Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 116
Filtrar
Más filtros

País/Región como asunto
Intervalo de año de publicación
1.
Artículo en Inglés | MEDLINE | ID: mdl-39078396

RESUMEN

Inflammation is an important physiological response of the organism to restore homeostasis upon pathogenic or damaging stimuli. However, persistence of the harmful trigger, or a deficient resolution of the process can evolve into a state of low-grade, chronic inflammation. This condition is strongly associated to the development of several increasingly prevalent and serious chronic conditions such as obesity, cancer and cardiovascular diseases, elevating overall morbidity and mortality worldwide. The current pandemic of chronic diseases underscores the need to address chronic inflammation, its pathogenic mechanisms and potential preventive measures to limit its current widespread impact. The present review discusses the current knowledge and research gaps regarding the association between low-grade chronic inflammation and chronic diseases, focusing on obesity, cardiovascular diseases, digestive diseases and cancer. We examine the state-of-the-art in selected aspects of the topic, and propose future directions and approaches for the field.

2.
Bioorg Chem ; 151: 107681, 2024 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-39106711

RESUMEN

Aberrant activation of the Hedgehog (Hh) signalling pathway has been associated with the development and progression of pancreatic cancer. For this reason, blockade of Hh pathway by inhibitors targeting the G protein-coupled receptor Smoothened (SMO) has been considered as a therapeutic target for the treatment of this cancer. In our previous work, we obtained a new SMO ligand based on a purine scaffold (compound I), which showed interesting antitumor activity in several cancer cell lines. In this work, we report the design and synthesis of 17 new purine derivatives, some of which showed high cytotoxic effect on Mia-PaCa-2 (Hh-dependent pancreatic cancer cell lines) and low toxicity on non-neoplastic HEK-293 cells compared with gemcitabine, such as 8f, 8g and 8h (IC50 = 4.56, 4.11 and 3.08 µM, respectively). Two of these purines also showed their ability to bind to SMO through NanoBRET assays (pKi = 5.17 for 8f and 5.01 for 8h), with higher affinities to compound I (pKi = 1.51). In addition, docking studies provided insight the purine substitution pattern is related to the affinity on SMO. Finally, studies of Hh inhibition for selected purines, using a transcriptional functional assay based on luciferase activity in NIH3T3 Shh-Light II cells, demonstrated that 8g reduced GLI activity with a IC50 = 6.4 µM as well as diminished the expression of Hh target genes in two specific Hh-dependent cell models, Med1 cells and Ptch1-/- mouse embryonic fibroblasts. Therefore, our results provide a platform for the design of SMO ligands that could be potential selective cytotoxic agents for the treatment of pancreatic cancer.

3.
J Nanobiotechnology ; 22(1): 248, 2024 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-38741193

RESUMEN

The use of nanomaterials in medicine offers multiple opportunities to address neurodegenerative disorders such as Alzheimer's and Parkinson's disease. These diseases are a significant burden for society and the health system, affecting millions of people worldwide without sensitive and selective diagnostic methodologies or effective treatments to stop their progression. In this sense, the use of gold nanoparticles is a promising tool due to their unique properties at the nanometric level. They can be functionalized with specific molecules to selectively target pathological proteins such as Tau and α-synuclein for Alzheimer's and Parkinson's disease, respectively. Additionally, these proteins are used as diagnostic biomarkers, wherein gold nanoparticles play a key role in enhancing their signal, even at the low concentrations present in biological samples such as blood or cerebrospinal fluid, thus enabling an early and accurate diagnosis. On the other hand, gold nanoparticles act as drug delivery platforms, bringing therapeutic agents directly into the brain, improving treatment efficiency and precision, and reducing side effects in healthy tissues. However, despite the exciting potential of gold nanoparticles, it is crucial to address the challenges and issues associated with their use in the medical field before they can be widely applied in clinical settings. It is critical to ensure the safety and biocompatibility of these nanomaterials in the context of the central nervous system. Therefore, rigorous preclinical and clinical studies are needed to assess the efficacy and feasibility of these strategies in patients. Since there is scarce and sometimes contradictory literature about their use in this context, the main aim of this review is to discuss and analyze the current state-of-the-art of gold nanoparticles in relation to delivery, diagnosis, and therapy for Alzheimer's and Parkinson's disease, as well as recent research about their use in preclinical, clinical, and emerging research areas.


