Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Chem Biol Interact ; 387: 110813, 2024 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-38006960

RESUMEN

Podocytes are of key interest for the prediction of nephrotoxicity as they are especially sensitive to toxic insults due to their central role in the glomerular filtration apparatus. However, currently, prediction of nephrotoxicity in humans remains insufficiently reliable, thus highlighting the need for advanced in vitro model systems using human cells with improved prediction capacity. Recent approaches for refining in vitro model systems focus on closely replicating physiological conditions as observed under the in vivo situation typical of the respective nephron section of interest. PODO/TERT256, a human immortalized podocyte cell line, were employed in a semi-static transwell system to evaluate its potential use as a human podocyte in vitro system for modelling potential human glomerular toxicity. Furthermore, the impact of routinely employed excessive oxygen tension (21 % - AtmOx), when compared to the physiological oxygen tensions (10 % - PhysOx) observed in vivo, was analyzed. Generally, cultured PODO/TERT256 formed a stable, contact-inhibited monolayer with typical podocyte morphology (large cell body, apical microvilli, finger-like cytoplasmic projections (reminiscent of foot processes), and interdigitating cell-cell junctions) and developed a size-selective filtration barrier. PhysOx, however, induced a more pronounced in vivo like phenotype, comprised of significantly larger cell bodies, significantly enhanced filtration barrier size-selectivity, and a remarkable re-localization of nephrin to the cell membrane, thus suggesting an improved in vitro replication of in vivo characteristics. Preliminary toxicity characterization with the known glomerulotoxin doxorubicin (DOX) suggested an increasing change in filtration permeability, already at the lowest DOX concentrations tested (0.01 µM) under PhysOx, whereas obvious changes under AtmOx were observed as of 0.16 µM and higher with a near all or nothing effect. The latter findings suggested that PODO/TERT256 could serve as an in vitro human podocyte model for studying glomerulotoxicity, whereby culturing at PhyOx tension appeared critical for an improved in vivo-like phenotype and functionality. Moreover, PODO/TERT256 could be incorporated into advanced human glomerulus systems in vitro, recapitulating microfluidic conditions and multiple cell types (endothelial and mesenchymal cells) that can even better predict human glomerular toxicity.


Asunto(s)
Enfermedades Renales , Podocitos , Humanos , Podocitos/metabolismo , Glomérulos Renales/metabolismo , Línea Celular , Enfermedades Renales/metabolismo , Membrana Celular/metabolismo , Doxorrubicina/farmacología , Doxorrubicina/metabolismo
2.
Chem Biol Interact ; 361: 109959, 2022 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-35533734

RESUMEN

Reliable prediction of compound mediated nephrotoxicity in humans is still unsatisfactory irrespective of the recent advancements in in silico, in vitro and in vivo models. Therefore, current in vitro approaches need refinement to better match the human in vivo situation, specifically with regard to the potential influence of other cell types (e.g. fibroblasts) and to the potential biases introduced by the excessive 21% O2 (AtmOx) as employed in routine cell culturing. We used a transwell co-culture model combining human renal proximal tubule epithelial cells (RPTEC/TERT1) and human fibroblasts (fHDF/TERT166) to compare the functional properties and expression of selected marker proteins at 21% O2 and at the physiologically normal 10% O2 tension (PhysOx) commensurate with in vivo conditions. Culturing at PhysOx and co-culturing with fibroblasts significantly improved epithelial barrier integrity, expression of transporters (e.g. aquaporin 2; OCT-MATE; MRP-OAT) and metabolism. Moreover, beyond culturing these human cells in co-culture for up to 41 days, we were able to demonstrate increased functionality of cation transport, as shown via ASP+ (OCT-MATE axis), and anion transport, as shown via LY (MRP-OAT axis). Thus, adjusting the in vitro system to near physiological conditions had a major impact on functionality and provides the basis for the future development of true flow-through microfluidic renal testing systems with better predictability of human renal proximal toxicity.


Asunto(s)
Túbulos Renales Proximales , Oxígeno , Línea Celular , Técnicas de Cocultivo , Células Epiteliales/metabolismo , Fibroblastos , Humanos , Oxígeno/metabolismo
3.
J Cell Biochem ; 122(5): 507-523, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33417272

RESUMEN

There are conflicting data about localization of poly(ADP-ribose)polymerase-1 and its product poly(ADP-ribose) in mitochondria. To finally clarify the discussion, we investigated with biochemical and cell biological methods the potential presence of poly(ADP-ribose) polymerase-1 in these organelles. Our data show that endogenous and overexpressed poly(ADP-ribose)polymerase 1 is only localized to the nucleus with a clear exclusion of cytosolic compartments. In addition, highly purified mitochondria devoid of nuclear contaminations do not contain poly(ADP-ribose)polymerase-1. Although no poly(ADP-ribose)polymerase-1 enzyme is detectable in mitochondria, a shorter variant of its product poly(ADP-ribose) is present, associated specifically with a small subset of mitochondrial proteins as revealed by immunoprecipitation and protein fingerprint analysis. These proteins are located at key-points of the Krebs-cycle, are chaperones involved in mitochondrial functionality and quality-control, and are RNA-binding proteins important for transcript stability, respectively. Of note, despite the fact that especially poly(ADP-ribose)polymerase-1 is its own major target for modification, we could not detect this enzyme by mass spectrometry in these organelles. These data suggests a new way of targeted nuclear-mitochondrial signaling, mediated by nuclear poly(ADP-ribosyl)ation dependent on poly(ADP-ribose)polymerase-1.


Asunto(s)
Núcleo Celular/metabolismo , Mitocondrias/metabolismo , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Poli Adenosina Difosfato Ribosa/metabolismo , Línea Celular Tumoral , Técnica del Anticuerpo Fluorescente , Humanos , Inmunoprecipitación
4.
Toxicology ; 420: 29-38, 2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-30940547

RESUMEN

Aristolochic acid (AA) dependent human nephropathy results either from environmental exposure to Aristolochiaceae plant subspecies or their use in traditional phytotherapy. The toxic components are structurally related nitrophenanthrene carboxylic acids, i.e. Aristolochic acid I (AAI) and II (AAII). AAI is considered to be the major cause of Aristolochic acid nephropathy, characterized by severe renal fibrosis and upper urothelial cancer. Following enzymatic activation in kidney and/or liver, AAI metabolites react with genomic DNA to form persistent DNA adducts with purines. To determine whether AAI can be activated in human renal cells to form DNA adducts, we exposed telomerase immortalized renal proximal tubular epithelial cells (RPTEC/TERT1), the human embryonic kidney (HEK293) cell line, as well as primary human kidney cells (pHKC) to AAI in vitro. We modified an isotope dilution ultra-performance liquid chromatography/tandem mass spectrometry (ID-UPLC-MS/MS) based method for the quantification of dA-AAI adducts in genomic DNA. In addition, time dependent accumulation of adducts in renal cortex and bladder tissue from AAI/II treated Eker rats were used to validate the detection method. AAI-induced toxicity in human renal cells was determined by dA-AAI adduct quantification, the impact on cell viability, and NQO1 expression and activity. Our findings demonstrated adduct formation in all cell lines, although only pHKC and RPTEC/TERT1 expressed NQO1. The highest adduct formation was detected in pHKC despite low NQO1 expression, while we observed much lower adduct levels in NQO1-negative HEK293 cells. Adduct formation and decreased cell viability correlated only weakly. Therefore, our data suggested that i.) enzymes other than NQO1 could be at least equally important for AA bioactivation in human renal proximal tubule cells, and ii.) the suggested correlation between adduct levels and viability appears to be questionable.


Asunto(s)
Ácidos Aristolóquicos/toxicidad , Aductos de ADN/metabolismo , Enfermedades Renales/inducido químicamente , Riñón/efectos de los fármacos , Activación Metabólica , Anciano , Animales , Supervivencia Celular/efectos de los fármacos , Femenino , Células HEK293 , Humanos , Riñón/metabolismo , Riñón/patología , Enfermedades Renales/genética , Enfermedades Renales/metabolismo , Enfermedades Renales/patología , Masculino , Mutación , NAD(P)H Deshidrogenasa (Quinona)/genética , NAD(P)H Deshidrogenasa (Quinona)/metabolismo , Cultivo Primario de Células , Ratas Transgénicas , Proteína 2 del Complejo de la Esclerosis Tuberosa/genética
5.
Cell Death Dis ; 9(2): 226, 2018 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-29445145

RESUMEN

Recent FDA Drug Safety Communications report an increased risk for acute kidney injury in patients treated with the gliflozin class of sodium/glucose co-transport inhibitors indicated for treatment of type 2 diabetes mellitus. To identify a potential rationale for the latter, we used an in vitro human renal proximal tubule epithelial cell model system (RPTEC/TERT1), physiologically representing human renal proximal tubule function. A targeted metabolomics approach, contrasting gliflozins to inhibitors of central carbon metabolism and mitochondrial function, revealed a double mode of action for canagliflozin, but not for its analogs dapagliflozin and empagliflozin. Canagliflozin inhibited the glutamate dehydrogenase (GDH) and mitochondrial electron transport chain (ETC) complex I at clinically relevant concentrations. This dual inhibition specifically prevented replenishment of tricarboxylic acid cycle metabolites by glutamine (anaplerosis) and thus altered amino acid pools by increasing compensatory transamination reactions. Consequently, canagliflozin caused a characteristic intracellular accumulation of glutamine, glutamate and alanine in confluent, quiescent RPTEC/TERT1. Canagliflozin, but none of the classical ETC inhibitors, induced cytotoxicity at particularly low concentrations in proliferating RPTEC/TERT1, serving as model for proximal tubule regeneration in situ. This finding is testimony of the strong dependence of proliferating cells on glutamine anaplerosis via GDH. Our discovery of canagliflozin-mediated simultaneous inhibition of GDH and ETC complex I in renal cells at clinically relevant concentrations, and their particular susceptibility to necrotic cell death during proliferation, provides a mechanistic rationale for the adverse effects observed especially in patients with preexisting chronic kidney disease or previous kidney injury characterized by sustained regenerative tubular epithelial cell proliferation.


Asunto(s)
Canagliflozina/efectos adversos , Ciclo del Ácido Cítrico/efectos de los fármacos , Complejo I de Transporte de Electrón/metabolismo , Células Epiteliales/efectos de los fármacos , Glutamato Deshidrogenasa/metabolismo , Hipoglucemiantes/efectos adversos , Alanina/metabolismo , Compuestos de Bencidrilo/metabolismo , Compuestos de Bencidrilo/farmacología , Muerte Celular/efectos de los fármacos , Línea Celular , Complejo I de Transporte de Electrón/antagonistas & inhibidores , Células Epiteliales/citología , Células Epiteliales/enzimología , Glucósidos/metabolismo , Glucósidos/farmacología , Glutamato Deshidrogenasa/antagonistas & inhibidores , Ácido Glutámico/metabolismo , Glutamina/metabolismo , Humanos , Túbulos Renales Proximales/citología , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/enzimología , Metabolómica/métodos , Mitocondrias/efectos de los fármacos , Mitocondrias/enzimología
6.
DNA Repair (Amst) ; 52: 12-23, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28216063

RESUMEN

NAD+ is an essential cofactor for enzymes catalyzing redox-reactions as well as an electron carrier in energy metabolism. Aside from this, NAD+ consuming enzymes like poly(ADP-ribose) polymerases and sirtuins are important regulators involved in chromatin-restructuring processes during repair and epigenetics/transcriptional adaption. In order to replenish cellular NAD+ levels after cleavage, synthesis starts from precursors such as nicotinamide, nicotinamide riboside or nicotinic acid to match the need for this essential molecule. In the present study, we investigated the impact of supplementation with nicotinic acid on resting and proliferating human mononuclear blood cells with a focus on DNA damage and repair processes. We observed that nicotinic acid supplementation increased NAD+ levels as well as DNA repair efficiency and enhanced genomic stability evaluated by micronucleus test after x-ray treatment. Interestingly, resting cells displayed lower basal levels of DNA breaks compared to proliferating cells, but break-induction rates were identical. Despite similar levels of p53 protein upregulation after irradiation, higher NAD+ concentrations led to reduced acetylation of this protein, suggesting enhanced SIRT1 activity. Our data reveal that even in normal primary human cells cellular NAD+ levels may be limiting under conditions of genotoxic stress and that boosting the NAD+ system with nicotinic acid can improve genomic stability.


Asunto(s)
Daño del ADN , Reparación del ADN , Leucocitos Mononucleares/metabolismo , NAD/metabolismo , Niacina/metabolismo , Radiación Ionizante , Acetilación , Adulto , ADN/metabolismo , ADN/efectos de la radiación , Suplementos Dietéticos , Humanos , Leucocitos Mononucleares/efectos de la radiación , Persona de Mediana Edad , NAD/análisis , Sirtuinas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Adulto Joven
7.
Nucleic Acids Res ; 44(21): 10386-10405, 2016 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-27694308

RESUMEN

Genotoxic stress activates PARP1, resulting in the post-translational modification of proteins with poly(ADP-ribose) (PAR). We genetically deleted PARP1 in one of the most widely used human cell systems, i.e. HeLa cells, via TALEN-mediated gene targeting. After comprehensive characterization of these cells during genotoxic stress, we analyzed structure-function relationships of PARP1 by reconstituting PARP1 KO cells with a series of PARP1 variants. Firstly, we verified that the PARP1\E988K mutant exhibits mono-ADP-ribosylation activity and we demonstrate that the PARP1\L713F mutant is constitutively active in cells. Secondly, both mutants exhibit distinct recruitment kinetics to sites of laser-induced DNA damage, which can potentially be attributed to non-covalent PARP1-PAR interaction via several PAR binding motifs. Thirdly, both mutants had distinct functional consequences in cellular patho-physiology, i.e. PARP1\L713F expression triggered apoptosis, whereas PARP1\E988K reconstitution caused a DNA-damage-induced G2 arrest. Importantly, both effects could be rescued by PARP inhibitor treatment, indicating distinct cellular consequences of constitutive PARylation and mono(ADP-ribosyl)ation. Finally, we demonstrate that the cancer-associated PARP1 SNP variant (V762A) as well as a newly identified inherited PARP1 mutation (F304L\V762A) present in a patient with pediatric colorectal carcinoma exhibit altered biochemical and cellular properties, thereby potentially supporting human carcinogenesis. Together, we establish a novel cellular model for PARylation research, by revealing strong structure-function relationships of natural and artificial PARP1 variants.


Asunto(s)
Poli(ADP-Ribosa) Polimerasa-1/química , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Nucleasas de los Efectores Tipo Activadores de la Transcripción/química , Nucleasas de los Efectores Tipo Activadores de la Transcripción/metabolismo , Animales , Línea Celular , Daño del ADN , Técnicas de Inactivación de Genes , Marcación de Gen , Variación Genética , Células HeLa , Humanos , NAD/metabolismo , Poli(ADP-Ribosa) Polimerasa-1/genética , Unión Proteica , Conformación Proteica , Proteínas Recombinantes , Eliminación de Secuencia , Relación Estructura-Actividad
8.
BMC Cancer ; 16: 469, 2016 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-27412346

RESUMEN

BACKGROUND: Chemotherapy is one of the major treatment modalities for cancer. Metal-based compounds such as derivatives of cisplatin are in the front line of therapy against a subset of cancers, but their use is restricted by severe side-effects and the induction of resistance in treated tumors. Subsequent research focused on development of cytotoxic metal-complexes without cross-resistance to cisplatin and reduced side-effects. This led to the discovery of first-generation titanium(IV)salan complexes, which reached clinical trials but lacked efficacy. New-generation titanium (IV)salan-complexes show promising anti-tumor activity in mice, but their molecular mechanism of cytotoxicity is completely unknown. METHODS: Four different human cell lines were analyzed in their responses to a toxic (Tc52) and a structurally highly related but non-toxic (Tc53) titanium(IV)salan complex. Viability assays were used to reveal a suitable treatment range, flow-cytometry analysis was performed to monitor the impact of dosage and treatment time on cell-cycle distribution and cell death. Potential DNA strand break induction and crosslinking was investigated by immunostaining of damage markers as well as automated fluorometric analysis of DNA unwinding. Changes in nuclear morphology were analyzed by DAPI staining. Acidic beta-galactosidase activity together with morphological changes was monitored to detect cellular senescence. Western blotting was used to analyze induction of pro-apoptotic markers such as activated caspase7 and cleavage of PARP1, and general stress kinase p38. RESULTS: Here we show that the titanium(IV)salan Tc52 is effective in inducing cell death in the lower micromolar range. Surprisingly, Tc52 does not target DNA contrary to expectations deduced from the reported activity of other titanium complexes. Instead, Tc52 application interferes with progression from G2-phase into mitosis and induces apoptotic cell death in tested tumor cells. Contrarily, human fibroblasts undergo senescence in a time and dose-dependent manner. As deduced from fluorescence studies, the potential cellular target seems to be the cytoskeleton. CONCLUSIONS: In summary, we could demonstrate in four different human cell lines that tumor cells were specifically killed without induction of major cytotoxicity in non-tumorigenic cells. Absence of DNA damaging activity and the cell-cycle block in G2 instead of mitosis makes Tc52 an attractive compound for further investigations in cancer treatment.


Asunto(s)
Apoptosis/efectos de los fármacos , Complejos de Coordinación/farmacología , Fase G2/efectos de los fármacos , Radioisótopos/farmacología , Titanio/farmacología , Antineoplásicos , Western Blotting , Caspasa 7/metabolismo , División Celular/efectos de los fármacos , Línea Celular Tumoral , Núcleo Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Senescencia Celular/efectos de los fármacos , Complejos de Coordinación/química , Citoesqueleto/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Citometría de Flujo , Células HEK293 , Células HeLa , Humanos , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Radioisótopos/química , Titanio/química , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
9.
Angew Chem Int Ed Engl ; 55(37): 11256-60, 2016 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-27468728

RESUMEN

Poly(ADP-ribos)ylation (PARylation) is an important posttranslational protein modification, and is involved in major cellular processes such as gene regulation and DNA repair. Its dysregulation has been linked to several diseases, including cancer. Despite its importance, methods to observe PARylation dynamics within cells are rare. By following a chemical biology approach, we developed a fluorescent NAD(+) analogue that proved to be a competitive building block for protein PARylation in vitro and in cells. This allowed us to directly monitor the turnover of PAR in living cells at DNA damage sites after near-infrared (NIR) microirradiation. Additionally, covalent and noncovalent interactions of selected target proteins with PAR chains were visualized in cells by using FLIM-FRET microscopy. Our results open up new opportunities for the study of protein PARylation in real time and in live cells, and will thus contribute to a better understanding of its significance in a cellular context.


Asunto(s)
Imagen Óptica , Poli Adenosina Difosfato Ribosa/metabolismo , Proteínas/metabolismo , Daño del ADN , Fluorescencia , Células HeLa , Histonas/química , Histonas/metabolismo , Humanos , Rayos Infrarrojos , Estructura Molecular , NAD/análogos & derivados , NAD/síntesis química , NAD/química , Poli Adenosina Difosfato Ribosa/química , Proteínas/química , Factores de Tiempo
10.
Biochem Pharmacol ; 94(3): 212-9, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25667043

RESUMEN

Depending on their genetic background (p53(wt) versus p53(null)), carcinoma cells are more or less sensitive to drug-induced cell cycle arrest and/or apoptosis. Among the members of the p53 family, p63 is characterized by two N-terminal isoforms, TAp63 and ΔNp63. TAp63 isoform has p53-like functions, while ΔNp63 acts as a dominant negative inhibitor of p53. We have previously published that TAp63 is involved in poly(ADP-ribose)polymerase-1 (PARP-1) signaling of DNA damage deriving from DNA topoisomerase I (TOP I) inhibition in carcinoma cells. In the present study, we treated MCF7 breast carcinoma cells (p53(+)/ΔNp63(-)) or SCC022 (p53(-)/ΔNp63(+)) squamous carcinoma cells with the TOP I inhibitor topotecan (TPT) and the PJ34 PARP inhibitor, to compare their effects in the two different cell contexts. In MCF7 cells, we found that PJ34 addition reverts TPT-dependent PARP-1 auto-modification and triggers caspase-dependent PARP-1 proteolysis. Moreover, TPT as single agent stimulates p53(ser15) phosphorylation, p53 PARylation and occupancy of the p21WAF promoter by p53 resulting in an increase of p21WAF expression. Interestingly, PJ34 in combination with TPT enhances p53 occupancy at the BAX promoter and is associated with increased BAX protein level. In SCC022 cells, instead, TPT+PJ34 combined treatment reduces the level of the anti-apoptotic ΔNp63α protein without inducing apoptosis. Remarkably, in such cells, either exogenous p53 or TAp63 can rescue the apoptotic program in response to the treatment. All together our results suggest that in cancer cells PARP inhibitor(s) can operate in the choice between growth arrest and apoptosis by modulating p53 family-dependent signal.


Asunto(s)
Supervivencia Celular/efectos de los fármacos , Proteínas de la Membrana/fisiología , Poli(ADP-Ribosa) Polimerasas/metabolismo , Inhibidores de Topoisomerasa I/farmacología , Proteína p53 Supresora de Tumor/fisiología , Secuencia de Bases , Supervivencia Celular/fisiología , Cartilla de ADN , Humanos , Células MCF-7 , Procesamiento Proteico-Postraduccional , Reacción en Cadena en Tiempo Real de la Polimerasa
11.
Biol Reprod ; 92(3): 80, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25673562

RESUMEN

Sperm are highly differentiated cells characterized by their species-specific nuclear shapes and extremely condensed chromatin. Abnormal head shapes represent a form of teratozoospermia that can impair fertilization capacity. This study shows that poly(ADP-ribose) polymerase-11 (ARTD11/PARP11), a member of the ADP-ribosyltransferase (ARTD) family, is expressed preferentially in spermatids undergoing nuclear condensation and differentiation. Deletion of the Parp11 gene results in teratozoospermia and male infertility in mice due to the formation of abnormally shaped fertilization-incompetent sperm, despite normal testis weights and sperm counts. At the subcellular level, PARP11-deficient elongating spermatids reveal structural defects in the nuclear envelope and chromatin detachment associated with abnormal nuclear shaping, suggesting functional relevance of PARP11 for nuclear envelope stability and nuclear reorganization during spermiogenesis. In vitro, PARP11 exhibits mono(ADP-ribosyl)ation activity with the ability to ADP-ribosylate itself. In transfected somatic cells, PARP11 colocalizes with nuclear pore components, such as NUP153. Amino acids Y77, Q86, and R95 in the N-terminal WWE domain, as well as presence of the catalytic domain, are essential for colocalization of PARP11 with the nuclear envelope, but catalytic activity of the protein is not required for colocalization with NUP153. This study demonstrates that PARP11 is a novel enzyme important for proper sperm head shaping and identifies it as a potential factor involved in idiopathic mammalian teratozoospermia.


Asunto(s)
Forma del Núcleo Celular/fisiología , Poli(ADP-Ribosa) Polimerasas/fisiología , Cabeza del Espermatozoide/fisiología , Espermátides/fisiología , Espermatogénesis/fisiología , Animales , Núcleo Celular/patología , Núcleo Celular/fisiología , Forma del Núcleo Celular/genética , Células Cultivadas , Modelos Animales de Enfermedad , Infertilidad Masculina/genética , Infertilidad Masculina/fisiopatología , Masculino , Ratones , Ratones Noqueados , Membrana Nuclear/fisiología , Fenotipo , Poli(ADP-Ribosa) Polimerasas/deficiencia , Poli(ADP-Ribosa) Polimerasas/genética , Capacitación Espermática/genética , Capacitación Espermática/fisiología , Cabeza del Espermatozoide/patología , Espermátides/citología , Espermatogénesis/genética
12.
Methods Mol Biol ; 1228: 67-81, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25311123

RESUMEN

Genomic DNA is always associated with proteins that modulate the accessibility of the genetic information. This chromatin is the essential structure in which all nuclear activity from regulation to replication, transcription, and repair takes place. This dynamic structure can be most efficiently analyzed by using the method of chromatin immunoprecipitation (ChIP), where application of cell-permeable cross-linkers to living cells induces covalent bridging between proteins and adjacent DNA in the nucleus. After fragmentation of the DNA, the complexed proteins are isolated by binding to specific antibodies. The attached DNA is isolated and can be analyzed. This method has been improved multiple times and adjusted to different experimental needs. This chapter describes a further advance based on the observation that the current standard method itself induces alterations in the chromatin.


Asunto(s)
Inmunoprecipitación de Cromatina/métodos , Daño del ADN , Transducción de Señal , Proteínas Bacterianas/metabolismo , Línea Celular , Proliferación Celular , Cloroformo/química , ADN/genética , ADN/aislamiento & purificación , Fragmentación del ADN , Humanos , Fenol/química , Reacción en Cadena de la Polimerasa
13.
FEBS J ; 281(16): 3625-41, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24953096

RESUMEN

Poly(ADP-ribose) (PAR) is a complex and reversible post-translational modification that controls protein function and localization through covalent modification of, or noncovalent binding to target proteins. Previously, we and others characterized the noncovalent, high-affinity binding of the key nucleotide excision repair (NER) protein XPA to PAR. In the present study, we address the functional relevance of this interaction. First, we confirm that pharmacological inhibition of cellular poly(ADP-ribosyl)ation (PARylation) impairs NER efficacy. Second, we demonstrate that the XPA-PAR interaction is mediated by specific basic amino acids within a highly conserved PAR-binding motif, which overlaps the DNA damage-binding protein 2 (DDB2) and transcription factor II H (TFIIH) interaction domains of XPA. Third, biochemical studies reveal a mutual regulation of PARP1 and XPA functions showing that, on the one hand, the XPA-PAR interaction lowers the DNA binding affinity of XPA, whereas, on the other hand, XPA itself strongly stimulates PARP1 enzymatic activity. Fourth, microirradiation experiments in U2OS cells demonstrate that PARP inhibition alters the recruitment properties of XPA-green fluorescent protein to sites of laser-induced DNA damage. In conclusion, our results reveal that XPA and PARP1 regulate each other in a reciprocal and PAR-dependent manner, potentially acting as a fine-tuning mechanism for the spatio-temporal regulation of the two factors during NER.


Asunto(s)
Poli Adenosina Difosfato Ribosa/metabolismo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Procesamiento Proteico-Postraduccional , Proteína de la Xerodermia Pigmentosa del Grupo A/metabolismo , Secuencia de Aminoácidos , Secuencia de Consenso , Reparación del ADN , Células HeLa , Humanos , Cinética , Datos de Secuencia Molecular , Poli(ADP-Ribosa) Polimerasa-1 , Poli(ADP-Ribosa) Polimerasas/química , Unión Proteica , Células Sf9 , Proteína de la Xerodermia Pigmentosa del Grupo A/química
14.
J Trace Elem Med Biol ; 28(2): 138-146, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23994116

RESUMEN

Thio-dimethylarsinic acid (thio-DMA(V)) has recently been identified as human metabolite after exposure toward both the human carcinogen inorganic arsenic and arsenosugars, which are the major arsenical constituents of marine algae. This study aims to get further insight in the toxic modes of action of thio-DMA(V) in cultured human urothelial cells. Among others effects of thio-DMA(V) on eight cell death related endpoints, cell cycle distribution, genotoxicity, cellular bioavailability as well as for the first time its impact on DNA damage induced poly(ADP-ribosyl)ation were investigated and compared to effects induced by arsenite. The data indicate that thio-DMA(V) exerts its cellular toxicity in a similar or even lower concentration range, however most likely via different mechanisms, than arsenite. Most interestingly, thio-DMA(V) decreased damage-induced cellular poly(ADP-ribosyl)ation by 35,000-fold lower concentrations than arsenite. The inhibition of this essential DNA-damage induced and DNA-repair related signaling reaction might contribute to inorganic arsenic induced toxicity, at least in the bladder. Therefore, and also because thio-DMA(V) is to date by far the most toxic human metabolite identified after arsenosugar intake, thio-DMA(V) should contemporary be fully (also in vivo) toxicologically characterized, to assess risks to human health related to inorganic arsenic but especially arsenosugar dietary intake.


Asunto(s)
Arseniatos/metabolismo , Arseniatos/toxicidad , Arsénico/toxicidad , Ácido Cacodílico/análogos & derivados , Monosacáridos/metabolismo , Monosacáridos/toxicidad , Compuestos de Sulfhidrilo/toxicidad , Vejiga Urinaria/efectos de los fármacos , Vejiga Urinaria/patología , Adenosina Difosfato Ribosa/metabolismo , Apoptosis/efectos de los fármacos , Apoptosis/genética , Arsenicales , Disponibilidad Biológica , Ácido Cacodílico/toxicidad , Caspasa 2/metabolismo , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Muerte Celular/efectos de los fármacos , Línea Celular , Roturas del ADN de Doble Cadena/efectos de los fármacos , Daño del ADN/efectos de los fármacos , Daño del ADN/genética , Regulación de la Expresión Génica , Glicósido Hidrolasas/genética , Glicósido Hidrolasas/metabolismo , Humanos , Peróxido de Hidrógeno/toxicidad , NAD/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Poli(ADP-Ribosa) Polimerasas/genética , Poli(ADP-Ribosa) Polimerasas/metabolismo , Proteínas Recombinantes/metabolismo
15.
Mol Nutr Food Res ; 57(7): 1255-69, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23495240

RESUMEN

SCOPE: In industrial countries dietary manganese (Mn) intake is well above the estimated average requirement. Moreover, exposure to high Mn levels is known to cause adverse neurological effects in humans, which are yet mechanistically not well understood. METHODS AND RESULTS: This study aimed to identify early modes of action of Mn induced toxicity in mammalian brain cells. In primary porcine brain capillary endothelial cells induction of reactive oxygen and nitrogen species was identified as the most sensitive endpoint (≥0.5 µM MnCl2 ). In cultured human astrocytes MnCl2 was rapidly bioavailable, induced a slight increase of cellular reactive oxygen and nitrogen species levels and a slight decrease of ATP levels (1-100 µM MnCl2 ), while no genotoxic effects were observed. However, MnCl2 (≥1 µM) efficiently disturbed DNA-damage-induced poly(ADP-ribosyl)ation in human astrocytes, which indicates sensitization of cells to genotoxic treatment. Additionally, we determined Mn levels in infant formula, which are generally massively supplemented with Mn and thus might pose an important source for Mn overexposure. CONCLUSION: The observed inhibition of DNA-damage-induced poly(ADP-ribosyl)ation in human astrocytes by exposure-relevant Mn concentrations indicate that in terms of Mn the existing guidelines for infant formula but also drinking water should be critically reconsidered.


Asunto(s)
Cloruros/toxicidad , Daño del ADN/efectos de los fármacos , Síndromes de Neurotoxicidad/fisiopatología , Especies de Nitrógeno Reactivo/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Disponibilidad Biológica , Encéfalo/citología , Encéfalo/efectos de los fármacos , Línea Celular Tumoral , Cloruros/administración & dosificación , Contaminación de Alimentos/análisis , Humanos , Fórmulas Infantiles/química , Compuestos de Manganeso/administración & dosificación , Síndromes de Neurotoxicidad/etiología , Poli(ADP-Ribosa) Polimerasa-1 , Poli(ADP-Ribosa) Polimerasas/genética , Poli(ADP-Ribosa) Polimerasas/metabolismo , Porcinos
16.
Cell Commun Signal ; 11(1): 11, 2013 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-23384168

RESUMEN

The determination of the cytotoxic potential of new and so far unknown compounds as well as their metabolites is fundamental in risk assessment. A variety of strategic endpoints have been defined to describe toxin-cell interactions, leading to prediction of cell fate. They involve measurement of metabolic endpoints, bio-energetic parameters or morphological cell modifications. Here, we evaluated alterations of the free cytosolic Ca2+ homeostasis using the Fluo-4 dye and compared results with the metabolic cell viability assay Alamar Blue. We investigated a panel of toxins (As2O3, gossypol, H2O2, staurosporine, and titanium(IV)-salane complexes) in four different mammalian cell lines covering three different species (human, mouse, and African green monkey). All tested compounds induced an increase in free cytosolic Ca2+ within the first 5 s after toxin application. Cytosolic Ca2+ shifts occurred independently of the chemical structure in all tested cell systems and were persistent up to 3 h. The linear increase of free cytosolic Ca2+ within the first 5 s of drug treatment correlates with the EC25 and EC75 values obtained in Alamar Blue assays one day after toxin exposure. Moreover, a rise of cytosolic Ca2+ was detectable independent of induced cell death mode as assessed by caspase and poly(ADP-ribose) polymerase (PARP) activity in HeLa versus MCF-7 cells at very low concentrations. In conclusion, a cytotoxicity assay based on Ca2+ shifts has a low limit of detection (LOD), is less time consuming (at least 24 times faster) compared to the cell viability assay Alamar Blue and is suitable for high-troughput-screening (HTS).

17.
Toxicology ; 303: 177-86, 2013 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-23178243

RESUMEN

Data on local genotoxicity after particle exposure are crucial to resolve mechanistic aspects such as the impact of chronic inflammation, types of DNA damage, and their role in lung carcinogenesis. We established immunohistochemical methods to quantify the DNA damage markers poly(ADP-ribose) (PAR), phosphorylated H2AX (γ-H2AX), 8-hydroxyguanosine (8-OH-dG), and 8-oxoguanine DNA glycosylase (OGG1) in paraffin-embedded tissue from particle-exposed rats. The study was based on lungs from a subchronic study that was part of an already published carcinogenicity study where rats had been intratracheally instilled with saline, quartz DQ12, amorphous silica (Aerosil(®) 150), or carbon black (Printex(®) 90) at monthly intervals for 3 months. Lung sections were stained immunohistochemically and markers were quantified in alveolar lining cells. Local genotoxicity was then correlated with already defined endpoints, i.e. mean inflammation score, bronchoalveolar lavage parameters, and carcinogenicity. Genotoxicity was most pronounced in quartz DQ12-treated rats, where all genotoxicity markers gave statistically significant positive results, indicating considerable genotoxic stress such as occurrence of DNA double-strand breaks (DSB), and oxidative damage with subsequent repair activity. Genotoxicity was less pronounced for Printex(®) 90, but significant increases in γ-H2AX- and 8-OH-dG-positive nuclei and OGG1-positive cytoplasm were nevertheless detected. In contrast, Aerosil(®) 150 significantly enhanced only 8-OH-dG-positive nuclei and oxidative damage-related repair activity (OGG1) in cytoplasm. In the present study, γ-H2AX was the most sensitive genotoxicity marker, differentiating best between the three types of particles. The mean number of 8-OH-dG-positive nuclei, however, correlated best with the mean inflammation score at the same time point. This methodological approach enables integration of local genotoxicity testing in subchronic inhalation studies and makes immunohistochemical detection, in particular of γ-H2AX and 8-hydroxyguanine, a very promising approach for local genotoxicity testing in lungs, with prognostic value for the long-term outcome of particle exposure.


Asunto(s)
Pulmón/efectos de los fármacos , Mutágenos/toxicidad , Cuarzo/toxicidad , Dióxido de Silicio/toxicidad , Hollín/toxicidad , Animales , Roturas del ADN de Doble Cadena/efectos de los fármacos , Daño del ADN/efectos de los fármacos , ADN Glicosilasas/metabolismo , Femenino , Guanosina/análogos & derivados , Guanosina/metabolismo , Histonas/metabolismo , Inmunohistoquímica , Pulmón/patología , Pruebas de Mutagenicidad , Tamaño de la Partícula , Fosfoproteínas/metabolismo , Poli Adenosina Difosfato Ribosa/metabolismo , Ratas , Ratas Wistar
18.
Front Genet ; 3: 169, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22969794

RESUMEN

The interaction of DNA with proteins in the context of chromatin has to be tightly regulated to achieve so different tasks as packaging, transcription, replication and repair. The very rapid and transient post-translational modification of proteins by poly(ADP-ribose) has been shown to take part in all four. Originally identified as immediate cellular answer to a variety of genotoxic stresses, already early data indicated the ability of this highly charged nucleic acid-like polymer to modulate nucleosome structure, the basic unit of chromatin. At the same time the enzyme responsible for synthesizing poly(ADP-ribose), the zinc-finger protein poly(ADP-ribose) polymerase-1 (PARP1), was shown to control transcription initiation as basic factor TFIIC within the RNA-polymerase II machinery. Later research focused more on PARP-mediated regulation of DNA repair and cell death, but in the last few years, transcription as well as chromatin modulation has re-appeared on the scene. This review will discuss the impact of PARP1 on transcription and transcription factors, its implication in chromatin remodeling for DNA repair and probably also replication, and its role in controlling epigenetic events such as DNA methylation and the functionality of the insulator protein CCCTC-binding factor.

19.
PLoS One ; 7(3): e32914, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22479348

RESUMEN

Chromatin-immunoprecipitation (ChIP) employs generally a mild formaldehyde cross-linking step, which is followed by isolation of specific protein-DNA complexes and subsequent PCR testing, to analyze DNA-protein interactions. Poly(ADP-ribosyl)ation, a posttranslational modification involved in diverse cellular functions like repair, replication, transcription, and cell death regulation, is most prominent after DNA damage. Poly(ADP-ribose)polymerase-1 is activated upon binding to DNA strand-breaks and coordinates repair by recruitment or displacement of proteins. Several proteins involved in different nuclear pathways are directly modified or contain poly(ADP-ribose)-interaction motifs. Thus, poly(ADP-ribose) regulates chromatin composition. In immunofluorescence experiments, we noticed artificial polymer-formation after formaldehyde-fixation of undamaged cells. Therefore, we analyzed if the formaldehyde applied during ChIP also induces poly(ADP-ribosyl)ation and its impact on chromatin composition. We observed massive polymer-formation in three different ChIP-protocols tested independent on the cell line. This was due to induction of DNA damage signaling as monitored by γH2AX formation. To abrogate poly(ADP-ribose) synthesis, we inhibited this enzymatic reaction either pharmacologically or by increased formaldehyde concentration. Both approaches changed ChIP-efficiency. Additionally, we detected specific differences in promoter-occupancy of tested transcription factors as well as the in the presence of histone H1 at the respective sites. In summary, we show here that standard ChIP is flawed by artificial formation of poly(ADP-ribose) and suppression of this enzymatic activity improves ChIP-efficiency in general. Also, we detected specific changes in promoter-occupancy dependent on poly(ADP-ribose). By preventing polymer synthesis with the proposed modifications in standard ChIP protocols it is now possible to analyze the natural chromatin-composition.


Asunto(s)
Inmunoprecipitación de Cromatina/métodos , Cromatina/metabolismo , Poli Adenosina Difosfato Ribosa/metabolismo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Células 3T3 , Animales , Línea Celular , Cromatina/efectos de los fármacos , Cromatina/genética , Daño del ADN , Fijadores/farmacología , Formaldehído/farmacología , Células HeLa , Histonas/metabolismo , Humanos , Ratones , Microscopía Fluorescente , Fenantrenos/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Reacción en Cadena de la Polimerasa , Unión Proteica/efectos de los fármacos
20.
Biochemistry ; 49(33): 7119-30, 2010 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-20669926

RESUMEN

Poly(ADP-ribose) polymerase-1 (PARP-1) is a molecular DNA damage sensor that catalyzes the synthesis of the complex biopolymer poly(ADP-ribose) (PAR) under consumption of NAD(+). PAR engages in fundamental cellular processes such as DNA metabolism and transcription and interacts noncovalently with specific binding proteins involved in DNA repair and regulation of chromatin structure. A factor implicated in DNA repair and chromatin organization is the DEK oncoprotein, an abundant and conserved constituent of metazoan chromatin, and the only member of its protein class. We have recently demonstrated that DEK, under stress conditions, is covalently modified with PAR by PARP-1, leading to a partial release of DEK into the cytoplasm. Additionally, we have also observed a noncovalent interaction between DEK and PAR, which we detail here. Using sequence alignment, we identify three functional PAR-binding sites in the DEK primary sequence and confirm their functionality in PAR binding studies. Furthermore, we show that the noncovalent binding to DEK is dependent on PAR chain length as revealed by an overlay blot technique and a PAR electrophoretic mobility shift assay. Intriguingly, DEK promotes the formation of a defined complex with a 54mer PAR (K(D) = 6 x 10(-8) M), whereas no specific interaction is detected with a short PAR chain (18mer). In stark contrast to covalent poly(ADP-ribosyl)ation of DEK, the noncovalent interaction does not affect the overall ability of DEK to bind to DNA. Instead the noncovalent interaction interferes with subsequent DNA-dependent multimerization activities of DEK, as seen in South-Western, electrophoretic mobility shift, topology, and aggregation assays. In particular, noncovalent attachment of PAR to DEK promotes the formation of DEK-DEK complexes by competing with DNA binding. This was seen by the reduced affinity of PAR-bound DEK for DNA templates in solution. Taken together, our findings deepen the molecular understanding of the DEK-PAR interplay and support the existence of a cellular "PAR code" represented by PAR chain length.


Asunto(s)
Proteínas Cromosómicas no Histona/metabolismo , Proteínas Oncogénicas/metabolismo , Poli Adenosina Difosfato Ribosa/metabolismo , Secuencia de Aminoácidos , Sitios de Unión , Proteínas Cromosómicas no Histona/química , ADN/metabolismo , Humanos , Datos de Secuencia Molecular , Proteínas Oncogénicas/química , Poli Adenosina Difosfato Ribosa/química , Proteínas de Unión a Poli-ADP-Ribosa , Multimerización de Proteína , Estructura Terciaria de Proteína , Alineación de Secuencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA