Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.420
Filtrar
1.
Int J Oral Sci ; 16(1): 35, 2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38719825

RESUMEN

The efficient clinical treatment of oral squamous cell carcinoma (OSCC) is still a challenge that demands the development of effective new drugs. Phenformin has been shown to produce more potent anti-tumor activities than metformin on different tumors, however, not much is known about the influence of phenformin on OSCC cells. We found that phenformin suppresses OSCC cell proliferation, and promotes OSCC cell autophagy and apoptosis to significantly inhibit OSCC cell growth both in vivo and in vitro. RNA-seq analysis revealed that autophagy pathways were the main targets of phenformin and identified two new targets DDIT4 (DNA damage inducible transcript 4) and NIBAN1 (niban apoptosis regulator 1). We found that phenformin significantly induces the expression of both DDIT4 and NIBAN1 to promote OSCC autophagy. Further, the enhanced expression of DDIT4 and NIBAN1 elicited by phenformin was not blocked by the knockdown of AMPK but was suppressed by the knockdown of transcription factor ATF4 (activation transcription factor 4), which was induced by phenformin treatment in OSCC cells. Mechanistically, these results revealed that phenformin triggers endoplasmic reticulum (ER) stress to activate PERK (protein kinase R-like ER kinase), which phosphorylates the transitional initial factor eIF2, and the increased phosphorylation of eIF2 leads to the increased translation of ATF4. In summary, we discovered that phenformin induces its new targets DDIT4 and especially NIBAN1 to promote autophagic and apoptotic cell death to suppress OSCC cell growth. Our study supports the potential clinical utility of phenformin for OSCC treatment in the future.


Asunto(s)
Autofagia , Carcinoma de Células Escamosas , Proliferación Celular , Estrés del Retículo Endoplásmico , Neoplasias de la Boca , Fenformina , Factores de Transcripción , Fenformina/farmacología , Estrés del Retículo Endoplásmico/efectos de los fármacos , Humanos , Neoplasias de la Boca/tratamiento farmacológico , Autofagia/efectos de los fármacos , Carcinoma de Células Escamosas/tratamiento farmacológico , Proliferación Celular/efectos de los fármacos , Línea Celular Tumoral , Factores de Transcripción/metabolismo , Factores de Transcripción/efectos de los fármacos , Ratones , Proteínas Reguladoras de la Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/metabolismo , Apoptosis/efectos de los fármacos , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Western Blotting
2.
Int J Oncol ; 60(3)2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35179222

RESUMEN

Regulator of ribosome synthesis 1 (RRS1) is a key factor in ribosome biosynthesis and other cellular functions. High level of RRS1 in breast cancer cell lines is associated with increased cell proliferation, invasion and migration. RRS1 controls the assembly of the 60s subunit and maturation of 25S rRNA during ribosome biosynthesis. In this study, lentiviral transfection of sh­RNA was used to knock down the level of RRS1, to detect the effect of RRS1 on cell function and to explore the specific mechanism of RRS1 affecting cell invasion and metastasis by COIP and dual­luciferase reporter gene assays. The present study found that RRS1 knockdown reduced the accumulation of ribosome protein L11 (RPL11) in the nucleolus, which then migrated to the nucleoplasm and bound to c­Myc. This inhibited trans­activation of SNAIL by c­Myc and eventually decreased the invasion and metastasis capacity of the human breast cancer cell line BT549. Taken together, RRS1 regulates invasion and metastasis of human breast cancer cells through the RPL11­c­Myc­SNAIL axis. The findings are of great significance for exploring the mechanism of breast cancer invasion and metastasis and the corresponding regulatory factors.


Asunto(s)
Regulación hacia Abajo/genética , Metástasis de la Neoplasia/genética , Proteínas de Unión al ARN/efectos de los fármacos , Línea Celular Tumoral/efectos de los fármacos , Línea Celular Tumoral/metabolismo , Proliferación Celular/genética , Proteínas de Unión al ADN/efectos de los fármacos , Proteínas de Unión al ADN/genética , Regulación hacia Abajo/efectos de los fármacos , Humanos , Metástasis de la Neoplasia/tratamiento farmacológico , Metástasis de la Neoplasia/prevención & control , Proteínas de Unión al ARN/antagonistas & inhibidores , Proteínas de Unión al ARN/genética , Factores de Transcripción de la Familia Snail/efectos de los fármacos , Factores de Transcripción de la Familia Snail/genética , Factores de Transcripción/efectos de los fármacos , Factores de Transcripción/genética
3.
Nat Cell Biol ; 24(1): 24-34, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-35027731

RESUMEN

SARS-CoV-2 infection of human cells is initiated by the binding of the viral Spike protein to its cell-surface receptor ACE2. We conducted a targeted CRISPRi screen to uncover druggable pathways controlling Spike protein binding to human cells. Here we show that the protein BRD2 is required for ACE2 transcription in human lung epithelial cells and cardiomyocytes, and BRD2 inhibitors currently evaluated in clinical trials potently block endogenous ACE2 expression and SARS-CoV-2 infection of human cells, including those of human nasal epithelia. Moreover, pharmacological BRD2 inhibition with the drug ABBV-744 inhibited SARS-CoV-2 replication in Syrian hamsters. We also found that BRD2 controls transcription of several other genes induced upon SARS-CoV-2 infection, including the interferon response, which in turn regulates the antiviral response. Together, our results pinpoint BRD2 as a potent and essential regulator of the host response to SARS-CoV-2 infection and highlight the potential of BRD2 as a therapeutic target for COVID-19.


Asunto(s)
Enzima Convertidora de Angiotensina 2/metabolismo , Antivirales/farmacología , Células Epiteliales/virología , SARS-CoV-2/metabolismo , Factores de Transcripción/efectos de los fármacos , Enzima Convertidora de Angiotensina 2/efectos de los fármacos , COVID-19/metabolismo , COVID-19/virología , Línea Celular , Células Epiteliales/metabolismo , Humanos , Glicoproteínas de Membrana/metabolismo , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/patogenicidad , Factores de Transcripción/metabolismo , Tratamiento Farmacológico de COVID-19
4.
Clin Transl Med ; 12(1): e718, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-35083874

RESUMEN

BACKGROUND: Chemoradiotherapy-induced PD-L1 upregulation leads to therapeutic resistance and treatment failure. The PD-1/PD-L1 blocking antibodies sensitize cancers to chemoradiotherapy by blocking extracellular PD-1 and PD-L1 binding without affecting the oncogenic function of intracellular PD-L1. Reversing the chemoradiation-induced PD-L1 expression could provide a new strategy to achieve a greater anti-tumour effect of chemoradiotherapy. Here, we aimed to identify candidate small molecular inhibitors that might boost the anti-tumour immunity of chemoradiotherapy by decreasing treatment-induced PD-L1 expression in non-small cell lung cancer (NSCLC). METHODS: A drug array was used to recognize compounds that can suppress the cisplatin-induced and radiation-induced PD-L1 expression in NSCLC via the flow cytometry-based assay. We examined whether and how targeting bromodomain containing 4 (BRD4) inhibits chemoradiation-induced PD-L1 expression and evaluated the effect of BRD4 inhibition and chemoradiation combination in vivo. RESULTS: BRD4 inhibitors JQ1 and ARV-771 were identified as the most promising drugs both in the cisplatin and radiation screening projects in two NSCLC cell lines. Targeting BRD4 was supposed to block chemoradiotherapy inducible PD-L1 expression by disrupting the recruitment of BRD4-IRF1 complex to PD-L1 promoter. A positive correlation between BRD4 and PD-L1 expression was observed in human NSCLC tissues. Moreover, BRD4 inhibition synergized with chemoradiotherapy and PD-1 blockade to show a robust anti-tumour immunity dependent on CD8+ T cell through limiting chemoradiation-induced tumour cell surface PD-L1 upregulation in vivo. Notably, the BRD4-targeted combinatory treatments did not show increased toxicities. CONCLUSION: The data showed that BRD4-targeted therapy synergized with chemoradiotherapy and anti-PD-1 antibody by boosting anti-tumour immunity in NSCLC.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Quimioradioterapia/normas , Transducción de Señal/genética , Animales , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/inmunología , Proteínas de Ciclo Celular/efectos de los fármacos , Proteínas de Ciclo Celular/genética , Quimioradioterapia/métodos , Quimioradioterapia/estadística & datos numéricos , Modelos Animales de Enfermedad , Expresión Génica/efectos de los fármacos , Expresión Génica/genética , Factor 1 Regulador del Interferón/efectos de los fármacos , Factor 1 Regulador del Interferón/genética , Ratones , Transducción de Señal/efectos de los fármacos , Factores de Transcripción/efectos de los fármacos , Factores de Transcripción/genética
5.
Biomed Pharmacother ; 147: 112652, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35065514

RESUMEN

Alterations in epigenetic marking, due to changes in expression or activity of epigenetic regulators, may affect cancer development and progression and thus, targeting epigenetic regulators provides potential avenues for cancer treatment. Bromodomain and extra terminal domain (BET) proteins, epigenetic readers recognizing histone acetylation, and Sirtuins (SIRT1-7), histone deacetylases or erasers, affect the chromatin acetylation status, and thus have a vital role in transcriptional regulation of a variety of cancer-related genes. Here, the effects of three BET inhibitors on SIRT expression were screened in a broad set of cancer cell lines to study the potential interplay of these distinct epigenetic factors in gene regulation. We show that BET inhibitors have distinct effects on SIRTs and their target gene expression in cancer cell lines derived from several solid tumour cancers. This functional link may open further avenues for epigenetic combination therapies for different cancers.


Asunto(s)
Azepinas/farmacología , Benzazepinas/farmacología , Benzodiazepinas/farmacología , Isoxazoles/farmacología , Proteínas/metabolismo , Sirtuinas/efectos de los fármacos , Triazoles/farmacología , Proteínas de Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Humanos , Proteínas/efectos de los fármacos , Factores de Transcripción/efectos de los fármacos
6.
BMC Plant Biol ; 22(1): 23, 2022 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-34998386

RESUMEN

BACKGROUND: Our previous study has demonstrated that the transcription of AchnKCS involved in suberin biosynthesis was up-regulated by exogenous abscisic acid (ABA) during the wound suberization of kiwifruit, but the regulatory mechanism has not been fully elucidated. RESULTS: Through subcellular localization analysis in this work, AchnbZIP29 and AchnMYB70 transcription factors were observed to be localized in the nucleus. Yeast one-hybrid and dual-luciferase assay proved the transcriptional activation of AchnMYB70 and transcriptional suppression of AchnbZIP29 on AchnKCS promoter. Furthermore, the transcription level of AchnMYB70 was enhanced by ABA during wound suberization of kiwifruit, but AchnbZIP29 transcription was reduced by ABA. CONCLUSIONS: Therefore, it was believed that ABA enhanced the transcriptional activation of AchnMYB70 on AchnKCS by increasing AchnMYB70 expression. On the contrary, ABA relieved the inhibitory effect of AchnbZIP29 on transcription of AchnKCS by inhibiting AchnbZIP29 expression. These results gave further insight into the molecular regulatory network of ABA in wound suberization of kiwifruit.


Asunto(s)
Ácido Abscísico/metabolismo , Actinidia/crecimiento & desarrollo , Actinidia/genética , Regulación de la Expresión Génica de las Plantas/efectos de los fármacos , Metabolismo de los Lípidos/genética , Reguladores del Crecimiento de las Plantas/metabolismo , Factores de Transcripción/efectos de los fármacos , Actinidia/efectos de los fármacos , Productos Agrícolas/efectos de los fármacos , Productos Agrícolas/genética , Productos Agrícolas/crecimiento & desarrollo , Frutas/efectos de los fármacos , Frutas/genética , Frutas/crecimiento & desarrollo , Reguladores del Crecimiento de las Plantas/genética
7.
Mol Plant ; 15(1): 167-178, 2022 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-34530166

RESUMEN

Nitrogen is an essential nutrient for plant growth and development, and plays vital roles in crop yield. Assimilation of nitrogen is thus fine-tuned in response to heterogeneous environments. However, the regulatory mechanism underlying this essential process remains largely unknown. Here, we report that a zinc-finger transcription factor, drought and salt tolerance (DST), controls nitrate assimilation in rice by regulating the expression of OsNR1.2. We found that loss of function of DST results in a significant decrease of nitrogen use efficiency (NUE) in the presence of nitrate. Further study revealed that DST is required for full nitrate reductase activity in rice and directly regulates the expression of OsNR1.2, a gene showing sequence similarity to nitrate reductase. Reverse genetics and biochemistry studies revealed that OsNR1.2 encodes an NADH-dependent nitrate reductase that is required for high NUE of rice. Interestingly, the DST-OsNR1.2 regulatory module is involved in the suppression of nitrate assimilation under drought stress, which contributes to drought tolerance. Considering the negative role of DST in stomata closure, as revealed previously, the positive role of DST in nitrogen assimilation suggests a mechanism coupling nitrogen metabolism and stomata movement. The discovery of this coupling mechanism will aid the engineering of drought-tolerant crops with high NUE in the future.


Asunto(s)
Adaptación Fisiológica/genética , Sequías , Nitrato-Reductasa/genética , Nitrato-Reductasa/metabolismo , Nitrógeno/metabolismo , Oryza/crecimiento & desarrollo , Oryza/genética , Oryza/metabolismo , Productos Agrícolas/genética , Productos Agrícolas/crecimiento & desarrollo , Productos Agrícolas/metabolismo , Regulación de la Expresión Génica de las Plantas , Genes de Plantas , Variación Genética , Genotipo , Tolerancia a la Sal/genética , Factores de Transcripción/efectos de los fármacos , Dedos de Zinc/efectos de los fármacos
8.
Adv Sci (Weinh) ; 9(4): e2103669, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34761556

RESUMEN

Transcription activator RamA is linked to multidrug resistance of Klebsiella pneumoniae through controlling genes that encode efflux pumps (acrA) and porin-regulating antisense RNA (micF). In bacteria, σ70 , together with activators, controls the majority of genes by recruiting RNA polymerase (RNAP) to the promoter regions. RNAP and σ70 form a holoenzyme that recognizes -35 and -10 promoter DNA consensus sites. Many activators bind upstream from the holoenzyme and can be broadly divided into two classes. RamA acts as a class I activator on acrA and class II activator on micF, respectively. The authors present biochemical and structural data on RamA in complex with RNAP-σ70 at the two promoters and the data reveal the molecular basis for how RamA assembles and interacts with core RNAP and activates transcription that contributes to antibiotic resistance. Further, comparing with CAP/TAP complexes reveals common and activator-specific features in activator binding and uncovers distinct roles of the two C-terminal domains of RNAP α subunit.


Asunto(s)
Antibacterianos/farmacología , Proteínas Bacterianas/metabolismo , Farmacorresistencia Bacteriana Múltiple/efectos de los fármacos , Klebsiella pneumoniae/efectos de los fármacos , Activación Transcripcional/efectos de los fármacos , Antibacterianos/metabolismo , Proteínas Bacterianas/efectos de los fármacos , Klebsiella pneumoniae/metabolismo , Transactivadores/metabolismo , Factores de Transcripción/efectos de los fármacos , Factores de Transcripción/metabolismo
9.
Brief Bioinform ; 23(1)2022 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-34849560

RESUMEN

Prostate cancer is the second leading cause of cancer-related death in men. Metastasis shows poor survival even though the recovery rate is high. In spite of numerous studies regarding prostate carcinoma, multiple questions are still unanswered. In this regards, gene regulatory network can uncover the mechanisms behind cancer progression, and metastasis. Under a feed forward loop, transcription factors (TFs) can be a good druggable candidate. We have proposed a computational model to study the uncertainty of TFs and suggest the appropriate cellular conditions for drug targeting. We have selected feed-forward loops depending on the shared list of the functional annotations among TFs, genes and miRNAs. From the potential feed forward loop cores, six TFs were identified as druggable targets, which include AR, CEBPB, CREB1, ETS1, NFKB1 and RELA. However, TFs are known for their Protein Moonlighting properties, which provide unrelated multi-functionalities within the same or different subcellular localizations. Following that, we have identified such functions that are suitable for drug targeting. On the other hand, we have tried to identify membraneless organelles for providing more specificity to the proposed time and space theory. The study has provided certain possibilities on TF-based therapeutics. The controlled dynamic nature of the TF may have enhanced the chances where TFs can be considered as one of the prime drug targets. Finally, the combination of membranless phase separation and protein moonlighting has provided possible druggable period within the biological clock.


Asunto(s)
Redes Reguladoras de Genes , Neoplasias de la Próstata , Factores de Transcripción , Regulación de la Expresión Génica , Redes Reguladoras de Genes/genética , Humanos , Masculino , MicroARNs/genética , MicroARNs/metabolismo , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/genética , Factores de Transcripción/efectos de los fármacos , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
10.
Plant Sci ; 313: 111093, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34763877

RESUMEN

Although several studies have confirmed that exogenous melatonin promotes anthocyanin accumulation, the molecular mechanism of this remains elusive. Here, the signaling cross-talk between melatonin and NADPH oxidase (RBOH) -mediated ROS during anthocyanin biosynthesis were investigated. We found that application of exogenous melatonin not only induced anthocyanin biosynthesis, but also increased endogenous H2O2 and O2‾ content in pear fruits. The effect of melatonin on anthocyanin biosynthesis was abolished by inhibitors of RBOH. We also observed that genes encoding RBOH (PuRBOHF) were ubiquitously and highly expressed after melatonin treatment. Transient PuRBOHF overexpression significantly enhanced anthocyanin accumulation and activated transcription of anthocyanin biosynthesis genes, whereas PuRBOHF silencing repressed melatonin-promoted anthocyanin accumulation and H2O2 production. Moreover, RBOH-derived H2O2 induced PuMYB10 transcription, and PuRBOHF enhanced the PuMYB10-induced activation of the PuUFGT promoter. PuMYB10, in turn, activated PuRBOHF transcription, revealing a positive feedback loop. These results provide molecular evidence supporting the essential roles of PuRBOHF-dependent H2O2 in melatonin-induced anthocyanin accumulation in pears.


Asunto(s)
Antocianinas/biosíntesis , Peróxido de Hidrógeno/metabolismo , Melatonina/metabolismo , Pyrus/genética , Pyrus/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Transcripción/efectos de los fármacos , Antocianinas/genética , China , Productos Agrícolas/genética , Productos Agrícolas/metabolismo , Frutas/genética , Frutas/metabolismo , Regulación de la Expresión Génica de las Plantas , Melatonina/genética
11.
Plant Sci ; 313: 111094, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34763879

RESUMEN

Anthocyanins are important flavonoid pigments involved in the colouring of flowers and fruits. They are synthesized on the cytoplasmic surface of the endoplasmic reticulum and transported into the vacuole for storage. Previous reports have suggested that glutathione S-transferase (GST) is involved in anthocyanin transport. However, due to the limitation of plant materials, most GSTs only participate in the cyanidin or delphinidin transport pathway. Here, an anthocyanin-related GST, ScGST3, was identified from the transcriptome of cineraria. The expression pattern of ScGST3 was highly consistent with anthocyanin accumulation in ray florets. Molecular complementation of Arabidopsis tt19 indicated that the overexpression of ScGST3 restores the anthocyanin-deficient phenotype of the mutant. Virus-induced gene silencing (VIGS) of ScGST3 in carmine and blue cineraria leaves could inhibit anthocyanin accumulation, further confirming the function of ScGST3 in anthocyanin accumulation. In vitro assays showed that ScGST3 increases the water solubility of cyanidin-3-O-glucoside (C3G) and delphinidin-3-O-glucosid (D3G). In addition, we also identified two anthocyanin-related MYB transcription factors, ScMYB3 and ScMYB6. The expression pattern of these two genes was also highly consistent with anthocyanin accumulation. Faded abaxial leaf phenotypes were observed after the silencing of ScMYB3 and ScMYB6, and the expression levels of partial structural genes were repressed. Based on the results from dual-luciferase assays and yeast one-hybrid assays, ScMYB3 can activate the promoter of ScGST3. Collectively, the transcription of ScGST3 is regulated by ScMYB3, which plays an important role in the transport of C3G and D3G in cineraria.


Asunto(s)
Antocianinas/biosíntesis , Antocianinas/genética , Flores/metabolismo , Glutatión Transferasa/metabolismo , Pigmentación/genética , Senecio/genética , Senecio/metabolismo , Factores de Transcripción/efectos de los fármacos , China , Flores/genética , Regulación de la Expresión Génica de las Plantas , Genes de Plantas , Variación Genética , Genotipo , Glutatión Transferasa/genética
12.
Mol Microbiol ; 116(5): 1378-1391, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34626146

RESUMEN

In Staphylococcus aureus, the two-component system SaeRS is responsible for regulating various virulence factors essential for the success of this pathogen. SaeRS can be stimulated by neutrophil-derived products but has also recently been shown to be inactivated by the presence of free fatty acids. A mechanism for how fatty acids negatively impacts SaeRS has not been described. We found that unsaturated fatty acids, as well as fatty acids not commonly found in Staphylococcal membranes, prevent the activation of SaeRS at a lower concentration than their saturated counterparts. These fatty acids can negatively impact SaeRS without altering the respiratory capacity of the bacterium. To uncover a potential mechanism for how fatty acids impact SaeRS function/activity, we utilized a naturally occurring point mutation found in S. aureus as well as chimeric SaeS proteins. Using these tools, we identified that the native transmembrane domains of SaeS dictate the transcriptional response to fatty acids in S. aureus. Our data support a model where free fatty acids alter the activity of the two-component system SaeRS directly through the sensor kinase SaeS and is dependent on the transmembrane domains of the protein.


Asunto(s)
Proteínas Bacterianas/efectos de los fármacos , Proteínas Bacterianas/metabolismo , Ácidos Grasos/farmacología , Proteínas Quinasas/efectos de los fármacos , Proteínas Quinasas/metabolismo , Staphylococcus aureus/metabolismo , Factores de Transcripción/efectos de los fármacos , Factores de Transcripción/metabolismo , Regulación Bacteriana de la Expresión Génica , Humanos , Respiración , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/enzimología , Staphylococcus aureus/patogenicidad , Virulencia
13.
BMC Cancer ; 21(1): 1061, 2021 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-34565342

RESUMEN

BACKGROUND: Neuroblastoma (NB) patients with MYCN amplification or overexpression respond poorly to current therapies and exhibit extremely poor clinical outcomes. PI3K-mTOR signaling-driven deregulation of protein synthesis is very common in NB and various other cancers that promote MYCN stabilization. In addition, both the MYCN and mTOR signaling axes can directly regulate a common translation pathway that leads to increased protein synthesis and cell proliferation. However, a strategy of concurrently targeting MYCN and mTOR signaling in NB remains unexplored. This study aimed to investigate the therapeutic potential of targeting dysregulated protein synthesis pathways by inhibiting the MYCN and mTOR pathways together in NB. METHODS: Using small molecule/pharmacologic approaches, we evaluated the effects of combined inhibition of MYCN transcription and mTOR signaling on NB cell growth/survival and associated molecular mechanism(s) in NB cell lines. We used two well-established BET (bromodomain extra-terminal) protein inhibitors (JQ1, OTX-015), and a clinically relevant mTOR inhibitor, temsirolimus, to target MYCN transcription and mTOR signaling, respectively. The single agent and combined efficacies of these inhibitors on NB cell growth, apoptosis, cell cycle and neurospheres were assessed using MTT, Annexin-V, propidium-iodide staining and sphere assays, respectively. Effects of inhibitors on global protein synthesis were quantified using a fluorescence-based (FamAzide)-based protein synthesis assay. Further, we investigated the specificities of these inhibitors in targeting the associated pathways/molecules using western blot analyses. RESULTS: Co-treatment of JQ1 or OTX-015 with temsirolimus synergistically suppressed NB cell growth/survival by inducing G1 cell cycle arrest and apoptosis with greatest efficacy in MYCN-amplified NB cells. Mechanistically, the co-treatment of JQ1 or OTX-015 with temsirolimus significantly downregulated the expression levels of phosphorylated 4EBP1/p70-S6K/eIF4E (mTOR components) and BRD4 (BET protein)/MYCN proteins. Further, this combination significantly inhibited global protein synthesis, compared to single agents. Our findings also demonstrated that both JQ1 and temsirolimus chemosensitized NB cells when tested in combination with cisplatin chemotherapy. CONCLUSIONS: Together, our findings demonstrate synergistic efficacy of JQ1 or OTX-015 and temsirolimus against MYCN-driven NB, by dual-inhibition of MYCN (targeting transcription) and mTOR (targeting translation). Additional preclinical evaluation is warranted to determine the clinical utility of targeted therapy for high-risk NB patients.


Asunto(s)
Acetanilidas/farmacología , Azepinas/farmacología , Compuestos Heterocíclicos con 3 Anillos/farmacología , Proteína Proto-Oncogénica N-Myc/antagonistas & inhibidores , Neuroblastoma/tratamiento farmacológico , Sirolimus/análogos & derivados , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Triazoles/farmacología , Proteínas Adaptadoras Transductoras de Señales/efectos de los fármacos , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Proteínas de Ciclo Celular/efectos de los fármacos , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Cisplatino/farmacología , Regulación hacia Abajo , Sinergismo Farmacológico , Factor 4E Eucariótico de Iniciación/efectos de los fármacos , Factor 4E Eucariótico de Iniciación/metabolismo , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Humanos , Proteína Proto-Oncogénica N-Myc/metabolismo , Neuroblastoma/metabolismo , Neuroblastoma/patología , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Quinasas S6 Ribosómicas 70-kDa/efectos de los fármacos , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Transducción de Señal , Sirolimus/farmacología , Esferoides Celulares/efectos de los fármacos , Serina-Treonina Quinasas TOR/metabolismo , Factores de Transcripción/efectos de los fármacos , Factores de Transcripción/metabolismo
14.
Genes (Basel) ; 12(9)2021 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-34573394

RESUMEN

In eukaryotes, ribosome biogenesis is driven by the synthesis of the ribosomal RNA (rRNA) by RNA polymerase I (Pol-I) and is tightly linked to cell growth and proliferation. The 3D-structure of the rDNA promoter plays an important, yet not fully understood role in regulating rRNA synthesis. We hypothesized that DNA intercalators/groove binders could affect this structure and disrupt rRNA transcription. To test this hypothesis, we investigated the effect of a number of compounds on Pol-I transcription in vitro and in cells. We find that intercalators/groove binders are potent inhibitors of Pol-I specific transcription both in vitro and in cells, regardless of their specificity and the strength of its interaction with DNA. Importantly, the synthetic ability of Pol-I is unaffected, suggesting that these compounds are not targeting post-initiating events. Notably, the tested compounds have limited effect on transcription by Pol-II and III, demonstrating the hypersensitivity of Pol-I transcription. We propose that stability of pre-initiation complex and initiation are affected as result of altered 3D architecture of the rDNA promoter, which is well in line with the recently reported importance of biophysical rDNA promoter properties on initiation complex formation in the yeast system.


Asunto(s)
Células Eucariotas/efectos de los fármacos , Sustancias Intercalantes/farmacología , ARN Ribosómico/biosíntesis , Iniciación de la Transcripción Genética/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Células Eucariotas/metabolismo , Células HCT116 , Células HeLa , Humanos , Unión Proteica/efectos de los fármacos , ARN Polimerasa I/efectos de los fármacos , ARN Polimerasa I/metabolismo , Factores de Transcripción/efectos de los fármacos , Factores de Transcripción/metabolismo
15.
Eur J Pharmacol ; 910: 174446, 2021 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-34461124

RESUMEN

Ponatinib is used for advanced treatment of chronic myeloid leukemia (CML), although low doses to prevent side effects do not suppress survival pathways and eradicate leukemia stem cells (LSCs). We evaluated the potential of ponatinib and PI3K/mTOR dual-inhibitor VS-5584 combination (PoVS) therapy to increase the anti-leukemic effects of ponatinib and investigated the underlying mechanisms at the molecular level. We measured the cytotoxicities of ponatinib, VS-5584, and PoVS (CCK-8 assay), and used the median-effect equation for combination analyses. We investigated the effects of inhibitory concentrations on apoptosis, cell viability and cell-cycle regulation (flow cytometry), protein levels (ELISA, Western blot), transcriptional activities (dual-luciferase reporter assay), gene expressions (qRT-PCR). VS-5584 exerted selective cytotoxic effects against CML and LSC cell lines. VS-5584 inhibited the PI3K/Akt/mTOR pathway, resulting in reduced cell viability, slightly induced caspase-independent apoptosis, prominent G0/G1 cell-cycle blockade that is not a consequence of quiescence. Normal hematopoietic stem cell line was the least affected. Moreover, ponatinib and VS-5584 mediated synergistic anti-leukemic effects on leukemic cells. VS-5584 reduced the ponatinib dose required to target leukemic cells. PoVS treatment inhibited PI3K/Akt/mTOR pathway more consistently than either of the two agents alone through reducing p-Akt, p-mTOR, p-S6K, p-PRAS40, p-S6. The subsequent downstream effects were an increase in C/EBP transcriptional activity and decreases in activities of E2F/DP1, Myc/Max, CREB, STAT3, NFκB, AP-1, Elk-1/SRF. Transcriptional regulation resulted in alterations in the expression levels of target mRNAs. Our results highlight PoVS can be a promising treatment strategy for eliminating CML cells and LSCs selectively, with the reduced ponatinib doses.


Asunto(s)
Antineoplásicos/farmacología , Imidazoles/farmacología , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Morfolinas/farmacología , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3/farmacología , Purinas/farmacología , Piridazinas/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Apoptosis/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Sinergismo Farmacológico , Puntos de Control de la Fase G1 del Ciclo Celular/efectos de los fármacos , Humanos , Concentración 50 Inhibidora , Quinasas Janus/genética , Quinasas Janus/metabolismo , Células K562 , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Proteínas Quinasas Activadas por Mitógenos/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , FN-kappa B/genética , FN-kappa B/metabolismo , Fosfatidilinositol 3-Quinasas/genética , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Factores de Transcripción STAT/genética , Factores de Transcripción STAT/metabolismo , Transducción de Señal/efectos de los fármacos , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Factores de Transcripción/efectos de los fármacos
16.
Plant Sci ; 310: 110983, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34315599

RESUMEN

The Arabidopsis thaliana R2R3-MYB transcription factor AtMYB32 and its homologs AtMYB4 and AtMYB7 play crucial roles in the regulation of phenylpropanoid metabolism. In addition, AtMYB4 and AtMYB7 are involved in the response to abiotic stress. However, the function of AtMYB32 remains unclear. In this study, we found that AtMYB32 is induced by abscisic acid (ABA) and repressed by drought stress. AtMYB32 positively regulates ABA-mediated seed germination and early seedling development. The expression of ABSCISIC ACID-INSENSITIVE 3 (ABI3), ABI4 and ABI5, which encode key positive regulators of ABA signaling, was upregulated in response to ABA in AtMYB32-overexpressing plants and downregulated in the atmyb32-1 mutant. In addition, we found that the atmyb32-1 mutant was drought resistant. Consistent with the drought-resistant phenotype, the transcript levels of C-repeat binding factor 4 (CBF4) were higher in the atmyb32-1 mutant in response to drought stress. Electrophoretic mobility shift assays (EMSAs) and chromatin immunoprecipitation (ChIP) assays revealed that AtMYB32 binds directly to the ABI3, ABI4, ABI5 and CBF4 promoters both in vitro and in vivo. Genetically, ABI4 was found to be epistatic to AtMYB32 for ABA-induced inhibition of seed germination and early seedling development. Taken together, our findings revealed that AtMYB32 regulates the ABA response by directly promoting ABI3, ABI4 and ABI5 expression and that the drought stress response likely occurs because of repression of CBF4 expression.


Asunto(s)
Ácido Abscísico/farmacología , Proteínas de Arabidopsis/metabolismo , Arabidopsis/efectos de los fármacos , Arabidopsis/metabolismo , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Proteínas de Arabidopsis/efectos de los fármacos , Proteínas de Arabidopsis/genética , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/efectos de los fármacos , Sequías , Regulación de la Expresión Génica de las Plantas/efectos de los fármacos , Regulación de la Expresión Génica de las Plantas/genética , Transactivadores/efectos de los fármacos , Factores de Transcripción/efectos de los fármacos
17.
Invest New Drugs ; 39(6): 1523-1537, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34213719

RESUMEN

Background Triptolide is an active natural product, which inhibits cell proliferation, induces cell apoptosis, suppresses tumor metastasis and improves the effect of other therapeutic treatments in several cancer cell lines by affecting multiple molecules and signaling pathways, such as caspases, heat-shock proteins, DNA damage and NF-ĸB. Purpose We investigated the effect of triptolide towards NF-ĸB and GATA1. Methods We used cell viability assay, compare and cluster analyses of microarray-based mRNA transcriptome-wide expression data, gene promoter binding motif analysis, molecular docking, Ingenuity pathway analysis, NF-ĸB reporter cell assay, and electrophoretic mobility shift assay (EMSA) of GATA1. Results Triptolide inhibited the growth of drug-sensitive (CCRF-CEM, U87.MG) and drug-resistant cell lines (CEM/ADR5000, U87.MGΔEGFR). Hierarchical cluster analysis showed six major clusters in dendrogram. The sensitive and resistant cell lines were statistically significant (p = 0.65 × 10-2) distributed. The binding motifs of NF-κB (Rel) and of GATA1 proteins were significantly enriched in regions of 25 kb upstream promoter of all genes. IPA showed the networks, biological functions, and canonical pathways influencing the activity of triptolide towards tumor cells. Interestingly, upstream analysis for the 40 genes identified by compare analysis revealed ZFPM1 (friend of GATA protein 1) as top transcription regulator. However, we did not observe any effect of triptolide to the binding of GATA1 in vitro. We confirmed that triptolide inhibited NF-κB activity, and it strongly bound to the pharmacophores of IκB kinase ß and NF-κB in silico. Conclusion Triptolide showed promising inhibitory effect toward NF-κB, making it a potential candidate for targeting NF-κB.


Asunto(s)
Diterpenos/farmacología , Factor de Transcripción GATA1/efectos de los fármacos , FN-kappa B/efectos de los fármacos , Farmacología en Red/métodos , Fenantrenos/farmacología , Unión Proteica/efectos de los fármacos , Factores de Transcripción/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Ensayo de Cambio de Movilidad Electroforética , Compuestos Epoxi/farmacología , Humanos , Simulación del Acoplamiento Molecular , ARN Mensajero
18.
Melanoma Res ; 31(4): 319-327, 2021 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-34054057

RESUMEN

Large/giant congenital nevi (L/GCMN) are benign neoplasms of the melanocytic neural crest lineage covering extensive areas of skin presenting risk for melanoma. Surgical resection often leads to scarring and trauma. Histone deacetylase inhibitors (iHDACs) as topical therapeutic agents may prove beneficial as an alternative/adjunct to surgery in this disease. Here we describe the effect of in vitro treatment of iHDACs drugs on primary nevocytes isolated from L/GCMN patients. Micropthalmia transcription factor (MITF) expression in L/GCMN patients' lesions was detected by immunohistochemistry, in cultured nevocytes by immunofluorescence, immunoblot and quantitative polymerase chain reaction. Cellular senescence was detected by SA-ß galactosidase activity. Markers for melanocytic differentiation were evaluated by immunoblot analysis and extracted melanin content was estimated spectrophotometrically. Cell death was measured by lactate dehydrogenase (LDH) assay and necrosis confirmed by polymerase (PARP) cleavage and acridine orange staining of the nuclei. MITF was expressed ubiquitously in nevocytes and melanocytes in patients' lesions. In culture, iHDAC treatment suppressed MITF protein and mRNA expression resulting in a senescent-like phenotype with positive ß-galactosidase staining, progressing to necrotic cell death as evidenced by increased LDH activity, appearance of cleaved PARP and necrotic nuclei. This is the first report showing evidence of iHDACs-induced MITF suppression in congenital nevocytes in vitro leading to a morphologic change with positive ß-galactosidase staining, followed by necrotic cell death in nevocytes, indicating that iHDAC drugs could be valuable therapeutic agents for treatment of L/GCMN lesions.


Asunto(s)
Inhibidores de Histona Desacetilasas/uso terapéutico , Nevo Pigmentado/tratamiento farmacológico , Neoplasias Cutáneas/tratamiento farmacológico , Factores de Transcripción/efectos de los fármacos , Vorinostat/uso terapéutico , Muerte Celular , Diferenciación Celular , Preescolar , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Lactante , Vorinostat/farmacología
19.
Nat Chem Biol ; 17(6): 711-717, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34035522

RESUMEN

The zinc-finger transcription factor Helios is critical for maintaining the identity, anergic phenotype and suppressive activity of regulatory T (Treg) cells. While it is an attractive target to enhance the efficacy of currently approved immunotherapies, no existing approaches can directly modulate Helios activity or abundance. Here, we report the structure-guided development of small molecules that recruit the E3 ubiquitin ligase substrate receptor cereblon to Helios, thereby promoting its degradation. Pharmacological Helios degradation destabilized the anergic phenotype and reduced the suppressive activity of Treg cells, establishing a route towards Helios-targeting therapeutics. More generally, this study provides a framework for the development of small-molecule degraders for previously unligandable targets by reprogramming E3 ligase substrate specificity.


Asunto(s)
Proteínas de Unión al ADN/efectos de los fármacos , Factor de Transcripción Ikaros/efectos de los fármacos , Linfocitos T Reguladores/efectos de los fármacos , Factores de Transcripción/efectos de los fármacos , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Línea Celular , Proteínas de Unión al ADN/genética , Humanos , Factor de Transcripción Ikaros/genética , Células Jurkat , Ratones , Modelos Moleculares , Estructura Molecular , Mutación/genética , Bibliotecas de Moléculas Pequeñas , Especificidad por Sustrato , Factores de Transcripción/genética , Ubiquitina-Proteína Ligasas/metabolismo
20.
Methods Mol Biol ; 2318: 337-346, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34019301

RESUMEN

Oncoproteins encoded by dominant oncogenes have long been considered as targets for chemotherapeutic intervention. However, oncogenic transcription factors have often been dismissed as "undruggable." Members of the Myc family of transcription factors have been identified as promising targets for cancer chemotherapy in multiple publications reporting the requirement of Myc proteins for maintenance of almost every type of tumor. Here, we describe cell-based approaches to identify c-Myc small molecule inhibitors by screening complex libraries of diverse small molecules based on Myc functionality and specificity.


Asunto(s)
Ensayos de Selección de Medicamentos Antitumorales/métodos , Genes myc/efectos de los fármacos , Proteínas Proto-Oncogénicas c-myc/antagonistas & inhibidores , Línea Celular Tumoral , Genes myc/genética , Genes myc/fisiología , Humanos , Proteínas Oncogénicas/efectos de los fármacos , Proteínas Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología , Factores de Transcripción/efectos de los fármacos , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...