Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 21(4)2020 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-32085611

RESUMEN

Annexin A6 (AnxA6) is the largest member of the annexin family of proteins present in matrix vesicles (MVs). MVs are a special class of extracellular vesicles that serve as a nucleation site during cartilage, bone, and mantle dentin mineralization. In this study, we assessed the localization of AnxA6 in the MV membrane bilayer using native MVs and MV biomimetics. Biochemical analyses revealed that AnxA6 in MVs can be divided into three distinct groups. The first group corresponds to Ca2+-bound AnxA6 interacting with the inner leaflet of the MV membrane. The second group corresponds to AnxA6 localized on the surface of the outer leaflet. The third group corresponds to AnxA6 inserted in the membrane's hydrophobic bilayer and co-localized with cholesterol (Chol). Using monolayers and proteoliposomes composed of either dipalmitoylphosphatidylcholine (DPPC) to mimic the outer leaflet of the MV membrane bilayer or a 9:1 DPPC:dipalmitoylphosphatidylserine (DPPS) mixture to mimic the inner leaflet, with and without Ca2+, we confirmed that, in agreement with the biochemical data, AnxA6 interacted differently with the MV membrane. Thermodynamic analyses based on the measurement of surface pressure exclusion (πexc), enthalpy (ΔH), and phase transition cooperativity (Δt1/2) showed that AnxA6 interacted with DPPC and 9:1 DPPC:DPPS systems and that this interaction increased in the presence of Chol. The selective recruitment of AnxA6 by Chol was observed in MVs as probed by the addition of methyl-ß-cyclodextrin (MßCD). AnxA6-lipid interaction was also Ca2+-dependent, as evidenced by the increase in πexc in negatively charged 9:1 DPPC:DPPS monolayers and the decrease in ΔH in 9:1 DPPC:DPPS proteoliposomes caused by the addition of AnxA6 in the presence of Ca2+ compared to DPPC zwitterionic bilayers. The interaction of AnxA6 with DPPC and 9:1 DPPC:DPPS systems was distinct even in the absence of Ca2+ as observed by the larger change in Δt1/2 in 9:1 DPPC:DPPS vesicles as compared to DPPC vesicles. Protrusions on the surface of DPPC proteoliposomes observed by atomic force microscopy suggested that oligomeric AnxA6 interacted with the vesicle membrane. Further work is needed to delineate possible functions of AnxA6 at its different localizations and ways of interaction with lipids.


Asunto(s)
Anexina A6/metabolismo , Calcificación Fisiológica , Matriz Extracelular/metabolismo , Vesículas Extracelulares/metabolismo , 1,2-Dipalmitoilfosfatidilcolina/química , Rastreo Diferencial de Calorimetría , Colesterol/metabolismo , Humanos , Membrana Dobles de Lípidos/metabolismo , Microdominios de Membrana/metabolismo , Microscopía de Fuerza Atómica , Proteolípidos/metabolismo
2.
Arch Biochem Biophys ; 667: 14-21, 2019 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-30998909

RESUMEN

Matrix vesicles (MVs) are a class of extracellular vesicles that initiate mineralization in cartilage, bone, and other vertebrate tissues by accumulating calcium ions (Ca2+) and inorganic phosphate (Pi) within their lumen and forming a nucleation core (NC). After further sequestration of Ca2+ and Pi, the NC transforms into crystalline complexes. Direct evidence of the existence of the NC and its maturation have been provided solely by analyses of dried samples. We isolated MVs from chicken embryo cartilage and used atomic force microscopy peak force quantitative nanomechanical property mapping (AFM-PFQNM) to measure the nanomechanical and morphological properties of individual MVs under both mineralizing (+Ca2+) and non-mineralizing (-Ca2+) fluid conditions. The elastic modulus of MVs significantly increased by 4-fold after incubation in mineralization buffer. From AFM mapping data, we inferred the morphological changes of MVs as mineralization progresses: prior to mineralization, a punctate feature, the NC, is present within MVs and this feature grows and stiffens during mineralization until it occupies most of the MV lumen. Dynamic light scattering showed a significant increase in hydrodynamic diameter and no change in the zeta potential of hydrated MVs after incubation with Ca2+. This validates that crystalline complexes, which are strongly negative relative to MVs, were forming within the lumen of MVs. These data were substantiated by transmission electron microscopy energy dispersive X-ray and Fourier transform infrared spectroscopic analyses of dried MVs, which provide evidence that the complexes increased in size, crystallinity, and Ca/P ratio within MVs during the mineralization process.


Asunto(s)
Biomineralización/fisiología , Vesículas Extracelulares/química , Vesículas Extracelulares/metabolismo , Microscopía de Fuerza Atómica/métodos , Animales , Fenómenos Biomecánicos , Cartílago/química , Cartílago/metabolismo , Cartílago/ultraestructura , Embrión de Pollo , Vesículas Extracelulares/ultraestructura , Microscopía Electrónica de Transmisión , Espectroscopía Infrarroja por Transformada de Fourier
3.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1862(9): 813-822, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28499815

RESUMEN

In this study we provide the first evidence of the interaction of a truncated-TRAF2 with lipid raft microdomains. We have analyzed this interaction by measuring the diffusion coefficient of the protein in large and giant unilamellar vesicles (LUVs and GUVs, respectively) obtained both from synthetic lipid mixtures and from natural extracts. Steady-state fluorescence measurements performed with synthetic vesicles indicate that this truncated form of TRAF2 displays a tighter binding to raft-like LUVs with respect to the control (POPC-containing LUVs), and that this process depends on the protein oligomeric state. Generalized Polarization measurements and spectral phasor analysis revealed that truncated-TRAF2 affects the membrane fluidity, especially when vesicles are heated up at physiological temperature. The addition of nanomolar concentration of TRAF2 in GUVs also seems to exert a mechanical action, as demonstrated by the formation of intraluminal vesicles, a process in which ganglioside GM1 plays a crucial role.


Asunto(s)
Membrana Dobles de Lípidos/química , Membrana Dobles de Lípidos/metabolismo , Lípidos/química , Microdominios de Membrana/química , Microdominios de Membrana/metabolismo , Factor 2 Asociado a Receptor de TNF/química , Factor 2 Asociado a Receptor de TNF/metabolismo , Difusión , Fluorescencia , Gangliósido G(M1)/metabolismo , Humanos , Fluidez de la Membrana/fisiología , Unión Proteica/fisiología , Dominios Proteicos , Liposomas Unilamelares/metabolismo
4.
Surg Innov ; 24(3): 268-275, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28178883

RESUMEN

BACKGROUND: A retained surgical item in patients (gossypiboma) is a persisting problem, despite consistent improvements and existing guidelines in counting instruments and sponges. Previous experiences with radiofrequency identification technology (RFID) tracking sponges show that it could represent an innovation, in order to reduce the criticism and increase the effectiveness during surgical procedures. We present an automated system that allows reduction of errors and improves safety in the operating room. METHODS: The system consists of 3 antennas, surgical sponges containing RFID tags, and dedicated software applications, with Wi-Fi real-time communication between devices. The first antenna provides the initial count of gauzes; the second a real-time counting during surgery, including the sponges thrown into the kick-bucket; and the third can be used in the event of uneven sponge count. The software allows management at all stages of the process. RESULTS: In vitro and in vivo tests were performed: the system provided excellent results in detecting sponges in patients' body. Hundred percent retained sponges were detected correctly, even when they were overlapped. No false positive or false negative was recorded. The counting procedure turned out to be more streamlined and efficient and it could save time in a standard procedure. CONCLUSIONS: The RFID system for sponge tracking was shown to be experimentally a reliable and feasible method to track sponges with a full detection accuracy in the operating room. The results indicate the system to be safe and effective with acceptable cost-effective parameters.


Asunto(s)
Cuerpos Extraños , Dispositivo de Identificación por Radiofrecuencia , Cirugía Asistida por Computador/instrumentación , Tapones Quirúrgicos de Gaza , Animales , Ingeniería Biomédica , Simulación por Computador , Diseño de Equipo , Cuerpos Extraños/diagnóstico , Cuerpos Extraños/prevención & control , Humanos , Internet , Fantasmas de Imagen , Programas Informáticos , Cirugía Asistida por Computador/métodos , Porcinos
5.
J Transl Med ; 14: 37, 2016 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-26847645

RESUMEN

BACKGROUND: Nitrobenzoxadiazole derivatives (NBDs), including NBDHEX and the recently developed MC3181, are promising anticancer agents able to target glutathione transferase and inhibit both its catalytic activity and ability to sequester TNF-receptor associated factor 2 (TRAF2) and c-Jun N-terminal kinase (JNK). NBDs have been shown to impair the growth and survival of a broad-spectrum of tumor types, in vitro and in vivo. Herein, we evaluated the effects of the new compound MC3181 on U-2OS osteosarcoma cells and investigated the impact of both NBDHEX and MC3181 on autophagy. METHODS: Cell viability was evaluated by sulforhodamine B assay. The dissociation of the TRAF2-GSTP1-1 complex was detected by proximity ligation assay, while the phospho-activation of JNK was assessed by western blotting. The effects of NBDs on autophagy were evaluated by GFP-LC3 puncta formation, western blotting for LC3-II and p62, and LC3 turnover assay in the presence of bafilomycin A1. The role of JNK in the reduction of autophagic flux caused by NBDs was investigated using JNK1 shRNA-transfected cells. Fluorogenic caspase activity assay and flow cytometric analysis of DNA content were used to determine the cytotoxic effects of NBDs on JNK1-silenced cells. RESULTS: Similar to NBDHEX, MC3181 reduced viability and activated TRAF2/JNK signaling in U-2OS cells. Moreover, NBDs induced the accumulation of autophagic vesicles and LC3-II while reducing both basal and nutritional stress-induced autophagic flux. Furthermore, increased levels of both LC3-II and the autophagy selective substrate p62 were observed in different tumor cell lines treated with NBDs, the concurrent increase of these markers being consistent with an impairment of autophagosome clearance. Autophagy inhibition by NBDs required JNK activity: NBDs caused autophagy inhibition and caspase-3 activation in JNK-positive U-2OS, but no autophagic flux inhibition or caspase-3 activation in JNK-silenced cells. CONCLUSIONS: Our demonstration that NBDs can act as late-phase autophagy inhibitors opens new opportunities to fully exploit their therapeutic potential. This may not rely solely on their effectiveness in inducing cell cycle arrest and apoptosis, but also on their ability to weaken the capacity of tumor cells to endure stress conditions via autophagy. In addition, this study provides evidence that JNK can participate in impairing autophagy.


Asunto(s)
Autofagia/efectos de los fármacos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Oxadiazoles/farmacología , Apoptosis/efectos de los fármacos , Caspasa 3/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Silenciador del Gen/efectos de los fármacos , Humanos , Concentración 50 Inhibidora , Proteínas Asociadas a Microtúbulos/metabolismo , Osteosarcoma/metabolismo , Osteosarcoma/patología , Fagosomas/efectos de los fármacos , Fagosomas/metabolismo , Proteínas de Unión al ARN/metabolismo , Transducción de Señal/efectos de los fármacos , Estrés Fisiológico/efectos de los fármacos , Factor 2 Asociado a Receptor de TNF/metabolismo
6.
Nanomedicine ; 12(2): 255-68, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26707894

RESUMEN

Cartilage undergoes drastic structural changes during the development of osteoarthritis and cannot heal itself due to a defective chondrocyte response. Thus, much effort has been invested in the development of disease modifying drugs able to block key mediators within the cartilage matrix and biochemical pathways inside chondrocytes. However, the delivery of therapeutic agents into cartilage is ineffective. This has led to the use of cartilage-targeted nanodrugs to accumulate therapeutic agents into specific cartilage sub-compartments. This review will describe the nanodrugs targeted to specific components of cartilage matrix to generate drug reservoirs within the cartilage. The nanodrugs used as chondrocyte-specific gene delivery systems are also described. Although the use of cartilage-targeted nanodrugs in osteoarthritis is still in its infancy, these studies lay the foundation for the development of novel approaches for preventing the progression of cartilage breakdown and improving the quality of life of patients with osteoarthritis. FROM THE CLINICAL EDITOR: Osteoarthritis is a degeneration of joint cartilage, which affects a large number of aging people. Current therapy for disease modification is often suboptimal. Recent research in nanomedicine has led to the design and use of nanodrugs with the aim to help reverse the disease process. In this comprehensive review, the authors described and discussed various nanodrugs in the hope that newer drugs could be discovered in the future.


Asunto(s)
Cartílago Articular/efectos de los fármacos , Portadores de Fármacos/química , Sistemas de Liberación de Medicamentos/métodos , Nanopartículas/química , Osteoartritis/tratamiento farmacológico , Preparaciones Farmacéuticas/administración & dosificación , Animales , Cartílago Articular/metabolismo , Cartílago Articular/patología , Condrocitos/efectos de los fármacos , Condrocitos/metabolismo , Condrocitos/patología , Humanos , Nanomedicina/métodos , Osteoartritis/metabolismo , Osteoartritis/patología
7.
Biochemistry ; 54(40): 6153-61, 2015 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-26390021

RESUMEN

TNF receptor-associated factors (TRAFs) are characterized by an oligomeric structure that plays a fundamental role in the binding process with membrane receptors. In this work, we studied the trimer-to-monomer (T ↔ 3M) equilibrium transition of the TRAF2 C-terminal domain using both chemical (dilution/guanidinium hydrochloride) and mechanical stress (high pressure) to induce the dissociation of the native protein into subunits. The experimental results and computer simulations indicate that stable monomers exist and that their population accounts for 15% of the total TRAF2 molecules already at a physiological intracellular concentration (≈1 µM), being instead the predominant species in the nanomolar concentration range. Because the total amount of TRAF2 changes during a cell cycle, the monomer-trimer equilibrium can be crucial for regulating the activities of TRAF2 in vivo.


Asunto(s)
Multimerización de Proteína , Factor 2 Asociado a Receptor de TNF/química , Línea Celular , Humanos , Simulación de Dinámica Molecular , Estabilidad Proteica , Factor 2 Asociado a Receptor de TNF/análisis
8.
Langmuir ; 31(27): 7572-80, 2015 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-26102092

RESUMEN

Several diseases are related to the lack or to the defective activity of a particular enzyme; therefore, these proteins potentially represent a very interesting class of therapeutics. However, their application is hampered by their rapid degradation and immunogenic side effects. Most attempts to increase the bioavailability of therapeutic enzymes are based on formulations in which the protein is entrapped within a scaffold structure but needs to be released to exert its activity. In this work, an alternative method will be described, designed to keep the enzyme in its active form inside a nanoparticle (NP) without the need to release it, thus maintaining the protective action of the nanoscaffold during the entire period of administration. In this approach, liposomes were used as nanotemplates for the synthesis of polyacrylamide hydrogel NPs under nondenaturing conditions, optimizing the polymer properties to obtain a mesh size small enough to limit the enzyme release while allowing the free diffusion of its substrates and products. The enzyme Cu, Zn-superoxide dismutase was chosen as a test case for this study, but our results indicate that the approach is generalizable to other enzymes. Biocompatible, size-tunable nanoparticles have been obtained, with a good encapsulation efficiency (37%), in which the enzyme maintains its activity. This system represents a promising tool for enzyme-based therapy, which would protect the protein from antibodies and degradation while allowing it to exert its catalytic activity.


Asunto(s)
Resinas Acrílicas/química , Hidrogel de Polietilenoglicol-Dimetacrilato/química , Nanopartículas/química , Superóxido Dismutasa/metabolismo , Resinas Acrílicas/síntesis química , Resinas Acrílicas/metabolismo , Biocatálisis , Activación Enzimática , Hidrogel de Polietilenoglicol-Dimetacrilato/síntesis química , Hidrogel de Polietilenoglicol-Dimetacrilato/metabolismo , Liposomas , Tamaño de la Partícula , Propiedades de Superficie
9.
J Nanosci Nanotechnol ; 14(1): 98-114, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24730253

RESUMEN

The recent advent of nanomedicine holds potential to revolutionize cancer therapy. This innovative discipline has paved the way for the emergence of a new class of drugs based on nanoengineered particles. These "nanodrugs" are designed to greatly enhance drug therapeutic indices. First-generation nanodrugs consisted of conventional anti-cancer drugs loaded into/onto nanoengineered particles (nanocarriers) devoid of targeting features (non-targeted nanodrugs). Non-targeted nanodrugs have provided the opportunity to carry large amounts of drugs, including poorly water-soluble and/or permeable drugs, to several types of tumors, improving the therapeutic index with respect to comparable free drugs. Although effective, the primary delivery mechanism of non-targeted nanodrugs was through passive tissue accumulation, due to pathophysiological differences between tumor-associated and healthy vessels, and through non-specific targeting of cell subsets, posing the danger of off-target binding and effects. Recently, the therapeutic indices of certain anti-cancer drugs were further improved by attaching targeting ligands to nanodrugs (targeted-nanodrugs). Targeted-nanodrugs selectively bind to cognate receptors expressed on target cells and enter cells more efficiently than non-targeted formulations. Although these advancements have been sufficiently beneficial to place targeted-nanodrugs into clinical development for use in cancer therapy, they also come at a price. The addition of ligands to drug-loaded nanocarriers often leads to additional synthesis steps and costs, and more complex biological performance relative to ligand-devoid nanodrugs. Here, we will discuss the benefits and challenges facing the addition of targeting features to nanodrugs for cancer therapy.


Asunto(s)
Antineoplásicos/administración & dosificación , Antineoplásicos/farmacocinética , Terapia Molecular Dirigida/métodos , Nanocápsulas/administración & dosificación , Nanocápsulas/química , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Animales , Antineoplásicos/química , Diseño de Fármacos , Humanos
10.
New Microbiol ; 37(4): 495-501, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25387286

RESUMEN

Urinary tract infection is a common disease diagnosed from symptoms and clinical signs, and bacterial count per volume of urine. This study have evaluated the BiesseBioscreen analyzer as a new way to analyze urine samples en- abling fast screening of urine, prior to reference standard methods currently utilized in microbiology analysis labo- ratory. We analyzed 962 urine samples from outpatients and inpatients of the Tor Vergata (TV) University Hospital of the University of Rome "Tor Vergata". All samples were processed both with the BiesseBioscreen and with the standard methodology adopted by the clinical microbiology laboratory of TV Hospital and the results were com- pared. Of the samples analyzed 54.9% were concordant negative with the reference method and 21.6% concordant positive, 23.3% resulted false positive and 0.2% false negative. The results obtained from BiesseBioscreen showed a sensitivity of 99.0%, indicating it as a system suitable to rule out urinary tract infection. BiesseBioscreen could represent a valid method for screening negative samples to exclude from culture test with a potential reduction in time, workload and costs of the diagnosis.


Asunto(s)
Bacterias/crecimiento & desarrollo , Bacteriuria/diagnóstico , Pruebas Diagnósticas de Rutina/métodos , Orina/microbiología , Bacterias/aislamiento & purificación , Bacteriuria/microbiología , Pruebas Diagnósticas de Rutina/instrumentación , Humanos , Juego de Reactivos para Diagnóstico
11.
Biochim Biophys Acta ; 1821(11): 1425-33, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22877990

RESUMEN

Fatty acid amide hydrolase (FAAH) is a membrane protein that plays a relevant role in the metabolism of fatty acid amides and esters. It degrades important neurotransmitters such as oleamide and anandamide, and it has been involved in a number of human pathological conditions, representing therefore a valuable target for biochemical and pharmacological research. In this study, we have investigated in vitro the structure-function relationship of rat and human FAAHs. In particular circular dichroism, fluorescence spectroscopy and light scattering measurements have been performed, in order to characterize the structural features of the two proteins, both in the presence and absence of the irreversible inhibitor methoxyarachidonyl-fluorophosphonate. The results demonstrate that the structural dynamics of the two FAAHs are different, despite their high sequence homology and overall similarity in temperature-dependence. Additionally, membrane binding and kinetic assays of both FAAHs indicate that also the functional properties of the two enzymes are different in their interaction with lipid bilayers and with exogenous inhibitors. These findings suggest that pre-clinical studies of FAAH-dependent human diseases based only on animal models should be interpreted with caution, and that the efficacy of new drugs targeted to FAAH should be tested in vitro, on both rat and human enzymes.


Asunto(s)
Amidohidrolasas/antagonistas & inhibidores , Amidohidrolasas/metabolismo , Ácidos Araquidónicos/farmacología , Inhibidores Enzimáticos/farmacología , Ácidos Grasos/metabolismo , Organofosfonatos/farmacología , Amidohidrolasas/química , Animales , Humanos , Cinética , Estabilidad Proteica , Estructura Secundaria de Proteína , Ratas , Especificidad por Sustrato
12.
Bioconjug Chem ; 24(6): 852-8, 2013 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-23682992

RESUMEN

Recent evidence regarding the role of regulatory T cells (Treg) in tumor development has suggested that the manipulation of Treg function selectively in the tumor microenvironment would be a desirable immunotherapy approach. Targeting intratumor immune populations would reduce side effects on peripheral healthy cells and increase antitumor efficacy of immunotherapies. However, no current approaches are available which enable selective in vivo targeting of intratumor Treg or other immune cell subpopulations. Herein, we investigated the ability of ligands against Treg-specific receptors to drive selective internalization of PEG-modified single-walled carbon nanotubes (PEG-SWCNTs) into Treg residing in the tumor microenvironment. We focused our attention on the glucocorticoid-induced TNFR-related receptor (GITR), as it showed higher overexpression on intratumor vs peripheral (i.e., splenic) Treg compared to other reported Treg-specific markers (folate receptor 4, CD103, and CD39). Ex vivo investigations showed that the Treg targeting efficiency and selectivity of PEG-SWCNTs depended on incubation time, dose, number of ligands per nanotube, and targeted surface marker. In vivo investigations showed that PEG-SWCNTs armed with GITR ligands targeted Treg residing in a B16 melanoma more efficiently then intratumor non-Treg or splenic Treg. The latter result was achieved by exploiting a combination of passive tumor targeting due to enhanced tumor vascular permeability, naturally increased intratumor Treg vs effector T cell (Teff) ratio, and active targeting of markers that are enriched in intratumor vs splenic Treg. We also found that PEG-SWCNTs loaded with GITR ligands were internalized by Treg through receptor-mediated endocytosis and transported into the cytoplasm and nucleus ex vivo and in vivo. This is the first example of intratumor immune cell targeting and we hope it will pave the way to innovative immunotherapies against cancer.


Asunto(s)
Nanotubos de Carbono/química , Neoplasias Experimentales/tratamiento farmacológico , Neoplasias Experimentales/inmunología , Polietilenglicoles/farmacología , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Animales , Relación Dosis-Respuesta a Droga , Humanos , Ratones , Ratones Endogámicos C57BL , Polietilenglicoles/química , Receptores de Antígenos de Linfocitos T/antagonistas & inhibidores , Receptores de Antígenos de Linfocitos T/inmunología , Relación Estructura-Actividad , Ensayos Antitumor por Modelo de Xenoinjerto
13.
J Extracell Biol ; 1(1): e34, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38938684

RESUMEN

Extracellular vesicles (EVs) are lipid bilayer-enclosed nanosized particles released by all cell types during physiological as well as pathophysiological processes to carry out diverse biological functions, including acting as sources of cellular dumping, signalosomes and mineralisation nanoreactors. The ability of EVs to perform specific biological functions is due to their biochemical machinery. Among the components of the EVs' biochemical machinery, surface proteins are of critical functional significance as they mediate the interactions of EVs with components of the extracellular milieu, the extracellular matrix and neighbouring cells. Surface proteins are thought to be native, that is, pre-assembled on the EVs' surface by the parent cells before the vesicles are released. However, numerous pieces of evidence have suggested that soluble proteins are acquired by the EVs' surface from the extracellular milieu and further modulate the biological functions of EVs during innate and adaptive immune responses, autoimmune disorders, complement activation, coagulation, viral infection and biomineralisation. Herein, we will describe the methods currently used to identify the EVs' surface proteins and discuss recent knowledge on the functional relevance of the soluble proteins acquired by EVs.

14.
Biomacromolecules ; 12(10): 3381-93, 2011 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-21916410

RESUMEN

Since their discovery at the end of the previous millennium, carbon nanotubes (CNTs) have been the object of thousands of papers describing their applications in fields ranging from physics to electronics, photonics, chemistry, biology, and medicine. The development of chemical approaches to modify their graphitic sidewalls enabled the generation of poly(ethylene glycol) (PEG)-modified CNTs and their exploration in multiple biomedical applications. Studies at the cellular and organism level revealed that PEG-modified CNTs have favorable pharmacokinetic and toxicology profiles. Recently, PEG-modified CNTs have been successfully tested in preclinical studies in the fields of oncology, neurology, vaccination, and imaging, suggesting that they are well suited for the generation of novel multifunctional nanodrugs. Here we will review published data about the application of PEG-modified CNTs as in vitro and in vivo therapeutic and imaging tools and describe what is known about the interaction between PEG-modified CNTs and biological systems. Although several pieces of the puzzle are still missing, we will also attempt to formulate a preliminary structure-function model for PEG-modified CNT cellular trafficking, disposition, and side effects.


Asunto(s)
Materiales Biocompatibles/farmacocinética , Sistemas de Liberación de Medicamentos/métodos , Imagen Molecular/métodos , Nanomedicina/métodos , Nanotubos de Carbono/química , Polietilenglicoles/química , Animales , Materiales Biocompatibles/química , Materiales Biocompatibles/metabolismo , Evaluación Preclínica de Medicamentos , Monitoreo de Drogas , Humanos , Ratones , Nanotubos de Carbono/ultraestructura , Polietilenglicoles/metabolismo , Relación Estructura-Actividad
15.
J Nanosci Nanotechnol ; 10(8): 5293-301, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21125885

RESUMEN

Nanotechnology-introduced materials have promising applications as nanocarriers for drugs, peptides, proteins and nucleic acids. Several studies showed that the geometry (shape and size) and chemical properties of nanoparticles affect the kinetics and pathways of cellular uptake and their intracellular trafficking and signaling. Accurate physico-chemical characterization of nanoparticles customarily precedes their use in cell biology and in vivo experiments. However, a fact that is easily overlooked is that nanomaterials decorated with organic matter or resuspended in aqueous buffers can be theoretically contaminated by fungal and bacterial microorganisms. While investigating the effects of extensively characterized PEGylated carbon nanotubes (PNTs) on T lymphocyte activation, we demonstrated bacterial contamination of PNTs, which correlated with low reproducibility and artifacts in cell signaling assays. Contamination and artifacts were easily eliminated by preparing the materials in sterile conditions. We propose that simple sterile preparation procedures should be adopted and sterility evaluation of nanoparticles should be customarily performed, prior to assessing nanoparticle intracellular internalization, trafficking and their effects on cells and entire organisms.


Asunto(s)
Portadores de Fármacos , Contaminación de Medicamentos , Nanoconjugados/microbiología , Nanotubos de Carbono/microbiología , Artefactos , Endocitosis/efectos de los fármacos , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos , Interleucina-2/genética , Interleucina-2/metabolismo , Células Jurkat , Microscopía de Fuerza Atómica , Microscopía Confocal , Nanoconjugados/química , Nanotubos de Carbono/química , Tamaño de la Partícula , Polietilenglicoles , Reproducibilidad de los Resultados , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo , Receptores Toll-Like/metabolismo
16.
ACS Omega ; 5(27): 16491-16501, 2020 Jul 14.
Artículo en Inglés | MEDLINE | ID: mdl-32685813

RESUMEN

Titanium oxide (TiO2) nano-/microparticles have been widely used in orthopedic and dental sciences because of their excellent mechanical properties, chemical stability, and ability to promote the osseointegration of implants. However, how the structure and crystallinity of TiO2 particles may affect their osteogenic activity remains elusive. Herein, we evaluated the osteogenic response to submicron amorphous, anatase, and rutile TiO2 particles with controlled size and morphology. First, the ability of TiO2 particles to precipitate apatite was assessed in an acellular medium by using a simulated body fluid (SBF). Three days after the addition to SBF, anatase and rutile TiO2 particles induced the precipitation of aggregates of nanoparticles with a platelike morphology, typical for biomimetic apatite. Conversely, amorphous TiO2 particles induced the precipitation of particles with poor Ca/P atomic ratio only after 14 days of exposure to SBF. Next, the osteogenic response to TiO2 particles was assessed in vitro by incubating MC3T3-E1 preosteoblasts with the particles. The viability and mineralization efficiency of osteoblastic cells were maintained in the presence of all the tested TiO2 particles despite the differences in the induction of apatite precipitation in SBF by TiO2 particles with different structures. Analysis of the particles' surface charge and of the proteins adsorbed onto the particles from the culture media suggested that all the tested TiO2 particles acquired a similar biological identity in the culture media. We posited that this phenomenon attenuated potential differences in osteoblast response to amorphous, anatase, and rutile particles. Our study provides an important insight into the complex relationship between the physicochemical properties and function of TiO2 particles and sheds light on their safe use in medicine.

17.
Bioconjug Chem ; 20(3): 427-31, 2009 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-19243140

RESUMEN

PEGylated-carbon nanotubes (PNTs) were evaluated as nanocarriers of antisense oligonucleotides into T-cells using protein tyrosine phosphatase N22 (PTPN22) as a model target gene. PTPN22 is an important predisposing gene and drug target in type 1 diabetes and several other human autoimmune diseases. Here, we generated the first anti-PTPN22 20-mer antisense oligonucleotides (ASOs) and tethered them to PNTs through a cleavable disulfide bond. Spectroscopic and atomic force microscopy analyses were used to determine the loading of ASO onto PNTs, whereas the cleavable nature of the disulfide bond connecting the oligonucleotide to the nanocarrier was confirmed by incubation with dithiothreitol followed by agarose gel electrophoresis. PNT-conjugated ASOs achieved efficient (>50%) knockdown of PTPN22 expression in T-lymphocytes in culture at the mRNA and protein level, as measured by quantitative real-time PCR and Western blotting, respectively. Considering the high biocompatibility and low in vivo toxicity of PNTs, we expect that our approach will be easily translated to achieve in vivo knockdown of PTPN22 and other T lymphocyte targets, thus enabling novel ASO-mediated immunotherapies for type 1 diabetes and other autoimmune diseases.


Asunto(s)
Nanotubos de Carbono/química , Oligonucleótidos Antisentido/administración & dosificación , Polietilenglicoles/química , Proteína Tirosina Fosfatasa no Receptora Tipo 22/genética , Linfocitos T/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , Células Jurkat , Oligonucleótidos Antisentido/química , Oligonucleótidos Antisentido/genética
18.
ACS Nano ; 13(1): 176-186, 2019 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-30592401

RESUMEN

It remains an unresolved challenge to achieve spatial and temporal monitoring of drug release from nanomedicines (NMs) in vivo, which is of crucial importance in disease treatment. To tackle this issue, we constructed core-satellite ICG/DOX@Gel-CuS NMs, which consist of gelatin (Gel) nanoparticles (NPs) with payloads of near-infrared fluorochrome indocyanine green (ICG) and chemo-drug doxorubicin (DOX) and surrounding CuS NPs. The fluorescence of ICG was initially shielded by satellite CuS NPs within the intact ICG/DOX@Gel-CuS NMs and increased in proportion to the amount of DOX released from NMs in response to enzyme-activated NMs degradation. For more comprehensive understanding of the drug-release profile, a theoretical model derived from computer simulation was employed to reconstruct the enzyme-activatable drug release of the ICG/DOX@Gel-CuS NMs, which demonstrated the underlying kinetics functional relationship between the released DOX amount and recovered ICG fluorescence intensity. The kinetics of drug release in vivo was assessed by administrating ICG/DOX@Gel-CuS NMs both locally and systemically into MDA-MB-231 tumor-bearing mice. Upon accumulation of ICG/DOX@Gel-CuS NMs in the tumor, overexpressed enzymes triggered the degradation of the gelatin scaffold as well as the release of DOX and ICG, which can be visually depicted with the ICG fluorescence signal increasing only in the tumor area by fluorescence imaging. Additionally, the photoacoustic signal from CuS NPs was independent from the physical status of ICG/DOX@Gel-CuS NMs and hence was utilized for real-time NMs tracking. Thus, by taking advantage of the core-satellite architecture and NMs degradability in tumor site, the DOX release profile of ICG/DOX@Gel-CuS NMs was monitored by fluorescence and photoacoustic dual-modal imaging in a real-time noninvasive manner.


Asunto(s)
Antineoplásicos/farmacocinética , Doxorrubicina/farmacocinética , Liberación de Fármacos , Verde de Indocianina/farmacocinética , Nanocápsulas/química , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Doxorrubicina/uso terapéutico , Fluorescencia , Gelatina/química , Humanos , Verde de Indocianina/química , Ratones , Neoplasias Experimentales/diagnóstico por imagen , Neoplasias Experimentales/tratamiento farmacológico , Imagen Óptica , Técnicas Fotoacústicas
19.
J Nanosci Nanotechnol ; 8(5): 2259-69, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18572636

RESUMEN

In this paper we report the fabrication of a multivalent, cell-type specific and cytoplasmic delivery system based on single-walled carbon nanotubes. The latter were functionalized through adsorption of phospholipids terminated by biotinylated PEG chains functionalized with fluorochrome-coupled neutravidin, and subsequently with antibodies (anti-CD3epsilon and anti-CD28) for T cell receptor post-signaling endocytosis and a synthetic fusogenic polymer for disruption of lysosomal compartments. The biomimetic nanoassemblies were composed by PEGylated individual/very small bundles of carbon nanotubes having an average length and a standard deviation of 176 nm and 77 nm, respectively. The nanoassemblies were stably dispersed under physiological conditions, visible by conventional optical and confocal microscopy and specifically targeted to T cells both in vitro and in living animals. The addition of a fusogenic polymer to the nanoassemblies did not affect the cellular uptake and allowed the release into the cytosol of the targeted cells both in vitro and in the animals. The present manuscript is the first report about the cytoplasmic delivery of carbon nanotubes in a specific cell type in intact animals and paves the way for their use as in vivo intracellular delivery systems.


Asunto(s)
Citoplasma/metabolismo , Nanotubos de Carbono , Polietilenglicoles/química , Animales , Endocitosis , Citometría de Flujo , Ratones , Microscopía de Fuerza Atómica , Microscopía Confocal , Espectrofotometría Ultravioleta , Linfocitos T/metabolismo
20.
ACS Appl Mater Interfaces ; 10(48): 41167-41177, 2018 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-30406646

RESUMEN

Suboptimal intratumor accumulation and poorly controllable release of encapsulated drugs remain unresolved challenges hampering further advancement of nanomedicines in cancer therapy. Herein, we conceived near-infrared (NIR) laser-triggered transformable BiS@HSA/DTX multiple nanorods (mNRs), which were made of small bundles of bismuth sulfide nanorods (BiS NRs) coated with docetaxel (DTX)-inlaid human serum albumin (HSA). The BiS@HSA/DTX mNRs had a lateral size of approximately 100 nm and efficiently accumulated in the tumor microenvironment upon systemic administration in tumor-bearing nude mice. NIR laser irradiation of the tumor area caused rapid disassembly of the BiS@HSA/DTX mNRs into individual HSA-coated BiS nanorods (BiS@HSA iNRs) and triggered the release of DTX from the HSA corona, due to the local temperature increase generated by BiS NRs via the photothermal effect. The laser-induced transformation into BiS@HSA iNRs facilitated their penetration and increased the retention time in tumor. The spatiotemporal delivery behavior of the BiS@HSA/DTX mNRs could be monitored by photoacoustic/computed tomography dual-modal imaging in vivo. Furthermore, because of the excellent photothermal conversion properties of BiS NRs and laser-triggered DTX release from BiS@HSA/DTX mNRs, efficient tumor combinatorial therapy was achieved via concurrent hyperthermia and chemotherapy in mice treated with BiS@HSA/DTX mNRs upon NIR laser irradiation.


Asunto(s)
Bismuto , Docetaxel , Hipertermia Inducida , Nanotubos/química , Neoplasias Experimentales , Técnicas Fotoacústicas , Fototerapia , Sulfuros , Tomografía , Animales , Bismuto/química , Bismuto/farmacocinética , Bismuto/farmacología , Línea Celular Tumoral , Docetaxel/química , Docetaxel/farmacocinética , Docetaxel/farmacología , Femenino , Humanos , Rayos Láser , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias Experimentales/diagnóstico por imagen , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Neoplasias Experimentales/terapia , Sulfuros/química , Sulfuros/farmacocinética , Sulfuros/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA