Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 91
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Int J Nanomedicine ; 19: 6999-7014, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39011386

RESUMEN

Introduction: Glioblastoma multiforme (GBM), a highly invasive and prognostically challenging brain cancer, poses a significant hurdle for current treatments due to the existence of the blood-brain barrier (BBB) and the difficulty to maintain an effective drug accumulation in deep GBM lesions. Methods: We present a biomimetic nanoplatform with angiopep-2-modified macrophage membrane, loaded with indocyanine green (ICG) templated self-assembly of SN38 (AM-NP), facilitating active tumor targeting and effective blood-brain barrier penetration through specific ligand-receptor interaction. Results: Upon accumulation at tumor sites, these nanoparticles achieved high drug concentrations. Subsequent combination of laser irradiation and release of chemotherapy agent SN38 induced a synergistic chemo-photothermal therapy. Compared to bare nanoparticles (NPs) lacking cell membrane encapsulation, AM-NPs significantly suppressed tumor growth, markedly enhanced survival rates, and exhibited excellent biocompatibility with minimal side effects. Conclusion: This NIR-activatable biomimetic camouflaging macrophage membrane-based nanoparticles enhanced drug delivery targeting ability through modifications of macrophage membranes and specific ligands. It simultaneously achieved synergistic chemo-photothermal therapy, enhancing treatment effectiveness. Compared to traditional treatment modalities, it provided a precise, efficient, and synergistic method that might have contributed to advancements in glioblastoma therapy.


Asunto(s)
Barrera Hematoencefálica , Neoplasias Encefálicas , Liberación de Fármacos , Glioblastoma , Verde de Indocianina , Nanopartículas , Terapia Fototérmica , Glioblastoma/terapia , Glioblastoma/tratamiento farmacológico , Glioblastoma/metabolismo , Animales , Verde de Indocianina/química , Verde de Indocianina/farmacocinética , Verde de Indocianina/farmacología , Neoplasias Encefálicas/terapia , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/metabolismo , Humanos , Línea Celular Tumoral , Ratones , Nanopartículas/química , Terapia Fototérmica/métodos , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Irinotecán/farmacocinética , Irinotecán/química , Irinotecán/farmacología , Péptidos/química , Péptidos/farmacología , Péptidos/farmacocinética , Rayos Infrarrojos , Materiales Biomiméticos/química , Materiales Biomiméticos/farmacocinética , Materiales Biomiméticos/farmacología , Sistemas de Liberación de Medicamentos/métodos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones Desnudos , Terapia Combinada/métodos
2.
Int J Nanomedicine ; 19: 5523-5544, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38882544

RESUMEN

Metal-organic frameworks (MOFs) are porous materials resulting from the coordination of metal clusters or ions with organic ligands, merging macromolecular and coordination chemistry features. Among these, zeolitic imidazolate framework-8 (ZIF-8) stands out as a widely utilized MOF known for its robust stability in aqueous environments owing to the robust interaction between its constituent zinc ions (Zn2+) and 2-methylimidazole (2-MIM). ZIF-8 readily decomposes under acidic conditions, serving as a promising candidate for pH-responsive drug delivery systems. Moreover, biomimetic materials typically possess good biocompatibility, reducing immune reactions. By mimicking natural structures or surface features within the body, they enhance the targeting of nanoparticles, prolong their circulation time, and increase their bioavailability in vivo. This review explores the latest advancements in biomimetic ZIF-8 nanoparticles for drug delivery, elucidating the primary obstacles and future prospects in utilizing ZIF-8 for drug delivery applications.


Asunto(s)
Materiales Biomiméticos , Sistemas de Liberación de Medicamentos , Imidazoles , Estructuras Metalorgánicas , Nanopartículas , Zeolitas , Estructuras Metalorgánicas/química , Estructuras Metalorgánicas/farmacocinética , Humanos , Materiales Biomiméticos/química , Materiales Biomiméticos/farmacocinética , Zeolitas/química , Zeolitas/farmacocinética , Nanopartículas/química , Sistemas de Liberación de Medicamentos/métodos , Imidazoles/química , Imidazoles/farmacocinética , Imidazoles/administración & dosificación , Animales , Zinc/química , Zinc/farmacocinética , Zinc/administración & dosificación , Biomimética/métodos , Portadores de Fármacos/química , Portadores de Fármacos/farmacocinética , Concentración de Iones de Hidrógeno
3.
Int J Nanomedicine ; 19: 5273-5295, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38859952

RESUMEN

Purpose: Reducing the first-pass hepatic effect via intestinal lymphatic transport is an effective way to increase the oral absorption of drugs. 2-Monoacylglycerol (2-MAG) as a primary digestive product of dietary lipids triglyceride, can be assembled in chylomicrons and then transported from the intestine into the lymphatic system. Herein, we propose a biomimetic strategy and report a 2-MAG mimetic nanocarrier to target the intestinal lymphatic system via the lipid absorption pathway and improve oral bioavailability. Methods: The 2-MAG mimetic liposomes were designed by covalently bonding serinol (SER) on the surface of liposomes named SER-LPs to simulate the structure of 2-MAG. Dihydroartemisinin (DHA) was chosen as the model drug because of its disadvantages such as poor solubility and high first-pass effect. The endocytosis and exocytosis mechanisms were investigated in Caco-2 cells and Caco-2 cell monolayers. The capacity of intestinal lymphatic transport was evaluated by ex vivo biodistribution and in vivo pharmacokinetic experiments. Results: DHA loaded SER-LPs (SER-LPs-DHA) had a particle size of 70 nm and a desirable entrapment efficiency of 93%. SER-LPs showed sustained release for DHA in the simulated gastrointestinal environment. In vitro cell studies demonstrated that the cellular uptake of SER-LPs primarily relied on the caveolae- rather than clathrin-mediated endocytosis pathway and preferred to integrate into the chylomicron assembly process through the endoplasmic reticulum/Golgi apparatus route. After oral administration, SER-LPs efficiently promoted drug accumulation in mesenteric lymphatic nodes. The oral bioavailability of DHA from SER-LPs was 10.40-fold and 1.17-fold larger than that of free DHA and unmodified liposomes at the same dose, respectively. Conclusion: SER-LPs improved oral bioavailability through efficient intestinal lymphatic transport. These findings of the current study provide a good alternative strategy for oral delivery of drugs with high first-pass hepatic metabolism.


Asunto(s)
Artemisininas , Disponibilidad Biológica , Liposomas , Animales , Liposomas/química , Liposomas/farmacocinética , Células CACO-2 , Humanos , Administración Oral , Artemisininas/farmacocinética , Artemisininas/química , Artemisininas/administración & dosificación , Absorción Intestinal/efectos de los fármacos , Masculino , Distribución Tisular , Tamaño de la Partícula , Ratones , Sistema Linfático/metabolismo , Sistema Linfático/efectos de los fármacos , Ratas Sprague-Dawley , Ratas , Materiales Biomiméticos/farmacocinética , Materiales Biomiméticos/química , Mucosa Intestinal/metabolismo
4.
Int J Nanomedicine ; 19: 6177-6199, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38911498

RESUMEN

Purpose: Ginsenoside Rg3 (Rg3) and Panax notoginseng saponins (PNS) can be used for ischemic stroke treatment, however, the lack of targeting to the ischemic region limits the therapeutic effect. To address this, we leveraged the affinity of macrophage membrane proteins for inflamed brain microvascular endothelial cells to develop a macrophage membrane-cloaked liposome loaded with Rg3 and PNS (MM-Lip-Rg3/PNS), which can precisely target brain lesion region through intranasal administration. Methods: MM-Lip-Rg3/PNS was prepared by co-extrusion method and was performed by characterization, stability, surface protein, and morphology. The cellular uptake, immune escape ability, and blood-brain barrier crossing ability of MM-Lip-Rg3/PNS were studied in vitro. The in vivo brain targeting, biodistribution and anti-ischemic efficacy of MM-Lip-Rg3/PNS were evaluated in MACO rats, and we determined the diversity of the nasal brain pathway through the olfactory nerve blockade model in rats. Finally, the pharmacokinetics and brain targeting index of MM-Lip-Rg3/PNS were investigated. Results: Our results indicated that MM-Lip-Rg3/PNS was spherical with a shell-core structure. MM-Lip-Rg3/PNS can avoid mononuclear phagocytosis, actively bind to inflammatory endothelial cells, and have the ability to cross the blood-brain barrier. Moreover, MM-Lip-Rg3/PNS could specifically target ischemic sites, even microglia, increase the cumulative number of drugs in the brain, improve the inflammatory environment of the brain, and reduce the infarct size. By comparing olfactory nerve-blocking rats with normal rats, it was found that there are direct and indirect pathways for nasal entry into the brain. Pharmacokinetics demonstrated that MM-Lip-Rg3/PNS exhibited stronger brain targeting and prolonged drug half-life. Conclusion: MM-Lip-Rg3/PNS might contribute to the accumulation of Rg3 and PNS in the ischemic brain area to improve treatment efficacy. This biomimetic nano-drug delivery system provides a new and promising strategy for the treatment of ischemic stroke.


Asunto(s)
Administración Intranasal , Barrera Hematoencefálica , Ginsenósidos , Accidente Cerebrovascular Isquémico , Liposomas , Macrófagos , Animales , Liposomas/química , Accidente Cerebrovascular Isquémico/tratamiento farmacológico , Ratas , Masculino , Ginsenósidos/farmacocinética , Ginsenósidos/química , Ginsenósidos/administración & dosificación , Ginsenósidos/farmacología , Barrera Hematoencefálica/efectos de los fármacos , Macrófagos/efectos de los fármacos , Sistemas de Liberación de Medicamentos/métodos , Ratas Sprague-Dawley , Distribución Tisular , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Materiales Biomiméticos/química , Materiales Biomiméticos/farmacocinética , Materiales Biomiméticos/administración & dosificación , Saponinas/farmacocinética , Saponinas/química , Saponinas/administración & dosificación , Saponinas/farmacología , Ratones
5.
J Nanobiotechnology ; 19(1): 374, 2021 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-34789284

RESUMEN

BACKGROUND: Intimal hyperplasia caused by vascular injury is an important pathological process of many vascular diseases, especially occlusive vascular disease. In recent years, Nano-drug delivery system has attracted a wide attention as a novel treatment strategy, but there are still some challenges such as high clearance rate and insufficient targeting. RESULTS: In this study, we report a biomimetic ROS-responsive MM@PCM/RAP nanoparticle coated with macrophage membrane. The macrophage membrane with the innate "homing" capacity can superiorly regulate the recruitment of MM@PCM/RAP to inflammatory lesion to enhance target efficacy, and can also disguise MM@PCM/RAP nanoparticle as the autologous cell to avoid clearance by the immune system. In addition, MM@PCM/RAP can effectively improve the solubility of rapamycin and respond to the high concentration level of ROS accumulated in pathological lesion for controlling local cargo release, thereby increasing drug availability and reducing toxic side effects. CONCLUSIONS: Our findings validate that the rational design, biomimetic nanoparticles MM@PCM/RAP, can effectively inhibit the pathological process of intimal injury with excellent biocompatibility.


Asunto(s)
Hiperplasia/metabolismo , Macrófagos/citología , Sistema de Administración de Fármacos con Nanopartículas , Especies Reactivas de Oxígeno/metabolismo , Túnica Íntima , Animales , Materiales Biomiméticos/química , Materiales Biomiméticos/farmacocinética , Materiales Biomiméticos/farmacología , Membrana Celular/química , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Sistema de Administración de Fármacos con Nanopartículas/química , Sistema de Administración de Fármacos con Nanopartículas/metabolismo , Sirolimus/química , Sirolimus/farmacocinética , Sirolimus/farmacología , Túnica Íntima/efectos de los fármacos , Túnica Íntima/metabolismo , Pez Cebra
6.
J Nanobiotechnology ; 19(1): 385, 2021 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-34809623

RESUMEN

Demineralization of hard tooth tissues leads to dental caries, which cause health problems and economic burdens throughout the world. A biomimetic mineralization strategy is expected to reverse early dental caries. Commercially available anti-carious mineralizing products lead to inconclusive clinical results because they cannot continuously replenish the required calcium and phosphate resources. Herein, we prepared a mineralizing film consisting of hydroxypropylmethylcellulose (HPMC) and polyaspartic acid-stabilized amorphous calcium phosphate (PAsp-ACP) nanoparticles. HPMC which contains multiple hydroxyl groups is a film-forming material that can be desiccated to form a dry film. In a moist environment, this film gradually changes into a gel. HPMC was used as the carrier of PAsp-ACP nanoparticles to deliver biomimetic mineralization. Our results indicated that the hydroxyl and methoxyl groups of HPMC could assist the stability of PAsp-ACP nanoparticles and maintain their biomimetic mineralization activity. The results further demonstrated that the bioinspired mineralizing film induced the early mineralization of demineralized dentin after 24 h with increasing mineralization of the whole demineralized dentin (3-4 µm) after 72-96 h. Furthermore, these results were achieved without any cytotoxicity or mucosa irritation. Therefore, this mineralizing film shows promise for use in preventive dentistry due to its efficient mineralization capability.


Asunto(s)
Materiales Biomiméticos , Fosfatos de Calcio , Caries Dental/metabolismo , Derivados de la Hipromelosa , Calcificación de Dientes/efectos de los fármacos , Animales , Materiales Biomiméticos/química , Materiales Biomiméticos/farmacocinética , Materiales Biomiméticos/farmacología , Fosfatos de Calcio/química , Fosfatos de Calcio/farmacocinética , Fosfatos de Calcio/farmacología , Células Cultivadas , Dentina/efectos de los fármacos , Humanos , Hidrogeles/química , Hidrogeles/farmacología , Derivados de la Hipromelosa/química , Derivados de la Hipromelosa/farmacología , Masculino , Ratones , Sistema de Administración de Fármacos con Nanopartículas , Nanopartículas , Conejos
7.
J Nanobiotechnology ; 19(1): 339, 2021 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-34689761

RESUMEN

BACKGROUND: Biomimetic nanotechnology-based RNA interference (RNAi) has been successful in improving theranostic efficacy in malignant tumors. Its integration with hybrid biomimetic membranes made of natural cell membranes fused with liposomal membranes is mutually beneficial and extends their biofunctions. However, limited research has focused on engineering such biomimetics to endow them with unique properties and functions, in particular, those essential for a "smart" drug delivery system, such as a tumor microenvironment (TME)-activated multifunctional biomimetic nanoplatform. RESULTS: Herein, we utilized an integrated hybrid nanovesicle composed of cancer cell membranes (Cm) and matrix metallopeptidase 9 (MMP-9)-switchable peptide-based charge-reversal liposome membranes (Lipm) to coat lipoic acid-modified polypeptides (LC) co-loaded with phosphoglycerate mutase 1 (PGAM1) siRNA (siPGAM1) and DTX. The nanovesicle presented a negatively charged coating (citraconic anhydride-grafted poly-L-lysine, PC) in the middle layer for pH-triggered charge conversion functionalization. The established chemotherapeutic drug (DTX) co-delivery system CLip-PC@CO-LC nanoparticles (NPs) have a particle size of ~ 193 nm and present the same surface proteins as the Cm. Confocal microscopy and flow cytometry results indicated a greater uptake of MMP-9-treated CLip-PC@CO-LC NPs compared with that of the CLip-PC@CO-LC NPs without MMP-9 pretreatment. The exposure to MMP-9 activated positively charged cell-penetrating peptides on the surface of the hybrid nanovesicles. Moreover, pH triggered membrane disruption, and redox triggered DTX and siRNA release, leading to highly potent target-gene silencing in glycolysis and chemotherapy with enhanced antiproliferation ability. The biodistribution results demonstrated that the CLip-PC@LC-DiR NPs accumulated in the tumor owing to a combination of long blood retention time, homologous targeting ability, and TME-activated characteristics. The CLip-PC@CO-LC NPs led to more effective tumor growth inhibition than the DTX and free siPGAM1 formulations. CONCLUSIONS: TME-activated cancer cell membrane-liposome integrated hybrid NPs provide an encouraging nanoplatform that combines RNAi with chemotherapy for precise treatment of non-small cell lung cancer.


Asunto(s)
Antineoplásicos , Materiales Biomiméticos , Membrana Celular/química , Liposomas , Microambiente Tumoral/efectos de los fármacos , Células A549 , Animales , Antineoplásicos/química , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Materiales Biomiméticos/química , Materiales Biomiméticos/farmacocinética , Materiales Biomiméticos/farmacología , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Sistemas de Liberación de Medicamentos , Glucólisis , Humanos , Liposomas/química , Liposomas/farmacocinética , Neoplasias Pulmonares/metabolismo , Ratones , Ratones Desnudos , Nanopartículas/química , Nanopartículas/metabolismo
8.
J Am Chem Soc ; 143(43): 18029-18040, 2021 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-34664942

RESUMEN

The chemical topology is a unique dimension for protein engineering, yet the topological diversity and architectural complexity of proteins remain largely untapped. Herein, we report the biosynthesis of complex topological proteins using a rationally engineered, cross-entwining peptide heterodimer motif derived from p53dim (an entangled homodimeric mutant of the tetramerization domain of the tumor suppressor protein p53). The incorporation of an electrostatic interaction at specific sites converts the p53dim homodimer motif into a pair of heterodimer motifs with high specificity for directing chain entanglement upon folding. Its combination with split-intein-mediated ligation and/or SpyTag/SpyCatcher chemistry facilitates the programmed synthesis of protein heterocatenane or [n]catenanes in cells, leading to a general and modular approach to complex protein catenanes containing various proteins of interest. Concatenation enhances not only the target protein's affinity but also the in vivo stability as shown by its prolonged circulation time in blood. As a proof of concept, artificial antibodies have been developed by embedding a human epidermal growth factor receptor 2-specific affibody onto the [n]catenane scaffolds and shown to exhibit a higher affinity and a better pharmacokinetic profile than the wild-type affibody. These results suggest that topology engineering holds great promise in the development of therapeutic proteins.


Asunto(s)
Anticuerpos/química , Materiales Biomiméticos/metabolismo , Catenanos/metabolismo , Fragmentos de Péptidos/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Secuencia de Aminoácidos , Animales , Materiales Biomiméticos/química , Materiales Biomiméticos/farmacocinética , Catenanos/química , Catenanos/farmacocinética , Línea Celular Tumoral , Femenino , Humanos , Ratones Endogámicos BALB C , Fragmentos de Péptidos/química , Fragmentos de Péptidos/farmacocinética , Prueba de Estudio Conceptual , Dominios Proteicos , Ingeniería de Proteínas , Estructura Cuaternaria de Proteína , Receptor ErbB-2/metabolismo , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/farmacocinética , Proteína p53 Supresora de Tumor/química , Proteína p53 Supresora de Tumor/farmacocinética
9.
Drug Deliv ; 28(1): 1237-1255, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34142930

RESUMEN

Nanotechnology provides synthetic carriers for cancer drug delivery that protect cargos from degradation, control drug release and increase local accumulation at tumors. However, these non-natural vehicles display poor tumor targeting and potential toxicity and are eliminated by the immune system. Recently, biomimetic nanocarriers have been widely developed based on the concept of 'mimicking nature.' Among them, cell-derived biomimetic vehicles have become the focus of bionics research because of their multiple natural functions, such as low immunogenicity, long circulation time and targeting ability. Cell membrane-coated carriers and extracellular vesicles are two widely used cell-based biomimetic materials. Here, this review summarizes the latest progress in the application of these two biomimetic carriers in targeted cancer therapy. Their properties and performance are compared, and their future challenges and development prospects are discussed.


Asunto(s)
Materiales Biomiméticos/farmacología , Biomimética/métodos , Membrana Celular/metabolismo , Portadores de Fármacos/farmacología , Vesículas Extracelulares/metabolismo , Materiales Biomiméticos/farmacocinética , Química Farmacéutica , Portadores de Fármacos/química , Portadores de Fármacos/farmacocinética , Humanos , Nanopartículas/química , Neoplasias/tratamiento farmacológico
10.
Eur J Pharm Biopharm ; 158: 382-389, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33309845

RESUMEN

Interaction of conventional drug delivery systems such as polymeric or lipid based nano- and microparticles with the in vivo milieu has garnered significant interest, primarily to orchestrate immune escape and/or improve targeting. Surface modification with targeting ligands has been heavily relied upon for the mentioned purpose in the recent years. However, the surface modified particles can also activate the immune system. Large-scale manufacturing can also be a challenge, as surface modification needs to be reproducible. Furthermore, in vivo, the targeting is dependent on the receptor expression density and number of target sites, which adds to the pharmacokinetic variability of the constructs. An evolving paradigm to overcome complications of surface functionalization is the incorporation of bio-inspired topographies into these conventional delivery systems to enable them to better interact with biological systems. Biomimetic delivery systems combine the unique surface composition of cells or cell membranes, and versatility of synthetic nanoparticles. In this review, we focus on one such delivery system, silica particles, and explore their interaction with different biological membranes.


Asunto(s)
Antineoplásicos/administración & dosificación , Materiales Biomiméticos/química , Portadores de Fármacos/química , Neoplasias/tratamiento farmacológico , Dióxido de Silicio/química , Animales , Antineoplásicos/farmacocinética , Materiales Biomiméticos/farmacocinética , Membrana Celular/metabolismo , Preparaciones de Acción Retardada/administración & dosificación , Preparaciones de Acción Retardada/farmacocinética , Modelos Animales de Enfermedad , Portadores de Fármacos/farmacocinética , Liberación de Fármacos , Humanos , Nanopartículas/química , Dióxido de Silicio/farmacocinética , Propiedades de Superficie
11.
Theranostics ; 10(25): 11607-11621, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33052236

RESUMEN

The hypoxic microenvironment in solid tumors severely limits the efficacy of photodynamic therapy (PDT). Therefore, the development of nanocarriers co-loaded with photosensitizers and oxygen, together with imaging guidance ability, is of great significance in cancer therapy. However, previously reported synthetic methods for these multi-functional probes are complicated, and the raw materials used are toxic. Methods: Herein, the human endogenous protein, hemoglobin (Hb), was used for the simultaneous biomimetic synthesis of Gd-based nanostructures and co-loading of Chlorine e6 (Ce6) and oxygen for alleviating the hypoxic environment of tumors and accomplishing magnetic resonance imaging (MRI)-guided enhanced PDT. The Gd@HbCe6-PEG nanoprobes were synthesized via a green and protein biomimetic approach. The physicochemical properties, including relaxivity, oxygen-carrying/release capability, and PDT efficacy of Gd@HbCe6-PEG, were measured in vitro and in vivo on tumor-bearing mice after intravenous injection. Morphologic and functional MRI were carried out to evaluate the efficacy of PDT. Results: The results demonstrated the successful synthesis of compact Gd@HbCe6-PEG nanostructures with desired multi-functionalities. Following treatment with the nanoparticles, the embedded MR moiety was effective in lighting tumor lesions and guiding therapy. The oxygen-carrying capability of Hb after biomimetic synthesis was confirmed by spectroscopic analysis and oxygen detector in vitro. Further, tumor oxygenation for alleviating tumor hypoxia in vivo after intravenous injection of Gd@HbCe6-PEG was verified by photoacoustic imaging and immunofluorescence staining. The potent treatment efficacy of PDT on early-stage was observed by the morphologic and functional MR imaging. Importantly, rapid renal clearance of the particles was observed after treatment. Conclusion: In this study, by using a human endogenous protein, we demonstrated the biomimetic synthesis of multi-functional nanoprobes for simultaneous tumor oxygenation and imaging-guided enhanced PDT. The therapeutic efficacy could be quantitatively confirmed at 6 h post PDT with diffusion-weighted imaging (DWI).


Asunto(s)
Antineoplásicos/administración & dosificación , Nanopartículas del Metal/administración & dosificación , Neoplasias/tratamiento farmacológico , Fotoquimioterapia/métodos , Nanomedicina Teranóstica/métodos , Animales , Antineoplásicos/síntesis química , Antineoplásicos/farmacocinética , Materiales Biomiméticos/administración & dosificación , Materiales Biomiméticos/síntesis química , Materiales Biomiméticos/farmacocinética , Línea Celular Tumoral/trasplante , Clorofilidas , Imagen de Difusión por Resonancia Magnética , Modelos Animales de Enfermedad , Femenino , Gadolinio/administración & dosificación , Gadolinio/química , Tecnología Química Verde , Hemoglobinas/administración & dosificación , Hemoglobinas/química , Humanos , Inyecciones Intravenosas , Nanopartículas del Metal/química , Ratones , Sondas Moleculares/administración & dosificación , Sondas Moleculares/síntesis química , Sondas Moleculares/farmacocinética , Neoplasias/diagnóstico por imagen , Oxígeno/administración & dosificación , Oxígeno/química , Técnicas Fotoacústicas , Porfirinas/administración & dosificación , Porfirinas/química , Hipoxia Tumoral/efectos de los fármacos , Microambiente Tumoral/efectos de los fármacos
12.
J Nanobiotechnology ; 18(1): 115, 2020 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-32819405

RESUMEN

BACKGROUND: Glucocorticoids (GCs) show powerful treatment effect on rheumatoid arthritis (RA). However, the clinical application is limited by their nonspecific distribution after systemic administration, serious adverse reactions during long-term administration. To achieve better treatment, reduce side effect, we here established a biomimetic exosome (Exo) encapsulating dexamethasone sodium phosphate (Dex) nanoparticle (Exo/Dex), whose surface was modified with folic acid (FA)-polyethylene glycol (PEG)-cholesterol (Chol) compound to attain FPC-Exo/Dex active targeting drug delivery system. RESULTS: The size of FPC-Exo/Dex was 128.43 ± 16.27 nm, with a polydispersity index (PDI) of 0.36 ± 0.05, and the Zeta potential was - 22.73 ± 0.91 mV. The encapsulation efficiency (EE) of the preparation was 10.26 ± 0.73%, with drug loading efficiency (DLE) of 18.81 ± 2.05%. In vitro study showed this system displayed enhanced endocytosis and excellent anti-inflammation effect against RAW264.7 cells by suppressing pro-inflammatory cytokines and increasing anti-inflammatory cytokine. Further biodistribution study showed the fluorescence intensity of FPC-Exo/Dex was stronger than other Dex formulations in joints, suggesting its enhanced accumulation to inflammation sites. In vivo biodistribution experiment displayed FPC-Exo/Dex could preserve the bone and cartilage of CIA mice better and significantly reduce inflamed joints. Next in vivo safety evaluation demonstrated this biomimetic drug delivery system had no obvious hepatotoxicity and exhibited desirable biocompatibility. CONCLUSION: The present study provides a promising strategy for using exosome as nanocarrier to enhance the therapeutic effect of GCs against RA.


Asunto(s)
Artritis Reumatoide/metabolismo , Materiales Biomiméticos , Dexametasona , Exosomas , Nanopartículas , Animales , Antiinflamatorios/química , Antiinflamatorios/farmacocinética , Artritis Reumatoide/patología , Materiales Biomiméticos/química , Materiales Biomiméticos/farmacocinética , Colesterol/análogos & derivados , Colesterol/química , Dexametasona/química , Dexametasona/farmacocinética , Exosomas/química , Exosomas/metabolismo , Ácido Fólico/química , Articulaciones/metabolismo , Articulaciones/patología , Masculino , Ratones , Nanopartículas/química , Nanopartículas/metabolismo , Polietilenglicoles/química , Células RAW 264.7
13.
J Nanobiotechnology ; 18(1): 71, 2020 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-32404183

RESUMEN

BACKGROUND: Although many therapeutic strategies for Alzheimer's disease (AD) have been explored, these strategies are seldom used in the clinic. Therefore, AD therapeutic research is still urgently needed. One major challenge in the field of nanotherapeutics is to increase the selective delivery of drugs to a targeted location. Herein, we devised and tested a strategy for delivery of nanoparticles to neurons to inhibit tau aggregation by directly targeting p-tau. RESULTS: Curcumin (CUR) is loaded onto red blood cell (RBC) membrane-coated PLGA particles bearing T807 molecules attached to the RBC membrane surface (T807/RPCNP). With the advantage of the suitable physicochemical properties of the PLGA nanoparticles and the unique biological functions of the RBC membrane, the RPCNP are stabilized and promote sustained CUR release, which provided improved biocompatibility and resulted in long-term presence in the circulation. Under the synergistic effects of T807, T807/RPCNP can not only effectively penetrate the blood-brain barrier (BBB), but they also possess high binding affinity to hyperphosphorylated tau in nerve cells where they inhibit multiple key pathways in tau-associated AD pathogenesis. When CUR was encapsulated, our data also demonstrated that CUR-loaded T807/RPCNP NPs can relieve AD symptoms by reducing p-tau levels and suppressing neuronal-like cells death both in vitro and in vivo. The memory impairment observed in an AD mouse model is significantly improved following systemic administration of CUR-loaded T807/RPCNP NPs. CONCLUSION: Intravenous neuronal tau-targeted T807-modified novel biomimetic nanosystems are a promising clinical candidate for the treatment of AD.


Asunto(s)
Enfermedad de Alzheimer , Materiales Biomiméticos , Curcumina , Portadores de Fármacos , Nanopartículas/química , Animales , Apoptosis/efectos de los fármacos , Materiales Biomiméticos/química , Materiales Biomiméticos/farmacocinética , Barrera Hematoencefálica/metabolismo , Línea Celular , Curcumina/química , Curcumina/farmacocinética , Curcumina/farmacología , Modelos Animales de Enfermedad , Portadores de Fármacos/química , Portadores de Fármacos/farmacocinética , Hipocampo/efectos de los fármacos , Hipocampo/patología , Humanos , Aprendizaje por Laberinto/efectos de los fármacos , Ratones , Neuronas/metabolismo , Sustancias Protectoras/química , Sustancias Protectoras/farmacocinética , Sustancias Protectoras/farmacología , Proteínas tau/metabolismo
14.
Biomolecules ; 10(5)2020 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-32443614

RESUMEN

A new six intraperitoneal injections insulin-mimetic vanadyl(IV) compounds [(VO)(FA)(AAn)] (where n = 1-6: AA1 = isoleucine, AA2 = threonine, AA3 = proline, AA4 = phenylalanine, AA5 = lysine, and AA6 = glutamine) were synthesized by the chemical reactions between folic acid (FA), VOSO4, and amino acids (AAn) with equal molar ratio 1:1:1 in neutralized media. These complexes were characterized by elemental analysis and estimation of vanadyl(IV) metal ions. The thermal stability behavior of these complexes was studied by TG-DTG-DTA analyses. The structures of these complexes were elucidated by spectroscopic methods like infrared, electron spin resonance (ESR), and solid reflectance spectroscopes. The powder X-ray diffraction (XRD) study suggested the crystalline nature of the complexes. Magnetic moments and electronic spectra revealed the square-pyramid geometrical structure of the complexes. The conductivity results refereed that all synthesized vanadyl(IV) complexes were of a non-electrolyte behavior. The infrared spectra assignments of these complexes revealed that the FAH2 and AAn chelates act as a bidentate ligation. The chelation towards vanadyl (IV) ions existed via deprotonation of one of the carboxylic groups of FAH2 drug ligand, and so amino acids act as bidentate ligands via N-amino and O-carboxylate groups. Both scanning and transmission electron microscope (SEM and TEM) techniques were used to investigate the surface morphology. The main task of this research is the aim of designing a new insulin alternative antidiabetic drug agent. The antidiabetic efficiency of these complexes was evaluated in streptozotocin-induced diabetic male albino rats. Liver and kidney functions, insulin and blood glucose levels, lipid profile, and superoxide dismutase antioxidant (SOD) are verified identifiers for the efficiency of VO(IV)/FA/AAn system compounds as antidiabetic drug agents.


Asunto(s)
Materiales Biomiméticos/síntesis química , Complejos de Coordinación/síntesis química , Diabetes Mellitus Experimental/tratamiento farmacológico , Ácido Fólico/análogos & derivados , Hipoglucemiantes/síntesis química , Insulina/química , Compuestos de Vanadio/química , Aminoácidos/química , Animales , Materiales Biomiméticos/farmacocinética , Materiales Biomiméticos/uso terapéutico , Complejos de Coordinación/farmacocinética , Complejos de Coordinación/uso terapéutico , Hipoglucemiantes/farmacocinética , Hipoglucemiantes/uso terapéutico , Riñón/efectos de los fármacos , Riñón/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratas
15.
Drug Deliv ; 27(1): 502-518, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32228100

RESUMEN

Reactive oxygen species (ROS)-induced neuronal mitochondrial dysfunction is a key pathologic factor in sporadic Alzheimer's disease (AD). Neuronal mitochondria have been proposed to be a promising therapeutic target for AD, especially for the failures of phase III clinical trials on conventional amyloid-ß (Aß) targeted therapy. However, the efficient intravenous delivery of therapeutic agents to neuronal mitochondria in the brain remains a major challenge due to the complicated physiological environment. Recently, biomaterials-based nanomedicine has been widely investigated for the treatment of AD. Herein, we devised a strategy for functional antioxidant delivery to neuronal mitochondria by loading antioxidants into red blood cell (RBC) membrane-coated nanostructured lipid carriers (NLC) bearing rabies virus glycoprotein (RVG29) and triphenylphosphine cation (TPP) molecules attached to the RBC membrane surface (RVG/TPP NPs@RBCm). With the advantage of suitable physicochemical properties of NLC and unique biological functions of the RBC membrane, RVG/TPP NPs@RBCm are stabilized and enabled sustained drug release, providing improved biocompatibility and long-term circulation. Under the synergistic effects of RVG29 and TPP, RVG/TPP NPs@RBCm can not only penetrate the blood-brain barrier (BBB) but also target neuron cells and further localize in the mitochondria. After encapsulating Resveratrol (RSV) as the model antioxidant, the data demonstrated that RVG/TPP-RSV NPs@RBCm can relieve AD symptoms by mitigating Aß-related mitochondrial oxidative stress both in vitro and in vivo. The memory impairment in APP/PS1 mice is significantly improved following the systemic administration of RVG/TPP-RSV NPs@RBCm. In conclusion, intravenous neuronal mitochondria-targeted dual-modified novel biomimetic nanosystems are a promising therapeutic candidate for ROS-induced mitochondrial dysfunction in AD.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Antioxidantes/administración & dosificación , Materiales Biomiméticos/química , Mitocondrias/efectos de los fármacos , Nanopartículas/química , Neuronas/efectos de los fármacos , Resveratrol/administración & dosificación , Administración Intravenosa , Enfermedad de Alzheimer/metabolismo , Animales , Antioxidantes/farmacocinética , Antioxidantes/uso terapéutico , Transporte Biológico/efectos de los fármacos , Materiales Biomiméticos/farmacocinética , Materiales Biomiméticos/uso terapéutico , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Línea Celular , Portadores de Fármacos/química , Portadores de Fármacos/farmacocinética , Portadores de Fármacos/uso terapéutico , Membrana Eritrocítica/química , Masculino , Ratones , Ratones Endogámicos ICR , Mitocondrias/metabolismo , Nanopartículas/metabolismo , Nanopartículas/uso terapéutico , Neuronas/metabolismo , Compuestos Organofosforados/farmacocinética , Compuestos Organofosforados/uso terapéutico , Estrés Oxidativo/efectos de los fármacos , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Resveratrol/farmacocinética , Resveratrol/uso terapéutico , Distribución Tisular
16.
Mater Sci Eng C Mater Biol Appl ; 109: 110566, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32228987

RESUMEN

Marine demosponges of the Verongiida order are considered a gold-mine for bioinspired materials science and marine pharmacology. The aim of this work was to simultaneously isolate selected bromotyrosines and unique chitinous structures from A. aerophoba and to propose these molecules and biomaterials for possible application as antibacterial and antitumor compounds and as ready-to-use scaffolds for cultivation of cardiomyocytes, respectively. Among the extracted bromotyrosines, the attention has been focused on aeroplysinin-1 that showed interesting unexpected growth inhibition properties for some Gram-negative clinical multi-resistant bacterial strains, such as A. baumannii and K. pneumoniae, and on aeroplysinin-1 and on isofistularin-3 for their anti-tumorigenic activity. For both compounds, the effects are cell line dependent, with significant growth inhibition activity on the neuroblastoma cell line SH-SY5Y by aeroplysinin-1 and on breast cancer cell line MCF-7 by isofistularin-3. In this study, we also compared the cultivation of human induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) on the A. aerophoba chitinous scaffolds, in comparison to chitin structures that were pre-coated with Geltrex™, an extracellular matrix mimetic which is used to enhance iPSC-CM adhesion. The iPSC-CMs on uncoated and pure chitin structures started contracting 24 h after seeding, with comparable behaviour observed on Geltrex-coated cell culture plates, confirming the biocompatibility of the sponge biomaterial with this cell type. The advantage of A. aerophoba is that this source organism does not need to be collected in large quantities to supply the necessary amount for further pre-clinical studies before chemical synthesis of the active compounds will be available. A preliminary analysis of marine sponge bioeconomy as a perspective direction for application of biomaterials and secondary bioactive metabolites has been finally performed for the first time.


Asunto(s)
Acetonitrilos , Alcaloides , Organismos Acuáticos/química , Materiales Biomiméticos , Ciclohexenos , Células Madre Pluripotentes Inducidas/metabolismo , Miocitos Cardíacos/metabolismo , Poríferos/química , Acetonitrilos/química , Acetonitrilos/farmacocinética , Acetonitrilos/farmacología , Alcaloides/química , Alcaloides/farmacocinética , Alcaloides/farmacología , Animales , Materiales Biomiméticos/química , Materiales Biomiméticos/farmacocinética , Materiales Biomiméticos/farmacología , Línea Celular Tumoral , Ciclohexenos/química , Ciclohexenos/farmacocinética , Ciclohexenos/farmacología , Preparaciones de Acción Retardada/química , Preparaciones de Acción Retardada/farmacocinética , Humanos , Células Madre Pluripotentes Inducidas/citología , Células MCF-7 , Miocitos Cardíacos/citología
17.
Nat Commun ; 11(1): 1126, 2020 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-32111847

RESUMEN

The efficacy of nano-mediated drug delivery has been impeded by multiple biological barriers such as the mononuclear phagocyte system (MPS), as well as vascular and interstitial barriers. To overcome the abovementioned obstacles, we report a nano-pathogenoid (NPN) system that can in situ hitchhike circulating neutrophils and supplement photothermal therapy (PTT). Cloaked with bacteria-secreted outer membrane vesicles inheriting pathogen-associated molecular patterns of native bacteria, NPNs are effectively recognized and internalized by neutrophils. The neutrophils migrate towards inflamed tumors, extravasate across the blood vessels, and penetrate through the tumors. Then NPNs are rapidly released from neutrophils in response to inflammatory stimuli and subsequently taken up by tumor cells to exert anticancer effects. Strikingly, due to the excellent targeting efficacy, cisplatin-loaded NPNs combined with PTT completely eradicate tumors in all treated mice. Such a nano-platform represents an efficient and generalizable strategy towards in situ cell hitchhiking as well as enhanced tumor targeted delivery.


Asunto(s)
Quimiotaxis de Leucocito , Sistemas de Liberación de Medicamentos , Nanopartículas/administración & dosificación , Neoplasias/terapia , Neutrófilos/fisiología , Fototerapia , Animales , Membrana Externa Bacteriana/química , Membrana Externa Bacteriana/inmunología , Materiales Biomiméticos/administración & dosificación , Materiales Biomiméticos/química , Materiales Biomiméticos/farmacocinética , Cisplatino/administración & dosificación , Cisplatino/química , Cisplatino/farmacocinética , Liberación de Fármacos , Vesículas Extracelulares/química , Vesículas Extracelulares/inmunología , Inmunoterapia Adoptiva , Inflamación/etiología , Ratones , Nanopartículas/química , Nanopartículas/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Activación Neutrófila , Infiltración Neutrófila , Neutrófilos/inmunología , Neutrófilos/metabolismo , Moléculas de Patrón Molecular Asociado a Patógenos/inmunología , Fototerapia/efectos adversos , Microambiente Tumoral/efectos de la radiación , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Nano Lett ; 20(3): 1637-1646, 2020 03 11.
Artículo en Inglés | MEDLINE | ID: mdl-32013452

RESUMEN

Nanotechnology-based RNA interference (RNAi) has shown great promise in overcoming the limitations of traditional clinical treatments for glioblastoma (GBM). However, because of the complexity of brain physiology, simple blood-brain barrier (BBB) penetration or tumor-targeting strategies cannot entirely meet the demanding requirements of different therapeutic delivery stages. Herein, we developed a charge conversional biomimetic nanoplatform with a three-layer core-shell structure to programmatically overcome persistent obstacles in siRNA delivery to GBM. The resulting nanocomplex presents good biocompatibility, prolonged blood circulation, high BBB transcytosis, effective tumor accumulation, and specific uptake by tumor cells in the brain. Moreover, red blood cell membrane (RBCm) disruption and effective siRNA release can be further triggered elegantly by charge conversion from negative to positive in the endo/lysosome (pH 5.0-6.5) of tumor cells, leading to highly potent target-gene silencing with a strong anti-GBM effect. Our study provides an intelligent biomimetic nanoplatform tailored for systemically siRNA delivery to GBM, leveraging Angiopep-2 peptide-modified, immune-free RBCm and charge conversional components. Improved therapeutic efficacy, higher survival rates, and minimized systemic side effects were achieved in orthotopic U87MG-luc human glioblastoma tumor-bearing nude mice.


Asunto(s)
Materiales Biomiméticos , Neoplasias Encefálicas , Glioblastoma , Interferencia de ARN , ARN Interferente Pequeño , Animales , Materiales Biomiméticos/química , Materiales Biomiméticos/farmacocinética , Materiales Biomiméticos/farmacología , Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/patología , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Glioblastoma/tratamiento farmacológico , Glioblastoma/metabolismo , Glioblastoma/patología , Ratones , Ratones Desnudos , ARN Interferente Pequeño/química , ARN Interferente Pequeño/farmacocinética , ARN Interferente Pequeño/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Nanoscale ; 12(5): 3317-3329, 2020 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-31976511

RESUMEN

Immune checkpoint blockade (ICB) has emerged as one of the breakthrough approaches for tumor immunotherapy. However, known as an immune "cold" tumor, breast cancer harbors an immunosuppressive tumor niche that compromises ICB-based therapy. Chemoimmunotherapy combines a chemotherapeutic with an immune-modulating agent, representing a promising tactic to combat cancers, while the lack of effectively targeted co-delivery strategy is one of the main obstacles to achieve the synergistic utilization. Herein, self-assembled PEGylated pure drug-based nanohybrids (DNH) were created, which could evoke immunogenic cell death (ICD), aiding ICB-based immunotherapy by controlling the spatiotemporal release of oxaliplatin (OXA) and small molecular inhibitor 1-methyl-d-tryptophan (1-MT). Furthermore, biomimetic functionalization was exploited by nature killer cell membrane camouflaging to target cancerous cells as well as by elicit immune response through inducing M1 macrophage polarization. The drug release profiles of the nanosystem were investigated in the presence of low pH and intracellular reductants. Systemic in vivo bio-behaviors were evaluated via pharmacokinetics and biodistribution. As an "all-in-one" pure drug-based codelivery system, our biomimetic nanoplatform possessed multiple immunomodulation functions, which markedly aided in increasing the frequency of immune responders and generate an immune "hot" breast tumor niche, and eventually allowed to boost breast cancer therapy.


Asunto(s)
Materiales Biomiméticos , Neoplasias de la Mama , Nanopartículas , Oxaliplatino , Triptófano/análogos & derivados , Microambiente Tumoral , Animales , Materiales Biomiméticos/farmacocinética , Materiales Biomiméticos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/patología , Femenino , Humanos , Células MCF-7 , Ratones Endogámicos BALB C , Nanopartículas/química , Nanopartículas/uso terapéutico , Oxaliplatino/farmacocinética , Oxaliplatino/farmacología , Triptófano/farmacocinética , Triptófano/farmacología , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Biomacromolecules ; 21(2): 688-700, 2020 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-31769678

RESUMEN

Bioinspired nonantibiotics can prove to be a better and an efficient tool to fight against antimicrobial resistance. In our study, biomaterial composed of zinc-carboxymethyl chitosan (CMC)-genipin was investigated for this purpose. Briefly, CMC was synthesized and transformed to porous scaffolds using the freeze drying method. The scaffolds were cross-linked and stabilized with genipin and zinc (2 M zinc acetate), respectively. FTIR spectroscopic data testified Zn complex formation and pointed out the absence of water molecule like that of zinc motif containing proteins. Hence, the complex may be termed as biomimetic. Genipin (0.5%) cross-linking appeared to contribute additively to the wet compressive strength of the zinc-CMC scaffolds. Biodegradation data revealed better stability of CMC-genipin-zinc scaffolds in enzymatic and nonenzymatic conditions than their redundant controls. The scaffolds seem to support adhesion and proliferation of human dental pulp stem cells and were hemocompatible to human red blood corpuscles, as revealed by scanning electron microscopy. The scaffolds were found to be antibacterial and mildly antibiofilm when tested against biofilm-forming bacteria, that is, Staphylococcus aureus (ATCC 9144), making it a potential nonantibiotic-like biomaterial. To conclude, this organometallic complex-based biomaterial may potentially serve as a weapon against antimicrobial resistance. Furthermore, the biomaterial potentially finds its application in dental, maxillofacial, and orthopedic tissue engineering applications.


Asunto(s)
Adhesivos/química , Materiales Biocompatibles/farmacocinética , Materiales Biomiméticos/farmacocinética , Quitosano/análogos & derivados , Iridoides/química , Zinc/química , Adhesivos/farmacocinética , Materiales Biocompatibles/química , Materiales Biomiméticos/química , Biomimética/métodos , Células Cultivadas , Quitosano/química , Quitosano/farmacocinética , Pulpa Dental/efectos de los fármacos , Pulpa Dental/metabolismo , Eritrocitos/efectos de los fármacos , Eritrocitos/metabolismo , Humanos , Iridoides/farmacocinética , Ensayo de Materiales/métodos , Pruebas de Sensibilidad Microbiana/métodos , Andamios del Tejido , Zinc/farmacocinética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...