Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
J Exp Clin Cancer Res ; 41(1): 64, 2022 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-35172855

RESUMEN

BACKGROUND: Ion channels are emerging as promising oncological targets. The potassium channels Kv1.3 and IKCa are highly expressed in the plasma membrane and mitochondria of human chronic lymphocytic leukemia (CLL) cells, compared to healthy lymphocytes. In vitro, inhibition of mitoKv1.3 by PAPTP was shown to kill ex vivo primary human CLL cells, while targeting IKCa with TRAM-34 decreased CLL cell proliferation. METHODS: Here we evaluated the effect of the above drugs in CLL cells from ibrutinib-resistant patients and in combination with Venetoclax, two drugs used in the clinical practice. The effects of the drugs were tested also in the Eµ-TCL1 genetic CLL murine model, characterized by a lympho-proliferative disease reminiscent of aggressive human CLL. Eµ-TCL1 mice showing overt disease state were treated with intraperitoneal injections of non-toxic 5 nmol/g PAPTP or 10 nmol/g TRAM-34 once a day and the number and percentage of pathological B cells (CD19+CD5+) in different, pathologically relevant body districts were determined. RESULTS: We show that Kv1.3 expression correlates with sensitivity of the human and mouse neoplastic cells to PAPTP. Primary CLL cells from ibrutinib-resistant patients could be killed with PAPTP and this drug enhanced the effect of Venetoclax, by acting on mitoKv1.3 of the inner mitochondrial membrane and triggering rapid mitochondrial changes and cytochrome c release. In vivo, after 2 week- therapy of Eµ-TCL1 mice harboring distinct CLL clones, leukemia burden was reduced by more than 85%: the number and percentage of CLL B cells fall in the spleen and peritoneal cavity and in the peripheral blood, without signs of toxicity. Notably, CLL infiltration into liver and spleen and splenomegaly were also drastically reduced upon PAPTP treatment. In contrast, TRAM-34 did not exert any beneficial effect when administered in vivo to Eµ-TCL1 mice at non-toxic concentration. CONCLUSION: Altogether, by comparing vehicle versus compound effect in different Eµ-TCL1 animals bearing unique clones similarly to CLL patients, we conclude that PAPTP significantly reduced leukemia burden in CLL-relevant districts, even in animals with advanced stage of the disease. Our results thus identify PAPTP as a very promising drug for CLL treatment, even for the chemoresistant forms of the disease.


Asunto(s)
Linfocitos B/metabolismo , Canal de Potasio Kv1.3/metabolismo , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Animales , Apoptosis , Modelos Animales de Enfermedad , Humanos , Ratones
2.
Am J Physiol Cell Physiol ; 322(1): C49-C62, 2022 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-34817270

RESUMEN

Administration of branched-chain amino acids (BCAA) has been suggested to enhance mitochondrial biogenesis, including levels of PGC-1α, which may, in turn, alter kynurenine metabolism. Ten healthy subjects performed 60 min of dynamic one-leg exercise at ∼70% of Wmax on two occasions. They were in random order supplied either a mixture of BCAA or flavored water (placebo) during the experiment. Blood samples were collected during exercise and recovery, and muscle biopsies were taken from both legs before, after, and 90 and 180 min following exercise. Ingestion of BCAA doubled their concentration in both plasma and muscle while causing a 30%-40% reduction (P < 0.05 vs. placebo) in levels of aromatic amino acids in both resting and exercising muscle during 3-h recovery period. The muscle concentration of kynurenine decreased by 25% (P < 0.05) during recovery, similar in both resting and exercising leg and with both supplements, although plasma concentration of kynurenine during recovery was 10% lower (P < 0.05) when BCAA were ingested. Ingestion of BCAA reduced the plasma concentration of kynurenic acid by 60% (P < 0.01) during exercise and recovery, whereas the level remained unchanged with placebo. Exercise induced a three- to fourfold increase (P < 0.05) in muscle content of PGC-1α1 mRNA after 90 min of recovery under both conditions, whereas levels of KAT4 mRNA and protein were unaffected by exercise or supplement. In conclusion, the reduction of plasma levels of kynurenine and kynurenic acid caused by BCAA were not associated with any changes in the level of muscle kynurenine, suggesting that kynurenine metabolism was altered in tissues other than muscle.


Asunto(s)
Aminoácidos de Cadena Ramificada/administración & dosificación , Ejercicio Físico/fisiología , Quinurenina/sangre , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Consumo de Oxígeno/efectos de los fármacos , Adulto , Femenino , Humanos , Quinurenina/metabolismo , Masculino , Consumo de Oxígeno/fisiología , Adulto Joven
3.
Cell Metab ; 33(11): 2215-2230.e8, 2021 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-34592133

RESUMEN

Endurance exercise promotes skeletal muscle vascularization, oxidative metabolism, fiber-type switching, and neuromuscular junction integrity. Importantly, the metabolic and contractile properties of the muscle fiber must be coupled to the identity of the innervating motor neuron (MN). Here, we show that muscle-derived neurturin (NRTN) acts on muscle fibers and MNs to couple their characteristics. Using a muscle-specific NRTN transgenic mouse (HSA-NRTN) and RNA sequencing of MN somas, we observed that retrograde NRTN signaling promotes a shift toward a slow MN identity. In muscle, NRTN increased capillary density and oxidative capacity and induced a transcriptional reprograming favoring fatty acid metabolism over glycolysis. This combination of effects on muscle and MNs makes HSA-NRTN mice lean with remarkable exercise performance and motor coordination. Interestingly, HSA-NRTN mice largely recapitulate the phenotype of mice with muscle-specific expression of its upstream regulator PGC-1ɑ1. This work identifies NRTN as a myokine that couples muscle oxidative capacity to slow MN identity.


Asunto(s)
Neuronas Motoras , Neurturina , Animales , Ratones , Ratones Transgénicos , Neuronas Motoras/metabolismo , Músculo Esquelético/metabolismo , Neurturina/genética , Neurturina/metabolismo , Neurturina/farmacología , Estrés Oxidativo
5.
Oxid Med Cell Longev ; 2021: 7658501, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34992716

RESUMEN

Pterostilbene (Pt) is a potentially beneficial plant phenol. In contrast to many other natural compounds (including the more celebrated resveratrol), Pt concentrations producing significant effects in vitro can also be reached with relative ease in vivo. Here we focus on some of the mechanisms underlying its activity, those involved in the activation of transcription factor EB (TFEB). A set of processes leading to this outcome starts with the generation of ROS, attributed to the interaction of Pt with complex I of the mitochondrial respiratory chain, and spreads to involve Ca2+ mobilization from the ER/mitochondria pool, activation of CREB and AMPK, and inhibition of mTORC1. TFEB migration to the nucleus results in the upregulation of autophagy and lysosomal and mitochondrial biogenesis. Cells exposed to several µM levels of Pt experience a mitochondrial crisis, an indication for using low doses in therapeutic or nutraceutical applications. Pt afforded significant functional improvements in a zebrafish embryo model of ColVI-related myopathy, a pathology which also involves defective autophagy. Furthermore, long-term supplementation with Pt reduced body weight gain and increased transcription levels of Ppargc1a and Tfeb in a mouse model of diet-induced obesity. These in vivo findings strengthen the in vitro observations and highlight the therapeutic potential of this natural compound.


Asunto(s)
Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Estilbenos/metabolismo , Animales , Modelos Animales de Enfermedad , Células HeLa , Humanos , Ratones , Factores de Transcripción , Pez Cebra
6.
Redox Biol ; 37: 101705, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33007503

RESUMEN

The potassium channel Kv1.3, involved in several important pathologies, is the target of a family of psoralen-based drugs whose mechanism of action is not fully understood. Here we provide evidence for a physical interaction of the mitochondria-located Kv1.3 (mtKv1.3) and Complex I of the respiratory chain and show that this proximity underlies the death-inducing ability of psoralenic Kv1.3 inhibitors. The effects of PAP-1-MHEG (PAP-1, a Kv1.3 inhibitor, with six monomeric ethylene glycol units attached to the phenyl ring of PAP-1), a more soluble novel derivative of PAP-1 and of its various portions on mitochondrial physiology indicate that the psoralenic moiety of PAP-1 bound to mtKv1.3 facilitates the diversion of electrons from Complex I to molecular oxygen. The resulting massive production of toxic Reactive Oxygen Species leads to death of cancer cells expressing Kv1.3. In vivo, PAP-1-MHEG significantly decreased melanoma volume. In summary, PAP-1-MHEG offers insights into the mechanisms of cytotoxicity of this family of compounds and may represent a valuable clinical tool.


Asunto(s)
Canal de Potasio Kv1.3 , Mitocondrias , Animales , Línea Celular Tumoral , Disección , Humanos , Canal de Potasio Kv1.3/antagonistas & inhibidores , Canal de Potasio Kv1.3/genética , Ratones Endogámicos C57BL , Especies Reactivas de Oxígeno
7.
Am J Physiol Cell Physiol ; 318(5): C818-C830, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32208989

RESUMEN

Tryptophan catabolism through the kynurenine pathway generates a variety of bioactive metabolites. Physical exercise can modulate kynurenine pathway metabolism in skeletal muscle and thus change the concentrations of select compounds in peripheral tissues and in the central nervous system. Here we review recent advances in our understanding of how exercise alters tryptophan-kynurenine metabolism in muscle and its subsequent local and distal effects. We propose that the effects of kynurenine pathway metabolites on skeletal muscle, adipose tissue, immune system, and the brain suggest that some of these compounds could qualify as exercise-induced myokines. Indeed, some of the more recently discovered biological activities for kynurenines include many of the best-known benefits of exercise: improved energy homeostasis, promotion of an anti-inflammatory environment, and neuroprotection. Finally, by considering the tissue expression of the different membrane and cytosolic receptors for kynurenines, we discuss known and potential biological activities for these tryptophan metabolites.


Asunto(s)
Sistema Nervioso Central/metabolismo , Quinurenina/metabolismo , Redes y Vías Metabólicas/fisiología , Músculo Esquelético/metabolismo , Encéfalo/metabolismo , Encéfalo/fisiología , Sistema Nervioso Central/fisiología , Ejercicio Físico/fisiología , Homeostasis/genética , Homeostasis/fisiología , Humanos , Sistema Inmunológico/metabolismo , Sistema Inmunológico/fisiología , Quinurenina/genética , Metabolismo/fisiología , Músculo Esquelético/fisiología , PPAR gamma/genética , Triptófano/metabolismo
8.
Int J Mol Sci ; 20(21)2019 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-31671737

RESUMEN

Obesity and related comorbidities are a major health concern. The drugs used to treat these conditions are largely inadequate or dangerous, and a well-researched approach based on nutraceuticals would be highly useful. Pterostilbene (Pt), i.e., 3,5-dimethylresveratrol, has been reported to be effective in animal models of obesity, acting on different metabolic pathways. We investigate here its ability to induce browning of white adipose tissue. Pt (5 µM) was first tested on 3T3-L1 mature adipocytes, and then it was administered (352 µmol/kg/day) to mice fed an obesogenic high-fat diet (HFD) for 30 weeks, starting at weaning. In the cultured adipocytes, the treatment elicited a significant increase of the levels of Uncoupling Protein 1 (UCP1) protein-a key component of thermogenic, energy-dissipating beige/brown adipocytes. In vivo administration antagonized weight increase, more so in males than in females. Analysis of inguinal White Adipose Tissue (WAT) revealed a trend towards browning, with significantly increased transcription of several marker genes (Cidea, Ebf2, Pgc1α, PPARγ, Sirt1, and Tbx1) and an increase in UCP1 protein levels, which, however, did not achieve significance. Given the lack of known side effects of Pt, this study strengthens the candidacy of this natural phenol as an anti-obesity nutraceutical.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Suplementos Dietéticos , Obesidad/metabolismo , Estilbenos/farmacología , Células 3T3-L1 , Adipocitos Marrones/metabolismo , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Tejido Adiposo Blanco/patología , Animales , Proteínas Reguladoras de la Apoptosis/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Peso Corporal , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/genética , PPAR gamma/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Sirtuina 1/genética , Proteínas de Dominio T Box/genética , Termogénesis/genética , Proteína Desacopladora 1/genética , Proteína Desacopladora 1/metabolismo
9.
Eur J Med Chem ; 181: 111557, 2019 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-31374419

RESUMEN

Many potential pharmacological targets are present in multiple subcellular compartments and have different pathophysiological roles depending on location. In these cases, selective targeting of a drug to the relevant subcellular domain(s) may help to sharpen its impact by providing topological specificity, thus limiting side effects, and to concentrate the compound where needed, thus increasing its effectiveness. We review here the state of the art in precision subcellular delivery. The major approaches confer "homing" properties to the active principle via permanent or reversible (in pro-drug fashion) modifications, or through the use of special-design nanoparticles or liposomes to ferry a drug(s) cargo to its desired destination. An assortment of peptides, substituents with delocalized positive charges, custom-blended lipid mixtures, pH- or enzyme-sensitive groups provide the main tools of the trade. Mitochondria, lysosomes and the cell membrane may be mentioned as the fronts on which the most significant advances have been made. Most of the examples presented here have to do with targeting natural compounds - in particular polyphenols, known as pleiotropic agents - to one or the other subcellular compartment.


Asunto(s)
Productos Biológicos/farmacología , Hipolipemiantes/farmacología , Polifenoles/farmacología , Animales , Productos Biológicos/química , Membrana Celular/efectos de los fármacos , Retículo Endoplásmico/efectos de los fármacos , Humanos , Hipolipemiantes/química , Liposomas/antagonistas & inhibidores , Mitocondrias/efectos de los fármacos , Polifenoles/química
10.
Cell Physiol Biochem ; 52(2): 232-239, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30816671

RESUMEN

BACKGROUND/AIMS: Pterostilbene (Pt; trans-3,5-dimethoxy-4'-hydroxystilbene) is a natural phenol found in blueberries and grapevines. It shows remarkable biomedical activities similar to those of resveratrol. Its high bioavailability is a major advantage for possible biomedical applications. The goal of the study was to evaluate the effects of chronic pterostilbene administration on cognitive performance in aged rats with mild cognitive impairment. METHODS: 18-month-old animals were subjected to behavioral tests to establish the "baseline", then divided into treatment and control groups. The former were chronically fed Pt (22.5 mg/kg-day) for 20 consecutive days. At the end of this period all animals were tested again and sacrificed. The dentate gyrus, the hippocampus and the prefrontal and perirhinal cortices were then collected, and RT-qPCR and/or Western blot analyses were performed on a few transcripts/proteins involved in synaptic remodeling. Mitochondrial content was also assessed. RESULTS: Pt administration improved performance in behavioral tests and positively affected memory consolidation. We found increased levels of REST, PSD-95 and mitochondrial porin1 in the dentate gyrus and a positive correlation between T-maze test score and levels of cAMP responsive element binding protein (CREB) phosphorylation. CONCLUSION: These results underscore the therapeutic potential of Pt supplementation for age-related cognitive decline.


Asunto(s)
Envejecimiento/metabolismo , Conducta Animal/efectos de los fármacos , Cognición/efectos de los fármacos , Aprendizaje por Laberinto/efectos de los fármacos , Estilbenos/farmacología , Animales , Proteína de Unión a CREB/metabolismo , Disfunción Cognitiva/tratamiento farmacológico , Disfunción Cognitiva/metabolismo , Disfunción Cognitiva/patología , Giro Dentado/metabolismo , Homólogo 4 de la Proteína Discs Large/biosíntesis , Ratas , Proteínas Represoras/biosíntesis
11.
Biochem Biophys Res Commun ; 500(1): 51-58, 2018 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-28642134

RESUMEN

Mitochondria are intracellular organelles involved in several processes from bioenergetics to cell death. In the latest years, ion channels are arising as new possible targets in controlling several cellular functions. The discovery that several plasma membrane located ion channels have intracellular counterparts, has now implemented this consideration and the number of studies enforcing the understanding of their role in different metabolic pathways. In this review, we will discuss the recent updates in the field, focusing our attention on the involvement of potassium channels during mitochondrial mediated apoptotic cell death. Since mitochondria are one of the key organelles involved in this process, it is not surprising that potassium channels located in their inner membrane could be involved in modulating mitochondrial membrane potential, ROS production, and respiratory chain complexes functions. Eventually, these events lead to changes in the mitochondrial fitness that prelude to the cytochrome c release and apoptosis. In this scenario, both the inhibition and the activation of mitochondrial potassium channels could cause cell death, and their targeting could be a novel pharmacological way to treat different human diseases.


Asunto(s)
Apoptosis/genética , Mitocondrias/metabolismo , Membranas Mitocondriales/metabolismo , Neoplasias/metabolismo , Canales de Potasio/metabolismo , Animales , Citocromos c/metabolismo , Metabolismo Energético/genética , Regulación de la Expresión Génica , Humanos , Transporte Iónico , Potencial de la Membrana Mitocondrial/fisiología , Mitocondrias/ultraestructura , Membranas Mitocondriales/ultraestructura , Neoplasias/genética , Neoplasias/patología , Canales de Potasio/clasificación , Canales de Potasio/genética , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal
12.
Neurosignals ; 25(1): 26-38, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28869943

RESUMEN

BACKGROUND/AIMS: Glioblastoma (GBM) is one of the most aggressive cancers, counting for a high number of the newly diagnosed patients with central nervous system (CNS) cancers in the United States and Europe. Major features of GBM include aggressive and invasive growth as well as a high resistance to treatment. Kv1.3, a potassium channel of the shaker family, is expressed in the inner mitochondrial membrane of many cancer cells. Inhibition of mitochondrial Kv1.3 was shown to induce apoptosis in several tumor cells at doses that were not lethal for normal cells. METHODS: We investigated the expression of Kv1.3 in different glioma cell lines by immunocytochemistry, western blotting and electron microscopy and analyzed the effect of newly synthesized, mitochondria-targeted, Kv1.3 inhibitors on the induction of cell death in these cells. Finally, we performed in vivo studies on glioma bearing mice. RESULTS: Here, we report that Kv1.3 is expressed in mitochondria of human and murine GL261, A172 and LN308 glioma cells. Treatment with the novel Kv1.3 inhibitors PAPTP or PCARBTP as well as with clofazimine induced massive cell death in glioma cells, while Psora-4 and PAP-1 were almost without effect. However, in vivo experiments revealed that the drugs had no effect on orthotopic brain tumors in vivo. CONCLUSION: These data serve as proof of principle that Kv1.3 inhibitors kills GBM cells, but drugs that act in vivo against glioblastoma must be developed to translate these findings in vivo.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Neoplasias Encefálicas/tratamiento farmacológico , Glioblastoma/tratamiento farmacológico , Canal de Potasio Kv1.3/antagonistas & inhibidores , Bloqueadores de los Canales de Potasio/farmacología , Animales , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Clofazimina/farmacología , Clofazimina/uso terapéutico , Cumarinas/farmacología , Cumarinas/uso terapéutico , Humanos , Inmunohistoquímica , Ratones , Compuestos Organofosforados/farmacología , Compuestos Organofosforados/uso terapéutico
13.
Ann N Y Acad Sci ; 1403(1): 27-37, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28675763

RESUMEN

Prodrugs of resveratrol are under development. Among the long-term goals, still largely elusive, are (1) modulating physical properties (e.g., water-soluble derivatives bearing polyethylene glycol chains), (2) changing distribution in the body (e.g., galactosyl derivatives restricted to the intestinal lumen), (3) increasing absorption from the gastrointestinal tract (e.g., derivatives imitating the natural substrates of endogenous transporters), and (4) hindering phase II metabolism (e.g., temporarily blocking the hydroxyls), all contributing to (5) increasing bioavailability. The chemical bonds that have been tested for functionalization include carboxyester, acetal, and carbamate groups. A second approach, which can be combined with the first, seeks to reinforce or modify the biochemical activities of resveratrol by concentrating the compound at specific subcellular sites. An example is provided by mitochondria-targeted derivatives. These proved to be pro-oxidant and cytotoxic in vitro, selectively killing fast-growing and tumor cells when supplied in the low micromolar range. This suggests the possibility of anticancer applications.


Asunto(s)
Estrés Oxidativo/efectos de los fármacos , Profármacos/farmacología , Especies Reactivas de Oxígeno/metabolismo , Estilbenos/farmacología , Animales , Humanos , Mitocondrias/metabolismo , Resveratrol
14.
Cancer Cell ; 31(4): 516-531.e10, 2017 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-28399409

RESUMEN

The potassium channel Kv1.3 is highly expressed in the mitochondria of various cancerous cells. Here we show that direct inhibition of Kv1.3 using two mitochondria-targeted inhibitors alters mitochondrial function and leads to reactive oxygen species (ROS)-mediated death of even chemoresistant cells independently of p53 status. These inhibitors killed 98% of ex vivo primary chronic B-lymphocytic leukemia tumor cells while sparing healthy B cells. In orthotopic mouse models of melanoma and pancreatic ductal adenocarcinoma, the compounds reduced tumor size by more than 90% and 60%, respectively, while sparing immune and cardiac functions. Our work provides direct evidence that specific pharmacological targeting of a mitochondrial potassium channel can lead to ROS-mediated selective apoptosis of cancer cells in vivo, without causing significant side effects.


Asunto(s)
Antineoplásicos/farmacología , Canal de Potasio Kv1.3/antagonistas & inhibidores , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Bloqueadores de los Canales de Potasio/farmacología , Anciano , Animales , Antineoplásicos/síntesis química , Antineoplásicos/química , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/patología , Estudios de Casos y Controles , Cumarinas/farmacología , Estabilidad de Medicamentos , Femenino , Humanos , Canal de Potasio Kv1.3/metabolismo , Leucemia Linfocítica Crónica de Células B/patología , Masculino , Melanoma/tratamiento farmacológico , Melanoma/patología , Ratones Endogámicos C57BL , Persona de Mediana Edad , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Terapia Molecular Dirigida , Compuestos Organofosforados/farmacología , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/patología , Bloqueadores de los Canales de Potasio/síntesis química , Bloqueadores de los Canales de Potasio/química
15.
Eur J Med Chem ; 135: 77-88, 2017 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-28433778

RESUMEN

Obesity and associated pathologies are a dramatically growing problem. New therapies to prevent and/or cure them are strongly needed. Adipose tissue is a logical target for pharmacological intervention, since it is now recognized to exert an important endocrine function, secreting a variety of adipokines affecting, for example, adiposity and insulin resistance. This proof of principle work focuses on the development of novel lipid-mimetic prodrugs reaching fat deposits by the same lymphatic absorption route followed by dietary triglycerides. Pterostilbene, a natural phenolic compound with potential anti-obesity effects, was used as model "cargo", attached via a carbamate group to an ω-aminodecanoate chain linked to either position 1 or position 2 of the glycerol moiety of synthetic triglycerides. The prodrugs underwent position-selective hydrolysis when challenged with pancreatic lipases in vitro. Pterostilbene-containing triglycerides as well as pterostilbene and its metabolites were present in the adipose tissue of mice fed an obesogenic diet containing one or the other of the derivatives. For the first time this approach is used to deliver an obesity antagonist to the adipose tissue. The results demonstrate the feasibility of delivering active compounds to adipose tissue by reversibly incorporating them into triglyceride-mimetic structures. Upon release in the target site these compounds are expected to exert their pharmacological activity precisely where needed.


Asunto(s)
Tejido Adiposo/efectos de los fármacos , Fármacos Antiobesidad/farmacología , Lípidos/farmacología , Obesidad/tratamiento farmacológico , Profármacos/farmacología , Estilbenos/farmacología , Animales , Fármacos Antiobesidad/síntesis química , Fármacos Antiobesidad/química , Relación Dosis-Respuesta a Droga , Lípidos/síntesis química , Lípidos/química , Ratones , Ratones Endogámicos C57BL , Estructura Molecular , Profármacos/síntesis química , Profármacos/química , Estilbenos/síntesis química , Estilbenos/química , Relación Estructura-Actividad
16.
Eur J Pharm Biopharm ; 115: 149-158, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28254379

RESUMEN

The biomedical effects of the natural phenol pterostilbene are of great interest but its bioavailability is negatively affected by the phenolic group in position 4' which is an ideal target for the conjugative enzymes of phase II metabolism. We report the synthesis and characterization of prodrugs in which the hydroxyl moiety is reversibly protected as a carbamate ester linked to the N-terminus of a natural amino acid. Prodrugs comprising amino acids with hydrophobic side chains were readily absorbed after intragastric administration to rats. The Area Under the Curve for pterostilbene in blood was optimal when prodrugs with isoleucine or ß-alanine were used. The prodrug incorporating isoleucine was used for further studies to map distribution into major organs. When compared to pterostilbene itself, administration of the isoleucine prodrug afforded increased absorption, reduced metabolism and higher concentrations of pterostilbene, sustained for several hours, in most of the organs examined. Experiments using Caco-2 cells as an in vitro model for human intestinal absorption suggest that the prodrug could have promising absorption profiles also in humans; its uptake is partly due to passive diffusion, and partly mediated by H+-dependent transporters expressed on the apical membrane of enterocytes, such as PepT1 and OATP.


Asunto(s)
Aminoácidos/química , Profármacos/química , Profármacos/farmacocinética , Estilbenos/química , Estilbenos/farmacocinética , Administración Oral , Animales , Disponibilidad Biológica , Transporte Biológico/efectos de los fármacos , Células CACO-2 , Carbamatos/química , Línea Celular Tumoral , Ésteres/química , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Absorción Intestinal/efectos de los fármacos , Masculino , Ratas , Ratas Wistar , Distribución Tisular/efectos de los fármacos
17.
Oncotarget ; 8(24): 38276-38293, 2017 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-27542263

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) represents the most common form of pancreatic cancer with rising incidence in developing countries. Unfortunately, the overall 5-year survival rate is still less than 5%. The most frequent oncogenic mutations in PDAC are loss-of function mutations in p53 and gain-of-function mutations in KRAS. Here we show that clofazimine (Lamprene), a drug already used in the clinic for autoimmune diseases and leprosy, is able to efficiently kill in vitro five different PDAC cell lines harboring p53 mutations. We provide evidence that clofazimine induces apoptosis in PDAC cells with an EC50 in the µM range via its specific inhibitory action on the potassium channel Kv1.3. Intraperitoneal injection of clofazimine resulted in its accumulation in the pancreas of mice 8 hours after administration. Using an orthotopic PDAC xenotransplantation model in SCID beige mouse, we show that clofazimine significantly and strongly reduced the primary tumor weight. Thus, our work identifies clofazimine as a promising therapeutic agent against PDAC and further highlights ion channels as possible oncological targets.


Asunto(s)
Antineoplásicos/farmacología , Carcinoma Ductal Pancreático/patología , Clofazimina/farmacología , Canal de Potasio Kv1.3/efectos de los fármacos , Neoplasias Pancreáticas/patología , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Humanos , Canal de Potasio Kv1.3/antagonistas & inhibidores , Ratones , Ratones SCID , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Biochim Biophys Acta ; 1863(10): 2515-30, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-26902508

RESUMEN

Over the past 30years the mitochondrial permeability transition - the permeabilization of the inner mitochondrial membrane due to the opening of a wide pore - has progressed from being considered a curious artifact induced in isolated mitochondria by Ca(2+) and phosphate to a key cell-death-inducing process in several major pathologies. Its relevance is by now universally acknowledged and a pharmacology targeting the phenomenon is being developed. The molecular nature of the pore remains to this day uncertain, but progress has recently been made with the identification of the FOF1 ATP synthase as the probable proteic substrate. Researchers sharing this conviction are however divided into two camps: these believing that only the ATP synthase dimers or oligomers can form the pore, presumably in the contact region between monomers, and those who consider that the ring-forming c subunits in the FO sector actually constitute the walls of the pore. The latest development is the emergence of a new candidate: Spastic Paraplegia 7 (SPG7), a mitochondrial AAA-type membrane protease which forms a 6-stave barrel. This review summarizes recent developments of research on the pathophysiological relevance and on the molecular nature of the mitochondrial permeability transition pore. This article is part of a Special Issue entitled: Mitochondrial Channels edited by Pierre Sonveaux, Pierre Maechler and Jean-Claude Martinou.


Asunto(s)
Proteínas de Transporte de Membrana Mitocondrial/metabolismo , ATPasas Asociadas con Actividades Celulares Diversas , Adenosina Trifosfato/metabolismo , Animales , Peptidil-Prolil Isomerasa F , Ciclofilinas/metabolismo , Dimerización , Humanos , Metaloendopeptidasas/genética , Metaloendopeptidasas/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/efectos de los fármacos , Poro de Transición de la Permeabilidad Mitocondrial , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Enfermedades Neurodegenerativas/metabolismo , ATPasas de Translocación de Protón/química , ATPasas de Translocación de Protón/metabolismo , Paraplejía Espástica Hereditaria/genética , Paraplejía Espástica Hereditaria/metabolismo
19.
Sci Rep ; 5: 15216, 2015 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-26463125

RESUMEN

Resveratrol (3, 5, 4'-trihydroxy-trans-stilbene), a plant polyphenol, has important drug-like properties, but its pharmacological exploitation in vivo is hindered by its rapid transformation via phase II conjugative metabolism. One approach to bypass this problem relies on prodrugs. We report here the synthesis, characterization, stability and in vivo pharmacokinetic behaviour of prodrugs of resveratrol in which the OH groups are engaged in an N-monosubstituted carbamate ester (-OC(O)NHR) linkage with a natural amino acid (Leu, Ile, Phe, Thr) to prevent conjugation and modulate the physicochemical properties of the molecule. We also report a convenient, high-yield protocol to obtain derivatives of this type. The new carbamate ester derivatives are stable at pH 1, while they undergo slow hydrolysis at physiological pH and hydrolyse with kinetics suitable for use in prodrugs in whole blood. After administration to rats by oral gavage the isoleucine-containing prodrug was significantly absorbed, and was present in the bloodstream as non-metabolized unaltered or partially deprotected species, demonstrating effective shielding from first-pass metabolism. We conclude that prodrugs based on the N-monosubstituted carbamate ester bond have the appropriate stability profile for the systemic delivery of phenolic compounds.


Asunto(s)
Aminoácidos/farmacocinética , Carbamatos/síntesis química , Carbamatos/farmacocinética , Profármacos/síntesis química , Profármacos/farmacocinética , Estilbenos/farmacocinética , Administración Oral , Aminoácidos/administración & dosificación , Aminoácidos/química , Animales , Carbamatos/administración & dosificación , Diseño de Fármacos , Masculino , Tasa de Depuración Metabólica , Profármacos/administración & dosificación , Ratas , Ratas Wistar , Resveratrol , Estilbenos/química
20.
Molecules ; 20(9): 16085-102, 2015 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-26404221

RESUMEN

Resveratrol is a natural polyphenol with many interesting biological activities. Its pharmacological exploitation in vivo is, however, hindered by its rapid elimination via phase II conjugative metabolism at the intestinal and, most importantly, hepatic levels. One approach to bypass this problem relies on prodrugs. We report here the synthesis, characterization, hydrolysis, and in vivo pharmacokinetic behavior of resveratrol prodrugs in which the OH groups are engaged in an N-monosubstituted carbamate ester linkage. As promoiety, methoxy-oligo(ethylene glycol) groups (m-OEG) (CH3-[OCH2CH2]n-) of defined chain length (n = 3, 4, 6) were used. These are expected to modulate the chemico-physical properties of the resulting derivatives, much like longer poly(ethylene glycol) (PEG) chains, while retaining a relatively low MW and, thus, a favorable drug loading capacity. Intragastric administration to rats resulted in the appearance in the bloodstream of the prodrug and of the products of its partial hydrolysis, confirming protection from first-pass metabolism during absorption.


Asunto(s)
Carbamatos/farmacocinética , Profármacos/síntesis química , Estilbenos/química , Animales , Carbamatos/síntesis química , Carbamatos/química , Estructura Molecular , Polietilenglicoles/química , Profármacos/química , Profármacos/farmacocinética , Ratas , Resveratrol
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA