Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 99
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Int J Pharm ; 657: 124131, 2024 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-38643811

RESUMEN

Immunization is a straightforward concept but remains for some pathogens like HIV-1 a challenge. Thus, new approaches towards increasing the efficacy of vaccines are required to turn the tide. There is increasing evidence that antigen exposure over several days to weeks induces a much stronger and more sustained immune response compared to traditional bolus injection, which usually leads to antigen elimination from the body within a couple of days. Therefore, we developed a poly(ethylene) glycol (PEG) hydrogel platform to investigate the principal feasibility of a sustained release of antigens to mimic natural infection kinetics. Eight-and four-armed PEG macromonomers of different MWs (10, 20, and 40 kDa) were end-group functionalized to allow for hydrogel formation via covalent cross-linking. An HIV-1 envelope (Env) antigen in its trimeric (Envtri) or monomeric (Envmono) form was applied. The soluble Env antigen was compared to a formulation of Env attached to silica nanoparticles (Env-SiNPs). The latter are known to have a higher immunogenicity compared to their soluble counterparts. Hydrogels were tunable regarding the rheological behavior allowing for different degradation times and release timeframes of Env-SiNPs over two to up to 50 days. Affinity measurements of the VCR01 antibody which specifically recognizes the CD4 binding site of Env, revealed that neither the integrity nor the functionality of Envmono-SiNPs (Kd = 2.1 ± 0.9 nM) and Envtri-SiNPs (Kd = 1.5 ± 1.3 nM), respectively, were impaired after release from the hydrogel (Kd before release: 2.1 ± 0.1 and 7.8 ± 5.3 nM, respectively). Finally, soluble Env and Env-SiNPs which are two physico-chemically distinct compounds, were co-delivered and shown to be sequentially released from one hydrogel which could be beneficial in terms of heterologous immunization or single dose vaccination. In summary, this study presents a tunable, versatile applicable, and effective delivery platform that could improve vaccination effectiveness also for other infectious diseases than HIV-1.

2.
Eur J Pharm Biopharm ; 198: 114240, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38437906

RESUMEN

The specific delivery of a drug to its site of action also known as targeted drug delivery is a topic in the field of pharmaceutics studied for decades. One approach extensively investigated in this context is the use ligand functionalized nanoparticles. These particles are modified to carry receptor specific ligands, enabling them to accumulate at a desired target site. However, while this concept initially appears straightforward to implement, in-depth research has revealed several challenges hindering target site specific particle accumulation - some of which remain unresolved to this day. One of these challenges consists in the still incomplete understanding of how nanoparticles interact with biological systems. This knowledge gap significantly compromises the predictability of particle distribution in biological systems, which is critical for therapeutic efficacy. One of the most crucial steps in delivery is the attachment of nanoparticles to cells at the target site. This attachment occurs via the formation of multiple ligand receptor bonds. A process also referred to as multivalent interaction. While multivalency has been described extensively for individual molecules and macromolecules respectively, little is known on the multivalent binding of nanoparticles to cells. Here, we will specifically introduce the concept of avidity as a measure for favorable particle membrane interactions. Also, an overview about nanoparticle and membrane properties affecting avidity will be given. Thereafter, we provide a thorough review on literature investigating the correlation between nanoparticle avidity and success in targeted particle delivery. In particular, we want to analyze the currently uncertain data on the existence and nature of the correlation between particle avidity and biodistribution.


Asunto(s)
Nanopartículas , Distribución Tisular , Ligandos , Incertidumbre , Nanopartículas/química , Sistemas de Liberación de Medicamentos
3.
Int J Pharm ; 647: 123453, 2023 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-37783283

RESUMEN

Pharmacotherapy is often limited by undesired side effects while insufficient drug reaches the site of action. Active-targeted nanotherapy should provide a solution for this problem, by using ligands in the nanoparticle corona for the identification of receptors on the target-cell surface. However, since receptor binding is directly associated with pharmacological responses, today's targeting concepts must be critically evaluated. We hypothesized that addressing ectoenzymes would help to overcome this problem, but it was not clear if particles would show sufficiently high avidity to provide us with a viable alternative to classical ligand-receptor concepts. We scrutinized this aspect by immobilizing the highly selective angiotensin-converting enzyme 2 (ACE2) inhibitor MLN-4760 in the corona of block-copolymer nanoparticles and investigated enzyme binding via microscale thermophoresis and flow cytometry. Excellent avidities with Kd values as low as 243 pM for soluble ACE2 and 306 pM for ACE2-positive cells were obtained. In addition, the inhibitory activity had an IC50 value of 2.88 nM. Reliable target cell identification could be proven in coculture experiments. High avidity is the basis for minimizing material loss to off-target sites and paves the way for a paradigm shift in nanoparticle targeting which does not trigger unintended side effects following target cell identification.


Asunto(s)
Enzima Convertidora de Angiotensina 2 , Nanopartículas , Polímeros/química , Nanopartículas/química , Ligandos , Unión Proteica
4.
J Control Release ; 362: 325-341, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37598888

RESUMEN

Interferon-γ (IFN-γ) is well known to reduce the infectivity of viral pathogens by altering their tissue tropism. This effect is induced by upregulation of cholesterol 25-hydroxylase (CH25H). Given the similarity of viral pathogens and ligand-functionalized nanoparticles in the underlying strategy of receptor-mediated cell recognition, it appears conceivable that IFN-γ exceeds similar effects on nanoparticles. Concretely, IFN-γ-induced activation of CH25H could decrease nanoparticle avidity for target cells via depletion of clathrin-coated pits. We hypothesized that this effect would cause deterioration of target-cell specific accumulation of nanoparticles. To prove our hypothesis, we investigated the cell tropism of angiotensin II functionalized nanoparticles (NPLys-Ang II) in a co-culture system of angiotensin II subtype 1 receptor (AT1R) positive rat mesangial target cells (rMCs) and AT1R-negative HeLa off-target cells. In the presence of IFN-γ we observed an up to 5-fold loss of target cell preference for NPLys-Ang II. Thus, our in vitro results suggest a strong influence of IFN-γ on nanoparticle distribution, which is relevant in the context of nanotherapeutic approaches to cancer treatment, as IFN-γ is strongly expressed in tumors. For the target cell tropism of viruses, our results provide a conclusive hypothesis for the underlying mechanism behind non-directed viral distribution in the presence of IFN-γ.

5.
ACS Appl Bio Mater ; 6(6): 2111-2121, 2023 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-37145591

RESUMEN

Atherosclerosis is one of the most urgent global health subjects, causes millions of deaths worldwide, and is associated with enormous healthcare costs. Macrophages are the root cause for inflammatory onset and progression of the disease but are not addressed by conventional therapy. Therefore, we used pioglitazone, which is a drug initially used for diabetes therapies, but at the same time has great potential regarding the mitigation of inflammation. As yet, this potential of pioglitazone cannot be exploited, as drug concentrations at the target site in vivo are not sufficient. To overcome this shortcoming, we established PEG-PLA/PLGA-based nanoparticles loaded with pioglitazone and tested them in vitro. Encapsulation of the drug was analyzed by HPLC and revealed an outstanding encapsulation efficiency of 59% into the nanoparticles, which were 85 nm in size and had a PDI of 0.17. Further, uptake of our loaded nanoparticles in THP-1 macrophages was comparable to the uptake of unloaded nanoparticles. On the mRNA level, pioglitazone-loaded nanoparticles were superior to the free drug by 32% in increasing the expression of the targeted receptor PPAR-γ. Thereby the inflammatory response in macrophages was ameliorated. In this study, we take the first step toward an anti-inflammatory, causal antiatherosclerotic therapy, using the potential of the already established drug pioglitazone, and enable it to enrich at the target site by using nanoparticles. An additional crucial feature of our nanoparticle platform is the versatile modifiability of ligands and ligand density, to achieve an optimal active targeting effect in the future.


Asunto(s)
Aterosclerosis , Nanopartículas , Humanos , Pioglitazona/farmacología , Pioglitazona/uso terapéutico , Polímeros/farmacología , Aterosclerosis/tratamiento farmacológico , Aterosclerosis/genética , Aterosclerosis/metabolismo , Macrófagos
6.
Drug Deliv Transl Res ; 13(11): 2807-2818, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37208562

RESUMEN

Inflammation and immune system activation are key pathologic events in the onset and escalation of diabetic retinopathy (DR). Both are driven by cytokines and complement originating from the retinal pigment epithelium (RPE). Despite the RPE's pivotal role, there is no therapeutic tool to specifically interfere with the RPE-related pathomechanism. A therapy that addresses RPE cells and counteracts inflammation and immune response would be of paramount value for the early treatment of DR, where currently are no specific therapies available. Here, we utilized lipoprotein-mimetic lipid nanocapsules to deliver the anti-inflammatory and immunosuppressive drug cyclosporin A (CsA) to RPE cells. Using a mouse model of DR that mirrors all pathologic aspects of human DR, we demonstrate that intravenously applied CsA-loaded lipid nanocapsules comprehensively counteract inflammation and immune system activation. One single injection suppressed the expression of pro-inflammatory cytokines, dampened macrophage infiltration, and prevented macrophage and microglia activation in eyes with DR. This work shows that CsA-loaded lipid nanocapsules can offer new avenues for the treatment of DR.


Asunto(s)
Diabetes Mellitus , Retinopatía Diabética , Nanocápsulas , Animales , Humanos , Retinopatía Diabética/tratamiento farmacológico , Ciclosporina/uso terapéutico , Nanocápsulas/uso terapéutico , Inyecciones Intravenosas , Inflamación/tratamiento farmacológico , Modelos Animales de Enfermedad , Citocinas , Sistema Inmunológico/metabolismo , Sistema Inmunológico/patología , Lípidos , Diabetes Mellitus/tratamiento farmacológico
7.
Mol Pharm ; 20(5): 2465-2476, 2023 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-36961431

RESUMEN

Water-free preparation of protein delivery systems has the potential to overcome the limitations of hydrogel depot systems such as off-target reactions, functional group hydrolysis, and limited loading capacity. However, a major roadblock in the development and use of these systems is administration as implantation is often required. In this study, we developed a biodegradable and water-free injectable protein delivery system via inverse electron demand Diels-Alder reaction between norbornene- and tetrazine-functionalized four-armed poly(ethylene glycol) macromonomers. 1:1 mixtures of these precursors gelled rapidly in situ, taking less than 11 s to reach their gelation point. Methyl substitution of tetrazine slowed the gelation time and increased the cross-linking density, whereas oxygen incorporation into norbornene changed the mechanical properties. Introduction of hydrolytically cleavable groups enabled biodegradability. Using phenyl carbamate and phenyl carbonate ester groups, we could tune the stability. Controlled release of the protein surrogate glucose oxidase was achieved over a period of 500 days. The novel preparation method presented here is a promising step toward the development of water-free injectable protein depots for controlled drug delivery.


Asunto(s)
Polietilenglicoles , Polímeros , Preparaciones de Acción Retardada , Hidrogeles , Sistemas de Liberación de Medicamentos , Proteínas
8.
Nanoscale Horiz ; 8(2): 256-269, 2023 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-36594629

RESUMEN

The paramount relevance of clathrin-coated pits (CCPs) to receptor-mediated endocytosis of nanoparticles, extracellular vesicles, and viruses has made them the focus of many studies; however, the role of CCP geometry in the ligand-receptor interactions between multivalent nanoparticles and cells has not been investigated. We hypothesized the general dependence of nanoparticle binding energy on local membrane curvature to be expandable to the specific case of ligand-functionalized nanoparticles binding cell membranes, in the sense that membrane structures whose curvature matches that of the particle (e.g., CCPs) signficantly contribute to binding avidity. We investigated this hypothesis with nanoparticles that bind multivalently to angiotensin II receptor type 1, which is subject to clathrin-mediated endocytosis. When we used cholesterol extraction to prevent the action of CCPs, we found a 67 to 100-fold loss in avidity. We created a theoretical model that predicts this decrease based on the loss of ligand-receptor interactions when CCPs, which perfectly match nanoparticle geometry, are absent. Our findings shed new light on how cells "see" nanoparticles. The presence or absence of CPPs is so influential on how cells interact with nanoparticles that the number of particles required to be visible to cells changes by two orders of magnitude depending on CCP presence.


Asunto(s)
Clatrina , Nanopartículas , Clatrina/metabolismo , Ligandos , Membrana Celular/metabolismo , Endocitosis
9.
Sci Adv ; 8(38): eabo6638, 2022 09 23.
Artículo en Inglés | MEDLINE | ID: mdl-36149956

RESUMEN

Retinopathy of prematurity (ROP) is a retinal disease that threatens the vision of prematurely born infants. Severe visual impairment up to complete blindness is caused by neovascularization and inflammation, progressively destroying the immature retina. ROP primarily affects newborns in middle- and low-income countries with limited access to current standard treatments such as intraocular drug injections and laser- or cryotherapy. To overcome these limitations, we developed a nanotherapeutic that effectively prevents ROP development with one simple intravenous injection. Its lipid nanocapsules transport the antiangiogenic and anti-inflammatory cyclosporin A efficiently into disease-driving retinal pigment epithelium cells. In a mouse model of ROP, a single intravenous injection of the nanotherapeutic prevented ROP and led to normal retinal development by counteracting neovascularization and inflammation. This nanotherapeutic approach has the potential to bring about a change of paradigm in ROP therapy and prevent millions of preterm born infants from developing ROP.


Asunto(s)
Nanocápsulas , Retinopatía de la Prematuridad , Animales , Ciclosporina/farmacología , Ciclosporina/uso terapéutico , Humanos , Recién Nacido , Inflamación/tratamiento farmacológico , Inyecciones Intravenosas , Lípidos , Ratones , Nanocápsulas/uso terapéutico , Retinopatía de la Prematuridad/tratamiento farmacológico , Retinopatía de la Prematuridad/prevención & control , Factor A de Crecimiento Endotelial Vascular
10.
Macromol Biosci ; 22(12): e2200226, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36112280

RESUMEN

Eight-armed poly(ethylene glycol) (PEG) hydrogels cross-linked via inverse electron demand Diels-Alder reaction between norbornene and tetrazine groups are promising materials for long-term protein delivery. While a controlled release over 265 days is achieved for 15% w/v hydrogels in the previous study, the material shows high stability over 500 days despite having cleavable ester linkages between the PEG macromonomers and their functionalities. In this study, the hydrolyzable ester linkers in the PEG-norbornene precursor structure are exchanged to reduce the degradation time. To this end, 3,6-epoxy-1,2,3,6-tetrahydrophthalimide, phenyl carbamate, carbonate ester, and phenyl carbonate ester are introduced as degradable functional groups. Oscillatory shear experiments reveal that they are not affected the in situ gelation. All hydrogel types have gel points of less than 20 s even at a low polymer concentration of 5% w/v. Hydrogels with varying polymer concentrations have similar mesh sizes, all of which fell in the range of 4-12 nm. The inclusion of phenyl carbonate ester accelerates degradation considerably, with complete dissolution of 15% w/v hydrogels after 302 days of incubation in phosphate buffer (pH 7.4). Controlled release of 150 kDa fluorescein isothiocyanate-dextran over a period of at least 150 days is achieved with 15% w/v hydrogels.


Asunto(s)
Electrones , Hidrogeles , Reacción de Cicloadición , Preparaciones de Acción Retardada/química , Hidrogeles/química , Polietilenglicoles/química , Norbornanos , Materiales Biocompatibles , Polímeros , Ésteres
11.
Drug Discov Today ; 27(9): 2497-2509, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35654389

RESUMEN

The retinal pigment epithelium (RPE) plays a crucial part in sight-threatening diseases. In this review, we shed light on the pivotal implication of the RPE in age-related macular degeneration, diabetic retinopathy and retinopathy of prematurity; and explain why a paradigm shift toward targeted RPE therapy is needed to efficiently fight these retinal diseases. We provide guidance for the development of RPE-specific nanotherapeutics by giving a comprehensive overview of the possibilities and challenges of drug delivery to the RPE and highlight successful nanotherapeutic approaches targeting the RPE.


Asunto(s)
Retinopatía Diabética , Degeneración Macular , Humanos , Recién Nacido , Epitelio Pigmentado de la Retina
12.
Nanomedicine ; 42: 102545, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35283290

RESUMEN

Nanoparticles hold great potential as vaccine carriers due to their highly versatile structure and the possibility to influence intracellular trafficking and antigen presentation by their design. In this study, we developed a nanoparticulate system with a new enzyme-triggered antigen release mechanism. For this novel approach, nanoparticle and model antigen ovalbumin were linked with a substrate of the early endosomal protease cathepsin S. This construct enabled the transfer of antigens delivered to bone marrow-derived dendritic cells from the endo-lysosomal compartments in the cytosol. Consecutively, our particles enhanced cross-presentation on dendritic cells and subsequently promoted a stronger activation of CD8+ T cells. Our findings suggest that enzyme-triggered antigen release allows the endosomal escape of the antigen, leading to increased MHC-I presentation. Since T cell immunity is central for the control of viral infections and cancer, this release mechanism offers a promising approach for the development of both prophylactic and therapeutic vaccines.


Asunto(s)
Reactividad Cruzada , Vacunas , Animales , Presentación de Antígeno , Antígenos , Linfocitos T CD8-positivos , Células Dendríticas , Ratones , Ratones Endogámicos C57BL , Ovalbúmina/química
13.
Biomacromolecules ; 22(8): 3223-3236, 2021 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-34270216

RESUMEN

Off-target interactions between reactive hydrogel moieties and drug cargo as well as slow reaction kinetics and the absence of controlled protein release over an extended period of time are major drawbacks of chemically cross-linked hydrogels for biomedical applications. In this study, the inverse electron demand Diels-Alder (iEDDA) reaction between norbornene- and tetrazine-functionalized eight-armed poly(ethylene glycol) (PEG) macromonomers was used to overcome these obstacles. Oscillatory shear experiments revealed that the gel point of a 15% (w/v) eight-armed PEG hydrogel with a molecular weight of 10 kDa was less than 15 s, suggesting the potential for fast in situ gelation. However, the high-speed reaction kinetics result in a risk of premature gel formation that complicates the injection process. Therefore, we investigated the effect of polymer concentration, temperature, and chemical structure on the gelation time. The cross-linking reaction was further characterized regarding bioorthogonality. Only 11% of the model protein lysozyme was found to be PEGylated by the iEDDA reaction, whereas 51% interacted with the classical Diels-Alder reaction. After determination of the mesh size, fluorescein isothiocyanate-dextran was used to examine the release behavior of the hydrogels. When glucose oxidase was embedded into 15% (w/v) hydrogels, a controlled release over more than 250 days was achieved. Overall, the PEG-based hydrogels cross-linked via the fast iEDDA reaction represent a promising material for the long-term administration of biologics.


Asunto(s)
Electrones , Hidrogeles , Peso Molecular , Polietilenglicoles , Proteínas
14.
Eur J Pharm Biopharm ; 166: 44-60, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34087354

RESUMEN

The development of nanomedical devices has led to a considerable number of clinically applied nanotherapeutics. Yet, the overall poor translation of nanoparticular concepts into marketable systems has not met the initial expectations and led to increasing criticism in recent years. Most novel nano approaches thereby use highly refined formulations including a plethora of active targeting sequences, but ultimately fail to reach their target due to a generally high off-target deposition in organs such as the liver or kidney. In this context, we argue that initial nanoparticle (NP) development should not entirely become set on conventional formulation aspects. In contrast, we propose a change of focus towards a prior analysis of general sites of NP in vivo deposition and an assessment of how accumulation in these organs or tissues can be harnessed to develop therapies for site-related pathologies. We therefore give a comprehensive overview of existing nanotherapeutic targeting strategies for specific cell types within three of the usual suspects, i.e. the liver, kidney and the vascular system. We discuss the physiological surroundings and relevant pathologies of described tissues as well as the implications for NP-mediated drug delivery. Additionally, successful cell-selective NP concepts using active targeting strategies are assessed. By bringing together both (patho)physiological aspects and concepts for cell-selective NP formulations, we hope to show a novel opportunity for the development of more promising nanotherapeutic devices.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Nanomedicina , Nanopartículas , Disponibilidad Biológica , Vasos Sanguíneos/citología , Vasos Sanguíneos/metabolismo , Humanos , Riñón/citología , Riñón/metabolismo , Hígado/citología , Hígado/metabolismo , Nanomedicina/métodos , Nanomedicina/tendencias , Nanopartículas/administración & dosificación , Nanopartículas/metabolismo , Distribución Tisular
15.
J Control Release ; 333: 536-559, 2021 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-33794270

RESUMEN

Atherosclerosis is the leading cause of death in developed countries. The pathogenetic mechanism relies on a macrophage-based immune reaction to low density lipoprotein (LDL) deposition in blood vessels with dysfunctional endothelia. Thus, atherosclerosis is defined as a chronic inflammatory disease. A plethora of cardiovascular drugs have been developed and are on the market, but the major shortcoming of standard medications is that they do not address the root cause of the disease. Statins and thiazolidinediones that have recently been recognized to exert specific anti-atherosclerotic effects represent a potential breakthrough on the horizon. But their whole potential cannot be realized due to insufficient availability at the pathological site and severe off-target effects. The focus of this review will be to elaborate how both groups of drugs could immensely profit from nanoparticulate carriers. This delivery principle would allow for their accumulation in target macrophages and endothelial cells of the atherosclerotic plaque, increasing bioavailability where it is needed most. Based on the analyzed literature we conclude design criteria for the delivery of statins and thiazolidinediones with nanoparticles for anti-atherosclerotic therapy. Nanoparticles need to be below a diameter of 100 nm to accumulate in the atherosclerotic plaque and should be fabricated using biodegradable materials. Further, the thiazolidinediones or statins must be encapsulated into the particle core, because especially for thiazolidindiones the uptake into cells is prerequisite for their mechanism of action. For optimal uptake into targeted macrophages and endothelial cells, the ideal particle should present ligands on its surface which bind specifically to scavenger receptors. The impact of statins on the lectin-type oxidized LDL receptor 1 (LOX1) seems particularly promising because of its outstanding role in the inflammatory process. Using this pioneering concept, it will be possible to promote the impact of statins and thiazolidinediones on macrophages and endothelial cells and significantly enhance their anti-atherosclerotic therapeutic potential.


Asunto(s)
Aterosclerosis , Fármacos Cardiovasculares , Placa Aterosclerótica , Aterosclerosis/tratamiento farmacológico , Células Endoteliales , Humanos , Lipoproteínas LDL , Nanotecnología
16.
Int J Mol Sci ; 22(5)2021 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-33806499

RESUMEN

Diabetic nephropathy (DN) ranks among the most detrimental long-term effects of diabetes, affecting more than 30% of all patients. Within the diseased kidney, intraglomerular mesangial cells play a key role in facilitating the pro-fibrotic turnover of extracellular matrix components and a progredient glomerular hyperproliferation. These pathological effects are in part caused by an impaired functionality of soluble guanylate cyclase (sGC) and a consequentially reduced synthesis of anti-fibrotic messenger 3',5'-cyclic guanosine monophosphate (cGMP). Bay 58-2667 (cinaciguat) is able to re-activate defective sGC; however, the drug suffers from poor bioavailability and its systemic administration is linked to adverse events such as severe hypotension, which can hamper the therapeutic effect. In this study, cinaciguat was therefore efficiently encapsulated into virus-mimetic nanoparticles (NPs) that are able to specifically target renal mesangial cells and therefore increase the intracellular drug accumulation. NP-assisted drug delivery thereby increased in vitro potency of cinaciguat-induced sGC stabilization and activation, as well as the related downstream signaling 4- to 5-fold. Additionally, administration of drug-loaded NPs provided a considerable suppression of the non-canonical transforming growth factor ß (TGF-ß) signaling pathway and the resulting pro-fibrotic remodeling by 50-100%, making the system a promising tool for a more refined therapy of DN and other related kidney pathologies.


Asunto(s)
Benzoatos/administración & dosificación , Sistemas de Liberación de Medicamentos , Células Mesangiales/efectos de los fármacos , Células Mesangiales/metabolismo , Guanilil Ciclasa Soluble/metabolismo , Animales , Benzoatos/farmacocinética , Materiales Biomiméticos , Células Cultivadas , GMP Cíclico/metabolismo , Nefropatías Diabéticas/tratamiento farmacológico , Nefropatías Diabéticas/metabolismo , Nefropatías Diabéticas/patología , Activación Enzimática/efectos de los fármacos , Estabilidad de Enzimas/efectos de los fármacos , Fibrosis , Humanos , Células Mesangiales/patología , Modelos Biológicos , Nanopartículas/administración & dosificación , Ratas , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta/metabolismo
17.
Pharmaceuticals (Basel) ; 15(1)2021 Dec 24.
Artículo en Inglés | MEDLINE | ID: mdl-35056075

RESUMEN

A root cause for the development and progression of primary open-angle glaucoma might be the loss of the Schlemm's canal (SC) cell function due to an impaired Angiopoietin-1 (Angpt-1)/Tie2 signaling. Current therapeutic options fail to restore the SC cell function. We propose Angpt-1 mimetic nanoparticles (NPs) that are intended to bind in a multivalent manner to the Tie2 receptor for successful receptor activation. To this end, an Angpt-1 mimetic peptide was coupled to a poly(ethylene glycol)-poly(lactic acid) (PEG-PLA) block co-polymer. The modified polymer allowed for the fabrication of Angpt-1 mimetic NPs with a narrow size distribution (polydispersity index < 0.2) and the size of the NPs ranging from about 120 nm (100% ligand density) to about 100 nm (5% ligand density). NP interaction with endothelial cells (HUVECs, EA.hy926) as surrogate for SC cells and fibroblasts as control was investigated by flow cytometry and confocal microscopy. The NP-cell interaction strongly depended on the ligand density and size of NPs. The cellular response to the NPs was investigated by a Ca2+ mobilization assay as well as by a real-time RT-PCR and Western blot analysis of endothelial nitric oxide synthase (eNOS). NPs with a ligand density of 25% opposed VEGF-induced Ca2+ influx in HUVECs significantly which could possibly increase cell relaxation and thus aqueous humor drainage, whereas the expression and synthesis of eNOS was not significantly altered. Therefore, we suggest Angpt-1 mimetic NPs as a first step towards a causative therapy to recover the loss of SC cell function during glaucoma.

18.
J Control Release ; 329: 552-569, 2021 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-33007365

RESUMEN

Viruses are nanomaterials with a number of properties that surpass those of many synthetic nanoparticles (NPs) for biomedical applications. They possess a rigorously ordered structure, come in a variety of shapes, and present unique surface elements, such as spikes. These attributes facilitate propitious biodistribution, the crossing of complex biological barriers and a minutely coordinated interaction with cells. Due to the orchestrated sequence of interactions of their stringently arranged particle corona with cellular surface receptors they effectively identify and infect their host cells with utmost specificity, while evading the immune system at the same time. Furthermore, their efficacy is enhanced by their response to stimuli and the ability to spread from cell to cell. Over the years, great efforts have been made to mimic distinct viral traits to improve biomedical nanomaterial performance. However, a closer look at the literature reveals that no comprehensive evaluation of the benefit of virus-mimetic material design on the targeting efficiency of nanomaterials exists. In this review we, therefore, elucidate the impact that viral properties had on fundamental advances in outfitting nanomaterials with the ability to interact specifically with their target cells. We give a comprehensive overview of the diverse design strategies and identify critical steps on the way to reducing them to practice. More so, we discuss the advantages and future perspectives of a virus-mimetic nanomaterial design and try to elucidate if viral mimicry holds the key for better NP targeting.


Asunto(s)
Nanopartículas , Nanoestructuras , Virus , Distribución Tisular
19.
Pharm Res ; 37(11): 217, 2020 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-33037505

RESUMEN

BACKGROUND: Leukaemia is the most prevalent form of cancer-causing death in a large number of populations and needs prompt and effective treatment. Chemotherapeutics can be used to treat leukaemia, but their pronounced killing effects to other living cells is still an issue. Active targeting to certain specific receptors in leukaemic cells is the best way to avoid damage to other living cells. Leukaemic cells can be targeted using novel nanoparticles (NPs) coated with a specific ligand, such as octreotide (OCD), to target somatostatin receptor type 2 (SSTR2), which is expressed in leukaemic cells. METHODS: Amino-PEGylated quantum dots (QDs) were chosen as model NPs. The QDs were first succinylated using succinic anhydride and then coated with OCD. The reactivity and selectivity of the formulated QDs-OCD were studied in cell lines with well-expressed SSTR2, while fluorescence was detected using confocal laser scanning microscopy (CLSM) and flow cytometry (FACS). Conclusively, QD-OCD targeting to blood cells was studied in vivo in mice and detected using inductively coupled plasma mass spectrometry and CLSM in tissues. RESULTS: Highly stable QDs coated with OCD were prepared. FACS and CLSM showed highly definite interactions with overexpressed SSTR2 in the investigated cell lines. Moreover, the in vivo results revealed a higher concentration of QDs-OCD in blood cells. The fluorescence intensity of the QDs-OCD was highly accumulated in blood cells, while the unmodified QDs did not accumulate significantly in blood cells. CONCLUSION: The formulated novel QDs-OCD can target SSTR2 overexpressed in blood cells with great potential for treating blood cancer.


Asunto(s)
Antineoplásicos/metabolismo , Colorantes Fluorescentes/química , Leucemia/metabolismo , Monocitos/metabolismo , Octreótido/metabolismo , Puntos Cuánticos , Receptores de Somatostatina/agonistas , Receptores de Somatostatina/metabolismo , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Composición de Medicamentos , Citometría de Flujo , Células HeLa , Humanos , Leucemia/tratamiento farmacológico , Leucemia/patología , Masculino , Ratones , Microscopía Confocal , Octreótido/química , Octreótido/farmacología
20.
ACS Appl Mater Interfaces ; 12(31): 34689-34702, 2020 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-32639709

RESUMEN

Viral infection patterns often rely on precisely coordinated sequences of distinct ligand-receptor interactions, leading in many cases to an outstanding target cell specificity. A successful mimicry of viral targeting strategies to create more site-specific nanoparticles (NPs) would therefore require particle-cell interactions to also be adequately controllable. In the present study, hetero-multivalent block-copolymer NPs present their attached ligands in a sterically controlled manner to create a sequential NP-cell interaction similar to the cell infiltration strategy of human adenovirus type 2. Targeting renal mesangial cells, particles therefore initially bind angiotensin II receptor type 1 (AT1r) on the cell surface via a structurally flexible AT1r antagonist. After a mandatory spatial approach, particle endocytosis is realized via binding of immobile αVß3 integrins with a previously concealed secondary ligand, thereby creating a stepwise particle-cell interplay of primary NP attachment and subsequent uptake. Manufactured adenovirus-mimetic NPs show great avidity for both target motifs in vitro, leading to a substantial binding as well as subsequent cell uptake into target mesangial cells. Additionally, steric shielding of secondary ligand visibility leads to a highly controllable, sequential ligand-receptor interaction, whereby hetero-functional NPs activate mesangial cell surface integrins only after a successful prior binding to the AT1r. This stepwise cell identification significantly enhances mesangial cell specificity in co-culture assays with different off-target cells. Additionally, described NPs display excellent in vivo robustness by efficiently accumulating in the mesangium upon injection, thereby opening new paths for possible drug delivery applications.


Asunto(s)
Nanopartículas/química , Receptor de Angiotensina Tipo 1/química , Línea Celular Tumoral , Colorantes Fluorescentes/química , Células HeLa , Humanos , Ligandos , Imagen Óptica , Tamaño de la Partícula , Propiedades de Superficie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...