Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nature ; 626(7998): 401-410, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38297129

RESUMO

Ferroptosis is a form of cell death that has received considerable attention not only as a means to eradicate defined tumour entities but also because it provides unforeseen insights into the metabolic adaptation that tumours exploit to counteract phospholipid oxidation1,2. Here, we identify proferroptotic activity of 7-dehydrocholesterol reductase (DHCR7) and an unexpected prosurvival function of its substrate, 7-dehydrocholesterol (7-DHC). Although previous studies suggested that high concentrations of 7-DHC are cytotoxic to developing neurons by favouring lipid peroxidation3, we now show that 7-DHC accumulation confers a robust prosurvival function in cancer cells. Because of its far superior reactivity towards peroxyl radicals, 7-DHC effectively shields (phospho)lipids from autoxidation and subsequent fragmentation. We provide validation in neuroblastoma and Burkitt's lymphoma xenografts where we demonstrate that the accumulation of 7-DHC is capable of inducing a shift towards a ferroptosis-resistant state in these tumours ultimately resulting in a more aggressive phenotype. Conclusively, our findings provide compelling evidence of a yet-unrecognized antiferroptotic activity of 7-DHC as a cell-intrinsic mechanism that could be exploited by cancer cells to escape ferroptosis.


Assuntos
Linfoma de Burkitt , Desidrocolesteróis , Ferroptose , Neuroblastoma , Animais , Humanos , Linfoma de Burkitt/metabolismo , Linfoma de Burkitt/patologia , Sobrevivência Celular , Desidrocolesteróis/metabolismo , Peroxidação de Lipídeos , Transplante de Neoplasias , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Oxirredução , Fenótipo , Reprodutibilidade dos Testes
2.
EMBO Mol Med ; 15(8): e18014, 2023 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-37435859

RESUMO

Ferroptosis has emerged as an attractive strategy in cancer therapy. Understanding the operational networks regulating ferroptosis may unravel vulnerabilities that could be harnessed for therapeutic benefit. Using CRISPR-activation screens in ferroptosis hypersensitive cells, we identify the selenoprotein P (SELENOP) receptor, LRP8, as a key determinant protecting MYCN-amplified neuroblastoma cells from ferroptosis. Genetic deletion of LRP8 leads to ferroptosis as a result of an insufficient supply of selenocysteine, which is required for the translation of the antiferroptotic selenoprotein GPX4. This dependency is caused by low expression of alternative selenium uptake pathways such as system Xc- . The identification of LRP8 as a specific vulnerability of MYCN-amplified neuroblastoma cells was confirmed in constitutive and inducible LRP8 knockout orthotopic xenografts. These findings disclose a yet-unaccounted mechanism of selective ferroptosis induction that might be explored as a therapeutic strategy for high-risk neuroblastoma and potentially other MYCN-amplified entities.


Assuntos
Ferroptose , Neuroblastoma , Humanos , Linhagem Celular Tumoral , Proteína Proto-Oncogênica N-Myc/genética , Proteína Proto-Oncogênica N-Myc/metabolismo , Neuroblastoma/genética , Neuroblastoma/tratamento farmacológico , Selenocisteína/uso terapêutico , Animais
3.
Redox Biol ; 62: 102639, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36958250

RESUMO

Despite a strong rationale for why cancer cells are susceptible to redox-targeting drugs, such drugs often face tumor resistance or dose-limiting toxicity in preclinical and clinical studies. An important reason is the lack of specific biomarkers to better select susceptible cancer entities and stratify patients. Using a large panel of lung cancer cell lines, we identified a set of "antioxidant-capacity" biomarkers (ACB), which were tightly repressed, partly by STAT3 and STAT5A/B in sensitive cells, rendering them susceptible to multiple redox-targeting and ferroptosis-inducing drugs. Contrary to expectation, constitutively low ACB expression was not associated with an increased steady state level of reactive oxygen species (ROS) but a high level of nitric oxide, which is required to sustain high replication rates. Using ACBs, we identified cancer entities with a high percentage of patients with favorable ACB expression pattern, making it likely that more responders to ROS-inducing drugs could be stratified for clinical trials.


Assuntos
Antioxidantes , Neoplasias Pulmonares , Humanos , Espécies Reativas de Oxigênio/metabolismo , Antioxidantes/metabolismo , Neoplasias Pulmonares/metabolismo , Oxirredução , Biomarcadores/metabolismo
4.
Nat Cancer ; 3(4): 471-485, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35484422

RESUMO

Aberrant expression of MYC transcription factor family members predicts poor clinical outcome in many human cancers. Oncogenic MYC profoundly alters metabolism and mediates an antioxidant response to maintain redox balance. Here we show that MYCN induces massive lipid peroxidation on depletion of cysteine, the rate-limiting amino acid for glutathione (GSH) biosynthesis, and sensitizes cells to ferroptosis, an oxidative, non-apoptotic and iron-dependent type of cell death. The high cysteine demand of MYCN-amplified childhood neuroblastoma is met by uptake and transsulfuration. When uptake is limited, cysteine usage for protein synthesis is maintained at the expense of GSH triggering ferroptosis and potentially contributing to spontaneous tumor regression in low-risk neuroblastomas. Pharmacological inhibition of both cystine uptake and transsulfuration combined with GPX4 inactivation resulted in tumor remission in an orthotopic MYCN-amplified neuroblastoma model. These findings provide a proof of concept of combining multiple ferroptosis targets as a promising therapeutic strategy for aggressive MYCN-amplified tumors.


Assuntos
Ferroptose , Neuroblastoma , Morte Celular , Criança , Cisteína/uso terapêutico , Ferroptose/genética , Glutationa/uso terapêutico , Humanos , Proteína Proto-Oncogênica N-Myc/genética , Neuroblastoma/genética
5.
Adv Exp Med Biol ; 1301: 1-5, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34370284

RESUMO

Ferroptosis is a newly discovered form of cell death that is rapidly becoming associated to a variety of diseases and explaining their pathological mechanisms. This book addresses new emerging topics in the field of ferroptosis, with special attention to diseases more recently explained through ferroptotic mechanisms, including infectious diseases and neurodegeneration. In this chapter, we will provide the readers with an introduction to the concepts and pathways involved in ferroptosis to further move into a more detailed exposition of the topics advertised in this book. In special, we aim for this book to broaden the perspectives on how ferroptosis is regulated and connected to human diseases and motivate new studies in this emerging field.


Assuntos
Ferroptose , Morte Celular , Humanos
6.
Commun Biol ; 1: 210, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30511023

RESUMO

Cytotoxic activities of several Golgi-dispersing compounds including AMF-26/M-COPA, brefeldin A and golgicide A have previously been shown to induce autophagy or apoptosis. Here, we demonstrate that these Golgi disruptors also trigger ferroptosis, a non-apoptotic form of cell death characterized by iron-dependent oxidative degradation of lipids. Inhibitors of ferroptosis not only counteract cell death, but they also protect from Golgi dispersal and inhibition of protein secretion in response to several Golgi stress agents. Furthermore, the application of sublethal doses of ferroptosis-inducers such as erastin and sorafenib, low cystine growth conditions, or genetic knockdown of SLC7A11 and GPX4 all similarly protect cells from Golgi stress and lead to modulation of ACSL4, SLC7A5, SLC7A11 or GPX4 levels. Collectively, this study suggests a previously unrecognized function of the Golgi apparatus, which involves cellular redox control and prevents ferroptotic cell death.

7.
Cell Death Dis ; 9(2): 51, 2018 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-29352207

RESUMO

Alcohol abuse is a global health problem causing a substantial fraction of chronic liver diseases. Abundant TGF-ß-a potent pro-fibrogenic cytokine-leads to disease progression. Our aim was to elucidate the crosstalk of TGF-ß and alcohol on hepatocytes. Primary murine hepatocytes were challenged with ethanol and TGF-ß and cell fate was determined. Fluidigm RNA analyses revealed transcriptional effects that regulate survival and apoptosis. Mechanistic insights were derived from enzyme/pathway inhibition experiments and modulation of oxidative stress levels. To substantiate findings, animal model specimens and human liver tissue cultures were investigated. RESULTS: On its own, ethanol had no effect on hepatocyte apoptosis, whereas TGF-ß increased cell death. Combined treatment led to massive hepatocyte apoptosis, which could also be recapitulated in human HCC liver tissue treated ex vivo. Alcohol boosted the TGF-ß pro-apoptotic gene signature. The underlying mechanism of pathway crosstalk involves SMAD and non-SMAD/AKT signaling. Blunting CYP2E1 and ADH activities did not prevent this effect, implying that it was not a consequence of alcohol metabolism. In line with this, the ethanol metabolite acetaldehyde did not mimic the effect and glutathione supplementation did not prevent the super-induction of cell death. In contrast, blocking GSK-3ß activity, a downstream mediator of AKT signaling, rescued the strong apoptotic response triggered by ethanol and TGF-ß. This study provides novel information on the crosstalk between ethanol and TGF-ß. We give evidence that ethanol directly leads to a boost of TGF-ß's pro-apoptotic function in hepatocytes, which may have implications for patients with chronic alcoholic liver disease.


Assuntos
Etanol/efeitos adversos , Hepatócitos/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Apoptose , Humanos , Camundongos , Camundongos Endogâmicos C57BL
8.
Mol Biol Cell ; 29(1): 42-52, 2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-29118074

RESUMO

The secretory pathway is a major determinant of cellular homoeostasis. While research into secretory stress signaling has so far mostly focused on the endoplasmic reticulum (ER), emerging data suggest that the Golgi itself serves as an important signaling hub capable of initiating stress responses. To systematically identify novel Golgi stress mediators, we performed a transcriptomic analysis of cells exposed to three different pharmacological compounds known to elicit Golgi fragmentation: brefeldin A, golgicide A, and monensin. Subsequent gene-set enrichment analysis revealed a significant contribution of the ETS family transcription factors ELK1, GABPA/B, and ETS1 to the control of gene expression following compound treatment. Induction of Golgi stress leads to a late activation of the ETS upstream kinases MEK1/2 and ERK1/2, resulting in enhanced ETS factor activity and the transcription of ETS family target genes related to spliceosome function and cell death induction via alternate MCL1 splicing. Further genetic analyses using loss-of-function and gain-of-function experiments suggest that these transcription factors operate in parallel.


Assuntos
Processamento Alternativo/genética , Complexo de Golgi/metabolismo , Sistema de Sinalização das MAP Quinases , Proteína de Sequência 1 de Leucemia de Células Mieloides/genética , Proteínas Proto-Oncogênicas c-ets/metabolismo , Estresse Fisiológico , Transcrição Gênica , Células A549 , Processamento Alternativo/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Brefeldina A/farmacologia , Citoproteção/efeitos dos fármacos , Perfilação da Expressão Gênica , Técnicas de Silenciamento de Genes , Complexo de Golgi/efeitos dos fármacos , Células HEK293 , Células HeLa , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Monensin/farmacologia , Piridinas/farmacologia , Quinolinas/farmacologia , Bibliotecas de Moléculas Pequenas/farmacologia , Spliceossomos/efeitos dos fármacos , Spliceossomos/metabolismo , Estresse Fisiológico/efeitos dos fármacos , Transcrição Gênica/efeitos dos fármacos , Transcriptoma/efeitos dos fármacos , Transcriptoma/genética , Regulação para Cima/efeitos dos fármacos
9.
Biochim Biophys Acta Mol Cell Res ; 1864(10): 1618-1630, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28651973

RESUMO

Prostate cell metabolism exhibits distinct profiles pre- and post-malignancy. The malignant metabolic shift converts prostate cells from "citrate-producing" to "citrate-oxidizing" cells, thereby enhancing glucose metabolism, a phenotype that contrasts classical tumoral Warburg metabolism. An on-line biosensor chip system (BIONAS 2500) was used to monitor metabolic changes (glycolysis and respiration) in response to the putative anti-cancer nutraceutical 1,25-dihydroxyvitamin D3 [1,25(OH)2D3], in different prostate cancer (PCa) cell lines (LNCaP, VCaP, DU145 and PC3). LNCaP cells exhibited profound metabolic responsiveness to the treatment and thus extensive analysis of metabolism-modulating effects of 1,25(OH)2D3 were performed, including mRNA expression analysis of key metabolic genes (e.g. GLUT1 and PDHK1), analysis of TCA cycle metabolites, glucose uptake/consumption measurements, ATP production, and mitochondrial biogenesis/activity. Altogether, data demonstrate a vivid disruption of glucose metabolism by 1,25(OH)2D3, illustrated by a decreased glucose uptake and an accumulation of citrate/isocitrate due to TCA cycle truncation. Depletion of glycolytic intermediates led to a consistent decrease in TXNIP expression in response to 1,25(OH)2D3, an effect that coincided with the activation of AMPK signaling and a reduction in c-MYC expression. Reduction in TXNIP levels in response to 1,25(OH)2D3 was rescued by an AMPK signaling inhibitor and mimicked by a MYC inhibitor highlighting the possible involvement of both pathways in mediating 1,25(OH)2D3's metabolic effects in PCa cells. Furthermore, pharmacological and genetic modulation of the androgen receptor showed similar and disparate effects on metabolic parameters compared to 1,25(OH)2D3 treatment, highlighting the AR-independent nature of 1,25(OH)2D3's metabolism-modulating effects.


Assuntos
Calcitriol/administração & dosagem , Proteínas de Transporte/genética , Neoplasias da Próstata/tratamento farmacológico , Proteínas Quinases/genética , Proteínas Proto-Oncogênicas c-myc/genética , Quinases Proteína-Quinases Ativadas por AMP , Técnicas Biossensoriais , Calcitriol/metabolismo , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glucose/metabolismo , Humanos , Masculino , Próstata/efeitos dos fármacos , Neoplasias da Próstata/genética , Neoplasias da Próstata/patologia , Receptores Androgênicos/metabolismo
10.
Front Physiol ; 7: 191, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27313533

RESUMO

Liver myofibroblasts (MFB) are crucial mediators of extracellular matrix (ECM) deposition in liver fibrosis. They arise mainly from hepatic stellate cells (HSCs) upon a process termed "activation." To a lesser extent, and depending on the cause of liver damage, portal fibroblasts, mesothelial cells, and fibrocytes may also contribute to the MFB population. Targeting MFB to reduce liver fibrosis is currently an area of intense research. Unfortunately, a clog in the wheel of antifibrotic therapies is the fact that although MFB are known to mediate scar formation, and participate in liver inflammatory response, many of their molecular portraits are currently unknown. In this review, we discuss recent understanding of MFB in health and diseases, focusing specifically on three evolving research fields: metabolism, autophagy, and epigenetics. We have emphasized on therapeutic prospects where applicable and mentioned techniques for use in MFB studies. Subsequently, we highlighted uncharted territories in MFB research to help direct future efforts aimed at bridging gaps in current knowledge.

11.
Traffic ; 17(9): 1042-53, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27306974

RESUMO

Morphogen gradients and concentration are critical features during early embryonic development and cellular differentiation. Previously we reported the preparation of biologically active, fluorescently labeled BMP2 and quantitatively analyzed their binding to the cell surface and followed BMP2 endocytosis over time on the level of single endosomes. Here we show that this internalized BMP2 can be transferred to neighboring cells and, moreover, also activates downstream BMP signaling in adjacent cells, indicated by Smad1/5/8 phosphorylation and activation of the downstream target gene id1. Using a 3D matrix to modulate cell-cell contacts in culture we could show that direct cell-cell contact significantly increased BMP2 transfer. Using inhibitors of vesicular transport, transfer was strongly inhibited. Interestingly, cotreatment with the physiological BMP inhibitor Noggin increased BMP2 uptake and transfer, albeit activation of Smad signaling in neighboring cells was completely suppressed. Our findings present a novel and interesting mechanism by which morphogens such as BMP2 can be transferred between cells and how this is modulated by BMP antagonists such as Noggin, and how this influences activation of Smad signaling by BMP2 in neighboring cells.


Assuntos
Proteína Morfogenética Óssea 2/metabolismo , Comunicação Celular , Endocitose , Transdução de Sinais , Animais , Proteína Morfogenética Óssea 2/antagonistas & inibidores , Proteína Morfogenética Óssea 2/genética , Proteína Morfogenética Óssea 2/farmacologia , Linhagem Celular , Técnicas de Cocultura , Endocitose/efeitos dos fármacos , Citometria de Fluxo , Células HeLa , Humanos , Camundongos , Microscopia de Fluorescência , Mioblastos/citologia , Mioblastos/metabolismo , Fosforilação , Transporte Proteico , Proteínas Recombinantes , Transdução de Sinais/efeitos dos fármacos , Proteína Smad1/metabolismo , Proteína Smad5/metabolismo , Proteína Smad8/metabolismo
12.
Biochem Biophys Res Commun ; 472(4): 585-91, 2016 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-26972255

RESUMO

Low extracellular pH (pHe) is not only the result of cancer metabolism, but a factor of anti-cancer drug efficacy and cancer immunity. In this study, the consequences of acidic stress were evaluated by applying STAP protocol on Jurkat T-lymphocytes (2.0 × 10(6) cells/ml, 25 min in 37 °C). We detected apoptotic process exclusively in pH 3.3 treated cells within 8 h with western blotting (WB). This programmed cell death led to significant drop of cell viability in 72 h measured by MTT assay resulting PI positive population on flow cytometry (FCM) at day 7. Quantified RT-PCR (qRT-PCR) data indicated that all of above mentioned responses are irrelevant to expression of OCT4 gene variants. Interestingly enough, pluripotent cells represented by positive alkaline phosphatase (AP) staining survived acidic stress and consequently proportion of AP positive cells was significantly increased after pH 3.3 treatment (day 7). In general, acidic treatment led to an apoptotic condition for Jurkat T-lymphocytes, which occurred independent of OCT4 induction.


Assuntos
Ácidos/metabolismo , Apoptose , Leucemia de Células T/metabolismo , Células-Tronco Neoplásicas/metabolismo , Estresse Fisiológico , Regulação Leucêmica da Expressão Gênica , Humanos , Concentração de Íons de Hidrogênio , Células Jurkat , Leucemia de Células T/genética , Leucemia de Células T/patologia , Células-Tronco Neoplásicas/patologia , Fator 3 de Transcrição de Octâmero/genética
13.
Mol Oncol ; 10(6): 806-24, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26887594

RESUMO

Pancreatic ductal adenocarcinoma (PDAC) clinically has a very poor prognosis. No small molecule is available to reliably achieve cures. Meisoindigo is chemically related to the natural product indirubin and showed substantial efficiency in clinical chemotherapy for CML in China. However, its effect on PDAC is still unknown. Our results showed strong anti-proliferation effect of meisoindigo on gemcitabine-resistant PDACs. Using a recently established primary PDAC cell line, called Jopaca-1 with a larger CSCs population as model, we observed a reduction of CD133+ and ESA+/CD44+/CD24+ populations upon treatment and concomitantly a decreased expression of CSC-associated genes, and reduced cellular mobility and sphere formation. Investigating basic cellular metabolic responses, we detected lower oxygen consumption and glucose uptake, while intracellular ROS levels increased. This was effectively neutralized by the addition of antioxidants, indicating an essential role of the cellular redox balance. Further analysis on energy metabolism related signaling revealed that meisoindigo inhibited LKB1, but activated AMPK. Both of them were involved in cellular apoptosis. Additional in situ hybridization in tissue sections of PDAC patients reproducibly demonstrated co-expression and -localization of LKB1 and CD133 in malignant areas. Finally, we detected that CD133+/CD44+ were more vulnerable to meisoindigo, which could be mimicked by LKB1 siRNAs. Our results provide the first evidence, to our knowledge, that LKB1 sustains the CSC population in PDACs and demonstrate a clear benefit of meisoindigo in treatment of gemcitabine-resistant cells. This novel mechanism may provide a promising new treatment option for PDAC.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Antineoplásicos/farmacologia , Carcinoma Ductal Pancreático/tratamento farmacológico , Células-Tronco Neoplásicas/efeitos dos fármacos , Pâncreas/efeitos dos fármacos , Neoplasias Pancreáticas/tratamento farmacológico , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Quinases Proteína-Quinases Ativadas por AMP , Apoptose/efeitos dos fármacos , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patologia , Linhagem Celular Tumoral , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Humanos , Indóis/farmacologia , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Pâncreas/metabolismo , Pâncreas/patologia , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Proteínas Serina-Treonina Quinases/metabolismo , Gencitabina , Neoplasias Pancreáticas
14.
J Med Chem ; 58(24): 9591-600, 2015 Dec 24.
Artigo em Inglês | MEDLINE | ID: mdl-26595649

RESUMO

Organometallics with N-heterocyclic carbene (NHC) ligands have triggered major interest in inorganic medicinal chemistry. Complexes of the type Rh(I)(NHC)(COD)X (where X is Cl or I, COD is cyclooctadiene, and NHC is a dimethylbenzimidazolylidene) represent a promising type of new metallodrugs that have been explored by advanced biomedical methods only recently. In this work, we have synthesized and characterized several complexes of this type. As observed by mass spectrometry, these complexes remained stable over at least 3 h in aqueous solution, after which hydrolysis of the halido ligands occurred and release of the NHC ligand was evident. Effects against mitochondria and general cell tumor metabolism were noted at higher concentrations, whereas phosphorylation of HSP27, p38, ERK1/2, FAK, and p70S6K was induced substantially already at lower exposure levels. Regarding the antiproliferative activity in tumor cells, a clear preference for iodido over chlorido secondary ligands was noted, as well as effects of the substituents of the NHC ligand.


Assuntos
Antineoplásicos/química , Benzimidazóis/química , Complexos de Coordenação/química , Ciclo-Octanos/química , Ródio , Antineoplásicos/síntese química , Antineoplásicos/farmacologia , Benzimidazóis/síntese química , Benzimidazóis/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Complexos de Coordenação/síntese química , Complexos de Coordenação/farmacologia , Ciclo-Octanos/síntese química , Ciclo-Octanos/farmacologia , Ensaios de Seleção de Medicamentos Antitumorais , Estabilidade de Medicamentos , Humanos , Hidrólise , Mitocôndrias/metabolismo , Transdução de Sinais/efeitos dos fármacos , Relação Estrutura-Atividade
15.
J Med Chem ; 58(15): 5742-50, 2015 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-26143659

RESUMO

The octamer-binding transcription factor 4 (Oct3/4) is a master gene in the transcriptional regulatory network of pluripotent cells. Repression of Oct3/4 in embryonic stem cells (ESCs) is associated with cell differentiation and loss of pluripotency, whereas forced overexpression in cooperation with other transcriptional factors, such as Nanog, Sox2, and Lin28, can reprogram somatic cells back into pluripotent cells, termed induced pluripotent stem cells (iPSCs). However, random integration and potential tumorigenic transformation caused by viral transduction limit the clinical application of iPSCs. By performing a cell-based high throughput screening (HTS) campaign, we identified several potential small molecules as inducers of Oct3/4 expression. Here we report a lead structure ethyl 2-((4-chlorophenyl)amino)-thiazole-4-carboxylate, termed O4I2, showing high activity in enforcing Oct3/4 expression. On the basis of chemical expansion, we further identified derivatives having increased activities toward Oct3/4 induction. Thus, O4I2 and its derivatives should provide a new class of small molecules suitable for iPSC generation.


Assuntos
Fator 3 de Transcrição de Octâmero/biossíntese , Tiazóis/farmacologia , Células HEK293 , Células HeLa , Humanos
16.
J Med Chem ; 58(12): 4976-83, 2015 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-25898186

RESUMO

Reprogramming somatic cells into induced-pluripotent cells (iPSCs) provides new access to all somatic cell types for clinical application without any ethical controversy arising from the use of embryonic stem cells (ESCs). Established protocols for iPSCs generation based on viral transduction with defined factors are limited by low efficiency and the risk of genetic abnormality. Several small molecules have been reported as replacements for defined transcriptional factors, but a chemical able to replace Oct3/4 allowing the generation of human iPSCs is still unavailable. Using a cell-based High Throughput Screening (HTS) campaign, we identified that 2-[4-[(4-methoxyphenyl)methoxy]phenyl]acetonitrile (1), termed O4I1, enhanced Oct3/4 expression. Structural verification and modification by chemical synthesis showed that O4I1 and its derivatives not only promoted expression and stabilization of Oct3/4 but also enhanced its transcriptional activity in diverse human somatic cells, implying the possible benefit from using this class of compounds in regenerative medicine.


Assuntos
Acetonitrilas/farmacologia , Fator 3 de Transcrição de Octâmero/genética , Éteres Fenílicos/farmacologia , Regulação para Cima/efeitos dos fármacos , Acetonitrilas/química , Células Cultivadas , Descoberta de Drogas , Células HEK293 , Humanos , Modelos Moleculares , Éteres Fenílicos/química
17.
Metallomics ; 7(2): 371-84, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25608481

RESUMO

Compounds which are able to destabilize the lysosomal membrane have been proposed as interesting candidates for targeted anticancer drugs due to the pronounced lysosomal changes in cancer cells. For this purpose, metallocene derivatives of a cell penetrating polyarginine peptide M­(Arg)9(Phe)2Lys­NH2 (where M = ferrocene carboxylate or ruthenocene carboxylate) were designed and their biological activities were investigated in detail. The ferrocenoyl- and ruthenocenoyl polyarginine bioconjugates were synthesized via Fmoc solid-phase peptide synthesis (SPPS) protocols on a microwave-assisted synthesizer. After HPLC purification >98% purity was observed for all conjugates. Their interaction with supported biomimetic membranes was investigated on a quartz crystal microbalance (QCM) and revealed a very strong binding of the metallocene peptides and their metal-free congeners to an artificial eukaryotic membrane model (DMPC­cholesterol). To demonstrate their antiproliferative utility as cytotoxic compounds for a targeted anticancer drug, cell viability (by the crystal violet assay), apoptosis (flow cytometry, Ann V/PI staining), induction of reactive oxygen species (ROS, by flow cytometry with dihydroethidium staining), and changes in cancer cell metabolism, e.g. respiration and glycolysis, were studied. Our results reveal only a weak toxicity for the metal-free polyarginine peptide, which could be significantly enhanced (to ca. 50 µM against HeLa cells in the best case) by coupling ferrocene or ruthenocene carboxylates to the N-terminus of the peptide. The investigation of the cellular uptake and intracellular localization by fluorescence microscopy revealed an enhanced vesicular disruption by the metallocene bioconjugate compared to the metal-free derivative which could be triggered by light and chemicals. Further studies of apoptosis, respiration, glycolysis and ROS formation reveal the superior characteristics of the metallocene compounds. While most cells remain viable even at 300 µM of the metal free bioconjugate 1, most cells are dead or in late stages of apoptosis at 200 µM of the ruthenocene derivative 3, and at 100 µM of the most active ferrocene derivative 2, however, all show very little sign of necrosis. Also, the metal free compound 1 does not induce ROS formation but both metallocene­polyarginine bioconjugates are clearly associated with enhanced intracellular ROS levels, with levels for the redox-active ferrocene derivative being two times higher than for the structurally very similar but redox-silent ruthenocene derivative. We propose that such metallocene­polyarginine peptides induce lysosomal membrane permeabilization and thereby could be developed towards targeted anticancer drugs.


Assuntos
Vesículas Citoplasmáticas/metabolismo , Lisossomos/metabolismo , Compostos Organometálicos/metabolismo , Peptídeos/metabolismo , Apoptose , Ciclo Celular , Fluoresceína-5-Isotiocianato/metabolismo , Violeta Genciana/metabolismo , Células HeLa , Células Hep G2 , Humanos , Concentração Inibidora 50 , Espécies Reativas de Oxigênio/metabolismo , Padrões de Referência
18.
Mol Cancer ; 13: 221, 2014 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-25253202

RESUMO

BACKGROUND: Cancer cells in the advanced stage show aberrant antioxidant capacity to detoxify excessive ROS resulting in the compensation for intrinsic oxidative stress and therapeutic resistance. PDAC is one of the most lethal cancers and often associated with a high accumulation of ROS. Recent studies identified gold(I) NHC complexes as potent TrxR inhibitors suppressing cell growth in a wide spectrum of human malignant cell lines at the low micromolar concentration. However, the mechanism of action is not completely elucidated yet. METHODS: To understand the biological function of gold(I) NHC complexes in PDAC, we used a recently published gold(I) NHC complex, MC3, and evaluated its anti-proliferative effect in four PDAC cell lines, determined by MTT and SRB assays. In further detailed analysis, we analyzed cellular ROS levels using the ROS indicator DHE and mitochondrial membrane potential indicated by the dye JC-1 in Panc1. We also analyzed cell cycle arrest and apoptosis by FACS. To elucidate the role of specific cell signaling pathways in MC3-induced cell death, co-incubation with ROS scavengers, a p38-MAPK inhibitor and siRNA mediated depletion of ASK1 were performed, and results were analyzed by immunoblotting, ELISA-microarrays, qRT-PCR and immunoprecipitation. RESULTS: Our data demonstrate that MC3 efficiently suppressed cell growth, and induced cell cycle arrest and apoptosis in pancreatic cancer cells, in particular in the gemcitabine-resistant cancer cells Panc1 and ASPC1. Treatment with MC3 resulted in a substantial alteration of the cellular redox homeostasis leading to increased ROS levels and a decrease in the mitochondrial membrane potential. ROS scavengers suppressed ROS formation and rescued cells from damage. On the molecular level, MC3 blocked the interaction of Trx with ASK1 and subsequently activated p38-associated signaling. Furthermore, inhibition of this pathway by using ASK1 siRNA or a p38 inhibitor clearly attenuated the effect of MC3 on cell proliferation in Panc1 and ASPC1. CONCLUSIONS: Our results confirm that MC3 is a TrxR inhibitor and show MC3 induced apoptosis in gemcitabine-resistant PDACs. MC3 mediated cell death could be blocked by using anti-oxidants, ASK1 siRNA or p38 inhibitor suggesting that the Trx-ASK1-p38 signal cascade played an important role in gold(I) NHC complexes-mediated cellular damage.


Assuntos
Apoptose/efeitos dos fármacos , MAP Quinase Quinase Quinase 5/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Compostos Organoáuricos/farmacologia , Neoplasias Pancreáticas/patologia , Tiorredoxina Dissulfeto Redutase/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Densitometria , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Sequestradores de Radicais Livres/farmacologia , Homeostase/efeitos dos fármacos , Humanos , Concentração Inibidora 50 , Oxirredução/efeitos dos fármacos , Neoplasias Pancreáticas/enzimologia , Ligação Proteica/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Tiorredoxinas/metabolismo , Gencitabina
19.
Molecules ; 19(9): 15088-102, 2014 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-25244288

RESUMO

We investigated the effect of the Arabidopsis thaliana-derived decapeptide OSIP108 on human cell tolerance to the chemotherapeutic agent cisplatin (Cp), which induces apoptosis and mitochondrial dysfunction. We found that OSIP108 increases the tolerance of HepG2 cells to Cp and prevents Cp-induced changes in basic cellular metabolism. More specifically, we demonstrate that OSIP108 reduces Cp-induced inhibition of respiration, decreases glycolysis and prevents Cp-uptake in HepG2 cells. Apart from its protective action against Cp in human cells, OSIP108 also increases the yeast Saccharomyces cerevisiae tolerance to Cp. A limited yeast-based study of OSIP108 analogs showed that cyclization does not severely affect its activity, which was further confirmed in HepG2 cells. Furthermore, the similarity in the activity of the D-stereoisomer (mirror image) form of OSIP108 with the L-stereoisomer suggests that its mode of action does not involve binding to a stereospecific receptor. In addition, as OSIP108 decreases Cp uptake in HepG2 cells and the anti-Cp activity of OSIP108 analogs without free cysteine is reduced, OSIP108 seems to protect against Cp-induced toxicity only partly via complexation. Taken together, our data indicate that OSIP108 and its cyclic derivatives can protect against Cp-induced toxicity and, thus, show potential as treatment options for mitochondrial dysfunction- and apoptosis-related conditions.


Assuntos
Antineoplásicos/toxicidade , Proteínas de Arabidopsis/farmacologia , Arabidopsis/fisiologia , Cisplatino/toxicidade , Mitocôndrias/efeitos dos fármacos , Células Hep G2 , Humanos , Mitocôndrias/fisiologia
20.
ChemMedChem ; 9(8): 1794-800, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24803348

RESUMO

Organometallic conjugates consisting of a gold(I) N-heterocyclic carbene (NHC) moiety and a naphthalimide were prepared and investigated as cytotoxic agents that interact with both DNA and the disulfide reductase enzyme thioredoxin reductase (TrxR). The complexes were potent DNA intercalators related to their naphthalimide partial structure, inhibited TrxR as a consequence of the incorporation of the gold(I) moiety, and triggered efficient cytotoxic effects in MCF-7 breast and HT-29 colon adenocarcinoma cells. Strong effects on tumor cell metabolism were noted for the most cytotoxic complex, chlorido[1-(3'-(4''-ethylthio-1'',8''-naphthalimid-N''-yl))-propyl-3-methyl-imidazol-2-ylidene]gold(I) (4 d). In conclusion, the conjugation of naphthalimides with gold(I) NHC moieties provided a useful strategy for the design of bioorganometallic anticancer agents with multiple modes of action.


Assuntos
Antineoplásicos/química , Complexos de Coordenação/química , DNA/química , Ouro/química , Substâncias Intercalantes/química , Naftalimidas/química , Tiorredoxina Dissulfeto Redutase/antagonistas & inibidores , Antineoplásicos/síntese química , Antineoplásicos/farmacologia , Proliferação de Células/efeitos dos fármacos , Complexos de Coordenação/síntese química , Complexos de Coordenação/farmacologia , DNA/metabolismo , Ensaios de Seleção de Medicamentos Antitumorais , Células HT29 , Compostos Heterocíclicos/química , Humanos , Substâncias Intercalantes/síntese química , Substâncias Intercalantes/farmacologia , Ligantes , Células MCF-7 , Metano/análogos & derivados , Metano/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...