Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.645
Filtrar
1.
Molecules ; 29(10)2024 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-38792269

RESUMEN

Quercetin, a flavonoid polyphenol found in many plants, has garnered significant attention due to its potential cancer chemoprevention. Our previous studies have shown that acetyl modification of the hydroxyl group of quercetin altered its antitumor effects in HepG2 cells. However, the antitumor effect in other cancer cells with different gene mutants remains unknown. In this study, we investigated the antitumor effect of quercetin and its methylated derivative 3,3',4',7-O-tetramethylquercetin (4Me-Q) and acetylated derivative 3,3',4',7-O-tetraacetylquercetin (4Ac-Q) on two human breast cancer cells, MCF-7 (wt-p53, caspase-3-ve) and MDA-MB-231 (mt-p53, caspase-3+ve). The results demonstrated that 4Ac-Q exhibited significant cell proliferation inhibition and apoptosis induction in both MCF-7 and MDA-MB-231 cells. Conversely, methylation of quercetin was found to lose the activity. The human apoptosis antibody array revealed that 4Ac-Q might induce apoptosis in MCF-7 cells via a p53-dependent pathway, while in MDA-MB-231 cells, it was induced via a caspase-3-dependent pathway. Furthermore, an evaluation using a superoxide inhibitor, MnTBAP, revealed 4Ac-Q-induced apoptosis in MCF-7 cells in a superoxide-independent manner. These findings provide valuable insights into the potential of acetylated quercetin as a new approach in cancer chemoprevention and offer new avenues for health product development.


Asunto(s)
Apoptosis , Neoplasias de la Mama , Proliferación Celular , Quercetina , Humanos , Quercetina/farmacología , Quercetina/análogos & derivados , Quercetina/química , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Acetilación/efectos de los fármacos , Apoptosis/efectos de los fármacos , Metilación , Femenino , Proliferación Celular/efectos de los fármacos , Células MCF-7 , Antineoplásicos/farmacología , Antineoplásicos/química , Línea Celular Tumoral , Proteína p53 Supresora de Tumor/metabolismo , Caspasa 3/metabolismo
2.
Biol Res ; 57(1): 33, 2024 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-38802872

RESUMEN

BACKGROUND: There is a need for novel treatments for neuroblastoma, despite the emergence of new biological and immune treatments, since refractory pediatric neuroblastoma is still a medical challenge. Phyto cannabinoids and their hemisynthetic derivatives have shown evidence supporting their anticancer potential. The aim of this research was to examine Phytocannabinoids or hemisynthetic cannabinoids, which reduce the SHSY-5Y, neuroblastoma cell line's viability. METHODS: Hexane and acetyl acetate extracts were produced starting with Cannabis sativa L. as raw material, then, 9-tetrahidrocannabinol, its acid counterpart and CBN were isolated. In addition, acetylated derivatives of THC and CBN were synthesized. The identification and purity of the chemicals was determined by High Performance Liquid Chromatography and 1H y 13C Magnetic Nuclear Resonance. Then, the capacity to affect the viability of SHSY-5Y, a neuroblastoma cell line, was examined using the resazurin method. Finally, to gain insight into the mechanism of action of the extracts, phytocannabinoids and acetylated derivatives on the examined cells, a caspase 3/7 determination was performed on cells exposed to these compounds. RESULTS: The structure and purity of the isolated compounds was demonstrated. The extracts, the phytocannabinoids and their acetylated counterparts inhibited the viability of the SHSY 5Y cells, being CBN the most potent of all the tested molecules with an inhibitory concentration of 50 percent of 9.5 µM. CONCLUSION: Each of the evaluated molecules exhibited the capacity to activate caspases 3/7, indicating that at least in part, the cytotoxicity of the tested phytocannabinoids and their hemi-synthetic derivatives is mediated by apoptosis.


Asunto(s)
Cannabinoides , Cannabis , Caspasa 3 , Supervivencia Celular , Neuroblastoma , Extractos Vegetales , Humanos , Cannabis/química , Extractos Vegetales/farmacología , Extractos Vegetales/química , Línea Celular Tumoral , Neuroblastoma/tratamiento farmacológico , Supervivencia Celular/efectos de los fármacos , Caspasa 3/metabolismo , Caspasa 3/efectos de los fármacos , Cannabinoides/farmacología , Cannabinoides/química , Caspasa 7/metabolismo , Apoptosis/efectos de los fármacos , Acetilación/efectos de los fármacos , Cromatografía Líquida de Alta Presión
3.
Biochem Biophys Res Commun ; 717: 150061, 2024 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-38718570

RESUMEN

Epithelial mesenchymal transition (EMT) is a critical process implicated in the pathogenesis of retinal fibrosis and the exacerbation of diabetic retinopathy (DR) within retinal pigment epithelium (RPE) cells. Apigenin (AP), a potential dietary supplement for managing diabetes and its associated complications, has demonstrated inhibitory effects on EMT in various diseases. However, the specific impact and underlying mechanisms of AP on EMT in RPE cells remain poorly understood. In this study, we have successfully validated the inhibitory effects of AP on high glucose-induced EMT in ARPE-19 cells and diabetic db/db mice. Notably, our findings have identified CBP/p300 as a potential therapeutic target for EMT in RPE cells and have further substantiated that AP effectively downregulates the expression of EMT-related genes by attenuating the activity of CBP/p300, consequently reducing histone acetylation alterations within the promoter region of these genes. Taken together, our results provide novel evidence supporting the inhibitory effect of AP on EMT in RPE cells, and highlight the potential of specifically targeting CBP/p300 as a strategy for inhibiting retinal fibrosis in the context of DR.


Asunto(s)
Apigenina , Transición Epitelial-Mesenquimal , Glucosa , Histonas , Epitelio Pigmentado de la Retina , Transición Epitelial-Mesenquimal/efectos de los fármacos , Epitelio Pigmentado de la Retina/efectos de los fármacos , Epitelio Pigmentado de la Retina/metabolismo , Epitelio Pigmentado de la Retina/patología , Animales , Apigenina/farmacología , Acetilación/efectos de los fármacos , Humanos , Glucosa/metabolismo , Glucosa/toxicidad , Histonas/metabolismo , Línea Celular , Ratones , Factores de Transcripción p300-CBP/metabolismo , Factores de Transcripción p300-CBP/antagonistas & inhibidores , Ratones Endogámicos C57BL , Retinopatía Diabética/metabolismo , Retinopatía Diabética/patología , Retinopatía Diabética/tratamiento farmacológico , Proteína p300 Asociada a E1A/metabolismo , Masculino , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/patología , Proteína de Unión a CREB/metabolismo , Proteína de Unión a CREB/genética
4.
Cancer Res Commun ; 4(5): 1307-1320, 2024 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-38669046

RESUMEN

Anticancer drug-tolerant persister (DTP) cells at an early phase of chemotherapy reshape refractory tumors. Aldehyde dehydrogenase 1 family member A3 (ALDH1A3) is commonly upregulated by various anticancer drugs in gastric cancer patient-derived cells (PDC) and promotes tumor growth. However, the mechanism underlying the generation of ALDH1A3-positive DTP cells remains elusive. Here, we investigated the mechanism of ALDH1A3 expression and a combination therapy targeting gastric cancer DTP cells. We found that gastric cancer tissues treated with neoadjuvant chemotherapy showed high ALDH1A3 expression. Chromatin immunoprecipitation (ChIP)-PCR and ChIP sequencing analyses revealed that histone H3 lysine 27 acetylation was enriched in the ALDH1A3 promoter in 5-fluorouracil (5-FU)-tolerant persister PDCs. By chemical library screening, we found that the bromodomain and extraterminal (BET) inhibitors OTX015/birabresib and I-BET-762/molibresib suppressed DTP-related ALDH1A3 expression and preferentially inhibited DTP cell growth. In DTP cells, BRD4, but not BRD2/3, was recruited to the ALDH1A3 promoter and BRD4 knockdown decreased drug-induced ALDH1A3 upregulation. Combination therapy with 5-FU and OTX015 significantly suppressed in vivo tumor growth. These observations suggest that BET inhibitors are efficient DTP cell-targeting agents for gastric cancer treatment. SIGNIFICANCE: Drug resistance hampers the cure of patients with cancer. To prevent stable drug resistance, DTP cancer cells are rational therapeutic targets that emerge during the early phase of chemotherapy. This study proposes that the epigenetic regulation by BET inhibitors may be a rational therapeutic strategy to eliminate DTP cells.


Asunto(s)
Resistencia a Antineoplásicos , Fluorouracilo , Histonas , Neoplasias Gástricas , Factores de Transcripción , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/patología , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/genética , Humanos , Animales , Histonas/metabolismo , Ratones , Acetilación/efectos de los fármacos , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Fluorouracilo/farmacología , Fluorouracilo/uso terapéutico , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Línea Celular Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Ratones Desnudos , Aldehído Oxidorreductasas/metabolismo , Aldehído Oxidorreductasas/genética , Proliferación Celular/efectos de los fármacos , Masculino , Femenino , Antineoplásicos/farmacología , Regiones Promotoras Genéticas/efectos de los fármacos , Ratones Endogámicos BALB C , Proteínas que Contienen Bromodominio
5.
Molecules ; 29(8)2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38675608

RESUMEN

Increased oxidative stress is one of the critical pathologies inducing age-related macular degeneration (AMD), characterized by retinal pigment epithelial (RPE) cell damage and death. The unbalanced acetylation and deacetylation of histones have been implicated in AMD pathogenesis or hydrogen peroxide (H2O2)-induced cell damage. Therefore, strategies aimed at controlling the balance between acetylation and deacetylation may effectively protect RPE cells from oxidative damage. Artemisinin is an antimalarial lactone drug derived from Artemisia annua, with antioxidant activity known to modulate histone acetylation in the brain, but its effect on the retina is unknown. In this study, we aimed to investigate whether Artemisinin exerts a cytoprotective effect on oxidative stress-induced apoptosis in RPE cells by regulating histone acetylation. We hypothesized that Artemisinin confers cytoprotection toward H2O2-induced apoptosis in RPE cells through this mechanism. In the present study, we found that Artemisinin at a sub-clinic dosage of 20 µM inhibited the H2O2-induced cell viability decrease and B-cell lymphoma 2 (Bcl-2) protein level decrease and attenuated the H2O2-induced decrease in the histone H4 lysine (Lys) 8 acetylation [Acetyl-H4 (Lys 8)] level in the retinal RPE cell line D407. As expected, histone deacetylase inhibitor Trichostatin A at the concentration of 250 nM increased the Acetyl-H4 (Lys 8) level in D407 cells and attenuated the H2O2-induced cell viability decrease and apoptosis. Similar findings were obtained using adult RPE (ARPE)19 cells, another human RPE cell line, and primary human RPE cell cultures. In conclusion, these results confirmed our hypothesis and indicated that Artemisinin attenuated H2O2-induced apoptosis in apparent correlation with the increase in the Acetyl-H4 (Lys 8) level, which is associated with gene transcription and cell survival. By modulating histone acetylation, Artemisinin may restore the balance between acetylation and deacetylation and enhance the resistance and survival of RPE cells under oxidative stress. Our study provides novel mechanistic insights into the effect of Artemisinin on histone acetylation and apoptosis in RPE cells and supports the potential application of Artemisinin in the prevention and/or treatment of AMD.


Asunto(s)
Apoptosis , Artemisininas , Supervivencia Celular , Histonas , Peróxido de Hidrógeno , Lisina , Estrés Oxidativo , Epitelio Pigmentado de la Retina , Humanos , Histonas/metabolismo , Apoptosis/efectos de los fármacos , Acetilación/efectos de los fármacos , Peróxido de Hidrógeno/farmacología , Artemisininas/farmacología , Epitelio Pigmentado de la Retina/efectos de los fármacos , Epitelio Pigmentado de la Retina/metabolismo , Epitelio Pigmentado de la Retina/citología , Lisina/metabolismo , Supervivencia Celular/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Línea Celular , Citoprotección/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo
6.
FEBS J ; 291(9): 1909-1924, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38380720

RESUMEN

Breast cancer is often treated with chemotherapy. However, the development of chemoresistance results in treatment failure. Long non-coding RNA nuclear paraspeckle assembly transcript 1 (NEAT1) has been shown to contribute to chemoresistance in breast cancer cells. In studying the transcriptional regulation of NEAT1 using multi-omics approaches, we showed that NEAT1 is up-regulated by 5-fluorouracil in breast cancer cells with wild-type cellular tumor antigen p53 but not in mutant-p53-expressing breast cancer cells. The regulation of NEAT1 involves mediator complex subunit 12 (MED12)-mediated repression of histone acetylation marks at the promoter region of NEAT1. Knockdown of MED12 but not coactivator-associated arginine methyltransferase 1 (CARM1) induced histone acetylation at the NEAT1 promoter, leading to elevated NEAT1 mRNAs, resulting in a chemoresistant phenotype. The MED12-dependent regulation of NEAT1 differs between wild-type and mutant p53-expressing cells. MED12 depletion led to increased expression of NEAT1 in a wild-type p53 cell line, but decreased expression in a mutant p53 cell line. Chemoresistance caused by MED12 depletion can be partially rescued by NEAT1 knockdown in p53 wild-type cells. Collectively, our study reveals a novel mechanism of chemoresistance dependent on MED12 transcriptional regulation of NEAT1 in p53 wild-type breast cancer cells.


Asunto(s)
Neoplasias de la Mama , Resistencia a Antineoplásicos , Regulación Neoplásica de la Expresión Génica , Complejo Mediador , ARN Largo no Codificante , Proteína p53 Supresora de Tumor , Humanos , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/tratamiento farmacológico , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Resistencia a Antineoplásicos/genética , Femenino , Complejo Mediador/genética , Complejo Mediador/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Fluorouracilo/farmacología , Línea Celular Tumoral , Regiones Promotoras Genéticas , Proteína-Arginina N-Metiltransferasas/genética , Proteína-Arginina N-Metiltransferasas/metabolismo , Acetilación/efectos de los fármacos , Histonas/metabolismo , Histonas/genética
7.
Circ Heart Fail ; 15(5): e008547, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35418250

RESUMEN

BACKGROUND: High doses of doxorubicin put cancer patients at risk for developing dilated cardiomyopathy. Previously, we showed that doxorubicin treatment decreases SIRT3 (sirtuin 3), the main mitochondrial deacetylase and increases protein acetylation in rat cardiomyocytes. Here, we hypothesize that SIRT3 expression can attenuate doxorubicin induced dilated cardiomyopathy in vivo by preventing the acetylation of mitochondrial proteins. METHODS: Nontransgenic, M3-SIRT3 (truncated SIRT3; short isoform), and M1-SIRT3 (full-length SIRT3; mitochondrial localized) transgenic mice were treated with doxorubicin for 4 weeks (8 mg/kg body weight per week). Echocardiography was performed to assess cardiac structure and function and validated by immunohistochemistry and immunofluorescence (n=4-10). Mass spectrometry was performed on cardiac mitochondrial peptides in saline (n=6) and doxorubicin (n=5) treated hearts. Validation was performed in doxorubicin treated primary rat and human induced stem cell derived cardiomyocytes transduced with adenoviruses for M3-SIRT3 and M1-SIRT3 and deacetylase deficient mutants (n=4-10). RESULTS: Echocardiography revealed that M3-SIRT3 transgenic mice were partially resistant to doxorubicin induced changes to cardiac structure and function whereas M1-SIRT3 expression prevented cardiac remodeling and dysfunction. In doxorubicin hearts, 37 unique acetylation sites on mitochondrial proteins were altered. Pathway analysis revealed these proteins are involved in energy production, fatty acid metabolism, and oxidative stress resistance. Increased M1-SIRT3 expression in primary rat and human cardiomyocytes attenuated doxorubicin-induced superoxide formation, whereas deacetylase deficient mutants were unable to prevent oxidative stress. CONCLUSIONS: Doxorubicin reduced SIRT3 expression and markedly affected the cardiac mitochondrial acetylome. Increased M1-SIRT3 expression in vivo prevented doxorubicin-induced cardiac dysfunction, suggesting that SIRT3 could be a potential therapeutic target for mitigating doxorubicin-induced dilated cardiomyopathy.


Asunto(s)
Cardiomiopatía Dilatada , Doxorrubicina , Estrés Oxidativo , Sirtuina 3 , Acetilación/efectos de los fármacos , Animales , Cardiomiopatía Dilatada/inducido químicamente , Cardiomiopatía Dilatada/genética , Cardiomiopatía Dilatada/patología , Cardiomiopatía Dilatada/prevención & control , Doxorrubicina/efectos adversos , Doxorrubicina/farmacología , Insuficiencia Cardíaca/metabolismo , Humanos , Ratones , Ratones Transgénicos , Mitocondrias Cardíacas/efectos de los fármacos , Mitocondrias Cardíacas/metabolismo , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Estrés Oxidativo/efectos de los fármacos , Ratas , Sirtuina 3/genética , Sirtuina 3/metabolismo
8.
Oxid Med Cell Longev ; 2022: 4592170, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35251473

RESUMEN

Lysine ß-hydroxybutyrylation (Kbhb) is a newly identified protein posttranslational modification (PTM) derived from ß-hydroxybutyrate (BHB), a product of ketone body metabolism in liver. BHB could serve as an energy source and play a role in the suppression of oxidative stress. The plasma concentration of BHB could increase up to 20 mM during starvation and in pathological conditions. Despite the progress, how the cells derived from extrahepatic tissues respond to elevated environmental BHB remains largely unknown. Given that BHB can significantly drive Kbhb, we characterized the BHB-induced lysine ß-hydroxybutyrylome and acetylome by quantitative proteomics. A total of 840 unique Kbhb sites on 429 proteins were identified, with 42 sites on 39 proteins increased by more than 50% in response to BHB. The results showed that the upregulated Kbhb induced by BHB was involved in aminoacyl-tRNA biosynthesis, 2-oxocarboxylic acid metabolism, citrate cycle, glycolysis/gluconeogenesis, and pyruvate metabolism pathways. Moreover, some BHB-induced Kbhb substrates were significantly involved in diseases such as cancer. Taken together, we investigate the dynamics of lysine ß-hydroxybutyrylome and acetylome induced by environmental BHB, which reveals the roles of Kbhb in regulating various biological processes and expands the biological functions of BHB.


Asunto(s)
Ácido 3-Hidroxibutírico/metabolismo , Ácido 3-Hidroxibutírico/farmacología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Lisina/metabolismo , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Proteoma/efectos de los fármacos , Proteómica/métodos , Transducción de Señal/efectos de los fármacos , Acetilación/efectos de los fármacos , Animales , Células Cultivadas , Ratones , Unión Proteica/efectos de los fármacos , Proteoma/metabolismo , Regulación hacia Arriba/efectos de los fármacos
9.
Cancer Control ; 29: 10732748221074051, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35067084

RESUMEN

INTRODUCTION: The prototype DNA hypomethylating agents 5-azacytidine (5AC) and decitabine (DAC) are currently FDA-approved for treatment of blood and bone marrow disorders like myelodysplastic syndrome. 5AC and DAC are considered similar drugs and were shown to induce histone modifications that modulate gene expression. The aim of this study is to compare the effect of both drugs on histone acetylation and methylation at multiple histone amino acids residues. METHODS: Mass spectrometry was used to compare the effect of both drugs on 95 different histone posttranslational modifications (PTMs) in leukemia cells. ChIP-Seq analysis was used to compare the impact of both drugs on the genome-wide acetylation of the H3K9 mark using primary leukemia cells from six de-identified AML patients. RESULTS: Both DAC and 5AC induced histone PTMs in different histone isoforms like H1.4, H2A, H3, H3.1, and H4. Changes in both histone methylation and acetylation were observed with both drugs; however, there were distinct differences in the histone modifications induced by the two drugs. Since both drugs were shown to increase the activity of the HDAC SIRT6 previously, we tested the effect of 5AC on the acetylation of H3K9, the physiological substrate SIRT6, using ChIP-Seq analysis and compared it to the previously published DAC-induced changes. Significant H3K9 acetylation changes (P< .05) were detected at 925 genes after 5AC treatment vs only 182 genes after DAC treatment. Nevertheless, the gene set modified by 5AC was different from that modified by DAC with only ten similar genes modulated by both drugs. CONCLUSION: Despite similarity in chemical structure and DNA hypomethylating activity, 5AC and DAC induced widely different histone PTMs and considering them interchangeable should be carefully evaluated. The mechanism of these histone PTM changes is not clear and may involve modulation of the activity or the expression of the enzymes inducing histone PTMs.


Asunto(s)
Acetilación/efectos de los fármacos , Azacitidina/farmacología , Metilación de ADN/efectos de los fármacos , Decitabina/farmacología , Histonas/efectos de los fármacos , Línea Celular Tumoral , Humanos , Leucemia/tratamiento farmacológico , Procesamiento Proteico-Postraduccional/efectos de los fármacos
10.
Toxicol Appl Pharmacol ; 436: 115882, 2022 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-35016910

RESUMEN

Oocyte maturation is essential for fertilization and early embryo development, and proper organelle functions guarantee this process to maintain high-quality oocytes. The type B trichothecene nivalenol (NIV) is a mycotoxin produced by Fusarium oxysporum and is commonly found in contaminated food. NIV intake affect growth, the immune system, and the female reproductive system. Here, we investigated NIV toxicity on mouse oocyte quality. Transcriptome analysis results showed that NIV exposure altered the expression of multiple genes involved in spindle formation and organelle function in mouse oocytes, indicating its toxicity on mouse oocyte maturation. Further analysis indicated that NIV exposure disrupted spindle structure and chromosome alignment, possibly through tubulin acetylation. NIV exposure induced aberrant mitochondria distribution and reduced mitochondria number, mitochondria membrane potential (MMP), and ATP levels. In addition, NIV caused the abnormal distribution of the Golgi apparatus and altered the expression of the vesicle trafficking protein Rab11. ER distribution was also disturbed under NIV exposure, indicating the effects of NIV on protein modification and transport in oocytes. Thus, our results demonstrated that NIV exposure affected spindle structure and organelles function in mouse oocytes.


Asunto(s)
Desarrollo Embrionario/efectos de los fármacos , Oocitos/efectos de los fármacos , Orgánulos/efectos de los fármacos , Huso Acromático/efectos de los fármacos , Tricotecenos/efectos adversos , Acetilación/efectos de los fármacos , Adenosina Trifosfato/metabolismo , Animales , Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Cromosomas/efectos de los fármacos , Femenino , Meiosis/efectos de los fármacos , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Ratones Endogámicos ICR , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Micotoxinas/efectos adversos , Oocitos/metabolismo , Oogénesis/efectos de los fármacos , Orgánulos/metabolismo , Huso Acromático/metabolismo , Transcriptoma/efectos de los fármacos , Tubulina (Proteína)/metabolismo
11.
J Med Chem ; 65(3): 2208-2224, 2022 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-35005974

RESUMEN

Glioma treatment remains a challenge with a low survival rate due to the lack of effective therapeutics. Monoamine oxidase A (MAO A) plays a role in glioma development, and MAO A inhibitors reduce glioma growth. Histone deacetylase (HDAC) inhibition has emerged as a promising therapy for various malignancies including gliomas. We have synthesized and evaluated N-methylpropargylamine-conjugated hydroxamic acids as dual inhibitors of MAO A and HDAC. Compounds display potent MAO A inhibition with IC50 from 0.03 to <0.0001 µM and inhibit HDAC isoforms and cell growth in the micromolar to nanomolar IC50 range. These selective MAO A inhibitors increase histone H3 and α-tubulin acetylation and induce cell death via nonapoptotic mechanisms. Treatment with 15 reduced tumor size, reduced MAO A activity in brain and tumor tissues, and prolonged the survival. This first report on dual inhibitors of MAO A and HDAC establishes the basis of translational research for an improved treatment of glioma.


Asunto(s)
Inhibidores Enzimáticos/química , Histona Desacetilasas/química , Ácidos Hidroxámicos/química , Monoaminooxidasa/química , Acetilación/efectos de los fármacos , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Diseño de Fármacos , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacología , Inhibidores Enzimáticos/uso terapéutico , Glioma/tratamiento farmacológico , Glioma/mortalidad , Histona Desacetilasas/metabolismo , Histonas/metabolismo , Humanos , Ácidos Hidroxámicos/metabolismo , Ácidos Hidroxámicos/farmacología , Ácidos Hidroxámicos/uso terapéutico , Isoenzimas/antagonistas & inhibidores , Isoenzimas/metabolismo , Estimación de Kaplan-Meier , Masculino , Ratones , Ratones Endogámicos C57BL , Monoaminooxidasa/metabolismo , Pargilina/análogos & derivados , Pargilina/química , Propilaminas/química , Relación Estructura-Actividad , Trasplante Heterólogo
12.
Lab Invest ; 102(1): 80-89, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34508164

RESUMEN

Ameloblastoma (AB) is the most common benign epithelial odontogenic tumor occurring in the jawbone. AB is a slowly growing tumor but sometimes shows a locally invasive and an aggressive growth pattern with a marked bone resorption. In addition, the local recurrence and distant metastasis of AB also sometimes occurs, which resembles one of the typical malignant potentials. From these points of view, to understand better the mechanisms of AB cell migration or invasion is necessary for the better clinical therapy and improvements of the patients' quality of life. Microtubules in eukaryotic cells reveal the shape of hollow cylinders made up of polymerized alpha (α)- and beta (ß)-tubulin dimers and form the cytoskeleton together with microfilaments and intermediate filaments. Microtubules play important roles in cell migration by undergoing assembly and disassembly with post-translational modifications. Stability of microtubules caused by their acetylation is involved in cell migration. In this study, we investigated the expression and distribution of acetylated α-tubulin and alpha-tubulin N-acetyltransferase 1 (αTAT1), an enzyme which acetylates Lys-40 in α-tubulin, in AB specimens, and analyzed how tubulin was acetylated by αTAT1 activation in a human AB cell line, AM-1. Finally, we clarified that TGF-ß-activated kinase1 (TAK1) was phosphorylated by TGF-ß stimulation, then, induced tubulin acetylation via αTAT1 activation, which subsequently activated the migration and invasion of AB cells.


Asunto(s)
Acetiltransferasas/metabolismo , Ameloblastoma/metabolismo , Movimiento Celular , Neoplasias Maxilomandibulares/metabolismo , Proteínas de Microtúbulos/metabolismo , Tubulina (Proteína)/metabolismo , Acetilación/efectos de los fármacos , Acetiltransferasas/genética , Adolescente , Adulto , Anciano , Ameloblastoma/genética , Ameloblastoma/patología , Línea Celular Tumoral , Femenino , Humanos , Inmunohistoquímica , Neoplasias Maxilomandibulares/genética , Neoplasias Maxilomandibulares/patología , Quinasas Quinasa Quinasa PAM/genética , Quinasas Quinasa Quinasa PAM/metabolismo , Masculino , Proteínas de Microtúbulos/genética , Persona de Mediana Edad , Invasividad Neoplásica , Interferencia de ARN , Factor de Crecimiento Transformador beta/farmacología , Adulto Joven
13.
J Oral Pathol Med ; 51(6): 553-562, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34661317

RESUMEN

AIM: To evaluate the potential use of Cephaeline as a therapeutic strategy to manage mucoepidermoid carcinomas (MEC) of the salivary glands. MATERIAL AND METHODS: UM-HMC-1, UM-HMC-2, and UM-HMC-3A MEC cell lines were used to establish the effects of Cephaeline over tumor viability determined by MTT assay. In vitro wound healing scratch assays were performed to address cellular migration while immunofluorescence staining for histone H3 lysine 9 (H3k9ac) was used to identify the acetylation status of tumor cells upon Cephaeline administration. The presence of cancer stem cells was evaluated by the identification of ALDH enzymatic activity by flow cytometry and through functional assays using in vitro tumorsphere formation. RESULTS: A single administration of Cephaeline resulted in reduced viability of MEC cells along with the halt on tumor growth and cellular migration potential. Administration of Cephaeline resulted in chromatin histone acetylation as judged by the increased levels of H3K9ac and disruption of tumorspheres formation. Interestingly, ALDH levels were increased in UM-HMC-1 and UM-HMC-3A cell lines, while UM-HMC-2 showed a reduced enzymatic activity. CONCLUSION: Cephaeline has shown anti-cancer properties in all MEC cell lines tested by regulating tumor cells' viability, migration, proliferation, and disrupting the ability of cancer cells to generate tumorspheres.


Asunto(s)
Carcinoma Mucoepidermoide , Acetilación/efectos de los fármacos , Carcinoma Mucoepidermoide/metabolismo , Línea Celular Tumoral , Emetina/análogos & derivados , Emetina/farmacología , Histonas/metabolismo , Humanos , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/patología
14.
J Clin Endocrinol Metab ; 107(2): e783-e792, 2022 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-34453169

RESUMEN

CONTEXT: Primary hyperparathyroidism (PHPT) results from the hypersecretion of parathyroid hormone from parathyroid tumors. A transcription factor, namely Paired box1 (PAX1), is active in parathyroid gland development. OBJECTIVE: We aimed to study potential epigenetic-mediated mechanism of PAX1 gene in sporadic parathyroid adenomas. METHODS: In parathyroid adenomas tissues, we analyzed the DNA methylation via bisulfite-specific polymerase chain reaction (BSP) and histone modifications via chromatin immunoprecipitation in regulating the differential expression of PAX1. RESULTS: The results showed that mRNA and protein expression of PAX1 was significantly reduced in parathyroid adenomas. Bisulfite sequencing demonstrated hypermethylation in the promoter region of PAX1 (35%; 14/40) and lower levels of histone 3 lysine 9 acetylation (H3K9ac) were observed on the promoter region of PAX1 (6-fold; P < .004) in parathyroid adenomas. Furthermore, upon treatment with a pharmacologic inhibitor, namely 5'aza-2 deoxycytidine, in rat parathyroid continuous cells, we found re-expression of PAX1 gene. CONCLUSION: Our study not only reveals expression of PAX1 is epigenetically deregulated but also paves a way for clinical and therapeutic implications in patients with PHPT.


Asunto(s)
Adenoma/genética , Carcinogénesis/genética , Epigénesis Genética/genética , Factores de Transcripción Paired Box/genética , Neoplasias de las Paratiroides/genética , Acetilación/efectos de los fármacos , Adenoma/patología , Adenoma/terapia , Adolescente , Adulto , Anciano , Animales , Carcinogénesis/efectos de los fármacos , Estudios de Casos y Controles , Línea Celular Tumoral , Metilación de ADN/efectos de los fármacos , Decitabina/farmacología , Decitabina/uso terapéutico , Epigénesis Genética/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Voluntarios Sanos , Código de Histonas/efectos de los fármacos , Humanos , Ácidos Hidroxámicos/farmacología , Ácidos Hidroxámicos/uso terapéutico , Masculino , Persona de Mediana Edad , Factores de Transcripción Paired Box/metabolismo , Glándulas Paratiroides/patología , Glándulas Paratiroides/cirugía , Neoplasias de las Paratiroides/patología , Neoplasias de las Paratiroides/terapia , Paratiroidectomía , Regiones Promotoras Genéticas/genética , Ratas , Adulto Joven
16.
Pharmacology ; 107(1-2): 1-13, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34915505

RESUMEN

BACKGROUND: The pathobiology of diabetes and associated complications has been widely researched in various countries, but effective prevention and treatment methods are still insufficient. Diabetes is a metabolic disorder of carbohydrates, fats, and proteins caused by an absence of insulin or insulin resistance, which mediates an increase of oxidative stress, release of inflammatory factors, and macro- or micro-circulation dysfunctions, ultimately developing into diverse complications. SUMMARY: In the last decade through pathogenesis research, epigenetics has been found to affect metabolic diseases. Particularly, DNA methylation, histone acetylation, and miRNAs promote or inhibit diabetes and complications by regulating the expression of related factors. Curcumin has a wide range of beneficial pharmacological activities, including anti-inflammatory, anti-oxidation, anticancer, anti-diabetes, anti-rheumatism, and increased immunity. Key Messages: In this review, we discuss the effects of curcumin and analogs on diabetes and associated complications through epigenetics, and we summarize the preclinical and clinical researches for curcumin and its analogs in terms of management of diabetes and associated complications, which may provide an insight into the development of targeted therapy of endocrine diseases.


Asunto(s)
Curcumina/farmacología , Curcumina/uso terapéutico , Complicaciones de la Diabetes/tratamiento farmacológico , Diabetes Mellitus/tratamiento farmacológico , Epigénesis Genética/efectos de los fármacos , Acetilación/efectos de los fármacos , Animales , Curcumina/análogos & derivados , Metilación de ADN/efectos de los fármacos , Complicaciones de la Diabetes/genética , Diabetes Mellitus/genética , Humanos , MicroARNs/efectos de los fármacos
17.
Cells ; 10(12)2021 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-34943918

RESUMEN

Experiments on Vicia faba root meristem cells exposed to 150 µM cadmium chloride (CdCl2) were undertaken to analyse epigenetic changes, mainly with respect to DNA replication stress. Histone modifications examined by means of immunofluorescence labeling included: (1) acetylation of histone H3 on lysine 56 (H3K56Ac), involved in transcription, S phase, and response to DNA damage during DNA biosynthesis; (2) dimethylation of histone H3 on lysine 79 (H3K79Me2), correlated with the replication initiation; (3) phosphorylation of histone H3 on threonine 45 (H3T45Ph), engaged in DNA synthesis and apoptosis. Moreover, immunostaining using specific antibodies against 5-MetC-modified DNA was used to determine the level of DNA methylation. A significant decrease in the level of H3K79Me2, noted in all phases of the CdCl2-treated interphase cell nuclei, was found to correspond with: (1) an increase in the mean number of intranuclear foci of H3K56Ac histones (observed mainly in S-phase), (2) a plethora of nuclear and nucleolar labeling patterns (combined with a general decrease in H3T45Ph), and (3) a decrease in DNA methylation. All these changes correlate well with a general viewpoint that DNA modifications and post-translational histone modifications play an important role in gene expression and plant development under cadmium-induced stress conditions.


Asunto(s)
Cadmio/toxicidad , Replicación del ADN/genética , Epigénesis Genética , Meristema/citología , Meristema/genética , Estrés Fisiológico/genética , Vicia faba/genética , 5-Metilcitosina/metabolismo , Acetilación/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Cromatina/metabolismo , Replicación del ADN/efectos de los fármacos , ADN de Plantas/metabolismo , Epigénesis Genética/efectos de los fármacos , Histonas/metabolismo , Lisina/metabolismo , Meristema/efectos de los fármacos , Metilación/efectos de los fármacos , Fosforilación/efectos de los fármacos , Estrés Fisiológico/efectos de los fármacos , Vicia faba/citología , Vicia faba/efectos de los fármacos
18.
Cells ; 10(12)2021 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-34943964

RESUMEN

People living with HIV (PLWH) have to take an antiretroviral therapy (ART) for life and show noncommunicable illnesses such as chronic inflammation, immune activation, and multiorgan dysregulation. Recent studies suggest that long-term use of ART induces comorbid conditions and is one of the leading causes of heart failure in PLWH. However, the molecular mechanism of antiretroviral drugs (ARVs) induced heart failure is unclear. To determine the mechanism of ARVs induced cardiac dysfunction, we performed global transcriptomic profiling of ARVs treated neonatal rat ventricular cardiomyocytes in culture. Differentially expressed genes were identified by RNA-sequencing. Our data show that ARVs treatment causes upregulation of several biological functions associated with cardiotoxicity, hypertrophy, and heart failure. Global gene expression data were validated in cardiac tissue isolated from HIV patients having a history of ART. Interestingly, we found that homeodomain-only protein homeobox (HOPX) expression was significantly increased in cardiomyocytes treated with ARVs and in the heart tissue of HIV patients. Furthermore, we found that HOPX plays a crucial role in ARVs mediated cellular hypertrophy. Mechanistically, we found that HOPX plays a critical role in epigenetic regulation, through deacetylation of histone, while the HDAC inhibitor, Trichostatin A, can restore the acetylation level of histone 3 in the presence of ARVs.


Asunto(s)
Cardiomegalia/genética , Infecciones por VIH/tratamiento farmacológico , Insuficiencia Cardíaca/genética , Proteínas de Homeodominio/genética , Proteínas Supresoras de Tumor/genética , Acetilación/efectos de los fármacos , Animales , Antirretrovirales/efectos adversos , Antirretrovirales/farmacología , Cardiomegalia/inducido químicamente , Cardiomegalia/patología , Modelos Animales de Enfermedad , Epigénesis Genética/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , VIH/efectos de los fármacos , VIH/patogenicidad , Infecciones por VIH/complicaciones , Infecciones por VIH/virología , Insuficiencia Cardíaca/inducido químicamente , Insuficiencia Cardíaca/tratamiento farmacológico , Insuficiencia Cardíaca/patología , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Ácidos Hidroxámicos/farmacología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , RNA-Seq , Ratas , Transcriptoma/efectos de los fármacos , Transcriptoma/genética
19.
Int J Mol Sci ; 22(23)2021 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-34884764

RESUMEN

Genetic and epigenetic changes alter gene expression, contributing to cancer. Epigenetic changes in cancer arise from alterations in DNA and histone modifications that lead to tumour suppressor gene silencing and the activation of oncogenes. The acetylation status of histones and non-histone proteins are determined by the histone deacetylases and histone acetyltransferases that control gene transcription. Organoselenium compounds have become promising contenders in cancer therapeutics. Apart from their anti-oxidative effects, several natural and synthetic organoselenium compounds and metabolites act as histone deacetylase inhibitors, which influence the acetylation status of histones and non-histone proteins, altering gene transcription. This review aims to summarise the effect of natural and synthetic organoselenium compounds on histone and non-histone protein acetylation/deacetylation in cancer therapy.


Asunto(s)
Inhibidores de Histona Desacetilasas/farmacología , Neoplasias/tratamiento farmacológico , Compuestos de Organoselenio/farmacología , Acetilación/efectos de los fármacos , Sistemas de Liberación de Medicamentos , Epigénesis Genética/efectos de los fármacos , Código de Histonas/efectos de los fármacos , Código de Histonas/genética , Inhibidores de Histona Desacetilasas/síntesis química , Inhibidores de Histona Desacetilasas/metabolismo , Histona Desacetilasas/metabolismo , Histonas/metabolismo , Humanos , Terapia Molecular Dirigida , Nanopartículas , Neoplasias/genética , Neoplasias/metabolismo , Compuestos de Organoselenio/síntesis química , Compuestos de Organoselenio/metabolismo , Procesamiento Proteico-Postraduccional/efectos de los fármacos
20.
Anticancer Res ; 41(12): 5987-5996, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34848452

RESUMEN

BACKGROUND/AIM: Simultaneous inhibition of histone deacetylase and proteasomes induces endoplasmic reticulum (ER) stress efficiently. RTS-V5 is the first dual histone deacetylase-proteasome inhibitor, and we anticipated that combining it with the cytochrome P450 family 3 subfamily A member 4 inhibitor ritonavir would enhance its activity in bladder cancer cells. MATERIALS AND METHODS: Using bladder cancer cells (human T-24, J-82, murine MBT-2), we evaluated the ability and mechanism by which the combination of RTS-V5 and ritonavir induced ER stress and killed cancer cells. RESULTS: The combination of RTS-V5 and ritonavir triggered robust apoptosis and inhibited bladder cancer growth effectively in vitro and in vivo. It caused ubiquitinated protein accumulation and induced ER stress synergistically. The combination inhibited the mammalian target of rapamycin pathway by increasing the expression of AMP-activated protein kinase. We also found that the combination caused histone and tubulin hyperacetylation. CONCLUSION: Ritonavir enhances the ability of RTS-V5 to cause ER stress in bladder cancer cells.


Asunto(s)
Antineoplásicos/farmacología , Estrés del Retículo Endoplásmico/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Inhibidores de Proteasoma/farmacología , Ritonavir/farmacología , Acetilación/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Histonas/metabolismo , Humanos , Ratones , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Neoplasias de la Vejiga Urinaria , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA