37 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. De novo missense variants in RRAGC lead to a fatal mTORopathy of early childhood
- Author
-
Margot R.F. Reijnders, Annette Seibt, Melanie Brugger, Ideke J.C. Lamers, Torsten Ott, Oliver Klaas, Judit Horváth, Ailsa M.S. Rose, Isabel M. Craghill, Theresa Brunet, Elisabeth Graf, Katharina Mayerhanser, Debby Hellebrekers, David Pauck, Eva Neuen-Jacob, Richard J.T. Rodenburg, Dagmar Wieczorek, Dirk Klee, Ertan Mayatepek, Gertjan Driessen, Robert Bindermann, Luisa Averdunk, Klaus Lohmeier, Margje Sinnema, Alexander P.A. Stegmann, Ronald Roepman, James A. Poulter, and Felix Distelmaier
- Subjects
Genetics (clinical) - Abstract
PURPOSE: Mechanistic target of rapamycin (mTOR) complex 1 (mTORC1) regulates cell growth in response to nutritional status. Central to the mTORC1 function is the Rag-GTPase heterodimer. One component of the Rag heterodimer is RagC (Ras-related GTP-binding protein C), which is encoded by the RRAGC gene. METHODS: Genetic testing via trio exome sequencing was applied to identify the underlying disease cause in 3 infants with dilated cardiomyopathy, hepatopathy, and brain abnormalities, including pachygyria, polymicrogyria, and septo-optic dysplasia. Studies in patient-derived skin fibroblasts and in a HEK293 cell model were performed to investigate the cellular consequences. RESULTS: We identified 3 de novo missense variants in RRAGC (NM_022157.4: c.269C>A, p.(Thr90Asn), c.353C>T, p.(Pro118Leu), and c.343T>C, p.(Trp115Arg)), which were previously reported as occurring somatically in follicular lymphoma. Studies of patient-derived fibroblasts carrying the p.(Thr90Asn) variant revealed increased cell size, as well as dysregulation of mTOR-related p70S6K (ribosomal protein S6 kinase 1) and transcription factor EB signaling. Moreover, subcellular localization of mTOR was decoupled from metabolic state. We confirmed the key findings for all RRAGC variants described in this study in a HEK293 cell model. CONCLUSION: The above results are in line with a constitutive overactivation of the mTORC1 pathway. Our study establishes de novo missense variants in RRAGC as cause of an early-onset mTORopathy with unfavorable prognosis.
- Published
- 2023
- Full Text
- View/download PDF
8. Biallelic variants in CRIPT cause a Rothmund-Thomson-like syndrome with increased cellular senescence
- Author
-
Luisa Averdunk, Maxim A Huetzen, Daniel Moreno-Andrés, Reinhard Kalb, Shane McKee, Tzung-Chien Hsieh, Annette Seibt, Marten Schouwink, Seema Lalani, Eissa Ali Faqeih, Theresa Brunet, Peter Boor, Kornelia Neveling, Alexander Hoischen, Barbara Hildebrandt, Elisabeth Graf, Linchao Lu, Weidong Jin, Joerg Schaper, Jamal A Omer, Tanguy Demaret, Nicole Fleischer, Detlev Schindler, Peter Krawitz, Ertan Mayatepek, Dagmar Wieczorek, Lisa L. Wang, Wolfram Antonin, Ron D. Jachimowicz, Verena von Felbert, and Felix Distelmaier
- Subjects
Genetics (clinical) - Abstract
PURPOSE: Rothmund-Thomson syndrome (RTS) is characterized by poikiloderma, sparse hair, small stature, skeletal defects, cancer, and cataracts, resembling features of premature aging. RECQL4 and ANAPC1 are the 2 known disease genes associated with RTS in >70% of cases. We describe RTS-like features in 5 individuals with biallelic variants in CRIPT (OMIM 615789). METHODS: Two newly identified and 4 published individuals with CRIPT variants were systematically compared with those with RTS using clinical data, computational analysis of photographs, histologic analysis of skin, and cellular studies on fibroblasts. RESULTS: All CRIPT individuals fulfilled the diagnostic criteria for RTS and additionally had neurodevelopmental delay and seizures. Using computational gestalt analysis, CRIPT individuals showed greatest facial similarity with individuals with RTS. Skin biopsies revealed a high expression of senescence markers (p53/p16/p21) and the senescence-associated ß-galactosidase activity was elevated in CRIPT-deficient fibroblasts. RECQL4- and CRIPT-deficient fibroblasts showed an unremarkable mitotic progression and unremarkable number of mitotic errors and no or only mild sensitivity to genotoxic stress by ionizing radiation, mitomycin C, hydroxyurea, etoposide, and potassium bromate. CONCLUSION: CRIPT causes an RTS-like syndrome associated with neurodevelopmental delay and epilepsy. At the cellular level, RECQL4- and CRIPT-deficient cells display increased senescence, suggesting shared molecular mechanisms leading to the clinical phenotypes.
- Published
- 2023
- Full Text
- View/download PDF
9. De Novo and Bi-allelic Pathogenic Variants in NARS1 Cause Neurodevelopmental Delay Due to Toxic Gain-of-Function and Partial Loss-of-Function Effects
- Author
-
Cheryl Cytrynbaum, Francesca Mattioli, Maria J. Guillen Sacoto, Federico Santoni, Rosanna Weksberg, Amina Nasar, Annemarie Fock, Henry Houlden, Shaikh Riazuddin, Tobias B. Haack, Roisin Sullivan, Mona Grimmel, Helen Griffin, Stylianos E. Antonarakis, Nuzhat Rana, Andreea Manole, Marisa I. Mendes, Ayca Dilruba Aslanger, Justyna Iwaszkiewicz, Julia Mohr, Rolph Pfundt, Muhammed Ilyas, Tina Duelund Hjortshøj, Kshitij Mankad, Muhammad Ansar, Katherine M. Christensen, Sonal Desai, Aida Telegrafi, Faisal Zafar, Helena Gásdal Karstensen, Dagan Jenkins, Yue Si, John F. Mantovani, Alice Goldenberg, Sylvain Debard, Muhammad T. Sarwar, Jagdeep S. Walia, Stephanie Efthymiou, Rita Horvath, Vincenzo Salpietro, Reza Maroofian, Jawad Ahmed, Joost Raaphorst, Lindsay B. Henderson, Benyekhlef Kara, Lauren Badalato, Adnan Y. Manzur, Desirée E.C. Smith, Ruben Portier, Marwan Shinawi, Marisa V. Andrews, Gajja S. Salomons, John B. Vincent, Amélie Piton, Felix Distelmaier, Emmanuelle Ranza, Jean-Louis Mandel, Sohail A. Paracha, Marybeth Hummel, Jürg Bähler, Dustin Baldridge, Muhammad A. Usmani, Lu Wang, Maria Rodriguez Lopez, Frédéric Fischer, Annette Seibt, Servi J. C. Stevens, Matthew J. Jennings, Majdi Kara, Amelia Kirby, Hubert Dominique Becker, Kristin W. Barañano, Christopher S. Francklyn, Saima Riazuddin, Rasim Ozgur Rosti, Emer O'Connor, Yalda Jamshidi, Barbara Oehl-Jaschkowitz, Ricardo Harripaul, Anne Marie Jelsig, Anna Sarkozy, Indran Davagnanam, Zubair M. Ahmed, David A. Koolen, Joseph G. Gleeson, Heinz Gabriel, Alkyoni Athanasiou-Fragkouli, Muhammad Ayub, Alejandro Horga, Conny van Ravenwaaij, Bruno Senger, Ingrid M. Wentzensen, Clinical Cognitive Neuropsychiatry Research Program (CCNP), Neurology, Laboratory Genetic Metabolic Diseases, ANS - Cellular & Molecular Mechanisms, AGEM - Amsterdam Gastroenterology Endocrinology Metabolism, ASLANGER, Ayça Dilruba, Université de Strasbourg (UNISTRA), MUMC+: DA KG Lab Centraal Lab (9), RS: FHML non-thematic output, Laboratory Medicine, AGEM - Endocrinology, metabolism and nutrition, AGEM - Inborn errors of metabolism, Amsterdam Neuroscience - Cellular & Molecular Mechanisms, Amsterdam Gastroenterology Endocrinology Metabolism, and Amsterdam Reproduction & Development (AR&D)
- Subjects
Male ,0301 basic medicine ,Microcephaly ,Developmental delay ,[SDV]Life Sciences [q-bio] ,Aspartate-tRNA Ligase ,TRANSFER-RNA SYNTHETASE ,RNA, Transfer, Amino Acyl ,0302 clinical medicine ,RNA, Transfer ,Loss of Function Mutation ,ComputingMilieux_MISCELLANEOUS ,Genetics (clinical) ,next generation sequencing ,chemistry.chemical_classification ,Genetics ,neurodevelopment ,Stem Cells ,Disorders of movement Donders Center for Medical Neuroscience [Radboudumc 3] ,Neural stem cell ,Pedigree ,Amino acid ,developmental delay ,Gain of Function Mutation ,Transfer RNA ,Female ,Amino Acyl ,medicine.symptom ,Rare cancers Radboud Institute for Health Sciences [Radboudumc 9] ,EXPRESSION ,Ataxia ,Biology ,Article ,Cell Line ,Amino Acyl-tRNA Synthetases ,03 medical and health sciences ,aminoacyl-tRNA synthetase ,epilepsy ,neuropathy ,Alleles ,Genetic Predisposition to Disease ,Humans ,Neurodevelopmental Disorders ,2 SIBLINGS ,medicine ,Allele ,Epilepsy ,Neurodevelopmental disorders Donders Center for Medical Neuroscience [Radboudumc 7] ,MUTATIONS ,medicine.disease ,Transfer ,030104 developmental biology ,Enzyme ,chemistry ,Aminoacyl-tRNA synthetase ,RNA ,030217 neurology & neurosurgery ,Function (biology) - Abstract
Aminoacyl-tRNA synthetases (ARSs) are ubiquitous, ancient enzymes that charge amino acids to cognate tRNA molecules, the essential first step of protein translation. Here, we describe 32 individuals from 21 families, presenting with microcephaly, neurodevelopmental delay, seizures, peripheral neuropathy, and ataxia, with de novo heterozygous and bi-allelic mutations in asparaginyl-tRNA synthetase (NARS1). We demonstrate a reduction in NARS1 mRNA expression as well as in NARS1 enzyme levels and activity in both individual fibroblasts and induced neural progenitor cells (iNPCs). Molecular modeling of the recessive c.1633C>T (p.Arg545Cys) variant shows weaker spatial positioning and tRNA selectivity. We conclude that de novo and bi-allelic mutations in NARS1 are a significant cause of neurodevelopmental disease, where the mechanism for de novo variants could be toxic gain-of-function and for recessive variants, partial loss-of-function.
- Published
- 2020
- Full Text
- View/download PDF
10. Validation of current and bathymetry measurements in the German Bight by airborne along-track interferometric SAR.
- Author
-
Roland Romeiser, Annette Seibt-Winckler, Martina Heineke, and Dieter Eppel
- Published
- 2002
- Full Text
- View/download PDF
11. Bi-Allelic COQ4 Variants Cause Adult-Onset Ataxia-Spasticity Spectrum Disease
- Author
-
Isabell Cordts, Luisa Semmler, Jannik Prasuhn, Annette Seibt, Diran Herebian, Tharsini Navaratnarajah, Joohyun Park, Natalie Deininger, Lucia Laugwitz, Sophia L. Göricke, Paul Lingor, Norbert Brüggemann, Alexander Münchau, Matthis Synofzik, Dagmar Timmann, Johannes A. Mayr, Tobias B. Haack, Felix Distelmaier, and Marcus Deschauer
- Subjects
Mitochondrial Diseases ,Cerebellar Ataxia ,Ubiquinone ,Medizin ,genetics [Mutation] ,deficiency [Ubiquinone] ,coenzyme Q10 deficiency ,Mitochondrial Proteins ,genetics [Ubiquinone] ,Humans ,ddc:610 ,hereditary spastic paraplegia ,metabolism [Ubiquinone] ,Muscle Weakness ,genetics [Cerebellar Ataxia] ,Brief Report ,Regular Issue Articles ,coenzyme Q ,cerebellar ataxia ,mitochondriopathy ,genetics [Ataxia] ,ddc ,Neurology ,Muscle Spasticity ,Mutation ,genetics [Mitochondrial Proteins] ,Ataxia ,Neurology (clinical) - Abstract
Background: COQ4 codes for a mitochondrial protein required for coenzyme Q₁₀ (CoQ₁₀) biosynthesis. Autosomal recessive COQ4-associated CoQ₁₀ deficiency leads to an early-onset mitochondrial multi-organ disorder. Methods: In-house exome and genome datasets (n = 14,303) were screened for patients with bi-allelic variants in COQ4. Work-up included clinical characterization and functional studies in patient-derived cell lines. Results: Six different COQ4 variants, three of them novel, were identified in six adult patients from four different families. Three patients had a phenotype of hereditary spastic paraparesis, two sisters showed a predominant cerebellar ataxia, and one patient had mild signs of both. Studies in patient-derived fibroblast lines revealed significantly reduced amounts of COQ4 protein, decreased CoQ₁₀ concentrations, and elevated levels of the metabolic intermediate 6-demethoxyubiquinone. Conclusion: We report bi-allelic variants in COQ4 causing an adult-onset ataxia-spasticity spectrum phenotype and a disease course much milder than previously reported. © 2022 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.
- Published
- 2021
- Full Text
- View/download PDF
12. 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
Electron Transport Complex I ,Mitochondrial Diseases ,Respiration ,Medicine (miscellaneous) ,Mesenchymal Stem Cells ,NADH Dehydrogenase ,Cell Biology ,Fibroblasts ,Biochemistry, Genetics and Molecular Biology (miscellaneous) ,DNA, Mitochondrial ,Antioxidants ,Cell Line ,Molecular Medicine ,Homeostasis ,Humans ,Reactive Oxygen Species ,Oxidation-Reduction - 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
- 2021
13. Bi-allelic Variants in RALGAPA1 Cause Profound Neurodevelopmental Disability, Muscular Hypotonia, Infantile Spasms, and Feeding Abnormalities
- Author
-
Thomas Meitinger, Ortal Barel, Ertan Mayatepek, Dirk Klee, Tim M. Strom, Dagmar Wieczorek, Felix Distelmaier, Fuad Al Mutairi, Yezmin Perilla-Young, Marc Remke, Fowzan S. Alkuraya, Laurie A. Demmer, Cynthia M. Powell, Annette Seibt, Yuliya Skorobogatko, Tharsini Navaratnarajah, Peter Lichtner, Hanan E. Shamseldin, Bader Alhaddad, Matias Wagner, Alan R. Saltiel, Chen Hoffmann, Gali Heimer, Yair Anikster, and Ben Pode-Shakked
- Subjects
Male ,0301 basic medicine ,GTPase-activating protein ,Infantile ,Medical and Health Sciences ,muscular hypotonia ,Spasms ,0302 clinical medicine ,Cell Movement ,Intellectual disability ,2.1 Biological and endogenous factors ,Aetiology ,Child ,Genetics (clinical) ,G alpha subunit ,Genetics & Heredity ,GTPase-Activating Proteins ,Cell migration ,Biological Sciences ,Phenotype ,RALA ,Child, Preschool ,030220 oncology & carcinogenesis ,Muscle Hypotonia ,Female ,Signal transduction ,Spasms, Infantile ,Intellectual and Developmental Disabilities (IDD) ,Nerve Tissue Proteins ,Biology ,Epilepsy ,Garnl1 ,Muscular Hypotonia ,Neurodevelopmental Disorder ,Rala Signaling ,Tulip1 ,West Syndrome ,Feeding and Eating Disorders ,03 medical and health sciences ,Rare Diseases ,Clinical Research ,Report ,TULIP1 ,Genetics ,medicine ,Humans ,Family ,Preschool ,Alleles ,Cell Proliferation ,Muscular hypotonia ,Neurosciences ,Infant ,RalA signaling ,West syndrome ,medicine.disease ,neurodevelopmental disorder ,Brain Disorders ,030104 developmental biology ,Neurodevelopmental Disorders ,Mutation ,epilepsy ,GARNL1 ,Neuroscience - Abstract
Ral (Ras-like) GTPases play an important role in the control of cell migration and have been implicated in Ras-mediated tumorigenicity. Recently, variants in RALA were also described as a cause of intellectual disability and developmental delay, indicating the relevance of this pathway to neuropediatric diseases. Here, we report the identification of bi-allelic variants in RALGAPA1 (encoding Ral GTPase activating protein catalytic alpha subunit 1) in four unrelated individuals with profound neurodevelopmental disability, muscular hypotonia, feeding abnormalities, recurrent fever episodes, and infantile spasms . Dysplasia of corpus callosum with focal thinning of the posterior part and characteristic facial features appeared to be unifying findings. RalGAPA1 was absent in the fibroblasts derived from two affected individuals suggesting a loss-of-function effect of the RALGAPA1 variants. Consequently, RalA activity was increased in these cell lines, which is in keeping with the idea that RalGAPA1 deficiency causes a constitutive activation of RalA. Additionally, levels of RalGAPB, a scaffolding subunit of the RalGAP complex, were dramatically reduced, indicating a dysfunctional RalGAP complex. Moreover, RalGAPA1 deficiency clearly increased cell-surface levels of lipid raft components in detached fibroblasts, which might indicate that anchorage-dependence of cell growth signaling is disturbed. Our findings indicate that the dysregulation of the RalA pathway has an important impact on neuronal function and brain development. In light of the partially overlapping phenotype between RALA- and RALGAPA1-associated diseases, it appears likely that dysregulation of the RalA signaling pathway leads to a distinct group of genetic syndromes that we suggest could be named RALopathies.
- Published
- 2020
- Full Text
- View/download PDF
14. Choline transporter-like 1 deficiency causes a new type of childhood-onset neurodegeneration
- Author
-
Dirk Klee, Eva Kildall Hejbøl, Ljubo Markovic, Marica Bakovic, Mark A. Tarnopolsky, Lauren Brady, Vernon W. Dolinsky, Maria Kibaek, Annette Seibt, Prasoon Agarwal, Else Gade, Rami Abou Jamra, Henrik Daa Schrøder, Martin Jakob Larsen, Adrian Taylor, Peter L. Nagy, Dagmar Wieczorek, Felix Distelmaier, Christina Fagerberg, and Nicholas A. Rouse
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Endoplasmic reticulum ,Neurodegeneration ,Substantia nigra ,Biology ,medicine.disease ,Choline transporter ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,Atrophy ,Endocrinology ,chemistry ,Phosphatidylcholine ,Internal medicine ,medicine ,Choline ,Neurology (clinical) ,Choline transport ,030217 neurology & neurosurgery - Abstract
Cerebral choline metabolism is crucial for normal brain function, and its homoeostasis depends on carrier-mediated transport. Here, we report on four individuals from three families with neurodegenerative disease and homozygous frameshift mutations (Asp517Metfs*19, Ser126Metfs*8, and Lys90Metfs*18) in the SLC44A1 gene encoding choline transporter-like protein 1. Clinical features included progressive ataxia, tremor, cognitive decline, dysphagia, optic atrophy, dysarthria, as well as urinary and bowel incontinence. Brain MRI demonstrated cerebellar atrophy and leukoencephalopathy. Moreover, low signal intensity in globus pallidus with hyperintensive streaking and low signal intensity in substantia nigra were seen in two individuals. The Asp517Metfs*19 and Ser126Metfs*8 fibroblasts were structurally and functionally indistinguishable. The most prominent ultrastructural changes of the mutant fibroblasts were reduced presence of free ribosomes, the appearance of elongated endoplasmic reticulum and strikingly increased number of mitochondria and small vesicles. When chronically treated with choline, those characteristics disappeared and mutant ultrastructure resembled healthy control cells. Functional analysis revealed diminished choline transport yet the membrane phosphatidylcholine content remained unchanged. As part of the mechanism to preserve choline and phosphatidylcholine, choline transporter deficiency was implicated in impaired membrane homeostasis of other phospholipids. Choline treatments could restore the membrane lipids, repair cellular organelles and protect mutant cells from acute iron overload. In conclusion, we describe a novel childhood-onset neurometabolic disease caused by choline transporter deficiency with autosomal recessive inheritance.
- Published
- 2019
- Full Text
- View/download PDF
15. Detection of 6-demethoxyubiquinone in CoQ10 deficiency disorders: Insights into enzyme interactions and identification of potential therapeutics
- Author
-
Daniela Karall, Felix Distelmaier, Anna Wredenberg, Anna Wedell, Luis C. López, Gisela Bünning, Annette Seibt, Christoph Freyer, Diran Herebian, Holger Prokisch, Ertan Mayatepek, and Sander H. J. Smits
- Subjects
0301 basic medicine ,Coenzyme Q10 ,chemistry.chemical_classification ,Chemistry ,Endocrinology, Diabetes and Metabolism ,Metabolite ,Oxidative phosphorylation ,Mitochondrion ,Biochemistry ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,Endocrinology ,Enzyme ,Biosynthesis ,Cell culture ,Genetics ,COQ7 ,Molecular Biology - Abstract
Coenzyme Q10 (CoQ10) is an essential cofactor of the mitochondrial oxidative phosphorylation (OXPHOS) system and its deficiency has important implications for several inherited metabolic disorders of childhood. The biosynthesis of CoQ10 is a complicated process, which involves at least 12 different enzymes. One of the metabolic intermediates that are formed during CoQ10 biosynthesis is the molecule 6-demethoxyubiquinone (6-DMQ). This CoQ precursor is processed at the level of COQ7 and COQ9. We selected this metabolite as a marker substance for metabolic analysis of cell lines with inherited genetic defects (COQ2, COQ4, COQ7 and COQ9) or siRNA knockdown in CoQ biosynthesis enzymes using ultra-performance liquid chromatography coupled to tandem mass spectrometry (UPLC-MS/MS). In COQ4, COQ7 and COQ9 deficient cell lines, we detected significantly elevated levels of 6-DMQ. This suggests a functional interplay of these proteins. However, additional siRNA studies demonstrated that elevated 6-DMQ levels are not an exclusive marker of the COQ7/COQ9 enzymatic step of CoQ10 biosynthesis but constitute a more general phenomenon that occurs in disorders impairing the function or stability of the CoQ-synthome. To further investigate the interdependence of CoQ10 biosynthesis enzyme expression, we performed immunoblotting in various cell lines with CoQ10 deficiency, indicating that COQ4, COQ7 and COQ9 protein expression levels are highly regulated depending on the underlying defect. Supplementation of cell lines with synthetic CoQ precursor compounds demonstrated beneficial effects of 2,4-dihydroxybenzoic acid in COQ7 and COQ9 deficiency. Moreover, vanillic acid selectively stimulated CoQ10 biosynthesis and improved cell viability in COQ9 deficiency. However, compounds tested in this study failed to rescue COQ4 deficiency.
- Published
- 2017
- Full Text
- View/download PDF
16. Coexisting variants in OSTM1 and MANEAL cause a complex neurodegenerative disorder with NBIA-like brain abnormalities
- Author
-
Thomas Meitinger, Tobias B. Haack, Diran Herebian, Stefani Harmsen, Ertan Mayatepek, Felix Distelmaier, Thomas Schwarzmayr, Annette Seibt, Bader Alhaddad, Ansgar Schulz, Katharina Danhauser, Tim M. Strom, and Dirk Klee
- Subjects
Male ,0301 basic medicine ,Candidate gene ,Pathology ,medicine.medical_specialty ,Neurodegeneration with brain iron accumulation ,Ubiquitin-Protein Ligases ,Short Report ,Context (language use) ,Biology ,Diagnosis, Differential ,03 medical and health sciences ,0302 clinical medicine ,Global brain atrophy ,Mannosidases ,Genetics ,medicine ,Humans ,Child ,Genetics (clinical) ,Exome sequencing ,Brain Diseases, Metabolic ,Neurodegeneration ,Brain ,Membrane Proteins ,Neurodegenerative Diseases ,medicine.disease ,Iron Metabolism Disorders ,030104 developmental biology ,Mutation ,Hereditary Diseases ,Mannose ,030217 neurology & neurosurgery ,Infantile malignant osteopetrosis - Abstract
Coexistence of different hereditary diseases is a known phenomenon in populations with a high consanguinity rate. The resulting clinical phenotypes are extremely challenging for physicians involved in the care of these patients. Here we describe a 6-year-old boy with co-occurrence of a homozygous splice defect in OSTM1, causing infantile malignant osteopetrosis, and a loss-of-function variant in MANEAL, which has not been associated with human disease so far. The child suffered from severe infantile-onset neurodegeneration that could not be stopped by bone marrow transplantation. Magnetic resonance imaging demonstrated global brain atrophy and showed hypointensities of globus pallidus, corpora mamillaria, and cerebral peduncles, which were comparable to findings in neurodegeneration with brain iron accumulation disorders. LC-MS/MS analysis of urine and cerebrospinal fluid samples revealed a distinct metabolic profile with accumulation of mannose tetrasaccharide molecules, suggestive of an oligosaccharide storage disease. Our results demonstrate that exome sequencing is a very effective tool in dissecting complex neurological diseases. Moreover, we suggest that MANEAL is an interesting candidate gene that should be considered in the context of neurological disorders with brain iron accumulation and/or indications of an oligosaccharide storage disease.
