Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Nanotechnol ; 19(5): 705-714, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38366225

RESUMEN

Graphene oxide nanomaterials are being developed for wide-ranging applications but are associated with potential safety concerns for human health. We conducted a double-blind randomized controlled study to determine how the inhalation of graphene oxide nanosheets affects acute pulmonary and cardiovascular function. Small and ultrasmall graphene oxide nanosheets at a concentration of 200 µg m-3 or filtered air were inhaled for 2 h by 14 young healthy volunteers in repeated visits. Overall, graphene oxide nanosheet exposure was well tolerated with no adverse effects. Heart rate, blood pressure, lung function and inflammatory markers were unaffected irrespective of graphene oxide particle size. Highly enriched blood proteomics analysis revealed very few differential plasma proteins and thrombus formation was mildly increased in an ex vivo model of arterial injury. Overall, acute inhalation of highly purified and thin nanometre-sized graphene oxide nanosheets was not associated with overt detrimental effects in healthy humans. These findings demonstrate the feasibility of carefully controlled human exposures at a clinical setting for risk assessment of graphene oxide, and lay the foundations for investigating the effects of other two-dimensional nanomaterials in humans. Clinicaltrials.gov ref: NCT03659864.


Asunto(s)
Grafito , Nanoestructuras , Humanos , Grafito/química , Masculino , Adulto , Femenino , Nanoestructuras/química , Adulto Joven , Método Doble Ciego , Frecuencia Cardíaca/efectos de los fármacos , Administración por Inhalación , Exposición por Inhalación/efectos adversos , Presión Sanguínea/efectos de los fármacos , Tamaño de la Partícula
2.
Nanoscale ; 15(26): 11038-11051, 2023 Jul 06.
Artículo en Inglés | MEDLINE | ID: mdl-37357917

RESUMEN

The spontaneous self-assembly of biomolecules around the surface of nanoparticles (NPs) once exposed to plasma and other biofluids, has been termed the 'biomolecule corona'. While the protein composition of the biomolecule corona has been widely characterised, the interaction of NPs with the plasma lipidome has not been fully investigated. Here, we use targeted and untargeted lipidomics to analyse a wide spectrum of bioactive lipids adsorbed onto the surface of liposome NPs post-incubation with human plasma. Our data indicate that the biomolecule corona contains a diverse mixture of simple and complex lipid species, including sphingolipids such as ceramides and sphingomyelins, glycerolipids, glycerophospholipids, cholesteryl esters, as well as oxylipin and N-acyl ethanolamine derivatives of fatty acids. Although the corona lipidomic profiles reflected the overall composition of the plasma lipidome, monohydroxy- and oxo-fatty acid oxylipins, mono-, di- and tri- acylglycerols, sphingomyelins and ceramides showed a preferential binding for liposome NP surface. Interestingly, the biomolecule corona lipid profiles appeared to mirror those of the lipoprotein lipid cargo, suggesting that lipid species may be carried within the lipoprotein complexes attached to the corona. Proteomic analysis of corona-associated proteins showed the presence of several apolipoproteins (A-I, A-II, A-IV, B, C-I, C-III, C-IV, C2-C4, D, E, L, M and lipoprotein Lp(A)), supporting this notion. Our findings reveal the wide lipid diversity of the biomolecule corona and indicate a potential lipoprotein-mediated adsorption mechanism of lipids onto liposome NPs, highlighting the importance of bridging proteomics with lipidomics to fully comprehend the interactions at the bio-nano interface.


Asunto(s)
Nanopartículas , Corona de Proteínas , Humanos , Liposomas/química , Lipidómica , Esfingomielinas , Proteómica , Lipoproteínas , Nanopartículas/química , Ceramidas , Corona de Proteínas/química
3.
Nat Rev Clin Oncol ; 19(8): 551-561, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35739399

RESUMEN

Over the past decade, the development of 'simple' blood tests that enable cancer screening, diagnosis or monitoring and facilitate the design of personalized therapies without the need for invasive tumour biopsy sampling has been a core ambition in cancer research. Data emerging from ongoing biomarker development efforts indicate that multiple markers, used individually or as part of a multimodal panel, are required to enhance the sensitivity and specificity of assays for early stage cancer detection. The discovery of cancer-associated molecular alterations that are reflected in blood at multiple dimensions (genome, epigenome, transcriptome, proteome and metabolome) and integration of the resultant multi-omics data have the potential to uncover novel biomarkers as well as to further elucidate the underlying molecular pathways. Herein, we review key advances in multi-omics liquid biopsy approaches and introduce the 'nano-omics' paradigm: the development and utilization of nanotechnology tools for the enrichment and subsequent omics analysis of the blood-circulating cancerome.


Asunto(s)
Neoplasias , Proteoma , Biomarcadores/análisis , Genoma , Humanos , Metaboloma , Nanotecnología , Neoplasias/diagnóstico , Neoplasias/genética , Neoplasias/terapia , Transcriptoma
4.
ACS Nano ; 15(4): 7357-7369, 2021 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-33730479

RESUMEN

Blood-circulating biomarkers have the potential to detect Alzheimer's disease (AD) pathology before clinical symptoms emerge and to improve the outcomes of clinical trials for disease-modifying therapies. Despite recent advances in understanding concomitant systemic abnormalities, there are currently no validated or clinically used blood-based biomarkers for AD. The extremely low concentration of neurodegeneration-associated proteins in blood necessitates the development of analytical platforms to address the "signal-to-noise" issue and to allow an in-depth analysis of the plasma proteome. Here, we aimed to discover and longitudinally track alterations of the blood proteome in a transgenic mouse model of AD, using a nanoparticle-based proteomics enrichment approach. We employed blood-circulating, lipid-based nanoparticles to extract, analyze and monitor AD-specific protein signatures and to systemically uncover molecular pathways associated with AD progression. Our data revealed the existence of multiple proteomic signals in blood, indicative of the asymptomatic stages of AD. Comprehensive analysis of the nanoparticle-recovered blood proteome by label-free liquid chromatography-tandem mass spectrometry resulted in the discovery of AD-monitoring signatures that could discriminate the asymptomatic phase from amyloidopathy and cognitive deterioration. While the majority of differentially abundant plasma proteins were found to be upregulated at the initial asymptomatic stages, the abundance of these molecules was significantly reduced as a result of amyloidosis, suggesting a disease-stage-dependent fluctuation of the AD-specific blood proteome. The potential use of the proposed nano-omics approach to uncover information in the blood that is directly associated with brain neurodegeneration was further exemplified by the recovery of focal adhesion cascade proteins. We herein propose the integration of nanotechnology with already existing proteomic analytical tools in order to enrich the identification of blood-circulating signals of neurodegeneration, reinvigorating the potential clinical utility of the blood proteome at predicting the onset and kinetics of the AD progression trajectory.


Asunto(s)
Enfermedad de Alzheimer , Nanopartículas , Enfermedad de Alzheimer/diagnóstico , Animales , Biomarcadores , Proteínas Sanguíneas , Ratones , Proteoma , Proteómica
5.
Adv Healthc Mater ; 10(1): e2001378, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33236524

RESUMEN

Sepsis is one of the leading causes of death worldwide with high mortality rates and a pathological complexity hindering early and accurate diagnosis. Today, laboratory culture tests are the epitome of pathogen recognition in sepsis. However, their consistency remains an issue of controversy with false negative results often observed. Clinically used blood markers, C reactive protein (CRP) and procalcitonin (PCT) are indicators of an acute-phase response and thus lack specificity, offering limited diagnostic efficacy. In addition to poor diagnosis, inefficient drug delivery and the increasing prevalence of antibiotic-resistant microorganisms constitute significant barriers in antibiotic stewardship and impede effective therapy. These challenges have prompted the exploration for alternative strategies that pursue accurate diagnosis and effective treatment. Nanomaterials are examined for both diagnostic and therapeutic purposes in sepsis. The nanoparticle (NP)-enabled capture of sepsis causative agents and/or sepsis biomarkers in biofluids can revolutionize sepsis diagnosis. From the therapeutic point of view, currently existing nanoscale drug delivery systems have proven to be excellent allies in targeted therapy, while many other nanotherapeutic applications are envisioned. Herein, the most relevant applications of nanomedicine for the diagnosis, prognosis, and treatment of sepsis is reviewed, providing a critical assessment of their potentiality for clinical translation.


Asunto(s)
Calcitonina , Sepsis , Antibacterianos/uso terapéutico , Biomarcadores , Proteína C-Reactiva/análisis , Humanos , Sepsis/diagnóstico , Sepsis/tratamiento farmacológico
6.
Nanoscale Horiz ; 5(11): 1476-1486, 2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-32853302

RESUMEN

The spontaneous adsorption of biomolecules onto the surface of nanoparticles (NPs) in complex physiological biofluids has been widely investigated over the last decade. Characterisation of the protein composition of the 'biomolecule corona' has dominated research efforts, whereas other classes of biomolecules, such as nucleic acids, have received no interest. Scarce, speculative statements exist in the literature about the presence of nucleic acids in the biomolecule corona, with no previous studies attempting to describe the contribution of genomic content to the blood-derived NP corona. Herein, we provide the first experimental evidence of the interaction of circulating cell-free DNA (cfDNA) with lipid-based NPs upon their incubation with human plasma samples, obtained from healthy volunteers and ovarian carcinoma patients. Our results also demonstrate an increased amount of detectable cfDNA in patients with cancer. Proteomic analysis of the same biomolecule coronas revealed the presence of histone proteins, suggesting an indirect, nucleosome-mediated NP-cfDNA interaction. The finding of cfDNA as part of the NP corona, offers a previously unreported new scope regarding the chemical composition of the 'biomolecule corona' and opens up new possibilities for the potential exploitation of the biomolecule corona for the enrichment and analysis of blood-circulating nucleic acids.


Asunto(s)
Ácidos Nucleicos Libres de Células/química , Lípidos/química , Nanopartículas/química , Adsorción , Anciano , Anciano de 80 o más Años , Femenino , Histonas , Humanos , Persona de Mediana Edad , Neoplasias Ováricas , Plasma , Proteómica
7.
J Intensive Care Soc ; 21(3): 241-246, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32782464

RESUMEN

The urgent need to start anti-infective therapeutic interventions in suspected sepsis, and the lack of specific time-critical diagnostic information often lead to the widespread administration of broad-spectrum antimicrobial therapies, increasing the risk of unwanted patient harms and contributing to rising pathogen antimicrobial resistance. Nanotechnology, which involves engineering at the nanoscale, allows for the bespoke development of diagnostic solutions with multi-functionality and high sensitivity that has the potential to help provide time-critical information to make more accurate diagnoses and treatment decisions for sepsis. Nanotechnologies also have the potential to improve upon the current strategies used for novel biomarker discovery. Here we describe some of the current limitations to identifying sepsis and explore the potential role for nanotechnology solutions.

8.
Nanoscale ; 12(18): 10240-10253, 2020 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-32356537

RESUMEN

Rapid and accurate diagnosis of sepsis remains clinically challenging. The lack of specific biomarkers that can differentiate sepsis from non-infectious systemic inflammatory diseases often leads to excessive antibiotic treatment. Novel diagnostic tests are urgently needed to rapidly and accurately diagnose sepsis and enable effective treatment. Despite investment in cutting-edge technologies available today, the discovery of disease-specific biomarkers in blood remains extremely difficult. The highly dynamic environment of plasma restricts access to vital diagnostic information that can be obtained by proteomic analysis. Here, we employed clinically used lipid-based nanoparticles (AmBisome®) as an enrichment platform to analyze the human plasma proteome in the setting of sepsis. We exploited the spontaneous interaction of plasma proteins with nanoparticles (NPs) once in contact, called the 'protein corona', to discover previously unknown disease-specific biomarkers for sepsis diagnosis. Plasma samples obtained from non-infectious acute systemic inflammation controls and sepsis patients were incubated ex vivo with AmBisome® liposomes, and the resultant protein coronas were thoroughly characterised and compared by mass spectrometry (MS)-based proteomics. Our results demonstrate that the proposed nanoparticle enrichment technology enabled the discovery of 67 potential biomarker proteins that could reproducibly differentiate non-infectious acute systemic inflammation from sepsis. This study provides proof-of-concept evidence that nanoscale-based 'omics' enrichment technologies have the potential to substantially improve plasma proteomics analysis and to uncover novel biomarkers in a challenging clinical setting.


Asunto(s)
Inflamación/diagnóstico , Corona de Proteínas/química , Proteoma/análisis , Proteómica/métodos , Sepsis/diagnóstico , Biomarcadores/sangre , Cromatografía Líquida de Alta Presión , Humanos , Lípidos/química , Liposomas/química , Nanopartículas/química , Proteoma/metabolismo , Espectrometría de Masas en Tándem
9.
Nanomedicine (Lond) ; 14(24): 3127-3142, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31855120

RESUMEN

Aim: To develop a nonviral tool for the delivery of siRNA to brain tumor cells using peptide nanofibers (PNFs). Materials & methods: Uptake of PNFs was evaluated by confocal microscopy and flow cytometry. Gene silencing was determined by RT-qPCR and cell invasion assay. Results: PNFs enter phagocytic (BV-2) and nonphagocytic (U-87 MG) cells via endocytosis and passive translocation. siPLK1 delivered using PNFs reduced the expression of polo-like kinase 1 mRNA and induced cell death in a panel of immortalized and glioblastoma-derived stem cells. Moreover, targeting MMP2 using PNF:siMMP2 reduced the invasion capacity of U-87 MG cells. We show that stereotactic intra-tumoral administration of PNF:siPLK1 significantly extends the survival of tumor bearing mice comparing with the untreated tumor bearing animals. Conclusion: Our results suggest that this nanomedicine-based RNA interference approach deserves further investigation as a potential brain tumor therapeutic tool.


Asunto(s)
Neoplasias Encefálicas/terapia , Proteínas de Ciclo Celular/fisiología , Movimiento Celular/fisiología , Proliferación Celular/fisiología , Nanofibras/química , Péptidos/química , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas Proto-Oncogénicas/fisiología , Animales , Neoplasias Encefálicas/metabolismo , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Citometría de Flujo , Terapia Genética/métodos , Humanos , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 2 de la Matriz/fisiología , Ratones , Ratones Desnudos , Microscopía Confocal , Nanomedicina/métodos , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas/genética , Quinasa Tipo Polo 1
10.
Adv Mater ; 31(4): e1803335, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30488990

RESUMEN

The self-assembled layered adsorption of proteins onto nanoparticle (NP) surfaces, once in contact with biological fluids, is termed the "protein corona" and it is gradually seen as a determinant factor for the overall biological behavior of NPs. Here, the previously unreported in vivo protein corona formed in human systemic circulation is described. The human-derived protein corona formed onto PEGylated doxorubicin-encapsulated liposomes (Caelyx) is thoroughly characterized following the recovery of liposomes from the blood circulation of ovarian carcinoma patients. In agreement with previous investigations in mice, the in vivo corona is found to be molecularly richer in comparison to its counterpart ex vivo corona. The intravenously infused liposomes are able to scavenge the blood pool and surface-capture low-molecular-weight, low-abundance plasma proteins that cannot be detected by conventional plasma proteomic analysis. This study describes the previously elusive or postulated formation of protein corona around nanoparticles in vivo in humans and illustrates that it can potentially be used as a novel tool to analyze the blood circulation proteome.


Asunto(s)
Liposomas/química , Polietilenglicoles/química , Corona de Proteínas/química , Adsorción , Doxorrubicina/química , Humanos , Nanopartículas/química
11.
Biomaterials ; 188: 118-129, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30343255

RESUMEN

The prominent discrepancy between the significant investment towards plasma biomarker discovery and the very low number of biomarkers currently in clinical use stresses the need for discovery technologies. The discovery of protein biomarkers present in human blood by proteomics is tremendously challenging, owing to the large dynamic concentration range of blood proteins. Here, we describe the use of blood-circulating lipid-based nanoparticles (NPs) as a scavenging tool to comprehensively analyse the blood proteome. We aimed to exploit the spontaneous interaction of NPs with plasma proteins once injected in the bloodstream, known as 'protein corona', in order to facilitate the capture of tumor-specific molecules. We employed two different tumor models, a subcutaneous melanoma model (B16-F10) and human lung carcinoma xenograft model (A549) and comprehensively compared by mass spectrometry the in vivo protein coronas formed onto clinically used liposomes, intravenously administered in healthy and tumor-bearing mice. The results obtained demonstrated that blood-circulating liposomes surface-capture and amplify a wide range of different proteins including low molecular weight (MW) and low abundant tumor specific proteins (intracellular products of tissue leakage) that could not be detected by plasma analysis, performed in comparison. Most strikingly, the NP (liposomal) corona formed in the xenograft model was found to consist of murine host response proteins, as well as human proteins released from the inoculated and growing human cancer cells. This study offers direct evidence that the in vivo NP protein corona could be deemed as a valuable tool to enrich the blood proteomic analysis and to allow the discovery of potential biomarkers in experimental disease models.


Asunto(s)
Biomarcadores de Tumor/sangre , Proteínas Sanguíneas/análisis , Liposomas/metabolismo , Neoplasias Pulmonares/sangre , Melanoma Experimental/sangre , Corona de Proteínas/análisis , Células A549 , Animales , Biomarcadores de Tumor/metabolismo , Proteínas Sanguíneas/metabolismo , Femenino , Humanos , Liposomas/sangre , Neoplasias Pulmonares/metabolismo , Melanoma Experimental/metabolismo , Ratones Endogámicos C57BL , Nanopartículas/metabolismo , Corona de Proteínas/metabolismo
12.
J Control Release ; 276: 157-167, 2018 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-29522832

RESUMEN

Thermally triggered drug release from temperature-sensitive liposomes (TSL) holds great promise for cancer therapy. Different types of TSL have been designed recently for heat triggered drug release inside tumor blood vessels or after accumulation into the tumor interstitium. However, justification of drug release profiles is for far mainly based on in vitro release data. While these methods could be good enough to give early indication about the thermal sensitivity of TSL, they are still far from being optimum. This is because these methods do not take into consideration the actual adsorption of proteins onto the surface of TSL after their in vivo administration, also known as "protein corona" and the influence this could have on drug release. Therefore, in this study we compared thermal triggered drug release profile of two different types of doxorubicin encapsulated TSL; namely the lysolipid-containing TSL (LTSL) and traditional TSL (TTSL) after their in vivo recovery from the blood circulation of CD-1 mice. Ex vivo release profile at 42 °C was then tested either in the presence of full plasma or after removal of unbound plasma proteins (i.e. protein corona coated TSL). Our data showed that the influence of the environment on drug release profile was very much dependent on the type of TSL. LTSL release profile was consistently characterized by ultrafast drug release independent on the conditions tested. On the contrary, TTSL release profile changed significantly. Doxorubicin release from in vivo recovered TTSL was slow and incomplete in the presence of unbound plasma proteins, whereas very rapid drug release was detected from in vivo recovered and purified protein corona-coated TTSL in the absence of unbound proteins. Using mass spectrometry and quantification of protein adsorption, we confirmed that this discrepancy is due to the changes in protein adsorption onto TTSL when heated in the presence of unbound proteins leading to reduction in drug release. In summary this study showed that the formation of the in vivo corona on TSL will have a dramatic impact on their release profile and is dependent on both their lipid composition and the protein content of the environment in which drug release is triggered.


Asunto(s)
Antibióticos Antineoplásicos/química , Doxorrubicina/química , Corona de Proteínas , Antibióticos Antineoplásicos/administración & dosificación , Proteínas Sanguíneas/química , Doxorrubicina/administración & dosificación , Liberación de Fármacos , Liposomas , Temperatura
13.
Nanoscale ; 10(3): 1256-1264, 2018 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-29292433

RESUMEN

The efficacy of drug delivery and other nanomedicine-related therapies largely relies on the ability of nanoparticles to reach the target organ. However, when nanoparticles are injected into the bloodstream, their surface is instantly modified upon interaction with blood components, principally with proteins. It is well known that a dynamic and multi-layered protein structure is formed spontaneously on the nanoparticle upon contact with physiological media, which has been termed protein corona. Although several determinant factors involved in protein corona formation have been identified from in vitro studies, specific relationships between the nanomaterial synthetic identity and its ensuing biological identity under realistic in vivo conditions remain elusive. We present here a detailed study of in vivo protein corona formation after blood circulation of anisotropic gold nanoparticles (nanorods and nanostars). Plasmonic gold nanoparticles of different shapes and sizes were coated with polyethyleneglycol, intravenously administered in CD-1 mice and subsequently recovered. The results from gel electrophoresis and mass spectrometry analysis revealed the formation of complex protein coronas, as early as 10 minutes post-injection. The total amount of protein adsorbed onto the particle surface and the protein corona composition were found to be affected by both the particle size and shape.


Asunto(s)
Oro , Nanopartículas del Metal , Corona de Proteínas , Animales , Ratones , Tamaño de la Partícula , Polietilenglicoles
15.
Nanoscale ; 8(13): 6948-57, 2016 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-26961355

RESUMEN

Nanoparticles (NPs) are instantly modified once injected in the bloodstream because of their interaction with the blood components. The spontaneous coating of NPs by proteins, once in contact with biological fluids, has been termed the 'protein corona' and it is considered to be a determinant factor for the pharmacological, toxicological and therapeutic profile of NPs. Protein exposure time is thought to greatly influence the composition of protein corona, however the dynamics of protein interactions under realistic, in vivo conditions remain unexplored. The aim of this study was to quantitatively and qualitatively investigate the time evolution of in vivo protein corona, formed onto blood circulating, clinically used, PEGylated liposomal doxorubicin. Protein adsorption profiles were determined 10 min, 1 h and 3 h post-injection of liposomes into CD-1 mice. The results demonstrated that a complex protein corona was formed as early as 10 min post-injection. Even though the total amount of protein adsorbed did not significantly change over time, the fluctuation of protein abundances observed indicated highly dynamic protein binding kinetics.


Asunto(s)
Circulación Sanguínea , Doxorrubicina/análogos & derivados , Nanopartículas/metabolismo , Corona de Proteínas/metabolismo , Adsorción , Animales , Disponibilidad Biológica , Doxorrubicina/sangre , Doxorrubicina/farmacocinética , Femenino , Ratones , Nanopartículas/química , Polietilenglicoles/farmacocinética , Corona de Proteínas/química , Factores de Tiempo , Distribución Tisular
16.
ACS Nano ; 9(8): 8142-56, 2015 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-26135229

RESUMEN

The adsorption of proteins and their layering onto nanoparticle surfaces has been called the "protein corona". This dynamic process of protein adsorption has been extensively studied following in vitro incubation of many different nanoparticles with plasma proteins. However, the formation of protein corona under dynamic, in vivo conditions remains largely unexplored. Extrapolation of in vitro formed protein coronas to predict the fate and possible toxicological burden from nanoparticles in vivo is of great interest. However, complete lack of such direct comparisons for clinically used nanoparticles makes the study of in vitro and in vivo formed protein coronas of great importance. Our aim was to study the in vivo protein corona formed onto intravenously injected, clinically used liposomes, based on the composition of the PEGylated liposomal formulation that constitutes the anticancer agent Doxil. The formation of in vivo protein corona was determined after the recovery of the liposomes from the blood circulation of CD-1 mice 10 min postinjection. In comparison, in vitro protein corona was formed by the incubation of liposomes in CD-1 mouse plasma. In vivo and in vitro formed protein coronas were compared in terms of morphology, composition and cellular internalization. The protein coronas on bare (non-PEGylated) and monoclonal antibody (IgG) targeted liposomes of the same lipid composition were also comparatively investigated. A network of linear fibrillary structures constituted the in vitro formed protein corona, whereas the in vivo corona had a different morphology but did not appear to coat the liposome surface entirely. Even though the total amount of protein attached on circulating liposomes correlated with that observed from in vitro incubations, the variety of molecular species in the in vivo corona were considerably wider. Both in vitro and in vivo formed protein coronas were found to significantly reduce receptor binding and cellular internalization of antibody-conjugated liposomes; however, the in vivo corona formation did not lead to complete ablation of their targeting capability.


