Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
J Nanobiotechnology ; 22(1): 175, 2024 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-38609947

RESUMO

Nonviral delivery of the CRISPR/Cas9 system provides great benefits for in vivo gene therapy due to the low risk of side effects. However, in vivo gene editing by delivering the Cas9 ribonucleoprotein (RNP) is challenging due to the poor delivery into target tissues and cells. Here, we introduce an effective delivery method for the CRISPR/Cas9 RNPs by finely tuning the formulation of ionizable lipid nanoparticles. The LNPs delivering CRISPR/Cas9 RNPs (CrLNPs) are demonstrated to induce gene editing with high efficiencies in various cancer cell lines in vitro. Furthermore, we show that CrLNPs can be delivered into tumor tissues with high efficiency, as well as induce significant gene editing in vivo. The current study presents an effective platform for nonviral delivery of the CRISPR/Cas9 system that can be applied as an in vivo gene editing therapeutic for treating various diseases such as cancer and genetic disorders.


Assuntos
Sistemas CRISPR-Cas , Edição de Genes , Lipossomos , Nanopartículas , Linhagem Celular , Ribonucleoproteínas/genética
2.
Mol Pharm ; 14(2): 423-430, 2017 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-27936751

RESUMO

Efficient delivery of drugs to the retina is critical but difficult to achieve with current methods. There have been a number of attempts to use intravitreal injection of liposomes, artificial vesicles composed of a phospholipid bilayer, to overcome the limitations of conventional intravitreal injection (short retention time, toxicity, poor penetration, etc.). Here, we report an optimal liposomal formulation that can diffuse through the vitreous humor, deliver the incorporated agents to all retinal layers effectively, and maintain them for a relatively long time. We first delivered lipophilic compounds and phospholipid-conjugated hydrophilic agents to the inner limiting membrane using engineered liposomes. Subsequently, the agents penetrated the retina deeply, presumably via extracellular vesicles, nanoscale vesicles secreted from retinal-associated cells. These results suggest that this engineered liposomal formulation can leverage the biological transport system for effective retinal penetration of lipophilic and lipid-conjugated agents.


Assuntos
Bicamadas Lipídicas/metabolismo , Lipídeos/química , Lipossomos/administração & dosagem , Lipossomos/química , Retina/efeitos dos fármacos , Retina/metabolismo , Animais , Química Farmacêutica/métodos , Portadores de Fármacos/administração & dosagem , Portadores de Fármacos/química , Feminino , Interações Hidrofóbicas e Hidrofílicas , Camundongos , Camundongos Endogâmicos C57BL , Fosfolipídeos/administração & dosagem , Fosfolipídeos/química , Corpo Vítreo/efeitos dos fármacos , Corpo Vítreo/metabolismo
3.
Nano Lett ; 15(5): 2938-44, 2015 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-25806671

RESUMO

Natural membrane vesicles (MVs) derived from various types of cells play an essential role in transporting biological materials between cells. Here, we show that exogenous compounds are packaged in the MVs by engineering the parental cells via liposomes, and the MVs mediate autonomous intercellular migration of the compounds through multiple cancer cell layers. Hydrophobic compounds delivered selectively to the plasma membrane of cancer cells using synthetic membrane fusogenic liposomes were efficiently incorporated into the membrane of MVs secreted from the cells and then transferred to neighboring cells via the MVs. This liposome-mediated MV engineering strategy allowed hydrophobic photosensitizers to significantly penetrate both spheroids and in vivo tumors, thereby enhancing the therapeutic efficacy. These results suggest that innate biological transport systems can be in situ engineered via synthetic liposomes to guide the penetration of chemotherapeutics across challenging tissue barriers in solid tumors.


Assuntos
Engenharia Celular , Lipossomos/química , Neoplasias/tratamento farmacológico , Transporte Biológico , Membrana Celular/química , Humanos , Interações Hidrofóbicas e Hidrofílicas , Lipossomos/ultraestrutura , Neoplasias/química , Células Tumorais Cultivadas
4.
Biomaterials ; 309: 122623, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38797121

RESUMO

Photobiomodulation (PBM), the use of biocompatible tissue-penetrating light to interact with intracellular chromophores to modulate the fates of cells and tissues, has emerged as a promising non-invasive approach to enhancing tissue regeneration. Unlike photodynamic or photothermal therapies that require the use of photothermal agents or photosensitizers, PBM treatment does not need external agents. With its non-harmful nature, PBM has demonstrated efficacy in enhancing molecular secretions and cellular functions relevant to tissue regeneration. The utilization of low-level light from various sources in PBM targets cytochrome c oxidase, leading to increased synthesis of adenosine triphosphate, induction of growth factor secretion, activation of signaling pathways, and promotion of direct or indirect gene expression. When integrated with stem cell populations, bioactive molecules or nanoparticles, or biomaterial scaffolds, PBM proves effective in significantly improving tissue regeneration. This review consolidates findings from in vitro, in vivo, and human clinical outcomes of both PBM alone and PBM-combined therapies in tissue regeneration applications. It encompasses the background of PBM invention, optimization of PBM parameters (such as wavelength, irradiation, and exposure time), and understanding of the mechanisms for PBM to enhance tissue regeneration. The comprehensive exploration concludes with insights into future directions and perspectives for the tissue regeneration applications of PBM.


Assuntos
Terapia com Luz de Baixa Intensidade , Regeneração , Humanos , Terapia com Luz de Baixa Intensidade/métodos , Animais , Regeneração/efeitos da radiação , Engenharia Tecidual/métodos , Materiais Biocompatíveis/química , Alicerces Teciduais/química
5.
ACS Biomater Sci Eng ; 9(4): 1919-1927, 2023 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-36921244

RESUMO

Nanoparticle-based drug delivery has been widely used for effective anticancer treatment. However, a key challenge restricting the efficacy of nanotherapeutics is limited tissue penetration within solid tumors. Here, we report a targeted fusogenic liposome (TFL) that can selectively deliver lipophilic cargo to the plasma membranes of tumor cells. TFL is prepared by directly attaching tumor-targeting peptides to the surface of FL instead of the cationic moieties. The lipophilic cargo loaded in the membrane of TFL is transferred to the plasma membranes of tumor cells and subsequently packaged in the extracellular vesicles (EVs) released by the cells. Systemically administered TFL accumulates in the perivascular region of tumors, where the lipophilic cargo is unloaded to the tumor cell membranes and distributed autonomously throughout the tumor tissue via extracellular vesicle-mediated intercellular transfer. When loaded with a lipophilic pro-apoptotic drug, thapsigargin (Tg), TFL significantly inhibits tumor growth in a mouse colorectal cancer model. Furthermore, the combination treatment with TFL (Tg) potentiates the antitumor efficacy of FDA-approved liposomal doxorubicin, whose therapeutic effect is limited to perivascular regions without significant toxicity.


Assuntos
Vesículas Extracelulares , Lipossomos , Camundongos , Animais , Sistemas de Liberação de Medicamentos , Peptídeos , Linhagem Celular Tumoral
6.
Mol Pharm ; 9(3): 539-45, 2012 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-22243419

RESUMO

Premature recognition and clearance of nanoparticulate imaging and therapeutic agents by macrophages in the tissues can dramatically reduce both the nanoparticle half-life and delivery to the diseased tissue. Grafting nanoparticles with hydrogels prevents nanoparticulate recognition by liver and spleen macrophages and greatly prolongs circulation times in vivo. Understanding the mechanisms by which hydrogels achieve this "stealth" effect has implications for the design of long-circulating nanoparticles. Thus, the role of plasma protein absorption in the hydrogel effect is not yet understood. Short-circulating dextran-coated iron oxide nanoparticles could be converted into stealth hydrogel nanoparticles by cross-linking with 1-chloro-2,3-epoxypropane. We show that hydrogelation did not affect the size, shape and zeta potential, but completely prevented the recognition and clearance by liver macrophages in vivo. Hydrogelation decreased the number of hydroxyl groups on the nanoparticle surface and reduced the binding of the anti-dextran antibody. At the same time, hydrogelation did not reduce the absorption of cationic proteins on the nanoparticle surface. Specifically, there was no effect on the binding of kininogen, histidine-rich glycoprotein, and protamine sulfate to the anionic nanoparticle surface. In addition, hydrogelation did not prevent activation of plasma kallikrein on the metal oxide surface. These data suggest that (a) a stealth hydrogel coating does not mask charge interactions with iron oxide surface and (b) the total blockade of plasma protein absorption is not required for maintaining iron oxide nanoparticles' long-circulating stealth properties. These data illustrate a novel, clinically promising property of long-circulating stealth nanoparticles.


Assuntos
Dextranos/química , Compostos Férricos/química , Hidrogel de Polietilenoglicol-Dimetacrilato/química , Nanopartículas/química , Animais , Feminino , Complexo Ferro-Dextran/química , Cininogênios/química , Camundongos , Camundongos Endogâmicos C57BL , Ligação Proteica
7.
ACS Biomater Sci Eng ; 8(10): 4140-4152, 2022 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-36210772

RESUMO

Porous silicon (pSi) nanoparticles are loaded with Immunoglobulin A-2 (IgA2) antibodies, and the assembly is coated with pH-responsive polymers on the basis of the Eudragit family of enteric polymers (L100, S100, and L30-D55). The temporal release of the protein from the nanocomposite formulations is quantified following an in vitro protocol simulating oral delivery: incubation in simulated gastric fluid (SGF; at pH 1.2) for 2 h, followed by a fasting state simulated intestinal fluid (FasSIF; at pH 6.8) or phosphate buffer solution (PBS; at pH 7.4). The nanocomposite formulations display a negligible release in SGF, while more than 50% of the loaded IgA2 is released in solutions at a pH of 6.8 (FasSIF) or 7.4 (PBS). Between 21 and 44% of the released IgA2 retains its functional activity. A capsule-based system is also evaluated, where the IgA2-loaded particles are packed into a gelatin capsule and the capsule is coated with either EudragitL100 or EudragitS100 polymer for a targeted release in the small intestine or the colon, respectively. The capsule-based formulations outperform polymer-coated nanoparticles in vitro, preserving 45-54% of the activity of the released protein.


Assuntos
Nanopartículas , Polímeros , Gelatina , Concentração de Íons de Hidrogênio , Imunoglobulina A , Intestino Delgado , Fosfatos , Porosidade , Silício , Solubilidade
8.
Nat Mater ; 8(4): 331-6, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19234444

RESUMO

Nanomaterials that can circulate in the body hold great potential to diagnose and treat disease. For such applications, it is important that the nanomaterials be harmlessly eliminated from the body in a reasonable period of time after they carry out their diagnostic or therapeutic function. Despite efforts to improve their targeting efficiency, significant quantities of systemically administered nanomaterials are cleared by the mononuclear phagocytic system before finding their targets, increasing the likelihood of unintended acute or chronic toxicity. However, there has been little effort to engineer the self-destruction of errant nanoparticles into non-toxic, systemically eliminated products. Here, we present luminescent porous silicon nanoparticles (LPSiNPs) that can carry a drug payload and of which the intrinsic near-infrared photoluminescence enables monitoring of both accumulation and degradation in vivo. Furthermore, in contrast to most optically active nanomaterials (carbon nanotubes, gold nanoparticles and quantum dots), LPSiNPs self-destruct in a mouse model into renally cleared components in a relatively short period of time with no evidence of toxicity. As a preliminary in vivo application, we demonstrate tumour imaging using dextran-coated LPSiNPs (D-LPSiNPs). These results demonstrate a new type of multifunctional nanostructure with a low-toxicity degradation pathway for in vivo applications.


Assuntos
Portadores de Fármacos , Nanopartículas , Silício , Animais , Antineoplásicos/administração & dosagem , Materiais Biocompatíveis , Doxorrubicina/administração & dosagem , Células HeLa , Humanos , Luminescência , Camundongos , Camundongos Endogâmicos BALB C , Microscopia Eletrônica de Varredura
9.
Drug Deliv Transl Res ; 10(6): 1876-1887, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32367425

RESUMO

The blood-brain barrier (BBB) impedes the efficient delivery of systemically administered drugs to brain tumors, thus reducing the therapeutic efficacy. To overcome the limitations of intravascular delivery, convention-enhanced delivery (CED) was introduced to infuse drugs directly into the brain tumor using a catheter with a continuous positive pressure. However, tissue distribution and retention of the infused drugs are significantly hindered by microenvironmental factors of the tumor such as the extracellular matrix and lymphatic drainage system in the brain. Here, we leveraged a liposomal formulation to simultaneously improve tissue distribution and retention of drugs infused in the brain tumor via the CED method. Various liposomal formulations with different surface charge, PEGylation, and transition temperature (Tm) were prepared to test the cellular uptake in vitro, and the tissue distribution and retention in the brain. In in vitro studies, PEGylated liposomal formulations with a positive surface charge and high Tm showed the most efficient cellular uptake among the tested formulations. In in vivo studies, the liposomal formulations were infused directly into the brain via the CED method. PEGylated liposomal formulations with a positive surface charge and high Tm showed more efficient distribution and retention in both normal and tumor tissues while only-PEGylated formulations displayed rapid clearance from the tissues to cervical lymph nodes. Furthermore, we demonstrated that the CED of liposomal everolimus prepared with the PEGylated formulation with a positive surface charge and high Tm resulted in superior therapeutic effects for glioblastoma treatment compared to other formulations. Graphical abstract.


Assuntos
Neoplasias Encefálicas , Sistemas de Liberação de Medicamentos , Glioblastoma , Animais , Neoplasias Encefálicas/tratamento farmacológico , Convecção , Feminino , Glioblastoma/tratamento farmacológico , Lipossomos/uso terapêutico , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL
10.
J Control Release ; 319: 77-86, 2020 03 10.
Artigo em Inglês | MEDLINE | ID: mdl-31843641

RESUMO

Recently, cyclodextrin (CD) has shown the potential for effective treatment of atherosclerotic plaques in mice by solubilizing plaque cholesterol. While promising as a new therapy for atherosclerosis, poor pharmacokinetics and ototoxicity of CD pose a therapeutic challenge. Thus far, however, there has been no attempts to overcome such limitations. Here, we showed that cyclodextrin polymer (CDP) with a diameter of ~ 10 nm exhibits outstanding pharmacokinetics and plaque targeting efficacy compared to a monomeric CD. Furthermore, we found out that CDP does not induce plasma membrane disruption as opposed to CD, which eliminated cytotoxicity and hemolytic activity of CD. In a mouse model of atherosclerosis, subcutaneous injections of beta-cyclodextrin polymer (ßCDP) significantly inhibited plaque growth compared to monomeric hydroxypropyl-beta-cyclodextrin (HPßCD) at the same dose (1 g/kg). More importantly, ßCDP did not induce significant ototoxicity at a high-dose (8 g/kg) where HPßCD reduced the outer hair cell content by 36%. These findings suggest that the polymerization of CD can overcome major limitations of CD therapy for treatment of atherosclerosis.


Assuntos
Aterosclerose , Ciclodextrinas , Ototoxicidade , 2-Hidroxipropil-beta-Ciclodextrina , Animais , Aterosclerose/tratamento farmacológico , Celulose , Camundongos
11.
J Drug Target ; 28(7-8): 780-788, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32116049

RESUMO

Despite the wide utility of gold nanorods (GNRs) in biomedical fields, only a few methods for modifying or coating the surface of GNRs suitable for biomedical applications are available. In this study, we report a new facile method that enables formation of an ultra-thin (nanometre-thickness) siloxane layer on GNRs with anti-biofouling properties and ligand functionalisation ability. A triblock random copolymer, poly(TMSMA-r-PEGMA-r-NAS), was used to coat GNRs. An ultrathin polymeric shell was formed surrounding GNRs through acid-catalysed crosslinking of silicates of TMSMA. The polymer-coated GNRs (p-GNRs) exhibited high colloidal stability in biological solutions of high ionic strength and long-term stability superior to that of PEG2k-S-GNRs. The functionalities of NAS were demonstrated using two methods for conjugating targeting ligands and loading doxorubicin via electrostatic interactions. The ligand-specific cancer-targeting ability and combinatorial chemo-photothermal anticancer effects were validated in vitro and in vivo, suggesting their potential utility in various fields.


Assuntos
Química Farmacêutica/métodos , Sistemas de Liberação de Medicamentos/métodos , Ouro/química , Nanotubos/química , Siloxanas/química , Animais , Linhagem Celular Tumoral , Doxorrubicina/administração & dosagem , Doxorrubicina/farmacocinética , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Polietilenoglicóis/química , Polímeros
12.
Cancer Chemother Pharmacol ; 83(3): 399-410, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30515553

RESUMO

BACKGROUND: Chemoresistance remains a major challenge for effective chemotherapy of non-small-cell lung carcinoma (NSCLC). CD44 expression is related to the susceptibility of various cancer cell types to anticancer drugs. Here, we systematically investigated the CD44-dependent chemoresistance of NSCLC cells and developed a liposomal siRNA delivery system to overcome this chemoresistance by the self-targeted downregulation of CD44. METHODS: We confirmed the relationship between the expression of CD44 and the chemosensitivity of NSCLC cells using flow cytometry and MTT assay. We then generated and characterized cisplatin-resistant cell lines and compared the expression of CD44 in resistant cells to that in parental cells using western blotting. To evaluate whether the chemosensitivity of resistant cells depends on CD44 expression, we performed CD44 knockdown using CD44 siRNA and detected the chemosensitivity of these cells. Additionally, we prepared hyaluronic acid (HA)-coated liposomes as a targeted delivery system to selectively deliver CD44-specific siRNA to chemoresistant NSCLC cells and observed whether the chemosensitivity of these cells was improved. RESULTS: We found that CD44 expression is inversely proportional to the degree of cellular response to cisplatin chemotherapy and that CD44 is overexpressed in chemoresistant NSCLC cells. By performing CD44 knockdown using siRNA, we reconfirmed that the chemosensitivity of resistant cells depends on CD44 expression. We also observed that HA-liposome-mediated siRNA delivery prior to cisplatin chemotherapy significantly reduced CD44 expression and enhanced cisplatin sensitivity in chemoresistant NSCLC cells. CONCLUSIONS: These results suggest that self-targeted downregulation of chemoresistance-associated cell surface proteins during chemotherapy is an effective therapeutic strategy for overcoming the chemoresistance of NSCLC cells.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos/genética , Receptores de Hialuronatos/genética , Neoplasias Pulmonares/tratamento farmacológico , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas/genética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Cisplatino/farmacologia , Cisplatino/uso terapêutico , Regulação para Baixo , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Sinergismo Farmacológico , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Humanos , Receptores de Hialuronatos/metabolismo , Ácido Hialurônico/química , Lipossomos , Neoplasias Pulmonares/genética , RNA Interferente Pequeno/administração & dosagem
13.
Bioconjug Chem ; 19(8): 1570-8, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18611045

RESUMO

The in vivo fate of nanomaterials strongly determines their biomedical efficacy. Accordingly, much effort has been invested into the development of library screening methods to select targeting ligands for a diversity of sites in vivo. Still, broad application of chemical and biological screens to the in vivo targeting of nanomaterials requires ligand attachment chemistries that are generalizable, efficient, covalent, orthogonal to diverse biochemical libraries, applicable under aqueous conditions, and stable in in vivo environments. To date, the copper(I)-catalyzed Huisgen 1,3-dipolar cycloaddition or "click" reaction has shown considerable promise as a method for developing targeted nanomaterials in vitro. Here, we investigate the utility of "click" chemistry for the in vivo targeting of inorganic nanoparticles to tumors. We find that "click" chemistry allows cyclic LyP-1 targeting peptides to be specifically linked to azido-nanoparticles and to direct their binding to p32-expressing tumor cells in vitro. Moreover, "click" nanoparticles are able to stably circulate for hours in vivo following intravenous administration (>5 h circulation time), extravasate into tumors, and penetrate the tumor interstitium to specifically bind p32-expressing cells in tumors. In the future, in vivo use of "click" nanomaterials should expedite the progression from ligand discovery to in vivo evaluation and diversify approaches toward multifunctional nanoparticle development.


Assuntos
Nanopartículas/química , Nanopartículas/uso terapêutico , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Peptídeos Cíclicos/química , Peptídeos Cíclicos/uso terapêutico , Alcinos/química , Aminas/química , Sequência de Aminoácidos , Animais , Azidas/química , Proteínas de Transporte , Bovinos , Linhagem Celular Tumoral , Fluorescência , Regulação da Expressão Gênica , Humanos , Magnetismo , Camundongos , Proteínas Mitocondriais/metabolismo , Peptídeos Cíclicos/metabolismo , Polímeros/química , Sensibilidade e Especificidade , Compostos de Sulfidrila/química
14.
Drug Deliv Transl Res ; 8(5): 1380-1388, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30027371

RESUMO

Borrelidin is an inhibitor of threonyl-tRNA synthetase with both anticancer and antiangiogenic activities. Although borrelidin could be a potent drug that can treat metastatic cancer through synergistic therapeutic effects, its severe liver toxicity has limited the use for cancer therapeutics. In this study, we developed a liposomal formulation of borrelidin to treat metastatic breast cancer effectively through its combined anticancer and antiangiogenic effects while reducing the potential liver toxicity. The liposomal formulation was optimized to maximize loading stability and efficiency of lipophilic borrelidin in the liposomal membrane and its delivery efficiency to primary tumor in a mouse model of metastatic breast cancer. Liposomal borrelidin showed significant in vitro therapeutic effects on proliferation and migration of tumor cells and angiogenesis of endothelial cells. Furthermore, liposomal borrelidin exhibited superior inhibitory effects on primary tumor growth and lung metastasis in vivo compared to free borrelidin. More importantly, liposomal borrelidin did not induce any significant systemic toxicity in the mouse model after multiple injections.


Assuntos
Inibidores da Angiogênese/administração & dosagem , Antineoplásicos/administração & dosagem , Neoplasias da Mama/tratamento farmacológico , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/secundário , Inibidores da Angiogênese/efeitos adversos , Inibidores da Angiogênese/química , Inibidores da Angiogênese/farmacologia , Animais , Antineoplásicos/efeitos adversos , Antineoplásicos/química , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Estabilidade de Medicamentos , Álcoois Graxos/administração & dosagem , Álcoois Graxos/efeitos adversos , Álcoois Graxos/química , Álcoois Graxos/farmacologia , Feminino , Células Endoteliais da Veia Umbilical Humana , Humanos , Lipossomos , Camundongos , Ensaios Antitumorais Modelo de Xenoenxerto
15.
Korean J Ophthalmol ; 32(5): 417-423, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30311465

RESUMO

PURPOSE: To improve the treatment efficiency of optic nerve diseases by delivering therapeutic materials to the optic nerve directly. METHODS: We tried to optimize liposomal composition to deliver a payload to the optic nerve efficiently when it is injected intravitreally. After loading dexamethasone into this liposome, we tested the therapeutic effect of liposomes in this treatment using a murine model of ischemic optic neuropathy. RESULTS: Our optimized liposome can deliver its payload to the optic nerve more efficiently than other tested compositions. Moreover, dexamethasone-loaded liposomes had a significant therapeutic effect in a murine model of ischemic optic neuropathy. CONCLUSIONS: Here, we demonstrate the optimal composition of liposomes that could efficiently deliver intravitreally injected exogenous compounds to the optic nerve. We expect that the intravitreal injection of liposomes with the suggested composition would improve the delivery efficacy of therapeutic compounds to the optic nerve.


Assuntos
Dexametasona/administração & dosagem , Nervo Óptico/efeitos dos fármacos , Neuropatia Óptica Isquêmica/tratamento farmacológico , Animais , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Feminino , Glucocorticoides/administração & dosagem , Injeções Intravítreas , Lipossomos , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal , Nervo Óptico/patologia , Neuropatia Óptica Isquêmica/diagnóstico , Células Ganglionares da Retina/efeitos dos fármacos , Células Ganglionares da Retina/patologia , Resultado do Tratamento
16.
ACS Appl Mater Interfaces ; 10(7): 6118-6123, 2018 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-29363943

RESUMO

Near-infrared photothermal therapy has been investigated extensively with regard to selective tumor eradication, yet its clinical translation has been limited because of the absence of FDA-approvable agents with effective phototherapeutic function and minimal systemic toxicity. In this work, we developed photothermally amplified therapeutic liposomes in an attempt to synergize chemotherapy and hyperthermia for effective cancer phototherapy. The anticancer drug cisplatin and the photothermal agent indocyanine green (ICG) were encapsulated in a thermosensitive liposomal formulation at the lipid/ICG ratio maximizing the ICG loading efficiency. These liposomes released cytotoxic cisplatin molecules selectively via ICG-mediated photothermal stimulation. In phototherapeutic studies, these liposomes amplified therapeutic effects both in vitro in cancer cells and in vivo in mouse tumor models significantly over chemotherapy or photothermal therapy alone. We believe that these photothermally amplified therapeutic liposomes composed solely of already FDA-approved components (cisplatin, ICG, and phospholipids) have enormous potential for clinical translation in treating various tumors compatible with laser irradiation.


Assuntos
Lipossomos , Animais , Hipertermia Induzida , Verde de Indocianina , Camundongos , Neoplasias , Fototerapia
17.
Sci Rep ; 8(1): 1899, 2018 01 30.
Artigo em Inglês | MEDLINE | ID: mdl-29382898

RESUMO

This work aimed at formulating paclitaxel (PTX) loaded cell penetrating peptide (CPP) coated Mn doped ZnS nanoparticles (Mn:ZnS NPs) for improved anti-cancer efficacy in vitro and in vivo. The developed PTX loaded Mn:ZnS NPs with different CPPs (PEN, pVEC and R9) showed enhanced anti-cancer effect compared to bare PTX, which has been validated by MTT assay followed by apoptosis assay and DNA fragmentation analysis. The in vivo bio-distribution and anti-cancer efficacy was studied on breast cancer xenograft model showing maximum tumor localization and enhanced therapeutic efficacy with R9 coated Mn:ZnS NPs (R9:Mn:ZnS NPs) and was confirmed by H/E staining. Thus, R9:Mn:ZnS NPs could be an ideal theranostic nano-carrier for PTX with enhanced  the rapeutic efficacy toward cancer cells, where penetration and sustainability of therapeutics are essential.


Assuntos
Peptídeos Penetradores de Células/química , Peptídeos Penetradores de Células/farmacologia , Manganês/química , Nanopartículas/química , Paclitaxel/química , Paclitaxel/farmacologia , Sulfetos/química , Compostos de Zinco/química , Animais , Antineoplásicos Fitogênicos/química , Antineoplásicos Fitogênicos/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias da Mama/tratamento farmacológico , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Portadores de Fármacos/química , Feminino , Células HeLa , Humanos , Polietilenoglicóis/química , Polímeros/química
18.
Nat Commun ; 9(1): 1969, 2018 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-29773788

RESUMO

The incidence of adverse effects and pathogen resistance encountered with small molecule antibiotics is increasing. As such, there is mounting focus on immunogene therapy to augment the immune system's response to infection and accelerate healing. A major obstacle to in vivo gene delivery is that the primary uptake pathway, cellular endocytosis, results in extracellular excretion and lysosomal degradation of genetic material. Here we show a nanosystem that bypasses endocytosis and achieves potent gene knockdown efficacy. Porous silicon nanoparticles containing an outer sheath of homing peptides and fusogenic liposome selectively target macrophages and directly introduce an oligonucleotide payload into the cytosol. Highly effective knockdown of the proinflammatory macrophage marker IRF5 enhances the clearance capability of macrophages and improves survival in a mouse model of Staphyloccocus aureus pneumonia.


Assuntos
Antibacterianos/farmacologia , Terapia Genética/métodos , Macrófagos/efeitos dos fármacos , Pneumonia Estafilocócica/terapia , Staphylococcus aureus/fisiologia , Animais , Antibacterianos/uso terapêutico , Citocinas/imunologia , Citocinas/metabolismo , Modelos Animais de Doenças , Farmacorresistência Bacteriana/genética , Técnicas de Silenciamento de Genes , Terapia Genética/efeitos adversos , Humanos , Fatores Reguladores de Interferon/genética , Fatores Reguladores de Interferon/imunologia , Lipossomos , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Nanopartículas/administração & dosagem , Nanopartículas/química , Peptídeos Cíclicos/administração & dosagem , Pneumonia Estafilocócica/imunologia , Pneumonia Estafilocócica/microbiologia , Pneumonia Estafilocócica/mortalidade , Células RAW 264.7 , Interferência de RNA/imunologia , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/genética , Staphylococcus aureus/efeitos dos fármacos , Análise de Sobrevida , Resultado do Tratamento
19.
Nat Commun ; 8: 15880, 2017 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-28627516

RESUMO

The targeted delivery of therapeutics using antibodies or nanomaterials has improved the precision and safety of cancer therapy. However, the paucity and heterogeneity of identified molecular targets within tumours have resulted in poor and uneven distribution of targeted agents, thus compromising treatment outcomes. Here, we construct a cooperative targeting system in which synthetic and biological nanocomponents participate together in the tumour cell membrane-selective localization of synthetic receptor-lipid conjugates (SR-lipids) to amplify the subsequent targeting of therapeutics. The SR-lipids are first delivered selectively to tumour cell membranes in the perivascular region using fusogenic liposomes. By hitchhiking with extracellular vesicles secreted by the cells, the SR-lipids are transferred to neighbouring cells and further spread throughout the tumour tissues where the molecular targets are limited. We show that this tumour cell membrane-targeted delivery of SR-lipids leads to uniform distribution and enhanced phototherapeutic efficacy of the targeted photosensitizer.


Assuntos
Membrana Celular/efeitos dos fármacos , Sistemas de Liberação de Medicamentos/métodos , Lipídeos/administração & dosagem , Neoplasias Experimentais/terapia , Fototerapia/métodos , Animais , Linhagem Celular Tumoral , Vesículas Extracelulares/química , Feminino , Células HeLa , Humanos , Lipídeos/química , Lipossomos/química , Lipossomos/farmacologia , Camundongos Endogâmicos BALB C , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
20.
Biomater Sci ; 5(8): 1670-1677, 2017 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-28715515

RESUMO

A surgical suture is a medical device to close the wound site of skin and organs but excessive inflammation surrounding the suture can disrupt the wound healing process. Although post-operative prescription of anti-inflammatory drugs is used to manage the inflammation, the need for local drug delivery systems has been rising because of low bioavailability and fast clearance of drugs. In this work, we proposed a new strategy for a local anti-inflammatory device by incorporating macrophage-targeted anti-inflammatory nanoparticles into the suture. For macrophage-targeted anti-inflammatory nanoparticles, poly(lactic-co-glycolic) nanoparticles were loaded with anti-inflammatory drug diclofenac and decorated with polyethylene glycol and macrophage-targeting ligand mannose. These anti-inflammatory nanoparticles released diclofenac sustainably, and targeted activated macrophages efficiently. After nanoparticle optimization, a suture was coated with multiple layers of macrophage-targeted anti-inflammatory nanoparticles using a dip coating process. The suture releasing macrophage-targeted anti-inflammatory nanoparticles showed an enhanced anti-inflammatory effect in both macrophage culture and excisional wound healing animal models compared to a free drug molecule-coated suture. These results suggest that anti-inflammatory nanoparticle-coated sutures have great potential as an effective local delivery system to reduce inflammation and pain at the wound site.


Assuntos
Anti-Inflamatórios/química , Anti-Inflamatórios/farmacologia , Portadores de Fármacos/química , Liberação Controlada de Fármacos , Macrófagos/metabolismo , Nanopartículas , Suturas , Animais , Diclofenaco/química , Diclofenaco/farmacologia , Portadores de Fármacos/metabolismo , Ácido Láctico/química , Camundongos , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Células RAW 264.7
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA