1
|
Dou Y, Zhang Y, Liu Y, Sun X, Liu X, Li B, Yang Q. Role of macrophage in intervertebral disc degeneration. Bone Res 2025; 13:15. [PMID: 39848963 PMCID: PMC11758090 DOI: 10.1038/s41413-024-00397-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 12/04/2024] [Accepted: 12/16/2024] [Indexed: 01/25/2025] Open
Abstract
Intervertebral disc degeneration is a degenerative disease where inflammation and immune responses play significant roles. Macrophages, as key immune cells, critically regulate inflammation through polarization into different phenotypes. In recent years, the role of macrophages in inflammation-related degenerative diseases, such as intervertebral disc degeneration, has been increasingly recognized. Macrophages construct the inflammatory microenvironment of the intervertebral disc and are involved in regulating intervertebral disc cell activities, extracellular matrix metabolism, intervertebral disc vascularization, and innervation, profoundly influencing the progression of disc degeneration. To gain a deeper understanding of the inflammatory microenvironment of intervertebral disc degeneration, this review will summarize the role of macrophages in the pathological process of intervertebral disc degeneration, analyze the regulatory mechanisms involving macrophages, and review therapeutic strategies targeting macrophage modulation for the treatment of intervertebral disc degeneration. These insights will be valuable for the treatment and research directions of intervertebral disc degeneration.
Collapse
Affiliation(s)
- Yiming Dou
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, 300211, China
| | - Yiming Zhang
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, 300211, China
- Clinical School of Orthopedics, Tianjin Medical University, Tianjin, 300070, China
| | - Yang Liu
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, 300211, China
| | - Xun Sun
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, 300211, China
| | - Xinyu Liu
- Department of Orthopaedics, Qilu Hospital of Shandong University, Jinan, 250012, China.
| | - Bin Li
- Orthopedic Institute, Department of Orthopedic Surgery, The First Affiliated Hospital, School of Biology & Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, 215007, China.
| | - Qiang Yang
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, 300211, China.
- Clinical School of Orthopedics, Tianjin Medical University, Tianjin, 300070, China.
| |
Collapse
|
2
|
Bermudez-Lekerika P, Crump KB, Wuertz-Kozak K, Le Maitre CL, Gantenbein B. Sulfated Hydrogels as Primary Intervertebral Disc Cell Culture Systems. Gels 2024; 10:330. [PMID: 38786247 PMCID: PMC11121347 DOI: 10.3390/gels10050330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 05/07/2024] [Accepted: 05/10/2024] [Indexed: 05/25/2024] Open
Abstract
The negatively charged extracellular matrix plays a vital role in intervertebral disc tissues, providing specific cues for cell maintenance and tissue hydration. Unfortunately, suitable biomimetics for intervertebral disc regeneration are lacking. Here, sulfated alginate was investigated as a 3D culture material due to its similarity to the charged matrix of the intervertebral disc. Precursor solutions of standard alginate, or alginate with 0.1% or 0.2% degrees of sulfation, were mixed with primary human nucleus pulposus cells, cast, and cultured for 14 days. A 0.2% degree of sulfation resulted in significantly decreased cell density and viability after 7 days of culture. Furthermore, a sulfation-dependent decrease in DNA content and metabolic activity was evident after 14 days. Interestingly, no significant differences in cell density and viability were observed between surface and core regions for sulfated alginate, unlike in standard alginate, where the cell number was significantly higher in the core than in the surface region. Due to low cell numbers, phenotypic evaluation was not achieved in sulfated alginate biomaterial. Overall, standard alginate supported human NP cell growth and viability superior to sulfated alginate; however, future research on phenotypic properties is required to decipher the biological properties of sulfated alginate in intervertebral disc cells.
Collapse
Affiliation(s)
- Paola Bermudez-Lekerika
- Tissue Engineering for Orthopaedics and Mechanobiology, Bone & Joint Program, Department for BioMedical Research (DBMR), Medical Faculty, University of Bern, 3008 Bern, Switzerland; (P.B.-L.); (K.B.C.)
- Graduate School for Cellular and Biomedical Sciences (GCB), University of Bern, 3012 Bern, Switzerland
| | - Katherine B. Crump
- Tissue Engineering for Orthopaedics and Mechanobiology, Bone & Joint Program, Department for BioMedical Research (DBMR), Medical Faculty, University of Bern, 3008 Bern, Switzerland; (P.B.-L.); (K.B.C.)
- Graduate School for Cellular and Biomedical Sciences (GCB), University of Bern, 3012 Bern, Switzerland
| | - Karin Wuertz-Kozak
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY 14623, USA;
- Spine Center, Schön Klinik München Harlaching Academic Teaching Hospital, Spine Research Institute, Paracelsus Private Medical University Salzburg (Austria), 81547 Munich, Germany
| | - Christine L. Le Maitre
- Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield S10 2TN, UK;
| | - Benjamin Gantenbein
- Tissue Engineering for Orthopaedics and Mechanobiology, Bone & Joint Program, Department for BioMedical Research (DBMR), Medical Faculty, University of Bern, 3008 Bern, Switzerland; (P.B.-L.); (K.B.C.)
- Inselspital, Department of Orthopedic Surgery & Traumatology, Medical Faculty, University of Bern, 3010 Bern, Switzerland
| |
Collapse
|
3
|
Chai Q, Zhang B, Da Y, Wang W, Gao Y, Yao M, Zhu H, Yang X, Zhu Y. Enhancement and Repair of Degenerative Intervertebral Disc in Rats Using Platelet-Rich Plasma/Ferulic Acid Hydrogel. Cartilage 2023; 14:506-515. [PMID: 36899464 PMCID: PMC10807731 DOI: 10.1177/19476035231157341] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 01/30/2023] [Accepted: 01/30/2023] [Indexed: 03/12/2023] Open
Abstract
BACKGROUND Intervertebral degenerative disc (IDD) disease is one of the most common clinical conditions causing low back pain. The main objective of this study was to investigate the repair effect of platelet-rich plasma (PRP) and ferulic acid (FA) hydrogel compound on degenerative discs in rats in combination with bioengineering technology, which may provide a strong theoretical basis for the future treatment of IDD. METHODS Forty-five male Sprague-Dawley rats were randomly divided into groups A-F; MRI was performed in each group at 0, 4, and 8 weeks after injection; and disc tissues were obtained after executing the animals. The histomorphology, apoptosis, and protein synthesis of intervertebral discs in each group were observed by hematoxylin-eosin, Masson, terminal deoxynucleotidyl transferase dUTP nick end labeling staining, and Western blot. RESULTS The release concentration of all groups reached the peak at 12 hours, and the highest concentration was found in the hydrogel/PRP/FA group at the same time. The MTT assay showed that hydrogel/PRP/FA is well-cytocompatible. The results of animal experiments show that hydrogel/PRP/FA has a good effect on degenerative intervertebral disc in rats. CONCLUSION PRP/FA-rich hydrogel compound plays an active role in promoting extracellular matrix synthesis, strengthening and repairing degenerated intervertebral discs in rats.
Collapse
Affiliation(s)
- Qiang Chai
- The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Baining Zhang
- The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Yifeng Da
- Department of Region A, Spinal Surgery Center, The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Wenlei Wang
- Department of Region B, Joint Surgery Center, The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Yidan Gao
- The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Mingyu Yao
- The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - He Zhu
- The Second Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Xuejun Yang
- Department of Orthopaedics, The Affiliated People’s Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Yong Zhu
- Department of Orthopaedics, The Affiliated People’s Hospital of Inner Mongolia Medical University, Hohhot, China
| |
Collapse
|
4
|
Tu Z, Han F, Zhu Z, Yu Q, Liu C, Bao Y, Li B, Zhou F. Sustained release of basic fibroblast growth factor in micro/nanofibrous scaffolds promotes annulus fibrosus regeneration. Acta Biomater 2023; 166:241-253. [PMID: 37230436 DOI: 10.1016/j.actbio.2023.05.034] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 05/14/2023] [Accepted: 05/18/2023] [Indexed: 05/27/2023]
Abstract
Tissue engineering has promising applications in the treatment of intervertebral disc degeneration (IDD). The annulus fibrosus (AF) is critical for maintaining the physiological function of the intervertebral disc (IVD), but the lack of vessels and nutrition in AF makes it difficult to repair. In this study, we used hyaluronan (HA) micro-sol electrospinning and collagen type I (Col-I) self-assembly techniques to fabricate layered biomimetic micro/nanofibrous scaffolds, which released basic fibroblast growth factor (bFGF) to promote AF repair and regeneration after discectomy and endoscopic transforaminal discectomy. The bFGF enveloped in the core of the poly-L-lactic-acid (PLLA) core-shell structure was released in a sustained manner and promoted the adhesion and proliferation of AF cells (AFCs). Col-I could self-assemble on the shell of the PLLA core-shell scaffold to mimic the extracellular matrix (ECM) microenvironment, providing structural and biochemical cues for the regeneration of AF tissue. The in vivo studies showed that the micro/nanofibrous scaffolds promoted the repair of AF defects by simulating the microstructure of native AF tissue and inducing endogenous regeneration mechanism. Taken together, the biomimetic micro/nanofibrous scaffolds have clinical potential for the treatment of AF defects caused by IDD. STATEMENT OF SIGNIFICANCE: The annulus fibrosus (AF) is essential for the intervertebral disc (IVD) physiological function, yet it lacks vascularity and nutrition, making repair difficult. Micro-sol electrospinning technology and collagen type I (Col-I) self-assembly technique were combined in this study to create a layered biomimetic micro/nanofibrous scaffold that releases basic fibroblast growth factor (bFGF) to promote AF repair and regeneration. Col-I could mimic the extracellular matrix (ECM) microenvironment, in vivo, offering structural and biochemical cues for AF tissue regeneration. This research indicates that micro/nanofibrous scaffolds have clinical potential for treating AF deficits induced by IDD.
Collapse
Affiliation(s)
- Zhengdong Tu
- Department of Orthopaedic Surgery, Orthopedic Institute, The First Affiliated Hospital, School of Biology & Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Feng Han
- Department of Orthopaedic Surgery, Orthopedic Institute, The First Affiliated Hospital, School of Biology & Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Zhuang Zhu
- Department of Orthopaedic Surgery, Orthopedic Institute, The First Affiliated Hospital, School of Biology & Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Qifan Yu
- Department of Orthopaedic Surgery, Orthopedic Institute, The First Affiliated Hospital, School of Biology & Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Changjiang Liu
- Department of Orthopaedic Surgery, Orthopedic Institute, The First Affiliated Hospital, School of Biology & Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Yu Bao
- Department of Orthopaedic Surgery, Orthopedic Institute, The First Affiliated Hospital, School of Biology & Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Bin Li
- Department of Orthopaedic Surgery, Orthopedic Institute, The First Affiliated Hospital, School of Biology & Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China.
| | - Feng Zhou
- Department of Orthopaedic Surgery, Orthopedic Institute, The First Affiliated Hospital, School of Biology & Basic Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
5
|
Du J, Long R, Nakai T, Sakai D, Benneker L, Zhou G, Li B, Eglin D, Iatridis J, Alini M, Grad S, Li Z. Functional cell phenotype induction with TGF-β1 and collagen-polyurethane scaffold for annulus fibrosus rupture repair. Eur Cell Mater 2020; 39:1-17. [PMID: 31899537 PMCID: PMC7027376 DOI: 10.22203/ecm.v039a01] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Appropriate cell sources, bioactive factors and biomaterials for generation of functional and integrated annulus fibrosus (AF) tissue analogues are still an unmet need. In the present study, the AF cell markers, collagen type I, cluster of differentiation 146 (CD146), mohawk (MKX) and smooth muscle protein 22α (SM22α) were found to be suitable indicators of functional AF cell induction. In vitro 2D culture of human AF cells showed that transforming growth factor β1 (TGF-β1) upregulated the expression of the functional AF markers and increased cell contractility, indicating that TGF-β1-pre-treated AF cells were an appropriate cell source for AF tissue regeneration. Furthermore, a tissue engineered construct, composed of polyurethane (PU) scaffold with a TGF-β1-supplemented collagen type I hydrogel and human AF cells, was evaluated with in vitro 3D culture and ex vivo preclinical bioreactor-loaded organ culture models. The collagen type I hydrogel helped maintaining the AF functional phenotype. TGF-β1 supplement within the collagen I hydrogel further promoted cell proliferation and matrix production of AF cells within in vitro 3D culture. In the ex vivo IVD organ culture model with physiologically relevant mechanical loading, TGF-β1 supplement in the transplanted constructs induced the functional AF cell phenotype and enhanced collagen matrix synthesis. In conclusion, TGF-β1-containing collagen-PU constructs can induce the functional cell phenotype of human AF cells in vitro and in situ. This combined cellular, biomaterial and bioactive agent therapy has a great potential for AF tissue regeneration and rupture repair.
Collapse
Affiliation(s)
- J. Du
- AO Research Institute Davos, Davos, Switzerland
| | - R.G. Long
- AO Research Institute Davos, Davos, Switzerland,Icahn School of Medicine at Mount Sinai, New York, USA,Collaborative Research Program Annulus Fibrosus Repair, AO Foundation, Davos, Switzerland
| | - T. Nakai
- Tokai University School of Medicine, Isehara, Japan,Collaborative Research Program Annulus Fibrosus Repair, AO Foundation, Davos, Switzerland
| | - D. Sakai
- Tokai University School of Medicine, Isehara, Japan,Collaborative Research Program Annulus Fibrosus Repair, AO Foundation, Davos, Switzerland
| | - L.M. Benneker
- Inselspital, University of Bern, Bern, Switzerland,Collaborative Research Program Annulus Fibrosus Repair, AO Foundation, Davos, Switzerland
| | - G. Zhou
- Shenzhen Key Laboratory of Anti-Aging and Regenerative Medicine, Department of Medical Cell Biology and Genetics, Health Sciences Centre, Shenzhen University, Shenzhen, China
| | - B. Li
- Orthopaedic Institute, Medical College, Soochow University, Suzhou, Jiangsu, China
| | - D. Eglin
- AO Research Institute Davos, Davos, Switzerland,Collaborative Research Program Annulus Fibrosus Repair, AO Foundation, Davos, Switzerland
| | - J.C. Iatridis
- Icahn School of Medicine at Mount Sinai, New York, USA,Collaborative Research Program Annulus Fibrosus Repair, AO Foundation, Davos, Switzerland
| | - M. Alini
- AO Research Institute Davos, Davos, Switzerland,Collaborative Research Program Annulus Fibrosus Repair, AO Foundation, Davos, Switzerland
| | - S. Grad
- AO Research Institute Davos, Davos, Switzerland,Collaborative Research Program Annulus Fibrosus Repair, AO Foundation, Davos, Switzerland
| | - Z. Li
- AO Research Institute Davos, Davos, Switzerland,Collaborative Research Program Annulus Fibrosus Repair, AO Foundation, Davos, Switzerland,Address for correspondence: Zhen Li, PhD, AO Research Institute Davos, Clavadelerstrasse 8, 7270, Davos Platz, Switzerland. Telephone number: +41 814142325
| |
Collapse
|
6
|
Saiyin W, Li L, Zhang H, Lu Y, Qin C. Inactivation of FAM20B causes cell fate changes in annulus fibrosus of mouse intervertebral disc and disc defects via the alterations of TGF-β and MAPK signaling pathways. Biochim Biophys Acta Mol Basis Dis 2019; 1865:165555. [PMID: 31513834 PMCID: PMC7194007 DOI: 10.1016/j.bbadis.2019.165555] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 09/05/2019] [Accepted: 09/07/2019] [Indexed: 01/30/2023]
Abstract
Intervertebral disc (IVD) disorder is often caused by the defect of annulus fibrosus (AF), especially that of the outer AF. Studies about the mechanisms governing the development of the outer AF are needed for a better understanding of pathogenesis of IVD defects. Glycosaminoglycans (GAGs) are essential components of extracellular matrix (ECM) in AF. FAM20B is a newly identified xylose kinase that catalyzes the biosynthesis of GAGs. In this study, we created Fam20B conditional knockout (cKO) mice in which FAM20B was inactivated in type I collagen-expressing cells, the main type of cells in the outer AF of IVD. The cKO mice showed severe spine deformity and remarkable IVD defects associated with AF malformation. The AF of cKO mice had a lower level of chondroitin sulfate and heparan sulfate, and the outer AF cells lost their normal fibroblast-like morphology and acquired chondrocyte phenotypes, expressing a higher level of Sox 9 and type II collagen along with a reduced level of type I collagen. The level of phospho-Smad 2 and phospho-Smad 3, and that of scleraxis, a downstream target molecule of canonical TGF-β signaling pathway were significantly lower in the AF of cKO mice. The AF in cKO mice also manifested altered levels in the molecules associated with the activations of MAPK pathway; the changes included the increase of phospho-P38 and phospho-ERK and a decrease of phospho-JNK. These results indicate that FAM20B plays an essential role in the development of AF by regulating the TGF-β signaling and MAPK signaling pathways.
Collapse
Affiliation(s)
- Wuliji Saiyin
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX 75246, USA
| | - Lili Li
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX 75246, USA
| | - Hua Zhang
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX 75246, USA
| | - Yongbo Lu
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX 75246, USA
| | - Chunlin Qin
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX 75246, USA.
| |
Collapse
|
7
|
Liu C, Jin Z, Ge X, Zhang Y, Xu H. Decellularized Annulus Fibrosus Matrix/Chitosan Hybrid Hydrogels with Basic Fibroblast Growth Factor for Annulus Fibrosus Tissue Engineering. Tissue Eng Part A 2019; 25:1605-1613. [PMID: 30929614 PMCID: PMC6919252 DOI: 10.1089/ten.tea.2018.0297] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Low back pain caused by degenerative disc disease affects many people worldwide and brings huge economical burden. Thus, attentions have focused on annulus fibrosus (AF) tissue engineering for treatment of intervertebral disc degeneration. To engineer a functional replacement for the AF, it is important to fabricate scaffolds that mimic the structural and mechanical properties of native tissue. AF-derived stem cells are promising seed cells for AF tissue engineering due to their tissue specificity. In the present study, decellularized AF matrix (DAFM)/chitosan hybrid hydrogels were fabricated using genipin as a crosslinker. AF stem cells were cultured on hydrogel scaffolds with or without basic fibroblast growth factor (bFGF), and cell proliferation, morphology, gene expression, and AF tissue synthesis were examined. Overall, more collagen-I, collagen-II, and aggrecan were secreted by AF stem cells grown on hydrogels with bFGF compared to those without. These results support the application of DAFM/chitosan hybrid hydrogels as an appropriate candidate for AF tissue engineering. Furthermore, incorporation of bFGF into hydrogels promoted AF-related tissue synthesis.
Collapse
Affiliation(s)
- Chen Liu
- Department of Orthopaedics, Yijishan Hospital of Wannan Medical College, Wuhu, China
| | - Zhongxing Jin
- Department of Orthopaedics, Yijishan Hospital of Wannan Medical College, Wuhu, China
| | - Xin Ge
- Department of Orthopaedics, Yijishan Hospital of Wannan Medical College, Wuhu, China
| | - Yu Zhang
- Department of Orthopaedics, Yijishan Hospital of Wannan Medical College, Wuhu, China
| | - Hongguang Xu
- Department of Orthopaedics, Yijishan Hospital of Wannan Medical College, Wuhu, China
| |
Collapse
|
8
|
Comparison of polysaccharides in articular cartilage regeneration associated with chondrogenic and autophagy-related gene expression. Int J Biol Macromol 2019; 146:922-930. [PMID: 31726172 DOI: 10.1016/j.ijbiomac.2019.09.215] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 09/11/2019] [Accepted: 09/22/2019] [Indexed: 01/01/2023]
Abstract
Articular cartilage exhibits reduced self-healing following degeneration. This research evaluated the effects of hydrogels derived from various polysaccharides-gellan gum (GG), alginate, and agarose-on cartilage regeneration compared with that of hyaluronic acid (HA), which is commonly used in cartilage tissue engineering. Chondrocytes were isolated from the articular cartilage of New Zealand White (NZW) rabbits and stimulated with IL-1β followed by incubation with polysaccharides. The expressions of NF-κB and Cox-2 were decreased and those of IκBα, Sox-9, aggrecan, and type II collagen were increased in HA, GG, and Alginate groups. Osteochondral defects in NZW rabbits were treated with intra-articular polysaccharide injections; all except alginate resulted in tissue regeneration. Significant improvements were observed in cartilage regeneration in the GG and agarose groups. These results show that GG and agarose improve cartilage regeneration by suppressing inflammatory mediators and inducing cartilage formation and autophagy-related gene expression, indicating their potential for cartilage tissue engineering.
Collapse
|
9
|
Chen S, Liu S, Ma K, Zhao L, Lin H, Shao Z. TGF-β signaling in intervertebral disc health and disease. Osteoarthritis Cartilage 2019; 27:1109-1117. [PMID: 31132405 DOI: 10.1016/j.joca.2019.05.005] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 05/02/2019] [Accepted: 05/14/2019] [Indexed: 02/02/2023]
Abstract
OBJECTIVE This paper aims to provide a comprehensive review of the changing role of transforming growth factor-β (TGF-β) signaling in intervertebral disc (IVD) health and disease. METHODS A comprehensive literature search was performed using PubMed terms 'TGF-β' and 'IVD'. RESULTS TGF-β signaling is necessary for the development and growth of IVD, and can play a protective role in the restoration of IVD tissues by stimulating matrix synthesis, inhibiting matrix catabolism, inflammatory response and cell loss. However, excessive activation of TGF-β signaling is detrimental to the IVD, and inhibition of the aberrant TGF-β signaling can delay IVD degeneration. CONCLUSIONS Activation of TGF-β signaling has a promising treatment prospect for IVD degeneration, while excessive activation of TGF-β signaling may contribute to the progression of IVD degeneration. Studies aimed at elucidating the changing role of TGF-β signaling in IVD at different pathophysiological stages and its specific molecular mechanisms are needed, and these studies will contribute to safe and effective TGF-β signaling-based treatments for IVD degeneration.
Collapse
Affiliation(s)
- S Chen
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - S Liu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - K Ma
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - L Zhao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - H Lin
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Z Shao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
10
|
Wang HC, Jin CH, Kong J, Yu T, Guo JW, Hu YG, Liu Y. The research of transgenic human nucleus pulposus cell transplantation in the treatment of lumbar disc degeneration. Kaohsiung J Med Sci 2019; 35:486-492. [PMID: 31091017 DOI: 10.1002/kjm2.12084] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 04/21/2019] [Indexed: 01/13/2023] Open
Abstract
The present study determines whether the in vivo injection of TGFβ1 and CTGF mediated by AAV2 to transfect nucleus pulposus cells in degenerative lumbar discs can reverse the biological effects of rhesus lumbar disc degeneration. A total of 42 lumbar discs obtained from six rhesus monkeys were classified into three groups: experimental group, control group, and blank group. Degenerative lumbar discs were respectively injected with double gene-transfected human nucleus pulposus cells using minimally invasive techniques. Immumohistochemical staining, RT-PCR, and western blot were performed to observe the biological effects of double gene-transfected human nucleus pulposus cells in degenerative lumbar discs on rhesus lumbar disc degeneration. At 4, 8, and 12 weeks after the transplantation of nucleus pulposus cells, the expression levels of TGF-ß1, CTGF, proteoglycan mRNA, and type-II collagen were detected by RT-PCR. The values of immumohistochemical staining and RT-PCR in the experimental group increased at 8 weeks, decreased with time at 12 weeks, and remained greater than the values in the control group, and the differences were statistically significant (P < .05). The western blot revealed that the values in the experimental group decreased with time, but remained greater than those in the PBS control group and blank control group, and the differences were statistically significant (P < .05). The double gene-transfection of human nucleus pulposus cells in degenerative lumbar discs mediated by rAAV2 can be continuously expressed in vivo after transplantation in lumbar discs of rhesus monkeys, and promotes the synthesis of proteoglycan and type II collagen, achieving the treatment purpose.
Collapse
Affiliation(s)
- Hua-Cong Wang
- Department of Orthopedic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, People's Republic of China
| | - Cang-Hai Jin
- Department of Minimally Invasive Spine Surgery, Qingdao Municipal Hospital, East Branch, Qingdao, Shandong, People's Republic of China
| | - Jie Kong
- Department of Orthopedic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, People's Republic of China
| | - Tao Yu
- Department of Orthopedic Surgery, Rushan City People Hospital, Rushan, Shandong, P.R China
| | - Jian-Wei Guo
- Department of Orthopedic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, People's Republic of China
| | - You-Gu Hu
- Department of Orthopedic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, People's Republic of China
| | - Yong Liu
- Department of Orthopedic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, People's Republic of China
| |
Collapse
|
11
|
Ishiguro H, Kaito T, Yarimitsu S, Hashimoto K, Okada R, Kushioka J, Chijimatsu R, Takenaka S, Makino T, Sakai Y, Moriguchi Y, Otsuru S, Hart DA, Fujie H, Nakamura N, Yoshikawa H. Intervertebral disc regeneration with an adipose mesenchymal stem cell-derived tissue-engineered construct in a rat nucleotomy model. Acta Biomater 2019; 87:118-129. [PMID: 30690206 DOI: 10.1016/j.actbio.2019.01.050] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 01/23/2019] [Accepted: 01/24/2019] [Indexed: 01/08/2023]
Abstract
Low back pain results in more global disabilities than any other condition, and intervertebral disc (IVD) degeneration is commonly involved in the etiology. Supplementation of IVDs with reparative cells is a rational strategy to address such clinical problems. We have previously developed a scaffold-free tissue-engineered construct (TEC) as a novel cell therapy system for repair of articular cartilage and meniscus. We now show the regenerative potential of adipose mesenchymal stem cells derived TEC (ADSC-TEC) for IVD degeneration using a rat tail model of total nucleotomy. The regenerative efficacy of ASDC-TEC was investigated structurally and biomechanically up to 6 months after implantation. ADSC-TEC implantation into IVDs preserved the disc height, endplate, and annulus fibrosus structure, and showed similar biomechanical characteristics to the sham group at postoperative 6 weeks. The structure of regenerated IVD was maintained until 6 months. Furthermore, ADSC-TEC implantation attenuated the impact of age-related biomechanical deterioration when assessed at 6 months post-implantation. These results demonstrate that use of ADSC-TECs can be an effective treatment for IVD degeneration. STATEMENT OF SIGNIFICANCE: We developed adipose mesenchymal stem cell-derived scaffold-free tissue engineered construct (ADSC-TEC) as a novel cell therapy system. The ADSC-TEC implantation into a rat total-nucleotomized disc space regenerated intervertebral discs (IVDs) histologically and biomechanically. The regenerative capacity of the ADSC-TEC was exerted by its trophic effects on annulus fibrosus cells and the load-sharing effect at intervertebral space. Interestingly, the regenerated IVDs by the ADSC-TEC was less susceptible to the age-related deterioration than the IVDs of normal rats. Thus, the application of ADSC-TEC into the degenerated disc can be an alternative therapy for various disease associated with structural and functional failure of IVDs.
Collapse
|
12
|
Elsaadany M, Winters K, Adams S, Stasuk A, Ayan H, Yildirim-Ayan E. Equiaxial Strain Modulates Adipose-derived Stem Cell Differentiation within 3D Biphasic Scaffolds towards Annulus Fibrosus. Sci Rep 2017; 7:12868. [PMID: 28993681 PMCID: PMC5634474 DOI: 10.1038/s41598-017-13240-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 09/19/2017] [Indexed: 12/14/2022] Open
Abstract
Recurrence of intervertebral disc (IVD) herniation is the most important factor leading to chronic low back pain and subsequent disability after discectomy. Efficacious annulus fibrosus (AF) repair strategy that delivers cells and biologics to IVD injury site is needed to limit the progression of disc degeneration and promote disc self-regeneration capacities after discectomy procedures. In this study, a biphasic mechanically-conditioned scaffold encapsulated with human adipose-derived stem cells (ASCs) is studied as a potential treatment strategy for AF defects. Equiaxial strains and frequencies were applied to ASCs-encapsulated scaffolds to identify the optimal loading modality to induce AF differentiation. Equiaxial loading resulted in 2–4 folds increase in secretion of extracellular matrix proteins and the reorganization of the matrix fibers and elongations of the cells along the load direction. Further, the equiaxial load induced region-specific differentiation of ASCs within the inner and outer regions of the biphasic scaffolds. Gene expression of AF markers was upregulated with 5–30 folds within the equiaxially loaded biphasic scaffolds compared to unstrained samples. The results suggest that there is a specific value of equiaxial strain favorable to differentiate ASCs towards AF lineage and that ASCs-embedded biphasic scaffold can potentially be utilized to repair the AF defects.
Collapse
Affiliation(s)
| | - Kayla Winters
- Department of Bioengineering, University of Toledo, Toledo, OH, USA
| | - Sarah Adams
- Department of Bioengineering, University of Toledo, Toledo, OH, USA
| | - Alexander Stasuk
- Department of Bioengineering, University of Toledo, Toledo, OH, USA
| | - Halim Ayan
- Department of Bioengineering, University of Toledo, Toledo, OH, USA
| | - Eda Yildirim-Ayan
- Department of Bioengineering, University of Toledo, Toledo, OH, USA.
| |
Collapse
|
13
|
Zhang H, Li J, Duan D, She W, Wang L, Zhang F. The role of lncRNA MALAT1 in intervertebral degenerative disc disease. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2017; 10:10611-10617. [PMID: 31966403 PMCID: PMC6965762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 08/17/2017] [Indexed: 06/10/2023]
Abstract
Intervertebral degenerative disc disease (IDDD) is a common disease in clinic that causes pain and heavy financial burden on patients with poor prognosis. However, the pathogenesis of IDDD is not clear. Long non-coding RNA (LncRNA) is involved in regulating various body growth and pathological processes by affecting cell proliferation, differentiation, and apoptosis. However, the role of lncRNAs in IDDD is rarely reported. This study aims to investigate the role and mechanism of lncRNA MALAT1 in the development of IDDD. The nucleus pulposus of the intervertebral disc were collected and the primary nucleus pulposus cells were isolated and cultured. The cells were divided into three groups, including IDDD group, empty plasmid group transfected by pcDNA3.1, or MALAT1 group transfected by pcDNA3.1-MALAT1. MALAT1 expression was detected by real-time PCR. Cell proliferation was assessed by MTT assay. Caspase 3 activity was tested by the activity detection kit. IL-1 and IL-6 levels were analyzed by ELISA. The expression of MALAT1 in IDDD nucleus pulposus cells was significantly lower than that in control group (P < 0.05). The expression of MALAT1 was significantly increased after transfection with pcDNA3.1-MALAT1 plasmid in IDDD nucleus pulposus cells, which obviously inhibited cell proliferation, enhanced Caspase 3 activity, and promoted the secretion of IL-1 and IL-6 compared with IDDD group (P < 0.05). MALAT1 level decreased in IDDD nucleus pulposus cells. Upregulation of MALAT1 expression restrained IDDD through suppressing inflammation; inhibiting nucleus pulposus cell apoptosis, and promoting cell proliferation.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Orthopedics, Gansu Provincial People’s HospitalLanzhou, Gansu, China
| | - Jinde Li
- Department of Orthopedics, The Second People’s Hospital of Lanzhou CityLanzhou, Gansu, China
| | - Dapeng Duan
- Department of Orthopedics, Shaanxi Provincial People’s HospitalXi’an, Shaanxi, China
| | - Wei She
- Department of Orthopedics, Gansu Provincial People’s HospitalLanzhou, Gansu, China
| | - Liguo Wang
- Department of Orthopedics, Gansu Provincial People’s HospitalLanzhou, Gansu, China
| | - Fuqiang Zhang
- Department of Orthopedics, Gansu Provincial People’s HospitalLanzhou, Gansu, China
| |
Collapse
|
14
|
Zou G, Li Y, Jin Y, Zhu X, Yang J, Wang S, You Q, Xiong H, Liu Y. [ In vitrodifferentiation of human amniotic mesenchymal stem cells into ligament fibroblasts after induced by transforming growth factor β 1 and vascular endothelial growth factor]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2017; 31:582-593. [PMID: 29798549 PMCID: PMC8498243 DOI: 10.7507/1002-1892.201612090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 04/07/2017] [Indexed: 11/03/2022]
Abstract
Objective To investigate whether human amniotic mesenchymal stem cells (hAMSCs) have the characteristics of mesenchymal stem cells (MSCs) and the differentiation capacity into ligament fibroblasts in vitro. Methods The hAMSCs were separated through trypsin and collagenase digestion from placenta, the phenotypic characteristics of hAMSCs were detected by flow cytometry, the cytokeratin-19 (CK-19) and vimentin expression of hAMSCs were tested through immunofluorescence staining. The hAMSCs at the 3rd passage were cultured with L-DMEM/F12 medium containing transforming growth factor β 1 (TGF-β 1) and vascular endothelial growth factor (VEGF) as the experimental group and with single L-DMEM/F12 medium as the control group. The morphology of hAMSCs was observed by inverted phase contrast microscope; the cellular activities and ability of proliferation were examined by cell counting kit-8 (CCK-8) method; the ligament fibroblasts related protein expressions including collagen type I, collagen type III, Fibronectin, and Tenascin-C were detected by immunofluorescence staining; specific mRNA expressions of ligament fibroblasts and angiogenesis including collagen type I, collagen type III, Fibronectin, α-smooth muscle actin (α-SMA), and VEGF were measured by real-time fluorescence quantitative PCR. Results The hAMSCs presented monolayer and adherent growth under inverted phase contrast microscope; the flow cytometry results demonstrated that hAMSCs expressed the MSCs phenotypes; the immunofluorescence staining results indicated the hAMSCs had high expression of the vimentin and low expression of CK-19; the hAMSCs possessed the differentiation ability into the osteoblasts, chondroblasts, and lipoblasts. The CCK-8 results displayed that cells reached the peak of growth curve at 7 days in each group, and the proliferation ability in the experimental group was significantly higher than that in the control group at 7 days ( P<0.05). The immunofluorescence staining results showed that the expressions of collagen type I, collagen type III, Fibronectin, and Tenascin-C in the experimental group were significantly higher than those in the control group at 5, 10, and15 days after culture ( P<0.05). The real-time fluorescence quantitative PCR results revealed that the mRNA relative expressions had an increasing tendency at varying degrees with time in the experimental group ( P<0.05). The relative mRNA expressions of collagen type I, collagen type III, Fibronectin, α-SMA, and VEGF in the experimental group were significantly higher than those in the control group at the other time points ( P<0.05), but no significant difference was found in the relative mRNA expressions of collagen type I, collagen type III, and VEGF between 2 groups at 5 days ( P>0.05). Conclusion The hAMSCs possesses the characteristics of MSCs and good proliferation ability which could be chosen as seed cell source in tissue engineering. The expressions of ligament fibroblasts and angiogenesis related genes could be up-regulated, after induction in vitro, and the synthesis of ligament fibroblasts related proteins could be strengthened. In addition, the application of TGF-β 1 and VEGF could be used as growth factors sources in constructing tissue engineered ligament.
Collapse
Affiliation(s)
- Gang Zou
- The First Department of Orthopaedics, the Affiliated Hospital of Zunyi Medical College, Zunyi Guizhou, 563000, P.R.China
| | | | - Ying Jin
- The First Department of Orthopaedics, the Affiliated Hospital of Zunyi Medical College, Zunyi Guizhou, 563000, P.R.China
| | - Xizhong Zhu
- The First Department of Orthopaedics, the Affiliated Hospital of Zunyi Medical College, Zunyi Guizhou, 563000, P.R.China
| | - Jibin Yang
- The First Department of Orthopaedics, the Affiliated Hospital of Zunyi Medical College, Zunyi Guizhou, 563000, P.R.China
| | - Shengmin Wang
- The First Department of Orthopaedics, the Affiliated Hospital of Zunyi Medical College, Zunyi Guizhou, 563000, P.R.China
| | - Qi You
- The First Department of Orthopaedics, the Affiliated Hospital of Zunyi Medical College, Zunyi Guizhou, 563000, P.R.China
| | - Huazhang Xiong
- The First Department of Orthopaedics, the Affiliated Hospital of Zunyi Medical College, Zunyi Guizhou, 563000, P.R.China
| | - Yi Liu
- The First Department of Orthopaedics, the Affiliated Hospital of Zunyi Medical College, Zunyi Guizhou, 563000,
| |
Collapse
|