Asunto(s)
Proteínas Sanguíneas/química , Inmunoglobulina G/química , Membrana Dobles de Lípidos/química , Liposomas/química , Nanopartículas/química , Adsorción , Animales , Antibióticos Antineoplásicos/química , Colesterol/química , Doxorrubicina/análogos & derivados , Doxorrubicina/química , Humanos , Inmunoconjugados/química , Inyecciones Intravenosas , Liposomas/ultraestructura , Células MCF-7 , Ratones , Nanopartículas/ultraestructura , Fosfatidilcolinas/química , Fosfatidiletanolaminas/química , Polietilenglicoles/química
17.
ACS Nano ; 9(2): 1137-49, 2015 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-25574683

RESUMEN

Peptide nanofibers (PNFs) are one-dimensional assemblies of amphiphilic peptides in a cylindrical geometry. We postulated that peptide nanofibers (PNFs) can provide the tools for genetic intervention and be used for delivery of siRNA, as they can be engineered with positively charged amino acids that can electrostatically bind siRNA. The aim of this work was to investigate the use of PNFs as vectors for siRNA delivery providing effective gene knockdown. We designed a surfactant-like peptide (palmitoyl-GGGAAAKRK) able to self-assemble into PNFs and demonstrated that complexes of PNF:siRNA are uptaken intracellularly and increase the residence time of siRNA in the brain after intracranial administration. The biological activity of the complexes was investigated in vitro by analyzing the down-regulation of the expression of a targeted protein (BCL2), as well as induction of apoptosis, as well as in vivo by analyzing the relative gene expression upon stereotactic administration into a deep rat brain structure (the subthalamic nucleus). Gene expression levels of BCL2 mRNA showed that PNF:siBCL2 constructs were able to silence the target BCL2 in specific loci of the brain. Silencing of the BCL2 gene resulted in ablation of neuronal cell populations, indicating that genetic interventions by PNF:siRNA complexes may lead to novel treatment strategies of CNS pathologies.


Asunto(s)
Encéfalo/metabolismo , Silenciador del Gen , Nanofibras/química , Péptidos/química , ARN Interferente Pequeño/química , ARN Interferente Pequeño/genética , Técnicas Estereotáxicas , Secuencia de Aminoácidos , Animales , Apoptosis/genética , Transporte Biológico , Encéfalo/cirugía , Línea Celular Tumoral , Portadores de Fármacos/química , Portadores de Fármacos/metabolismo , Humanos , Masculino , Péptidos/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/deficiencia , Proteínas Proto-Oncogénicas c-bcl-2/genética , Ratas , Ratas Sprague-Dawley
18.
J Liposome Res ; 23(2): 94-100, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23294394

RESUMEN

Dimethoxycurcumin (DMC) is a lipophilic analog of curcumin found in Curcuma longa Linn., which is known to possess significant activity against various cancer cell lines. The purpose of this study was to develop suitable liposomal formulations in order to overcome DMC's poor water solubility and to study the aggregation kinetic profile using the fractal analysis. DMC was incorporated into liposomal formulations composed of DPPC, DPPC:DPPG:chol (9:1:1 molar ratio) and DPPC:DODAP:chol (9:1:1 molar ratio) liposomes. Light scattering techniques were used to elucidate the physicochemical parameters of the liposomal formulations with and without DMC. The structural characteristics of the incorporated molecule were found to be crucial and promote the aggregation mechanism depending also on the liposomes' composition. The results of our study contribute to the overall scientific efforts to prepare efficient carriers for DMC and could be a useful tool in order to study more efficiently the kinetics of the aggregation process of the liposomal carriers.


Asunto(s)
Curcumina/análogos & derivados , Fractales , Liposomas , Curcumina/química , Cinética , Dispersión de Radiación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...