Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
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
2.
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
3.
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
4.
J Neurosci ; 30(19): 6559-76, 2010 May 12.
Article in English | MEDLINE | ID: mdl-20463219

ABSTRACT

Guanylate cyclase activating protein 2 (GCAP2) is a recoverin-like Ca2+-sensor protein known to modulate guanylate cyclase activity in photoreceptor outer segments. GCAP2 is also present in photoreceptor ribbon synapses where its function is unknown. Synaptic ribbons are active zone-associated presynaptic structures in the tonically active photoreceptor ribbon synapses and contain RIBEYE as a unique and major protein component. In the present study, we demonstrate by various independent approaches that GCAP2 specifically interacts with RIBEYE in photoreceptor synapses. We show that the flexible hinge 2 linker region of RIBEYE(B) domain that connects the nicotinamide adenine dinucleotide (NADH)-binding subdomain with the substrate-binding subdomain (SBD) binds to the C terminus of GCAP2. We demonstrate that the RIBEYE-GCAP2 interaction is induced by the binding of NADH to RIBEYE. RIBEYE-GCAP2 interaction is modulated by the SBD. GCAP2 is strongly expressed in synaptic terminals of light-adapted photoreceptors where GCAP2 is found close to synaptic ribbons as judged by confocal microscopy and proximity ligation assays. Virus-mediated overexpression of GCAP2 in photoreceptor synaptic terminals leads to a reduction in the number of synaptic ribbons. Therefore, GCAP2 is a prime candidate for mediating Ca2+-dependent dynamic changes of synaptic ribbons in photoreceptor synapses.


Subject(s)
Guanylate Cyclase-Activating Proteins/metabolism , NAD/metabolism , Photoreceptor Cells, Vertebrate/metabolism , Synapses/metabolism , Alcohol Oxidoreductases , Amino Acid Sequence , Animals , Cattle , Cells, Cultured , Co-Repressor Proteins , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Guanylate Cyclase-Activating Proteins/genetics , In Vitro Techniques , Mice , Phosphoproteins/chemistry , Phosphoproteins/genetics , Phosphoproteins/metabolism , Presynaptic Terminals/metabolism , Protein Binding , Protein Conformation , Retina/metabolism , Two-Hybrid System Techniques
5.
Pharmaceutics ; 12(10)2020 09 29.
Article in English | MEDLINE | ID: mdl-33003607

ABSTRACT

Osteochondral defects involve both the articular cartilage and the underlying subchondral bone. If left untreated, they may lead to osteoarthritis. Advanced biomaterial-guided delivery of gene vectors has recently emerged as an attractive therapeutic concept for osteochondral repair. The goal of this review is to provide an overview of the variety of biomaterials employed as nonviral or viral gene carriers for osteochondral repair approaches both in vitro and in vivo, including hydrogels, solid scaffolds, and hybrid materials. The data show that a site-specific delivery of therapeutic gene vectors in the context of acellular or cellular strategies allows for a spatial and temporal control of osteochondral neotissue composition in vitro. In vivo, implantation of acellular hydrogels loaded with nonviral or viral vectors has been reported to significantly improve osteochondral repair in translational defect models. These advances support the concept of scaffold-mediated gene delivery for osteochondral repair.

6.
Nanomaterials (Basel) ; 10(9)2020 Aug 29.
Article in English | MEDLINE | ID: mdl-32872542

ABSTRACT

Injectable nanobioplatforms capable of locally fighting the inflammation in osteoarticular diseases, by reducing the number of administrations and prolonging the therapeutic effect is highly challenging. -Cyclodextrin cationic polymers are promising cartilage-penetrating candidates by intra-articular injection due to the high biocompatibility and ability to entrap multiple therapeutic and diagnostic agents, thus monitoring and mitigating inflammation. In this study, nanoassemblies based on poly--amino-cyclodextrin (PolyCD) loaded with the non-steroidal anti-inflammatory drug diclofenac (DCF) and linked by supramolecular interactions with a fluorescent probe (adamantanyl-Rhodamine conjugate, Ada-Rhod) were developed to manage inflammation in osteoarticular diseases. PolyCD@Ada-Rhod/DCF supramolecular nanoassemblies were characterized by complementary spectroscopic techniques including UV-Vis, steady-state and time-resolved fluorescence, DLS and ζ-potential measurement. Stability and DCF release kinetics were investigated in medium mimicking the physiological conditions to ensure control over time and efficacy. Biological experiments evidenced the efficient cellular internalization of PolyCD@Ada-Rhod/DCF (within two hours) without significant cytotoxicity in primary human bone marrow-derived mesenchymal stromal cells (hMSCs). Finally, polyCD@Ada-Rhod/DCF significantly suppressed IL-1 production in hMSCs, revealing the anti-inflammatory properties of these nanoassemblies. With these premises, this study might open novel routes to exploit original CD-based nanobiomaterials for the treatment of osteoarticular diseases.

7.
Tissue Eng Regen Med ; 16(4): 345-355, 2019 08.
Article in English | MEDLINE | ID: mdl-31413939

ABSTRACT

Background: Viral vector-based therapeutic gene therapy is a potent strategy to enhance the intrinsic reparative abilities of human orthopaedic tissues. However, clinical application of viral gene transfer remains hindered by detrimental responses in the host against such vectors (immunogenic responses, vector dissemination to nontarget locations). Combining viral gene therapy techniques with tissue engineering procedures may offer strong tools to improve the current systems for applications in vivo. Methods: The goal of this work is to provide an overview of the most recent systems exploiting biomaterial technologies and therapeutic viral gene transfer in human orthopaedic regenerative medicine. Results: Integration of tissue engineering platforms with viral gene vectors is an active area of research in orthopaedics as a means to overcome the obstacles precluding effective viral gene therapy. Conclusions: In light of promising preclinical data that may rapidly expand in a close future, biomaterial-guided viral gene therapy has a strong potential for translation in the field of human orthopaedic regenerative medicine.


Subject(s)
Genetic Therapy/methods , Orthopedics/methods , Regenerative Medicine/methods , Animals , Gene Transfer Techniques , Genes, Viral/genetics , Genetic Vectors/genetics , Humans , Oncolytic Virotherapy/methods , Tissue Engineering/methods
8.
J Clin Med ; 8(9)2019 Aug 28.
Article in English | MEDLINE | ID: mdl-31466339

ABSTRACT

The application of chondrogenic gene sequences to human bone marrow-derived mesenchymal stromal cells (hMSCs) is an attractive strategy to activate the reparative activities of these cells as a means to enhance the processes of cartilage repair using indirect cell transplantation procedures that may improve the repopulation of cartilage lesions. In the present study, we examined the feasibility of co-delivering the highly competent transforming growth factor beta (TGF-ß) with the insulin-like growth factor I (IGF-I) in hMSCs via recombinant adeno-associated virus (rAAV) vector-mediated gene transfer prior to implantation in a human model of osteochondral defect (OCD) ex vivo that provides a microenvironment similar to that of focal cartilage lesions. The successful co-overexpression of rAAV TGF-ß/IGF-I in implanted hMSCs promoted the durable remodeling of tissue injury in human OCDs over a prolonged period of time (21 days) relative to individual gene transfer and the control (reporter lacZ gene) treatment, with enhanced levels of cell proliferation and matrix deposition (proteoglycans, type-II collagen) both in the lesions and at a distance, while hypertrophic, osteogenic, and catabolic processes could be advantageously delayed. These findings demonstrate the value of indirect, progenitor cell-based combined rAAV gene therapy to treat human focal cartilage defects in a natural environment as a basis for future clinical applications.

9.
J Exp Orthop ; 4(1): 4, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28176272

ABSTRACT

BACKGROUND: Transplantation of genetically modified bone marrow concentrates is an attractive approach to conveniently activate the chondrogenic differentiation processes as a means to improve the intrinsic repair capacities of damaged articular cartilage. METHODS: Human bone marrow aspirates were co-transduced with recombinant adeno-associated virus (rAAV) vectors to overexpress the pleiotropic transformation growth factor beta (TGF-ß) and the cartilage-specific transcription factor sox9 as a means to enhance the chondroreparative processes in conditions of specific lineage differentiation. RESULTS: Successful TGF-ß/sox9 combined gene transfer and overexpression via rAAV was achieved in chondrogenically induced human bone marrow aspirates for up to 21 days, the longest time point evaluated, leading to increased proliferation, matrix synthesis, and chondrogenic differentiation relative to control treatments (reporter lacZ treatment, absence of vector application) especially when co-applying the candidate vectors at the highest vector doses tested. Optimal co-administration of TGF-ß with sox9 also advantageously reduced hypertrophic differentiation in the aspirates. CONCLUSIONS: These findings report the possibility of directly modifying bone marrow aspirates by combined therapeutic gene transfer as a potent and convenient future approach to improve the repair of articular cartilage lesions.

10.
Stem Cells Transl Med ; 6(1): 249-260, 2017 01.
Article in English | MEDLINE | ID: mdl-28170175

ABSTRACT

Transplantation of genetically modified peripheral blood aspirates that carry chondrogenically competent progenitor cells may offer new, convenient tools to treat articular cartilage lesions compared with the more complex and invasive application of bone marrow concentrates or of bone marrow-derived mesenchymal stem cells. Here, we show that recombinant adeno-associated viral (rAAV) vectors are powerful gene vehicles capable of successfully targeting primary human peripheral blood aspirates in a stable and safe manner, allowing for an efficient and long-term transgene expression in such samples (up to 63 days with use of a lacZ reporter gene and for at least 21 days with application of the pleiotropic, chondrogenic factor transforming growth factor-ß [TGF-ß]). rAAV-mediated overexpression of TGF-ß enhanced both the proliferative and metabolic properties of the peripheral blood aspirates, also increasing the chondrogenic differentiation processes in these samples. Hypertrophy and osteogenic differentiation events were also activated by production of TGF-ß via rAAV, suggesting that translation of the current approach in vivo will probably require close regulation of expression of this candidate gene. However, these results support the concept of directly modifying peripheral blood as a novel approach to conveniently treat articular cartilage lesions in patients. Stem Cells Translational Medicine 2017;6:249-260.


Subject(s)
Cartilage, Articular/pathology , Chondrogenesis , Dependovirus/genetics , Gene Transfer Techniques , Genetic Vectors/metabolism , Transforming Growth Factor beta/genetics , Wound Healing , Cell Differentiation , Cell Proliferation , Humans , Hypertrophy , Immunophenotyping , Middle Aged , Osteogenesis , Suction , Transforming Growth Factor beta/metabolism , Transgenes
11.
Tissue Eng Part A ; 21(17-18): 2460-71, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26123891

ABSTRACT

Direct therapeutic gene transfer in marrow concentrates is an attractive strategy to conveniently enhance the chondrogenic differentiation processes as a means to improve the healing response of damaged articular cartilage upon reimplantation in sites of injury. In the present study, we evaluated the ability of the clinically adapted recombinant adeno-associated virus (rAAV) vectors to mediate overexpression of the insulin-like growth factor I (IGF-I) in human bone marrow aspirates that may modulate the proliferative, anabolic activities, and chondrogenic differentiation potential in such samples in vitro. The results demonstrate that successful, significant rAAV-mediated IGF-I gene transfer and expression were achieved in transduced aspirates (up to 105.9±35.1 pg rhIGF-I/mg total proteins) over time (21 days) at very high levels (∼80% of cells expressing the candidate IGF-I transgene), leading to increased levels of proliferation, matrix synthesis, and chondrogenic differentiation over time compared with the control (lacZ) condition. Treatment with the candidate IGF-I vector also stimulated the hypertrophic and osteogenic differentiation processes in the aspirates, suggesting that the regulation of IGF-I expression through rAAV will be a prerequisite for future translation of the approach in vivo. However, these findings show the possible benefits of this vector class to directly modify marrow concentrates as a convenient tool for strategies that aim at improving the repair of articular cartilage lesions.


Subject(s)
Bone Marrow/metabolism , Cell Differentiation , Chondrogenesis , Dependovirus/metabolism , Gene Transfer Techniques , Insulin-Like Growth Factor I/genetics , Adipogenesis , Aged , Cell Proliferation , Humans , Hypertrophy , Real-Time Polymerase Chain Reaction , Suction , Transduction, Genetic
12.
Hum Gene Ther ; 25(12): 1050-60, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25333854

ABSTRACT

Genetic modification of bone marrow-derived mesenchymal stem cells (MSCs) for use in transplantation settings may be a valuable strategy to enhance the repair processes in articular cartilage defects. Here, we evaluated the potential of overexpressing the transforming growth factor (TGF)-ß via recombinant adeno-associated viral (rAAV) vector-mediated gene transfer to promote the chondrogenic differentiation of human MSCs (hMSCs). A human TGF-ß sequence was delivered to undifferentiated and chondrogenically induced primary hMSCs, using rAAV vectors to test the efficacy and duration of transgene expression and its effects on the chondrogenic, osteogenic, and adipogenic differentiation patterns of the cells compared with control (lacZ) treatment after 21 days in vitro. Significant, durable TGF-ß expression was noted both in undifferentiated and chondrogenically induced hMSCs transduced with the candidate rAAV-hTGF-ß vector for up to 21 days compared with rAAV-lacZ treatment, allowing for increased proliferative, metabolic, and chondrogenic activities via stimulation of the critical SOX9 (SRY [sex-determining region Y]-related HMG [high-mobility group] box 9) chondrogenic pathway. Overexpression of TGF-ß under the conditions applied here also activated the hypertrophic and osteogenic differentiation processes in the treated cells. Such effects were noted in association with enhanced levels of ß-catenin and Indian hedgehog and decreased parathyroid hormone-related protein expression. The current findings show that rAAV vectors provide advantageous vehicles for gene- and stem cell-based approaches to treat articular cartilage defects, requiring tight regulation of TGF-ß expression to avoid hypertrophy as candidate treatment for future applications in clinically relevant animal models in vivo.


Subject(s)
Cell Differentiation/genetics , Dependovirus/genetics , Mesenchymal Stem Cells , Transforming Growth Factor beta/biosynthesis , Animals , Bone Marrow Cells , Cartilage/growth & development , Cell Proliferation/genetics , Chondrogenesis/genetics , Gene Expression Regulation , Genetic Vectors , Humans , Osteogenesis/genetics , Transforming Growth Factor beta/genetics
13.
Stem Cell Res Ther ; 5(4): 103, 2014 Aug 27.
Article in English | MEDLINE | ID: mdl-25163769

ABSTRACT

INTRODUCTION: The transplantation of genetically modified progenitor cells such as bone marrow-derived mesenchymal stem cells (MSCs) is an attractive strategy to improve the natural healing of articular cartilage defects. In the present study, we examined the potential benefits of sustained overexpression of the mitogenic and pro-anabolic insulin-like growth factor I (IGF-I) via gene transfer upon the biological activities of human MSCs (hMSCs). METHODS: Recombinant adeno-associated vectors (rAAV) were used to deliver a human IGF-I coding sequence in undifferentiated and chondrogenically-induced primary hMSCs in order to determine the efficacy and duration of transgene expression and the subsequent effects of the genetic modification upon the chondrogenic versus osteogenic differentiation profiles of the cells relative to control (lacZ) treatment after 21 days in vitro. RESULTS: Significant and prolonged expression of IGF-I was evidenced in undifferentiated and most importantly in chondrogenically-induced hMSCs transduced with the candidate rAAV-hIGF-I vector for up to 21 days, leading to enhanced proliferative, biosynthetic, and chondrogenic activities compared with rAAV-lacZ treatment. Overexpression of IGF-I as achieved in the conditions applied here also increased the expression of hypertrophic and osteogenic markers in the treated cells. CONCLUSIONS: These results suggest that a tight regulation of rAAV expression may be necessary for further translation of the approach in clinically relevant animal models in vivo. However, the current findings support the concept of using this type of vector as an effective tool to treat articular cartilage defects via gene- and stem cell-based procedures.


Subject(s)
Cell Differentiation/genetics , Insulin-Like Growth Factor I/metabolism , Mesenchymal Stem Cells/metabolism , Protein Precursors/metabolism , Transgenes , Cartilage, Articular/pathology , Cell Culture Techniques , Cell Proliferation , Gene Transfer Techniques , Genetic Vectors , Humans , Immunohistochemistry , Insulin-Like Growth Factor I/genetics , Osteogenesis/genetics , Protein Precursors/genetics , Recombinant Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL