Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Small ; 20(10): e2306892, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-37867244

RESUMO

Poly(I:C) is a synthetic analogue of dsRNA capable of activating both TLR3 and RLRs, such as MDA-5 and RIG-I, as pathogen recognition receptors. While poly(I:C) is known to provoke a robust type I IFN, type III IFN, and Th1 cytokine response, its therapeutic use as a vaccine adjuvant is limited due to its vulnerability to nucleases and poor uptake by immune cells. is encapsulated poly(I:C) into lipid nanoparticles (LNPs) containing an ionizable cationic lipid that can electrostatically interact with poly(I:C). LNP-formulated poly(I:C) triggered both lysosomal TLR3 and cytoplasmic RLRs, in vitro and in vivo, whereas poly(I:C) in an unformulated soluble form only triggered endosomal-localized TLR3. Administration of LNP-formulated poly(I:C) in mouse models led to efficient translocation to lymphoid tissue and concurrent innate immune activation following intramuscular (IM) administration, resulting in a significant increase in innate immune activation compared to unformulated soluble poly(I:C). When used as an adjuvant for recombinant full-length SARS-CoV-2 spike protein, LNP-formulated poly(I:C) elicited potent anti-spike antibody titers, surpassing those of unformulated soluble poly(I:C) by orders of magnitude and offered complete protection against a SARS-CoV-2 viral challenge in vivo, and serum from these mice are capable of significantly reducing viral infection in vitro.


Assuntos
Lipossomos , Nanopartículas , Poli I-C , Glicoproteína da Espícula de Coronavírus , Receptor 3 Toll-Like , Animais , Camundongos , Humanos , Receptor 3 Toll-Like/genética , Receptor 3 Toll-Like/metabolismo , Adjuvantes Imunológicos/farmacologia
2.
Mol Ther ; 29(4): 1370-1381, 2021 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-33484964

RESUMO

Synthetic mRNAs are an appealing platform with multiple biomedical applications ranging from protein replacement therapy to vaccination. In comparison with conventional mRNA, synthetic self-amplifying mRNAs (sa-mRNAs) are gaining interest because of their higher and longer-lasting expression. However, sa-mRNAs also elicit an innate immune response, which may complicate their clinical application. Approaches to reduce the innate immunity of sa-mRNAs have not been studied in detail. Here we investigated, in vivo, the effect of several innate immune inhibitors and a novel cellulose-based mRNA purification approach on the type I interferon (IFN) response and the translation and vaccination efficacy of our formerly developed sa-mRNA vaccine against Zika virus. Among the investigated inhibitors, we found that corticosteroids and especially topical application of clobetasol at the sa-mRNA injection site was the most efficient in suppressing the type I IFN response and increasing the translation of sa-mRNA. However, clobetasol prevented formation of antibodies against sa-mRNA-encoded antigens and should therefore be avoided in a vaccination context. Residual dsRNA by-products of the in vitro transcription reaction are known inducers of immediate type I IFN responses. We additionally demonstrate a drastic reduction of these dsRNA by-products upon cellulose-based purification, reducing the innate immune response and improving sa-mRNA vaccination efficacy.


Assuntos
Imunidade Inata/genética , RNA Mensageiro/genética , Vacinação , Infecção por Zika virus/tratamento farmacológico , Corticosteroides/química , Celulose/química , Clobetasol/farmacologia , Regulação da Expressão Gênica/genética , Humanos , Imunidade Inata/efeitos dos fármacos , Imunidade Inata/imunologia , Interferon Tipo I/genética , Interferon Tipo I/imunologia , Biossíntese de Proteínas/efeitos dos fármacos , Biossíntese de Proteínas/imunologia , RNA Mensageiro/síntese química , RNA Mensageiro/química , RNA Mensageiro/farmacologia , Zika virus/efeitos dos fármacos , Zika virus/patogenicidade , Infecção por Zika virus/imunologia , Infecção por Zika virus/virologia
3.
J Am Chem Soc ; 143(26): 9872-9883, 2021 07 07.
Artigo em Inglês | MEDLINE | ID: mdl-34166595

RESUMO

Small-molecular Toll-like receptor 7/8 (TLR7/8) agonists hold promise as immune modulators for a variety of immune therapeutic purposes including cancer therapy or vaccination. However, due to their rapid systemic distribution causing difficult-to-control inflammatory off-target effects, their application is still problematic, in particular systemically. To address this problem, we designed and robustly fabricated pH-responsive nanogels serving as versatile immunodrug nanocarriers for safe delivery of TLR7/8-stimulating imidazoquinolines after intravenous administration. To this aim, a primary amine-reactive methacrylamide monomer bearing a pendant squaric ester amide is introduced, which is polymerized under controlled RAFT polymerization conditions. Corresponding PEG-derived squaric ester amide block copolymers self-assemble into precursor micelles in polar protic solvents. Their cores are amine-reactive and can sequentially be transformed by acid-sensitive cross-linkers, dyes, and imidazoquinolines. Remaining squaric ester amides are hydrophilized affording fully hydrophilic nanogels with profound stability in human plasma but stimuli-responsive degradation upon exposure to endolysosomal pH conditions. The immunomodulatory behavior of the imidazoquinolines alone or conjugated to the nanogels was demonstrated by macrophages in vitro. In vivo, however, we observed a remarkable impact of the nanogel: After intravenous injection, a spatially controlled immunostimulatory activity was evident in the spleen, whereas systemic off-target inflammatory responses triggered by the small-molecular imidazoquinoline analogue were absent. These findings underline the potential of squaric ester-based, pH-degradable nanogels as a promising platform to permit intravenous administration routes of small-molecular TLR7/8 agonists and, thus, the opportunity to explore their adjuvant potency for systemic vaccination or cancer immunotherapy purposes.


Assuntos
Adjuvantes Imunológicos/química , Ésteres/química , Nanogéis/química , Receptor 7 Toll-Like/agonistas , Receptor 8 Toll-Like/agonistas , Animais , Portadores de Fármacos/química , Liberação Controlada de Fármacos , Humanos , Concentração de Íons de Hidrogênio , Imunoterapia , Camundongos Endogâmicos BALB C , Micelas , Imagem Óptica , Polimerização , Polímeros/química
4.
Angew Chem Int Ed Engl ; 60(17): 9467-9473, 2021 04 19.
Artigo em Inglês | MEDLINE | ID: mdl-33464672

RESUMO

The search for vaccines that protect from severe morbidity and mortality because of infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the virus that causes coronavirus disease 2019 (COVID-19) is a race against the clock and the virus. Here we describe an amphiphilic imidazoquinoline (IMDQ-PEG-CHOL) TLR7/8 adjuvant, consisting of an imidazoquinoline conjugated to the chain end of a cholesterol-poly(ethylene glycol) macromolecular amphiphile. It is water-soluble and exhibits massive translocation to lymph nodes upon local administration through binding to albumin, affording localized innate immune activation and reduction in systemic inflammation. The adjuvanticity of IMDQ-PEG-CHOL was validated in a licensed vaccine setting (quadrivalent influenza vaccine) and an experimental trimeric recombinant SARS-CoV-2 spike protein vaccine, showing robust IgG2a and IgG1 antibody titers in mice that could neutralize viral infection in vitro and in vivo in a mouse model.


Assuntos
Adjuvantes Imunológicos/uso terapêutico , Vacinas contra COVID-19/uso terapêutico , COVID-19/prevenção & controle , Imidazóis/uso terapêutico , Imunidade Inata/efeitos dos fármacos , Quinolinas/uso terapêutico , Animais , Vacinas contra COVID-19/imunologia , Colesterol/análogos & derivados , Colesterol/imunologia , Colesterol/uso terapêutico , Feminino , Humanos , Imidazóis/imunologia , Vírus da Influenza A Subtipo H1N1/efeitos dos fármacos , Vacinas contra Influenza/imunologia , Vacinas contra Influenza/uso terapêutico , Influenza Humana/prevenção & controle , Glicoproteínas de Membrana/agonistas , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Polietilenoglicóis/uso terapêutico , Quinolinas/imunologia , Proteínas Recombinantes/imunologia , SARS-CoV-2/efeitos dos fármacos , Glicoproteína da Espícula de Coronavírus/imunologia , Tensoativos/uso terapêutico , Receptor 7 Toll-Like/agonistas , Receptor 8 Toll-Like/agonistas
5.
J Am Chem Soc ; 142(28): 12133-12139, 2020 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-32524819

RESUMO

Synthetic immune-stimulatory drugs such as agonists of the Toll-like receptors (TLR) 7/8 are potent activators of antigen-presenting cells (APCs), however, they also induce severe side effects due to leakage from the site of injection into systemic circulation. Here, we report on the design and synthesis of an amphiphilic polymer-prodrug conjugate of an imidazoquinoline TLR7/8 agonist that in aqueous medium forms vesicular structures of 200 nm. The conjugate contains an endosomal enzyme-responsive linker enabling degradation of the vesicles and release of the TLR7/8 agonist in native form after endocytosis, which results in high in vitro TLR agonist activity. In a mouse model, locally administered vesicles provoke significantly more potent and long-lasting immune stimulation in terms of interferon expression at the injection site and in draining lymphoid tissue compared to a nonamphiphilic control and the native TLR agonist. Moreover, the vesicles induce robust activation of dendritic cells in the draining lymph node in vivo.


Assuntos
Imidazóis/farmacologia , Glicoproteínas de Membrana/agonistas , Pró-Fármacos/farmacologia , Quinolinas/farmacologia , Receptor 7 Toll-Like/agonistas , Receptor 8 Toll-Like/agonistas , beta-Galactosidase/imunologia , Animais , Imidazóis/química , Imidazóis/metabolismo , Imunidade Inata/efeitos dos fármacos , Imunidade Inata/imunologia , Glicoproteínas de Membrana/imunologia , Camundongos , Estrutura Molecular , Tamanho da Partícula , Polietilenoglicóis/química , Polietilenoglicóis/metabolismo , Polietilenoglicóis/farmacologia , Pró-Fármacos/química , Pró-Fármacos/metabolismo , Quinolinas/química , Quinolinas/metabolismo , Propriedades de Superfície , Receptor 7 Toll-Like/imunologia , Receptor 8 Toll-Like/imunologia , beta-Galactosidase/química , beta-Galactosidase/metabolismo
6.
Angew Chem Int Ed Engl ; 58(43): 15390-15395, 2019 10 21.
Artigo em Inglês | MEDLINE | ID: mdl-31397948

RESUMO

Uncontrolled systemic inflammatory immune triggering has hampered the clinical translation of several classes of small-molecule immunomodulators, such as imidazoquinoline TLR7/8 agonists for vaccine design and cancer immunotherapy. By taking advantage of the inherent serum-protein-binding property of lipid motifs and their tendency to accumulate in lymphoid tissue, we designed amphiphilic lipid-polymer conjugates that suppress systemic inflammation but provoke potent lymph-node immune activation. This work provides a rational basis for the design of lipid-polymer amphiphiles for optimized lymphoid targeting.


Assuntos
Imunidade Inata , Receptor 7 Toll-Like/agonistas , Receptor 8 Toll-Like/agonistas , Animais , Colesterol/química , Imidazóis/química , Imunidade Inata/efeitos dos fármacos , Fatores Imunológicos/química , Fatores Imunológicos/metabolismo , Fatores Imunológicos/farmacologia , Lipídeos/química , Linfonodos/efeitos dos fármacos , Linfonodos/imunologia , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , NF-kappa B/metabolismo , Polímeros/química , Quinolinas/química , Quinolinas/farmacologia , Células RAW 264.7 , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia , Receptor 7 Toll-Like/metabolismo , Receptor 8 Toll-Like/metabolismo
7.
J Am Chem Soc ; 140(43): 14300-14307, 2018 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-30277761

RESUMO

Small molecule immuno-modulators such as agonists of Toll-like receptors (TLRs) are attractive compounds to stimulate innate immune cells toward potent antiviral and antitumor responses. However, small molecules rapidly enter the systemic circulation and cause "wasted inflammation". Hence, synthetic strategies to confine their radius of action to lymphoid tissue are of great relevance, to both enhance their efficacy and concomitantly limit toxicity. Here, we demonstrate that covalent conjugation of a small molecule TLR7/8 agonist immunomodulatory to a micelle-forming amphiphilic block copolymer greatly alters the pharmacokinetic profile, resulting in highly efficient lymphatic delivery. Moreover, we designed amphiphilic block copolymers in such a way to form thermodynamically stable micelles through π-π stacking between aromatic moieties, and we engineered the block copolymers to undergo an irreversible amphiphilic to hydrophilic transition in response to the acidic endosomal pH.


Assuntos
Linfonodos/efeitos dos fármacos , Polímeros/farmacologia , Tensoativos/farmacologia , Receptor 7 Toll-Like/agonistas , Receptor 8 Toll-Like/agonistas , Humanos , Concentração de Íons de Hidrogênio , Linfonodos/imunologia , Micelas , Modelos Moleculares , Estrutura Molecular , Polímeros/química , Tensoativos/química , Termodinâmica , Receptor 7 Toll-Like/imunologia , Receptor 8 Toll-Like/imunologia
9.
J Mater Chem B ; 12(16): 3927-3946, 2024 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-38563779

RESUMO

Messenger RNA (mRNA) based vaccines have been introduced worldwide to combat the Covid-19 pandemic. These vaccines consist of non-amplifying mRNA formulated in lipid nanoparticles (LNPs). Consequently, LNPs are considered benchmark non-viral carriers for nucleic acid delivery. However, the formulation and manufacturing of these mRNA-LNP nanoparticles are expensive and time-consuming. Therefore, we used self-amplifying mRNA (saRNA) and synthesized novel polymers as alternative non-viral carrier platform to LNPs, which enable a simple, rapid, one-pot formulation of saRNA-polyplexes. Our novel polymer-based carrier platform consists of randomly concatenated ethylenimine and propylenimine comonomers, resulting in linear, poly(ethylenimine-ran-propylenimine) (L-PEIx-ran-PPIy) copolymers with controllable degrees of polymerization. Here we demonstrate in multiple cell lines, that our saRNA-polyplexes show comparable to higher in vitro saRNA transfection efficiencies and higher cell viabilities compared to formulations with Lipofectamine MessengerMAX™ (LFMM), a commercial, lipid-based carrier considered to be the in vitro gold standard carrier. This is especially true for our in vitro best performing saRNA-polyplexes with N/P 5, which are characterised with a size below 100 nm, a positive zeta potential, a near 100% encapsulation efficiency, a high retention capacity and the ability to protect the saRNA from degradation mediated by RNase A. Furthermore, an ex vivo hemolysis assay with pig red blood cells demonstrated that the saRNA-polyplexes exhibit negligible hemolytic activity. Finally, a bioluminescence-based in vivo study was performed over a 35-day period, and showed that the polymers result in a higher and prolonged bioluminescent signal compared to naked saRNA and L-PEI based polyplexes. Moreover, the polymers show different expression profiles compared to those of LNPs, with one of our new polymers (L-PPI250) demonstrating a higher sustained expression for at least 35 days after injection.


Assuntos
Polietilenoimina , RNA Mensageiro , Transfecção , Animais , Transfecção/métodos , Polietilenoimina/química , Humanos , RNA Mensageiro/genética , Camundongos , Polipropilenos/química , Polímeros/química , Portadores de Fármacos/química , SARS-CoV-2/efeitos dos fármacos , Nanopartículas/química
10.
J Fluoresc ; 23(5): 909-20, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23579930

RESUMO

In vivo optical imaging has become a popular tool in animal laboratories. Currently, many in vivo optical imaging systems are available on the market, which often makes it difficult for research groups to decide which system fits their needs best. In this work we compared different commercially available systems, which can measure both bioluminescent and fluorescent light. The systems were tested for their bioluminescent and fluorescent sensitivity both in vitro and in vivo. The IVIS Lumina II was found to be most sensitive for bioluminescence imaging, with the Photon Imager a close second. Contrary, the Kodak system was, in vitro, the most sensitive system for fluorescence imaging. In vivo, the fluorescence sensitivity of the systems was similar. Finally, we examined the added value of spectral unmixing algorithms for in vivo optical imaging and demonstrated that spectral unmixing resulted in at least a doubling of the in vivo sensitivity. Additionally, spectral unmixing also enabled separate imaging of dyes with overlapping spectra which were, without spectral unmixing, not distinguishable.


Assuntos
Luminescência , Algoritmos , Animais , Temperatura Corporal , Corantes/química , Escherichia coli/química , Feminino , Lipossomos/química , Camundongos , Camundongos Endogâmicos , Camundongos Nus , Imagem Óptica/instrumentação
11.
Mol Pharm ; 9(8): 2136-45, 2012 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-22676473

RESUMO

Complexes between mRNA and GL67:DOPE:DMPE-PEG5000 (GL67) liposomes were formulated and characterized. Subsequently, the in vitro and in vivo expression characteristics of mRNA/GL67 complexes and pDNA/GL67 complexes, each produced at their optimal ratio, were compared in respiratory cells. Transfection of A549 cells with mRNA/GL67 complexes resulted in a much faster expression than after transfection with pDNA/GL67 complexes. The percentage of GFP-positive cells after mRNA and pDNA transfection peaked after 8 and 24 h, respectively. At these time points the percentage of GFP-positive cells was two times higher after mRNA transfection than after pDNA transfection. Furthermore, the efficacy of mRNA/GL67 complexes was independent of the cell cycle. This was in sharp contrast with pDNA/GL67 complexes that caused only a weak expression in nondividing cells. This confirms that the nuclear barrier is a crucial obstacle for pDNA but not for mRNA. Finally, mRNA/GL67 and pDNA/GL67 complexes encoding luciferase were administered intranasally to the lungs of mice. The mRNA/GL67 complexes did not give rise to a measurable luciferase expression in the murine lungs. In contrast, a detectable bioluminescent signal was present in the lungs of mice that received the pDNA/GL67 complexes. We showed that mRNA/GL67 complexes have a lower stability in biological fluids. Consequently, this may be an explanation for their lower performance in vivo.


Assuntos
Lipossomos/química , RNA Mensageiro/química , RNA Mensageiro/genética , Animais , Linhagem Celular , Escherichia coli/genética , Escherichia coli/metabolismo , Técnicas de Transferência de Genes , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Transfecção/métodos
12.
Mol Pharm ; 8(6): 2244-51, 2011 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-22014166

RESUMO

Microbubbles are Food and Drug Administration (FDA) approved contrast agents for ultrasound imaging. It has been reported that applying ultrasound on drug-loaded microbubbles facilitates drug uptake by cells, due to so-named sonoporation. However, the biophysics behind sonoporation are not fully understood. It is believed that sonoporation results in a "direct" delivery of drugs in the cytoplasm of cells, though it has been suggested as well that sonoporation facilitates endocytosis which would improve the internalization of drugs by cells. To get a better understanding of sonoporation, this study reports on the ultrasound assisted delivery of adeno-associated virus (AAV) loaded on the surface of microbubbles. AAVs rely on endocytosis for efficient transduction of cells and are, consequently, an elegant tool to evaluate whether endocytosis is involved in ultrasound-induced sonoporation. Applying ultrasound on AAV-loaded microbubbles clearly improved the internalization of AAVs by cells, though transduction of the cells did not occur, indicating that by sonoporation substances become directly delivered in the cytosol of cells.


Assuntos
Dependovirus , Sistemas de Liberação de Medicamentos , Microbolhas , Terapia por Ultrassom , Linhagem Celular Tumoral , Meios de Contraste/química , Endossomos/diagnóstico por imagem , Endossomos/metabolismo , Vetores Genéticos/farmacologia , Humanos , Microscopia Confocal , Modelos Biológicos , Estrutura Molecular , Polietilenoglicóis/química , Sonicação , Ultrassonografia
13.
Mol Ther ; 18(1): 101-8, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19623162

RESUMO

Drug delivery with microbubbles and ultrasound is gaining more and more attention in the drug delivery field due to its noninvasiveness, local applicability, and proven safety in ultrasonic imaging techniques. In this article, we tried to improve the cytotoxicity of doxorubicin (DOX)-containing liposomes by preparing DOX-liposome-containing microbubbles for drug delivery with therapeutic ultrasound. In this way, the DOX release and uptake can be restricted to ultrasound-treated areas. Compared to DOX-liposomes, DOX-loaded microbubbles killed at least two times more melanoma cells after exposure to ultrasound. After treatment of the melanoma cells with DOX-liposome-loaded microbubbles and ultrasound, DOX was mainly present in the nuclei of the cancer cells, whereas it was mainly detected in the cytoplasm of cells treated with DOX-liposomes. Exposure of cells to DOX-liposome-loaded microbubbles and ultrasound caused an almost instantaneous cellular entry of the DOX. At least two mechanisms were identified that explain the fast uptake of DOX and the superior cell killing of DOX-liposome-loaded microbubbles and ultrasound. First, exposure of DOX-liposome-loaded microbubbles to ultrasound results in the release of free DOX that is more cytotoxic than DOX-liposomes. Second, the cellular entry of the released DOX is facilitated due to sonoporation of the cell membranes. The in vitro results shown in this article indicate that DOX-liposome-loaded microbubbles could be a very interesting tool to obtain an efficient ultrasound-controlled DOX delivery in vivo.


Assuntos
Doxorrubicina/administração & dosagem , Doxorrubicina/farmacologia , Microbolhas , Animais , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Doxorrubicina/química , Lipossomos/administração & dosagem , Lipossomos/química , Melanoma/patologia , Camundongos , Microscopia Confocal , Modelos Teóricos
14.
Front Immunol ; 12: 768397, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34925337

RESUMO

Approximately 9 out of 10 adults have some form of periodontal disease, an infection-induced inflammatory disease of the tooth-supporting tissues. The initial form, gingivitis, often remains asymptomatic, but this can evolve into periodontitis, which is typically associated with halitosis, oral pain or discomfort, and tooth loss. Furthermore, periodontitis may contribute to systemic disorders like cardiovascular disease and type 2 diabetes mellitus. Control options remain nonspecific, time-consuming, and costly; largely relying on the removal of dental plaque and calculus by mechanical debridement. However, while dental plaque bacteria trigger periodontal disease, it is the host-specific inflammatory response that acts as main driver of tissue destruction and disease progression. Therefore, periodontal disease control should aim to alter the host's inflammatory response as well as to reduce the bacterial triggers. Vaccines may provide a potent adjunct to mechanical debridement for periodontal disease prevention and treatment. However, the immunopathogenic complexity and polymicrobial aspect of PD appear to complicate the development of periodontal vaccines. Moreover, a successful periodontal vaccine should induce protective immunity in the oral cavity, which proves difficult with traditional vaccination methods. Recent advances in mucosal vaccination may bridge the gap in periodontal vaccine development. In this review, we offer a comprehensive overview of mucosal vaccination strategies to induce protective immunity in the oral cavity for periodontal disease control. Furthermore, we highlight the need for additional research with appropriate and clinically relevant animal models. Finally, we discuss several opportunities in periodontal vaccine development such as multivalency, vaccine formulations, and delivery systems.


Assuntos
Mucosa Bucal/imunologia , Mucosa Nasal/imunologia , Doenças Periodontais/prevenção & controle , Vacinação , Animais , Humanos , Doenças Periodontais/terapia , Desenvolvimento de Vacinas
15.
Biochim Biophys Acta ; 1768(3): 571-9, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17188643

RESUMO

Several reports have shown a fast and efficient translocation of TAT-modified lipoplexes and particles into the cell cytoplasm. However, neither the uptake mechanism nor the biological effect of TAT-modified lipoplexes has been studied in detail. In this report we show that the increase in gene transfer of TAT-modified lipoplexes depends on the amount of cationic lipid in the lipoplexes and on the way TAT was coupled to the lipoplexes. We demonstrate that the cellular uptake of both TAT-modified and unmodified lipoplexes is very fast and, in contrast to previous publications, temperature-dependent. Additionally, after internalization TAT-modified as well as unmodified lipoplexes end up in lysosomal vesicles, indicating the involvement of clathrin-mediated endocytosis. Furthermore, chlorpromazine, a specific inhibitor of clathrin-dependent endocytosis, strongly inhibits the cellular uptake and biological activity of both the TAT-modified and unmodified lipoplexes. We also found that the uptake and biological activity of these lipoplexes are diminished when cholesterol in the cell membrane was bound by filipin, an inhibitor of the lipid-raft mediated pathway. Considering these data, we conclude that TAT-modified and unmodified lipoplexes are mainly internalized via a cholesterol-dependent clathrin-mediated pathway.


Assuntos
Endocitose/fisiologia , Produtos do Gene tat/química , Técnicas de Transferência de Genes , Lipossomos/metabolismo , Animais , Células COS , Chlorocebus aethiops , Colesterol/metabolismo , Clatrina/química , Clatrina/metabolismo , Clatrina/fisiologia , Ácidos Graxos Monoinsaturados/química , Filipina/metabolismo , Filipina/farmacologia , Fluoresceína-5-Isotiocianato , Corantes Fluorescentes , Terapia Genética , Lipossomos/química , Lisossomos/metabolismo , Microdomínios da Membrana/efeitos dos fármacos , Microdomínios da Membrana/metabolismo , Modelos Moleculares , Estrutura Molecular , Fosfatidiletanolaminas/química , Polietilenoglicóis/química , Compostos de Amônio Quaternário/química , Temperatura , Fatores de Tempo
16.
J Gene Med ; 10(7): 783-94, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18470950

RESUMO

BACKGROUND: Small interfering (si)RNA mediated inhibition of oncogenes or viral genes may offer great opportunities for the treatment of several diseases such as hepatocellular carcinoma and viral hepatitis. However, the development of siRNAs as therapeutic agents strongly depends on the availability of safe and effective intracellular delivery systems. Poly(beta-amino esters) (PbAEs) are, in contrast to many other cationic polymers evaluated in siRNA delivery, biodegradable into smaller, nontoxic molecules. METHODS AND RESULTS: We show for the first time that PbAE : siRNA complexes, containing 1,4-butanediol (PbAE1) or 1,6-hexanediol (PbAE2) diacrylate-based polymers, induced efficient gene silencing in both hepatoma cells and primary hepatocytes without causing significant cytotoxicity. Furthermore, carriers that slowly release the siRNA into the cytoplasm and hence induce a prolonged gene silencing are of major clinical interest, especially in fast dividing tumour cells. Therefore, we also studied the duration of gene silencing in the hepatoma cells and found that it was maintained for at least 5 days after siRNA delivery with PbAE2, the polymer with the slowest degradation kinetics. CONCLUSIONS: From the time-dependent cellular distribution of these PbAE : siRNA complexes, we suggest that the slowly degrading PbAE2 causes a sustained endosomal release of siRNA during a much longer period than PbAE1. This may support the hypothesis that the endosomal release mechanism of PbAE : siRNA complexes is based on an increase of osmotic pressure in the endosomal vesicles after polymer hydrolysis. In conclusion, our results show that both PbAEs, and especially PbAE2, open up new perspectives for the development of efficient biodegradable siRNA carriers suitable for clinical applications.


Assuntos
Carcinoma Hepatocelular/terapia , Inativação Gênica , Técnicas de Transferência de Genes , Terapia Genética/métodos , RNA Interferente Pequeno/genética , Adenoviridae , Animais , Butileno Glicóis , Linhagem Celular Tumoral , Ésteres/química , Glicóis , Hepatócitos , Humanos , Microscopia Confocal , Estrutura Molecular , Polímeros/química , Ratos
17.
J Control Release ; 115(3): 335-43, 2006 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-17010468

RESUMO

Plasmid DNA (pDNA) can occur in the compact supercoiled (SC) form, the relaxed open circular (OC) form and the linearized form. In this paper we investigated the transfection efficiency of SC, OC and linearized pDNA complexed to DOTAP/DOPE liposomes in Vero cells. Only DOTAP/DOPE liposomes containing SC pDNA showed protein expression while DOTAP/DOPE liposomes loaded with OC or linearized pDNA failed. First we questioned if the better transfection properties of the SC pDNA-containing lipoplexes could be due to a better transcription of SC pDNA in the nuclei of the cells, compared to OC and linearized pDNA. However, microinjecting (naked) SC, OC or linearized pDNA in the nuclei of the Vero cells revealed that the transcription efficiency was independent on the pDNA topology but did depend on the intranuclear concentration of the pDNA. As the amount of pDNA that reaches the nucleus is determined by the amount of pDNA that arrives in the cytosol it could be hypothesized that SC pDNA is more efficiently released from the DOTAP/DOPE liposomes when compared to OC and linearized pDNA. However, microinjecting comparable concentrations of the pDNA topologies in the cytoplasm still resulted in a significantly higher transfection in the case of SC pDNA, especially in cells that underwent cell division in the period after injection. It seems that, compared to OC and linearized pDNA, SC pDNA is better suited to reach the perinuclear region, a prerequisite to become entrapped in the nuclei of the cells during cell division.


Assuntos
DNA/química , Ácidos Graxos Monoinsaturados/química , Lipossomos , Conformação de Ácido Nucleico , Fosfatidiletanolaminas/química , Compostos de Amônio Quaternário/química , Transfecção , Transporte Ativo do Núcleo Celular , Animais , Núcleo Celular/metabolismo , Chlorocebus aethiops , DNA/genética , DNA/metabolismo , DNA Circular/química , DNA Circular/genética , DNA Circular/metabolismo , DNA Super-Helicoidal/química , DNA Super-Helicoidal/genética , DNA Super-Helicoidal/metabolismo , Desoxirribonuclease I/metabolismo , Genes Reporter , Proteínas de Fluorescência Verde , Substâncias Luminescentes , Microinjeções , Plasmídeos , Transcrição Gênica , Células Vero
18.
Invest Ophthalmol Vis Sci ; 46(10): 3553-61, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16186333

RESUMO

PURPOSE: Intravitreal injection of therapeutic DNA, complexed to nonviral carriers such as cationic liposomes, may be promising in the treatment of many severe retinal eye diseases. However, after intravitreal injection, such DNA/cationic liposome complexes-called lipoplexes (LPXs)-which are typically hundreds of nanometers in size, must first diffuse through the vitreous before they can reach the retina. The aim of this study was to elucidate whether vitreous is a barrier for the LPXs and to find strategies to overcome this barrier. METHODS: Fluorescent polystyrene nanospheres and LPXs were mixed with vitreous, and their mobility was monitored by fluorescence recovery after photobleaching (FRAP), a microscopy-based technique. The stability of LPXs and naked plasmid DNA in vitreous was studied by gel electrophoresis. RESULTS: We showed that polystyrene nanospheres, in our first experiments used as a model for the LPXs, do not diffuse freely into the vitreous but adhere to fibrillar structures in the vitreous, most likely to collagen fibers. Making the surfaces of the polystyrene nanospheres hydrophilic by attaching hydrophilic polyethylene glycol (PEG) chains at their surfaces circumvented the binding to fibrillar structures in the vitreous. FRAP revealed that "pegylated" polystyrene nanospheres, as long as they are smaller than 500 nm, are indeed mobile in the vitreous. It was further demonstrated that LPXs severely aggregate in vitreous and strongly bind to biopolymers in the vitreous, which immobilizes them completely. However, as observed for the polystyrene nanospheres, coating of the LPXs with PEG averted their aggregation in the vitreous and their binding to fibrillar structures. CONCLUSIONS: Modifying the surfaces of LPXs with hydrophilic PEG chains prevents them from aggregating in vitreous. In this way, LPXs are obtained that can freely move in vitreous, an absolute criterion for reaching the retina after intravitreal injection.


Assuntos
DNA/metabolismo , Dextranos/metabolismo , Fluoresceína-5-Isotiocianato/análogos & derivados , Terapia Genética , Metabolismo dos Lipídeos , Polietilenoglicóis/metabolismo , Corpo Vítreo/fisiologia , Animais , Bovinos , Eletroforese em Gel de Ágar , Fluoresceína-5-Isotiocianato/metabolismo , Técnicas de Transferência de Genes , Ácido Hialurônico/metabolismo , Microesferas , Poliestirenos/metabolismo
19.
J Control Release ; 167(2): 157-66, 2013 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-23402727

RESUMO

In this report we show that carrier-mediated delivery of mRNA may activate TLR3 signaling in respiratory cells. This activation of the innate immune system was accompanied with a massive production of type 1 interferons and other immunostimulating cytokines. The recognition of mRNA by the innate immune system was also associated with cell death, which proceeded in human respiratory cells via pyroptosis, a form of programmed cell death mediated by substantial overexpression of caspase-1. This indicated that the delivered mRNA is most likely also recognized by NOD-like receptors which regulate caspase-1 production. The viability of murine respiratory cells was less affected by mRNA transfection, which is in line with the lower transfection efficiency, lower innate immune response and the absence of a massive caspase-1 upregulation in these cells. Finally, we also demonstrated that the recognition of the delivered mRNA by the innate immune system had a negative effect on mRNA translation.


Assuntos
Imunidade Inata , RNA Mensageiro/administração & dosagem , Receptor 3 Toll-Like/metabolismo , Animais , Apoptose , Linhagem Celular Tumoral , Citocinas/metabolismo , DNA/administração & dosagem , Células HEK293 , Humanos , Lipossomos , Luciferases/metabolismo , Pulmão/citologia , Camundongos , Camundongos Endogâmicos BALB C , Transfecção
20.
J Control Release ; 172(3): 885-93, 2013 Dec 28.
Artigo em Inglês | MEDLINE | ID: mdl-24075924

RESUMO

Local extravasation and triggered drug delivery by use of ultrasound and microbubbles is a promising strategy to target drugs to their sites of action. In the past we have developed drug loaded microbubbles by coupling drug containing liposomes to the surface of microbubbles. Until now the advantages of this drug loading strategy have only been demonstrated in vitro. Therefore, in this paper, microbubbles with indocyanine green (ICG) containing liposomes at their surface or a mixture of ICG-liposomes and microbubbles was injected intravenously in mice. Immediately after injection the left hind leg was exposed to 1 MHz ultrasound and the ICG deposition was monitored 1, 4 and 7 days post-treatment by in vivo fluorescence imaging. In mice that received the ICG-liposome loaded microbubbles the local ICG deposition was, at each time point, about 2-fold higher than in mice that received ICG-liposomes mixed with microbubbles. We also showed that the perforations in the blood vessels allow the passage of ICG-liposomes up to 5h after microbubble and ultrasound treatment. An increase in tissue temperature to 41°C was observed in all ultrasound treated mice. However, ultrasound tissue heating was excluded to cause the local ICG deposition. We concluded that coupling of drug containing liposomes to microbubbles may increase ultrasound mediated drug delivery in vivo.


Assuntos
Sistemas de Liberação de Medicamentos/instrumentação , Verde de Indocianina/administração & dosagem , Lipossomos/química , Microbolhas , Ultrassom/instrumentação , Animais , Feminino , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA