Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
Add more filters

Publication year range
1.
Chem Res Toxicol ; 33(3): 834-848, 2020 03 16.
Article in English | MEDLINE | ID: mdl-32041405

ABSTRACT

The ongoing developments in chemical risk assessment have led to new concepts building on integration of sophisticated nonanimal models for hazard characterization. Here we explore a pragmatic approach for implementing such concepts, using a case study of three triazole fungicides, namely, flusilazole, propiconazole, and cyproconazole. The strategy applied starts with evaluating the overall level of concern by comparing exposure estimates to toxicological potential, followed by a combination of in silico tools and literature-derived high-throughput screening assays and computational elaborations to obtain insight into potential toxicological mechanisms and targets in the organism. Additionally, some targeted in vitro tests were evaluated for their utility to confirm suspected mechanisms of toxicity and to generate points of departure. Toxicological mechanisms instead of the current "end point-by-end point" approach should guide the selection of methods and assays that constitute a toolbox for next-generation risk assessment. Comparison of the obtained in silico and in vitro results with data from traditional in vivo testing revealed that, overall, nonanimal methods for hazard identification can produce adequate qualitative hazard information for risk assessment. Follow-up studies are needed to further refine the proposed approach, including the composition of the toolbox, toxicokinetics models, and models for exposure assessment.


Subject(s)
Fungicides, Industrial/toxicity , High-Throughput Screening Assays , Silanes/toxicity , Toxicity Tests , Triazoles/toxicity , Humans , Molecular Structure , Risk Assessment
2.
Arch Toxicol ; 94(8): 2749-2767, 2020 08.
Article in English | MEDLINE | ID: mdl-32533217

ABSTRACT

Accurate prediction of drug- and chemical-induced hepatotoxicity remains to be a problem for pharmaceutical companies as well as other industries and regulators. The goal of the current study was to develop an in vitro/in silico method for the rapid and accurate prediction of drug- and chemical-induced hepatocyte injury in humans. HepaRG cells were employed for high-throughput imaging in combination with phenotypic profiling. A reference set of 69 drugs and chemicals was screened at a range of 7 concentrations, and the cellular response values were used for training a supervised classifier and for determining assay performance by using tenfold cross-validation. The results showed that the best performing phenotypic features were related to nuclear translocation of RELA (RELA proto-oncogene, NF-kB subunit; also known as NF-kappa B p65), DNA organization, and the F-actin cytoskeleton. Using a subset of 30 phenotypic features, direct hepatocyte toxicity in humans could be predicted with a test sensitivity, specificity and balanced accuracy of 73%, 92%, and 83%, respectively. The method was applied to another set of 26 drugs and chemicals with unclear annotation and their hepatocyte toxicity in humans was predicted. The results also revealed that the identified discriminative phenotypic changes were related to cell death and cellular senescence. Whereas cell death-related endpoints are widely applied in in vitro toxicology, cellular senescence-related endpoints are not, although cellular senescence can be induced by various drugs and other small molecule compounds and plays an important role in liver injury and disease. These findings show how phenotypic profiling can reveal unexpected chemical-induced mechanisms in toxicology.


Subject(s)
Chemical and Drug Induced Liver Injury/etiology , Hepatocytes/drug effects , High-Throughput Screening Assays , Machine Learning , Microscopy, Fluorescence , Toxicity Tests , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/metabolism , Actin Cytoskeleton/pathology , Cell Death/drug effects , Cellular Senescence/drug effects , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , DNA Damage , Dose-Response Relationship, Drug , Hep G2 Cells , Hepatocytes/metabolism , Hepatocytes/pathology , Humans , Image Processing, Computer-Assisted , Phenotype , Primary Cell Culture , Proto-Oncogene Mas , Risk Assessment , Transcription Factor RelA/metabolism
3.
Part Fibre Toxicol ; 16(1): 26, 2019 Jun 27.
Article in English | MEDLINE | ID: mdl-31248442

ABSTRACT

After the publication of this article [1] it was hihglighted that the number of deaths related to natural disasters was incorrectly reported in the second paragraph of the Hazards from Natural particulates and the evolution of the biosphere section. This correction article shows the correct and incorrect statement. This correction does not change the idea presented in the article that from an evolutionary view point, natural disasters account only for a small fraction of the people on the planet. The original article has been updated.

4.
Part Fibre Toxicol ; 16(1): 19, 2019 04 23.
Article in English | MEDLINE | ID: mdl-31014371

ABSTRACT

BACKGROUND: Particles and fibres affect human health as a function of their properties such as chemical composition, size and shape but also depending on complex interactions in an organism that occur at various levels between particle uptake and target organ responses. While particulate pollution is one of the leading contributors to the global burden of disease, particles are also increasingly used for medical purposes. Over the past decades we have gained considerable experience in how particle properties and particle-bio interactions are linked to human health. This insight is useful for improved risk management in the case of unwanted health effects but also for developing novel medical therapies. The concepts that help us better understand particles' and fibres' risks include the fate of particles in the body; exposure, dosimetry and dose-metrics and the 5 Bs: bioavailability, biopersistence, bioprocessing, biomodification and bioclearance of (nano)particles. This includes the role of the biomolecule corona, immunity and systemic responses, non-specific effects in the lungs and other body parts, particle effects and the developing body, and the link from the natural environment to human health. The importance of these different concepts for the human health risk depends not only on the properties of the particles and fibres, but is also strongly influenced by production, use and disposal scenarios. CONCLUSIONS: Lessons learned from the past can prove helpful for the future of the field, notably for understanding novel particles and fibres and for defining appropriate risk management and governance approaches.


Subject(s)
Air Pollutants/toxicity , Inhalation Exposure/adverse effects , Mineral Fibers/toxicity , Nanoparticles/toxicity , Particulate Matter/toxicity , Air Pollutants/chemistry , Humans , Nanoparticles/chemistry , Particle Size , Particulate Matter/chemistry , Risk Assessment , Risk Management , Surface Properties
5.
Arch Toxicol ; 93(1): 221-223, 2019 01.
Article in English | MEDLINE | ID: mdl-30328497

ABSTRACT

Rapid progress is made in the development of high-content screening assays for the prediction of nephrotoxicity. The findings from different laboratories are consistent with respect to endpoints and concentration ranges screened. Discrepancies regarding compound annotation and the predictive performance analysis are discussed.


Subject(s)
Drug Discovery , Biological Assay , Cell Survival
6.
Altern Lab Anim ; 45(5): 241-252, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29112452

ABSTRACT

The Lush Science Prize 2016 was awarded to Daniele Zink and Lit-Hsin Loo for the interdisciplinary and collaborative work between their research groups in developing alternative methods for the prediction of nephrotoxicity in humans. The collaboration has led to the establishment of a series of pioneering alternative methods for nephrotoxicity prediction, which includes: predictive gene expression markers based on pro-inflammatory responses; predictive in vitro cellular models based on pluripotent stem cell-derived proximal tubular-like cells; and predictive cellular phenotypic markers based on chromatin and cytoskeletal changes. A high-throughput method was established for chemical testing, which is currently being used to predict the potential human nephrotoxicity of ToxCast compounds in collaboration with the US Environmental Protection Agency. Similar high-throughput imaging-based methodologies are currently being developed and adapted by the Zink and Loo groups, to include other human organs and cell types. The ultimate goal is to develop a portfolio of methods accepted for the accurate prediction of human organ-specific toxicity and the consequent replacement of animal experiments.


Subject(s)
Kidney/drug effects , High-Throughput Screening Assays , Humans , In Vitro Techniques , Intersectoral Collaboration , Kidney Tubules, Proximal/drug effects , Phenotype
7.
Arch Toxicol ; 90(11): 2793-2808, 2016 Nov.
Article in English | MEDLINE | ID: mdl-26612367

ABSTRACT

The kidney is a major target for xenobiotics, which include drugs, industrial chemicals, environmental toxicants and other compounds. Accurate methods for screening large numbers of potentially nephrotoxic xenobiotics with diverse chemical structures are currently not available. Here, we describe an approach for nephrotoxicity prediction that combines high-throughput imaging of cultured human renal proximal tubular cells (PTCs), quantitative phenotypic profiling, and machine learning methods. We automatically quantified 129 image-based phenotypic features, and identified chromatin and cytoskeletal features that can predict the human in vivo PTC toxicity of 44 reference compounds with ~82 % (primary PTCs) or 89 % (immortalized PTCs) test balanced accuracies. Surprisingly, our results also revealed that a DNA damage response is commonly induced by different PTC toxicants that have diverse chemical structures and injury mechanisms. Together, our results show that human nephrotoxicity can be predicted with high efficiency and accuracy by combining cell-based and computational methods that are suitable for automation.


Subject(s)
Chromatin Assembly and Disassembly/drug effects , Cytoskeleton/drug effects , Kidney Tubules, Proximal/drug effects , Models, Molecular , Mutagens/toxicity , Xenobiotics/toxicity , Automation, Laboratory , Cell Death/drug effects , Cell Line, Transformed , Cells, Cultured , Computational Biology , DNA Damage , Drug Evaluation, Preclinical , Feasibility Studies , High-Throughput Screening Assays , Humans , Kidney Tubules, Proximal/cytology , Machine Learning , Molecular Structure , Mutagens/chemistry , Osmolar Concentration , Small Molecule Libraries , Xenobiotics/chemistry
10.
BMC Bioinformatics ; 15 Suppl 16: S16, 2014.
Article in English | MEDLINE | ID: mdl-25521947

ABSTRACT

BACKGROUND: Drug-induced nephrotoxicity causes acute kidney injury and chronic kidney diseases, and is a major reason for late-stage failures in the clinical trials of new drugs. Therefore, early, pre-clinical prediction of nephrotoxicity could help to prioritize drug candidates for further evaluations, and increase the success rates of clinical trials. Recently, an in vitro model for predicting renal-proximal-tubular-cell (PTC) toxicity based on the expression levels of two inflammatory markers, interleukin (IL)-6 and -8, has been described. However, this and other existing models usually use linear and manually determined thresholds to predict nephrotoxicity. Automated machine learning algorithms may improve these models, and produce more accurate and unbiased predictions. RESULTS: Here, we report a systematic comparison of the performances of four supervised classifiers, namely random forest, support vector machine, k-nearest-neighbor and naive Bayes classifiers, in predicting PTC toxicity based on IL-6 and -8 expression levels. Using a dataset of human primary PTCs treated with 41 well-characterized compounds that are toxic or not toxic to PTC, we found that random forest classifiers have the highest cross-validated classification performance (mean balanced accuracy = 87.8%, sensitivity = 89.4%, and specificity = 85.9%). Furthermore, we also found that IL-8 is more predictive than IL-6, but a combination of both markers gives higher classification accuracy. Finally, we also show that random forest classifiers trained automatically on the whole dataset have higher mean balanced accuracy than a previous threshold-based classifier constructed for the same dataset (99.3% vs. 80.7%). CONCLUSIONS: Our results suggest that a random forest classifier can be used to automatically predict drug-induced PTC toxicity based on the expression levels of IL-6 and -8.


Subject(s)
Algorithms , Interleukin-6/metabolism , Interleukin-8/metabolism , Kidney Diseases/chemically induced , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Models, Theoretical , Pharmaceutical Preparations/metabolism , Artificial Intelligence , Bayes Theorem , Humans , ROC Curve , Support Vector Machine
11.
Mol Pharm ; 11(7): 1933-48, 2014 Jul 07.
Article in English | MEDLINE | ID: mdl-24502545

ABSTRACT

The kidney is a major target for drug-induced toxicity. Drug-induced nephrotoxicity remains a major problem in the clinical setting, where the use of nephrotoxic drugs is often unavoidable. This leads frequently to acute kidney injury, and current problems are discussed. One strategy to avoid such problems would be the development of drugs with decreased nephrotoxic potential. However, the prediction of nephrotoxicity during preclinical drug development is difficult and nephrotoxicity is typically detected only late. Also, the nephrotoxic potential of newly approved drugs is often underestimated. Regulatory approved or validated in vitro models for the prediction of nephrotoxicity are currently not available. Here, we will review current approaches on the development of such models. This includes a discussion of three-dimensional and microfluidic models and recently developed stem cell based approaches. Most in vitro models have been tested with a limited number of compounds and are of unclear predictivity. However, some studies have tested larger numbers of compounds and the predictivity of the respective in vitro model had been determined. The results showed that high predictivity can be obtained by using primary or stem cell derived human renal cells in combination with appropriate end points.


Subject(s)
Acute Kidney Injury/chemically induced , Drug-Related Side Effects and Adverse Reactions/prevention & control , Kidney/drug effects , Animals , Drug Approval/methods , Drug Evaluation, Preclinical/methods , Humans , In Vitro Techniques/methods
12.
Mol Pharm ; 11(7): 1982-90, 2014 Jul 07.
Article in English | MEDLINE | ID: mdl-24495215

ABSTRACT

The kidney is a major target for drug-induced toxicity, and the renal proximal tubule is frequently affected. Nephrotoxicity is typically detected only late during drug development, and the nephrotoxic potential of newly approved drugs is often underestimated. A central problem is the lack of preclinical models with high predictivity. Validated in vitro models for the prediction of nephrotoxicity are not available. Major problems are related to the identification of appropriate cell models and end points. As drug-induced kidney injury is associated with inflammatory reactions, we explored the expression of inflammatory markers as end point for renal in vitro models. In parallel, we developed a new cell model. Here, we combined these approaches and developed an in vitro model with embryonic stem-cell-derived human renal proximal tubular-like cells that uses the expression of interleukin (IL)-6 and IL-8 as end points. The predictivity of the model was evaluated with 41 well-characterized compounds. The results revealed that the model predicts proximal tubular toxicity in humans with high accuracy. In contrast, the predictivity was low when well-established standard in vitro assays were used. Together, the results show that high predictivity can be obtained with in vitro models employing pluripotent stem cell-derived human renal proximal tubular-like cells.


Subject(s)
Acute Kidney Injury/chemically induced , Drug-Related Side Effects and Adverse Reactions/metabolism , Embryonic Stem Cells/drug effects , Kidney Tubules, Proximal/drug effects , Kidney/drug effects , Pharmaceutical Preparations/administration & dosage , Acute Kidney Injury/metabolism , Biomarkers/metabolism , Cell Line , Embryonic Stem Cells/metabolism , Humans , Inflammation/chemically induced , Inflammation/metabolism , Interleukin-6/metabolism , Interleukin-8/metabolism , Kidney Tubules, Proximal/metabolism
13.
Biomacromolecules ; 15(6): 2067-78, 2014 Jun 09.
Article in English | MEDLINE | ID: mdl-24762265

ABSTRACT

Clinical and industrial applications of human pluripotent stem cells (hPSC) require large amounts of cells that have been expanded under defined conditions. Labor-intensive techniques and ill-defined or expensive compounds and substrates are not applicable. Here we describe a chemically defined synthetic substrate consisting of polysulfone (PSF) membranes coated with polymerized 3,4-dihydroxy-l-phenylalanine (DOPA). DOPA/PSF is inexpensive and can be easily produced at various shapes and sizes. DOPA/PSF supports long-term self-renewal of undifferentiated human embryonic (hESC) and human induced pluripotent stem cells (hiPSC) under defined conditions. Pluripotency is maintained for at least 10 passages. Adhesion of hPSC to DOPA/PSF is mainly mediated by a specific integrin heterodimer. Proliferation and gene expression patterns on DOPA/PSF and control substrates are comparable. Labor-intensive cultivation methods and use of serum or coating with proteins are not required. Together, these features make DOPA/PSF attractive for applications where large-scale expansion of human pluripotent stem cells under defined conditions is essential.


Subject(s)
Cell Culture Techniques/methods , Cost-Benefit Analysis , Dihydroxyphenylalanine/chemistry , Induced Pluripotent Stem Cells/drug effects , Polymers/chemistry , Sulfones/chemistry , Cell Culture Techniques/economics , Cell Proliferation/drug effects , Cell Proliferation/physiology , Cost-Benefit Analysis/methods , Dihydroxyphenylalanine/economics , Dihydroxyphenylalanine/pharmacology , Humans , Induced Pluripotent Stem Cells/metabolism , Polymers/economics , Polymers/pharmacology , Substrate Specificity/drug effects , Substrate Specificity/physiology , Sulfones/economics
14.
J Cell Mol Med ; 17(4): 497-507, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23480720

ABSTRACT

Treatment with bioartificial kidneys had beneficial effects in animal experiments and improved survival of critically ill patients with acute kidney injury in a Phase II clinical trial. However, a Phase II b clinical trial failed. This and other results suggested various problems with the current design of bioartificial kidneys. We propose a novel design to improve various properties of device, including haemocompatibility and cell performance. An important feature of the novel design is confinement of the blood to the lumina of the hollow fibre membranes. This avoids exposure of the blood to the non-haemocompatible outer surfaces of hollow fibre membranes, which usually occurs in bioartificial kidneys. We use these outer surfaces as substrate for cell growth. Our results show that commercial hollow fibre membranes can be directly applied in the bioreactor when human primary renal proximal tubular cells are grown in this configuration, and no coatings are required for the formation of robust and functional renal epithelia. Furthermore, we demonstrate that the bioreactor unit produces significant amounts of interleukins. This result helps to understand the immunomodulatory effects of bioartificial kidneys, which have been observed previously. The novel bioartificial kidney design outlined here and the results obtained would be expected to improve the safety and performance of bioartificial kidneys and to contribute to a better understanding of their effects.


Subject(s)
Kidney Tubules, Proximal/cytology , Kidneys, Artificial , Animals , Bioreactors , Creatinine/metabolism , Epithelium/metabolism , Gene Expression , Hemofiltration , Humans , Interleukins/metabolism , Materials Testing , Membranes, Artificial , Mice , NIH 3T3 Cells , Permeability , Sus scrofa , Urea/metabolism , gamma-Glutamyltransferase/metabolism
15.
Kidney Int ; 83(4): 593-603, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23389418

ABSTRACT

Renal cells are used in basic research, disease models, tissue engineering, drug screening, and in vitro toxicology. In order to provide a reliable source of human renal cells, we developed a protocol for the differentiation of human embryonic stem cells into renal epithelial cells. The differentiated stem cells expressed markers characteristic of renal proximal tubular cells and their precursors, whereas markers of other renal cell types were not expressed or expressed at low levels. Marker expression patterns of these differentiated stem cells and in vitro cultivated primary human renal proximal tubular cells were comparable. The differentiated stem cells showed morphological and functional characteristics of renal proximal tubular cells, and generated tubular structures in vitro and in vivo. In addition, the differentiated stem cells contributed in organ cultures for the formation of simple epithelia in the kidney cortex. Bioreactor experiments showed that these cells retained their functional characteristics under conditions as applied in bioartificial kidneys. Thus, our results show that human embryonic stem cells can differentiate into renal proximal tubular-like cells. Our approach would provide a source for human renal proximal tubular cells that are not affected by problems associated with immortalized cell lines or primary cells.


Subject(s)
Bioartificial Organs , Cell Differentiation , Embryonic Stem Cells/physiology , Epithelial Cells/physiology , Kidney Tubules, Proximal/physiology , Tissue Engineering , Activins/pharmacology , Animals , Biomarkers/metabolism , Bioreactors , Bone Morphogenetic Protein 2/pharmacology , Bone Morphogenetic Protein 7/pharmacology , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Shape , Cells, Cultured , Dose-Response Relationship, Drug , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/transplantation , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/transplantation , Gene Expression Regulation, Developmental , Humans , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/transplantation , Mice , Mice, SCID , Organ Culture Techniques , Time Factors , Tissue Engineering/methods , Tretinoin/pharmacology
16.
Nat Rev Cancer ; 4(9): 677-87, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15343274

ABSTRACT

Nuclear architecture - the spatial arrangement of chromosomes and other nuclear components - provides a framework for organizing and regulating the diverse functional processes within the nucleus. There are characteristic differences in the nuclear architectures of cancer cells, compared with normal cells, and some anticancer treatments restore normal nuclear structure and function. Advances in understanding nuclear structure have revealed insights into the process of malignant transformation and provide a basis for the development of new diagnostic tools and therapeutics.


Subject(s)
Cell Nucleus/ultrastructure , Chromatin/ultrastructure , Neoplasms/pathology , Nuclear Matrix , Humans , Neoplasms/diagnosis
17.
Nucleic Acids Res ; 39(15): 6489-99, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21511815

ABSTRACT

DNA double-strand breaks (DSBs) can induce chromosomal aberrations and carcinogenesis and their correct repair is crucial for genetic stability. The cellular response to DSBs depends on damage signaling including the phosphorylation of the histone H2AX (γH2AX). However, a lack of γH2AX formation in heterochromatin (HC) is generally observed after DNA damage induction. Here, we examine γH2AX and repair protein foci along linear ion tracks traversing heterochromatic regions in human or murine cells and find the DSBs and damage signal streaks bending around highly compacted DNA. Given the linear particle path, such bending indicates a relocation of damage from the initial induction site to the periphery of HC. Real-time imaging of the repair protein GFP-XRCC1 confirms fast recruitment to heterochromatic lesions inside murine chromocenters. Using single-ion microirradiation to induce localized DSBs directly within chromocenters, we demonstrate that H2AX is early phosphorylated within HC, but the damage site is subsequently expelled from the center to the periphery of chromocenters within ∼ 20 min. While this process can occur in the absence of ATM kinase, the repair of DSBs bordering HC requires the protein. Finally, we describe a local decondensation of HC at the sites of ion hits, potentially allowing for DSB movement via physical forces.


Subject(s)
DNA Breaks, Double-Stranded , DNA Repair , Euchromatin/metabolism , Heterochromatin/metabolism , Histones/metabolism , Animals , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/metabolism , Cells, Cultured , Chromobox Protein Homolog 5 , Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins/metabolism , HeLa Cells , Humans , Kinetics , Mice , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , X-ray Repair Cross Complementing Protein 1
18.
Biomater Sci ; 11(13): 4675-4683, 2023 Jun 27.
Article in English | MEDLINE | ID: mdl-37219049

ABSTRACT

Toxicity towards non-tumor cells during anticancer therapy can be reduced by using nanoscale systems for anticancer drug delivery. Usually only the loaded drug has anticancer activity. Recently, micellar nanocomplexes (MNCs) comprising green tea catechin derivatives for the delivery of the anticancer proteins, such as Herceptin, have been developed. Herceptin as well as the MNCs without the drug were effective against HER2/neu-overexpressing human tumor cells and had synergistic anticancer effects in vitro and in vivo. It remained unclear which kinds of negative effects the MNCs had on tumor cells exactly, and which of their components mediated them. Also, it was unclear if MNC has any toxicity effects on the normal cells of vital human organ systems. Herein we examined the effects of Herceptin-MNCs and their individual components on human breast cancer cells and on normal primary human endothelial and kidney proximal tubular cells. We applied a novel in vitro model that predicts nephrotoxicity in humans with high accuracy, as well as high-content screening and microfluidic mono- and co-culture models to thoroughly address effects on various cell types. The results showed that MNCs alone were profoundly toxic for breast cancer cells, and induced apoptosis regardless of HER2/neu expression levels. Apoptosis was induced by both green tea catechin derivatives contained within MNCs. In contrast, MNCs were not toxic for normal human cells, and the probability was low that MNCs would be nephrotoxic in humans. Together, the results supported the hypothesis that green tea catechin derivative-based MNCs could improve efficacy and safety of therapies with anticancer proteins.


Subject(s)
Breast Neoplasms , Catechin , Humans , Female , Micelles , Trastuzumab , Tea
19.
J Cell Biochem ; 113(8): 2607-21, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22422629

ABSTRACT

The nuclear positioning of mammalian genes often correlates with their functional state. For instance, the human cystic fibrosis transmembrane conductance regulator (CFTR) gene associates with the nuclear periphery in its inactive state, but occupies interior positions when active. It is not understood how nuclear gene positioning is determined. Here, we investigated trichostatin A (TSA)-induced repositioning of CFTR in order to address molecular mechanisms controlling gene positioning. Treatment with the histone deacetylase (HDAC) inhibitor TSA induced increased histone acetylation and CFTR repositioning towards the interior within 20 min. When CFTR localized in the nuclear interior (either after TSA treatment or when the gene was active) consistent histone H3 hyperacetylation was observed at a CTCF site close to the CFTR promoter. Knockdown experiments revealed that CTCF was essential for perinuclear CFTR positioning and both, CTCF knockdown as well as TSA treatment had similar and CFTR-specific effects on radial positioning. Furthermore, knockdown experiments revealed that also A-type lamins were required for the perinuclear positioning of CFTR. Together, the results showed that CTCF, A-type lamins and an active HDAC were essential for perinuclear positioning of CFTR and these components acted on a CTCF site adjacent to the CFTR promoter. The results are consistent with the idea that CTCF bound close to the CFTR promoter, A-type lamins and an active HDAC form a complex at the nuclear periphery, which becomes disrupted upon inhibition of the HDAC, leading to the observed release of CFTR.


Subject(s)
Cell Nucleus/metabolism , Cystic Fibrosis/metabolism , Histone Deacetylases/metabolism , Lamins/metabolism , Acetylation , Cell Line , Chromatin Immunoprecipitation , Cystic Fibrosis/genetics , Cystic Fibrosis Transmembrane Conductance Regulator , HeLa Cells , Histone Deacetylases/genetics , Humans , Immunoblotting , Immunoprecipitation , Lamins/genetics , Polymerase Chain Reaction , RNA Interference
20.
Am J Physiol Renal Physiol ; 302(8): F1055-62, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22319059

ABSTRACT

Interactions between renal tubular epithelial cells and adjacent endothelial cells are essential for normal renal functions but also play important roles in renal disease and repair. Here, we investigated cocultures of human primary renal proximal tubular cells (HPTC) and human primary endothelial cells to address the cross talk between these cell types. HPTC showed improved proliferation, marker gene expression, and enzyme activity in cocultures. Also, the long-term maintenance of epithelia formed by HPTC was improved, which was due to the secretion of transforming growth factor-ß1 and its antagonist α2-macroglobulin. HPTC induced endothelial cells to secrete increased amounts of these factors, which balanced each other functionally and only displayed in combination the observed positive effects. In addition, in the presence of HPTC endothelial cells expressed increased amounts of hepatocyte growth factor and vascular endothelial growth factor, which have well-characterized effects on renal tubular epithelial cells as well as on endothelial cells. Together, the results showed that HPTC stimulated endothelial cells to express a functionally balanced combination of various factors, which in turn improved the performance of HPTC. The results give new insights into the cross talk between renal epithelial and endothelial cells and suggest that cocultures could be also useful models for the analysis of cellular communication in renal disease and repair. Furthermore, the characterization of defined microenvironments, which positively affect HPTC, will be helpful for improving the performance of this cell type in in vitro applications including in vitro toxicology and kidney tissue engineering.


Subject(s)
Kidney Glomerulus/metabolism , Kidney Tubules, Proximal/metabolism , Cell Communication/physiology , Cell Line , Cell Proliferation , Coculture Techniques , Gene Expression , Hepatocyte Growth Factor/biosynthesis , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/physiology , Humans , Kidney Glomerulus/physiology , Kidney Tubules, Proximal/physiology , Transforming Growth Factor beta1/antagonists & inhibitors , Transforming Growth Factor beta1/metabolism , Vascular Endothelial Growth Factor A/biosynthesis , alpha-Macroglobulins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL