neurology & neurosurgery ,metabolism [Carrier Proteins] - Abstract
To safeguard the cell from the accumulation of potentially harmful metabolic intermediates, specific repair mechanisms have evolved. APOA1BP, now renamed NAXE, encodes an epimerase essential in the cellular metabolite repair for NADHX and NADPHX. The enzyme catalyzes the epimerization of NAD(P)HX, thereby avoiding the accumulation of toxic metabolites. The clinical importance of the NAD(P)HX repair system has been unknown. Exome sequencing revealed pathogenic biallelic mutations in NAXE in children from four families with (sub-) acute-onset ataxia, cerebellar edema, spinal myelopathy, and skin lesions. Lactate was elevated in cerebrospinal fluid of all affected individuals. Disease onset was during the second year of life and clinical signs as well as episodes of deterioration were triggered by febrile infections. Disease course was rapidly progressive, leading to coma, global brain atrophy, and finally to death in all affected individuals. NAXE levels were undetectable in fibroblasts from affected individuals of two families. In these fibroblasts we measured highly elevated concentrations of the toxic metabolite cyclic-NADHX, confirming a deficiency of the mitochondrial NAD(P)HX repair system. Finally, NAD or nicotinic acid (vitamin B3) supplementation might have therapeutic implications for this fatal disorder.
- Published
- 2016
- Full Text
- View/download PDF
9. Transcellular migration of neutrophil granulocytes through the blood-cerebrospinal fluid barrier after infection with Streptococcus suis
- Author
-
Lilo Greune, Christian Schwerk, Hans-Joachim Galla, M. Alexander Schmidt, Corinna Wewer, Tobias Tenenbaum, Hartwig Wolburg, Annette Seibt, Ulrike Quitsch, Horst Schroten, and Jürgen Berger
- Subjects
Streptococcus suis ,Neutrophils ,Swine ,Immunology ,Vascular Cell Adhesion Molecule-1 ,CD18 ,lcsh:RC346-429 ,Tight Junctions ,Cellular and Molecular Neuroscience ,Transcellular Cell Migration ,Electric Impedance ,Animals ,Humans ,Transcellular ,Barrier function ,Cells, Cultured ,Cytoskeleton ,lcsh:Neurology. Diseases of the nervous system ,CD11b Antigen ,biology ,Tight junction ,Cell adhesion molecule ,General Neuroscience ,Research ,Epithelial Cells ,Apical membrane ,biology.organism_classification ,Intercellular Adhesion Molecule-1 ,Actins ,Cell biology ,Neurology ,Blood-Brain Barrier ,Paracellular transport ,CD18 Antigens ,Choroid Plexus - Abstract
Background A critical point during the course of bacterial meningitis is the excessive influx of polymorphnuclear neutrophils (PMNs) from the blood into the brain. Both paracellular and transcellular routes of leukocyte transmigration through the blood-brain barrier have been described in CNS diseases so far. Thus, we investigated the mechanism of PMN transmigration through the blood-CSF barrier under inflammatory conditions. Methods In an "inverted" Transwell culture model of the blood-CSF barrier, the zoonotic agent Streptococcus suis (S. suis) was used to stimulate porcine choroid plexus epithelial cells (PCPECs) specifically from the physiologically relevant basolateral side. Barrier function was analyzed by measuring TEER and TR-dextran-flux, and tight junction morphology was investigated by immunofluorescence. Route and mechanism of PMN transmigration were determined by immunofluorescence, electron microscopy and FACS analysis. Quantitative real time-PCR was used to determine expression levels of ICAM-1 and VCAM-1. Results Here, we show that the transmigration of PMNs through PCPECs was significantly higher after stimulation with TNFα or infection with S. suis strain 10 compared to its non-encapsulated mutant. Barrier function was not significantly affected by PMN migration alone, but in combination with S. suis infection. Tight junction and cytoskeletal actin reorganisation were also observed after stimulation with S. suis or TNFα. Most strikingly, PMNs preferentially migrated across PCPECs via the transcellular route. Extensive sequential analyses of the PMN transmigration process with Apotome®-imaging and electron microscopy revealed that paracellular migrating PMNs stop just before tight junctions. Interestingly, PMNs subsequently appeared to proceed by transcellular migration via funnel-like structures developing from the apical membrane. It is noteworthy that some PMNs contained bacteria during the transmigration process. Flow cytometric and transmigration inhibition studies with integrin-specific antibodies showed that PMN traversal is dependent on CD11b/CD18. Analysis of cell adhesion molecules in PCPECs revealed a significant increase of ICAM-1 and VCAM-1 expression after TNFα and S. suis stimulation. Conclusion Our data underline the relevance of the blood-CSF barrier as a gate for leukocyte entry into the CNS and suggest a novel transcellular migration step during the pathogenesis of bacterial meningitis.
- Published
- 2011
10. TNF𝛼 Induces Choroid Plexus Epithelial Cell Barrier Alterations by Apoptotic and Nonapoptotic Mechanisms
- Author
-
Horst Schroten, Patrick Zeni, Christian Schwerk, Marie-Louise Mölleken, Kasia Rybarczyk, Frank Essmann, Annette Seibt, and Tobias Tenenbaum
- Subjects
Article Subject ,Swine ,Health, Toxicology and Mutagenesis ,lcsh:Biotechnology ,lcsh:Medicine ,Apoptosis ,Cycloheximide ,Biology ,Blood–brain barrier ,lcsh:Chemical technology ,lcsh:Technology ,chemistry.chemical_compound ,lcsh:TP248.13-248.65 ,Genetics ,medicine ,Choroid Plexus Epithelium ,Electric Impedance ,Animals ,lcsh:TP1-1185 ,Viability assay ,Molecular Biology ,Barrier function ,Cells, Cultured ,Tumor Necrosis Factor-alpha ,lcsh:T ,lcsh:R ,Epithelial Cells ,General Medicine ,Caspase Inhibitors ,Epithelium ,Cell biology ,medicine.anatomical_structure ,chemistry ,Blood-Brain Barrier ,Caspases ,Choroid Plexus ,Molecular Medicine ,Choroid plexus ,Biotechnology ,Research Article ,Signal Transduction - Abstract
The choroid plexus epithelium constitutes the structural basis of the blood-cerebrospinal fluid barrier. Since the cytokine TNFalpha is markedly increased during inflammatory diseases in the blood and the central nervous system, we investigated by which mechanisms TNFalpha induces barrier alteration in porcine choroid plexus epithelial cells. We found a dose-dependent decrease of transepithelial electrical resistance, increase of paracellular inulin-flux, and induction of histone-associated DNA fragmentation and caspase-3 activation after TNFalpha stimulation. This response was strongly aggravated by the addition of cycloheximide and could partially be inhibited by the NF-kappaB inhibitor CAPE, but most effectively by the pan-caspase-inhibitor zVAD-fmk and not by the JNK inhibitor SP600125. Partial loss of cell viability could also be attenuated by CAPE. Immunostaining showed cell condensation and nuclear binding of high-mobility group box 1 protein as a sign of apoptosis after TNFalpha stimulation. Taken together our findings indicate that TNFalpha compromises PCPEC barrier function by caspase and NF-kappaB dependent mechanisms.
- Published
- 2010
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.