Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 81
Filtrar
1.
Food Chem Toxicol ; 182: 114196, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37992955

RESUMEN

α-Amanitin, the primary lethal toxin of Amanita, specifically targets the liver, causing oxidative stress, hepatocyte apoptosis, and irreversible liver damage. As little as 0.1 mg/kg of α-amanitin can be lethal for humans, and there is currently no effective antidote for α-amanitin poisoning. Cannabidiol is a non-psychoactive natural compound derived from Cannabis sativa that exhibits a wide range of anti-inflammatory, antioxidant, and anti-apoptotic effects. It may play a protective role in preventing liver damage induced by α-amanitin. To investigate the potential protective effects of cannabidiol on α-amanitin-induced hepatocyte apoptosis and oxidative stress, we established α-amanitin exposure models using C57BL/6J mice and L-02 cells in vitro. Our results showed that α-amanitin exposure led to oxidative stress, apoptosis, and DNA damage in both mouse hepatocytes and L-02 cells, resulting in the death of mice. We also found that cannabidiol upregulated the level of Nrf2 and antioxidant enzymes, alleviating apoptosis, and oxidative stress in mouse hepatocytes and L-02 cells and increasing the survival rate of mice. Our findings suggest that cannabidiol has hepatoprotective effects through the regulation of Nrf2 and antioxidant enzymes and may be a potential therapeutic drug for Amanita poisoning.


Asunto(s)
Alfa-Amanitina , Cannabidiol , Humanos , Animales , Ratones , Alfa-Amanitina/metabolismo , Alfa-Amanitina/farmacología , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Cannabidiol/farmacología , Cannabidiol/metabolismo , Antioxidantes/farmacología , Antioxidantes/metabolismo , Ratones Endogámicos C57BL , Hígado , Apoptosis , Estrés Oxidativo , Hepatocitos
2.
Int J Mol Sci ; 24(22)2023 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-38003593

RESUMEN

α-Amanitin is one of the primary toxins produced by the poisonous mushroom genus, Amanita. Because it is odorless and tasteless, it is an important cause of death from the consumption of misidentified mushrooms. To study the thermal stability of α-amanitin, novel cell-based assays were developed to measure the toxin's activity, based on the inhibition of RNA polymerase II by α-amanitin. First, an MTT-formazan cell viability assay was used to measure the biological activity of α-amanitin through the inhibition of cellular activity. This method can detect 10 µg/mL of α-amanitin in a time-dependent manner. Second, a more sensitive quantitative PCR approach was developed to examine its inhibition of viral replication. The new RT-qPCR assay enabled the detection of 100 ng/mL. At this level, α-amanitin still significantly reduced adenovirus transcription. Third, a simpler GFP expression-based assay was developed with an equal sensitivity to the RT-qPCR assay. With this assay, aqueous α-amanitin heated at 90 °C for 16 h or treated in the microwave for 3 min retained its biological activity when tested in HEK293 cells, but a slight reduction was observed when tested in Vero cells. Beyond detecting the activity of α-amanitin, the new method has a potential application for detecting the activity of other toxins that are RNA polymerase inhibitors.


Asunto(s)
Alfa-Amanitina , ARN Polimerasa II , Animales , Chlorocebus aethiops , Humanos , Alfa-Amanitina/farmacología , Células Vero , Células HEK293 , Amanita
3.
STAR Protoc ; 4(3): 102534, 2023 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-37656628

RESUMEN

Data normalization is critical to the process of estimating RNA degradation by analyzing RNA levels when transcription is blocked. Here, we present a protocol for measuring mRNA degradation rates, optimized for mouse embryonic stem cells, using α-amanitin inhibitor. We describe steps for a time course α-amanitin treatment, RNA-seq, and alignment; we then detail procedures for analyzing data and sequence enrichment. Our method relies on large-scale normalization of stable transcripts in genomic RNA-seq measurements, providing reliable readouts. For complete details on the use and execution of this protocol, please refer to Viegas et al.1.


Asunto(s)
Alfa-Amanitina , Células Madre Embrionarias de Ratones , Animales , Ratones , Alfa-Amanitina/farmacología , Genómica , ARN/genética , RNA-Seq
4.
Nat Commun ; 14(1): 2241, 2023 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-37193694

RESUMEN

The "death cap", Amanita phalloides, is the world's most poisonous mushroom, responsible for 90% of mushroom-related fatalities. The most fatal component of the death cap is α-amanitin. Despite its lethal effect, the exact mechanisms of how α-amanitin poisons humans remain unclear, leading to no specific antidote available for treatment. Here we show that STT3B is required for α-amanitin toxicity and its inhibitor, indocyanine green (ICG), can be used as a specific antidote. By combining a genome-wide CRISPR screen with an in silico drug screening and in vivo functional validation, we discover that N-glycan biosynthesis pathway and its key component, STT3B, play a crucial role in α-amanitin toxicity and that ICG is a STT3B inhibitor. Furthermore, we demonstrate that ICG is effective in blocking the toxic effect of α-amanitin in cells, liver organoids, and male mice, resulting in an overall increase in animal survival. Together, by combining a genome-wide CRISPR screen for α-amanitin toxicity with an in silico drug screen and functional validation in vivo, our study highlights ICG as a STT3B inhibitor against the mushroom toxin.


Asunto(s)
Hexosiltransferasas , Micotoxinas , Humanos , Masculino , Animales , Ratones , Alfa-Amanitina/farmacología , Verde de Indocianina/farmacología , Antídotos , Amanita , Proteínas de la Membrana
5.
Int J Mol Sci ; 23(20)2022 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-36293151

RESUMEN

The well-known hepatotoxicity mechanism resulting from alpha-amanitin (α-AMA) exposure arises from RNA polymerase II (RNAP II) inhibition. RNAP Ⅱ inhibition occurs through the dysregulation of mRNA synthesis. However, the signaling pathways in hepatocytes that arise from α-AMA have not yet been fully elucidated. Here, we identified that the RAS/RAF/ERK signaling pathway was activated through quantitative phosphoproteomic and molecular biological analyses in Huh-7 cells. Bioinformatics analysis showed that α-AMA exposure increased protein phosphorylation in a time-dependent α-AMA exposure. In addition, phosphorylation increased not only the components of the ERK signaling pathway but also U2AF65 and SPF45, known splicing factors. Therefore, we propose a novel mechanism of α-AMA as follows. The RAS/RAF/ERK signaling pathway involved in aberrant splicing events is activated by α-AMA exposure followed by aberrant splicing events leading to cell death in Huh-7 cells.


Asunto(s)
Alfa-Amanitina , ARN Polimerasa II , Alfa-Amanitina/farmacología , Sistema de Señalización de MAP Quinasas/fisiología , Fosforilación , Factores de Empalme de ARN , ARN Mensajero
6.
Plant Mol Biol ; 107(4-5): 387-404, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34189708

RESUMEN

KEY MESSAGE: The moss Pseudocrossidium replicatum is a desiccation-tolerant species that uses an inducible system to withstand severe abiotic stress in both protonemal and gametophore tissues. Desiccation tolerance (DT) is the ability of cells to recover from an air-dried state. Here, the moss Pseudocrossidium replicatum was identified as a fully desiccation-tolerant (FDT) species. Its gametophores rapidly lost more than 90% of their water content when exposed to a low-humidity atmosphere [23% relative humidity (RH)], but abscisic acid (ABA) pretreatment diminished the final water loss after equilibrium was reached. P. replicatum gametophores maintained good maximum photosystem II (PSII) efficiency (Fv/Fm) for up to two hours during slow dehydration; however, ABA pretreatment induced a faster decrease in the Fv/Fm. ABA also induced a faster recovery of the Fv/Fm after rehydration. Protein synthesis inhibitor treatment before dehydration hampered the recovery of the Fv/Fm when the gametophores were rehydrated after desiccation, suggesting the presence of an inducible protective mechanism that is activated in response to abiotic stress. This observation was also supported by accumulation of soluble sugars in gametophores exposed to ABA or NaCl. Exogenous ABA treatment delayed the germination of P. replicatum spores and induced morphological changes in protonemal cells that resembled brachycytes. Transcriptome analyses revealed the presence of an inducible molecular mechanism in P. replicatum protonemata that was activated in response to dehydration. This study is the first RNA-Seq study of the protonemal tissues of an FDT moss. Our results suggest that P. replicatum is an FDT moss equipped with an inducible molecular response that prepares this species for severe abiotic stress and that ABA plays an important role in this response.


Asunto(s)
Adaptación Fisiológica/genética , Bryopsida/genética , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica de las Plantas/genética , Ácido Abscísico/farmacología , Adaptación Fisiológica/efectos de los fármacos , Alfa-Amanitina/farmacología , Bryopsida/metabolismo , Cicloheximida/farmacología , Deshidratación , Regulación de la Expresión Génica de las Plantas/efectos de los fármacos , Geografía , México , Inhibidores de la Síntesis del Ácido Nucleico/farmacología , Reguladores del Crecimiento de las Plantas/farmacología , Inhibidores de la Síntesis de la Proteína/farmacología , RNA-Seq/métodos , Estrés Fisiológico , Factores de Tiempo
7.
J Vet Med Sci ; 83(1): 158-161, 2021 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-33250482

RESUMEN

Alpha-amanitin, one of the amatoxins in egg amanita, has a cyclic peptide structure, and was reported as having antiviral activity against several viruses. We investigated whether α-amanitin has antiviral activity against feline immunodeficiency virus (FIV). FL-4 cells persistently infected with FIV Petaluma were cultured with α-amanitin. Reverse transcriptase (RT) activity in the supernatant of FL-4 cells was significantly inhibited by α-amanitin. In addition, the production of FIV core protein in FL-4 cells was inhibited by α-amanitin when analyzed by western blotting. Furthermore, α-amanitin inhibited the transcription of FIV in real-time RT-PCR. These data suggested that α-amanitin showed anti-FIV activity by inhibiting the RNA transcription level.


Asunto(s)
Virus de la Inmunodeficiencia Felina , Alfa-Amanitina/farmacología , Animales , Antivirales/farmacología , Gatos
8.
Development ; 146(19)2019 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-31570370

RESUMEN

Histone post-translational modifications are key gene expression regulators, but their rapid dynamics during development remain difficult to capture. We applied a Fab-based live endogenous modification labeling technique to monitor the changes in histone modification levels during zygotic genome activation (ZGA) in living zebrafish embryos. Among various histone modifications, H3 Lys27 acetylation (H3K27ac) exhibited most drastic changes, accumulating in two nuclear foci in the 64- to 1k-cell-stage embryos. The elongating form of RNA polymerase II, which is phosphorylated at Ser2 in heptad repeats within the C-terminal domain (RNAP2 Ser2ph), and miR-430 transcripts were also concentrated in foci closely associated with H3K27ac. When treated with α-amanitin to inhibit transcription or JQ-1 to inhibit binding of acetyl-reader proteins, H3K27ac foci still appeared but RNAP2 Ser2ph and miR-430 morpholino were not concentrated in foci, suggesting that H3K27ac precedes active transcription during ZGA. We anticipate that the method presented here could be applied to a variety of developmental processes in any model and non-model organisms.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Genoma , Histonas/metabolismo , Lisina/metabolismo , Transcripción Genética , Pez Cebra/embriología , Pez Cebra/genética , Cigoto/metabolismo , Acetilación/efectos de los fármacos , Alfa-Amanitina/farmacología , Animales , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Embrión no Mamífero/efectos de los fármacos , Embrión no Mamífero/metabolismo , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Código de Histonas/efectos de los fármacos , ARN Polimerasa II/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transcripción Genética/efectos de los fármacos , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , Cigoto/efectos de los fármacos
9.
Sci Rep ; 9(1): 7779, 2019 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-31123282

RESUMEN

Despite the great success of small molecule inhibitors in the treatment of patients with BRAFV600E mutated melanoma, the response to these drugs remains transient and patients eventually relapse within a few months, highlighting the need to develop novel combination therapies based on the understanding of the molecular changes induced by BRAFV600E inhibitors. The acute inhibition of oncogenic signaling can rewire entire cellular signaling pathways and thereby create novel cancer cell vulnerabilities. Here, we demonstrate that inhibition of BRAFV600E oncogenic signaling in melanoma cell lines leads to destabilization of the large subunit of RNA polymerase II POLR2A (polymerase RNA II DNA-directed polypeptide A), thereby preventing its binding to the unconventional prefoldin RPB5 interactor (URI1) chaperone complex and the successful assembly of RNA polymerase II holoenzymes. Furthermore, in melanoma cell lines treated with mitogen-activated protein kinase (MAPK) inhibitors, α-amanitin, a specific and irreversible inhibitor of RNA polymerase II, induced massive apoptosis. Pre-treatment of melanoma cell lines with MAPK inhibitors significantly reduced IC50 values to α-amanitin, creating a state of collateral vulnerability similar to POLR2A hemizygous deletions. Thus, the development of melanoma specific α-amanitin antibody-drug conjugates could represent an interesting therapeutic approach for combination therapies with BRAFV600E inhibitors.


Asunto(s)
Alfa-Amanitina/farmacología , Muerte Celular/efectos de los fármacos , Melanoma/metabolismo , Inhibidores de la Síntesis del Ácido Nucleico/farmacología , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , ARN Polimerasa II/metabolismo , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Humanos , Transducción de Señal/efectos de los fármacos
10.
Sci Rep ; 9(1): 4868, 2019 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-30890736

RESUMEN

Transcription and DNA damage repair act in a coordinated manner. Recent studies have shown that double-strand DNA breaks (DSBs) are repaired in a transcription-coupled manner. Active transcription results in a faster recruitment of DSB repair factors and expedites DNA repair. On the other hand, transcription is repressed by DNA damage through multiple mechanisms. We previously reported that TLP, a TATA box-binding protein (TBP) family member that functions as a transcriptional regulator, is also involved in DNA damage-induced apoptosis. However, the mechanism by which TLP affects DNA damage response was largely unknown. Here we show that TLP-mediated global transcriptional repression after DSBs is crucial for apoptosis induction by DNA-damaging agents such as etoposide and doxorubicin. Compared to control cells, TLP-knockdown cells were resistant to etoposide-induced apoptosis and exhibited an elevated level of global transcription after etoposide exposure. DSBs were efficiently removed in transcriptionally hyperactive TLP-knockdown cells. However, forced transcriptional shutdown using transcriptional inhibitors α-amanitin and 5,6-dichloro-1-ß-D-ribofuranosylbenzimidazole (DRB) slowed down DSB repair and resensitized TLP-knockdown cells to etoposide. Taken together, these results indicate that TLP is a critical determinant as to how cells respond to DSBs and triggers apoptosis to cells that have sustained DNA damage.


Asunto(s)
Apoptosis/genética , Proteínas Relacionadas con la Autofagia/genética , Roturas del ADN de Doble Cadena/efectos de los fármacos , Transcripción Genética/efectos de los fármacos , Proteínas de Transporte Vesicular/genética , Alfa-Amanitina/farmacología , Apoptosis/efectos de los fármacos , Proteínas Relacionadas con la Autofagia/antagonistas & inhibidores , Daño del ADN/efectos de los fármacos , Daño del ADN/genética , Reparación del ADN/efectos de los fármacos , Diclororribofuranosil Benzoimidazol/farmacología , Doxorrubicina/farmacología , Etopósido/farmacología , Técnicas de Silenciamiento del Gen , Humanos , Transcripción Genética/genética , Proteínas de Transporte Vesicular/antagonistas & inhibidores
11.
Mol Cell ; 72(5): 888-901.e7, 2018 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-30344095

RESUMEN

Safeguarding cell function and identity following a genotoxic stress challenge entails a tight coordination of DNA damage signaling and repair with chromatin maintenance. How this coordination is achieved and with what impact on chromatin integrity remains elusive. Here, we address these questions by investigating the mechanisms governing the distribution in mammalian chromatin of the histone variant H2A.X, a central player in damage signaling. We reveal that H2A.X is deposited de novo at sites of DNA damage in a repair-coupled manner, whereas the H2A.Z variant is evicted, thus reshaping the chromatin landscape at repair sites. Our mechanistic studies further identify the histone chaperone FACT (facilitates chromatin transcription) as responsible for the deposition of newly synthesized H2A.X. Functionally, we demonstrate that FACT potentiates H2A.X-dependent signaling of DNA damage. We propose that new H2A.X deposition in chromatin reflects DNA damage experience and may help tailor DNA damage signaling to repair progression.


Asunto(s)
Reparación del ADN , Proteínas de Unión al ADN/genética , ADN/genética , Proteínas del Grupo de Alta Movilidad/genética , Histonas/genética , Factores de Elongación Transcripcional/genética , Alfa-Amanitina/farmacología , Animales , Proteínas de la Ataxia Telangiectasia Mutada/antagonistas & inhibidores , Proteínas de la Ataxia Telangiectasia Mutada/genética , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Línea Celular Tumoral , Ensamble y Desensamble de Cromatina/efectos de los fármacos , ADN/metabolismo , Daño del ADN , Proteínas de Unión al ADN/metabolismo , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Regulación de la Expresión Génica , Proteínas del Grupo de Alta Movilidad/metabolismo , Histonas/metabolismo , Humanos , Ratones , Morfolinas/farmacología , Células 3T3 NIH , Nucleosomas/química , Nucleosomas/efectos de los fármacos , Nucleosomas/metabolismo , Venenos/farmacología , Pirimidinas/farmacología , Pironas/farmacología , Transducción de Señal , Factores de Elongación Transcripcional/metabolismo
12.
World J Gastroenterol ; 24(34): 3834-3848, 2018 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-30228778

RESUMEN

Colorectal cancer (CRC) is often diagnosed at an advanced stage when tumor cell dissemination has taken place. Chemo- and targeted therapies provide only a limited increase of overall survival for these patients. The major reason for clinical outcome finds its origin in therapy resistance. Escape mechanisms to both chemo- and targeted therapy remain the main culprits. Here, we evaluate major resistant mechanisms and elaborate on potential new therapies. Amongst promising therapies is α-amanitin antibody-drug conjugate targeting hemizygous p53 loss. It becomes clear that a dynamic interaction with the tumor microenvironment exists and that this dictates therapeutic outcome. In addition, CRC displays a limited response to checkpoint inhibitors, as only a minority of patients with microsatellite instable high tumors is susceptible. In this review, we highlight new developments with clinical potentials to augment responses to checkpoint inhibitors.


Asunto(s)
Antineoplásicos Inmunológicos/farmacología , Neoplasias Colorrectales/tratamiento farmacológico , Inmunoconjugados/farmacología , Escape del Tumor/efectos de los fármacos , Alfa-Amanitina/farmacología , Alfa-Amanitina/uso terapéutico , Antineoplásicos Inmunológicos/uso terapéutico , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/mortalidad , Receptores Coestimuladores e Inhibidores de Linfocitos T/antagonistas & inhibidores , Receptores Coestimuladores e Inhibidores de Linfocitos T/inmunología , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Resistencia a Antineoplásicos/inmunología , Humanos , Inmunoconjugados/uso terapéutico , Inmunoterapia/métodos , Inestabilidad de Microsatélites/efectos de los fármacos , Inhibidores de la Síntesis del Ácido Nucleico/farmacología , Inhibidores de la Síntesis del Ácido Nucleico/uso terapéutico , ARN Polimerasa II/antagonistas & inhibidores , Resultado del Tratamiento , Escape del Tumor/genética , Escape del Tumor/inmunología , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología , Proteína p53 Supresora de Tumor/genética
13.
Syst Biol Reprod Med ; 64(5): 389-398, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30136857

RESUMEN

The episodic pattern of gonadotropin-releasing hormone (GnRH) secretion from the hypothalamus is driven by an integrated network of cells termed the GnRH pulse generator. Cultured and immortalized GnRH neurons also produce a pulsatile pattern of GnRH secretions when grown in the absence of other cell types, suggesting the presence of an intrinsic oscillator mediating GnRH secretion. The mechanisms underlying such pulsatility comprise one of the most tantalizing problems in contemporary neuroendocrinology. In order to study the mechanism by which GnRH is produced in a pulsatile fashion, the autocrine effect of GnRH on GnRH-producing neurons must be eliminated. This may be performed by downregulating the expression of the GnRH receptor. Treatment with three 21-mer exogenous phosphorothioates and transient transfections with an inducible plasmid containing an antisense construct to the GnRH receptor gene decreased GnRH receptor expression further. This resulted in less cytotoxicity compared to inhibition of RNA or protein synthesis with actinomycin D, α-amanitin, puromycin, and cycloheximide. This study shows methods and optimized conditions established for the generation of a stable GT1-7 cell line containing an inducible construct allowing the downregulation of GnRH receptor expression. ABBREVIATIONS: ANOVA: analysis of the variance; DMEM: Dulbecco's modified Eagle's medium; GnRH: gonadotropin-releasing hormone; RXR: retinoid X receptor.


Asunto(s)
Supervivencia Celular/efectos de los fármacos , Oligodesoxirribonucleótidos Antisentido/farmacología , Receptores LHRH/metabolismo , Alfa-Amanitina/farmacología , Animales , Línea Celular Transformada , Medios de Cultivo , Ciclofosfamida/farmacología , Dactinomicina/farmacología , Regulación hacia Abajo , Técnicas de Silenciamiento del Gen , Hormona Liberadora de Gonadotropina/biosíntesis , Hormona Liberadora de Gonadotropina/metabolismo , Ratones , Plásmidos , Inhibidores de la Síntesis de la Proteína/farmacología , Puromicina/farmacología , Receptores LHRH/antagonistas & inhibidores , Receptores LHRH/genética , Transfección
14.
Int J Mol Sci ; 19(7)2018 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-30029518

RESUMEN

In the rapidly developing field of targeted cancer therapy there is growing interest towards therapeutics combining two or more compounds to achieve synergistic action and minimize the chance of cancer resistance to treatment. We developed a fibroblast growth factor 2 (FGF2)-conjugate bearing two cytotoxic drugs with independent mode of action: α-amanitin and monomethyl auristatin E. Drugs are covalently attached to the targeting protein in a site-specific manner via maleimide-thiol conjugation and Cu(I)-catalyzed alkyne-azide cycloaddition. The dual warhead conjugate binds to FGF receptor 1 (FGFR1) and utilizes receptor-mediated endocytosis for selective internalization into cancer cells with FGFR1. The developed conjugate displays high cytotoxicity towards all tested FGFR1-positive cell lines. Most importantly, the improved cytotoxic effect of both drugs is observed for lung cancer cell line NCI-H446. The single drug-FGF2 conjugates have no impact on the viability of NCI-H446 cells, whereas the dual warhead-FGF2 conjugate selectively and efficiently kills these FGFR1 positive cancer cells. Due to the diversified mode of action the dual warhead-FGF2 conjugate may overcome the potential acquired resistance of FGFR1-overproducing cancer cells towards single cytotoxic drugs.


Asunto(s)
Alfa-Amanitina/farmacología , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Oligopéptidos/farmacología , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Alfa-Amanitina/química , Animales , Línea Celular Tumoral , Endocitosis , Factor 2 de Crecimiento de Fibroblastos/química , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Humanos , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Ratones , Modelos Biológicos , Células 3T3 NIH , Oligopéptidos/química , Estructura Secundaria de Proteína , Transducción de Señal
15.
J Am Chem Soc ; 140(21): 6513-6517, 2018 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-29561592

RESUMEN

α-Amanitin is an extremely toxic bicyclic octapeptide isolated from the death-cap mushroom, Amanita phalloides. As a potent inhibitor of RNA polymerase II, α-amanitin is toxic to eukaryotic cells. Recent interest in α-amanitin arises from its promise as a payload for antibody-drug conjugates. For over 60 years, A. phalloides has been the only source of α-amanitin. Here we report a synthesis of α-amanitin, which surmounts the key challenges for installing the 6-hydroxy-tryptathionine sulfoxide bridge, enantioselective synthesis of (2 S,3 R,4 R)-4,5-dihydroxy-isoleucine, and diastereoselective sulfoxidation.


Asunto(s)
Agaricales/química , Alfa-Amanitina/síntesis química , Micotoxinas/síntesis química , Alfa-Amanitina/química , Alfa-Amanitina/farmacología , Animales , Células CHO , Cricetulus , Relación Dosis-Respuesta a Droga , Modelos Moleculares , Conformación Molecular , Micotoxinas/química , Micotoxinas/farmacología , Relación Estructura-Actividad
16.
J Biol Chem ; 293(19): 7189-7194, 2018 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-29550768

RESUMEN

RNA polymerase II (Pol II) is the central enzyme that transcribes eukaryotic protein-coding genes to produce mRNA. The mushroom toxin α-amanitin binds Pol II and inhibits transcription at the step of RNA chain elongation. Pol II from yeast binds α-amanitin with micromolar affinity, whereas metazoan Pol II enzymes exhibit nanomolar affinities. Here, we present the high-resolution cryo-EM structure of α-amanitin bound to and inhibited by its natural target, the mammalian Pol II elongation complex. The structure revealed that the toxin is located in a pocket previously identified in yeast Pol II but forms additional contacts with metazoan-specific residues, which explains why its affinity to mammalian Pol II is ∼3000 times higher than for yeast Pol II. Our work provides the structural basis for the inhibition of mammalian Pol II by the natural toxin α-amanitin and highlights that cryo-EM is well suited to studying interactions of a small molecule with its macromolecular target.


Asunto(s)
Alfa-Amanitina/química , Inhibidores Enzimáticos/química , ARN Polimerasa II/antagonistas & inhibidores , ARN Polimerasa II/química , Elongación de la Transcripción Genética/efectos de los fármacos , Alfa-Amanitina/farmacología , Secuencia de Aminoácidos , Animales , Sitios de Unión , Microscopía por Crioelectrón , Inhibidores Enzimáticos/farmacología , Enlace de Hidrógeno , Conformación Proteica , Homología de Secuencia de Aminoácido , Porcinos
17.
Sci Rep ; 7(1): 15763, 2017 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-29150675

RESUMEN

To study the relationship between chromatin condensation, gene transcription and developmental competence during oocyte maturation and to explore the mechanisms by which meiotic arrest maintenance (MAM) and sexual maturity improve oocyte competence, we examined effects of MAM with roscovitine or db-cAMP on chromatin condensation, gene transcription and developmental potential of NSN or SN oocytes from prepubertal or adult mice. MAM with roscovitine improved the developmental competence and global gene transcription of prepubertal NSN (prep-NSN) and adult-SN oocytes while having no effect on those of prep-SN oocytes. MAM with db-cAMP facilitated neither development nor transcription in any type of oocytes. MAM with either roscovitine or db-cAMP promoted chromatin condensation of prep-NSN oocytes. MAM with roscovitine promoted gene transcription and chromatin condensation simultaneously through inhibiting cyclin-dependent kinase (CDK) 5 and 2, respectively. The results suggested that MAM with roscovitine improved oocyte competence by promoting gene transcription via inhibiting CDK5. Oocyte cytoplasmic maturation is correlated with gene transcription but not with chromatin condensation. The difference in developmental competence between prepubertal NSN and SN oocytes and between prepubertal and adult SN oocytes was because while the former had not, the latter had completed or acquired the ability for transcription of important genes.


Asunto(s)
Puntos de Control del Ciclo Celular , Meiosis , Oocitos/citología , Maduración Sexual , Alfa-Amanitina/farmacología , Animales , Bucladesina/farmacología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Nucléolo Celular/efectos de los fármacos , Nucléolo Celular/metabolismo , Cromatina/metabolismo , Células del Cúmulo/citología , Células del Cúmulo/efectos de los fármacos , Células del Cúmulo/metabolismo , Quinasa 5 Dependiente de la Ciclina/antagonistas & inhibidores , Quinasa 5 Dependiente de la Ciclina/metabolismo , Citoplasma/efectos de los fármacos , Citoplasma/metabolismo , Desarrollo Embrionario/efectos de los fármacos , Femenino , Isoquinolinas/farmacología , Ratones , Oocitos/efectos de los fármacos , Oocitos/metabolismo , ARN/genética , ARN/metabolismo , ARN Polimerasa II/metabolismo , Roscovitina/farmacología , Maduración Sexual/efectos de los fármacos , Sulfonamidas/farmacología , Transcripción Genética/efectos de los fármacos
18.
Nucleic Acids Res ; 45(22): 12715-12722, 2017 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-29036442

RESUMEN

H2A.Z histone variant is an important regulator of gene transcription, which is enriched at regulatory regions but is also found within gene bodies. Recent evidence suggests that active recruitment of H2A.Z within gene bodies is required to induce gene repression. In contrast to this view, we show that global inhibition of transcription results in H2A.Z accumulation at gene transcription start sites, as well as within gene bodies. Our results indicate that accumulation of H2A.Z within repressed genes can also be a consequence of the repression of gene transcription rather than an active mechanism required to establish the repression.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/genética , Histonas/genética , Sitio de Iniciación de la Transcripción , Iniciación de la Transcripción Genética , Adenosina Trifosfatasas/genética , Adenosina Trifosfatasas/metabolismo , Alfa-Amanitina/farmacología , Flavonoides/farmacología , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HCT116 , Histonas/metabolismo , Humanos , Modelos Genéticos , Nucleosomas/genética , Nucleosomas/metabolismo , Piperidinas/farmacología
19.
J Neurosci ; 37(40): 9675-9685, 2017 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-28887385

RESUMEN

Reactivated memories can be modified during reconsolidation, making this process a potential therapeutic target for posttraumatic stress disorder (PTSD), a mental illness characterized by the recurring avoidance of situations that evoke trauma-related fears. However, avoidance memory reconsolidation depends on a set of still loosely defined boundary conditions, limiting the translational value of basic research. In particular, the involvement of the hippocampus in fear-motivated avoidance memory reconsolidation remains controversial. Combining behavioral and electrophysiological analyses in male Wistar rats, we found that previous learning of relevant nonaversive information is essential to elicit the participation of the hippocampus in avoidance memory reconsolidation, which is associated with an increase in theta- and gamma-oscillation power and cross-frequency coupling in dorsal CA1 during reactivation of the avoidance response. Our results indicate that the hippocampus is involved in memory reconsolidation only when reactivation results in contradictory representations regarding the consequences of avoidance and suggest that robust nesting of hippocampal theta-gamma rhythms at the time of retrieval is a specific reconsolidation marker.SIGNIFICANCE STATEMENT Posttraumatic stress disorder (PTSD) is characterized by maladaptive avoidance responses to stimuli or behaviors that represent or bear resemblance to some aspect of a traumatic experience. Disruption of reconsolidation, the process by which reactivated memories become susceptible to modifications, is a promising approach for treating PTSD patients. However, much of what is known about fear-motivated avoidance memory reconsolidation derives from studies based on fear conditioning instead of avoidance-learning paradigms. Using a step-down inhibitory avoidance task in rats, we found that the hippocampus is involved in memory reconsolidation only when the animals acquired the avoidance response in an environment that they had previously learned as safe and showed that increased theta- and gamma-oscillation coupling during reactivation is an electrophysiological signature of this process.


Asunto(s)
Reacción de Prevención/fisiología , Hipocampo/fisiología , Consolidación de la Memoria/fisiología , Alfa-Amanitina/farmacología , Animales , Reacción de Prevención/efectos de los fármacos , Ondas Encefálicas/efectos de los fármacos , Ondas Encefálicas/fisiología , Hipocampo/efectos de los fármacos , Aprendizaje/efectos de los fármacos , Aprendizaje/fisiología , Masculino , Consolidación de la Memoria/efectos de los fármacos , Inhibidores de la Síntesis del Ácido Nucleico/farmacología , Ratas , Ratas Wistar
20.
Dev Genes Evol ; 227(4): 279-287, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28624889

RESUMEN

Early embryos of the clitellate annelid Tubifex (oligochaete) undergo a series of unequal spiral cell divisions before the descendants of the D quadrant micromeres (cells 2d and 4d) divide bilaterally. Here, we show that inhibition of zygotic transcription by microinjection of α-amanitin (transcription inhibitor) exclusively converts unequal cleavage in cell 2d11 (granddaughter of 2d) into equal cleavage while other unequal cleavages and ensuing bilateral cleavages in cells 4d and 2d111 (great-granddaughter of 2d) all proceed in a normal fashion in the presence of this inhibitor. These results differ significantly from those reported for embryos of another clitellate annelid Helobdella (leech), in which inhibition of transcription converts bilateral (symmetric) cleavages in cells DNOPQ"' and DM" (equivalent to 2d111 and 4d) into unequal (asymmetric) cleavages while having no apparent effect on unequal cleavage in DNOPQ" (equivalent to 2d11). These differences imply distinct mechanisms for the control of the unequal-to-bilateral transition in the two clitellate annelids.


Asunto(s)
Oligoquetos/crecimiento & desarrollo , Oligoquetos/genética , Alfa-Amanitina/farmacología , Animales , Tipificación del Cuerpo , División Celular , Tamaño de la Célula , Embrión no Mamífero/citología , Sanguijuelas/citología , Sanguijuelas/genética , Sanguijuelas/crecimiento & desarrollo , Oligoquetos/citología , Transcripción Genética/efectos de los fármacos , Cigoto/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...