31 results on '"Philipp Treskes"'
Search Results
2. Chlorpromazine toxicity is associated with disruption of cell membrane integrity and initiation of a pro-inflammatory response in the HepaRG hepatic cell line
- Author
-
Katie Morgan, Nicole Martucci, Ada Kozlowska, Wesam Gamal, Filip Brzeszczyński, Philipp Treskes, Kay Samuel, Peter Hayes, Lenny Nelson, Pierre Bagnaninchi, Joanna Brzeszczynska, and John Plevris
- Subjects
Chlorpromazine ,HepaRG ,Cell membrane ,Tight junctions ,ECIS ,Pro-inflammatory ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Chlorpromazine (CPZ) is a neuroleptic drug and prototype compound used to study intrahepatic cholestasis. The exact mechanisms of CPZ induced cholestasis remain unclear. Rat hepatocytes, or a sandwich culture of rat and human hepatocytes, have been the most commonly used models for studying CPZ toxicity in vitro. However, to better predict outcomes in pre-clinical trials where cholestasis may be an unwanted consequence, a human in vitro model, based on human HepaRG cells, capable of real-time, non-invasive and label free monitoring, alongside molecular investigations would be beneficial. To address this we used the human hepatic HepaRG cell line, and established concentrations of CPZ ranging from sub-toxic, 25 μM and 50 μM, to toxic 100 μM and compared them with untreated control. To assess the effect of this range of CPZ concentrations we employed electrical cell-substrate impedance sensing (ECIS) to measure viability and cell membrane interactions alongside traditional viability assays, immunocytostaining and qRT-PCR to assess genes of interest within adaptive and inflammatory pathways. Using these methods, we show a concentration dependant response to CPZ involving pro-inflammatory pathway, loss of tight junctions and membrane integrity, and an adaptive response mediated by Cytochrome P450 (CYP) enzyme activation and up-regulation of membrane phospholipid and xenobiotic transporters. In conclusion, structural changes within the membrane caused by sub-toxic and toxic concentrations of CPZ negatively impact the function of the cellular membrane. Damage to efflux transport proteins caused by CPZ induce cholestasis alongside downstream inflammation, which activates compensatory responses for cell survival. Lay summary: Chlorpromazine is a drug used to treat patients with schizophrenia, which has a known association with liver damage. Here we show that it causes inflammation and alters the cell membranes in liver and bile duct cells similar to what is seen within a human population. The initiation of the inflammatory response and changes to cellular structure may provide insight into the damage and disease process and inform medical treatment.
- Published
- 2019
- Full Text
- View/download PDF
3. HepaRG-Progenitor Cell Derived Hepatocytes Cultured in Bioartificial Livers Are Protected from Healthy- and Acute Liver Failure-Plasma Induced Toxicity
- Author
-
Martien van Wenum, Philipp Treskes, Aziza A.A. Adam, Vincent. A van der Mark, Aldo Jongejan, Perry D. Moerland, Thomas M. van Gulik, Ronald P.J. Oude Elferink, Robert A.F.M. Chamuleau, and Ruurdtje Hoekstra
- Subjects
Liver failure ,Bioartifcial liver ,Dedifferentiation ,3D culture ,Plasma toxicity ,HepaRG ,Physiology ,QP1-981 ,Biochemistry ,QD415-436 - Abstract
Background/Aims: For applicability of cell-based therapies aimed at the treatment of liver failure, such as bioartificial livers (BALs) and hepatocyte transplantation, it is essential that the applied hepatocytes tolerate exposure to the patient plasma. However, plasma from both healthy donors and acute liver failure (ALF) patients is detrimental to hepatocytes and hepatic cell lines, such as HepaRG. We aimed to elucidate the underlying mechanisms of plasma-induced toxicity against HepaRG cells in order to ultimately develop methods to reduce this toxicity and render HepaRG-BAL treatment more effective. Methods: Differentiated HepaRG cells cultured in monolayers and laboratory-scale BALs were exposed to culture medium, healthy human plasma, healthy porcine plasma and ALF porcine plasma. Healthy human plasma was fractionated based on size- and polarity, albumin depleted and heat treated to characterize the toxic fraction. The cells were assessed for viability by total protein content and trypan blue staining. Their hepatic differentiation was assessed on transcript level through qRT-PCR and microarray analysis, and on functional level for Cytochrome P450 3A4 activity and ammonia elimination. Mitochondrial damage was assessed by JC-1 staining and mitochondrial gene transcription. Results: Sixteen hours of healthy human plasma exposure did not affect viability, however, hepatic gene-transcript levels decreased dramatically and dose-dependently within four hours of exposure. These changes were associated with early NF-kB signaling and a shift from mitochondrial energy metabolism towards glycolysis. Healthy human plasma-toxicity was associated with the dose-dependent presence of heat-resistant, albumin-bound and (partly) hydrophobic toxic compound(s). HepaRG cells cultured in BALs were partially protected from plasma-toxicity, which was mainly attributable to medium perfusion and/or 3D configuration applied during BAL culturing. The detrimental human plasma effects were reversible in BAL-cultured cells. Porcine ALF-plasma elicited mitotoxicity additional to the basal detrimental effect of porcine healthy plasma, which were only partially reversible. Conclusion: A specific fraction of human plasma reduces hepatic differentiation of HepaRG cultures, in association with early NF-κB activation. In addition, ALF-plasma elicits mitotoxic effects. These findings allow for a targeted approach in preventing plasma-induced cell damage.
- Published
- 2018
- Full Text
- View/download PDF
4. Dynamic Support Culture of Murine Skeletal Muscle-Derived Stem Cells Improves Their Cardiogenic Potential In Vitro
- Author
-
Klaus Neef, Philipp Treskes, Guoxing Xu, Florian Drey, Sureshkumar Perumal Srinivasan, Tomo Saric, Erastus Nembo, Judith Semmler, Filomain Nguemo, Christof Stamm, Douglas B. Cowan, Antje-Christin Deppe, Maximilian Scherner, Thorsten Wittwer, Jürgen Hescheler, Thorsten Wahlers, and Yeong-Hoon Choi
- Subjects
Internal medicine ,RC31-1245 - Abstract
Ischemic heart disease is the main cause of death in western countries and its burden is increasing worldwide. It typically involves irreversible degeneration and loss of myocardial tissue leading to poor prognosis and fatal outcome. Autologous cells with the potential to regenerate damaged heart tissue would be an ideal source for cell therapeutic approaches. Here, we compared different methods of conditional culture for increasing the yield and cardiogenic potential of murine skeletal muscle-derived stem cells. A subpopulation of nonadherent cells was isolated from skeletal muscle by preplating and applying cell culture conditions differing in support of cluster formation. In contrast to static culture conditions, dynamic culture with or without previous hanging drop preculture led to significantly increased cluster diameters and the expression of cardiac specific markers on the protein and mRNA level. Whole-cell patch-clamp studies revealed similarities to pacemaker action potentials and responsiveness to cardiac specific pharmacological stimuli. This data indicates that skeletal muscle-derived stem cells are capable of adopting enhanced cardiac muscle cell-like properties by applying specific culture conditions. Choosing this route for the establishment of a sustainable, autologous source of cells for cardiac therapies holds the potential of being clinically more acceptable than transgenic manipulation of cells.
- Published
- 2015
- Full Text
- View/download PDF
5. Noninvasive in Vivo Tracking of Mesenchymal Stem Cells and Evaluation of Cell Therapeutic Effects in a Murine Model Using a Clinical 3.0 T MRI
- Author
-
Florian Drey, Yeong-Hoon Choi, Klaus Neef, Birgit Ewert, Arne Tenbrock, Philipp Treskes, Henning Bovenschulte, Oliver J. Liakopoulos, Meike Brenkmann, Christof Stamm, Thorsten Wittwer, and Thorsten Wahlers
- Subjects
Medicine - Abstract
Cardiac cell therapy with mesenchymal stem cells (MSCs) represents a promising treatment approach for endstage heart failure. However, little is known about the underlying mechanisms and the fate of the transplanted cells. The objective of the presented work is to determine the feasibility of magnetic resonance imaging (MRI) and in vivo monitoring after transplantation into infarcted mouse hearts using a clinical 3.0 T MRI device. The labeling procedure of bone marrow-derived MSCs with micron-sized paramagnetic iron oxide particles (MPIOs) did not affect the viability of the cells and their cell type-defining properties when compared to unlabeled cells. Using a clinical 3.0 T MRI scanner equipped with a dedicated small animal solenoid coil, 10 5 labeled MSCs could be detected and localized in the mouse hearts for up to 4 weeks after intramyocardial transplantation. Weekly ECG-gated scans using T1-weighted sequences were performed, and left ventricular function was assessed. Histological analysis of hearts confirmed the survival of labeled MSCs in the target area up to 4 weeks after transplantation. In conclusion, in vivo tracking of labeled MSCs using a clinical 3.0 T MRI scanner is feasible. In combination with assessment of heart function, this technology allows the monitoring of the therapeutic efficacy of regenerative therapies in a small animal model.
- Published
- 2013
- Full Text
- View/download PDF
6. Oxygen Plasma Substrate and Specific Nanopattern Promote Early Differentiation of HepaRG Progenitors
- Author
-
Katie Morgan, Steven D. Morley, Leonard J Nelson, Kay Samuel, Peter C. Hayes, John N. Plevris, Joanna Brzeszczynska, Anna Bryans, Filip Brzeszczyński, Nikolaj Gadegaard, and Philipp Treskes
- Subjects
Plasma Gases ,0206 medical engineering ,Biomedical Engineering ,Cell Culture Techniques ,Bioengineering ,02 engineering and technology ,Biochemistry ,Cell Line ,Biomaterials ,03 medical and health sciences ,Tissue culture ,chemistry.chemical_compound ,Humans ,Progenitor cell ,030304 developmental biology ,Progenitor ,0303 health sciences ,Chemistry ,Dimethyl sulfoxide ,Substrate (chemistry) ,Cell Differentiation ,020601 biomedical engineering ,Cell biology ,Oxygen ,Cell culture ,Hepatic stellate cell ,Hepatocytes ,Function (biology) - Abstract
Fully differentiated HepaRG™ cells are the hepatic cell line of choice for in vitro study in toxicology and drug trials. They are derived from a hepatoblast-like progenitor (HepaRG-P) that differentiates into a coculture of hepatocyte-like and cholangiocyte-like cells. This process that requires 2 weeks of proliferation followed by 2 weeks of differentiation using dimethyl sulfoxide (DMSO) can be time consuming and costly. Identifying a method to accelerate HepaRG-Ps toward a mature lineage would save both time and money. The ability to do this in the absence of DMSO would remove the possibility of confounding toxicology results caused by DMSO induction of CYP pathways. It has been shown that tissue culture substrates play an important role in the development and maturity of a cell line, and this is particularly important for progenitor cells, which retain some form of plasticity. Oxygen plasma treatment is used extensively to modify cell culture substrates. There is also evidence that patterned rather than planar surfaces have a positive effect on proliferation and differentiation. In this study, we compared the effect of standard tissue culture plastic (TCP), oxygen plasma coated (OPC), and nanopatterned substrates (NPS) on early differentiation and function of HepaRG-P cells. Since NPS were OPC we initially compared the effect of TCP and OPC to enable comparison between all three culture surfaces using OPC as control to asses if patterning further enhanced early differentiation and functionality. The results show that HepaRG-P's grown on OPC substrate exhibited earlier differentiation, proliferation, and function compared with TCP. Culturing HepaRG-P's on OPC with the addition of NPS did not confer any additional advantage. In conclusion, OPC surface appeared to enhance hepatic differentiation and functionality and could replace traditional methods of differentiating HepaRG-P cells into fully differentiated and functional HepaRGs earlier than standard methods. Impact statement We show significantly earlier differentiation and function of HepaRG progenitor cells when grown in dimethyl sulfoxide-free medium on oxygen plasma substrates versus standard tissue culture plastic. Further investigation showed that nanopatterning of oxygen plasma substrates did not confer any additional advantage over smooth oxygen plasma, although one pattern (DSQ120) showed comparable early differentiation and function.
- Published
- 2020
7. HepaRG-Progenitor Cell Derived Hepatocytes Cultured in Bioartificial Livers Are Protected from Healthy- and Acute Liver Failure-Plasma Induced Toxicity
- Author
-
Ronald P.J. Oude Elferink, Thomas M. van Gulik, Vincent A. van der Mark, Philipp Treskes, Robert A. F. M. Chamuleau, Martien van Wenum, Aziza A. A. Adam, Aldo Jongejan, Ruurdtje Hoekstra, Perry D. Moerland, Graduate School, Gastroenterology and Hepatology, AGEM - Endocrinology, metabolism and nutrition, Epidemiology and Data Science, APH - Methodology, Surgery, AGEM - Re-generation and cancer of the digestive system, Tytgat Institute for Liver and Intestinal Research, and APH - Personalized Medicine
- Subjects
Male ,3D culture ,0301 basic medicine ,Swine ,Physiology ,Cell ,Cell Culture Techniques ,Pharmacology ,lcsh:Physiology ,lcsh:Biochemistry ,Plasma ,03 medical and health sciences ,medicine ,Animals ,Cytochrome P-450 CYP3A ,Humans ,lcsh:QD415-436 ,Glycolysis ,Progenitor cell ,Cell damage ,Membrane Potential, Mitochondrial ,Arginase ,lcsh:QP1-981 ,CYP3A4 ,Chemistry ,Stem Cells ,Liver failure ,Albumin ,Cell Differentiation ,Liver Failure, Acute ,medicine.disease ,Liver, Artificial ,Culture Media ,Up-Regulation ,Plasma toxicity ,030104 developmental biology ,medicine.anatomical_structure ,Bioartifcial liver ,Toxicity ,Hepatocytes ,Hepatic stellate cell ,Female ,Dedifferentiation ,HepaRG - Abstract
Background/Aims: For applicability of cell-based therapies aimed at the treatment of liver failure, such as bioartificial livers (BALs) and hepatocyte transplantation, it is essential that the applied hepatocytes tolerate exposure to the patient plasma. However, plasma from both healthy donors and acute liver failure (ALF) patients is detrimental to hepatocytes and hepatic cell lines, such as HepaRG. We aimed to elucidate the underlying mechanisms of plasma-induced toxicity against HepaRG cells in order to ultimately develop methods to reduce this toxicity and render HepaRG-BAL treatment more effective. Methods: Differentiated HepaRG cells cultured in monolayers and laboratory-scale BALs were exposed to culture medium, healthy human plasma, healthy porcine plasma and ALF porcine plasma. Healthy human plasma was fractionated based on size- and polarity, albumin depleted and heat treated to characterize the toxic fraction. The cells were assessed for viability by total protein content and trypan blue staining. Their hepatic differentiation was assessed on transcript level through qRT-PCR and microarray analysis, and on functional level for Cytochrome P450 3A4 activity and ammonia elimination. Mitochondrial damage was assessed by JC-1 staining and mitochondrial gene transcription. Results: Sixteen hours of healthy human plasma exposure did not affect viability, however, hepatic gene-transcript levels decreased dramatically and dose-dependently within four hours of exposure. These changes were associated with early NF-kB signaling and a shift from mitochondrial energy metabolism towards glycolysis. Healthy human plasma-toxicity was associated with the dose-dependent presence of heat-resistant, albumin-bound and (partly) hydrophobic toxic compound(s). HepaRG cells cultured in BALs were partially protected from plasma-toxicity, which was mainly attributable to medium perfusion and/or 3D configuration applied during BAL culturing. The detrimental human plasma effects were reversible in BAL-cultured cells. Porcine ALF-plasma elicited mitotoxicity additional to the basal detrimental effect of porcine healthy plasma, which were only partially reversible. Conclusion: A specific fraction of human plasma reduces hepatic differentiation of HepaRG cultures, in association with early NF-κB activation. In addition, ALF-plasma elicits mitotoxic effects. These findings allow for a targeted approach in preventing plasma-induced cell damage.
- Published
- 2018
8. Scaling-up of a HepaRG progenitor cell based bioartificial liver: optimization for clinical application and transport
- Author
-
Thomas M. van Gulik, Martien van Wenum, Sandrine Camus, Chung-Yin Tang, Ruurdtje Hoekstra, Koen Jansen, Philipp Treskes, Robert A. F. M. Chamuleau, Esmee J Coppens, Erik J. Hendriks, Graduate School, Other departments, Amsterdam Gastroenterology Endocrinology Metabolism, Surgery, Tytgat Institute for Liver and Intestinal Research, and Gastroenterology and Hepatology
- Subjects
0301 basic medicine ,Time Factors ,Carcinogenesis ,Biomedical Engineering ,Cell Culture Techniques ,Mice, Nude ,Bioengineering ,Transportation ,Mice, SCID ,Biochemistry ,Cryopreservation ,law.invention ,Biomaterials ,03 medical and health sciences ,Tissue engineering ,law ,medicine ,Animals ,Humans ,Progenitor cell ,Cell Proliferation ,Tissue Engineering ,Chemistry ,Cell growth ,Stem Cells ,Bioartificial liver device ,Temperature ,General Medicine ,Liver, Artificial ,Cell biology ,030104 developmental biology ,medicine.anatomical_structure ,Cell culture ,Hepatocyte ,Stem cell ,Biotechnology ,HeLa Cells - Abstract
A new generation of bioartificial livers, based on differentiated proliferative hepatocyte sources, has been developed. Several practicable and regulatory demands have to be addressed before these can be clinically evaluated. We identified three main hurdles: (1) expansion and preservation of the biocomponent, (2) development of scaled-up culture conditions and (3) transport of the device to the bedside. In this study we address these three issues for the HepaRG-progenitor cell line-loaded AMC-Bioartificial Liver. (1) HepaRG cells were expanded in large quantities and then cryopreserved or loaded directly into bioreactors. After 3 weeks of culture, key hepatic functions (ammonia/lactate elimination, apolipoprotein A1 synthesis and cytochrome P450 3A4 activity) did not differ significantly between the two groups. (2) Bioartificial livers were scaled up from 9 ml to 540 ml priming volume, with preservation of normalized hepatic functionality. Quantification of amino acid consumption revealed rapid depletion of several amino acids. (3) Whole-device cryopreservation and cooled preservation induced significant loss of hepatic functionality, whereas simulated transport from culture-facility to the bedside in a clinical-grade transport unit with controlled temperature maintenance, medium perfusion and gas supply did not affect functionality. In addition, we assessed tumorigenicity of HepaRG cells in immune-incompetent mice and found no tumor formation of HepaRG cells (n = 12), while HeLa cells induced formation of carcinomas in eight out of 12 mice in 140 days.
- Published
- 2017
9. Noninvasive in Vivo Tracking of Mesenchymal Stem Cells and Evaluation of Cell Therapeutic Effects in a Murine Model Using a Clinical 3.0 T MRI
- Author
-
H. Bovenschulte, Oliver J. Liakopoulos, Birgit Ewert, Christof Stamm, F. Drey, Thorsten Wittwer, Meike Brenkmann, Klaus Neef, Philipp Treskes, Thorsten Wahlers, Yeong-Hoon Choi, and A. Tenbrock
- Subjects
Cardiac function curve ,Pathology ,medicine.medical_specialty ,Time Factors ,Cellular differentiation ,Cell ,Myocardial Infarction ,Biomedical Engineering ,Contrast Media ,lcsh:Medicine ,Mesenchymal Stem Cell Transplantation ,Ferric Compounds ,Mice ,Osteogenesis ,In vivo ,medicine ,Animals ,Transplantation, Homologous ,Magnetite Nanoparticles ,Transplantation ,medicine.diagnostic_test ,business.industry ,Myocardium ,lcsh:R ,Mesenchymal stem cell ,Cell Differentiation ,Heart ,Mesenchymal Stem Cells ,Magnetic resonance imaging ,Cell Biology ,medicine.disease ,Magnetic Resonance Imaging ,Mice, Inbred C57BL ,Radiography ,Disease Models, Animal ,medicine.anatomical_structure ,Cell Tracking ,Heart failure ,business ,Chondrogenesis - Abstract
Cardiac cell therapy with mesenchymal stem cells (MSCs) represents a promising treatment approach for endstage heart failure. However, little is known about the underlying mechanisms and the fate of the transplanted cells. The objective of the presented work is to determine the feasibility of magnetic resonance imaging (MRI) and in vivo monitoring after transplantation into infarcted mouse hearts using a clinical 3.0 T MRI device. The labeling procedure of bone marrow-derived MSCs with micron-sized paramagnetic iron oxide particles (MPIOs) did not affect the viability of the cells and their cell type-defining properties when compared to unlabeled cells. Using a clinical 3.0 T MRI scanner equipped with a dedicated small animal solenoid coil, 105 labeled MSCs could be detected and localized in the mouse hearts for up to 4 weeks after intramyocardial transplantation. Weekly ECG-gated scans using T1-weighted sequences were performed, and left ventricular function was assessed. Histological analysis of hearts confirmed the survival of labeled MSCs in the target area up to 4 weeks after transplantation. In conclusion, in vivo tracking of labeled MSCs using a clinical 3.0 T MRI scanner is feasible. In combination with assessment of heart function, this technology allows the monitoring of the therapeutic efficacy of regenerative therapies in a small animal model.
- Published
- 2013
10. Acetaminophen cytotoxicity is ameliorated in a human liver organotypic co-culture model
- Author
-
Peter C. Hayes, María Luisa Navarro, Olga Tura-Ceide, Philipp Treskes, Leonard J Nelson, Kay Samuel, Steven D. Morley, and John N. Plevris
- Subjects
Multidisciplinary ,Liver cytology ,Cell Survival ,Cellular polarity ,Pharmacology ,Biology ,medicine.disease_cause ,Article ,Coculture Techniques ,Mitochondria ,Drug development ,Liver ,In vivo ,Toxicity ,Pyruvic Acid ,Toxicity Tests ,medicine ,Human Umbilical Vein Endothelial Cells ,Lactates ,Humans ,Cytotoxicity ,Drug metabolism ,Oxidative stress ,Acetaminophen - Abstract
Organotypic liver culture models for hepatotoxicity studies that mimic in vivo hepatic functionality could help facilitate improved strategies for early safety risk assessment during drug development. Interspecies differences in drug sensitivity and mechanistic profiles, low predictive capacity and limitations of conventional monocultures of human hepatocytes, with high attrition rates remain major challenges. Herein, we show stable, cell-type specific phenotype/cellular polarity with differentiated functionality in human hepatocyte-like C3A cells (enhanced CYP3A4 activity/albumin synthesis) when in co-culture with human vascular endothelial cells (HUVECs), thus demonstrating biocompatibility and relevance for evaluating drug metabolism and toxicity. In agreement with in vivo studies, acetaminophen (APAP) toxicity was most profound in HUVEC mono-cultures; whilst in C3A:HUVEC co-culture, cells were less susceptible to the toxic effects of APAP, including parameters of oxidative stress and ATP depletion, altered redox homeostasis and impaired respiration. This resistance to APAP is also observed in a primary human hepatocyte (PHH) based co-culture model, suggesting bidirectional communication/stabilization between different cell types. This simple and easy-to-implement human co-culture model may represent a sustainable and physiologically-relevant alternative cell system to PHHs, complementary to animal testing, for initial hepatotoxicity screening or mechanistic studies of candidate compounds differentially targeting hepatocytes and endothelial cells.
- Published
- 2015
- Full Text
- View/download PDF
11. Dynamic Support Culture of Murine Skeletal Muscle-derived Stem Cells Improves Their Cardiogenic Potential In Vitro
- Author
-
Douglas B. Cowan, Filomain Nguemo, Judith Semmler, Philipp Treskes, Antje-Christin Deppe, Klaus Neef, Yeong-Hoon Choi, Erastus Nembu Nembo, F. Drey, Christof Stamm, Thorsten Wittwer, Guoxing Xu, Thorsten Wahlers, Sureshkumar Perumal Srinivasan, Jürgen Hescheler, Maximilian Scherner, and Tomo Saric
- Subjects
Pathology ,medicine.medical_specialty ,lcsh:Internal medicine ,Article Subject ,Transgene ,Cell ,Cardiac muscle ,Skeletal muscle ,Cell Biology ,Disease ,Biology ,600 Technik, Medizin, angewandte Wissenschaften::610 Medizin und Gesundheit ,In vitro ,medicine.anatomical_structure ,Cell culture ,medicine ,Cancer research ,Stem cell ,lcsh:RC31-1245 ,Molecular Biology ,Research Article - Abstract
Ischemic heart disease is the main cause of death in western countries and its burden is increasing worldwide. It typically involves irreversible degeneration and loss of myocardial tissue leading to poor prognosis and fatal outcome. Autologous cells with the potential to regenerate damaged heart tissue would be an ideal source for cell therapeutic approaches. Here, we compared different methods of conditional culture for increasing the yield and cardiogenic potential of murine skeletal muscle-derived stem cells. A subpopulation of nonadherent cells was isolated from skeletal muscle by preplating and applying cell culture conditions differing in support of cluster formation. In contrast to static culture conditions, dynamic culture with or without previous hanging drop preculture led to significantly increased cluster diameters and the expression of cardiac specific markers on the protein and mRNA level. Whole-cell patch-clamp studies revealed similarities to pacemaker action potentials and responsiveness to cardiac specific pharmacological stimuli. This data indicates that skeletal muscle-derived stem cells are capable of adopting enhanced cardiac muscle cell-like properties by applying specific culture conditions. Choosing this route for the establishment of a sustainable, autologous source of cells for cardiac therapies holds the potential of being clinically more acceptable than transgenic manipulation of cells.
- Published
- 2015
12. Towards non-invasive 3D hepatotoxicity assays with optical coherence phase microscopy
- Author
-
Andreas Koulovasilopoulos, Pierre Bagnaninchi, John N. Plevris, Peter C. Hayes, Philipp Treskes, and Leonard J Nelson
- Subjects
Programmed cell death ,medicine.diagnostic_test ,Chemistry ,business.industry ,Hepatotoxin ,Tissue culture ,Optics ,Optical coherence tomography ,Cell culture ,Microscopy ,Toxicity ,medicine ,Viability assay ,business ,Biomedical engineering - Abstract
Three-dimensional tissue-engineered models are increasingly recognised as more physiologically-relevant than standard 2D cell culture for pre-clinical drug toxicity testing. However, many types of conventional toxicity assays are incompatible with dense 3D tissues. This study investigated the use of optical coherence phase microscopy (OCPM) as a novel approach to assess cell death in 3D tissue culture. For 3D micro-spheroid formation Human hepatic C3A cells were encapsulated in hyaluronic acid gels and cultured in 100μl MEME/10%FBS in 96-well plates. After spheroid formation the 3D liver constructs were exposed to acetaminophen on culture day 8. Acetaminophen hepatotoxicity in 3D cultures was evaluated using standard biochemical assays. An inverted OCPM in common path configuration was developed with a Callisto OCT engine (Thorlabs), centred at 930nm and a custom scanning head. Intensity data were used to perform in-depth microstructural imaging. In addition, phase fluctuations were measured by collecting several successive B scans at the same location, and statistics on the first time derivative of the phase, i.e. time fluctuations, were analysed over the acquisition time interval to retrieve overall cell viability. OCPM intensity (cell cluster size) and phase fluctuation statistics were directly compared with biochemical assays. In this study, we investigated optical coherence phase tomography to assess cell death in a 3d liver model after exposure to a prototypical hepatotoxin, acetaminophen. We showed that OCPM has the potential to assess noninvasively and label-free drug toxicity in 3D tissue models.
- Published
- 2015
13. Preconditioning of skeletal myoblast-based engineered tissue constructs enables functional coupling to myocardium in vivo
- Author
-
Navid Madershahian, Christof Stamm, Klaus Neef, Thorsten Wahlers, Marcel Halbach, Maximilian Scherner, Douglas B. Cowan, Sureshkumar Perumal Srinivasan, Jürgen Hescheler, Thorsten Wittwer, Yeong-Hoon Choi, and Philipp Treskes
- Subjects
Pulmonary and Respiratory Medicine ,Time Factors ,Cell Survival ,Myoblasts, Skeletal ,Cell Culture Techniques ,Connexin ,Action Potentials ,Cell Communication ,Article ,Cell therapy ,chemistry.chemical_compound ,Tissue engineering ,Y Chromosome ,medicine ,Animals ,Myocytes, Cardiac ,DAPI ,Cells, Cultured ,Excitation Contraction Coupling ,Tissue Engineering ,Myogenesis ,business.industry ,Skeletal muscle ,Anatomy ,Chromosomes, Mammalian ,Myocardial Contraction ,Cell biology ,Transplantation ,Mice, Inbred C57BL ,medicine.anatomical_structure ,chemistry ,Cell culture ,Connexin 43 ,Surgery ,Female ,Stress, Mechanical ,Cardiology and Cardiovascular Medicine ,business ,Biomarkers - Abstract
Skeletal myoblasts fuse to form functional syncytial myotubes as an integral part of the skeletal muscle. During this differentiation process, expression of proteins for mechanical and electrical integration is seized, which is a major drawback for the application of skeletal myoblasts in cardiac regenerative cell therapy, because global heart function depends on intercellular communication.Mechanically preconditioned engineered tissue constructs containing neonatal mouse skeletal myoblasts were transplanted epicardially. A Y-chromosomal specific polymerase chain reaction (PCR) was undertaken up to 10 weeks after transplantation to confirm the presence of grafted cells. Histologic and electrophysiologic analyses were carried out 1 week after transplantation.Cells within the grafted construct expressed connexin 43 at the interface to the host myocardium, indicating electrical coupling, confirmed by sharp electrode recordings. Analyses of the maximum stimulation frequency (5.65 ± 0.37 Hz), conduction velocity (0.087 ± 0.011 m/s) and sensitivity for pharmacologic conduction block (0.736 ± 0.080 mM 1-heptanol) revealed effective electrophysiologic coupling between graft and host cells, although significantly less robust than in native myocardial tissue (maximum stimulation frequency, 11.616 ± 0.238 Hz, P .001; conduction velocity, 0.300 ± 0.057 m/s, P .01; conduction block, 1.983 ± 0.077 mM 1-heptanol, P .001).Although untreated skeletal myoblasts cannot couple to cardiomyocytes, we confirm that mechanical preconditioning enables transplanted skeletal myoblasts to functionally interact with cardiomyocytes in vivo and, thus, reinvigorate the concept of skeletal myoblast-based cardiac cell therapy.
- Published
- 2014
14. Electrophysiologically competent skeletal myoblast based engineered tissue constructs show cardiac-like force generation
- Author
-
Christof Stamm, Thorsten Wahlers, Philipp Treskes, Antje-Christin Deppe, Klaus Neef, Sureshkumar Perumal Srinivasan, and Yeong-Hoon Choi
- Subjects
Pulmonary and Respiratory Medicine ,Force generation ,business.industry ,Skeletal Myoblasts ,Medicine ,Surgery ,Anatomy ,Cardiology and Cardiovascular Medicine ,business ,Engineered tissue ,Cell biology - Published
- 2014
15. P1085 : Mechanistic insight into acetaminophen-induced hepatic tight junction disruption using a human HepaRG-based liverbiochip impedance biosensor
- Author
-
Katie Morgan, Philipp Treskes, V. Shevchenko, C. Chesne, Pierre Bagnaninchi, Wesam Gamal, Lenny Nelson, and John N. Plevris
- Subjects
Impedance biosensor ,Hepatology ,Biochemistry ,Tight junction ,Chemistry ,Biophysics ,medicine ,Acetaminophen ,medicine.drug - Published
- 2015
16. Profiling the impact of medium formulation on morphology and functionality of primary hepatocytes in vitro
- Author
-
Leonard J Nelson, Philipp Treskes, John N. Plevris, Peter C. Hayes, A. Forbes Howie, and S W Walker
- Subjects
Organelles ,Multidisciplinary ,Cell Survival ,Swine ,Cell Culture Techniques ,Galactose ,Reproducibility of Results ,Biology ,In vitro ,Article ,Cell biology ,Culture Media ,Biochemistry ,Cytochrome P-450 Enzyme System ,Cell culture ,Hepatocytes ,Animals ,Humans ,Urea ,Cell survival ,Cells, Cultured ,Serum Albumin - Abstract
The characterization of fully-defined in vitro hepatic culture systems requires testing of functional and morphological variables to obtain the optimal trophic support, particularly for cell therapeutics including bioartificial liver systems (BALs). Using serum-free fully-defined culture medium formulations, we measured synthetic, detoxification and metabolic variables of primary porcine hepatocytes (PPHs) - integrated these datasets using a defined scoring system and correlated this hepatocyte biological activity index (HBAI) with morphological parameters. Hepatic-specific functions exceeded those of both primary human hepatocytes (PHHs) and HepaRG cells, whilst retaining biotransformation potential and in vivo-like ultrastructural morphology, suggesting PPHs as a potential surrogate for PHHs in various biotech applications. The HBAI permits assessment of global functional capacity allowing the rational choice of optimal trophic support for a defined operational task (including BALs, hepatocellular transplantation, and cytochrome P450 (CYP450) drug metabolism studies), mitigates risk associated with sub-optimal culture systems, and reduces time and cost of research and therapeutic applications.
- Published
- 2013
17. ELEctrophysiological coupling OF skeletal myoblast based engineered tissue constructs IN VIVO
- Author
-
S Kumar, Klaus Neef, Thorsten Wittwer, Philipp Treskes, OJ Liakopoulos, Yeong-Hoon Choi, Navid Madershahian, Markus Khalil, and Thorsten Wahlers
- Subjects
Pulmonary and Respiratory Medicine ,Pathology ,medicine.medical_specialty ,Skeletal Myoblasts ,business.industry ,Coupling (electronics) ,Electrophysiology ,In vivo ,Biophysics ,Medicine ,Surgery ,Cardiology and Cardiovascular Medicine ,business ,Engineered tissue - Published
- 2013
18. Establishing the Edinburgh Model: Dose to Injury Response in a Large Animal Infusion Model of Paracetamol-Induced Acute Liver Failure
- Author
-
K. Borland, B. Few, Peter C. Hayes, Guen Bradbury, B. Vliegenthart, A. Hay, John N. Plevris, Michael Eddleston, E. Clutton, M. Van Wenum, and Philipp Treskes
- Subjects
Hepatology ,business.industry ,Anesthesia ,Liver failure ,Medicine ,business ,Injury response ,Large animal - Published
- 2016
19. Effects of Healthy- and Acute Liver Failure Plasma on Differentiated Human Heparg Progenitor Cells in Monolayers and Bioartificial Livers
- Author
-
Ruurdtje Hoekstra, Perry D. Moerland, M. Van Wenum, S. Meisner, Aldo Jongejan, Philipp Treskes, R.A.F.M. Chamuleau, T.M. van Gulik, and Erik J. Hendriks
- Subjects
0301 basic medicine ,03 medical and health sciences ,Pathology ,medicine.medical_specialty ,030104 developmental biology ,Hepatology ,law ,Chemistry ,Liver failure ,Bioartificial liver device ,medicine ,Progenitor cell ,law.invention - Published
- 2016
20. Enhanced gap junction expression in myoblast-containing engineered tissue
- Author
-
Klaus Neef, Douglas B. Cowan, Yeong-Hoon Choi, Elmar W. Kuhn, Christof Stamm, Philipp Treskes, Thorsten Wahlers, Thorsten Wittwer, Sureshkumar Perumal Srinivasan, Ingo Slottosch, Navid Madershahian, and Oliver J. Liakopoulos
- Subjects
Tissue Engineering ,Myogenesis ,Chemistry ,Myoblasts, Skeletal ,Biophysics ,Gap junction ,Connexin ,Gap Junctions ,Cell Biology ,Anatomy ,Cell Separation ,Biochemistry ,Article ,Cell biology ,Transplantation ,Mice, Inbred C57BL ,Weight-Bearing ,Mice ,Tissue engineering ,Animals, Newborn ,Myocyte ,Animals ,Desmin ,Mechanotransduction ,Molecular Biology - Abstract
Transplantation of skeletal myoblasts (SMs) has been investigated as a potential cardiac cell therapy approach. SM are available autologously, predetermined for muscular differentiation and resistant to ischemia. Major hurdles for their clinical application are limitations in purity and yield during cell isolation as well as the absence of gap junction expression after differentiation into myotubes. Furthermore, transplanted SMs do not functionally or electrically integrate with the host myocardium. Here, we describe an efficient method for isolating homogeneous SM populations from neonatal mice and demonstrate persistent gap junction expression in an engineered tissue. This method resulted in a yield of 1.4 × 108 high-purity SMs (>99% desmin positive) after 10 days in culture from 162.12 ± 11.85 mg muscle tissue. Serum starvation conditions efficiently induced differentiation into spontaneously contracting myotubes that coincided with loss of gap junction expression. For mechanical conditioning, cells were integrated into engineered tissue constructs. SMs within tissue constructs exhibited long term survival, ordered alignment, and a preserved ability to differentiate into contractile myotubes. When the tissue constructs were subjected to passive longitudinal tensile stress, the expression of gap junction and cell adherence proteins was maintained or increased throughout differentiation. Our studies demonstrate that mechanical loading of SMs may provide for improved electromechanical integration within the myocardium, which could lead to more therapeutic opportunities.
- Published
- 2012
21. Enrichment of cardiogenic cell populations from murine skeletal muscle
- Author
-
Yh Choi, Christof Stamm, Klaus Neef, S Kumar, Thorsten Wittwer, Thorsten Wahlers, Oj Liakopoulos, Pb Rahmanian, and Philipp Treskes
- Subjects
Pulmonary and Respiratory Medicine ,Pathology ,medicine.medical_specialty ,medicine.anatomical_structure ,business.industry ,Cell ,Medicine ,Skeletal muscle ,Surgery ,Cardiology and Cardiovascular Medicine ,business - Published
- 2012
22. Mechanical preconditioning enables electrophysiological coupling of skeletal myoblast cells to myocardium
- Author
-
Christof Stamm, Yeong-Hoon Choi, Thorsten Wahlers, Philipp Treskes, Markus Khalil, Roland Adelmann, Klaus Neef, S Kumar, and Thorsten Wittwer
- Subjects
Pulmonary and Respiratory Medicine ,Coupling (electronics) ,Electrophysiology ,Skeletal Myoblasts ,business.industry ,Biophysics ,Medicine ,Surgery ,Cardiology and Cardiovascular Medicine ,business - Published
- 2012
23. Cardiogenic progenitor cells derived from skeletal muscle - A promising autologous cell source for cardiac cell therapy
- Author
-
S Kumar, OJ Liakopoulos, Klaus Neef, Yeong-Hoon Choi, Christof Stamm, Philipp Treskes, Thorsten Wittwer, and Thorsten Wahlers
- Subjects
Pulmonary and Respiratory Medicine ,Autologous cell ,medicine.anatomical_structure ,business.industry ,Cancer research ,Medicine ,Skeletal muscle ,Surgery ,Progenitor cell ,Cardiology and Cardiovascular Medicine ,business ,Cardiac cell - Published
- 2011
24. P333 HUMAN HEPATIC HepaRG CO-CULTURE MODEL AS A SENSITIVE AND NON-INVASIVE TOXICOLOGICAL PLATFORM USING ECIS (ELECTRICAL CELL-SUBSTRATE IMPEDANCE SENSING) BIOSENSORS
- Author
-
John N. Plevris, Pierre Bagnaninchi, Philipp Treskes, Lenny Nelson, C. Chesne, and Wesam Gamal
- Subjects
Hepatology ,Chemistry ,business.industry ,Cell substrate ,Non invasive ,Impedance sensing ,Nanotechnology ,business ,Biosensor ,Biotechnology - Published
- 2014
25. P331 HUMAN HEPATIC HepaRG CELLS MAINTAIN HIGH INTRINSIC CYP450 ACTIVITY/METABOLISM AND SIGNIFICANTLY OUTPERFORM STANDARD HepG2/C3A CELLS USED IN DRUG PHARMACOLOGY APPLICATIONS
- Author
-
John N. Plevris, Natalie Z.M. Homer, Lenny Nelson, Katie Morgan, Philipp Treskes, C. Lebled, Mary Grant, and Catherine Henderson
- Subjects
Hepatology ,CYP3A4 ,Cell ,CYP1A2 ,Pharmacology ,Biology ,In vitro ,medicine.anatomical_structure ,Phenacetin ,Cell culture ,medicine ,Hepatic stellate cell ,Drug metabolism ,medicine.drug - Abstract
Introduction: Conventional in vitro human hepatic models for drug testing are based on the use of cell lines or primary human hepatocytes (PHHs). However, limited availability, inter-donor functional variability and early phenotypic alterations of PHHs in vitro restrict their use; whilst cell lines such as HepG2/C3As lack a substantial and variable set of liver-specific functions, specifically, CYP450 activity. In this study we compared CYP450 activity/ metabolism between HepG2/C3A and human HepaRG cells as hepatic models for pre-clinical drug testing. Methods: Human hepatic cell lines [HepG2/C3A or HepaRG] were grown to >80% confluence on collagen-I-coated plates and treated (in triplicates) 24 h with prototypical inducers rifampicin (CYP3A4) and omeprazole (CYP1A2), [n=3]. 50μM testosterone or phenacetin were added and supernatant and cell samples taken after 2 hours of incubation at 37°C. CYP1A2/3A4 activity [P450-Glo™-Luminometry; Promega] was determined (Figure 1). Relative turnover of testosterone [HPLC] and phenacetin [LC-MS/MS] metabolites was also measured. Cell phenotype was assessed by light-microscopy, histology (PAS-Glycogen), CYP3A4, F-actin/phalloidin, and JC-1 fluorescent-staining. Results: Figure 1 shows HepaRG CYP1A2/3A4 activity was 40-80x fold >> HepG2/C3A cells [P
- Published
- 2014
26. O038 : Investigating paracetamol toxicity in HepaRG-based 3D human hepatic organotypic models with non-invasive optical coherence phase microscopy (OCPM)
- Author
-
Pierre Bagnaninchi, Katie Morgan, Lenny Nelson, C. Chesne, Wesam Gamal, Philipp Treskes, and John N. Plevris
- Subjects
Materials science ,Hepatology ,law ,Phase contrast microscopy ,Non invasive ,Toxicity ,Coherence (statistics) ,Pharmacology ,Biomedical engineering ,law.invention - Published
- 2015
27. P332 DEVELOPMENT OF HUMAN HEPATIC HepaRG CELL-BASED LiverBioChip PROTOTYPES USING ELECTRON BEAM LITHOGRAPHY DIRECTED-NANOPATTERNING FOR HTS-COMPATIBLE PRE-CLINICAL DRUG EVALUATION APPLICATIONS
- Author
-
Nikolaj Gadegaard, Lenny Nelson, María Luisa Navarro, Philipp Treskes, C. Lebled, John N. Plevris, and M. Thompson
- Subjects
Materials science ,Hepatology ,Heparg cell ,Nanotechnology ,Electron-beam lithography - Published
- 2014
28. P337 HEPATOPROTECTIVE EFFECT OF HUVEC CELLS IN AN IN VITRO HEPATIC CO-CULTURE MODEL OF ACETAMINOPHEN TOXICITY
- Author
-
Kay Samuel, Philipp Treskes, P. C. Hayes, J. N. Plevris, Lenny Nelson, and María Luisa Navarro
- Subjects
Hepatology ,Chemistry ,HUVEC Cells ,Pharmacology ,ACETAMINOPHEN TOXICITY ,In vitro - Published
- 2014
29. P336 HEPATOCYTES IMPROVE THEIR DRUG METABOLIC ACTIVITY WITH THE PRESENCE OF ENDOTHELIAL CELLS IN AN IN VITRO HEPATIC CO-CULTURE MODEL OF ACETAMINOPHEN TOXICITY
- Author
-
Lenny Nelson, Philipp Treskes, J. N. Plevris, María Luisa Navarro, P. C. Hayes, and Kay Samuel
- Subjects
Drug ,Hepatology ,Chemistry ,media_common.quotation_subject ,Pharmacology ,Metabolic activity ,ACETAMINOPHEN TOXICITY ,In vitro ,media_common - Published
- 2014
30. Erratum to: Fibrinogen production is enhanced in an in-vitro model of non-alcoholic fatty liver disease: an isolated risk factor for cardiovascular events?
- Author
-
Emily N. W. Yeung, Philipp Treskes, Sarah F. Martin, Jonathan R. Manning, Donald R. Dunbar, Sophie M. Rogers, Thierry Le Bihan, K. Ann Lockman, Steven D. Morley, Peter C. Hayes, Leonard J. Nelson, and John N. Plevris
- Subjects
Proteomics ,Biochemistry, medical ,Endocrinology, Diabetes and Metabolism ,Biochemistry (medical) ,Clinical Biochemistry ,Fibrinogen ,030209 endocrinology & metabolism ,030204 cardiovascular system & hematology ,Models, Biological ,Up-Regulation ,03 medical and health sciences ,Farnesyl-Diphosphate Farnesyltransferase ,0302 clinical medicine ,Endocrinology ,Cardiovascular Diseases ,Non-alcoholic Fatty Liver Disease ,Risk Factors ,Cell Line, Tumor ,Humans ,Erratum ,Genetic Association Studies ,Oligonucleotide Array Sequence Analysis ,Signal Transduction - Abstract
Cardiovascular disease (CVD) remains the major cause of excess mortality in patients with non-alcoholic fatty liver disease (NAFLD). The aim of this study was to investigate the individual contribution of NAFLD to CVD risk factors in the absence of pathogenic influences from other comorbidities often found in NAFLD patients, by using an established in-vitro model of hepatic steatosis.Histopathological events in non-alcoholic fatty liver disease were recapitulated by focused metabolic nutrient overload of hepatoblastoma C3A cells, using oleate-treated-cells and untreated controls for comparison. Microarray and proteomic data from cell culture experiments were integrated into a custom-built systems biology database and proteogenomics analysis performed. Candidate genes with significant dysregulation and concomitant changes in protein abundance were identified and STRING association and enrichment analysis performed to identify putative pathogenic pathways.The search strategy yielded 3 candidate genes that were specifically and significantly up-regulated in nutrient-overloaded cells compared to untreated controls: fibrinogen alpha chain (2.2 fold), fibrinogen beta chain (2.3 fold) and fibrinogen gamma chain (2.1 fold) (all rank products pfp0.05). Fibrinogen alpha and gamma chain also demonstrated significant concomitant increases in protein abundance (3.8-fold and 2.0-fold, respectively, p0.05).In-vitro modelling of NAFLD and reactive oxygen species formation in nutrient overloaded C3A cells, in the absence of pathogenic influences from other comorbidities, suggests that NAFLD is an isolated determinant of CVD. Nutrient overload-induced up-regulation of all three fibrinogen component subunits of the coagulation cascade provides a possible mechanism to explain the excess CVD mortality observed in NAFLD patients.
- Full Text
- View/download PDF
31. Scaling-up of a HepaRG progenitor cell based bioartificial liver: optimization for clinical application and transport.
- Author
-
Martien van Wenum, Philipp Treskes, Chung-Yin Tang, Esmee J Coppens, Koen Jansen, Erik J Hendriks, Sandrine Camus, Thomas M van Gulik, Robert A F M Chamuleau, and Ruurdtje Hoekstra
- Published
- 2017
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.