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.
Keywords: osteoarthritis, gene therapy, expression vectors, CRISPR/Cas9
Introduction
Osteoarthritis (OA) is a degenerative joint disease that causes pain and disability in 250 million people.1,2 Rather than defined as a single mechanically induced disease as before, OA has been increasingly considered to be a complex inflammatory and metabolic syndrome involving the entire joint tissue.3 The most significant change of OA is degeneration in articular cartilage. Components derived from degraded cartilage trigger foreign body reaction in synoviocytes to induce synovitis. Synovitis leads to the production of metalloproteinases and inflammatory factors and synovial angiogenesis, which further aggravates cartilage destruction.4 Current treatment approaches, which focus on alleviating pain and restoring motor function, include pharmacotherapy, non-pharmacotherapy, patient education and self-management.5 Pharmacotherapy is indicated for moderate and severe OA patients to alleviate pain, improve joint function and quality of life. Non-pharmacological treatments mainly refer to arthroplasty, which is the most effective treatment for patients with end-stage OA.6 The disadvantage of drug therapy is that patients need to take long-term drugs that pose a high risk to the gastrointestinal and cardiovascular systems such as non-steroidal anti-inflammatory drugs (NSAIDs) and cyclooxygenase 2 (COX2).7 Although surgical treatment can significantly improve patients' motor function, postoperative bleeding and infection as well as traumatic damage caused by surgery are still of concern. Due to the change in perception of OA, the therapeutic principle of OA has shifted from support to regeneration or prevention. In order to slow down or reverse the process of cartilage degeneration, disease-modifying osteoarthritis drugs (DMOADs) have been proposed due to their effect of alleviating structural exacerbation of OA while relieving symptoms.8
As a result of advances in biological sciences, genetic engineering has recently evolved into an effective method for cartilage regeneration. Compared with traditional treatment methods, gene therapy has advantages such as effective targeting, prolonged half-life and minor side effects. In addition, gene therapy, either alone or in combination with cells and scaffolds, can alleviate local inflammation and metabolism in OA9 and even promote cartilage repair.10 Therefore, the study of gene therapy in OA will contribute to the development of DMOADs and application of long-term clinical translation. In this review, we summarize the current applications of gene therapy in OA, and discuss the choice of vector, gene, and mode of delivery for gene therapy. Finally, we highlight the development and application of CRISPR/Cas9 in OA therapy and indicate the difficulties and solutions for extending OA gene therapy to clinical applications.
History of gene therapy and its development in OA
Gene therapy is described by the US Food and Drug Administration (FDA) as “items that mediate their effects through transcriptional and/or translational transfer of genetic material and/or integration into the host genome, including nucleic acids, viruses, or genetically modified microbes”. These items can be used to modify cells in vivo directly or ex vivo cells before delivering cells to recipients.11 In 1989, Rosenberg et al. used lentiviruses to genetically modify tumor-infiltrating lymphocytes (TILs) and thereafter transfused transgenic TILs back into patients. It was the first ever approved trial to transfer the gene into a human, demonstrating the feasibility of retroviral gene transfer in human gene therapy.12,13 In 1990, the FDA approved the first clinical trial of therapeutic gene transfer in humans. Blaese et al. used retroviruses to transfer genes mediating adenosine deaminase (ADA) expression into T cells from two children with severe combined immunodeficiency (ADA-SCID).14 The study has been a driving force in the application of gene therapy in clinical trials.
Interleukin-1 (IL-1), which is induced in OA-associated cartilage and synovial tissue, is one of the main factors in the disease. IL-1 enhances cartilage catabolism by up-regulating extracellular proteolytic enzymes, accelerates the inflammatory response of OA, and inhibits cartilage anabolism by down-regulating collagen and proteoglycan production.15,16 The endogenous IL-1 receptor antagonist (IL-1Ra), which competes with IL-1 for IL-1 receptor binding, is one of the most potent inhibitors of the IL-1 pathway as it blocks the initiation of the signal transduction.17 The earliest gene therapy for OA was focused on inhibiting the IL-1 pathway in articular cartilage and synovium. The initial study was to transduce IL-1Ra cDNA into rabbit synovial fibroblasts using retroviruses, and then transgenic synoviocytes were transplanted into rabbit knees by intra-articular injection. It was shown that the transgenic synovial cells could protect against damage triggered by exogenous IL-1β injections .18 Baragi et al. transfected human articular cartilage monolayer culture with adenovirus vector carrying human IL-1Ra gene, and found that the genetically modified chondrocytes were resistant to IL-1β-induced degradation of the extracellular matrix (ECM).19 Pelletier et al. demonstrated for the first time in vivo that transplantation of synoviocytes transduced with the IL-1Ra gene following intra-articular injection delayed the progression of early anterior cruciate ligament transection (ACLT) model induced OA.20 The results showed that human IL-1Ra gene could be effectively introduced into canine synoviocytes by retrovirus infection and produce high levels of IL-1Ra in transgenic knee joints under the experimental conditions. In summary, these early studies collectively indicated gene therapy as a promising treatment option for OA, setting the stage for a large number of subsequent studies.
Targets
Unlike gene therapy for inherited diseases, which is dedicated to replacing defective genes with normal ones, gene therapy for OA is aimed at overexpressing therapeutic genes or downregulating the expression of deleterious genes by direct or indirect means. Currently, the function of genes used in the treatment of OA typically includes (1) promotion of chondrocyte proliferation and growth; (2) inhibition of chondrocyte senescence and apoptosis; (3) elevation of anabolism resulting in more ECM production; (4) prevention of inflammation, catabolism, and synthesis of matrix degrading enzymes.
Transcription factors
The main transcription factors involved in the progression of OA include Runt–related transcription factor (Runx1,2,3), Sex-determining region Y (Sry)-box-containing family (Sox5,6,9), CCAAT/enhancer binding protein (C/EBPβ) and Hypoxia-inducible factor (HIF-1α,2α).21 Runx2 is a transcription factor essential for chondrocyte maturation and bone formation in normal cartilage. It promotes the expression of catabolic factors into the extracellular matrix of cartilage and is upregulated in human osteoarthritic cartilage and in mouse articular cartilage after joint injury.22 Runx3 acts similarly to Runx2 and also regulates chondrocyte hypertrophy, although Runx2 has a more pronounced effect.23 In contrast, upregulation of Runx1 mRNA by intra-articular injection ameliorates the progression of OA in mice, including osteophyte formation and cartilage destruction.24 Runx2 and C/EBP work closely together physically and functionally to stimulate the production of matrix metallopeptidase-13 (MMP13), the main collagenase of type 2 collagen, leading to the progress of OA.25 Sox9 is a transcription factor responsible for chondrogenesis and promotes the expression of Coll II. Delivery of the Sox9 gene via the recombinant adeno-associated virus (rAAV) vector durably enhances the repair process at sites of osteochondral damage in sheep and counteracts the progression of focal OA in adjacent joints.26 HIF-1α promotes cartilage formation through upregulation of Sox9 expression. In addition, HIF-1α protects articular cartilage by promoting chondrocyte differentiation, maintaining chondrocyte viability, and supporting metabolic adaptation to hypoxic environments. In contrast to HIF-1α, HIF-2α directly induces chondrocyte catabolism, mediates chondrocyte apoptosis and regulates mature chondrocyte autophagy.27,28
Growth factors
Insulin-like growth factor 1(IGF-1) is closely involved in cartilage formation and maintenance. IGF-1 promotes chondrocyte proliferation, enhances ECM production, and inhibits ECM degradation and chondrocyte apoptosis. However, under OA condition, the chondrocytes produce excessive IGF receptor binding protein. In addition, the arthritis related inflammation environment accelerates IGF-1 degradation. Thus, direct injection of IGF-1 into the joint cannot achieve the expected effect in most cases.29 Implantation of chondrocytes overexpressing IGF-1 at the site of injury has shown to improve predominance and type 2 collagen production in articular cartilage.30 The transforming growth factor-β (TGFβ) is also implicated in OA and cartilage biology. TGFβ superfamily consists of nearly forty ligands, and in cartilage, the main representatives of this superfamily are TGF-β, bone morphogenetic protein (BMP) and growth/differentiation factor-5 (GDF‐5). All of them are essential for normal joint development and homeostasis, and have been implicated in the pathogenesis of OA.31 TGF-β is beneficial to cartilage because it stimulates chondrocytes in vitro to induce an increase in proteoglycan and type 2 collagen production. In addition, TGF-β can also resist IL-1β and TNF-α induced catabolism.32 BMP is generally thought to have a protective effect on articular cartilage, but it has also shown to be involved in chondrocyte hypertrophy and ECM degradation. Steinert et al. transferred genes for BMP-2 and BMP-4 into human bone marrow-derived mesenchymal stem cells (MSCs) and found that BMP promoted upregulation of various cartilage hypertrophy and maturation indicators. This suggests that the therapeutic use of BMPs for OA should be used cautiously.33 GDF-5 can stimulate the expression of aggrecan (ACAN) and Sox9 in human chondrocytes, and thus promote anabolism. In addition, GDF-5 also inhibits the expression of ECM degrading enzyme MMP-13 and disintegrin and metalloproteinase with thrombospondin motifs-4 (ADAMTS4).34 Fibroblast growth factor 2(FGF-2) and FGF-18 of the fibroblast growth factor family also have regulatory effects on cartilage. FGF-18 is an anabolic factor that promotes articular cartilage repair and cartilage formation. The role of FGF-2 in cartilage metabolism is currently controversial. Some reports suggest that FGF-2 is involved in catabolism, while others show that it inhibits ADAMTS-5 and delays cartilage degradation in murine OA.35,36 Indeed, co-transfection of multiple growth factor genes into cells has a better therapeutic effect on OA than single gene transfer into cells. For example, transfer of IGF-1 combined with FGF into rabbit knee joint via adenoviral vector induced more type 2 collagen expression and reduced ECM enzyme expression.37
Inflammation-related cytokines
The disruption of cytokine homeostasis is one of the most prominent contributors in the pathogenesis of OA. This is primarily because the process favors the generation of pro-inflammatory cytokines that degrade cartilage and other intra-articular structures by activating catabolic enzymes. Among all the cytokines, IL-1β, TNF-α, and IL-6 are the most significant inflammatory mediators in the etiology of OA.38 IL-4 and IL-10 are major anti-inflammatory factors in the pathology of OA and have significant chondroprotective effects, alleviating chondrocyte apoptosis either alone or in combination.39 Therefore, down-regulation of pro-inflammatory factors or up-regulation of anti-inflammatory factors by gene therapy is a promising treatment for OA. Intra-articular overexpression of IL-1β and TNF-α antagonists via viral vectors has shown to reduce ECM degradation, attenuate chondrocyte apoptosis, and slow the progression of OA. In addition, completed or ongoing clinical trials support the effectiveness of this approach for the treatment of OA.16,40 MSCs spheroids with the expression of IL-4 gene have also been reported to alleviate painful symptoms of OA in rats and provide durable protection against cartilage.41 Intra-articular administration of AAV5-IL-10 in horses achieved sustained local IL-10 expression in the joint and resulted in a significant decrease in IL-1β and IL-6 expression.42 It is worth mentioning that the combination of anti-inflammatory factors is more helpful in terms of therapeutic effect than the single use.43
Non-coding RNAs
With the rising number of high-throughput profiling studies and mechanistic studies of microRNAs (miRNAs), long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs) in joint tissues and biological fluids, non-coding RNAs (ncRNAs) have been shown to possess a significant regulatory role in the pathogenesis of OA. ncRNAs regulate a wide range of physiological processes (inflammation, senescence, oxidative stress, chondrogenic differentiation, autophagy, methylation), signaling molecules (TGF-β, NFκB, Wnt–β-catenin) and mediators (FGF18, Sox9, Sox5, Hedgehog). These regulatory roles of ncRNAs directly or indirectly influence the inflammatory response, cell death and extracellular matrix production in OA pathology.44 A variety of ncRNAs can inhibit IL-1β and TNF-α-induced inflammatory responses in chondrocytes by acting upstream and downstream of the NFκB pathway or on pathway-related molecules. Inhibition of IL-1β-mediated cartilage inflammatory responses is executed by targeting connexin 43 (CX43) miR-382–3p via TLR4/MyD88/NF-κB signaling pathway.45 LncRNA SNHG1 attenuates IL-1β-induced OA by inhibiting miR-16–5p-mediated NF-κB signaling pathway.46 The effect of ncRNAs on apoptosis and proliferation of chondrocytes usually occurs via the phosphoinositide 3-kinase (PI3K)-AKT signaling pathway. For example, overexpression of miR-455–3p in chondrocytes ameliorates apoptosis and mitigates OA progression by modulating the PI3K/AKT pathway.47 In addition, miR-20 blocks chondrocyte proliferation and autophagy by targeting ATG10 via the PI3K/AKT/mTOR signaling pathway.48 Furthermore, ncRNAs in most cases affect the progression of OA by regulating ECM enzymes. For example, both circCDR1as and circTMBIM6 can upregulate MMP13 expression through the mechanism of competing endogenous RNAs (ceRNAs).49,50 CircRNA has been studied as a biomarker for various diseases due to its higher stability compared to other ncRNAs. For example, circ_016901, circ_0032131, circ_0044235 and circ0000175 in peripheral blood have been suggested as markers for the early diagnosis of arthritis.51,52
Small nucleolar RNA (snoRNA) is a class of ncRNAs ranging from 60 to 300 nt in length and is closely related to the splicing and processing of ribosomal RNA (rRNA) precursors, post-transcriptional modification processes and ribosome biosynthesis.53 Studies have shown that snoRNAs can also be used as diagnostic biomarkers and therapeutic targets for joint aging and OA. The results of SnoRNASeq showed 27 differential snoRNAs in serum from young and aged mice and 18 differential snoRNAs in serum from sham-operated groups and experimental OA group.54 The results of snoRNA matrix analysis of cartilage from young, old and OA patients showed that SNORD26 and SNORD96A were associated with aging while SNORD26 and SNORD116 were associated with the development of OA.55
Gene delivery vectors
A gene must be put into a vector for proper expression in a target cell. The ideal vector is chosen based on two criteria: (1) effective gene transfer with simple access to target cells and steady gene transcription after entering the nucleus; and (2) safety with minimal consequences of infection, mutation, or immunogenicity.56 Currently, two types of vectors are commonly used, viral vectors and non-viral vectors, which have different advantages and disadvantages.
Viral vectors
The transmission of particular cell types with extreme specificity and dependability is the major benefit of viral vectors.57 On the other hand, viral vectors also have drawbacks, including limited carrying capacity of therapeutic genetic material, invasive administration, and the possibility of unfavorable immune reactions or subsequent genetic impacts.58,59 Typically, three recombinant viral vectors are used as appropriate means of delivering therapeutic genes directly and specifically into cells, including retroviruses, adenoviruses and AAV.60
Retrovirus
The vectors of the retroviral family are spherical enclosed viruses with a diameter of about 100 nm. Their genomes are made up of two copies of sense (+) strand RNA. Retrovirus is often identified by the ability of reverse transcribing its RNA genome into cDNA, and permanently integrates these cDNA copies into the genome of the host cell. This allows maintenance of gene expression over a longer time span.61 The most often used retroviral vectors are gamma retroviral vectors and lentiviral vectors. These vectors are characterized by their efficient gene transfer capabilities and ability to integrate into the cellular genome so as to achieve stable genetic modification of the target cells.62 Among retroviruses, lentiviral vectors are more suitable for transfection of non-dividing cells. Studies showed that the efficiency of transfection of chondrocytes using lentivirus was up to 95% and that of MSCs was 70%.63,64 The biggest drawback of retroviral applications is the possibility of insertional mutations and oncogene formation.65,66 Therefore, the use of retroviruses for gene therapy in OA patients requires further investigation.
Adenovirus
At present, the most commonly used adenoviral vectors are based on human adenovirus type 5 (Ad5), with 36 kb long genome and linear double-stranded DNA structure. Adenoviruses exist in the form of loose bodies in cells.67 The advantages of adenoviral vectors include a wide range of tropism, effective transduction of dividing and non-dividing cells, high transduction efficiency and rapid onset of action and relatively large packaging capacity.68 Adenoviruses have been used to deliver genes encoding growth factors,37,69 transcription factors,28,70–75 enzymes,76–80 and cytokines81,82 to chondrocytes, synovial fibroblasts, and MSCs.
Because of the strong host humoral and cellular immune responses to adenoviral gene products, the use of adenoviral vectors in clinical settings has received serious attention. Using vectors that only contain genes, packaging sequences and flanking viral end duplications can help reduce immune responses, which is one approach to addressing this issue. However, this strategy requires the use of another helper virus to achieve cell transduction.40
Helper-dependent adenovirus (HDV) vectors lack the coding sequences of the main components of adenoviruses and contain only about 500 bp cis-acting elements for adenoviral DNA replication and packaging. Therefore, in addition to helper viruses, the process also requires all proteins required for adenoviral vector packaging. Compared with adenoviral vectors, HDV vectors have advantages such as long transgene expression time, large transgene capacity, excellent safety and less side effects.83 Previous studies have shown that HDV can transduce chondrocytes and synoviocytes more efficiently compared to AAV. Additionally, HDV-transduced genes might continue to express for more than a year after being injected into the knee joints of normal mice.84 FX201, constructed based on the HDV vector of human serotype 5, is an innovative intra-articular therapeutic agent for IL-1Ra. Using FX201, the gene is delivered to joint cells and produces IL-1Ra under the control of an inflammation-sensitive promoter.84 Senter et al. demonstrated that a single intra-articular injection of FX201 7 days after ACLT model relieved osteoarthritic changes in cartilage, bone, and synovium 12 weeks after surgery.85 Moreover, FX201 persisted in the joint for at least 3 months without any adverse effects. Currently, clinical trials investigating the safety and tolerability of FX201 in patients with knee OA are ongoing.
Adeno-associated virus
AAV is a single-stranded DNA parvovirus that is non-pathogenic and non-enveloped. The primary benefits of AAV vectors include their small size, low immunogenicity, and inability of viral infection. Therefore, AAV is currently the most commonly used viral vector.86 AAV can be modified to remove viral genetic material so that it can produce targeted gene transcription without generating viral proteins.87 The most prevalent AAV serotype is AAV-2, which has a wide tissue tropism and a respectable cell infection effectiveness. Different serotypes of AAV exhibit different transduction efficiencies in chondrocytes and MSCs. Serotypes of AAV2, AAV5, AAV6 and AAV6.2 are appropriate for human chondrocyte in vitro culture systems. Different AAV serotypes also differ in tissue specificity. For example, AAV2 and AAV6.2 are better choices for hard cartilage tissue as they have a higher transduction efficiency on chondrocytes than other AAV serotypes. The high transduction efficacy of AAV2 and AAV6.2 in human chondrocytes and MSCs suggests that these viruses have stronger chondrocyte selectivity both in vitro and in vivo.88
In the current field of gene therapy, the most used AAV vectors are the rAAV vector family or self-complementary AAV (ScAAV). The rAAV cannot be integrated into genome; thus, it can be used to infect both dividing and non-dividing cells. The rAAV is thought to be the least harmful viral system because it causes relatively little innate and adaptive immune reactions to transduced cells.89 Even though rAAV's genome is still an episome, it can support in vivo long-term transgene expression in dormant cells.90 In multiple clinical studies, rAAV is the vector system of choice for human protocols involving in vivo gene delivery due to the safty.68 ScAAV complementary vectors package an inverted repeat genome that can fold into dsDNA without DNA synthesis or base pairing between multiple vector genomes, thus improving transduction efficiency.91 When testing the ability of scAAV vectors to deliver genes in joints, researchers found that in the presence of the same number of viral genomes in infected cells, the transgene expression of scAAV vectors was earlier and the gene product was approximately 25-fold higher than traditional AAV.92 The drawback of scAAV in comparison to standard AAV is its inability of carrying larger genes. Numerous studies of OA have focused on the use of rAAV and scAAV in the studies of mammals, including humans,93,94 horses,95,96 dogs,97 rabbits,92 sheep26,98 and pigs.99,100
For gene therapy, the efficiency of vector delivery has always been one of the most important concerns of researchers. Studies have shown that synovial tissue has better delivery efficiency than cartilage when adenovirus is injected directly into the joint as a vector. To assess the dose-response and cellular distribution of transduction, Ruan et al. evaluated the knee joints of mice injected with 109 or 108 HDV viral particles.84 These doses were much less than the maximum tolerated systemic dose in humans. At the higher doses, HDV transduced superficial chondrocytes and synovial cells, whereas at the lower doses only synovial cells were transduced.
Although there are still no customized strategies for cartilage and synovial gene therapy, what has been reported can provide us with some reference. For cartilage, HDV with the same titer has a higher transduction efficiency than many popular AAVs (including AAV2, AAV2.5 and AAV6).84 Combining HDV with an alpha-10 integrin monoclonal antibody (a10mab) will reposition chondrocytes, and this modified HDV can reduce the effective treatment dosage by 10-fold compared to normal HDV.101 For synovium, AAV serotype DJ (AAV-DJ) is a recombinant virus of AAV2 and AAV5, which has a better ability to evade immune neutralization and is suitable for in vitro and in vivo DNA therapy, with a higher transfection efficiency for synovial cells and a higher expression of the transfected protein in the joint fluid.102 In addition to using a different delivery vehicle, changing the timing of gene delivery can also have an impact on the site of treatment. When AAV was injected before injury, AAV transgene expression was localized to the soft tissues of uninjured and injured joints; whereas when injected after injury, AAV was able to transduce articular chondrocytes within the cartilage matrix.103
Non-viral vectors
There are two categories of design of non-viral gene delivery vectors: the delivery of DNA or RNA packaged in a vector, and bare-leakage delivery of DNA or RNA.104 The former normally utilizes physical methods (gene gun, laser, ultrasound, electroporation, microinjection) to physically deliver the target gene into the target cell. The latter involves use of chemical methods (lipofection, lipoplexes, exosomes, vesicles, nanoparticles) to penetrate cell membranes by packaging target genes into components such as lipophilic particles.105 The design of non-viral vectors has received increasing attention because they are inexpensive, easy to handle, less toxic and minor immunogenic than viral vectors.
Electroporation is a physical transfection method that promotes cellular uptake of nucleic acids or drugs by applying a high-voltage pulse to the cell membrane for a short period of time. Earlier reports suggested that gene transfer into articular cartilage and synovial cells can be performed using electroporation.106,107 Grossin et al. delivered a plasmid containing the heat shock proteins70 (HSP70) gene into rat knee cartilage by electroporation.108 Their study found that 50% of chondrocytes exhibited HSP70 positivity the day after in vivo use of electroporation; and after 3 months, long-term expression of the transgene was seen only in the deeper layers (20%–30% of positive cells). They isolated rat chondrocytes for testing at different time points of gene introduction and showed that electrical pulses do not affect ECM synthesis in articular cartilage, indicating that this method is safe for in vivo application. Another report suggests that intramuscular electroporation has a better therapeutic effect and duration for arthritis than intra-articular plasmid injection for electroporation.109
Cationic liposomes and other lipid-based systems are easy and safe to prepare. Moreover, the transport of this type of vector is not limited by the size of the delivered DNA.110 However, lipid-based systems have several disadvantages compared to viral vectors, including the low transfection efficiency, which can be affected by the amount of lipid reagent per DNA.111 FuGENE6 is a commercially available lipid transfection reagent that was used to transfect chondrocytes in vitro. The results showed that the efficiency of FuGENE6 is 41% in normal bovine articular cartilage, 20.7% in normal human articular cartilage, and 7.8% in cartilage of patients with OA.112 The integration of plasmids, and biocompatible and better-targeted components into nanoparticles can produce nanovectors with higher transfection efficiency.113 Chitosan (CS) has superior biocompatibility and biodegradability and can easily pass through cell membranes and bind to negatively-charged genes. The combination of CS, sodium hyaluronate (HA), chondroitin sulfate (CHS) and plasmid produces nanospheres, which have high stability, elevated transfection efficiency and low cytotoxicity.114 Chen et al. used the above nanoparticle-packed GDF-5 plasmid to transfect rabbit chondrocytes in vitro, to achieve a transfection rate of 60%.115
Exosomes are membrane-bound extracellular vesicles surrounded by a phospholipid bilayer and released by cells in normal or pathological states into body fluids such as blood, saliva, urine and synovial fluid.116 Cell-derived exosomes can be modified to carry exogenous genes as a non-viral delivery system.117 Exosome-cell responses and interactions are based on recognition of membrane receptors. Activation upon receptors triggers certain signaling pathways, which in turn leads to the fusion of the exosome with the plasma membrane or the exocytosis of the exosome. Exosomes can release their cargo into the receiving cell through the internalization pathway mentioned above. Therefore, exosomes as gene carriers are characterized by high targeting efficacy, good safety and low xenobiotics.118 Currently, a great number of studies have been focusing on the use of engineered exosomes for the treatment of OA.119–121
Gene delivery methods
At present, the most commonly used delivery strategies can be summarized into two categories: (1) direct delivery of the target gene to the lesion site by surgical incision or local injection (in vivo); (2) in vitro transfection or infection of the cells, followed by delivery of the cells to the target tissue (ex vivo) (Fig. 1). In addition, protocols using natural or synthetic scaffolds to carry genes or cells prior to performing the above two delivery methods have increasingly gained popularity in the field.
Figure 1.
Overview of osteoarthritis gene therapy targets, vectors, delivery methods and treatment processes. Commonly used genes include transcription factors, growth factors, inflammation-associated cytokines, and non-coding RNAs. Delivery vectors consist of viral vectors (including adenovirus, adeno-associated virus, γ-retrovirus and lentivirus) and non-viral vectors (including plasmids, exosomes, nanoparticles). Direct delivery (in vivo) refers to the direct delivery of the vector carrying the gene to the target region. Indirect delivery (ex vivo) is the genetic modification of cells (including chondrocytes, mesenchymal stem cells, induced pluripotent stem cells and fibroblasts) before delivery to the target site. TFs: transcription factors, GFs: growth factors, ncRNAs: non-coding RNAs, MSC: mesenchymal stem cell, iPS: induced pluripotent stem cell, IA: intra-articular.
In vivo methods
Direct delivery is convenient, relatively cheap and less time-consuming. However, the delivered gene via this method cannot be individually targeted to a particular cell, resulting in non-specific targeting in intra-articular cells spread, leading to potential off-target effects. Articular chondrocytes are surrounded by a dense ECM, and the rAAV vector is considered to be the most suitable option for in vivo transfection up to this point.122 At present, direct delivery methods are more commonly used to deliver anti-inflammatory genes,123 growth factors124 and ncRNAs.125
Ex vivo methods
Ex vivo gene therapy has better safety and fewer off-target effects; however, it is involved with more technical steps and is less applicable because of the technical complexity and the more stringent environmental requirements for in vitro cell amplification. Indirect gene therapy strategies for OA typically employ cells such as chondrocytes126,127 and MSCs.128,129 Chondrocytes are the only cell type in articular cartilage that can produce ECM to maintain basic joint homeostasis, and autologous articular cells are used for autologous chondrocyte implantation in cartilage repair. The source used for gene therapy in OA is mostly allogeneic cartilage. Chondrocytes isolated from allogeneic cartilage are first modified in vitro with an anti-inflammatory gene (IL-1Ra) or growth factor (TGF-β), and then injected into the patient's joint cartilage.130 TissueGene-C (TG-C), a novel gene therapy for OA, includes a mixture of human allogeneic chondrocytes and irradiated cells modified to overexpress TGF-β1. Currently, TG-C has been used in numerous animal studies and clinical trials. Transformed chondrocytes can adhere to cartilage and continuously secrete TGF-β1. Animal studies have also shown that transformed chondrocytes can promote M2 polarization in rat OA macrophages, which provides an anti-inflammatory microenvironment, thereby relieving pain and improving joint structure.131,132 Several clinical studies have shown that TG-C improves the International Knee Documentation Committee (IKDC) and Visual Analog Scale (VAS) scores in patients with chronic degenerative knee OA.126,133,134 Other sources like induced pluripotent stem cells (iPSCs) and MSCs have also been used in preclinical or clinical trials (NCT01448434, NCT01448434). Due to the chondrogenic potential, iPSCs can be an alternative if there is a shortage of autologous and allogeneic chondrocyte sources.135 MSCs are self-renewing and directionally differentiated cells, which have chondrogenic differentiation potential and can secrete inflammatory regulators for immune regulation in OA. MSCs can exert a better therapeutic effect on OA after the treatment with growth factors, inflammatory regulators and transcription factors.136 One study showed that a mixture of MSCs and chondrocytes in equal proportions promoted cartilage repair in destabilization of the medial meniscus (DMM)-induced OA compared to single cell types alone.137 Therefore, the use of genetically modified MSCs and chondrocytes together for ex vivo gene therapy may be more promising for the treatment of OA. We summarize the current gene types, vectors, expression regulation types, species, model construction methods, delivery methods and treatment effects of OA gene therapy in Table 1.
Table 1.
Genes, vectors, animal models, and outcomes in OA gene therapy.
| Genes | Vectors | Expression | Species | Model of OA | Delivery methods | Outcome | References |
|---|---|---|---|---|---|---|---|
| TFs | |||||||
| Sox9 | rAAV | Up | Sheep | Osteochondral defects | In vivo | Reduced histological signs of perifocal OA changes | 26 |
| Sox5,6,9 | Retrovirus | Up | Rat | ACLT | Ex vivo (ASCs) | Prevented the progression of degenerative changes | 138 |
| KLF2/KLF4 | AAV | Up | Mouse | DMM | In vivo | Reduced severity of OA-associated changes in cartilage, meniscus and synovium, and improved pain behaviors | 70 |
| Mohawk | Adenovirus | Up | Mouse | DMM | In vivo | Suppressed meniscus and cartilage damage, reduced OA severity | 139 |
| BATF | Adenovirus | Up | Mouse | DMM | In vivo | Caused cartilage destruction and synovial inflammation | 140 |
| GFs | |||||||
| IGF-1 | Plasmid | Up | Mouse | DMM | Ex vivo (ADSCs) | Reduced histological OARSI score and decreased loss of cartilage ECM | 141 |
| IGF-1 + bFGF | Adenovirus | Up | Rabbit | ACLT | In vivo | Enhanced chondrocyte proliferation, proteoglycan synthesis and type 2 collagen expression | 37 |
| TGF-β | Plasmid | UP | Mouse | DMM | Ex vivo (MSCs) | Enhanced the repair of damaged cartilage | 113 |
| GDF-5 | NMPs | Up | Rabbit | DMM + ACLT | In vivo | Increased the expression of ECM proteins in chondrocytes | 115 |
| VEGF | Lentivirus | Down | Rat | DMM | In vivo | Enhanced chondrogenesis and prevented OA progression | 142 |
| Inflammation-related cytokines | |||||||
| IL-1Ra | AAV | Up | Equine | OCF | In vivo | Reduced joint effusion and synovitis, and improved repair of the osteochondral lesion | 143 |
| HDAd | Up | Mouse/horse | ACLT/OCF | In vivo | Prevented development of cartilage damage, osteophytes, and synovitis. Improved lameness parameters. | 86 | |
| IL-10 | Adenovirus | Up | Mouse | CIA | Ex vivo (MSCs) | Induced long-term reduction in activated T cells | 81 |
| IL-1Ra + IL-10 | Retrovirus | Up | Rabbit | Excision of the medial collateral ligament and medial meniscectomy. | Ex vivo (synoviocytes) | Inhibition of cartilage breakdown | 144 |
| IL-4 | Plasmid | Up | Rat | ACLT | Ex vivo (MSCs) | Improved cartilage protection and pain relief function | 41 |
| ncRNAs | |||||||
| miR-140 | Exosomes | Up | Rat | DMM | In vivo | Inhibited cartilage-degrading proteases, and alleviated OA progression | 119 |
| miR-101 | Adenovirus | Down | Rat | MIA | In vivo | Inhibition of cartilage degradation | 145 |
| circRNA.33 186 | Lentivirus | Down | Mouse | DMM | In vivo | Increased type 2 collagen expression and decreased MMP-13 expression | 146 |
| circPDE4D | AAV | Up | Mouse | DMM | In vivo | Suppressed ECM catabolism and alleviated OA progression. | 125 |
| lncRNA PILA | Adenovirus | Up | Mouse | DMM | In vivo | Triggered spontaneous cartilage loss and exacerbated OA | 147 |
OA: osteoarthritis, TFs: transcription factors, KLF2/4: Krϋppel-like factor 2/4, AAV: Adeno-Associated virus, GFs: growth factors, IGF-1: Insulin-like growth factor 1, TGF-β: Transforming growth factor-β, GDF-5: Growth/differentiation factor-5, NMPs: nano-and microplastic particles, HDAd: Helper-dependent adenovirus, ACLT: anterior cruciate ligament transection ASCs: adipose stem cells, ADSCs: adipose derived stem cells, DMM: destabilization of the medial meniscus, VEGF: vascular endothelial growth factor, OCF: osteochondral fragmentation, CIA: collagenase-induced osteoarthritis, MIA: Mono-iodoacetate-induced arthritis
Gene-editing for OA
The ability to precisely edit mammalian cell genomes allows us to gain greater insight into deeper mechanisms while minimizing off-target effects. The recently developed CRISPR/Cas9 technology provides a simple and efficient option for gene editing.148,149 CRISPR/Cas consists of a single guide RNA (sgRNA), targeting ends (including base pairs complementary to the nucleic acid to be edited) and Cas proteins. The use of different target ends enables precise editing of specific genes. After a double-stranded break caused by a Cas proteolytic cleavage of a gene, the cell employs non-homologous end-joining (NHEJ) to repair the broken DNA, thus achieving a knockout. On the other hand, the introduction of exogenous DNA templates into cells can lead to the occurrence of homologous recombination repair (HDR), resulting in gene knock-in.150 An earlier report transfected human articular chondrocytes with ribonucleoprotein complexes (RNP) containing Cas9 and crisprRNA targeting exon 2 of MMP13. The results showed that the editing efficiency of MMP13 was 63%–74% and the level and activity of secreted MMP13 protein was significantly reduced.151 This suggests that gene editing within chondrocytes is feasible.
Current applications of CRISPR-Cas9 technology in OA therapy focus on reducing the inflammatory response. Cell transplantation therapy is effective in the treatment of OA. However, inflammatory factors can lead to failure of cartilage differentiation of transplanted cells. Karlsen et al. used CRISPR/Cas9 to knock out the IL-1β receptor (IL-1R1) in human chondrocytes and inserted a puromycin resistance gene to screen the cells.152 The results showed that deletion or knockdown of IL-1R1 improved cell therapy outcomes in OA patients as well as cytokine stimulation by TNF-α independent of IL-1β signaling. The implanted chondrocytes secrete a large amount of cytokines, which in turn leads to the invasion of M1 macrophages in the grafted cartilage, thus reducing the therapeutic effect. To address this situation, knockdown of the TGF-β-activated kinase 1 (TAK1) gene in chondrocytes by CRISPR-Cas9 technology significantly reduced M1 macrophage infiltration and inflammatory factor secretion in transplanted cartilage.153 CRISPR-Cas9 system is also applied to target growth factor in the treatment to OA. Nerve growth factor (NGF) is upregulated in OA and associated with the onset of pain. However, reduced NGF can also lead to bone destruction.154 Thus, gene therapy of NGF alone cannot achieve the best therapeutic effect. In this case, simultaneous deletion of NGF, MMP13 and IL-1β genes by intra-articular injection of AAV expressing CRISPR/Cas9 components alleviates pain and structural damage in a post-traumatic OA (PTOA) model.155
Although numerous scaffolds can prolong the expression of the target gene, neither the delivery method nor the delivery of any gene can achieve self-regulated, dynamic and feedback release of the target gene. The pathological changes in OA are progressive, and exogenous gene expression usually begins to decline after reaching the peak, which does not meet the subsequent regulatory requirements. Guilak's team has come up with a promising solution to this situation.156,157 One of their studies packaged a synthetic promoter consisting of NFκB recognition gene sequences (including INF-β1, IL-6, CCL2, Adamts5, and Cxcl10), along with the IL-1Ra gene, into a lentiviral vector. Thereafter they transfected lentiviruses into iPSCs and induced iPSCs into chondrocytes. Such chondrocytes respond in a time- and dose-dependent manner to IL-1α stimulation, rapidly producing therapeutic levels of IL-1Ra to counteract the effects of inflammation. This method may be broadly relevant to the creation of auto-regulatory biological systems for the delivery of gene and tissue engineering in response to a variety of disease.158 The chemokine (C-C) ligand 2 (CCl2) gene product attracts a variety of inflammatory cells, leading to increased inflammation. The activation of CCL2 depends on the continuous expression of IL-1 and TNF-α, and the expression of CCL2 gene decreases along with alleviated inflammation.159 Based on this property, Choi et al. used CRISPR-Cas9 technology to insert the IL-1Ra gene after the CCL2 promoter in iPSCs (Fig. 2).160 This synthetic gene circuit can sense changes in endogenous inflammatory cytokine levels, thereby triggering a proportional treatment response and blocking IL-1Ra production when inflammation resolves. Gene-edited iPSCs delivered to OA cartilage significantly reduced joint pain, structural damage, and systemic and local inflammation in the mouse model with inflammatory arthritis.
Figure 2.
CRIPSR/Cas9-mediated expression of IL-1Ra or sTNFR1 genes downstream of the CCL2 promoter. Introduction of CRISPR/Cas9 into iPSCs by plasmid allows insertion of IL-1Ra or sTNFR1 genes into CCL2 after the promoter. Binding of exogenous IL-1 and IL-1 receptor induces intracellular NFκB pathway activation, which in turn induces CCL2 promoter activation. Similarly, binding of exogenous TNF-α and TNFR1 activates NFκB pathway, subsequently leading the binding of transcription factor NFκB to the CCL2 promoter. Activation of the CCL2 gene leads to the translation of IL-1Ra and sTNFR1 proteins. IL-1Ra binds IL-1R1 subunit with higher affinity than IL-1α or IL-1β, but does not recruit IL-1RAcP. sTNFR1 binds TNF-α without generating downstream intracellular signaling. The gene circuit formed in this way is capable of feedback release of anti-inflammatory factors. sTNFR1: soluble tumour necrosis factor receptor-1, IL-1RAcP: IL-1 receptor accessory protein, CCL2: C-C Motif Chemokine Ligand 2.
CRISPRa system is a modified CRISPR-Cas9 system for upregulation of genes. It utilizes nuclease-inactivated Streptococcus pyogenes Cas9 (dCas9), fused to transcriptional activation domains (VP64, p65 and Rta). When paired with a guide RNA targeting a gene near the promoter region, the natural transcriptional start site of the gene is activated.161 Farhang et al. used this system to successfully upregulate the gene transcription of ACAN and COL2A1 in pellet-cultured human adipose stem cells (hADSC), which mediated the deposition of sulfated glycosaminoglycan (sGAG) and type II collagen.162 Compared to the use of growth factors to induce differentiation of stem cells into natural cell types, this approach overcomes the effect of the musculoskeletal microenvironment on growth factors in vivo and allows for more precise and controlled production of matrix proteins.
Among the factors associated with the development of OA, genetic factors account for approximately 50% of the total risk of disease in individuals. Currently, more than 100 polymorphic DNA variants have been associated with OA. Although data from OA genetics studies have not yet led directly to new treatments, a number of proteins encoded by genes associated with OA have potential for therapeutic applications.163 Point mutations or exogenous knock-in using the CRISPR-Cas9 system can better explore the impact of risk variants in joint development on OA pathogenesis. Dicks et al. explains how functionally acquired mutations in transient receptor potential vanilloid 4 (TRPV4) can lead to skeletal dysplasia of varying severity by generating CRISPR-Cas9 gene edited hiPSC cell lines with V620I or T89I TRPV4 mutations.164 Their results suggest that TRPV4 mutations alter BMP signaling in chondrocytes and prevent chondrocyte hypertrophy. Similarly, Klein et al. knocked the single nucleotide polymorphism (SNP) rs4730222 into the osteosarcoma cell line.165 The results demonstrate that rs4730222 leads to increased expression of alternative transcripts of disease-associated alleles, followed by reduced expression of typical HMG box protein 1 (HBP1), leading to the development of OA.
Existing issues and future directions
The number of clinical trials of gene therapy for OA is relatively small compared to other diseases. A search on the website clinicaltrial.gov shows that 12 clinical trials (including TissueGene-C, FX201, ICM-203, XT-150, Invossa K) for OA and degenerative arthritis gene therapy are currently enrolled, ongoing, or completed. We categorized these clinical trials in Table 2 according to intervention name, identification number, target, delivery mode, disease type, trial type, and evaluated subjects. We believe that in addition to the high cost of gene therapy itself and the obstacle of commercializing therapeutic products, there are three major issues that need to be addressed.
Table 2.
Completed, in-recruit and out-recruit clinical trials for OA gene therapy.
| Intervention | Identifier | Target | Method of delivery | Conditions | Study | Trial status | Evaluations |
|---|---|---|---|---|---|---|---|
| TissueGene-C | NCT02341378 | TGF-β1 | Retrovirus, ex vivo | Degenerative arthritis | Randomized, Multicenter, Single-blind, Phase 2A | Completed | Efficacy and safety |
| NCT00599248 | TGF-β1 | Retrovirus, ex vivo | OA | Randomized, Single-blind, Phase1 | Completed | Safety and activity | |
| NCT02341391 | TGF-β1 | Retrovirus, ex vivo | Degenerative arthritis | Randomized, Dose-escalating, Single-center, Phase 1 | Completed | Safety and efficacy | |
| NCT02072070 | TGF-β1 | Retrovirus, ex vivo | Degenerative arthritis | Randomized, Placebo Controlled, Double-blind, Multi-center Phase 3 | Completed | Efficacy and safety | |
| NCT01671072 | TGF-β1 | Retrovirus, ex vivo | Degenerative arthritis | Randomized, Placebo Controlled, Single-blind, Multi-center Phase 2B | Completed | Safety and efficacy | |
| NCT01825811 | TGF-β1 | Retrovirus, ex vivo | Degenerative arthritis | Single-blind, Randomized, Parallel-group, Multi-center Phase 2 | Completed | Efficacy and safety | |
| Invossa K | NCT03412864 | TGF-β1 | Retrovirus, ex vivo | OA | Observational | Not recruiting | Efficacy and safety |
| FX201 | NCT04119687 | IL-1Ra | HDAd, in vivo | OA | Non-Randomized, Open-Label, Single Ascending Dose, Phase 1 | Not recruiting | Safety and tolerability |
| ICM-203 | NCT04875754 | Nkx3.2 | AAV, in vivo | OA | Randomized, Double-Blind, Placebo-Controlled Single Dose Escalation, Phase 1/2a | Recruiting | Safety, tolerability, and range |
| NCT05454566 | Nkx3.2 | AAV, in vivo | OA | Randomized, Single Dose Escalation, Phase1/2 | Not recruiting | Safety, tolerability, and activity | |
| XT-150 | NCT03477487 | IL-10 | Plasmid, in vivo | OA | Non-Randomized, Ascending dose-ranging, Phase 1 | Completed | Safety, tolerability, and efficacy |
| NCT04124042 | IL-10 | Plasmid, in vivo | OA | Randomized, Double-Blind, Placebo-Controlled, Phase 2 | Completed | Efficacy and safety |
The first and most important issue is the safety of gene therapy. In some cases, viral vector activation of the immune system led to death of patients due to multiple organ failure. These cases serve as a constant reminder of the awareness and caution of the safety of gene therapy.166 Although there are no serious incidents in gene therapy for OA, the immune activation and tumorigenic side effects of viral vectors still demand a more rational use of gene therapy. The use of non-integrated and low-immunogenic rAAV or safer non-viral vectors may be a more promising direction for future research.
Second, the existing OA-related clinical trials all focus on in vivo suppression or overexpression of a single gene. However, the functions of genes are diverse and can lead to unpredictable harmful pathological processes. For example, the NF-κB family member RelA/p65 is a potent transcriptional activator of ADAMTS5 in chondrocytes, while it can also induce anti-apoptotic genes to protect chondrocytes from apoptosis during the development of OA.167 In articular cartilage, heterozygous knockout of Rela caused a considerable acceleration of OA by increasing chondrocyte death, despite reduced expression of catabolic genes.168 In this case, the use of multigene regulation instead of a single gene may be a more practical approach. Simultaneous regulation of NGF, MMP13 and IL-1β genes not only addressed the structural damage caused by deletion of NGF genes, but also alleviated the pain symptoms in OA.155 In summary, the use of multiple gene regulation can effectively exploit the therapeutic effects of genes while avoiding harmful effects.
Lastly, gene expression requires dynamic homeostasis and should be maintained within an appropriate range. Otherwise, unintended adverse effects may occur. In the treatment of OA, it is not the continuous inhibition of inflammatory factor expression or the continuous promotion of growth factor expression that provides the best therapeutic effect. Excessive TGF-β in OA can also lead to synovial fibrosis, osteophyte formation and subchondral bone changes.169 Potent overexpression of TGFβ1 in rabbit joints with experimental OA resulted in a significant reduction in cartilage ECM.170 The above-mentioned self-feedback gene regulation circuit using CRISPR/Cas9 technology has an environment-dependent feature and can automatically regulate gene expression on demand. Future clinical trials should consider incorporating this technique, which may improve treatment efficacy and mitigate negative side effects.
Conclusions
The current dilemma in OA treatment is that there is no effective way to stop the continued destruction of joint structures and no plausible method to regenerate natural joint structures in vivo. Gene therapy offers a new strategy to alleviate symptoms and promote cartilage regeneration and joint function recovery in patients with OA. Meanwhile, gene therapy can lead to more sustained, targeted, site-specific expression of therapeutic proteins in a more physiologically relevant manner than traditional approaches. Future research should focus on safer, more convenient and less expensive gene delivery vectors, delivery methods and gene editing technologies. In addition, current gene therapy does not titrate the appropriate vector dosage to mimic the in vivo expression of delivered genes, which requires further investigation in the future. Deepening the understanding of OA pathophysiology and optimizing gene therapy approaches will shed light on the treatment of OA.
Abbreviations
OA: Osteoarthritis; DMOADs: Disease-modifying osteoarthritis drugs; TILs: Tumor-infiltrating lymphocytes; FDA: Food and Drug Administration; ADA: Adenosine deaminase; IL-1: Interleukin-1; IL-1Ra: IL-1 receptor antagonist; ECM: Extracellular matrix; ACLT: Anterior cruciate ligament transection; Runx: Runt–related transcription factor; Sox: Sex-determining Region Y-related HMG-box; C/EBPβ: CCAAT/enhancer binding protein; HIF: Hypoxia-inducible factor; MMP13: Matrix metallopeptidase-13; BMP: Bone morphogenetic protein; IGF-1: Insulin-like growth factor 1; AAV: Adeno-associated virus; ADAMTS: Thrombospondin Motif; MSCs: Mesenchymal stem cells; TGFβ: Transforming growth factor-β; GDF‐5: Growth/differentiation factor-5; FGF: Fibroblast growth factor; HDAd: Helper-dependent adenovirus; HSP70: Heat shock proteins70; TG-C: TissueGene-C; iPSCs: Induced pluripotent stem cells; DMM: Destabilization of the medial meniscus; NGF: Nerve growth factor; PTOAPost-traumatic OA;CCL2: Post-traumatic OA;CCL2Chemokine (C-C) ligand 2
Authors' contributions
ZYH conceived of the work. XYL wrote the draft manuscript. LYS, ZHD, and ZYH performed critical revisions of the manuscript. XYL, LYS, ZHD, and ZYH take responsibility for the integrity of the work as a whole. All authors read and approved the final manuscript.
Acknowledgement
This work was supported by the National Natural Science Foundation of China (NSFC) to ZYH (Grants No. 92049101, 81972097, and 81702185). This research was also supported by Sichuan Science and Technology Program to ZYH (Grant No. 2022YFH0101). This work was also supported by Guangdong Basic and Applied Basic Research Foundation (Grant No. 2021A1515220030), Shenzhen Science and Technology Program (Grant No. RCYX20210609103902019). We would like to thank Biorender company for their schematic diagram in this review.
Contributor Information
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.
Zhenghan Deng, Department of Sports Medicine, the First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen 518035, China.
Zeyu Huang, Department of Orthopaedic Surgery, West China Hospital, West China Medical School, Sichuan University, Chengdu 610041, China.
Conflict of interest
The authors declare that they have no conflict of interest.
References
- 1. Hunter DJ, Bierma-Zeinstra S. Osteoarthritis. Lancet. 2019;393:1745–59.. doi: 10.1016/S0140-6736(19)30417-9. [DOI] [PubMed] [Google Scholar]
- 2. Quicke JG, Conaghan PG, Corp Net al. Osteoarthritis year in review 2021: Epidemiology & therapy. Osteoarthritis Cartilage. 2022;30:196–206.. doi: 10.1016/j.joca.2021.10.003. [DOI] [PubMed] [Google Scholar]
- 3. Chen D, Shen J, Zhao Wet al. Osteoarthritis: Toward a comprehensive understanding of pathological mechanism. Bone Res. 2017;5:16044. doi: 10.1038/boneres.2016.44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Abramoff B, Caldera FE.. Osteoarthritis: Pathology, diagnosis, and treatment options. Med Clin North Am. 2020;104:293–311.. doi: 10.1016/j.mcna.2019.10.007. [DOI] [PubMed] [Google Scholar]
- 5. Yao Q, Wu X, Tao Cet al. Osteoarthritis: Pathogenic signaling pathways and therapeutic targets. Signal Transduct Target Ther. 2023;8:56. doi: 10.1038/s41392-023-01330-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Katz JN, Arant KR, Loeser RF.. Diagnosis and treatment of hip and knee osteoarthritis: A review. JAMA. 2021;325:568–78.. doi: 10.1001/jama.2020.22171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Richard MJ, Driban JB, McAlindon TE.. Pharmaceutical treatment of osteoarthritis. Osteoarthritis Cartilage. 2023;31:458–66.. doi: 10.1016/j.joca.2022.11.005. [DOI] [PubMed] [Google Scholar]
- 8. Oo WM, Yu SPC, Daniel MSet al. Disease-modifying drugs in osteoarthritis: Current understanding and future therapeutics. Expert Opin Emerg Drugs. 2018;23:331–47.. doi: 10.1080/14728214.2018.1547706. [DOI] [PubMed] [Google Scholar]
- 9. Dunbar CE, High KA, Joung JKet al. Gene therapy comes of age. Science. 2018;359:eaan4672. doi: 10.1126/science.aan4672. [DOI] [PubMed] [Google Scholar]
- 10. Chen T, Weng W, Liu Yet al. Update on novel non-operative treatment for osteoarthritis: Current status and future trends. Front Pharmacol. 2021;12:755230. doi: 10.3389/fphar.2021.755230. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Wirth T, Parker N, Ylä-Herttuala S.. History of gene therapy. Gene. 2013;525:162–9.. doi: 10.1016/j.gene.2013.03.137. [DOI] [PubMed] [Google Scholar]
- 12. Rosenberg SA, Packard BS, Aebersold PMet al. Use of tumor-infiltrating lymphocytes and interleukin-2 in the immunotherapy of patients with metastatic melanoma. A preliminary report. N Engl J Med. 1988;319:1676–80.. doi: 10.1056/NEJM198812223192527. [DOI] [PubMed] [Google Scholar]
- 13. Rosenberg SA, Aebersold P, Cornetta Ket al. Gene transfer into humans–immunotherapy of patients with advanced melanoma, using tumor-infiltrating lymphocytes modified by retroviral gene transduction. N Engl J Med. 1990;323:570–8.. doi: 10.1056/NEJM199008303230904. [DOI] [PubMed] [Google Scholar]
- 14. Blaese RM, Culver KW, Miller ADet al. T lymphocyte-directed gene therapy for ADA- SCID: Initial trial results after 4 years. Science. 1995;270:475–80.. doi: 10.1126/science.270.5235.475. [DOI] [PubMed] [Google Scholar]
- 15. Sanchez-Lopez E, Coras R, Torres Aet al. Synovial inflammation in osteoarthritis progression. Nat Rev Rheumatol. 2022;18:258–75.. doi: 10.1038/s41584-022-00749-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Kapoor M, Martel-Pelletier J, Lajeunesse Det al. Role of proinflammatory cytokines in the pathophysiology of osteoarthritis. Nat Rev Rheumatol. 2011;7:33–42.. doi: 10.1038/nrrheum.2010.196. [DOI] [PubMed] [Google Scholar]
- 17. Arend WP, Welgus HG, Thompson RCet al. Biological properties of recombinant human monocyte-derived interleukin 1 receptor antagonist. J Clin Invest. 1990;85:1694–7.. doi: 10.1172/JCI114622. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Bandara G, Mueller GM, Galea-Lauri Jet al. Intraarticular expression of biologically active interleukin 1-receptor-antagonist protein by ex vivo gene transfer. Proc Natl Acad Sci U S A. 1993;90:10764–8.. doi: 10.1073/pnas.90.22.10764. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Baragi VM, Renkiewicz RR, Jordan Het al. Transplantation of transduced chondrocytes protects articular cartilage from interleukin 1-induced extracellular matrix degradation. J Clin Invest. 1995;96:2454–60.. doi: 10.1172/JCI118303. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Pelletier JP, Caron JP, Evans Cet al. In vivo suppression of early experimental osteoarthritis by interleukin-1 receptor antagonist using gene therapy. Arthritis Rheum. 1997;40:1012–9.. doi: 10.1002/art.1780400604. [DOI] [PubMed] [Google Scholar]
- 21. Nishimura R, Hata K, Takahata Yet al. Role of signal transduction pathways and transcription factors in cartilage and joint diseases. Int J Mol Sci. 2020;21:1340. doi: 10.3390/ijms21041340. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Catheline SE, Hoak D, Chang Met al. Chondrocyte-specific RUNX2 overexpression accelerates post-traumatic osteoarthritis progression in adult mice. J Bone Miner Res. 2019;34:1676–89.. doi: 10.1002/jbmr.3737. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Yoshida CA, Yamamoto H, Fujita Tet al. Runx2 and Runx3 are essential for chondrocyte maturation, and Runx2 regulates limb growth through induction of Indian hedgehog. Genes Dev. 2004;18:952–63.. doi: 10.1101/gad.1174704. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Aini H, Itaka K, Fujisawa Aet al. Messenger RNA delivery of a cartilage-anabolic transcription factor as a disease-modifying strategy for osteoarthritis treatment. Sci Rep. 2016;6:18743. doi: 10.1038/srep18743. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Hirata M, Kugimiya F, Fukai Aet al. C/EBPβ and RUNX2 cooperate to degrade cartilage with MMP-13 as the target and HIF-2α as the inducer in chondrocytes. Hum Mol Genet. 2012;21:1111–23.. doi: 10.1093/hmg/ddr540. [DOI] [PubMed] [Google Scholar]
- 26. Lange C, Madry H, Venkatesan JKet al. rAAV-mediated overexpression improves the repair of osteochondral defects in a clinically relevant large animal model over time in vivo and reduces perifocal osteoarthritic changes. Am J Sports Med. 2021;49:3696–707.. doi: 10.1177/03635465211049414. [DOI] [PubMed] [Google Scholar]
- 27. Zhang F-J, Luo W, Lei G-H.. Role of HIF-1α and HIF-2α in osteoarthritis. Joint Bone Spine. 2015;82:144–7.. doi: 10.1016/j.jbspin.2014.10.003 [DOI] [PubMed] [Google Scholar]
- 28. Oh H, Kwak JS, Yang Set al. Reciprocal regulation by hypoxia-inducible factor-2α and the NAMPT-NAD(+)-SIRT axis in articular chondrocytes is involved in osteoarthritis. Osteoarthritis Cartilage. 2015;23:2288–96.. doi: 10.1016/j.joca.2015.07.009. [DOI] [PubMed] [Google Scholar]
- 29. Wen C, Xu L, Xu Xet al. Insulin-like growth factor-1 in articular cartilage repair for osteoarthritis treatment. Arthritis Res Ther. 2021;23:277. doi: 10.1186/s13075-021-02662-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Ortved KF, Begum L, Mohammed HOet al. Implantation of rAAV5-IGF-I transduced autologous chondrocytes improves cartilage repair in full-thickness defects in the equine model. Mol Ther. 2015;23:363–73.. doi: 10.1038/mt.2014.198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Baugé C, Girard N, Lhuissier Eet al. Regulation and role of tgfβ signaling pathway in aging and osteoarthritis joints. Aging Dis. 2014;5:394–405.. doi: 10.14336/AD.2014.0500394. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Blaney Davidson EN, van der Kraan PM, van den Berg WB. TGF-beta and osteoarthritis. Osteoarthritis Cartilage. 2007;15:597–604.. doi: 10.1016/j.joca.2007.02.005. [DOI] [PubMed] [Google Scholar]
- 33. Steinert AF, Proffen B, Kunz Met al. Hypertrophy is induced during the in vitro chondrogenic differentiation of human mesenchymal stem cells by bone morphogenetic protein-2 and bone morphogenetic protein-4 gene transfer. Arthritis Res Ther. 2009;11:R148. doi: 10.1186/ar2822. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Sun K, Guo J, Yao Xet al. Growth differentiation factor 5 in cartilage and osteoarthritis: A possible therapeutic candidate. Cell Prolif. 2021;54:e12998. doi: 10.1111/cpr.12998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Chia S-L, Sawaji Y, Burleigh Aet al. Fibroblast growth factor 2 is an intrinsic chondroprotective agent that suppresses ADAMTS-5 and delays cartilage degradation in murine osteoarthritis. Arthritis Rheum. 2009;60:2019–27.. doi: 10.1002/art.24654. [DOI] [PubMed] [Google Scholar]
- 36. Nummenmaa E, Hämäläinen M, Moilanen Tet al. Effects of FGF-2 and FGF receptor antagonists on MMP enzymes, aggrecan, and type II collagen in primary human OA chondrocytes. Scand J Rheumatol. 2015;44:321–30.. doi: 10.3109/03009742.2014.1000372. [DOI] [PubMed] [Google Scholar]
- 37. Chen B, Qin J, Wang Het al. Effects of adenovirus-mediated bFGF, IL-1Ra and IGF-1 gene transfer on human osteoarthritic chondrocytes and osteoarthritis in rabbits. Exp Mol Med. 2010;42:684–95.. doi: 10.3858/emm.2010.42.10.067. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Molnar V, Matišić V, Kodvanj Iet al. Cytokines and chemokines involved in osteoarthritis pathogenesis. Int J Mol Sci. 2021;22:9208. doi: 10.3390/ijms22179208. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. van Meegeren MER, Roosendaal G, Jansen NWDet al. IL-4 alone and in combination with IL-10 protects against blood-induced cartilage damage. Osteoarthritis Cartilage. 2012;20:764–72.. doi: 10.1016/j.joca.2012.04.002. [DOI] [PubMed] [Google Scholar]
- 40. Bellavia D, Veronesi F, Carina Vet al. Gene therapy for chondral and osteochondral regeneration: Is the future now?. Cell Mol Life Sci. 2018;75:649–67.. doi: 10.1007/s00018-017-2637-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Song SY, Hong J, Go Set al. Interleukin-4 gene transfection and spheroid formation potentiate therapeutic efficacy of mesenchymal stem cells for osteoarthritis. Adv Healthc Mater. 2020;9:e1901612. doi: 10.1002/adhm.201901612. [DOI] [PubMed] [Google Scholar]
- 42. Moss KL, Jiang Z, Dodson MEet al. Sustained interleukin-10 transgene expression following intra-articular AAV5-IL-10 administration to horses. Hum Gene Ther. 2020;31:110–8.. doi: 10.1089/hum.2019.195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Knedla A, Riepl B, Lefèvre Set al. The therapeutic use of osmotic minipumps in the severe combined immunodeficiency (SCID) mouse model for rheumatoid arthritis. Ann Rheum Dis. 2009;68:124–9.. doi: 10.1136/ard.2007.086116. [DOI] [PubMed] [Google Scholar]
- 44. Ali SA, Peffers MJ, Ormseth MJet al. The non-coding RNA interactome in joint health and disease. Nat Rev Rheumatol. 2021;17:692–705.. doi: 10.1038/s41584-021-00687-y. [DOI] [PubMed] [Google Scholar]
- 45. Lei J, Fu Y, Zhuang Yet al. miR-382-3p suppressed IL-1β induced inflammatory response of chondrocytes via the TLR4/MyD88/NF-κb signaling pathway by directly targeting CX43. J Cell Physiol. 2019;234:23160–8.. doi: 10.1002/jcp.28882. [DOI] [PubMed] [Google Scholar]
- 46. Lei J, Fu Y, Zhuang Yet al. LncRNA SNHG1 alleviates IL-1β-induced osteoarthritis by inhibiting miR-16-5p-mediated p38 MAPK and NF-κb signaling pathways. Biosci Rep. 2019;39:BSR20191523. doi: 10.1042/BSR20191523. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Wen X, Li H, Sun Het al. MiR-455-3p reduces apoptosis and alleviates degeneration of chondrocyte through regulating PI3K/AKT pathway. Life Sci. 2020;253:117718. doi: 10.1016/j.lfs.2020.117718. [DOI] [PubMed] [Google Scholar]
- 48. He W, Cheng Y.. Inhibition of miR-20 promotes proliferation and autophagy in articular chondrocytes by PI3K/AKT/mTOR signaling pathway. Biomed Pharmacother. 2018;97:607–15.. doi: 10.1016/j.biopha.2017.10.152. [DOI] [PubMed] [Google Scholar]
- 49. Zhang W, Zhang C, Hu Cet al. Circular RNA-CDR1as acts as the sponge of microRNA-641 to promote osteoarthritis progression. J Inflamm (Lond). 2020;17:8. doi: 10.1186/s12950-020-0234-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Bai ZM, Kang MM, Zhou XFet al. CircTMBIM6 promotes osteoarthritis-induced chondrocyte extracellular matrix degradation via miR-27a/MMP13 axis. Eur Rev Med Pharmacol Sci. 2020;24:7927–36.. doi: 10.26355/eurrev_202008_22475. [DOI] [PubMed] [Google Scholar]
- 51. Du M, Fan S, Liu Yet al. The application of circRNA-016901 in improving the diagnostic accuracy of osteoarthritis. Biomed Res Int. 2022;2022:1158562. doi: 10.1155/2022/1158562. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Wang Y, Wu C, Yang Yet al. Preliminary exploration of hsa_circ_0032131 levels in peripheral blood as a potential diagnostic biomarker of osteoarthritis. Genet Test Mol Biomarkers. 2019;23:717–21.. doi: 10.1089/gtmb.2019.0036. [DOI] [PubMed] [Google Scholar]
- 53. Xiao H, Feng X, Liu Met al. SnoRNA and lncSNHG: Advances of nucleolar small RNA host gene transcripts in anti-tumor immunity. Front Immunol. 2023;14:1143980. doi: 10.3389/fimmu.2023.1143980. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Steinbusch MMF, Fang Y, Milner PIet al. Serum snoRNAs as biomarkers for joint ageing and post traumatic osteoarthritis. Sci Rep. 2017;7:43558. doi: 10.1038/srep43558. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Peffers MJ, Chabronova A, Balaskas Pet al. SnoRNA signatures in cartilage ageing and osteoarthritis. Sci Rep. 2020;10:10641. doi: 10.1038/s41598-020-67446-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Zavvar M, Assadiasl S, Soleimanifar Net al. Gene therapy in rheumatoid arthritis: Strategies to select therapeutic genes. J Cell Physiol. 2019;234:16913–24.. doi: 10.1002/jcp.28392. [DOI] [PubMed] [Google Scholar]
- 57. Naso MF, Tomkowicz B, Perry WLet al. Adeno-associated virus (AAV) as a vector for gene therapy. BioDrugs. 2017;31:317–34.. doi: 10.1007/s40259-017-0234-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Mingozzi F, High KA. Immune responses to AAV vectors: Overcoming barriers to successful gene therapy. Blood. 2013;122:23–36.. doi: 10.1182/blood-2013-01-306647. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Pena SA, Iyengar R, Eshraghi RSet al. Gene therapy for neurological disorders: Challenges and recent advancements. J Drug Target. 2020;28:111–28.. doi: 10.1080/1061186X.2019.1630415. [DOI] [PubMed] [Google Scholar]
- 60. Gruntman AM, Flotte TR.. The rapidly evolving state of gene therapy. FASEB J. 2018;32:1733–40.. doi: 10.1096/fj.201700982R. [DOI] [PubMed] [Google Scholar]
- 61. Ueblacker P, Wagner B, Vogt Set al. In vivo analysis of retroviral gene transfer to chondrocytes within collagen scaffolds for the treatment of osteochondral defects. Biomaterials. 2007;28:4480–7.. doi: 10.1016/j.biomaterials.2007.06.027. [DOI] [PubMed] [Google Scholar]
- 62. Morgan MA, Galla M, Grez Met al. Retroviral gene therapy in Germany with a view on previous experience and future perspectives. Gene Ther. 2021;28:494–512.. doi: 10.1038/s41434-021-00237-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Meyerrose TE, Roberts M, Ohlemiller KKet al. Lentiviral-transduced human mesenchymal stem cells persistently express therapeutic levels of enzyme in a xenotransplantation model of human disease. Stem Cells. 2008;26:1713–22.. doi: 10.1634/stemcells.2008-0008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Gouze E, Pawliuk R, Pilapil Cet al. In vivo gene delivery to synovium by lentiviral vectors. Mol Ther. 2002;5:397–404.. doi: 10.1006/mthe.2002.0562. [DOI] [PubMed] [Google Scholar]
- 65. Cesana D, Ranzani M, Volpin Met al. Uncovering and dissecting the genotoxicity of self-inactivating lentiviral vectors in vivo. Mol Ther. 2014;22:774–85.. doi: 10.1038/mt.2014.3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Schambach A, Zychlinski D, Ehrnstroem Bet al. Biosafety features of lentiviral vectors. Hum Gene Ther. 2013;24:132–42.. doi: 10.1089/hum.2012.229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Crystal RG. Adenovirus: The first effective in vivo gene delivery vector. Hum Gene Ther. 2014;25:3–11.. doi: 10.1089/hum.2013.2527. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Watson-Levings RS, Palmer GD, Levings PPet al. Gene therapy in orthopaedics: Progress and challenges in pre-clinical development and translation. Front Bioeng Biotechnol. 2022;10:901317. doi: 10.3389/fbioe.2022.901317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Scharstuhl A, Vitters EL, van der Kraan PMet al. Reduction of osteophyte formation and synovial thickening by adenoviral overexpression of transforming growth factor beta/bone morphogenetic protein inhibitors during experimental osteoarthritis. Arthritis Rheum. 2003;48:3442–51.. doi: 10.1002/art.11328. [DOI] [PubMed] [Google Scholar]
- 70. Kawata M, Teramura T, Ordoukhanian Pet al. Krüppel-like factor-4 and Krüppel-like factor-2 are important regulators of joint tissue cells and protect against tissue destruction and inflammation in osteoarthritis. Ann Rheum Dis. 2022;81:1179–88.. doi: 10.1136/annrheumdis-2021-221867. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Gao X, Jiang S, Du Zet al. KLF2 protects against osteoarthritis by repressing oxidative response through activation of Nrf2/ARE signaling in vitro and in vivo. Oxid Med Cell Longev. 2019;2019:8564681. doi: 10.1155/2019/8564681. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Yang S, Kim J, Ryu J-Het al. Hypoxia-inducible factor-2alpha is a catabolic regulator of osteoarthritic cartilage destruction. Nat Med. 2010;16:687–93.. doi: 10.1038/nm.2153. [DOI] [PubMed] [Google Scholar]
- 73. Ryu JH, Shin Y, Huh YHet al. Hypoxia-inducible factor-2α regulates Fas-mediated chondrocyte apoptosis during osteoarthritic cartilage destruction. Cell Death Differ. 2012;19:440–50.. doi: 10.1038/cdd.2011.111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Bondeson J, Lauder S, Wainwright Set al. Adenoviral gene transfer of the endogenous inhibitor IkappaBalpha into human osteoarthritis synovial fibroblasts demonstrates that several matrix metalloproteinases and aggrecanases are nuclear factor-kappaB-dependent. J Rheumatol. 2007;34:523–33. [PubMed] [Google Scholar]
- 75. Takahata Y, Nakamura E, Hata Ket al. Sox4 is involved in osteoarthritic cartilage deterioration through induction of ADAMTS4 and ADAMTS5. FASEB J. 2019;33:619–30.. doi: 10.1096/fj.201800259R. [DOI] [PubMed] [Google Scholar]
- 76. Dong Y, Wang P, Yang Yet al. PRMT5 inhibition attenuates cartilage degradation by reducing MAPK and NF-κb signaling. Arthritis Res Ther. 2020;22:201. doi: 10.1186/s13075-020-02304-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Jeon J, Noh H-J, Lee Het al. TRIM24-RIP3 axis perturbation accelerates osteoarthritis pathogenesis. Ann Rheum Dis. 2020;79:1635–43.. doi: 10.1136/annrheumdis-2020-217904. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Choi W-S, Yang J-I, Kim Wet al. Critical role for arginase II in osteoarthritis pathogenesis. Ann Rheum Dis. 2019;78:421–8.. doi: 10.1136/annrheumdis-2018-214282. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Yang J-I, Chun J-S. Upregulated endonuclease Regnase-1 suppresses osteoarthritis by forming a negative feedback loop of catabolic signaling in chondrocytes. Arthritis Res Ther. 2021;23:114. doi: 10.1186/s13075-021-02485-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Ishizuka S, Tsuchiya S, Ohashi Yet al. Hyaluronan synthase 2 (HAS2) overexpression diminishes the procatabolic activity of chondrocytes by a mechanism independent of extracellular hyaluronan. J Biol Chem. 2019;294:13562–79.. doi: 10.1074/jbc.RA119.008567. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Farrell E, Fahy N, Ryan AEet al. vIL-10-overexpressing human mscs modulate naïve and activated T lymphocytes following induction of collagenase-induced osteoarthritis. Stem Cell Res Ther. 2016;7:74. doi: 10.1186/s13287-016-0331-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. van Geffen EW, van Caam APM, van Beuningen HMet al. IL37 dampens the IL1β-induced catabolic status of human OA chondrocytes. Rheumatology (Oxford). 2017;56:351–61.. doi: 10.1093/rheumatology/kew411. [DOI] [PubMed] [Google Scholar]
- 83. Parks RJ, Chen L, Anton Met al. A helper-dependent adenovirus vector system: Removal of helper virus by Cre-mediated excision of the viral packaging signal. Proc Natl Acad Sci U S A. 1996;93:13565–70.. doi: 10.1073/pnas.93.24.13565. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Ruan MZC, Erez A, Guse Ket al. Proteoglycan 4 expression protects against the development of osteoarthritis. Sci Transl Med. 2013;5:176ra34. doi: 10.1126/scitranslmed.3005409. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Senter R, Boyce R, Repic Met al. Efficacy and safety of FX201, a novel intra-articular IL-1Ra gene therapy for osteoarthritis treatment, in a rat model. Hum Gene Ther. 2022;33:541–9.. doi: 10.1089/hum.2021.131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Nixon AJ, Goodrich LR, Scimeca MSet al. Gene therapy in musculoskeletal repair. Ann N Y Acad Sci. 2007;1117:310–27.. doi: 10.1196/annals.1402.065. [DOI] [PubMed] [Google Scholar]
- 87. Chen X, Gonçalves MAFV.. Engineered viruses as genome editing devices. Mol Ther. 2016;24:447–57.. doi: 10.1038/mt.2015.164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Yoon DS, Lee K-M, Cho Set al. Cellular and tissue selectivity of aav serotypes for gene delivery to chondrocytes and cartilage. Int J Med Sci. 2021;18:3353–60.. doi: 10.7150/ijms.56760. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Wang D, Tai PWL, Gao G.. Adeno-associated virus vector as a platform for gene therapy delivery. Nat Rev Drug Discov. 2019;18:358–78.. doi: 10.1038/s41573-019-0012-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Verdera HC, Kuranda K, Mingozzi F.. AAV vector immunogenicity in humans: A long journey to successful gene transfer. Mol Ther. 2020;28:723–46.. doi: 10.1016/j.ymthe.2019.12.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. McCarty DM. Self-complementary AAV vectors; advances and applications. Mol Ther. 2008;16:1648–56.. doi: 10.1038/mt.2008.171. [DOI] [PubMed] [Google Scholar]
- 92. Kay JD, Gouze E, Oligino TJet al. Intra-articular gene delivery and expression of interleukin-1Ra mediated by self-complementary adeno-associated virus. J Gene Med. 2009;11:605–14.. doi: 10.1002/jgm.1334. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Rey-Rico A, Venkatesan JK, Schmitt Get al. Effective remodelling of human osteoarthritic cartilage by sox9 gene transfer and overexpression upon delivery of rAAV vectors in polymeric micelles. Mol Pharm. 2018;15:2816–26.. doi: 10.1021/acs.molpharmaceut.8b00331. [DOI] [PubMed] [Google Scholar]
- 94. Weimer A, Madry H, Venkatesan JKet al. Benefits of recombinant adeno-associated virus (rAAV)-mediated insulinlike growth factor I (IGF-I) overexpression for the long-term reconstruction of human osteoarthritic cartilage by modulation of the IGF-I axis. Mol Med. 2012;18:346–58.. doi: 10.2119/molmed.2011.00371. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Hemphill DD, McIlwraith CW, Slayden RAet al. Adeno-associated virus gene therapy vector scAAVIGF-I for transduction of equine articular chondrocytes and RNA-seq analysis. Osteoarthritis Cartilage. 2016;24:902–11.. doi: 10.1016/j.joca.2015.12.001. [DOI] [PubMed] [Google Scholar]
- 96. Watson Levings RS, Broome TA, Smith ADet al. Gene therapy for osteoarthritis: Pharmacokinetics of intra-articular self-complementary adeno-associated virus interleukin-1 receptor antagonist delivery in an equine model. Hum Gene Ther Clin Dev. 2018;29:90–100.. doi: 10.1089/humc.2017.142. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Kyostio-Moore S, Berthelette P, Cornell CSet al. Hyaluronic acid synthase-2 gene transfer into the joints of Beagles by use of recombinant adeno-associated viral vectors. Am J Vet Res. 2018;79:505–17.. doi: 10.2460/ajvr.79.5.505. [DOI] [PubMed] [Google Scholar]
- 98. Morscheid YP, Venkatesan JK, Schmitt Get al. rAAV-mediated human FGF-2 gene therapy enhances osteochondral repair in a clinically relevant large animal model over time in vivo. Am J Sports Med. 2021;49:958–69.. doi: 10.1177/0363546521988941. [DOI] [PubMed] [Google Scholar]
- 99. Maihöfer J, Madry H, Rey-Rico Aet al. Hydrogel-guided, rAAV-mediated IGF-I overexpression enables long-term cartilage repair and protection against perifocal osteoarthritis in a large-animal full-thickness chondral defect model at one year in vivo. Adv Mater. 2021;33:e2008451. doi: 10.1002/adma.202008451. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Cucchiarini M, Asen A-K, Goebel Let al. Effects of TGF-β overexpression via rAAV gene transfer on the early repair processes in an osteochondral defect model in minipigs. Am J Sports Med. 2018;46:1987–96.. doi: 10.1177/0363546518773709. [DOI] [PubMed] [Google Scholar]
- 101. Ruan MZ, Cerullo V, Cela Ret al. Treatment of osteoarthritis using a helper-dependent adenoviral vector retargeted to chondrocytes. Mol Ther Methods Clin Dev. 2016;3:16008. doi: 10.1038/mtm.2016.8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. Martinez-Redondo P, Guillen-Guillen I, Davidsohn Net al. αKLOTHO and sTGFβR2 treatment counteract the osteoarthritic phenotype developed in a rat model. Protein Cell. 2020;11:219–26.. doi: 10.1007/s13238-019-00685-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Lee HH, O'Malley MJ, Friel NAet al. Persistence, localization, and external control of transgene expression after single injection of adeno-associated virus into injured joints. Hum Gene Ther. 2013;24:457–66.. doi: 10.1089/hum.2012.118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Patil S, Gao Y-G, Lin Xet al. The development of functional non-viral vectors for gene delivery. Int J Mol Sci. 2019;20:5491. doi: 10.3390/ijms20215491. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105. Gantenbein B, Tang S, Guerrero Jet al. Non-viral gene delivery methods for bone and joints. Front Bioeng Biotechnol. 2020;8:598466. doi: 10.3389/fbioe.2020.598466. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Grossin L, Cournil-Henrionnet C, Mir LMet al. Direct gene transfer into rat articular cartilage by in vivo electroporation. FASEB J. 2003;17:829–35.. doi: 10.1096/fj.02-0518com. [DOI] [PubMed] [Google Scholar]
- 107. Ohashi S, Kubo T, Kishida Tet al. Successful genetic transduction in vivo into synovium by means of electroporation. Biochem Biophys Res Commun. 2002;293:1530–5.. doi: 10.1016/S0006-291X(02)00386-8. [DOI] [PubMed] [Google Scholar]
- 108. Grossin L, Cournil-Henrionnet C, Pinzano Aet al. Gene transfer with HSP 70 in rat chondrocytes confers cytoprotection in vitro and during experimental osteoarthritis. FASEB J. 2006;20:65–75.. doi: 10.1096/fj.04-2889com. [DOI] [PubMed] [Google Scholar]
- 109. Khoury M, Bigey P, Louis-Plence Pet al. A comparative study on intra-articular versus systemic gene electrotransfer in experimental arthritis. J Gene Med. 2006;8:1027–36.. doi: 10.1002/jgm.922. [DOI] [PubMed] [Google Scholar]
- 110. Caplen NJ, Alton EW, Middleton PGet al. Liposome-mediated CFTR gene transfer to the nasal epithelium of patients with cystic fibrosis. Nat Med. 1995;1:39–46.. doi: 10.1038/nm0195-39. [DOI] [PubMed] [Google Scholar]
- 111. Stern M, Sorgi F, Hughes Cet al. The effects of jet nebulisation on cationic liposome-mediated gene transfer in vitro. Gene Ther. 1998;5:583–93.. doi: 10.1038/sj.gt.3300629. [DOI] [PubMed] [Google Scholar]
- 112. Madry H, Trippel SB.. Efficient lipid-mediated gene transfer to articular chondrocytes. Gene Ther. 2000;7:286–91.. doi: 10.1038/sj.gt.3301086. [DOI] [PubMed] [Google Scholar]
- 113. Cai Y, Wu C, Ou Qet al. Enhanced osteoarthritis therapy by nanoengineered mesenchymal stem cells using biomimetic CuS nanoparticles loaded with plasmid DNA encoding TGF-β1. Bioact Mater. 2023;19:444–57.. doi: 10.1016/j.bioactmat.2022.04.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Lu H-D, Zhao H-Q, Wang Ket al. Novel hyaluronic acid-chitosan nanoparticles as non-viral gene delivery vectors targeting osteoarthritis. Int J Pharm. 2011;420:358–65.. doi: 10.1016/j.ijpharm.2011.08.046. [DOI] [PubMed] [Google Scholar]
- 115. Chen Z, Deng S, Yuan D-Cet al. Novel nano-microspheres containing chitosan, hyaluronic acid, and chondroitin sulfate deliver growth and differentiation factor-5 plasmid for osteoarthritis gene therapy. J Zhejiang Univ Sci B. 2018;19:910–23.. doi: 10.1631/jzus.B1800095. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. Wu X, Wang Y, Xiao Yet al. Extracellular vesicles: Potential role in osteoarthritis regenerative medicine. J Orthop Translat. 2020;21:73–80.. doi: 10.1016/j.jot.2019.10.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117. Gallego-Perez D, Pal D, Ghatak Set al. Topical tissue nano-transfection mediates non-viral stroma reprogramming and rescue. Nat Nanotechnol. 2017;12:974–9.. doi: 10.1038/nnano.2017.134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. Zhou Q, Cai Y, Jiang Yet al. Exosomes in osteoarthritis and cartilage injury: Advanced development and potential therapeutic strategies. Int J Biol Sci. 2020;16:1811–20.. doi: 10.7150/ijbs.41637. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Liang Y, Xu X, Li Xet al. Chondrocyte-targeted microRNA delivery by engineered exosomes toward a cell-free osteoarthritis therapy. ACS Appl Mater Interfaces. 2020;12:36938–47.. doi: 10.1021/acsami.0c10458. [DOI] [PubMed] [Google Scholar]
- 120. Liang Y, Xu X, Xu Let al. Chondrocyte-specific genomic editing enabled by hybrid exosomes for osteoarthritis treatment. Theranostics. 2022;12:4866–78.. doi: 10.7150/thno.69368. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121. Liu Y, Zeng Y, Si H-Bet al. Exosomes derived from human urine-derived stem cells overexpressing miR-140-5p alleviate knee osteoarthritis through downregulation of VEGFA in a rat model. Am J Sports Med. 2022;50:1088–105.. doi: 10.1177/03635465221073991. [DOI] [PubMed] [Google Scholar]
- 122. Evans CH, Ghivizzani SC, Robbins PD.. Gene delivery to joints by intra-articular injection. Hum Gene Ther. 2018;29:2–14.. doi: 10.1089/hum.2017.181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123. Goodrich LR, Grieger JC, Phillips JNet al. scAAVIL-1ra dosing trial in a large animal model and validation of long-term expression with repeat administration for osteoarthritis therapy. Gene Ther. 2015;22:536–45.. doi: 10.1038/gt.2015.21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124. Izal I, Acosta CA, Ripalda Pet al. IGF-1 gene therapy to protect articular cartilage in a rat model of joint damage. Arch Orthop Trauma Surg. 2008;128:239–47.. doi: 10.1007/s00402-007-0407-7. [DOI] [PubMed] [Google Scholar]
- 125. Wu Y, Hong Z, Xu Wet al. Circular RNA circPDE4D protects against osteoarthritis by binding to miR-103a-3p and regulating FGF18. Mol Ther. 2021;29:308–23.. doi: 10.1016/j.ymthe.2020.09.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. Cherian JJ, Parvizi J, Bramlet Det al. Preliminary results of a phase II randomized study to determine the efficacy and safety of genetically engineered allogeneic human chondrocytes expressing TGF-β1 in patients with grade 3 chronic degenerative joint disease of the knee. Osteoarthritis Cartilage. 2015;23:2109–18.. doi: 10.1016/j.joca.2015.06.019. [DOI] [PubMed] [Google Scholar]
- 127. Ha C-W, Noh MJ, Choi KBet al. Initial phase I safety of retrovirally transduced human chondrocytes expressing transforming growth factor-beta-1 in degenerative arthritis patients. Cytotherapy. 2012;14:247–56.. doi: 10.3109/14653249.2011.629645. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128. Geng Y, Chen J, Alahdal Met al. Intra-articular injection of hUC-mscs expressing miR-140-5p induces cartilage self-repairing in the rat osteoarthritis. J Bone Miner Metab. 2020;38:277–88.. doi: 10.1007/s00774-019-01055-3. [DOI] [PubMed] [Google Scholar]
- 129. Lee S-Y, Lee SH, Na HSet al. The therapeutic effect of STAT3 signaling-suppressed MSC on pain and articular cartilage damage in a rat model of monosodium iodoacetate-induced osteoarthritis. Front Immunol. 2018;9:2881. doi: 10.3389/fimmu.2018.02881. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130. Li K-C, Hu Y-C.. Cartilage tissue engineering: Recent advances and perspectives from gene regulation/therapy. Adv Healthc Mater. 2015;4:948–68.. doi: 10.1002/adhm.201400773. [DOI] [PubMed] [Google Scholar]
- 131. Lee H, Kim H, Seo Jet al. TissueGene-C promotes an anti-inflammatory micro-environment in a rat monoiodoacetate model of osteoarthritis via polarization of M2 macrophages leading to pain relief and structural improvement. Inflammopharmacology. 2020;28:1237–52.. doi: 10.1007/s10787-020-00738-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132. Lee B, Parvizi J, Bramlet Det al. Results of a phase II study to determine the efficacy and safety of genetically engineered allogeneic human chondrocytes expressing TGF-β1. J Knee Surg. 2020;33:167–72.. doi: 10.1055/s-0038-1676803. [DOI] [PubMed] [Google Scholar]
- 133. Lee MC, Ha CW, Elmallah RKet al. A placebo-controlled randomised trial to assess the effect of TGF-ß1-expressing chondrocytes in patients with arthritis of the knee. Bone Joint J. 2015;97-B:924–32.. doi: 10.1302/0301-620X.97B7.35852. [DOI] [PubMed] [Google Scholar]
- 134. Ha C-W, Cho JJ, Elmallah RKet al. A multicenter, single-blind, phase iia clinical trial to evaluate the efficacy and safety of a cell-mediated gene therapy in degenerative knee arthritis patients. Hum Gene Ther Clin Dev. 2015;26:125–30.. doi: 10.1089/humc.2014.145. [DOI] [PubMed] [Google Scholar]
- 135. Adkar SS, Wu C-L, Willard VPet al. Step-wise chondrogenesis of human induced pluripotent stem cells and purification via a reporter allele generated by CRISPR-Cas9 genome editing. Stem Cells. 2019;37:65–76.. doi: 10.1002/stem.2931. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136. Attia N, Mashal M, Puras Get al. Mesenchymal stem cells as a gene delivery tool: Promise, problems, and prospects. Pharmaceutics. 2021;13:843. doi: 10.3390/pharmaceutics13060843. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137. Prasadam I, Akuien A, Friis TEet al. Mixed cell therapy of bone marrow-derived mesenchymal stem cells and articular cartilage chondrocytes ameliorates osteoarthritis development. Lab Invest. 2018;98:106–16.. doi: 10.1038/labinvest.2017.117. [DOI] [PubMed] [Google Scholar]
- 138. Lee J-M, Im G-I.. SOX trio-co-transduced adipose stem cells in fibrin gel to enhance cartilage repair and delay the progression of osteoarthritis in the rat. Biomaterials. 2012;33:2016–24.. doi: 10.1016/j.biomaterials.2011.11.050. [DOI] [PubMed] [Google Scholar]
- 139. Lee KI, Gamini R, Olmer Met al. Mohawk is a transcription factor that promotes meniscus cell phenotype and tissue repair and reduces osteoarthritis severity. Sci Transl Med. 2020;12:eaan7967. doi: 10.1126/scitranslmed.aan7967. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140. Rhee J, Park S-H, Kim S-Ket al. Inhibition of BATF/JUN transcriptional activity protects against osteoarthritic cartilage destruction. Ann Rheum Dis. 2017;76:427–34.. doi: 10.1136/annrheumdis-2015-208953. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141. Wu H, Peng Z, Xu Yet al. Engineered adipose-derived stem cells with IGF-1-modified mRNA ameliorates osteoarthritis development. Stem Cell Res Ther. 2022;13:19. doi: 10.1186/s13287-021-02695-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142. Zhang X, Crawford R, Xiao Y.. Inhibition of vascular endothelial growth factor with shRNA in chondrocytes ameliorates osteoarthritis. J Mol Med (Berl). 2016;94:787–98.. doi: 10.1007/s00109-016-1425-0. [DOI] [PubMed] [Google Scholar]
- 143. Watson Levings RS, Smith AD, Broome TAet al. Self-complementary adeno-associated virus-mediated interleukin-1 receptor antagonist gene delivery for the treatment of osteoarthritis: Test of efficacy in an equine model. Hum Gene Ther Clin Dev. 2018;29:101–12.. doi: 10.1089/humc.2017.143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144. Zhang X, Mao Z, Yu C.. Suppression of early experimental osteoarthritis by gene transfer of interleukin-1 receptor antagonist and interleukin-10. J Orthop Res. 2004;22:742–50.. doi: 10.1016/j.orthres.2003.12.007. [DOI] [PubMed] [Google Scholar]
- 145. Dai L, Zhang X, Hu Xet al. Silencing of miR-101 prevents cartilage degradation by regulating extracellular matrix-related genes in a rat model of osteoarthritis. Mol Ther. 2015;23:1331–40.. doi: 10.1038/mt.2015.61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146. Zhou Z-B, Huang G-X, Fu Qet al. circRNA.33186 contributes to the pathogenesis of osteoarthritis by sponging miR-127-5p. Mol Ther. 2019;27:531–41.. doi: 10.1016/j.ymthe.2019.01.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147. Tang Sa, Cao Y, Cai Zet al. The lncRNA PILA promotes NF-κb signaling in osteoarthritis by stimulating the activity of the protein arginine methyltransferase PRMT1. Sci Signal. 2022;15:eabm6265. doi: 10.1126/scisignal.abm6265. [DOI] [PubMed] [Google Scholar]
- 148. Tanikella AS, Hardy MJ, Frahs SMet al. Emerging gene-editing modalities for osteoarthritis. Int J Mol Sci. 2020;21:6046. doi: 10.3390/ijms21176046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149. Liu WY, Li LX, Jiang JXet al. Applications and challenges of CRISPR-Cas gene-editing to disease treatment in clinics. Precis Clin Med. 2021;4:179–91.. doi: 10.1093/pcmedi/pbab014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150. Zhang F, Wen Y, Guo X. CRISPR/Cas9 for genome editing: Progress, implications and challenges. Hum Mol Genet. 2014;23:R40–6.. doi: 10.1093/hmg/ddu125. [DOI] [PubMed] [Google Scholar]
- 151. Seidl CI, Fulga TA, Murphy CL.. CRISPR-Cas9 targeting of MMP13 in human chondrocytes leads to significantly reduced levels of the metalloproteinase and enhanced type II collagen accumulation. Osteoarthritis Cartilage. 2019;27:140–7.. doi: 10.1016/j.joca.2018.09.001. [DOI] [PubMed] [Google Scholar]
- 152. Karlsen TA, Pernas PF, Staerk Jet al. Generation of IL1β-resistant chondrocytes using CRISPR-CAS genome editing. Osteoarthritis Cartilage. 2016;24:S325. doi: 10.1016/j.joca.2016.01.581. [Google Scholar]
- 153. Bonato A, Fisch P, Ponta Set al. Engineering inflammation-resistant cartilage: Bridging gene therapy and tissue engineering. Adv Healthc Mater. 2023;2023:e2202271. doi: 10.1002/adhm.202202271. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154. Seidel MF, Wise BL, Lane NE.. Nerve growth factor: An update on the science and therapy. Osteoarthritis Cartilage. 2013;21:1223–8.. doi: 10.1016/j.joca.2013.06.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155. Zhao L, Huang J, Fan Yet al. Exploration of CRISPR/Cas9-based gene editing as therapy for osteoarthritis. Ann Rheum Dis. 2019;78:676–82.. doi: 10.1136/annrheumdis-2018-214724. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156. Coleman RM. Engineering closed-loop, autoregulatory gene circuits for osteoarthritis cell-based therapies. Curr Rheumatol Rep. 2022;24:96–110.. doi: 10.1007/s11926-022-01061-x. [DOI] [PubMed] [Google Scholar]
- 157. Brunger JM, Zutshi A, Willard VPet al. Genome engineering of stem cells for autonomously regulated, closed-loop delivery of biologic drugs. Stem Cell Reports. 2017;8:1202–13.. doi: 10.1016/j.stemcr.2017.03.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158. Pferdehirt L, Ross AK, Brunger JMet al. A synthetic gene circuit for self-regulating delivery of biologic drugs in engineered tissues. Tissue Eng Part A. 2019;25:809–20.. doi: 10.1089/ten.TEA.2019.0027 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159. Deshmane SL, Kremlev S, Amini Set al. Monocyte chemoattractant protein-1 (MCP-1): An overview. J Interferon Cytokine Res. 2009;29:313–26.. doi: 10.1089/jir.2008.0027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160. Choi Y-R, Collins KH, Springer LEet al. A genome-engineered bioartificial implant for autoregulated anticytokine drug delivery. Sci Adv. 2021;7:eabj1414. doi: 10.1126/sciadv.abj1414. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161. Ding X, Yu L, Chen Let al. Recent progress and future prospect of CRISPR/Cas-derived transcription activation (CRISPRa) system in plants. Cells. 2022;11:3045. doi: 10.3390/cells11193045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162. Farhang N, Davis B, Weston Jet al. Synergistic CRISPRa-regulated chondrogenic extracellular matrix deposition without exogenous growth factors. Tissue Eng Part A. 2020;26:1169–79.. doi: 10.1089/ten.TEA.2020.0062. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163. Aubourg G, Rice SJ, Bruce-Wootton Pet al. Genetics of osteoarthritis. Osteoarthritis Cartilage. 2022;30:636–49.. doi: 10.1016/j.joca.2021.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164. Dicks AR, Maksaev GI, Harissa Zet al. Skeletal dysplasia-causing TRPV4 mutations suppress the hypertrophic differentiation of human iPSC-derived chondrocytes. Elife. 2023;12:e71154. doi: 10.7554/eLife.71154 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165. Klein JC, Keith A, Rice SJet al. Functional testing of thousands of osteoarthritis-associated variants for regulatory activity. Nat Commun. 2019;10:2434. doi: 10.1038/s41467-019-10439-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166. Stolberg SG. The biotech death of Jesse Gelsinger. N Y Times Mag. 1999;136-140:149–150. [PubMed] [Google Scholar]
- 167. Kobayashi H, Hirata M, Saito Tet al. Transcriptional induction of ADAMTS5 protein by nuclear factor-κb (NF-κB) family member RelA/p65 in chondrocytes during osteoarthritis development. J Biol Chem. 2013;288:28620–9.. doi: 10.1074/jbc.M113.452169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168. Kobayashi H, Chang SH, Mori Det al. Biphasic regulation of chondrocytes by Rela through induction of anti-apoptotic and catabolic target genes. Nat Commun. 2016;7:13336. doi: 10.1038/ncomms13336. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169. van der Kraan PM. The changing role of tgfβ in healthy, ageing and osteoarthritic joints. Nat Rev Rheumatol. 2017;13:155–63.. doi: 10.1038/nrrheum.2016.219. [DOI] [PubMed] [Google Scholar]
- 170. Zhang P, Zhong ZH, Yu HTet al. Exogenous expression of IL-1Ra and TGF-β1 promotes in vivo repair in experimental rabbit osteoarthritis. Scand J Rheumatol. 2015;44:404–11.. doi: 10.3109/03009742.2015.1009942. [DOI] [PubMed] [Google Scholar]


