1
|
Huang G, Shen H, Xu K, Shen Y, Jiale Jin, Chu G, Xing H, Feng Z, Wang Y. Single-Cell Microgel Encapsulation Improves the Therapeutic Efficacy of Mesenchymal Stem Cells in Treating Intervertebral Disc Degeneration via Inhibiting Pyroptosis. RESEARCH (WASHINGTON, D.C.) 2024; 7:0311. [PMID: 38371273 PMCID: PMC10871001 DOI: 10.34133/research.0311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 01/14/2024] [Indexed: 02/20/2024]
Abstract
While mesenchymal stem cell (MSC) shows great potentials in treating intervertebral disc degeneration, most MSC die soon after intradiscal transplantation, resulting in inferior therapeutic efficacy. Currently, bulk hydrogels are the common solution to improve MSC survival in tissues, although hydrogel encapsulation impairs MSC migration and disrupts extracellular microenvironment. Cell hydrogel encapsulation has been proposed to overcome the limitation of traditional bulk hydrogels, yet this technique has not been used in treating disc degeneration. Using a layer-by-layer self-assembly technique, we fabricated alginate and gelatin microgel to encapsulate individual MSC for treating disc degeneration. The small size of microgel allowed intradiscal injection of coated MSC. We demonstrated that pyroptosis was involved in MSC death under oxidative stress stimulation, and microgel coating suppressed pyroptosis activation by maintaining mitochondria homeostasis. Microgel coating protected MSC in the harsh disc microenvironment, while retaining vital cellular functions such as migration, proliferation, and differentiation. In a rat model of disc degeneration, coated MSC exhibits prolonged retention in the disc and better efficacy of attenuating disc degeneration, as compared with bare MSC treatment alone. Further, microgel-coated MSC exhibited improved therapeutic effects in treating disc degeneration via suppressing the activation of pyroptosis in the disc. For the first time, microgel-encapsulated MSC was used to treat disc degeneration and obtain encouraging outcomes. The developed biocompatible single-cell hydrogel is an effective strategy to protect MSC and maintain cellular functions and may be an efficacious approach to improving the efficacy of MSC therapy in treating disc degeneration. The objective of this study is to improve the efficacy of cell therapy for treating disc degeneration using single-cell hydrogel encapsulation and further to understand related cytoprotective mechanisms.
Collapse
Affiliation(s)
- Guanrui Huang
- Department of Orthopedic Surgery, The First Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Haotian Shen
- Department of Orthopedic Surgery, The First Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Kaiwang Xu
- Zhejiang University, Hangzhou 310058, China
| | - Yifan Shen
- Department of Orthopedic Surgery, The First Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Jiale Jin
- Department of Orthopedic Surgery, The First Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Guangyu Chu
- Department of Orthopedic Surgery, The First Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Hongyuan Xing
- Department of Orthopedic Surgery, The First Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Zhiyun Feng
- Department of Orthopedic Surgery, The First Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Yue Wang
- Department of Orthopedic Surgery, The First Affiliated Hospital,
Zhejiang University School of Medicine, Hangzhou 310003, China
| |
Collapse
|
2
|
Han Y, Ouyang Z, Wawrose RA, Chen SR, Hallbaum M, Dong Q, Dando E, Tang Y, Wang B, Lee JY, Shaw JD, Kang JD, Sowa GA, Vo NN. ISSLS prize in basic science 2021: a novel inducible system to regulate transgene expression of TIMP1. 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 2021; 30:1098-1107. [PMID: 33523281 PMCID: PMC8550652 DOI: 10.1007/s00586-021-06728-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 12/14/2020] [Accepted: 01/09/2021] [Indexed: 11/28/2022]
Abstract
Purpose Inflammatory and oxidative stress upregulates matrix metalloproteinase (MMP) activity, leading to intervertebral disc degeneration (IDD). Gene therapy using human tissue inhibitor of metalloproteinase 1 (hTIMP1) has effectively treated IDD in animal models. However, persistent unregulated transgene expression may have negative side effects. We developed a recombinant adeno-associated viral (AAV) gene vector, AAV-NFκB-hTIMP1, that only expresses the hTIMP1 transgene under conditions of stress. Methods Rabbit disc cells were transfected or transduced with AAV-CMV-hTIMP1, which constitutively expresses hTIMP1, or AAV-NFκB-hTIMP1. Disc cells were selectively treated with IL-1β. NFκB activation was verified by nuclear translocation. hTIMP1 mRNA and protein expression were measured by RT-PCR and ELISA, respectively. MMP activity was measured by following cleavage of a fluorogenic substrate. Results IL-1β stimulation activated NFκB demonstrating that IL-1β was a surrogate for inflammatory stress. Stimulating AAV-NFκB-hTIMP1 cells with IL-1β increased hTIMP1 expression compared to unstimulated cells. AAV-CMV-hTIMP1 cells demonstrated high levels of hTIMP1 expression regardless of IL-1β stimulation. hTIMP1 expression was comparable between IL-1β stimulated AAV-NFκB-hTIMP1 cells and AAV-CMV-hTIMP1 cells. MMP activity was decreased in AAV-NFκB-hTIMP1 cells compared to baseline levels or cells exposed to IL-1β. Conclusion AAV-NFκB-hTIMP1 is a novel inducible transgene delivery system. NFκB regulatory elements ensure that hTIMP1 expression occurs only with inflammation, which is central to IDD development. Unlike previous inducible systems, the AAV-NFκB-hTIMP1 construct is dependent on endogenous factors, which minimizes potential side effects caused by constitutive transgene overexpression. It also prevents the unnecessary production of transgene products in cells that do not require therapy.
Collapse
Affiliation(s)
- Yingchao Han
- Department of Spine Surgery, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai, China.,Department of Spine Surgery, School of Medicine, Shanghai East Hospital, Tongji University, Shanghai, China.,Ferguson Laboratory for Orthopaedic and Spine Research, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Zhihua Ouyang
- Ferguson Laboratory for Orthopaedic and Spine Research, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA.,Department of Spine Surgery, The First Affiliated Hospital of Nanhua University, Hengyang, China
| | - Richard A Wawrose
- Ferguson Laboratory for Orthopaedic and Spine Research, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Stephen R Chen
- Ferguson Laboratory for Orthopaedic and Spine Research, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Maximiliane Hallbaum
- Ferguson Laboratory for Orthopaedic and Spine Research, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Qing Dong
- Ferguson Laboratory for Orthopaedic and Spine Research, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Emily Dando
- Ferguson Laboratory for Orthopaedic and Spine Research, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ying Tang
- Molecular Therapeutics Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Bing Wang
- Molecular Therapeutics Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Joon Y Lee
- Ferguson Laboratory for Orthopaedic and Spine Research, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jeremy D Shaw
- Ferguson Laboratory for Orthopaedic and Spine Research, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - James D Kang
- Department of Orthopaedics, School of Medicine, Brigham and Women's Hospital, Harvard University, Boston, MA, USA
| | - Gwendolyn A Sowa
- Ferguson Laboratory for Orthopaedic and Spine Research, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA. .,Department of Physical Medicine and Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | - Nam N Vo
- Ferguson Laboratory for Orthopaedic and Spine Research, Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
3
|
Dombrowski ME, Olsen AS, Vaudreuil N, Couch BK, Dong Q, Tucci M, Lee JY, Vo NV, Sowa G. Rabbit Annulus Fibrosus Cells Express Neuropeptide Y, Which Is Influenced by Mechanical and Inflammatory Stress. Neurospine 2020; 17:69-76. [PMID: 32252156 PMCID: PMC7136102 DOI: 10.14245/ns.2040046.023] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 02/26/2020] [Indexed: 12/17/2022] Open
Abstract
Objective Rabbit annulus fibrosus (AF) cells were exposed to isolated or combined mechanical and inflammatory stress to examine the expression of neuropeptide Y (NPY). This study aims to explore the ability of AF cells to produce NPY in response to mechanical and inflammatory stress.
Methods Lumbar AF cells of 6- to 8-month-old female New Zealand white rabbits were harvested and exposed to combinations of inflammatory (interleukin-1β) and mechanical (6% or 18%) tensile stress using the Flexcell System. NPY concentrations were measured in the media via enzyme-linked immunosorbent assay. The presence of NPY receptor-type 1 (NPY-1R) in AF cells of rabbit intervertebral discs was also analyzed via immunohistochemistry and immunofluorescence.
Results Exposure to inflammatory stimuli showed a significant increase in the amount of NPY expression compared to control AF cells. Mechanical strain alone did not result in a significant difference in NPY expression. While combined inflammatory and mechanical stress did not demonstrate an increase in NPY expression at low (6%) levels of strain, at 18% strain, there was a large—though not statistically significant—increase in NPY expression under conditions of inflammatory stress. Lastly, immunofluorescence and immunohistochemistry of AF cells and tissue, respectively, demonstrated the presence of NPY-1R.
Conclusion These findings demonstrate that rabbit AF cells are capable of expressing NPY, and expression is enhanced in response to inflammatory and mechanical stress. Because both inflammatory and mechanical stress contribute to intervertebral disc degeneration (IDD), this observation raises the potential of a mechanistic link between low back pain and IDD.
Collapse
Affiliation(s)
- Malcolm E Dombrowski
- Department of Orthopaedic Surgery, Ferguson Laboratory for Orthopaedic Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Adam S Olsen
- Department of Orthopaedic Surgery, Ferguson Laboratory for Orthopaedic Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Nicholas Vaudreuil
- Department of Orthopaedic Surgery, Ferguson Laboratory for Orthopaedic Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Brandon K Couch
- Department of Orthopaedic Surgery, Ferguson Laboratory for Orthopaedic Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Qing Dong
- Department of Orthopaedic Surgery, Ferguson Laboratory for Orthopaedic Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Michelle Tucci
- Department of Anesthesiology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Joon Y Lee
- Department of Orthopaedic Surgery, Ferguson Laboratory for Orthopaedic Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Nam V Vo
- Department of Orthopaedic Surgery, Ferguson Laboratory for Orthopaedic Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Gwendolyn Sowa
- Department of Orthopaedic Surgery, Ferguson Laboratory for Orthopaedic Research, University of Pittsburgh, Pittsburgh, PA, USA.,Department of Physical Medicine and Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
4
|
Rider SM, Mizuno S, Kang JD. Molecular Mechanisms of Intervertebral Disc Degeneration. Spine Surg Relat Res 2019; 3:1-11. [PMID: 31435545 PMCID: PMC6690117 DOI: 10.22603/ssrr.2017-0095] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 01/24/2018] [Indexed: 12/25/2022] Open
Abstract
Intervertebral disc degeneration is a well-known cause of disability, the result of which includes neck and back pain with associated mobility limitations. The purpose of this article is to provide an overview of the known molecular mechanisms through which intervertebral disc degeneration occurs as a result of complex interactions of exogenous and endogenous stressors. This review will focus on some of the identified molecular changes leading to the deterioration of the extracellular matrix of both the annulus fibrosus and nucleus pulposus. In addition, we will provide a summation of our current knowledge supporting the role of associated DNA and intracellular damage, cellular senescence's catabolic effects, oxidative stress, and the cell's inappropriate response to damage in contributing to intervertebral disc degeneration. Our current understanding of the molecular mechanisms through which intervertebral disc degeneration occurs provides us with abundant insight into how physical and chemical changes exacerbate the degenerative process of the entire spine. Furthermore, we will describe some of the related molecular targets and therapies that may contribute to intervertebral repair and regeneration.
Collapse
Affiliation(s)
- Sean M Rider
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Shuichi Mizuno
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - James D Kang
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
5
|
Rustenburg CM, Emanuel KS, Peeters M, Lems WF, Vergroesen PA, Smit TH. Osteoarthritis and intervertebral disc degeneration: Quite different, quite similar. JOR Spine 2018; 1:e1033. [PMID: 31463450 PMCID: PMC6686805 DOI: 10.1002/jsp2.1033] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 08/14/2018] [Accepted: 08/17/2018] [Indexed: 02/06/2023] Open
Abstract
Intervertebral disc degeneration describes the vicious cycle of the deterioration of intervertebral discs and can eventually result in degenerative disc disease (DDD), which is accompanied by low-back pain, the musculoskeletal disorder with the largest socioeconomic impact world-wide. In more severe stages, intervertebral disc degeneration is accompanied by loss of joint space, subchondral sclerosis, and osteophytes, similar to osteoarthritis (OA) in the articular joint. Inspired by this resemblance, we investigated the analogy between human intervertebral discs and articular joints. Although embryonic origin and anatomy suggest substantial differences between the two types of joint, some features of cell physiology and extracellular matrix in the nucleus pulposus and articular cartilage share numerous parallels. Moreover, there are great similarities in the response to mechanical loading and the matrix-degrading factors involved in the cascade of degeneration in both tissues. This suggests that the local environment of the cell is more important to its behavior than embryonic origin. Nevertheless, OA is widely regarded as a true disease, while intervertebral disc degeneration is often regarded as a radiological finding and DDD is undervalued as a cause of chronic low-back pain by clinicians, patients and society. Emphasizing the similarities rather than the differences between the two diseases may create more awareness in the clinic, improve diagnostics in DDD, and provide cross-fertilization of clinicians and scientists involved in both intervertebral disc degeneration and OA.
Collapse
Affiliation(s)
- Christine M.E. Rustenburg
- Department or Orthopaedic SurgeryAmsterdam Movement Sciences, Amsterdam UMC, University of AmsterdamAmsterdamThe Netherlands
| | - Kaj S. Emanuel
- Department or Orthopaedic SurgeryAmsterdam Movement Sciences, Amsterdam UMC, University of AmsterdamAmsterdamThe Netherlands
| | - Mirte Peeters
- Department or Orthopaedic SurgeryAmsterdam Movement Sciences, Amsterdam UMC, University of AmsterdamAmsterdamThe Netherlands
| | - Willem F. Lems
- Department of RheumatologyAmsterdam Movement Sciences, Amsterdam UMC, Vrije Universiteit AmsterdamAmsterdamThe Netherlands
| | | | - Theodoor H. Smit
- Department or Orthopaedic SurgeryAmsterdam Movement Sciences, Amsterdam UMC, University of AmsterdamAmsterdamThe Netherlands
- Department of Medical BiologyAmsterdam UMC, University of AmsterdamAmsterdamThe Netherlands
| |
Collapse
|
6
|
Hartman R, Patil P, Tisherman R, St Croix C, Niedernhofer LJ, Robbins PD, Ambrosio F, Van Houten B, Sowa G, Vo N. Age-dependent changes in intervertebral disc cell mitochondria and bioenergetics. Eur Cell Mater 2018; 36:171-183. [PMID: 30334244 PMCID: PMC9972500 DOI: 10.22203/ecm.v036a13] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Robust cellular bioenergetics is vital in the energy-demanding process of maintaining matrix homeostasis in the intervertebral disc. Age-related decline in disc cellular bioenergetics is hypothesised to contribute to the matrix homeostatic perturbation observed in intervertebral disc degeneration. The present study aimed to measure how ageing impacted disc cell mitochondria and bioenergetics. Age-related changes measured included matrix content and cellularity in disc tissue, as well as matrix synthesis, cell proliferation and senescence markers in cell cultures derived from annulus fibrosus (AF) and nucleus pulposus (NP) isolated from the discs of young (6-9 months) and older (36-50 months) New Zealand White rabbits. Cellular bioenergetic parameters were measured using a Seahorse XFe96 Analyzer, in addition to quantitating mitochondrial morphological changes and membrane potential. Ageing reduced mitochondrial number and membrane potential in both cell types. Also, it significantly reduced glycolytic capacity, mitochondrial reserve capacity, maximum aerobic capacity and non-glucose-dependent respiration in NP. Moreover, NP cells exhibited age-related decline in matrix synthesis and reduced cellularity in older tissues. Despite a lack of changes in mitochondrial respiration with age, AF cells showed an increase in glycolysis and altered matrix production. While previous studies report age-related matrix degenerative changes in disc cells, the present study revealed, for the first time, that ageing affected mitochondrial number and function, particularly in NP cells. Consequently, age-related bioenergetic changes may contribute to the functional alterations in aged NP cells that underlie disc degeneration.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - N Vo
- Ferguson Laboratory for Orthopaedic Research, 200 Lothrop St., E1648 University of Pittsburgh, Pittsburgh, PA 15213,
| |
Collapse
|
7
|
Xu Y, Xu S, Gao Z, Xiao L, Xiao F, Xu H, Zhang X. Degree of endplate chondrocyte degeneration in different tension regions during mechanical stimulation. Mol Med Rep 2018; 17:4415-4421. [PMID: 29344659 PMCID: PMC5802215 DOI: 10.3892/mmr.2018.8435] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 11/16/2017] [Indexed: 01/15/2023] Open
Abstract
The aim of this study was to explore the degree of degeneration of endplate chondrocytes in different tension regions induced by intermittent cyclic mechanical tension (ICMT) in vitro. Rat endplate chondrocytes were harvested and treated with 10% ICMT for 8 h/day with a frequency of 0.5 Hz. A cartilage degeneration model was induced using an FX‑5000T cell strain‑loading system. The experiment was divided into the central region and the peripheral region, according to the contact area between the loading post and the six‑well flexible silicone rubber BioFlex plates. Toluidine blue and phalloidin staining were used to observe the morphological changes of cells following mechanical stimulation. Apoptosis was detected by flow cytometry and the mRNA and protein expression levels of collagen type II α1, aggrecan, SRY‑box 9 and matrix metalloproteinase 13 were detected by reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR) and western blotting, respectively. Endplate chondrocytes exhibited degenerative alterations under mechanical conditions of 10% ICMT and 0.5 Hz at 8 h/day. Toluidine blue and phalloidin staining demonstrated that the cells in the peripheral region were more slender compared with cells in the central region, but RT‑qPCR and western blotting results demonstrated that the degree of cell degeneration between the two groups was not statistically differences. So that cell morphological alteration does not imply that cells have undergone degeneration.
Collapse
Affiliation(s)
- Yongming Xu
- Department of Orthopedic Surgery, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui 241001, P.R. China
| | - Shujuan Xu
- Department of Surgery, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui 241001, P.R. China
| | - Zhi Gao
- Department of Orthopedic Surgery, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui 241001, P.R. China
| | - Liang Xiao
- Department of Orthopedic Surgery, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui 241001, P.R. China
| | - Fei Xiao
- Department of Orthopedic Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Hongguang Xu
- Department of Orthopedic Surgery, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui 241001, P.R. China
| | - Xiaoling Zhang
- Department of Orthopedic Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| |
Collapse
|
8
|
Nguyen QT, Jacobsen TD, Chahine NO. Effects of Inflammation on Multiscale Biomechanical Properties of Cartilaginous Cells and Tissues. ACS Biomater Sci Eng 2017; 3:2644-2656. [PMID: 29152560 PMCID: PMC5686563 DOI: 10.1021/acsbiomaterials.6b00671] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 01/24/2017] [Indexed: 12/20/2022]
Abstract
![]()
Cells
within cartilaginous tissues are mechanosensitive and thus
require mechanical loading for regulation of tissue homeostasis and
metabolism. Mechanical loading plays critical roles in cell differentiation,
proliferation, biosynthesis, and homeostasis. Inflammation is an important
event occurring during multiple processes, such as aging, injury,
and disease. Inflammation has significant effects on biological processes
as well as mechanical function of cells and tissues. These effects
are highly dependent on cell/tissue type, timing, and magnitude. In
this review, we summarize key findings pertaining to effects of inflammation
on multiscale mechanical properties at subcellular, cellular, and
tissue level in cartilaginous tissues, including alterations in mechanotransduction
and mechanosensitivity. The emphasis is on articular cartilage and
the intervertebral disc, which are impacted by inflammatory insults
during degenerative conditions such as osteoarthritis, joint pain,
and back pain. To recapitulate the pro-inflammatory cascades that
occur in vivo, different inflammatory stimuli have been used for in
vitro and in situ studies, including tumor necrosis factor (TNF),
various interleukins (IL), and lipopolysaccharide (LPS). Therefore,
this review will focus on the effects of these stimuli because they
are the best studied pro-inflammatory cytokines in cartilaginous tissues.
Understanding the current state of the field of inflammation and cell/tissue
biomechanics may potentially identify future directions for novel
and translational therapeutics with multiscale biomechanical considerations.