- Published
- 2017
- Full Text
- View/download PDF
17. Modification of the fatty acid composition of an obesogenic diet improves the maternal and placental metabolic environment in obese pregnant mice
- Author
-
Raphaela Kübeck, Eckhard Wolf, Martina Gimpfl, Annette Seibt, Martin Klingenspor, Maik Dahlhoff, Andreas Blutke, Birgit Rathkolb, Soner Öner-Sieben, Jan Rozman, Regina Ensenauer, Martin Hrabě de Angelis, and Adelbert A. Roscher
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Placenta ,Perilipin 2 ,Mice, Obese ,Pregnancy Proteins ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,Pregnancy ,Internal medicine ,Adipocyte ,medicine ,Animals ,Obesity ,Molecular Biology ,chemistry.chemical_classification ,ACACA ,030109 nutrition & dietetics ,biology ,Leptin ,Gene Expression Regulation, Developmental ,Fatty acid ,obesity ,nutrition ,medium-chain fatty acids ,n-3 fatty acids ,placenta ,Pregnancy Complications ,Fatty acid synthase ,030104 developmental biology ,Endocrinology ,chemistry ,Lipogenesis ,Fatty Acids, Unsaturated ,biology.protein ,Molecular Medicine ,Female ,Polyunsaturated fatty acid - Abstract
Peri-conceptional exposure to maternal obesogenic nutrition is associated with in utero programming of later-life overweight and metabolic disease in the offspring. We aimed to investigate whether dietary intervention with a modified fatty acid quality in an obesogenic high-calorie (HC) diet during the preconception and gestational phases can improve unfavourable effects of an adipogenic maternal environment. In NMRI mice, peri-conceptional and gestational obesity was induced by feeding a HC diet (controls), and they were compared with dams on a fat-modified (Fat-mod) HC diet of the same energy content but enriched with medium-chain fatty acids (MCFAs) and adjusted to a decreased ratio of n-6 to n-3 long-chain polyunsaturated fatty acids (LC-PUFAs). Effects on maternal and placental outcomes at delivery (day 17.5 post coitum) were investigated. Despite comparable energy assimilation between the two groups of dams, the altered fatty acid composition of the Fat-mod HC diet induced lower maternal body weight, weights of fat depots, adipocyte size, and hepatic fat accumulation compared to the unmodified HC diet group. Further, there was a trend towards lower fasting glucose, insulin and leptin concentrations in dams fed the Fat-mod HC diet. Phenotypic changes were accompanied by inhibition of transcript and protein expression of genes involved in hepatic de novo lipogenesis comprising PPARG2 and its target genes Fasn, Acaca, and Fabp4, whereas regulation of other lipogenic factors (Srebf1, Nr1h3, Abca1) appeared to be more complex. The modified diet led to a sex-specific placental response by upregulating PPARG-dependent fatty acid transport gene expression in female versus male placentae. Qualitative modification of the fatty acid spectrum of a high-energy maternal diet, using a combination of both MCFAs and n-3 LC-PUFAs, seems to be a promising interventional approach to ameliorate the adipogenic milieu of mice before and during gestation.
- Published
- 2017
- Full Text
- View/download PDF
18. Neonatal encephalocardiomyopathy caused by mutations in VARS2
- Author
-
Thomas Meitinger, Sonja Budaeus, Tim M. Strom, Annette Seibt, Bader Alhaddad, Felix Distelmaier, Jörg Schaper, Holger Prokisch, Ertan Mayatepek, Tobias B. Haack, and Fabian Baertling
- Subjects
Male ,0301 basic medicine ,Mitochondrial encephalomyopathy ,Pathology ,medicine.medical_specialty ,Valine-tRNA Ligase ,Mitochondrial disease ,DNA Mutational Analysis ,Cardiomyopathy ,Biology ,Compound heterozygosity ,Corpus callosum ,Biochemistry ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,HLA Antigens ,Mitochondrial Encephalomyopathies ,medicine ,Humans ,Exome ,Exome sequencing ,Epilepsy ,Cardiomyopathy, Hypertrophic ,medicine.disease ,VARS2 ,OXPHOS ,Aminoacyl tRNA synthtetas ,Hypoplasia ,030104 developmental biology ,Mutation ,Neurology (clinical) ,Severe lactic acidosis - Abstract
VARS2 encodes a mitochondrial aminoacyl-tRNA-synthetase. Mutations in VARS2 have recently been identified as a cause of mitochondrial encephalomyopathy in three individuals. However, clinical information remained scarce. Exome sequencing lead us to identify compound heterozygous pathogenic VARS2 variants in a boy presenting with severe lactic acidosis, hypertrophic cardiomyopathy, epilepsy, and abnormalities on brain imaging including hypoplasia of corpus callosum and cerebellum as well as a massive lactate peak on MR-spectroscopy. Studies in patient-derived fibroblasts confirmed the functional relevance of the identified VARS2 variants. Our report expands the phenotypic spectrum associated with this rare mitochondrial defect, in that VARS2 deficiency may also cause severe neonatal presentations with cardiac involvement and structural brain abnormalities.
- Published
- 2016
- Full Text
- View/download PDF
19. Biallelic DMXL2 mutations impair autophagy and cause Ohtahara syndrome with progressive course
- Author
-
Matias Wagner, Hava Hafner, Erez Y. Levanon, Alessandro Esposito, Antonio Falace, Silvia Giovedì, Antonio De Fusco, Tiziana Pisano, Sarit Ravid, Hanna Mandel, Davide Aprile, Ayelet Eran, Sarah L. Stenton, Saskia B. Wortmann, Annette Seibt, Maria Sabina Cerullo, Davide Mei, Valerio Conti, Daniella Magen, Carla Marini, Chiara Fiorillo, Felix Distelmaier, Ertan Mayatepek, Fabio Benfenati, Tilman Polster, Moran Gal, Anna Fassio, and Renzo Guerrini
- Subjects
0301 basic medicine ,Male ,medicine.medical_specialty ,Ohtahara syndrome ,Synaptogenesis ,Nerve Tissue Proteins ,Compound heterozygosity ,autophagy ,developmental and epileptic encephalopathy ,neuropathy ,progressive disorder ,Leukoencephalopathy ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Exome Sequencing ,Autophagy ,Medicine ,Missense mutation ,Humans ,Child ,Developmental And Epileptic Encephalopathy ,Neuropathy ,Ohtahara Syndrome ,Progressive Disorder ,Loss function ,Exome sequencing ,Adaptor Proteins, Signal Transducing ,business.industry ,Brain ,Infant ,Electroencephalography ,medicine.disease ,Magnetic Resonance Imaging ,Pedigree ,030104 developmental biology ,Endocrinology ,Child, Preschool ,Mutation ,Disease Progression ,Female ,Neurology (clinical) ,business ,Lysosomes ,Spasms, Infantile ,030217 neurology & neurosurgery - Abstract
Ohtahara syndrome, early infantile epileptic encephalopathy with a suppression burst EEG pattern, is an aetiologically heterogeneous condition starting in the first weeks or months of life with intractable seizures and profound developmental disability. Using whole exome sequencing, we identified biallelic DMXL2 mutations in three sibling pairs with Ohtahara syndrome, belonging to three unrelated families. Siblings in Family 1 were compound heterozygous for the c.5135C>T (p.Ala1712Val) missense substitution and the c.4478C>G (p.Ser1493*) nonsense substitution; in Family 2 were homozygous for the c.4478C>A (p.Ser1493*) nonsense substitution and in Family 3 were homozygous for the c.7518-1G>A (p.Trp2507Argfs*4) substitution. The severe developmental and epileptic encephalopathy manifested from the first day of life and was associated with deafness, mild peripheral polyneuropathy and dysmorphic features. Early brain MRI investigations in the first months of life revealed thin corpus callosum with brain hypomyelination in all. Follow-up MRI scans in three patients revealed progressive moderate brain shrinkage with leukoencephalopathy. Five patients died within the first 9 years of life and none achieved developmental, communicative or motor skills following birth. These clinical findings are consistent with a developmental brain disorder that begins in the prenatal brain, prevents neural connections from reaching the expected stages at birth, and follows a progressive course. DMXL2 is highly expressed in the brain and at synaptic terminals, regulates v-ATPase assembly and activity and participates in intracellular signalling pathways; however, its functional role is far from complete elucidation. Expression analysis in patient-derived skin fibroblasts demonstrated absence of the DMXL2 protein, revealing a loss of function phenotype. Patients' fibroblasts also exhibited an increased LysoTracker® signal associated with decreased endolysosomal markers and degradative processes. Defective endolysosomal homeostasis was accompanied by impaired autophagy, revealed by lower LC3II signal, accumulation of polyubiquitinated proteins, and autophagy receptor p62, with morphological alterations of the autolysosomal structures on electron microscopy. Altered lysosomal homeostasis and defective autophagy were recapitulated in Dmxl2-silenced mouse hippocampal neurons, which exhibited impaired neurite elongation and synaptic loss. Impaired lysosomal function and autophagy caused by biallelic DMXL2 mutations affect neuronal development and synapse formation and result in Ohtahara syndrome with profound developmental impairment and reduced life expectancy.
- Published
- 2019
20. MRPS22 mutation causes fatal neonatal lactic acidosis with brain and heart abnormalities
- Author
-
Berit Hadzik, Fabian Baertling, Holger Prokisch, Annette Seibt, Felix Distelmaier, Ertan Mayatepek, Richard J. Rodenburg, Tobias B. Haack, Gündüz Selcan, Jörg Schaper, Tim M. Strom, and Thomas Meitinger
- Subjects
Male ,Ribosomal Proteins ,Pathology ,medicine.medical_specialty ,Mitochondrial Diseases ,Heart malformation ,Mitochondrial disease ,Oxidative phosphorylation ,Biology ,medicine.disease_cause ,Mitochondrial Proteins ,Cellular and Molecular Neuroscience ,Fatal Outcome ,Genetics ,Mitochondrial ribosome ,medicine ,Humans ,Agenesis of the corpus callosum ,Frameshift Mutation ,Genetics (clinical) ,Mutation ,Myocardium ,Infant, Newborn ,Brain ,Metabolic Disorders Radboud Institute for Molecular Life Sciences [Radboudumc 6] ,Fibroblasts ,medicine.disease ,Molecular medicine ,Lactic acidosis ,Acidosis, Lactic - Abstract
The mitochondrial ribosomes are required for the synthesis of mitochondrial DNA-encoded subunits of the oxidative phosphorylation (OXPHOS) system. Here, we present a neonate with fatal lactic acidosis and combined OXPHOS deficiency caused by a homozygous mutation in MRPS22, a gene encoding a mitochondrial ribosomal small subunit protein. Brain imaging revealed several structural abnormalities, including agenesis of the corpus callosum, multiple periventricular cysts, and suspected intracerebral calcifications. Moreover, echocardiography demonstrated atrial and ventricular septal defects as well as a coronary artery fistula. Our report expands the clinical spectrum of this rare mitochondrial disorder and confirms the severe clinical phenotype associated with this defect.
- Published
- 2015
21. 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
22. Detection of 6-demethoxyubiquinone in CoQ
- Author
-
Diran, Herebian, Annette, Seibt, Sander H J, Smits, Gisela, Bünning, Christoph, Freyer, Holger, Prokisch, Daniela, Karall, Anna, Wredenberg, Anna, Wedell, Luis C, López, Ertan, Mayatepek, and Felix, Distelmaier
- Subjects
Vanillic Acid ,Mitochondrial Diseases ,Muscle Weakness ,Cell Survival ,Ubiquinone ,Fibroblasts ,Oxidative Phosphorylation ,Cell Line ,Mitochondria ,Mitochondrial Proteins ,Tandem Mass Spectrometry ,Hydroxybenzoates ,Humans ,Ataxia - Abstract
Coenzyme Q
- Published
- 2017
23. 4-Hydroxybenzoic acid restores CoQ(10) biosynthesis in human COQ2 deficiency
- Author
-
Annette Seibt, Diran Herebian, Felix Distelmaier, Richard J. Rodenburg, Sander H. J. Smits, and Ertan Mayatepek
- Subjects
0301 basic medicine ,General Neuroscience ,In silico ,Endogeny ,Metabolic Disorders Radboud Institute for Molecular Life Sciences [Radboudumc 6] ,Biology ,Brief Communication ,Ligand (biochemistry) ,Phenotype ,Cell biology ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,Biochemistry ,Biosynthesis ,chemistry ,Cell culture ,Mitochondrial matrix ,Neurology (clinical) ,Viability assay ,Brief Communications ,GeneralLiterature_REFERENCE(e.g.,dictionaries,encyclopedias,glossaries) ,030217 neurology & neurosurgery - Abstract
The clinical phenotypes of human CoQ10‐deficiency caused by COQ2 mutations range from fatal neonatal disease to adult‐onset multisystem atrophy. So far, treatment options for these diseases are unsatisfactory. Here, we demonstrate that supplementation of 4‐hydroxybenzoic acid (4‐HBA) fully restores endogenous CoQ10‐biosynthesis in COQ2‐deficient cell lines. This was accompanied by increased protein expression of CoQ10‐biosynthesis‐enzymes as well as a rescue of cell viability during stress conditions. In silico analysis suggested a ligand transportation path for 4‐HBA through the COQ2 protein towards the mitochondrial matrix side. This process is apparently hindered by disease‐causing mutations, which can be overcome by increasing 4‐HBA concentrations.
- Published
- 2017
24. Pre-exercise medium-chain triglyceride application prevents acylcarnitine accumulation in skeletal muscle from very-long-chain acyl-CoA-dehydrogenase-deficient mice
- Author
-
Sara Tucci, Annette Seibt, Diran Herebian, Frank ter Veld, Sonja Primassin, Ute Spiekerkoetter, and Lars Hoffmann
- Subjects
medicine.medical_specialty ,Physical exercise ,Biology ,Long-chain acyl-CoA dehydrogenase ,Mice ,chemistry.chemical_compound ,Carnitine ,Physical Conditioning, Animal ,Internal medicine ,Oxazines ,Genetics ,medicine ,Animals ,Medium-chain triglyceride ,Muscle, Skeletal ,Beta oxidation ,Triglycerides ,Genetics (clinical) ,Mice, Knockout ,Triglyceride ,Acyl-CoA Dehydrogenase, Long-Chain ,Skeletal muscle ,medicine.disease ,Animal Feed ,Lipids ,Oxygen ,Endocrinology ,medicine.anatomical_structure ,chemistry ,Acyl Coenzyme A ,Rhabdomyolysis ,medicine.drug - Abstract
Dietary modification with medium-chain triglyceride (MCT) supplementation is one crucial way of treating children with long-chain fatty acid oxidation disorders. Recently, supplementation prior to exercise has been reported to prevent muscular pain and rhabdomyolysis. Systematic studies to determine when MCT supplementation is most beneficial have not yet been undertaken. We studied the effects of an MCT-based diet compared with MCT administration only prior to exercise in very-long-chain acyl-CoA dehydrogenase (VLCAD) knockout (KO) mice. VLCAD KO mice were fed an MCT-based diet in same amounts as normal mouse diet containing long-chain triglycerides (LCT) and were exercised on a treadmill. Mice fed a normal LCT diet received MCT only prior to exercise. Acylcarnitine concentration, free carnitine concentration, and acyl-coenzyme A (CoA) oxidation capacity in skeletal muscle as well as hepatic lipid accumulation were determined. Long-chain acylcarnitines significantly increased in VLCAD-deficient skeletal muscle with an MCT diet compared with an LCT diet with MCT bolus prior to exercise, whereas an MCT bolus treatment significantly decreased long-chain acylcarnitines after exercise compared with an LCT diet. C8-carnitine was significantly increased in skeletal muscle after MCT bolus treatment and exercise compared with LCT and long-term MCT treatment. Increased hepatic lipid accumulation was observed in long-term MCT-treated KO mice. MCT seems most beneficial when given in a single dose directly prior to exercise to prevent acylcarnitine accumulation. In contrast, continuous MCT treatment produces a higher skeletal muscle content of long-chain acylcarnitines after exercise and increases hepatic lipid storage in VLCAD KO mice.
- Published
- 2010
- Full Text
- View/download PDF
25. Polar bacterial invasion and translocation ofStreptococcus suisacross the blood-cerebrospinal fluid barrierin vitro
- Author
-
Hans-Joachim Galla, Corinna Wewer, Dorothee Gellrich, Annette Seibt, Rüdiger Adam, Ulrike Friedrichs, Tobias Tenenbaum, Christian Schwerk, Thalia Papandreou, and Horst Schroten
- Subjects
Streptococcus suis ,Swine ,Immunology ,Chromosomal translocation ,Vacuole ,Biology ,Microbiology ,Phosphatidylinositol 3-Kinases ,chemistry.chemical_compound ,Microscopy, Electron, Transmission ,Virology ,Animals ,Phosphatidylinositol ,Transcellular ,Cells, Cultured ,Barrier function ,Phosphoinositide-3 Kinase Inhibitors ,Kinase ,Epithelial Cells ,biology.organism_classification ,Cell biology ,chemistry ,Blood-Brain Barrier ,Choroid Plexus ,Vacuoles ,Choroid plexus - Abstract
Previous experimental studies in a standard Transwell culture system have shown Streptococcus suis ability to compromise barrier function of porcine choroid plexus epithelial cells (PCPEC). The development of an 'inverted' Transwell filter system of PCPEC enables us now for the first time to investigate bacterial invasion and translocation from the physiologically relevant basolateral (blood) to the apical (cerebrospinal fluid) side. Most importantly, we observed specific invasion and translocation of S. suis across the PCPEC exclusively from the basolateral side. During this process, bacterial viability and the presence of a capsule as well as cytoskeletal regulation of PCPEC seemed to play an important role. No loss of barrier function was observed. Bacterial translocation could be significantly inhibited by the phosphatidylinositol 3-kinase inhibitor LY294002, but not by its inactive analogue Ly303511 or dexamethasone. Apotome imaging as well as electron microscopy revealed intracellular bacteria often in cell vacuoles. Thus, possibly regulated by the presence of a capsule, S. suis induces signals that depend on the lipid kinase phosphatidylinositol 3-kinase pathway, which paves the way for cellular uptake during the bacterial transcellular translocation process. Taken together, our data underline the relevance of the blood-cerebrospinal fluid barrier as a gate for bacterial entry into the central nervous system.
- Published
- 2009
- Full Text
- View/download PDF
26. 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
27. EARS2 mutations cause fatal neonatal lactic acidosis, recurrent hypoglycemia and agenesis of corpus callosum
- Author
-
Marlen Melcher, Tobias B. Haack, Katharina Danhauser, Dirk Klee, Tim M. Strom, Bader Alhaddad, Thomas Meitinger, Holger Prokisch, Annette Seibt, Ertan Mayatepek, and Felix Distelmaier
- Subjects
0301 basic medicine ,Male ,medicine.medical_specialty ,Mitochondrial Diseases ,Mitochondrial translation ,Mitochondrial disease ,Recurrent hypoglycemia ,Oxidative phosphorylation ,Biology ,Corpus callosum ,Biochemistry ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,chemistry.chemical_compound ,0302 clinical medicine ,Fatal Outcome ,Recurrence ,Internal medicine ,medicine ,Humans ,Ultrasonography ,Genetics ,Aminoacyl tRNA synthetase ,Infant, Newborn ,Brain ,medicine.disease ,Hypoglycemia ,Glutamate-tRNA Ligase ,030104 developmental biology ,Endocrinology ,chemistry ,Lactic acidosis ,Agenesis ,Acidosis, Lactic ,Neurology (clinical) ,Agenesis of Corpus Callosum ,030217 neurology & neurosurgery - Abstract
Mitochondrial aminoacyl tRNA synthetases are essential for organelle protein synthesis. Genetic defects affecting the function of these enzymes may cause pediatric mitochondrial disease. Here, we report on a child with fatal neonatal lactic acidosis and recurrent hypoglycemia caused by mutations in EARS2, encoding mitochondrial glutamyl-tRNA synthetase 2. Brain ultrasound revealed agenesis of corpus callosum. Studies on patient-derived skin fibroblasts showed severely decreased EARS2 protein levels, elevated reactive oxygen species (ROS) production, and altered mitochondrial morphology. Our report further illustrates the clinical spectrum of the severe neonatal-onset form of EARS2 mutations. Moreover, in this case the live-cell parameters appeared to be more sensitive to mitochondrial dysfunction compared to standard diagnostics, which indicates the potential relevance of fibroblast studies in children with mitochondrial diseases.
- Published
- 2015
28. Cell death, caspase activation, and HMGB1 release of porcine choroid plexus epithelial cells during Streptococcus suis infection in vitro
- Author
-
Annette Seibt, Horst Schroten, Hans-Joachim Galla, Gerd E. K. Novotny, Tobias Tenenbaum, Frank Essmann, Rüdiger Adam, and Reiner U. Jänicke
- Subjects
Programmed cell death ,Necrosis ,Streptococcus suis ,Swine ,Apoptosis ,Enzyme-Linked Immunosorbent Assay ,DNA Fragmentation ,Biology ,Cerebrospinal fluid ,Microscopy, Electron, Transmission ,Streptococcal Infections ,medicine ,Choroid Plexus Epithelium ,Animals ,Viability assay ,HMGB1 Protein ,Molecular Biology ,Cells, Cultured ,Cell Nucleus ,Cell Death ,L-Lactate Dehydrogenase ,General Neuroscience ,Epithelial Cells ,biology.organism_classification ,Immunohistochemistry ,Molecular biology ,Enzyme Activation ,Caspases ,Choroid Plexus ,Immunology ,Choroid plexus ,Neurology (clinical) ,medicine.symptom ,Developmental Biology - Abstract
The choroid plexus epithelium constitutes the structural basis of the blood-cerebrospinal fluid barrier. We previously demonstrated that Streptococcus suis (S. suis), a relevant cause of bacterial meningitis in pigs and humans, affects porcine choroid plexus epithelial cell (PCPEC) barrier function and integrity. We now characterized PCPEC cell death and investigated whether apoptosis or necrosis is responsible for the cytotoxicity after infection with different S. suis isolates. We found S. suis strain-dependent histone associated DNA-fragments quantified by ELISA. This response could partially be inhibited by cylcoheximide, cytochalasin D, dexamethasone, herbimycin A, but most effectively by the pan-caspase inhibitor zVAD-fmk. We further detected caspase-3 and -9 activation after infection with all tested S. suis isolates that could also be blocked by zVAD-fmk. However, we found a significantly stronger caspase activity with the protein kinase inhibitor staurosporine. All tested S. suis isolates induced loss of cell viability in PCPEC as shown with the Live/Dead assay, but strain dependent lactate dehydrogenase-release. Both parameters could not be influenced by zVAD-fmk. Immunostaining showed release of high-mobility group box 1 (HMGB1) protein from the nucleus, indicative of necrosis. Transmission electron microscopy showed cell swelling, cytoplasmic vacuolization, loss of membrane integrity, nuclear fermentation but no nuclear condensation, indices for a primarily necrotic cell morphology. Taken together, our findings indicate that S. suis causes cell death in PCPEC by different mechanisms. Although apoptosis may be involved in the process of PCPEC cell death, necrosis seems to be the predominant mechanism. Through inflammation in the choroid plexus during bacterial meningitis, the blood-cerebrospinal fluid barrier function will be compromised.
- Published
- 2006
- Full Text
- View/download PDF
29. Polyethylene Glycol-Conjugated Adenosine Deaminase (ADA) Therapy Provides Temporary Immune Reconstitution to a Child with Delayed-Onset ADA Deficiency
- Author
-
Elke Lainka, Michael S. Hershfield, W. Friedrich, Ines Santisteban, Pawan Bali, Annette Seibt, Jennifer Neubert, and Tim Niehues
- Subjects
CD4-Positive T-Lymphocytes ,Microbiology (medical) ,Interleukin 2 ,Purine-Pyrimidine Metabolism, Inborn Errors ,medicine.medical_specialty ,Adenosine Deaminase ,medicine.medical_treatment ,Lymphocyte ,Clinical Biochemistry ,Immunology ,Apoptosis ,Biology ,Lymphocyte Activation ,Polyethylene Glycols ,Mice ,Leukocyte Count ,Immune system ,Adenosine deaminase ,T-Lymphocyte Subsets ,Cellular Immunology ,Internal medicine ,medicine ,Animals ,Humans ,Immunology and Allergy ,Letters to the Editor ,Cells, Cultured ,Infant ,Genetic Therapy ,Enzymes, Immobilized ,Purine/pyrimidine metabolism ,medicine.disease ,Adenosine deaminase deficiency ,Endocrinology ,Cytokine ,medicine.anatomical_structure ,biology.protein ,Cytokines ,Female ,Cytokine secretion ,Stem Cell Transplantation ,medicine.drug - Abstract
We describe the effects of polyethylene glycol-conjugated adenosine deaminase (ADA) replacement therapy on lymphocyte counts, activation, apoptosis, proliferation, and cytokine secretion in a 14-month-old girl with “delayed-onset” ADA deficiency and marked immunodysregulation. Pretreatment lymphopenia affected T cells (CD4, 150/μl; CD8, 459/μl), B cells (16/μl), and NK cells (55/μl). T cells were uniformly activated and largely apoptotic (CD4, 59%; CD8, 82%); and T-cell-dependent cytokine levels in plasma were elevated, including the levels of interleukin 2 (IL-2; 26 pg/ml), IL-4 (81 pg/ml), IL-5 (46 pg/ml), gamma interferon (1,430 pg/ml), tumor necrosis factor alpha (210 pg/ml), and IL-10 (168 pg/ml). Mitogen-stimulated peripheral blood mononuclear cells show reduced IL-2 secretion and proliferation. During the first 5 months of therapy there was clinical improvement and partial immune reconstitution, with nearly normal lymphocyte subset numbers, reduced T-cell activation and CD4-cell apoptosis, and decreased plasma cytokine levels. In parallel, IL-2 secretion and the lymphocyte mitogenic response improved. Between 4 and 7 months, immunoglobulin G antibodies to bovine ADA developed and resulted in the complete reversal of immune recovery.
- Published
- 2005
- Full Text
- View/download PDF
30. Strain-dependent disruption of bloodâcerebrospinal fluid barrier byStreptoccocus suisin vitro
- Author
-
David Matalon, Annette Seibt, Hans-Joachim Galla, Ingo Eggelnpöhler, Rüdiger Adam, Tobias Tenenbaum, Horst Schroten, and Gerd E. K. Novotny
- Subjects
Microbiology (medical) ,Cell Membrane Permeability ,Streptococcus suis ,Sus scrofa ,Immunology ,Virulence ,In Vitro Techniques ,Microbiology ,Membrane Potentials ,chemistry.chemical_compound ,Species Specificity ,Streptococcal Infections ,Lactate dehydrogenase ,Choroid Plexus Epithelium ,Animals ,Humans ,Immunology and Allergy ,Mannitol ,Serotyping ,Cells, Cultured ,Barrier function ,Swine Diseases ,biology ,Epithelial Cells ,General Medicine ,biology.organism_classification ,In vitro ,Microscopy, Electron ,Infectious Diseases ,chemistry ,Blood-Brain Barrier ,Paracellular transport ,Choroid Plexus ,Choroid plexus - Abstract
Streptococcus suis capsular type 2 is an important agent of diseases including meningitis among pigs worldwide, and is also a zoonotic agent. The barrier function of the choroid plexus epithelium that constitutes the structural basis for the blood-cerebrospinal fluid (CSF) barrier has not been elucidated yet in bacterial meningitis. We investigated the influence of various S. suis isolates on the barrier function of cultured porcine choroid plexus epithelial cells with respect to the transepithelial resistance and paracellular [(3)H]-mannitol flux. Preferentially apical application of S. suis isolates significantly decreased transepithelial resistance and significantly increased paracellular [(3)H]-mannitol flux in a time-, dose- and strain-dependent manner. Viable S. suis isolates caused cytotoxicity determined by lactate dehydrogenase assay and electron microscopy, whereas S. suis sonicates and UV-inactivated S. suis did not cause cytotoxicity. The observed effects on porcine choroid plexus epithelial cells barrier function could not exclusively be ascribed to known virulence factors of S. suis such as suilysin. In conclusion, S. suis isolates induce loss of blood-cerebrospinal fluid barrier function in an in vitro model. Thus, S. suis may facilitate trafficking of bacteria and leucocytes across the blood-cerebrospinal fluid barrier. The underlying mechanisms for the barrier breakdown have yet to be determined.
- Published
- 2005
- Full Text
- View/download PDF
31. Mitochondrial dysfunction in primary human fibroblasts triggers an adaptive cell survival program that requires AMPK-alpha
- Author
-
Annette Seibt, Richard J. Rodenburg, Felix Distelmaier, Katharina Danhauser, Mariusz R. Wieckowski, Peter H.G.M. Willems, Werner J.H. Koopman, Jack A.M. Fransen, Magdalena Lebiedzinska, Jaap Keijer, Dania C. Liemburg-Apers, Jan A.M. Smeitink, Sandra G. Heil, Frank N. Gellerich, Benoit Viollet, Hiromi Imamura, and Federica Valsecchi
- Subjects
mammalian-cells ,MFN2 ,activated protein-kinase ,AMP-Activated Protein Kinases ,Endoplasmic Reticulum ,Mice ,Sirtuin 1 ,Cell Line, Transformed ,Membrane Potential, Mitochondrial ,Mice, Knockout ,calcium homeostasis ,TOR Serine-Threonine Kinases ,Metabolic Disorders Radboud Institute for Molecular Life Sciences [Radboudumc 6] ,Respirometry ,glycolysis ,Mitochondria ,cancer-cells ,human nadh ,Human and Animal Physiology ,Molecular Medicine ,ubiquinone oxidoreductase deficiency ,Signal Transduction ,medicine.medical_specialty ,Cell Survival ,Calcium pump ,respiratory-chain dysfunction ,SOD1 ,SOD2 ,Oxidative phosphorylation ,Biology ,energy-metabolism ,Chlorides ,Internal medicine ,medicine ,Animals ,Humans ,redox signaling ,Molecular Biology ,PI3K/AKT/mTOR pathway ,atp production ,AMPK ,Fibroblasts ,complex-i deficiency ,mitochondrial dynamics ,Oxidative Stress ,Endocrinology ,Electron Transport Chain Complex Proteins ,Mitochondrial biogenesis ,WIAS ,Fysiologie van Mens en Dier ,Calcium ,oxidative-phosphorylation ,metabolic regulation ,Nanomedicine Radboud Institute for Molecular Life Sciences [Radboudumc 19] ,Transcription Factors - Abstract
Contains fulltext : 153406.pdf (Publisher’s version ) (Closed access) Dysfunction of complex I (CI) of the mitochondrial electron transport chain (ETC) features prominently in human pathology. Cell models of ETC dysfunction display adaptive survival responses that still are poorly understood but of relevance for therapy development. Here we comprehensively examined how primary human skin fibroblasts adapt to chronic CI inhibition. CI inhibition triggered transient and sustained changes in metabolism, redox homeostasis and mitochondrial (ultra)structure but no cell senescence/death. CI-inhibited cells consumed no oxygen and displayed minor mitochondrial depolarization, reverse-mode action of complex V, a slower proliferation rate and futile mitochondrial biogenesis. Adaptation was neither prevented by antioxidants nor associated with increased PGC1-alpha/SIRT1/mTOR levels. Survival of CI-inhibited cells was strictly glucose-dependent and accompanied by increased AMPK-alpha phosphorylation, which occurred without changes in ATP or cytosolic calcium levels. Conversely, cells devoid of AMPK-alpha died upon CI inhibition. Chronic CI inhibition did not increase mitochondrial superoxide levels or cellular lipid peroxidation and was paralleled by a specific increase in SOD2/GR, whereas SOD1/CAT/Gpx1/Gpx2/Gpx5 levels remained unchanged. Upon hormone stimulation, fully adapted cells displayed aberrant cytosolic and ER calcium handling due to hampered ATP fueling of ER calcium pumps. It is concluded that CI dysfunction triggers an adaptive program that depends on extracellular glucose and AMPK-alpha. This response avoids cell death by suppressing energy crisis, oxidative stress induction and substantial mitochondrial depolarization.
- Published
- 2015
32. Anti-ganglioside monoclonal antibody AA4 selectively inhibits IgE-induced signal transduction pathways in rat basophilic leukemia cells
- Author
-
Annette Seibt, V. Stephan, Elsa H. Berenstein, Reuben P. Siraganian, V. Wahn, Dusan Dukanovic, William D. Swaim, and Marius Skasa
- Subjects
Cytoplasm ,Serotonin ,Immunology ,Phosphatidylinositols ,Immunoglobulin E ,chemistry.chemical_compound ,Gangliosides ,Tumor Cells, Cultured ,Animals ,Mast Cells ,Phosphorylation ,Phosphotyrosine ,Receptor ,Molecular Biology ,Ganglioside ,biology ,Receptors, IgE ,Degranulation ,Antibodies, Monoclonal ,Muscarinic acetylcholine receptor M3 ,Tyrosine phosphorylation ,Transfection ,Phosphoproteins ,Molecular biology ,Rats ,chemistry ,biology.protein ,Calcium ,Signal transduction ,Signal Transduction - Abstract
In rat basophilic leukemia 2H3 (RBL-2H3) cells, mAb AA4 binds to two derivatives of ganglioside GD1b that associate with the Src family kinase p53/56lyn and a serine kinase. Pre-incubation of cells with mAb AA4 blocks immunoglobulin E (IgE) mediated histamine release. In the present study we investigated the effect of incubation with mAb AA4 on signal transduction events. In addition to stimulation of the high affinity IgE receptor (Fc epsilonRI), cells were also activated by the calcium ionophore A23187 and the acetylcholine agonist carbachol in RBL-2H3 cells transfected with the G protein-coupled m3 muscarinic receptor. Incubation of cells with mAb AA4 in a dose-dependent manner inhibited the following Fc epsilonRI-induced signal transduction events: the increase of intracellular free calcium, phosphoinositol breakdown, tyrosine phosphorylation of proteins including the beta- of Fc epsilonRI and secretion. However, there was no inhibition of degranulation or of these biochemical events when cells were stimulated with calcium ionophore or activated via a G protein-coupled pathway. Our results demonstrate that mAb AA4 selectively blocks Fc epsilonRI-induced cell activation at a very early step upstream of receptor tyrosine phosphorylation. As mAb AA4 has previously been found to bind to gangliosides associated with Fc epsilonRI, inhibition of very early biochemical events may be due to interaction of mAb AA4 with the Fc epsilonRI induced signal transduction cascade at the receptor level.
- Published
- 1997
- Full Text
- View/download PDF
33. Monounsaturated 14:1n-9 and 16:1n-9 fatty acids but not 18:1n-9 induce apoptosis and necrosis in murine HL-1 cardiomyocytes
- Author
-
Lars Hoffmann, Diran Herebian, Ute Spiekerkoetter, and Annette Seibt
- Subjects
medicine.medical_specialty ,Cell Survival ,Gene Expression ,Carnitine-acylcarnitine translocase ,Apoptosis ,Biology ,Biochemistry ,Mice ,Necrosis ,Internal medicine ,Cell Line, Tumor ,medicine ,Myocyte ,Animals ,Myocytes, Cardiac ,Carnitine ,Beta oxidation ,Triglycerides ,chemistry.chemical_classification ,Membrane Potential, Mitochondrial ,L-Lactate Dehydrogenase ,Reverse Transcriptase Polymerase Chain Reaction ,Organic Chemistry ,Acyl-CoA Dehydrogenase, Long-Chain ,Fatty Acids ,Fatty acid ,Acyl CoA dehydrogenase ,Cell Biology ,Toll-Like Receptor 4 ,Endocrinology ,Lipotoxicity ,chemistry ,Microscopy, Fluorescence ,biology.protein ,Fatty Acids, Unsaturated ,medicine.drug - Abstract
Patients with inborn errors of long-chain fatty acid oxidation accumulate disease-specific acylcarnitines and triacylglycerols in various tissues. Some of these patients present significant cardiac diseases such as arrhythmias and cardiomyopathy. The mechanism of how fatty acid accumulation is involved in disease pathogenesis is still unclear but apoptosis of cardiomyocytes has been suggested to be one possible mechanism of cardiomyopathy development. In this study, we measured lipid uptake and intracellular lipid accumulation after incubation of HL1 cardiomyocytes with different saturated and monounsaturated long- and medium-chain fatty acid species for various time periods and at different physiological concentrations. We assessed apoptosis induction by analyzing the mitochondrial membrane potential and TLR-4 expression as well as the composition of the accumulating triacylglycerols. We identified only 14:1 and 16:1 monounsaturated fatty acids potentially leading to an increase in TLR-4 expression and disruption of the mitochondrial membrane potential, resulting in apoptosis and necrosis in cultured cardiomyocytes. This study demonstrates significant toxicity of especially those fatty acid species in vitro that significantly accumulate in fatty acid oxidation defects presenting with cardiac disease such as very long-chain acyl-CoA dehydrogenase, carnitine acylcarnitine translocase and carnitine palmitoyl-CoA transferase deficiencies.
- Published
- 2013
34. 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
35. 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
36. Dexamethasone prevents alteration of tight junction-associated proteins and barrier function in porcine choroid plexus epithelial cells after infection with Streptococcus suis in vitro
- Author
-
Christian Schwerk, David Matalon, Corinna Wewer, Hans-Joachim Galla, Rüdiger Adam, Tobias Tenenbaum, Annette Seibt, and Horst Schroten
- Subjects
Pathology ,medicine.medical_specialty ,Time Factors ,Cell Survival ,Swine ,Streptococcaceae ,Anti-Inflammatory Agents ,Streptococcus suis ,Occludin ,Tritium ,Cell junction ,Dexamethasone ,Capillary Permeability ,medicine ,Choroid Plexus Epithelium ,Animals ,Mannitol ,Molecular Biology ,Barrier function ,Cells, Cultured ,Radiation ,Tight junction ,biology ,Dose-Response Relationship, Drug ,General Neuroscience ,Membrane Proteins ,Epithelial Cells ,biology.organism_classification ,Actin cytoskeleton ,Cell biology ,Gene Expression Regulation ,Choroid Plexus ,Choroid plexus ,Neurology (clinical) ,Developmental Biology ,Signal Transduction - Abstract
Apart from antibiotic treatment in bacterial meningitis supportive therapy including dexamethasone is widely used. In investigations on the pathogenesis of bacterial meningitis we previously demonstrated that Streptococcus suis (S. suis), a relevant cause of bacterial meningitis in pigs and humans, affects porcine choroid plexus epithelial cell (PCPEC) barrier function. The choroid plexus epithelium constitutes the structural basis of the blood-CSF barrier. Now, we investigated the role of tight junction proteins and the actin cytoskeleton of PCPEC in correlation to barrier function after S. suis infection and analyzed the influence of dexamethasone. S. suis caused massive rearrangement of the tight junction proteins ZO-1, occludin and claudin-1, caused loss of actin at the apical cell pole and induced basolateral stress fiber formation. Moreover, tight junctions were shifted from the Triton X insoluble to the Triton X soluble fraction, and additionally occludin was dephosphorylated and degraded. Infection with S. suis leads to an inflammatory response exemplified by the induction of tumor necrosis factor (TNF) alpha and matrix metalloproteinase (MMP)-3 gene activation, which correlated with phosphorylation of extracellular signal regulated kinases (ERKs). Importantly, dexamethasone significantly prevented S.suis-induced protein and morphological tight junction alterations and attenuated ERK activation and MMP-3 expression. It especially improved the barrier function by preventing tight junction protein reorganization and degradation. In the pathogenesis of bacterial meningitis protection of blood-CSF barrier by dexamethasone may prevent the penetration of bacteria and leukocytes into the CSF.
- Published
- 2008
37. Expression of mRNA for the proto-oncogene c-fos in rat basophilic leukaemia cells
- Author
-
Volker Stephan, Volker Wahn, Dieter Körholz, and Annette Seibt
- Subjects
G protein ,Cell Degranulation ,Gene Expression ,Biology ,Immunoglobulin E ,Proto-Oncogene Mas ,Proto-Oncogene c-Fos ,Mice ,Tumor Cells, Cultured ,Animals ,Humans ,RNA, Messenger ,Enzyme Inhibitors ,Receptor ,Protein kinase C ,Protein Kinase C ,Messenger RNA ,Receptors, IgE ,Cell Biology ,3T3 Cells ,Protein-Tyrosine Kinases ,Molecular biology ,Genistein ,Isoflavones ,Culture Media ,Rats ,Leukemia, Basophilic, Acute ,biology.protein ,Calcium ,Signal transduction ,Proto-Oncogene Proteins c-fos - Abstract
Recently, the expression of the mRNA for the proto-oncogene c-fos following activation of the high-affinity receptor for immunoglobulin E in rodent mast cells has been reported. In the present study we investigated different biochemical events that may play a role in signal transduction pathways culminating in the expression of c-fos mRNA in rat basophilic leukaemia cells. Similar to IgE-mediated cell degranulation we demonstrated inhibition of the c-fos signal in the absence of calcium and after preincubation of cells with the protein tyrosine kinase inhibitor genistein. Activation of RBL-2H3 cells by short term PMA treatment failed to induce cell degranulation or expression of mRNA for c-fos. Depletion of protein kinase C by PMA pre-treatment resulted in substantial inhibition of the c-fos signal. In contrast to IgE-mediated cell degranulation, expression of mRNA for c-fos was not dependent on continued receptor aggregation. In addition, we demonstrate that c-fos mRNA expression is not restricted to FcϵRI activation but can be induced by a variety of IgE independent mechanisms including calcium influx by ionophore A 23187 and stimulation of G proteins.
- Published
- 1997
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.