Asunto(s)
Oro , Nanopartículas del Metal , Enfermedades Neurodegenerativas , alfa-Sinucleína , Proteínas tau , Humanos , Oro/química , Nanopartículas del Metal/química , Nanopartículas del Metal/uso terapéutico , Proteínas tau/metabolismo , Animales , Enfermedades Neurodegenerativas/tratamiento farmacológico , Enfermedades Neurodegenerativas/diagnóstico , Enfermedad de Parkinson/diagnóstico , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/diagnóstico , Sistemas de Liberación de Medicamentos/métodos , Biomarcadores
4.
J Neuroinflammation ; 20(1): 66, 2023 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-36895046

RESUMEN

BACKGROUND: Helicobacter pylori (Hp) infects the stomach of 50% of the world's population. Importantly, chronic infection by this bacterium correlates with the appearance of several extra-gastric pathologies, including neurodegenerative diseases. In such conditions, brain astrocytes become reactive and neurotoxic. However, it is still unclear whether this highly prevalent bacterium or the nanosized outer membrane vesicles (OMVs) they produce, can reach the brain, thus affecting neurons/astrocytes. Here, we evaluated the effects of Hp OMVs on astrocytes and neurons in vivo and in vitro. METHODS: Purified OMVs were characterized by mass spectrometry (MS/MS). Labeled OMVs were administered orally or injected into the mouse tail vein to study OMV-brain distribution. By immunofluorescence of tissue samples, we evaluated: GFAP (astrocytes), ßIII tubulin (neurons), and urease (OMVs). The in vitro effect of OMVs in astrocytes was assessed by monitoring NF-κB activation, expression of reactivity markers, cytokines in astrocyte-conditioned medium (ACM), and neuronal cell viability. RESULTS: Urease and GroEL were prominent proteins in OMVs. Urease (OMVs) was present in the mouse brain and its detection coincided with astrocyte reactivity and neuronal damage. In vitro, OMVs induced astrocyte reactivity by increasing the intermediate filament proteins GFAP and vimentin, the plasma membrane αVß3 integrin, and the hemichannel connexin 43. OMVs also produced neurotoxic factors and promoted the release of IFNγ in a manner dependent on the activation of the transcription factor NF-κB. Surface antigens on reactive astrocytes, as well as secreted factors in response to OMVs, were shown to inhibit neurite outgrowth and damage neurons. CONCLUSIONS: OMVs administered orally or injected into the mouse bloodstream reach the brain, altering astrocyte function and promoting neuronal damage in vivo. The effects of OMVs on astrocytes were confirmed in vitro and shown to be NF-κB-dependent. These findings suggest that Hp could trigger systemic effects by releasing nanosized vesicles that cross epithelial barriers and access the CNS, thus altering brain cells.


Asunto(s)
Helicobacter pylori , Ratones , Animales , Helicobacter pylori/metabolismo , Astrocitos , Ureasa/metabolismo , Ureasa/farmacología , FN-kappa B/metabolismo , Factor B del Complemento/metabolismo , Factor B del Complemento/farmacología , Modelos Animales de Enfermedad , Espectrometría de Masas en Tándem , Neuronas
5.
J Nanobiotechnology ; 21(1): 115, 2023 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-36978078

RESUMEN

BACKGROUND: The lack of predictive models that mimic the blood-brain barrier (BBB) hinders the development of effective drugs for neurodegenerative diseases. Animal models behave differently from humans, are expensive and have ethical constraints. Organ-on-a-chip (OoC) platforms offer several advantages to resembling physiological and pathological conditions in a versatile, reproducible, and animal-free manner. In addition, OoC give us the possibility to incorporate sensors to determine cell culture features such as trans-endothelial electrical resistance (TEER). Here, we developed a BBB-on-a-chip (BBB-oC) platform with a TEER measurement system in close distance to the barrier used for the first time for the evaluation of the permeability performance of targeted gold nanorods for theranostics of Alzheimer's disease. GNR-PEG-Ang2/D1 is a therapeutic nanosystem previously developed by us consisting of gold nanorods (GNR) functionalized with polyethylene glycol (PEG), angiopep-2 peptide (Ang2) to overcome the BBB and the D1 peptide as beta amyloid fibrillation inhibitor, finally obtaining GNR-PEG-Ang2/D1 which showed to be useful for disaggregation of the amyloid in in vitro and in vivo models. In this work, we evaluated its cytotoxicity, permeability, and some indications of its impact on the brain endothelium by employing an animal-free device based on neurovascular human cells. RESULTS: In this work, we fabricated a BBB-oC with human astrocytes, pericytes and endothelial cells and a TEER measuring system (TEER-BBB-oC) integrated at a micrometric distance of the endothelial barrier. The characterization displayed a neurovascular network and the expression of tight junctions in the endothelium. We produced GNR-PEG-Ang2/D1 and determined its non-cytotoxic range (0.05-0.4 nM) for plated cells included in the BBB-oC and confirmed its harmless effect at the highest concentration (0.4 nM) in the microfluidic device. The permeability assays revealed that GNR-PEG-Ang2/D1 cross the BBB and this entry is facilitated by Ang2 peptide. Parallel to the permeability analysis of GNR-PEG-Ang2/D1, an interesting behavior of the TJs expression was observed after its administration probably related to the ligands on the nanoparticle surface. CONCLUSIONS: BBB-oC with a novel TEER integrated setup which allow a correct read-out and cell imaging monitoring was proven as a functional and throughput platform to evaluate the brain permeability performance of nanotherapeutics in a physiological environment with human cells, putting forward a viable alternative to animal experimentation.


Asunto(s)
Enfermedad de Alzheimer , Barrera Hematoencefálica , Humanos , Barrera Hematoencefálica/metabolismo , Células Endoteliales/metabolismo , Enfermedad de Alzheimer/tratamiento farmacológico , Enfermedad de Alzheimer/metabolismo , Impedancia Eléctrica , Oro/farmacología , Astrocitos/metabolismo , Péptidos beta-Amiloides/metabolismo , Endotelio/metabolismo , Permeabilidad , Dispositivos Laboratorio en un Chip
6.
Chem Soc Rev ; 51(7): 2601-2680, 2022 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-35234776

RESUMEN

Recent advances in technology are expected to increase our current understanding of neuroscience. Nanotechnology and nanomaterials can alter and control neural functionality in both in vitro and in vivo experimental setups. The intersection between neuroscience and nanoscience may generate long-term neural interfaces adapted at the molecular level. Owing to their intrinsic physicochemical characteristics, gold nanostructures (GNSs) have received much attention in neuroscience, especially for combined diagnostic and therapeutic (theragnostic) purposes. GNSs have been successfully employed to stimulate and monitor neurophysiological signals. Hence, GNSs could provide a promising solution for the regeneration and recovery of neural tissue, novel neuroprotective strategies, and integrated implantable materials. This review covers the broad range of neurological applications of GNS-based materials to improve clinical diagnosis and therapy. Sub-topics include neurotoxicity, targeted delivery of therapeutics to the central nervous system (CNS), neurochemical sensing, neuromodulation, neuroimaging, neurotherapy, tissue engineering, and neural regeneration. It focuses on core concepts of GNSs in neurology, to circumvent the limitations and significant obstacles of innovative approaches in neurobiology and neurochemistry, including theragnostics. We will discuss recent advances in the use of GNSs to overcome current bottlenecks and tackle technical and conceptual challenges.


Asunto(s)
Nanoestructuras , Neurociencias , Oro , Nanoestructuras/uso terapéutico , Nanotecnología , Ingeniería de Tejidos
7.
Int J Mol Sci ; 24(4)2023 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-36835401

RESUMEN

Melphalan (Mel) is an antineoplastic widely used in cancer and other diseases. Its low solubility, rapid hydrolysis, and non-specificity limit its therapeutic performance. To overcome these disadvantages, Mel was included in ß-cyclodextrin (ßCD), which is a macromolecule that increases its aqueous solubility and stability, among other properties. Additionally, the ßCD-Mel complex has been used as a substrate to deposit silver nanoparticles (AgNPs) through magnetron sputtering, forming the ßCD-Mel-AgNPs crystalline system. Different techniques showed that the complex (stoichiometric ratio 1:1) has a loading capacity of 27%, an association constant of 625 M-1, and a degree of solubilization of 0.034. Added to this, Mel is partially included, exposing the NH2 and COOH groups that stabilize AgNPs in the solid state, with an average size of 15 ± 3 nm. Its dissolution results in a colloidal solution of AgNPs covered by multiple layers of the ßCD-Mel complex, with a hydrodynamic diameter of 116 nm, a PDI of 0.4, and a surface charge of 19 mV. The in vitro permeability assays show that the effective permeability of Mel increased using ßCD and AgNPs. This novel nanosystem based on ßCD and AgNPs is a promising candidate as a Mel nanocarrier for cancer therapy.


Asunto(s)
Nanopartículas del Metal , beta-Ciclodextrinas , Melfalán , Plata , beta-Ciclodextrinas/química , Solubilidad
8.
Nanomedicine ; 44: 102569, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35595016

RESUMEN

The ß-amyloid (Aß) peptide is one of the key etiological agents in Alzheimer's disease (AD). The in vivo detection of Aß species is challenging in all stages of the illness. Currently, the development of fluorescent probes allows the detection of Aß in animal models in the near-infrared region (NIR). However, considering future applications in biomedicine, it is relevant to develop strategies to improve detection of amyloid aggregates using NIR probes. An innovative approach to increase the fluorescence signal of these fluorophores is the use of plasmonic gold nanoparticles (surface-enhanced fluorescence effect). In this work, we improved the detection of Aß aggregates in C. elegans and mouse models of AD by co-administering functionalized gold nanorods (GNRs-PEG-D1) with the fluorescent probes CRANAD-2 or CRANAD-58, which bind selectively to different amyloid species (soluble and insoluble). This work shows that GNRs improve the detection of Aß using NIR probes in vivo.


Asunto(s)
Enfermedad de Alzheimer , Nanopartículas del Metal , Enfermedad de Alzheimer/diagnóstico por imagen , Péptidos beta-Amiloides/metabolismo , Animales , Caenorhabditis elegans , Colorantes Fluorescentes/química , Oro , Nanopartículas del Metal/química , Ratones
9.
Chemistry ; 27(14): 4670-4675, 2021 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-33368712

RESUMEN

Cytosolic protein delivery remains elusive. The inability of most proteins to cross the cellular membrane is a huge hurdle. Here we explore the unique photothermal properties of gold nanorods (AuNRs) to trigger cytosolic delivery of proteins. Both partners, protein and AuNRs, are modified with a protease-resistant cell-penetrating peptide with nuclear targeting properties to induce internalization. Once internalized, spatiotemporal control of protein release is achieved by near-infrared laser irradiation in the safe second biological window. Importantly, catalytic amounts of AuNRs are sufficient to trigger cytosolic protein delivery. To the best of our knowledge, this is the first time that AuNRs with their maximum of absorption in the second biological window are used to deliver proteins into the intracellular space. This strategy represents a powerful tool for the cytosolic delivery of virtually any class of protein.


Asunto(s)
Nanopartículas del Metal , Nanotubos , Línea Celular Tumoral , Oro , Fototerapia
10.
Sensors (Basel) ; 21(6)2021 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-33809416

RESUMEN

Alzheimer's disease (AD), considered a common type of dementia, is mainly characterized by a progressive loss of memory and cognitive functions. Although its cause is multifactorial, it has been associated with the accumulation of toxic aggregates of the amyloid-ß peptide (Aß) and neurofibrillary tangles (NFTs) of tau protein. At present, the development of highly sensitive, high cost-effective, and non-invasive diagnostic tools for AD remains a challenge. In the last decades, nanomaterials have emerged as an interesting and useful tool in nanomedicine for diagnostics and therapy. In particular, plasmonic nanoparticles are well-known to display unique optical properties derived from their localized surface plasmon resonance (LSPR), allowing their use as transducers in various sensing configurations and enhancing detection sensitivity. Herein, this review focuses on current advances in in vitro sensing techniques such as Surface-enhanced Raman scattering (SERS), Surface-enhanced fluorescence (SEF), colorimetric, and LSPR using plasmonic nanoparticles for improving the sensitivity in the detection of main biomarkers related to AD in body fluids. Additionally, we refer to the use of plasmonic nanoparticles for in vivo imaging studies in AD.


Asunto(s)
Enfermedad de Alzheimer , Nanopartículas del Metal , Enfermedad de Alzheimer/diagnóstico , Péptidos beta-Amiloides , Humanos , Espectrometría Raman , Resonancia por Plasmón de Superficie
11.
Int J Mol Sci ; 22(12)2021 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-34208594

RESUMEN

This article describes the synthesis and characterization of ß-cyclodextrin-based nano-sponges (NS) inclusion compounds (IC) with the anti-tumor drugs melphalan (MPH) and cytoxan (CYT), and the addition of gold nanoparticles (AuNPs) onto both systems, for the potential release of the drugs by means of laser irradiation. The NS-MPH and NS-CYT inclusion compounds were characterized using scanning electron microscopy (SEM), X-ray powder diffraction (XRPD), energy dispersive spectroscopy (EDS), thermogravimetric analysis (TGA), UV-Vis, and proton nuclear magnetic resonance (1H-NMR). Thus, the inclusion of MPH and CYT inside the cavities of NSs was confirmed. The association of AuNPs with the ICs was confirmed by SEM, EDS, TEM, and UV-Vis. Drug release studies using NSs synthesized with different molar ratios of ß-cyclodextrin and diphenylcarbonate (1:4 and 1:8) demonstrated that the ability of NSs to entrap and release the drug molecules depends on the crosslinking between the cyclodextrin monomers. Finally, irradiation assays using a continuous laser of 532 nm showed that photothermal drug release of both MPH and CYT from the cavities of NSs via plasmonic heating of AuNPs is possible.


Asunto(s)
Ciclodextrinas , Ciclofosfamida/administración & dosificación , Portadores de Fármacos , Oro , Melfalán/administración & dosificación , Nanopartículas del Metal , Técnicas de Química Sintética , Ciclodextrinas/química , Portadores de Fármacos/química , Sistemas de Liberación de Medicamentos , Liberación de Fármacos/efectos de la radiación , Oro/química , Luz , Espectroscopía de Resonancia Magnética , Nanopartículas del Metal/química , Nanopartículas del Metal/ultraestructura , Temperatura , Termogravimetría , Tocoferoles , Difracción de Rayos X
12.
J Nanobiotechnology ; 18(1): 20, 2020 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-31973696

RESUMEN

BACKGROUND: Extracellular vesicles (EVs) have shown great potential for targeted therapy, as they have a natural ability to pass through biological barriers and, depending on their origin, can preferentially accumulate at defined sites, including tumors. Analyzing the potential of EVs to target specific cells remains challenging, considering the unspecific binding of lipophilic tracers to other proteins, the limitations of fluorescence for deep tissue imaging and the effect of external labeling strategies on their natural tropism. In this work, we determined the cell-type specific tropism of B16F10-EVs towards cancer cell and metastatic tumors by using fluorescence analysis and quantitative gold labeling measurements. Surface functionalization of plasmonic gold nanoparticles was used to promote indirect labeling of EVs without affecting size distribution, polydispersity, surface charge, protein markers, cell uptake or in vivo biodistribution. Double-labeled EVs with gold and fluorescent dyes were injected into animals developing metastatic lung nodules and analyzed by fluorescence/computer tomography imaging, quantitative neutron activation analysis and gold-enhanced optical microscopy. RESULTS: We determined that B16F10 cells preferentially take up their own EVs, when compared with colon adenocarcinoma, macrophage and kidney cell-derived EVs. In addition, we were able to detect the preferential accumulation of B16F10 EVs in small metastatic tumors located in lungs when compared with the rest of the organs, as well as their precise distribution between tumor vessels, alveolus and tumor nodules by histological analysis. Finally, we observed that tumor EVs can be used as effective vectors to increase gold nanoparticle delivery towards metastatic nodules. CONCLUSIONS: Our findings provide a valuable tool to study the distribution and interaction of EVs in mice and a novel strategy to improve the targeting of gold nanoparticles to cancer cells and metastatic nodules by using the natural properties of malignant EVs.


Asunto(s)
Antineoplásicos/química , Vesículas Extracelulares/química , Oro/química , Neoplasias Pulmonares/diagnóstico por imagen , Neoplasias Pulmonares/metabolismo , Melanoma/química , Nanopartículas del Metal/química , Adenocarcinoma/diagnóstico por imagen , Adenocarcinoma/terapia , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral , Permeabilidad de la Membrana Celular , Neoplasias del Colon/diagnóstico por imagen , Neoplasias del Colon/terapia , Colorantes Fluorescentes/química , Humanos , Pulmón/metabolismo , Melanoma Experimental/diagnóstico por imagen , Melanoma Experimental/terapia , Ratones , Ratones Endogámicos C57BL , Imagen Óptica , Propiedades de Superficie , Distribución Tisular
13.
Mediators Inflamm ; 2020: 8680692, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32410869

RESUMEN

Oncolytic virus therapy has been tested against cancer in preclinical models and clinical assays. Current evidence shows that viruses induce cytopathic effects associated with fusogenic protein-mediated syncytium formation and immunogenic cell death of eukaryotic cells. We have previously demonstrated that tumor cell bodies generated from cells expressing the fusogenic protein of the infectious salmon anemia virus (ISAV-F) enhance crosspriming and display prophylactic antitumor activity against melanoma tumors. In this work, we evaluated the effects of the expression of ISAV-F on the B16 melanoma model, both in vitro and in vivo, using chitosan nanoparticles as transfection vehicle. We confirmed that the transfection of B16 tumor cells with chitosan nanoparticles (NP-ISAV) allows the expression of a fusogenically active ISAV-F protein and decreases cell viability because of syncytium formation in vitro. However, the in vivo transfection induces a delay in tumor growth, without inducing changes on the lymphoid populations in the tumor and the spleen. Altogether, our observations show that expression of ISAV fusion protein using chitosan nanoparticles induces cell fusion in melanoma cells and slight antitumor response.


Asunto(s)
Antineoplásicos/farmacología , Quitosano/química , Melanoma/tratamiento farmacológico , Nanopartículas/química , Viroterapia Oncolítica/métodos , Neoplasias Cutáneas/tratamiento farmacológico , Animales , Línea Celular Tumoral , Supervivencia Celular , Quitosano/metabolismo , ADN Complementario/metabolismo , Células Gigantes/metabolismo , Humanos , Isavirus/genética , Linfocitos/citología , Melanoma Experimental , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Nanomedicina/métodos , Infecciones por Orthomyxoviridae/genética , Proteínas Recombinantes de Fusión/química , Propiedades de Superficie , Transfección
14.
Sensors (Basel) ; 20(21)2020 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-33142935

RESUMEN

Prothrombin-related thrombophilia is a genetic disorder produced by a substitution of a single DNA base pair, replacing guanine with adenine, and is detected mainly by polymerase chain reaction (PCR). A suitable alternative that could detect the single point mutation without requiring sample amplification is the surface plasmon resonance (SPR) technique. SPR biosensors are of great interest: they offer a platform to monitor biomolecular interactions, are highly selective, and enable rapid analysis in real time. Oligonucleotide-based SPR biosensors can be used to differentiate complementary sequences from partially complementary or noncomplementary strands. In this work, a glass chip covered with an ultrathin (50 nm) gold film was modified with oligonucleotide strands complementary to the mutated or normal (nonmutated) DNA responsible for prothrombin-related thrombophilia, forming two detection platforms called mutated thrombophilia (MT) biosensor and normal thrombophilia (NT) biosensor. The results show that the hybridization response is obtained in 30 min, label free and with high reproducibility. The sensitivity obtained in both systems was approximately 4 ΔµRIU/nM. The dissociation constant and limits of detection calculated were 12.2 nM and 20 pM (3 fmol), respectively, for the MT biosensor, and 8.5 nM and 30 pM (4.5 fmol) for the NT biosensor. The two biosensors selectively recognize their complementary strand (mutated or normal) in buffer solution. In addition, each platform can be reused up to 24 times when the surface is regenerated with HCl. This work contributes to the design of the first SPR biosensor for the detection of prothrombin-related thrombophilia based on oligonucleotides with single point mutations, label-free and without the need to apply an amplification method.


Asunto(s)
Técnicas Biosensibles , Oligonucleótidos , Protrombina/genética , Trombofilia , Humanos , Oligonucleótidos/genética , Mutación Puntual , Reproducibilidad de los Resultados , Resonancia por Plasmón de Superficie , Trombofilia/diagnóstico , Trombofilia/genética
15.
Addict Biol ; 24(5): 994-1007, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-30239077

RESUMEN

Chronic ethanol consumption leads to brain oxidative stress and neuroinflammation, conditions known to potentiate and perpetuate each other. Several studies have shown that neuroinflammation results in increases in chronic ethanol consumption. Recent reports showed that the intra-cerebroventricular administration of mesenchymal stem cells to rats consuming alcohol chronically markedly inhibited oxidative-stress, abolished neuroinflammation and greatly reduced chronic alcohol intake and post deprivation relapse-like alcohol intake. However, the intra-cerebroventricular administration of living cells is not suitable as a treatment of a chronic condition. The present study aimed at inhibiting ethanol intake by the non-invasive intranasal administration of human mesenchymal stem cell products: exosomes, microvesicles (40 to 150 nm) with marked antioxidant activity extruded from mesenchymal stem cells. The exosome membrane can fuse with the plasma membrane of cells in different tissues, thus delivering their content intracellularly. The study showed that the weekly intranasal administration of mesenchymal stem cell-derived exosomes to rats consuming alcohol chronically (1) inhibited their ethanol intake by 84 percent and blunted the relapse-like 'binge' drinking that follows an alcohol deprivation period and ethanol re-access. (2) Intranasally administered exosomes were found in the brain within 24 hours; (3) fully reversed both alcohol-induced hippocampal oxidative-stress, evidenced by a lower ratio of oxidized to reduced glutathione, and neuroinflammation, shown by a reduced astrocyte activation and microglial density; and (4) increased glutamate transporter GLT1 expression in nucleus accumbens, counteracting the inhibition of glutamate transporter activity, reportedly depressed under oxidative-stress conditions. Possible translational implications are envisaged.


Asunto(s)
Consumo de Bebidas Alcohólicas/prevención & control , Exosomas/trasplante , Trasplante de Células Madre Mesenquimatosas/métodos , Estrés Oxidativo/fisiología , Administración Intranasal , Consumo de Bebidas Alcohólicas/fisiopatología , Sistema de Transporte de Aminoácidos X-AG/metabolismo , Animales , Astrocitos/fisiología , Enfermedad Crónica , Femenino , Hipocampo/metabolismo , Humanos , Inyecciones Intraventriculares , Microglía/fisiología , Núcleo Accumbens/metabolismo , Corteza Prefrontal/metabolismo , ARN Mensajero/metabolismo , Ratas Wistar , Prevención Secundaria , Grasa Subcutánea/citología , Síndrome de Abstinencia a Sustancias/prevención & control
16.
J Nanobiotechnology ; 16(1): 60, 2018 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-30097010

RESUMEN

BACKGROUND: Stable and non-toxic fluorescent markers are gaining attention in molecular diagnostics as powerful tools for enabling long and reliable biological studies. Such markers should not only have a long half-life under several assay conditions showing no photo bleaching or blinking but also, they must allow for their conjugation or functionalization as a crucial step for numerous applications such as cellular tracking, biomarker detection and drug delivery. RESULTS: We report the functionalization of stable fluorescent markers based on nanodiamonds (NDs) with a bifunctional peptide. This peptide is made of a cell penetrating peptide and a six amino acids long ß-sheet breaker peptide that is able to recognize amyloid ß (Aß) aggregates, a biomarker for the Alzheimer disease. Our results indicate that functionalized NDs (fNDs) are not cytotoxic and can be internalized by the cells. The fNDs allow ultrasensitive detection (at picomolar concentrations of NDs) of in vitro amyloid fibrils and amyloid aggregates in AD mice brains. CONCLUSIONS: The fluorescence of functionalized NDs is more stable than that of fluorescent markers commonly used to stain Aß aggregates such as Thioflavin T. These results pave the way for performing ultrasensitive and reliable detection of Aß aggregates involved in the pathogenesis of the Alzheimer disease.


Asunto(s)
Enfermedad de Alzheimer/diagnóstico , Amiloide/análisis , Colorantes Fluorescentes/química , Nanodiamantes/química , Amiloide/metabolismo , Péptidos beta-Amiloides/análisis , Péptidos beta-Amiloides/metabolismo , Animales , Benzotiazoles/química , Benzotiazoles/toxicidad , Biomarcadores/análisis , Línea Celular , Supervivencia Celular/efectos de los fármacos , Péptidos de Penetración Celular/química , Colorantes Fluorescentes/toxicidad , Humanos , Ratones Transgénicos , Nanodiamantes/toxicidad , Agregado de Proteínas
17.
Arch Pharm (Weinheim) ; 351(5): e1800024, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29611620

RESUMEN

With the purpose of expanding the structural variety of chemical compounds available as pharmacological tools for the treatment of Alzheimer's disease, we synthesized and evaluated a novel series of indole-benzoxazinones (Family I) and benzoxazine-arylpiperazine derivatives (Family II) for potential human acetylcholinesterase (hAChE) inhibitory properties. The most active compounds 7a and 7d demonstrated effective inhibitory profiles with Ki values of 20.3 ± 0.9 µM and 20.2 ± 0.9 µM, respectively. Kinetic inhibition assays showed non-competitive inhibition of AChE by the tested compounds. According to our docking studies, the most active compounds from both series (Families I and II) showed a binding mode similar to donepezil and interact with the same residues.


Asunto(s)
Acetilcolinesterasa/efectos de los fármacos , Benzoxazinas/farmacología , Inhibidores de la Colinesterasa/farmacología , Piperazinas/farmacología , Acetilcolinesterasa/metabolismo , Enfermedad de Alzheimer/tratamiento farmacológico , Benzoxazinas/síntesis química , Benzoxazinas/química , Inhibidores de la Colinesterasa/síntesis química , Inhibidores de la Colinesterasa/química , Donepezilo , Diseño de Fármacos , Humanos , Indanos/farmacología , Simulación del Acoplamiento Molecular , Piperazinas/síntesis química , Piperazinas/química , Piperidinas/farmacología , Unión Proteica , Relación Estructura-Actividad
18.
Molecules ; 23(7)2018 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-29973523

RESUMEN

The antibacterial effects against Staphylococcus epidermidis of five silver carboxylate complexes with anti-inflammatory ligands were studied in order to analyze and compare them in terms of stability (in solution and after exposure to UV light), and their antibacterial and morphological differences. Four effects of the Ag-complexes were evidenced by transmission electronic microscopy (TEM) and scanning electronic microscopy (SEM): DNA condensation, membrane disruption, shedding of cytoplasmic material and silver compound microcrystal penetration of bacteria. 5-Chlorosalicylic acid (5Cl) and sodium 4-aminosalicylate (4A) were the most effective ligands for synthesizing silver complexes with high levels of antibacterial activity. However, Ag-5Cl was the most stable against exposure UV light (365 nm). Cytotoxic effects were tested against two kinds of eukaryotic cells: murine fibroblast cells (T10 1/2) and human epithelial ovarian cancer cells (A2780). The main objective was to identify changes in their antibacterial properties associated with potential decomposition and the implications for clinical applications.


Asunto(s)
Antibacterianos/síntesis química , Complejos de Coordinación/síntesis química , Plata/química , Staphylococcus epidermidis/efectos de los fármacos , Ácidos Aminosalicílicos/química , Animales , Antibacterianos/química , Antibacterianos/farmacología , Línea Celular , Complejos de Coordinación/química , Complejos de Coordinación/farmacología , Estabilidad de Medicamentos , Humanos , Ratones , Pruebas de Sensibilidad Microbiana , Microscopía Electrónica de Rastreo , Microscopía Electrónica de Transmisión , Salicilatos/química
19.
Molecules ; 23(5)2018 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-29783629

RESUMEN

Here we report the incorporation of gold nanostructures (nanospheres or nanorods, functionalized with carboxylate-end PEG) and curcumin oil-in-water (O/W) nanoemulsions (CurNem) into alginate microgels using the dripping technique. While gold nanostructures are promising nanomaterials for photothermal therapy applications, CurNem possess important pharmacological activities as reported here. In this sense, we evaluated the effect of CurNem on cell viability of both cancerous and non-cancerous cell lines (AGS and HEK293T, respectively), demonstrating preferential toxicity in cancer cells and safety for the non-cancerous cells. After incorporating gold nanostructures and CurNem together into the microgels, microstructures with diameters of 220 and 540 µm were obtained. When stimulating microgels with a laser, the plasmon effect promoted a significant rise in the temperature of the medium; the temperature increase was higher for those containing gold nanorods (11⁻12 °C) than nanospheres (1⁻2 °C). Interestingly, the incorporation of both nanosystems in the microgels maintains the photothermal properties of the gold nanostructures unmodified and retains with high efficiency the curcumin nanocarriers. We conclude that these results will be of interest to design hydrogel formulations with therapeutic applications.


Asunto(s)
Portadores de Fármacos/química , Oro/química , Nanosferas/química , Nanotubos/química , Antineoplásicos/administración & dosificación , Antineoplásicos/química , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Curcumina/administración & dosificación , Curcumina/química , Liberación de Fármacos , Emulsiones , Geles , Células HEK293 , Humanos , Rayos Láser , Tamaño de la Partícula , Fotoquimioterapia/métodos , Polietilenglicoles/química , Solubilidad , Propiedades de Superficie
20.
Nanomedicine ; 13(5): 1645-1652, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28285163

RESUMEN

We studied the effect of gold nanoparticle (AuNP) size, surface charge, concentration and morphology on the integrity of the blood-brain barrier (BBB) in a well-established in vitro model set-up. We focused on the effect of peptide functionalized hollow gold nanospheres and gold nanorods, which selectively bind to amyloidogenic ß-amyloid structures. These AuNP conjugates have already been successfully tested as photothermal absorbers for potential application in Alzheimer's disease (AD) therapy in an in vitro set-up, but may exhibit a low passage through the BBB due to their overall negative charge. Our results show that: (i) small (1.4 nm) AuNPs strongly affects the BBB integrity, (ii) negative surface charge impedes BBB passage, and (iii) this charge effect caused by the peptide is compensated by covalent coupling to a polyethylene glycol ligand stabilizing the particles in diluted manner.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Barrera Hematoencefálica , Nanopartículas del Metal , Péptidos beta-Amiloides , Transporte Biológico , Oro , Humanos , Péptidos , Unión Proteica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA