Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 24(8)2023 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-37108337

RESUMO

Gastrointestinal stromal tumor (GIST), the most common sarcoma, is mainly caused by an oncogenic mutation in the KIT receptor tyrosine kinase. Targeting KIT using tyrosine kinase inhibitors, such as imatinib and sunitinib, provides substantial benefit; however, in most patients, the disease will eventually progress due to KIT secondary mutations leading to treatment failure. Understanding how GIST cells initially adapt to KIT inhibition should guide the selection of appropriate therapies to overcome the emergence of resistance. Several mechanisms have been broadly implicated in the resistance to imatinib anti-tumoral effects, including the reactivation of MAPK signaling upon KIT/PDGFRA targeted inhibition. This study provides evidence that LImb eXpression 1 (LIX1), a protein we identified as a regulator of the Hippo transducers YAP1 and TAZ, is upregulated upon imatinib or sunitinib treatment. LIX1 silencing in GIST-T1 cells impaired imatinib-induced MAPK signaling reactivation and enhanced imatinib anti-tumor effect. Our findings identified LIX1 as a key regulator of the early adaptative response of GIST cells to targeted therapies.


Assuntos
Antineoplásicos , Tumores do Estroma Gastrointestinal , Humanos , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Proteínas Relacionadas à Autofagia/genética , Resistencia a Medicamentos Antineoplásicos/genética , Tumores do Estroma Gastrointestinal/tratamento farmacológico , Tumores do Estroma Gastrointestinal/genética , Tumores do Estroma Gastrointestinal/patologia , Mesilato de Imatinib/farmacologia , Mesilato de Imatinib/uso terapêutico , Mutação , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-kit/metabolismo , Sunitinibe/farmacologia , Sunitinibe/uso terapêutico , Sistema de Sinalização das MAP Quinases
2.
J Nanobiotechnology ; 19(1): 236, 2021 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-34380479

RESUMO

Recently, we designed novel amphipathic cell-penetrating peptides, called WRAP, able to transfer efficiently siRNA molecules into cells. In order to gain more information about the relationship between amino acid composition, nanoparticle formation and cellular internalization of these peptides composed of only three amino acids (leucine, arginine and tryptophan), we performed a structure-activity relationship (SAR) study. First, we compared our WRAP1 and WRAP5 peptides with the C6M1 peptide also composed of the same three amino acids and showing similar behaviors in siRNA transfection. Afterwards, to further define the main determinants in the WRAP activity, we synthesized 13 new WRAP analogues harboring different modifications like the number and location of leucine and arginine residues, the relative location of tryptophan residues, as well as the role of the α-helix formation upon proline insertions within the native WRAP sequence. After having compared the ability of these peptides to form peptide-based nanoparticles (PBNs) using different biophysical methods and to induce a targeted gene silencing in cells, we established the main sequential requirements of the amino acid composition of the WRAP peptide. In addition, upon measuring the WRAP-based siRNA transfection ability into cells compared to several non-peptide transfection agents available on the markets, we confirmed that WRAP peptides induced an equivalent level of targeted gene silencing but in most of the cases with lower cell toxicity as clearly shown in clonogenic assays.


Assuntos
Bandagens Compressivas , Lipídeos/química , Nanopartículas/química , RNA Interferente Pequeno/química , Transfecção , Sequência de Aminoácidos , Linhagem Celular Tumoral , Peptídeos Penetradores de Células , Sistemas de Liberação de Medicamentos , Inativação Gênica/efeitos dos fármacos , Glioblastoma , Humanos , Indicadores e Reagentes/química , RNA Interferente Pequeno/farmacologia , Relação Estrutura-Atividade
3.
Pharmaceutics ; 13(5)2021 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-34069377

RESUMO

Small interfering RNA (siRNA) exhibits a high degree of specificity for targeting selected genes. They are efficient on cells in vitro, but in vivo siRNA therapy remains a challenge for solid tumor treatment as siRNAs display difficulty reaching their intracellular target. The present study was designed to show the in vivo efficiency of a new peptide (WRAP5), able to form peptide-based nanoparticles (PBN) that can deliver siRNA to cancer cells in solid tumors. WRAP5:siRNA nanoparticles targeting firefly luciferase (Fluc) were formulated and assayed on Fluc-expressing U87 glioblastoma cells. The mode of action of WRAP5:siRNA by RNA interference was first confirmed in vitro and then investigated in vivo using a combination of bioluminescent reporter genes. Finally, histological analyses were performed to elucidate the cell specificity of this PBN in the context of brain tumors. In vitro and in vivo results showed efficient knock-down of Fluc expression with no toxicity. WRAP5:siFluc remained in the tumor for at least 10 days in vivo. Messenger RNA (mRNA) analyses indicated a specific decrease in Fluc mRNA without affecting tumor growth. Histological studies identified PBN accumulation in the cytoplasm of tumor cells but also in glial and neuronal cells. Through in vivo molecular imaging, our findings established the proof of concept for specific gene silencing in solid tumors. The evidence generated could be translated into therapy for any specific gene in different types of tumors without cell type specificity but with high molecular specificity.

4.
J Vis Exp ; (166)2020 12 19.
Artigo em Inglês | MEDLINE | ID: mdl-33393518

RESUMO

Cell-penetrating peptides (CPPs) are defined as carriers that are able to cross the plasma membrane and to transfer a cargo into cells. One of the main common features required for this activity resulted from the interactions of CPPs with the plasma membrane (lipids) and more particularly with components of the extracellular matrix of the membrane itself (heparan sulphate). Indeed, independent of the direct translocation or the endocytosis-dependent internalization, lipid bilayers are involved in the internalization process both at the level of the plasma membrane and at the level of intracellular traffic (endosomal vesicles). In this article, we present a detailed protocol describing the different steps of a large unilamellar vesicles (LUVs) formulation, purification, characterization, and application in fluorescence leakage assay in order to detect possible CPP-membrane destabilization/interaction and to address their role in the internalization mechanism. LUVs with a lipid composition reflecting the plasma membrane content are generated in order to encapsulate both a fluorescent dye and a quencher. The addition of peptides in the extravesicular medium and the induction of peptide-membrane interactions on the LUVs might thus induce in a dose-dependent manner a significant increase in fluorescence revealing a leakage. Examples are provided here with the recently developed tryptophan (W)- and arginine (R)-rich Amphipathic Peptides (WRAPs), which showed a rapid and efficient siRNA delivery in various cell lines. Finally, the nature of these interactions and the affinity for lipids are discussed to understand and to improve the membrane translocation and/or the endosomal escape.


Assuntos
Bioensaio/métodos , Membrana Celular/metabolismo , Peptídeos Penetradores de Células/química , Arginina/química , Fluorescência , Bicamadas Lipídicas/química , Nanopartículas/química , Peptídeos/metabolismo , RNA Interferente Pequeno/metabolismo , Lipossomas Unilamelares/química
5.
Bioconjug Chem ; 30(3): 592-603, 2019 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-30586303

RESUMO

Delivery of small interfering RNA (siRNA) as a therapeutic tool is limited due to critical obstacles such as the cellular barrier, the negative charges of the siRNA molecule, and its instability in serum. Several siRNA delivery systems have been constructed using cell-penetrating peptides (CPPs) since the CPPs have shown a high potential for oligonucleotide delivery into the cells, especially by forming nanoparticles. In this study, we have developed a new family of short (15mer or 16mer) tryptophan-(W) and arginine-(R) rich Amphipathic Peptides (WRAP) able to form stable nanoparticles and to enroll siRNA molecules into cells. The lead peptides, WRAP1 and WRAP5, form defined nanoparticles smaller than 100 nm as characterized by biophysical methods. Furthermore, they have several benefits as oligonucleotide delivery tools such as the rapid encapsulation of the siRNA, the efficient siRNA delivery in several cell types, and the high gene silencing activity, even in the presence of serum. In conclusion, we have designed a new family of CPPs specifically dedicated for siRNA delivery through nanoparticle formation. Our results indicate that the WRAP family has significant potential for the safe, efficient, and rapid delivery of siRNA for diverse applications.


Assuntos
Peptídeos Penetradores de Células/química , Nanopartículas/química , Interferência de RNA , RNA Interferente Pequeno/administração & dosagem , Sequência de Aminoácidos , Linhagem Celular Tumoral , Humanos , Modelos Moleculares , RNA Interferente Pequeno/genética , Transfecção
6.
J Nanobiotechnology ; 15(1): 34, 2017 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-28454579

RESUMO

BACKGROUND: Small interfering RNAs (siRNAs) are powerful tools to control gene expression. However, due to their poor cellular permeability and stability, their therapeutic development requires a specific delivery system. Among them, cell-penetrating peptides (CPP) have been shown to transfer efficiently siRNA inside the cells. Recently we developed amphipathic peptides able to self-assemble with siRNAs as peptide-based nanoparticles and to transfect them into cells. However, despite the great potential of these drug delivery systems, most of them display a low resistance to proteases. RESULTS: Here, we report the development and characterization of a new CPP named RICK corresponding to the retro-inverso form of the CADY-K peptide. We show that RICK conserves the main biophysical features of its L-parental homologue and keeps the ability to associate with siRNA in stable peptide-based nanoparticles. Moreover the RICK:siRNA self-assembly prevents siRNA degradation and induces inhibition of gene expression. CONCLUSIONS: This new approach consists in a promising strategy for future in vivo application, especially for targeted anticancer treatment (e.g. knock-down of cell cycle proteins). Graphical abstract RICK-based nanoparticles: RICK peptides and siRNA self-assemble in peptide-based nanoparticles to penetrate into the cells and to induce target protein knock-down.


Assuntos
Peptídeos Penetradores de Células/química , Nanopartículas/química , Interferência de RNA , RNA Interferente Pequeno/administração & dosagem , Transfecção , Linhagem Celular Tumoral , Peptídeos Penetradores de Células/metabolismo , Genes Reporter , Humanos , Nanopartículas/metabolismo , Nanopartículas/ultraestrutura , Estabilidade de RNA , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo
7.
J Control Release ; 256: 79-91, 2017 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-28411182

RESUMO

Small interfering RNAs (siRNAs) present a strong therapeutic potential because of their ability to inhibit the expression of any desired protein. Recently, we developed the retro-inverso amphipathic RICK peptide as novel non-covalent siRNA carrier. This peptide is able to form nanoparticles (NPs) by self-assembling with the siRNA resulting in the fully siRNA protection based on its protease resistant peptide sequence. With regard to an in vivo application, we investigated here the influence of the polyethylene glycol (PEG) grafting to RICK NPs on their in vitro and in vivo siRNA delivery properties. A detailed structural study shows that PEGylation did not alter the NP formation (only decrease in zeta potential) regardless of the used PEGylation rates. Compared to the native RICK:siRNA NPs, low PEGylation rates (≤20%) of the NPs did not influence their cellular internalization capacity as well as their knock-down specificity (over-expressed or endogenous system) in vitro. Because the behavior of PEGylated NPs could differ in their in vivo application, we analyzed the repartition of fluorescent labeled NPs injected at the one-cell stage in zebrafish embryos as well as their pharmacokinetic (PK) profile after administration to mice. After an intra-cardiac injection of the PEGylated NPs, we could clearly determine that 20% PEG-RICK NPs reduce significantly liver and kidney accumulation. NPs with 20% PEGylation constitutes a modular, easy-to-handle drug delivery system which could be adapted to other types of functional moieties to develop safe and biocompatible delivery systems for the clinical application of RNAi-based cancer therapeutics.


Assuntos
Peptídeos Penetradores de Células/administração & dosagem , Nanopartículas/administração & dosagem , Polietilenoglicóis/administração & dosagem , RNA Interferente Pequeno/administração & dosagem , Proteína Serina-Treonina Quinase 2 de Interação com Receptor/administração & dosagem , Animais , Peptídeos Penetradores de Células/química , Cisteína/administração & dosagem , Cisteína/química , Embrião não Mamífero , Luciferases/genética , Masculino , Camundongos Endogâmicos C57BL , Nanopartículas/química , Polietilenoglicóis/química , RNA Interferente Pequeno/química , Proteína Serina-Treonina Quinase 2 de Interação com Receptor/química , Propriedades de Superfície , Peixe-Zebra
8.
Int J Pharm ; 509(1-2): 71-84, 2016 07 25.
Artigo em Inglês | MEDLINE | ID: mdl-27224007

RESUMO

RNA interference provides a powerful technology for specific gene silencing. Therapeutic applications of small interfering RNA (siRNA) however require efficient vehicles for stable complexation and intracellular delivery. In order to enhance their cell delivery, short amphipathic peptides called cell-penetrating peptides (CPPs) have been intensively developed for the last two decades. In this context, the secondary amphipathic peptide CADY has shown to form stable siRNA complexes and to improve their cellular uptake independent of the endosomal pathway. In the present work, we have described the parameters influencing CADY nanoparticle formation (buffers, excipients, presence of serum, etc.), and have followed in details the CPP:siRNA self-assembly. Once optimal conditions were determined, we have compared the ability of seven different CADY analogues to form siRNA-loaded nanoparticles compared to CADY:siRNA. First of all, we were able to show by biophysical methods that structural polymorphism (α-helix) is an important prerequisite for stable nanoparticle formation independently of occurring sequence mutations. Luciferase assays revealed that siRNA complexed to CADY-K (shorter version) shows better knock-down efficiency on Neuro2a-Luc(+) and B16-F10-Luc(+) cells compared to CADY:siRNA. Altogether, CADY-K is an ideal candidate for further application especially with regards to ex vivo or in vivo applications.


Assuntos
Peptídeos Penetradores de Células/química , Animais , Linhagem Celular Tumoral , Portadores de Fármacos/química , Inativação Gênica/efeitos dos fármacos , Interações Hidrofóbicas e Hidrofílicas , Camundongos , Nanopartículas/química , Interferência de RNA/fisiologia , RNA Interferente Pequeno/metabolismo
9.
Biochim Biophys Acta ; 1828(2): 499-509, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23000699

RESUMO

CADY is a cell-penetrating peptide spontaneously making non-covalent complexes with Short interfering RNAs (siRNAs) in water. Neither the structure of CADY nor that of the complexes is resolved. We have calculated and analyzed 3D models of CADY and of the non-covalent CADY-siRNA complexes in order to understand their formation and stabilization. Data from the ab initio calculations and molecular dynamics support that, in agreement with the experimental data, CADY is a polymorphic peptide partly helical. Taking into consideration the polymorphism of CADY, we calculated and compared several complexes with peptide/siRNA ratios of up to 40. Four complexes were run by using molecular dynamics. The initial binding of CADYs is essentially due to the electrostatic interactions of the arginines with siRNA phosphates. Due to a repetitive arginine motif (XLWR(K)) in CADY and to the numerous phosphate moieties in the siRNA, CADYs can adopt multiple positions at the siRNA surface leading to numerous possibilities of complexes. Nevertheless, several complex properties are common: an average of 14±1 CADYs is required to saturate a siRNA as compared to the 12±2 CADYs experimentally described. The 40 CADYs/siRNA that is the optimal ratio for vector stability always corresponds to two layers of CADYs per siRNA. When siRNA is covered by the first layer of CADYs, the peptides still bind despite the electrostatic repulsion. The peptide cage is stabilized by hydrophobic CADY-CADY contacts thanks to CADY polymorphism. The analysis demonstrates that the hydrophobicity, the presence of several positive charges and the disorder of CADY are mandatory to make stable the CADY-siRNA complexes.


Assuntos
Peptídeos Penetradores de Células/química , Peptídeos/química , RNA Interferente Pequeno/metabolismo , Motivos de Aminoácidos , Arginina/química , Vetores Genéticos , Interações Hidrofóbicas e Hidrofílicas , Modelos Moleculares , Conformação Molecular , Simulação de Dinâmica Molecular , Ligação Proteica , Estrutura Secundária de Proteína , Eletricidade Estática , Termodinâmica , Fatores de Tempo
10.
Curr Pharm Des ; 19(16): 2869-77, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23140452

RESUMO

Although siRNA consist in very promising therapeutics, their clinical development is limited by several biological barriers including low cellular permeability, poor stability and lack of tissue specificity. Therefore the Achilles' heel for siRNA-based therapy is directly related to the lack of efficient system to promote their delivery. During the last two decades, cell-penetrating peptides (CPPs) have been widely developed to enhance the cellular delivery of therapeutics. In this context we have elaborated a new strategy based on self-assembling peptide-based nanoparticles. The CADY peptide is a 20-residue secondary amphipathic peptide which is able to spontaneously self associate with siRNA with a strong affinity, by combining both electrostatic and hydrophobic interactions, to form stable nanoparticles. Investigations of both physico-chemical properties and cellular siRNA delivery revealed that the CADY/siRNA complexes were able to enter a wide variety of cell lines by a mechanism independent of any endocytotic pathway. In addition a deeper understanding of the self assembly of CADY molecules around siRNA leads to a "raspberry"-like nanoparticle architecture which provides new perspectives for the CADY/siRNA formulations. Finally the robustness of the biological response infers that peptide-based nanoparticle technology holds a strong promise for therapeutic applications. The present review deals with most of the biophysical characteristics as well as the cellular mechanism and cellular applications of CADY/siRNA nanoparticles.


Assuntos
Peptídeos Penetradores de Células/metabolismo , Peptídeos/metabolismo , RNA Interferente Pequeno/administração & dosagem , Animais , Técnicas de Transferência de Genes , Humanos , Interações Hidrofóbicas e Hidrofílicas , Nanopartículas , Eletricidade Estática
12.
Chem Biol ; 18(5): 631-41, 2011 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-21609844

RESUMO

A recent screen for compounds that selectively targeted pancreatic cancer cells isolated UA62784. We found that UA62784 inhibits microtubule polymerization in vitro. UA62784 interacts with tubulin dimers ten times more potently than colchicine, vinblastine, or nocodazole. Competition experiments revealed that UA62784 interacts with tubulin at or near the colchicine-binding site. Nanomolar doses of UA62784 promote the accumulation of mammalian cells in mitosis, due to aberrant mitotic spindles, as shown by immunofluorescence and live cell imaging. Treatment of cancerous cell lines with UA62784 is lethal, following activation of apoptosis signaling. By monitoring mitotic spindle perturbations and apoptosis, we found that the effects of UA62784 and of some known microtubule-depolymerizing drugs are additive. Finally, high content screening of H2B-GFP HeLa cells revealed that low doses of UA62784 and vinblastine potentiate each other to inhibit proliferation.


Assuntos
Microtúbulos/efeitos dos fármacos , Oxazóis/toxicidade , Moduladores de Tubulina/toxicidade , Tubulina (Proteína)/química , Xantonas/toxicidade , Apoptose , Sítios de Ligação , Proteínas Cromossômicas não Histona/antagonistas & inibidores , Proteínas Cromossômicas não Histona/metabolismo , Colchicina/farmacologia , Dimerização , Células HeLa , Humanos , Mitose , Nocodazol/farmacologia , Oxazóis/química , Fuso Acromático/efeitos dos fármacos , Tubulina (Proteína)/metabolismo , Moduladores de Tubulina/química , Vimblastina/farmacologia , Xantonas/química
13.
Methods Mol Biol ; 683: 41-56, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21053121

RESUMO

Due to the poor permeability of the plasma membrane, several strategies are designed to enhance the transfer of therapeutics into cells. Over the last 20 years, small peptides called Cell-Penetrating Peptides (CPPs) have been widely developed to improve the cellular delivery of biomolecules. These small peptides derive from protein transduction domains, chimerical constructs, or model sequences. Several CPPs are primary or secondary amphipathic peptides, depending on whether the distribution of their hydrophobic and hydrophilic domains occurs from their amino-acid sequence or through α-helical folding. Most of the CPPs are able to deliver different therapeutics such as nucleic acids or proteins in vitro and in vivo. Although their mechanisms of internalization are varied and controversial, the understanding of the intrinsic features of CPPs is essential for future developments. This chapter describes several protocols for the investigation of biophysical properties of amphipathic CPPs. Surface physics approaches are specifically applied to characterize the interactions of amphipathic peptides with model membranes. Circular dichroism and infra-red spectroscopy allow the identification of their structural state. These methods are exemplified by the analyses of the main biophysical features of the cell-penetrating peptides MPG, Pep-1, and CADY.


Assuntos
Biofísica/métodos , Membrana Celular/metabolismo , Peptídeos Penetradores de Células/química , Peptídeos Penetradores de Células/metabolismo , Interações Hidrofóbicas e Hidrofílicas , Ar , Sequência de Aminoácidos , Membrana Celular/química , Dicroísmo Circular , Cisteamina/análogos & derivados , Cisteamina/química , Cisteamina/metabolismo , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/metabolismo , Dados de Sequência Molecular , Peptídeos/química , Peptídeos/metabolismo , Fosfolipídeos/química , Fosfolipídeos/metabolismo , Ligação Proteica , Espectroscopia de Infravermelho com Transformada de Fourier , Lipossomas Unilamelares/química , Lipossomas Unilamelares/metabolismo , Água/química
14.
Biochemistry ; 49(16): 3393-402, 2010 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-20302329

RESUMO

Delivery of siRNA remains a major limitation to their clinical application, and several technologies have been proposed to improve their cellular uptake. We recently described a peptide-based nanoparticle system for efficient delivery of siRNA into primary cell lines: CADY. CADY is a secondary amphipathic peptide that forms stable complexes with siRNA and improves their cellular uptake independently of the endosomal pathway. In the present work, we have combined molecular modeling, spectroscopy, and membrane interaction approaches in order to gain further insight into CADY/siRNA particle mechanism of interaction with biological membrane. We demonstrate that CADY forms stable complexes with siRNA and binds phospholipids tightly, mainly through electrostatic interactions. Binding to siRNA or phospholipids triggers a conformational transition of CADY from an unfolded state to an alpha-helical structure, thereby stabilizing CADY/siRNA complexes and improving their interactions with cell membranes. Therefore, we propose that CADY cellular membrane interaction is driven by its structural polymorphism which enables stabilization of both electrostatic and hydrophobic contacts with surface membrane proteoglycan and phospholipids.


Assuntos
Peptídeos/química , RNA Interferente Pequeno/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Transporte Biológico , Linhagem Celular , Membrana Celular/metabolismo , Dicroísmo Circular , Glicosaminoglicanos/química , Glicosaminoglicanos/metabolismo , Lipossomos/química , Lipossomos/metabolismo , Micelas , Modelos Moleculares , Dados de Sequência Molecular , Distribuição Normal , Oligorribonucleotídeos/química , Peptídeos/síntese química , Peptídeos/metabolismo , Fosfolipídeos/química , Fosfolipídeos/metabolismo , Conformação Proteica , Estrutura Secundária de Proteína/genética , RNA Interferente Pequeno/metabolismo
15.
Biochim Biophys Acta ; 1798(6): 1119-28, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20214875

RESUMO

The clinical use of efficient therapeutic agents is often limited by the poor permeability of the biological membranes. In order to enhance their cell delivery, short amphipathic peptides called cell-penetrating peptides (CPPs) have been intensively developed for the last two decades. CPPs are based either on protein transduction domains, model peptide or chimeric constructs and have been used to deliver cargoes into cells through either covalent or non-covalent strategies. Although several parameters are simultaneously involved in their internalization mechanism, recent focuses on CPPs suggested that structural properties and interactions with membrane phospholipids could play a major role in the cellular uptake mechanism. In the present work, we report a comparative analysis of the structural plasticity of 10 well-known CPPs as well as their ability to interact with phospholipid membranes. We propose a new classification of CPPs based on their structural properties, affinity for phospholipids and internalization pathways already reported in the literature.


Assuntos
Permeabilidade da Membrana Celular , Membrana Celular/química , Sistemas de Liberação de Medicamentos , Peptídeos/química , Fosfolipídeos/química , Animais , Membrana Celular/metabolismo , Humanos , Peptídeos/metabolismo , Fosfolipídeos/metabolismo , Relação Estrutura-Atividade
17.
Curr Pharm Des ; 14(34): 3656-65, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-19075741

RESUMO

One of the major challenges for new therapeutics molecules to enter the clinic remains improving their bioavailability and cellular uptake. Therefore, delivery has become a key stone in therapeutic development and several technologies have been designed to improve cellular uptake of therapeutic molecules, including cell-penetrating peptides (CPPs) or protein transduction domain (PTD). PTDs or CPPs were discovered twenty years ago, based on the potency of several proteins to enter cells and nowadays, numerous peptide carriers have been described and successfully applied for ex vivo and in vivo delivery of varying therapeutic molecules. Two CPP-strategies have been reported; the first one requires chemical linkage between the drug and the carrier for cellular drug internalization and the second is based on the formation of stable complexes with drugs depending on their chemical nature. Peptide-Based-Nanoparticle Devices (PBND), correspond to short amphipathic peptides able to form stable nanoparticles with proteins and/or nucleic acids. Three PBND-families, PEP, MPG and CADY have been described, these carriers mainly enter cells independently of the endosomal pathway and efficiently deliver cargoes in a large variety of challenging cell lines as well as in animal models. This review will focus on the structure/function relationship of the PBND: CADY, PEP and MPG, in the general context of drug delivery. It will also highlight the requirement of primary or secondary amphipathic carriers for in vitro and in vivo delivery of therapeutic molecules and provide an update of their pre-clinical evaluation.


Assuntos
Portadores de Fármacos , Nanopartículas , Peptídeos/administração & dosagem , Sequência de Aminoácidos , Dados de Sequência Molecular , Ácidos Nucleicos/administração & dosagem , Peptídeos/química
18.
Biol Cell ; 100(4): 201-17, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18341479

RESUMO

The recent discovery of new potent therapeutic molecules which do not reach the clinic due to poor delivery and low bioavailability have made the delivery of molecules a keystone in therapeutic development. Several technologies have been designed to improve cellular uptake of therapeutic molecules, including CPPs (cell-penetrating peptides), which represent a new and innovative concept to bypass the problem of bioavailability of drugs. CPPs constitute very promising tools and have been successfully applied for in vivo. Two CPP strategies have been described to date; the first one requires chemical linkage between the drug and the carrier for cellular drug internalization, and the second is based on the formation of stable complexes with drugs, depending on their chemical nature. The Pep and MPG families are short amphipathic peptides, which form stable nanoparticles with proteins and nucleic acids respectively. MPG- and Pep-based nanoparticles enter cells independently of the endosomal pathway and efficiently deliver cargoes, in a fully biologically active form, into a large variety of cell lines, as well as in animal models. This review focuses on the structure-function relationship of non-covalent MPG and Pep-1 strategies, and their requirement for cellular uptake of biomolecules and applications in cultured cells and animal models.


Assuntos
Portadores de Fármacos/química , Peptídeos/farmacocinética , Sequência de Aminoácidos , Animais , Disponibilidade Biológica , Sistemas de Liberação de Medicamentos , Humanos , Dados de Sequência Molecular , Nanopartículas , Oligopeptídeos/química , Ácidos Nucleicos Peptídicos/administração & dosagem , Ácidos Nucleicos Peptídicos/uso terapêutico , Peptídeos/química , Peptídeos/uso terapêutico , Relação Estrutura-Atividade
19.
Adv Drug Deliv Rev ; 60(4-5): 537-47, 2008 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-18037526

RESUMO

The recent discovery of new potent therapeutic molecules which do not reach the clinic due to poor delivery and low bioavailability have made of delivery a key stone in therapeutic development. Several technologies have been designed to improve cellular uptake of therapeutic molecules, including cell-penetrating peptides (CPPs), which have been successfully applied for in vivo delivery of biomolecules and constitute very promising tools. Distinct families of CPPs have been described; some require chemical linkage between the drug and the carrier for cellular drug internalization while others like Pep-and MPG-families, form stable complexes with drugs depending on their chemical nature. Pep and MPG are short amphipathic peptides, which form stable nanoparticles with proteins and nucleic acids respectively. MPG and Pep based nanoparticles enter cells independently of the endosomal pathway and efficiently deliver cargoes in a fully biologically active form into a large variety of cell lines as well as in animal models. This review will focus on the mechanisms of non-covalent MPG and Pep-1 strategies and their applications in cultured cells and animal models.


Assuntos
Sistemas de Liberação de Medicamentos/métodos , Ácidos Nucleicos/administração & dosagem , Peptídeos/administração & dosagem , Proteínas/química , Animais , Membrana Celular/metabolismo , Permeabilidade da Membrana Celular , Sistemas de Liberação de Medicamentos/tendências , Humanos , Ácidos Nucleicos/química , Ácidos Nucleicos/farmacocinética , Peptídeos/química , Peptídeos/metabolismo , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/química , RNA Interferente Pequeno/farmacocinética
20.
Methods Mol Biol ; 386: 299-308, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-18604951

RESUMO

Control of gene expression using RNA interference (RNAi) technology constitutes a method of choice for investigating gene function in mammalian cells. However, like most oligonucleotide-based strategies, the major limitation of interfering RNA is their poor cellular uptake due to low permeability of the cell membrane to nucleic acids. Several strategies have been developed to improve delivery of oligonucleotides both in cultured cells and in vivo. So far, there is no universal method for their delivery, as they all present several limitations. Peptide-based strategies have been demonstrated to improve the cellular uptake of nucleic acids both in cultured cell and in vivo. This chapter describes a new peptide-based gene delivery system, MPG, which forms stable noncovalent complexes with oligonucleotides and promotes their delivery into a large panel of cell lines without the need for prior chemical covalent coupling. Protocols are described for both adherent and suspension cell lines.


Assuntos
Portadores de Fármacos/química , Ácidos Nucleicos/administração & dosagem , Peptídeos/química , Sequência de Aminoácidos , Animais , Linhagem Celular , Proteínas de Ligação a DNA/química , Sistemas de Liberação de Medicamentos , Células HeLa , Humanos , Células Jurkat , Lipídeos/química , Substâncias Macromoleculares/química , Camundongos , Biologia Molecular/métodos , Dados de Sequência Molecular , Ácidos Nucleicos/genética , Interferência de RNA , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA