Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 90
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 175(6): 1467-1480.e13, 2018 11 29.
Artigo em Inglês | MEDLINE | ID: mdl-30500534

RESUMO

Liquid-liquid phase separation plays a key role in the assembly of diverse intracellular structures. However, the biophysical principles by which phase separation can be precisely localized within subregions of the cell are still largely unclear, particularly for low-abundance proteins. Here, we introduce an oligomerizing biomimetic system, "Corelets," and utilize its rapid and quantitative light-controlled tunability to map full intracellular phase diagrams, which dictate the concentrations at which phase separation occurs and the transition mechanism, in a protein sequence dependent manner. Surprisingly, both experiments and simulations show that while intracellular concentrations may be insufficient for global phase separation, sequestering protein ligands to slowly diffusing nucleation centers can move the cell into a different region of the phase diagram, resulting in localized phase separation. This diffusive capture mechanism liberates the cell from the constraints of global protein abundance and is likely exploited to pattern condensates associated with diverse biological processes. VIDEO ABSTRACT.


Assuntos
Materiais Biomiméticos , Citoplasma/metabolismo , Animais , Materiais Biomiméticos/farmacocinética , Materiais Biomiméticos/farmacologia , Células HEK293 , Células HeLa , Humanos , Camundongos , Microscopia de Fluorescência/métodos , Células NIH 3T3
2.
J Am Chem Soc ; 143(43): 18029-18040, 2021 11 03.
Artigo em Inglês | MEDLINE | ID: mdl-34664942

RESUMO

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.


Assuntos
Anticorpos/química , Materiais Biomiméticos/metabolismo , Catenanos/metabolismo , Fragmentos de Peptídeos/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Sequência de Aminoácidos , Animais , Materiais Biomiméticos/química , Materiais Biomiméticos/farmacocinética , Catenanos/química , Catenanos/farmacocinética , Linhagem Celular Tumoral , Feminino , Humanos , Camundongos Endogâmicos BALB C , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/farmacocinética , Estudo de Prova de Conceito , Domínios Proteicos , Engenharia de Proteínas , Estrutura Quaternária de Proteína , Receptor ErbB-2/metabolismo , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/farmacocinética , Proteína Supressora de Tumor p53/química , Proteína Supressora de Tumor p53/farmacocinética
3.
J Nanobiotechnology ; 19(1): 385, 2021 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-34809623

RESUMO

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.


Assuntos
Materiais Biomiméticos , Fosfatos de Cálcio , Cárie Dentária/metabolismo , Derivados da Hipromelose , Calcificação de Dente/efeitos dos fármacos , Animais , Materiais Biomiméticos/química , Materiais Biomiméticos/farmacocinética , Materiais Biomiméticos/farmacologia , Fosfatos de Cálcio/química , Fosfatos de Cálcio/farmacocinética , Fosfatos de Cálcio/farmacologia , Células Cultivadas , Dentina/efeitos dos fármacos , Humanos , Hidrogéis/química , Hidrogéis/farmacologia , Derivados da Hipromelose/química , Derivados da Hipromelose/farmacologia , Masculino , Camundongos , Sistemas de Liberação de Fármacos por Nanopartículas , Nanopartículas , Coelhos
4.
J Nanobiotechnology ; 19(1): 339, 2021 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-34689761

RESUMO

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.


Assuntos
Antineoplásicos , Materiais Biomiméticos , Membrana Celular/química , Lipossomos , Microambiente Tumoral/efeitos dos fármacos , Células A549 , Animais , Antineoplásicos/química , Antineoplásicos/farmacocinética , Antineoplásicos/farmacologia , Materiais Biomiméticos/química , Materiais Biomiméticos/farmacocinética , Materiais Biomiméticos/farmacologia , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Sistemas de Liberação de Medicamentos , Glicólise , Humanos , Lipossomos/química , Lipossomos/farmacocinética , Neoplasias Pulmonares/metabolismo , Camundongos , Camundongos Nus , Nanopartículas/química , Nanopartículas/metabolismo
5.
J Nanobiotechnology ; 19(1): 374, 2021 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-34789284

RESUMO

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.


Assuntos
Hiperplasia/metabolismo , Macrófagos/citologia , Sistemas de Liberação de Fármacos por Nanopartículas , Espécies Reativas de Oxigênio/metabolismo , Túnica Íntima , Animais , Materiais Biomiméticos/química , Materiais Biomiméticos/farmacocinética , Materiais Biomiméticos/farmacologia , Membrana Celular/química , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Sistemas de Liberação de Fármacos por Nanopartículas/química , Sistemas de Liberação de Fármacos por Nanopartículas/metabolismo , Sirolimo/química , Sirolimo/farmacocinética , Sirolimo/farmacologia , Túnica Íntima/efeitos dos fármacos , Túnica Íntima/metabolismo , Peixe-Zebra
6.
Nano Lett ; 20(3): 1637-1646, 2020 03 11.
Artigo em Inglês | MEDLINE | ID: mdl-32013452

RESUMO

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.


Assuntos
Materiais Biomiméticos , Neoplasias Encefálicas , Glioblastoma , Interferência de RNA , RNA Interferente Pequeno , Animais , Materiais Biomiméticos/química , Materiais Biomiméticos/farmacocinética , Materiais Biomiméticos/farmacologia , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/patologia , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Glioblastoma/tratamento farmacológico , Glioblastoma/metabolismo , Glioblastoma/patologia , Camundongos , Camundongos Nus , RNA Interferente Pequeno/química , RNA Interferente Pequeno/farmacocinética , RNA Interferente Pequeno/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Biomacromolecules ; 21(2): 688-700, 2020 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-31769678

RESUMO

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.


Assuntos
Adesivos/química , Materiais Biocompatíveis/farmacocinética , Materiais Biomiméticos/farmacocinética , Quitosana/análogos & derivados , Iridoides/química , Zinco/química , Adesivos/farmacocinética , Materiais Biocompatíveis/química , Materiais Biomiméticos/química , Biomimética/métodos , Células Cultivadas , Quitosana/química , Quitosana/farmacocinética , Polpa Dentária/efeitos dos fármacos , Polpa Dentária/metabolismo , Eritrócitos/efeitos dos fármacos , Eritrócitos/metabolismo , Humanos , Iridoides/farmacocinética , Teste de Materiais/métodos , Testes de Sensibilidade Microbiana/métodos , Alicerces Teciduais , Zinco/farmacocinética
8.
J Nanobiotechnology ; 18(1): 115, 2020 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-32819405

RESUMO

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.


Assuntos
Artrite Reumatoide/metabolismo , Materiais Biomiméticos , Dexametasona , Exossomos , Nanopartículas , Animais , Anti-Inflamatórios/química , Anti-Inflamatórios/farmacocinética , Artrite Reumatoide/patologia , Materiais Biomiméticos/química , Materiais Biomiméticos/farmacocinética , Colesterol/análogos & derivados , Colesterol/química , Dexametasona/química , Dexametasona/farmacocinética , Exossomos/química , Exossomos/metabolismo , Ácido Fólico/química , Articulações/metabolismo , Articulações/patologia , Masculino , Camundongos , Nanopartículas/química , Nanopartículas/metabolismo , Polietilenoglicóis/química , Células RAW 264.7
9.
J Nanobiotechnology ; 18(1): 71, 2020 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-32404183

RESUMO

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.


Assuntos
Doença de Alzheimer , Materiais Biomiméticos , Curcumina , Portadores de Fármacos , Nanopartículas/química , Animais , Apoptose/efeitos dos fármacos , Materiais Biomiméticos/química , Materiais Biomiméticos/farmacocinética , Barreira Hematoencefálica/metabolismo , Linhagem Celular , Curcumina/química , Curcumina/farmacocinética , Curcumina/farmacologia , Modelos Animais de Doenças , Portadores de Fármacos/química , Portadores de Fármacos/farmacocinética , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , Humanos , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos , Neurônios/metabolismo , Substâncias Protetoras/química , Substâncias Protetoras/farmacocinética , Substâncias Protetoras/farmacologia , Proteínas tau/metabolismo
10.
Nano Lett ; 19(7): 4470-4477, 2019 07 10.
Artigo em Inglês | MEDLINE | ID: mdl-31244234

RESUMO

Neutrophils are implicated in numerous inflammatory diseases, and especially in acute ischemic stroke (AIS). The unchecked migration of neutrophils into cerebral ischemic regions, and their subsequent release of reactive oxygen species, are considered the primary causes of reperfusion injury following AIS. Reducing the infiltration of inflammatory neutrophils may therefore be a useful therapy for AIS. Here, inspired by the specific cell-cell recognition that occurs between platelets and inflammatory neutrophils, we describe platelet-mimetic nanoparticles (PTNPs) that can be used to directly recognize, intervene, and monitor inflammatory neutrophils in the AIS treatment and therapeutic evaluation. We demonstrate that PTNPs, coloaded with piceatannol, a selective spleen tyrosine kinase inhibitor, and superparamagnetic iron oxide (SPIO), a T2 contrast agent, can successfully recognize adherent neutrophils via platelet membrane coating. The loaded piceatannol could then be delivered to adherent neutrophils and detach them into circulation, thus decreasing neutrophil infiltration and reducing infarct size. Moreover, when coupled with magnetic resonance imaging, internalized SPIO could be used to monitor the inflammatory neutrophils, associated with therapeutic effects, in real time. This approach is an innovative method for both the treatment and therapeutic evaluation of AIS, and provides new insights into how to treat and monitor neutrophil-associated diseases.


Assuntos
Materiais Biomiméticos , Plaquetas , Isquemia Encefálica , Rastreamento de Células , Meios de Contraste , Nanopartículas de Magnetita , Neutrófilos/metabolismo , Estilbenos , Acidente Vascular Cerebral , Animais , Materiais Biomiméticos/química , Materiais Biomiméticos/farmacocinética , Materiais Biomiméticos/farmacologia , Encéfalo/diagnóstico por imagem , Encéfalo/metabolismo , Isquemia Encefálica/diagnóstico por imagem , Isquemia Encefálica/metabolismo , Meios de Contraste/química , Meios de Contraste/farmacocinética , Meios de Contraste/farmacologia , Nanopartículas de Magnetita/química , Nanopartículas de Magnetita/uso terapêutico , Camundongos , Células RAW 264.7 , Estilbenos/química , Estilbenos/farmacocinética , Estilbenos/farmacologia , Acidente Vascular Cerebral/diagnóstico por imagem , Acidente Vascular Cerebral/metabolismo
11.
Blood ; 127(13): 1633-41, 2016 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-26626991

RESUMO

ACE910 is a recombinant humanized bispecific antibody that binds to activated factor IX and factor X and mimics the cofactor function of factor VIII (FVIII). This first-in-human study examined the safety, tolerability, pharmacokinetics (PK), and pharmacodynamics (PD) of ACE910 in healthy male adults. A total of 40 Japanese and 24 white subjects were randomized to receive a single subcutaneous injection of ACE910 (Japanese: 0.001, 0.01, 0.1, 0.3, or 1 mg/kg; white: 0.1, 0.3, or 1 mg/kg; n = 6 per dose group) or placebo (n = 2 per dose group). ACE910 exhibited a linear PK profile and had a half-life of ∼4 to 5 weeks. In FVIII-neutralized plasma, ACE910 shortened activated partial thromboplastin time and increased peak height of thrombin generation in a dose-dependent manner. All adverse events were nonserious and did not lead to any subject's withdrawal. Neither clinical findings nor laboratory abnormalities indicating hypercoagulability were observed. Two of 48 subjects receiving ACE910 (1 Japanese and 1 white) were positive for anti-ACE910 antibodies (anti-drug antibodies [ADAs]). One subject tested positive for ADAs both before and after ACE910 administration, whereas the other became ADA positive after receiving ACE910. The PK and PD profiles of ACE910 were similar in healthy Japanese and white subjects and suggest that ACE910 will be an effective and convenient prophylactic treatment of hemophilia A. This trial was registered at www.clinicaltrials.jp as #JapicCTI-121934.


Assuntos
Anticorpos Biespecíficos/administração & dosagem , Anticorpos Monoclonais Humanizados/administração & dosagem , Adulto , Anticorpos Biespecíficos/efeitos adversos , Anticorpos Biespecíficos/farmacocinética , Anticorpos Monoclonais Humanizados/efeitos adversos , Anticorpos Monoclonais Humanizados/farmacocinética , Materiais Biomiméticos/administração & dosagem , Materiais Biomiméticos/efeitos adversos , Materiais Biomiméticos/farmacocinética , Relação Dose-Resposta a Droga , Fator VIII/uso terapêutico , Voluntários Saudáveis , Humanos , Injeções Subcutâneas , Masculino , Dose Máxima Tolerável , Adulto Jovem
12.
J Labelled Comp Radiopharm ; 61(11): 857-863, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29972867

RESUMO

Discoidal lipid nanoparticles mimicking native high-density lipoproteins (HDL) are promising delivery vehicles of drugs and/or imaging agents. However, little is known about the in vivo biodistribution of such discoidal lipid nanoparticles compared to liposomes, clinically available spherical lipid nanoparticles. Recently, it has been reported that synthetic polymers instead of apolipoproteins can be complexed with phospholipid to form discoidal nanoparticles. In the present study, with the aim of developing phospholipid-synthetic polymer complexes for future clinical applications, the biodistribution of such particles in normal mice was investigated. Lipid nanoparticles comprising 1-palmitoyl-2-oleoyl-glycero-3-phosphocholine (POPC) and styrene maleic acid copolymer (SMA), having sizes similar to native HDL, were prepared using the freeze-sonication method. POPC-SMA complexes remained stable at 37°C for at least 3 days in buffer. By devising ways to avoid detrimental effects accompanied by pH reduction and nonspecific binding of 111 In to SMA, POPC-SMA complexes were successfully labeled with 111 In without affecting particle integrity. The biodistribution of POPC-SMA complexes in normal mice was similar to that of discoidal lipid nanoparticles composed of POPC and apolipoprotein A-I, the major protein constituent of native HDL. Unlike liposomes, the accumulation of POPC-SMA complexes in the spleen was low, suggesting that these complexes are not recognized as foreign substances. To the best of our knowledge, this is the first in vivo study of HDL-mimicking phospholipid-synthetic polymer complexes.


Assuntos
Materiais Biomiméticos/química , Materiais Biomiméticos/farmacocinética , Radioisótopos de Índio , Lipoproteínas HDL/metabolismo , Maleatos/química , Maleatos/farmacocinética , Fosfatidilcolinas/química , Poliestirenos/química , Poliestirenos/farmacocinética , Animais , Marcação por Isótopo , Masculino , Camundongos , Nanopartículas/química , Distribuição Tecidual
13.
AAPS PharmSciTech ; 19(6): 2629-2638, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29922876

RESUMO

The aim of this work was to predict the permeability of two model drugs, sulfamerazine (SMR) and indomethacin (INM), and to determine the effect on their apparent permeabilities by complexation with cyclodextrins and/or meglumine or incorporation in microemulsions. Permeation experiments were performed using two-chamber diffusion cells with a new composition of bio-mimetic membrane composed of 80% of Lipoid® S100 and 20% of cholesterol in n-octanol 10% w/w solution, at 37 ± 0.5°C and 14,000 rpm. The predictive capacity of the permeability of passive diffusion absorbed compounds was evaluated using 20 drug standards and showed an exponential correlation between the apparent permeability coefficients (Papp) and the fraction absorbed percentages in humans (Fa%), with an R2 value of 0.67942 and a constant value of - 4.1 ± 0.8. SMR and INM were classified as Class II and I, respectively, according to the Biopharmaceutical Classification System. These drugs were complexed and incorporated in microemulsions. The Fa% from all the drug products was higher than 90%. SMR in the complexes and both drugs in microemulsions were classified as highly soluble. Thus, SMR and INM incorporated in these pharmaceutical products could be classified as Class I.


Assuntos
Materiais Biomiméticos/química , Materiais Biomiméticos/farmacocinética , Emulsões/química , Emulsões/farmacocinética , Membranas Artificiais , Biomimética/métodos , Ciclodextrinas/química , Ciclodextrinas/farmacocinética , Difusão , Indometacina/química , Indometacina/farmacocinética , Permeabilidade/efeitos dos fármacos , Solubilidade
14.
Exp Brain Res ; 235(10): 3033-3048, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28725925

RESUMO

Systemic administration of a Connexin43 mimetic peptide, Peptide5, has been shown to reduce secondary tissue damage and improve functional recovery after spinal cord injury (SCI). This study investigated safety measures and potential off-target effects of Peptide5 systemic administration. Rats were subjected to a mild contusion SCI using the New York University impactor. One cohort was injected intraperitoneally with a single dose of fluorescently labelled Peptide5 and euthanised at 2 or 4 h post-injury for peptide distribution analysis. A second cohort received intraperitoneal injections of Peptide5 or a scrambled peptide and was culled at 8 or 24 h post-injury for the analysis of connexin proteins and systemic cytokine profile. We found that Peptide5 did not cross the blood-spinal cord barrier in control animals, but reached the lesion area in the spinal cord-injured animals without entering non-injured tissue. There was no evidence that the systemic administration of Peptide5 modulates Connexin43 protein expression or hemichannel closure in the heart and lung tissue of SCI animals. The expression levels of other major connexin proteins including Connexin30 in astrocytes, Connexin36 in neurons and Connexin47 in oligodendrocytes were also unaltered by systemic delivery of Peptide5 in either the injured or non-injured spinal cords. In addition, systemic delivery of Peptide5 had no significant effect on the plasma levels of cytokines, chemokines or growth factors. These data indicate that the systemic delivery of Peptide5 is unlikely to cause any off-target or adverse effects and may thus be a safe treatment option for traumatic SCI.


Assuntos
Materiais Biomiméticos/farmacologia , Conexina 43/farmacologia , Traumatismos da Medula Espinal/tratamento farmacológico , Animais , Materiais Biomiméticos/administração & dosagem , Materiais Biomiméticos/efeitos adversos , Materiais Biomiméticos/farmacocinética , Conexina 43/administração & dosagem , Conexina 43/efeitos adversos , Conexina 43/farmacocinética , Modelos Animais de Doenças , Feminino , Ratos , Ratos Sprague-Dawley
15.
Nanotechnology ; 27(8): 085106, 2016 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-26820630

RESUMO

Suppression of the reticuloendothelial system (RES) uptake is one of the most challenging tasks in nanomedicine. Coating stratagems using polymers, such as poly(ethylene glycol) (PEG), have led to great success in this respect. Nevertheless, recent observations of immunological response toward these synthetic polymers have triggered a search for better alternatives. In this work, natural red blood cell (RBC) membranes are camouflaged on the surface of Fe3O4 nanoparticles for reducing the RES uptake. In vitro macrophage uptake, in vivo biodistribution and pharmacokinetic studies demonstrate that the RBC membrane is a superior alternative to the current gold standard PEG for nanoparticle 'stealth'. Furthermore, we systematically investigate the in vivo potential toxicity of RBC membrane-coated nanoparticles by blood biochemistry, whole blood panel examination and histology analysis based on animal models. The combination of synthetic nanoparticles and natural cell membranes embodies a novel and biomimetic nanomaterial design strategy and presents a compelling property of functional materials for a broad range of biomedical applications.


Assuntos
Materiais Biomiméticos/farmacocinética , Portadores de Fármacos/farmacocinética , Membrana Eritrocítica/química , Óxido Ferroso-Férrico/farmacocinética , Nanopartículas Metálicas/química , Animais , Transporte Biológico , Materiais Biomiméticos/síntese química , Linhagem Celular , Portadores de Fármacos/síntese química , Ferro/análise , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Nanopartículas Metálicas/administração & dosagem , Camundongos , Camundongos Endogâmicos ICR , Sistema Fagocitário Mononuclear/fisiologia , Polietilenoglicóis/química , Espectrofotometria Atômica
16.
Anal Chem ; 86(3): 1534-42, 2014 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-24397447

RESUMO

MRX34, a microRNA (miRNA)-based therapy for cancer, has recently entered clinical trials as the first clinical candidate in its class. It is a liposomal nanoparticle loaded with a synthetic mimic of the tumor suppressor miRNA miR-34a as the active pharmaceutical ingredient. To understand the pharmacokinetic properties of the drug and to rationalize an optimal dosing regimen in the clinic, a method is needed to quantitatively detect the miRNA mimic. Here, we report the development and qualification of a quantitative reverse transcription-polymerase chain reaction (qRT-PCR) assay in support of pharmacokinetic and toxicokinetic assessments in the nonhuman primate. Detection and quantification were performed on total ribonucleic acid (RNA) isolated from whole blood. The qualified range of the standard curve spans 6 orders of magnitude from 2.5 × 10(-7) to 2.5 × 10(-1) ng per reverse transcription (RT) reaction, corresponding to an estimated blood concentration from 6.2 × 10(-5) to 6.2 × 10(1) ng/mL. Our results demonstrate that endogenous as well as the exogenous miR-34a can be accurately and precisely quantified. The assay was used to establish the pharmacokinetic profile of MRX34, showing a favorable residence time and exposure of the miRNA mimic in whole blood from nonhuman primates.


Assuntos
Materiais Biomiméticos/análise , Macaca fascicularis/sangue , MicroRNAs/sangue , Animais , Materiais Biomiméticos/farmacocinética , Materiais Biomiméticos/uso terapêutico , Calibragem , Estabilidade de Medicamentos , Armazenamento de Medicamentos , Congelamento , Limite de Detecção , MicroRNAs/uso terapêutico
17.
J Nanosci Nanotechnol ; 14(1): 115-25, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24730254

RESUMO

Nanoparticles are diligently crafted with exacting control over size, shape, and composition. The pristine nanoparticles are rigorously characterized in vitro by numerous physical and materials science techniques. Immediately after being exposed to body fluids, nanoparticles interact with a heterogeneous mixture of proteins and numerous different cell types that modify the nanoparticle surface and affect their bioavailability. Understanding the mechanisms behind the recognition and elimination of these modified nanoparticles is the key to the successful translation of nanomaterials from preclinical to clinical applications. This paper reviews the anatomy of the primary organs (kidney, liver, and spleen) responsible for nanoparticle bioelimination and the components of the innate immune system (complement system and scavenger receptors) that indirectly and directly recognize nanoparticles as foreign. Recent results using PEG as a steric barrier, generating biomimetic nanoparticles, and the effect of nanoparticle material properties to increase the bioavailability of nanoparticles are presented.


Assuntos
Materiais Biomiméticos/química , Materiais Biomiméticos/farmacocinética , Desenho de Fármacos , Rim/metabolismo , Fígado/metabolismo , Nanopartículas/química , Baço/metabolismo , Animais , Disponibilidade Biológica , Humanos , Rim/química , Fígado/química , Baço/química
18.
J Nanosci Nanotechnol ; 14(6): 4007-13, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24738344

RESUMO

Extracellular matrix (ECM) plays a fundamental role in regulating cell attachment, proliferation, migration and differentiation. Both synthetic and biologically derived materials have been explored as an ECM in regenerative medicine and tissue engineering. To biomimick the extracellular matrix, we combined the advantages of the biological properties of nanofibrous scaffolds and the fusion protein to apply for the culture of human mesenchymal stem cells in vitro. In this study, we fabricated well random-oriented/aligned nanofibrous scaffolds with PCL, modified with hE-cadherin-Fc fusion protein and studied the synergistic effect of the scaffolds. The random-oriented/aligned architecture was observed in the nanofibrous scaffolds by SEM. XPS and WCA measurements evidenced that hE-cadherin-Fc was successfully modified on the PCL nanofibrous scaffolds and hydrophilicity of the scaffolds was well improved after fusion protein coating. The hE-cadherin-Fc modified markedly promoted the adhesion and proliferation of hMSCs and guided hMSCs to a spindlier morphology compared with unmodified nanofibrous scaffolds. Furthermore, hMSCs on the hE-cadherin-Fc-coated nanofibrous scaffolds also had differentiation potential. These results suggested that the combination of PCL nanofibrous scaffolds and hE-cadherin-Fc fusion protein may be a promising artificial ECM for the behavior of hMSCs in vitro.


Assuntos
Caderinas/farmacocinética , Moléculas de Adesão Celular/farmacocinética , Proteínas da Matriz Extracelular/farmacocinética , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/fisiologia , Poliésteres/química , Alicerces Teciduais , Materiais Biomiméticos/química , Materiais Biomiméticos/farmacocinética , Caderinas/química , Caderinas/genética , Adesão Celular/fisiologia , Moléculas de Adesão Celular/química , Moléculas de Adesão Celular/genética , Diferenciação Celular/fisiologia , Proliferação de Células , Células Cultivadas , Materiais Revestidos Biocompatíveis/química , Materiais Revestidos Biocompatíveis/farmacocinética , Desenho de Equipamento , Análise de Falha de Equipamento , Proteínas da Matriz Extracelular/química , Humanos , Teste de Materiais , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/farmacocinética , Engenharia Tecidual/instrumentação
19.
Proc Natl Acad Sci U S A ; 108(27): 10980-5, 2011 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-21690347

RESUMO

Efforts to extend nanoparticle residence time in vivo have inspired many strategies in particle surface modifications to bypass macrophage uptake and systemic clearance. Here we report a top-down biomimetic approach in particle functionalization by coating biodegradable polymeric nanoparticles with natural erythrocyte membranes, including both membrane lipids and associated membrane proteins for long-circulating cargo delivery. The structure, size and surface zeta potential, and protein contents of the erythrocyte membrane-coated nanoparticles were verified using transmission electron microscopy, dynamic light scattering, and gel electrophoresis, respectively. Mice injections with fluorophore-loaded nanoparticles revealed superior circulation half-life by the erythrocyte-mimicking nanoparticles as compared to control particles coated with the state-of-the-art synthetic stealth materials. Biodistribution study revealed significant particle retention in the blood 72 h following the particle injection. The translocation of natural cellular membranes, their associated proteins, and the corresponding functionalities to the surface of synthetic particles represents a unique approach in nanoparticle functionalization.


Assuntos
Sistemas de Liberação de Medicamentos , Membrana Eritrocítica/química , Nanopartículas/administração & dosagem , Nanopartículas/química , Animais , Materiais Biomiméticos/administração & dosagem , Materiais Biomiméticos/química , Materiais Biomiméticos/farmacocinética , Corantes Fluorescentes/administração & dosagem , Ácido Láctico/química , Masculino , Camundongos , Camundongos Endogâmicos ICR , Nanopartículas/ultraestrutura , Tamanho da Partícula , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Distribuição Tecidual
20.
Int J Nanomedicine ; 19: 5273-5295, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38859952

RESUMO

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.


Assuntos
Artemisininas , Disponibilidade Biológica , Lipossomos , Animais , Lipossomos/química , Lipossomos/farmacocinética , Células CACO-2 , Humanos , Administração Oral , Artemisininas/farmacocinética , Artemisininas/química , Artemisininas/administração & dosagem , Absorção Intestinal/efeitos dos fármacos , Masculino , Distribuição Tecidual , Tamanho da Partícula , Camundongos , Sistema Linfático/metabolismo , Sistema Linfático/efeitos dos fármacos , Ratos Sprague-Dawley , Ratos , Materiais Biomiméticos/farmacocinética , Materiais Biomiméticos/química , Mucosa Intestinal/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA