1
|
Vlashi R, Zhang X, Li H, Chen G. Potential therapeutic strategies for osteoarthritis via CRISPR/Cas9 mediated gene editing. Rev Endocr Metab Disord 2024; 25:339-367. [PMID: 38055160 DOI: 10.1007/s11154-023-09860-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/28/2023] [Indexed: 12/07/2023]
Abstract
Osteoarthritis (OA) is an incapacitating and one of the most common physically degenerative conditions with an assorted etiology and a highly complicated molecular mechanism that to date lacks an efficient treatment. The capacity to design biological networks and accurately modify existing genomic sites holds an apt potential for applications across medical and biotechnological sciences. One of these highly specific genomes editing technologies is the CRISPR/Cas9 mechanism, referred to as the clustered regularly interspaced short palindromic repeats, which is a defense mechanism constituted by CRISPR associated protein 9 (Cas9) directed by small non-coding RNAs (sncRNA) that bind to target DNA through Watson-Crick base pairing rules where subsequent repair of the target DNA is initiated. Up-to-date research has established the effectiveness of the CRISPR/Cas9 mechanism in targeting the genetic and epigenetic alterations in OA by suppressing or deleting gene expressions and eventually distributing distinctive anti-arthritic properties in both in vitro and in vivo osteoarthritic models. This review aims to epitomize the role of this high-throughput and multiplexed gene editing method as an analogous therapeutic strategy that could greatly facilitate the clinical development of OA-related treatments since it's reportedly an easy, minimally invasive technique, and a comparatively less painful method for osteoarthritic patients.
Collapse
Affiliation(s)
- Rexhina Vlashi
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Xingen Zhang
- Department of Orthopedics, Jiaxing Key Laboratory for Minimally Invasive Surgery in Orthopaedics & Skeletal Regenerative Medicine, Zhejiang Rongjun Hospital, Jiaxing, 314001, China
| | - Haibo Li
- The Central Laboratory of Birth Defects Prevention and Control, Ningbo Women and Children's Hospital, Ningbo, China.
- Ningbo Key Laboratory for the Prevention and Treatment of Embryogenic Diseases, Ningbo Women and Children's Hospital, Ningbo, China.
| | - Guiqian Chen
- College of Life Science and Medicine, Zhejiang Provincial Key Laboratory of Silkworm Bioreactor and Biomedicine, Zhejiang Sci-Tech University, Hangzhou, 310018, China.
| |
Collapse
|
2
|
Li X, Shen L, Deng Z, Huang Z. New treatment for osteoarthr: pbad014itis: Gene therapy. PRECISION CLINICAL MEDICINE 2023; 6:pbad014. [PMID: 37333626 PMCID: PMC10273835 DOI: 10.1093/pcmedi/pbad014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 05/24/2023] [Indexed: 06/20/2023] Open
Abstract
Osteoarthritis is a complex degenerative disease that affects the entire joint tissue. Currently, non-surgical treatments for osteoarthritis focus on relieving pain. While end-stage osteoarthritis can be treated with arthroplasty, the health and financial costs associated with surgery have forced the search for alternative non-surgical treatments to delay the progression of osteoarthritis and promote cartilage repair. Unlike traditional treatment, the gene therapy approach allows for long-lasting expression of therapeutic proteins at specific sites. In this review, we summarize the history of gene therapy in osteoarthritis, outlining the common expression vectors (non-viral, viral), the genes delivered (transcription factors, growth factors, inflammation-associated cytokines, non-coding RNAs) and the mode of gene delivery (direct delivery, indirect delivery). We highlight the application and development prospects of the gene editing technology CRISPR/Cas9 in osteoarthritis. Finally, we identify the current problems and possible solutions in the clinical translation of gene therapy for osteoarthritis.
Collapse
Affiliation(s)
- Xinyu Li
- Department of Orthopaedic Surgery, West China Hospital, West China Medical School, Sichuan University, Chengdu 610041, China
| | - Leyao Shen
- School of Dentistry, University of Michigan, Ann Arbor, MI 48109-1078, USA
| | | | | |
Collapse
|
3
|
Graceffa V, Vinatier C, Guicheux J, Evans CH, Stoddart M, Alini M, Zeugolis DI. State of art and limitations in genetic engineering to induce stable chondrogenic phenotype. Biotechnol Adv 2018; 36:1855-1869. [DOI: 10.1016/j.biotechadv.2018.07.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 05/16/2018] [Accepted: 07/12/2018] [Indexed: 12/18/2022]
|
4
|
Raftery RM, Walsh DP, Castaño IM, Heise A, Duffy GP, Cryan SA, O'Brien FJ. Delivering Nucleic-Acid Based Nanomedicines on Biomaterial Scaffolds for Orthopedic Tissue Repair: Challenges, Progress and Future Perspectives. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2016; 28:5447-5469. [PMID: 26840618 DOI: 10.1002/adma.201505088] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 11/27/2015] [Indexed: 06/05/2023]
Abstract
As well as acting to fill defects and allow for cell infiltration and proliferation in regenerative medicine, biomaterial scaffolds can also act as carriers for therapeutics, further enhancing their efficacy. Drug and protein delivery on scaffolds have shown potential, however, supraphysiological quantities of therapeutic are often released at the defect site, causing off-target side effects and cytotoxicity. Gene therapy involves the introduction of foreign genes into a cell in order to exert an effect; either replacing a missing gene or modulating expression of a protein. State of the art gene therapy also encompasses manipulation of the transcriptome by harnessing RNA interference (RNAi) therapy. The delivery of nucleic acid nanomedicines on biomaterial scaffolds - gene-activated scaffolds -has shown potential for use in a variety of tissue engineering applications, but as of yet, have not reached clinical use. The current state of the art in terms of biomaterial scaffolds and delivery vector materials for gene therapy is reviewed, and the limitations of current procedures discussed. Future directions in the clinical translation of gene-activated scaffolds are also considered, with a particular focus on bone and cartilage tissue regeneration.
Collapse
Affiliation(s)
- Rosanne M Raftery
- Tissue Engineering Research Group (TERG), Dept. of Anatomy, Royal College of Surgeons in Ireland (RCSI), 123, St. Stephens Green, Dublin 2, Dublin, Ireland
- Trinity Centre for Bioengineering (TCBE), Trinity College Dublin, Dublin 2, Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland
- Drug Delivery and Advanced Materials Research Team, School of Pharmacy, Royal College of Surgeons in Ireland, 123, St. Stephens Green, Dublin 2, Dublin, Ireland
- Centre for Research in Medical Devices (CÚRAM), National University of Ireland Galway, Galway, Ireland
| | - David P Walsh
- Tissue Engineering Research Group (TERG), Dept. of Anatomy, Royal College of Surgeons in Ireland (RCSI), 123, St. Stephens Green, Dublin 2, Dublin, Ireland
- Trinity Centre for Bioengineering (TCBE), Trinity College Dublin, Dublin 2, Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland
- Drug Delivery and Advanced Materials Research Team, School of Pharmacy, Royal College of Surgeons in Ireland, 123, St. Stephens Green, Dublin 2, Dublin, Ireland
- Centre for Research in Medical Devices (CÚRAM), National University of Ireland Galway, Galway, Ireland
| | - Irene Mencía Castaño
- Tissue Engineering Research Group (TERG), Dept. of Anatomy, Royal College of Surgeons in Ireland (RCSI), 123, St. Stephens Green, Dublin 2, Dublin, Ireland
- Trinity Centre for Bioengineering (TCBE), Trinity College Dublin, Dublin 2, Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland
| | - Andreas Heise
- Tissue Engineering Research Group (TERG), Dept. of Anatomy, Royal College of Surgeons in Ireland (RCSI), 123, St. Stephens Green, Dublin 2, Dublin, Ireland
| | - Garry P Duffy
- Tissue Engineering Research Group (TERG), Dept. of Anatomy, Royal College of Surgeons in Ireland (RCSI), 123, St. Stephens Green, Dublin 2, Dublin, Ireland
- Trinity Centre for Bioengineering (TCBE), Trinity College Dublin, Dublin 2, Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland
| | - Sally-Ann Cryan
- Tissue Engineering Research Group (TERG), Dept. of Anatomy, Royal College of Surgeons in Ireland (RCSI), 123, St. Stephens Green, Dublin 2, Dublin, Ireland
- Trinity Centre for Bioengineering (TCBE), Trinity College Dublin, Dublin 2, Dublin, Ireland
- Drug Delivery and Advanced Materials Research Team, School of Pharmacy, Royal College of Surgeons in Ireland, 123, St. Stephens Green, Dublin 2, Dublin, Ireland
- Centre for Research in Medical Devices (CÚRAM), National University of Ireland Galway, Galway, Ireland
| | - Fergal J O'Brien
- Tissue Engineering Research Group (TERG), Dept. of Anatomy, Royal College of Surgeons in Ireland (RCSI), 123, St. Stephens Green, Dublin 2, Dublin, Ireland
- Trinity Centre for Bioengineering (TCBE), Trinity College Dublin, Dublin 2, Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland
| |
Collapse
|
5
|
Sun Z, Yin H, Yu X, Sun X, Xiao B, Xu Y, Yuan Z, Meng H, Peng J, Yu C, Wang Y, Guo Q, Wang A, Lu S. Inhibition of Osteoarthritis in Rats by Electroporation with Interleukin-1 Receptor Antagonist. ACTA ACUST UNITED AC 2016. [DOI: 10.4236/jbise.2016.97027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
6
|
Vonk LA, Kragten AHM, Dhert WJA, Saris DBF, Creemers LB. Overexpression of hsa-miR-148a promotes cartilage production and inhibits cartilage degradation by osteoarthritic chondrocytes. Osteoarthritis Cartilage 2014; 22:145-53. [PMID: 24269634 DOI: 10.1016/j.joca.2013.11.006] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Revised: 10/27/2013] [Accepted: 11/09/2013] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Hsa-miR-148a expression is decreased in Osteoarthritis (OA) cartilage, but its functional role in cartilage has never been studied. Therefore, our aim was to investigate the effects of overexpressing hsa-miR-148a on cartilage metabolism of OA chondrocytes. DESIGN OA chondrocytes were transfected with a miRNA precursor for hsa-miR-148a or a miRNA precursor negative control. After 3, 7, 14 and 21 days, real-time PCR was performed to examine gene expression levels of aggrecan (ACAN), type I, II, and X collagen (COL1A1, COL2A1, COl10A1), matrix metallopeptidase 13 (MMP13), a disintegrin and metalloproteinase with thrombospondin motifs 5 (ADAMTS5) and the serpin peptidase inhibitor, clade H (heat shock protein 47), member 1 (SERPINH1). After 3 weeks, DNA content and proteoglycan and collagen content and release were determined. Type II collagen was analyzed at the protein level by Western blot. RESULTS Overexpression of hsa-miR-148a had no effect on ACAN, COL1A1 and SERPINH1 gene expression, but increased COL2A1 and decreased COL10A1, MMP13 and ADAMTS5 gene expression. Luciferase reporter assay confirmed direct interaction of miR-148a and COL10A1, MMP13 and ADAMTS5. The matrix deposited by the miR-148a overexpressing cells contained more proteoglycans and collagen, in particular type II collagen. Proteoglycan and collagen release into the culture medium was inhibited, but total collagen production was increased. CONCLUSION Overexpression of hsa-miR-148a inhibits hypertrophic differentiation and increases the production and deposition of type II collagen by OA chondrocytes, which is accompanied by an increased retention of proteoglycans. Hsa-miR-148a might be a potential disease-modifying compound in OA, as it promotes hyaline cartilage production.
Collapse
Affiliation(s)
- L A Vonk
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands.
| | - A H M Kragten
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - W J A Dhert
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands; Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - D B F Saris
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands; Tissue Regeneration, MIRA Institute, University Twente, Enschede, The Netherlands
| | - L B Creemers
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
7
|
Bistolfi F. Evidence of interlinks between bioelectromagnetics and biomechanics: from biophysics to medical physics. Phys Med 2012; 22:71-95. [PMID: 17664154 DOI: 10.1016/s1120-1797(06)80002-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2005] [Revised: 05/29/2006] [Accepted: 06/12/2006] [Indexed: 01/22/2023] Open
Abstract
A vast literature on electromagnetic and mechanical bioeffects at the bone and soft tissue level, as well as at the cellular level (osteoblasts, osteoclasts, keratinocytes, fibroblasts, chondrocytes, nerve cells, endothelial and muscle cells) has been reviewed and analysed in order to show the evident connections between both types of physical energies. Moreover, an intimate link between the two is suggested by transduction phenomena (electromagnetic-acoustic transduction and its reverse) occurring in living matter, as a sound biophysical literature has demonstrated. However, electromagnetic and mechanical signals are not always interchangeable, depending on their respective intensity. Calculations are reported in order to show in which cases (read: for which values of electric field in V/m and of mechanical pressure in Pa) a given electromagnetic or mechanical bioeffect is only due to the directly impinging energy or even to the indirect transductional energy. The relevance of the treated item for the applications of medical physics to regenerative medicine is stressed.
Collapse
Affiliation(s)
- F Bistolfi
- Radiotherapy Department, Galliera Hospital, Genova (Italy)
| |
Collapse
|
8
|
Tabler JM, Liu KJ. Electroporation of craniofacial mesenchyme. J Vis Exp 2011:e3381. [PMID: 22143372 PMCID: PMC3308610 DOI: 10.3791/3381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Electroporation is an efficient method of delivering DNA and other charged macromolecules into tissues at precise time points and in precise locations. For example, electroporation has been used with great success to study neural and retinal development in Xenopus, chicken and mouse (1-10). However, it is important to note that in all of these studies, investigators were not targeting soft tissues. Because we are interested in craniofacial development, we adapted a method to target facial mesenchyme. When we searched the literature, we found, to our surprise, very few reports of successful gene transfer into cartilaginous tissue. The majority of these studies were gene therapy studies, such as siRNA or protein delivery into chondrogenic cell lines, or, animal models of arthritis (11-13). In other systems, such as chicken or mouse, electroporation of facial mesenchyme has been challenging (personal communications, Dept of Craniofacial Development, KCL). We hypothesized that electroporation into procartilaginous and cartilaginous tissues in Xenopus might work better. In our studies, we show that gene transfer into the facial cartilages occurs efficiently at early stages (28), when the facial primordium is still comprised of soft tissue prior to cartilage differentiation. Xenopus is a very accessible vertebrate system for analysis of craniofacial development. Craniofacial structures are more readily visible in Xenopus than in any other vertebrate model, primarily because Xenopus embryos are fertilized externally, allowing analyses of the earliest stages, and facilitating live imaging at single cell resolution, as well as reuse of the mothers (14). Among vertebrate models developing externally, Xenopus is more useful for craniofacial analysis than zebrafish, as Xenopus larvae are larger and easier to dissect, and the developing facial region is more accessible to imaging than the equivalent region in fish. In addition, Xenopus is evolutionarily closer to humans than zebrafish (˜100 million years closer) (15). Finally, at these stages, Xenopus tadpoles are transparent, and concurrent expression of fluorescent proteins or molecules will allow easy visualization of the developing cartilages. We anticipate that this approach will allow us to rapidly and efficiently test candidate molecules in an in vivo model system.
Collapse
|
9
|
Yao S, Gutierrez DL, Ring S, Liu D, Wise GE. Electroporation to deliver plasmid DNA into rat dental tissues. J Gene Med 2011; 12:981-9. [PMID: 21157822 DOI: 10.1002/jgm.1521] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Delivery of DNA into the target tissues is an important technique in gene function studies and gene therapy. Surgical treatment of tooth eruption disorders, such as impacted third molars, is a major healthcare cost. Because the dental follicle (DF) is essential for regulating tooth eruption, establishment of local gene transfer protocols is needed to determine the effect of various genes on eruption and to develop gene therapy approaches for inducing the eruption of impacted molars. METHODS Plasmids containing lacZ reporter gene were injected into rat mandibles and then electroporated at the designated settings. Mandibles were collected 24 h after electroporation for X-gal staining to evaluate the transfection efficiency. Tissues were collected at various days post-electroporation to determine the expression of the transgene. RESULTS For the DF, depth of injection and pulse number appear to be important. Six pulses can achieve above 80% transfection of the DF at 50 V or 120 V. For alveolar bone (AB) transfection, voltages are important, with 120 V being optimal. Regarding pulse durations, we determined that durations of 20 and 30 ms achieve the maximum transfection in AB and DF, respectively. CONCLUSIONS The present study demonstrates for the first time the feasibility of electroporation to locally deliver plasmids into dental tissues. Parameters affecting electroporation to deliver plasmids into the dental tissues were optimized. This protocol could be used to deliver short hairpin RNA or genes of interest into the dental tissues to regulate tooth eruption. Thus, it may be possible to develop nonsurgical treatments for inducing the eruption of impacted teeth.
Collapse
Affiliation(s)
- Shaomian Yao
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA.
| | | | | | | | | |
Collapse
|
10
|
Denys A, Thiolat A, Descamps D, Lemeiter D, Benihoud K, Bessis N, Boissier MC. Intra-articular electrotransfer of mouse soluble tumour necrosis factor receptor in a murine model of rheumatoid arthritis. J Gene Med 2010; 12:659-68. [PMID: 20623491 DOI: 10.1002/jgm.1482] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Rheumatoid arthritis (RA) is a chronic autoimmune disease that causes inflammation and destruction of the joints. In the collagen-induced arthritis mouse model of RA, we developed a nonviral gene therapy method designed to block in situ the main cytokine tumour necrosis factor (TNF)-alpha METHODS Electrotransfer was used to deliver a plasmid encoding extracellular domain of mouse soluble TNF-alpha receptor type I fused to the Fc fragment of mouse immunoglobulin (Ig)G1 (pTNFR-Is) corresponding to a dimeric TNF-alpha soluble receptor fusion protein (mTNFR-Is/Ig). RESULTS Delivery of the plasmid into the knees at symptom onset improved the histological inflammation and destruction not only at the knees, but also at the ankles, indicating a local and a regional therapeutic effect. The plasmid was detected in synovial membrane and meniscus specimens from the injected joints. In the synovial membrane, 15 days post-injection, interleukin (IL)-17 and TNF-alpha mRNAs expression were increased, whereas IL-10 mRNA was unchanged. However, the empty plasmid exerted a pro-inflammatory effect 30 days post-injection. CONCLUSIONS These data indicate that local nonviral gene therapy against TNF-alpha is effective, although further work is needed to decrease plasmid induced inflammation.
Collapse
Affiliation(s)
- Anne Denys
- EA4222, Li2P, University of Paris 13, Bobigny, France.
| | | | | | | | | | | | | |
Collapse
|
11
|
Jorgensen C, Apparailly F. Prospects for gene therapy in inflammatory arthritis. Best Pract Res Clin Rheumatol 2010; 24:541-52. [DOI: 10.1016/j.berh.2010.02.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
12
|
Ramachandran N, Jaroszeski M, Hoff AM. Molecular delivery to cells facilitated by corona ion deposition. IEEE Trans Nanobioscience 2008; 7:233-9. [PMID: 18779104 DOI: 10.1109/tnb.2008.2002290] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
A novel method of inducing the delivery of nonpermeant molecules to the cytosol of cells is presented in this paper. Corona discharge in air was utilized to produce ions that in turn were deposited onto the liquid surface of media containing cultured cells. Murine B16 melanoma cells were used to demonstrate the molecular delivery of fluorescent dye calcein, the drug bleomycin, and a nucleic acid stain SYTOX-green. None of these molecules penetrate cells with intact membranes. Following the corona treatment, cells were observed to admit significant quantities of these molecules from the culture media, relative to control samples. Further, greater than 95% viability of treated cells was observed by Trypan Blue assay. This method may provide an attractive alternative to electroporation where a physical contact between electrodes and cells is needed to deliver molecules to the cytosol.
Collapse
Affiliation(s)
- Niraj Ramachandran
- Department of Chemical Engineering, University of South Florida, Tampa, FL 33620 USA
| | | | | |
Collapse
|
13
|
Bloquel C, Denys A, Boissier MC, Apparailly F, Bigey P, Scherman D, Bessis N. Intra-articular electrotransfer of plasmid encoding soluble TNF receptor variants in normal and arthritic mice. J Gene Med 2008; 9:986-93. [PMID: 17912759 DOI: 10.1002/jgm.1088] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND Anti-inflammatory gene therapy is promising in inflammatory diseases such as rheumatoid arthritis (RA). We have previously demonstrated that intra-muscular (i.m.) electrotransfer (ET) of plasmids encoding three different human tumor necrosis factor-alpha-soluble receptor I variants (hTNFR-Is) exert protective effects in an experimental RA model. However, such a systemic approach could be responsible for side effects. The present study aimed at performing an intra-articular (i.a.) gene therapy by electrotransfer using the hTNFR-Is plasmids. METHODS AND RESULTS We evaluated targeting of mice joints by CCD optical imaging after i.a. ET of a luciferase-encoding plasmid and we showed that ET led to strongly increased transgene expression in a plasmid dose-dependent manner. Moreover, articular and seric hTNFR-Is was detectable for 2 weeks. As expected, systemic hTNFR-Is rates were lower after i.a. ET than after i.m. ET. A longer protein secretion could be achieved with several i.a. ETs. Also, we observed that hTNFR-Is expression within arthritic joints was slightly higher than in normal joints. CONCLUSIONS In collagen-induced arthritis (CIA), a mouse model for RA, we demonstrated that hTNFR-Is/mIgG1-encoding plasmid i.a. ET decreased joint destruction in the ankles. In conclusion, our results suggest that local TNFR-Is gene therapy may play a role in decreasing joint destruction in CIA.
Collapse
Affiliation(s)
- C Bloquel
- Inserm, Eri-18, F-93017 Bobigny, France
| | | | | | | | | | | | | |
Collapse
|
14
|
Laroui H, Grossin L, Léonard M, Stoltz JF, Gillet P, Netter P, Dellacherie E. Hyaluronate-covered nanoparticles for the therapeutic targeting of cartilage. Biomacromolecules 2007; 8:3879-85. [PMID: 18039001 DOI: 10.1021/bm700836y] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Hyaluronic acid (HA) has a high affinity for the CD44 receptor present at the surface of articular cells, particularly of chondrocytes. HA-covered polylactide nanoparticles containing bioactive compounds such as HA and chondroitin sulfate (CS) were thus prepared in order to achieve a controlled delivery targeted to cartilage cells after injection near articular alterations/erosions. Such nanoparticles (diameter = 700 nm) were prepared by double emulsion/solvent evaporation, using amphiphilic derivatives of HA, as stabilizer of the secondary emulsion. These nanoparticles were incubated with articular cells, and several tests were carried out. First, they proved that the nanospheres provoked no decrease in cell viability, even after 72 h of contact. Second, a confocal microscopy analysis on fluorescent HA-covered particles showed that they were captured by articular cells, while with those covered with poly(vinyl alcohol), the uptake was far lower. Third, a scattering electron microscopy analysis proved that the HA-coated nanoparticles were localized in the cell intracytoplasmic area.
Collapse
Affiliation(s)
- Hamed Laroui
- Laboratoire de Chimie Physique Macromoléculaire, UMR CNRS-INPL 7568, BP 20451, 54001 Nancy Cedex, France
| | | | | | | | | | | | | |
Collapse
|
15
|
Pringle IA, McLachlan G, Collie DDS, Sumner-Jones SG, Lawton AE, Tennant P, Baker A, Gordon C, Blundell R, Varathalingam A, Davies LA, Schmid RA, Cheng SH, Porteous DJ, Gill DR, Hyde SC. Electroporation enhances reporter gene expression following delivery of naked plasmid DNA to the lung. J Gene Med 2007; 9:369-80. [PMID: 17410613 DOI: 10.1002/jgm.1026] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Existing methods of non-viral airway gene transfer suffer from low levels of efficiency. Electroporation has been used to enhance gene transfer in a range of tissues. Here we assess the usefulness of electroporation for enhancing gene transfer in the lungs of mice and sheep. METHODS Naked plasmid DNA (pDNA) expressing either luciferase or green fluorescent protein (GFP) was delivered to mouse lungs by instillation. Following surgical visualisation, the lungs were directly electroporated and the level and duration of luciferase activity was assessed and cell types that were positive for GFP were identified in lung cryosections. Naked pDNA was nebulised to the sheep lung and electrodes attached to the tip of a bronchoscope were used to electroporate airway segment bifurcations, Luciferase activity was assessed in electroporated and control non-electroporated regions, after 24 h. RESULTS Following delivery of naked pDNA to the mouse lung, electroporation resulted in up to 400-fold higher luciferase activity than naked pDNA alone when luciferase was under the control of a cytomegalovirus (CMV) promoter. Following delivery of a plasmid containing the human polyubiquitin C (UbC) promoter, electroporation resulted in elevated luciferase activity for at least 28 days. Visualisation of GFP indicated that electroporation resulted in increased GFP detection compared with non-electroporated controls. In the sheep lung electroporation of defined sites in the airways resulted in luciferase activity 100-fold greater than naked pDNA alone. CONCLUSIONS These results indicate that electroporation can be used to enhance gene transfer in the lungs of mice and sheep without compromising the duration of expression.
Collapse
Affiliation(s)
- Ian A Pringle
- GeneMedicine Research Group, Nuffield Department of Clinical Laboratory Sciences, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Abstract
OBJECTIVES To provide a comprehensive literature review describing recent developments of the recombinant adeno-associated virus (rAAV) vector and exploring the therapeutic application of rAAV for bone defects, cartilage lesions and rheumatoid arthritis. DESIGN Narrative review. RESULT The review outlines the serotypes and genome of AAV, integration and life cycle of the rAAV vectors, the immune response and regulating system for AAV gene therapy. Furthermore, the advancements of rAAV gene therapy for bone growth together with cartilage repair are summarized. CONCLUSION Recombinant adeno-associated virus vector is perceived to be one of the most promising vector systems for bone and cartilage gene therapy approaches and further investigations need to be carried out for craniofacial research.
Collapse
Affiliation(s)
- Juan Dai
- The Biomedical and Tissue Engineering Group, Department of Orthodontics, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China
| | | |
Collapse
|
17
|
van de Loo FAJ, Geurts J, van den Berg WB. Gene therapy works in animal models of rheumatoid arthritis...so what! Curr Rheumatol Rep 2006; 8:386-93. [PMID: 16973113 DOI: 10.1007/s11926-006-0070-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Rheumatoid arthritis (RA) is a systemic disease with polyarticular manifestation of chronic inflammation in the knees and small joints of hand and feet. The current systemic anti-tumor necrosis factor (TNF)-alpha therapies with biologics ameliorate disease in 60% to 70% of RA patients. However, biologics must be given systemically in relatively high dosages to achieve constant therapeutic levels in the joints, and side effects have been reported. To this end, local gene delivery can provide an alternative approach to achieve high, long-term expression of biologics, optimizing the therapeutic efficacy and minimizing systemic exposure. Evidence from animal models convincingly supports the application of local gene therapy in rheumatoid arthritis, but preclinical studies remain necessary to evaluate the merge of cell-specific targeting, viral vector development, and disease-regulated transgene expression to optimize efficacy and safety.
Collapse
Affiliation(s)
- Fons A J van de Loo
- Rheumatology Research and Advanced Therapeutics, Department of Rheumatology, Radboud University Nijmegen Medical Centre, Nijmegen Centre for Molecular Life Sciences, Geert Grooteplein 28, 6525 GA Nijmegen, The Netherlands.
| | | | | |
Collapse
|
18
|
Bloquel C, Trollet C, Pradines E, Seguin J, Scherman D, Bureau MF. Optical imaging of luminescence for in vivo quantification of gene electrotransfer in mouse muscle and knee. BMC Biotechnol 2006; 6:16. [PMID: 16524461 PMCID: PMC1431530 DOI: 10.1186/1472-6750-6-16] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2005] [Accepted: 03/08/2006] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Optical imaging is an attractive non-invasive way to evaluate the expression of a transferred DNA, mainly thanks to its lower cost and ease of realization. In this study optical imaging was evaluated for monitoring and quantification of the mouse knee joint and tibial cranial muscle electrotransfer of a luciferase encoding plasmid. Optical imaging was applied to study the kinetics of luciferase expression in both tissues. RESULTS The substrate of luciferase (luciferin) was injected either intraperitonealy (i.p.) or in situ into the muscle or the knee joint. Luminescence resulting from the luciferase-luciferin reaction was measured in vivo with a cooled CCD camera and/or in vitro on tissue lysate. Maximal luminescence of the knee joint and muscle after i.p. (2.5 mg) or local injection of luciferin (50 microg in the knee joint, 100 microg in the muscle) were highly correlated. With the local injection procedure adopted, in vivo and in vitro luminescences measured on the same muscles significantly correlated. Luminescence measurements were reproducible and the signal level was proportional to the amount of plasmid injected. In vivo luciferase activity in the electrotransfered knee joint was detected for two weeks. Intramuscular electrotransfer of 0.3 or 3 microg of plasmid led to stable luciferase expression for 62 days, whereas injecting 30 microg of plasmid resulted in a drop of luminescence three weeks after electrotransfer. These decreases were partially associated with the development of an immune response. CONCLUSION A particular advantage of the i.p. injection of substrate is a widespread distribution at luciferase production sites. We have also highlighted advantages of local injection as a more sensitive detection method with reduced substrate consumption. Besides, this route of injection is relatively free of uncontrolled parameters, such as diffusion to the target organ, crossing of biological barriers and evidencing variations in local enzymatic kinetics, probably related to the reaction medium in the targeted organ. Optical imaging was shown to be a sensitive and relevant technique to quantify variations of luciferase activity in vivo. Further evaluation of the effective amount of luciferase in a given tissue by in vivo optical imaging relies on conditions of the enzymatic reaction and light absorption and presently requires in vitro calibration for each targeted organ.
Collapse
Affiliation(s)
- C Bloquel
- Inserm, U640, Paris, F-75006 France; CNRS, UMR8151, Paris, F-75006 France; Université Paris Descartes, Faculté de Pharmacie, Chemical and Genetic Pharmacology Laboratory, Paris, F-75270 France; Ecole Nationale Supérieure de Chimie de Paris, Paris, F-75005, France
| | - C Trollet
- Inserm, U640, Paris, F-75006 France; CNRS, UMR8151, Paris, F-75006 France; Université Paris Descartes, Faculté de Pharmacie, Chemical and Genetic Pharmacology Laboratory, Paris, F-75270 France; Ecole Nationale Supérieure de Chimie de Paris, Paris, F-75005, France
| | - E Pradines
- Inserm, U640, Paris, F-75006 France; CNRS, UMR8151, Paris, F-75006 France; Université Paris Descartes, Faculté de Pharmacie, Chemical and Genetic Pharmacology Laboratory, Paris, F-75270 France; Ecole Nationale Supérieure de Chimie de Paris, Paris, F-75005, France
| | - J Seguin
- Inserm, U640, Paris, F-75006 France; CNRS, UMR8151, Paris, F-75006 France; Université Paris Descartes, Faculté de Pharmacie, Chemical and Genetic Pharmacology Laboratory, Paris, F-75270 France; Ecole Nationale Supérieure de Chimie de Paris, Paris, F-75005, France
| | - D Scherman
- Inserm, U640, Paris, F-75006 France; CNRS, UMR8151, Paris, F-75006 France; Université Paris Descartes, Faculté de Pharmacie, Chemical and Genetic Pharmacology Laboratory, Paris, F-75270 France; Ecole Nationale Supérieure de Chimie de Paris, Paris, F-75005, France
| | - MF Bureau
- Inserm, U640, Paris, F-75006 France; CNRS, UMR8151, Paris, F-75006 France; Université Paris Descartes, Faculté de Pharmacie, Chemical and Genetic Pharmacology Laboratory, Paris, F-75270 France; Ecole Nationale Supérieure de Chimie de Paris, Paris, F-75005, France
| |
Collapse
|
19
|
Grossin L, Cournil-Henrionnet C, Pinzano A, Gaborit N, Dumas D, Etienne S, Stoltz JF, Terlain B, Netter P, Mir LM, Gillet P. Gene transfer with HSP 70 in rat chondrocytes confers cytoprotection in vitro and during experimental osteoarthritis. FASEB J 2006; 20:65-75. [PMID: 16394269 DOI: 10.1096/fj.04-2889com] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Osteoarthritis is characterized by a gradual degradation of extracellular matrix, resulting from an excess of chondrocyte cell death, mainly due to an increase in apoptotis. Recent studies have revealed the essential role of HSP70 in protecting cells from stressful stimuli. Therefore, overexpressing HSP70 in chondrocytes could represent a good strategy to prevent extracellular matrix destruction. To this end, we have developed a vector carrying HSP70/GFP, and transduced chondrocytes were thus more resistant to cell death induced by mono-iodoacetate (MIA). To overcome the barrier-effect of matrix, we investigated the efficacy of plasmid delivery by electroporation (EP) in rat patellar cartilage. Two days after EP, 50% of patellar chondrocytes were HSP/GFP+. After 3 months, long-term expression of transgene was only depicted in the deep layer (20-30% positive cells). HSP70 overexpression inhibited the natural endochondral ossification in the deep layer, thus leading to a lesser decrease in chondrocyte distribution. Moreover, overexpression of HSP70, after a preventive EP transfer in rat patella, was sufficient to decrease the severity of osteoarthritis-induced lesions, as demonstrated histologically and biochemically. In conclusion, intracellular overexpression of HSP70, through EP delivery, could protect chondrocytes from cellular injuries and thus might be a novel chondroprotective modality in rat OA.
Collapse
Affiliation(s)
- Laurent Grossin
- Unité Mixte de Recherches 7561, Centre National de la Recherche Scientifique-Université Henri Poincaré Nancy 1, Faculté de Médecine, Vandoeuvre lès Nancy, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Khoury M, Bigey P, Louis-Plence P, Noel D, Rhinn H, Scherman D, Jorgensen C, Apparailly F. A comparative study on intra-articular versus systemic gene electrotransfer in experimental arthritis. J Gene Med 2006; 8:1027-36. [PMID: 16733831 DOI: 10.1002/jgm.922] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Electric pulse mediated gene transfer has been applied successfully in vivo for increasing naked DNA administration in various tissues. To achieve non-viral gene transfer into arthritic joint tissue, we investigated the use of electrotransfer (ET). Because anti-inflammatory cytokine strategies have proven efficient in experimental models of arthritis, we compared the therapeutic efficiency of local versus systemic delivery of the interleukin-10 (IL-10) using in vivo ET. METHODS A plasmid vector expressing IL-10 was transferred into DBA/1 mouse knee joints by ET with 12 pulses of variable duration and voltage. The kinetics of transgene expression were analyzed by specific enzyme-linked immunosorbent assay (ELISA) in sera and knees. Optimal conditions were then used to deliver increasing amounts of IL-10 plasmid intra-articularly (i.a.) in the collagen-induced arthritis (CIA) mouse model. The therapeutic efficiency was compared with the potency of intra-muscular (i.m.) ET. RESULTS Following i.a. ET, local IL-10 secretion peaked on day 7 and dropped 2 weeks after. A second ET produced the same kinetics without enhancing gene transfer efficiency, while transgene was still detected in injected muscles 4 weeks after ET. Only the i.m. ET of 25 microg of IL-10 significantly inhibited all the clinical and biological features of arthritis. The i.a. ET only showed mild improvement of arthritis when 100 microg of IL-10 plasmid were electrotransfered weekly from day 18 following arthritis induction. CONCLUSIONS The present results suggest that gene transfer into arthritic joints by ET is an effective means to deliver anti-inflammatory cytokines. However, short duration of transgene expression impedes a significant effect for the treatment of arthritis, making i.m. ET more potent than i.a. ET for clinical benefit in CIA.
Collapse
Affiliation(s)
- M Khoury
- Inserm, U 475, F-34000 Montpellier, France
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Galois L, Freyria AM, Herbage D, Mainard D. Ingénierie tissulaire du cartilage : état des lieux et perspectives. ACTA ACUST UNITED AC 2005; 53:590-8. [PMID: 16364811 DOI: 10.1016/j.patbio.2004.12.019] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2004] [Accepted: 12/09/2004] [Indexed: 11/24/2022]
Abstract
Lesions of the articular cartilage have a large variety of causes among which traumatic damage, osteoarthritis and osteochondritis dissecans are the most frequent. Returning damaged cartilage in articular joints back to a functionally normal state has been a major challenge for orthopaedic surgeons. This interest results in large part because cartilage defects cannot adequately heal themselves. Current techniques used in orthopaedic practice to repair cartilage give variable and unpredictable results. Bone marrow stimulation techniques such as abrasion arthroplasty, drilling and microfracture produce mostly fibrocartilage. Autologous osteochondral transplant systems (mosaicplasty) have shown encouraging results. Autologous chondrocyte transplantation has led to a hyaline articular cartilage repair but little is known about the predictability and reliability of the procedure. The rapidly emerging field of tissue engineering promises creation of viable substitutes for failing cartilage tissue. Current tissue engineering approaches are mainly focused on the restoration of pathologically altered tissue structure based on the transplantation of cells in combination with supportive matrices and molecules. Among natural and synthetic matrices, collagen and polysaccharidic biomaterials have been extensively used with promising results. Recently, interest has switched to the use of mesenchymal stem cells instead of chondrocytes. Tissue engineering offers the possibility to treat localised cartilage lesions. Genetic engineering techniques using genetically modified chondrocytes offer also the opportunity to treat diffuse cartilage lesions occurring in osteoarthritis or inflammatory joint diseases. Electroporation is specially a reliable and inexpensive technique that shares with electrochemotherapy an ability to target the chondrocytes despite the barrier effect of the extracellular matrix without viral vectors. The authors review recent research achievements and highlight the potential clinical applications of new technologies in the treatment of patients with cartilage injuries.
Collapse
Affiliation(s)
- L Galois
- Département de chirurgie orthopédique et traumatologique, hôpital central, 29, avenue Maréchal-de-Lattre-de-Tassigny, 54035 Nancy cedex, France.
| | | | | | | |
Collapse
|
22
|
Bakker JM, Bleeker WK, Parren PWHI. Therapeutic antibody gene transfer: an active approach to passive immunity. Mol Ther 2005; 10:411-6. [PMID: 15336642 DOI: 10.1016/j.ymthe.2004.06.865] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2004] [Revised: 06/18/2004] [Accepted: 06/18/2004] [Indexed: 01/08/2023] Open
Abstract
Advances in gene transfer approaches are enabling the possibility of applying therapeutic antibodies using DNA. In particular gene transfer in combination with electroporation is promising and can result in generating in vivo antibody concentrations in the low therapeutic range. However, several important problems need to be dealt with before antibody gene transfer can become a valuable supplement to the current therapies. As antibody production following gene transfer is difficult to control, the danger of inducing autoimmune conditions or uncontrollable side effects occurs in cases in which autologous antigens are targeted. It is suggested that the most promising area of application therefore appears to be infectious disease in which heterologous antigens are targeted and concerns for long-term antibody exposure are minimal. Finally, genes encoding fully human antibodies will enhance long-term expression and decrease problems linked to immunogenicity.
Collapse
Affiliation(s)
- Joost M Bakker
- Genmab B.V., Yalelaan 60, P.O. Box 85199, 3508 AD Utrecht, The Netherlands
| | | | | |
Collapse
|
23
|
Goto T, Nishi T, Kobayashi O, Tamura T, Dev SB, Takeshima H, Kochi M, Kuratsu JI, Sakata T, Ushio Y. Combination electro-gene therapy using herpes virus thymidine kinase and interleukin-12 expression plasmids is highly efficient against murine carcinomas in vivo. Mol Ther 2004; 10:929-37. [PMID: 15509510 DOI: 10.1016/j.ymthe.2004.07.028] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2004] [Accepted: 07/27/2004] [Indexed: 10/26/2022] Open
Abstract
We report the use of plasmid DNA-mediated combination gene therapy for tumor-bearing mice using in vivo electroporation, also called electro-gene therapy (EGT), that resulted in uncomplicated and complete cures in more than 90% of the mice. Subcutaneously inoculated CT26 tumors in syngeneic BALB/c mice were subjected to repeated EGT treatments consisting of intratumoral co-injection of naked plasmids encoding the cytokine interleukin-12 (IL-12) (p35 and p40 subunits) and the suicide gene herpes simplex virus thymidine kinase (HSV-tk), followed by in vivo electroporation. The early anti-tumor effect was always stronger, and the rate of cure, as seen in the long-term follow-up, was always greater in the groups treated with combination EGT than in those treated with IL-12 or HSV-tk EGT alone. Systemic levels of IL-12 and IFN-gamma increased in both combination and IL-12-alone EGT-treated groups. Moreover, combination EGT for established subcutaneous tumors strongly reduced hematogenous lung metastases and increased survival time when live CT26 tumor cells were injected through the tail vein. Limited experiments on C57/B16 mice with murine melanoma also showed very similar trends. These results suggest that this simple and safe method of plasmid-mediated combination EGT may provide a potentially effective gene therapy for cancer.
Collapse
Affiliation(s)
- Tomoaki Goto
- Department of Neurosurgery, Saiseikai Kumamoto Hospital, 5-3-1 Chikami, Kumamoto 861-4193, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Mohan RM, Golding S, Heaton DA, Danson EJ, Paterson DJ. Targeting neuronal nitric oxide synthase with gene transfer to modulate cardiac autonomic function. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2004; 84:321-44. [PMID: 14769442 DOI: 10.1016/j.pbiomolbio.2003.11.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Microdomains of neuronal nitric oxide synthase (nNOS) are spatially localised within both autonomic neurons innervating the heart and post-junctional myocytes. This review examines the use of gene transfer to investigate the role of nNOS in cardiac autonomic control. Furthermore, it explores techniques that may be used to improve upon gene delivery to the cardiac autonomic nervous system, potentially allowing more specific delivery of genes to the target neurons/myocytes. This may involve modification of the tropism of the adenoviral vector, or the use of alternative viral and non-viral gene delivery mechanisms to minimise potential immune responses in the host. Here we show that adenoviral vectors provide an efficient method of gene delivery to cardiac-neural tissue. Functionally, adenovirus-nNOS can increase cardiac vagal responsiveness by facilitating cholinergic neurotransmission and decrease beta-adrenergic excitability. Whether gene transfer remains the preferred strategy for targeting cardiac autonomic impairment will depend on site-specific promoters eliciting sustained gene expression that results in restoration of physiological function.
Collapse
Affiliation(s)
- R M Mohan
- University Laboratory of Physiology, University of Oxford, Parks Road, Oxford OX1 3PT, UK
| | | | | | | | | |
Collapse
|
25
|
Abstract
Osteoarthritis (OA) is a major health problem in urgent need of better treatment. Gene therapy offers to meet this need. Of the different strategies for using gene therapy in OA, local gene transfer to synovium is in the most advanced stage of development. Local gene transfer brings several advantages, including a focused, local therapy that promises greater efficacy with reduced side-effects, potentially at far lower cost. Moreover, its clinical feasibility has already been confirmed in two Phase I studies of gene therapy for rheumatoid arthritis. Although there are numerous candidate genes of potential use in treating OA genetically, considerable evidence identifies interleukin-1 (IL-1) as a key target. The existence of a natural antagonist, the IL-1 receptor antagonist (IL-1Ra), provides a means with which to inhibit its biologic actions. Clinical studies are suggested in which IL-1Ra complementary DNA is transferred to knee joints shortly before they are surgically replaced with prostheses. This will permit the ready assessment of the safety and efficiency of gene transfer and expression in the human OA knee, as well as permitting preliminary functional data to be obtained, as a prelude to phase II efficacy studies. At this point, the major barriers to progress are financial rather than intellectual or technical.
Collapse
Affiliation(s)
- Christopher H Evans
- Center for Molecular Orthopedics, Harvard Medical School, 221 Longwood Avenue, BLI-152, Boston, MA 02115, USA.
| |
Collapse
|
26
|
Grossin L, Gaborit N, Mir L, Netter P, Gillet P. Gene therapy in cartilage using electroporation. Joint Bone Spine 2003; 70:480-2. [PMID: 14667558 DOI: 10.1016/j.jbspin.2003.07.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Affiliation(s)
- Laurent Grossin
- UMR 7561 CNRS, Laboratoire de pharmacologie, faculté de médecine de Nancy, Université Nancy I, avenue de la Foret-de-Haye BP 184, 54505 Vandoeuvre-les-Nancy, France
| | | | | | | | | |
Collapse
|