Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Clin Sleep Med ; 19(7): 1191-1198, 2023 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-36856062

RESUMEN

STUDY OBJECTIVES: Obstructive sleep apnea (OSA) is a risk factor for a major public health problem, car crashes, due to excessive daytime sleepiness (EDS). Commercial vehicle driving (CVD) is a hazardous occupation, having a high fatality rate worldwide. There have been no studies on EDS and OSA in Zambia despite the high rate of annual road traffic accidents (RTAs). We aim to determine the prevalence of EDS and OSA risk among CVDs in Lusaka, Zambia, to assess the impact of OSA on high RTA rates. METHODS: This was a cross-sectional study. The STOP BANG questionnaire and the Epworth Sleepiness Scale were used. Consecutive sampling of drivers was done who were divided into low and high risk of OSA (HROSA). The risk factors associated with OSA in the bivariate analyses were subjected to a multivariate logistic regression model. RESULTS: One hundred thirty-six drivers participated in the study (all male) with a mean age of 48 ± 5 years. The prevalence of HROSA was 22.8% out of whom 67.7% also had a EDS. Only 9.6% of the total cohort had EDS without HROSA. Using Fisher's exact test, HROSA was significantly associated with older age (> 50 years, P < .001), obesity (body mass index >30, P < .001), neck circumference of > 40 cm (P = .032), and hypertension (P < .001). Snoring and EDS were significantly associated with RTAs (P < .0001 and P = .007, respectively). CONCLUSIONS: High risk of OSA and EDS are common among CMV drivers in Zambia and underdiagnosed. The risk factors for OSA are amenable to preventive interventions. CITATION: Simpamba K, May JL, Waghat A, Attarian H, Mateyo K. Obstructive sleep apnea and excessive daytime sleepiness among commercial motor vehicle drivers in Lusaka, Zambia. J Clin Sleep Med. 2023;19(7):1191-1198.


Asunto(s)
Conducción de Automóvil , Trastornos de Somnolencia Excesiva , Apnea Obstructiva del Sueño , Humanos , Masculino , Adulto , Persona de Mediana Edad , Estudios Transversales , Zambia/epidemiología , Trastornos de Somnolencia Excesiva/epidemiología , Trastornos de Somnolencia Excesiva/diagnóstico , Apnea Obstructiva del Sueño/complicaciones , Apnea Obstructiva del Sueño/epidemiología , Apnea Obstructiva del Sueño/diagnóstico , Vehículos a Motor
3.
Sci Adv ; 5(5): eaaw4543, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31131326

RESUMEN

Isocitrate dehydrogenases (IDHs) are critical metabolic enzymes that catalyze the oxidative decarboxylation of isocitrate to α-ketoglutarate (αKG), NAD(P)H, and CO2. IDHs epigenetically control gene expression through effects on αKG-dependent dioxygenases, maintain redox balance and promote anaplerosis by providing cells with NADPH and precursor substrates for macromolecular synthesis, and regulate respiration and energy production through generation of NADH. Cancer-associated mutations in IDH1 and IDH2 represent one of the most comprehensively studied mechanisms of IDH pathogenic effect. Mutant enzymes produce (R)-2-hydroxyglutarate, which in turn inhibits αKG-dependent dioxygenase function, resulting in a global hypermethylation phenotype, increased tumor cell multipotency, and malignancy. Recent studies identified wild-type IDHs as critical regulators of normal organ physiology and, when transcriptionally induced or down-regulated, as contributing to cancer and neurodegeneration, respectively. We describe how mutant and wild-type enzymes contribute on molecular levels to disease pathogenesis, and discuss efforts to pharmacologically target IDH-controlled metabolic rewiring.


Asunto(s)
Isocitrato Deshidrogenasa/genética , Mutación , Neoplasias/genética , Sitio Alostérico , Animales , Dominio Catalítico , Ciclo del Ácido Cítrico , Citoplasma/metabolismo , Metilación de ADN , Epigénesis Genética , Glutaratos/metabolismo , Homeostasis , Humanos , Sistema Inmunológico , Concentración 50 Inhibidora , Isocitrato Deshidrogenasa/metabolismo , Ratones , Mitocondrias/metabolismo , NADP/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Oxidación-Reducción , Fenotipo
4.
Sci Adv ; 5(1): eaat0456, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30613765

RESUMEN

Mutation or transcriptional up-regulation of isocitrate dehydrogenases 1 and 2 (IDH1 and IDH2) promotes cancer progression through metabolic reprogramming and epigenetic deregulation of gene expression. Here, we demonstrate that IDH3α, a subunit of the IDH3 heterotetramer, is elevated in glioblastoma (GBM) patient samples compared to normal brain tissue and promotes GBM progression in orthotopic glioma mouse models. IDH3α loss of function reduces tricarboxylic acid (TCA) cycle turnover and inhibits oxidative phosphorylation. In addition to its impact on mitochondrial energy metabolism, IDH3α binds to cytosolic serine hydroxymethyltransferase (cSHMT). This interaction enhances nucleotide availability during DNA replication, while the absence of IDH3α promotes methionine cycle activity, S-adenosyl methionine generation, and DNA methylation. Thus, the regulation of one-carbon metabolism via an IDH3α-cSHMT signaling axis represents a novel mechanism of metabolic adaptation in GBM.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , Glicina Hidroximetiltransferasa/metabolismo , Isocitrato Deshidrogenasa/metabolismo , Animales , Neoplasias Encefálicas/genética , Línea Celular Tumoral , Ciclo del Ácido Cítrico/genética , Citosol/metabolismo , Metilación de ADN/genética , Femenino , Glioblastoma/genética , Células HEK293 , Xenoinjertos , Humanos , Isocitrato Deshidrogenasa/genética , Ratones , Ratones SCID , Fosforilación Oxidativa , Puntos de Control de la Fase S del Ciclo Celular , Transfección
5.
Cell Rep ; 19(9): 1858-1873, 2017 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-28564604

RESUMEN

Oncogenic mutations in two isocitrate dehydrogenase (IDH)-encoding genes (IDH1 and IDH2) have been identified in acute myelogenous leukemia, low-grade glioma, and secondary glioblastoma (GBM). Our in silico and wet-bench analyses indicate that non-mutated IDH1 mRNA and protein are commonly overexpressed in primary GBMs. We show that genetic and pharmacologic inactivation of IDH1 decreases GBM cell growth, promotes a more differentiated tumor cell state, increases apoptosis in response to targeted therapies, and prolongs the survival of animal subjects bearing patient-derived xenografts (PDXs). On a molecular level, diminished IDH1 activity results in reduced α-ketoglutarate (αKG) and NADPH production, paralleled by deficient carbon flux from glucose or acetate into lipids, exhaustion of reduced glutathione, increased levels of reactive oxygen species (ROS), and enhanced histone methylation and differentiation marker expression. These findings suggest that IDH1 upregulation represents a common metabolic adaptation by GBMs to support macromolecular synthesis, aggressive growth, and therapy resistance.


Asunto(s)
Resistencia a Antineoplásicos , Glioblastoma/enzimología , Glioblastoma/patología , Isocitrato Deshidrogenasa/genética , Terapia Molecular Dirigida , Mutación/genética , Animales , Apoptosis/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Progresión de la Enfermedad , Resistencia a Antineoplásicos/efectos de los fármacos , Clorhidrato de Erlotinib/farmacología , Clorhidrato de Erlotinib/uso terapéutico , Factores de Transcripción Forkhead/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Glioblastoma/tratamiento farmacológico , Glioblastoma/genética , Histonas/metabolismo , Isocitrato Deshidrogenasa/metabolismo , Ácidos Cetoglutáricos/metabolismo , Lípidos/biosíntesis , Metilación , Ratones , Ratones SCID , NADP/metabolismo , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/patología , Inhibidores de Proteínas Quinasas/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Especies Reactivas de Oxígeno/metabolismo
6.
Proc Natl Acad Sci U S A ; 114(16): 4129-4134, 2017 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-28373576

RESUMEN

RNA interference (RNAi)-based gene regulation platforms have shown promise as a novel class of therapeutics for the precision treatment of cancer. Techniques in preclinical evaluation of RNAi-based nanoconjugates have yet to allow for optimization of their gene regulatory activity. We have developed spherical nucleic acids (SNAs) as a blood-brain barrier-/blood-tumor barrier-penetrating nanoconjugate to deliver small interfering (si) and micro (mi)RNAs to intracranial glioblastoma (GBM) tumor sites. To identify high-activity SNA conjugates and to determine optimal SNA treatment regimens, we developed a reporter xenograft model to evaluate SNA efficacy in vivo. Engrafted tumors stably coexpress optical reporters for luciferase and a near-infrared (NIR) fluorescent protein (iRFP670), with the latter fused to the DNA repair protein O6-methylguanine-DNA-methyltransferase (MGMT). Using noninvasive imaging of animal subjects bearing reporter-modified intracranial xenografts, we quantitatively assessed MGMT knockdown by SNAs composed of MGMT-targeting siRNA duplexes (siMGMT-SNAs). We show that systemic administration of siMGMT-SNAs via single tail vein injection is capable of robust intratumoral MGMT protein knockdown in vivo, with persistent and SNA dose-dependent MGMT silencing confirmed by Western blotting of tumor tissue ex vivo. Analyses of SNA biodistribution and pharmacokinetics revealed rapid intratumoral uptake and significant intratumoral retention that increased the antitumor activity of coadministered temozolomide (TMZ). Our study demonstrates that dual noninvasive bioluminescence and NIR fluorescence imaging of cancer xenograft models represents a powerful in vivo strategy to identify RNAi-based nanotherapeutics with potent gene silencing activity and will inform additional preclinical and clinical investigations of these constructs.


Asunto(s)
Neoplasias Encefálicas/tratamiento farmacológico , Metilasas de Modificación del ADN/antagonistas & inhibidores , Enzimas Reparadoras del ADN/antagonistas & inhibidores , Glioblastoma/tratamiento farmacológico , Nanoconjugados/administración & dosificación , ARN Interferente Pequeño/genética , Proteínas Supresoras de Tumor/antagonistas & inhibidores , Animales , Antineoplásicos Alquilantes/farmacología , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Metilasas de Modificación del ADN/genética , Enzimas Reparadoras del ADN/genética , Dacarbazina/análogos & derivados , Dacarbazina/farmacología , Femenino , Fluorescencia , Glioblastoma/genética , Glioblastoma/patología , Humanos , Ratones , Ratones SCID , Nanoconjugados/química , Interferencia de ARN , Temozolomida , Células Tumorales Cultivadas , Proteínas Supresoras de Tumor/genética , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Nanomaterials (Basel) ; 6(8)2016 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-28335271

RESUMEN

This work compares intravenous (IV) versus fluoroscopy-guided transarterial intra-catheter (IC) delivery of iron oxide core-titanium dioxide shell nanoparticles (NPs) in vivo in VX2 model of liver cancer in rabbits. NPs coated with glucose and decorated with a peptide sequence from cortactin were administered to animals with developed VX2 liver cancer. Two hours after NPs delivery tumors, normal liver, kidney, lung and spleen tissues were harvested and used for a series on histological and elemental analysis tests. Quantification of NPs in tissues was done both by bulk inductively coupled plasma mass spectrometry (ICP-MS) analysis and by hard X-ray fluorescence microscopy. Both IV and IC NPs injection are feasible modalities for delivering NPs to VX2 liver tumors with comparable tumor accumulation. It is possible that this is an outcome of the fact that VX2 tumors are highly vascularized and hemorrhagic, and therefore enhanced permeability and retention (EPR) plays the most significant role in accumulation of nanoparticles in tumor tissue. It is, however, interesting to note that IV delivery led to increased sequestration of NPs by spleen and normal liver tissue, while IC delivery lead to more NP positive Kupffer cells. This difference is most likely a direct outcome of blood flow dynamics. Armed with this knowledge about nanoparticle delivery, we plan to test them as radiosensitizers in the future.

8.
Theranostics ; 3(9): 719-28, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24019856

RESUMEN

Metabolomic profiling is ideally suited for the analysis of cardiac metabolism in healthy and diseased states. Here, we show that systematic discovery of biomarkers of ischemic preconditioning using metabolomics can be translated to potential nanotheranostics. Thirty-three patients underwent percutaneous coronary intervention (PCI) after myocardial infarction. Blood was sampled from catheters in the coronary sinus, aorta and femoral vein before coronary occlusion and 20 minutes after one minute of coronary occlusion. Plasma was analysed using GC-MS metabolomics and iTRAQ LC-MS/MS proteomics. Proteins and metabolites were mapped into the Metacore network database (GeneGo, MI, USA) to establish functional relevance. Expression of 13 proteins was significantly different (p<0.05) as a result of PCI. Included amongst these was CD44, a cell surface marker of reperfusion injury. Thirty-eight metabolites were identified using a targeted approach. Using PCA, 42% of their variance was accounted for by 21 metabolites. Multiple metabolic pathways and potential biomarkers of cardiac ischemia, reperfusion and preconditioning were identified. CD44, a marker of reperfusion injury, and myristic acid, a potential preconditioning agent, were incorporated into a nanotheranostic that may be useful for cardiovascular applications. Integrating biomarker discovery techniques into rationally designed nanoconstructs may lead to improvements in disease-specific diagnosis and treatment.


Asunto(s)
Biomarcadores/sangre , Metaboloma , Isquemia Miocárdica/diagnóstico , Isquemia Miocárdica/fisiopatología , Proteoma , Puntos Cuánticos/metabolismo , Silicio/metabolismo , Bioingeniería/métodos , Cromatografía Liquida , Cromatografía de Gases y Espectrometría de Masas , Humanos , Plasma/química , Puntos Cuánticos/química , Silicio/química , Espectrometría de Masas en Tándem
9.
Integr Biol (Camb) ; 5(1): 144-50, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23014624

RESUMEN

The development of multimodal nanoparticle platforms is desirable for cancer nanotechnology applications. Creating single nanoplatforms with both plasmonic and photoluminescent optical properties has remained a challenge, because combining discrete entities each having one of these unique properties typically results in the attenuation of one of the desirable properties. Here, we overcome challenges associated with combining plasmonic gold with luminescent silicon nanocrystals for biological imaging applications by incorporating multiple silicon quantum dots into the core of a micelle and then depositing gold on the surface of the nanostructure. Within the newly developed nanoconstruct, the gold shell exhibits plasmonic light scattering properties useful for dark field imaging, while the silicon nanocrystals maintain their photoluminescence. The result is a nanoplatform with both plasmonic and luminescent properties in a useful form. Multimodal imaging of pancreatic cancer cells demonstrates overlap of luminescence from the silicon quantum dots with light scattering from the gold shell. This approach can be tailored to other formulations and address the challenge of fluorescence attenuation that is typically observed when quantum dots are combined with plasmonic materials. The usefulness of these particles may eventually extend beyond multimodal imaging to include photothermal treatment.


Asunto(s)
Oro/química , Mediciones Luminiscentes/métodos , Microscopía Fluorescente/métodos , Nanopartículas/química , Neoplasias Experimentales/patología , Resonancia por Plasmón de Superficie/métodos , Línea Celular Tumoral , Humanos , Puntos Cuánticos , Técnica de Sustracción
10.
Nanoscale ; 4(17): 5483-9, 2012 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-22854899

RESUMEN

Luminescent imaging agents and MRI contrast agents are desirable components in the rational design of multifunctional nanoconstructs for biological imaging applications. Luminescent biocompatible silicon quantum dots (SiQDs) and gadolinium chelates can be applied for fluorescence microscopy and MRI, respectively. Here, we report the first synthesis of a nanocomplex incorporating SiQDs and gadolinium ions (Gd³âº) for biological applications. The nanoconstruct is composed of a PEGylated micelle, with hydrophobic SiQDs in its core, covalently bound to DOTA-chelated Gd³âº. Dynamic light scattering reveals a radius of 85 nm for these nanoconstructs, which is consistent with the electron microscopy results depicting radii ranging from 25 to 60 nm. Cellular uptake of the probes verified that they maintain their optical properties within the intracellular environment. The magnetic resonance relaxivity of the nanoconstruct was 2.4 mM⁻¹ s⁻¹ (in terms of Gd³âº concentration), calculated to be around 6000 mM⁻¹ s⁻¹ per nanoconstruct. These desirable optical and relaxivity properties of the newly developed probe open the door for use of SiQDs in future multimodal applications such as tumour imaging.


Asunto(s)
Gadolinio/química , Puntos Cuánticos , Silicio/química , Animales , Línea Celular Tumoral , Complejos de Coordinación/química , Iones/química , Sustancias Luminiscentes/química , Ratones , Micelas , Microscopía Confocal
11.
Nanoscale ; 4(16): 5163-8, 2012 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-22802158

RESUMEN

Quantum dots are known for their superior optical properties; however, when transferred into aqueous media, their luminescent properties are frequently compromised. When encapsulated in micelles for bioimaging applications, luminescent silicon quantum dots can lose as much as 50% of their luminescence depending on the formulation used. Here, we create an energy transfer micelle platform that combines silicon quantum dots with an anthracene-based dye in the hydrophobic core of 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[methoxy(polyethylene glycol)-2000] (DSPE-PEG) micelles. These phospholipid micelles are water dispersible, stable, and surrounded by a PEGylated layer with modifiable functional groups. The spectroscopic properties of energy transfer between the anthracene donors and silicon quantum dot acceptors were analyzed based on the observed dependence of the steady-state emission spectrum on concentration ratio, excitation wavelength, pH, and temperature. The luminescence of silicon quantum dots from the core of a 150 nm micelle is enhanced by more than 80% when the anthracene dye is added. This work provides a simple yet readily applicable solution to the long-standing problem of luminescence enhancement of silicon quantum dots and can serve as a template for improving the quantum dot emission yield for biological applications where luminescence signal enhancements are desirable and for solar applications where energy transfer plays a critical role in device performance.


Asunto(s)
Puntos Cuánticos , Silicio/química , Antracenos/química , Transferencia de Energía , Concentración de Iones de Hidrógeno , Interacciones Hidrofóbicas e Hidrofílicas , Micelas , Fosfatidiletanolaminas/química , Polietilenglicoles/química , Temperatura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...