Collapse
Affiliation(s)
- Q T Nguyen
- Bioengineering-Biomechanics Laboratory The Feinstein Institute for Medical Research, Northwell Health System, Manhasset, New York 11030, United States
| | - T D Jacobsen
- Bioengineering-Biomechanics Laboratory The Feinstein Institute for Medical Research, Northwell Health System, Manhasset, New York 11030, United States.,Hofstra Northwell School of Medicine, Hempstead, New York 11549, United States
| | - N O Chahine
- Bioengineering-Biomechanics Laboratory The Feinstein Institute for Medical Research, Northwell Health System, Manhasset, New York 11030, United States.,Hofstra Northwell School of Medicine, Hempstead, New York 11549, United States
| |
Collapse
|
9
|
Krupkova O, Hlavna M, Amir Tahmasseb J, Zvick J, Kunz D, Ito K, Ferguson SJ, Wuertz-Kozak K. An Inflammatory Nucleus Pulposus Tissue Culture Model to Test Molecular Regenerative Therapies: Validation with Epigallocatechin 3-Gallate. Int J Mol Sci 2016; 17:ijms17101640. [PMID: 27689996 PMCID: PMC5085673 DOI: 10.3390/ijms17101640] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2016] [Revised: 09/15/2016] [Accepted: 09/19/2016] [Indexed: 12/18/2022] Open
Abstract
Organ cultures are practical tools to investigate regenerative strategies for the intervertebral disc. However, most existing organ culture systems induce severe tissue degradation with only limited representation of the in vivo processes. The objective of this study was to develop a space- and cost-efficient tissue culture model, which represents degenerative processes of the nucleus pulposus (NP). Intact bovine NPs were cultured in a previously developed system using Dyneema jackets. Degenerative changes in the NP tissue were induced either by the direct injection of chondroitinase ABC (1-20 U/mL) or by the diffusion of interleukin-1 beta (IL-1β) and tumor necrosis factor alpha (TNF-α) (both 100 ng/mL) from the culture media. Extracellular matrix composition (collagens, proteoglycans, water, and DNA) and the expression of inflammatory and catabolic genes were analyzed. The anti-inflammatory and anti-catabolic compound epigallocatechin 3-gallate (EGCG, 10 µM) was employed to assess the relevance of the degenerative NP model. Although a single injection of chondroitinase ABC reduced the proteoglycan content in the NPs, it did not activate cellular responses. On the other hand, IL-1β and TNF-α significantly increased the mRNA expression of inflammatory mediators IL-6, IL-8, inducible nitric oxide synthase (iNOS), prostaglandin-endoperoxide synthase 2 (PTGS2) and matrix metalloproteinases (MMP1, MMP3, and MMP13). The cytokine-induced gene expression in the NPs was ameliorated with EGCG. This study provides a proof of concept that inflammatory NP cultures, with appropriate containment, can be useful for the discovery and evaluation of molecular therapeutic strategies against early degenerative disc disease.
Collapse
Affiliation(s)
- Olga Krupkova
- Institute for Biomechanics, ETH Zurich, Hoenggerbergring 64, CH-8093 Zurich, Switzerland.
| | - Marian Hlavna
- Institute for Biomechanics, ETH Zurich, Hoenggerbergring 64, CH-8093 Zurich, Switzerland.
| | - Julie Amir Tahmasseb
- Institute for Biomechanics, ETH Zurich, Hoenggerbergring 64, CH-8093 Zurich, Switzerland.
| | - Joel Zvick
- Institute for Biomechanics, ETH Zurich, Hoenggerbergring 64, CH-8093 Zurich, Switzerland.
| | - Dominik Kunz
- Institute for Biomechanics, ETH Zurich, Hoenggerbergring 64, CH-8093 Zurich, Switzerland.
- Health Department, ZHAW-Zurich University of Applied Sciences, Technikumstrasse 71, CH-8401 Winterthur, Switzerland.
| | - Keita Ito
- Department of Biomedical Engineering, Eindhoven University of Technology, Postbus 513, 5600 MB Eindhoven, The Netherlands.
| | - Stephen J Ferguson
- Institute for Biomechanics, ETH Zurich, Hoenggerbergring 64, CH-8093 Zurich, Switzerland.
- Competence Center for Applied Biotechnology and Molecular Medicine, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland.
| | - Karin Wuertz-Kozak
- Institute for Biomechanics, ETH Zurich, Hoenggerbergring 64, CH-8093 Zurich, Switzerland.
- Competence Center for Applied Biotechnology and Molecular Medicine, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland.
| |
Collapse
|
10
|
Vo NV, Hartman RA, Patil PR, Risbud MV, Kletsas D, Iatridis JC, Hoyland JA, Le Maitre CL, Sowa GA, Kang JD. Molecular mechanisms of biological aging in intervertebral discs. J Orthop Res 2016; 34:1289-306. [PMID: 26890203 PMCID: PMC4988945 DOI: 10.1002/jor.23195] [Citation(s) in RCA: 280] [Impact Index Per Article: 31.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 02/03/2016] [Indexed: 02/04/2023]
Abstract
Advanced age is the greatest risk factor for the majority of human ailments, including spine-related chronic disability and back pain, which stem from age-associated intervertebral disc degeneration (IDD). Given the rapid global rise in the aging population, understanding the biology of intervertebral disc aging in order to develop effective therapeutic interventions to combat the adverse effects of aging on disc health is now imperative. Fortunately, recent advances in aging research have begun to shed light on the basic biological process of aging. Here we review some of these insights and organize the complex process of disc aging into three different phases to guide research efforts to understand the biology of disc aging. The objective of this review is to provide an overview of the current knowledge and the recent progress made to elucidate specific molecular mechanisms underlying disc aging. In particular, studies over the last few years have uncovered cellular senescence and genomic instability as important drivers of disc aging. Supporting evidence comes from DNA repair-deficient animal models that show increased disc cellular senescence and accelerated disc aging. Additionally, stress-induced senescent cells have now been well documented to secrete catabolic factors, which can negatively impact the physiology of neighboring cells and ECM. These along with other molecular drivers of aging are reviewed in depth to shed crucial insights into the underlying mechanisms of age-related disc degeneration. We also highlight molecular targets for novel therapies and emerging candidate therapeutics that may mitigate age-associated IDD. © 2016 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 34:1289-1306, 2016.
Collapse
Affiliation(s)
- Nam V. Vo
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Robert A. Hartman
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Prashanti R. Patil
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Makarand V. Risbud
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Dimitris Kletsas
- Laboratory of Cell Proliferation and Ageing, Institute of Biosciences and Applications, National Centre for Scientific Research “Demokritos”, Athens, Greece
| | - James C. Iatridis
- Leni & Peter W May Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Judith A. Hoyland
- Centre for Tissue Injury and Repair, Faculty of Medical and Human Sciences, University of Manchester M13 9PT and NIHR Manchester Musculoskeletal Biomedical Research Unit, Manchester Academic Health Science Centre, Manchester, UK
| | - Christine L. Le Maitre
- Musculoskeletal and Regenerative Medicine Research Group, Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield, S1 1WB, UK
| | - Gwendolyn A. Sowa
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - James D. Kang
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
11
|
Walter B, Purmessur D, Moon A, Occhiogrosso J, Laudier D, Hecht A, Iatridis J. Reduced tissue osmolarity increases TRPV4 expression and pro-inflammatory cytokines in intervertebral disc cells. Eur Cell Mater 2016; 32:123-36. [PMID: 27434269 PMCID: PMC5072776 DOI: 10.22203/ecm.v032a08] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The mechanical behaviour and cellular metabolism of intervertebral discs (IVDs) and articular cartilage are strongly influenced by their proteoglycan content and associated osmotic properties. This osmotic environment is a biophysical signal that changes with disease and may contribute to the elevated matrix breakdown and altered biologic response to loading observed in IVD degeneration and osteoarthritis. This study tested the hypothesis that changes in osmo-sensation by the transient receptor potential vallinoid-4 (TRPV4) ion channel occur with disease and contribute to the inflammatory environment found during degeneration. Immunohistochemistry on bovine IVDs from an inflammatory organ culture model were used to investigate if TRPV4 is expressed in the IVD and how expression changes with degeneration. Western blot, live-cell calcium imaging, and qRT-PCR were used to investigate whether osmolarity changes or tumour necrosis factor α (TNFα) regulate TRPV4 expression, and how altered TRPV4 expression influences calcium signalling and pro-inflammatory cytokine expression. TRPV4 expression correlated with TNFα expression, and was increased when cultured in reduced medium osmolarity and unaltered with TNFα-stimulation. Increased TRPV4 expression increased the calcium flux following TRPV4 activation and increased interleukin-1β (IL-1β) and IL-6 gene expression in IVD cells. TRPV4 expression was qualitatively elevated in regions of aggrecan depletion in degenerated human IVDs. Collectively, results suggest that reduced tissue osmolarity, likely following proteoglycan degradation, can increase TRPV4 signalling and enhance pro-inflammatory cytokine production, suggesting changes in TRPV4 mediated osmo-sensation may contribute to the progressive matrix breakdown in disease.
Collapse
Affiliation(s)
- B.A. Walter
- Leni & Peter W. May Department of Orthopaedics at the Icahn School of Medicine at Mount Sinai, New York, NY, USA,Department of Biomedical Engineering, The City College of New York, New York, NY, USA
| | - D Purmessur
- Leni & Peter W. May Department of Orthopaedics at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - A. Moon
- Leni & Peter W. May Department of Orthopaedics at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - J. Occhiogrosso
- Leni & Peter W. May Department of Orthopaedics at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - D.M. Laudier
- Leni & Peter W. May Department of Orthopaedics at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - A.C. Hecht
- Leni & Peter W. May Department of Orthopaedics at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - J.C. Iatridis
- Leni & Peter W. May Department of Orthopaedics at the Icahn School of Medicine at Mount Sinai, New York, NY, USA,Address for correspondence: James C. Iatridis Leni & Peter W. May Department of Orthopaedics, Box 1188, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA, Telephone Number: 1-212-241-1517, FAX Number: 1-212-876-3168 www.ecmjournal.org
| |
Collapse
|
12
|
NF-κB Signaling Pathway in Controlling Intervertebral Disk Cell Response to Inflammatory and Mechanical Stressors. Phys Ther 2016; 96:704-11. [PMID: 26893504 PMCID: PMC4858661 DOI: 10.2522/ptj.20150045] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Accepted: 01/21/2016] [Indexed: 12/18/2022]
Abstract
BACKGROUND Intervertebral disk degeneration (IDD) has a greater than 90% lifetime incidence and is one of the leading causes of chronic back pain in the United States. Despite the high societal cost of IDD, there is limited understanding of the biological effects of mechanical overloading on further degeneration. The transcription factor NF-κB (nuclear factor κB) has been implicated as a key mediator of disk cell response to inflammatory and mechanical stresses and represents a potential control point. OBJECTIVE The study objective was to measure the effect of NF-κB signaling pathway inhibition on annulus fibrosus (AF) cell matrix synthesis and gene expression under conditions of combined inflammatory and mechanical stimulation. METHODS Annulus fibrosus cells were harvested from rabbit intervertebral disks and grown in vitro on flexible plates. The cells were exposed to inflammatory and mechanical stimulation for 24 hours with and without NF-κB inhibition. Nuclear translocation of NF-κB was measured via immunofluorescent staining. Intervertebral disk cell homeostasis was assessed via inflammatory, anabolic, and catabolic gene expression and via matrix synthetic ability. RESULTS NF-κB nuclear translocation in response to interleukin-1 beta (IL-1β) was reversed with exposure to NF-κB inhibition. NF-κB inhibition decreased matrix metalloproteinase-3, inducible nitric oxide synthase, and cyclooxygenase-2 gene expression and prostaglandin E2 production response to combined inflammatory and mechanical stimulation. Proteoglycan and collagen synthesis were decreased by combined stimulation, but this effect was not reversed by NF-κB inhibition. LIMITATIONS In vitro modeling of conditions within the disk may not fully reflect the response that AF cells have in native matrix. CONCLUSIONS NF-κB signaling mediates catabolic and inflammatory responses to inflammatory and mechanical stimulation but does not mediate the decrease in matrix synthesis under combined harmful stimulation. Identification of key control points in the cellular responses to inflammatory and mechanical stimuli will facilitate rational design of exercise-based therapies and facilitate synergistic treatments of novel biochemical treatments with rehabilitation regimens.
Collapse
|
13
|
Likhitpanichkul M, Torre OM, Gruen J, Walter BA, Hecht AC, Iatridis JC. Do mechanical strain and TNF-α interact to amplify pro-inflammatory cytokine production in human annulus fibrosus cells? J Biomech 2016; 49:1214-1220. [PMID: 26924657 DOI: 10.1016/j.jbiomech.2016.02.029] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2015] [Revised: 02/09/2016] [Accepted: 02/12/2016] [Indexed: 12/20/2022]
Abstract
During intervertebral disc (IVD) injury and degeneration, annulus fibrosus (AF) cells experience large mechanical strains in a pro-inflammatory milieu. We hypothesized that TNF-α, an initiator of IVD inflammation, modifies AF cell mechanobiology via cytoskeletal changes, and interacts with mechanical strain to enhance pro-inflammatory cytokine production. Human AF cells (N=5, Thompson grades 2-4) were stretched uniaxially on collagen-I coated chambers to 0%, 5% (physiological) or 15% (pathologic) strains at 0.5Hz for 24h under hypoxic conditions with or without TNF-α (10ng/mL). AF cells were treated with anti-TNF-α and anti-IL-6. ELISA assessed IL-1β, IL-6, and IL-8 production and immunocytochemistry measured F-actin, vinculin and α-tubulin in AF cells. TNF-α significantly increased AF cell pro-inflammatory cytokine production compared to basal conditions (IL-1β:2.0±1.4-84.0±77.3, IL-6:10.6±9.9-280.9±214.1, IL-8:23.9±26.0-5125.1±4170.8pg/ml for basal and TNF-α treatment, respectively) as expected, but mechanical strain did not. Pathologic strain in combination with TNF-α increased IL-1β, and IL-8 but not IL-6 production of AF cells. TNF-α treatment altered F-actin and α-tubulin in AF cells, suggestive of altered cytoskeletal stiffness. Anti-TNF-α (infliximab) significantly inhibited pro-inflammatory cytokine production while anti-IL-6 (atlizumab) did not. In conclusion, TNF-α altered AF cell mechanobiology with cytoskeletal remodeling that potentially sensitized AF cells to mechanical strain and increased TNF-α-induced pro-inflammatory cytokine production. Results suggest an interaction between TNF-α and mechanical strain and future mechanistic studies are required to validate these observations.
Collapse
Affiliation(s)
- Morakot Likhitpanichkul
- Leni and Peter W. May Department of Orthopedics, Icahn School of Medicine at Mount Sinai, Box 1188, New York, NY 10029, United States.
| | - Olivia M Torre
- Leni and Peter W. May Department of Orthopedics, Icahn School of Medicine at Mount Sinai, Box 1188, New York, NY 10029, United States.
| | - Jadry Gruen
- Leni and Peter W. May Department of Orthopedics, Icahn School of Medicine at Mount Sinai, Box 1188, New York, NY 10029, United States.
| | - Benjamin A Walter
- Leni and Peter W. May Department of Orthopedics, Icahn School of Medicine at Mount Sinai, Box 1188, New York, NY 10029, United States; Department of Biomedical Engineering, The City College of New York, New York, NY, United States.
| | - Andrew C Hecht
- Leni and Peter W. May Department of Orthopedics, Icahn School of Medicine at Mount Sinai, Box 1188, New York, NY 10029, United States.
| | - James C Iatridis
- Leni and Peter W. May Department of Orthopedics, Icahn School of Medicine at Mount Sinai, Box 1188, New York, NY 10029, United States.
| |
Collapse
|
14
|
Rehabilomics research: a model for translational rehabilitation and comparative effectiveness rehabilitation research. Am J Phys Med Rehabil 2014; 93:913-6. [PMID: 24901762 DOI: 10.1097/phm.0000000000000114] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
15
|
Cuesta A, Del Valle ME, García-Suárez O, Viña E, Cabo R, Vázquez G, Cobo JL, Murcia A, Alvarez-Vega M, García-Cosamalón J, Vega JA. Acid-sensing ion channels in healthy and degenerated human intervertebral disc. Connect Tissue Res 2014; 55:197-204. [PMID: 24432912 DOI: 10.3109/03008207.2014.884083] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Acid-sensing ion channels (ASICs) are a family of H(+)-gated voltage-insensitive ion channels that respond to extracellular acidification by regulating transmembrane Ca(2+) flux. Moreover, ASICs can also be gated by mechanical forces and may function as mechanosensors. The cells of the intervertebral disc (IVD) have an unusual acidic and hyperosmotic microenvironment. Changes in the pH and osmolarity determine the viability of IVD cells and the composition of the extracellular matrix, and both are the basis of IVD degeneration. In this study, the expression of ASICs (ASIC1, ASIC2, ASIC3 and ASIC4) mRNAs and proteins in human healthy and degenerated IVD was evaluated by quantitative reverse transcription-quantitative polymerase chain reaction and Western blot. The distribution of ASIC proteins was determined by immunohistochemistry. The mRNAs for all ASICs were detected in normal human IVD, and significantly increased levels were found in degenerated IVD. Western blots demonstrated the presence of proteins with estimated molecular weights of approximately 68-72 kDa. In both the annulus fibrosus (AF) and nucleus pulposus (NP) of normal IVD, ASIC2 is the most frequently expressed ASIC followed by ASIC3, ASIC1 and ASIC4. In the AF of degenerated IVD, there was a significant increase in the number of ASIC1 and ASIC4 positive cells, whereas in the NP, we found significant increase of expression of ASIC1, ASIC2 and ASIC3. These results describe the occurrence and localization of different ASICs in human healthy IVD, and their increased expression in degenerated IVD, thus suggesting that ASICs may be involved in IVD degeneration.
Collapse
|
16
|
Spyropoulou A, Basdra EK. Mechanotransduction in bone: Intervening in health and disease. World J Exp Med 2013; 3:74-86. [DOI: 10.5493/wjem.v3.i4.74] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Revised: 09/06/2013] [Accepted: 11/03/2013] [Indexed: 02/06/2023] Open
Abstract
Mechanotransduction has been proven to be one of the most significant variables in bone remodeling and its alterations have been shown to result in a variety of bone diseases. Osteoporosis, Paget’s disease, orthopedic disorders, osteopetrosis as well as hyperparathyroidism and hyperthyroidism all comprise conditions which have been linked with deregulated bone remodeling. Although the significance of mechanotransduction for bone health and disease is unquestionable, the mechanisms behind this important process have not been fully understood. This review will discuss the molecules that have been found to be implicated in mechanotransduction, as well as the mechanisms underlying bone health and disease, emphasizing on what is already known as well as new molecules potentially taking part in conveying mechanical signals from the cell surface towards the nucleus under physiological or pathologic conditions. It will also focus on the model systems currently used in mechanotransduction studies, like osteoblast-like cells as well as three-dimensional constructs and their applications among others. It will also examine the role of mechanostimulatory techniques in preventing and treating bone degenerative diseases and consider their applications in osteoporosis, craniofacial development, skeletal deregulations, fracture treatment, neurologic injuries following stroke or spinal cord injury, dentistry, hearing problems and bone implant integration in the near future.
Collapse
|
17
|
Wuertz K, Haglund L. Inflammatory mediators in intervertebral disk degeneration and discogenic pain. Global Spine J 2013; 3:175-84. [PMID: 24436868 PMCID: PMC3854585 DOI: 10.1055/s-0033-1347299] [Citation(s) in RCA: 161] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Accepted: 04/09/2013] [Indexed: 01/07/2023] Open
Abstract
Although degeneration of the intervertebral disk has historically been described as a misbalance between anabolic and catabolic factors, the role of inflammatory mediators has long been neglected. However, past research clearly indicates that inflammatory mediators such as interleukin (IL)-1β, IL-6, IL-8 and tumor necrosis factor-α are expressed at higher levels in "diseased" intervertebral disks. Both disk cells as well as invading macrophages can be the source of the detected cytokines. Importantly, occurrence of inflammatory mediators in the disk can worsen the progress of degeneration by inducing the expression of matrix degrading enzymes as well as by inhibiting extracellular matrix synthesis. In addition, inflammatory mediators play a crucial role in pain development during intervertebral disk herniation (i.e., sciatica) and disk degeneration (i.e., discogenic pain). This review provides information on the most relevant inflammatory mediators during different types of disk diseases and explains how these factors can induce disk degeneration and the development of discogenic and sciatic/radiculopathic pain.
Collapse
Affiliation(s)
- Karin Wuertz
- Institute for Biomechanics, D-HEST, ETH Zurich, Zurich, Switzerland,Competence Center for Applied Biotechnology and Molecular Medicine, University of Zurich, Zurich, Switzerland,AOSpine Research Network, Duebendorf, Switzerland,Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland,Address for correspondence Dr. Karin Wuertz, PhD Institute for Biomechanics, D-HESTETH Zurich, Schafmattstrasse 30, HPP-O12, 8093 ZurichSwitzerland
| | - Lisbet Haglund
- Competence Center for Applied Biotechnology and Molecular Medicine, University of Zurich, Zurich, Switzerland,Orthopeadic Research Laboratory, Division of Orthopedic Surgery, McGill University, Montreal, Canada,Dr. Lisbet Haglund, PhD Orthopaedic Research Laboratory, Montreal General HospitalRoom C9.173, 1650 Cedar Avenue, Montreal, QCCanada H3G 1A4
| |
Collapse
|