Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 126
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Nanobiotechnology ; 22(1): 10, 2024 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-38166940

RESUMEN

BACKGROUND: Intravenous administration of fibrinolytic drugs, such as recombinant tissue plasminogen activator (rtPA) is the standard treatment of acute thrombotic diseases. However, current fibrinolytics exhibit limited clinical efficacy because of their short plasma half-lives and risk of hemorrhagic transformations. Platelet membrane-based nanocarriers have received increasing attention for ischemic stroke therapies, as they have natural thrombus-targeting activity, can prolong half-life of the fibrinolytic therapy, and reduce side effects. In this study we have gone further in developing platelet-derived nanocarriers (defined as cellsomes) to encapsulate and protect rtPA from degradation. Following lyophilization and characterization, their formulation properties, biocompatibility, therapeutic effect, and risk of hemorrhages were later investigated in a thromboembolic model of stroke in mice. RESULTS: Cellsomes of 200 nm size and loaded with rtPA were generated from membrane fragments of human platelets. The lyophilization process did not influence the nanocarrier size distribution, morphology, and colloidal stability conferring particle preservation and long-term storage. Encapsulated rtPA in cellsomes and administered as a single bolus showed to be as effective as a continuous clinical perfusion of free rtPA at equal concentration, without increasing the risk of hemorrhagic transformations or provoking an inflammatory response. CONCLUSIONS: This study provides evidence for the safe and effective use of lyophilized biomimetic platelet-derived nanomedicine for precise thrombolytic treatment of acute ischemic stroke. In addition, this new nanoformulation could simplify the clinical use of rtPA as a single bolus, being easier and less time-consuming in an emergency setting than a treatment perfusion, particularly in stroke patients. We have successfully addressed one of the main barriers to drug application and commercialization, the long-term storage of nanomedicines, overcoming the potential chemical and physical instabilities of nanomedicines when stored in an aqueous buffer.


Asunto(s)
Isquemia Encefálica , Accidente Cerebrovascular Isquémico , Accidente Cerebrovascular , Humanos , Ratones , Animales , Activador de Tejido Plasminógeno , Fibrinolíticos/farmacología , Fibrinolíticos/uso terapéutico , Terapia Trombolítica/efectos adversos , Accidente Cerebrovascular/tratamiento farmacológico , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/etiología
2.
Anal Bioanal Chem ; 413(20): 5135-5146, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34173039

RESUMEN

Drug safety assessment in the early phases of drug discovery is critical to facilitate the rapid development of novel therapeutics. Recently, teleost zebrafish (Danio rerio) has emerged as a promising vertebrate model for the assessment of drug safety. Zebrafish is a convenient model because of its small size, high fecundity, embryo transparency, and ex utero development. In this study, we developed a matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI MSI) method applied to zebrafish larvae to investigate safety and metabolism of sahaquine (Sq), an anticancer agent inhibiting histone deacetylase 6. This technique improves on prior studies using liquid chromatography-mass spectrometry (LC-MS) by adding analysis of the drug spatial distribution. Using this method, it was determined that Sq dissolved in fish water (1-2000 µM) did not reach the larval body and was mainly distributed throughout the yolk. High Sq concentration (800 µM) administered intravenously allowed the compound to reach the larval body but did not induce phenotypic abnormalities. Sq was metabolized into its glucuronidated form within 24 h and was excreted within 72 h. MALDI MSI was instrumental in showing that Sq-glucuronide was mainly formed in the gut and slightly in yolk syncytial layer, and provided valuable insights into xenobiotics elimination in zebrafish larvae. This study indicates that Sq has a good safety profile and merits further investigations in other disease models. In addition, the optimized MALDI MSI protocol provided here can be widely applied to study distribution and metabolic fate of other structurally related molecules.


Asunto(s)
Espectrometría de Masas/métodos , Animales , Línea Celular Tumoral , Embrión no Mamífero/efectos de los fármacos , Humanos , Larva/efectos de los fármacos , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Pez Cebra
3.
Nanomedicine ; 37: 102441, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34302989

RESUMEN

A fundamental problem in oncology is that anticancer chemotherapeutics kill both cancer and healthy cells in the surrounding tissues. Resveratrol is a natural antioxidant with intriguing and opposing biological properties: it reduces viability of some cancer cells but not of non-transformed ones (in equimolar concentrations). Therefore, we examined resveratrol in human non-transformed primary astrocytes and astrocytoma. Resveratrol reduced reactive oxygen species in astrocytes, but not in astrocytoma. Such cell-type dependent response is particularly evident with analyses at the single cell level showing clear population difference in high and low glutathione levels. Due to resveratrol's poor aqueous solubility that limits its use in clinics, we incorporated it into stimulus-responsive micelles assembled from miktoarm polymers. This could be an attractive chemotherapeutic delivery strategy in nano-oncology. As a proof of principle, we show that these formulations containing resveratrol markedly decrease astrocytoma viability, particularly in combination with temozolomide, a first line chemotherapeutic for astrocytoma.


Asunto(s)
Astrocitos/efectos de los fármacos , Astrocitoma/tratamiento farmacológico , Nanopartículas/química , Resveratrol/farmacología , Antioxidantes/química , Antioxidantes/farmacología , Astrocitoma/metabolismo , Astrocitoma/patología , Línea Celular Tumoral , Humanos , Nanopartículas/uso terapéutico , Cultivo Primario de Células , Especies Reactivas de Oxígeno/metabolismo , Resveratrol/química , Análisis de la Célula Individual
4.
Int J Mol Sci ; 22(5)2021 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-33670858

RESUMEN

Interleukin-6 (IL-6) is involved in physiological and pathological processes. Different pharmacological agents have been developed to block IL-6 deleterious effects and to recover homeostatic IL-6 signaling. One of the proposed nanostructures in pre-clinical investigations which reduced IL-6 concentrations is polyglycerol dendrimer, a nano-structure with multiple sulfate groups. The aim of the present study was to uncover the type of binding between critical positions in the human IL-6 structure available for binding dPGS and compare it with heparin sulfate binding. We studied these interactions by performing docking simulations of dPGS and heparins with human IL-6 using AutoDock Vina. These molecular docking analyses indicate that the two ligands have comparable affinities for the positively charged positions on the surface of IL-6. All-atom molecular dynamics simulations (MD) employing Gromacs were used to explore the binding sites and binding strengths. Results suggest two major binding sites and show that the strengths of binding are similar for heparin and dPGS (-5.5-6.4 kcal/ mol). dPGS or its analogs could be used in the therapeutic intervention in sepsis and inflammatory disorders to reduce unbound IL-6 in the plasma or tissues and its binding to the receptors. We propose that analogs of dPGS could specifically block IL-6 binding in the desired signaling mode and would be valuable new probes to establish optimized therapeutic intervention in inflammation.


Asunto(s)
Dendrímeros/química , Glicerol/farmacología , Interleucina-6/antagonistas & inhibidores , Modelos Moleculares , Polímeros/farmacología , Antiinflamatorios/farmacología , Heparina , Humanos , Inflamación , Ligandos , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Unión Proteica
5.
Angew Chem Int Ed Engl ; 60(8): 3882-3904, 2021 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-32589355

RESUMEN

The counterions neutralizing the charges on polyelectrolytes such as DNA or heparin may dissociate in water and greatly influence the interaction of such polyelectrolytes with biomolecules, particularly proteins. In this Review we give an overview of studies on the interaction of proteins with polyelectrolytes and how this knowledge can be used for medical applications. Counterion release was identified as the main driving force for the binding of proteins to polyelectrolytes: Patches of positive charge become multivalent counterions of the polyelectrolyte and lead to the release of counterions from the polyelectrolyte and a concomitant increase in entropy. This is shown from investigations on the interaction of proteins with natural and synthetic polyelectrolytes. Special emphasis is paid to sulfated dendritic polyglycerols (dPGS). The Review demonstrates that we are moving to a better understanding of charge-charge interactions in systems of biological relevance. Research along these lines will aid and promote the design of synthetic polyelectrolytes for medical applications.


Asunto(s)
ADN/química , Polielectrolitos/química , Proteínas/química , Antineoplásicos/química , Antineoplásicos/farmacología , Supervivencia Celular/efectos de los fármacos , ADN/metabolismo , Portadores de Fármacos/química , Glicosaminoglicanos/química , Glicosaminoglicanos/metabolismo , Humanos , Polielectrolitos/metabolismo , Unión Proteica , Proteínas/metabolismo , Termodinámica
6.
Molecules ; 25(19)2020 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-33007959

RESUMEN

Nanostructured hyperbranched macromolecules have been extensively studied at the chemical, physical and morphological levels. The cellular structural and functional complexity of neural cells and their cross-talk have made it rather difficult to evaluate dendrimer effects in a mixed population of glial cells and neurons. Thus, we are at a relatively early stage of bench-to-bedside translation, and this is due mainly to the lack of data valuable for clinical investigations. It is only recently that techniques have become available that allow for analyses of biological processes inside the living cells, at the nanoscale, in real time. This review summarizes the essential properties of neural cells and dendrimers, and provides a cross-section of biological, pre-clinical and early clinical studies, where dendrimers were used as nanocarriers. It also highlights some examples of biological studies employing dendritic polyglycerol sulfates and their effects on glia and neurons. It is the aim of this review to encourage young scientists to advance mechanistic and technological approaches in dendrimer research so that these extremely versatile and attractive nanostructures gain even greater recognition in translational medicine.


Asunto(s)
Encéfalo/citología , Dendrímeros/farmacología , Animales , Antiinflamatorios/farmacología , Neoplasias Encefálicas/patología , Dendrímeros/química , Humanos , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/metabolismo
7.
Molecules ; 25(4)2020 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-32075239

RESUMEN

An architectural polymer containing hydrophobic isoxazole-based dendron and hydrophilic polyethylene glycol linear tail is prepared by a combination of the robust ZnCl2 catalyzed alkyne-nitrile oxide 1,3-dipolar cycloaddition and esterification chemistry. This water soluble amphiphilic telodendrimer acts as a macromolecular biologically active agent and shows concentration dependent reduction of glioblastoma (U251) cell survival.


Asunto(s)
Antineoplásicos/síntesis química , Dendrímeros/síntesis química , Isoxazoles/síntesis química , Neuroglía/efectos de los fármacos , Polietilenglicoles/química , Alquinos/química , Antineoplásicos/farmacología , Catálisis , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Cloruros/química , Reacción de Cicloadición , Dendrímeros/farmacología , Relación Dosis-Respuesta a Droga , Diseño de Fármacos , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Isoxazoles/farmacología , Neuroglía/patología , Nitrilos/química , Óxidos/química , Solubilidad , Agua/química , Compuestos de Zinc/química
8.
J Cell Mol Med ; 23(4): 2890-2900, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30710421

RESUMEN

Both type 2 diabetes (T2D) and obesity are characterized by excessive hyperlipidaemia and subsequent lipid droplet (LD) accumulation in adipose tissue. To investigate whether LDs also accumulate in ß-cells of T2D patients, we assessed the expression of PLIN2, a LD-associated protein, in non-diabetic (ND) and T2D pancreata. We observed an up-regulation of PLIN2 mRNA and protein in ß-cells of T2D patients, along with significant changes in the expression of lipid metabolism, apoptosis and oxidative stress genes. The increased LD buildup in T2D ß-cells was accompanied by inhibition of nuclear translocation of TFEB, a master regulator of autophagy and by down-regulation of lysosomal biomarker LAMP2. To investigate whether LD accumulation and autophagy were influenced by diabetic conditions, we used rat INS-1 cells to model the effects of hyperglycaemia and hyperlipidaemia on autophagy and metabolic gene expression. Consistent with human tissue, both LD formation and PLIN2 expression were enhanced in INS-1 cells under hyperglycaemia, whereas TFEB activation and autophagy gene expression were significantly reduced. Collectively, these results suggest that lipid clearance and overall homeostasis is markedly disrupted in ß-cells under hyperglycaemic conditions and interventions ameliorating lipid clearance could be beneficial in reducing functional impairments in islets caused by glucolipotoxicity.


Asunto(s)
Autofagia , Diabetes Mellitus Tipo 2/fisiopatología , Regulación de la Expresión Génica , Hiperglucemia/patología , Hiperlipidemias/patología , Células Secretoras de Insulina/patología , Lípidos/análisis , Animales , Apoptosis , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Estudios de Casos y Controles , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/metabolismo , Humanos , Hiperglucemia/etiología , Hiperglucemia/metabolismo , Hiperlipidemias/etiología , Hiperlipidemias/metabolismo , Células Secretoras de Insulina/metabolismo , Insulinoma/genética , Insulinoma/metabolismo , Insulinoma/patología , Proteína 2 de la Membrana Asociada a los Lisosomas/genética , Proteína 2 de la Membrana Asociada a los Lisosomas/metabolismo , Lisosomas/metabolismo , Lisosomas/patología , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Perilipina-2/genética , Perilipina-2/metabolismo , Ratas , Células Tumorales Cultivadas
9.
Nanomedicine ; 22: 102083, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31404650

RESUMEN

Gold nanoparticles have excellent potential for theranostic applications, but their impact on living cells is only partially understood. Many gold nanoparticles enter cells through endosomes/lysosomes which are linked to different cell organelles and compartments. Our study focuses on the unfolded protein response (UPR) in the endoplasmic reticulum (ER), cytoplasmic RNA-granules and proteostasis, because they are established indicators of cell stress and key regulators of cellular homeostasis. Using HeLa and renal proximal tubule cells as model systems, we show that gold nanourchins reduce cell proliferation, cause ER stress and impair proteostasis. Specifically, gold nanourchins activate the PERK-branch of the UPR, promote RNA oxidation, enhance P-body formation, and accumulate the oxidative stress marker Nrf2 and NFκB in nuclei. Taken together, our study demonstrates that gold nanourchins compromise ER, redox, protein, and RNA homeostasis. These insights provide new information on the cellular responses and molecular changes that gold nanourchins elicit in mammalian cells.


Asunto(s)
Oro/toxicidad , Nanopartículas del Metal/toxicidad , Proteostasis/efectos de los fármacos , ARN/genética , Estrés Fisiológico/efectos de los fármacos , Animales , Antioxidantes/farmacología , Apoptosis/efectos de los fármacos , Biomarcadores/metabolismo , Proliferación Celular/efectos de los fármacos , Citoplasma/metabolismo , Estrés del Retículo Endoplásmico/efectos de los fármacos , Células HeLa , Humanos , Células LLC-PK1 , Modelos Biológicos , Factor 2 Relacionado con NF-E2/metabolismo , FN-kappa B/metabolismo , Oxidación-Reducción , Estrés Oxidativo/efectos de los fármacos , Tamaño de la Partícula , Porcinos , Respuesta de Proteína Desplegada/efectos de los fármacos
10.
Nat Methods ; 12(3): 195-8, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25622108

RESUMEN

We have developed a versatile new class of genetically encoded fluorescent biosensor based on reversible exchange of the heterodimeric partners of green and red dimerization-dependent fluorescent proteins. We demonstrate the use of this strategy to construct both intermolecular and intramolecular ratiometric biosensors for qualitative imaging of caspase activity, Ca(2+) concentration dynamics and other second-messenger signaling activities.


Asunto(s)
Técnicas Biosensibles/métodos , Proteínas Fluorescentes Verdes/metabolismo , Proteínas Luminiscentes/metabolismo , Calcio/metabolismo , Calmodulina/metabolismo , Caspasa 3/genética , Proteínas Fluorescentes Verdes/genética , Células HeLa , Humanos , Proteínas Luminiscentes/genética , Imagen Molecular/métodos , Multimerización de Proteína , Proteína Fluorescente Roja
11.
Mol Pharm ; 14(8): 2607-2615, 2017 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-28520445

RESUMEN

New therapeutics for glioblastoma multiforme and our ability to deliver them using efficient nanocarriers constitute topical areas of research. We report a comparative study of temozolomide and quercetin in the treatment of glioblastoma (GBM) in three-dimensions, and their incorporation into micelles obtained from synthetically articulated architectural copolymers, and a commercially available linear polymer poly(ethylene glycol)-poly(lactic-co-glycolic acid) (PEG-PLGA). A versatile synthetic methodology to telodendrimers, which can be easily adapted to the needs of other therapeutic interventions, is presented. These dendritic block copolymers self-assemble into micelles and offer a platform for single or combination drug therapy. Telodendrimer micelles loaded with quercetin did not exhibit superior cell killing effect over the free drug, but acetazolamide, an inhibitor carbonic anhydrase IX, significantly reduced GBM cell viability in 3D spheroids. Results from these studies show that high loading of drugs into telodendrimer micelles requires a physical fit between the biologically active agent and telodendrimer nanocarrier, and points toward new possibilities for incorporation of chemotherapeutic and other agents to enhance their effectiveness.


Asunto(s)
Dacarbazina/análogos & derivados , Lactatos/química , Poliésteres/química , Polietilenglicoles/química , Acetazolamida/química , Acetazolamida/farmacología , Anhidrasa Carbónica IX/antagonistas & inhibidores , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Dacarbazina/química , Dacarbazina/farmacología , Dendrímeros/química , Sistemas de Liberación de Medicamentos/métodos , Glioblastoma/metabolismo , Humanos , Micelas , Quercetina/química , Quercetina/farmacología , Esferoides Celulares/citología , Esferoides Celulares/metabolismo , Temozolomida
12.
Int J Mol Sci ; 18(2)2017 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-28125025

RESUMEN

Air pollution of anthropogenic origin is largely from the combustion of biomass (e.g., wood), fossil fuels (e.g., cars and trucks), incinerators, landfills, agricultural activities and tobacco smoke. Air pollution is a complex mixture that varies in space and time, and contains hundreds of compounds including volatile organic compounds (e.g., benzene), metals, sulphur and nitrogen oxides, ozone and particulate matter (PM). PM0.1 (ultrafine particles (UFP)), those particles with a diameter less than 100 nm (includes nanoparticles (NP)) are considered especially dangerous to human health and may contribute significantly to the development of numerous respiratory and cardiovascular diseases such as chronic obstructive pulmonary disease (COPD) and atherosclerosis. Some of the pathogenic mechanisms through which PM0.1 may contribute to chronic disease is their ability to induce inflammation, oxidative stress and cell death by molecular mechanisms that include transcription factors such as nuclear factor κB (NF-κB) and nuclear factor (erythroid-derived 2)-like 2 (Nrf2). Epigenetic mechanisms including non-coding RNA (ncRNA) may also contribute towards the development of chronic disease associated with exposure to PM0.1. This paper highlights emerging molecular concepts associated with inhalational exposure to PM0.1 and their ability to contribute to chronic respiratory and systemic disease.


Asunto(s)
Contaminantes Atmosféricos/efectos adversos , Contaminación del Aire/efectos adversos , Susceptibilidad a Enfermedades , Material Particulado/efectos adversos , Animales , Antioxidantes/metabolismo , Biomarcadores , Muerte Celular , Epigénesis Genética , Predisposición Genética a la Enfermedad , Humanos , Inflamación/etiología , Inflamación/metabolismo , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Enfermedades Respiratorias/etiología , Enfermedades Respiratorias/metabolismo , Transducción de Señal
13.
J Neuroinflammation ; 13(1): 116, 2016 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-27220286

RESUMEN

BACKGROUND: Organelle remodeling processes are evolutionarily conserved and involved in cell functions during development, aging, and cell death. Some endogenous and exogenous molecules can modulate these processes. Docosahexaenoic acid (DHA), an omega-3 polyunsaturated fatty acid, has mainly been considered as a modulator of plasma membrane fluidity in brain development and aging, while DHA's role in organelle remodeling in specific neural cell types at the ultrastructural level remains largely unexplored. DHA is notably incorporated into dynamic organelles named lipid bodies (LBs). We hypothesized that DHA could attenuate the inflammatory response in lipopolysaccharide (LPS)-activated microglia by remodeling LBs and altering their functional interplay with mitochondria and other associated organelles. RESULTS: We used electron microscopy to analyze at high spatial resolution organelle changes in N9 microglial cells exposed to the proinflammogen LPS, with or without DHA supplementation. Our results revealed that DHA reverses several effects of LPS in organelles. In particular, a large number of very small and grouped LBs was exclusively found in microglial cells exposed to DHA. In contrast, LBs in LPS-stimulated cells in the absence of DHA were sparse and large. LBs formed in the presence of DHA were generally electron-dense, suggesting DHA incorporation into these organelles. The accumulation of LBs in microglial cells from mouse and human was confirmed in situ. In addition, DHA induced numerous contacts between LBs and mitochondria and reversed the frequent disruption of mitochondrial integrity observed upon LPS stimulation. Dilation of the endoplasmic reticulum lumen was also infrequent following DHA treatment, suggesting that DHA reduces oxidative stress and protein misfolding. Lipidomic analysis in N9 microglial cells treated with DHA revealed an increase in phosphatidylserine, indicating the role of this phospholipid in normalization and maintenance of physiological membrane functions. This finding was supported by a marked reduction of microglial filopodia and endosome number and significant reduction of LPS-induced phagocytosis. CONCLUSIONS: DHA attenuates the inflammatory response in LPS-stimulated microglial cells by remodeling LBs and altering their interplay with mitochondria and other associated organelles. Our findings point towards a mechanism by which omega-3 DHA participates in organelle reorganization and contributes to the maintenance of neural cell homeostasis.


Asunto(s)
Ácidos Docosahexaenoicos/farmacología , Gotas Lipídicas/efectos de los fármacos , Microglía/efectos de los fármacos , Animales , Línea Celular Transformada , Citocinas/metabolismo , Citocinas/ultraestructura , Relación Dosis-Respuesta a Droga , Ácidos Grasos/metabolismo , Gotas Lipídicas/ultraestructura , Lipopolisacáridos/farmacología , Ratones , Microglía/ultraestructura , Microscopía Electrónica de Transmisión , Orgánulos/efectos de los fármacos , Orgánulos/ultraestructura , Fagocitosis/efectos de los fármacos , Factores de Tiempo , Triglicéridos/metabolismo
14.
J Neuroinflammation ; 12: 34, 2015 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-25889069

RESUMEN

BACKGROUND: Recent studies have revealed that excessive activation of microglia and inflammation-mediated neurotoxicity are implicated in the progression of several neurological disorders. In particular, chronic inflammation in vivo and exposure of cultured brain cells to lipopolysaccharide (LPS) in vitro can adversely change microglial morphology and function. This can have both direct and indirect effects on synaptic structures and functions. The integrity of dendritic spines, the postsynaptic component of excitatory synapses, dictates synaptic efficacy. Interestingly, dysgenesis of dendritic spines has been found in many neurological diseases associated with ω-3 polyunsaturated fatty acid (PUFA) deficiency and cognitive decline. In contrast, supplemented ω-3 PUFAs, such as docosahexaenoic acid (DHA), can partly correct spine defects. Hence, we hypothesize that DHA directly affects synaptic integrity and indirectly through neuron-glia interaction. Strong activation of microglia by LPS is accompanied by marked release of nitric oxide and formation of lipid bodies (LBs), both dynamic biomarkers of inflammation. Here we investigated direct effects of DHA on synaptic integrity and its indirect effects via microglia in the hippocampal CA1 region. METHODS: Microglia (N9) and organotypic hippocampal slice cultures were exposed to the proinflammagen LPS (100 ng/ml) for 24 h. Biochemical and morphological markers of inflammation were investigated in microglia and CA1 regions of hippocampal slices. As biomarkers of hyperactive microglia, mitochondrial function, nitric oxide release and LBs (number, size, LB surface-associated proteins) were assessed. Changes in synaptic transmission of CA1 pyramidal cells were determined following LPS and DHA (25-50 µM) treatments by recording spontaneous AMPA-mediated miniature excitatory postsynaptic currents (mEPSCs). RESULTS: Microglia responded to LPS stimulation with a significant decrease of mitochondrial function, increased nitric oxide production and an increase in the formation of large LBs. LPS treatment led to a significant reduction of dendritic spine densities and an increase in the AMPA-mediated mEPSC inter-event interval (IEI). DHA normalized the LPS-induced abnormalities in both neurons and microglia, as revealed by the restoration of synaptic structures and functions in hippocampal CA1 pyramidal neurons. CONCLUSION: Our findings indicate that DHA can prevent LPS-induced abnormalities (neuroinflammation) by reducing inflammatory biomarkers, thereby normalizing microglia activity and their effect on synaptic function.


Asunto(s)
Espinas Dendríticas/efectos de los fármacos , Ácidos Docosahexaenoicos/farmacología , Hipocampo/citología , Microglía/efectos de los fármacos , Neuronas/citología , Neuronas/efectos de los fármacos , Potenciales de Acción/efectos de los fármacos , Animales , Animales Recién Nacidos , Bencimidazoles/metabolismo , Carbocianinas/metabolismo , Células Cultivadas , Citocromos c/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Técnicas In Vitro , Gotas Lipídicas/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Ratones Transgénicos , Neuronas/fisiología , Óxido Nítrico/metabolismo , Técnicas de Cultivo de Órganos , Perilipina-2 , Polisacáridos/farmacología
15.
Biomacromolecules ; 16(9): 3073-82, 2015 Sep 14.
Artículo en Inglés | MEDLINE | ID: mdl-26218295

RESUMEN

Hyperactivity of microglia and loss of functional circuitry is a common feature of many neurological disorders including those induced or exacerbated by inflammation. Herein, we investigate the response of microglia and changes in hippocampal dendritic postsynaptic spines by dendritic polyglycerol sulfate (dPGS) treatment. Mouse microglia and organotypic hippocampal slices were exposed to dPGS and an inflammogen (lipopolysaccharides). Measurements of intracellular fluorescence and confocal microscopic analyses revealed that dPGS is avidly internalized by microglia but not CA1 pyramidal neurons. Concentration and time-dependent response studies consistently showed no obvious toxicity of dPGS. The adverse effects induced by proinflammogen LPS exposure were reduced and dendritic spine morphology was normalized with the addition of dPGS. This was accompanied by a significant reduction in nitrite and proinflammatory cytokines (TNF-α and IL-6) from hyperactive microglia suggesting normalized circuitry function with dPGS treatment. Collectively, these results suggest that dPGS acts anti-inflammatory, inhibits inflammation-induced degenerative changes in microglia phenotype and rescues dendritic spine morphology.


Asunto(s)
Región CA1 Hipocampal/metabolismo , Espinas Dendríticas/metabolismo , Glicerol/farmacología , Microglía/metabolismo , Polímeros/farmacología , Células Piramidales/metabolismo , Animales , Línea Celular , Espinas Dendríticas/patología , Inflamación/inducido químicamente , Inflamación/metabolismo , Inflamación/patología , Interleucina-6/metabolismo , Lipopolisacáridos/toxicidad , Ratones , Ratones Transgénicos , Microglía/patología , Enfermedades del Sistema Nervioso/inducido químicamente , Enfermedades del Sistema Nervioso/metabolismo , Enfermedades del Sistema Nervioso/patología , Células Piramidales/patología , Factor de Necrosis Tumoral alfa/metabolismo
16.
Cell Mol Life Sci ; 71(21): 4259-73, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24740795

RESUMEN

Gold nanoparticles have emerged as promising tools for cancer research and therapy, where they can promote thermal killing. The molecular mechanisms underlying these events are not fully understood. The geometry and size of gold nanoparticles can determine the severity of cellular damage. Therefore, small and big gold nanospheres as well as gold nanoflowers were evaluated side-by-side. To obtain quantitative data at the subcellular and molecular level, we assessed how gold nanoparticles, either alone or in combination with mild hyperthermia, altered the physiology of cultured human breast cancer cells. Our analyses focused on the nucleus, because this organelle is essential for cell survival. We showed that all the examined gold nanoparticles associated with nuclei. However, their biological effects were quantitatively different. Thus, depending on the shape and size, gold nanoparticles changed multiple nuclear parameters. They redistributed stress-sensitive regulators of nuclear biology, altered the nuclear morphology, reorganized nuclear laminae and envelopes, and inhibited nucleolar functions. In particular, gold nanoparticles reduced the de novo biosynthesis of RNA in nucleoli, the subnuclear compartments that produce ribosomes. While small gold nanospheres and nanoflowers, but not big gold nanospheres, damaged the nucleus at normal growth temperature, several of these defects were further exacerbated by mild hyperthermia. Taken together, the toxicity of gold nanoparticles correlated with changes in nuclear organization and function. These results emphasize that the cell nucleus is a prominent target for gold nanoparticles of different morphologies. Moreover, we demonstrated that RNA synthesis in nucleoli provides quantitative information on nuclear damage and cancer cell survival.


Asunto(s)
Neoplasias de la Mama/terapia , Nanopartículas del Metal/química , Nanotecnología/métodos , Núcleo Celular/metabolismo , Proliferación Celular , Supervivencia Celular , Femenino , Colorantes Fluorescentes/química , Calor , Humanos , Hipertermia Inducida , Células MCF-7 , Microscopía Confocal , Ribosomas/metabolismo , Sales de Tetrazolio/química , Tiazoles/química
17.
Acc Chem Res ; 46(3): 672-80, 2013 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-22775328

RESUMEN

The dramatic increase in the use of nanoparticles (NP) in industry and research has raised questions about the potential toxicity of such materials. Unfortunately, not enough is known about how the novel, technologically-attractive properties of NPs correlate with the interactions that may take place at the nano/bio interface. The academic, industrial, and regulatory communities are actively seeking answers to the growing concerns on the impact of nanotechnology on humans. In this Account we adopt quantum dots (QDs) as an illustrative example of the difficulties associated with the development of a rational science-based approach to nanotoxicology. The optical properties of QDs are far superior to those of organic dyes in terms of emission and absorption bandwidths, quantum yield, and resistance to photobleaching. Moreover, QDs may be decorated with targeting moieties or drugs and, therefore, are candidates for site-specific medical imaging and for drug delivery, for example in cancer treatment. Earlier this year researchers demonstrated that QD-based imaging using monkeys caused no adverse effects although QDs accumulated in lymph nodes, bone marrow, liver, and spleen for up to 3 months after injection. Such persistence of QDs in live animals does, however, raise concerns about the safety of using QDs both in the laboratory and in the clinic. Researchers anticipate that QDs will be increasingly used not only in clinical applications but also in various manufactured products. For example, QD-solar cells have emerged as viable contenders to complement or replace dye-sensitized solar cells; CdTe/CdS thin film cells have already captured approximately 10 percent of the global market, and in addition, QDs can serve as components of sensors and as emitting materials in LEDs. Given the clear indications that QDs will inevitably become components of a wide range of manufactured and consumer products, researchers and policy makers need to understand the possible health risks associated with exposure to QDs. In this Account, we initially review the known mechanisms by which QDs can damage cells, including oxidative stress elicited by reactive oxygen species (ROS). We discuss lesser-known impairments induced in cells by nanomolar to picomolar concentrations of QDs, which imply that cadmium-containing QDs can exert genotoxic, epigenetic, and metalloestrogenic effects. These observations strongly suggest that minute concentrations of QDs could be sufficient to cause long lasting, even transgenerational, effects. We also consider various modes by which humans could be exposed to QDs in their work or through the environment. Although considerable advances have been made in enhancing the stability and overall quality of QDs, over time they can partially degrade in the environment or in biological systems, and eventually cause small, but cumulative undesirable effects. A combination of toxicological, genetic, epigenetic and imaging approaches is required to create comprehensive guidelines for evaluating the nanotoxicity of nanomaterials, including QDs. Prior to biological investigations with these materials, an indispensible step must be the full characterization of NPs by complementary techniques. Specifically, the concentration, size, charge, and ligand stability of NPs in biological media must be known if we are to understand fully how the properties of nanoparticles and of their biological environment contribute to cytotoxicity.


Asunto(s)
Puntos Cuánticos/toxicidad , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Contaminantes Ambientales/toxicidad , Humanos
18.
Aging Cell ; 23(5): e14178, 2024 05.
Artículo en Inglés | MEDLINE | ID: mdl-38685568

RESUMEN

Senescence is a heterogenous and dynamic process in which various cell types undergo cell-cycle arrest due to cellular stressors. While senescence has been implicated in aging and many human pathologies, therapeutic interventions remain inadequate due to the absence of a comprehensive set of biomarkers in a context-dependent manner. Polyphenols have been investigated as senotherapeutics in both preclinical and clinical settings. However, their use is hindered by limited stability, toxicity, modest bioavailability, and often inadequate concentration at target sites. To address these limitations, nanocarriers such as polymer nanoparticles and lipid vesicles can be utilized to enhance the efficacy of senolytic polyphenols. Focusing on widely studied senolytic agents-specifically fisetin, quercetin, and resveratrol-we provide concise summaries of their physical and chemical properties, along with an overview of preclinical and clinical findings. We also highlight common signaling pathways and potential toxicities associated with these agents. Addressing challenges linked to nanocarriers, we present examples of senotherapeutic delivery to various cell types, both with and without nanocarriers. Finally, continued research and development of senolytic agents and nanocarriers are encouraged to reduce the undesirable effects of senescence on different cell types and organs. This review underscores the need for establishing reliable sets of senescence biomarkers that could assist in evaluating the effectiveness of current and future senotherapeutic candidates and nanocarriers.


Asunto(s)
Portadores de Fármacos , Nanopartículas , Polifenoles , Senoterapéuticos , Humanos , Polifenoles/farmacología , Polifenoles/química , Nanopartículas/química , Nanopartículas/uso terapéutico , Senoterapéuticos/farmacología , Portadores de Fármacos/química , Animales , Senescencia Celular/efectos de los fármacos , Quercetina/farmacología , Quercetina/química
19.
ACS Pharmacol Transl Sci ; 7(3): 680-692, 2024 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-38481701

RESUMEN

While stroke represents one of the main causes of death worldwide, available effective drug treatment options remain limited to classic thrombolysis with recombinant tissue plasminogen activator (rtPA) for arterial-clot occlusion. Following stroke, multiple pathways become engaged in producing a vicious proinflammatory cycle through the release of damage-associated molecular patterns (DAMPs) such as high-mobility group box 1 (HMGB1) and heat shock protein 70 kDa (HSP72). HMGB1, in particular, can activate proinflammatory cytokine production when acetylated (AcHMGB1), a form that prefers cytosolic localization and extracellular release. This study aimed at determining how HMGB1 and HSP72 are modulated and affected following treatment with the anti-inflammatory compound resveratrol and novel platelet membrane-derived nanocarriers loaded with rtPA (CSM@rtPA) recently developed by our group for ischemic artery recanalization. Under ischemic conditions of oxygen-glucose deprivation (OGD), nuclear abundance of HMGB1 and AcHMGB1 in microglia and macrophages decreased, whereas treatment with CSM@rtPA did not alter nuclear or cytosolic abundance. Resveratrol treatment markedly increased the cytosolic abundance of HSP72 in microglia. Using proximity ligation assays, we determined that HSP72 interacted with HMGB1 and with acetylated HMGB1. The interaction was differentially affected under the OGD conditions. Resveratrol treatment under the OGD further decreased HSP72-HMGB1 interactions, whereas, in contrast, treatment increased HSP72-AcHMGB1 interactions in microglia. This study points out a salient molecular interaction suited for a two-pronged nanotherapeutic intervention in stroke: enhancement of rtPA's thrombolytic activity and modulation of cytosolic interactions between HMGB1 and HSP72 by resveratrol.

20.
Biochim Biophys Acta ; 1821(4): 607-17, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22289388

RESUMEN

Lipid droplets (LDs) are neutral lipid-rich organelles involved in many cellular processes. A well-known example is their accumulation in leukocytes upon activation by pro-inflammatory stimuli such as lipopolysaccharides (LPS) derived from gram-negative bacteria. A role of LDs and LD-associated proteins during inflammation in the brain is unknown, however. We have now studied their dynamics and regulation in microglia, the resident immune cells in the brain. We find that LPS treatment of microglia leads to the accumulation in them of LDs, and enhancement of the size of LDs. This induction of LDs was abolished by triacsin C, an inhibitor of triglyceride biosynthesis. LPS strongly activated c-Jun N-terminal kinase (JNK) and p38 MAPK stress signaling pathways and increased the expression of LD-associated protein perilipin-2 (ADRP) in a time-dependent manner. Immunostaining showed that perilipin-2 in LPS-treated microglia predominantly colocalized with LDs. Inhibitors of p38 α/ß (SB203580) and PI3K/Akt pathway (LY294002), but not that of JNK (SP600125), reduced LPS-induced LD accumulation and eliminated the activating effect of LPS on perilipin-2. In addition, cytosolic phospholipase A(2) (cPLA(2)-α), a key enzyme for arachidonic acid release, colocalized with LPS-induced LDs. These observations suggest that LDs may play an important role in eicosanoid synthesis in activated microglia; they provide a novel insight into the regulation of LDs in inflammatory cells of the brain and point to a potential role of p38 α/ß in LPS-induced LD accumulation. Collectively, our findings imply that LD formation and perilipin-2 induction could be microglial biomarkers of inflammation in the central nervous system.


Asunto(s)
Gránulos Citoplasmáticos/efectos de los fármacos , Lípidos/química , Lipopolisacáridos/farmacología , Microglía/efectos de los fármacos , Animales , Antracenos/farmacología , Western Blotting , Células Cultivadas , Cromonas/farmacología , Gránulos Citoplasmáticos/metabolismo , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Fosfolipasas A2 Grupo IV/metabolismo , Imidazoles/farmacología , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Cinética , Proteínas de la Membrana/metabolismo , Ratones , Microglía/citología , Microglía/metabolismo , Microscopía Confocal , Modelos Biológicos , Morfolinas/farmacología , Ácido Oléico/farmacología , Perilipina-2 , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Piridinas/farmacología , Factores de Tiempo , Triglicéridos/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA