Abstract
Degenerative disc disease (DDD) is a painful, chronic and progressive disease, which is characterized by inflammation, structural and biological deterioration of the intervertebral disc (IVD) tissues. DDD is specified as cell-, age-, and genetic-dependent degenerative process that can be accelerated by environmental factors. It is one of the major causes of chronic back pain and disability affecting millions of people globally. Current treatment options, such as physical rehabilitation, pain management, and surgical intervention, can provide only temporary pain relief. Different animal models have been used to study the process of IVD degeneration and develop therapeutic options that may restore the structure and function of degenerative discs. Several research works have depicted considerable progress in understanding the biological basis of disc degeneration and the therapeutic potentials of cell transplantation, gene therapy, applications of supporting biomaterials and bioactive factors, or a combination thereof. Since animal models play increasingly significant roles in treatment approaches of DDD, we conducted an electronic database search on Medline through June 2020 to identify, compare, and discuss publications regarding biological therapeutic approaches of DDD that based on intradiscal treatment strategies. We provide an up-to-date overview of biological treatment strategies in animal models including mouse, rat, rabbit, porcine, bovine, ovine, caprine, canine, and primate models. Although no animal model could profoundly reproduce the clinical conditions in humans; animal models have played important roles in specifying our knowledge about the pathophysiology of DDD. They are crucial for developing new therapy approaches for clinical applications.
Keywords: Degenerative disc disorders, chronic back pain, animal models of disc degeneration, biological treatment approaches, intradiscal regenerative strategies, biological restoration of intervertebral disc tissues
Impact statement
Degenerative disc disease (DDD) in human is a complex and multifactorial process involving for example cellular senescence, mechanical stresses, nutritional deprivation, and genetic predisposition. The complex and multifactorial process has been studied in different animal models. Little progress has been achieved from biological treatment approaches in animal models to clinical trials. Nevertheless, animal models play increasingly significant roles in treatment approaches of DDD. Therefore, we provide here an up-to-date overview of biological treatment strategies in degenerative DDD using small and large animal models from mouse to non-human primate. This review may help the specification of our knowledge about the pathophysiology and treatment modalities of DDD, in order to develop new and hopeful therapy approaches for future clinical applications.
Introduction
Structure and function of intervertebral discs
Intervertebral discs (IVDs) separate adjacent vertebra in the spinal column that provides the base body support and protects the spinal cord. IVDs are fibrocartilaginous articulations that are made up of the soft jelly-like inner core, the nucleus pulposus (NP), and the outer fibrous ring, the anulus fibrosus (AF), which are interfaced with the vertebra by the vertebral end plates (VEPs).1–3 The highly hydrated NP is mainly composed of hyaluronan and proteoglycan that are entangled in a network of fine type II collagen; whereas the AF is mainly composed of the radially aligned type I collagen fibers. The VEP is made of the cartilaginous layer, which is fused with the IVD as well as with the bony endplate layer attached to the vertebra. The permeability of the VEP allows the diffusion of water, nutrition, and oxygen to the predominantly avascular IVDs.1 IVDs have, due to their particular load-bearing organization, the ability to withstand biomechanical stress and to provide tensile strength, stability, and flexibility of the spine by spreading load evenly on the vertebral bodies and resisting spinal compression. During loading the swelling of the hydrated NP exerts a radial and axial hydrostatic pressure on AF and VEP, while AF and VEP resist and distribute the compressive loading on the adjacent vertebral bodies.2,3 IVDs have aneural environment, as the sinovertebral nerves (the neural supply) emerge from the dorsal root ganglion and extend only to the outer layer of anulus. The avascular environment of IVDs with low supply of nutrient and oxygen promotes degeneration and impairs the regeneration process of degenerative IVDs, especially in the center of the IVDs.4–9 The structure of human vertebra is illustrated in Figure 1.
Figure 1.
Structure of human vertebra. (a) Spinal column illustrating the vertebral bodies separated by intervertebral discs. (b) Lumbar spine (L3–L5) with L3/L4 and L4/L5 discs along with nerve roots and the spinal canal enclosing the spinal cord (© Mern DS).
Degenerative intervertebral disc disease
IVD degeneration is a normal process of aging that can be accelerated by different environmental and biological factors. These include for example (a) unfavorable mechanical stresses that damage the structure of the extracellular matrix (ECM), (b) genetic risk factors that contribute to IVD degeneration by affecting the biomechanical strength of the ECM and lead to low tolerance for mechanical stress, and (c) calcification of the vertebral endplates (VEPs) along with atherosclerotic obstruction of the lumbar arteries that impair nutrient supply to IVDs and affect the intradiscal cell viability and ECM synthesis.10–15
IVD degeneration associated with pain, referred to as degenerative disc disease (DDD), can considerably affect patients’ quality of life with a substantial socioeconomic impact. It can impel disabilities, social isolations, and cause huge socioeconomic cost in terms of medication, disability benefits, and lost productivity.16–18 IVD degeneration leading to backpain is caused by structural failure of the disc affecting the biomechanics of the spine and it can be aggravated by inflammatory ingrowth of blood vessels and nerves into the inner layers of the torn IVDs19,20 (Figure 2). Current treatment modalities of DDD are based on: (a) conservative measures as bed rest, non-steroidal anti-inflammatory medication, analgesia, and physical therapy, (b) interventional measures as epidural steroid injections and ablation techniques, and (c) surgical procedures as discectomy, fusion, and artificial disc replacement. Those treatment modalities are restricted on or put the emphasis on the management of DDD symptoms. They do not address options of restoring the structural or biological deterioration of the discs as the underlying problem.21,22 Therefore, biological treatment approaches of DDD that may decelerate disc degradation or restore the structure and function of IVDs have gained increasingly greater consideration over the last years.
Figure 2.
The course of degenerative disc disease. Causative factors such as aging, mechanical stress, genetic, etc. upregulate the expression of inflammatory cytokines in NPCs. The inflammatory environment leads to the overexpression of catabolic genes that degrade ECM proteins in NP tissue. Continuous degradation of the ECM proteins results in mechanical instability, disintegration of the disc tissue, annular tears, and disc herniation. Ingrowth of blood vessels and nerves into the inner layers of the torn disc releasing chemokines can promote activation and infiltration of leukocytes. These in turn amplify the inflammatory response and reinforce degeneration with back and radicular pain causing DDD (© Mern DS).
Biological treatment approaches of degenerative disc disease
Various in vivo studies have demonstrated promising biological strategies that may decelerate disc degeneration or restore the structure and function of the discs. The different biological strategies include cell transplantation, gene therapy, applications of bioactive factors and bioscaffolds, or a combination thereof.23–27 Imbalances between pro-inflammatory (catabolic) cytokines and anabolic factors within IVDs are the major aggravating factors of IVD degeneration. The imbalances could lead to loss of proteoglycan content, loss of structural organization, and loss of hydration and affect the load-bearing ability of the IVDs. Enhanced expression of inflammatory cytokines, such as tumor necrosis factor alfa (TNFα), interleukin-1β (IL-1β), interleukin-6 (IL-6), interleukin-17, and cell-surface heparan sulfate, can be induced by initiating factors of IVD degeneration. The inflammatory cytokines have been shown to enhance the expression levels of catabolic enzymes, such as disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS4, ADAMTS5), matrix metalloproteinases (MMP1, MMP3), and metalloproteinase inhibitors (TIMP1, TIMP3), which in turn substantially affect the matrix turnover within the IVDs. Loss of matrix turnover can result in reduced disc height, anulus tears, and collapsed disc space that may sensitize the spinal nerves and trigger radicular pain28–34 (Figure 2). Several studies have attempted to deregulate the imbalances by downregulating the inflammatory and catabolic factors or by upregulating the anti-catabolic and anabolic factors that inhibit the degradation of ECM or boost its synthesis within IVDs. Moreover, in various in vivo studies intradiscal upregulation of matrix anabolism has been attempted using cell transplantation combined with growth factors, such as transforming growth factor beta (TGF-β1, TGF-β3), insulin-like growth factor 1 (IGF-1), and bone morphogenetic proteins (BMP2, BMP4, BMP6, BMP7).27,35–39
Different animal models of disc degeneration have been used for regenerative approaches of cell transplantation and gene therapy with applications of bioactive factors and bioscaffolds.40–156 Autologous or allogeneic transplantations of stem cells or chondrocytes with or without genetic modifications of the cells have been often combined with the use of biodegradable scaffolds.40–52,68–82,107–114,124–127,136–140 In addition, transplantations of human stem cells or human chondrocytes have been performed in some animal models of disc degeneration.40,41,44,52,77,82,112 Stem cells and chondrocytes can be harvested, in vitro expanded, genetically modified and injected intradiscally for regeneration attempts of degenerated discs. The genetic modification of the cells can include inhibition of inflammatory and catabolic genes or overexpression of anti-catabolic and anabolic genes, or a combination thereof. The combination of both approaches can be more important to achieve additive outcomes, namely inhibition of ECM degradation along with its restoration. However, gene therapeutic approaches depend not only on the identification of the relevant target genes but also on the optimal selection and application of gene delivery systems. In vivo gene therapeutic approaches have been presented in different animal models of disc degeneration by using retrovirus, lentivirus (LV), adenovirus, and adeno-associated virus (AAV) gene delivery vectors.53–58,83–89,107,110,136,139,148 Viral vector-based gene therapeutic treatments of DDD can be performed: (I) by direct injection of viral vectors into degenerative IVDs leading to the expression of the therapeutic gene by disc cells transduced within the IVDs; (II) by injection of genetically modified cells into degenerative IVDs that are able to express the therapeutic gene in vivo; and (III) by transplanting bioscaffolds that are seeded with genetically modified cells enabling the in vivo expression of the therapeutic gene. Biological treatment approaches of DDD are schematically illustrated in Figure 3.
Figure 3.
Schematic illustration of procedures for biological treatment approaches of degenerative disc disease. Tissue isolated and expanded stem cells and chondrocytes can be used for autologous or allogeneic transplantations with or without genetical modification or loaded on bioscaffolds. Beyond that, therapeutic vectors, bioactive factors, and bioscaffolds can be injected into degenerative IVDs for inhibition of disc degeneration or boosting of its regeneration (© Mern DS).
Analyses of animal models in biological treatment approaches of DDD
Various animal models belonging to different species, which have been found to reproduce the degenerative changes in human IVDs, have been used for biological treatment approaches of DDD. Therefore, we conducted an electronic database search on Medline through May 2020 to identify, compare, and discuss findings associated with intradiscal biological treatment approaches of DDD in various animal models of different species, including mouse, rat, rabbit, porcine, bovine, ovine, caprine, canine, and non-human primate models. Inclusion criteria for selection of the articles were studies that based on intradiscal applications of biological factors in an attempt to achieve IVD regeneration in animal models. Exclusion criteria were the use of non-biological factors or non-intradiscal application routes of biological factors, such as intravenous, subcutaneous, intraperitoneal, or oral routes. Searches were performed in MEDLINE by all authors of the manuscript for in vivo biological treatment approaches of IVD by using multiple search terms such as: intradiscal treatment and IVD regeneration, IVD degeneration and cell transplantation, gene therapy and IVD regeneration, biomaterials and IVD repair, etc.
Rodent models for biological treatment approaches of DDD
Primarily rats and rarely mice have been used for biological treatment approaches of DDD. They are considered to be desirable models, since these models have the advantages of giving advanced options for genetic manipulation as well as easiness of use and husbandry with low costs. In addition, they provide readily accessible tail discs for easy induction of mechanical trauma, asymmetrical compression, and easy administration of bioactive agents. However, the small size and geometry of their discs that reduces the nutritional challenge, the different mechanical loading, and the persistent notochordal cells encompass the disadvantages of those models to mimic the degenerative or regenerative processes in human IVDs. Nonetheless, different regenerative treatment approaches have been developed using rodent models of disc degeneration.
Cell-based in vivo therapy approaches in rodent models of disc degeneration
Different rat models have been used for stem cell or chondrocyte-based treatments of degenerative discs with or without gene modification and mostly in combination with appropriate biomaterials that may maintain the viability and function of the transplanted cells40–52 (Table 1). Human mesenchymal stem cells (hMSCs) harvested from bone marrow specimens of iliac crests have been tested as cell-based therapies in a rat tail models of IVD degeneration.40–42 To explore the feasibility of MSC delivery, their retention, and survival in NP, rat coccygeal discs were injected with 15% hyaluronan gel that was seeded with MSCs expressing green fluorescent protein (GFP). After injection discs were weekly assessed for 28 days using radiography and histology. Injected MSCs could maintain viability and proliferate within the rat IVDs with 100% viability over 28 days. Moreover, a trend of increased disc height was observed as compared to blank gel injection.40 In another study, the direct treatment of denucleated rat coccygeal discs was tested with bilaminar cocultured pellets (BCPs) of human MSCs and bovine NP cells (bNPCs) that were loaded in fibrin sealant (FS) carrier. Between two and five weeks after injury, BCP treatment increased disc height over time with high cell retention as compared to control discs that were treated only with MSCs, NPCs, or FS.41 Since hypoxic and hypertonic microenvironment in IVDs can decrease the treatment effects of cell transplantation, hypoxic-preconditioned rat MSCs that express GFP (H-MSC-GFP) were tested in needle-punctured coccygeal discs (Co5/Co6 and Co6/Co7). Hypoxic-precondition was generated by culturing of MSCs in a complete medium with cobalt chloride (CoCl2) at the concentration up to 0.3 mmol/L for 48 h. Cell transplantation (2 × 104 H-MSC-GFP dissolved in 2.0 μL PBS) was performed at two weeks after initial surgery, and radiographs were taken every two weeks till six weeks. The survival, migration, and differentiation of transplanted MSCs as well as matrix protein expression were directly evaluated in freshly harvested discs. Treatments with H-MSC-GFP showed extensively increased disc height, higher cell population in NP and AF regions, and enhanced collagen II and aggrecan expressions as compared to discs treated with cells without hypoxic-preconditioning (MSC-GFP). The results indicated that hypoxic-preconditioning enhances the capacity of MSCs to repair degenerated discs.42
Table 1.
Cell transplantation approaches in rodent models of disc degeneration.
| Animal model | Transplanted cell/Gene modification | Implanted biomaterial | Disc regeneration effect | Ref. |
|---|---|---|---|---|
| Rat: Caudal disc puncture injury | hMSC/AdV-GFP | Hyaluronan gel (15%) | Increased disc height, maintained cell viability, and proliferation | 40 |
| Rat: Caudal disc puncture injury | BCP-hMSC-bNPCs | FS carrier | High cell retention, increased disc height, and proteoglycan content | 41 |
| Rat: Caudal disc puncture injury | Allogeneic hMSC/AdV-GFP | Improved cell population, disc high, collagen II, and aggrecan levels | 42 | |
| Rat: Caudal disc puncture injury | hADSC | PLGA-TGFβ3 | Enhanced disc height, MRI signal intensity, and proteoglycan content | 43 |
| Rat: Caudal disc puncture injury | hADSCs-NP-lC | Heparin-PEAD- GDF5 | Improved disc height, NP cell proliferation, and ECM synthesis | 44 |
| Rat: Caudal disc puncture injury | Autologous iPSC-NP-lC | Polymeric GM-GDF5 | Enhanced disc height, NP cell proliferation, and ECM formation | 45 |
| Rat: Lumbar disc puncture injury | Allogeneic stem cell from the olfactory mucosa / RV-GFP | Successful engraftment of donor cells expressing collagen II and chondroitin sulfate proteoglycan | 46 | |
| Rat: Caudal disc puncture injury | Allogeneic NPMSCs / LV-mCherry | RGD-peptide-hydrogel | Decreased cell apoptosis rate with restored ECM content and structure | 47 |
| Rat: Caudal disc puncture injury | Ovine AF cells | Riboflavin-HDC gel | Accelerated AF reparative sealing, better disc height, and NP hydration | 48–50 |
| Rat: Lumbar disc puncture injury | Allogeneic NPC/AdV-Luc Allogeneic ADSC/AdV-Luc Allogeneic MSC/AdV-Luc |
Alginate bead Alginate bead Alginate bead |
Highest noninvasive track of AdV-Luc gene expression within IVDs after injection with NPCs | 51 |
| Mouse: Biglycan knockout (Bgn-/0) | hADSCs | High survival of hADSCs able to express biglycan with improved MRI index and aggrecan level | 52 |
hMSC: human mesenchymal stem cell; AdV: adenoviral vector; GFP: green fluorescent protein; BCP: bilaminar cocultured pellets; bNPC: bovine nucleus pulposus cell; hADSCs: human adipose-derived stem cells; NP-lC: nucleus pulposus-like cell; iPSCs: induced pluripotent stem cells; RV: retroviral vector; NPMSCs: nucleus pulposus-derived mesenchymal stem cells; LV: lentiviral vector; NP: nucleus pulposus; AF: anulus fibrosus; Luc: luciferase; MRI: magnetic resonance imaging; ECM: extracellular matrix; IVD: intervertebral disc.
Stem cell- or chondrocyte-based treatments of degenerative discs with or without gene modification or in combination with biomaterials have been attempted.
Human adipose-derived stem cells (hADSCs), induced pluripotent stem cells (iPSCs), and multipotent stem cells from the olfactory mucosa have been used for therapeutic approaches in rat models of disc degeneration.43–46 Human ADSCs that were seeded in nano-structured three-dimensional poly-lactide-co-glycolide (PLGA) microspheres, which could dually release dexamethasone and TGF-β3, were used in needle-punctured rat coccygeal discs (Co7/Co8 and Co8/Co9). Discs were injected with 2 µL of PLGA microspheres (5 mg/mL) containing ADSCs (1 × 106 cells/mL; PMA group) at two weeks after puncture injury, and weekly analyzed by magnetic resonance imaging (MRI), radiographical, histological, immunohistochemical, and biochemical methods till 24 weeks after injection. The PMA group showed improved disc height values and MRI signal intensities with proteoglycan accumulation as compared to punctured discs treated only with 2 µL PLGA microspheres (PM groups) or untreated punctured discs (DC group).43 Furthermore, ADSCs, which were seeded on a polymer heparin-PEAD (heparin-poly(ethylene argininylaspartate diglyceride)) and differentiated into an NP-like phenotype using growth and differentiation factor-5 (GDF5), were therapeutically tested in the same manner as above in needle-punctured rat coccygeal discs. Sustainable release of GDF5, enhanced disc height, increased water content, and improved NPC proliferation were obtained with enhanced ECM synthesis.44 In the same manner, the regenerative potentials of autologous iPSCs, which were seeded on a polymeric gelatin microsphere (GM) and differentiated into NP-like cells (NP-LCs) using GDF5, were examined in needle-punctured rat coccygeal discs. Similar results with sustainable GDF5 release, enhanced disc height, increased water content, and improved NP cell (NPC) proliferation with enhanced ECM formation were demonstrated.45 In addition, the therapeutic potentials of human multipotent stem cells from the olfactory mucosa were investigated in view of disc repair after needle aspiration of the NP in lumbar discs of rats. Two weeks after injury discs were injected with undifferentiated olfactory neurosphere-derived cells (1 × 104 cells/μL) that were transduced with retroviral vectors encoding for GFP (RV-GFP). Immunocytochemical and microscopical analyses of the disc tissues at three weeks after injection showed the survival of GFP-positive donor-derived cells within the NP of transplanted discs. They were able to express NP distinctive markers such as collagen II and chondroitin sulfate proteoglycan, indicating the potential of stem cells from the olfactory mucosa to repair injured IVDs.46
Chondrocytes from NP and annulus fibrosus (AF) have been used usually in combination with biomaterials to restore the proteoglycan content in the NP or to repair anular defects that could prevent re-herniation and further degeneration.47–51 NPCs from rats expressing mCherry that were cultivated on RGD-peptide modified polysaccharide hydrogel in culture medium of MSCs (referred to as nucleus pulposus-derived mesenchymal stem cells: NPMSCs) were used for regeneration approaches in needle-punctured coccygeal discs (Co5/Co6). Two weeks after puncture, the hydrogel-loaded NPMSCs were injected into the discs and these were analyzed by MRI, radiographical, and histological analyses at one, two, four, and eight weeks after injection. Significantly higher ECM content with partly restored ECM structure and decrease cell apoptosis rate were determined in hydrogel-loaded NPMSC discs until eight weeks after injection as compared with discs that obtained only hydrogel or NPMSC injection.47 The utilities of AF cells that were loaded in riboflavin-crosslinked high-density collagen gel (HDC gel) were examined to repair anular defects and prevent re-herniation in rat tail discs. Degeneration was induced by needle puncture of coccygeal discs (Co5/Co6) that was subsequently followed by the injection HDC gel loaded with ovine AF cells. X-rays, MRI, and histological assessments conducted over five weeks showed significantly better retention of disc height, NP size, and hydration with accelerated reparative sealing after treatment with HDC gel loaded with AF cells as compared to discs with acellular treatment (HDC gel alone) or without treatment.48–50 Furthermore, genetically modified rat NPCs, ADSCs, and MSCs that were transduced with adenovirus vectors expressing luciferase reporter gene (AdV-Luc) were used to investigate the feasibility and usefulness of in vivo bioluminescent imaging of transplanted cells in degenerative rat lumbar discs. Lumbar discs were needle punctured and directly injected with transduced ADSCs, MSCs or NPCs (1 × 105 cells in 10 µL alginate bead). In vivo bioluminescent imaging and gene expression in lumbar IVDs were assessed at several time points up to 14 days after surgery. Although luciferase expression was detected in all ADSCs-, MSCs-, or NPCs-injected discs at all imaging time points, the highest luciferase expression was found in discs injected with NPCs. Moreover, higher expression of luciferase was found with the transplanted cells than with the direct in situ injection of the adenovirus vectors expressing the luciferase gene (3 × 107 AdV-luc pfu in 10 µL solution). The results presented the feasibility to noninvasive monitoring of therapeutic gene expression within IVDs of small animals.51
Moreover, knockout mice of biglycan gene (Bgn−/0) that undergo spontaneous disc degeneration with aging have been used to examine the regenerative potential of hADSCs. Bgn−/0 mice of 16 months age that exhibited severe disc degeneration on MRI were intradiscally injected with human ADSCs (8 × 104 cells) at lumbar level L1/L2. MRI, histological, and immunohistochemical evaluations until 12 weeks after transplantation displayed in hADSCs-treated discs increased MRI signal intensity and surviving ADSCs with expression of biglycan that led to enhanced level of aggrecan as compared to untreated or PBS-treated discs.52
In situ gene therapy approaches in rodent models of disc degeneration
In situ gene therapy approaches have been attempted by direct injection of recombinant viral vectors into degenerative IVDs of rats. Recombinant LV, adenoviral (AdV), or AAV vectors were injected into degenerative lumbar or coccygeal discs to examine the therapeutic relevance of target genes in DDD53–58 (Table 2).
Table 2.
In situ gene therapy approaches in rodent models of disc degeneration.
| Animal model | In situ gene delivery | Disc regeneration effect | Ref. |
|---|---|---|---|
| Rat: Caudal disc puncture injury | LV-TFEB | Delayed loss of disc height with delayed MRI and histopathological changes | 53 |
| Rat: Caudal disc puncture injury | LV-IGFBP5 | Increased NP cell proliferation and decreased NP cell apoptosis | 54 |
| Rat: Lumbar disc loading injury | LV-CHOP-shRNA | Improved MRI and histologic disc scores and inhibited apoptosis of NP cells | 55 |
| Rat: Lumbar disc puncture injury | AdV-Mfn2 | Impaired NP cell apoptosis with impaired autophagic flux and mitochondrial dysfunction | 56 |
| Rat: Caudal disc puncture injury | AAV2-BMP2 | Improved disc height, inhibited cell apoptosis with upregulated collagen II, aggrecan, SOX9 and downregulated MMP13 and CTX II | 57 |
| Mouse: Lumbar disc puncture injury | AdV-GDF5 | Increased disc height, improved MRI index with enhanced GAG level, and DNA content | 58 |
LV: lentiviral vector; TFEB: transcription factor EB; LV-IGFBP5: insulin-like growth factor binding protein 5; shRNA: short hairpin RNA; AdV: adenoviral vector; Mfn2: mitofusin 2; AAV-2: adeno-associated virus vector serotype 2; BMP2: bone morphogenetic protein 2; GDF5: differentiation factor 5; MRI: magnetic resonance imaging; MMP: matrix metalloproteinase; GAG: glycosaminoglycan.
In situ gene therapies have been attempted using lentiviral, adenoviral, and adeno-associated viral vectors carrying therapeutically relevant genes.
Lentiviral vectors encoding the transcription factor EB (TFEB), which is known as a master regulator of autophagic flux, have been used to investigate the therapeutic potential of the transcription factor. Three microliters of recombinant LV vectors (1 × 106 pfu) expressing TFEB (LV-TFEB) or normal control (LV-NC) were injected into the NP of coccygeal discs directly after needle puncture of the discs. The MRI, radiographical, and histological analyses performed at four or eight weeks after surgery showed that LV-mediated overexpression of TFEB could decelerate the loss of disc height, improve MRI signal intensity, and delay histopathological changes as compared to injured control discs.53 Similarly, 10 µL of recombinant LV vectors (1 × 106 pfu) expressing the insulin-like growth factor binding protein 5 (LV-IGFBP5) or normal control (LV-NC) were injected into the NP of coccygeal discs at seven days after needle puncture. Histological and biochemical evaluations of the discs at eight weeks after initial surgery displayed that LV-IGFBP5 injection significantly inhibited the progression of IVD degeneration by promoting NPC proliferation and inhibiting apoptosis.54 These results suggest that TFEB and IGFBP5 could be seen as potential therapeutic targets in the pathogenesis of IVD degeneration.53,54 Furthermore, the therapeutic potential of LV-mediated inhibition of the pro-apoptotic transcription factor CHOP was investigated in degenerative lumbar discs using anti-CHOP shRNA. A 2 µL injection of lentiviral vectors (1 × 106 pfu) carrying a CHOP inhibitory shRNA (LV-CHOP-shRNA) or normal control (LV-NC) was carried out at one week after mechanical induction of disc degeneration. Lumbar disc degeneration was induced by applying unbalanced dynamic and static forces on the lumbar spine for a week. MRI, radiological, histological, and TUNEL staining evaluations of the lumbar spines at one, three, five, and seven weeks after injection revealed that injection of LV-CHOP-shRNA could decelerate disc degeneration by inhibiting NPC apoptosis and by improving the MRI and histologic scores as compared to control discs. However, no significant change in disc height was observed until seven weeks after injection.55
Adenoviral vectors expressing mitofusin 2 (AdV-Mfn2), a dynamin-like GTPase playing a central role in regulating mitochondrial fusion and cell metabolism, were applied in degenerative lumbar discs. The aim was to investigate, whether the low abandoned of Mfn2 in degenerative NPCs is implicated in the pathogenesis of IVD degeneration. A 3 μL of AdV-Mfn2 (1 × 106 pfu) or normal control (AdV-NC) was directly injected after induction of needle-punctured lumbar disc degeneration (L4–L6). At eight weeks after surgery, discs were evaluated using MRI, histology, immunofluorescence, transmission electron microscopy, TUNEL staining, and immunoblotting. In discs injected with AdV-Mfn2, the development of IVD degeneration was ameliorated by improving the impairment of autophagic flux, mitochondrial dysfunction, and cellular apoptosis in NP. The results indicated that Mfn2 repression is deeply involved in the pathogenesis of IVD degeneration and its overexpression could be a promising therapeutic target.56 Moreover, AAV vectors of serotype 2 (AAV2) expressing the BMP2 were tested in degenerative coccygeal discs. A 5 µL of AAV2 expressing BMP2 (AAV2-BMP2) or normal control (AAV2-NC) at different concentrations (1 × 106, 1 × 108 or 1 × 1010 pfu) was injected two weeks after needle puncture of the discs Co6/Co7 and Co8/Co9. At eight weeks after surgery, the discs assessed by MRI, histological, and biochemical methods showed that AAV2-BMP2 treatment could significantly decelerate loss of disc height, inhibit NPC apoptosis, upregulate the expression levels of collagen II, aggrecan, and SOX9 and downregulate the levels of MMP-13 and CTX II in a dose-dependent manner.57
Furthermore, a mouse model of disc degeneration has been used to examine the regenerative effect of AdV vectors carrying the GDF5 (AdV-GDF5). Needle-punctured lumbar discs were directly injected with a 5 µL of AdV-GDF5 (1.8 × 1011 pfu/mL) or 5 µL of AdV-Luc (2 × 1011 pfu/mL) vectors that had been engineered to express the luciferase gene. Bioluminescent imaging, MRI scanning, histological, and biochemical analyses performed at one, two, four, or eight weeks after injection showed that discs treated with AdV-GDF5 could significantly increase the disc height index, the glycosaminoglycan level (GAG), the DNA content, and improve the MRI signal intensity as compared with injured discs that were treated with AdV-Luc or left untreated.58
Intradiscal application of bioactive factors in rodent models of disc degeneration
Bioactive factors, such as cytokines, antibodies, kinases, shRNAs, exosomes, lignans, and growth factors have been applied in degenerative rat discs to explore their therapeutic effects59–65 (Table 3). To investigate the regenerative potential of the anti-inflammatory cytokine, interleukin-10 (IL-10), 20 μL (10 ng/mL) of exogenous IL-10 peptide was injected into degenerative coccygeal discs (Co7/8-Co9/10) one week after needle puncture injury. Discs were evaluated using MRI, histological, and immunohistochemical methods at one, two, and four weeks after injection. In discs treated with IL-10 peptide improved MRI signal intensity, increased expression levels of sox-9, collagen II, and aggrecan with decreased expression levels of IL-1β, TNFα, and collagen X and reduced phosphorylation level of p38 MAPK were determined. The data indicated that IL-10 could have the potential to be used for anti-inflammation therapy of DDD.59 Moreover, an inhibition of the inflammatory cytokine, TNFα, was assessed in needle-punctured lumbar discs (L3–L6) using anti-TNFɑ monoclonal antibody. Punctured discs were directly injected with 2.5 µL anti-TNFɑ monoclonal antibody (0.5 mg/kg) or 2.5 µL TNFα peptide (0.25 ng) and pain behavior of the rats was measured at one and six weeks after surgery using hindpaw mechanical hyperalgesia test. In addition, disc height, disc morphology, and expression levels of pro-inflammatory cytokine were evaluated at six weeks after surgery using radiographical, histological, and biochemical methods. As expected, the intradiscal injection of TNFα peptide increased pain and aggravated disc degeneration; whereas pain threshold was linearly associated with the loss of disc height and enhanced levels of TNFα and IL-1β. In contrast, the injection of anti-TNFα antibody mitigated disc degeneration and alleviated pain to sham level.60 Likewise, the therapeutic effect of the peptide jagged-2 (JAG2), a ligand that activate notch signaling pathway, was investigated in needle-punctured lumbar discs (L4/5). Punctured discs were injected with 2 µL of JAG2 peptide (1.6 ng) and evaluated by MRI, histological, and biochemical methods at three and nine weeks after surgery. Intradiscal injection of JAG2 peptide increased the MRI signal intensity, induced Notch2 expression, and enhanced proliferation of NPC by inhibiting TNFα-induced apoptosis. Quite contrary to that a 2 µL injection of anti-Notch2-shRNA (1 × 106 pfu) decreased the MRI signal intensity and downregulated Notch2 expression with induction of NPC cycle arrest.61
Table 3.
Intradiscal applications of bioactive factors in rodent models of disc degeneration.
| Animal model | Intradiscal injected bioactive factor | Disc regeneration effect | Ref. |
|---|---|---|---|
| Rat: Caudal disc puncture injury | IL-10 peptide | Improved MRI signal intensity with enhanced levels of aggrecan, collagen II, and sox-9 plus decreased levels of IL-1β, TNFα, and collagen X | 59 |
| Rat: Lumbar disc puncture injury | Anti-TNFɑ monoclonal antibody | Alleviated pain, delayed loss of disc height, and decreased levels of TNFα and IL-1β | 60 |
| Rat: Lumbar disc puncture injury | JAG2 peptide | Enhanced MRI signal intensity with raised NP cell proliferation and Notch2 expression level | 61 |
| Rat: Caudal disc puncture injury | hMSC-exosomes | Retarded progression of disc degeneration with decreased apoptotic rate of NP cells | 62 |
| Rat: Caudal disc puncture injury | Simvastatin loaded in PEG-PLGA-PEG gel | Recovered NP weight, MRI signal intensity, and raised sGAG, BMP2, aggrecan, collagen II levels | 63 |
| Rat: Caudal disc puncture injury | Sesamin | Improved MRI signal, aggrecan, and collagen II levels and reduced ADAMTS5 and MMP3 levels | 64 |
| Mouse: Caudal disc loading injury | GDF-5 IGF-1 bFGF TGF-β1 |
Improved cell density with enhanced levels of aggrecan and collagen II levels in each growth factor treatment, but increased disc height only in GDF-5 treatments | 65 |
IL-10: interleukin 10; TNFɑ: tumor necrosis factor alfa; JAG2: jagged-2; hMSC: human mesenchymal stem cell; PEG-PLGA-PEG: poly(ethyleneglycol)-poly(lactic acid-co-glycolic acid)-poly(ethylene glycol); GDF-5: differentiation factor-5; IGF1: insulin-like growth factor-1; bFGF: basic fibroblast growth factor; TGF-β1: transforming growth factor-beta1; MRI: magnetic resonance imaging; NP: nucleus pulposus; GAG: glycosaminoglycan; ADAMTS: metalloproteinase with thrombospondin motifs; MMP: matrix metalloproteinase.
Intradiscal injections of bioactive factors, such as cytokines, antibodies, kinases, shRNAs, exosomes, lignans, and growth factors have been applied aiming at regenerative effects.
It is apparently suggested that the therapeutic effects of mesenchymal stem cells (MSCs) are largely mediated by paracrine factors such as exosomes. Exosomes are very small membrane-bound vesicles functioning as paracrine factors as mediators of cell-to-cell communication. The therapeutic effect of exosomes that were isolated from hMSCs (hMSC-exosomes) and loaded in AGEs (advanced glycation end products) was examined in needle-punctured coccygeal discs (Co7/8, Co8/9, and Co9/10). After surgery the punctured discs were injected every two weeks for eight weeks with 2 µL mixture of AGEs plus exosomes (200 μg/mL plus 100 μg/mL) or 2 µL of AGEs alone (200 μg/mL) and 2 µL of PBS, respectively. Histological and biochemical analyses showed retarded progression disc degeneration and decreased apoptotic rate of NPC after two months of treatment with MSC-exosomes and not with AGEs or PBS alone.62 Furthermore, the regenerative potential of lignans, such as simvastatin and sesamin, has been assessed in rat models of disc degeneration. To investigate the therapeutic role for simvastatin, a cholesterol-lowering medicament, injured coccygeal discs (Co5/Co6, Co7/Co8) were injected four weeks after needle puncture either with PEG-PLGA-PEG (poly(ethylene glycol)-poly(lactic acid-co-glycolic acid)-poly(ethylene glycol)) gel that was loaded with 2 μL of simvastatin (5 mg/mL) or with a gel alone. MRI, histological, and biochemical evaluations of the discs performed at two weeks after injection showed significantly retarded progression of disc degeneration by injection of simvastatin. It reverted the decreased NP weight, delayed histological changes, and improved the MRI signal intensity as well as the levels of aggrecan, sGAG, BMP-2, and collagen type II.63 Similarly, a 2 μL injection of the major lignan extracted from sesame seed (sesamin; 0.1 M) into needle-punctured coccygeal discs (Co4/Co5, Co6/Co7) resulted in retarded progression of disc degeneration. Discs treated with sesamin showed enhanced MRI signal intensity and increased levels of aggrecan and collagen II along with decreased levels of MMP-3 and ADAMTS-5.64 Moreover, regenerative effects of various growth factors, such as human GDF5, IGF-1, basic fibroblast growth factor (bFGF), and TGF-β1, have been examined in a mouse model of disc degeneration. After induction of caudal disc degeneration using static compression for seven days and a three-week period of no compression, degenerative discs were injected with 8 µL of GDF-5 (1 µg/mL), or IGF-1(1 µg/mL), or bFGF (1 µg/mL), or TGF-β1 (200 ng/mL). In situ hybridization, histological, and immunohistochemical assessments of the discs performed up to four weeks after injection showed improved cell density with increased levels of aggrecan and collagen II in all discs treated with each growth factor. However, only treatments with GDF-5 showed significantly increased disc height at four weeks after injection.65
Rabbit models for biological treatment approaches of DDD
Rabbits have been frequently used to study the process of IVD degeneration and the regenerative effects of intradiscal therapeutical approaches. Compared to the rodent models, rabbit models exhibit higher degree of structural and size homology to human spine. Due to the appearance of facet joints, paravertebral muscles, ligaments, and closed growth plates of the vertebral bodies in the rabbit spine, the biomechanical behavior is more comparable with the human spine contrary to mouse and rat tails. Moreover, in contrast to discs of mouse and rat tails, rabbit discs contain much less notochordal cells after their maturation age of 10 months.66,67 Beyond that the use of rabbit models is more cost-effective than that of larger animal models such as porcine, ovine, caprine, bovine, canine, and non-human primate models.
Cell-based in vivo therapy approaches in rabbit models of disc degeneration
To retard the process IVD degeneration or maintain the structure as well as the function of degenerative discs, different rabbit models of disc degeneration have been used with applications of autologous, allogeneic or human derived stem cells as well as chondrocytes with or without genetic modification of the cells or in combination with bioscaffolds68–82 (Table 4). Bone marrow-derived MSCs, synovial MSCs, umbilical cord tissue-derived MSCs, adipose-derived stem cells (ADSCs), and embryonic stem cells (ESCs) have been used with or without in vitro differentiation toward a chondrocyte lineage.68–78 The regenerative effect of bone morphogenic protein 2 (BMP2), which is known to have biological effects on the differentiation of MSCs into chondrocyte-like cells in platelet-rich plasma (PRP) gel culture, was investigated in a rabbit model of disc degeneration. Autologous bone marrow-derived MSCs transduced with AdV vectors encoding for BMP2 (AdV-BMP2) and loaded in PRP gel (MSC-AdV-BMP2-PRP; 1 × 106 cells/20 µL) were directly injected into anular-punctured lumbar discs. MRI, histological, and immunohistochemical evaluations at 6 and 12 weeks after transplantation showed that discs treated with MSC-AdV-BMP2-PRP could retain relatively well-preserved NP structure, significantly higher MRI signal intensity, and a greater amount of ECM proteins as compared with control discs treated with MSC-PRP or PRP alone.68 Likewise, autologous MSCs transduced with AdV vectors encoding for the transcription factor Sox9 (AdV-Sox9), which is also known to have biological effects on the differentiation of MSC into chondrocyte-like cells in chitosan-glycerophosphate (C/Gp) gel culture, were used in a rabbit model of disc degeneration. Following anulus puncture, lumbar discs were injected with MSCs transduced with Sox9 and loaded in C/Gp gel (MSC-AdV-Sox9-C/Gp). Treatment with MSC-AdV-Sox9-C/Gp showed similar effects as above at 6 and 12 weeks after injection as shown by the MRI, CT, histological, immunohistochemical, and biochemical evaluations.69 Furthermore, the application of autologous MSCs transduced with AdV vectors encoding for tissue inhibitor of metalloproteinase 1 (AdV-TIMP1) also showed regenerative effect in a rabbit model disc degeneration as determined by radiographical, immunohistochemical, and biochemical analyses. Anular-punctured lumbar discs injected with 30 µL MSC-AdV-TIMP1 (1 × 106 cells) showed significantly improved disc space and ECM content at 12 weeks after surgery as compared to discs treated with unmodified MSCs.70 Moreover, autologous MSCs transduced with lentiviral vectors encoding for bone morphogenetic protein-7 (LV-BMP7) and loaded in thiol-modified hyaluronic acid (TMHA) hydrogel were used to investigate its therapeutic efficacy and feasibility in a rabbit model disc degeneration. For that MSC-LV-BMP7-TMHA (1 × 106 cells/20 µL) was transplanted in injured discs at two weeks after needle aspiration of the NP and discs were analyzed using histological and immunohistological methods at 6 and 12 weeks after transplantation. Stimulated cell differentiation, enhanced cell proliferation, and cell survival with greater ECM homeostasis were confirmed in discs treated with MSC-AdV-BMP7-TMHA, indicating its potential for the treatment of DDD.71 Similarly, transplantation autologous MSCs (1 × 106 cells/20 µL) transduced with retroviral vectors encoding for GFP (MSC-RV-GFP) and embedded in 20 µL atelocollagen gel was performed two weeks after needle aspiration of lumbar NP. Immunohistochemical and biochemical evaluations of the lumbar discs at 2, 4, 8, 16, 24, and 48 weeks after transplantation displayed detectable GFP positive cells up to 48 weeks, which were able to express matrix molecules such as collagen II, keratan sulfate, chondroitin sulfate, aggrecan, hypoxia inducible factor 1 alpha, glutamine transporter 1, and MMP2; while the MSCs did not show significant expression of these molecules before transplantation.72 Moreover, hypoxic (1% O2) and normoxic (air) expanded allogeneic MSCs were examined to compare their in vivo therapeutic effects in anular-punctured lumbar discs of rabbits. At three to six weeks after puncture injury, discs were injected either with MSCs (1 × 106 cells) embedded in photopolymerizable biogel scaffold (25 μL), 10% w/v methacrylated gelatin biogel plus 0.15% w/v lithium phenyl 2,4,6-trimethylbenzoylphosphinate (LAP)73 or with MSCs (1 × 106 cells) suspended in 0.1 mL PBS or PBS alone.74 Discs were assessed up to 12 weeks after initial surgery using MRI, radiographical, histological, and immunohistochemical methods. As compared with controls and with discs treated with normoxic MSCs, the discs treated with hypoxic MSCs, especially if combined with gel, showed less disc space narrowing, significantly better histological scores, enhanced ECM deposition of collagen II and collagen XI, and increased CD105 and BMP7 expressions.73,74
Table 4.
Approaches of cell transplantation in rabbit models of disc degeneration.
| Animal model | Transplanted cell/Gene modification | Implanted biomaterial | Disc regeneration effect | Ref. |
|---|---|---|---|---|
| Rabbit: Lumbar disc puncture injury | Autologous MSC /AdV-BMP2 | Platelet-rich plasma gel | Improved MRI signal intensity, NP structure, ECM content | 68 |
| Rabbit: Lumbar disc puncture injury | Autologous MSC /AdV-Sox9 | Chitosanglycero-phosphate gel | Improved MRI signal intensity, NP structure, ECM content | 69 |
| Rabbit: Lumbar disc puncture injury | Autologous MSC /AdV-TIMP1 | Improved disc space with increased ECM content | 70 | |
| Rabbit: Lumbar disc NP aspiration | Autologous MSC /LV-BMP7 | Thiol-modified hyaluronic acid hydrogel | Stimulated cell differentiation, cell proliferation and survival with ECM homeostasis | 71 |
| Rabbit: Lumbar disc NP aspiration | Autologous MSC /RV-GFP | Atelocollagen gel | Promoted cell differentiation, survival and ECM homeostasis | 72 |
| Rabbit: Lumbar disc puncture injury | Allogeneic hypoxic-conditioned MSC | Photopolymerizable gel: methacrylated gelatin plus LAP | Improved disc space, cellularity, histological scores and ECM content with enhanced collagen II/XI, CD105, and BMP7 levels | 73,74 |
| Rabbit: Lumbar disc puncture injury | Allogeneic ADSC | NPCS: Bioscaffold from decellularized fresh porcine NP | NP-like differentiation, repaired NP/ECM structure with restored disc height, MRI signal intensity | 75 |
| Rabbit: Lumbar disc NP aspiration | Allogeneic synovial MSC/AdV-GFP | Improved disc height, level of collagen II, MRI signal intensity | 76 | |
| Rabbit: Lumbar disc puncture injury | Human umbilical tissue-derived MSC | Hydrogel carrier | Enhanced disc cellularity, ECM architecture, and viscoelasticity | 77 |
| Rabbit: Lumbar disc puncture injury | Murine ESC /AdV-GFP | Growth of new notochordal cell populations in degenerated NP | 78 | |
| Rabbit: Lumbar disc nucleotomy | Allogeneic NP cells | Tri-copolymer scaffold: CII/HyA/CS | Increased cell population, ECM production with improved disc space and MRI signal intensity | 79 |
| Rabbit: Lumbar disc nucleotomy | Autologous NP cells activated by AF cells | Improved cell population, ECM production, restored disc space | 80 | |
| Rabbit: Lumbar disc puncture injury | Allogeneic NP cells /AdV-BMP2 | No effect on disc height and disc degeneration grade | 81 | |
| Rabbit: Lumbar disc puncture injury | Human NP cells | Sodium hyaluronate gel (1%) | Restored disc height and ECM architecture | 82 |
MSC: mesenchymal stem cell; AdV: adenoviral vector; BMP2: bone morphogenetic protein 2; TIMP1: metalloproteinase inhibitor 1; RV: retroviral vector; GFP: green fluorescent protein; ADSC: adipose-derived stem cell; ESC: embryonic stem cells; NP: nucleus pulposus; AF: anulus fibrosus; MRI: magnetic resonance imaging; ECM: extracellular matrix.
Regenerative treatments of degenerative discs were based on stem cell and NP cell transplantation with or without genetic modification or in combination with biomaterials.
Furthermore, allogeneic ADSCs, allogeneic synovial MSCs, human umbilical cord tissue-derived MSCs, and murine ESCs that were expanded and differentiation toward a chondrocyte lineage have been therapeutically analyzed in rabbit models of disc degeneration.75–78 Allogeneic ADSCs were applied in combination with an injectable bioscaffold (NPCS), which was made-up by decellularization of fresh porcine NP. NPCS is able to retain the biochemical composition, the microstructure, and the mechanical properties of the native NP that could have stimulatory effects on the NP-like differentiation of ADSCs. A 30 µL ADSCs (2 × 106 cells) loaded in the NPCS or medium (DMEM) alone was injection of lumbar discs at four weeks after anular needle puncture. Evaluation of the discs was performed up to 16 weeks after injection using MRI, radiographical, histological, biochemical, and immunohistochemical methods. Higher NP-LC differentiation, restoration of NP/ECM structure with improved disc height, and MRI signal intensity could be detected in discs treated with ADSCs loaded in NPCS.75 In addition, synovial MSCs expressing GFP were directly transplanted into degenerative lumbar discs of rabbits after needle aspiration of the NP. The postoperative disc evaluations at 2, 4, 6, 8, 16, and 24 weeks were performed using MRI, radiographical and histological methods. As compared to untreated injured discs, synovial MSCs-treated discs showed improved disc height, MRI signal intensity, and level of type II collagen with 24 weeks intradiscal existence of the transplanted GFP positive cells.76 Moreover, human umbilical cord tissue-derived MSCs embedded in a hydrogel carrier, which were transplanted in needle-punctured rabbit lumbar discs could also show regenerative effects with improved values of disc cellularity, ECM architecture, and viscoelastic property as determined by MRI, histological, and uniaxial compression analyses at 3, 6, and 12 weeks after transplantation.77 Additionally, anular needle-punctured rabbit lumbar discs were directly treated with 20 µL of murine ESCs (1 × 106 ESCs) that were transduced with AdV vectors encoding for GFP (ESC-AdV-GFP) and cultured with cis-retinoic acid, TGF-β1, ascorbic acid, and IGF-1 for inducing differentiation toward a chondrocyte lineage. Evaluations of the discs were completed at two, six, and eight weeks after transplantation using MRI, confocal fluorescent microscopy, histochemical, and immunohistochemical methods. The results revealed the existence of notochordal-type cells and islands of notochordal cell growth in discs treated with ESCs; while untreated punctured discs and non-punctured discs were negative for notochordal cells indicating that new notochordal cell populations can be generated in degenerated discs using ESCs. However, immune response to a xenograft of mouse cells was lacking in an immunocompetent rabbit model.78
In addition to stem cells autologous, allogeneic, or xenogeneic (human origin) NPC have been used in rabbit models of disc degeneration to evaluate their therapeutic potential of retarding disc degeneration or maintaining the structure and function of degenerative discs.79–82 Allogeneic NPC (1 × 105 cells), which were seeded in tri-copolymer scaffold-construct, consisting of collagen II, hyaluronan, and chondroitin-6-sulfate (CII/HyA/CS scaffold: 6 mm in diameter, 1.5 mm thick) and cultured for one week, were transplanted in lumbar discs directly after nucleotomy. Assessments of the discs were performed up to 24 weeks after nucleotomy using MRI, radiographical, gross morphological, and histological methods plus cell viability assays. Higher T2-weighted signal intensity, retarded narrowing of the disc space, and higher number of viable NPCs with higher ECM production were detected in discs transplanted with cell-seeded scaffold than in discs transplanted with cell-free scaffold or untransplanted nucleotomy.79 Likewise, the direct reinsertion of autologous NPCs that were activated by coculture with AF cells showed comparable results as stated above.80 Contrary to that, transplantation of allogeneic NPCs that were transduced with AdV vectors encoding for bone morphogenetic protein-2 (AdV-BMP2) could not show any regeneration effect, since similar disc height and disc degeneration grade were determined using MRI and histology in all injured discs with or without cell transplantation.81 On the other hand, anular-punctured lumbar discs that were transplanted with adult human NPCs loaded in sodium hyaluronate (1%) gel showed restored disc height and disc tissue architecture as compared to injured discs with only gel treatment or without treatment.82
In situ gene therapy approaches in rabbit models of disc degeneration
In view of restoring IVD structure and function, intradiscal injection of recombinant LV, AdV, or AAV vectors encoding for target genes have been used for in situ gene therapy approaches in rabbit models of disc degeneration83–89 (Table 5).
Table 5.
In situ gene therapy approaches in rabbit models of disc degeneration.
| Animal model | In situ gene delivery | Disc regeneration effect | Ref. |
|---|---|---|---|
| Rabbit: Lumbar disc puncture injury | LV-TGFβ3-P2A-CTGF-T2A-TIMP1 | Enhanced MRI signal intensity with increased levels of aggrecan and collagen II | 83 |
| Rabbit: Lumbar disc puncture injury | LV-Survivin-P2A-TGF β3-T2A-TIMP1 | Inhibited cell apoptosis with improved MRI signal intensity, collagen II, and aggrecan levels | 84 |
| Rabbit: Lumbar disc puncture injury | AdV-TGFβ1 | Increased TGFβ1 and proteoglycan synthesis with improved MRI and histological indexes | 85,86 |
| Rabbit: Lumbar disc puncture injury | AdV-Sox9 | Maintained chondrocytic phenotype and preserved architecture of NP tissue | 87 |
| Rabbit: Lumbar disc Fn-f injection injury | AAV2-TGFβ1 | Rapid and prolonged TGFβ1 expressions with increased proteoglycan and collagen II synthesis | 88 |
| Rabbit: Lumbar disc puncture injury | AAV2-BMP2 or AAV2-TMP1 | Increased collagen II content with improved viscoelasticity, MRI, and histological indexes | 89 |
LV: lentiviral vector; TGFβ3: transforming growth factor beta 3; CTGF: connective tissue growth factor; TIMP1: metalloproteinase inhibitor 1; AdV: adenoviral vector; AAV2: adeno-associated virus vector serotype 2; BMP2: bone morphogenetic protein 2; NP: nucleus pulposus; MRI: magnetic resonance imaging.
Recombinant lentiviral, adenoviral, and adeno-associated viral constructs encoding for therapeutic genes have been used for intradiscal injections.
Anular-punctured lumbar discs of rabbits were injected at four weeks after puncture surgery with recombinant LV vectors encoding for three genes: TGFβ3, the connective tissue growth factor (CTGF), and the tissue inhibitor of metalloproteinase 1 (TIMP1) that are separated by 2A-self-cleaving-peptides (LV-TGFβ3-P2A-CTGF-T2A-TIMP1). Treatments of degenerated discs with 20 µL of LV-TGFβ3-P2A-CTGF-T2A-TIMP1 (2 × 108 pfu) showed improved MRI signal intensity and enhanced levels of aggrecan and collagen II as determined by MRI and biochemical analyses at 16 and 20 weeks after initial surgery.83 Similarly, recombinant LV vectors encoding for the anti-apoptosis gene Survivin, the anti-catabolic gene TIMP1, and the anabolic gene TGF-β3 (LV-Survivin-P2A-TGFβ3-T2A-TIMP1) were injected into anular-punctured lumbar discs of rabbits three weeks after puncture injury. Caspase-3 activity assays as well as MRI, histological, and biochemical assessments of the discs were performed at 3 and 12 weeks after injection. As above, treatments of degenerated discs with 20 µL of LV-survivin-P2A-TGFβ3-T2A-TIMP1 (2 × 108 pfu) could enhance the levels of aggrecan and collagen II with improved MRI signal intensity as well as inhibited apoptosis of NPCs.84
Furthermore, 20 µL of recombinant AdV vectors (6 × 106 pfu) encoding for the human TGFβ1 (AdV-TGFβ1) gene or luciferase reporter (AdV-LUC) were injected directly after anular puncture of rabbit lumbar discs. Immunohistochemical and gene expression assays performed at one week after injection displayed a 30-fold increase in active TGFβ1 production and a 100% increase in proteoglycan synthesis in AdV-TGFβ1-treated discs as compared to AdV-LUC treated or untreated punctured discs.85 Likewise, anular-punctured rabbit lumbar discs injected with 20 µL of AdV-TGFβ1 (10 × 106 pfu) or AdV-LacZ at eight weeks after surgery showed improved MRI and histopathological indexes at three weeks when injected with AdV-TGFβ1 as determined by MRI, histological, and immunohistochemical evaluations.86 Also, a 10 µL injection of AdV vectors (1 × 109 pfu) encoding for a chondrocyte-specific transcription factor Sox9 (AdV-Sox9) directly after anular puncture of lumbar discs resulted in disc regeneration at five weeks after injection. Histological and immunohistochemical assessments displayed maintained chondrocytic phenotype and preserved architecture of the NP as compared with discs treated with AdV-GFP (1 × 109 pfu/10 µL) or untreated injured discs.87
Moreover, the serotype 2 of AAV2 vectors encoding for TGF (AAV2-TGFβ1), bone morphogenetic protein-2 (AAV2-BMP2), or tissue inhibitor of metalloproteinase 1 (AAV2-TIMP1) were examined in rabbit models disc degeneration.88,89 After stimulation of lumbar disc degeneration by injection the fibronectin fragment (Fn-f), lumbar discs were instantly treated with 10 µL injection of AAV2-TGFβ1 or AAV2-GFP (1 × 109 pfu) or PBS alone as control. The efficiency and effect of AAV2-mediated gene transfer was assessed at 1, 4, 8, and 12-week time points after injection using immunohistochemical staining, fluorescence observation, immunoblotting, and 35S-sulfate incorporation assay. Treatment with AAV2-TGFβ1 exhibited rapid and prolonged TGFβ1 expression and increased ECM production with significantly enhanced levels of proteoglycan and collagen II, as compared to the control discs that were treated with AAV2-GFP or PBS.88 Similarly, a 10 µL injection of AAV2-BMP2 or AAV2-TIMP1 (1 × 109 pfu) into anular-punctured lumbar discs directly after surgery led to increased level collagen II and enhanced viscoelastic properties at 12 weeks after surgery as determined by MRI, uniaxial load-normalized displacement tests, and histology.89
Intradiscal applications of bioactive factors in rabbit models of disc degeneration
Bioactive factors, such as PRP, platelet-derived growth factor (PDGF), serum immersed hydrogels, growth factor analogs, small interfering RNAs (siRNAs), short peptides, antibodies, and hormones have been intradiscally applied in rabbit models of disc degeneration to examine their in vivo therapeutic potentials in DDDs90–106 (Table 6).
Table 6.
Intradiscal application of bioactive factors as regenerative therapy approaches in rabbit models of disc degeneration.
| Animal model | Intradiscal injected bioactive factor | Disc regeneration effect | Ref. |
|---|---|---|---|
| Rabbit: Lumbar disc puncture injury | Autologous platelet-rich plasma (PRP) | Restoration of disc height with better MRI signal intensity and number of chondrocyte-like cells | 90 |
| Rabbit: Lumbar disc NP aspiration | PRP-GHMs: PRP-impregnated gelatin hydrogel microspheres | Improved disc height, preserved water content, higher levels of proteoglycan, and collagen II with lower NP cell apoptosis | 91 |
| Rabbit: Lumbar disc puncture injury | Platelet-derived growth factor BB (PDGF-BB) in TMHA gel | Improved compressive strength with improved disc area, MRI index, and deposition of ECM as well as lower disc cell apoptosis | 92 |
| Rabbit: Lumbar disc microdiscectomy | Allogeneic serum immersed PGA implant | Enhanced cell migration into the defect area with improved MRI signal intensity and disc height | 93 |
| Rabbit: Lumbar disc microdiscectomy | Allogeneic serum immersed PVA implant | Enhanced cell migration into the defect area with improved MRI signal intensity and disc height | 94 |
| Rabbit: Lumbar disc puncture injury | Osteogenic protein-1 (OP-1) | Improved disc height, MRI signal intensity, proteoglycan content, and elastoviscous property | 95–97 |
| Rabbit: Lumbar disc puncture injury | Growth differentiation factor-5 (GDF-5) | Dose-dependent restoration of disc height with improved MRI and histological grading scores | 98 |
| Rabbit: Lumbar disc puncture injury | Anti-caspase3 siRNA | Reduced level of caspase 3 and apoptotic NP cells with improved MRI and histological scores | 99 |
| Rabbit: Lumbar disc puncture injury | Anti-ADAMTS5 siRNA | Reduced level of ADAMTS5 with improved MRI and histological scores, but no disc height change | 100 |
| Rabbit: Lumbar disc puncture injury | Anti-Caspase3 / anti-ADAMTS5 siRNAs | Synergistic effect with improved disc cellularity, ECM formation, histological, and MRI scores | 101 |
| Rabbit: Lumbar disc puncture injury | Short link N peptide | Increased disc height, GAG, and aggrecan levels with reduced levels of ADAMTS4/5 and MMP3 | 102,103 |
| Rabbit: Lumbar disc puncture injury | Biglycan-derived peptide (P2K) | Increased disc height, MRI signal intensity, PG content with improved disc tissue architecture | 104 |
| Rabbit: Lumbar disc puncture injury | CDH2 antibody/RES embedded PLGA-NBs | Retarded disc degeneration with increased anti-catabolic activity and accelerated RES release | 105 |
| Rabbit: Lumbar disc puncture injury | Melatonin | Improved MRI signal intensity and NP cell viability with enhanced levels of aggrecan and collagen II as well as restrained cell arrest | 106 |
PRP: platelet-rich plasma; GHM: gelatin hydrogel microspheres; PDGF: platelet-derived growth factor; TMHA: thiol-modified hyaluronic acid; PGA: poly(glycolic acid); PVA: polyvinyl alcohol; OP-1: osteogenic protein-1; GDF-5: differentiation factor-5; siRNA: small interfering RNA, ADAMTS5: disintegrin and metalloproteinase with thrombospondin motifs 5; CDH2: cadherin-2; RES: resveratrol; PLGA: poly L-lactic-co-glycolic acid; NB: nanobubble; MRI: magnetic resonance imaging; ECM: extracellular matrix; GAG: glycosaminoglycan; MMP: matrix metalloproteinase; PG: proteoglycan; NP: nucleus pulposus.
Bioactive factors, such as platelet-rich plasma, platelet-derived growth factor, serum immersed hydrogels, growth factor analogs, siRNAs, short peptides, antibodies, and hormones have been intradiscally applied.
PRP, PDGF, and serum have been described to stimulate the metabolism in IVD cells.90–94 To study the regenerative effect of PRP (a fraction of plasma in which several growth factors are concentrated at high levels) and platelet-poor plasma (PPP), a 20 µL of autologous PRP or PPP was injected into anular-punctured rabbit lumbar discs at four weeks after the puncture injury. Restoration of disc height, improvement MRI signal intensity, and growing number of chondrocyte-like cells were verified in PRP-treated discs related to PPP- or PBS-treated punctured discs as determined by MRI, histological, and biochemical analyses.90 Moreover, a 20 µL injection of PRB-impregnated gelatin hydrogel microspheres (PRP-GHMs) in degenerative lumbar discs at two weeks after partial aspiration of the NP showed significantly restored disc height, preserved water content, higher expression of proteoglycan, and collagen II with lower NPC apoptose. The results determined by MRI, immunohistochemistry, TUNEL staining, and gene expression assays at two, four, and eight weeks after injection showed the greater potential of PRP-GHMs treatment as compared to control discs that were treated with PBS-GHMs or PRP without impregnation of GHMs.91 Furthermore, recombinant human platelet-derived growth factor BB (PDGF-BB), a primary component of PDGF, was intradiscally injected in combination with TMHA gel into rabbit lumbar discs after four weeks of anular puncture. Injured lumbar discs were injected with either 20 µL of PDGF-BB (1 ng/mL) in TMHA gel (PDGF-TMHA-treated group), PDGF-BB (1 ng/mL) without TMHA gel (PDGF-treated control group), TMHA without PDGF (TMHA-treated control group), or PBS alone. Treatment efficacies were determined using x-ray, MRI, histology, and biomechanical testing at four and eight weeks after injection. Lower cell apoptosis, improved deposition of ECM in NP and AF, restored compressive strength with improved disc area and MRI index were found in PDGF-TMHA-treated or PDGF-treated groups, suggesting that PDGF-BB significantly decreases disc degeneration.92 In addition, serum-immersed polyglycolic/hyaluronic acid (PGA) hydrogel implant and serum immersed rod-shaped polyvinyl alcohol (PVA) hydrogel implant were examined for stimulations of IVD tissue repair directly after microdiscectomy of lumbar discs. Allogeneic serum-immersed PGA implant93 or PVA implant94 was inserted into injured discs, and transplanted discs were assessed using MRI and histology between 1 and 24 weeks after surgery. Improved MRI signal intensity, disc height, and cell migration into the defect area were confirmed in PGA or PVA implanted discs as compared to injured discs without implant.93,94
Furthermore, recombinant human growth factors, such as osteogenic protein-1 and growth and differentiation factor-5 (rhGDF-5) that are known to stimulate proteoglycans and collagen synthesis in cells, have been intradiscally applied to study their ability of restoring IVD tissues.95–98 A 10 µL injection of OP-1 solution with different concentrations (100 µg OP-1 in 10 µL 5% lactose)95–96 or (2 µg OP-1 in 10 µL PBS)97 have been performed into anular-punctured lumbar discs four weeks after puncture injury. The MRI, radiographical, histological, biochemical, and biomechanical analyses that were made between 2 and 24 weeks after injection revealed that the injection of OP-1 in each concentration could induce restoration of disc height, improvement of MRI signal intensity, enhancement of proteoglycan content for the entire experimental period up to 24 weeks. The dynamic viscoelastic property test (0.05 to 2 Hz) showed significantly higher elastic and viscous moduli after eight weeks in all discs that were treated with OP-1.95–97 Similarly, a 10 µL injection of rhGDF-5 solution at different dosages (10 ng, 1 µg or 100 µg in 10 µL PBS) into anular-punctured lumbar discs also showed that a single injection of rhGDF-5 at all tested dosages could have a reparative capacity through noticeable restoration of disc height, improvement of MRI, and histological grading scores with statistical significance P < 0.05 (10 ng and 1 µg rhGDF-5 injection) and P < 0.01 (100 µg rhGDF-5 injection).98
Additionally, siRNAs were tested to inhibit one of the major caspases in cell apoptosis (caspase 3) and one of the major aggrecanases responsible for disc degradation (ADAMTS5).99–101 Anular-punctured lumbar discs were injected one week after puncture surgery either with 100 µg anti-caspase3 siRNAs labeled with red fluorescence Alexa Fluor 555,99 anti-ADAMTS5 siRNAs,100 or dual siRNAs (anti-caspase3 plus anti-ADAMTS5 siRNAs).101 Discs were evaluated using MRI, histological, immunohistochemical, fluorescence staining, gene expression assay, and TUNEL staining techniques until eight weeks after the injection. Significantly improved MRI and histological scores, lower number apoptotic NPCs as well as lower expression of caspase 3 at mRNA and protein levels were verified in discs treated with the anti-caspase 3 siRNA as compared to control siRNA or PBS-treated discs.99 The anti-ADAMTS5 siRNA-treated discs also showed improved MRI and histological scores, but no significantly changed disc height.100 Synergistic effect was observed in discs treated with the dual siRNA formulation (anti-caspase 3 plus anti-ADAMTS5 siRNAs) with improved MRI and histopathological scores, disc cellularity as well as ECM formation at eight weeks after injection.101 Moreover, to evaluate the restoration in degenerative discs the short Link N (sLN) was used, the fragment of the link N peptide consisting of 16 amino acid sequence released from the N-terminus as a proteolysis product, which is supposed to play a role in matrix homeostasis and stabilization of the proteoglycan aggregates in cartilage and discs.102,103 A 10 μL injection of sLN solution (25 μg in PBS) or 10 μL PBS alone was performed in anular-punctured lumbar discs at two weeks after puncture surgery. Evaluations of the discs were carried out using radiographical, histological, and biochemical methods up to 12 weeks after injection. The results exhibited in sLN-treated discs decelerated loss of disc height, increased levels of GAG, and aggrecan with decreased levels of proteinases such as ADAMTS4, ADAMTS5, and MMP3 as compared to PBS alone-treated discs.102,103 In a similar way the biglycan-derived peptide (Peniel 2000 or P2K), which is known as a regulator of TGF-β1 signaling on ECM metabolism, was tested in anular-punctured lumbar discs at four weeks after surgery (10 μg P2K in 15 μL of 5% lactose or 15 µL 5% lactose). At 12 weeks after treatment MRI, radiographical, biochemical, and histological evaluations of the discs showed improved disc height, MRI signal intensity, proteoglycan content, and disc architecture in discs treated with P2K as compared to discs treated with lactose alone.104 Besides, the application of ultrasound (US) has been suggested as an effective means to advance gene and drug delivery by using echogenic nanoparticles, such as nanoparticles made from poly lactic-co-glycolic acid (PLGA), echogenic PLGA-nanobubbles (PLGA-NBs) combined with US. In order to enhance the intradiscal gene delivery in NPCs in vivo, PLGA-NBs embedded with resveratrol (RES) and conjugated with NP-cell-targeting-biomarker CDH2-antibody (AbCDH2) were tested in rabbit models of disc degeneration (US-mediated RES/AbCDH2 PLGA-NBs). Anular-punctured lumbar discs were injected with (US-mediated RES/AbCDH2 PLGA-NBs) four weeks after puncture surgery, where the therapeutic US was irradiated on the surface of injected discs. US-mediated RES/AbCDH2 PLGA-NBs treatment resulted in retarded disc degeneration with accelerated RES release and increased anti-catabolic activity as determined by histological and biochemical methods up to 12 weeks after injection. The results suggest that the combination of US irradiation and drug delivery through RES/AbCDH2 PLGA-NBs can be considered as a therapeutic option for DDD.105 In addition to that, melatonin, a neuroendocrine hormone secreted by the pineal body, is supposed to activate the ERK signaling pathway and promote the proliferation of NPC and the expression of aggrecan and collagen II. Thus melatonin was examined in anular-punctured lumbar discs to prove its in vivo regenerative potential. Discs were injected at four weeks after injury with a 20 μL of melatonin (2 mg/mL) or melatonin plus U0126 as ERK inhibitor (2 mg/mL plus 0.4 mg/kg) or PBS alone. MRI, histological, and biochemical evaluations of the discs at four and eight weeks after injection demonstrated that treatment with melatonin alone could improve MRI signal intensity, promote NPC viability, and inhibit cell arrest as well as upregulate aggrecan and collagen II with down regulation of collagen X as compared with discs treated melatonin plus U0126 or with PBS alone. Moreover, inhibition of the ERK1/2 signals by U0126 that reversed the role of melatonin in the regulation of NPC proliferation indicated that melatonin affected disc degeneration by activating ERK1/2 signaling pathway.106
Porcine models for biological treatment approaches of DDD
In relation to small animal models like rodents and rabbits, porcine models of disc degeneration have been used for intradiscal therapeutic approaches due to their IVD size and shape, which are more similar to that of humans. However, the abundant of notochordal cells in porcine IVDs and the higher costs related to small animal models should be taken into consideration.
Intradiscal biological therapy approaches in porcine models of disc degeneration
Cell transplantation, gene therapy, application of bioactive factors, and bioscaffolds have been used for in vivo therapy approaches in porcine models of disc degeneration. Bone marrow-derived autologous, allogeneic or xenogenic (human derived) MSCs, ADSCs, iPSCs, and chondrocytes have been implanted with or without gene modification or in combination with bioscaffolds107–117 (Table 7). To achieve anterior intradiscal fusion in porcine model of disc degeneration using minimally invasive techniques, autologous MSCs that were transduced with AdV-BMP2 were used. After removal of 1 cm tissue from each disc, four thoracic disc spaces in each porcine were injected thoracoscopically with autologous MSCs as followed: two of the discs were injected with MSCs transduced with AdV-BMP2 (MSC-AdV-BMP2), the third disc was injected with MSCs transduced with AdV-betagal as control, and the fourth disc served as the sham-operated control. At six weeks after the implantation, animals were euthanized and the spines were evaluated using CT and histology. Anterior spinal fusion and bridging bone from end-plate to end-plate were found in disc spaces treated with MSC-AdV-BMP2, while control disc spaces had shown no intervening or bridging bone. The results indicated that MSCs transduced with AdV-BMP2 could differentiate into osteoblasts and induce anterior spinal fusion in porcine model.107 In addition, autologous MSCs transduced with AdV-BMP2 (MSC-AdV-BMP2) and loaded in hydrogel were used in porcine model of disc degeneration to investigate their integration level and repair efficacy. After induction of lumbar disc degeneration by percutaneous delivery of vaporizing laser light at four lumbar discs L2–L5 (dehydration of NP using laser vaporization of the disc with an energy pulse of 1000 J over 1 min), MSC-AdV-BMP2 (1 × 106 cells) loaded in HyStem hydrogel were delivered into the disc space using the percutaneous approach. L5/L6 discs were left as non-transplantation controls and L1/L2 discs were the non-injury controls. Assessment of the discs at four and eight weeks after transplantation using MRI and histological methods showed deceleration of disc degeneration with improved MRI signal intensity and ECM architecture at the damage site as compared to non-transplantation controls.108 Also, four lumbar discs were implanted with autologous MSCs loaded in fibrin matrix at 12 weeks after partial nucleotomy. Two discs were injected with MSCs loaded in fibrin matrix (2 × 106 cells in 150 µL fibrin) and remaining two discs with acellular fibrin matrix, and discs were evaluated by micro-CT, histology, and gene expression analyses at 1 and 12 weeks after injection. Discs treated with MSCs loaded in fibrin matrix showed retarded disc degeneration with lower expression levels of IL-1ß, higher expression level of BMP2 in NP, and collagen I in AF as compared to discs treated with acellular fibrin matrix. However, regenerative changes, such as restoration of disc height, were not significant.109 Similarly, autologous MSCs transduced with retroviral vectors encoding for the reporter luciferase gene (MSC-RV-LUC) and loaded in albumin-hyaluronan hydrogel were applied in a porcine nucleotomy model. Directly after partial nucleotomy of two lumbar discs (∼10% of total nucleus volume), MSC-RV-LUC (3.3 × 106 cells) that were seeded in albumin-hyaluronan hydrogel and cultured under low levels of oxygen and glucose were injected (1 mL cell suspension) into injured discs. After three days of postoperative survival time, a short-term follow-up of intradiscal cell therapy, results of micro-CT, and biochemical evaluations demonstrated an in vivo evidence for hydrogel compression with implant volume shrank to 61% and luciferase expression of the implanted MSCs, emphasizing implant retainment inside the disc and persistence of metabolically active MSCs.110 Furthermore, allogeneic MSCs and allogeneic juvenile articular chondrocytes (JCs) seeded in fibrin matrix have been examined in a porcine model disc degeneration to compare their restoration capacities in injured IVDs. After induction of lumbar disc degeneration by nucleotomy at three randomized levels between L1 and L5 discs (one level left uninjured), denucleated discs were transplanted either with allogeneic MSCs in fibrin matrix or with allogeneic JCs in fibrin matrix at a volume of 0.5–0.7 mL cell suspension (1 × 107 cells) or with fibrin matrix alone. Radiological, histological, immunohistochemical, and biochemical analyses of the lumbar spines were performed at 3, 6, and 12 months after surgery. Discs treated with JCs exhibited abundant cartilage formation with collagen II rich ECM at 3 months and to a lesser extent at 6 and 12 months. Viable JCs were observed at all time points, whereas no evidence of viable MSCs was found. These results support the premise that committed chondrocytes are more appropriate for use in disc repair, as they are uniquely suited for survival in the ischemic disc microenvironment.111 Likewise, xenotransplantation of hMSCs or human-derived chondrocytes (hCs) in combination with hyaluronan gel (Durolane®) (Smith & Nephew, Inc. Mississauge, Ontario, Canada) were performed in porcine model of disc degeneration. Two weeks after partial nucleotomy of four lumbar discs (L1–L5), discs were injected either with hMSCs/gel or hCs/gel mix (0.5 × 106 cells), where the third disc was injected with only Durolane® gel and the fourth disc was left untreated. At 12 and 24 weeks after implantation assessments of the discs were completed using MRI, histology and immunohistochemistry, where xenotransplanted cells were traced with anti-human antibodies. Although xenotransplanted hMSCs and hCs could survive for 24 weeks and produce collagen II in all animals, more pronounced endplate changes (bone mineralization) and greater degenerative changes at MRI were observed in MSCs/gel, hCs/gel or gel alone-transplanted discs than in untransplanted injured discs. The results indicate that hyaluronan gel (Durolane®) is not suitable for use in cell therapy of injured IVDs. The high cell proliferation observed in vitro in the gel (Durolane®) could have been a negative factor in vivo.112 Since ADSCs within micro fragmented adipose tissue (MFAT) are assumed to be a promising agent for a wide range of applications in regenerative medicine, autologous MFAT that were co-cultured with nucleus pulposus cells (NPCs) were examined in porcine model of disc degeneration. At four weeks after needle puncture injury of lumbar discs L2–L5, the L4/L5 discs were injected with 200 μL of autologous ADSCs (2 × 105), the L3/L4 discs with 200 μL of autologous MFAT, the L2/L3 discs with 200 μL of DMEM and the L1/L2 discs were left untreated as stab control. Radiographical and MRI analyses performed at 0, 4, 8, and 16 weeks and followed by histological, immunohistochemical, and biochemical analyses at 16 weeks after injection showed that implantation of autologous MFAT co-cultured with NPCs led to significantly improved disc height, disc water content, and ECM structure in NP as compared to untreated, DMEM-treated or ADSCs-treated discs. The results verified that MFAT could promote the function of NPCs and NPCs could stimulate the NP-like differentiation of ADSCs.113 Furthermore, iPSCs generated from normal human dermal fibroblasts and co-cultured with MSCs in hypoxic condition (2% O2) and defined growth media for differentiation towards notochordal-like cells (NCs) were used in a porcine model of disc degeneration. The co-cultured iPSCs or MSCs alone (1 × 106 cells) suspended in 100 µL hydrogel (Geltrex™) or hydrogel alone were injected into lumbar discs at four weeks after anular puncture injury. MRI, histological, gene expression, and quantitative chemical exchange saturation transfer analyses at 12 weeks after injection displayed higher expression levels of the notochordal markers such as keratin 8, keratin 18, keratin 19, CD24, Gal3, and CTGF as well as improved MRI and histological indexes with minor change of pH level in discs treated with the co-cultured iPSCs. The results confirm that iPSCs can be differentiated into NCs and able to retard IVD degeneration.114 Moreover, non-cell-based implantation of biomaterials such as thiol-modified hyaluronan elastin-like polypeptide composite (TMHA/EP),115 hyaluronan collagen gel matrix composite,116 or gelfoam117 have been used in porcine model of disc degeneration as replacement agent for NP or to prevent recurrent disc herniation. Although the disc integrity of the implanted discs was better than that of bone-cement, tissue-glue, or platinum-coil implanted discs, in all cases disc injuries could not be recovered between 8 and 24 weeks after transplantation. The mechanical behavior of implanted discs was restored only in early-stage of degeneration, indicating the importance of cell or bioactive factors in transplantation for long-term and effective recovery of IVD injury.115–117
Table 7.
Biological treatment approaches of degenerative discs in porcine models of disc degeneration.
| Animal model | Transplanted cell/Gene modification | Implanted biomaterial | Disc regeneration effect | Ref. |
|---|---|---|---|---|
| Pig: Thoracic disc discectomy | Autologous MSC/AdV-BMP2 | Anterior spinal fusion, bridging bone from end plate to end plate | 107 | |
| Pig: Lumbar disc laser vaporization | Autologous MSC | HyStem hydrogel | Improved MRI signal intensity and damage site ECM formation | 108 |
| Pig: Lumbar disc partial nucleotomy | Autologous MSC | Fibrin matrix | Enhanced collagen I in AF and BMP2 in NP with lesser IL-1β | 109 |
| Pig: Lumbar disc partial nucleotomy | Autologous MSC /RV-LUC | Albumin-hyaluron-hydrogel | Implant retainment with in vivo activity of the implanted cells | 110 |
| Pig: Lumbar disc partial nucleotomy | Allogeneic MSC or Allogeneic JC |
Fibrin matrix Fibrin matrix |
JC enhanced ECM and cartilage formation with vigorous viability | 111 |
| Pig: Lumbar disc partial nucleotomy | Human MSC or Human chondrocytes |
Durolane gel Durolane gel |
Not suitable for use in vivo due to endplate bone mineralization | 112 |
| Pig: Lumbar disc puncture injury | Autologous ADSC within MFAT | Enhanced disc height, NP water content and ECM structure | 113 | |
| Pig: Lumbar disc puncture injury | Hypoxic iPSC cocultured with MSC | Geltrex hydrogel | Improved levels of notochordal markers with greater MRI index | 114 |
| Pig: Lumbar disc CABC inject injury | TMHA/EP matrix composite | Good disc integrity, but without long-term disc recovery | 115 | |
| Pig: Lumbar disc partial nucleotomy | Collagen-hyaluronan matrix composite | Good disc integrity, but without long-term disc recovery | 116 | |
| Pig: Lumbar disc puncture injury | Gelfoam | Good disc integrity, but without long-term disc recovery | 117 |
MSC: mesenchymal stem cell; AdV: adenoviral vector; BMP2: bone morphogenetic protein 2; Luc: luciferase; JC: juvenile articular chondrocyte; ADSC: adipose-derived stem cell; MFAT: micro fragmented adipose tissue; iPSC: induced pluripotent stem cell; MRI: magnetic resonance imaging; ECM: extracellular matrix; NP: nucleus pulposus; AF: anulus fibrosus.
Intradiscal treatments include cell transplantation with or without gene modification as well as cell-based or non-cell-based implantation of biomaterials.
In vivo treatment approaches of DDD in a bovine model
Bovines are mostly used as large animal IVD organ donors for ex vivo study of disc biomechanical behavior. They are used to test the effect of stress under different loading conditions and the regenerative effect of different materials in an IVD-organ culture. Nevertheless, preliminary results involving the feasibility of the runt cow for in vivo testing of a spinal interbody prosthesis have been published. Transosseous implantations of a human-sized disc prosthesis were performed in lumbar spines (L4/L5) of mature runt cows (Corrientes breed). At 16 and 24 weeks after implantation surgery, the lumbar spines were harvested and evaluated based on healing process, motion segment mobility, and ability to remove the implant nondestructively. The results showed that all animals had successful implantation with surgical healing and intended device motion. Moreover, bone ingrowth into the device's porous plates was confirmed with trabecular bone increase adjacent to the implants. The results of this study have specified the possibility of using mature runt cow for in vivo testing of human-sized lumbar interbody implants and the ability of performing common analyses of explants.118
Ovine and caprine models for biological treatment approaches of DDD
Ovine and caprine have been used as large animal models in translational research, especially for testing the functionality of implants in regenerative therapy approaches. Ovine and caprine belong to animal species that lose their notochordal cells in early adulthood, predisposing their IVDs to degeneration similar to humans.67 Moreover, mature ovine and cappine have roughly similar weight to humans and their discs have anatomical similarities regarding size and shape to human IVDs. Despite their quadrupedal stature, their IVDs are exposed to comparable mechanical stresses and show qualitatively similar craniocaudal variation in range of motion (ROM) in all load directions as human IVDs. They are resilient animals to endure invasive surgical interventions.119–123
Intradiscal biological therapy in ovine and caprine models of disc degeneration
Cell transplantation and implantation of supporting bioscaffolds have been used to repair disc injuries in ovine and caprin models of disc degeneration. Allogeneic bone marrow-derived MSCs, allogeneic bone marrow-derived mesenchymal progenitor cells (MPCs), autologous bone marrow-derived mononuclear cells (BMCs); as well as bioscaffolds such as riboflavin-crosslinked HDC gel, gelatin/fibrin scaffold (SCAF), dodecyl-amide of hyaluronic acid-based hydrogel (DDAHA), polyglycolic acid/polyvinylidene fluoride (PVDF), and small intestinal submucosa (SIS)-based scaffold (SIS-based patch and plug scaffold) have been used in ovine models of annulotomy or nucleotomy124–130 (Table 8). Caprine models of disc degeneration have been used to investigate the regenerative potential of growth factors, such as BMP2, BMP7, IGFI, and TGFβ1 in combination with bioresorbable scaffolds or cages in monosegmental spinal fusion approaches131–133 (Table 8).
Table 8.
Biological approaches of disc repair in ovine and caprine models of disc degeneration.
| Animal model | Transplanted cell/Gene modification | Implanted biomaterial | Disc regeneration effect | Ref. |
|---|---|---|---|---|
| Sheep: Lumbar disc anulotomy | Allogeneic MSC | Restored disc height, GAG level, and MRI / histological grade | 124 | |
| Sheep: Lumbar disc anulotomy | Allogeneic MSC | HDC gel/riboflavin crosslinked | Improved disc height and MRI index with restored AF tissue | 125 |
| Sheep: Lumbar disc anulotomy | Allogeneic MPC | SCAF scaffold mixed with PPS | Restored disc morphology, disc height, and proteoglycan content | 126 |
| Sheep: Lumbar disc nucleotomy | Autologous BMC | iGG-MA/DDAHA/ antiangiogenic PEP | Only slight NP restoration with DDAHA gel loaded with BMC | 127 |
| Sheep: Lumbar disc anulotomy | PVDF scaffold | Repaired AF providing effective barrier for herniating NP | 128 | |
| Sheep: Lumbar disc anulotomy | SIS-based patch and plug scaffold | Improved MRI index with ECM building and functional recovery | 129 | |
| Sheep: Lumbar disc puncture injury | Recombinant BMP13 peptide | Enhanced NP hydration, NP cell population, and ECM formation | 130 | |
| Sheep: Lumbar disc CABC injection injury | BMP2 and BMP2/7 conjugated FB/HA hydrogel/TGmoiety | Conjugated BMP2 and BMP2/7 appeared safe, but without any observed regeneration effect | 131 | |
| Goat: Cervical disc discectomy | IGF-I, TGF-β1, and hydroxyapatite coated HCIF-Cage | Improved stiffness in flexion, extension, and lateral bending with higher DSH, IVA, and LA | 132 | |
| Goat: Lumbar disc discectomy | Bioresorbable PLLA cage | More complete and faster fusion with unchanged grafting height, trabecular bone replaced PLLA | 133 |
MSC: mesenchymal stem cell; MPC: mesenchymal progenitor cell; BMC: bone marrow derived mononuclear cell; HDC: high-density collagen; SCAF: gelatin/fibrin scaffold; PPS: polyurethane porous scaffolds; iGG-MA: hydrogels made of ionic crosslinked methacrylated gellan gum; DDAHA: dodecyl-amide of hyaluronic acid; PEP: anti-angiogenic peptide; PVDF: polyglycolic acid/polyvinylidene fluoride; SIS: small intestinal submucosa; BMP: bone morphogenetic protein; FB/HA: fibrin/hyaluronic acid; IGF-I: insulin-like growth factor 1; TGF-β1: transforming growth factor beta 1; HCIFC: hat-type cervical intervertebral fusion cage; PLLA: poly(L-lactide); GAG: glycosaminoglycan; MRI: magnetic resonance imaging; ECM: extracellular matrix; DSH: disc space height; IVA: intervertebral angle; LA: lordosis angle.
Disc repair approaches include intradiscal applications of cells, growth factors, and bioscaffolds or cages.
To investigate their healing capacity in postero-lateral anular lesions, allogeneic bone marrow-derived MSCs were used in ovine model of disc degeneration. Scalpel blade postero-lateral anulotomy was performed in three lumbar discs (L3–L6) to induce disc injury. At 28 weeks after anulotomy, two discs were injected with allogeneic MSCs (1 × 106 in 200 mL PBS) either into the previously incised AF (AFI) or into the NP (NPI) and one disc was left uninjected as a negative control (Neg. C); whereas the NP of the superior adjacent uninjured disc (L2/L3) was injected with 200 mL PBS as a positive control (PC). Radiographs and MRI scans were obtained at baseline, 6, 9, and 12 months after injection and lumbar spines were harvested at 12 months for biochemical and histological analyses. Disc height, MRI, and histopathological grades were improved in AFI and NPI treatment groups with enhanced GAG content after six months. Both AFI and NPI groups showed repair of the postero-lateral anular lesion, while consistent disc degeneration was observed in control groups without MSC injection.124 In addition, allogeneic bone marrow-derived MSCs have been used in combination with riboflavin-crosslinked HDC gel in an attempt to improve the in vivo anular repair capacity in an ovine model of annular injury. Scalpel blade lumbar disc injury was made (3 mm × 1 cm anulotomy plus 100 mg discectomy) at 12 discs from L1 to L6 in three sheeps, whereby 3 of the 15 randomized discs left intact without injury as controls. Directly after anular injury four discs were treated with HDC gel seeded with MSCs (1 × 106 cell/mL), four discs with acellular HDC gel, and four discs left untreated. Animals were euthanized at six weeks after surgery and lumbar discs were assessed by MRI, radiographical, and histological methods. Although all treated discs retained gel plugs on gross assessment, discs treated with HDC gel seeded with MSCs showed significantly improved disc height and MRI index as compared with discs treated with acellular HDC gel or left untreated. The treatment of anular injury with HDC gel seeded with MSCs could improve the disc tissue (AF/NP) reconstitution and organization.125 Furthermore, formulations of allogeneic MPCs and pentosan polysulfate (PPS) were used in combination with SCAF at the time of microdiscectomy to improve the capacity of disc restoration in ovine model. Scalpel blade lumbar disc injury was made (5 mm × 3 cm anulotomy plus 200 mg discectomy) at lumbar discs (L2–L5) and injured discs levels were randomized. Randomized discs were treated either with MPC + PPS + SCAF (100 μL containing 5 × 106 MPCs + 125 μg PPS loaded in SCAF piece [5 × 5 × 3 mm]) or treated with SCAF piece alone (5 × 5 × 3 mm) as positive control or left untreated as negative control. Animals were euthanized at six months after surgery for radiological, biochemical, and histological analyses of the lumbar spines. The capacity of disc repair was improved by restoring disc height, disc morphology, and proteoglycan content in discs treated with MPC + PPS + SCAF as compared with that in both positive and negative controls.126 In addition, two different hydrogels as NP replacement were tested with joint use of an anti-angiogenic peptide or autologous BMCs to restore the biofunctionality of nucleotomized IVDs in an ovine model. For the anti-angiogenic peptide (PEP) approach a hydrogel made of ionic crosslinked methacrylated gellan gum (iGG-MA) was functionalized with non-cytotoxic polylysine-based VEGF-blockers (iGG-MA + PEP), and for the BMC approach a hydrogel made of DDAHA was used (DDAHA + BMC). The harvesting of bone marrow from the iliac crest as well as the isolation of BMCs and the orthotopic implantation of DDAHA + BMCs were combined in a one-step surgical procedure. Directly after scalpel blade neuclotomy of lumbar discs (L1–L6) with removal of 200 mg NP, discs were treated in an alternating sequence with iGG-MA, iGG-MA + PEP, DDAHA or DDAHA + BMC, or left untreated as neuclotomy control. Animals were euthanized after 6 weeks for only histological analysis and after 12 weeks for histological and biomechanical analyses. The six-week group for biomechanics was omitted, because anulur healing was not to be expected at this time period. For biomechanical and histological comparison with intact discs native lumbar spines and lumbar segments from independent comparable sheep were used. Albeit not significant, the DDAHA + BMC treatment slightly tended to decelerate degeneration between 6 and 12 weeks. Apart from that no marked differences in degeneration and biomechanical properties were observed between any of implanted or nucleotomized discs.127
Furthermore, cell free biointegrative anulus implants such as polyglycolic acid/PVDF and SIS were used for anular defect closure in ovine models of anulotomy.128,129 To investigate the efficacy of PVDF implant standardized box-shaped scalpel blade anular defects (3.5 × 3.5 mm) were made in lumbar discs (L3–L5) and PVDF implant was randomly inserted.128 Animals were euthanized at 2, 6, and 12 weeks after surgery and explanted lumbar spines were directly analyzed using biomechanical and histological methods. A provocative pressure testing in implanted discs at a mean pressure of 0.53 MPa showed a contrast media leakage between different groups or within each group without any significant differences at 2, 6, and 12 weeks after implantation. However, the intact control discs did not show any contrast media leakage up to the pressure limit of 4.8 MPa. Nevertheless, the PVDF implant consistently provided an effective barrier for herniating NP without any implant dislocation at all time-points. Repair tissue thickness was significantly stronger with the PVDF implant as compared to defect control without implant.128 To investigate the efficacy of a SIS-based patch and plug scaffold, lumbar discs (L2–L5) were randomly assigned to one of the three treatments after anulotomy (4 × 8 mm lesion with removal of AF to the level of NP): (a) exposure-only (no additional procedure), (b) box anulotomy with screw placement only (anulotomy-only), and (c) box anulotomy with placement of SIS (25 × 14 mm) and titanium screw fixation.129 At 26 weeks after surgery, pressure-volume testing, MRI, and undecalcified histological evaluation were performed with randomized motion segments. As compared to the anulotomy-only group, the SIS-treated group showed significantly improved MRI signal intensity with new tissue formation that was integrated well with the native AF. The SIS patch and plug reduced the cascade of functional degeneration with substantial functional recovery (66% of its capacity to develop internal pressure).129 In addition to that, a recombinant BMP13 was used in an ovine model to test its therapeutic effect on injured IVDs.130 Lumbar discs were directly injected after annular puncture either with BMP-13 (300 μg/70 μL PBS) or 70 μL PBS alone. Discs were analyzed by X-ray at baseline, 2, and 16 weeks after surgery that was followed by evaluations by CT, MRI, and histology immediately after euthanasia at 16 weeks after surgery. Discs injected with BMP-13 showed greater retention of hydration and retarded histological changes with enhanced NPC population and ECM formation as compared with PBS-injected control discs.130
Furthermore BMP-2 and BMP-7 that were conjugated to a fibrin/hyaluronic acid (FB/HA) hydrogel allowing a slow release of the BMPs were used in a caprine model of disc degeneration. Mild IVD degeneration in lumbar discs (L1–L6) was induced by injection chondroitinase ABC (CABC; 0.25 unit/mL: CABC dissolved in PBS), whereas the disc (T13/L1) was left as a healthy control. At 12 weeks after CABC injection, injured discs were treated with injection of the FB/HA hydrogels supplemented with either BMP-2 monomer or BMP-2/BMP-7 dimer in two concentrations (1 or 5 μg/mL) or with injection of the FB/HA hydrogel alone as control. At 12 weeks after treatment spines (T13–L6) were harvested for radiographical, MRI, biochemical, and histological analyses. Radiographs showed significant disc height loss upon induction of mild degeneration that correlated with MRI, biochemical, and histological results. Surprisingly, no difference could be confirmed in any parameter between intervention groups. The conjugated BMP-2 and BMP-2/BMP-/7 appeared safe, but no disc regeneration was observed. Possible explanations could be too low dosages and/or insufficient release of the conjugated BMPs.131 In addition to that of IGF-I and TGF-β1 in combination with a hydroxyapatite (HA)-coated cage (HCIFC: hat-type cervical intervertebral fusion cage) were used in a treatment approach of goat cervical discectomy model. Goats underwent C3/C4 discectomy and intervertebral fusion was performed using: (a) HCIFC coated with HA plus IGF-I and TGF-β1, (b) HCIFC coated with HA, (c) HCIFC only, or (d) autologous tricortical iliac crest bone graft. Radiography and measurements of disc space height (DSH), intervertebral angle (IVA), and lordosis angle (LA) were performed before surgery and at 0, 1, 2, 4, 8, and 12 weeks after surgery. Fused segments were harvested at 12 weeks after surgery to characterize the biomechanics in flexion, extension, axial rotation, and lateral bending with a nondestructive stiffness method. The stiffness of HCIFC coated with HA plus IGF-I and TGF-β1 in flexion, extension, and lateral bending was significantly greater than that of the other groups, and the stiffness of HCIFC coated with HA in extension, axial rotation, and lateral bending was significantly greater than that of fusion with the bone graft and cage (HCIFC) only. All three cage-treated groups showed significantly higher values for DSH, IVA, and LA as well as better fusion compared with the autologous bone graft group. The data indicate that HA plus IGF-I and TGF-β1 coating can improve the fusion effect of the cervical intervertebral cage by enhancing bone fusion.132 Furthermore, two bioresorbable poly(L-lactic acid) (PLLA) cages of the same size (10 × 10 × 18 mm) with the respective axial compression stiffness of 2 kN/mm (wall thickness of 0.75 mm) or 4 kN/mm (wall thickness of 1.5 mm) were applied in goat model of lumbar discectomy for monosegmental spinal fusion. Their applicabilities were compared with titanium cages with axial compression stiffness of 700 kN/mm, but with the same dimension and wall thickness as the stiff PLLA cages, respectively. PLLA and titanium cages were packed with autogenous iliac bone graft and randomly inserted directly after discectomy of the lumbar disc L3/L4. To estimate interbody fusion within the cages, radiographs were taken immediately after surgery and at the time of euthanasia in a three-year follow-up in vivo study. Before euthanasia bones were labeled with tetracycline and calcein to analyze bone remodeling and bone formation within the cages followed by micro-CT and histological analyses of the spinal segments. The three-year follow-up study demonstrated a significantly faster and more complete fusion using PLLA cages as compared to titanium cages of the same dimensions. The PLLA group showed unchanged intervertebral grafting height and progressive replacement of PLLA by trabecular bone. A complete bone remodeling within the cage was achieved after two years and degradation of PLLA with mild tissue reaction was completed at three years after implantation.133
Canine models for biological treatment approaches of DDD
IVD degeneration in canines is a prevalent disorder. Diseases in canines that are associated with disc degeneration are among the major reasons for euthanasia of canines younger than 10 years old. Disc degeneration-associated diseases in canines include disc herniation, degenerative lumbosacral stenosis (DLSS), and cervical spondylomyelopathy. Several similarities have been shown between IVD degenerations in humans and in chondrodystrophoid breed canines: Similar to that in humans, degeneration of IVD with or without clinical signs of disease is prevalent in chondrodystrophoid canines, and it is more common in older canines than in younger onces; chondrodystrophoid canines exhibit loss of notochordal disc cells at the early young age and suffer spontaneous disc degeneration with reduction in proteoglycan content, gross pathology, and chronic low back pain as humans. Thus, typically chondrodystrophoid canines have been used to study the processes of IVD degeneration and to prove regenerative effects of therapeutic approaches.134,135
Intradiscal biological therapy approaches in canine models of disc degeneration
Autologous or allogeneic cells isolated from bone marrow (MSCs) and from NP tissue (NPCs), or from human umbilical cord matrix (WJCs: Wharton’s jelly cells) were applied in degenerative discs of canines. Moreover, supporting biomaterials without any supplement or in combination with cells or bioactive factors have been intradiscally applied136–147 (Table 9).
Table 9.
Biological approaches of disc repair in canine models of disc degeneration.
| Animal Model | Transplanted cell/Gene modification | Implanted biomaterial | Disc regeneration effect | Ref. |
|---|---|---|---|---|
| Dog: Lumbar disc NP aspiration | Autologous MSC/RV-GFP (1 × 105, 106, 107 cell/disc) | Well preserved disc height and ECM structure, but highest cell survival rate at 1 × 106 MSCs | 136 | |
| Dog: Lumbar disc natural degenerated | Autologous MSC | TGF-β1 crosslinked collagen µ-carriers | Marked clinical improvement, without restoration of disc tissue | 137 |
| Dog: Lumbar disc nucleotomy or natural degenerated | Allogeneic MSC | NCM: Notochordal cell-derived matrix of healthy porcine | NCM improved disc height, MRI index, collagen II level, and ameliorated local inflammation | 138 |
| Dog: Lumbar disc NP aspiration | Autologous normal or cryopreserved NPC /AdV-GFP, activated with MSCs | Improved MRI signal intensity, disc height, enhanced levels of proteoglycan, collagen II with normal, or cryopreserved NPCs | 139 | |
| Dog: Lumbar disc nucleotomy | Allogeneic NPC, PHK-26 marked | PLGA scaffold | Improved disc height, MRI index, and segmental stability | 140 |
| Dog: Lumbar disc NP aspiration | Wharton's jelly cells (WJCs/AAV2-GFP) | Improved disc tissue structure, disc height, stability, MRI grade | 141 | |
| Dog: Lumbar disc natural degenerated | PEAMs: Polyester amide microspheres | Good compatibility for safe and sustained release of therapeutics | 142 | |
| Dog: Lumbar disc NP aspiration | PEAMs loaded with celecoxib (CXB) | Decreased PGE2 level and NGF immunopositivity with boosted proteoglycan level in NP | 143 | |
| Dog: Lumbar disc nucleotomy | PEAMs loaded with triamcinolone-acetonide (TAA) | Lower NGF immunopositivity, but absence of beneficial effects on disc tissue regeneration | 144 | |
| Dog: Lumbar disc natural degenerated | PCLA-PEG-PCLA hydrogel loaded by celecoxib (CXB) | Clinical improvement with recurred or reduced back pain, but no disc tissue regeneration | 145 | |
| Dog: Lumbar disc natural degenerated | rhBMP-7: dose/disc 2.5, 25, and 250 μg | No regenerative effect at all and wide extradiscal bone formation | 146 | |
| Dog: Lumbar disc discectomy | γ-irradiated disc allografts (18, 25 or 50 kGy) | Dose-dependent allografts decellularization with increased segmental degeneration | 147 |
MSC: mesenchymal stem cell; RV: retroviral vector; GFP: green fluorescent protein; NPC: nucleus pulposus cell; AdV: adenoviral vector; WJC: Wharton’s jelly cell; AAV2: adeno-associated virus vector serotype 2; PLGA: poly L-lactic-co-glycolic acid; PEAMs: polyester amide microspheres; TAA: triamcinolone-acetonide; PCLA-PEG-PCLA: poly(ε-caprolactone-co-lactide)-b-poly(ethyleneglycol)-b-poly(ε-caprolactone-co-lactide); CXB: celecoxib; ECM: extracellular matrix; MRI: magnetic resonance imaging; PGE2: prostaglandin E2; NGF: nerve growth factor.
Treatment approaches include intradiscal application of cells, bioactive factors, biomaterial, or a combination thereof.
Autologous MSCs of different concentrations (1 × 105, 106, or 107 per disc) were transplanted into degenerative discs of beagle dogs to determine the optimal donor cell number for maximum benefit of cell transplantation. At four weeks after induction of lumbar disc degeneration (L3–L6) by aspiration of NP tissue (15 mg NP), the different amount of MSCs that were transduced with RV-GFP were transplanted into the injured discs in a randomized sequence. Radiographs and MRIs were taken at baseline, 4, 8, and 12 weeks after initial surgery that was followed by gross anatomical, histological, and apoptotical assessments at 12 weeks. As compared with injured discs without cell transplantation, discs transplanted with MSCs exhibited better preservation of the disc height and anular structure. However, higher number of remaining transplanted MSC cells and higher survival rate of NPCs with ECM maintenance were found in discs transplanted with 1 × 106 MSCs. Less viable cells were detected in discs transplanted with 1 × 105 MSCs, and more apoptotic cells in discs transplanted with 1 × 107 MSCs.136 Furthermore, the suitability of intradiscal application of autologous MSCs was proved in a canine model of disc degeneration that recapitulates the spontaneous natural IVD degeneration occurring in humans. The study involved client-owned dogs with clinical signs of low back pain and MRI findings that confirmed the diagnosis of DLSS caused by a degenerated and centrally protruded L7–S1 disc. Dog breeds included: Belgian Malinois, Labrador Retriever, German Shepherd, Berger Blanc Suisse, Rhodesian Ridgeback, German Longhair, and Dalmatian. The dogs underwent decompressive spinal surgery and received an intradiscal injection of autologous bone marrow-derived MSCs that were attached on collagen microcarriers crosslinked with TGF-β1 (3 × 106 MSCs/250 mg collagen microcarriers plus100 ng/mL of TGF-β1) or without TGF-β1 crosslinking in 500 µL PBS suspension, along with a control suspension of microcarriers alone. Clinical performance and Pfirrmann grading of MRI were completed at 10 months after injection. Although marked clinical improvement was observed in all groups with clinical functioning restored back to normality, neither microcarriers nor MSCs could restore the structure of degenerated discs. Postoperative Pfirrmann grade remained equivalent in all dogs, and formation of Schmorl’s nodes was detected in 11% of dogs.137 In addition, allogeneic MSCs in combination with notochordal cell-derived matrix (NCM) of healthy porcine were used in two beagle dog models of disc degeneration: (a) a spontaneous natural degeneration model (mild degeneration) and (b) a neuclotomy induced degeneration model (moderate degeneration). Five lumbar discs (L1–L6) were injected with NCM, MSCs, NCM-MSCs, or left uninjected at six weeks after neuclotomy; whereas two NCM-treated IVDs per dog received an additional NCM injection at three months after the first injection. Radiological, macroscopical, histological, and biochemical analyses were performed at six months after the first injection to evaluate the effects of treatments on mildly and moderately degenerated discs. Injection of NCM in moderately (induced) degenerated discs stimulated collagen type II rich ECM production, improved the disc height, MRI signal intensity, and ameliorated local inflammation. But injection of MSCs exerted no (additive) effects.138 Moreover, activated normal NPCs and cryopreserved NPCs have been used in a dog model of disc degeneration to compare their disc regeneration effect after transplantation. If NPCs that are cryopreserved show regeneration ability, then autologous cell transplantation could be offered to patients as and when required. Activated NPCs were obtained by coculturing of NPCs and bone marrow MSCs using the indirect cell-to-cell contact method. Disc degeneration (L2–L5) was induced by NP aspiration (15 mg NP) and bone marrow blood was aspirated at the same time from the iliac crest to isolate MSCs. Isolated NPCs that were cryopreserved for two weeks and normal NPCs were activated by coculturing with MSCs and transduced with AdV vectors encoding for GFP (AdV-GFP). At three weeks after the first surgery, cells were transplanted into one of the injured discs (L2–L5; 1 × 106 cells/100 µL of medium), where L5/L6 disc served as uninjured control. Radiological, MRI, gross anatomical, histological, and immunohistochemical examinations performed up to 12 weeks after initial surgery showed a similar attenuation of IVD degeneration in discs transplanted with cryopreserved or normal NPCs. Disc height, MRI signal intensity, and contents of proteoglycan and collagen II were significantly improved with improved histological findings in both cell transplantation groups as compared with injured untreated group.139 Additionally, expanded and PHK-26 marked allogeneic NPCs were transplanted in combination with a three-dimensional porous PLGA scaffold into lumbar discs of beagle dogs after nucleotomy. Directly following nucleotomy injured discs were randomly injected either with NPC-PLGA composite (1 × 106 cells/100 µL) or with PLGA scaffold alone (100 µL) or left untreated. The regenerative effect of the composite was evaluated up to eight weeks after injection using X-rays and MRIs. Discs treated with the NPC-PLGA composite showed well-preserved disc height, segmental stability, and improved MRI signal intensity as compared with discs treated with PLGA alone or left untreated. Moreover, PHK-26-marked NPCs were found within the area of the NP up to eight weeks after treatment.140 Furthermore, the survival and regenerative ability of WJCs were examined after intradiscal transplantation in beagles. At four weeks after injury operation (aspiration of NP) of the lumbar discs (L4–L7), the L6/L7 discs were injected with WJCs (1 × 106 cells/100 µL PBS) that were transduced with AAV2 vectors encoding for GFP (AAV2-GFP), while L5/L6 discs were injected with 100 µL PBS, L4/L5 discs were left untreated and L3/L4 intact discs served as control. Radiological and MRI analyses were performed at baseline, 4, 8, 12, and 24 weeks after the initial surgery that were followed by biomechanical, histological, immunohistochemical, macroscopical, and cell survival analyses at the 24th week. As compared with untreated or PBS-treated discs, WJCs-treated discs showed upregulation of aggrecan, collagen II, and SOX-9 with significantly smaller reduction of disc height and MRI signal intensity. Improved spinal segmental stability, preserved disc tissue structure, and enhanced survival of transplanted cells were confirmed up to 24 weeks.141
Biomaterials that have a good cytocompatibility and biocompatibility are considered as safe and sustained release systems for intradiscal delivery of bioactive factors that support disc regeneration or pain relief. Polyester amide microspheres (PEAMs), that were tested for safety of intradiscal injection and biocompatibility at long-term (24 weeks) follow-up in a canine model predisposed to spontaneous natural disc degeneration, did not induce degenerative changes in T12–L5 discs of beagle dogs after injection of 40 µL PEAMs. The results determined by MRI, macroscopical, histopathological, and biochemical methods showed a good cytocompatibility and biocompatibility of PEAMs, suggesting them as safe and sustained release system for intradiscal delivery of bioactive factors.142 Accordingly, PEAMs were used for intradiscal delivery and controlled release of celecoxib (CXB), the nonsteroidal anti-inflammatory cyclooxygenase-2 (COX-2) inhibitor. At four weeks after needle aspiration of the NP at five alternating levels (T12–T13, L1–L2, L3–L4, L5–L6, and L7–S1), CXB-loaded PEAMs or unloaded PEAMs were injected in a random fashion: 40 μL of low dose CXB-PEAMs (0.72 mg/mL particles plus 8.4 μg CXB) or 40 μL high dose CXB-PEAMs (72 mg/mL particles plus 280 μg CXB) both as duplicates, or 40 μL of PEAMs without CXB (70 mg/mL in PBS). At 12 weeks after treatment MRI, CT, macroscopical, histopathological, immunohistochemical, and biochemical evaluations were performed. The results showed no evidence of adverse effects by intradiscal and sustained delivery of CXB. Quite the contrary, the local and sustained delivery of CXB prevented progression of IVD degeneration by enhancing the proteoglycan content and decreasing the PGE2 level and NGF immunopositivity in NP. The results provided evidence that intradiscal delivery of CXB could address the pain-related IVD degeneration.143 Likewise, PEAMs were used for intradiscal delivery and controlled release of triamcinolone acetonide (TAA), an anti-inflammatory corticosteroid. The purpose was to achieve a prolonged release of TAA with pain relief and to circumvent adverse effects usually caused by high dosages of corticosteroids, which are used to relieve symptoms of chronic low back pain. At four weeks after nucleotomy that was performed at five levels (T12–T13, L1–L2, L3–L4, L5–L6, and L7–S1), randomized lumbar discs were injected with 40 µL of low dose TAA-PEAMs (0.72 mg/mL particles; 8.4 µg TAA) or 40 µL of high dose TAA-PEAMs (72 mg/mL particles; 0.84 mg TAA) both as duplicates, or 40 µL of PEAMs alone (70 mg/mL in PBS). All adjacent non-nucleotomized noninjected lumbar discs were included as controls. At 12 weeks after treatment, discs were evaluated using MRI, CT, histopathological, immunohistochemical, and biochemical methods. The results showed that the low dosage of TAA-PEAMs with sustained intradiscal release of TAA significantly reduced NGF immunopositivity in degenerated NP tissue, suggesting its potential applicability for pain relief. However, discs treated with TAA-PEAMs did not show any beneficial effect on tissue regeneration. They showed no effect on disc height, MRI index, DNA content, and levels of prostaglandin E2 (PGE2), collagen, and GAG.144 In addition, the COX-2 inhibitor (CXB) was also used in combination with a thermoresponsive PCLA-PEG-PCLA hydrogel (poly(ε-caprolactone-co-lactide)-b-poly(ethylene glycol)-b-poly(ε-caprolactone-co-lactide hydrogel) in a canine model of spontaneous natural IVD degeneration with chronic low back pain. Since previously IVD samples that were surgically obtained from canine patients showed increased COX-2 expression that specified the role of COX-2 in clinical IVD disease, patient dogs were selected to receive an intradiscal injection of CXB-loaded hydrogel (PCLA-PEG-PCLA). The long-term follow-up (24 weeks) after intradiscal injection showed no adverse reactions and no influence on MRI signal intensity. However, clinical examinations and owner questionnaires discovered that the back pain was recurred after three months in 3 of 10 dogs, and a reduction of back pain was achieved with clinical improvement in 9 of 10 dogs.145 In another long-term follow-up (24 weeks) study, the regenerative effect of human bone morphogenetic protein-7 (rhBMP-7) was examined in a canine model of spontaneous natural disc degeneration (T13–L6) at three different dosages (2.5, 25, and 250 μg administered in a 40 µL injection volume per disc). MRI scans were completed in the course of the 24 weeks that were followed by postmortem evaluations of IVD tissues using radiographical, histological, and biochemical methods. The results exhibited no regenerative effect in all discs treated with different dosages of rhBMP-7. On the contrary, treatments with rhBMP-7 resulted in extensive extradiscal bone formation, suggesting that injection of rhBMP-7 should not be used for treatment of IVD degeneration.146
Moreover, a canine model of spontaneous IVD degeneration was used to explore the regenerative potential of decellularized disc allografts treated by 6°Co gamma irradiation for biological treatment of DDD. Beagle dogs received single-level allografted disc treated with different doses of gamma irradiation (irradiated with 18, 25 or 50-kGy doses and 0-kGy control group with no irradiation), where the cell viability and biomechanical properties of the allograft discs were previously in vitro determined among the dosage groups. After random removal of a disc between L2 and L6 levels, a disc allograft of the most compatible size was selected, trimmed according to the transplanted segmental space and positioned into the slot of the excised disc. Radiologic assessments were performed before the transplantation and at baseline, one, three, and six months after transplantation that were followed by biomechanical testing, macroscopical, and morphological evaluations at six months after operation. There were significant differences of cell viabilities among the various dosage groups, but there were no obvious differences of the biomechanical properties among the various dosage groups. The disc height and ROM decreased significantly in all groups as time went on. The disc height index, ROM, hydration status as well as segmental regeneration in irradiated groups were inferior to those in the control group, but they were better in 18-kGy group than those in 25 and 50-kGy groups.147
Non-human primate models for biological treatment approaches of DDD
Non-human primates are the closest animal models to humans regarding anatomy, genetics, physiology, and behavior, but due to ethical restriction they have been more rarely used for therapeutic studies of DDDs. Although they are quadrupedal for locomotion, they spend plenty of time in semi-upright or upright positions and their discs are more exposed to mechanical stresses similarly to human discs. Although the methods to accelerate progressions of disc degeneration are established in non-human primate models such as rhesus macaques and baboons, regenerative approaches of DDD have been sparsely attempted.148–156
In vivo regenerative approaches in non-human primate models of disc degeneration
Intradiscal gene therapeutic approach mediated by AAV2 encoding for CTGF and TIMP1 as well as applications of biocompatible or biodegradable scaffold, such as fresh frozen disc allograft, PVA hydrogels, and HA-coated porous ingrowth surfaces, have been tried in degenerative discs of rhesus macaques and baboons148–152 (Table 10). A gene therapeutic approach of degenerative discs was attempted in rhesus macaques by intradiscal injection AAV2 vectors encoding for CTGF and TIMP1 genes (AAV2-CTGF-TIMP1). At 12 weeks after induction of lumbar disc degeneration (L1–L6) by annular needle puncture, where the success degeneration was proved by MRI at 4, 8, and 12 weeks after surgery, the L1–L4 discs were injected with 50 μL of AAV2-CTGF-TIMP1 (1 × 1012 pfu/L) as gene therapy group, while the L4–L6 discs were assigned as negative control group and injected with 50 μL PBS alone. The L6–L7 and L7–S1 discs that received neither needle puncture nor treatment were assigned as blank control group. At 8, 16, and 24 weeks after injection animals were euthanized to acquire the lumbar discs (L1–S1) and evaluate them using histological, immunohistochemical, and biochemical methods. An intradiscal injection of AAV2-CTGF-TIMP1 could enhance the mRNA expression levels of CTGF and TIMP1with higher synthesis of collagen II and aggrecan in NP tissue, indicating the regenerative effects of AAV2-CTGF-TIMP1 in degenerated lumbar discs of rhesus macaques.148 Furthermore, rhesus macaques were used for examining the possibility of using fresh frozen IVD allograft in disc transplantation. Examinations were associated with the allograft survival, its maintenance of disc tissue metabolism, segmental mobility, and problems of rejection in short- and long-term follow-up after transplantation. Lumbar discs of donor monkeys were isolated and directly fresh deep-frozen. The fresh frozen allografts were randomly divided into a short-term group (followed up for 2, 4, 6, and 8 weeks, respectively), a midterm group (6 months) and a long-term group (24 months). Lumbar disc (L5/L6) replacement in recipient monkeys with a complete discectomy was performed, and after disc replacement the changes in fresh frozen disc allografts were evaluated using radiographical, histological, biochemical, and biomechanical methods. The allografts were able to survive through a deep freezing protocol and maintain cell viability after transplantation without significant immunoreaction. The biomechanical properties of stability and mobility of the transplanted allografts were similar to those of the controls. However, the radiographical and histological examinations showed severe disc degeneration at 24 months of follow-up with disc height decrease mainly in the early postoperative stage. Similarly, decrease in hydration as well as in proteoglycan and hydroxyproline contents of the allografts were confirmed at 6 and 24 months of follow-up. Taken together, the allografts were able to maintain stability and mobility of the functional spinal unit, but degeneration of the allograft was inevitable.149 Moreover, baboons have been used for preclinical evaluation of a PVA hydrogel implant as a replacement for a diseased NP. A single-level discectomy was performed at lumbar discs (L3/L4 or L4/L5) with either cup forceps (2.5 mm tip diameter) or IVD rongeurs with a rectangular cross section (maximum dimension 6.0 mm) and the nuclear cavity was left empty. A hydrogel implant was injected into the nuclear cavity by means of a custom insertion device (PVA treatment group), and then animals were housed individually in indoor cages for the first month (4 weeks) and returned to gang cages for the rest of the study period (23 months). Follow-up evaluations over 24 months after surgery included: radiographical analyses (before surgery, immediately after surgery and at the time of euthanasia), MRI (at 2 weeks after surgery and at 1–3 days before euthanasia) as well as postmortem gross pathological and histopathological analyses of both local and remote tissues. The PVA implants were well tolerated over 24 months with no evidence of implant-related pathology in the adjacent disc tissue, spinal cord, or remote tissues. However, additional studies are needed to verify the efficacy of the implant in its intended application as a prosthetic replacement for the diseased NP.150 In addition, baboons were used for an in vivo comparison of two different types of unconstrained disc prostheses owing alternate in-growth surfaces for total disc replacement arthroplasty: (a) the AcroFlex device (consisted of sintered titanium beaded HA-coated ingrowth surfaces, bound together by a hexene-based polyolefin rubber core) and (b) the SB Charitè device (consisted of prosthetic VEPs from cobalt chrome, covered by two layers of thin titanium with an electrochemically bonded HA coating and an ultra-high molecular weight polyethylene core). After a complete discectomy of L5/L6 lumbar discs (AF/NP) and burr decortication of the VEPs, one of the implant devices per animal was inserted using a customized distractor and implant position was verified with anteroposterior and lateral fluoroscopic images. Following six-month survival periods the implants were evaluated using multidirectional flexibility testing, radiographical, histological, and immunohistological methods. Compared to the AcroFlex treatments, the SB Charitè treatments showed greater ROM under axial compression, flexion-extension, and lateral bending, but it exhibited similar ROM to the intact non-surgical controls. No loosening of any metallic prosthetic VEP was seen and excellent ingrowth at the level of the implant–bone interface was demonstrated on both AcroFlex and SB Charité prosthesis. Moreover, no local and systemic accumulation of particulate wear debris (titanium, polyethylene or cobalt chrome) or cytokines (TNFα, PGE2, IL-1, IL-2 or IL-6) were detected. The porous ingrowth calculations showed the mean ingrowth (linear apposition) ranging from 47.9 ± 9.12% for the SB Charitè device and 54.59 ± 13.24% for the AcroFlex device, which is more favorable for total disc replacement as compared to that affirmed for cementless total joint components in the appendicular skeleton (range 10–30%).151,152 Above and beyond, baboons were used for in vivo testing of a technique that was able to sensitively monitor dynamic changes in strain and load during extreme activity. The technique was suggested to provide key biomechanical information that could be essential for designing of suitable implants. For that an intradiscal implant-load-cell connected to a telemetry transmitter was surgically placed in L4/L5 disc space of baboons to measure real-time in vivo loads that were imposed on the lumbar spine. Implants placed into the disc space and the transmitter in the flank allowed the collection of load data from the animals during activities.153
Table 10.
Intradiscal regenerative approaches in non-human primate models.
| Animal Model | Implanted material | Disc regeneration effect | Ref. |
|---|---|---|---|
| Rhesus macaque: Lumbar disc puncture injury | AAV2-CTGF-TIMP1 | Enhanced expression levels of CTGF and TIMP1 with increased synthesis of collagen II and aggrecan in NP tissue | 148 |
| Rhesus macaque: Lumbar disc complete discectomy | Fresh frozen IVD allograft | Maintained stability and mobility of the spinal unit with high cell viability, but severe allograft degeneration was certain in long-term follow-up | 149 |
| Baboon: Lumbar disc complete nucleotomy | PVA hydrogel implant | No implant-related pathology in the adjacent disc tissue, spinal cord, or remote tissues, but the ability as prosthetic NP replacement not proved | 150 |
| Baboon: Lumbar disc complete discectomy with EP decortication | AcroFlex and SB Charitè implants with HA-coated porous ingrowth surfaces | Excellent ingrowth at the level of the implant-bone interface, with no accumulation of wear debris or inflammatory cytokines | 151,152 |
AAV2: adeno-associated virus vector serotype 2; CTGF: connective tissue growth factor; TIMP1: metalloproteinase inhibitor 1; IVD: intervertebral disc; PVA: polyvinyl alcohol; HA: hydroxyapatite; NP: nucleus pulposus.
Treatment approaches include applications of recombinant AAV2, fresh frozen disc allograft, PVA hydrogel implant, and hydroxyapatite coated porous ingrowth surfaces on disc prostheses.
Discussions
For choosing appropriate animal models of disc degeneration and regeneration important points should be considered that might reflect the clinical facts of DDD in humans. Considerations for choosing an appropriate animal model include for example: the intradiscal deficiency of notochordal cells, the size of the animal and its IVD dimension relative to humans, the biomechanical stress exerting on the IVDs, and ethical concerns. Moreover, the significance of spontaneous IVD degeneration (example: aging or genetic predisposition) in comparison to mechanistical induced IVD degeneration (chemical, mechanical or surgical induced injuries) must be well thought out during the design of research projects involving animal models.
Notochordal cell loss in IVD is thought to precede the onset of IVD degeneration in humans, although the vital role of notochordal cells in initiating IVD degeneration it is still uncertain and their function in healthy discs during childhood are not yet clearly understood. Nonetheless, IVD degeneration that is characterized by changes in the mechanical integrity and biochemical composition in discs, especially in NP, is thought to be associated with the disappearance of notochordal cells in adult IVDs. In some animal species, notochordal cells steadily disappear during aging alike in humans, while in other species they persist for a long period of time after maturity or throughout their matured life. Different animal models of IVD degeneration, which differ significantly in the status of notochordal cell disappearance, have been used for regenerative approaches of DDD. Based on the literature, matured chondrodystrophic dogs, goats, sheeps, and non-human primates are the most suitable species regarding the cell populations mimicking those found in adult humans. Nevertheless, animal models that retain their notochordal cells could also be beneficial to understand the cell biology during development, maturation, degeneration, and maintenance of IVDs.67,157–158
Initiations of IVD degeneration in most animal models have been achieved using chemical, mechanical, or surgical interventions, whereas in some animals it occurs spontaneously. The success of regenerative treatment approaches could significantly depend on the standardization and replicability of the degeneration (intervention) mechanisms along with the severity and duration of the degeneration. Regarding spontaneous IVD degeneration in animals, the chondrodystrophoid canine,134,159 non-human primates160,161 along with genetically altered rabbits,162,163 mice52,164,165, and sand rats166,167 are well described. However, the use of spontaneous models of disc degeneration is often limited by the long time course of degeneration and unanticipated progression of degeneration, which is combined with the high ethical consideration of using non-human primates and canine.
Different chemicals, for example chymopapain, collagenase, CABC, and Fn-f, have been used to induce the pathophysiological process of disc degeneration. Chemical agents can induce IVD degeneration by promoting the breakdown of GAG chains of proteoglycans that impair structural integrity of the IVDs. However, some chemical agents like CABC can affect the viability of disc cells, which in turn limits the restoration of the disc ECM.88,168–171
Mechanical models of disc degeneration are more suitable to ensure the benefit of introducing highly replicable degenerative cascade at a defined time point. Mechanical models of disc degeneration include the applications of static and cyclical compression or bending. They are mostly applied on rodent tails by using external fixation device (Ilizarov-type apparatus), which allows the application of mechanical force across the tail IVDs, or by attachment of an external loading device that allows the application of compressive force on the lumbar spine of rats or rabbits.55,65,172–176
The most commonly used and well-established methods for initiation of IVD degeneration are surgical interventions that introduce injuries to AF or NP. Partial thickness anular injuries are mainly consisting of a smaller incision depth (≤5 mm) or a smaller size anular injury that do not damage the inner AF and NP at the time of injury, and they persuade slower degenerative processes. Conversely, full thickness anular injuries are consisting of annular incisions accompanied by partial removal of AF and NP tissues, and they induce NP avulsion with hastier degeneration. However, great care must be taken to create a highly replicable disc injury.121,177–180
Initiation and progression IVD degeneration in animal models that can be confirmed by radiological, histological, biochemical, and biomechanical findings are essential to comprehend the process of disc degeneration on the bases of particular provocations. Although such findings are crucial markers of disc degeneration, their relation to pain is not yet well established in most animal models, which may affect their relevance for clinical translation. Pain assessments have been performed mainly in rodent models of disc degeneration. These include for example an intensified grooming, wet-dog-shake, declined locomotor capability, declined rotarod performance, sensitivity to thermal or mechanical stimuli (hind paw mechanical sensitivity) and declined general condition of the animals.177,181–184 Pain assessments, especially in large animal models of disc degeneration, could significantly improve our current knowledge about the progression and therapy of DDD.
Intradiscal biological treatment approaches have been tried in different animal models of disc degeneration. The animal models used to test new biological therapeutic approaches include mice, rats, rabbits, porcine, ovine, caprine, canine, rhesus macaques, and baboons, which had developed disc degeneration either spontaneously or after chemical, mechanical, or surgical interventions. The animal experiments included: cell transplantation approaches with or without gene modification, in situ gene therapeutical approaches, intradiscal applications of bioactive factors and bioscaffolds or a combination thereof. These preclinical treatment approaches and their effects of disc regeneration in different animal models are promising (Tables 1 to 10) and may help to formulate regenerative therapy strategies for clinical applications. Different types of autologous, allogeneic, or human derived cells were intradiscally tested. These included: chondrocytes (NP and AF cells), bone marrow-derived MSCs, bone marrow-derived mesenchymal precursor cells, ADSCs, iPSCs, ESCs, multipotent stem cell from the olfactory mucosa, and umbilical cord tissue-derived MSCs. The intradiscal applications of bioactive factors included: cytokines, antibodies, kinases, shRNAs, exosomes, lignans, and growth factors. Regenerative in situ gene therapy approaches included: retroviral, LV, AdV, and AAV vectors carrying candidate therapeutic genes. However, safety assessments, especially during in situ gene therapy approaches with certain viral constructs, are essential, because misdirected injection, incorrect dose, and immune reactions near to sensitive neural structure could provoke toxicity and immunological side-effects with neurological deficits.
IVD degeneration in human is a complex and multifactorial process involving for example cellular senescence, mechanical stresses, nutritional deprivation, and genetical predisposition. The complex and multifactorial process indicates that no animal model is able to profoundly reproduce the clinical condition in humans. Nevertheless, progresses have been achieved from biological treatment approaches in animal models to clinical trials.185–188 For example, transplantation of chondrocytes, harvested from damaged cartilaginous tissues and expanded in culture under controlled and defined conditions, was performed in a controlled, randomized and multicenter study (EuroDISC), which compared the safety and efficacy of autologous disc chondrocyte transplantation in patients undergoing single level discectomy. The interim analysis of 28 patients that was performed after two years could show improved long-term pain relief, maintained disc height, and enhanced hydration, as compared to discs that had undergone discectomy without cell transplantation.185 Furthermore, in two small noncontrolled clinical trials expanded autologous bone marrow-derived MSCs were percutaneously injected in 12 patients, which had low back pain of DDD and unsuccessful conservative treatment. At two years after transplantation, the clinical studies showed similar results: improved long-term pain relief, maintained disc height, and enhanced hydration.186,187 Moreover, a multicenter, randomized, and controlled trial of a phase II study was performed with allogeneic mesenchymal precursor cells (MPCs). The trial included 100 patients with moderate to severe low back pain that persisted for more than six months and caused by early disc degeneration. The results after 12 months showed that intradiscal injection of allogeneic MPCs was well tolerated and reduced back pain with improved disc functions. In addition, it reduced opioid use and the need for additional interventions.188
Conclusions
This review article provides a detailed and an up-to-date overview of biological treatment strategies in all small and large animal models used thus far in regenerative research of degenerative discs. Biological treatment strategies include applications of cell transplantation, gene therapy, bioscaffolds, and bioactive factors, or a combination thereof, which may able to address options of restoring the structural or biological deterioration of degenerative discs. However, it is limited to in vivo and intradiscal biological treatment strategies in research animal models and does not include studies based on non-intradiscal, non-biological, or in vitro strategies. In conclusion, although no animal model is able to profoundly reproduce the clinical conditions in humans, the various animal models have played important roles in specifying our knowledge about the pathophysiology of DDD. They are crucial for developing new and hopeful therapy approaches for clinical applications.
Authors’ contributions
All authors participated in the design, interpretation of the studies, and analysis of the data and review of the manuscript. DSM did the conception and design of the article, DSM, TW, and AB performed the acquisition of data, and DSM, TW, AB, and CT completed the analysis and interpretation of data, revision, and final approval of the article.
Footnotes
Declaration OF CONFLICTING INTERESTS: The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.
Funding: The author(s) received no financial support for the research, authorship, and/or publication of this article.
ORCID iD: Demissew Shenegelegn Mern https://orcid.org/0000-0001-5893-981X
References
- 1.Roughley PJ. Biology of intervertebral disc aging and degeneration: involvement of the extracellular matrix. Spine 2004; 29:2691–9 [DOI] [PubMed] [Google Scholar]
- 2.Adams MA, Dolan P. Spine biomechanics. J Biomech 2005; 38:1972–83 [DOI] [PubMed] [Google Scholar]
- 3.Adams MA, Roughley PJ. What is intervertebral disc degeneration, and what causes it? Spine 2006; 31:2151–61 [DOI] [PubMed] [Google Scholar]
- 4.Sivakamasundari V, Lufkin T. Stemming the degeneration: IVD stem cells and stem cell regenerative therapy for degenerative disc disease. Adv Stem Cells 2013; 2013:724547. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Oehme D, Goldschlager T, Ghosh P, Rosenfeld JV, Jenkin G. Cell-based therapies used to treat lumbar degenerative disc disease: a systematic review of animal studies and human clinical trials. Stem Cells Int 2015; 2015:946031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Roberts S, Evans H, Trivedi J, Menage J. Histology and pathology of the human intervertebral disc. J Bone Joint Surg Am 2006; 88(Suppl 2):10–4 [DOI] [PubMed] [Google Scholar]
- 7.Melrose J, Smith SM, Little CB, Moore RJ, Vernon-Roberts B, Fraser RD. Recent advances in annular pathobiology provide insights into rim-lesion mediated intervertebral disc degeneration and potential new approaches to annular repair strategies. Eur Spine J 2008; 17:1131–48 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Roberts S, Eisenstein SM, Menage J, Evans EH, Ashton IK. Mechanoreceptors in intervertebral discs. Morphology, distribution, and neuropeptides. Spine 1995; 20:2645–51 [DOI] [PubMed] [Google Scholar]
- 9.Urban JPG, Roberts S. Degeneration of the intervertebral disc. Arthritis Res Ther 2003; 5:120–30 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Urban JP, Holm S, Maroudas A, Nachemson A. Nutrition of the intervertebral disk. An in vivo study of solute transport. Clin Orthop Relat Res 1977; 129:101–14 [PubMed] [Google Scholar]
- 11.Benneker LM, Heini PF, Alini M, Anderson SE, Ito K. Young investigator award winner: vertebral endplate marrow contact channel occlusions and intervertebral disc degeneration. Spine 2004; 30:167–73 [DOI] [PubMed] [Google Scholar]
- 12.Battié MC, Videman T, Kaprio J, Gibbons LE, Gill K, Manninen H, Saarela J, Peltonen L. The twin spine study: contributions to a changing view of disc degeneration. Spine J 2009; 9:47–59 [DOI] [PubMed] [Google Scholar]
- 13.Kalb S, Martirosyan NL, Kalani MYS, Broc GG, Theodore N. Genetics of the gegenerated intervertebral disc. World Neurosurg 2012; 77:491–501 [DOI] [PubMed] [Google Scholar]
- 14.Setton LA, Chen J. Mechanobiology of the intervertebral disc and relevance to disc degeneration. J Bone Joint Surg Am 2006; 88(Suppl 2):52–7 [DOI] [PubMed] [Google Scholar]
- 15.Videman T, Sarna S, Battié MC, Koskinen S, Gill K, Paananen H, Gibbons L. The long-term effects of physical loading and exercise lifestyles on back-related symptoms, disability and spinal pathology among men. Spine 1995; 20:699–709 [DOI] [PubMed] [Google Scholar]
- 16.Martin BI, Deyo RA, Mirza SK, Turner JA, Comstock BA, Hollingworth W, Sullivan SD. Expenditures and health status among adults with back and neck problems. JAMA 2008; 299:656–64 [DOI] [PubMed] [Google Scholar]
- 17.Walker BF. The prevalence of low back pain: a systematic review of the literature from 1966 to 1998. J Spinal Disord 2000; 13:205–17 [DOI] [PubMed] [Google Scholar]
- 18.Stewart WF, Ricci JA, Chee E, Morganstein D, Lipton R. Lost productive time and cost due to common pain conditions in the US workforce. JAMA 2003; 290:2443–54 [DOI] [PubMed] [Google Scholar]
- 19.Freemont AJ, Peacock TE, Goupille P, Hoyland JA, O'Brien J, Jayson MI. Nerve ingrowth into diseased intervertebral disc in chronic back pain. Lancet 1997; 350:178–81 [DOI] [PubMed] [Google Scholar]
- 20.Peng B, Wu W, Hou S, Li P, Zhang C, Yang Y. The pathogenesis of discogenic low back pain. J Bone Joint Surg Br 2005; 87:62–7 [PubMed] [Google Scholar]
- 21.Taher F, Essig D, Lebl DR, Hughes AP, Sama AA, Cammisa FP, Girardi FP. Lumbar degenerative disc disease: current and future concepts of diagnosis and management. Adv Orthop 2012; 2012:970752. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Van Uden S, Silva-Correia J, Oliveira JM, Reis RL. Current strategies for treatment of intervertebral disc degeneration: substitution and regeneration possibilities. Biomater Res 2017; 21:22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Kandel R, Roberts S, Urban JPG. Tissue engineering and the intervertebral disc: the challenges. Eur Spine J 2008; 17(Suppl 4):480–91 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Hegewald AA, Ringe J, Sittinger M, Thomè C. Regenerative treatment strategies in spinal surgery. Front Biosci 2008; 13:1507–25 [DOI] [PubMed] [Google Scholar]
- 25.An HS, Masuda K, Cs-Szabo G, Zhang Y, Chee A, Andersson GBJ, Im HJ, Thonar EJMA, Kwon YM. Biologic repair and regeneration of the intervertebral disk. J Am Acad Orthop Surg 2011; 19:450–2 [PubMed] [Google Scholar]
- 26.Leung VYL, Tam V, Chan D, Chan BP, Cheung KMC. Tissue engineering for intervertebral disk degeneration. Orthop Clin North Am 2011; 42:575–83 [DOI] [PubMed] [Google Scholar]
- 27.Mern DS, Beierfuß A, Thomé C, Hegewald AA. Enhancing human nucleus pulposus cells for biological treatment approaches of degenerative intervertebral disc diseases: a systematic review. J Tissue Eng Regen Med 2014; 8:925–36 [DOI] [PubMed] [Google Scholar]
- 28.Millward-Sadler SJ, Costello PW, Freemont AJ, Hoyland JA. Regulation of catabolic gene expression in normal and degenerate human intervertebral disc cells: implications for the pathogenesis of intervertebral disc degeneration. Arthritis Res Ther 2009; 11:R65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Kang JD, Stefanovic-Racic M, McIntyre LA, Georgescu HI, Evans CH. Toward a biochemical understanding of human intervertebral disc degeneration and herniation. Contributions of nitric oxide, interleukins, prostaglandin E2, and matrix metalloproteinases. Spine 1997; 22:1065–73 [DOI] [PubMed] [Google Scholar]
- 30.Wang J, Markova D, Anderson DG, Zheng Z, Shapiro IM, Risbud MV. TNF-α and IL-1β promote a disintegrin-like and metalloprotease with thrombospondin type I motif-5-mediated aggrecan degradation through syndecan-4 in intervertebral disc. J Biol Chem 2011; 286:39738–49 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Le Maitre CL, Freemont AF, Hoyland JA. The role of interleukin-1 in the pathogenesis of human intervertebral disc degeneration. Arthritis Res Ther 2005; 7:R732–45 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Sedowofia KA, Tomlinson IW, Weiss JB, Hilton RC, Jayson MI. Collagenolytic enzyme systems in human intervertebral disc: their control, mechanism, and their possible role in the initiation of biomechanical failure. Spine 1982; 7:213–22 [PubMed] [Google Scholar]
- 33.Sternlicht MD, Werb Z. How matrix metalloproteinases regulate cell behavior. Annu Rev Cell Dev Biol 2001; 17:463–516 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Liu J, Roughley PJ, Mort JS. Identification of human intervertebral disc stromelysin and its involvement in matrix degradation. J Orthop Res 1991; 9:568–75 [DOI] [PubMed] [Google Scholar]
- 35.Hohaus C, Ganey TM, Minkus Y, Meisel HJ. Cell transplantation in lumbar spine disc degeneration disease. Eur Spine J 2008; 17(Suppl 4):492–503 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Sakai D. Future perspectives of cell-based therapy for intervertebral disc disease. Eur Spine J 2008; 17(Suppl 4):452–8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Le Maitre CL, Baird P, Freemont AJ, Hoyland JA. An in vitro study investigating the survival and phenotype of mesenchymal stem cells following injection into nucleus pulposus tissue. Arthritis Res Ther 2009; 11:R20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Yim RLH, Lee JTY, Bow CH, Meij B, Leung V, Cheung KMC, Vavken P, Samartzis D. A systematic review of the safety and efficacy of mesenchymal stem cells for disc degeneration: insights and future directions for regenerative therapeutics. Stem Cells Dev 2014; 23:2553–67 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Iwashina T, Mochida J, Sakai D, Yamamoto Y, Miyazaki T, Ando K, Hotta T. Feasibility of using a human nucleus pulposus cell line as a cell source in cell transplantation therapy for intervertebral disc degeneration. Spine 2006; 31:1177–86 [DOI] [PubMed] [Google Scholar]
- 40.Crevensten G, Walsh AJ, Ananthakrishnan D, Page P, Wahba GM, Lotz JC, Berven S. Intervertebral disc cell therapy for regeneration: mesenchymal stem cell implantation in rat intervertebral discs. Ann Biomed Eng 2004; 32:430–4 [DOI] [PubMed] [Google Scholar]
- 41.Allon AA, Aurouer N, Yoo BB, Liebenberg EC, Buser Z, Lotz JC. Structured coculture of stem cells and disc cells prevent disc degeneration in a rat model. Spine J 2010; 10:1089–97 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Wang W, Wang Y, Deng G, Ma J, Huang X, Yu J, Xi Y, Ye X. Transplantation of hypoxic-preconditioned bone mesenchymal stem cells retards intervertebral disc degeneration via enhancing implanted cell survival and migration in rats. Stem Cells Int 2018; 2018:7564159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Liang CZ, Li H, Tao YQ, Peng LH, Gao JQ, Wu JJ, Li FC, Hua JM, Chen QX. Dual release of dexamethasone and TGF-β3 from polymeric microspheres for stem cell matrix accumulation in a rat disc degeneration model. Acta Biomater 2013; 9:9423–33 [DOI] [PubMed] [Google Scholar]
- 44.Zhu J, Xia K, Yu W, Wang Y, Hua J, Liu B, Gong Z, Wang J, Xu A, You Z, Chen Q, Li F, Tao H, Liang C. Sustained release of GDF5 from a designed coacervate attenuates disc degeneration in a rat model. Acta Biomater 2019; 86:300–11 [DOI] [PubMed] [Google Scholar]
- 45.Xia K, Zhu J, Hua J, Gong Z, Yu C, Zhou X, Wang J, Huang X, Yu W, Li L, Gao J, Chen Q, Li F, Liang C. Intradiscal injection of induced pluripotent stem cell-derived nucleus pulposus-like cell-seeded polymeric microspheres promotes rat disc regeneration. Stem Cells Int 2019; 2019:6806540. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Murrell W, Sanford E, Anderberg L, Cavanagh B, Mackay-Sim A. Olfactory stem cells can be induced to express chondrogenic phenotype in a rat intervertebral disc injury model. Spine J 2009; 9:585–94 [DOI] [PubMed] [Google Scholar]
- 47.Wang F, Nan LP, Zhou SF, Liu Y, Wang ZY, Wang JC, Feng XM, Zhang L. Injectable hydrogel combined with nucleus pulposus-derived mesenchymal stem cells for the treatment of degenerative intervertebral disc in rats. Stem Cells Int 2019; 2019:8496025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Moriguchi Y, Borde B, Berlin C, Wipplinger C, Sloan SR, Kirnaz S, Pennicooke B, Navarro-Ramirez R, Khair T, Grunert P, Kim E, Bonassar L, Härtl R. In vivo annular repair using high-density collagen gel seeded with annulus fibrosus cells. Acta Biomater 2018; 79:230–8 [DOI] [PubMed] [Google Scholar]
- 49.Grunert P, Borde BH, Towne SB, Moriguchi Y, Hudson KD, Bonassar LJ, Härtl R. Riboflavin crosslinked high-density collagen gel for the repair of annular defects in intervertebral discs: an in vivo study. Acta Biomater 2015; 26:215–24 [DOI] [PubMed] [Google Scholar]
- 50.Grunert P, Borde BH, Hudson KD, Macielak MR, Bonassar LJ, Härtl R. Annular repair using high-density collagen gel: a rat-tail in vivo model. Spine 2014; 39:198–206 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Leo BM, Li X, Balian G, Anderson DG. In vivo bioluminescent imaging of virus-mediated gene transfer and transduced cell transplantation in the intervertebral disc. Spine 2004; 29:838–44 [DOI] [PubMed] [Google Scholar]
- 52.Marfia G, Campanella R, Navone SE, Zucca I, Scotti A, Figini M, Di Vito C, Alessandri G, Riboni L, Parati E. Potential use of human adipose mesenchymal stromal cells for intervertebral disc regeneration: a preliminary study on biglycan-deficient murine model of chronic disc degeneration. Arthritis Res Ther 2014; 16:457. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Zheng G, Pan Z, Zhan Y, Tang Q, Zheng F, Zhou Y, Wu Y, Zhou Y, Chen D, Chen J, Wang X, Gao W, Xu H, Tian N, Zhang X. TFEB protects nucleus pulposus cells against apoptosis and senescence via restoring autophagic flux. Osteoarthritis Cartilage 2019; 27:347–57 [DOI] [PubMed] [Google Scholar]
- 54.Chen Z, Zhang W, Zhang N, Zhou Y, Hu G, Xue M, Liu J, Li Y. Down-regulation of insulin-like growth factor binding protein 5 is involved in intervertebral disc degeneration via the ERK signalling pathway. J Cell Mol Med 2019; 23:6368–77 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Zhang YH, Zhao CQ, Jiang LS, Dai LY. Lentiviral shRNA silencing of CHOP inhibits apoptosis induced by cyclic stretch in rat annular cells and attenuates disc degeneration in the rats. Apoptosis 2011; 16:594–605 [DOI] [PubMed] [Google Scholar]
- 56.Chen Y, Lin J, Chen J, Huang C, Zhang Z, Wang J, Wang K, Wang X. Mfn2 is involved in intervertebral disc degeneration through autophagy modulation. Osteoarthritis Cartilage 2020; 28:363–74 [DOI] [PubMed] [Google Scholar]
- 57.Tan Y, Yao X, Dai Z, Wang Y, Lv G. Bone morphogenetic protein 2 alleviated intervertebral disc degeneration through mediating the degradation of ECM and apoptosis of nucleus pulposus cells via the PI3K/Akt pathway. Int J Mol Med 2019; 43:583–92 [DOI] [PubMed] [Google Scholar]
- 58.Liang H, Ma SY, Feng G, Shen FH, Joshua Li X. Therapeutic effects of adenovirus-mediated growth and differentiation factor-5 in a mice disc degeneration model induced by annulus needle puncture. Spine J 2010; 10:32–41 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Ge J, Yan Q, Wang Y, Cheng X, Song D, Wu C, Yu H, Yang H, Zou J. IL-10 delays the degeneration of intervertebral discs by suppressing the p38 MAPK signaling pathway. Free Radic Biol Med 2019; 147:262–70 [DOI] [PubMed] [Google Scholar]
- 60.Evashwick-Rogler TW, Lai A, Watanabe H, Salandra JM1, Winkelstein BA, Cho SK, Hecht AC, Iatridis JC. Inhibiting tumor necrosis factor-alpha at time of induced intervertebral disc injury limits long-term pain and degeneration in a rat model. JOR Spine 2018; 1:e1014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Long J, Wang X, Du X, Pan H, Wang J, Li Z, Liu H, Li X, Zheng Z. JAG2/Notch2 inhibits intervertebral disc degeneration by modulating cell proliferation, apoptosis, and extracellular matrix. Arthritis Res Ther 2019; 21:213. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Liao Z, Luo R, Li G, Song Y, Zhan S, Zhao K, Hua W, Zhang Y, Wu X, Yang C. Exosomes from mesenchymal stem cells modulate endoplasmic reticulum stress to protect against nucleus pulposus cell death and ameliorate intervertebral disc degeneration in vivo. Theranostics 2019; 9:4084–100 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Zhang H, Wang L, Park JB, Park P, Yang VC, Hollister SJ, La Marca F, Lin CY. Intradiscal injection of simvastatin retards progression of intervertebral disc degeneration induced by stab injury. Arthritis Res Ther 2009; 11:R172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Li K, Lv C. Intradiscal injection of sesamin protects from lesion-induced degeneration. Connect Tissue Res 2019; 2:1–10 [DOI] [PubMed] [Google Scholar]
- 65.Walsh AJ, Bradford DS, Lotz JC. In vivo growth factor treatment of degenerated intervertebral discs. Spine 2004; 29:156–63 [DOI] [PubMed] [Google Scholar]
- 66.Kroeber MW, Unglaub F, Wang H, Schmid C, Thomsen M, Nerlich A, Richter W. New in vivo animal model to create intervertebral disc degeneration and to investigate the effects of therapeutic strategies to stimulate disc regeneration. Spine 2002; 27:2684–90 [DOI] [PubMed] [Google Scholar]
- 67.Hunter CJ, Matyas JR, Duncan NA. Cytomorphology of notochordal and chondrocytic cells from the nucleus pulposus: a species comparison. J Anat 2004; 205:357–62 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Hou Y, Shi G, Shi J, Xu G, Guo Y, Xu P. Study design: in vitro and in vivo assessment of bone morphogenic protein 2 combined with platelet-rich plasma on treatment of disc degeneration. Int Orthop 2016; 40:1143–55 [DOI] [PubMed] [Google Scholar]
- 69.Sun W, Zhang K, Liu G, Ding W, Zhao C, Xie Y, Yuan J, Sun X, Li H, Liu C, Tang T, Zhao J. Sox9 gene transfer enhanced regenerative effect of bone marrow mesenchymal stem cells on the degenerated intervertebral disc in a rabbit model. PLoS One 2014; 9:e93570. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Yi Z, Guanjun T, Lin C, Zifeng P. Effects of transplantation of hTIMP1-expressing bone marrow mesenchymal stem cells on the extracellular matrix of degenerative intervertebral discs in an in vivo rabbit model. Spine 2014; 39:E669–E675 [DOI] [PubMed] [Google Scholar]
- 71.Xu J, Xiao-Qiang E, Wang NX, Wang MN, Xie HX, Cao YH, Sun LH, Tian J, Chen HJ, Yan JL. BMP7 enhances the effect of BMSCs on extracellular matrix remodeling in a rabbit model of intervertebral disc degeneration. FEBS J 2016; 283:1689–700 [DOI] [PubMed] [Google Scholar]
- 72.Sakai D, Mochida J, Iwashina T, Watanabe T, Nakai T, Ando K, Hotta T. Differentiation of mesenchymal stem cells transplanted to a rabbit degenerative disc model: potential and limitations for stem cell therapy in disc regeneration. Spine 2005; 30:2379–87 [DOI] [PubMed] [Google Scholar]
- 73.Vaudreuil N, Henrikson K, Pohl P, Lee A, Lin H, Olsen A, Dong Q, Dombrowski M, Kang J, Vo N, Lee J, Sowa G. Photopolymerizable biogel scaffold seeded with mesenchymal stem cells: safety and efficacy evaluation of novel treatment for intervertebral disc degeneration. J Orthop Res 2019; 37:1451–9 [DOI] [PubMed] [Google Scholar]
- 74.Chiang ER, Ma HL, Wang JP, Chang MC, Liu CL, Chen TH, Hung SC. Use of allogeneic hypoxic mesenchymal stem cells for treating disc degeneration in rabbits. J Orthop Res 2019; 37:1440–50 [DOI] [PubMed] [Google Scholar]
- 75.Zhou X, Wang J, Huang X, Fang W, Tao Y, Zhao T, Liang C, Hua J, Chen Q, Li F. Injectable decellularized nucleus pulposus-based cell delivery system for differentiation of adipose-derived stem cells and nucleus pulposus regeneration. Acta Biomater 2018; 81:115–28 [DOI] [PubMed] [Google Scholar]
- 76.Miyamoto T, Muneta T, Tabuchi T, Matsumoto K, Saito H, Tsuji K, Sekiya I. Intradiscal transplantation of synovial mesenchymal stem cells prevents intervertebral disc degeneration through suppression of matrix metalloproteinase-related genes in nucleus pulposus cells in rabbits. Arthritis Res Ther 2010; 12:R206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Leckie SK, Sowa GA, Bechara BP, Hartman RA, Coelho JP, Witt WT, Dong QD, Bowman BW, Bell KM, Vo NV, Kramer BC, Kang JD. Injection of human umbilical tissue-derived cells into the nucleus pulposus alters the course of intervertebral disc degeneration in vivo. Spine J 2013; 13:263–72 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Sheikh H, Zakharian K, De La Torre RP, Facek C, Vasquez A, Chaudhry GR, Svinarich D, Perez-Cruet MJ. In vivo intervertebral disc regeneration using stem cell-derived chondroprogenitors. J Neurosurg Spine 2009; 10:265–72 [DOI] [PubMed] [Google Scholar]
- 79.Huang B, Zhuang Y, Li CQ, Liu LT, Zhou Y. Regeneration of the intervertebral disc with nucleus pulposus cell-seeded collagen II/hyaluronan/chondroitin-6-sulfate tri-copolymer constructs in a rabbit disc degeneration model. Spine 2011; 36:2252–9 [DOI] [PubMed] [Google Scholar]
- 80.Okuma M, Mochida J, Nishimura K, Sakabe K, Seiki K. Reinsertion of stimulated nucleus pulposus cells retards intervertebral disc degeneration: an in vitro and in vivo experimental study. J Orthop Res 2000; 18:988–97 [DOI] [PubMed] [Google Scholar]
- 81.Kong MH, Do DH, Miyazaki M, Wei F, Yoon SH, Wang JC. Rabbit model for in vivo study of intervertebral disc degeneration and regeneration. J Korean Neurosurg Soc 2008; 44:327–33 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Silverman LI, Dulatova G, Tandeski T, Erickson IE, Lundell B, Toplon D, Wolff T, Howard A, Chintalacharuvu S, Foley KT. In vitro and in vivo evaluation of discogenic cells, an investigational cell therapy for disc degeneration. Spine J 2020; 20:138–49 [DOI] [PubMed] [Google Scholar]
- 83.Liu Y, Yu T, Ma XX, Xiang HF, Hu YG, Chen BH. Lentivirus-mediated TGF-β3, CTGF and TIMP1 gene transduction as a gene therapy for intervertebral disc degeneration in an in vivo rabbit model. Exp Ther Med 2016; 11:1399–404 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Yue B, Lin Y, Ma X, Xiang H, Qiu C, Zhang J, Li L, Chen B. Survivin-TGFB3-TIMP1 gene therapy via lentivirus vector slows the course of intervertebral disc degeneration in an in vivo rabbit model. Spine 2016; 41:926–34 [DOI] [PubMed] [Google Scholar]
- 85.Nishida K, Kang JD, Gilbertson LG, Moon SH, Suh JK, Vogt MT, Robbins PD, Evans CH. Modulation of the biologic activity of the rabbit intervertebral disc by gene therapy: an in vivo study of adenovirus-mediated transfer of the human transforming growth factor beta 1 encoding gene. Spine 1999; 24:2419–25 [DOI] [PubMed] [Google Scholar]
- 86.Zhan Z, Shao Z, Xiong X, Yang S, Du J, Zheng Q, Wang H, Guo X, Liu Y. Ad/CMV-hTGF-beta1 treats rabbit intervertebral discs degeneration in vivo. J Huazhong Univ Sci Technol Med Sci 2004; 24:599–601,624 [DOI] [PubMed] [Google Scholar]
- 87.Paul R, Haydon RC, Cheng H, Ishikawa A, Nenadovich N, Jiang W, Zhou L, Breyer B, Feng T, Gupta P, He TC, Phillips FM. Potential use of Sox9 gene therapy for intervertebral degenerative disc disease. Spine 2003; 28:755–63 [PMC free article] [PubMed] [Google Scholar]
- 88.Liu HF, Ning B, Zhang H, Wang DC, Hu YL, Qiao GX, Zhao YP., Hu YG. Effect of rAAV2-hTGFβ1 gene transfer on matrix synthesis in an in vivo rabbit disk degeneration model. Clin Spine Surg 2016; 29:E127–34 [DOI] [PubMed] [Google Scholar]
- 89.Leckie SK, Bechara BP, Hartman RA, Sowa GA, Woods BI, Coelho JP, Witt WT, Dong QD, Bowman BW, Bell KM, Vo NV, Wang B, Kang JD. Injection of AAV2-BMP2 and AAV2-TIMP1 into the nucleus pulposus slows the course of intervertebral disc degeneration in an in vivo rabbit model. Spine J 2012; 12:7–20 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Obata S, Akeda K, Imanishi T, Masuda K, Bae W, Morimoto R, Asanuma Y, Kasai Y, Uchida A, Sudo A. Effect of autologous platelet-rich plasma-releasate on intervertebral disc degeneration in the rabbit anular puncture model: a preclinical study. Arthritis Res Ther 2012; 14:R241. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Sawamura K, Ikeda T, Nagae M, Okamoto S, Mikami Y, Hase H, Ikoma K, Yamada T, Sakamoto H, Matsuda K, Tabata Y, Kawata M, Kubo T. Characterization of in vivo effects of platelet-rich plasma and biodegradable gelatin hydrogel microspheres on degenerated intervertebral discs. Tissue Eng Part A 2009; 15:3719–27 [DOI] [PubMed] [Google Scholar]
- 92.Paglia DN, Singh H, Karukonda T, Drissi H, Moss IL. PDGF-BB delays degeneration of the intervertebral discs in a rabbit preclinical model. Spine 2016; 41:E449–58 [DOI] [PubMed] [Google Scholar]
- 93.Abbushi A, Endres M, Cabraja M, Kroppenstedt SN, Thomale UW, Sittinger M, Hegewald AA, Morawietz L, Lemke AJ, Bansemer VG, Kaps C, Woiciechowsky C. Regeneration of intervertebral disc tissue by resorbable cell-free polyglycolic acid-based implants in a rabbit model of disc degeneration. Spine 2008; 33:1527–32 [DOI] [PubMed] [Google Scholar]
- 94.So K, Takemoto M, Fujibayashi S, Neo M, Kyomoto M, Hayami T, Hyon SH, Nakamura T. Antidegenerative effects of partial disc replacement in an animal surgery model. Spine 2007; 32:1586–91 [DOI] [PubMed] [Google Scholar]
- 95.Masuda K, Imai Y, Okuma M, Muehleman C, Nakagawa K, Akeda K, Thonar E, Andersson G, An HS. Osteogenic protein-1 injection into a degenerated disc induces the restoration of disc height and structural changes in the rabbit anular puncture model. Spine 2006; 31:742–54 [DOI] [PubMed] [Google Scholar]
- 96.Miyamoto K, Masuda K, Kim JG, Inoue N, Akeda K, Andersson GB, An HS. Intradiscal injections of osteogenic protein-1 restore the viscoelastic properties of degenerated intervertebral discs. Spine J 2006; 6:692–703 [DOI] [PubMed] [Google Scholar]
- 97.Kroeber M, Unglaub F, Guehring T, Nerlich A, Hadi T, Lotz J, Carstens C. Effects of controlled dynamic disc distraction on degenerated intervertebral discs: an in vivo study on the rabbit lumbar spine model. Spine 2005; 30:181–7 [DOI] [PubMed] [Google Scholar]
- 98.Chujo T, An HS, Akeda K, Miyamoto K, Muehleman C, Attawia M, Andersson G, Masuda K. Effects of growth differentiation factor-5 on the intervertebral disc: in vitro bovine study and in vivo rabbit disc degeneration model study. Spine 2006; 31:2909–17 [DOI] [PubMed] [Google Scholar]
- 99.Sudo H, Minami A. Caspase 3 as a therapeutic target for regulation of intervertebral disc degeneration in rabbits. Arthritis Rheum 2011; 63:1648–57 [DOI] [PubMed] [Google Scholar]
- 100.Seki S, Asanuma-Abe Y, Masuda K, Kawaguchi Y, Asanuma K, Muehleman C, Iwai A, Kimura T. Effect of small interference RNA (siRNA) for ADAMTS5 on intervertebral disc degeneration in the rabbit anular needle-puncture model. Arthritis Res Ther 2009; 11:R166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Banala RR, Vemuri SK, Dar GH, Palanisamy V, Penkulinti M, Surekha MV, Gurava Reddy AV, Nalam MR, Subbaiah G. Efficiency of dual siRNA-mediated gene therapy for intervertebral disc degeneration (IVDD). Spine J 2019; 19:896–904 [DOI] [PubMed] [Google Scholar]
- 102.Mwale F, Masuda K, Pichika R, Epure LM, Yoshikawa T, Hemmad A, Roughley PJ, Antoniou J. The efficacy of link N as a mediator of repair in a rabbit model of intervertebral disc degeneration. Arthritis Res Ther 2011; 13:R120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Mwale F, Masuda K, Grant MP, Epure LM, Kato K, Miyazaki S, Cheng K, Yamada J, Bae WC, Muehleman C, Roughley PJ, Antoniou J. Short link N promotes disc repair in a rabbit model of disc degeneration. Arthritis Res Ther 2018; 20:201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Kwon YJ, Lee JW, Moon EJ, Chung YG., Kim OS, Kim HJ. Anabolic effects of peniel 2000, a peptide that regulates TGF-β1 signaling on intervertebral disc degeneration. Spine 2013; 38:E49–58 [DOI] [PubMed] [Google Scholar]
- 105.Shen J, Zhuo N, Xu S, Song Z, Hu Z, Hao J, Guo X. Resveratrol delivery by ultrasound-mediated nanobubbles targeting nucleus pulposus cells. Nanomedicine 2018; 13:1433–46 [DOI] [PubMed] [Google Scholar]
- 106.Ge J, Zhou Q, Niu J, Wang Y, Yan Q, Wu C, Qian J, Yang H, Zou J. Melatonin protects intervertebral disc from degeneration by improving cell survival and function via activation of the ERK1/2 signaling pathway. Oxid Med Cell Longev 2019; 2019:5120275. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Riew KD, Lou J, Wright NM, Cheng SL, Bae KT, Avioli LV. Thoracoscopic intradiscal spine fusion using a minimally invasive gene-therapy technique. J Bone Joint Surg Am 2003; 85:866–71 [DOI] [PubMed] [Google Scholar]
- 108.Barczewska M, Jezierska-Wozniak K, Habich A, Lipinski S, Holak P, Maksymowicz W, Wojtkiewicz J. Evaluation of regenerative processes in the pig model of intervertebral disc degeneration after transplantation of bone marrow-derived mesenchymal stem cells. Folia Neuropathol 2018; 56:124–32 [DOI] [PubMed] [Google Scholar]
- 109.Omlor GW, Lorenz S, Nerlich AG, Guehring T, Richter W. Disc cell therapy with bone-marrow-derived autologous mesenchymal stromal cells in a large porcine disc degeneration mode. Eur Spine J 2018; 27:2639–49 [DOI] [PubMed] [Google Scholar]
- 110.Omlor GW, Fischer J, Kleinschmitt K, Benz K, Holschbach J, Brohm K, Anton M, Guehring T, Richter W. Short-term follow-up of disc cell therapy in a porcine nucleotomy model with an albumin-hyaluronan hydrogel: in vivo and in vitro results of metabolic disc cell activity and implant distribution. Eur Spine J 2014; 23:1837–47 [DOI] [PubMed] [Google Scholar]
- 111.Acosta FL, Jr., Metz L, Adkisson HD, Liu J, Carruthers-Liebenberg E, Milliman C, Maloney M, Lotz JC. Porcine intervertebral disc repair using allogeneic juvenile articular chondrocytes or mesenchymal stem cells. Tissue Eng Part A 2011; 17:3045–55 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Henriksson HB, Hagman M, Horn M, Lindahl A, Brisby H. Investigation of different cell types and gel carriers for cell-based intervertebral disc therapy, in vitro and in vivo studies. J Tissue Eng Regen Med 2012; 6:738–47 [DOI] [PubMed] [Google Scholar]
- 113.Zhou X, Zhang F, Wang D, Wang J, Wang C, Xia K, Ying L, Huang X, Tao Y, Chen S, Xue D, Hua J, Liang C, Chen Q, Li F. Micro fragmented adipose tissue promotes the matrix synthesis function of nucleus pulposus cells and regenerates degenerated intervertebral disc in a pig model. Cell Transplant 2020; 29:963689720905798. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Sheyn D, Ben-David S, Tawackoli W, Zhou Z, Salehi K, Bez M, De Mel S, Chan V, Roth J, Avalos P, Giaconi JC, Yameen H, Hazanov L, Seliktar D, Li D, Gazit D, Gazit Z. Human iPSCs can be differentiated into notochordal cells that reduce intervertebral disc degeneration in a porcine model. Theranostics 2019; 9:7506–24 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Leckie AE, Akens MK, Woodhouse KA, Yee AJ, Whyne CM. Evaluation of thiol-modified hyaluronan and elastin-like polypeptide composite augmentation in early-stage disc degeneration: comparing 2 minimally invasive techniques. Spine 2012; 37:E1296–303 [DOI] [PubMed] [Google Scholar]
- 116.Omlor GW, Nerlich AG, Lorenz H, Bruckner T, Richter W, Pfeiffer M, Gühring T. Injection of a polymerized hyaluronic acid/collagen hydrogel matrix in an in vivo porcine disc degeneration model. Eur Spine J 2012; 21:1700–8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Wang YH, Kuo TF, Wang JL. The implantation of non-cell-based materials to prevent the recurrent disc herniation: an in vivo porcine model using quantitative discomanometry examination. Eur Spine J 2007; 16:1021–7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Buttermann GR, Mendenhall HV. Feasibility of the runt cow for in vivo testing of a spinal interbody prosthesis with preliminary results. J Invest Surg 2012; 25:112–22 [DOI] [PubMed] [Google Scholar]
- 119.Wilke HJ, Kettler A, Claes LE. Are sheep spines a valid biomechanical model for human spines? Spine 1997; 22:2365–74 [DOI] [PubMed] [Google Scholar]
- 120.Kandziora F, Pflugmacher R, Scholz M, Schnake K, Lucke M, Schröder R, Mittlmeier T. Comparison between sheep and human cervical spines: an anatomic, radiographic, bone mineral density, and biomechanical study. Spine 2001; 26:1028–37 [DOI] [PubMed] [Google Scholar]
- 121.Zhang Y, Drapeau S, An HS, Markova D, Lenart BA, Anderson DG. Histological features of the degenerating intervertebral disc in a goat disc-injury model. Spine 2011; 36:1519–27 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Qin J, He X, Wang D, Qi P, Guo L, Huang S, Cai X, Li H, Wang R. Artificial cervical vertebra and intervertebral complex replacement through the anterior approach in animal model: a biomechanical and in vivo evaluation of a successful goat model. PLoS One 2012; 7:e52910. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Hoogendoorn RJW, Helder MN, Kroeze RJ, Bank RA, Smit TH, Wuisman PIJM. Reproducible long-term disc degeneration in a large animal model. Spine 2008; 33:949–54 [DOI] [PubMed] [Google Scholar]
- 124.Freeman BJ, Kuliwaba JS, Jones CF, Shu CC, Colloca CJ, Zarrinkalam MR, Mulaibrahimovic A, Gronthos S, Zannettino AC, Howell S. Allogeneic mesenchymal stem cells promote healing in postero-lateral annular lesions and improve indices of lumbar intervertebral disc degeneration in an ovine model. Spine 2016; 41:1331–9 [DOI] [PubMed] [Google Scholar]
- 125.Hussain I, Sloan SR, Wipplinger C, Navarro-Ramirez R, Zubkov M, Kim E, Kirnaz S, Bonassar LJ, Härtl R. Mesenchymal stem cell-seeded high-density collagen gel for annular repair: 6-week results from in vivo sheep models. Neurosurgery 2019; 85:E350–E359 [DOI] [PubMed] [Google Scholar]
- 126.Oehme D, Ghosh P, Shimmon S, Wu J, McDonald C, Troupis JM, Goldschlager T, Rosenfeld JV, Jenkin G. Mesenchymal progenitor cells combined with pentosan polysulfate mediating disc regeneration at the time of microdiscectomy: a preliminary study in an ovine model. J Neurosurg Spine 2014; 20:657–69 [DOI] [PubMed] [Google Scholar]
- 127.Reitmaier S, Kreja L, Gruchenberg K, Kanter B, Silva-Correia J, Oliveira JM, Reis RL, Perugini V, Santin M, Ignatius A, Wilke HJ. In vivo biofunctional evaluation of hydrogels for disc regeneration. Eur Spine J 2014; 23:19–26 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Hegewald AA, Medved F, Feng D, Tsagogiorgas C, Beierfuß A, Schindler GA, Trunk M, Kaps C, Mern DS, Thomé C. Enhancing tissue repair in annulus fibrosus defects of the intervertebral disc: analysis of a bio-integrative annulus implant in an in-vivo ovine model. J Tissue Eng Regen Med 2015; 9:405–14 [DOI] [PubMed] [Google Scholar]
- 129.Ledet EH, Jeshuran W, Glennon JC, Shaffrey C, De Deyne P, Belden C, Kallakury B, Carl AL. Small intestinal submucosa for anular defect closure: long-term response in an in vivo sheep model. Spine 2009; 34:1457–63 [DOI] [PubMed] [Google Scholar]
- 130.Wei A, Williams LA, Bhargav D, Shen B, Kishen T, Duffy N, Diwan AD. BMP13 prevents the effects of annular injury in an ovine model. Int J Biol Sci 2009; 5:388–96 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Peeters M, Detiger SE, Karfeld-Sulzer LS, Smit TH, Yayon A, Weber FE, Helder MN. BMP-2 and BMP-2/7 heterodimers conjugated to a fibrin/hyaluronic acid hydrogel in a large animal model of mild intervertebral disc degeneration. Biores Open Access 2015; 4:398–406 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Gu Y, Zhang F, Lineaweaver WC, Zhang J, Jia L, Qi J, Wang J, Zhen X. In vivo study of hydroxyapatite-coated hat type cervical intervertebral fusion cage combined with IGF-I and TGF-β1 in the goat model. Clin Spine Surg 2016; 29:E267–75 [DOI] [PubMed] [Google Scholar]
- 133.Wuisman PI, van Dijk M, Smit TH. Resorbable cages for spinal fusion: an experimental goat model. J Neurosurg 2002; 97:433–9 [DOI] [PubMed] [Google Scholar]
- 134.Bergknut N, Auriemma E, Wijsman S, Voorhout G, Hagman R, Lagerstedt AS, Hazewinkel HAW, Meij BP. Evaluation of intervertebral disk degeneration in chondrodystrophic and nonchondrodystrophic dogs by use of Pfirrmann grading of images obtained with low-field magnetic resonance imaging. Am J Vet Res 2011; 72:893–8 [DOI] [PubMed] [Google Scholar]
- 135.Willems N, Tellegen AR, Bergknut N, Creemers LB, Wolfswinkel J, Freudigmann C, Benz K, Grinwis GCM, Tryfonidou MA, Meij BP. Inflammatory profiles in canine intervertebral disc degeneration. BMC Vet Res 2016; 12:10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Serigano K, Sakai D, Hiyama A, Tamura F, Tanaka M, Mochida J. Effect of cell number on mesenchymal stem cell transplantation in a canine disc degeneration model. J Orthop Res 2010; 28:1267–75 [DOI] [PubMed] [Google Scholar]
- 137.Steffen F, Bertolo A, Affentranger R, Ferguson SJ, Stoyanov J. Treatment of naturally degenerated canine lumbosacral intervertebral discs with autologous mesenchymal stromal cells and collagen microcarriers: a prospective clinical study. Cell Transplant 2019; 28:201–11 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Bach FC, Tellegen AR, Beukers M, Miranda-Bedate A, Teunissen M, de Jong WAM, de Vries SAH, Creemers LB, Benz K, Meij BP, Ito K, Tryfonidou MA. Biologic canine and human intervertebral disc repair by notochordal cell-derived matrix: from bench towards bedside. Oncotarget 2018; 9:26507–26 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Nukaga T, Sakai D, Tanaka M, Hiyama A, Nakai T, Mochida J. Transplantation of activated nucleus pulposus cells after cryopreservation: efficacy study in a canine disc degeneration model. Eur Cell Mater 2016; 31:95–106 [DOI] [PubMed] [Google Scholar]
- 140.Ruan DK, Xin H, Zhang C, Wang C, Xu C, Li C, He Q. Experimental intervertebral disc regeneration with tissue-engineered composite in a canine model. Tissue Eng Part A 2010; 16:2381–9 [DOI] [PubMed] [Google Scholar]
- 141.Zhang Y, Tao H, Gu T, Zhou M, Jia Z, Jiang G, Chen C, Han Z, Xu C, Wang D, He Q, Ruan D. The effects of human Wharton's jelly cell transplantation on the intervertebral disc in a canine disc degeneration model. Stem Cell Res Ther 2015; 6:154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Willems N, Mihov G, Grinwis GC, van Dijk M, Schumann D, Bos C, Strijkers GJ, Dhert WJ, Meij BP, Creemers LB, Tryfonidou MA. Safety of intradiscal injection and biocompatibility of polyester amide microspheres in a canine model predisposed to intervertebral disc degeneration. J Biomed Mater Res B Appl Biomater 2017; 105:707–14 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Tellegen AR, Rudnik-Jansen I, Beukers M, Miranda-Bedate A, Bach FC, de Jong W, Woike N, Mihov G, Thies JC, Meij BP, Creemers LB, Tryfonidou MA. Intradiscal delivery of celecoxib-loaded microspheres restores intervertebral disc integrity in a preclinical canine model. J Control Release 2018; 286:439–50 [DOI] [PubMed] [Google Scholar]
- 144.Rudnik-Jansen I, Tellegen A, Beukers M, Öner F, Woike N, Mihov G, Thies J, Meij B, Tryfonidou M, Creemers L. Safety of intradiscal delivery of triamcinolone acetonide by a poly(esteramide) microsphere platform in a large animal model of intervertebral disc degeneration. Spine J 2019; 19:905–19 [DOI] [PubMed] [Google Scholar]
- 145.Tellegen AR, Willems N, Beukers M, Grinwis GCM, Plomp SGM, Bos C, van Dijk M, de Leeuw M, Creemers LB, Tryfonidou MA, Meij BP. Intradiscal application of a PCLA-PEG-PCLA hydrogel loaded with celecoxib for the treatment of back pain in canines: what's in it for humans? J Tissue Eng Regen Med 2018; 12:642–52 [DOI] [PubMed] [Google Scholar]
- 146.Willems N, Bach FC, Plomp SG, van Rijen MH, Wolfswinkel J, Grinwis GC, Bos C, Strijkers GJ, Dhert WJ, Meij BP, Creemers LB, Tryfonidou MA. Intradiscal application of rhBMP-7 does not induce regeneration in a canine model of spontaneous intervertebral disc degeneration. Arthritis Res Ther 2015; 17:137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Ding Y, Ruan D, Luk KD, He Q, Wang C. The effect of gamma irradiation on the biological properties of intervertebral disc allografts: in vitro and in vivo studies in a beagle model. PLoS One 2014; 9:e100304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Liu Y, Kong J, Xi YM, Yu T, Wu XL, Hu YG. Effects of AAV2-mediated co-transfection of CTGF and TIMP1 genes on degenerative lumbar intervertebral discs in rhesus monkeys in vivo. Am J Transl Res 2018; 10:1085–96 [PMC free article] [PubMed] [Google Scholar]
- 149.Luk KD, Ruan DK, Lu DS, Fei ZQ. Fresh frozen intervertebral disc allografting in a bipedal animal model. Spine 2003; 28:864–9, discussion 870 [DOI] [PubMed] [Google Scholar]
- 150.Allen MJ, Schoonmaker JE, Bauer TW, Williams PF, Higham PA, Yuan HA. Preclinical evaluation of a poly (vinyl alcohol) hydrogel implant as a replacement for the nucleus pulposus. Spine 2004; 29:515–23 [DOI] [PubMed] [Google Scholar]
- 151.Cunningham BW, Dmitriev AE, Hu N, McAfee PC. General principles of total disc replacement arthroplasty: seventeen cases in a nonhuman primate model. Spine 2003; 28:S118–24 [DOI] [PubMed] [Google Scholar]
- 152.Cunningham BW, Lowery GL, Serhan HA, Dmitriev AE, Orbegoso CM, McAfee PC, Fraser RD, Ross RE, Kulkarni SS. Total disc replacement arthroplasty using the AcroFlex lumbar disc: a non-human primate model. Eur Spine J 2002; 11:S115–23 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Ledet EH, Sachs BL, Brunski JB, Gatto CE, Donzelli PS. Real-time in vivo loading in the lumbar spine: part 1. Interbody implant: load cell design and preliminary results. Spine 2000; 25:2595–600 [DOI] [PubMed] [Google Scholar]
- 154.Wei F, Zhong R, Wang L, Zhou Z, Pan X, Cui S, Sun H, Zou X, Gao M, Jiang B, Chen W, Zhuang W, Sun H, Liu S. Pingyangmycin-induced in vivo lumbar disc degeneration model of rhesus monkeys. Spine 2015; 40:E199–210 [DOI] [PubMed] [Google Scholar]
- 155.Wei F, Zhong R, Zhou Z, Wang L, Pan X, Cui S, Zou X, Gao M, Sun H, Chen W, Liu S. In vivo experimental intervertebral disc degeneration induced by bleomycin in the rhesus monkey. BMC Musculoskelet Disord 2014; 15:340. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Stern WE, Coulson WF. Effects of collagenase upon the intervertebral disc in monkeys. J Neurosurg 1976; 44:32–44 [DOI] [PubMed] [Google Scholar]
- 157.Hayes AJ, Benjamin M, Ralphs JR. Extracellular matrix in development of the intervertebral disc. Matrix Biol 2001; 20:107–21 [DOI] [PubMed] [Google Scholar]
- 158.Trout JJ, Buckwalter JA, Moore KC, Landas SK. Ultrastructure of the human intervertebral disc. I. Changes in notochordal cells with age. Tissue Cell 1982; 14:359–69 [DOI] [PubMed] [Google Scholar]
- 159.Bergknut N, Rutges JP, Kranenburg HJ, Smolders LA, Hagman R, Smidt HJ, Lagerstedt AS, Penning LC, Voorhout G, Hazewinkel HA, Grinwis GC, Creemers LB, Meij BP, Dhert WJ. The dog as an animal model for intervertebral disc degeneration? Spine 2012; 37:351–8 [DOI] [PubMed] [Google Scholar]
- 160.Platenberg RC, Hubbard GB, Ehler WJ, Hixson CJ. Spontaneous disc degeneration in the baboon model: magnetic resonance imaging and histopathologic correlation. J Med Primatol 2001; 30:268–72 [DOI] [PubMed] [Google Scholar]
- 161.Nuckley DJ, Kramer PA, Del Rosario A, Fabro N, Baran S, Ching RP. Intervertebral disc degeneration in a naturally occurring primate model: radiographic and biomechanical evidence. J Orthop Res 2008; 26:1283–8 [DOI] [PubMed] [Google Scholar]
- 162.Beierfuß A, Dietrich H, Kremser C, Hunjadi M, Ritsch A, Rülicke T, Thomé C, Mern DS. Knockout of apolipoprotein E in rabbit promotes premature intervertebral disc degeneration: a new in vivo model for therapeutic approaches of spinal disc disorders. PLoS One 2017; 12:e0187564. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Beierfuß A, Hunjadi M, Ritsch A, Kremser C, Thomé C, Mern DS. APOE-knockout in rabbits causes loss of cells in nucleus pulposus and enhances the levels of inflammatory catabolic cytokines damaging the intervertebral disc matrix. PLoS One 2019; 14:e0225527. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Sahlman J, Inkinen R, Hirvonen T, Lammi MJ, Lammi PE, Nieminen J, Lapveteläinen T, Prockop DJ, Arita M, Li SW, Hyttinen MM, Helminen HJ, Puustjärvi K. Premature vertebral endplate ossification and mild disc degeneration in mice after inactivation of one allele belonging to the Col2a1 gene for type II collagen. Spine 2001; 26:2558–65 [DOI] [PubMed] [Google Scholar]
- 165.Kimura T, Nakata K, Tsumaki N, Miyamoto S, Matsui Y, Ebara S, Ochi T. Progressive degeneration of articular cartilage and intervertebral discs. An experimental study in transgenic mice bearing a type IX collagen mutation. Int Orthop 1996; 20:177–81 [DOI] [PubMed] [Google Scholar]
- 166.Silberberg R, Aufdermaur M, Adler JH. Degeneration of the intervertebral disks and spondylosis in aging sand rats. Arch Pathol Lab Med 1979; 103:231–5 [PubMed] [Google Scholar]
- 167.Moskowitz RW, Ziv I, Denko CW, Boja B, Jones PK, Adler JH. Spondylosis in sand rats: a model of intervertebral disc degeneration and hyperostosis. J Orthop Res 1990; 8:401–11 [DOI] [PubMed] [Google Scholar]
- 168.Smith L, Brown JE. Treatment of lumbar intervertebral disc lesions by direct injection of chymopapain. J Bone Joint Surg Br 1967; 49:502–19 [PubMed] [Google Scholar]
- 169.Zook BC, Kobrine AI. Effects of collagenase and chymopapain on spinal nerves and intervertebral discs of cynomolgus monkeys. J Neurosurg 1986; 64:474–83 [DOI] [PubMed] [Google Scholar]
- 170.Kiester DP, Williams JM, Andersson GB, Thonar EJ, McNeill TW. The dose-related effect of intradiscal chymopapain on rabbit intervertebral discs. Spine 1994; 19:747–51 [DOI] [PubMed] [Google Scholar]
- 171.Hoogendoorn RJ, Wuisman PI, Smit TH, Everts VE, Helder MN. Experimental intervertebral disc degeneration induced by chondroitinase ABC in the goat. Spine 2007; 32:1816–25 [DOI] [PubMed] [Google Scholar]
- 172.Lindblom K. Intervertebral-disc degeneration considered as a pressure atrophy. J Bone Joint Surg Am 1957; 39-A:933–45 [PubMed] [Google Scholar]
- 173.Ching CTS, Chow DHK, Yao FYD, Holmes AD. The effect of cyclic compression on the mechanical properties of the inter-vertebral disc: an in vivo study in a rat tail model. Clin Biomech 2003; 18:182–9 [DOI] [PubMed] [Google Scholar]
- 174.Court C, Colliou OK, Chin JR, Liebenberg E, Bradford DS, Lotz JC. The effect of static in vivo bending on the murine intervertebral disc. Spine J 2001; 1:239–45 [DOI] [PubMed] [Google Scholar]
- 175.Iatridis JC, Mente PL, Stokes IA, Aronsson DD, Alini M. Compression-induced changes in intervertebral disc properties in a rat tail model. Spine 1999; 24:996–1002 [DOI] [PubMed] [Google Scholar]
- 176.Kroeber MW, Unglaub F, Wang H, Schmid C, Thomsen M, Nerlich A, Richter W. New in vivo animal model to create intervertebral disc degeneration and to investigate the effects of therapeutic strategies to stimulate disc regeneration. Spine 2002; 27:2684–90 [DOI] [PubMed] [Google Scholar]
- 177.Rousseau MA, Ulrich JA, Bass EC, Rodriguez AG, Liu JJ, Lotz JC. Stab incision for inducing intervertebral disc degeneration in the rat. Spine 2007; 32:17–24 [DOI] [PubMed] [Google Scholar]
- 178.Masuda K, Aota Y, Muehleman C, Imai Y, Okuma M, Thonar EJ, Andersson GB, An HS. A novel rabbit model of mild, reproducible disc degeneration by an anulus needle puncture: correlation between the degree of disc injury and radiological and histological appearances of disc degeneration. Spine 2005; 30:5–14 [DOI] [PubMed] [Google Scholar]
- 179.Osti OL, Vernon-Roberts B, Fraser RD. Volvo award in experimental studies. Anulus tears and intervertebral disc degeneration. An experimental study using an animal model. Spine 1990; 15:762–7 [DOI] [PubMed] [Google Scholar]
- 180.Oehme D, Goldschlager T, Rosenfeld J, Danks A, Ghosh P, Gibbon A, Jenkin G. Lateral surgical approach to lumbar intervertebral discs in an ovine model. Sci World J 2012; 2012:873726. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Millecamps M, Czerminski JT, Mathieu AP, Stone LS. Behavioral signs of axial low back pain and motor impairment correlate with the severity of intervertebral disc degeneration in a mouse model. Spine J 2015; 15:2524–37 [DOI] [PubMed] [Google Scholar]
- 182.Lai A, Moon A, Purmessur D, Skovrlj B, Winkelstein BA, Cho SK, Hecht AC, Iatridis JC. Assessment of functional and behavioral changes sensitive to painful disc degeneration. J Orthop Res 2015; 33:755–64 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Olmarker K. Puncture of a lumbar intervertebral disc induces changes in spontaneous pain behavior: an experimental study in rats. Spine 2008; 33:850–5 [DOI] [PubMed] [Google Scholar]
- 184.Omarker K, Myers RR. Pathogenesis of sciatic pain: role of herniated nucleus pulposus and deformation of spinal nerve root and dorsal root ganglion. Pain 1998; 78:99–105 [DOI] [PubMed] [Google Scholar]
- 185.Meisel HJ, Siodla V, Ganey T, Minkus Y, Hutton WC, Alasevic OJ. Clinical experience in cell-based therapeutics: disc chondrocyte transplantation a treatment for degenerated or damaged intervertebral disc. Biomol Eng 2007; 24:5–21 [DOI] [PubMed] [Google Scholar]
- 186.Orozco L, Soler R, Morera C, Alberca M, Sánchez A, García-Sancho J. Intervertebral disc repair by autologous mesenchymal bone marrow cells: a pilot study. Transplantation 2011; 92:822–8 [DOI] [PubMed] [Google Scholar]
- 187.Yoshikawa T, Ueda Y, Miyazaki K, Koizumi M, Takakura Y. Disc regeneration therapy using marrow mesenchymal cell transplantation: a report of two case studies. Spine 2010; 35:E475–80 [DOI] [PubMed] [Google Scholar]
- 188.Bae HW, Amirdelfan K, Coric D, McJunkin TL, Pettine KA, Hong HJ, DePalma MJ, Kim KD, Beckworth WJ, Oehme D, Goldschlager T, Brown RD. A phase II study demonstrating efficacy and safety of mesenchymal precursor cells in low back pain due to disc degeneration. Spine J 2014; 14:31–223602377 [Google Scholar]



