Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
1.
J Am Chem Soc ; 144(41): 19127-19136, 2022 10 19.
Article in English | MEDLINE | ID: mdl-36206443

ABSTRACT

We present herein the "vermellogens", a new class of pH-responsive viologen analogues, which replace the direct linking between para-substituted pyridinium moieties within those by a hydrazone functional group. A series of such compounds have been efficiently synthesized in aqueous media by hydrazone exchange reactions, displaying a marked pH-responsivity. Furthermore, the parent N,N'-dimethylated "vermellogen": the "red thread", an analogue of the herbicide paraquat and used herein as a representative model of the series, showed anion-recognition abilities, non-reversible electrochemical behavior, and non-toxicity of the modified bis-pyridinium core. The host-guest chemistry for the "red thread" with the CB[7,8] macrocyclic receptors has been extensively studied experimentally and by dispersion corrected density functional theory methods, showing a parallel behavior to that previously described for the herbicide but, crucially, swapping the well-known redox reactive capabilities of the viologen-based inclusion complexes by acid-base supramolecular responsiveness.


Subject(s)
Herbicides , Viologens , Paraquat/toxicity , Anions , Hydrogen-Ion Concentration , Hydrazones
2.
FASEB J ; 32(10): 5298-5311, 2018 10.
Article in English | MEDLINE | ID: mdl-29688810

ABSTRACT

The continuous presence of TGF-ß is critically important to induce effective chondrogenesis. To investigate chondrogenesis in a cartilage defect, we tested the hypothesis that the implantation of TGF-ß1-releasing scaffolds improves very early cartilage repair in vivo. Spatiotemporal controlled release of TGF-ß1 was achieved from multiblock scaffolds that were implanted in osteochondral defects in the medial femoral condyles of adult minipigs. We observed a sustained presence of TGF-ß1 at 4 wk in vivo, which significantly promoted structural aspects of early overall cartilage repair, especially cellularity, cellular morphology, and safranin O staining intensity. Furthermore, early aggrecan and type II collagen production were both increased in specific topographic patterns in cartilaginous repair tissue. Sustained release of TGF-ß1 also increased cell numbers and proliferation, staining intensities for the stem cell surface marker, CD105, and number of stromal cell-derived factor-1 (SDF-1) -positive cells within cartilaginous repair tissue. These data identify a mechanism by which TGF-ß1 modulates early chondrogenesis by primarily increasing the number of progenitor cells arising from the subchondral bone marrow compartment via the SDF-1/chemokine (CXC motif) receptor 4 pathway, their proliferation, differentiation, and extracellular matrix deposition in specific topographic patterns, highlighting the pivotal role played by TGF-ß1 during this crucial phase.-Asen, A.-K., Goebel, L., Rey-Rico, A., Sohier, J., Zurakowski, D., Cucchiarini, M., Madry, H. Sustained spatiotemporal release of TGF-ß1 confers enhanced very early chondrogenic differentiation during osteochondral repair in specific topographic patterns.


Subject(s)
Cartilage , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Chondrogenesis/drug effects , Transforming Growth Factor beta , Animals , Cartilage/injuries , Cartilage/metabolism , Cartilage/physiology , Chemokine CXCL12/metabolism , Drug Implants , Endoglin/metabolism , Receptors, CXCR4/metabolism , Swine , Swine, Miniature , Transforming Growth Factor beta/pharmacokinetics , Transforming Growth Factor beta/pharmacology
3.
Int J Mol Sci ; 20(10)2019 May 27.
Article in English | MEDLINE | ID: mdl-31137788

ABSTRACT

Application of chondroreparative gene vectors in cartilage defects is a powerful approach to directly stimulate the regenerative activities of bone-marrow-derived mesenchymal stem cells (MSCs) that repopulate such lesions. Here, we investigated the ability of combined recombinant adeno-associated virus (rAAV) vector-mediated delivery of the potent transforming growth factor beta (TGF-ß) and insulin-like growth factor I (IGF-I) to enhance the processes of chondrogenic differentiation in human MSCs (hMSCs) relative to individual candidate treatments and to reporter (lacZ) gene condition. The rAAV-hTGF-ß and rAAV-hIGF-I vectors were simultaneously provided to hMSC aggregate cultures (TGF-ß/IGF-I condition) in chondrogenic medium over time (21 days) versus TGF-ß/lacZ, IGF-I/lacZ, and lacZ treatments at equivalent vector doses. The cultures were then processed to monitor transgene (co)-overexpression, the levels of biological activities in the cells (cell proliferation, matrix synthesis), and the development of a chondrogenic versus osteogenic/hypertrophic phenotype. Effective, durable co-overexpression of TGF-ß with IGF-I via rAAV enhanced the proliferative, anabolic, and chondrogenic activities in hMSCs versus lacZ treatment and reached levels that were higher than those achieved upon single candidate gene transfer, while osteogenic/hypertrophic differentiation was delayed over the period of time evaluated. These findings demonstrate the potential of manipulating multiple therapeutic rAAV vectors as a tool to directly target bone-marrow-derived MSCs in sites of focal cartilage defects and to locally enhance the endogenous processes of cartilage repair.


Subject(s)
Chondrocytes/metabolism , Chondrogenesis , Insulin-Like Growth Factor I/genetics , Mesenchymal Stem Cells/metabolism , Transforming Growth Factor beta/genetics , Cell Differentiation , Cells, Cultured , Chondrocytes/cytology , Gene Transfer Techniques , Genetic Vectors/genetics , Humans , Insulin-Like Growth Factor I/metabolism , Mesenchymal Stem Cells/cytology , Parvovirinae/genetics , Transforming Growth Factor beta/metabolism
4.
Mol Pharm ; 15(7): 2816-2826, 2018 07 02.
Article in English | MEDLINE | ID: mdl-29763553

ABSTRACT

Recombinant adeno-associated virus (rAAV) vectors are well suited carriers to provide durable treatments for human osteoarthritis (OA). Controlled release of rAAV from polymeric micelles was already shown to increase both the stability and bioactivity of the vectors while overcoming barriers, precluding effective gene transfer. In the present study, we examined the convenience of delivering rAAV vectors via poly(ethylene oxide) (PEO) and poly(propylene oxide) (PPO) polymeric (PEO-PPO-PEO) micelles to transfer and overexpress the transcription factor SOX9 in monolayers of human OA chondrocytes and in experimentally created human osteochondral defects. Human osteoarthritic (OA) chondrocytes and human osteochondral defect models were produced using human OA cartilage obtained from patients subjected to total knee arthroplasty. Samples were genetically modified by adding a rAAV-FLAG-h sox9 vector in its free form or via polymeric micelles for 10 days relative to control conditions (unmodified cells). The effects of sox9 overexpression in human OA cartilage samples were monitored by biochemical, histological, and immunohistochemical analyses. Delivery of rAAV-FLAG-h sox9 via polymeric micelles enhanced the levels of sox9 expression compared with free vector administration, resulting in increased proteoglycan deposition and in a stimulated cell proliferation index in OA chondrocytes. Moreover, higher production of type II collagen and decreased hypertrophic events were noted in osteochondral defect cultures when compared with control conditions. Controlled therapeutic rAAV sox9 gene delivery using PEO-PPO-PEO micelles is a promising, efficient tool to promote the remodelling of human OA cartilage.


Subject(s)
Chondrocytes/metabolism , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Osteoarthritis/therapy , SOX9 Transcription Factor/genetics , Cartilage, Articular/cytology , Cartilage, Articular/metabolism , Cells, Cultured , Dependovirus/genetics , Genetic Vectors/genetics , Humans , Micelles , Osteoarthritis/pathology , Polyethylene Glycols/chemistry , Polymers/chemistry , Primary Cell Culture , Propylene Glycols/chemistry , Transduction, Genetic/methods
5.
Int J Mol Sci ; 19(3)2018 Mar 08.
Article in English | MEDLINE | ID: mdl-29518011

ABSTRACT

Lineal (poloxamers or Pluronic®) or X-shaped (poloxamines or Tetronic®) amphiphilic tri-block copolymers of poly(ethylene oxide) and poly(propylene oxide) (PEO-PPO-PEO) have been broadly explored for controlled drug delivery in different regenerative medicine approaches. The ability of these copolymers to self-assemble as micelles and to undergo sol-to-gel transitions upon heating has endowed the denomination of "smart" or "intelligent" systems. The use of PEO-PPO-PEO copolymers as gene delivery systems is a powerful emerging strategy to improve the performance of classical gene transfer vectors. This review summarizes the state of art of the application of PEO-PPO-PEO copolymers in both nonviral and viral gene transfer approaches and their potential as gene delivery systems in different regenerative medicine approaches.


Subject(s)
Drug Delivery Systems/methods , Gene Transfer Techniques , Polyethylene Glycols/chemistry , Propylene Glycols/chemistry , Regenerative Medicine/methods , Humans , Micelles , Polyethylene Glycols/adverse effects , Propylene Glycols/adverse effects
6.
Int J Mol Sci ; 19(9)2018 Sep 05.
Article in English | MEDLINE | ID: mdl-30189664

ABSTRACT

The repair of focal articular cartilage defects remains a problem. Combining gene therapy with tissue engineering approaches using bone marrow-derived mesenchymal stem cells (MSCs) may allow the development of improved options for cartilage repair. Here, we examined whether a three-dimensional fibrin-polyurethane scaffold provides a favorable environment for the effective chondrogenic differentiation of human MSCs (hMSCs) overexpressing the cartilage-specific SOX9 transcription factor via recombinant adeno-associated virus (rAAV) -mediated gene transfer cultured in a hydrodynamic environment in vitro. Sustained SOX9 expression was noted in the constructs for at least 21 days, the longest time point evaluated. Such spatially defined SOX9 overexpression enhanced proliferative, metabolic, and chondrogenic activities compared with control (reporter lacZ gene transfer) treatment. Of further note, administration of the SOX9 vector was also capable of delaying premature hypertrophic and osteogenic differentiation in the constructs. This enhancement of chondrogenesis by spatially defined overexpression of human SOX9 demonstrate the potential benefits of using rAAV-modified hMSCs seeded in fibrin-polyurethane scaffolds as a promising approach for implantation in focal cartilage lesions to improve cartilage repair.


Subject(s)
Cell Differentiation , Chondrogenesis , Fibrin/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Polyurethanes/metabolism , Cell Differentiation/genetics , Chondrogenesis/genetics , Dependovirus/genetics , Gene Expression , Genetic Vectors/genetics , Humans , Hydrodynamics , SOX9 Transcription Factor/genetics
7.
J Cell Mol Med ; 21(11): 2748-2758, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28467017

ABSTRACT

Implantation of peripheral blood aspirates induced towards chondrogenic differentiation upon genetic modification in sites of articular cartilage injury may represent a powerful strategy to enhance cartilage repair. Such a single-step approach may be less invasive than procedures based on the use of isolated or concentrated MSCs, simplifying translational protocols in patients. In this study, we provide evidence showing the feasibility of overexpressing the mitogenic and pro-anabolic insulin-like growth factor I (IGF-I) in human peripheral blood aspirates via rAAV-mediated gene transfer, leading to enhanced proliferative and chondrogenic differentiation (proteoglycans, type-II collagen, SOX9) activities in the samples relative to control (reporter rAAV-lacZ) treatment over extended periods of time (at least 21 days, the longest time-point evaluated). Interestingly, IGF-I gene transfer also triggered hypertrophic, osteo- and adipogenic differentiation processes in the aspirates, suggesting that careful regulation of IGF-I expression may be necessary to contain these events in vivo. Still, the current results demonstrate the potential of targeting human peripheral blood aspirates via therapeutic rAAV transduction as a novel, convenient tool to treat articular cartilage injuries.


Subject(s)
Chondrocytes/metabolism , Chondrogenesis/genetics , Dependovirus/genetics , Insulin-Like Growth Factor I/genetics , Mesenchymal Stem Cells/metabolism , Biomarkers/metabolism , Cell Differentiation , Cell Proliferation , Cell- and Tissue-Based Therapy , Chondrocytes/cytology , Collagen Type II/genetics , Collagen Type II/metabolism , Dependovirus/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Humans , Insulin-Like Growth Factor I/metabolism , Lac Operon , Mesenchymal Stem Cells/cytology , Primary Cell Culture , Proteoglycans/genetics , Proteoglycans/metabolism , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Transduction, Genetic/methods , Transgenes
8.
Connect Tissue Res ; 58(3-4): 317-328, 2017.
Article in English | MEDLINE | ID: mdl-28051883

ABSTRACT

The meniscus plays a pivotal role to preserve the knee joint homeostasis. Lesions to the meniscus are frequent, have a reduced ability to heal, and may induce tibiofemoral osteoarthritis. Current reconstructive therapeutic options mainly focus on the treatment of lesions in the peripheral vascularized region. In contrast, few approaches are capable of stimulating repair of damaged meniscal tissue in the central, avascular portion. Tissue engineering approaches are of high interest to repair or replace damaged meniscus tissue in this area. Hydrogel-based biomaterials are of special interest for meniscus repair as its inner part contains relatively high proportions of proteoglycans which are responsible for the viscoelastic compressive properties and hydration grade. Hydrogels exhibiting high water content and providing a specific three-dimensional (3D) microenvironment may be engineered to precisely resemble this topographical composition of the meniscal tissue. Different polymers of both natural and synthetic origins have been manipulated to produce hydrogels hosting relevant cell populations for meniscus regeneration and provide platforms for meniscus tissue replacement. So far, these compounds have been employed to design controlled delivery systems of bioactive molecules involved in meniscal reparative processes or to host genetically modified cells as a means to enhance meniscus repair. This review describes the most recent advances on the use of hydrogels as platforms for precision meniscus tissue engineering.


Subject(s)
Hydrogels/pharmacology , Meniscus/physiology , Tissue Engineering/methods , Animals , Drug Delivery Systems , Genetic Therapy , Humans , Meniscus/drug effects , Meniscus/pathology , Meniscus/surgery , Regeneration/drug effects
9.
J Cell Mol Med ; 20(3): 430-40, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26808466

ABSTRACT

Genetic modification of marrow concentrates may provide convenient approaches to enhance the chondrogenic differentiation processes and improve the repair capacities in sites of cartilage defects following administration in the lesions. Here, we provided clinically adapted recombinant adeno-associated virus (rAAV) vectors to human bone marrow aspirates to promote the expression of the potent transforming growth factor beta (TGF-ß) as a means to regulate the biological and chondrogenic activities in the samples in vitro. Successful TGF-ß gene transfer and expression via rAAV was reached relative to control (lacZ) treatment (from 511.1 to 16.1 pg rhTGF-ß/mg total proteins after 21 days), allowing to durably enhance the levels of cell proliferation, matrix synthesis, and chondrogenic differentiation. Strikingly, in the conditions applied here, application of the candidate TGF-ß vector was also capable of reducing the hypertrophic and osteogenic differentiation processes in the aspirates, showing the potential benefits of using this particular vector to directly modify marrow concentrates to generate single-step, effective approaches that aim at improving articular cartilage repair in vivo.


Subject(s)
Bone Marrow Cells/metabolism , Chondrogenesis , Dependovirus/genetics , Transforming Growth Factor beta/genetics , Cell Proliferation , Cells, Cultured , Gene Expression , Genetic Vectors , Humans , Transduction, Genetic , Transforming Growth Factor beta/biosynthesis
10.
Int Orthop ; 38(9): 1931-6, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24957484

ABSTRACT

PURPOSE: Our aim was to study the effect of three-dimensional (3D) environment and overexpression of human fibroblast growth factor 2 (FGF-2) on meniscal fibrochondrocytes in vitro. METHODS: Human meniscal fibrochondrocytes were transfected with expression plasmid vectors carrying the Photinus pyralis luciferase gene, the Escherichia coli ß-galactosidase gene or a human FGF-2 cDNA. Modified fibrochondrocytes were cultivated in 3D alginate hydrogel or cell pellets or in 2D monolayer culture. RESULTS: The levels of luciferase activity showed a peak at day two and returned to baseline levels by day 11, regardless of the type of cultivation. Both 3D environments supported the secretion of human FGF-2 protein upon FGF-2 transfection. Overexpression of human FGF-2 by genetically modified human meniscal fibrochondrocytes stimulated proliferation but not glycosaminoglycan synthesis only in 3D culture. Culture in alginate spheres resulted in a larger difference in cell numbers compared with pellet cultures. CONCLUSIONS: Three-dimensional alginate spheres are well suited for the culture of genetically modified human meniscal fibrochondrocytes. These data are of value for cell-based approaches to meniscal repair using genetically modified human meniscal fibrochondrocytes overexpressing human FGF-2.


Subject(s)
Cell Culture Techniques/methods , Cell Transplantation/methods , Chondrocytes/transplantation , Fibroblast Growth Factor 2/metabolism , Gene Transfer Techniques , Menisci, Tibial/surgery , Alginates , Animals , Cell Survival , Cells, Cultured , Chondrocytes/cytology , Chondrocytes/metabolism , Escherichia coli/genetics , Fibroblast Growth Factor 2/genetics , Fireflies/genetics , Genetic Therapy/methods , Genetic Vectors , Glucuronic Acid , Hexuronic Acids , Humans , In Vitro Techniques , Menisci, Tibial/cytology , Menisci, Tibial/metabolism , Up-Regulation
11.
J Transl Med ; 11: 211, 2013 Sep 13.
Article in English | MEDLINE | ID: mdl-24034904

ABSTRACT

BACKGROUND: Therapeutic gene transfer is of significant value to elaborate efficient, durable treatments against human osteoarthritis (OA), a slow, progressive, and irreversible disorder for which there is no cure to date. METHODS: Here, we directly applied a recombinant adeno-associated virus (rAAV) vector carrying a human transforming growth factor beta (TGF-ß) gene sequence to primary human normal and OA chondrocytes in vitro and cartilage explants in situ to monitor the stability of transgene expression and the effects of the candidate pleiotropic factor upon the regenerative cellular activities over time. RESULTS: Efficient, prolonged expression of TGF-ß achieved via rAAV gene transfer enhanced both the proliferative, survival, and anabolic activities of cells over extended periods of time in all the systems evaluated (at least for 21 days in vitro and for up to 90 days in situ) compared with control (reporter) vector delivery, especially in situ where rAAV-hTGF-ß allowed for a durable remodeling of OA cartilage. Notably, sustained rAAV production of TGF-ß in OA cartilage advantageously reduced the expression of key OA-associated markers of chondrocyte hypertrophic and terminal differentiation (type-X collagen, MMP-13, PTHrP, ß-catenin) while increasing that of protective TIMPs and of the TGF-ß receptor I in a manner that restored a favorable ALK1/ALK5 balance. Of note, the levels of activities in TGF-ß-treated OA cartilage were higher than those of normal cartilage, suggesting that further optimization of the candidate treatment (dose, duration, localization, presence of modulating co-factors) will most likely be necessary to reproduce an original cartilage surface in relevant models of experimental OA in vivo without triggering potentially adverse effects. CONCLUSIONS: The present findings show the ability of rAAV-mediated TGF-ß gene transfer to directly remodel human OA cartilage by activating the biological, reparative activities and by regulating hypertrophy and terminal differentiation in damaged chondrocytes as a potential treatment for OA or for other disorders of the cartilage that may require transplantation of engineered cells.


Subject(s)
Cartilage, Articular/pathology , Dependovirus/metabolism , Osteoarthritis/therapy , Recombination, Genetic , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/therapeutic use , Aged , Apoptosis/drug effects , Cell Proliferation/drug effects , Chondrocytes/drug effects , Chondrocytes/pathology , Humans , Protective Agents/pharmacology , Recombination, Genetic/genetics , Signal Transduction/drug effects , Transduction, Genetic , Transforming Growth Factor beta/pharmacology
12.
J Control Release ; 362: 606-619, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37678437

ABSTRACT

Articular cartilage injuries are very frequent lesions that if left untreated may degenerate into osteoarthritis. Gene transfer to mesenchymal stem cells (MSCs) provides a powerful approach to treat these lesions by promoting their chondrogenic differentiation into the appropriate cartilage phenotype. Non-viral vectors constitute the safest gene transfer tools, as they avoid important concerns of viral systems including immunogenicity and insertional mutagenesis. However, non-viral gene transfer usually led to lower transfection efficiencies when compared with their viral counterparts. Biomaterial-guided gene delivery has emerged as a promising alternative to increase non-viral gene transfer efficiency by achieving sustained delivery of the candidate gene into cellular microenvironment. In the present study, we designed hyaluronic acid-based gene-activated cryogels (HACGs) encapsulating a novel formulation of non-viral vectors based on niosomes (P80PX) to promote MSCs in situ transfection. The developed HACG P80PX systems showed suitable physicochemical properties to promote MSCs in situ transfection with very low cytotoxicity. Incorporation of a plasmid encoding for the transcription factor SOX9 (psox9) into HACG P80PX systems led to an effective MSCs chondrogenic differentiation with reduced expression of fibrocartilage and hypertrophic markers. The capacity of the developed systems to restore cartilage extracellular matrix was further confirmed in an ex vivo model of chondral defect.

13.
J Biol Eng ; 17(1): 49, 2023 Jul 25.
Article in English | MEDLINE | ID: mdl-37491322

ABSTRACT

In recent decades, human mesenchymal stem cells (hMSCs) have gained momentum in the field of cell therapy for treating cartilage and bone injuries. Despite the tri-lineage multipotency, proliferative properties, and potent immunomodulatory effects of hMSCs, their clinical potential is hindered by donor variations, limiting their use in medical settings. To address this challenge, gene delivery technologies have emerged as a promising approach to modulate the phenotype and commitment of hMSCs towards specific cell lineages, thereby enhancing osteochondral repair strategies. This review provides a comprehensive overview of current non-viral gene delivery approaches used to engineer MSCs, highlighting key factors such as the choice of nucleic acid or delivery vector, transfection strategies, and experimental parameters. Additionally, it outlines various protocols and methods for qualitative and quantitative evaluation of their therapeutic potential as a delivery system in osteochondral regenerative applications. In summary, this technical review offers a practical guide for optimizing non-viral systems in osteochondral regenerative approaches. hMSCs constitute a key target population for gene therapy techniques. Nevertheless, there is a long way to go for their translation into clinical treatments. In this review, we remind the most relevant transfection conditions to be optimized, such as the type of nucleic acid or delivery vector, the transfection strategy, and the experimental parameters to accurately evaluate a delivery system. This survey provides a practical guide to optimizing non-viral systems for osteochondral regenerative approaches.

14.
Mol Ther Nucleic Acids ; 32: 302-317, 2023 Jun 13.
Article in English | MEDLINE | ID: mdl-37096164

ABSTRACT

Senescence is a process characterized by a prolonged irreversible cell-cycle arrest. The accumulation of senescent cells in tissues is related to aging and to the development of age-related diseases. Recently, gene therapy has emerged as a powerful tool for treating age-associated diseases by the transference of specific genes into the target cell population. However, the high sensitivity of senescent cells significantly precludes their genetic modification via classical viral and non-viral systems. Niosomes are self-assembled non-viral nanocarriers that exhibit important advantages due to their elevated cytocompatibility, versatility, and cost-efficiency, arising as a new alternative for genetic modification of senescent cells. In this work, we explore for the first time the use of niosomes for genetic modification of senescent umbilical cord-derived mesenchymal stem cells. We report that niosome composition greatly affected transfection efficiency; those formulations prepared in medium with sucrose and containing cholesterol as helper lipid being the most suitable to transfect senescent cells. Moreover, resulting niosome formulations exhibited a superior transfection efficiency with a markedly less cytotoxicity than the commercial reagent Lipofectamine. These findings highlight the potentiality of niosomes as effective vectors for genetic modification of senescent cells, providing new tools for the prevention and/or treatment of age-related diseases.

15.
Pharmaceutics ; 14(11)2022 Oct 28.
Article in English | MEDLINE | ID: mdl-36365145

ABSTRACT

Gene transfer to mesenchymal stem cells constitutes a powerful approach to promote their differentiation into the appropriate cartilage phenotype. Although viral vectors represent gold standard vehicles, because of their high efficiency, their use is precluded by important concerns including an elevated immunogenicity and the possibility of insertional mutagenesis. Therefore, the development of new and efficient non-viral vectors is under active investigation. In the present study, we developed new non-viral carriers based on niosomes to promote the effective chondrogenesis of human MSCs. Two different niosome formulations were prepared by varying their composition on non-ionic surfactant, polysorbate 80 solely (P80), or combined with poloxamer 407 (P80PX). The best niosome formulation was proven to transfer a plasmid, encoding for the potent chondrogenic transcription factor SOX9 in hMSC aggregate cultures. Transfection of hMSC aggregates via nioplexes resulted in an increased chondrogenic differentiation with reduced hypertrophy. These results highlight the potential of niosome formulations for gene therapy approaches focused on cartilage repair.

16.
Eur Cell Mater ; 21: 317-40, 2011 Apr 11.
Article in English | MEDLINE | ID: mdl-21484703

ABSTRACT

In situ gelling solutions for minimally invasive local application of bone growth factors are attracting increasing attention as efficient and patient-friendly alternative to bone grafts and solid scaffolds for repairing bone defects. Poloxamines, i.e., X-shaped poly(ethylene oxide)-poly(propylene oxide) block copolymers with an ethylenediamine core (Tetronic®), were evaluated both as an active osteogenic component and as a vehicle for rhBMP-2 injectable implants. After cytotoxicity screening of various poloxamine varieties, Tetronic 908, 1107, 1301 and 1307 solutions were chosen as the most cytocompatible and their sol-to-gel transitions were rheologically characterized. Viscoelastic gels, formed at 37 ºC, sustained protein release under physiological-like conditions. Formulations of rhBMP-2 led to differentiation of mesenchymal stem cells to osteoblasts, quantified as alkaline phosphatase activity with a maximum at day 7, and to mineralized nodules. Interestingly, poloxamine solely gels led to an initial proliferation of the mesenchymal stem cells (first week), followed by differentiation to osteoblasts (second to third week). Histochemical analysis revealed that Tetronic 908 is only osteoinductive; Tetronic 1107 is mostly osteoinductive, although its use leads to a minor differentiation to adipocytes; Tetronic 1307, solely or loaded with rhBMP-2, causes differentiation of both osteoblasts and adipocytes. Enhanced expression levels of CBFA-1 and collagen type I were observed for Tetronic 908, 1107 and 1307, both solely and combined with rhBMP-2. The intrinsic osteogenic activity of poloxamines (not observed for Pluronic F127) offers novel perspectives for bone regeneration using minimally invasive procedures (i.e., injectable scaffolds) and overcoming the safety and the cost/effectiveness concerns associated with large scale clinical use of recombinant growth factors.


Subject(s)
Bone Morphogenetic Protein 2/pharmacology , Ethylenediamines/pharmacology , Osteoblasts/drug effects , Polyethylene Glycols/pharmacology , Adipocytes/cytology , Adipocytes/drug effects , Adipocytes/metabolism , Alkaline Phosphatase/metabolism , Animals , BALB 3T3 Cells , CHO Cells , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Collagen Type I/genetics , Core Binding Factor Alpha 1 Subunit/genetics , Cricetinae , Cricetulus , Ethylenediamines/chemistry , Gels/chemistry , Gene Expression/drug effects , Humans , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice , Osteoblasts/cytology , Osteoblasts/metabolism , Osteogenesis/drug effects , Polyethylene Glycols/chemistry , Surface-Active Agents/chemistry , Surface-Active Agents/pharmacology
17.
Mater Sci Eng C Mater Biol Appl ; 128: 112307, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34474858

ABSTRACT

Gene transfer to mesenchymal stem cells (MSCs) has arisen as a powerful approach to increase the therapeutic potential of this effective cell population. Over recent years, niosomes have emerged as self-assembled carriers with promising performance for gene delivery. The aim of our work was to develop effective niosomes-based DNA delivery platforms for targeting MSCs. Niosomes based on 1,2-di-O-octadecenyl-3-trimethylammonium propane (DOTMA; 0, 7 or 15%) as cationic lipid, cholesterol as helper lipid, and polysorbate 60 as non-ionic surfactant, were prepared using a reverse phase evaporation technique. Niosomes dispersions (filtered or not) and their corresponding nioplexes with a lacZ plasmid were characterized in terms of size, charge, protection, and complexation abilities. DOTMA concentration had a large influence on the physicochemical properties of resulting nioplexes. Transfection efficiency and cytotoxic profiles were assessed in two immortalized cell lines of MSCs. Niosomes formulated with 15% DOTMA provided the highest values of ß-galactosidase activity, being similar to those achieved with Lipofectamine®, but showed less cytotoxicity. Filtration of niosomes dispersions before adding to the cells resulted in a loss of their biological activities. Storage of niosomes formulations (for 30 days at room temperature) caused minor modification of their physicochemical properties but also attenuated the transfection capability of the nioplexes. Differently, addition of the lysosomotropic agent sucrose into the culture medium during transfection or to the formulation itself improved the transfection performance of non-filtered niosomes. Indeed, steam heat-sterilized niosomes prepared in sucrose medium demonstrated transfection capability.


Subject(s)
Liposomes , Mesenchymal Stem Cells , Gene Transfer Techniques , Humans , Plasmids/genetics , Transfection
18.
ACS Biomater Sci Eng ; 7(9): 4102-4127, 2021 09 13.
Article in English | MEDLINE | ID: mdl-34137581

ABSTRACT

A plethora of applications using polysaccharides have been developed in recent years due to their availability as well as their frequent nontoxicity and biodegradability. These polymers are usually obtained from renewable sources or are byproducts of industrial processes, thus, their use is collaborative in waste management and shows promise for an enhanced sustainable circular economy. Regarding the development of novel delivery systems for biotherapeutics, the potential of polysaccharides is attractive for the previously mentioned properties and also for the possibility of chemical modification of their structures, their ability to form matrixes of diverse architectures and mechanical properties, as well as for their ability to maintain bioactivity following incorporation of the biomolecules into the matrix. Biotherapeutics, such as proteins, growth factors, gene vectors, enzymes, hormones, DNA/RNA, and antibodies are currently in use as major therapeutics in a wide range of pathologies. In the present review, we summarize recent progress in the development of polysaccharide-based hydrogels of diverse nature, alone or in combination with other polymers or drug delivery systems, which have been implemented in the delivery of biotherapeutics in the pharmaceutical and biomedical fields.


Subject(s)
Hydrogels , Polysaccharides , Drug Delivery Systems , Polymers , Proteins
19.
Hum Gene Ther ; 32(17-18): 895-906, 2021 09.
Article in English | MEDLINE | ID: mdl-33573471

ABSTRACT

Scaffold-guided viral gene therapy is a novel, powerful tool to enhance the processes of tissue repair in articular cartilage lesions by the delivery and overexpression of therapeutic genes in a noninvasive, controlled release manner based on a procedure that may protect the gene vehicles from undesirable host immune responses. In this study, we examined the potential of transferring a recombinant adeno-associated virus (rAAV) vector carrying a sequence for the highly chondroregenerative transforming growth factor beta (TGF-ß), using poly(ɛ-caprolactone) (PCL) films functionalized by the grafting of poly(sodium styrene sulfonate) (pNaSS) in chondrogenically competent bone marrow aspirates as future targets for therapy in cartilage lesions. Effective overexpression of TGF-ß in the aspirates by rAAV was achieved upon delivery using pNaSS-grafted and ungrafted PCL films for up to 21 days (the longest time point evaluated), with superior levels using the grafted films, compared with respective conditions without vector coating. The production of rAAV-mediated TGF-ß by pNaSS-grafted and ungrafted PCL films significantly triggered the biological activities and chondrogenic processes in the samples (proteoglycan and type-II collagen deposition and cell proliferation), while containing premature mineralization and hypertrophy relative to the other conditions, with overall superior effects supported by the pNaSS-grafted films. These observations demonstrate the potential of PCL film-assisted rAAV TGF-ß gene transfer as a convenient, off-the-shelf technique to enhance the reparative potential of the bone marrow in patients in future approaches for improved cartilage repair.


Subject(s)
Bone Marrow , Transforming Growth Factor beta , Cell Differentiation , Chondrogenesis , Genetic Therapy , Genetic Vectors/genetics , Humans , Transforming Growth Factor beta/genetics
20.
Adv Mater ; 33(16): e2008451, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33734514

ABSTRACT

The regeneration of focal articular cartilage defects is complicated by the reduced quality of the repair tissue and the potential development of perifocal osteoarthritis (OA). Biomaterial-guided gene therapy may enhance cartilage repair by controlling the release of therapeutic sequences in a spatiotemporal manner. Here, the benefits of delivering a recombinant adeno-associated virus (rAAV) vector coding for the human insulin-like growth factor I (IGF-I) via an alginate hydrogel (IGF-I/AlgPH155) to enhance repair of full-thickness chondral defects following microfracture surgery after one year in minipigs versus control (lacZ/AlgPH155) treatment are reported. Sustained IGF-I overexpression is significantly achieved in the repair tissue of defects treated with IGF-I/AlgPH155 versus those receiving lacZ/AlgPH155 for one year and in the cartilage surrounding the defects. Administration of IGF-I/AlgPH155 significantly improves parameters of cartilage repair at one year relative to lacZ/AlgPH155 (semiquantitative total histological score, cell densities, matrix deposition) without deleterious or immune reactions. Remarkably, delivery of IGF-I/AlgPH155 also significantly reduces perifocal OA and inflammation after one year versus lacZ/AlgPH155 treatment. Biomaterial-guided rAAV gene transfer represents a valuable clinical approach to promote cartilage repair and to protect against OA.


Subject(s)
Cartilage, Articular/metabolism , Dependovirus/genetics , Insulin-Like Growth Factor I/genetics , Animals , Gene Expression , Genetic Therapy , Humans , Hydrogels/metabolism , Osteoarthritis , Swine , Swine, Miniature
SELECTION OF CITATIONS
SEARCH DETAIL