1
|
Xue P, Wang Y, Lv L, Wang D, Wang Y. Roles of Chemokines in Intervertebral Disk Degeneration. Curr Pain Headache Rep 2024; 28:95-108. [PMID: 37976014 DOI: 10.1007/s11916-023-01188-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/08/2023] [Indexed: 11/19/2023]
Abstract
PURPOSE OF REVIEW Intervertebral disc degeneration is the primary etiology of low back pain and radicular pain. This review examines the roles of crucial chemokines in different stages of degenerative disc disease, along with interventions targeting chemokine function to mitigate disc degeneration. RECENT FINDINGS The release of chemokines from degenerated discs facilitates the infiltration and activation of immune cells, thereby intensifying the inflammatory cascade response. The migration of immune cells into the venous lumen is concomitant with the emergence of microvascular tissue and nerve fibers. Furthermore, the presence of neurogenic factors secreted by disc cells and immune cells stimulates the activation of pain-related cation channels in the dorsal root ganglion, potentially exacerbating discogenic and neurogenic pain and intensifying the degenerative cascade response mediated by chemokines. Gaining a deeper comprehension of the functions of chemokines and immune cells in these processes involving catabolism, angiogenesis, and injury detection could offer novel therapeutic avenues for managing symptomatic disc disease.
Collapse
Affiliation(s)
- Pengfei Xue
- Medical School of Southeast University, Nanjing, Jiangsu, 210009, China
- Central Laboratory, Gaochun Hospital Affiliated to Jiangsu University, Nanjing, Jiangsu, 211300, China
| | - Yi Wang
- Department of Orthopaedics, Jiujiang Traditional Chinese Medicine Hospital, Jiujiang, Jiangxi, 332000, China
| | - Long Lv
- Central Laboratory, Gaochun Hospital Affiliated to Jiangsu University, Nanjing, Jiangsu, 211300, China
| | - Dongming Wang
- Central Laboratory, Gaochun Hospital Affiliated to Jiangsu University, Nanjing, Jiangsu, 211300, China.
| | - Yuntao Wang
- Medical School of Southeast University, Nanjing, Jiangsu, 210009, China.
- Department of Spine Center, Zhongda Hospital, Southeast University, Nanjing, Jiangsu, 210009, China.
| |
Collapse
|
2
|
Zhang A, Cheng Z, Chen Y, Shi P, Gan W, Zhang Y. Emerging tissue engineering strategies for annulus fibrosus therapy. Acta Biomater 2023:S1742-7061(23)00337-9. [PMID: 37330029 DOI: 10.1016/j.actbio.2023.06.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 05/31/2023] [Accepted: 06/12/2023] [Indexed: 06/19/2023]
Abstract
Low back pain is a major public health concern experienced by 80% of the world's population during their lifetime, which is closely associated with intervertebral disc (IVD) herniation. IVD herniation manifests as the nucleus pulposus (NP) protruding beyond the boundaries of the intervertebral disc due to disruption of the annulus fibrosus (AF). With a deepening understanding of the importance of the AF structure in the pathogenesis of intervertebral disc degeneration, numerous advanced therapeutic strategies for AF based on tissue engineering, cellular regeneration, and gene therapy have emerged. However, there is still no consensus concerning the optimal approach for AF regeneration. In this review, we summarized strategies in the field of AF repair and highlighted ideal cell types and pro-differentiation targeting approaches for AF repair, and discussed the prospects and difficulties of implant systems combining cells and biomaterials to guide future research directions. STATEMENT OF SIGNIFICANCE: Low back pain is a major public health concern experienced by 80% of the world's population during their lifetime, which is closely associated with intervertebral disc (IVD) herniation. However, there is still no consensus concerning the optimal approach for annulus fibrosus (AF) regeneration. In this review, we summarized strategies in the field of AF repair and highlighted ideal cell types and pro-differentiation targeting approaches for AF repair, and discussed the prospects and difficulties of implant systems combining cells and biomaterials to guide future research directions.
Collapse
Affiliation(s)
- Anran Zhang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zhangrong Cheng
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yuhang Chen
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Pengzhi Shi
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Weikang Gan
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yukun Zhang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
3
|
Hornung AL, Baker JD, Mallow GM, Sayari AJ, Albert HB, Tkachev A, An HS, Samartzis D. Resorption of Lumbar Disk Herniation: Mechanisms, Clinical Predictors, and Future Directions. JBJS Rev 2023; 11:01874474-202301000-00001. [PMID: 36722839 DOI: 10.2106/jbjs.rvw.22.00148] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
INTRODUCTION Resorption after lumbar disk herniation is a common yet unpredictable finding. It is hypothesized that nearly 70% of lumbar herniated nucleus pulposus (HNP) undergo the resorption to a significant degree after acute herniation, which has led to nonoperative management before surgical planning. METHODS This narrative review on the literature from 4 databases (MEDLINE, Cumulative Index to Nursing and Allied Health Literature, Scopus, and Cochrane) examines historical and recent advancements related to disk resorption. Studies were appraised for their description of the predictive factor (e.g., imaging or morphologic factors), pathophysiology, and treatment recommendations. OBSERVATIONS We reviewed 68 articles considering the possibility of resorption of lumbar HNP. Recent literature has proposed various mechanisms (inflammation and neovascularization, dehydration, and mechanical traction) of lumbar disk resorption; however, consensus has yet to be established. Current factors that increase the likelihood of resorption include the initial size of the herniation, sequestration, percentage of rim enhancement on initial gadolinium-based magnetic resonance imaging (MRI), composition of inflammatory mediators, and involvement of the posterior longitudinal ligament. CONCLUSION Heterogeneity in imaging and morphologic factors has led to uncertainty in the identification of which lumbar herniations will resorb. Current factors that increase the likelihood of disk resorption include the initial size of the herniation, sequestration, percentage of rim enhancement on initial MRI, composition of cellular and inflammatory mediators present, and involvement of the posterior longitudinal ligament. This review article highlights the role of disk resorption after herniation without surgical intervention and questions the role of traditional noninflammatory medications after acute herniation. Further research is warranted to refine the ideal patient profile for disk resorption to ultimately avoid unnecessary treatment, thus individualizing patient care.
Collapse
Affiliation(s)
| | - James D Baker
- Department of Orthopaedic Surgery, Rush University Medical Center, Chicago
| | - G Michael Mallow
- Department of Orthopaedic Surgery, Rush University Medical Center, Chicago
| | - Arash J Sayari
- Department of Orthopaedic Surgery, Rush University Medical Center, Chicago
| | | | - Alexander Tkachev
- Department of Orthopaedic Surgery, Rush University Medical Center, Chicago
| | - Howard S An
- Department of Orthopaedic Surgery, Rush University Medical Center, Chicago
| | - Dino Samartzis
- Department of Orthopaedic Surgery, Rush University Medical Center, Chicago
| |
Collapse
|
4
|
Yang X, Li B, Tian H, Cheng X, Zhou T, Zhao J. Curcumenol Mitigates the Inflammation and Ameliorates the Catabolism Status of the Intervertebral Discs In Vivo and In Vitro via Inhibiting the TNFα/NFκB Pathway. Front Pharmacol 2022; 13:905966. [PMID: 35795557 PMCID: PMC9252100 DOI: 10.3389/fphar.2022.905966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 05/11/2022] [Indexed: 11/25/2022] Open
Abstract
Low back pain (LBP) caused by intervertebral disc degeneration (IVDD) is accredited to the release of inflammatory cytokines followed by biomechanical and structural deterioration. In our study, we used a plant-derived medicine, curcumenol, to treat IVDD. A cell viability test was carried out to evaluate the possibility of using curcumenol. RNA-seq was used to determine relative pathways involved with curcumenol addition. Using TNFα as a trigger of inflammation, the activation of the NF-κB signaling pathway and expression of the MMP family were determined by qPCR and western blotting. Nucleus pulposus (NP) cells and the rats’ primary NP cells were cultured. The catabolism status was evaluated by an ex vivo model. A lumbar instability mouse model was carried out to show the effects of curcumenol in vivo. In general, RNA-seq revealed that multiple signaling pathways changed with curcumenol addition, especially the TNFα/NF-κB pathway. So, the NP cells and primary NP cells were induced to suffer inflammation with the activated TNFα/NF-κB signaling pathway and increased expression of the MMP family, such as MMP3, MMP9, and MMP13, which would be mitigated by curcumenol. Owing to the protective effects of curcumenol, the height loss and osteophyte formation of the disc could be prevented in the lumbar instability mouse model in vivo.
Collapse
Affiliation(s)
| | | | | | | | | | - Jie Zhao
- *Correspondence: Tangjun Zhou, ; Jie Zhao,
| |
Collapse
|
5
|
Yang JE, Zhao KH, Qu Y, Zou YC. Increased serum CXCL10 levels are associated with clinical severity and radiographic progression in patients with lumbar disc degeneration. Clin Chim Acta 2021; 525:15-22. [PMID: 34902344 DOI: 10.1016/j.cca.2021.12.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 11/16/2021] [Accepted: 12/07/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND Lumbar intervertebral degenerative disc disease (IDD) is a multifaceted progressive condition that commonly occurs in conjunction with lumbar disc herniation (LDH). CXCL10 mRNA appears to be increased in both IDD and LHD. OBJECTIVE This study was performed to identify the relationship between serum CXCL10 levels and disease severity in patients with IDD. METHODS 136 IDD patients with low back pain, 127 asymptomatic volunteers and 120 healthy controls were enrolled. Serum CXCL10 protein concentrations were detected using commercial human CXCL10 ELISA Kits. Serum CXCL10 mRNA were examined using qRT-PCR. Clinical severity was assessed using the visual analog scale (VAS) and Oswestry Disability Index(ODI) scores. Radiographic severity was defined using the MRI-based Pfirrmann classification of disc degeneration. Receiver operating characteristic (ROC) curve analysis was used in estimating the correlation between CXCL10 and Pfirrmann grade. The cross-sectional area (CSA) of the lumbar multifidus muscle (LMM) and psoas major (PM) were calculated, and fat infiltration was evaluated by Ropponen-Kjaer criteria. RESULTS Serum CXCL10 concentrations were markedly raised in IDD patients with low back pain in contrast to asymptomatic individuals and healthy controls. Serum CXCL10 levels were positively associated with Pfirrmann grade. ROC curve analysis indicated that serum CXCL10 correlated well with Pfirrmann grade. In addition, serum CXCL10 concentrations were significantly higher in IDD patients with LMM and PM degeneration compared with IDD patients without degeneration. Increased CXCL10 levels positively correlated with VAS and ODI scores, as well as decreased CSA and fat filtration of the LMM and PM. CONCLUSION Increased serum CXCL10 levels correspond to clinical severity and radiographic progression in IDD patients.
Collapse
Affiliation(s)
- Jia-En Yang
- Department of Rehabilitation Medicine, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China; College of Rehabilitation Medicine, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China; Sichuan Provincial Key Laboratory of Rehabilitation Medicine, Sichuan University, Chengdu 610041, Sichuan, China; Department of Rehabilitation Medicine, Affiliated Foshan Gaoming Hospital of Guangdong Medical University, Foshan 528000, Guangdong, China
| | - Ke-Hong Zhao
- Department of Rehabilitation Medicine, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China; College of Rehabilitation Medicine, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China; Sichuan Provincial Key Laboratory of Rehabilitation Medicine, Sichuan University, Chengdu 610041, Sichuan, China
| | - Yun Qu
- Department of Rehabilitation Medicine, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China; College of Rehabilitation Medicine, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China; Sichuan Provincial Key Laboratory of Rehabilitation Medicine, Sichuan University, Chengdu 610041, Sichuan, China.
| | - Yu-Cong Zou
- Department of Rehabilitation,The third Affiliated Hospital,Southern Medical University ,Guangzhou 510630, GuangDong, China.
| |
Collapse
|
6
|
Peredo AP, Gullbrand SE, Smith HE, Mauck RL. Putting the Pieces in Place: Mobilizing Cellular Players to Improve Annulus Fibrosus Repair. TISSUE ENGINEERING. PART B, REVIEWS 2021; 27:295-312. [PMID: 32907498 PMCID: PMC10799291 DOI: 10.1089/ten.teb.2020.0196] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The intervertebral disc (IVD) is an integral load-bearing tissue that derives its function from its composite structure and extracellular matrix composition. IVD herniations involve the failure of the annulus fibrosus (AF) and the extrusion of the nucleus pulposus beyond the disc boundary. Disc herniations can impinge the neural elements and cause debilitating pain and loss of function, posing a significant burden on individual patients and society as a whole. Patients with persistent symptoms may require surgery; however, surgical intervention fails to repair the ruptured AF and is associated with the risk for reherniation and further disc degeneration. Given the limitations of AF endogenous repair, many attempts have been made toward the development of effective repair approaches that reestablish IVD function. These methods, however, fail to recapitulate the composition and organization of the native AF, ultimately resulting in inferior tissue mechanics and function over time and high rates of reherniation. Harnessing the cellular function of cells (endogenous or exogenous) at the repair site through the provision of cell-instructive cues could enhance AF tissue regeneration and, ultimately, improve healing outcomes. In this study, we review the diverse approaches that have been developed for AF repair and emphasize the potential for mobilizing the appropriate cellular players at the site of injury to improve AF healing. Impact statement Conventional treatments for intervertebral disc herniation fail to repair the annulus fibrosus (AF), increasing the risk for recurrent herniation. The lack of repair devices in the market has spurred the development of regenerative approaches, yet most of these rely on a scarce endogenous cell population to repair large injuries, resulting in inadequate regeneration. This review identifies current and developing strategies for AF repair and highlights the potential for harnessing cellular function to improve AF regeneration. Ideal cell sources, differentiation strategies, and delivery methods are discussed to guide the design of repair systems that leverage specialized cells to achieve superior outcomes.
Collapse
Affiliation(s)
- Ana P. Peredo
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, Pennsylvania, USA
| | - Sarah E. Gullbrand
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, Pennsylvania, USA
| | - Harvey E. Smith
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, Pennsylvania, USA
| | - Robert L. Mauck
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Veterans Affairs Medical Center, Philadelphia, Pennsylvania, USA
| |
Collapse
|
7
|
Dai F, Dai YX, Jiang H, Yu PF, Liu JT. Non-surgical treatment with XSHHD for ruptured lumbar disc herniation: a 3-year prospective observational study. BMC Musculoskelet Disord 2020; 21:690. [PMID: 33076896 PMCID: PMC7574183 DOI: 10.1186/s12891-020-03723-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 10/15/2020] [Indexed: 12/02/2022] Open
Abstract
Background Lumbar disc herniation (LDH) is mainly caused by annular fiber disruption with a discrete leakage of nucleus pulposus pressing on a nerve, resulting in back pain and radiating pain. Most patients with LDH can be treated conservatively, but there are many different conservative treatments. Furthermore, most previous studies did not evaluate the long-term efficacy of these treatments and the prognosis. Therefore, an effective and safe therapeutic strategy is lacking for patients with LDH. In this study, we evaluated Xiao Sui Hua He decoction (XSHHD) in the treatment of LDH. Methods This was a rigorous prospective observational 3-year follow-up study. We recruited 69 participants with ruptured lumbar disc herniation (RLDH) between February 2014 and February 2016. Patients took XSHHD orally twice a day for 6 months. The primary outcome measurements were visual analogue scale (VAS) pain score, Oswestry disability index (ODI) and straight leg raising test (SLRT). The secondary outcome measurements was nucleus pulposus protrusion volume on magnetic resonance imaging (MRI). Clinical outcomes were measured at baseline (Visit 1), and at 3, 6, 12, and 36 months (Visit 2, 3, 4, and 5, respectively).. Results Sixty-three patients were followed-up for 3 years after treatment. SLRT and ODI after non-surgical treatment improved significantly compared with baseline (P < .001). There were no statistically significant differences at 6 months vs 36 months for SLRT and ODI. VAS scores (leg, back) after 3 years of treatment were statistically significantly different compared with baseline (P < .001; Z = − 6.93, − 6.637). The baseline protrusion volume was 2018.61 ± 601.16 mm3, and the volume decreased significantly to 996.51 ± 387.42 mm3 at 36 months (t = 12.863; P < .001). The volume of protrusion resorption rate (VPRR) at 36 months was 47.24 ± 23.99%, with significant resorption in 23 cases, partial resorption in 23 cases, no resorption in 15 cases, and increased volume in 2 cases. Conclusions This study showed that non-surgical treatment with XSHHD was effective, and the study clarified the natural outcomes in LDH.
Collapse
Affiliation(s)
- Feng Dai
- Department of Orthopedics, Suzhou TCM Hospital affiliated to Nanjing University of Traditional Chinese Medicine, Suzhou, 215009, Jiangsu Province, China
| | - Yu Xiang Dai
- Department of Orthopedics, Suzhou TCM Hospital affiliated to Nanjing University of Traditional Chinese Medicine, Suzhou, 215009, Jiangsu Province, China
| | - Hong Jiang
- Department of Orthopedics, Suzhou TCM Hospital affiliated to Nanjing University of Traditional Chinese Medicine, Suzhou, 215009, Jiangsu Province, China
| | - Peng Fei Yu
- Department of Orthopedics, Suzhou TCM Hospital affiliated to Nanjing University of Traditional Chinese Medicine, Suzhou, 215009, Jiangsu Province, China
| | - Jin Tao Liu
- Department of Orthopedics, Suzhou TCM Hospital affiliated to Nanjing University of Traditional Chinese Medicine, Suzhou, 215009, Jiangsu Province, China.
| |
Collapse
|
8
|
D'Este M, Eglin D, Alini M. Lessons to be learned and future directions for intervertebral disc biomaterials. Acta Biomater 2018; 78:13-22. [PMID: 30092378 DOI: 10.1016/j.actbio.2018.08.004] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 07/16/2018] [Accepted: 08/04/2018] [Indexed: 02/07/2023]
Abstract
Biomaterials science has achieved significant advancements for the replacement, repair and regeneration of intervertebral disc tissues. However, the translation of this research to the clinic presents hurdles. The goal of this paper is to identify strategies to recapitulate the intrinsic complexities of the intervertebral disc, to highlight the unresolved issues in basic knowledge hindering the clinical translation, and finally to report on the emerging technologies in the biomaterials field. On this basis, we identify promising research directions, with the hope of stimulating further debate and advances for resolving clinical problems such as cervical and low back pain using biomaterial-based approaches. STATEMENT OF SIGNIFICANCE Although not life-threatening, intervertebral disc disorders have enormous impact on life quality and disability. Disc function within the human body is mainly mechanical, and therefore the use of biomaterials to rescue disc function and alleviate pain is logical. Despite intensive research, the clinical translation of biomaterial-based therapies is hampered by the intrinsic complexity of this organ. After decades of development, artificial discs or tissue replacements are still niche applications given their issues of integration and displacement with detrimental consequences. The struggles of biological therapies and tissue engineering are therefore understandable. However, recent advances in biomaterial science give new hope. In this paper we identify the most promising new directions for intervertebral disc biomaterials.
Collapse
|
9
|
Stich S, Möller A, Cabraja M, Krüger JP, Hondke S, Endres M, Ringe J, Sittinger M. Chemokine CCL25 Induces Migration and Extracellular Matrix Production of Anulus Fibrosus-Derived Cells. Int J Mol Sci 2018; 19:ijms19082207. [PMID: 30060561 PMCID: PMC6121557 DOI: 10.3390/ijms19082207] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Revised: 07/23/2018] [Accepted: 07/26/2018] [Indexed: 12/17/2022] Open
Abstract
Intervertebral disc degeneration is a major source of back pain. For intervertebral disc regeneration after herniation a fast closure of anulus fibrosus (AF) defects is crucial. Here, the use of the C-C motif chemokine ligand 25 (CCL)25 in comparison to differentiation factors such as transforming growth factor (TGF)β3, bone morphogenetic protein (BMP)2, BMP7, BMP12, and BMP14 (all in concentrations of 10, 50 and 100 ng/mL) was tested in an in vitro micro mass pellet model with isolated and cultivated human AF-cells (n = 3) to induce and enhance AF-matrix formation. The pellets were differentiated (serum-free) with supplementation of the factors. After 28 days all used factors induced proteoglycan production (safranin O staining) and collagen type I production (immunohistochemical staining) in at least one of the tested concentrations. Histomorphometric scoring revealed that TGFβ3 delivered the strongest induction of proteoglycan production in all three concentrations. Furthermore, it was the only factor able to facilitate collagen type II production, even higher than in native tissue samples. CCL25 was also able to induce proteoglycan and collagen type I production comparable to several BMPs. CCL25 could additionally induce migration of AF-cells in a chemotaxis assay and therefore possibly aid in regeneration processes after disc herniation by recruiting AF-cells.
Collapse
Affiliation(s)
- Stefan Stich
- Tissue Engineering Laboratory, Berlin-Brandenburg Center for Regenerative Therapies, and Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universitätzu Berlin and Berlin Institute of Health, 10117 Berlin, Germany.
| | - Anke Möller
- Tissue Engineering Laboratory, Berlin-Brandenburg Center for Regenerative Therapies, and Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universitätzu Berlin and Berlin Institute of Health, 10117 Berlin, Germany.
| | - Mario Cabraja
- Department of Spinal Surgery, VivantesAuguste-Viktoria-Hospital, 12157 Berlin, Germany.
| | | | - Sylvia Hondke
- TransTissue Technologies GmbH, 10117 Berlin, Germany.
| | | | - Jochen Ringe
- Tissue Engineering Laboratory, Berlin-Brandenburg Center for Regenerative Therapies, and Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universitätzu Berlin and Berlin Institute of Health, 10117 Berlin, Germany.
| | - Michael Sittinger
- Tissue Engineering Laboratory, Berlin-Brandenburg Center for Regenerative Therapies, and Department of Rheumatology and Clinical Immunology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universitätzu Berlin and Berlin Institute of Health, 10117 Berlin, Germany.
| |
Collapse
|
10
|
Schubert AK, Smink JJ, Arp M, Ringe J, Hegewald AA, Sittinger M. Quality Assessment of Surgical Disc Samples Discriminates Human Annulus Fibrosus and Nucleus Pulposus on Tissue and Molecular Level. Int J Mol Sci 2018; 19:ijms19061761. [PMID: 29899321 PMCID: PMC6032144 DOI: 10.3390/ijms19061761] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 06/11/2018] [Accepted: 06/11/2018] [Indexed: 01/07/2023] Open
Abstract
A discrimination of the highly specialised annulus fibrosus (AF) and nucleus pulposus (NP) cells in the mature human intervertebral disc (IVD) is thus far still not possible in a reliable way. The aim of this study was to identify molecular markers that distinguish AF and NP cells in human disc tissue using microarray analysis as a screening tool. AF and NP samples were obtained from 28 cervical discs. First, all samples underwent quality sorting using two novel scoring systems for small-sized disc tissue samples including macroscopic, haptic and histological evaluation. Subsequently, samples with clear disc characteristics of either AF or NP that were free from impurities of foreign tissue (IVD score) and with low signs of disc degeneration on cellular level (DD score) were selected for GeneChip analysis (HGU1332P). The 11 AF and 9 NP samples showed distinctly different genome-wide transcriptomes. The majority of differentially expressed genes (DEGs) could be specifically assigned to the AF, whereas no DEG was exclusively expressed in the NP. Nevertheless, we identified 11 novel marker genes that clearly distinguished AF and NP, as confirmed by quantitative gene expression analysis. The novel established scoring systems and molecular markers showed the identity of AF and NP in disc starting material and are thus of great importance in the quality assurance of cell-based therapeutics in regenerative treatment of disc degeneration.
Collapse
Affiliation(s)
- Ann-Kathrin Schubert
- Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Tissue Engineering Laboratory and Berlin-Brandenburg Center for Regenerative Therapies, 13353 Berlin, Germany.
- CO.DON AG, 14513 Teltow, Germany.
| | | | - Mirko Arp
- Department of Neurosurgery, University Medical Center Mannheim, Heidelberg University, 68167 Mannheim, Germany.
| | - Jochen Ringe
- Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Tissue Engineering Laboratory and Berlin-Brandenburg Center for Regenerative Therapies, 13353 Berlin, Germany.
| | - Aldemar A Hegewald
- Department of Neurosurgery, University Medical Center Mannheim, Heidelberg University, 68167 Mannheim, Germany.
- Department of Neurosurgery and Spine Surgery, Helios Baltic Sea Hospital Damp, 24351 Damp, Germany.
| | - Michael Sittinger
- Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Tissue Engineering Laboratory and Berlin-Brandenburg Center for Regenerative Therapies, 13353 Berlin, Germany.
| |
Collapse
|
11
|
Furman BD, Kent CL, Huebner JL, Kraus VB, McNulty AL, Guilak F, Olson SA. CXCL10 is upregulated in synovium and cartilage following articular fracture. J Orthop Res 2018; 36:1220-1227. [PMID: 28906016 PMCID: PMC5851826 DOI: 10.1002/jor.23735] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 09/06/2017] [Indexed: 02/04/2023]
Abstract
The objective of this study was to investigate the expression of the chemokine CXCL10 and its role in joint tissues following articular fracture. We hypothesized that CXCL10 is upregulated following articular fracture and contributes to cartilage degradation associated with post-traumatic arthritis (PTA). To evaluate CXCL10 expression following articular fracture, gene expression was quantified in synovial tissue from knee joints of C57BL/6 mice that develop PTA following articular fracture, and MRL/MpJ mice that are protected from PTA. CXCL10 protein expression was assessed in human cartilage in normal, osteoarthritic (OA), and post-traumatic tissue using immunohistochemistry. The effects of exogenous CXCL10, alone and in combination with IL-1, on porcine cartilage explants were assessed by quantifying the release of catabolic mediators. Synovial tissue gene expression of CXCL10 was upregulated by joint trauma, peaking one day in C57BL/6 mice (25-fold) versus 3 days post-fracture in MRL/MpJ mice (15-fold). CXCL10 protein in articular cartilage was most highly expressed following trauma compared with normal and OA tissue. In a dose dependent manner, exogenous CXCL10 significantly reduced total matrix metalloproteinase (MMP) and aggrecanase activity of culture media from cartilage explants. CXCL10 also trended toward a reduction in IL-1α-stimulated total MMP activity (p = 0.09) and S-GAG (p = 0.09), but not NO release. In conclusion, CXCL10 was upregulated in synovium and chondrocytes following trauma. However, exogenous CXCL10 did not induce a catabolic response in cartilage. CXCL10 may play a role in modulating the chondrocyte response to inflammatory stimuli associated with joint injury and the progression of PTA. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:1220-1227, 2018.
Collapse
Affiliation(s)
- Bridgette D. Furman
- Department of Orthopaedic Surgery, Duke University Medical Center, Durham, NC 27710
| | - Collin L. Kent
- Department of Orthopaedic Surgery, Duke University Medical Center, Durham, NC 27710
| | | | | | - Amy L. McNulty
- Department of Orthopaedic Surgery, Duke University Medical Center, Durham, NC 27710
| | - Farshid Guilak
- Department of Orthopaedic Surgery, Washington University in St. Louis, St. Louis, MO 63110,Shriners’ Hospital for Children-St. Louis, St. Louis, MO 63110
| | - Steven A. Olson
- Department of Orthopaedic Surgery, Duke University Medical Center, Durham, NC 27710
| |
Collapse
|
12
|
Liu W, Liu D, Zheng J, Shi P, Chou PH, Oh C, Chen D, An HS, Chee A. Annulus fibrosus cells express and utilize C-C chemokine receptor 5 (CCR5) for migration. Spine J 2017; 17:720-726. [PMID: 28108404 PMCID: PMC5673099 DOI: 10.1016/j.spinee.2017.01.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 12/12/2016] [Accepted: 01/12/2017] [Indexed: 02/03/2023]
Abstract
BACKGROUND CONTEXT Disc degeneration is associated with the progressive loss of the proteoglycan content of the intervertebral disc, decreased matrix synthesis, higher concentrations of proteolytic enzymes, and increased levels of proinflammatory cytokines. In previous studies, we have shown that C-C chemokine ligand (CCL)2, CCL3, and CCL5 are highly expressed by cultured nucleus pulposus (NP) and annulus fibrosus (AF) cells that have been treated by interleukin-1. The major function of these chemokines is to recruit immune cells into the disc. It is unclear if disc cells can respond to these chemokines. Recent studies by Phillips et al. (2015) showed that NP cells express a number of cytokines and chemokine receptors. PURPOSE The purpose of this study is to determine the gene and protein expression of C-C chemokine receptor (CCR)1, CCR2, and CCR5 in NP and AF cells, and to test if these receptors can respond to their ligands in these cells by cell signaling and migration. STUDY DESIGN/SETTING This is an in vitro study. METHODS For RNA, surface expression, and cell signaling studies, human cells were isolated from the NP and AF tissues collected after spine surgery or from donated spine segments (Gift of Hope Human Donor & Tissue Network of Illinois) and cultured in monolayer. The gene expression of human CCR1, CCR2, and CCR5 was analyzed using real-time polymerase chain reaction. The surface expression of CCR1, CCR2, and CCR5 was analyzed using flow cytometry and fluorescently tagged antibodies specific for these proteins. Extracellular signal-regulated kinase (ERK) phosphorylation was analyzed from the cell lysates of NP and AF cells treated with CCL2 and CCL5 for 1 hour using enzyme-linked immunosorbent assay. Migration of primary rabbit AF cells was assayed using 8-µm Corning Transwell inserts in the presence or absence of CCL5. This study was partially funded by a North American Spine Society 2014 Basic Research Grant Award ($50,000). RESULTS RNA analysis showed that gene expression of CCR1, CCR2, and CCR5 was evident in human NP and AF cells (n=6). Only a small population of NP and AF cells expressed CCR1 (1.9% and 1.2%, respectively) and CCR2 (0.8% and 1.4%, respectively) on the cell surface, whereas a larger percentage expressed CCR5 (12.7% and 11.6%, respectively). Significantly higher levels of ERK phosphorylation were detected in AF cells after treatment with CCL5 and not CCL2. Treatment with either chemokine did not cause significantly higher ERK phosphorylation in NP cells. There was an increase in average AF cell migration in the presence of CCL5. The increase was significant when the migration was induced with CCL5 (500 ng/mL) at both 2- and 6-hour time points. CONCLUSIONS CCR5 is expressed at the RNA level and on the cell surface of NP and AF cells. In the presence of CCL5, we detected increased levels of ERK phosphorylation and AF cell migration, suggesting that the CCR5 receptors in AF cells are functional. These data suggest that AF cells may have the ability to migrate in response to disc damage or inflammation.
Collapse
Affiliation(s)
- Weijun Liu
- Department of Orthopedic Surgery, Rush University Medical Center, 1611 W Harrison St, Suite 300, Chicago, IL 60612, USA,Department of Orthopedics, Wuhan Pu’Ai Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 473 Hanzheng St, Wuhan 430033, China
| | - David Liu
- Department of Orthopedic Surgery, Rush University Medical Center, 1611 W Harrison St, Suite 300, Chicago, IL 60612, USA,College of Pharmacy, Rosalind Franklin University of Medicine and Science, 3333 Green Bay Rd, North Chicago, IL 60064, USA
| | - Justin Zheng
- Department of Orthopedic Surgery, Rush University Medical Center, 1611 W Harrison St, Suite 300, Chicago, IL 60612, USA,University of Illinois College of Medicine at Peoria, 1 Illini Dr, Peoria, IL 61605, USA
| | - Peng Shi
- Department of Orthopedic Surgery, Rush University Medical Center, 1611 W Harrison St, Suite 300, Chicago, IL 60612, USA
| | - Po-Hsin Chou
- Department of Orthopedic Surgery, Rush University Medical Center, 1611 W Harrison St, Suite 300, Chicago, IL 60612, USA,Department of Orthopaedic and Traumatology, Taipei Veterans General Hospital, No. 201, Section 2, Shipai Rd, Beitou District, Taipei City 112, Taiwan,School of Medicine, National Yang-Ming University, No. 155, Section 2, Linong St, Taipei City 112, Taiwan
| | - Chundo Oh
- Department of Biochemistry, Rush University Medical Center, 1735 W. Harrison St, Chicago, IL 60612, USA
| | - Di Chen
- Department of Orthopedic Surgery, Rush University Medical Center, 1611 W Harrison St, Suite 300, Chicago, IL 60612, USA,Department of Biochemistry, Rush University Medical Center, 1735 W. Harrison St, Chicago, IL 60612, USA
| | - Howard S. An
- Department of Orthopedic Surgery, Rush University Medical Center, 1611 W Harrison St, Suite 300, Chicago, IL 60612, USA,Corresponding author. Department of Orthopedic Surgery, Rush University Medical Center, 1611 W Harrison St, Suite 300, Chicago, IL 60612, USA. Tel.: (312) 432-2354; fax: (708) 492-5354. (H.S. An)
| | - Ana Chee
- Department of Orthopedic Surgery, Rush University Medical Center, 1611 W Harrison St, Suite 300, Chicago, IL 60612, USA
| |
Collapse
|
13
|
Gruber HE, Marrero E, Ingram JA, Hoelscher GL, Hanley EN. The chemokine, CXCL16, and its receptor, CXCR6, are constitutively expressed in human annulus fibrosus and expression of CXCL16 is up-regulated by exposure to IL-1ß in vitro. Biotech Histochem 2016; 92:7-14. [PMID: 27869573 DOI: 10.1080/10520295.2016.1237672] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Chemokines are an important group of soluble molecules with specialized functions in inflammation. The roles of many specialized chemokines and their receptors remain poorly understood in the human intervertebral disc. We investigated CXCL16 and its receptor, CXCR6, to determine their immunolocalization in disc tissue and their presence following exposure of cultured human annulus fibrosus cells to proinflammatory cytokines. CXCL16 is a marker for inflammation; it also can induce hypoxia-inducible factor 1α (HIF-1α), which is a phenotypic marker of heathy nucleus pulposus tissue. We found CXCL16 and CXCR6 immunostaining in many cells of the annulus portion of the disc. Molecular studies showed that annulus fibrosus cells exposed to IL-1ß, but not TNF-α, exhibited significant up-regulation of CXCL16 expression vs. control cells. There was no significant difference in the percentage of annulus cells that exhibited immunolocalization of CXCL16 in grade I/II, grade III or grade IV/V specimens. The presence of CXCL16 and its receptor, CXCR6, in the annulus in vivo suggests the need for future research concerning the role of this chemokine in proinflammatory functions, HIF-1α expression and disc vascularization.
Collapse
Affiliation(s)
- H E Gruber
- a Department of Orthopaedic Surgery , Carolinas Medical Center , Charlotte , North Carolina
| | - E Marrero
- a Department of Orthopaedic Surgery , Carolinas Medical Center , Charlotte , North Carolina
| | - J A Ingram
- a Department of Orthopaedic Surgery , Carolinas Medical Center , Charlotte , North Carolina
| | - G L Hoelscher
- a Department of Orthopaedic Surgery , Carolinas Medical Center , Charlotte , North Carolina
| | - E N Hanley
- a Department of Orthopaedic Surgery , Carolinas Medical Center , Charlotte , North Carolina
| |
Collapse
|
14
|
Phillips KLE, Cullen K, Chiverton N, Michael ALR, Cole AA, Breakwell LM, Haddock G, Bunning RAD, Cross AK, Le Maitre CL. Potential roles of cytokines and chemokines in human intervertebral disc degeneration: interleukin-1 is a master regulator of catabolic processes. Osteoarthritis Cartilage 2015; 23:1165-77. [PMID: 25748081 DOI: 10.1016/j.joca.2015.02.017] [Citation(s) in RCA: 119] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Revised: 12/23/2014] [Accepted: 02/13/2015] [Indexed: 02/02/2023]
Abstract
OBJECTIVE These studies investigated cytokine and chemokine receptor profiles in nucleus pulposus (NP) cells, and the effects of receptor stimulation on mRNA levels of extracellular matrix (ECM) components, degrading enzymes and cytokine and chemokine expression. METHOD Immunohistochemistry (IHC) was performed to localise expression of CD4, CCR1, CXCR1 and CXCR2 in human NP tissue samples. Effects of cytokine and chemokine stimulation was performed to investigate effects related to ECM remodelling and modulation of cytokine and chemokine mRNA expression. RESULTS IHC identified CD4, CCR1, CXCR1 and CXCR2 expression by NP cells. Differential expression profiles were observed for CD4 and CXCR2 in tissue samples from degenerate and infiltrated IVDs. In vitro stimulations of primary human NP cultures with IL-16, CCL2, CCL3, CCL7 or CXCL8 did not identify any modulatory effects on parameters associated with ECM remodelling or expression of other cytokines and chemokines. Conversely, IL-1 was seen to modulate ECM remodelling and expression of all other cytokines and chemokines investigated. CONCLUSION This study demonstrates for the first time that NP cells express a number of cytokine and chemokine receptors and thus could respond in an autocrine or paracrine manner to cytokines and chemokines produced by NP cells, particularly during tissue degeneration. However, this study failed to demonstrate regulatory effects on ECM genes and degradative enzymes or other cytokines and chemokines for any target investigated, with the exception of IL-1. This suggests that IL-1 is a master regulator within the IVD and may exert regulatory potential over a plethora of other cytokines and chemokines.
Collapse
Affiliation(s)
- K L E Phillips
- Biomedical Research Centre, Sheffield Hallam University, Sheffield, UK.
| | - K Cullen
- Transplant Immunology, St James's University Hospital, Leeds, UK.
| | - N Chiverton
- Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK.
| | - A L R Michael
- Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK.
| | - A A Cole
- Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK.
| | - L M Breakwell
- Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK.
| | - G Haddock
- Biomedical Research Centre, Sheffield Hallam University, Sheffield, UK.
| | - R A D Bunning
- Biomedical Research Centre, Sheffield Hallam University, Sheffield, UK.
| | - A K Cross
- Biomedical Research Centre, Sheffield Hallam University, Sheffield, UK.
| | - C L Le Maitre
- Biomedical Research Centre, Sheffield Hallam University, Sheffield, UK.
| |
Collapse
|
15
|
Endres M, Zenclussen M, Casalis P, Freymann U, Gil Garcia S, Krueger J, Thomale UW, Woiciechowsky C, Kaps C. Augmentation and repair tissue formation of the nucleus pulposus after partial nucleotomy in a rabbit model. Tissue Cell 2014; 46:505-13. [DOI: 10.1016/j.tice.2014.09.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Revised: 09/10/2014] [Accepted: 09/10/2014] [Indexed: 02/04/2023]
|
16
|
Hegewald AA, Cluzel J, Krüger JP, Endres M, Kaps C, Thomé C. Effects of initial boost with TGF-beta 1 and grade of intervertebral disc degeneration on 3D culture of human annulus fibrosus cells. J Orthop Surg Res 2014; 9:73. [PMID: 25116605 PMCID: PMC4143571 DOI: 10.1186/s13018-014-0073-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2014] [Accepted: 07/25/2014] [Indexed: 12/01/2022] Open
Abstract
Background Three-dimensional (3D) culture in porous biomaterials as well as stimulation with growth factors are known to be supportive for intervertebral disc cell differentiation and tissue formation. Unless sophisticated releasing systems are used, however, effective concentrations of growth factors are maintained only for a very limited amount of time in in vivo applications. Therefore, we investigated, if an initial boost with transforming growth factor-beta 1 (TGF-beta 1) is capable to induce a lasting effect of superior cartilaginous differentiation in slightly and severely degenerated human annulus fibrosus (AF) cells. Methods Human AF tissue was harvested during surgical treatment of six adult patients with lumbar spinal diseases. Grading of disc degeneration was performed with magnet resonance imaging. AF cells were isolated and expanded in monolayer culture and rearranged three-dimensionally in a porous biomaterial consisting of stepwise absorbable poly-glycolic acid and poly-(lactic-co-glycolic) acid and a supportive fine net of non-absorbable polyvinylidene fluoride. An initial boost of TGF-beta 1 or TGF-beta 1 and hyaluronan was applied and compared with controls. Matrix formation was assessed at days 7 and 21 by (1) histological staining of the typical extracellular matrix molecules proteoglycan and type I and type II collagens and by (2) real-time gene expression analysis of aggrecan, decorin, biglycan, type I, II, III, and X collagens as well as of catabolic matrix metalloproteinases MMP-2 and MMP-13. Results An initial boost with TGF-beta 1 or TGF-beta 1 and hyaluronan did not enhance the expression of characteristic AF matrix molecules in our 3D culture system. AF cells showed high viability in the progressively degrading biomaterial. Stratification by grade of intervertebral disc degeneration showed that AF cells from both, slightly degenerated, or severely degenerated tissue are capable of significant up-regulations of characteristic matrix molecules in 3D culture. AF cells from severely degenerated tissue, however, displayed significantly lower up-regulations in some matrix molecules such as aggrecan. Conclusions We failed to show a supportive effect of an initial boost with TGF-beta 1 in our 3D culture system. This underlines the need for further investigations on growth factor releasing systems.
Collapse
Affiliation(s)
- Aldemar Andres Hegewald
- Department of Neurosurgery, University Medical Center Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, Mannheim 68167, Germany.
| | | | | | | | | | | |
Collapse
|
17
|
Jin L, Liu Q, Scott P, Zhang D, Shen F, Balian G, Li X. Annulus fibrosus cell characteristics are a potential source of intervertebral disc pathogenesis. PLoS One 2014; 9:e96519. [PMID: 24796761 PMCID: PMC4010482 DOI: 10.1371/journal.pone.0096519] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Accepted: 04/08/2014] [Indexed: 01/07/2023] Open
Abstract
In the end stage of intervertebral disc degeneration, cartilage, bone, endothelial cells, and neurons appear in association with the worsening condition. The origin of the abnormal cells is not clear. This study investigated the properties of progenitor cells in the annulus fibrosus (AF) using one in vitro and two in vivo models. Cultivation of rabbit AF cells with chondrogenic media significantly increased expressions of collagen and aggrecan. Upon exposure to osteogenic conditions, the cultures showed increased mineralization and expression of osteopontin, runx2, and bmp2 genes. Two models were used in the in vivo subcutaneous implantation experiments: 1) rabbit AF tissue in a demineralized bone matrix (DBM) cylinder (DBM/AF), and, 2) rat intact and needle punctured lumbar discs. Bone formation in the AF tissue was detected and hypertrophic chondrocytes and osteoblasts were present 1 month after implantation of the DBM/AF to nude mice. In addition to collagen I and II, immunostaining shows collagen X and osteocalcin expression in DBM/AF specimens 4 months after implantation. Similar changes were detected in the injured discs. Almost the entire needle punctured disc had ossified at 6 months. The results suggest that AF cells have characteristics of progenitor cells and, under appropriate stimuli, are capable of differentiating into chondrocytes and osteoblasts in vitro as well as in vivo. Importantly, these cells may be a target for biological treatment of disc degeneration.
Collapse
Affiliation(s)
- Li Jin
- Department of Orthopaedic Surgery, University of Virginia, Charlottesville, Virginia, United States of America
- * E-mail:
| | - Qihai Liu
- Department of Orthopaedic Surgery, University of Virginia, Charlottesville, Virginia, United States of America
| | - Phillip Scott
- Department of Orthopaedic Surgery, University of Virginia, Charlottesville, Virginia, United States of America
| | - Dawei Zhang
- Department of Orthopaedic Surgery, University of Virginia, Charlottesville, Virginia, United States of America
| | - Francis Shen
- Department of Orthopaedic Surgery, University of Virginia, Charlottesville, Virginia, United States of America
| | - Gary Balian
- Department of Orthopaedic Surgery, University of Virginia, Charlottesville, Virginia, United States of America
| | - Xudong Li
- Department of Orthopaedic Surgery, University of Virginia, Charlottesville, Virginia, United States of America
| |
Collapse
|
18
|
Hegewald AA, Medved F, Feng D, Tsagogiorgas C, Beierfuß A, Schindler GAK, 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 2013; 9:405-14. [DOI: 10.1002/term.1831] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2012] [Revised: 05/05/2013] [Accepted: 09/02/2013] [Indexed: 11/12/2022]
Affiliation(s)
- Aldemar Andres Hegewald
- Department of Neurosurgery University Medical Centre Mannheim, Heidelberg University Mannheim Germany
- Department of Neurosurgery Innsbruck Medical University Innsbruck Austria
| | - Fabian Medved
- Department of Neurosurgery University Medical Centre Mannheim, Heidelberg University Mannheim Germany
| | - Daxiong Feng
- Department of Neurosurgery University Medical Centre Mannheim, Heidelberg University Mannheim Germany
- Spinal Surgery Department The Affiliated Hospital of Luzhou Medical College Luzhou Sichuan Province P R China
| | - Charalambos Tsagogiorgas
- Department of Anaesthesiology and Critical Care Medicine University Medical Centre Mannheim, Heidelberg University Mannheim Germany
| | - Anja Beierfuß
- Department of Small Animal Surgery and Ophthalmology Ludwig Maximilian's University Munich Munich Germany
| | | | - Marcus Trunk
- Institute of Pathology University Medical Centre Mannheim, Heidelberg University Mannheim Germany
| | | | - Demissew Shenegelegn Mern
- Department of Neurosurgery University Medical Centre Mannheim, Heidelberg University Mannheim Germany
| | - Claudius Thomé
- Department of Neurosurgery Innsbruck Medical University Innsbruck Austria
| |
Collapse
|
19
|
The challenge and advancement of annulus fibrosus tissue engineering. EUROPEAN SPINE JOURNAL : OFFICIAL PUBLICATION OF THE EUROPEAN SPINE SOCIETY, THE EUROPEAN SPINAL DEFORMITY SOCIETY, AND THE EUROPEAN SECTION OF THE CERVICAL SPINE RESEARCH SOCIETY 2013; 22:1090-100. [PMID: 23361531 DOI: 10.1007/s00586-013-2663-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2012] [Revised: 12/26/2012] [Accepted: 01/07/2013] [Indexed: 02/07/2023]
Abstract
BACKGROUND Intervertebral disc degeneration, a main cause of back pain, is an endemic problem and a big economic burden for the health care system. Current treatments are symptom relieving but do not address underlying problems-biological and structural deterioration of the disc. Tissue engineering is an emerging approach for the treatment of intervertebral disc degeneration since it restores the functionality of native tissues. Although numerous studies have focused on the nucleus pulposus tissue engineering and achieved successes in laboratory settings, disc tissue engineering without annulus fibrosus for the end stage of disc degeneration is deemed to fail. The purpose of this article is to review the advancement of annulus fibrosus tissue engineering. MATERIAL AND METHODS Relevant articles regarding annulus fibrosus tissue engineering were identified in PubMed and Medline databases. RESULTS The ideal strategy for disc regeneration is to restore the function and integrity of the disc by using biomaterials, native matrices, growth factors, and cells that producing matrices. In the past decades there are tremendous advancement in annulus fibrosus tissue engineering including cell biology, biomaterials, and whole disc replacement. The recent promising results on whole disc tissue engineering-a composite of annulus fibrosus and nucleus pulposus-make the tissue engineering approach more appealing. CONCLUSION Despite the promising results in disc tissue engineering, there is still much work to be done regarding the clinical application.
Collapse
|
20
|
Guterl CC, See EY, Blanquer SB, Pandit A, Ferguson SJ, Benneker LM, Grijpma DW, Sakai D, Eglin D, Alini M, Iatridis JC, Grad S. Challenges and strategies in the repair of ruptured annulus fibrosus. Eur Cell Mater 2013; 25:1-21. [PMID: 23283636 PMCID: PMC3655691 DOI: 10.22203/ecm.v025a01] [Citation(s) in RCA: 163] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Lumbar discectomy is the surgical procedure most frequently performed for patients suffering from low back pain and sciatica. Disc herniation as a consequence of degenerative or traumatic processes is commonly encountered as the underlying cause for the painful condition. While discectomy provides favourable outcome in a majority of cases, there are conditions where unmet requirements exist in terms of treatment, such as large disc protrusions with minimal disc degeneration; in these cases, the high rate of recurrent disc herniation after discectomy is a prevalent problem. An effective biological annular repair could improve the surgical outcome in patients with contained disc herniations but otherwise minor degenerative changes. An attractive approach is a tissue-engineered implant that will enable/stimulate the repair of the ruptured annulus. The strategy is to develop three-dimensional scaffolds and activate them by seeding cells or by incorporating molecular signals that enable new matrix synthesis at the defect site, while the biomaterial provides immediate closure of the defect and maintains the mechanical properties of the disc. This review is structured into (1) introduction, (2) clinical problems, current treatment options and needs, (3) biomechanical demands, (4) cellular and extracellular components, (5) biomaterials for delivery, scaffolding and support, (6) pre-clinical models for evaluation of newly developed cell- and material-based therapies, and (7) conclusions. This article highlights that an interdisciplinary approach is necessary for successful development of new clinical methods for annulus fibrosus repair. This will benefit from a close collaboration between research groups with expertise in all areas addressed in this review.
Collapse
Affiliation(s)
- Clare C. Guterl
- Department of Orthopaedics, Mount Sinai Medical Centre, New York, NY, USA,Collaborative Research Partner Annulus Fibrosus Rupture Program of AO Foundation, Davos, Switzerland
| | - Eugene Y. See
- Network of Excellence for Functional Biomaterials, National University of Ireland, Galway, Ireland,Collaborative Research Partner Annulus Fibrosus Rupture Program of AO Foundation, Davos, Switzerland
| | - Sebastien B.G. Blanquer
- Department of Biomaterials Science and Technology, University of Twente, Enschede, The Netherlands,Collaborative Research Partner Annulus Fibrosus Rupture Program of AO Foundation, Davos, Switzerland
| | - Abhay Pandit
- Network of Excellence for Functional Biomaterials, National University of Ireland, Galway, Ireland,Collaborative Research Partner Annulus Fibrosus Rupture Program of AO Foundation, Davos, Switzerland
| | - Stephen J. Ferguson
- Institute for Biomechanics, ETH Zurich, Zurich, Switzerland,Collaborative Research Partner Annulus Fibrosus Rupture Program of AO Foundation, Davos, Switzerland
| | - Lorin M. Benneker
- Department of Orthopaedic Surgery, University of Bern, Bern, Switzerland,Collaborative Research Partner Annulus Fibrosus Rupture Program of AO Foundation, Davos, Switzerland
| | - Dirk W. Grijpma
- Department of Biomaterials Science and Technology, University of Twente, Enschede, The Netherlands,Department of Biomedical Engineering, University Medical Centre Groningen and University of Groningen, Groningen, The Netherlands,Collaborative Research Partner Annulus Fibrosus Rupture Program of AO Foundation, Davos, Switzerland
| | - Daisuke Sakai
- Department of Orthopaedic Surgery, Tokai University School of Medicine, Isehara, Kanagawa, Japan,Collaborative Research Partner Annulus Fibrosus Rupture Program of AO Foundation, Davos, Switzerland
| | - David Eglin
- AO Research Institute Davos, Davos, Switzerland,Collaborative Research Partner Annulus Fibrosus Rupture Program of AO Foundation, Davos, Switzerland
| | - Mauro Alini
- AO Research Institute Davos, Davos, Switzerland,Collaborative Research Partner Annulus Fibrosus Rupture Program of AO Foundation, Davos, Switzerland
| | - James C. Iatridis
- Department of Orthopaedics, Mount Sinai Medical Centre, New York, NY, USA,Collaborative Research Partner Annulus Fibrosus Rupture Program of AO Foundation, Davos, Switzerland
| | - Sibylle Grad
- AO Research Institute Davos, Davos, Switzerland,Collaborative Research Partner Annulus Fibrosus Rupture Program of AO Foundation, Davos, Switzerland,Address for correspondence: Sibylle Grad, PhD, AO Research Institute Davos, Clavadelerstrasse 8, 7270 Davos, Switzerland, Telephone Number: +41 81 414 2480, FAX Number: +41 81 414 2288,
| |
Collapse
|
21
|
Towards biological anulus repair: TGF-β3, FGF-2 and human serum support matrix formation by human anulus fibrosus cells. Tissue Cell 2012; 45:68-76. [PMID: 23122986 DOI: 10.1016/j.tice.2012.09.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2012] [Revised: 09/13/2012] [Accepted: 09/23/2012] [Indexed: 01/04/2023]
Abstract
Closure and biological repair of anulus fibrosus (AF) defects in intervertebral disc diseases is a therapeutic challenge. The aim of our study was to evaluate the anabolic properties of bioactive factors on cartilaginous matrix formation by AF cells. Human AF cells were harvested from degenerated lumbar AF tissue and expanded in monolayer culture. AF cell differentiation and matrix formation was initiated by forming pellet cultures and stimulation with hyaluronic acid (HA), human serum (HS), fibroblast growth factor-2 (FGF-2), transforming growth factor-β3 (TGF-β3) and TGF-β3/FGF-2 for up to 4 weeks. Matrix formation was assessed histologically by staining of proteoglycan, type I and type II collagens and by gene expression analysis of typical extracellular matrix molecules and of catabolic matrix metalloproteinases MMP-2 and MMP-13. AF cells, stimulated with HS, FGF-2 and most pronounced with TGF-β3 or TGF-β3/FGF-2 formed a cartilaginous matrix with significantly enhanced expression of matrix molecules and of MMP-13. Stimulation of AF cells with TGF-β3 was accompanied by induction of type X collagen, known to occur in hypertrophic cartilage cells having mineralizing potential. HA did not show any chondro-inductive characteristics. These findings suggest human serum, FGF-2 and TGF-β3 as possible candidates to support biological treatment strategies of AF defects.
Collapse
|
22
|
Woiciechowsky C, Abbushi A, Zenclussen ML, Casalis P, Krüger JP, Freymann U, Endres M, Kaps C. Regeneration of nucleus pulposus tissue in an ovine intervertebral disc degeneration model by cell-free resorbable polymer scaffolds. J Tissue Eng Regen Med 2012; 8:811-20. [PMID: 22865642 DOI: 10.1002/term.1582] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2011] [Revised: 05/25/2012] [Accepted: 06/26/2012] [Indexed: 01/07/2023]
Abstract
Degeneration of intervertebral discs (IVDs) occurs frequently and is often associated with lower back pain. Recent treatment options are limited and treat the symptoms rather than regenerate the degenerated disc. Cell-free, freeze-dried resorbable polyglycolic acid (PGA)-hyaluronan implants were used in an ovine IVD degeneration model. The nucleus pulposus of the IVD was partially removed, endoscopically. PGA-hyaluronan implants were immersed in autologous sheep serum and implanted into the disc defect. Animals with nucleotomy only served as controls. The T2-weighted/fat suppression sequence signal intensity index of the operated discs, as assessed by magnetic resonance imaging (MRI), showed that implantation of the PGA-hyaluronan implant improved (p = 0.0066) the MRI signal compared to controls at 6 months after surgery. Histological analysis by haematoxylin and eosin and safranin O staining showed the ingrowth of cells with typical chondrocytic morphology, even cell distribution, and extracellular matrix rich in proteoglycan. Histomorphometric analyses confirmed that the implantation of the PGA-hyaluronan scaffolds improved (p = 0.027) the formation of regenerated tissue after nucleotomy. Disc heights remained stable in discs with nucleotomy only as well as after implantation of the implant. In conclusion, implantation of cell-free polymer-based implants after nucleotomy induces nucleus pulposus tissue regeneration and improves disc water content in the ovine model.
Collapse
Affiliation(s)
- Christian Woiciechowsky
- Tissue Engineering Laboratory, Campus Charité Mitte, Charité-Universitätsmedizin Berlin, Germany; Spine Centre Berlin, Germany
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Cabraja M, Endres M, Hegewald AA, Vetterlein S, Thomé C, Woiciechowsky C, Kaps C. A 3D environment for anulus fibrosus regeneration. J Neurosurg Spine 2012; 17:177-83. [PMID: 22632174 DOI: 10.3171/2012.4.spine111095] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT Biological repair strategies for the treatment of degenerated intervertebral discs are of growing interest. In addition to the development of nucleus pulposus implants to restore disc height and relieve pain, there is growing demand for an appropriate method for reconstructing the anulus fibrosus (AF). The aim of this pilot study was to evaluate the applicability of a resorbable 3D polymer of pure polyglycolic acid (PGA) combined with hyaluronan for the use in cell-free and cell-based regeneration and repair of the AF. METHODS Adult human AF cells were expanded in vitro using human serum and rearranged three dimensionally in hyaluronan-PGA scaffolds that were stabilized with fibrin for in vitro analyses. The capacity of dedifferentiated AF cells to redifferentiate was evaluated after 2 weeks of culture, using propidium iodide/fluorescein diacetate staining, gene expression analysis of typical marker genes, and histological staining of proteoglycans. RESULTS The propidium iodide/fluorescein diacetate staining demonstrated that vital human AF cells were evenly distributed within the construct. The induction of typical AF marker genes such as collagen Types I-III indicated the initiation of AF redifferentiation by 3D assembly in hyaluronan-PGA. Histological analysis of the constructs showed initial formation of an AF-like matrix comprising proteoglycans. CONCLUSIONS The results suggest that the 3D arrangement of human AF cells in resorbable hyaluronan-PGA scaffolds cultured in the presence of human serum is an excellent system for AF cell redifferentiation.
Collapse
Affiliation(s)
- Mario Cabraja
- Department of Neurosurgery, Charité University Medicine, Berlin
| | | | | | | | | | | | | |
Collapse
|