Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
Blood ; 142(25): 2175-2191, 2023 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-37756525

RESUMEN

ABSTRACT: Growth factor independence 1 (GFI1) is a DNA-binding transcription factor and a key regulator of hematopoiesis. GFI1-36N is a germ line variant, causing a change of serine (S) to asparagine (N) at position 36. We previously reported that the GFI1-36N allele has a prevalence of 10% to 15% among patients with acute myeloid leukemia (AML) and 5% to 7% among healthy Caucasians and promotes the development of this disease. Using a multiomics approach, we show here that GFI1-36N expression is associated with increased frequencies of chromosomal aberrations, mutational burden, and mutational signatures in both murine and human AML and impedes homologous recombination (HR)-directed DNA repair in leukemic cells. GFI1-36N exhibits impaired binding to N-Myc downstream-regulated gene 1 (Ndrg1) regulatory elements, causing decreased NDRG1 levels, which leads to a reduction of O6-methylguanine-DNA-methyltransferase (MGMT) expression levels, as illustrated by both transcriptome and proteome analyses. Targeting MGMT via temozolomide, a DNA alkylating drug, and HR via olaparib, a poly-ADP ribose polymerase 1 inhibitor, caused synthetic lethality in human and murine AML samples expressing GFI1-36N, whereas the effects were insignificant in nonmalignant GFI1-36S or GFI1-36N cells. In addition, mice that received transplantation with GFI1-36N leukemic cells treated with a combination of temozolomide and olaparib had significantly longer AML-free survival than mice that received transplantation with GFI1-36S leukemic cells. This suggests that reduced MGMT expression leaves GFI1-36N leukemic cells particularly vulnerable to DNA damage initiating chemotherapeutics. Our data provide critical insights into novel options to treat patients with AML carrying the GFI1-36N variant.


Asunto(s)
Proteínas de Unión al ADN , Leucemia Mieloide Aguda , Humanos , Ratones , Animales , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Temozolomida , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Daño del ADN , Reparación del ADN , Células Germinativas/metabolismo , ADN , Factores de Transcripción/genética
3.
Cancers (Basel) ; 15(15)2023 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-37568590

RESUMEN

Overcoming PARPi resistance is a high clinical priority. We established and characterized comparative in vitro models of acquired PARPi resistance, derived from either a BRCA1-proficient or BRCA1-deficient isogenic background by long-term exposure to olaparib. While parental cell lines already exhibited a certain level of intrinsic activity of multidrug resistance (MDR) proteins, resulting PARPi-resistant cells from both models further converted toward MDR. In both models, the PARPi-resistant phenotype was shaped by (i) cross-resistance to other PARPis (ii) impaired susceptibility toward the formation of DNA-platinum adducts upon exposure to cisplatin, which could be reverted by the drug efflux inhibitors verapamil or diphenhydramine, and (iii) reduced PARP-trapping activity. However, the signature and activity of ABC-transporter expression and the cross-resistance spectra to other chemotherapeutic drugs considerably diverged between the BRCA1-proficient vs. BRCA1-deficient models. Using dual-fluorescence co-culture experiments, we observed that PARPi-resistant cells had a competitive disadvantage over PARPi-sensitive cells in a drug-free medium. However, they rapidly gained clonal dominance under olaparib selection pressure, which could be mitigated by the MRP1 inhibitor MK-751. Conclusively, we present a well-characterized in vitro model, which could be instrumental in dissecting mechanisms of PARPi resistance from HR-proficient vs. HR-deficient background and in studying clonal dynamics of PARPi-resistant cells in response to experimental drugs, such as novel olaparib-sensitizers.

4.
Cancers (Basel) ; 13(24)2021 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-34944784

RESUMEN

To improve the treatment of pancreatic ductal adenocarcinoma (PDAC), a promising strategy consists of personalized chemotherapy based on gene expression profiles. Investigating a panel of PDAC-derived human cell lines, we found that their sensitivities towards cisplatin fall in two distinct classes. The platinum-sensitive class is characterized by the expression of GATA6, miRNA-200a, and miRNA-200b, which might be developable as predictive biomarkers. In the case of resistant PDAC cells, we identified a synergism of cisplatin with HSP90 inhibitors. Mechanistic explanations of this synergy include the degradation of Fanconi anemia pathway factors upon HSP90 inhibition. Treatment with the drug combination resulted in increased DNA damage and chromosome fragmentation, as we have reported previously for ovarian cancer cells. On top of this, HSP90 inhibition also enhanced the accumulation of DNA-bound platinum. We next investigated an orthotopic syngeneic animal model consisting of tumors arising from KPC cells (LSL-KrasG12D/+; LSL-Trp53R172H/+; Pdx-1-Cre, C57/BL6 genetic background). Here again, when treating established tumors, the combination of cisplatin with the HSP90 inhibitor onalespib was highly effective and almost completely prevented further tumor growth. We propose that the combination of platinum drugs and HSP90 inhibitors might be worth testing in the clinics for the treatment of cisplatin-resistant PDACs.

5.
Cancers (Basel) ; 13(3)2021 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-33530588

RESUMEN

The prognosis of late-stage epithelial ovarian cancer (EOC) patients is affected by chemotherapy response and the malignant potential of the tumor cells. In earlier work, we identified hypermethylation of the runt-related transcription factor 3 gene (RUNX3) as a prognostic biomarker and contrary functions of transcript variants (TV1 and TV2) in A2780 and SKOV3 cells. The aim of the study was to further validate these results and to increase the knowledge about RUNX3 function in EOC. New RUNX3 overexpression models of high-grade serous ovarian cancer (HGSOC) were established and analyzed for phenotypic (IC50 determination, migration, proliferation and angiogenesis assay, DNA damage analysis) and transcriptomic consequences (NGS) of RUNX3 TV1 and TV2 overexpression. Platinum sensitivity was affected by a specific transcript variant depending on BRCA background. RUNX3 TV2 induced an increased sensitivity in BRCA1wt cells (OVCAR3), whereas TV1 increased the sensitivity and induced a G2/M arrest under treatment in BRCA1mut cells (A13-2-12). These different phenotypes relate to differences in DNA repair: homologous recombination deficient A13-2-12 cells show less γH2AX foci despite higher levels of Pt-DNA adducts. RNA-Seq analyses prove transcript variant and cell-line-specific RUNX3 effects. Pathway analyses revealed another clinically important function of RUNX3-regulation of angiogenesis. This was confirmed by thrombospondin1 analyses, HUVEC spheroid sprouting assays and proteomic profiling. Importantly, conditioned media (CM) from RUNX3 TV1 overexpressing A13-2-12 cells induced an increased HUVEC sprouting. Altogether, the presented data support the hypothesis of different functions of RUNX3 transcript variants related to the clinically relevant processes-platinum resistance and angiogenesis.

6.
Histochem Cell Biol ; 153(5): 367-377, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32125512

RESUMEN

A major limitation in the pharmacological treatment of clinically detectable primary cancers and their metastases is their limited accessibility to anti-cancer drugs (cytostatics, inhibitory antibodies, small-molecule inhibitors) critically impairing therapeutic efficacies. Investigations on the tissue distribution of such drugs are rare and have only been based on fresh frozen material or methanol-fixed cell culture cells so far. In this paper, we expand the detection of cisplatin-induced DNA adducts and anthracyclines as well as therapeutic antibodies to routinely prepared formalin-fixed, paraffin-embedded sections (FFPE). Using pre-treated cell lines prepared as FFPE samples comparable to tissues from routine analysis, we demonstrate that our method allows for the detection of chemotherapeutics (anthracyclines by autofluorescence, cisplatin by immune detection of DNA adducts) as well as therapeutic antibodies. This methodology thus allows for analyzing archival FFPE tissues, as demonstrated here for the detection of cisplatin, doxorubicin and trastuzumab in FFPE sections of tumor xenografts from drug-treated mice. Analyzing human tumor samples, this will lead to new insights into the tissue penetration of drugs.


Asunto(s)
Antineoplásicos/análisis , Cetuximab/análisis , Cisplatino/análisis , Doxorrubicina/análisis , Neoplasias/patología , Adhesión en Parafina , Rituximab/análisis , Trastuzumab/análisis , Antineoplásicos/uso terapéutico , Cetuximab/uso terapéutico , Cisplatino/uso terapéutico , Doxorrubicina/uso terapéutico , Formaldehído/química , Humanos , Neoplasias/tratamiento farmacológico , Rituximab/uso terapéutico , Fijación del Tejido , Trastuzumab/uso terapéutico , Células Tumorales Cultivadas
7.
Mol Oncol ; 14(4): 686-703, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32037720

RESUMEN

Platinum-based compounds remain a well-established chemotherapy for cancer treatment despite their adverse effects which substantially restrict the therapeutic windows of the drugs. Both the cell type-specific toxicity and the clinical responsiveness of tumors have been associated with mechanisms that alter drug entry and export. We sought to identify pharmacological agents that promote cisplatin (CP) efficacy by augmenting the levels of drug-induced DNA lesions in malignant cells and simultaneously protecting normal tissues from accumulating such damage and from functional loss. Formation and persistence of platination products in the DNA of individual nuclei were measured in drug-exposed cell lines, in primary human tumor cells and in tissue sections using an immunocytochemical method. Using a mouse model of CP-induced toxicity, the antihistaminic drug diphenhydramine (DIPH) and two methylated derivatives decreased DNA platination in normal tissues and also ameliorated nephrotoxicity, ototoxicity, and neurotoxicity. In addition, DIPH sensitized multiple cancer cell types, particularly ovarian cancer cells, to CP by increasing intracellular uptake, DNA platination, and/or apoptosis in cell lines and in patient-derived primary tumor cells. Mechanistically, DIPH diminished transport capacity of CP efflux pumps MRP2, MRP3, and MRP5 particularly in its C2+C6 bimethylated form. Overall, we demonstrate that DIPH reduces side effects of platinum-based chemotherapy and simultaneously inhibits key mechanisms of platinum resistance. We propose that measuring DNA platination after ex vivo exposure may predict the responsiveness of individual tumors to DIPH-like modulators.


Asunto(s)
Antineoplásicos/farmacología , Cisplatino/farmacología , Difenhidramina/farmacología , Antagonistas de los Receptores Histamínicos H1/farmacología , Neoplasias Ováricas/tratamiento farmacológico , Animales , Antineoplásicos/toxicidad , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Cisplatino/toxicidad , Aductos de ADN/metabolismo , Resistencia a Antineoplásicos/efectos de los fármacos , Sinergismo Farmacológico , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Modelos Moleculares , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología
8.
Sci Rep ; 9(1): 1282, 2019 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-30718758

RESUMEN

Cisplatin is an extensively used chemotherapeutic drug for lung cancer, but the development of resistance decreases its effectiveness in the treatments of non-small cell lung cancer (NSCLC). In this study, we examined the effects of metformin, a widely used antidiabetic drug, on cisplatin radiosensitization in NSCLC cell lines. Human NSCLC cell lines, A549 (cisplatin-resistant) and H460 (cisplatin-sensitive), were treated with metformin, cisplatin or a combination of both drugs before ionizing radiation. Cell proliferation, clonogenic assays, western blotting, cisplatin-DNA adduct formation and immunocytochemistry were used to characterize the treatments effects. Metformin increased the radiosensitivity of NSCLC cells. Metformin showed additive and over-additive effects in combination with cisplatin and the radiation response in the clonogenic assay in H460 and A549 cell lines (p = 0.018 for the interaction effect between cisplatin and metformin), respectively. At the molecular level, metformin led to a significant increase in cisplatin-DNA adduct formation compared with cisplatin alone (p < 0.01, ANOVA-F test). This was accompanied by a decreased expression of the excision repair cross-complementation 1 expression (ERCC1), a key enzyme in nucleotide excision repair pathway. Furthermore, compared with each treatment alone metformin in combination with cisplatin yielded the lowest level of radiation-induced Rad51 foci, an essential protein of homologous recombination repair. Ionizing radiation-induced γ-H2AX and 53BP1 foci persisted longer in both cell lines in the presence of metformin. Pharmacological inhibition of AMP-activated protein kinase (AMPK) demonstrated that metformin enhances the radiosensitizing effect of cisplatin through an AMPK-dependent pathway only in H460 but not in A549 cells. Our results suggest that metformin can enhance the effect of combined cisplatin and radiotherapy in NSCLC and can sensitize these cells to radiation that are not sensitized by cisplatin alone.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Cisplatino/farmacología , Regulación Neoplásica de la Expresión Génica , Neoplasias Pulmonares , Metformina/farmacología , Proteínas de Neoplasias/biosíntesis , Tolerancia a Radiación/efectos de los fármacos , Células A549 , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Carcinoma de Pulmón de Células no Pequeñas/radioterapia , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de la radiación , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Neoplasias Pulmonares/radioterapia
9.
Oncogene ; 38(19): 3616-3635, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30651597

RESUMEN

The melanocytic lineage, which is prominently exposed to ultraviolet radiation (UVR) and radiation-independent oxidative damage, requires specific DNA-damage response mechanisms to maintain genomic and transcriptional homeostasis. The coordinate lineage-specific regulation of intricately intertwined DNA repair and transcription is incompletely understood. Here we demonstrate that the Microphthalmia-associated transcription factor (MITF) directly controls general transcription and UVR-induced nucleotide excision repair by transactivation of GTF2H1 as a core element of TFIIH. Thus, MITF ensures the rapid resumption of transcription after completion of strand repair and maintains transcriptional output, which is indispensable for survival of the melanocytic lineage including melanoma in vitro and in vivo. Moreover, MITF controls c-MYC implicated in general transcription by transactivation of far upstream binding protein 2 (FUBP2/KSHRP), which induces c-MYC pulse regulation through TFIIH, and experimental depletion of MITF results in consecutive loss of CDK7 in the TFIIH-CAK subcomplex. Targeted for proteasomal degradation, CDK7 is dependent on transactivation by MITF or c-MYC to maintain a steady state. The dependence of TFIIH-CAK on sequence-specific MITF and c-MYC constitutes a previously unrecognized mechanism feeding into super-enhancer-driven or other oncogenic transcriptional circuitries, which supports the concept of a transcription-directed therapeutic intervention in melanoma.


Asunto(s)
Reparación del ADN/fisiología , Factor de Transcripción Asociado a Microftalmía/metabolismo , Fosfoproteínas/metabolismo , Factor de Transcripción TFIIH/metabolismo , Factores de Transcripción TFII/metabolismo , Animales , Células Cultivadas , Reparación del ADN/efectos de la radiación , Receptor con Dominio Discoidina 1/genética , Receptor con Dominio Discoidina 1/metabolismo , Femenino , Genes myc , Humanos , Melanocitos/fisiología , Melanocitos/efectos de la radiación , Melanoma/metabolismo , Melanoma/patología , Ratones SCID , Factor de Transcripción Asociado a Microftalmía/genética , Fosfoproteínas/genética , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Factor de Transcripción TFIIH/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Factores de Transcripción TFII/genética , Transcripción Genética , Rayos Ultravioleta
10.
Methods Mol Biol ; 1655: 351-358, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-28889396

RESUMEN

Immunocytological staining with adduct-specific antibodies allows the visualization and measurement of structurally defined types of DNA damage in the nuclei of individual cells. Here we describe an immunocytological assay (ICA) procedure for the localization and quantification of such damage, in particular induced by platinum-based anticancer drugs, in cell lines , in primary cell suspensions and in frozen tissue sections.


Asunto(s)
Aductos de ADN , Daño del ADN , Inmunohistoquímica/métodos , Platino (Metal) , Análisis de la Célula Individual , Núcleo Celular , Técnica del Anticuerpo Fluorescente , Humanos , Procesamiento de Imagen Asistido por Computador , Microscopía Confocal , Microscopía Fluorescente , Platino (Metal)/química , Análisis de la Célula Individual/métodos
11.
Mol Pain ; 13: 1744806917746565, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29166837

RESUMEN

Platinum-based chemotherapeutic agents, such as cisplatin, are still frequently used for treating various types of cancer. Besides its high effectiveness, cisplatin has several serious side effects. One of the most common side effects is dorsal root ganglion (DRG) neurotoxicity. However, the mechanisms underlying this neurotoxicity are still unclear and controversially discussed. Cisplatin-mediated modulation of voltage-gated calcium channels (VGCCs) in the DRG neurons has been shown to alter intracellular calcium homeostasis, a process critical for the induction of neurotoxicity. Using the whole-cell patch-clamp technique, immunostaining, behavioural experiments and electron microscopy (EM) of rat DRGs, we here demonstrate that cisplatin-induced neurotoxicity is due to functional alteration of VGCC, but not due to morphological damage. In vitro application of cisplatin (0.5 µM) increased N-type VGCC currents ( ICa(V)) in small DRG neurons. Repetitive in vivo administration of cisplatin (1.5 mg/kg, cumulative 12 mg/kg) increased the protein level of N-type VGCC over 26 days, with the protein level being increased for at least 14 days after the final cisplatin administration. Behavioural studies revealed that N-type VGCCs are crucial for inducing symptoms of cisplatin-related neuropathic pain, such as thermal and mechanical hyperalgesia. EM and histology showed no evidence of any structural damage, apoptosis or necrosis in DRG cells after cisplatin exposure for 26 days. Furthermore, no nuclear DNA damage in sensory neurons was observed. Here, we provide evidence for a mainly functionally driven induction of neuropathic pain by cisplatin.


Asunto(s)
Canales de Calcio Tipo N/metabolismo , Cisplatino/farmacología , Activación del Canal Iónico/efectos de los fármacos , Células Receptoras Sensoriales/metabolismo , Células Receptoras Sensoriales/patología , Animales , Apoptosis/efectos de los fármacos , Conducta Animal , Cisplatino/administración & dosificación , Daño del ADN , Femenino , Ganglios Espinales/efectos de los fármacos , Ganglios Espinales/metabolismo , Ganglios Espinales/patología , Ganglios Espinales/ultraestructura , Masculino , Neuralgia/complicaciones , Neuralgia/patología , Ratas Wistar , Células Receptoras Sensoriales/efectos de los fármacos , Células Receptoras Sensoriales/ultraestructura
12.
Mol Pharm ; 14(12): 4454-4461, 2017 12 04.
Artículo en Inglés | MEDLINE | ID: mdl-29129076

RESUMEN

Despite their immense and rapidly increasing importance as analytical tools or therapeutic drugs, the detailed structural features of particular monoclonal antibodies are widely unknown. Here, an antibody already in use for diagnostic purposes and for molecular dosimetry studies in cancer therapy with very high affinity and specificity for cisplatin-induced DNA modifications was studied extensively. The molecular structure and modifications as well as the antigen specificity were investigated mainly by mass spectrometry. Using nano electrospray ionization mass spectrometry, it was possible to characterize the antibody in its native state. Tandem-MS experiments not only revealed specific fragments but also gave information on the molecular structure. The detailed primary structure was further elucidated by proteolytic treatment with a selection of enzymes and high resolution tandem-MS. The data were validated by comparison with known antibody sequences. Then, the complex glycan structures bound to the antibody were characterized in all detail. The Fc-bound oligosaccharides were released enzymatically and studied by matrix-assisted laser desorption/ionization mass spectrometry. Overall 16 different major glycan structures were identified. The binding specificity of the antibody was investigated by applying synthetic single and double stranded DNA oligomers harboring distinct Pt adducts. The antibody-antigen complexes were analyzed by mass spectrometry under native conditions. The stability of the complex with double stranded DNA was also investigated.


Asunto(s)
Anticuerpos Monoclonales/química , Complejo Antígeno-Anticuerpo/química , Aductos de ADN/inmunología , Anticuerpos Monoclonales/inmunología , Cisplatino/farmacología , Aductos de ADN/efectos de los fármacos , Epítopos/efectos de los fármacos , Epítopos/inmunología , Estructura Molecular , Oligosacáridos/química , Polisacáridos/química , Espectrometría de Masa por Ionización de Electrospray , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Espectrometría de Masas en Tándem
13.
Food Chem Toxicol ; 107(Pt A): 226-236, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28669851

RESUMEN

Strategies to minimize the nephrotoxicity of platinated antineoplastics without affecting its antitumour efficacy are strongly necessary to improve the pharmacotoxicological profile of these drugs. The natural flavonoid quercetin has been shown to afford nephroprotection without affecting cisplatin antitumour effect. The purpose of the present study has been to assess the differential mechanisms of action of cisplatin and quercetin on kidney and tumour tissues that could explain these effects. Wistar rats bearing subcutaneous tumours were treated with cisplatin and quercetin (and the appropriate controls). Tumour size and renal function evolution was monitored during 6 days. Platinum and quercetin content were also determined in both tissues. All the parameters studied, including blood supply, inflammation, apoptosis, critical MAPK signaling and oxidative stress in the cisplatin-treated animals are almost normalized by quercetin in the kidneys, but unaffected in the tumours. Our results suggest that in a cancer model in vivo, the protection exerted by quercetin on cisplatin nephrotoxicity is related to its antioxidant, vascular, anti-inflammatory and antiapoptotic effects, but these properties do not affect the mechanisms responsible for the antitumour effect of cisplatin.


Asunto(s)
Antineoplásicos/toxicidad , Cisplatino/toxicidad , Neoplasias Renales/tratamiento farmacológico , Quercetina/administración & dosificación , Animales , Antineoplásicos/administración & dosificación , Antioxidantes/administración & dosificación , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Cisplatino/administración & dosificación , Humanos , Riñón/efectos de los fármacos , Riñón/metabolismo , Riñón/fisiopatología , Neoplasias Renales/metabolismo , Neoplasias Renales/fisiopatología , Masculino , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Endogámicas F344 , Ratas Wistar
14.
Int J Cancer ; 141(8): 1600-1614, 2017 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-28670762

RESUMEN

Aim was to identify methylated genes with functional involvement in cisplatin-resistance development of epithelial ovarian cancer (EOC). Genome-wide analyses of hypermethylated CpG-islands in resistant cell lines in combination with qRT-PCR analyses were used to identify epigenetically silenced genes. EOC-Type-II tumors were analyzed for gene methylation and expression and TCGA data were interrogated in-silico. Experiments revealed 37 commonly hypermethylated genes in resistant cells of which Tribbles 2 (TRIB2) showed the most pronounced downregulation on mRNA level and was characterized further. TRIB2 showed a reactivation after 5'-Aza-Cytidine treatment in resistant cells but a cisplatin-dependent, prominent upregulation on mRNA level in sensitive cells, only. Re-expression in resistant A2780 cells increased the sensitivity to cisplatin and other DNA-damaging agents, but not taxanes. Contrary, knockdown of TRIB2 increased resistance to cisplatin in sensitive cells. TRIB2 was involved in the induction of a cisplatin-dependent cell cycle arrest and apoptosis by influencing p21 and survivin expression. An increased Pt-DNA-adduct formation in TRIB2 re-expressing cells did not translate in higher levels of dsDNA damage (yH2AX-foci). Thus, TRIB2 is potentially involved in the signal transduction from nucleotide excision repair of intrastrand cross links. Importantly, patient stratification of two homogenous cohorts of EOC-Type-II patients from Jena (n = 38) and the TCGA (n = 149) by TRIB2 mRNA expression consistently revealed a significantly decreased PFS for patients with low TRIB2 levels (log-rank p < 0.05). Tumors from resistant patients expressed the lowest levels of TRIB2. Downregulation of TRIB2 contributes to platin-resistance and TRIB2 expression should be validated as prognostic and predictive marker for EOC.


Asunto(s)
Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Cisplatino/farmacología , Daño del ADN , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neoplasias Glandulares y Epiteliales/tratamiento farmacológico , Neoplasias Glandulares y Epiteliales/genética , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/genética , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Proteínas Quinasas Dependientes de Calcio-Calmodulina/biosíntesis , Proteínas Quinasas Dependientes de Calcio-Calmodulina/genética , Carcinoma Epitelial de Ovario , Línea Celular Tumoral , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/biosíntesis , Aductos de ADN/biosíntesis , Metilación de ADN , Resistencia a Antineoplásicos/genética , Femenino , Fase G2 , Humanos , Péptidos y Proteínas de Señalización Intracelular/biosíntesis , Péptidos y Proteínas de Señalización Intracelular/genética , Puntos de Control de la Fase M del Ciclo Celular , Neoplasias Glandulares y Epiteliales/metabolismo , Neoplasias Ováricas/metabolismo , Proteoma/metabolismo , Células Tumorales Cultivadas
15.
Exp Neurol ; 288: 62-74, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27823926

RESUMEN

Cisplatin is important in the treatment of various types of cancer. Although it is highly effective, it also has severe side effects, with neurotoxicity in dorsal root ganglion (DRG) neurons being one of the most common. The key mechanisms of neurotoxicity are still controversially discussed; however, disturbances of the calcium homeostasis in DRG neurons have been suggested to mediate cisplatin neurotoxicity. By using the whole-cell patch-clamp technique, immunostaining and behavioral experiments with Sprague-Dawley rats, we examined the influence of short- and long-term exposure to cisplatin on voltage-gated calcium channel (VGCC) currents (ICa(V)) in small DRG neurons. In vitro exposure to cisplatin reduced ICa(V) in a concentration-dependent manner (0.01-50µM; 13.8-77.3%; IC50 5.07µM). Subtype-specific measurements of VGCCs showed differential effects on ICa(V). While the ICa(V) of P/Q-, L- and T-type VGCCs were reduced, ICa(V) of N-type VGCCs were increased by 30.3% during depolarization to 0mV. Exposure of DRG neurons to cisplatin (0.5 or 5µM) for 24-48h in vitro significantly increased a CaMK II-mediated ICa(V) current density. Immunostaining and western blot analysis revealed an increase of N-type VGCC protein level in DRG neurons 24h after cisplatin exposure. Cisplatin-mediated activation of caspase-3 was prevented by inhibition of N-type VGCCs using Ɯ-conotoxin MVIIA. Behavioral experiments showed that Ɯ-conotoxin MVIIA treatment prevented neuropathic syndromes in vivo by inhibiting upregulation of the N-type protein level. Here we show evidence for the first time for a crucial role of N-type VGCC in the genesis of cisplatin-induced polyneuropathy.


Asunto(s)
Canales de Calcio Tipo N/metabolismo , Cisplatino/farmacología , Ganglios Espinales/citología , Neuralgia/inducido químicamente , Células Receptoras Sensoriales/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos , Animales , Antineoplásicos/farmacología , Bencilaminas/farmacología , Bloqueadores de los Canales de Calcio/farmacología , Canales de Calcio Tipo N/genética , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Masculino , Potenciales de la Membrana/efectos de los fármacos , Actividad Motora/efectos de los fármacos , Neuralgia/fisiopatología , Dimensión del Dolor , Ratas , Ratas Sprague-Dawley , Ratas Wistar , Células Receptoras Sensoriales/metabolismo , Sulfonamidas/farmacología , Factores de Tiempo
16.
Mol Cancer Res ; 14(11): 1110-1123, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27514406

RESUMEN

KRAS-mutant lung adenocarcinoma is among the most common cancer entities and, in advanced stages, typically displays poor prognosis due to acquired resistance against chemotherapy, which is still largely based on cisplatin-containing combination regimens. Mechanisms of cisplatin resistance have been extensively investigated, and ERCC1 has emerged as a key player due to its central role in the repair of cisplatin-induced DNA lesions. However, clinical data have not unequivocally confirmed ERCC1 status as a predictor of the response to cisplatin treatment. Therefore, we employed an autochthonous mouse model of Kras-driven lung adenocarcinoma resembling human lung adenocarcinoma to investigate the role of Ercc1 in the response to cisplatin treatment. Our data show that Ercc1 deficiency in Tp53-deficient murine lung adenocarcinoma induces a more aggressive tumor phenotype that displays enhanced sensitivity to cisplatin treatment. Furthermore, tumors that relapsed after cisplatin treatment in our model develop a robust etoposide sensitivity that is independent of the Ercc1 status and depends solely on previous cisplatin exposure. Our results provide a solid rationale for further investigation of the possibility of preselection of lung adenocarcinoma patients according to the functional ERCC1- and mutational TP53 status, where functionally ERCC1-incompetent patients might benefit from sequential cisplatin and etoposide chemotherapy. IMPLICATIONS: This study provides a solid rationale for the stratification of lung adenocarcinoma patients according to the functional ERCC1- and mutational TP53 status, where functionally ERCC1-incompetent patients could benefit from sequential cisplatin and etoposide chemotherapy. Mol Cancer Res; 14(11); 1110-23. ©2016 AACR.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Cisplatino/administración & dosificación , Proteínas de Unión al ADN/deficiencia , Endonucleasas/deficiencia , Neoplasias Pulmonares/tratamiento farmacológico , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteína p53 Supresora de Tumor/genética , Adenocarcinoma/genética , Adenocarcinoma del Pulmón , Animales , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Cisplatino/farmacología , Resistencia a Antineoplásicos , Etopósido/administración & dosificación , Etopósido/farmacología , Humanos , Neoplasias Pulmonares/genética , Ratones , Mutación , Medicina de Precisión , Células Tumorales Cultivadas
17.
Proc Natl Acad Sci U S A ; 113(30): E4311-9, 2016 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-27407148

RESUMEN

DNA damage tolerance facilitates the progression of replication forks that have encountered obstacles on the template strands. It involves either translesion DNA synthesis initiated by proliferating cell nuclear antigen monoubiquitination or less well-characterized fork reversal and template switch mechanisms. Herein, we characterize a novel tolerance pathway requiring the tumor suppressor p53, the translesion polymerase ι (POLι), the ubiquitin ligase Rad5-related helicase-like transcription factor (HLTF), and the SWI/SNF catalytic subunit (SNF2) translocase zinc finger ran-binding domain containing 3 (ZRANB3). This novel p53 activity is lost in the exonuclease-deficient but transcriptionally active p53(H115N) mutant. Wild-type p53, but not p53(H115N), associates with POLι in vivo. Strikingly, the concerted action of p53 and POLι decelerates nascent DNA elongation and promotes HLTF/ZRANB3-dependent recombination during unperturbed DNA replication. Particularly after cross-linker-induced replication stress, p53 and POLι also act together to promote meiotic recombination enzyme 11 (MRE11)-dependent accumulation of (phospho-)replication protein A (RPA)-coated ssDNA. These results implicate a direct role of p53 in the processing of replication forks encountering obstacles on the template strand. Our findings define an unprecedented function of p53 and POLι in the DNA damage response to endogenous or exogenous replication stress.


Asunto(s)
Daño del ADN , Replicación del ADN , ADN Polimerasa Dirigida por ADN/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Línea Celular Tumoral , Células Cultivadas , ADN/química , ADN/genética , ADN/metabolismo , ADN Helicasas/genética , ADN Helicasas/metabolismo , Reparación del ADN , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , ADN Polimerasa Dirigida por ADN/genética , Recombinación Homóloga , Humanos , Células K562 , Conformación de Ácido Nucleico , Interferencia de ARN , Proteína de Replicación A/genética , Proteína de Replicación A/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteína p53 Supresora de Tumor/genética , ADN Polimerasa iota
18.
Nucleic Acids Res ; 44(7): 3204-18, 2016 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-26819410

RESUMEN

TP63, a member of the p53 gene family gene, encodes the ΔNp63 protein and is one of the most frequently amplified genes in squamous cell carcinomas (SCC) of the head and neck (HNSCC) and lungs (LUSC). Using an epiallelic series of siRNAs with intrinsically different knockdown abilities, we show that the complete loss of ΔNp63 strongly impaired cell proliferation, whereas partial ΔNp63 depletion rendered cells hypersensitive to cisplatin accompanied by an accumulation of DNA damage. Expression profiling revealed wide-spread transcriptional regulation of DNA repair genes and in particular Fanconi anemia (FA) pathway components such as FANCD2 and RAD18 - known to be crucial for the repair of cisplatin-induced interstrand crosslinks. In SCC patients ΔNp63 levels significantly correlate with FANCD2 and RAD18 expression confirming ΔNp63 as a key activator of the FA pathway in vivo Mechanistically, ΔNp63 bound an upstream enhancer of FANCD2 inactive in primary keratinocytes but aberrantly activated by ΔNp63 in SCC. Consistently, depletion of FANCD2 sensitized to cisplatin similar to depletion of ΔNp63. Together, our results demonstrate that ΔNp63 directly activates the FA pathway in SCC and limits the efficacy of cisplatin treatment. Targeting ΔNp63 therefore would not only inhibit SCC proliferation but also sensitize tumors to chemotherapy.


Asunto(s)
Antineoplásicos/uso terapéutico , Carcinoma de Células Escamosas/genética , Cisplatino/uso terapéutico , Reparación del ADN , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Células Cultivadas , Proteínas de Unión al ADN/metabolismo , Resistencia a Antineoplásicos , Elementos de Facilitación Genéticos , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/genética , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/metabolismo , Humanos , Factores de Transcripción/fisiología , Activación Transcripcional , Proteínas Supresoras de Tumor/fisiología , Ubiquitina-Proteína Ligasas/metabolismo
19.
Toxicol Appl Pharmacol ; 292: 103-14, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26739623

RESUMEN

The platinating agent cisplatin (CisPt) is commonly used in the therapy of various types of solid tumors. The anticancer efficacy of CisPt largely depends on the formation of bivalent DNA intrastrand crosslinks, which stimulate mechanisms of the DNA damage response (DDR), thereby triggering checkpoint activation, gene expression and cell death. The clinically most relevant adverse effect associated with CisPt treatment is nephrotoxicity that results from damage to renal tubular epithelial cells. Here, we addressed the question whether the HMG-CoA-reductase inhibitor lovastatin affects the DDR of renal cells by employing rat renal proximal tubular epithelial (NRK-52E) cells as in vitro model. The data show that lovastatin has extensive inhibitory effects on CisPt-stimulated DDR of NRK-52E cells as reflected on the levels of phosphorylated ATM, Chk1, Chk2, p53 and Kap1. Mitigation of CisPt-induced DDR by lovastatin was independent of the formation of DNA damage as demonstrated by (i) the analysis of Pt-(GpG) intrastrand crosslink formation by Southwestern blot analyses and (ii) the generation of DNA strand breaks as analyzed on the level of nuclear γH2AX foci and employing the alkaline comet assay. Lovastatin protected NRK-52E cells from the cytotoxicity of high CisPt doses as shown by measuring cell viability, cellular impedance and flow cytometry-based analyses of cell death. Importantly, the statin also reduced the level of kidney DNA damage and apoptosis triggered by CisPt treatment of mice. The data show that the lipid-lowering drug lovastatin extensively counteracts pro-apoptotic signal mechanisms of the DDR of tubular epithelial cells following CisPt injury.


Asunto(s)
Apoptosis/efectos de los fármacos , Cisplatino/toxicidad , Daño del ADN/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Túbulos Renales/efectos de los fármacos , Lovastatina/farmacología , Animales , Apoptosis/fisiología , Línea Celular , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Cisplatino/antagonistas & inhibidores , Daño del ADN/fisiología , Células Epiteliales/metabolismo , Túbulos Renales/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratas
20.
Biochim Biophys Acta ; 1853(3): 685-98, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25565603

RESUMEN

BACKGROUND: Platinum compounds are potent anticancer drugs but also evoke considerable normal tissue damage. Here, we elucidate the molecular mechanisms contributing to the nephrotoxic effects of cisplatin. METHODS: We comparatively investigated the stress responses of rat kidney tubular (NRK-52E) and glomerular cells (RGE) following treatment with cisplatin (CisPt), oxaliplatin (OxaliPt) and carboplatin (CarboPt). To this end, cell viability, apoptosis, cell cycle progression, DNA damage response (DDR) and repair of DNA adducts were investigated. RESULTS: CisPt reduced the viability of tubular NRK-52E and glomerular RGE cells most efficiently. Cytotoxicity evoked by CarboPt occurred with a delay, which might be related to a retarded formation of Pt-(GpG) intrastrand crosslinks. RGE cells were more sensitive towards all platinum compounds than NRK-52E cells. Platinum drugs efficiently induced caspase-mediated apoptosis in tubular cells, while RGE cells favored G2/M arrest when treated with equitoxic platinum doses. Mitotic index of NKR-52E and RGE cells was worst affected by OxaliPt. Activation of the DDR was strikingly agent- and cell type-specific. Most comprehensive and substantial stimulation of DDR mechanisms was provoked by CisPt. Repair of Pt-(GpG) intrastrand crosslinks was best in RGE, which was reflected by high mRNA expression of nucleotide excision repair (NER) factors. CONCLUSIONS: There are substantial differences regarding the cause of sensitivity and mechanisms of DDR between tubular and glomerular cells following platinum injury. CisPt is the most potent stimulator of the DDR. We hypothesize that specific DNA adducts and thereby forcefully activated pro-toxic DDR mechanisms contribute to the exceptionally high acute nephrotoxicity of CisPt.


Asunto(s)
Antineoplásicos/toxicidad , Cisplatino/toxicidad , Daño del ADN , Glomérulos Renales/efectos de los fármacos , Túbulos Renales/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Células Cultivadas , Aductos de ADN/efectos de los fármacos , Aductos de ADN/metabolismo , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Glomérulos Renales/citología , Glomérulos Renales/metabolismo , Túbulos Renales/citología , Túbulos Renales/metabolismo , Compuestos Organoplatinos/toxicidad , Oxaliplatino , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...