Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros










Intervalo de año de publicación
1.
Angew Chem Int Ed Engl ; 63(12): e202319925, 2024 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-38286754

RESUMEN

Anaerobes dominate the microbiota of the gastrointestinal (GI) tract, where a significant portion of small molecules can be degraded or modified. However, the enormous metabolic capacity of gut anaerobes remains largely elusive in contrast to aerobic bacteria, mainly due to the requirement of sophisticated laboratory settings. In this study, we employed an in silico machine learning platform, MoleculeX, to predict the metabolic capacity of a gut anaerobe, Clostridium sporogenes, against small molecules. Experiments revealed that among the top seven candidates predicted as unstable, six indeed exhibited instability in C. sporogenes culture. We further identified several metabolites resulting from the supplementation of everolimus in the bacterial culture for the first time. By utilizing bioinformatics and in vitro biochemical assays, we successfully identified an enzyme encoded in the genome of C. sporogenes responsible for everolimus transformation. Our framework thus can potentially facilitate future understanding of small molecules metabolism in the gut, further improve patient care through personalized medicine, and guide the development of new small molecule drugs and therapeutic approaches.


Asunto(s)
Clostridium , Everolimus , Humanos , Everolimus/metabolismo , Clostridium/metabolismo , Bacterias Anaerobias
2.
Exp Biol Med (Maywood) ; 248(23): 2440-2448, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38158699

RESUMEN

The mammalian target of rapamycin (mTOR) inhibitors, everolimus (but not dactolisib), is frequently associated with lung injury in clinical therapies. However, the underlying mechanisms remain unclear. Endothelial cell barrier dysfunction plays a major role in the pathogenesis of the lung injury. This study hypothesizes that everolimus increases pulmonary endothelial permeability, which leads to lung injury. We tested the effects of everolimus on human pulmonary microvascular endothelial cell (HPMEC) permeability and a mouse model of intraperitoneal injection of everolimus was established to investigate the effect of everolimus on pulmonary vascular permeability. Our data showed that everolimus increased human pulmonary microvascular endothelial cell (HPMEC) permeability which was associated with MLC phosphorylation and F-actin stress fiber formation. Furthermore, everolimus induced an increasing concentration of intracellular calcium Ca2+ leakage in HPMECs and this was normalized with ryanodine pretreatment. In addition, ryanodine decreased everolimus-induced phosphorylation of PKCα and MLC, and barrier disruption in HPMECs. Consistent with in vitro data, everolimus treatment caused a visible lung-vascular barrier dysfunction, including an increase in protein in BALF and lung capillary-endothelial permeability, which was significantly attenuated by pretreatment with an inhibitor of PKCα, MLCK, and ryanodine. This study shows that everolimus induced pulmonary endothelial hyper-permeability, at least partly, in an MLC phosphorylation-mediated EC contraction which is influenced in a Ca2+-dependent manner and can lead to lung injury through mTOR-independent mechanisms.


Asunto(s)
Células Endoteliales , Lesión Pulmonar , Animales , Ratones , Humanos , Células Endoteliales/metabolismo , Everolimus/farmacología , Everolimus/metabolismo , Lesión Pulmonar/patología , Endotelio Vascular , Proteína Quinasa C-alfa/metabolismo , Proteína Quinasa C-alfa/farmacología , Rianodina/metabolismo , Rianodina/farmacología , Pulmón/metabolismo , Fosforilación , Células Cultivadas , Serina-Treonina Quinasas TOR/metabolismo , Mamíferos
3.
FASEB J ; 37(8): e23082, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37462506

RESUMEN

Brain white matter injury (WMI) is a serious disease of the central nervous system. Pleiotrophin (PTN) promotes the differentiation and myelination of oligodendrocytes (OLs) in vitro. However, the role of PTN in WMI remains unknown. Therefore, this study aimed to investigate the neuroprotective role and potential mechanisms of PTN function in neonatal rats with WMI. The PTN and mammalian target of rapamycin (mTOR) inhibitor everolimus was used to treat a WMI model in postnatal day 3 Sprague-Dawley rats, in which the right common carotid arteries of these rats were isolated, ligated, and exposed to a hypoxic environment (6% O2 + 94% N2 ) for 2 h. OL differentiation and myelination, as well as the spatial learning and memory abilities of the rats were evaluated to examine the effects of PTN. Two proteins of the mTOR signaling pathway, YingYang1 (YY1) and inhibitor of DNA binding 4 (Id4), were detected and were used to explore the potential mechanisms of PTN in rat WMI experiment and oxygen glucose deprivation (OGD) model. We found that the differentiation and myelination of OLs were impaired after WMI. PTN administration rescued this injury by activating mTOR/YY1 and inhibiting Id4. Everolimus administration inhibited mTOR/YY1 and activated Id4, which blocked the neuroprotective role of PTN in WMI. PTN plays a neuroprotective role in neonatal rats with WMI, which could be involved in the mTOR/YY1/Id4 signaling pathway.


Asunto(s)
Lesiones Encefálicas , Sustancia Blanca , Animales , Ratas , Animales Recién Nacidos , Sustancia Blanca/metabolismo , Ratas Sprague-Dawley , Everolimus/farmacología , Everolimus/metabolismo , Transducción de Señal , Lesiones Encefálicas/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Mamíferos/metabolismo
4.
Sci Rep ; 13(1): 5032, 2023 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-36977745

RESUMEN

Hutchinson-Gilford Progeria Syndrome (HGPS) is a rare, fatal genetic disease that accelerates atherosclerosis. With a limited pool of HGPS patients, clinical trials face unique challenges and require reliable preclinical testing. We previously reported a 3D tissue engineered blood vessel (TEBV) microphysiological system fabricated with iPSC-derived vascular cells from HGPS patients. HGPS TEBVs exhibit features of HGPS atherosclerosis including loss of smooth muscle cells, reduced vasoactivity, excess extracellular matrix (ECM) deposition, inflammatory marker expression, and calcification. We tested the effects of HGPS therapeutics Lonafarnib and Everolimus separately and together, currently in Phase I/II clinical trial, on HGPS TEBVs. Everolimus decreased reactive oxygen species levels, increased proliferation, reduced DNA damage in HGPS vascular cells, and improved vasoconstriction in HGPS TEBVs. Lonafarnib improved shear stress response of HGPS iPSC-derived endothelial cells (viECs) and reduced ECM deposition, inflammation, and calcification in HGPS TEBVs. Combination treatment with Lonafarnib and Everolimus produced additional benefits such as improved endothelial and smooth muscle marker expression and reduced apoptosis, as well as increased TEBV vasoconstriction and vasodilation. These results suggest that a combined trial of both drugs may provide cardiovascular benefits beyond Lonafarnib, if the Everolimus dose can be tolerated.


Asunto(s)
Aterosclerosis , Calcinosis , Células Madre Pluripotentes Inducidas , Progeria , Humanos , Progeria/genética , Everolimus/farmacología , Everolimus/uso terapéutico , Everolimus/metabolismo , Células Endoteliales/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Aterosclerosis/metabolismo , Calcinosis/metabolismo , Lamina Tipo A/genética
5.
Bioengineered ; 13(5): 11361-11372, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35506423

RESUMEN

Helicobacter pylori (H.pylori) infection caused by gastric mucosal inflammation plays a pivotal role in the progression of gastric diseases. The recruitment and attachment of monocytes to the gastric mucosal epithelium are a major event in the early stages of H. pylori-associated gastric diseases. Everolimus is a mechanistic/mammalian target of rapamycin (mTOR) inhibitor used to prevent tumor growth by inhibiting the PI3K signaling pathway. Here, we examined the pharmacological role of Everolimus against H.pylori-induced damage in gastric epithelial cells. Firstly, we found that Everolimus ameliorated H.pylori-induced oxidative stress by reducing reactive oxygen species (ROS) and malondialdehyde (MDA). Secondly, Everolimus significantly reduced the expressions of the pro-inflammatory cytokines interleukin (IL)-6, tumor necrosis factor-α (TNF-α), and IL-8. Moreover, it decreased the production of the pro-inflammatory chemokines C-X-C motif ligand 1 (CXCL1) and macrophage chemoattractant protein-1 (MCP-1). Importantly, Everolimus suppressed the induction of the adhesion molecule intracellular adhesion molecule-1 (ICAM-1) and the attachment of THP-1 monocytes to gastric epithelial AGS cells. Our data also shows that Everolimus inhibited the activation of the NF-κB signaling pathway. Therefore, we conclude that Everolimus could protect gastric epithelial cells by mitigating H.pylori-induced inflammatory response and the attachment of monocytes to epithelial cells.


Asunto(s)
Infecciones por Helicobacter , Helicobacter pylori , Células Epiteliales/metabolismo , Everolimus/metabolismo , Everolimus/farmacología , Infecciones por Helicobacter/complicaciones , Infecciones por Helicobacter/tratamiento farmacológico , Infecciones por Helicobacter/metabolismo , Helicobacter pylori/metabolismo , Humanos , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Interleucina-8/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo
6.
Hum Gene Ther ; 33(13-14): 740-756, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35331006

RESUMEN

Intravenous onasemnogene abeparvovec is approved for the treatment of spinal muscular atrophy in children < 2 years. For later-onset patients, intrathecal onasemnogene abeparvovec may be advantageous over intravenous administration. Recently, microscopic dorsal root ganglion (DRG) changes were observed in nonhuman primates (NHPs) following intrathecal onasemnogene abeparvovec administration. To characterize these DRG findings, two NHP studies evaluating intrathecal onasemnogene abeparvovec administration were conducted: a 12-month study with a 6-week interim cohort and a 13-week study with a 2-week interim cohort. The latter investigated the potential impact of prednisolone or rituximab plus everolimus on DRG toxicity. An additional 6-month, single-dose, intravenous NHP study conducted in parallel evaluated onasemnogene abeparvovec safety (including DRG toxicity) with or without prednisolone coadministration. Intrathecal onasemnogene abeparvovec administration was well tolerated and not associated with clinical observations. Microscopic onasemnogene abeparvovec-related changes were observed in the DRG and trigeminal ganglion (TG) and included mononuclear cell inflammation and/or neuronal degeneration, which was colocalized with high vector transcript expression at 6 weeks postdose. Incidence and severity of DRG changes were generally decreased after 52 weeks compared with 6 weeks postdose. Other onasemnogene abeparvovec-related microscopic findings of axonal degeneration, mononuclear cell infiltrates and/or gliosis in the spinal cord, dorsal spinal nerve root/spinal nerves, and/or peripheral nerves were absent or found at decreased incidences and/or severities after 52 weeks. DRG and/or TG microscopic findings following intravenous onasemnogene abeparvovec dosing included minimal to slight neuronal degeneration and mononuclear cell inflammation at 6 weeks and 6 months postdose. Nervous system microscopic findings following intrathecal onasemnogene abeparvovec (≥1.2 × 1013 vg/animal) trended toward resolution after 52 weeks, supporting nonprogression of changes, including in the DRG. Onasemnogene abeparvovec-related DRG findings were not associated with electrophysiology changes and were not ameliorated by prednisolone or rituximab plus everolimus coadministration. The pathogenesis is possibly a consequence of increased vector genome transduction and/or transgene expression.


Asunto(s)
Everolimus , Ganglios Espinales , Animales , Everolimus/metabolismo , Ganglios Espinales/metabolismo , Humanos , Inflamación/metabolismo , Macaca fascicularis , Prednisolona/metabolismo , Prednisolona/uso terapéutico , Rituximab/metabolismo
7.
Biomark Med ; 14(12): 1165-1181, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32969246

RESUMEN

Liver transplantation has been widely accepted as an effective intervention for end-stage liver diseases and early hepatocellular carcinomas. However, a variety of postoperative complications and adverse reactions have baffled medical staff and patients. Currently, transplantation monitoring relies primarily on nonspecific biochemical tests, whereas diagnosis of multiple complications depends on invasive pathological examination. Therefore, a noninvasive monitoring method with high selectivity and specificity is desperately needed. This review summarized the potential of endogenous small-molecule metabolites as biomarkers for assessing graft function, ischemia-reperfusion injury and liver rejection. Exogenous metabolites, mainly those immunosuppressive agents with high intra- and inter-individual variability, were also discussed for transplantation monitoring.


Asunto(s)
Trasplante de Hígado/efectos adversos , Metaboloma , Complicaciones Posoperatorias/diagnóstico , Complicaciones Posoperatorias/etiología , Biomarcadores/metabolismo , Carcinoma Hepatocelular/cirugía , Everolimus/metabolismo , Everolimus/uso terapéutico , Rechazo de Injerto/metabolismo , Humanos , Inmunosupresores , Neoplasias Hepáticas/cirugía , Ácido Micofenólico/metabolismo , Complicaciones Posoperatorias/metabolismo , Pronóstico , Daño por Reperfusión , Tacrolimus/metabolismo , Tacrolimus/uso terapéutico
8.
Arterioscler Thromb Vasc Biol ; 38(9): 2217-2224, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30026269

RESUMEN

Objective- Drug-eluting stents eluting canonical mTOR (mammalian target of rapamycin) inhibitors are widely used to treat coronary artery disease but accelerate the development of atherosclerosis within the stent (neoatherosclerosis)-a leading cause of late stent failure. We recently showed that canonical mTOR inhibitors bind FKBP12.6 (12.6-kDa FK506-binding protein 12), displace it from calcium release channels, resulting in activation of PKCα (protein kinase Cα) and dissociation of p-120-catenin (p120) from VE-CAD (vascular endothelial cadherin; promoting endothelial barrier dysfunction [EBD]). However, the relevance of these findings to drug-eluting stents remains unknown. Newer generation direct mTOR kinase inhibitors do not bind FKBP12.6 and offer the potential of improving endothelial barrier function while maintaining antirestenotic efficacy, but their actual effects are unknown. We examined the effects of 2 different pharmacological targeting strategies-canonical mTOR inhibitor everolimus and mTOR kinase inhibitors Torin-2-on EBD after stenting. Approach and Results- Using the rabbit model of stenting and a combination of Evans blue dye, confocal and scanning electron microscopy studies, everolimus-eluting stents resulted in long-term EBD compared with bare metal stents. EBD was mitigated by using stents that eluted mTOR kinase inhibitors (Torin-2-eluting stent). At 60 days after stent placement, everolimus-eluting stents demonstrated large areas of Evans blue dye staining and evidence of p120 VE-CAD dissociation consistent with EBD. These findings were absent in bare metal stents and significantly attenuated in Torin-2-eluting stent. As proof of concept of the role of EBD in neoatherosclerosis, 100 days after stenting, animals were fed an enriched cholesterol diet for an additional 30 days. Everolimus-eluting stents demonstrated significantly more macrophage infiltration (consistent with neoatherosclerosis) compared with both bare metal stents and Torin-2-eluting stent. Conclusions- Our results pinpoint interactions between FKBP12.6 and canonical mTOR inhibitors as a major cause of vascular permeability and neoatherosclerosis, which can be overcome by using mTOR kinase inhibitors. Our study suggests further refinement of molecular targeting of the mTOR complex may be a promising strategy (Graphic Abstract).


Asunto(s)
Permeabilidad Capilar/efectos de los fármacos , Stents Liberadores de Fármacos , Endotelio Vascular/metabolismo , Everolimus/farmacología , Naftiridinas/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Animales , Antígenos CD/metabolismo , Cadherinas/metabolismo , Cateninas/metabolismo , Activación Enzimática , Everolimus/metabolismo , Masculino , Modelos Animales , Naftiridinas/metabolismo , Prueba de Estudio Conceptual , Proteína Quinasa C-alfa/metabolismo , Conejos , Serina-Treonina Quinasas TOR/metabolismo , Proteínas de Unión a Tacrolimus/metabolismo , Catenina delta
10.
Sci Rep ; 6: 35869, 2016 10 24.
Artículo en Inglés | MEDLINE | ID: mdl-27775055

RESUMEN

This study aimed to compare the mid-term adverse cardiovascular outcomes associated with Coronary Artery Bypass Surgery (CABG) and Percutaneous Coronary Intervention (PCI) with Everolimus Eluting Stents (EES). Electronic databases were searched for studies comparing the mid-term (>1 year) adverse cardiovascular outcomes between CABG and PCI with EES. Odd Ratios (OR) with 95% Confidence Intervals (CIs) were calculated and the pooled analyses were performed with RevMan 5.3 software. A total number of 5207 patients were involved in this analysis. No significant difference was observed in mortality between CABG and EES with OR: 0.90, 95% CI: 0.73-1.10; P = 0.30. Moreover, CABG was associated with a high stroke rate, with OR: 0.73, 95% CI: 0.45-1.17; P = 0.19, without any statistical significant. CABG was associated with significantly lower Major Adverse Cardiac Events and Myocardial Infarction with OR: 1.46, 95% CI: 1.05-2.04; P = 0.03 and OR: 1.46, 95% CI: 1.01-2.12; P = 0.05 respectively whereas PCI was associated with a significantly higher repeated revascularization with OR: 2.21; 95% CI: 1.76-2.77; P = 0.00001. In conclusion, significant differences were noted in several subgroups analyzing the mid-term cardiovascular outcomes between CABG and EES.


Asunto(s)
Puente de Arteria Coronaria/efectos adversos , Everolimus/metabolismo , Inmunosupresores/metabolismo , Intervención Coronaria Percutánea/efectos adversos , Stents , Humanos , Accidente Cerebrovascular/epidemiología , Análisis de Supervivencia , Resultado del Tratamiento
11.
J Control Release ; 220(Pt A): 503-514, 2015 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-26578440

RESUMEN

Metastatic melanoma has a high mortality rate due to lymphatic progression of the disease. Current treatment is surgery followed by radiation and intravenous chemotherapy. However, drawbacks for current chemotherapeutics lie in the fact that they develop resistance and do not achieve therapeutic concentrations in the lymphatic system. We hypothesize that a three-drug nanoscale drug delivery system, tailored for lymphatic uptake, administered subcutaneously, will have decreased drug resistance and therefore offer better therapeutic outcomes. We prepared and characterized nanoparticles (NPs) with docetaxel, everolimus, and LY294002 in polyethyleneglycol-block-poly(ε-caprolactone) (PEG-PCL) polymer with different charge distributions by modifying the ratio of anionic and neutral end groups on the PEG block. These NPs are similarly sized (~48 nm), with neutral, partially charged, or fully charged surface. The NPs are able to load ~2mg/mL of each drug and are stable for 24h. The NPs are assessed for safety and efficacy in two transgenic metastatic melanoma mouse models. All the NPs were safe in both models based on general appearance, weight changes, death, and blood biochemical analyses. The partially charged NPs are most effective in decreasing the number of melanocytes at both the proximal (sentinel) lymph node (LN) and the distal LN from the injection site. The neutral NPs are efficacious at the proximal LN, while the fully charged NPs have no effect on either LNs. Thus, our data indicates that the NP surface charge and lymphatic efficacy are closely tied to each other and the partially charged NPs have the highest potential in treating metastatic melanoma.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Cromonas/administración & dosificación , Portadores de Fármacos , Everolimus/administración & dosificación , Lactonas/química , Ganglios Linfáticos/efectos de los fármacos , Melanoma/tratamiento farmacológico , Morfolinas/administración & dosificación , Nanopartículas , Polietilenglicoles/química , Neoplasias Cutáneas/tratamiento farmacológico , Taxoides/administración & dosificación , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/química , Protocolos de Quimioterapia Combinada Antineoplásica/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Cromonas/química , Cromonas/metabolismo , Docetaxel , Composición de Medicamentos , Resistencia a Antineoplásicos , Everolimus/química , Everolimus/metabolismo , Humanos , Ganglios Linfáticos/metabolismo , Ganglios Linfáticos/patología , Melanocitos/efectos de los fármacos , Melanocitos/patología , Melanoma/genética , Melanoma/metabolismo , Melanoma/secundario , Ratones Transgénicos , Morfolinas/química , Morfolinas/metabolismo , Nanotecnología , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Solubilidad , Propiedades de Superficie , Taxoides/química , Taxoides/metabolismo , Factores de Tiempo , Carga Tumoral/efectos de los fármacos
12.
Expert Opin Drug Saf ; 14(7): 1055-70, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25912929

RESUMEN

INTRODUCTION: The inhibitors of the mammalian target of rapamycin (mTOR) sirolimus and everolimus are used not only as immunosuppressants after organ transplantation in combination with calcineurin inhibitors (CNIs) but also as proliferation signal inhibitors coated on drug-eluting stents and in cancer therapy. Notwithstanding their related chemical structures, both have distinct pharmacokinetic, pharmacodynamic and toxicodynamic properties. AREAS COVERED: The additional hydroxyethyl group at the C(40) of the everolimus molecule results in different tissue and subcellular distribution, different affinities to active drug transporters and drug-metabolizing enzymes as well as differences in drug-target protein interactions including a much higher potency in terms of interacting with the mTOR complex 2 than sirolimus. Said mechanistic differences as well as differences found in clinical trials in transplant patients are reviewed. EXPERT OPINION: In comparison to sirolimus, everolimus has higher bioavailability, a shorter terminal half-life, different blood metabolite patterns, the potential to antagonize the negative effects of CNIs on neuronal and kidney cell metabolism (which sirolimus enhances), the ability to stimulate mitochondrial oxidation (which sirolimus inhibits) and to reduce vascular inflammation to a greater extent. A head-to-head, randomized trial comparing the safety and tolerability of these two mTOR inhibitors in solid organ transplant recipients is merited.


Asunto(s)
Everolimus , Riñón/metabolismo , Mitocondrias/efectos de los fármacos , Complejos Multiproteicos , Neuronas/metabolismo , Sirolimus , Serina-Treonina Quinasas TOR , Disponibilidad Biológica , Everolimus/metabolismo , Everolimus/farmacocinética , Rechazo de Injerto/metabolismo , Rechazo de Injerto/prevención & control , Humanos , Inmunosupresores/metabolismo , Inmunosupresores/farmacocinética , Diana Mecanicista del Complejo 2 de la Rapamicina , Complejos Multiproteicos/antagonistas & inhibidores , Complejos Multiproteicos/metabolismo , Sirolimus/metabolismo , Sirolimus/farmacocinética , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...