Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Nature ; 526(7571): 118-21, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-26374997

RESUMO

Development of functional nanoparticles can be encumbered by unanticipated material properties and biological events, which can affect nanoparticle effectiveness in complex, physiologically relevant systems. Despite the advances in bottom-up nanoengineering and surface chemistry, reductionist functionalization approaches remain inadequate in replicating the complex interfaces present in nature and cannot avoid exposure of foreign materials. Here we report on the preparation of polymeric nanoparticles enclosed in the plasma membrane of human platelets, which are a unique population of cellular fragments that adhere to a variety of disease-relevant substrates. The resulting nanoparticles possess a right-side-out unilamellar membrane coating functionalized with immunomodulatory and adhesion antigens associated with platelets. Compared to uncoated particles, the platelet membrane-cloaked nanoparticles have reduced cellular uptake by macrophage-like cells and lack particle-induced complement activation in autologous human plasma. The cloaked nanoparticles also display platelet-mimicking properties such as selective adhesion to damaged human and rodent vasculatures as well as enhanced binding to platelet-adhering pathogens. In an experimental rat model of coronary restenosis and a mouse model of systemic bacterial infection, docetaxel and vancomycin, respectively, show enhanced therapeutic efficacy when delivered by the platelet-mimetic nanoparticles. The multifaceted biointerfacing enabled by the platelet membrane cloaking method provides a new approach in developing functional nanoparticles for disease-targeted delivery.


Assuntos
Antibacterianos/administração & dosagem , Plaquetas/citologia , Membrana Celular/metabolismo , Sistemas de Liberação de Medicamentos/métodos , Nanopartículas/administração & dosagem , Nanopartículas/química , Adesividade Plaquetária , Animais , Antibacterianos/farmacocinética , Vasos Sanguíneos/citologia , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/patologia , Colágeno/química , Colágeno/imunologia , Ativação do Complemento/imunologia , Reestenose Coronária/sangue , Reestenose Coronária/tratamento farmacológico , Reestenose Coronária/metabolismo , Modelos Animais de Doenças , Docetaxel , Humanos , Macrófagos/imunologia , Masculino , Camundongos , Polímeros/química , Ratos , Ratos Sprague-Dawley , Infecções Estafilocócicas/sangue , Infecções Estafilocócicas/tratamento farmacológico , Infecções Estafilocócicas/metabolismo , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/citologia , Staphylococcus aureus/metabolismo , Taxoides/administração & dosagem , Taxoides/farmacocinética , Lipossomas Unilamelares/química , Vancomicina/administração & dosagem , Vancomicina/farmacocinética
2.
Proc Natl Acad Sci U S A ; 111(49): 17600-5, 2014 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-25422427

RESUMO

Helicobacter pylori infection is marked by a vast prevalence and strong association with various gastric diseases, including gastritis, peptic ulcers, and gastric cancer. Because of the rapid emergence of H. pylori strains resistant to existing antibiotics, current treatment regimens show a rapid decline of their eradication rates. Clearly, novel antibacterial strategies against H. pylori are urgently needed. Here, we investigated the in vivo therapeutic potential of liposomal linolenic acid (LipoLLA) for the treatment of H. pylori infection. The LipoLLA formulation with a size of ∼ 100 nm was prone to fusion with bacterial membrane, thereby directly releasing a high dose of linolenic acids into the bacterial membrane. LipoLLA penetrated the mucus layer of mouse stomach, and a significant portion of the administered LipoLLA was retained in the stomach lining up to 24 h after the oral administration. In vivo tests further confirmed that LipoLLA was able to kill H. pylori and reduce bacterial load in the mouse stomach. LipoLLA treatment was also shown to reduce the levels of proinflammatory cytokines including interleukin 1ß, interleukin 6, and tumor necrosis factor alpha, which were otherwise elevated because of the H. pylori infection. Finally, a toxicity test demonstrated excellent biocompatibility of LipoLLA to normal mouse stomach. Collectively, results from this study indicate that LipoLLA is a promising, effective, and safe therapeutic agent for the treatment of H. pylori infection.


Assuntos
Infecções por Helicobacter/tratamento farmacológico , Helicobacter pylori/efeitos dos fármacos , Inflamação/tratamento farmacológico , Lipossomos/química , Ácido alfa-Linolênico/administração & dosagem , Animais , Antibacterianos , Carcinoma/tratamento farmacológico , Carcinoma/microbiologia , Linhagem Celular Tumoral , Membrana Celular/efeitos dos fármacos , Análise Custo-Benefício , Citocinas/metabolismo , Relação Dose-Resposta a Droga , Farmacorresistência Bacteriana , Ácidos Graxos não Esterificados/química , Mucosa Gástrica/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Nanomedicina , Células-Tronco , Estômago/microbiologia , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/microbiologia
3.
Angew Chem Int Ed Engl ; 56(8): 2156-2161, 2017 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-28105785

RESUMO

The highly acidic gastric environment creates a physiological barrier for using therapeutic drugs in the stomach. While proton pump inhibitors have been widely used for blocking acid-producing enzymes, this approach can cause various adverse effects. Reported herein is a new microdevice, consisting of magnesium-based micromotors which can autonomously and temporally neutralize gastric acid through efficient chemical propulsion in the gastric fluid by rapidly depleting the localized protons. Coating these micromotors with a cargo-containing pH-responsive polymer layer leads to autonomous release of the encapsulated payload upon gastric-acid neutralization by the motors. Testing in a mouse model demonstrate that these motors can safely and rapidly neutralize gastric acid and simultaneously release payload without causing noticeable acute toxicity or affecting the stomach function, and the normal stomach pH is restored within 24 h post motor administration.


Assuntos
Preparações de Ação Retardada/química , Ácido Gástrico/química , Magnésio/química , Polímeros/química , Animais , Liberação Controlada de Fármacos , Corantes Fluorescentes/administração & dosagem , Ouro/química , Concentração de Íons de Hidrogênio , Camundongos , Ácidos Polimetacrílicos/química , Rodaminas/administração & dosagem
4.
Small ; 11(34): 4309-13, 2015 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-26044721

RESUMO

The synthesis of biomimetic hydrogel nanoparticles coated with a natural cell membrane is described. Compared to the existing strategy of wrapping cell membranes onto pre-formed nanoparticle substrates, this new approach forms the cell membrane-derived vesicles first, followed by growing nanoparticle cores in situ. It adds significant controllability over the nanoparticle properties and opens unique opportunities for a broad range of biomedical applications.


Assuntos
Membrana Eritrocítica/metabolismo , Polietilenoglicóis/síntese química , Polietilenoimina/síntese química , Polimerização , Animais , Óxidos N-Cíclicos/síntese química , Óxidos N-Cíclicos/química , Membrana Eritrocítica/ultraestrutura , Masculino , Camundongos Endogâmicos ICR , Nanogéis , Polietilenoglicóis/química , Polietilenoimina/química
5.
Small ; 9(4): 511-7, 2013 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-23109494

RESUMO

Spontaneous formation of heterogeneous patches on the surface of lipid-based nanoparticles (NPs) and microparticles (MPs) due to the segregation of two different functional groups. Patch formation is observed when tracing the functional groups with quantum dots, gold nanoparticles, and fluorescent dyes. This discovery could have important implications for the future design of self-assembled NPs and MPs for different biomedical applications.


Assuntos
Lipídeos/química , Nanopartículas/química , Nanotecnologia/métodos , Polímeros/química , Tamanho da Partícula , Propriedades de Superfície
6.
Langmuir ; 29(39): 12228-33, 2013 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-23987129

RESUMO

We report a novel pH-responsive gold nanoparticle-stabilized liposome system for gastric antimicrobial delivery. By adsorbing small chitosan-modified gold nanoparticles (diameter ~10 nm) onto the outer surface of negatively charged phospholipid liposomes (diameter ~75 nm), we show that at gastric pH the liposomes have excellent stability with limited fusion ability and negligible cargo releases. However, when the stabilized liposomes are present in an environment with neutral pH, the gold stabilizers detach from the liposomes, resulting in free liposomes that can actively fuse with bacterial membranes. Using Helicobacter pylori as a model bacterium and doxycycline as a model antibiotic, we demonstrate such pH-responsive fusion activity and drug release profile of the nanoparticle-stabilized liposomes. Particularly, at neutral pH the gold nanoparticles detach, and thus the doxycycline-loaded liposomes rapidly fuse with bacteria and cause superior bactericidal efficacy as compared to the free doxycycline counterpart. Our results suggest that the reported liposome system holds a substantial potential for gastric drug delivery; it remains inactive (stable) in the stomach lumen but actively interacts with bacteria once it reaches the mucus layer of the stomach where the bacteria may reside.


Assuntos
Antibacterianos/farmacologia , Doxiciclina/farmacologia , Sistemas de Liberação de Medicamentos , Helicobacter pylori/efeitos dos fármacos , Lipossomos/química , Estômago/efeitos dos fármacos , Antibacterianos/química , Membrana Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Doxiciclina/química , Ouro/química , Helicobacter pylori/citologia , Humanos , Concentração de Íons de Hidrogênio , Lipossomos/síntese química , Nanopartículas Metálicas/química , Testes de Sensibilidade Microbiana , Tamanho da Partícula , Estômago/química , Estômago/microbiologia , Relação Estrutura-Atividade , Propriedades de Superfície
7.
mSphere ; 4(3)2019 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-31092603

RESUMO

Intraocular infections are prevalent after traumatic injuries or after common ocular surgeries. Infections cause inflammation that can damage the retina and architecture of the eye, often resulting in poor visual outcomes. Severe cases may result in blindness or require enucleation of the eye. Treatments for intraocular infections include intravitreal antibiotics and corticosteroids or surgical vitrectomy in serious cases. The increase in multidrug-resistant infections calls for novel treatment options. In the present study, a biomimetic erythrocyte-derived nanosponge was tested for the ability to neutralize pore-forming toxins from the most frequent Gram-positive bacterial causes of intraocular infections (Staphylococcus aureus, Enterococcus faecalis, Streptococcus pneumoniae, and Bacillus cereus). Nanosponge pretreatment of supernatants reduced hemolytic activity in vitro. In a murine sterile endophthalmitis model, nanosponge pretreatment of injected supernatants resulted in greater retinal function and less ocular pathology compared to that in eyes injected with untreated supernatants from all pathogens except methicillin-resistant S. aureus In a murine bacterial endophthalmitis model, treatment with gatifloxacin and gatifloxacin-nanosponges reduced intraocular bacterial burdens, except in the case of methicillin-sensitive S. aureus For all pathogens, eyes in both treatment groups showed decreased ocular pathology and inflammation. Overall, reductions in retinal function loss afforded by gatifloxacin-nanosponge treatment were significant for E. faecalis, S. pneumoniae, and methicillin-resistant S. aureus but not for B. cereus and methicillin-sensitive S. aureus These results suggest that clinical improvements in intraocular infections following nanosponge treatment were dependent on the complexity and types of toxins produced. Nanosponges might serve as an adjunctive therapy for the treatment of ocular infections.IMPORTANCE Endophthalmitis is a blinding consequence of bacterial invasion of the interior of the eye. Because of increases in the numbers of ocular surgeries and intraocular injections, the incidence of endophthalmitis is steadily increasing. Staphylococcus aureus, Enterococcus faecalis, Streptococcus pneumoniae, and Bacillus cereus are leading causes of infection following ocular procedures and trauma and are increasingly more difficult to treat due to multidrug resistance. Each of these pathogens produces pore-forming toxins that contribute to the pathogenesis of endophthalmitis. Treatment of these infections with antibiotics alone is insufficient to prevent damage to the retina and vision loss. Therefore, novel therapeutics are needed that include agents that neutralize bacterial pore-forming toxins. Here, we demonstrate that biomimetic nanosponges neutralize pore-forming toxins from these ocular pathogens and aid in preserving retinal function. Nanosponges may represent a new form of adjunct antitoxin therapy for serious potentially blinding intraocular infections.


Assuntos
Toxinas Bacterianas/antagonistas & inibidores , Materiais Biomiméticos , Infecções Oculares Bacterianas/tratamento farmacológico , Nanoestruturas/uso terapêutico , Animais , Eritrócitos/química , Gatifloxacina/uso terapêutico , Infecções por Bactérias Gram-Positivas/tratamento farmacológico , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Nanoestruturas/química , Nanotecnologia , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/química , Polímeros/química , Coelhos , Infecções Estafilocócicas/tratamento farmacológico , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/efeitos dos fármacos
8.
ACS Nano ; 12(1): 109-116, 2018 01 23.
Artigo em Inglês | MEDLINE | ID: mdl-29216423

RESUMO

Cardiovascular disease represents one of the major causes of death across the global population. Atherosclerosis, one of its most common drivers, is characterized by the gradual buildup of arterial plaque over time, which can ultimately lead to life-threatening conditions. Given the impact of the disease on public health, there is a great need for effective and noninvasive imaging modalities that can provide valuable information on its biological underpinnings during development. Here, we leverage the role of platelets in atherogenesis to design nanocarriers capable of targeting multiple biological elements relevant to plaque development. Biomimetic nanoparticles are prepared by coating platelet membrane around a synthetic nanoparticulate core, the product of which is capable of interacting with activated endothelium, foam cells, and collagen. The effects are shown to be exclusive to platelet membrane-coated nanoparticles. These biomimetic nanocarriers are not only capable of efficiently localizing to well-developed atherosclerotic plaque, but can also target subclinical regions of arteries susceptible to plaque formation. Using a commonly employed magnetic resonance imaging contrast agent, live detection is demonstrated using an animal model of atherosclerosis. Ultimately, this strategy may be leveraged to better assess the development of atherosclerosis, offering additional information to help clinicians better manage the disease.


Assuntos
Aterosclerose/diagnóstico por imagem , Aterosclerose/metabolismo , Plaquetas/metabolismo , Membrana Celular/metabolismo , Materiais Revestidos Biocompatíveis/metabolismo , Nanopartículas/metabolismo , Animais , Aterosclerose/patologia , Materiais Biomiméticos/metabolismo , Linhagem Celular , Células Espumosas/metabolismo , Células Espumosas/patologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Imageamento por Ressonância Magnética/métodos , Camundongos , Camundongos Endogâmicos C57BL , Nanopartículas/ultraestrutura , Nanotecnologia/métodos , Imagem Óptica/métodos , Placa Aterosclerótica/diagnóstico por imagem , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/patologia
9.
ACS Appl Mater Interfaces ; 8(28): 18367-74, 2016 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-27352845

RESUMO

Effective antibacterial treatment at the infection site associated with high shear forces remains challenging, owing largely to the lack of durably adhesive and safe delivery platforms that can enable localized antibiotic accumulation against bacterial colonization. Inspired by delivery systems mimicking marine mussels for adhesion, herein, we developed a bioadhesive nanoparticle-hydrogel hybrid (NP-gel) to enhance localized antimicrobial drug delivery. Antibiotics were loaded into polymeric nanoparticles and then embedded into a 3D hydrogel network that confers adhesion to biological surfaces. The combination of two distinct delivery platforms, namely, nanoparticles and hydrogel, allows the hydrogel network properties to be independently tailored for adhesion while maintaining controlled and prolonged antibiotic release profile from the nanoparticles. The bioadhesive NP-gel developed here showed superior adhesion and antibiotic retention under high shear stress on a bacterial film, a mammalian cell monolayer, and mouse skin tissue. Under a flow environment, the NP-gel inhibited the formation of an Escherichia coli bacterial film. When applied on mouse skin tissue for 7 consecutive days, the NP-gel did not generate any observable skin reaction or toxicity, implying its potential as a safe and effective local delivery platform against microbial infections.


Assuntos
Antibacterianos/administração & dosagem , Antibacterianos/química , Sistemas de Liberação de Medicamentos/métodos , Hidrogéis/química , Nanopartículas/química , Acrilamidas/química , Adesivos/administração & dosagem , Adesivos/química , Animais , Biofilmes/efeitos dos fármacos , Ciprofloxacina/administração & dosagem , Ciprofloxacina/química , Dopamina/química , Sistemas de Liberação de Medicamentos/instrumentação , Escherichia coli/efeitos dos fármacos , Células HEK293 , Humanos , Hidrogéis/administração & dosagem , Ácido Láctico/administração & dosagem , Ácido Láctico/química , Camundongos , Camundongos Endogâmicos ICR , Nanopartículas/administração & dosagem , Ácido Poliglicólico/administração & dosagem , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Pele/efeitos dos fármacos
10.
Biomaterials ; 111: 116-123, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27728811

RESUMO

Immune thrombocytopenia purpura (ITP) is characterized by the production of pathological autoantibodies that cause reduction in platelet counts. The disease can have serious medical consequences, leading to uncontrolled bleeding that can be fatal. Current widely used therapies for the treatment of ITP are non-specific and can, at times, result in complications that are more burdensome than the disease itself. In the present study, the use of platelet membrane-coated nanoparticles (PNPs) as a platform for the specific clearance of anti-platelet antibodies is explored. The nanoparticles, whose outer layer displays the full complement of native platelet surface proteins, act as decoys that strongly bind pathological anti-platelet antibodies in order to minimize disease burden. Here, we study the antibody binding properties of PNPs and assess the ability of the nanoparticles to neutralize antibody activity both in vitro and in vivo. Ultimately, we leverage the neutralization capacity of PNPs to therapeutically treat a murine model of antibody-induced thrombocytopenia and demonstrate considerable efficacy as shown in a bleeding time assay. PNPs represent a promising platform for the specific treatment of antibody-mediated immune thrombocytopenia by acting as an alternative target for anti-platelet antibodies, thus preserving circulating platelets with the potential of leaving broader immune function intact.


Assuntos
Autoanticorpos/imunologia , Materiais Revestidos Biocompatíveis/administração & dosagem , Nanopartículas/administração & dosagem , Glicoproteínas da Membrana de Plaquetas/administração & dosagem , Púrpura Trombocitopênica Idiopática/tratamento farmacológico , Púrpura Trombocitopênica Idiopática/imunologia , Animais , Materiais Revestidos Biocompatíveis/química , Masculino , Camundongos , Nanopartículas/química , Glicoproteínas da Membrana de Plaquetas/química , Resultado do Tratamento
11.
Nanoscale ; 8(30): 14411-9, 2016 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-27411852

RESUMO

Lipid-polymer hybrid nanoparticles, consisting of a polymeric core coated by a layer of lipids, are a class of highly scalable, biodegradable nanocarriers that have shown great promise in drug delivery applications. Here, we demonstrate the facile synthesis of ultra-small, sub-25 nm lipid-polymer hybrid nanoparticles using an adapted nanoprecipitation approach and explore their utility for targeted delivery of a model chemotherapeutic. The fabrication process is first optimized to produce a monodisperse population of particles that are stable under physiological conditions. It is shown that these ultra-small hybrid nanoparticles can be functionalized with a targeting ligand on the surface and loaded with drug inside the polymeric matrix. Further, the in vivo fate of the nanoparticles after intravenous injection is characterized by examining the blood circulation and biodistribution. In a final proof-of-concept study, targeted ultra-small hybrid nanoparticles loaded with the cancer drug docetaxel are used to treat a mouse tumor model and demonstrate improved efficacy compared to a clinically available formulation of the drug. The ability to synthesize a significantly smaller version of the established lipid-polymer hybrid platform can ultimately enhance its applicability across a wider range of applications.


Assuntos
Sistemas de Liberação de Medicamentos , Lipídeos/química , Nanopartículas , Neoplasias Experimentais/tratamento farmacológico , Animais , Linhagem Celular Tumoral , Docetaxel , Feminino , Masculino , Camundongos , Camundongos Nus , Polímeros , Taxoides/administração & dosagem , Distribuição Tecidual
13.
ACS Nano ; 9(1): 117-23, 2015 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-25549040

RESUMO

Artificial micromotors, operating on locally supplied fuels and performing complex tasks, offer great potential for diverse biomedical applications, including autonomous delivery and release of therapeutic payloads and cell manipulation. Various types of synthetic motors, utilizing different propulsion mechanisms, have been fabricated to operate in biological matrices. However, the performance of these man-made motors has been tested exclusively under in vitro conditions (outside the body); their behavior and functionalities in an in vivo environment (inside the body) remain unknown. Herein, we report an in vivo study of artificial micromotors in a living organism using a mouse model. Such in vivo evaluation examines the distribution, retention, cargo delivery, and acute toxicity profile of synthetic motors in mouse stomach via oral administration. Using zinc-based micromotors as a model, we demonstrate that the acid-driven propulsion in the stomach effectively enhances the binding and retention of the motors as well as of cargo payloads on the stomach wall. The body of the motors gradually dissolves in the gastric acid, autonomously releasing their carried payloads, leaving nothing toxic behind. This work is anticipated to significantly advance the emerging field of nano/micromotors and to open the door to in vivo evaluation and clinical applications of these synthetic motors.


Assuntos
Portadores de Fármacos/química , Portadores de Fármacos/metabolismo , Transferência de Energia , Mucosa Gástrica/metabolismo , Microtecnologia/métodos , Movimento (Física) , Animais , Compostos Bicíclicos Heterocíclicos com Pontes/química , Portadores de Fármacos/toxicidade , Ouro/química , Masculino , Nanopartículas Metálicas/química , Camundongos , Polímeros/química , Zinco/química
14.
ACS Nano ; 8(3): 2900-7, 2014 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-24483239

RESUMO

Adsorbing small charged nanoparticles onto the outer surfaces of liposomes has become an effective strategy to stabilize liposomes against fusion prior to "seeing" target bacteria, yet allow them to fuse with the bacteria upon arrival at the infection sites. As a result, nanoparticle-stabilized liposomes have become an emerging drug delivery platform for treatment of various bacterial infections. To facilitate the translation of this platform for clinical tests and uses, herein we integrate nanoparticle-stabilized liposomes with hydrogel technology for more effective and sustained topical drug delivery. The hydrogel formulation not only preserves the structural integrity of the nanoparticle-stabilized liposomes, but also allows for controllable viscoeleasticity and tunable liposome release rate. Using Staphylococcus aureus bacteria as a model pathogen, we demonstrate that the hydrogel formulation can effectively release nanoparticle-stabilized liposomes to the bacterial culture, which subsequently fuse with bacterial membrane in a pH-dependent manner. When topically applied onto mouse skin, the hydrogel formulation does not generate any observable skin toxicity within a 7-day treatment. Collectively, the hydrogel containing nanoparticle-stabilized liposomes hold great promise for topical applications against various microbial infections.


Assuntos
Anti-Infecciosos/administração & dosagem , Portadores de Fármacos/química , Ouro/química , Hidrogel de Polietilenoglicol-Dimetacrilato/química , Lipossomos/química , Nanopartículas Metálicas/química , Administração Tópica , Adsorção , Animais , Anti-Infecciosos/química , Portadores de Fármacos/toxicidade , Concentração de Íons de Hidrogênio , Lipossomos/toxicidade , Camundongos , Pele/efeitos dos fármacos
15.
Adv Healthc Mater ; 2(10): 1322-8, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23495239

RESUMO

Propionibacterium acnes (P. acnes) is a Gram-positive bacterium strongly associated with acne infection. While many antimicrobial agents have been used in clinic to treat acne infection by targeting P. acnes, these existing anti-acne agents usually produce considerable side effects. Herein, the development and evaluation of liposomal lauric acids (LipoLA) is reported as a new, effective and safe therapeutic agent for the treatment of acne infection. By incorporating lauric acids into the lipid bilayer of liposomes, it is observed that the resulting LipoLA readily fuse with bacterial membranes, causing effective killing of P. acnes by disrupting bacterial membrane structures. Using a mouse ear model, we demonstrated that the bactericidal property of LipoLA against P. acne is well preserved at physiological conditions. Topically applying LipoLA in a gel form onto the infectious sites leads to eradication of P. acnes bacteria in vivo. Further skin toxicity studies show that LipoLA does not induce acute toxicity to normal mouse skin, while benzoyl peroxide and salicylic acid, the two most popular over-the-counter acne medications, generate moderate to severe skin irritation within 24 h. These results suggest that LipoLA hold a high therapeutic potential for the treatment of acne infection and other P. acnes related diseases.


Assuntos
Antibacterianos/farmacologia , Ácidos Láuricos/farmacologia , Lipossomos/química , Propionibacterium acnes/efeitos dos fármacos , Animais , Antibacterianos/administração & dosagem , Antibacterianos/química , Modelos Animais de Doenças , Infecções por Bactérias Gram-Positivas/tratamento farmacológico , Infecções por Bactérias Gram-Positivas/microbiologia , Ácidos Láuricos/administração & dosagem , Ácidos Láuricos/química , Camundongos , Pele/efeitos dos fármacos , Pele/patologia , Dermatopatias/tratamento farmacológico , Dermatopatias/microbiologia
17.
Nanoscale ; 5(19): 8884-8, 2013 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-23907698

RESUMO

RBC membrane-cloaked polymeric nanoparticles represent an emerging nanocarrier platform with extended circulation in vivo. A lipid-insertion method is employed to functionalize these nanoparticles without the need for direct chemical conjugation. Insertion of both folate and the nucleolin-targeting aptamer AS1411 shows receptor-specific targeting against model cancer cell lines.


Assuntos
Membrana Eritrocítica/química , Lipídeos/química , Nanopartículas/química , Aptâmeros de Nucleotídeos/química , Sequência de Bases , Linhagem Celular Tumoral , Membrana Eritrocítica/metabolismo , Citometria de Fluxo , Fluoresceína-5-Isotiocianato/química , Ácido Fólico/química , Ácido Fólico/metabolismo , Humanos , Fosfoproteínas/química , Fosfoproteínas/metabolismo , Polietilenoglicóis/química , Proteínas de Ligação a RNA/química , Proteínas de Ligação a RNA/metabolismo , Nucleolina
18.
Biomaterials ; 32(26): 6324-32, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21624651

RESUMO

Gene therapy is a promising method for osteoarthritis and cartilage injury. However, specifically delivering target genes into chondrocytes is a great challenge because of their non-vascularity and the dense extracellular matrix of cartilage. In our study, we identified a chondrocyte-affinity peptide (CAP, DWRVIIPPRPSA) by phage display technology. Subsequent analysis suggests that the peptide can efficiently interact specifically with chondrocytes without any species specificity. Polyethylenimine (PEI) was covalently modified with CAP to construct a non-viral vector for cartilage-targeted therapy. To investigate the cartilage-targeting property of the CAP-modified vector, FITC-labeled CAP conjugated PEI/DNA particles were injected into rabbit knee joints, and visualized under confocal microscope. Higher concentrations of CAP-modified vector were detected in the cartilage and specifically taken up by chondrocytes compared with a randomly scrambled peptide (SP)-modified vector. To evaluate cartilage-targeting transfection efficiency, the GFP and luciferase genes were delivered into knee joints using CAP- and SP-modified PEI. Cartilage transfections mediated by CAP-modified PEI were much more efficient and specific than those by SP-modified PEI. This result suggests that CAP-modified PEI could be used as a specific cartilage-targeting vector for cartilage disorders.


Assuntos
Cartilagem/metabolismo , Condrócitos/metabolismo , Vetores Genéticos/administração & dosagem , Vetores Genéticos/química , Biblioteca de Peptídeos , Peptídeos/química , Animais , Células Cultivadas , Imunofluorescência , Humanos , Masculino , Pessoa de Meia-Idade , Peptídeos/síntese química , Polietilenoimina/química , Coelhos
19.
J Biomed Nanotechnol ; 7(6): 807-12, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22416580

RESUMO

Noise induced hearing loss (NIHL) is a complex occupational hazard caused by an interaction between genetic and environmental factors. Millions of Chinese industrial people are daily exposed to high level of noise. Although the environmental risk factors have been studied extensively, the nature of the genetic factors contributing to HIHL has not yet been clarified. In this study, we investigated 15 single nucleotide polymorphisms (SNPs) in 6 candidate genes influence susceptibility to noise in Chinese noise-exposed workers. Data from 3-dimensional polyacrylamide gel-based microarray platforms were analyzed. 103 blood samples were collected from noise-exposed laborers in Ningbo, Zhejiang, China. Subsequently, the interaction between noise exposure and genotypes and their effect on NIHL were analysed using logistic regression. Two interesting results were observed between noise exposure levels and genotypes of three SNPs, hence confirming that they are NIHL susceptibility genes in Chinese population.


Assuntos
Resinas Acrílicas/química , Perda Auditiva Provocada por Ruído/genética , Sequência de Bases , China , Frequência do Gene , Predisposição Genética para Doença , Humanos , Modelos Logísticos , Dados de Sequência Molecular , Ruído Ocupacional , Exposição Ocupacional , Análise de Sequência com Séries de Oligonucleotídeos/instrumentação , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Polimorfismo de Nucleotídeo Único
20.
Biomaterials ; 32(26): 6226-33, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21658757

RESUMO

The engineering of drug-encapsulated targeted nanoparticles (NPs) has the potential to revolutionize drug therapy. A major challenge for the smooth translation of targeted NPs to the clinic has been developing methods for the prediction and optimization of the NP surface composition, especially when targeting ligands (TL) of different chemical properties are involved in the NP self-assembly process. Here we investigated the self-assembly and properties of two different targeted NPs decorated with two widely used TLs that have different water solubilities, and developed methods to characterize and optimize NP surface composition. We synthesized two different biofunctional polymers composed of poly(lactide-co-glycolide)-b-polyethyleneglycol-RGD (PLGA-PEG-RGD, high water solubility TL) and PLGA-PEG-Folate (low water solubility TL). Targeted NPs with different ligand densities were prepared by mixing TL-conjugated polymers with non-conjugated PLGA-PEG at different ratios through nanoprecipitation. The NP surface composition was quantified and the results revealed two distinct nanoparticle assembly behaviors: for the case of PLGA-PEG-RGD, nearly all RGD molecules conjugated to the polymer were found to be on the surface of the NPs. In contrast, only ∼20% of the folate from PLGA-PEG-Folate was present on the NP surface while the rest remained presumably buried in the PLGA NP core due to hydrophobic interactions of PLGA and folate. Finally, in vitro phagocytosis and cell targeting of NPs were investigated, from which a window of NP formulations exhibiting minimum uptake by macrophages and maximum uptake by targeted cells was determined. These results underscore the impact that the ligand chemical properties have on the targeting capabilities of self-assembled targeted nanoparticles and provide an engineering strategy for improving their targeting specificity.


Assuntos
Nanopartículas/química , Polímeros/química , Água/química , Animais , Linhagem Celular , Ácido Fólico/química , Humanos , Interações Hidrofóbicas e Hidrofílicas , Ácido Láctico/química , Camundongos , Polietilenoglicóis/química , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Solubilidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA