1
|
Xie X, Zhang G, Liu N. Comprehensive analysis of abnormal methylation modification differential expression mRNAs between low-grade and high-grade intervertebral disc degeneration and its correlation with immune cells. Ann Med 2024; 56:2357742. [PMID: 38819022 PMCID: PMC11146251 DOI: 10.1080/07853890.2024.2357742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 04/10/2024] [Indexed: 06/01/2024] Open
Abstract
BACKGROUND Intervertebral disc degeneration (IDD) is an important cause of low back pain. The aim of this study is to identify the potential molecular mechanism of abnormal methylation-modified DNA in the progression of IDD, hoping to contribute to the diagnosis and management of IDD. METHODS Low-grade IDD (grade I-II) and high-grade IDD (grade III-V) data were downloaded from GSE70362 and GSE129789 datasets. The abnormally methylated modified differentially expressed mRNAs (DEmRNAs) were identified by differential expression analysis (screening criteria were p < .05 and |logFC| > 1) and differential methylation analysis (screening criteria were p < .05 and |δβ| > 0.1). The classification models were constructed, and the receiver operating characteristic analysis was also carried out. In addition, functional enrichment analysis and immune correlation analysis were performed and the miRNAs targeted for the abnormally methylated DEmRNAs were predicted. Finally, expression validation was performed using real-time PCR. RESULTS Compared with low-grade IDD, seven abnormal methylation-modified DEmRNAs (AOX1, IBSP, QDPR, ABLIM1, CRISPLD2, ACTC1 and EMILIN1) were identified in high-grade IDD, and the classification models of random forests (RF) and support vector machine (SVM) were constructed. Moreover, seven abnormal methylation-modified DEmRNAs and classification models have high diagnostic accuracy (area under the curve [AUC] > 0.8). We also found that AUC values of single abnormal methylation-modified DEmRNA were all lower than those of RF and SVM classification models. Pearson correlation analysis found that macrophages M2 and EMILIN1 had significant negative correlation, while macrophages M2 and IBSP had significant positive correlation. In addition, four targeted relationship pairs (hsa-miR-4728-5p-QDPR, hsa-miR-4533-ABLIM1, hsa-miR-4728-5p-ABLIM1 and hsa-miR-4534-CRISPLD2) and multiple signalling pathways (for example, PI3K-AKT signalling pathway, osteoclast differentiation and calcium signalling pathway) were also identified that may be involved in the progression of IDD. CONCLUSION The identification of abnormal methylation-modified DEmRNAs and the construction of classification models in this study were helpful for the diagnosis and management of IDD progression.
Collapse
Affiliation(s)
- Xuehu Xie
- Department of Orthopedics, Beijing Friendship Hospital, Capital Medical University, Xicheng District, Beijing, China
| | - Guoqiang Zhang
- Department of Orthopedics, Beijing Friendship Hospital, Capital Medical University, Xicheng District, Beijing, China
| | - Ning Liu
- Department of Orthopedics, Beijing Friendship Hospital, Capital Medical University, Xicheng District, Beijing, China
| |
Collapse
|
2
|
Wakefield B, Tang J, Hutchinson JL, Kanji R, Brooks C, Grol MW, Séguin CA, Penuela S, Beier F. Pannexin 3 deletion in mice results in knee osteoarthritis and intervertebral disc degeneration after forced treadmill running. J Orthop Res 2024; 42:1696-1709. [PMID: 38499500 DOI: 10.1002/jor.25830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 11/10/2023] [Accepted: 02/26/2024] [Indexed: 03/20/2024]
Abstract
Pannexin 3 (Panx3) is a glycoprotein that forms mechanosensitive channels expressed in chondrocytes and annulus fibrosus cells of the intervertebral disc (IVD). Evidence suggests Panx3 plays contrasting roles in traumatic versus aging osteoarthritis (OA) and intervertebral disc degeneration (IDD). However, whether its deletion influences the response of joint tissue to forced use is unknown. The purpose of this study was to determine if Panx3 deletion in mice causes increased knee joint OA and IDD after forced treadmill running. Male and female wildtype (WT) and Panx3 knockout (KO) mice were randomized to either a no-exercise group (sedentary; SED) or daily forced treadmill running (forced exercise; FEX) from 24 to 30 weeks of age. Knee cartilage and IVD histopathology were evaluated by histology, while tibial secondary ossification centers were analyzed using microcomputed tomography (µCT). Both male and female Panx3 KO mice developed larger superficial defects of the tibial cartilage after forced treadmill running compared with SED WT mice. Additionally, Panx3 KO mice developed reduced bone volume, and female PANX3 KO mice had lengthening of the lateral tubercle at the intercondylar eminence. In the lower lumbar spine, both male and female Panx3 KO mice developed histopathological features of IDD after running compared to SED WT mice. These findings suggest that the combination of deleting Panx3 and forced treadmill running induces OA and causes histopathological changes associated with the degeneration of the IVDs in mice.
Collapse
Affiliation(s)
- Brent Wakefield
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
- Western's Bone and Joint Institute, The Dr. Sandy Kirkley Centre for Musculoskeletal Research, University Hospital, London, Ontario, Canada
| | - Justin Tang
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
- Western's Bone and Joint Institute, The Dr. Sandy Kirkley Centre for Musculoskeletal Research, University Hospital, London, Ontario, Canada
| | - Jeffrey L Hutchinson
- Western's Bone and Joint Institute, The Dr. Sandy Kirkley Centre for Musculoskeletal Research, University Hospital, London, Ontario, Canada
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
| | - Rehanna Kanji
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Courtney Brooks
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
| | - Matthew W Grol
- Western's Bone and Joint Institute, The Dr. Sandy Kirkley Centre for Musculoskeletal Research, University Hospital, London, Ontario, Canada
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
| | - Cheryle A Séguin
- Western's Bone and Joint Institute, The Dr. Sandy Kirkley Centre for Musculoskeletal Research, University Hospital, London, Ontario, Canada
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
| | - Silvia Penuela
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
- Western's Bone and Joint Institute, The Dr. Sandy Kirkley Centre for Musculoskeletal Research, University Hospital, London, Ontario, Canada
| | - Frank Beier
- Western's Bone and Joint Institute, The Dr. Sandy Kirkley Centre for Musculoskeletal Research, University Hospital, London, Ontario, Canada
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
3
|
Tan Z, Chen P, Dong X, Guo S, Leung VYL, Cheung JPY, Chan D, Richardson SM, Hoyland JA, To MKT, Cheah KSE. Progenitor-like cells contributing to cellular heterogeneity in the nucleus pulposus are lost in intervertebral disc degeneration. Cell Rep 2024; 43:114342. [PMID: 38865240 DOI: 10.1016/j.celrep.2024.114342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 03/14/2024] [Accepted: 05/23/2024] [Indexed: 06/14/2024] Open
Abstract
The nucleus pulposus (NP) in the intervertebral disc (IVD) arises from embryonic notochord. Loss of notochordal-like cells in humans correlates with onset of IVD degeneration, suggesting that they are critical for healthy NP homeostasis and function. Comparative transcriptomic analyses identified expression of progenitor-associated genes (GREM1, KRT18, and TAGLN) in the young mouse and non-degenerated human NP, with TAGLN expression reducing with aging. Lineage tracing using Tagln-CreERt2 mice identified peripherally located proliferative NP (PeriNP) cells in developing and postnatal NP that provide a continuous supply of cells to the entire NP. PeriNP cells were diminished in aged mice and absent in puncture-induced degenerated discs. Single-cell transcriptomes of postnatal Tagln-CreERt2 IVD cells indicate enrichment for TGF-β signaling in Tagln descendant NP sub-populations. Notochord-specific removal of TGF-β/BMP mediator Smad4 results in loss of Tagln+ cells and abnormal NP morphologies. We propose Tagln+ PeriNP cells are potential progenitors crucial for NP homeostasis.
Collapse
Affiliation(s)
- Zhijia Tan
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China; Department of Orthopaedics and Traumatology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China; Shenzhen Clinical Research Center for Rare Diseases, The University of Hong Kong - Shenzhen Hospital, Shenzhen, China; Department of Orthopaedics and Traumatology, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Peikai Chen
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China; Department of Orthopaedics and Traumatology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China; Shenzhen Clinical Research Center for Rare Diseases, The University of Hong Kong - Shenzhen Hospital, Shenzhen, China; Artificial Intelligence and Big Data Lab, The University of Hong Kong - Shenzhen Hospital, Shenzhen, China
| | - Xiaonan Dong
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Shuang Guo
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Victor Y L Leung
- Department of Orthopaedics and Traumatology, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Jason P Y Cheung
- Department of Orthopaedics and Traumatology, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Danny Chan
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Stephen M Richardson
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester M13 9PT, UK
| | - Judith A Hoyland
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester M13 9PT, UK
| | - Michael K T To
- Department of Orthopaedics and Traumatology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China; Shenzhen Clinical Research Center for Rare Diseases, The University of Hong Kong - Shenzhen Hospital, Shenzhen, China; Department of Orthopaedics and Traumatology, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Kathryn S E Cheah
- School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
4
|
Warin J, Vedrenne N, Tam V, Zhu M, Yin D, Lin X, Guidoux-D’halluin B, Humeau A, Roseiro L, Paillat L, Chédeville C, Chariau C, Riemers F, Templin M, Guicheux J, Tryfonidou MA, Ho JW, David L, Chan D, Camus A. In vitro and in vivo models define a molecular signature reference for human embryonic notochordal cells. iScience 2024; 27:109018. [PMID: 38357665 PMCID: PMC10865399 DOI: 10.1016/j.isci.2024.109018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 11/13/2023] [Accepted: 01/22/2024] [Indexed: 02/16/2024] Open
Abstract
Understanding the emergence of human notochordal cells (NC) is essential for the development of regenerative approaches. We present a comprehensive investigation into the specification and generation of bona fide NC using a straightforward pluripotent stem cell (PSC)-based system benchmarked with human fetal notochord. By integrating in vitro and in vivo transcriptomic data at single-cell resolution, we establish an extended molecular signature and overcome the limitations associated with studying human notochordal lineage at early developmental stages. We show that TGF-β inhibition enhances the yield and homogeneity of notochordal lineage commitment in vitro. Furthermore, this study characterizes regulators of cell-fate decision and matrisome enriched in the notochordal niche. Importantly, we identify specific cell-surface markers opening avenues for differentiation refinement, NC purification, and functional studies. Altogether, this study provides a human notochord transcriptomic reference that will serve as a resource for notochord identification in human systems, diseased-tissues modeling, and facilitating future biomedical research.
Collapse
Affiliation(s)
- Julie Warin
- Nantes Université, Oniris, CHU Nantes, Inserm, Regenerative Medicine and Skeleton, RMeS, UMR 1229, 44000 Nantes, France
| | - Nicolas Vedrenne
- Nantes Université, Oniris, CHU Nantes, Inserm, Regenerative Medicine and Skeleton, RMeS, UMR 1229, 44000 Nantes, France
- Inserm, Univ. Limoges, Pharmacology & Transplantation, U1248, CHU Limoges, Service de Pharmacologie, toxicologie et pharmacovigilance, FHU SUPORT, 87000 Limoges, France
| | - Vivian Tam
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Mengxia Zhu
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Danqing Yin
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
- Laboratory of Data Discovery for Health Limited (D24H), Hong Kong Science Park, Hong Kong SAR, China
| | - Xinyi Lin
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
- Laboratory of Data Discovery for Health Limited (D24H), Hong Kong Science Park, Hong Kong SAR, China
| | - Bluwen Guidoux-D’halluin
- Nantes Université, Oniris, CHU Nantes, Inserm, Regenerative Medicine and Skeleton, RMeS, UMR 1229, 44000 Nantes, France
| | - Antoine Humeau
- Inserm, Univ. Limoges, Pharmacology & Transplantation, U1248, CHU Limoges, Service de Pharmacologie, toxicologie et pharmacovigilance, FHU SUPORT, 87000 Limoges, France
| | - Luce Roseiro
- Nantes Université, Oniris, CHU Nantes, Inserm, Regenerative Medicine and Skeleton, RMeS, UMR 1229, 44000 Nantes, France
| | - Lily Paillat
- Nantes Université, Oniris, CHU Nantes, Inserm, Regenerative Medicine and Skeleton, RMeS, UMR 1229, 44000 Nantes, France
| | - Claire Chédeville
- Nantes Université, Oniris, CHU Nantes, Inserm, Regenerative Medicine and Skeleton, RMeS, UMR 1229, 44000 Nantes, France
| | - Caroline Chariau
- Nantes Université, CHU Nantes, Inserm, CNRS, BioCore, 44000 Nantes, France
| | - Frank Riemers
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Markus Templin
- NMI Natural and Medical Sciences Institute, Markwiesenstraße 55, 72770 Reutlingen, Germany
| | - Jérôme Guicheux
- Nantes Université, Oniris, CHU Nantes, Inserm, Regenerative Medicine and Skeleton, RMeS, UMR 1229, 44000 Nantes, France
| | - Marianna A. Tryfonidou
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Joshua W.K. Ho
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
- Laboratory of Data Discovery for Health Limited (D24H), Hong Kong Science Park, Hong Kong SAR, China
| | - Laurent David
- Nantes Université, CHU Nantes, Inserm, CNRS, BioCore, 44000 Nantes, France
- Nantes Université, CHU Nantes, Inserm, CR2TI, 44000 Nantes, France
| | - Danny Chan
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Anne Camus
- Nantes Université, Oniris, CHU Nantes, Inserm, Regenerative Medicine and Skeleton, RMeS, UMR 1229, 44000 Nantes, France
| |
Collapse
|
5
|
Zhuang Y, Liu L, Liu M, Fu J, Ai X, Long D, Leng X, Zhang Y, Gong X, Shang X, Li C, Huang B, Zhou Y, Ning X, Dong S, Feng C. The sonic hedgehog pathway suppresses oxidative stress and senescence in nucleus pulposus cells to alleviate intervertebral disc degeneration via GPX4. Biochim Biophys Acta Mol Basis Dis 2024; 1870:166961. [PMID: 37979732 DOI: 10.1016/j.bbadis.2023.166961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 11/06/2023] [Accepted: 11/07/2023] [Indexed: 11/20/2023]
Abstract
Disruption of intervertebral disc (IVD) homeostasis caused by oxidative stress and nucleus pulposus cell (NPC) senescence is a main cause of intervertebral disc degeneration (IDD). The sonic hedgehog (Shh) pathway plays an important role in IVD development, but its roles in IDD are unknown. This study aimed to investigate the effects of the Shh pathway on the alleviation of IDD and the related mechanisms. In vivo, the effect of the Shh pathway on IVD homeostasis was studied by intraperitoneal injection of recombinant Shh (rShh) and GANT61 based on puncture-induced IDD. GANT61, lentivirus-coated sh-Gli1 and rShh were used to investigate the role and mechanism of the Shh pathway in NPCs based on senescence induced by Braco19 and oxidative stress induced by TBHP. Shh pathway expression decreased, and senescence and oxidative stress increased with age. Intraperitoneal injection of rShh activated the Shh pathway to suppress oxidative stress and NPC senescence and consequently alleviated needle puncture-induced IDD. In vitro, the Shh pathway upregulated glutathione peroxidase 4 (GPX4) expression to suppress oxidative stress and senescence in NPCs. Moreover, GPX4 suppression in NPCs by si-GPX4 significantly reduced the protective effect of the Shh pathway on oxidative stress and senescence in NPCs. Our results demonstrate for the first time that the Shh pathway plays a key role in the alleviation of IDD by suppressing oxidative stress and cell senescence in NP tissues. This study provides a new potential target for the prevention and reversal of IDD.
Collapse
Affiliation(s)
- Yong Zhuang
- Department of Orthopedics, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, PR China
| | - Libangxi Liu
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, Chongqing 400037, PR China
| | - Miao Liu
- Department of Orthopedics, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, PR China
| | - Jiawei Fu
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, Chongqing 400037, PR China
| | - Xuezheng Ai
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, Chongqing 400037, PR China
| | - Dan Long
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, Chongqing 400037, PR China
| | - Xue Leng
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, Chongqing 400037, PR China
| | - Yang Zhang
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, Chongqing 400037, PR China
| | - Xunren Gong
- Xinqiao Hospital, Army Medical University, Chongqing 400037, PR China
| | - Xianwen Shang
- Department of Orthopedics, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, PR China
| | - Changqing Li
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, Chongqing 400037, PR China
| | - Bo Huang
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, Chongqing 400037, PR China
| | - Yue Zhou
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, Chongqing 400037, PR China
| | - Xu Ning
- Department of Orthopedics, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, PR China.
| | - Shiwu Dong
- Department of Biomedical Materials Science, School of Biomedical Engineering, Third Military Medical University, Chongqing 400038, PR China; State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University, Chongqing 400038, PR China.
| | - Chencheng Feng
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, Chongqing 400037, PR China.
| |
Collapse
|
6
|
Liu L, Zhang Y, Fu J, Ai X, Long D, Leng X, Zhang Y, Huang B, Li C, Zhou Y, Feng C. Gli1 depletion induces oxidative stress and apoptosis of nucleus pulposus cells via Fos in intervertebral disc degeneration. J Orthop Translat 2023; 40:116-131. [PMID: 37457313 PMCID: PMC10338909 DOI: 10.1016/j.jot.2023.05.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 04/01/2023] [Accepted: 05/19/2023] [Indexed: 07/18/2023] Open
Abstract
Background Intervertebral disc degeneration (IDD) is the most common chronic disease. Oxidative stress and apoptosis of nucleus pulposus (NP) cells disrupt intervertebral disc (IVD) homeostasis, which is the main cause of IDD. Glioma-associated oncogene 1 (Gli1) is an important transcription factor in the Hedgehog (Hh) pathway. Depletion of Gli1 accelerates the occurrence and development of degenerative diseases. This study aimed to explore the role of aging related Gli1 depletion in the progression of IDD. Methods The relationship between aging related Gli1 depletion and IDD was studied in the NP tissues of human and rats of different ages, and the levels of oxidative stress and NP cell apoptosis during IDD were explored. Gli1 depletion of NP cells were established by targeting inhibitor GANT61 or lentivirus-coated Gli1 sh-RNA (sh-Gli1) to explore the role of Gli1 in NP cells and underlying mechanism. Exogenous Gli1 depletion induced IDD of rats was established by intraperitoneal injection of GANT61. Also, the roles of Fos in the Gli1 depletion induced NP cell oxidative stress, apoptosis and IDD were investigated. Results Gli1 was down-regulated in the tissues of degenerative NP, and the level of Gli1 was negatively correlated with the severity of aging related IDD in human and rats. Furthermore, we found enhanced oxidative stress and apoptosis in degenerative NP tissues. Gli1 depletion promoted oxidative stress and apoptosis of NP cells and resulted in the degradation of extracellular matrix (ECM) and decreased ECM synthesis. Transcriptome sequencing showed that Gli1 depletion caused Fos activation in NP cells. the effect of Gli1 depletion on the oxidative stress and apoptosis of NP cells were retarded by Fos inhibitor. In vivo, Fos inhibition alleviated the IDD induced by exogenous Gli1 depletion. Conclusions This study revealed for the first time that Gli1 is gradually depleted in NP with IDD progression. Exogenous Gli1 depletion causes oxidative stress and apoptosis of NP cells both in vivo and in vitro. Fos suppression effectively retards the destructive effects of Gli1 depletion on IVD homoeostasis.The translational potential of this article: This study may provide new potential targets for preventing and reversing IDD. Maintaining Gli1 expression in NP and suppressing Fos activation may be an effective treatment strategy for IDD.
Collapse
|
7
|
Fine N, Lively S, Séguin CA, Perruccio AV, Kapoor M, Rampersaud R. Intervertebral disc degeneration and osteoarthritis: a common molecular disease spectrum. Nat Rev Rheumatol 2023; 19:136-152. [PMID: 36702892 DOI: 10.1038/s41584-022-00888-z] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/05/2022] [Indexed: 01/27/2023]
Abstract
Intervertebral disc degeneration (IDD) and osteoarthritis (OA) affecting the facet joint of the spine are biomechanically interdependent, typically occur in tandem, and have considerable epidemiological and pathophysiological overlap. Historically, the distinctions between these degenerative diseases have been emphasized. Therefore, research in the two fields often occurs independently without adequate consideration of the co-dependence of the two sites, which reside within the same functional spinal unit. Emerging evidence from animal models of spine degeneration highlight the interdependence of IDD and facet joint OA, warranting a review of the parallels between these two degenerative phenomena for the benefit of both clinicians and research scientists. This Review discusses the pathophysiological aspects of IDD and OA, with an emphasis on tissue, cellular and molecular pathways of degeneration. Although the intervertebral disc and synovial facet joint are biologically distinct structures that are amenable to reductive scientific consideration, substantial overlap exists between the molecular pathways and processes of degeneration (including cartilage destruction, extracellular matrix degeneration and osteophyte formation) that occur at these sites. Thus, researchers, clinicians, advocates and policy-makers should consider viewing the burden and management of spinal degeneration holistically as part of the OA disease continuum.
Collapse
Affiliation(s)
- Noah Fine
- Osteoarthritis Research Program, Division of Orthopaedics, Schroeder Arthritis Institute, University Health Network, Toronto, Ontario, Canada.,Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Starlee Lively
- Osteoarthritis Research Program, Division of Orthopaedics, Schroeder Arthritis Institute, University Health Network, Toronto, Ontario, Canada.,Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Cheryle Ann Séguin
- Department of Physiology & Pharmacology, Schulich School of Medicine & Dentistry, Bone and Joint Institute, University of Western Ontario London, London, Ontario, Canada
| | - Anthony V Perruccio
- Osteoarthritis Research Program, Division of Orthopaedics, Schroeder Arthritis Institute, University Health Network, Toronto, Ontario, Canada.,Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.,Institute of Health Policy, Management and Evaluation, Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada.,Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Mohit Kapoor
- Osteoarthritis Research Program, Division of Orthopaedics, Schroeder Arthritis Institute, University Health Network, Toronto, Ontario, Canada.,Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.,Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Raja Rampersaud
- Osteoarthritis Research Program, Division of Orthopaedics, Schroeder Arthritis Institute, University Health Network, Toronto, Ontario, Canada. .,Krembil Research Institute, University Health Network, Toronto, Ontario, Canada. .,Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
8
|
Whittal MC, Poynter SJ, Samms K, Briar KJ, Sinopoli SI, Millecamps M, Stone LS, DeWitte-Orr SJ, Gregory DE. TAK-242 treatment and its effect on mechanical properties and gene expression associated with IVD degeneration in SPARC-null mice. 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 2022; 31:2801-2811. [PMID: 35816198 DOI: 10.1007/s00586-022-07310-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 05/09/2022] [Accepted: 06/24/2022] [Indexed: 06/15/2023]
Abstract
PURPOSE Intervertebral disc (IVD) degeneration is accompanied by mechanical and gene expression changes to IVDs. SPARC-null mice display accelerated IVD degeneration, and treatment with (toll-like receptor 4 (TLR4) inhibitor) TAK-242 decreases proinflammatory cytokines and pain. This study examined if chronic TAK-242 treatment impacts mechanical properties and gene expression associated with IVD degeneration in SPARC-null mice. METHODS Male and female SPARC-null and WT mice aged 7-9 months were given intraperitoneal injections with TAK-242 or an equivalent saline vehicle for 8 weeks (3x/per week, M-W-F). L2-L5 spinal segments were tested in cyclic axial tension and compression. Gene expression analysis (RT-qPCR) was performed on male IVD tissues using Qiagen RT2 PCR arrays. RESULTS SPARC-null mice had decreased NZ length (p = 0.001) and increased NZ stiffness (p < 0.001) compared to WT mice. NZ length was not impacted by TAK-242 treatment (p = 0.967) despite increased hysteresis energy (p = 0.024). Tensile stiffness was greater in SPARC-null mice (p = 0.018), and compressive (p < 0.001) stiffness was reduced from TAK-242 treatment in WT but not SPARC-null mice (p = 0.391). Gene expression analysis found upregulation of 13 ECM and 5 inflammatory genes in SPARC-null mice, and downregulation of 2 inflammatory genes after TAK-242 treatment. CONCLUSIONS TAK-242 had limited impacts on SPARC-null mechanical properties and did not attenuate NZ mechanical changes associated with IVD degeneration. Expression analysis revealed an increase in ECM and inflammatory gene expression in SPARCnull mice with a reduction in inflammatory expression due to TAK-242 treatment. This study provides insight into the role of TLR4 in SPARC-null mediated IVD degeneration.
Collapse
Affiliation(s)
- Mitchel C Whittal
- Department of Kinesiology and Physical Education, Wilfrid Laurier University, 75 University Ave W, Waterloo, ON, N2L 3C5, Canada
| | - Sarah J Poynter
- Department of Health Sciences, Wilfrid Laurier University, 75 University Ave W, Waterloo, ON, N2L 3C5, Canada
| | - Kayla Samms
- Department of Health Sciences, Wilfrid Laurier University, 75 University Ave W, Waterloo, ON, N2L 3C5, Canada
| | - K Josh Briar
- Department of Kinesiology and Physical Education, Wilfrid Laurier University, 75 University Ave W, Waterloo, ON, N2L 3C5, Canada
| | - Sabrina I Sinopoli
- Department of Kinesiology and Physical Education, Wilfrid Laurier University, 75 University Ave W, Waterloo, ON, N2L 3C5, Canada
| | - Magali Millecamps
- McGill University, 845 Sherbrooke Street West, Montréal, QC, H3A 0G4, Canada
| | - Laura S Stone
- McGill University, 845 Sherbrooke Street West, Montréal, QC, H3A 0G4, Canada
- University of Minnesota, 321 Church Street SE, Minneapolis, MN, 55455, USA
| | - Stephanie J DeWitte-Orr
- Department of Health Sciences, Wilfrid Laurier University, 75 University Ave W, Waterloo, ON, N2L 3C5, Canada
| | - Diane E Gregory
- Department of Kinesiology and Physical Education, Wilfrid Laurier University, 75 University Ave W, Waterloo, ON, N2L 3C5, Canada.
- Department of Health Sciences, Wilfrid Laurier University, 75 University Ave W, Waterloo, ON, N2L 3C5, Canada.
| |
Collapse
|
9
|
Choi MR, Cho S, Kim DJ, Choi JS, Jin YB, Kim M, Chang HJ, Jeon SH, Yang YD, Lee SR. Effects of Ethanol on Expression of Coding and Noncoding RNAs in Murine Neuroblastoma Neuro2a Cells. Int J Mol Sci 2022; 23:ijms23137294. [PMID: 35806296 PMCID: PMC9267046 DOI: 10.3390/ijms23137294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/26/2022] [Accepted: 06/28/2022] [Indexed: 11/19/2022] Open
Abstract
Excessive use of alcohol can induce neurobiological and neuropathological alterations in the brain, including the hippocampus and forebrain, through changes in neurotransmitter systems, hormonal systems, and neuroimmune processes. We aimed to investigate the effects of ethanol on the expression of coding and noncoding RNAs in a brain-derived cell line exposed to ethanol. After exposing Neuro2a cells, a neuroblastoma cell line, to ethanol for 24 and 72 h, we observed cell proliferation and analyzed up- and downregulated mRNAs and long noncoding RNAs (lncRNAs) using total RNA-Seq technology. We validated the differential expression of some mRNAs and lncRNAs by RT-qPCR and analyzed the expression of Cebpd and Rnu3a through knock-down of Cebpd. Cell proliferation was significantly reduced in cells exposed to 100 mM ethanol for 72 h, with 1773 transcripts up- or downregulated by greater than three-fold in ethanol-treated cells compared to controls. Of these, 514 were identified as lncRNAs. Differentially expressed mRNAs and lncRNAs were mainly observed in cells exposed to ethanol for 72 h, in which Atm and Cnr1 decreased, but Trib3, Cebpd, and Spdef increased. On the other hand, lncRNAs Kcnq1ot1, Tug1, and Xist were changed by ethanol, and Rnu3a in particular was greatly increased by chronic ethanol treatment through inhibition of Cebpd. Our results increase the understanding of cellular and molecular mechanisms related to coding and noncoding RNAs in an in vitro model of acute and chronic exposure to ethanol.
Collapse
Affiliation(s)
- Mi Ran Choi
- Laboratory Animal Research Center, Ajou University School of Medicine, Suwon 16499, Korea;
| | - Sinyoung Cho
- Department of Pharmacy, College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Pocheon 11160, Korea; (S.C.); (S.H.J.)
| | - Dai-Jin Kim
- Department of Psychiatry, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea;
| | - Jung-Seok Choi
- Department of Psychiatry, Samsung Medical Center, Seoul 06351, Korea;
| | - Yeung-Bae Jin
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Korea;
| | - Miran Kim
- Department of Obstetrics and Gynecology, Ajou University School of Medicine, Suwon 16499, Korea; (M.K.); (H.J.C.)
| | - Hye Jin Chang
- Department of Obstetrics and Gynecology, Ajou University School of Medicine, Suwon 16499, Korea; (M.K.); (H.J.C.)
| | - Seong Ho Jeon
- Department of Pharmacy, College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Pocheon 11160, Korea; (S.C.); (S.H.J.)
| | - Young Duk Yang
- Department of Pharmacy, College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Pocheon 11160, Korea; (S.C.); (S.H.J.)
- Correspondence: (Y.D.Y.); (S.-R.L.); Tel.: +82-31-881-7170 (Y.D.Y.); +82-31-219-4499 (S.-R.L.)
| | - Sang-Rae Lee
- Laboratory Animal Research Center, Ajou University School of Medicine, Suwon 16499, Korea;
- Department of Pharmacology, Ajou University School of Medicine, Suwon 16499, Korea
- Correspondence: (Y.D.Y.); (S.-R.L.); Tel.: +82-31-881-7170 (Y.D.Y.); +82-31-219-4499 (S.-R.L.)
| |
Collapse
|
10
|
Importance of Matrix Cues on Intervertebral Disc Development, Degeneration, and Regeneration. Int J Mol Sci 2022; 23:ijms23136915. [PMID: 35805921 PMCID: PMC9266338 DOI: 10.3390/ijms23136915] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 06/17/2022] [Accepted: 06/20/2022] [Indexed: 01/25/2023] Open
Abstract
Back pain is one of the leading causes of disability worldwide and is frequently caused by degeneration of the intervertebral discs. The discs’ development, homeostasis, and degeneration are driven by a complex series of biochemical and physical extracellular matrix cues produced by and transmitted to native cells. Thus, understanding the roles of different cues is essential for designing effective cellular and regenerative therapies. Omics technologies have helped identify many new matrix cues; however, comparatively few matrix molecules have thus far been incorporated into tissue engineered models. These include collagen type I and type II, laminins, glycosaminoglycans, and their biomimetic analogues. Modern biofabrication techniques, such as 3D bioprinting, are also enabling the spatial patterning of matrix molecules and growth factors to direct regional effects. These techniques should now be applied to biochemically, physically, and structurally relevant disc models incorporating disc and stem cells to investigate the drivers of healthy cell phenotype and differentiation. Such research will inform the development of efficacious regenerative therapies and improved clinical outcomes.
Collapse
|
11
|
Single-Cell RNA-Seq Analysis of Cells from Degenerating and Non-Degenerating Intervertebral Discs from the Same Individual Reveals New Biomarkers for Intervertebral Disc Degeneration. Int J Mol Sci 2022; 23:ijms23073993. [PMID: 35409356 PMCID: PMC8999935 DOI: 10.3390/ijms23073993] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 03/31/2022] [Accepted: 04/01/2022] [Indexed: 02/07/2023] Open
Abstract
In this study, we used single-cell transcriptomic analysis to identify new specific biomarkers for nucleus pulposus (NP) and inner annulus fibrosis (iAF) cells, and to define cell populations within non-degenerating (nD) and degenerating (D) human intervertebral discs (IVD) of the same individual. Cluster analysis based on differential gene expression delineated 14 cell clusters. Gene expression profiles at single-cell resolution revealed the potential functional differences linked to degeneration, and among NP and iAF subpopulations. GO and KEGG analyses discovered molecular functions, biological processes, and transcription factors linked to cell type and degeneration state. We propose two lists of biomarkers, one as specific cell type, including C2orf40, MGP, MSMP, CD44, EIF1, LGALS1, RGCC, EPYC, HILPDA, ACAN, MT1F, CHI3L1, ID1, ID3 and TMED2. The second list proposes predictive IVD degeneration genes, including MT1G, SPP1, HMGA1, FN1, FBXO2, SPARC, VIM, CTGF, MGST1, TAF1D, CAPS, SPTSSB, S100A1, CHI3L2, PLA2G2A, TNRSF11B, FGFBP2, MGP, SLPI, DCN, MT-ND2, MTCYB, ADIRF, FRZB, CLEC3A, UPP1, S100A2, PRG4, COL2A1, SOD2 and MT2A. Protein and mRNA expression of MGST1, vimentin, SOD2 and SYF2 (p29) genes validated our scRNA-seq findings. Our data provide new insights into disc cells phenotypes and biomarkers of IVD degeneration that could improve diagnostic and therapeutic options.
Collapse
|
12
|
Intervertebral disc repair and regeneration: Insights from the notochord. Semin Cell Dev Biol 2021; 127:3-9. [PMID: 34865989 DOI: 10.1016/j.semcdb.2021.11.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 11/05/2021] [Accepted: 11/11/2021] [Indexed: 12/25/2022]
Abstract
The vertebrate notochord plays an essential role in patterning multiple structures during embryonic development. In the early 2000s, descendants of notochord cells were demonstrated to form the entire nucleus pulposus of the intervertebral disc in addition to their key role in embryonic patterning. The nucleus pulposus undergoes degeneration during postnatal life, which can lead to back pain. Recently, gene and protein profiles of notochord and nucleus pulposus cells have been identified. These datasets, coupled with the ability to differentiate human induced pluripotent stem cells (iPSCs) into cells that resemble nucleus pulposus cells, provide the possibility of generating a cell-based therapy to halt and/or reverse disc degeneration.
Collapse
|
13
|
Novais EJ, Tran VA, Johnston SN, Darris KR, Roupas AJ, Sessions GA, Shapiro IM, Diekman BO, Risbud MV. Long-term treatment with senolytic drugs Dasatinib and Quercetin ameliorates age-dependent intervertebral disc degeneration in mice. Nat Commun 2021; 12:5213. [PMID: 34480023 PMCID: PMC8417260 DOI: 10.1038/s41467-021-25453-2] [Citation(s) in RCA: 183] [Impact Index Per Article: 45.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 08/04/2021] [Indexed: 02/07/2023] Open
Abstract
Intervertebral disc degeneration is highly prevalent within the elderly population and is a leading cause of chronic back pain and disability. Due to the link between disc degeneration and senescence, we explored the ability of the Dasatinib and Quercetin drug combination (D + Q) to prevent an age-dependent progression of disc degeneration in mice. We treated C57BL/6 mice beginning at 6, 14, and 18 months of age, and analyzed them at 23 months of age. Interestingly, 6- and 14-month D + Q cohorts show lower incidences of degeneration, and the treatment results in a significant decrease in senescence markers p16INK4a, p19ARF, and SASP molecules IL-6 and MMP13. Treatment also preserves cell viability, phenotype, and matrix content. Although transcriptomic analysis shows disc compartment-specific effects of the treatment, cell death and cytokine response pathways are commonly modulated across tissue types. Results suggest that senolytics may provide an attractive strategy to mitigating age-dependent disc degeneration.
Collapse
Affiliation(s)
- Emanuel J. Novais
- grid.265008.90000 0001 2166 5843Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, USA ,grid.265008.90000 0001 2166 5843Graduate Program in Cell Biology and Regenerative Medicine, Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, USA ,grid.10328.380000 0001 2159 175XLife and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal ,grid.10328.380000 0001 2159 175XICVS/3B’s—PT Government Associate Laboratory, Braga, Portugal
| | - Victoria A. Tran
- grid.265008.90000 0001 2166 5843Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, USA
| | - Shira N. Johnston
- grid.265008.90000 0001 2166 5843Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, USA ,grid.265008.90000 0001 2166 5843Graduate Program in Cell Biology and Regenerative Medicine, Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, USA
| | - Kayla R. Darris
- grid.10698.360000000122483208Thurston Arthritis Research Center, University of North Carolina School of Medicine, Chapel Hill, NC USA ,grid.40803.3f0000 0001 2173 6074Department of Biomedical Engineering, University of North Carolina, Chapel Hill, NC, and North Carolina State University, Raleigh, NC USA
| | - Alex J. Roupas
- grid.10698.360000000122483208Thurston Arthritis Research Center, University of North Carolina School of Medicine, Chapel Hill, NC USA ,grid.40803.3f0000 0001 2173 6074Department of Biomedical Engineering, University of North Carolina, Chapel Hill, NC, and North Carolina State University, Raleigh, NC USA
| | - Garrett A. Sessions
- grid.10698.360000000122483208Thurston Arthritis Research Center, University of North Carolina School of Medicine, Chapel Hill, NC USA ,grid.40803.3f0000 0001 2173 6074Department of Biomedical Engineering, University of North Carolina, Chapel Hill, NC, and North Carolina State University, Raleigh, NC USA
| | - Irving M. Shapiro
- grid.265008.90000 0001 2166 5843Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, USA ,grid.265008.90000 0001 2166 5843Graduate Program in Cell Biology and Regenerative Medicine, Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, USA
| | - Brian O. Diekman
- grid.10698.360000000122483208Thurston Arthritis Research Center, University of North Carolina School of Medicine, Chapel Hill, NC USA ,grid.40803.3f0000 0001 2173 6074Department of Biomedical Engineering, University of North Carolina, Chapel Hill, NC, and North Carolina State University, Raleigh, NC USA
| | - Makarand V. Risbud
- grid.265008.90000 0001 2166 5843Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, USA ,grid.265008.90000 0001 2166 5843Graduate Program in Cell Biology and Regenerative Medicine, Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, USA
| |
Collapse
|
14
|
Zhang Y, Han S, Kong M, Tu Q, Zhang L, Ma X. Single-cell RNA-seq analysis identifies unique chondrocyte subsets and reveals involvement of ferroptosis in human intervertebral disc degeneration. Osteoarthritis Cartilage 2021; 29:1324-1334. [PMID: 34242803 DOI: 10.1016/j.joca.2021.06.010] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 05/28/2021] [Accepted: 06/25/2021] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Nucleus pulposus (NP) plays a central role in disc degeneration pathogenesis, however, as a heterogeneous tissue, cell subsets in NP and their corresponding biological process in intervertebral disc degeneration (IVDD) are unreported. METHOD Nucleus pulposus were isolated from normal control and IVDD, and then subjected to single-cell RNA sequencing (scRNA-seq). Unsupervised clustering of the cells based on the gene expression profiles using the Seurat package and passed to tSNE for clustering visualization. Rat model of disc degeneration was built to validate the pathways identified by scRNA-Seq. RESULTS Seven chondrocyte subsets were revealed in NP based on differential gene expression, among which 4 subsets (C1-C4) were reported for the first time. Furthermore, GO and KEGG analyses discovered that ferroptosis pathways were enriched. Rat model of disc degeneration was built (n = 6/group, control vs. model) to validate the pathways identified by scRNA-Seq. Iron levels of NP were significantly higher in model group than control group (means 0.712 vs. 0.248, respectively, mg/gpro, p = 0.0026), and the levels of Heme Oxygenase 1 (HO-1) were also elevated in model group (means 14.33 vs. 5.16 IOD, respectively, p = 0.0002). However, the levels of ferritin light chain (FTL) were significantly decreased in model group compared to control group (means 26.17 vs. 9.00 FTL+ cell number, respectively, p = 0.0011). CONCLUSIONS Novel chondrocyte subsets in nucleus pulposus were discovered through scRNA-Seq, which provided novel insight to understand the pathological change during the development of IVDD. Ferroptosis participated in disc degeneration pathogenesis and it might serve as a new target for intervening IVDD.
Collapse
Affiliation(s)
- Y Zhang
- Shandong Institute of Orthopaedics and Traumatology, Medical Research Center, Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - S Han
- Department of Spinal Surgery, Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - M Kong
- Department of Spinal Surgery, Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Q Tu
- Department of Spinal Surgery, Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - L Zhang
- Systems Biology & Medicine Center for Complex Diseases, Affiliated Hospital of Qingdao University, Qingdao, 266003, China.
| | - X Ma
- Department of Spinal Surgery, Affiliated Hospital of Qingdao University, Qingdao, 266003, China.
| |
Collapse
|
15
|
Kudelko M, Chen P, Tam V, Zhang Y, Kong OY, Sharma R, Au TY, To MKT, Cheah KS, Chan WC, Chan D. PRIMUS: Comprehensive proteomics of mouse intervertebral discs that inform novel biology and relevance to human disease modelling. Matrix Biol Plus 2021; 12:100082. [PMID: 34409283 PMCID: PMC8361275 DOI: 10.1016/j.mbplus.2021.100082] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 07/08/2021] [Accepted: 07/15/2021] [Indexed: 12/21/2022] Open
Abstract
Proteomics of healthy mouse IVDs differentiating compartments and spine levels. NP cells feature vacuoles with lysosomal, transport and cell–cell communication functions. Collagen XII, decorin and other ECM proteins contribute to function of the AF. Distinct proteomics between lumbar and tail discs. Mouse is a relevant model for human disc biology but care is needed in its use.
Mice are commonly used to study intervertebral disc (IVD) biology and related diseases such as IVD degeneration. Discs from both the lumbar and tail regions are used. However, little is known about compartmental characteristics in the different regions, nor their relevance to the human setting, where a functional IVD unit depends on a homeostatic proteome. Here, we address these major gaps through comprehensive proteomic profiling and in-depth analyses of 8-week-old healthy murine discs, followed by comparisons with human. Leveraging on a dataset of over 2,700 proteins from 31 proteomic profiles, we identified key molecular and cellular differences between disc compartments and spine levels, but not gender. The nucleus pulposus (NP) and annulus fibrosus (AF) compartments differ the most, both in matrisome and cellularity contents. Differences in the matrisome are consistent with the fibrous nature required for tensile strength in the AF and hydration property in the NP. Novel findings for the NP cells included an enrichment in cell junction proteins for cell–cell communication (Cdh2, Dsp and Gja1) and osmoregulation (Slc12a2 and Wnk1). In NP cells, we detected heterogeneity of vacuolar organelles; where about half have potential lysosomal function (Vamp3, Copb2, Lamp1/2, Lamtor1), some contain lipid droplets and others with undefined contents. The AF is enriched in proteins for the oxidative stress responses (Sod3 and Clu). Interestingly, mitochondrial proteins are elevated in the lumbar than tail IVDs that may reflect differences in metabolic requirement. Relative to the human, cellular and structural information are conserved for the AF. Even though the NP is more divergent between mouse and human, there are similarities at the level of cell biology. Further, common cross-species markers were identified for both NP (KRT8/19, CD109) and AF (COL12A1). Overall, mouse is a relevant model to study IVD biology, and an understanding of the limitation will facilitate research planning and data interpretation, maximizing the translation of research findings to human IVDs.
Collapse
Affiliation(s)
- Mateusz Kudelko
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong
| | - Peikai Chen
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong
- Department of Orthopaedics Surgery and Traumatology, The University of Hong Kong -Shenzhen Hospital (HKU-SZH), Shenzhen, China
| | - Vivian Tam
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong
| | - Ying Zhang
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong
| | - Oi-Yin Kong
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong
| | - Rakesh Sharma
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong
- Proteomics and Metabolomics Core Facility, The University of Hong Kong, Pokfulam, Hong Kong
| | - Tiffany Y.K. Au
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong
| | - Michael Kai-Tsun To
- Department of Orthopaedics Surgery and Traumatology, The University of Hong Kong -Shenzhen Hospital (HKU-SZH), Shenzhen, China
- Department of Orthopaedics and Traumatology, The University of Hong Kong, Hong Kong
| | - Kathryn S.E. Cheah
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong
| | - Wilson C.W. Chan
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong
- Department of Orthopaedics Surgery and Traumatology, The University of Hong Kong -Shenzhen Hospital (HKU-SZH), Shenzhen, China
| | - Danny Chan
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong
- Department of Orthopaedics Surgery and Traumatology, The University of Hong Kong -Shenzhen Hospital (HKU-SZH), Shenzhen, China
- Corresponding author at: School of Biomedical Sciences, Faculty of Medicine Building, 21 Sassoon Road, Pokfulam, Hong Kong.
| |
Collapse
|
16
|
O'Donnell BL, Penuela S. Pannexin 3 channels in health and disease. Purinergic Signal 2021; 17:577-589. [PMID: 34250568 DOI: 10.1007/s11302-021-09805-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 06/30/2021] [Indexed: 01/07/2023] Open
Abstract
Pannexin 3 (PANX3) is a member of the pannexin family of single membrane channel-forming glycoproteins. Originally thought to have a limited localization in cartilage, bone, and skin, PANX3 has now been detected in a variety of other tissues including skeletal muscle, mammary glands, the male reproductive tract, the cochlea, blood vessels, small intestines, teeth, and the vomeronasal organ. In many cell types of the musculoskeletal system, such as osteoblasts, chondrocytes, and odontoblasts, PANX3 has been shown to regulate the balance of proliferation and differentiation. PANX3 can be induced during progenitor cell differentiation, functioning at the cell surface as a conduit for ATP and/or in the endoplasmic reticulum as a calcium leak channel. Evidence in osteoblasts and monocytes also highlight a role for PANX3 in purinergic signalling through its function as an ATP release channel. PANX3 is critical in the development and ageing of bone and cartilage, with its levels temporally regulated in other tissues such as skeletal muscle, skin, and the cochlea. In diseases such as osteoarthritis and intervertebral disc degeneration, PANX3 can have either protective or detrimental roles depending on if the disease is age-related or injury-induced. This review will discuss PANX3 function in tissue growth and regeneration, its role in cellular differentiation, and how it becomes dysregulated in disease conditions such as obesity, Duchenne's muscular dystrophy, osteosarcoma, and non-melanoma skin cancer, where most of the findings on PANX3 function can be attributed to the characterization of Panx3 KO mouse models.
Collapse
Affiliation(s)
- Brooke L O'Donnell
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada
| | - Silvia Penuela
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada.
- Department of Oncology, Division of Experimental Oncology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, N6A 5C1, Canada.
| |
Collapse
|
17
|
The Cellular Composition of Bovine Coccygeal Intervertebral Discs: A Comprehensive Single-Cell RNAseq Analysis. Int J Mol Sci 2021. [DOI: 10.3390/ijms22094917
expr 996488947 + 961598850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Abstract
Intervertebral disc (IVD) degeneration and its medical consequences is still one of the leading causes of morbidity worldwide. To support potential regenerative treatments for degenerated IVDs, we sought to deconvolute the cell composition of the nucleus pulposus (NP) and the annulus fibrosus (AF) of bovine intervertebral discs. Bovine calf tails have been extensively used in intervertebral disc research as a readily available source of NP and AF material from healthy and young IVDs. We used single-cell RNA sequencing (scRNAseq) coupled to bulk RNA sequencing (RNAseq) to unravel the cell populations in these two structures and analyze developmental changes across the rostrocaudal axis. By integrating the scRNAseq data with the bulk RNAseq data to stabilize the clustering results of our study, we identified 27 NP structure/tissue specific genes and 24 AF structure/tissue specific genes. From our scRNAseq results, we could deconvolute the heterogeneous cell populations in both the NP and the AF. In the NP, we detected a notochordal-like cell cluster and a progenitor stem cell cluster. In the AF, we detected a stem cell-like cluster, a cluster with a predominantly fibroblast-like phenotype and a potential endothelial progenitor cluster. Taken together, our results illustrate the cell phenotypic complexity of the AF and NP in the young bovine IVDs.
Collapse
|
18
|
Calió M, Gantenbein B, Egli M, Poveda L, Ille F. The Cellular Composition of Bovine Coccygeal Intervertebral Discs: A Comprehensive Single-Cell RNAseq Analysis. Int J Mol Sci 2021; 22:ijms22094917. [PMID: 34066404 PMCID: PMC8124861 DOI: 10.3390/ijms22094917] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/26/2021] [Accepted: 04/27/2021] [Indexed: 02/07/2023] Open
Abstract
Intervertebral disc (IVD) degeneration and its medical consequences is still one of the leading causes of morbidity worldwide. To support potential regenerative treatments for degenerated IVDs, we sought to deconvolute the cell composition of the nucleus pulposus (NP) and the annulus fibrosus (AF) of bovine intervertebral discs. Bovine calf tails have been extensively used in intervertebral disc research as a readily available source of NP and AF material from healthy and young IVDs. We used single-cell RNA sequencing (scRNAseq) coupled to bulk RNA sequencing (RNAseq) to unravel the cell populations in these two structures and analyze developmental changes across the rostrocaudal axis. By integrating the scRNAseq data with the bulk RNAseq data to stabilize the clustering results of our study, we identified 27 NP structure/tissue specific genes and 24 AF structure/tissue specific genes. From our scRNAseq results, we could deconvolute the heterogeneous cell populations in both the NP and the AF. In the NP, we detected a notochordal-like cell cluster and a progenitor stem cell cluster. In the AF, we detected a stem cell-like cluster, a cluster with a predominantly fibroblast-like phenotype and a potential endothelial progenitor cluster. Taken together, our results illustrate the cell phenotypic complexity of the AF and NP in the young bovine IVDs.
Collapse
Affiliation(s)
- Martina Calió
- Tissue Engineering for Orthopaedics & Mechanobiology (TOM), Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland; (M.C.); (B.G.)
- Department of Orthopaedic Surgery and Traumatology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland
- Space Biology Group, Institute of Medical Engineering, School of Engineering and Architecture, Lucerne University of Applied Sciences and Arts, 6052 Hergiswil, Switzerland;
| | - Benjamin Gantenbein
- Tissue Engineering for Orthopaedics & Mechanobiology (TOM), Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland; (M.C.); (B.G.)
- Department of Orthopaedic Surgery and Traumatology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland
| | - Marcel Egli
- Space Biology Group, Institute of Medical Engineering, School of Engineering and Architecture, Lucerne University of Applied Sciences and Arts, 6052 Hergiswil, Switzerland;
| | - Lucy Poveda
- Functional Genomics Center Zurich, Swiss Federal Institute of Technology, University of Zurich, 8057 Zurich, Switzerland;
| | - Fabian Ille
- Space Biology Group, Institute of Medical Engineering, School of Engineering and Architecture, Lucerne University of Applied Sciences and Arts, 6052 Hergiswil, Switzerland;
- Correspondence: ; Tel.: +41-41-349-36-15
| |
Collapse
|
19
|
Serjeant M, Moon PM, Quinonez D, Penuela S, Beier F, Séguin CA. The Role of Panx3 in Age-Associated and Injury-Induced Intervertebral Disc Degeneration. Int J Mol Sci 2021; 22:ijms22031080. [PMID: 33499145 PMCID: PMC7865929 DOI: 10.3390/ijms22031080] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 01/06/2021] [Accepted: 01/19/2021] [Indexed: 01/08/2023] Open
Abstract
Pannexin 3 (Panx3) is a mechanosensitive, channel-forming glycoprotein implicated in the progression of post-traumatic osteoarthritis. Despite evidence for Panx3 expression in the intervertebral disc (IVD), its function in this cartilaginous joint structure remained unknown. Using Panx3 knockout mice, this study investigated the role of Panx3 in age-associated IVD degeneration and degeneration induced by annulus fibrosus (AF) needle puncture. Loss of Panx3 did not significantly impact the progression of age-associated histopathological IVD degeneration; however, loss of Panx3 was associated with decreased gene expression of Acan, Col1a1, Mmp13 and Runx2 and altered localization of COLX in the IVD at 19 months-of-age. Following IVD injury in the caudal spine, histological analysis of wild-type mice revealed clusters of hypertrophic cells in the AF associated with increased pericellular proteoglycan accumulation, disruptions in lamellar organization and increased lamellar thickness. In Panx3 knockout mice, hypertrophic AF cells were rarely detected and AF structure was largely preserved post-injury. Interestingly, uninjured IVDs adjacent to the site of injury more frequently showed evidence of early nucleus pulposus degeneration in Panx3 knockout mice but remained healthy in wild-type mice. These findings suggest a role for Panx3 in mediating the adaptive cellular responses to altered mechanical stress in the IVD, which may buffer aberrant loads transferred to adjacent motion segments.
Collapse
Affiliation(s)
- Meaghan Serjeant
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON N6A 5C1, Canada; (M.S.); (P.M.M.); (D.Q.); (F.B.)
- Bone and Joint Institute, The University of Western Ontario, London, ON N6A 5C1, Canada;
| | - Paxton M. Moon
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON N6A 5C1, Canada; (M.S.); (P.M.M.); (D.Q.); (F.B.)
- Bone and Joint Institute, The University of Western Ontario, London, ON N6A 5C1, Canada;
| | - Diana Quinonez
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON N6A 5C1, Canada; (M.S.); (P.M.M.); (D.Q.); (F.B.)
| | - Silvia Penuela
- Bone and Joint Institute, The University of Western Ontario, London, ON N6A 5C1, Canada;
- Department of Anatomy and Cell Biology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON N6A 5C1, Canada
| | - Frank Beier
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON N6A 5C1, Canada; (M.S.); (P.M.M.); (D.Q.); (F.B.)
- Bone and Joint Institute, The University of Western Ontario, London, ON N6A 5C1, Canada;
| | - Cheryle A. Séguin
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, ON N6A 5C1, Canada; (M.S.); (P.M.M.); (D.Q.); (F.B.)
- Bone and Joint Institute, The University of Western Ontario, London, ON N6A 5C1, Canada;
- Correspondence:
| |
Collapse
|
20
|
Tam V, Chen P, Yee A, Solis N, Klein T, Kudelko M, Sharma R, Chan WC, Overall CM, Haglund L, Sham PC, Cheah KSE, Chan D. DIPPER, a spatiotemporal proteomics atlas of human intervertebral discs for exploring ageing and degeneration dynamics. eLife 2020; 9:64940. [PMID: 33382035 PMCID: PMC7857729 DOI: 10.7554/elife.64940] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 12/30/2020] [Indexed: 12/11/2022] Open
Abstract
The spatiotemporal proteome of the intervertebral disc (IVD) underpins its integrity and function. We present DIPPER, a deep and comprehensive IVD proteomic resource comprising 94 genome-wide profiles from 17 individuals. To begin with, protein modules defining key directional trends spanning the lateral and anteroposterior axes were derived from high-resolution spatial proteomes of intact young cadaveric lumbar IVDs. They revealed novel region-specific profiles of regulatory activities and displayed potential paths of deconstruction in the level- and location-matched aged cadaveric discs. Machine learning methods predicted a ‘hydration matrisome’ that connects extracellular matrix with MRI intensity. Importantly, the static proteome used as point-references can be integrated with dynamic proteome (SILAC/degradome) and transcriptome data from multiple clinical samples, enhancing robustness and clinical relevance. The data, findings, and methodology, available on a web interface (http://www.sbms.hku.hk/dclab/DIPPER/), will be valuable references in the field of IVD biology and proteomic analytics. The backbone of vertebrate animals consists of a series of bones called vertebrae that are joined together by disc-like structures that allow the back to move and distribute forces to protect it during daily activities. It is common for these intervertebral discs to degenerate with age, resulting in back pain and severely reducing quality of life. The mechanical features of intervertebral discs are the result of their proteins. These include extracellular matrix proteins, which form the external scaffolding that binds cells together in a tissue, and signaling proteins, which allow cells to communicate. However, how the levels of different proteins in each region of the disc vary with time has not been fully examined. To establish how protein composition changes with age, Tam, Chen et al. quantified the protein levels and gene activity (which leads to protein production) of intervertebral discs from young and old deceased individuals. They found that the position of different mixtures of proteins in the intervertebral disc changes with age, and that young people have high levels of extracellular matrix proteins and signaling proteins. Levels of these proteins decreased as people got older, as did the amount of proteins produced. To determine which region of the intervertebral disc different proteins were in, Tam, Chen et al. also performed magnetic resonance imaging (MRI) of the samples to correlate image intensity (which represents water content) with the corresponding protein signature. The data obtained provides a high-quality map of how the location of different proteins changes with age, and is available online under the name DIPPER. This database is an informative resource for research into skeletal biology, and it will likely advance the understanding of intervertebral disc degeneration in humans and animals, potentially leading to the development of new treatment strategies for this condition.
Collapse
Affiliation(s)
- Vivian Tam
- School of Biomedical Sciences,, The University of Hong Kong, Hong Kong.,The University of Hong Kong Shenzhen of Research Institute and Innovation (HKU-SIRI), Shenzhen, China
| | - Peikai Chen
- School of Biomedical Sciences,, The University of Hong Kong, Hong Kong
| | - Anita Yee
- School of Biomedical Sciences,, The University of Hong Kong, Hong Kong
| | - Nestor Solis
- Centre for Blood Research, Faculty of Dentistry, University of British Columbia, Vancouver, Canada
| | - Theo Klein
- Centre for Blood Research, Faculty of Dentistry, University of British Columbia, Vancouver, Canada
| | - Mateusz Kudelko
- School of Biomedical Sciences,, The University of Hong Kong, Hong Kong
| | - Rakesh Sharma
- Proteomics and Metabolomics Core Facility, The University of Hong Kong, Hong Kong
| | - Wilson Cw Chan
- School of Biomedical Sciences,, The University of Hong Kong, Hong Kong.,The University of Hong Kong Shenzhen of Research Institute and Innovation (HKU-SIRI), Shenzhen, China.,Department of Orthopaedics Surgery and Traumatology, HKU-Shenzhen Hospital, Shenzhen, China
| | - Christopher M Overall
- Centre for Blood Research, Faculty of Dentistry, University of British Columbia, Vancouver, Canada
| | - Lisbet Haglund
- Department of Surgery, McGill University, Montreal, Canada
| | - Pak C Sham
- Centre for PanorOmic Sciences (CPOS), The University of Hong Kong, Hong Kong
| | | | - Danny Chan
- School of Biomedical Sciences,, The University of Hong Kong, Hong Kong.,The University of Hong Kong Shenzhen of Research Institute and Innovation (HKU-SIRI), Shenzhen, China
| |
Collapse
|
21
|
Kim MKM, Burns MJ, Serjeant ME, Séguin CA. The mechano-response of murine annulus fibrosus cells to cyclic tensile strain is frequency dependent. JOR Spine 2020; 3:e21114. [PMID: 33392464 PMCID: PMC7770207 DOI: 10.1002/jsp2.1114] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 05/20/2020] [Accepted: 06/29/2020] [Indexed: 12/11/2022] Open
Abstract
The intervertebral disk (IVD) is a composite structure essential for spine stabilization, load bearing, and movement. Biomechanical factors are important contributors to the IVD microenvironment regulating joint homeostasis; however, the cell type-specific effectors of mechanotransduction in the IVD are not fully understood. The current study aimed to determine the effects of cyclic tensile strain (CTS) on annulus fibrosus (AF) cells and identify mechano-sensitive pathways. Using a cell-type specific reporter mouse to differentiation NP and AF cells from the murine IVD, we characterized AF cells in dynamic culture exposed to CTS (6% strain) at specific frequencies (0.1 Hz, 1.0 Hz, or 2.0 Hz). We demonstrate that our culture model maintains the phenotype of primary AF cells and that the bioreactor system delivers uniform biaxial strain across the cell culture surface. We show that exposure of AF cells to CTS induces cytoskeleton reorganization resulting in stress fiber formation, with acute exposure to CTS at 2.0 Hz inducing a significant yet transient increase ERK1/2 pathway activation. Using SYBPR-based qPCR to assess the expression of extracellular matrix (ECM) genes, ECM-remodeling genes, candidate mechano-sensitive genes, inflammatory cytokines and cell surface receptors, we demonstrated that exposure of AF cells to CTS at 0.1 Hz increased Acan, Prg4, Col1a1 and Mmp3 expression. AF cells exposed to CTS at 1.0 Hz showed a significant increase in the expression of Acan, Myc, and Tnfα. Exposure of AF cells to CTS at 2.0 Hz induced a significant increase in Acan, Prg4, Cox2, Myc, Fos, and Tnfα expression. Among the cell surface receptors assessed, AF cells exposed to CTS at 2.0 Hz showed a significant increase in Itgβ1, Itgα5, and Trpv4 expression. Our findings demonstrate that the response of AF cells to CTS is frequency dependent and suggest that mechanical loading may directly contribute to matrix remodeling and the onset of local tissue inflammation in the murine IVD.
Collapse
Affiliation(s)
- Min Kyu M. Kim
- Department of Physiology and PharmacologySchulich School of Medicine & Dentistry, The University of Western OntarioLondonOntarioCanada
- Bone and Joint Institute, The University of Western OntarioLondonOntarioCanada
| | - Marissa J. Burns
- Department of Physiology and PharmacologySchulich School of Medicine & Dentistry, The University of Western OntarioLondonOntarioCanada
| | - Meaghan E. Serjeant
- Department of Physiology and PharmacologySchulich School of Medicine & Dentistry, The University of Western OntarioLondonOntarioCanada
- Bone and Joint Institute, The University of Western OntarioLondonOntarioCanada
| | - Cheryle A. Séguin
- Department of Physiology and PharmacologySchulich School of Medicine & Dentistry, The University of Western OntarioLondonOntarioCanada
- Bone and Joint Institute, The University of Western OntarioLondonOntarioCanada
| |
Collapse
|
22
|
Fernandes LM, Khan NM, Trochez CM, Duan M, Diaz-Hernandez ME, Presciutti SM, Gibson G, Drissi H. Single-cell RNA-seq identifies unique transcriptional landscapes of human nucleus pulposus and annulus fibrosus cells. Sci Rep 2020; 10:15263. [PMID: 32943704 PMCID: PMC7499307 DOI: 10.1038/s41598-020-72261-7] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 08/19/2020] [Indexed: 12/29/2022] Open
Abstract
Intervertebral disc (IVD) disease (IDD) is a complex, multifactorial disease. While various aspects of IDD progression have been reported, the underlying molecular pathways and transcriptional networks that govern the maintenance of healthy nucleus pulposus (NP) and annulus fibrosus (AF) have not been fully elucidated. We defined the transcriptome map of healthy human IVD by performing single-cell RNA-sequencing (scRNA-seq) in primary AF and NP cells isolated from non-degenerated lumbar disc. Our systematic and comprehensive analyses revealed distinct genetic architecture of human NP and AF compartments and identified 2,196 differentially expressed genes. Gene enrichment analysis showed that SFRP1, BIRC5, CYTL1, ESM1 and CCNB2 genes were highly expressed in the AF cells; whereas, COL2A1, DSC3, COL9A3, COL11A1, and ANGPTL7 were mostly expressed in the NP cells. Further, functional annotation clustering analysis revealed the enrichment of receptor signaling pathways genes in AF cells, while NP cells showed high expression of genes related to the protein synthesis machinery. Subsequent interaction network analysis revealed a structured network of extracellular matrix genes in NP compartments. Our regulatory network analysis identified FOXM1 and KDM4E as signature transcription factor of AF and NP respectively, which might be involved in the regulation of core genes of AF and NP transcriptome.
Collapse
Affiliation(s)
- Lorenzo M Fernandes
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA, 30033, USA.,Atlanta VA Medical Center, Decatur, GA, USA
| | - Nazir M Khan
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA, 30033, USA.,Atlanta VA Medical Center, Decatur, GA, USA
| | - Camila M Trochez
- Center for Integrative Genomics, Georgia Institute of Technology, Atlanta, GA, USA
| | - Meixue Duan
- Center for Integrative Genomics, Georgia Institute of Technology, Atlanta, GA, USA
| | - Martha E Diaz-Hernandez
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA, 30033, USA.,Atlanta VA Medical Center, Decatur, GA, USA
| | - Steven M Presciutti
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA, 30033, USA.,Atlanta VA Medical Center, Decatur, GA, USA
| | - Greg Gibson
- Center for Integrative Genomics, Georgia Institute of Technology, Atlanta, GA, USA
| | - Hicham Drissi
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA, 30033, USA. .,Atlanta VA Medical Center, Decatur, GA, USA.
| |
Collapse
|
23
|
Piprode V, Mohanty S, Bonavita R, Loh S, Anbazhagan R, Saini C, Pinelli R, Pricop P, Dahia CL. An optimized step-by-step protocol for isolation of nucleus pulposus, annulus fibrosus, and end plate cells from the mouse intervertebral discs and subsequent preparation of high-quality intact total RNA. JOR Spine 2020; 3:e1108. [PMID: 33015579 PMCID: PMC7524240 DOI: 10.1002/jsp2.1108] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 05/27/2020] [Accepted: 06/29/2020] [Indexed: 12/14/2022] Open
Abstract
Intervertebral disc degeneration is the most significant, and least understood, cause of chronic back pain, affecting almost one in seven individuals at some point of time. Each intervertebral disc has three components; central nucleus pulposus (NP), concentric layers of annulus fibrosus (AF), and a pair of end plate (EP) that connects the disc to the vertebral bodies. Understanding the molecular and cellular basis of intervertebral disc growth, health, and aging will generate significant information for developing therapeutic approaches. Rapid and efficient preparations of homogeneous and pure cells are crucial for meaningful and rigorous downstream analysis at the cellular, molecular, and biochemical level. Cross-sample contamination may influence the interpretation of the results. In addition to altering gene expression, slow or delayed isolation procedures will also cause the degradation of cells and biomolecules that create a bias in the outcomes of the study. The mouse model system is extensively used to understand the intervertebral disc biology. Here we describe two protocols: (a) for efficient isolation of pure NP, AF, and EP cells from mouse lumbar intervertebral disc. We validated the purity of the NP and AF cells using Shh Cre/+ ; R26 mT/mG/+ dual-fluorescent reporter mice where all NP cells are GPF+ve, and by the sensitive approach of qPCR analysis using TaqMan probes for Shh, and Brachyury as NP-specific markers, Tenomodulin as AF-specific marker, and Osteocalcin as bone-specific marker. (b) For isolation of high-quality intact RNA with RIN of 9.3 to 10 from disc cells. These methods will be useful for the rigorous analysis of NP and AF cells, and improve our understanding of intervertebral disc biology.
Collapse
Affiliation(s)
| | | | | | - Sarah Loh
- Hospital for Special Surgery New York New York USA
| | | | | | | | - Paul Pricop
- Hospital for Special Surgery New York New York USA
| | - Chitra L Dahia
- Hospital for Special Surgery New York New York USA
- Department of Cell and Developmental Biology, Weill Cornell Medicine Graduate School of Medical Sciences New York New York USA
| |
Collapse
|
24
|
Veras MA, Lim YJ, Kuljanin M, Lajoie GA, Urquhart BL, Séguin CA. Protocol for parallel proteomic and metabolomic analysis of mouse intervertebral disc tissues. JOR Spine 2020; 3:e1099. [PMID: 33015574 PMCID: PMC7524214 DOI: 10.1002/jsp2.1099] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 04/25/2020] [Accepted: 05/14/2020] [Indexed: 01/07/2023] Open
Abstract
The comprehensiveness of data collected by "omics" modalities has demonstrated the ability to drastically transform our understanding of the molecular mechanisms of chronic, complex diseases such as musculoskeletal pathologies, how biomarkers are identified, and how therapeutic targets are developed. Standardization of protocols will enable comparisons between findings reported by multiple research groups and move the application of these technologies forward. Herein, we describe a protocol for parallel proteomic and metabolomic analysis of mouse intervertebral disc (IVD) tissues, building from the combined expertise of our collaborative team. This protocol covers dissection of murine IVD tissues, sample isolation, and data analysis for both proteomics and metabolomics applications. The protocol presented below was optimized to maximize the utility of a mouse model for "omics" applications, accounting for the challenges associated with the small starting quantity of sample due to small tissue size as well as the extracellular matrix-rich nature of the tissue.
Collapse
Affiliation(s)
- Matthew A Veras
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry The University of Western Ontario London Ontario Canada
- Bone and Joint Institute The University of Western Ontario London Ontario Canada
| | - Yong J Lim
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry The University of Western Ontario London Ontario Canada
| | - Miljan Kuljanin
- Department of Cell Biology Harvard Medical School Boston Massachusetts USA
| | - Gilles A Lajoie
- Department of Biochemistry, Schulich School of Medicine & Dentistry The University of Western Ontario London Ontario Canada
| | - Bradley L Urquhart
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry The University of Western Ontario London Ontario Canada
| | - Cheryle A Séguin
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry The University of Western Ontario London Ontario Canada
- Bone and Joint Institute The University of Western Ontario London Ontario Canada
| |
Collapse
|
25
|
Impaired chondrocyte U3 snoRNA expression in osteoarthritis impacts the chondrocyte protein translation apparatus. Sci Rep 2020; 10:13426. [PMID: 32778764 PMCID: PMC7417995 DOI: 10.1038/s41598-020-70453-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 07/23/2020] [Indexed: 12/18/2022] Open
Abstract
Although pathways controlling ribosome activity have been described to regulate chondrocyte homeostasis in osteoarthritis, ribosome biogenesis in osteoarthritis is unexplored. We hypothesized that U3 snoRNA, a non-coding RNA involved in ribosomal RNA maturation, is critical for chondrocyte protein translation capacity in osteoarthritis. U3 snoRNA was one of a number of snoRNAs with decreased expression in osteoarthritic cartilage and osteoarthritic chondrocytes. OA synovial fluid impacted U3 snoRNA expression by affecting U3 snoRNA gene promoter activity, while BMP7 was able to increase its expression. Altering U3 snoRNA expression resulted in changes in chondrocyte phenotype. Interference with U3 snoRNA expression led to reduction of rRNA levels and translational capacity, whilst induced expression of U3 snoRNA was accompanied by increased 18S and 28S rRNA levels and elevated protein translation. Whole proteome analysis revealed a global impact of reduced U3 snoRNA expression on protein translational processes and inflammatory pathways. For the first time we demonstrate implications of a snoRNA in osteoarthritis chondrocyte biology and investigated its role in the chondrocyte differentiation status, rRNA levels and protein translational capacity.
Collapse
|
26
|
Diekman BO. Dynamic interplay between connective tissues and the surrounding environment during aging. Connect Tissue Res 2020; 61:1-3. [PMID: 31782325 DOI: 10.1080/03008207.2020.1682282] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- Brian O'Callaghan Diekman
- Thurston Arthritis Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.,Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA and North Carolina State University, Raleigh, NC, USA
| |
Collapse
|
27
|
Bratsman A, Couasnay G, Elefteriou F. A step-by-step protocol for isolation of murine nucleus pulposus cells. JOR Spine 2019; 2:e1073. [PMID: 31891122 PMCID: PMC6920701 DOI: 10.1002/jsp2.1073] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 10/28/2019] [Accepted: 11/07/2019] [Indexed: 02/06/2023] Open
Abstract
The intervertebral disc (IVD) is composed of three separate tissues with distinct origins and properties. Elucidating changes occurring in these tissues in response to injury or age is paramount to identify new therapies to better manage disc and spine degenerative conditions, including low back pain. Despite their small size and different mechanical load pattern compared to higher species, the use of mouse models represents a cost-effective and powerful approach to better understand the formation, maintenance, and degeneration of the IVD. However, the isolation of the different compartments of the IVD is complicated by their diminutive size. Here, we describe a simple, step-by-step protocol for the isolation of the nucleus pulposus (NP) tissues that can then be processed for further analyses. Analysis from mouse NP tissues shows sufficient quantities of RNAs, purity of the NP fraction, and overall RNA quality for gene expression studies, and reveals no increase in expression of disc degeneration markers, including TNFa, IL1b, and Mmp1 up to 15 months of age in C57BL6 wildtype mice.
Collapse
Affiliation(s)
- Andrew Bratsman
- Department of Orthopedic SurgeryBaylor College of MedicineHoustonTexas
| | - Greig Couasnay
- Department of Orthopedic SurgeryBaylor College of MedicineHoustonTexas
| | - Florent Elefteriou
- Department of Orthopedic SurgeryBaylor College of MedicineHoustonTexas
- Department of Molecular and Human GeneticsBaylor College of MedicineHoustonTexas
| |
Collapse
|