Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 373
Filtrar
1.
Neurotoxicology ; 103: 230-255, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38955288

RESUMEN

The 3,4-methylenedioxy-alpha-pyrrolidinohexanophenone (MDPHP) is a synthetic cathinone closely related to 3,4-methylenedioxypyrovalerone (MDPV), one of the most common synthetic cathinones present in the "bath salts". MDPHP has recently gained attention due to increasing seizures and involvement in human intoxications which occurred in Europe and Italy in the last years, but currently there is a lack of information about its pharmaco-toxicological effects. With the aim at filling this gap, the present study is endeavoured to (i) evaluate the effects of acute administration of MDPHP (0.01-20 mg/kg; i.p.) on behaviour, cardiorespiratory and cardiovascular parameters in CD-1 male mice, comparing them to those observed after administration of MDPV; (ii) predict the ADMET profile of the two analogues using the Plus ADMET Predictor®; (iii) present clinical data related to MDPHP and MDPV-induced intoxications recorded between 2011 and 2023 by the Pavia Poison Control Centre (PCC) - National Toxicology Information Centre (Istituti Clinici Scientifici Maugeri, IRCCS Pavia, Italy). Our results substantiated that MDPHP and MDPV similarly affect sensorimotor and behavioural responses in mice, importantly increased locomotion and induced aggressive behaviour, and, at higher dosage, increased heart rate and blood pressure. These findings are in line with those observed in humans, revealing severe toxidromes typically characterized by Central Nervous System (CNS) alterations (behavioural/neuropsychiatric symptoms), including psychomotor agitation and aggressiveness, cardiovascular and respiratory disorders (e.g. tachycardia, hypertension, dyspnoea), and other peripheral symptoms (e.g. hyperthermia, acidosis, rhabdomyolysis).


Asunto(s)
Benzodioxoles , Pirrolidinas , Cathinona Sintética , Animales , Pirrolidinas/toxicidad , Pirrolidinas/farmacocinética , Pirrolidinas/química , Masculino , Benzodioxoles/química , Ratones , Alcaloides/toxicidad , Alcaloides/química , Alcaloides/farmacocinética , Humanos , Frecuencia Cardíaca/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Conducta Animal/efectos de los fármacos , Simulación por Computador , Presión Sanguínea/efectos de los fármacos
2.
Cell Stem Cell ; 31(8): 1175-1186.e7, 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-38876106

RESUMEN

Organoids and organs-on-a-chip have emerged as powerful tools for modeling human gut physiology and disease in vitro. Although physiologically relevant, these systems often lack the environmental milieu, spatial organization, cell type diversity, and maturity necessary for mimicking human intestinal mucosa. To instead generate models closely resembling in vivo tissue, we herein integrated organoid and organ-on-a-chip technology to develop an advanced human organoid model, called "mini-colons." By employing an asymmetric stimulation with growth factors, we greatly enhanced tissue longevity and replicated in vivo-like diversity and patterning of proliferative and differentiated cell types. Mini-colons contain abundant mucus-producing goblet cells and, signifying mini-colon maturation, single-cell RNA sequencing reveals emerging mature and functional colonocytes. This methodology is expanded to generate microtissues from the small intestine and incorporate additional microenvironmental components. Finally, our bioengineered organoids provide a precise platform to systematically study human gut physiology and pathology, and a reliable preclinical model for drug safety assessment.


Asunto(s)
Bioingeniería , Colon , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Organoides , Organoides/citología , Organoides/fisiología , Colon/citología , Colon/efectos de los fármacos , Colon/fisiología , Pirrolidinas/toxicidad , para-Aminobenzoatos/toxicidad , Proliferación Celular , Diferenciación Celular , Intestino Delgado/citología
3.
Br J Pharmacol ; 181(15): 2413-2428, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38770951

RESUMEN

BACKGROUND AND PURPOSE: Cystic fibrosis (CF) patients are living longer and healthier due to improved treatments, e.g. cystic fibrosis transmembrane conductance regulator (CFTR) modulator therapy elexacaftor/tezacaftor/ivacaftor (ETI), with treatment possibly occurring in pregnancy. The risk of ETI to foetuses remain unknown. Thus the effect of maternally administered ETI on foetal genetic and structural development was investigated. EXPERIMENTAL APPROACH: Pregnant Sprague Dawley rats were orally treated with ETI (6.7 mg·kg-1·day-1 elexacaftor + 3.5 mg·kg-1·day-1 tezacaftor + 25 mg·kg-1·day-1 ivacaftor) for 7 days from E12 to E19. Tissue samples collected at E19 were analysed using histology and RNA sequencing. Histological changes and differentially expressed genes (DEG) were assessed. KEY RESULTS: No overt structural abnormalities were found in foetal pancreas, liver, lung and small intestine after 7-day ETI exposure. Very few non-functionally associated DEG in foetal liver, lung and small intestine were identified using RNA-seq. 29 DEG were identified in thymus (27 up-regulated and two down-regulated) and most were functionally linked to each other. Gene ontology enrichment analysis revealed that multiple muscle-related terms were significantly enriched. Many more DEG were identified in cortex (44 up-regulated and four down-regulated) and a group of these were involved in central nervous system and brain development. CONCLUSION AND IMPLICATION: Sub-chronic ETI treatment in late pregnancy does not appear to pose a significant risk to the genetic and structural development of many foetal tissues. However, significant gene changes in foetal thymic myoid cells and cortical neuronal development requires future follow-up studies to assess the risk to these organs.


Asunto(s)
Aminofenoles , Benzodioxoles , Combinación de Medicamentos , Indoles , Pirazoles , Piridinas , Ratas Sprague-Dawley , Femenino , Animales , Embarazo , Aminofenoles/toxicidad , Aminofenoles/administración & dosificación , Ratas , Pirazoles/administración & dosificación , Pirazoles/toxicidad , Benzodioxoles/administración & dosificación , Indoles/administración & dosificación , Indoles/toxicidad , Piridinas/toxicidad , Piridinas/administración & dosificación , Quinolonas/toxicidad , Quinolonas/administración & dosificación , Pirroles/administración & dosificación , Pirroles/toxicidad , Pirrolidinas/administración & dosificación , Pirrolidinas/toxicidad , Pirrolidinas/farmacología , Feto/efectos de los fármacos , Feto/metabolismo , Exposición Materna/efectos adversos , Quinolinas
4.
Ann Pharm Fr ; 81(5): 801-813, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-36931432

RESUMEN

BACKGROUND: An array of commercially viable intermediate molecules necessary for the synthesis of a variety of bioactive molecules are chemically synthesized by pyrrolidine and its derivatives, which play a significant role in drug design and development process. AIM: The aim of the present research work was to explore the synthesis of some new pyrrolidine derivatives and to perform their in silico studies and finally evaluation of analgesic and anti-inflammatory activity. OBJECTIVE: The purpose of this study was to synthesis new pyrrolidine derivatives, examine how they affected the COX-1 and COX-2 enzymes computationally, and to screen their in vivo analgesic and anti-inflammatory activity on laboratory animals. METHOD: The new pyrrolidine derivatives were synthesized by condensing N-(3-acetylphenyl)-2-(pyrrolidin-1-yl)acetamide with substituted aniline in ethanol in the presence of catalytic amounts of glacial acetic acid. The structures of novel pyrrolidine derivatives were characterised using IR, NMR, and mass spectroscopy. Several molecular properties of the newly synthesized derivatives were calculated in order to evaluate the nature of the drug-like candidate. A specific reference cyclooxygenase-1 (COX-1) and cyclooxygenase-2 (COX-2) enzyme was used to dock the newly synthesized pyrrolidine derivatives. RESULTS: From the observed data, it was noted that amongst all newly synthesized compounds, A-1 and A-4 exhibited the highest anti-inflammatory and analgesic effects, respectively. CONCLUSION: On the basis of findings of present research, it was concluded that A-1 and A-4 might be utilized as a promising new lead compound for Non-Steroidal Anti-Inflammatory Drug (NSAIDs) development.


Asunto(s)
Analgésicos , Antiinflamatorios no Esteroideos , Pirrolidinas , Simulación por Computador , Pirrolidinas/síntesis química , Pirrolidinas/farmacología , Pirrolidinas/toxicidad , Analgésicos/síntesis química , Analgésicos/farmacología , Analgésicos/toxicidad , Antiinflamatorios no Esteroideos/síntesis química , Antiinflamatorios no Esteroideos/farmacología , Antiinflamatorios no Esteroideos/toxicidad , Masculino , Femenino , Animales , Ratas , Inhibidores de la Ciclooxigenasa/síntesis química , Inhibidores de la Ciclooxigenasa/farmacología , Inhibidores de la Ciclooxigenasa/toxicidad
5.
Int J Mol Sci ; 22(15)2021 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-34361071

RESUMEN

3,4-Methylenedioxypyrovalerone (MDPV) is a new psychoactive substance (NPS) and the most widespread and life-threatening synthetic cathinone of the "bath salts". Preclinical research has proven the cocaine-like psychostimulant effects of MDPV and its potential for abuse. Cannabidiol (CBD) is a non-psychotropic phytocannabinoid that has emerged as a new potential treatment for drug addiction. Here, we tested the effects of CBD (20 mg/kg) on MDPV (2 mg/kg)-induced conditioned place preference and MDPV (0.05 and 0.075 mg/kg/infusion) self-administration paradigms. In addition, we assessed the effects of the co-administration of CBD and MDPV (3 and 4 mg/kg) on anxiety-like behaviour using the elevated plus maze (EPM). CBD mitigated the MDPV-induced conditioned place preference. On the contrary, CBD administration throughout the MDPV (0.075 mg/kg/infusion) self-administration increased drug-seeking and taking behaviours, but only in the high-responders group of mice. Furthermore, CBD exerted anxiolytic-like effects, exclusively in MDPV-treated mice. Taken together, our results indicate that CBD modulation of MDPV-induced motivational responses in mice varies depending on the requirements of the learning task, resulting in a complex response. Therefore, further research attempting to decipher the behavioural and molecular interactions between CBD and MDPV is needed.


Asunto(s)
Ansiedad/tratamiento farmacológico , Conducta Animal/efectos de los fármacos , Benzodioxoles/toxicidad , Cannabidiol/farmacología , Comportamiento de Búsqueda de Drogas/efectos de los fármacos , Pirrolidinas/toxicidad , Inhibidores de Captación Adrenérgica/toxicidad , Animales , Anticonvulsivantes/farmacología , Ansiedad/inducido químicamente , Ansiedad/patología , Condicionamiento Clásico/efectos de los fármacos , Masculino , Ratones , Cathinona Sintética
6.
Int J Mol Sci ; 22(12)2021 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-34204826

RESUMEN

Mexedrone, α-PVP and α-PHP are synthetic cathinones. They can be considered amphetamine-like substances with a stimulating effect. Actually, studies showing their impact on DNA are totally absent. Therefore, in order to fill this gap, aim of the present work was to evaluate their mutagenicity on TK6 cells. On the basis of cytotoxicity and cytostasis results, we selected the concentrations (35-100 µM) to be used in the further analysis. We used the micronucleus (MN) as indicator of genetic damage and analyzed the MNi frequency fold increase by flow cytometry. Mexedrone demonstrated its mutagenic potential contrary to the other two compounds; we then proceeded by repeating the analyzes in the presence of extrinsic metabolic activation in order to check if it was possible to totally exclude the mutagenic capacity for α-PVP and α-PHP. The results demonstrated instead the mutagenicity of their metabolites. We then evaluated reactive oxygen species (ROS) induction as a possible mechanism at the basis of the highlighted effects but the results did not show a statistically significant increase in ROS levels for any of the tested substances. Anyway, our outcomes emphasize the importance of mutagenicity evaluation for a complete assessment of the risk associated with synthetic cathinones exposure.


Asunto(s)
Alcaloides/toxicidad , Metanfetamina/análogos & derivados , Mutágenos/toxicidad , Pentanonas/toxicidad , Pirrolidinas/toxicidad , Apoptosis/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Humanos , Metanfetamina/toxicidad , Micronúcleo Germinal/efectos de los fármacos , Micronúcleo Germinal/metabolismo , Especies Reactivas de Oxígeno/metabolismo
7.
Biomolecules ; 11(7)2021 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-34202146

RESUMEN

Liver malignant tumors (LMTs) represent a serious adverse drug event associated with drug-induced liver injury. Increases in endocrine-disrupting chemicals (EDCs) have attracted attention in recent years, due to their liver function-inhibiting abilities. Exposure to EDCs can induce nonalcoholic fatty liver disease and nonalcoholic steatohepatitis, which are major etiologies of LMTs, through interaction with nuclear receptors (NR) and stress response pathways (SRs). Therefore, exposure to potential EDC drugs could be associated with drug-induced LMTs. However, the drug classes associated with LMTs and the molecular initiating events (MIEs) that are specific to these drugs are not well understood. In this study, using the Food and Drug Administration Adverse Event Reporting System, we detected LMT-inducing drug signals based on adjusted odds ratios. Furthermore, based on the hypothesis that drug-induced LMTs are triggered by NR and SR modulation of potential EDCs, we used the quantitative structure-activity relationship platform for toxicity prediction to identify potential MIEs that are specific to LMT-inducing drug classes. Events related to cell proliferation and apoptosis, DNA damage, and lipid accumulation were identified as potential MIEs, and their relevance to LMTs was supported by the literature. The findings of this study may contribute to drug development and research, as well as regulatory decision making.


Asunto(s)
Sistemas de Registro de Reacción Adversa a Medicamentos/estadística & datos numéricos , Enfermedad Hepática Inducida por Sustancias y Drogas/epidemiología , Bases de Datos Factuales/estadística & datos numéricos , Neoplasias Hepáticas/epidemiología , United States Food and Drug Administration/estadística & datos numéricos , Carbamatos/efectos adversos , Carbamatos/toxicidad , Enfermedad Hepática Inducida por Sustancias y Drogas/diagnóstico , Enfermedad Hepática Inducida por Sustancias y Drogas/genética , Predicción , Humanos , Imidazoles/efectos adversos , Imidazoles/toxicidad , Isoquinolinas/efectos adversos , Isoquinolinas/toxicidad , Neoplasias Hepáticas/inducido químicamente , Neoplasias Hepáticas/diagnóstico , Neoplasias Hepáticas/genética , Inhibidores de Proteasas/efectos adversos , Inhibidores de Proteasas/toxicidad , Pirrolidinas/efectos adversos , Pirrolidinas/toxicidad , Receptores de Calcitriol/genética , Receptores de Estrógenos/genética , Sulfonamidas/efectos adversos , Sulfonamidas/toxicidad , Estados Unidos/epidemiología , Valina/efectos adversos , Valina/análogos & derivados , Valina/toxicidad
8.
Angew Chem Int Ed Engl ; 60(43): 23299-23305, 2021 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-34240523

RESUMEN

Development of proteolysis targeting chimeras (PROTACs) is emerging as a promising strategy for targeted protein degradation. However, the drug development using the heterobifunctional PROTAC molecules is generally limited by poor membrane permeability, low in vivo efficacy and indiscriminate distribution. Herein an aptamer-PROTAC conjugation approach was developed as a novel strategy to improve the tumor-specific targeting ability and in vivo antitumor potency of conventional PROTACs. As proof of concept, the first aptamer-PROTAC conjugate (APC) was designed by conjugating a BET-targeting PROTAC to the nucleic acid aptamer AS1411 (AS) via a cleavable linker. Compared with the unmodified BET PROTAC, the designed molecule (APR) showed improved tumor targeting ability in a MCF-7 xenograft model, leading to enhanced in vivo BET degradation and antitumor potency and decreased toxicity. Thus, the APC strategy may pave the way for the design of tumor-specific targeting PROTACs and have broad applications in the development of PROTAC-based drugs.


Asunto(s)
Antineoplásicos/uso terapéutico , Aptámeros de Nucleótidos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Oligodesoxirribonucleótidos/uso terapéutico , Proteolisis/efectos de los fármacos , Animales , Antineoplásicos/síntesis química , Antineoplásicos/toxicidad , Aptámeros de Nucleótidos/síntesis química , Aptámeros de Nucleótidos/toxicidad , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Disulfuros/síntesis química , Disulfuros/uso terapéutico , Disulfuros/toxicidad , Compuestos Heterocíclicos con 3 Anillos/síntesis química , Compuestos Heterocíclicos con 3 Anillos/uso terapéutico , Compuestos Heterocíclicos con 3 Anillos/toxicidad , Humanos , Ratones , Oligodesoxirribonucleótidos/síntesis química , Oligodesoxirribonucleótidos/toxicidad , Prueba de Estudio Conceptual , Pirrolidinas/síntesis química , Pirrolidinas/uso terapéutico , Pirrolidinas/toxicidad , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Physiol Res ; 70(4): 523-531, 2021 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-34062075

RESUMEN

The role of opioid kappa1 and kappa2 receptors in reperfusion cardiac injury was studied. Male Wistar rats were subjected to a 45-min coronary artery occlusion followed by a 120-min reperfusion. Opioid kappa receptor agonists were administered intravenously 5 min before the onset of reperfusion, while opioid receptor antagonists were given 10 min before reperfusion. The average value of the infarct size/area at risk (IS/AAR) ratio was 43 - 48% in untreated rats. Administration of the opioid kappa1 receptor agonist (-)-U-50,488 (1 mg/kg) limited the IS/AAR ratio by 42%. Administration of the opioid kappa receptor agonist ICI 199,441 (0.1 mg/kg) limited the IS/AAR ratio by 41%. The non-selective opioid kappa receptor agonist (+)-U-50,488 (1 mg/kg) with low affinity for opioid kappa receptor, the peripherally acting opioid kappa2 receptor agonist ICI 204,448 (4 mg/kg) and the selective opioid ?2 receptor agonist GR89696 (0.1 mg/kg) had no effect on the IS/AAR ratio. Pretreatment with naltrexone, the peripherally acting opioid receptor antagonist naloxone methiodide, or the selective opioid kappa2 receptor antagonist nor-binaltorphimine completely abolished the infarct-reducing effect of (-)-U-50,488 and ICI 199,441. Pretreatment with the selective opioid ? receptor antagonist TIPP[psi] and the selective opioid µ receptor antagonist CTAP did not alter the infarct reducing effect of (-)-U-50,488 and ICI 199,441. Our study is the first to demonstrate the following: (a) the activation of opioid kappa2 receptor has no effect on cardiac tolerance to reperfusion; (b) peripheral opioid kappa1 receptor stimulation prevents reperfusion cardiac injury; (c) ICI 199,441 administration resulted in an infarct-reducing effect at reperfusion; (e) bradycardia induced by opioid kappa receptor antagonists is not dependent on the occupancy of opioid kappa receptor.


Asunto(s)
3,4-Dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclohexil)-bencenacetamida, (trans)-Isómero/administración & dosificación , Analgésicos Opioides/administración & dosificación , Infarto del Miocardio/prevención & control , Daño por Reperfusión Miocárdica/prevención & control , Miocitos Cardíacos/efectos de los fármacos , Pirrolidinas/administración & dosificación , Receptores Opioides kappa/agonistas , 3,4-Dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclohexil)-bencenacetamida, (trans)-Isómero/toxicidad , Administración Intravenosa , Analgésicos Opioides/toxicidad , Animales , Arritmias Cardíacas/inducido químicamente , Arritmias Cardíacas/fisiopatología , Modelos Animales de Enfermedad , Frecuencia Cardíaca/efectos de los fármacos , Masculino , Infarto del Miocardio/metabolismo , Infarto del Miocardio/patología , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Antagonistas de Narcóticos/administración & dosificación , Piperazinas/administración & dosificación , Pirrolidinas/toxicidad , Ratas Wistar , Receptores Opioides kappa/metabolismo , Transducción de Señal
10.
Future Oncol ; 17(25): 3309-3319, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-33993741

RESUMEN

Aim: To determine a recommended Phase II dose of the oral fluoropyrimidine trifluridine/tipiracil (FTD/TPI) combined with the multi-kinase inhibitor regorafenib (REG) in refractory metastatic colorectal cancer patients. Materials & methods: A conventional 3 + 3 dose finding design was used. FTD/TPI was administered on days 1-5 and 8-12 of a 28-day cycle, REG on days 2-22. Two dose levels were used: FTD/TPI 25 mg/m2 b.i.d. + REG 120 mg/d, then escalated to FTD/TPI 35 mg/m2 b.i.d. + REG 120 mg/d. Results: In total, 12 patients were treated at two dose levels. Three dose-limiting toxicities were observed; all were grade 3 hypertension causally attributed to REG. Recommended Phase II dose is FTD/TPI 25 mg/m2 b.i.d. + REG 120 mg/d. Median progression-free survival was 3.81 months (95% CI: 1.51-5.29), median OS 11.1 months (95% CI: 2.3-18.2). Conclusion: The combination of REG and FTD/TPI is feasible and safe. Efficacy signals exceed that of the single agents at acceptable toxicity levels and are clinically meaningful.


Lay abstract Many patients with metastatic colorectal cancer need a sequence of different treatments over time. Regorafenib and trifluridine/tipiracil (also called TAS-102) are two drugs which are both used late in this sequence of treatments, but there is no rule as to which should be used first. Both drugs have very different mechanisms of action, and it might be beneficial to patients to administer them both at the same time as a combination treatment, instead of sequential treatment. We therefore conducted a Phase Ib study with a small number of patients to investigate whether this combined treatment would be feasible and safe. The study was designed to test the drug combination at different doses, and we found that treatment with trifluridine/tipiracil at 25 mg/m2 twice daily combined with regorafenib at 120 mg daily had acceptable side effects and is likely to be safe for use in future clinical trials. Efficacy results suggest that combined treatment with both drugs may extend patient's life span. However, these observations are preliminary and need testing in further clinical trials. Clinical trial registration: EudraCT 2016-001968-11; NCT03305913 (ClinicalTrials.gov).


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Neoplasias Colorrectales/tratamiento farmacológico , Hipertensión/epidemiología , Compuestos de Fenilurea/administración & dosificación , Piridinas/administración & dosificación , Pirrolidinas/administración & dosificación , Timina/administración & dosificación , Trifluridina/administración & dosificación , Administración Oral , Adulto , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/toxicidad , Neoplasias Colorrectales/diagnóstico , Neoplasias Colorrectales/mortalidad , Neoplasias Colorrectales/patología , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Combinación de Medicamentos , Resistencia a Antineoplásicos , Estudios de Factibilidad , Femenino , Humanos , Hipertensión/inducido químicamente , Masculino , Dosis Máxima Tolerada , Persona de Mediana Edad , Compuestos de Fenilurea/toxicidad , Supervivencia sin Progresión , Piridinas/toxicidad , Pirrolidinas/toxicidad , Criterios de Evaluación de Respuesta en Tumores Sólidos , Timina/toxicidad , Trifluridina/toxicidad
11.
Neurotox Res ; 39(4): 1360-1371, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34043181

RESUMEN

Chronic exposure to cathinone derivatives increases the risk of severe health hazards, whereas little is known about the detailed pathogenic mechanisms triggered by the derivatives. We have recently shown that treatment with α-pyrrolidinononanophenone (α-PNP, a highly lipophilic cathinone derivative possessing a long hydrocarbon main chain) provokes neuronal cell apoptosis and its 4'-fluorinated analog (F-α-PNP) potently augments the apoptotic effect. In this study, we found that neuronal SK-N-SH cell damage elicited by F-α-PNP treatment is augmented most potently by pre-incubation with an AKR1B1 inhibitor tolrestat, among specific inhibitors of four aldo-keto reductase (AKR) family members (1B1, 1C1, 1C2, and 1C3) expressed in the neuronal cells. In addition, forced overexpression of AKR1B1 remarkably lowered the cell sensitivity to F-α-PNP toxicity, clearly indicating that AKR1B1 protects from neurotoxicity of the derivative. Treatment of SK-N-SH cells with F-α-PNP resulted in a dose-dependent up-regulation of AKR1B1 expression and activation of its transcription factor NF-E2-related factor 2. Metabolic analyses using liquid chromatography/mass spectrometry/mass spectrometry revealed that AKR1B1 is hardly involved in the F-α-PNP metabolism. The F-α-PNP treatment resulted in production of reactive oxygen species and lipid peroxidation byproduct 4-hydroxy-2-nonenal (HNE) in the cells. The enhanced HNE level was reduced by overexpression of AKR1B1, which also lessened the cell damage elicited by HNE. These results suggest that the AKR1B1-mediated neuronal cell protection is due to detoxification of HNE formed by F-α-PNP treatment, but not to metabolism of the derivative.


Asunto(s)
Aldehído Reductasa/biosíntesis , Butirofenonas/toxicidad , Drogas de Diseño/toxicidad , Neuronas/efectos de los fármacos , Neuronas/enzimología , Neuroprotección/fisiología , Pirrolidinas/toxicidad , Aldehído Reductasa/antagonistas & inhibidores , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Humanos , Naftalenos/farmacología , Neuronas/patología
12.
Sci Rep ; 11(1): 8844, 2021 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-33893330

RESUMEN

A mouse model of human Familial Adenomatous Polyposis responds favorably to pharmacological inhibition of 5'-methylthioadenosine phosphorylase (MTAP). Methylthio-DADMe-Immucillin-A (MTDIA) is an orally available, transition state analogue inhibitor of MTAP. 5'-Methylthioadenosine (MTA), the substrate for MTAP, is formed in polyamine synthesis and is recycled by MTAP to S-adenosyl-L-methionine (SAM) via salvage pathways. MTDIA treatment causes accumulation of MTA, which inhibits growth of human head and neck (FaDu) and lung (H359, A549) cancers in immunocompromised mouse models. We investigated the efficacy of oral MTDIA as an anti-cancer therapeutic for intestinal adenomas in immunocompetent APCMin/+ mice, a murine model of human Familial Adenomatous Polyposis. Tumors in APCMin/+ mice were decreased in size by MTDIA treatment, resulting in markedly improved anemia and doubling of mouse lifespan. Metabolomic analysis of treated mice showed no changes in polyamine, methionine, SAM or ATP levels when compared with control mice but indicated an increase in MTA, the MTAP substrate. Generation of an MTDIA-resistant cell line in culture showed a four-fold amplification of the methionine adenosyl transferase (MAT2A) locus and expression of this enzyme. MAT2A is downstream of MTAP action and catalyzes synthesis of the SAM necessary for methylation reactions. Immunohistochemical analysis of treated mouse intestinal tissue demonstrated a decrease in symmetric dimethylarginine, a PRMT5-catalyzed modification. The anti-cancer effects of MTDIA indicate that increased cellular MTA inhibits PRMT5-mediated methylations resulting in attenuated tumor growth. Oral dosing of MTDIA as monotherapy has potential for delaying the onset and progression of colorectal cancers in Familial Adenomatous Polyposis (FAP) as well as residual duodenal tumors in FAP patients following colectomy. MTDIA causes a physiologic inactivation of MTAP and may also have efficacy in combination with inhibitors of MAT2A or PRMT5, known synthetic-lethal interactions in MTAP-/- cancer cell lines.


Asunto(s)
Genes APC , Longevidad/genética , Purina-Nucleósido Fosforilasa/metabolismo , Adenina/análogos & derivados , Adenina/farmacología , Adenina/uso terapéutico , Adenina/toxicidad , Poliposis Adenomatosa del Colon/tratamiento farmacológico , Poliposis Adenomatosa del Colon/enzimología , Poliposis Adenomatosa del Colon/genética , Animales , Modelos Animales de Enfermedad , Metabolómica , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteína-Arginina N-Metiltransferasas/metabolismo , Purina-Nucleósido Fosforilasa/antagonistas & inhibidores , Purina-Nucleósido Fosforilasa/genética , Pirrolidinas/farmacología , Pirrolidinas/uso terapéutico , Pirrolidinas/toxicidad , Análisis de Supervivencia
13.
Food Chem Toxicol ; 150: 112065, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33596453

RESUMEN

Stilbenes are secondary metabolites of great interest produced by many plant species due to their important bioactive properties. These phytochemicals have become of increasing interest in the wine industry as a natural alternative to sulphur dioxide, which has been associated with human health risks. However, there is still little toxicological information on stilbenes and the results thus far have been contradictory. Considering the key role of genotoxicity in risk assessment and the need to offer safe products in the market, the aim of this study was to assess the mutagenic and genotoxic potential of a stilbene extract with 99% purity (ST-99 extract). A complete series of different in vitro tests (Ames test, micronucleus (MN) test, and standard and enzyme-modified comet assays) was performed before its use as a preservative in wines. The ST-99 extract induces a significant increase of binucleated cells with micronuclei only in presence of the metabolic fraction S9 at the highest concentration assayed. Neither the Ames test nor the comet assay revealed the extract's genotoxic potential. Further studies are necessary, including in vivo assays, to ensure consumer safety before it can be used.


Asunto(s)
Estilbenos/toxicidad , Células CACO-2 , Daño del ADN , Células Hep G2 , Humanos , Peróxido de Hidrógeno/toxicidad , Técnicas In Vitro , Luz , Pruebas de Mutagenicidad , Pirrolidinas/toxicidad , Quinolizinas/toxicidad , Salmonella typhimurium
14.
Arterioscler Thromb Vasc Biol ; 41(2): 668-682, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33297751

RESUMEN

OBJECTIVE: Current antiplatelet medications increase the risk of bleeding, which leads to a clear clinical need in developing novel mechanism-based antiplatelet drugs. TYMP (Thymidine phosphorylase), a cytoplasm protein that is highly expressed in platelets, facilitates multiple agonist-induced platelet activation, and enhances thrombosis. Tipiracil hydrochloride (TPI), a selective TYMP inhibitor, has been approved by the Food and Drug Administration for clinical use. We tested the hypothesis that TPI is a safe antithrombotic medication. Approach and Results: By coexpression of TYMP and Lyn, GST (glutathione S-transferase) tagged Lyn-SH3 domain or Lyn-SH2 domain, we showed the direct evidence that TYMP binds to Lyn through both SH3 and SH2 domains, and TPI diminished the binding. TYMP deficiency significantly inhibits thrombosis in vivo in both sexes. Pretreatment of platelets with TPI rapidly inhibited collagen- and ADP-induced platelet aggregation. Under either normal or hyperlipidemic conditions, treating wild-type mice with TPI via intraperitoneal injection, intravenous injection, or gavage feeding dramatically inhibited thrombosis without inducing significant bleeding. Even at high doses, TPI has a lower bleeding side effect compared with aspirin and clopidogrel. Intravenous delivery of TPI alone or combined with tissue plasminogen activator dramatically inhibited thrombosis. Dual administration of a very low dose of aspirin and TPI, which had no antithrombotic effects when used alone, significantly inhibited thrombosis without disturbing hemostasis. CONCLUSIONS: This study demonstrated that inhibition of TYMP, a cytoplasmic protein, attenuated multiple signaling pathways that mediate platelet activation, aggregation, and thrombosis. TPI can be used as a novel antithrombotic medication without the increase in risk of bleeding.


Asunto(s)
Plaquetas/efectos de los fármacos , Trombosis de las Arterias Carótidas/prevención & control , Inhibidores Enzimáticos/farmacología , Fibrinolíticos/farmacología , Activación Plaquetaria/efectos de los fármacos , Inhibidores de Agregación Plaquetaria/farmacología , Pirrolidinas/farmacología , Timidina Fosforilasa/antagonistas & inhibidores , Timina/farmacología , Animales , Aspirina/farmacología , Plaquetas/enzimología , Células COS , Trombosis de las Arterias Carótidas/sangre , Trombosis de las Arterias Carótidas/enzimología , Trombosis de las Arterias Carótidas/genética , Chlorocebus aethiops , Modelos Animales de Enfermedad , Terapia Antiplaquetaria Doble , Inhibidores Enzimáticos/toxicidad , Femenino , Fibrinolíticos/toxicidad , Hemorragia/inducido químicamente , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Adhesividad Plaquetaria/efectos de los fármacos , Agregación Plaquetaria/efectos de los fármacos , Inhibidores de Agregación Plaquetaria/toxicidad , Unión Proteica , Pirrolidinas/toxicidad , Transducción de Señal , Timidina Fosforilasa/genética , Timidina Fosforilasa/metabolismo , Timina/toxicidad , Dominios Homologos src , Familia-src Quinasas/genética , Familia-src Quinasas/metabolismo
15.
Arch Toxicol ; 95(2): 509-527, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33215236

RESUMEN

3,4-Methylenedioxypyrovalerone (MDPV) is consumed worldwide, despite its potential to cause toxicity in several organs and even death. There is a recognized need to clarify the biological pathways through which MDPV elicits general and target-organ toxicity. In this work, a comprehensive untargeted GC-MS-based metabolomics analysis was performed, aiming to detect metabolic changes in putative target organs (brain, heart, kidneys and liver) but also in urine of mice after acute exposure to human-relevant doses of MDPV. Male CD-1 mice received binge intraperitoneal administrations of saline or MDPV (2.5 mg/kg or 5 mg/kg) every 2 h, for a total of three injections. Twenty-four hours after the first administration, target organs, urine and blood samples were collected for metabolomics, biochemical and histological analysis. Hepatic and renal tissues of MDPV-treated mice showed moderate histopathological changes but no significant differences were found in plasma and tissue biochemical markers of organ injury. In contrast, the multivariate analysis significantly discriminated the organs and urine of MDPV-treated mice from the control (except for the lowest dose in the brain), allowing the identification of a panoply of metabolites. Those levels were significantly deviated in relation to physiological conditions and showed an organ specific response towards the drug. Kidneys and liver showed the greatest metabolic changes. Metabolites related with energetic metabolism, antioxidant defenses and inflammatory response were significantly changed in the liver of MDPV-dosed animals, while the kidneys seem to have developed an adaptive response against oxidative stress caused by MDPV. On the other hand, the dysregulation of metabolites that contribute to metabolic acidosis was also observed in this organ. The heart showed an increase of fatty acid biosynthesis, possibly as an adaptation to maintain the cardiac energy homeostasis. In the brain, changes in 3-hydroxybutyric acid levels may reflect the activation of a neurotoxic pathway. However, the increase in metabolites with neuroprotective properties seems to counteract this change. Metabolic profiling of urine from MDPV-treated mice suggested that glutathione-dependent antioxidant pathways may be particularly involved in the compensatory mechanism to counteract oxidative stress induced by MDPV. Overall, this study reports, for the first time, the metabolic profile of liver, kidneys, heart, brain, and urine of MDPV-dosed mice, providing unique insights into the biological pathways of toxicity. Our findings also underline the value of toxicometabolomics as a robust and sensitive tool for detecting adaptive/toxic cellular responses upon exposure to a physiologically relevant dose of a toxic agent, earlier than conventional toxicity tests.


Asunto(s)
Benzodioxoles/metabolismo , Benzodioxoles/toxicidad , Encéfalo/metabolismo , Riñón/metabolismo , Hígado/metabolismo , Miocardio/metabolismo , Pirrolidinas/metabolismo , Pirrolidinas/toxicidad , Ácido 3-Hidroxibutírico/biosíntesis , Animales , Biomarcadores , Análisis Químico de la Sangre , Relación Dosis-Respuesta a Droga , Ácidos Grasos/biosíntesis , Cromatografía de Gases y Espectrometría de Masas , Homeostasis/efectos de los fármacos , Humanos , Riñón/patología , Hígado/patología , Masculino , Metaboloma , Ratones , Orina/química , Cathinona Sintética
16.
Toxicology ; 441: 152503, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32470494

RESUMEN

3,4-Methylenedioxypyrovalerone (MDPV) is one of the most popular cathinone derivatives worldwide and has recently been associated with several intoxications and deaths, in which, similarly to amphetamines, hyperthermia appears to play a prominent role. However, there remains a huge information gap underlying the mechanisms associated with its hepatotoxicity, namely under hyperthermic conditions. Here, we use a sensitive untargeted metabolomic approach based on gas chromatography-mass spectrometry (GC-MS) to investigate the effect of subtoxic and toxic concentrations of MDPV on the metabolic profile of primary mouse hepatocytes (PMH), under normothermic and hyperthermic conditions. For this purpose, hepatocytes were exposed to increasing concentrations of MDPV (LC01, LC10 and LC30) for 24 h, at 37 °C or 40.5 °C, and alterations on both intracellular metabolome and extracellular volatilome were evaluated. Multivariate analysis showed a clear separation between MDPV exposed cells and control cells in normothermic conditions, even at subtoxic concentrations (LC01 and LC10). In normothermia, there was a significant dysregulation of pathways associated with ascorbate metabolism, tricarboxylic acid (TCA) cycle and pyruvate metabolism. These metabolic changes were significantly increased at 40.5 °C, and several other pathways appear to be affected with the evolution of toxicity caused by MDPV under hyperthermic conditions, namely aspartate and glutamate metabolism, phenylalanine and tyrosine biosynthesis, aminoacyl-tRNA biosynthesis, butanoate metabolism, among others. Overall, our findings provide novel insights into the mechanism of hepatotoxicity triggered by MDPV and highlight the higher risks that may occur under hyperthermic conditions.


Asunto(s)
Benzodioxoles/toxicidad , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Hígado/efectos de los fármacos , Metaboloma/efectos de los fármacos , Pirrolidinas/toxicidad , Animales , Relación Dosis-Respuesta a Droga , Hepatocitos/efectos de los fármacos , Calor , Hígado/citología , Redes y Vías Metabólicas/efectos de los fármacos , Ratones , Temperatura , Cathinona Sintética
17.
Toxicol Lett ; 331: 42-52, 2020 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-32464236

RESUMEN

Synthetic cathinones abuse remains a serious public health problem. Kidney injury has been reported in intoxications associated with synthetic cathinones, but the molecular mechanisms involved have not been explored yet. In this study, the potential in vitro nephrotoxic effects of four commonly abused cathinone derivatives, namely pentedrone, 3,4-dimethylmethcatinone (3,4-DMMC), methylone and 3,4-methylenedioxypyrovalerone (MDPV), were assessed in the human kidney HK-2 cell line. All four derivatives elicited cell death in a concentration- and time-dependent manner, in the following order of potency: 3,4-DMMC >> MDPV > methylone ≈ pentedrone. 3,4-DMMC and methylone were selected to further elucidate the mechanisms behind synthetic cathinones-induced cell death. Both drugs elicited apoptotic cell death and prompted the formation of acidic vesicular organelles and autophagosomes in HK-2 cells. Moreover, the autophagy inhibitor 3-methyladenine significantly potentiated cell death, indicating that autophagy may serve as a cell survival mechanism that protects renal cells against synthetic cathinones toxicity. Both drugs triggered a rise in reactive oxygen and nitrogen species formation, which was completely prevented by antioxidant treatment with N­acetyl­L­cysteine or ascorbic acid. Importantly, these antioxidant agents significantly aggravated renal cell death induced by cathinone derivatives, most likely due to their autophagy-blocking properties. Taken together, our results support an intricate control of cell survival/death modulated by oxidative stress, apoptosis and autophagy in synthetic cathinones-induced renal injury.


Asunto(s)
Alcaloides/toxicidad , Apoptosis/efectos de los fármacos , Autofagia/efectos de los fármacos , Drogas Ilícitas/toxicidad , Riñón/efectos de los fármacos , Alcaloides/química , Benzodioxoles/química , Benzodioxoles/toxicidad , Técnicas de Cultivo de Célula , Línea Celular , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Humanos , Riñón/metabolismo , Riñón/patología , Metanfetamina/análogos & derivados , Metanfetamina/química , Metanfetamina/toxicidad , Metilaminas/química , Metilaminas/toxicidad , Pentanonas/química , Pentanonas/toxicidad , Pirrolidinas/química , Pirrolidinas/toxicidad , Especies de Nitrógeno Reactivo/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Factores de Tiempo , Cathinona Sintética
18.
Eur J Med Chem ; 190: 112100, 2020 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-32018095

RESUMEN

Parasitic roundworms (nematodes) are significant pathogens of humans and animals and cause substantive socioeconomic losses due to the diseases that they cause. The control of nematodes in livestock animals relies heavily on the use of anthelmintic drugs. However, their extensive use has led to a widespread problem of drug resistance in these worms. Thus, the discovery and development of novel chemical entities for the treatment of parasitic worms of humans and animals is needed. Herein, we describe our medicinal chemistry optimization efforts of a phenotypic hit against Haemonchus contortus based on a pyrrolidine-oxadiazole scaffold. This led to the identification of compounds with potent inhibitory activities (IC50 = 0.78-22.4 µM) on the motility and development of parasitic stages of H. contortus, and which were found to be highly selective in a mammalian cell counter-screen. These compounds could be used as suitable chemical tools for drug target identification or as lead compounds for further optimization.


Asunto(s)
Antihelmínticos/farmacología , Haemonchus/efectos de los fármacos , Oxadiazoles/farmacología , Pirrolidinas/farmacología , Animales , Antihelmínticos/síntesis química , Antihelmínticos/toxicidad , Línea Celular , Humanos , Estructura Molecular , Actividad Motora/efectos de los fármacos , Oxadiazoles/síntesis química , Oxadiazoles/toxicidad , Pruebas de Sensibilidad Parasitaria , Pirrolidinas/síntesis química , Pirrolidinas/toxicidad , Estereoisomerismo , Relación Estructura-Actividad
19.
Eur J Med Chem ; 188: 111986, 2020 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-31884407

RESUMEN

Breast cancer is the second leading cause of deaths in women globally. Present communication deals with design and synthesis of a few diarylnaphthyls as possible anti-breast cancer agents. Among the thirty three representatives with significant antiproliferative activity compounds 23 and 50 were quite efficacious against human breast cancer cells. Compound 50 induced apoptosis in both MCF-7 and MDA-MB-231 cells and exerted S phase and G2/M phase arrest respectively via distinct mechanistic pathways. It showed moderate microtubule destabilization. Further, it exhibited DNA topoisomerase-II inhibition effect in MCF-7 cells. It was well tolerable and found safe up to 300 mg/kg dose in Swiss albino mice. The dual action antiproliferative effect of compound 50 is quite interesting and warrants for future development.


Asunto(s)
Antineoplásicos/farmacología , Naftalenos/farmacología , Pirrolidinas/farmacología , Inhibidores de Topoisomerasa II/farmacología , Moduladores de Tubulina/farmacología , Animales , Antineoplásicos/síntesis química , Antineoplásicos/toxicidad , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/tratamiento farmacológico , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Puntos de Control de la Fase G2 del Ciclo Celular/efectos de los fármacos , Humanos , Masculino , Ratones , Simulación del Acoplamiento Molecular , Naftalenos/síntesis química , Naftalenos/toxicidad , Pirrolidinas/síntesis química , Pirrolidinas/toxicidad , Inhibidores de Topoisomerasa II/síntesis química , Inhibidores de Topoisomerasa II/toxicidad , Moduladores de Tubulina/síntesis química , Moduladores de Tubulina/toxicidad
20.
J Neuroinflammation ; 16(1): 173, 2019 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-31470863

RESUMEN

BACKGROUND: Disturbances in clock genes affect almost all patients with Alzheimer's disease (AD), as evidenced by their altered sleep/wake cycle, thermoregulation, and exacerbation of cognitive impairment. As microglia-mediated neuroinflammation proved to be a driver of AD rather than a result of the disease, in this study, we evaluated the relationship between clock gene disturbance and neuroinflammation in microglia and their contribution to the onset of AD. METHODS: In this study, the expression of clock genes and inflammatory-related genes was examined in MACS microglia isolated from 2-month-old amyloid precursor protein knock-in (APP-KI) and wild-type (WT) mice using cap analysis gene expression (CAGE) deep sequencing and RT-PCR. The effects of clock gene disturbance on neuroinflammation and relevant memory changes were examined in 2-month-old APP-KI and WT mice after injection with SR9009 (a synthetic agonist for REV-ERB). The microglia morphology was studied by staining, neuroinflammation was examined by Western blotting, and cognitive changes were examined by Y-maze and novel object recognition tests. RESULTS: CLOCK/BMAL1-driven transcriptional negative feedback loops were impaired in the microglia from 2-month-old APP-KI mice. Pro-inflammatory genes in microglia isolated from APP-KI mice were significantly higher than those isolated from WT mice at Zeitgeber time 14. The expression of pro-inflammatory genes was positively associated with NF-κB activation and negatively associated with the BMAL1 expression. SR9009 induced the activation of microglia, the increased expression of pro-inflammatory genes, and cognitive decline in 2-month-old APP-KI mice. CONCLUSION: Clock gene disturbance in microglia is involved in the early onset of AD through the induction of chronic neuroinflammation, which may be a new target for preventing or slowing AD.


Asunto(s)
Precursor de Proteína beta-Amiloide/metabolismo , Encéfalo/metabolismo , Proteínas CLOCK/metabolismo , Técnicas de Sustitución del Gen/métodos , Mediadores de Inflamación/metabolismo , Microglía/metabolismo , Precursor de Proteína beta-Amiloide/genética , Animales , Encéfalo/efectos de los fármacos , Proteínas CLOCK/antagonistas & inhibidores , Proteínas CLOCK/genética , Inflamación/genética , Inflamación/metabolismo , Mediadores de Inflamación/agonistas , Locomoción/efectos de los fármacos , Locomoción/fisiología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/efectos de los fármacos , Pirrolidinas/toxicidad , Tiofenos/toxicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA