1
|
Xu X, Liu Y, Jiang C, Jia P, Cao P, He Y, Zhang Y. Mechanism of microRNA-124-3p targeting calpain-1 to affect the function of intervertebral disc nucleus pulposus cells. Cytotechnology 2025; 77:53. [PMID: 39897108 PMCID: PMC11785900 DOI: 10.1007/s10616-024-00693-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 12/19/2024] [Indexed: 02/04/2025] Open
Abstract
Intervertebral disc degeneration (IVDD) represents a major cause of lower back pain, whose prevalence rises with age. This study probed into the mechanism of microRNA (miR)-124-3p regulating function of nucleus pulposus cells (NPCs) by targeting calpain-1 (CAPN1). Rat IVD NPCs were cultured in vitro and transfected with miR-124-3p mimics, miR-124-3p inhibitor, oe-CAPN1 and their negative controls. The mRNA levels of miR-124-3p and CAPN1 were assessed by RT-qPCR. Cell proliferation, apoptosis and migration were evaluated by CCK-8, flow cytometry and Transwell assays. Levels of CAPN1 protein, apoptosis-related proteins (BAX, Cleaved-Caspase3, BCL-2) and extracellular matrix (ECM) proteins (Collagen II, Aggrecan, Fibronectin, Collagen I, matrix metalloproteinase [MMP]-13) were determined by Western blot. The target binding relationship between miR-124-3p and CAPN1 was verified by dual-luciferase assay. miR-124-3p overexpression facilitated NPC function and the maintenance of ECM homeostasis, as evidenced by increased NPC proliferation and migration, decreased apoptosis, elevated apoptosis-related protein BCL-2 level, diminished BAX and Cleaved-Caspase3 levels, reduced levels of ECM homeostasis-associated factors Collagen I and MMP-13 proteins, as well as raised levels of Collagen II, Aggrecan and Fibronectin proteins. Conversely, miR-124-3p knockdown brought about the opposite results. miR-124-3p targeted CAPN1. Furthermore, overexpression of CAPN1 partially reversed the regulatory effects of miR-124-3p on the ECM homeostasis, proliferation and migration in NPCs, and promoted apoptosis. miR-124-3p contributed to proliferation and migration of IVD NPCs, and reduced their apoptosis by inhibiting CAPN1 expression, thereby modulating ECM homeostasis and maintaining the function of IVD NPCs.
Collapse
Affiliation(s)
- Xunan Xu
- Department of Orthopedics, People’s Hospital, Suzhou High-tech Zone, No.95 Huashan Road, Suzhou, 215129 Jiangsu China
| | - Yong Liu
- Department of Orthopedics, People’s Hospital, Suzhou High-tech Zone, No.95 Huashan Road, Suzhou, 215129 Jiangsu China
| | - Chun Jiang
- Department of Orthopedics, People’s Hospital, Suzhou High-tech Zone, No.95 Huashan Road, Suzhou, 215129 Jiangsu China
| | - Peng Jia
- Department of Orthopedics, People’s Hospital, Suzhou High-tech Zone, No.95 Huashan Road, Suzhou, 215129 Jiangsu China
| | - Pengfei Cao
- Department of Orthopedics, People’s Hospital, Suzhou High-tech Zone, No.95 Huashan Road, Suzhou, 215129 Jiangsu China
| | - Yi He
- Department of Orthopedics, People’s Hospital, Suzhou High-tech Zone, No.95 Huashan Road, Suzhou, 215129 Jiangsu China
| | - Yin Zhang
- Department of Orthopedics, People’s Hospital, Suzhou High-tech Zone, No.95 Huashan Road, Suzhou, 215129 Jiangsu China
| |
Collapse
|
2
|
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
|
3
|
Yu H, Khanshour AM, Ushiki A, Otomo N, Koike Y, Einarsdottir E, Fan Y, Antunes L, Kidane YH, Cornelia R, Sheng RR, Zhang Y, Pei J, Grishin NV, Evers BM, Cheung JPY, Herring JA, Terao C, Song YQ, Gurnett CA, Gerdhem P, Ikegawa S, Rios JJ, Ahituv N, Wise CA. Association of genetic variation in COL11A1 with adolescent idiopathic scoliosis. eLife 2024; 12:RP89762. [PMID: 38277211 PMCID: PMC10945706 DOI: 10.7554/elife.89762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2024] Open
Abstract
Adolescent idiopathic scoliosis (AIS) is a common and progressive spinal deformity in children that exhibits striking sexual dimorphism, with girls at more than fivefold greater risk of severe disease compared to boys. Despite its medical impact, the molecular mechanisms that drive AIS are largely unknown. We previously defined a female-specific AIS genetic risk locus in an enhancer near the PAX1 gene. Here, we sought to define the roles of PAX1 and newly identified AIS-associated genes in the developmental mechanism of AIS. In a genetic study of 10,519 individuals with AIS and 93,238 unaffected controls, significant association was identified with a variant in COL11A1 encoding collagen (α1) XI (rs3753841; NM_080629.2_c.4004C>T; p.(Pro1335Leu); p=7.07E-11, OR = 1.118). Using CRISPR mutagenesis we generated Pax1 knockout mice (Pax1-/-). In postnatal spines we found that PAX1 and collagen (α1) XI protein both localize within the intervertebral disc-vertebral junction region encompassing the growth plate, with less collagen (α1) XI detected in Pax1-/- spines compared to wild-type. By genetic targeting we found that wild-type Col11a1 expression in costal chondrocytes suppresses expression of Pax1 and of Mmp3, encoding the matrix metalloproteinase 3 enzyme implicated in matrix remodeling. However, the latter suppression was abrogated in the presence of the AIS-associated COL11A1P1335L mutant. Further, we found that either knockdown of the estrogen receptor gene Esr2 or tamoxifen treatment significantly altered Col11a1 and Mmp3 expression in chondrocytes. We propose a new molecular model of AIS pathogenesis wherein genetic variation and estrogen signaling increase disease susceptibility by altering a PAX1-COL11a1-MMP3 signaling axis in spinal chondrocytes.
Collapse
Affiliation(s)
- Hao Yu
- Center for Translational Research, Scottish Rite for ChildrenDallasUnited States
| | - Anas M Khanshour
- Center for Translational Research, Scottish Rite for ChildrenDallasUnited States
| | - Aki Ushiki
- Department of Bioengineering and Therapeutic Sciences, University of California, San FranciscoSan FranciscoUnited States
- Institute for Human Genetics, University of California, San FranciscoSan FranciscoUnited States
| | - Nao Otomo
- Laboratory of Bone and Joint Diseases, RIKEN Center for Integrative Medical SciencesTokyoJapan
| | - Yoshinao Koike
- Laboratory of Bone and Joint Diseases, RIKEN Center for Integrative Medical SciencesTokyoJapan
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical SciencesYokohamaJapan
| | - Elisabet Einarsdottir
- Science for Life Laboratory, Department of Gene Technology, KTH-Royal Institute of TechnologySolnaSweden
| | - Yanhui Fan
- School of Biomedical Sciences, The University of Hong KongHong Kong SARChina
| | - Lilian Antunes
- Department of Neurology, Washington University in St. LouisSt. LouisUnited States
| | - Yared H Kidane
- Center for Translational Research, Scottish Rite for ChildrenDallasUnited States
| | - Reuel Cornelia
- Center for Translational Research, Scottish Rite for ChildrenDallasUnited States
| | - Rory R Sheng
- Department of Bioengineering and Therapeutic Sciences, University of California, San FranciscoSan FranciscoUnited States
- Institute for Human Genetics, University of California, San FranciscoSan FranciscoUnited States
| | - Yichi Zhang
- Department of Bioengineering and Therapeutic Sciences, University of California, San FranciscoSan FranciscoUnited States
- Institute for Human Genetics, University of California, San FranciscoSan FranciscoUnited States
- School of Pharmaceutical Sciences, Tsinghua UniversityBeijingChina
| | - Jimin Pei
- Department of Biophysics, University of Texas Southwestern Medical CenterDallasUnited States
| | - Nick V Grishin
- Department of Biophysics, University of Texas Southwestern Medical CenterDallasUnited States
| | - Bret M Evers
- Department of Pathology, University of Texas Southwestern Medical CenterDallasUnited States
- Department of Ophthalmology, University of Texas Southwestern Medical CenterDallasUnited States
| | - Jason Pui Yin Cheung
- Department of Orthopaedics and Traumatology LKS Faculty of Medicine, The University of Hong KongHong Kong SARChina
| | - John A Herring
- Department of Orthopedic Surgery, Scottish Rite for ChildrenDallasUnited States
- Department of Orthopaedic Surgery, University of Texas Southwestern Medical CenterDallasUnited States
| | - Chikashi Terao
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical SciencesYokohamaJapan
| | - You-qiang Song
- School of Biomedical Sciences, The University of Hong KongHong Kong SARChina
| | - Christina A Gurnett
- Department of Neurology, Washington University in St. LouisSt. LouisUnited States
| | - Paul Gerdhem
- Department of Surgical Sciences, Uppsala UniversityUppsalaSweden
- Department of Orthopaedics and Hand Surgery, Uppsala University HospitalUppsalaSweden
- Department of Clinical Science, Intervention & Technology (CLINTEC), Karolinska Institutet, Stockholm, Uppsala UniversityUppsalaSweden
| | - Shiro Ikegawa
- Laboratory of Bone and Joint Diseases, RIKEN Center for Integrative Medical SciencesTokyoJapan
| | - Jonathan J Rios
- Center for Translational Research, Scottish Rite for ChildrenDallasUnited States
- Department of Orthopaedic Surgery, University of Texas Southwestern Medical CenterDallasUnited States
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical CenterDallasUnited States
- Department of Pediatrics, University of Texas Southwestern Medical CenterDallasUnited States
| | - Nadav Ahituv
- Department of Bioengineering and Therapeutic Sciences, University of California, San FranciscoSan FranciscoUnited States
- Institute for Human Genetics, University of California, San FranciscoSan FranciscoUnited States
| | - Carol A Wise
- Center for Translational Research, Scottish Rite for ChildrenDallasUnited States
- Department of Orthopaedic Surgery, University of Texas Southwestern Medical CenterDallasUnited States
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical CenterDallasUnited States
- Department of Pediatrics, University of Texas Southwestern Medical CenterDallasUnited States
| |
Collapse
|
4
|
Novais EJ, Narayanan R, Canseco JA, van de Wetering K, Kepler CK, Hilibrand AS, Vaccaro AR, Risbud MV. A new perspective on intervertebral disc calcification-from bench to bedside. Bone Res 2024; 12:3. [PMID: 38253615 PMCID: PMC10803356 DOI: 10.1038/s41413-023-00307-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 12/04/2023] [Accepted: 12/05/2023] [Indexed: 01/24/2024] Open
Abstract
Disc degeneration primarily contributes to chronic low back and neck pain. Consequently, there is an urgent need to understand the spectrum of disc degeneration phenotypes such as fibrosis, ectopic calcification, herniation, or mixed phenotypes. Amongst these phenotypes, disc calcification is the least studied. Ectopic calcification, by definition, is the pathological mineralization of soft tissues, widely studied in the context of conditions that afflict vasculature, skin, and cartilage. Clinically, disc calcification is associated with poor surgical outcomes and back pain refractory to conservative treatment. It is frequently seen as a consequence of disc aging and progressive degeneration but exhibits unique molecular and morphological characteristics: hypertrophic chondrocyte-like cell differentiation; TNAP, ENPP1, and ANK upregulation; cell death; altered Pi and PPi homeostasis; and local inflammation. Recent studies in mouse models have provided a better understanding of the mechanisms underlying this phenotype. It is essential to recognize that the presentation and nature of mineralization differ between AF, NP, and EP compartments. Moreover, the combination of anatomic location, genetics, and environmental stressors, such as aging or trauma, govern the predisposition to calcification. Lastly, the systemic regulation of calcium and Pi metabolism is less important than the local activity of PPi modulated by the ANK-ENPP1 axis, along with disc cell death and differentiation status. While there is limited understanding of this phenotype, understanding the molecular pathways governing local intervertebral disc calcification may lead to developing disease-modifying drugs and better clinical management of degeneration-related pathologies.
Collapse
Affiliation(s)
- Emanuel J Novais
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
- Unidade Local de Saúde do Litoral Alentejano, Orthopedic Department, Santiago do Cacém, Portugal
| | - Rajkishen Narayanan
- Rothman Orthopedic Institute at Thomas Jefferson University, Philadelphia, PA, USA
| | - Jose A Canseco
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
- Rothman Orthopedic Institute at Thomas Jefferson University, Philadelphia, PA, USA
| | - Koen van de Wetering
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Christopher K Kepler
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
- Rothman Orthopedic Institute at Thomas Jefferson University, Philadelphia, PA, USA
| | - Alan S Hilibrand
- Rothman Orthopedic Institute at Thomas Jefferson University, Philadelphia, PA, USA
| | - Alexander R Vaccaro
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
- Rothman Orthopedic Institute at Thomas Jefferson University, Philadelphia, PA, USA
| | - Makarand V Risbud
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
5
|
Leão Monteiro R. Future of low back pain: unravelling IVD components and MSCs' potential. CELL REGENERATION (LONDON, ENGLAND) 2024; 13:1. [PMID: 38227139 PMCID: PMC10792145 DOI: 10.1186/s13619-023-00184-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 11/27/2023] [Indexed: 01/17/2024]
Abstract
Low back pain (LBP) mainly emerges from intervertebral disc (IVD) degeneration. However, the failing mechanism of IVD ́s components, like the annulus fibrosus (AF) and nucleus pulposus (NP), leading to IVD degeneration/herniation is still poorly understood. Moreover, the specific role of cellular populations and molecular pathways involved in the inflammatory process associated with IVD herniation remains to be highlighted. The limited knowledge of inflammation associated with the initial steps of herniation and the lack of suitable models to mimic human IVD ́s complexity are some of the reasons for that. It has become essential to enhance the knowledge of cellular and molecular key players for AF and NP cells during inflammatory-driven degeneration. Due to unique properties of immunomodulation and pluripotency, mesenchymal stem cells (MSCs) have attained diverse recognition in this field of bone and cartilage regeneration. MSCs therapy has been particularly valuable in facilitating repair of damaged tissues and may benefit in mitigating inflammation' degenerative events. Therefore, this review article conducts comprehensive research to further understand the intertwine between the mechanisms of action of IVD components and therapeutic potential of MSCs, exploring their characteristics, how to optimize their use and establish them safely in distinct settings for LPB treatment.
Collapse
|
6
|
Yu H, Khanshour AM, Ushiki A, Otomo N, Koike Y, Einarsdottir E, Fan Y, Antunes L, Kidane YH, Cornelia R, Sheng R, Zhang Y, Pei J, Grishin NV, Evers BM, Cheung JPY, Herring JA, Terao C, Song YQ, Gurnett CA, Gerdhem P, Ikegawa S, Rios JJ, Ahituv N, Wise CA. Association of genetic variation in COL11A1 with adolescent idiopathic scoliosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.26.542293. [PMID: 37292598 PMCID: PMC10245954 DOI: 10.1101/2023.05.26.542293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Adolescent idiopathic scoliosis (AIS) is a common and progressive spinal deformity in children that exhibits striking sexual dimorphism, with girls at more than five-fold greater risk of severe disease compared to boys. Despite its medical impact, the molecular mechanisms that drive AIS are largely unknown. We previously defined a female-specific AIS genetic risk locus in an enhancer near the PAX1 gene. Here we sought to define the roles of PAX1 and newly-identified AIS-associated genes in the developmental mechanism of AIS. In a genetic study of 10,519 individuals with AIS and 93,238 unaffected controls, significant association was identified with a variant in COL11A1 encoding collagen (α1) XI (rs3753841; NM_080629.2_c.4004C>T; p.(Pro1335Leu); P=7.07e-11, OR=1.118). Using CRISPR mutagenesis we generated Pax1 knockout mice (Pax1-/-). In postnatal spines we found that PAX1 and collagen (α1) XI protein both localize within the intervertebral disc (IVD)-vertebral junction region encompassing the growth plate, with less collagen (α1) XI detected in Pax1-/- spines compared to wildtype. By genetic targeting we found that wildtype Col11a1 expression in costal chondrocytes suppresses expression of Pax1 and of Mmp3, encoding the matrix metalloproteinase 3 enzyme implicated in matrix remodeling. However, this suppression was abrogated in the presence of the AIS-associated COL11A1P1335L mutant. Further, we found that either knockdown of the estrogen receptor gene Esr2, or tamoxifen treatment, significantly altered Col11a1 and Mmp3 expression in chondrocytes. We propose a new molecular model of AIS pathogenesis wherein genetic variation and estrogen signaling increase disease susceptibility by altering a Pax1-Col11a1-Mmp3 signaling axis in spinal chondrocytes.
Collapse
Affiliation(s)
- Hao Yu
- Center for Pediatric Bone Biology and Translational Research, Scottish Rite for Children, Dallas, TX, USA
| | - Anas M Khanshour
- Center for Pediatric Bone Biology and Translational Research, Scottish Rite for Children, Dallas, TX, USA
| | - Aki Ushiki
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA
| | - Nao Otomo
- Laboratory of Bone and Joint Diseases, RIKEN Center for Integrative Medical Sciences, Tokyo, JP
| | - Yoshinao Koike
- Laboratory of Bone and Joint Diseases, RIKEN Center for Integrative Medical Sciences, Tokyo, JP
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, JP
| | - Elisabet Einarsdottir
- Science for Life Laboratory, Department of Gene Technology, KTH-Royal Institute of Technology, Solna, SE
| | - Yanhui Fan
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, CN
| | - Lilian Antunes
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
| | - Yared H Kidane
- Center for Pediatric Bone Biology and Translational Research, Scottish Rite for Children, Dallas, TX, USA
| | - Reuel Cornelia
- Center for Pediatric Bone Biology and Translational Research, Scottish Rite for Children, Dallas, TX, USA
| | - Rory Sheng
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA
| | - Yichi Zhang
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, CN
| | - Jimin Pei
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Nick V Grishin
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Bret M Evers
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jason Pui Yin Cheung
- Department of Orthopaedics and Traumatology LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, CN
| | - John A Herring
- Department of Orthopedic Surgery, Scottish Rite for Children, Dallas, TX, USA
- Department of Orthopaedic Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Chikashi Terao
- Laboratory for Statistical and Translational Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, JP
| | - You-Qiang Song
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, CN
| | - Christina A Gurnett
- Department of Neurology, Washington University in St. Louis, St. Louis, MO, USA
| | - Paul Gerdhem
- Department of Clinical Science, Intervention & Technology (CLINTEC), Karolinska Institutet, Stockholm, Uppsala University, Uppsala, SE
- Department of Surgical Sciences, Uppsala University and
- Department of Orthopaedics and Hand Surgery, Uppsala University Hospital, Uppsala, SE
| | - Shiro Ikegawa
- Laboratory of Bone and Joint Diseases, RIKEN Center for Integrative Medical Sciences, Tokyo, JP
| | - Jonathan J Rios
- Center for Pediatric Bone Biology and Translational Research, Scottish Rite for Children, Dallas, TX, USA
- Department of Orthopaedic Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Nadav Ahituv
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA
| | - Carol A Wise
- Center for Pediatric Bone Biology and Translational Research, Scottish Rite for Children, Dallas, TX, USA
- Department of Orthopaedic Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
7
|
Xu M, Huang J, Jin M, Jiang W, Luo F, Tan Q, Zhang R, Luo X, Kuang L, Zhang D, Liang S, Qi H, Chen H, Ni Z, Su N, Yang J, Du X, Chen B, Deng C, Xie Y, Chen L. Expansion of FGFR3-positive nucleus pulposus cells plays important roles in postnatal nucleus pulposus growth and regeneration. Stem Cell Res Ther 2022; 13:227. [PMID: 35659742 PMCID: PMC9166488 DOI: 10.1186/s13287-022-02903-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 11/29/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Intervertebral disc degeneration (IVDD) can cause low back pain, a major public health concern. IVDD is characterized with loss of cells especially those in nucleus pulposus (NP), due to the limited proliferative potential and regenerative ability. Few studies, however, have been carried out to investigate the in vivo proliferation events of NP cells and the cellular contribution of a specific subpopulation of NP during postnatal growth or regeneration. METHODS We generated FGFR3-3*Flag-IRES-GFP mice and crossed FGFR3-CreERT2 mice with Rosa26-mTmG, Rosa26-DTA and Rosa26-Confetti mice, respectively, to perform inducible genetic tracing studies. RESULTS Expression of FGFR3 was found in the outer region of NP with co-localized expressions of proliferating markers. By fate mapping studies, FGFR3-positive (FGFR3+) NP cells were found proliferate from outer region to inner region of NP during postnatal growth. Clonal lineage tracing by Confetti mice and ablation of FGFR3·+ NP cells by DTA mice further revealed that the expansion of the FGFR3+ cells was required for the morphogenesis and homeostasis of postnatal NP. Moreover, in degeneration and regeneration model of mouse intervertebral disc, FGFR3+ NP cells underwent extensive expansion during the recovery stage. CONCLUSION Our present work demonstrates that FGFR3+ NP cells are novel subpopulation of postnatal NP with long-existing proliferative capacity shaping the adult NP structure and participating in the homeostasis maintenance and intrinsic repair of NP. These findings may facilitate the development of new therapeutic approaches for IVD regeneration.
Collapse
Affiliation(s)
- Meng Xu
- Laboratory of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing, 400042, China.,Department of Rehabilitation Medicine, General Hospital of Central Theater Command of Chinese People's Liberation Army, Wuhan, China
| | - Junlan Huang
- Laboratory of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Min Jin
- Laboratory of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Wanling Jiang
- Laboratory of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Fengtao Luo
- Laboratory of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Qiaoyan Tan
- Laboratory of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Ruobin Zhang
- Laboratory of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Xiaoqing Luo
- Laboratory of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Liang Kuang
- Laboratory of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Dali Zhang
- Laboratory of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Sen Liang
- Laboratory of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Huabing Qi
- Laboratory of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Hangang Chen
- Laboratory of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Zhenhong Ni
- Laboratory of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Nan Su
- Laboratory of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Jing Yang
- Laboratory of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Xiaolan Du
- Laboratory of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Bo Chen
- Department of Spine Surgery, Institute of Surgery Research, Daping Hospital, Army Medical University, Chongqing, China
| | - Chuxia Deng
- Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Yangli Xie
- Laboratory of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing, 400042, China.
| | - Lin Chen
- Laboratory of Wound Repair and Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Daping Hospital, Army Medical University, Chongqing, 400042, China.
| |
Collapse
|
8
|
Bach FC, Poramba-Liyanage DW, Riemers FM, Guicheux J, Camus A, Iatridis JC, Chan D, Ito K, Le Maitre CL, Tryfonidou MA. Notochordal Cell-Based Treatment Strategies and Their Potential in Intervertebral Disc Regeneration. Front Cell Dev Biol 2022; 9:780749. [PMID: 35359916 PMCID: PMC8963872 DOI: 10.3389/fcell.2021.780749] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 11/15/2021] [Indexed: 12/20/2022] Open
Abstract
Chronic low back pain is the number one cause of years lived with disability. In about 40% of patients, chronic lower back pain is related to intervertebral disc (IVD) degeneration. The standard-of-care focuses on symptomatic relief, while surgery is the last resort. Emerging therapeutic strategies target the underlying cause of IVD degeneration and increasingly focus on the relatively overlooked notochordal cells (NCs). NCs are derived from the notochord and once the notochord regresses they remain in the core of the developing IVD, the nucleus pulposus. The large vacuolated NCs rapidly decline after birth and are replaced by the smaller nucleus pulposus cells with maturation, ageing, and degeneration. Here, we provide an update on the journey of NCs and discuss the cell markers and tools that can be used to study their fate and regenerative capacity. We review the therapeutic potential of NCs for the treatment of IVD-related lower back pain and outline important future directions in this area. Promising studies indicate that NCs and their secretome exerts regenerative effects, via increased proliferation, extracellular matrix production, and anti-inflammatory effects. Reports on NC-like cells derived from embryonic- or induced pluripotent-stem cells claim to have successfully generated NC-like cells but did not compare them with native NCs for phenotypic markers or in terms of their regenerative capacity. Altogether, this is an emerging and active field of research with exciting possibilities. NC-based studies demonstrate that cues from developmental biology can pave the path for future clinical therapies focused on regenerating the diseased IVD.
Collapse
Affiliation(s)
- Frances C. Bach
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | | | - Frank M. Riemers
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Jerome Guicheux
- UMR 1229-RMeS, Regenerative Medicine and Skeleton, Université de Nantes, ONIRIS, Nantes, France
- UFR Odontologie, Université de Nantes, Nantes, France
- PHU4 OTONN, CHU Nantes, Nantes, France
| | - Anne Camus
- UMR 1229-RMeS, Regenerative Medicine and Skeleton, Université de Nantes, ONIRIS, Nantes, France
| | - James C. Iatridis
- Leni and Peter W. May Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Danny Chan
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - Keita Ito
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, Netherlands
- Department of Orthopedics, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Christine L. Le Maitre
- Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield, United Kingdom
| | - Marianna A. Tryfonidou
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
- *Correspondence: Marianna A. Tryfonidou,
| |
Collapse
|
9
|
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
|
10
|
Gan Y, He J, Zhu J, Xu Z, Wang Z, Yan J, Hu O, Bai Z, Chen L, Xie Y, Jin M, Huang S, Liu B, Liu P. Spatially defined single-cell transcriptional profiling characterizes diverse chondrocyte subtypes and nucleus pulposus progenitors in human intervertebral discs. Bone Res 2021; 9:37. [PMID: 34400611 PMCID: PMC8368097 DOI: 10.1038/s41413-021-00163-z] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 04/30/2021] [Accepted: 06/10/2021] [Indexed: 02/07/2023] Open
Abstract
A comprehensive understanding of the cellular heterogeneity and molecular mechanisms underlying the development, homeostasis, and disease of human intervertebral disks (IVDs) remains challenging. Here, the transcriptomic landscape of 108 108 IVD cells was mapped using single-cell RNA sequencing of three main compartments from young and adult healthy IVDs, including the nucleus pulposus (NP), annulus fibrosus, and cartilage endplate (CEP). The chondrocyte subclusters were classified based on their potential regulatory, homeostatic, and effector functions in extracellular matrix (ECM) homeostasis. Notably, in the NP, a PROCR+ resident progenitor population showed enriched colony-forming unit-fibroblast (CFU-F) activity and trilineage differentiation capacity. Finally, intercellular crosstalk based on signaling network analysis uncovered that the PDGF and TGF-β cascades are important cues in the NP microenvironment. In conclusion, a single-cell transcriptomic atlas that resolves spatially regulated cellular heterogeneity together with the critical signaling that underlies homeostasis will help to establish new therapeutic strategies for IVD degeneration in the clinic.
Collapse
Affiliation(s)
- Yibo Gan
- grid.410570.70000 0004 1760 6682Department of Spine Surgery, Center of Orthopedics, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China ,grid.410570.70000 0004 1760 6682State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University (Third Military Medical University), Chongqing, China
| | - Jian He
- grid.410740.60000 0004 1803 4911State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China
| | - Jun Zhu
- grid.410570.70000 0004 1760 6682Department of Spine Surgery, Center of Orthopedics, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Zhengyang Xu
- grid.410740.60000 0004 1803 4911State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China
| | - Zhong Wang
- grid.410570.70000 0004 1760 6682Department of Spine Surgery, Center of Orthopedics, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Jing Yan
- grid.410740.60000 0004 1803 4911State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China
| | - Ou Hu
- grid.410570.70000 0004 1760 6682Department of Spine Surgery, Center of Orthopedics, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Zhijie Bai
- grid.410740.60000 0004 1803 4911State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China
| | - Lin Chen
- grid.410570.70000 0004 1760 6682Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Laboratory for the Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yangli Xie
- grid.410570.70000 0004 1760 6682Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Laboratory for the Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Min Jin
- grid.410570.70000 0004 1760 6682Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Laboratory for the Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Shuo Huang
- grid.410570.70000 0004 1760 6682Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Laboratory for the Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Bing Liu
- grid.410740.60000 0004 1803 4911State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China ,grid.11135.370000 0001 2256 9319State Key Laboratory of Experimental Hematology, Institute of Hematology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China ,grid.258164.c0000 0004 1790 3548Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, China
| | - Peng Liu
- grid.410570.70000 0004 1760 6682Department of Spine Surgery, Center of Orthopedics, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China ,grid.410570.70000 0004 1760 6682State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University (Third Military Medical University), Chongqing, China
| |
Collapse
|
11
|
Gan Y, He J, Zhu J, Xu Z, Wang Z, Yan J, Hu O, Bai Z, Chen L, Xie Y, Jin M, Huang S, Liu B, Liu P. Spatially defined single-cell transcriptional profiling characterizes diverse chondrocyte subtypes and nucleus pulposus progenitors in human intervertebral discs. Bone Res 2021; 9:37. [PMID: 34400611 PMCID: PMC8368097 DOI: 10.1038/s41413-021-00163-z+10.1038/s41413-021-00163-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 04/30/2021] [Accepted: 06/10/2021] [Indexed: 01/21/2024] Open
Abstract
A comprehensive understanding of the cellular heterogeneity and molecular mechanisms underlying the development, homeostasis, and disease of human intervertebral disks (IVDs) remains challenging. Here, the transcriptomic landscape of 108 108 IVD cells was mapped using single-cell RNA sequencing of three main compartments from young and adult healthy IVDs, including the nucleus pulposus (NP), annulus fibrosus, and cartilage endplate (CEP). The chondrocyte subclusters were classified based on their potential regulatory, homeostatic, and effector functions in extracellular matrix (ECM) homeostasis. Notably, in the NP, a PROCR+ resident progenitor population showed enriched colony-forming unit-fibroblast (CFU-F) activity and trilineage differentiation capacity. Finally, intercellular crosstalk based on signaling network analysis uncovered that the PDGF and TGF-β cascades are important cues in the NP microenvironment. In conclusion, a single-cell transcriptomic atlas that resolves spatially regulated cellular heterogeneity together with the critical signaling that underlies homeostasis will help to establish new therapeutic strategies for IVD degeneration in the clinic.
Collapse
Affiliation(s)
- Yibo Gan
- Department of Spine Surgery, Center of Orthopedics, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University (Third Military Medical University), Chongqing, China
| | - Jian He
- State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China
| | - Jun Zhu
- Department of Spine Surgery, Center of Orthopedics, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Zhengyang Xu
- State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China
| | - Zhong Wang
- Department of Spine Surgery, Center of Orthopedics, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Jing Yan
- State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China
| | - Ou Hu
- Department of Spine Surgery, Center of Orthopedics, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Zhijie Bai
- State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China
| | - Lin Chen
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Laboratory for the Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yangli Xie
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Laboratory for the Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Min Jin
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Laboratory for the Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Shuo Huang
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Laboratory for the Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Bing Liu
- State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China.
- State Key Laboratory of Experimental Hematology, Institute of Hematology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China.
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, China.
| | - Peng Liu
- Department of Spine Surgery, Center of Orthopedics, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China.
- State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University (Third Military Medical University), Chongqing, China.
| |
Collapse
|
12
|
Spatially defined single-cell transcriptional profiling characterizes diverse chondrocyte subtypes and nucleus pulposus progenitors in human intervertebral discs. Bone Res 2021; 9:37. [PMID: 34400611 PMCID: PMC8368097 DOI: 10.1038/s41413-021-00163-z 10.1038/s41413-021-00163-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2023] Open
Abstract
A comprehensive understanding of the cellular heterogeneity and molecular mechanisms underlying the development, homeostasis, and disease of human intervertebral disks (IVDs) remains challenging. Here, the transcriptomic landscape of 108 108 IVD cells was mapped using single-cell RNA sequencing of three main compartments from young and adult healthy IVDs, including the nucleus pulposus (NP), annulus fibrosus, and cartilage endplate (CEP). The chondrocyte subclusters were classified based on their potential regulatory, homeostatic, and effector functions in extracellular matrix (ECM) homeostasis. Notably, in the NP, a PROCR+ resident progenitor population showed enriched colony-forming unit-fibroblast (CFU-F) activity and trilineage differentiation capacity. Finally, intercellular crosstalk based on signaling network analysis uncovered that the PDGF and TGF-β cascades are important cues in the NP microenvironment. In conclusion, a single-cell transcriptomic atlas that resolves spatially regulated cellular heterogeneity together with the critical signaling that underlies homeostasis will help to establish new therapeutic strategies for IVD degeneration in the clinic.
Collapse
|
13
|
Ismail SMY, Murray CM, Stevenson MA, Hung-Hsun Y, Davies HMS. Variation in the seventh lumbar vertebra and the lumbosacral junction morphometry associated with the sacrocaudal fusion in greyhounds. Anat Histol Embryol 2021; 50:668-677. [PMID: 33878214 DOI: 10.1111/ahe.12675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 04/01/2021] [Indexed: 11/28/2022]
Abstract
The lumbosacral joint is where the 7th lumbar vertebra (L.7) articulates within the sacrum. It is a clinically important area in the dog because of its relatively large range of motion. The current study aims to determine the possible differences in the length of the L.7 vertebra and the angle of the lumbosacral junction among greyhounds of standard and those of fused sacra, and to determine the potential association of sex, body mass and type of fused sacrum (standard and fused) on the morphology of the L.7 vertebra and the angle of the lumbosacral junction. Radiographs of 55 greyhound cadavers were used for radiographing; all radiographic images were stored and measured using X-ray acquisition software, and then analysed using descriptive statistics, multiple linear regression and logistic regression. The results of this study showed a significant increase (p < .008) in the length of the L.7 vertebra and the angle of the lumbosacral junction (p < .028) in greyhounds with fused sacra comparing with those of standard sacra, but the L.6 length was not significant (p = .431). Differences have been found in the length of L.7 vertebra and the angle of the lumbosacral junction in greyhounds. It was found that in greyhounds, any variation in the sacrum's anatomical features may alter the structure of the surrounding anatomical structures such as the L.7 vertebra and lumbosacral junction.
Collapse
Affiliation(s)
- Sa'ad M Y Ismail
- Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Parkville, Vic., Australia
| | - Christina M Murray
- Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Parkville, Vic., Australia
| | - Mark A Stevenson
- Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Parkville, Vic., Australia
| | - Yen Hung-Hsun
- Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Parkville, Vic., Australia
| | - Helen M S Davies
- Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Parkville, Vic., Australia
| |
Collapse
|
14
|
Baumgartner L, Wuertz-Kozak K, Le Maitre CL, Wignall F, Richardson SM, Hoyland J, Ruiz Wills C, González Ballester MA, Neidlin M, Alexopoulos LG, Noailly J. Multiscale Regulation of the Intervertebral Disc: Achievements in Experimental, In Silico, and Regenerative Research. Int J Mol Sci 2021; 22:E703. [PMID: 33445782 PMCID: PMC7828304 DOI: 10.3390/ijms22020703] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 12/22/2020] [Accepted: 12/24/2020] [Indexed: 12/17/2022] Open
Abstract
Intervertebral disc (IVD) degeneration is a major risk factor of low back pain. It is defined by a progressive loss of the IVD structure and functionality, leading to severe impairments with restricted treatment options due to the highly demanding mechanical exposure of the IVD. Degenerative changes in the IVD usually increase with age but at an accelerated rate in some individuals. To understand the initiation and progression of this disease, it is crucial to identify key top-down and bottom-up regulations' processes, across the cell, tissue, and organ levels, in health and disease. Owing to unremitting investigation of experimental research, the comprehension of detailed cell signaling pathways and their effect on matrix turnover significantly rose. Likewise, in silico research substantially contributed to a holistic understanding of spatiotemporal effects and complex, multifactorial interactions within the IVD. Together with important achievements in the research of biomaterials, manifold promising approaches for regenerative treatment options were presented over the last years. This review provides an integrative analysis of the current knowledge about (1) the multiscale function and regulation of the IVD in health and disease, (2) the possible regenerative strategies, and (3) the in silico models that shall eventually support the development of advanced therapies.
Collapse
Affiliation(s)
- Laura Baumgartner
- BCN MedTech, Department of Information and Communication Technologies, Universitat Pompeu Fabra, 08018 Barcelona, Spain; (L.B.); (C.R.W.); (M.A.G.B.)
| | - Karin Wuertz-Kozak
- Department of Biomedical Engineering, Rochester Institute of Technology (RIT), Rochester, NY 14623, USA;
- Schön Clinic Munich Harlaching, Spine Center, Academic Teaching Hospital and Spine Research Institute of the Paracelsus Medical University Salzburg (Austria), 81547 Munich, Germany
| | - Christine L. Le Maitre
- Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield S1 1WB, UK;
| | - Francis Wignall
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Sciences Centre, Oxford Road, Manchester M13 9PT, UK; (F.W.); (S.M.R.); (J.H.)
| | - Stephen M. Richardson
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Sciences Centre, Oxford Road, Manchester M13 9PT, UK; (F.W.); (S.M.R.); (J.H.)
| | - Judith Hoyland
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Sciences Centre, Oxford Road, Manchester M13 9PT, UK; (F.W.); (S.M.R.); (J.H.)
| | - Carlos Ruiz Wills
- BCN MedTech, Department of Information and Communication Technologies, Universitat Pompeu Fabra, 08018 Barcelona, Spain; (L.B.); (C.R.W.); (M.A.G.B.)
| | - Miguel A. González Ballester
- BCN MedTech, Department of Information and Communication Technologies, Universitat Pompeu Fabra, 08018 Barcelona, Spain; (L.B.); (C.R.W.); (M.A.G.B.)
- Catalan Institution for Research and Advanced Studies (ICREA), Pg. Lluis Companys 23, 08010 Barcelona, Spain
| | - Michael Neidlin
- Department of Mechanical Engineering, National Technical University of Athens, 15780 Athens, Greece; (M.N.); (L.G.A.)
| | - Leonidas G. Alexopoulos
- Department of Mechanical Engineering, National Technical University of Athens, 15780 Athens, Greece; (M.N.); (L.G.A.)
| | - Jérôme Noailly
- BCN MedTech, Department of Information and Communication Technologies, Universitat Pompeu Fabra, 08018 Barcelona, Spain; (L.B.); (C.R.W.); (M.A.G.B.)
| |
Collapse
|
15
|
Xu H, Sun M, Wang C, Xia K, Xiao S, Wang Y, Ying L, Yu C, Yang Q, He Y, Liu A, Chen L. Growth differentiation factor-5-gelatin methacryloyl injectable microspheres laden with adipose-derived stem cells for repair of disc degeneration. Biofabrication 2020; 13:015010. [PMID: 33361566 DOI: 10.1088/1758-5090/abc4d3] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Nucleus pulposus (NP) degeneration is the major cause of degenerative disc disease (DDD). This condition cannot be treated or attenuated by traditional open or minimally invasive surgical options. However, a combination of stem cells, growth factors (GFs) and biomaterials present a viable option for regeneration. Injectable biomaterials act as carriers for controlled release of GFs and deliver stem cells to target tissues through a minimally invasive approach. In this study, injectable gelatin methacryloyl microspheres (GMs) with controllable, uniform particle sizes were rapidly biosynthesized through a low-cost electrospraying method. The GMs were used as delivery vehicles for cells and GFs, and they exhibited good mechanical properties and biocompatibility and enhanced the in vitro differentiation of laden cells into NP-like phenotypes. Furthermore, this integrated system attenuated the in vivo degeneration of rat intervertebral discs, maintained NP tissue integrity and accelerated the synthesis of extracellular matrix. Therefore, this novel therapeutic system is a promising option for the treatment of DDD.
Collapse
Affiliation(s)
- Haibin Xu
- Department of Orthopedic Surgery, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou 310009, Zhejiang, People's Republic of China. Department of Orthopedic Research, Institute of Zhejiang University, Hangzhou 310009, Zhejiang, People's Republic of China. These two authors contributed equally to this work
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Palaeontological evidence reveals convergent evolution of intervertebral joint types in amniotes. Sci Rep 2020; 10:14106. [PMID: 32839497 PMCID: PMC7445751 DOI: 10.1038/s41598-020-70751-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 07/23/2020] [Indexed: 11/08/2022] Open
Abstract
The intervertebral disc (IVD) has long been considered unique to mammals. Palaeohistological sampling of 17 mostly extinct clades across the amniote tree revealed preservation of different intervertebral soft tissue types (cartilage, probable notochord) seen in extant reptiles. The distribution of the fossilised tissues allowed us to infer the soft part anatomy of the joint. Surprisingly, we also found evidence for an IVD in fossil reptiles, including non-avian dinosaurs, ichthyosaurs, plesiosaurs, and marine crocodiles. Based on the fossil dataset, we traced the evolution of the amniote intervertebral joint through ancestral character state reconstruction. The IVD evolved at least twice, in mammals and in extinct diapsid reptiles. From this reptilian IVD, extant reptile groups and some non-avian dinosaurs independently evolved a synovial ball-and-socket joint. The unique birds dorsal intervertebral joint evolved from this dinosaur joint. The tuatara and some geckos reverted to the ancestral persisting notochord.
Collapse
|
17
|
Wise CA, Sepich D, Ushiki A, Khanshour AM, Kidane YH, Makki N, Gurnett CA, Gray RS, Rios JJ, Ahituv N, Solnica-Krezel L. The cartilage matrisome in adolescent idiopathic scoliosis. Bone Res 2020; 8:13. [PMID: 32195011 PMCID: PMC7062733 DOI: 10.1038/s41413-020-0089-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 01/27/2020] [Indexed: 12/13/2022] Open
Abstract
The human spinal column is a dynamic, segmented, bony, and cartilaginous structure that protects the neurologic system and simultaneously provides balance and flexibility. Children with developmental disorders that affect the patterning or shape of the spine can be at risk of neurologic and other physiologic dysfunctions. The most common developmental disorder of the spine is scoliosis, a lateral deformity in the shape of the spinal column. Scoliosis may be part of the clinical spectrum that is observed in many developmental disorders, but typically presents as an isolated symptom in otherwise healthy adolescent children. Adolescent idiopathic scoliosis (AIS) has defied understanding in part due to its genetic complexity. Breakthroughs have come from recent genome-wide association studies (GWAS) and next generation sequencing (NGS) of human AIS cohorts, as well as investigations of animal models. These studies have identified genetic associations with determinants of cartilage biogenesis and development of the intervertebral disc (IVD). Current evidence suggests that a fraction of AIS cases may arise from variation in factors involved in the structural integrity and homeostasis of the cartilaginous extracellular matrix (ECM). Here, we review the development of the spine and spinal cartilages, the composition of the cartilage ECM, the so-called "matrisome" and its functions, and the players involved in the genetic architecture of AIS. We also propose a molecular model by which the cartilage matrisome of the IVD contributes to AIS susceptibility.
Collapse
Affiliation(s)
- Carol A. Wise
- Center for Pediatric Bone Biology and Translational Research, Texas Scottish Rite Hospital for Children, 2222 Welborn St., Dallas, TX 75219 USA
- McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX 75235 USA
- Departments of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75235 USA
- Orthopaedic Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75235 USA
| | - Diane Sepich
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110 USA
| | - Aki Ushiki
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA 94158 USA
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA 94158 USA
| | - Anas M. Khanshour
- Center for Pediatric Bone Biology and Translational Research, Texas Scottish Rite Hospital for Children, 2222 Welborn St., Dallas, TX 75219 USA
| | - Yared H. Kidane
- Center for Pediatric Bone Biology and Translational Research, Texas Scottish Rite Hospital for Children, 2222 Welborn St., Dallas, TX 75219 USA
| | - Nadja Makki
- Department of Anatomy and Cell Biology, University of Florida, College of Medicine, Gainesville, FL 32610 USA
| | - Christina A. Gurnett
- Departments of Neurology, Washington University School of Medicine, St Louis, MO 63110 USA
- Pediatrics, Washington University School of Medicine, St Louis, MO 63110 USA
- Orthopaedic Surgery, Washington University School of Medicine, St Louis, MO 63110 USA
| | - Ryan S. Gray
- Department of Pediatrics, Dell Pediatric Research Institute, University of Texas at Austin Dell Medical School, Austin, TX 78723 USA
| | - Jonathan J. Rios
- Center for Pediatric Bone Biology and Translational Research, Texas Scottish Rite Hospital for Children, 2222 Welborn St., Dallas, TX 75219 USA
- McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, TX 75235 USA
- Departments of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75235 USA
- Orthopaedic Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75235 USA
| | - Nadav Ahituv
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA 94158 USA
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA 94158 USA
| | - Lila Solnica-Krezel
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110 USA
| |
Collapse
|
18
|
Department of Histology and Embryology, Bogomolets National Medical University, Kyiv, Ukraine, Chaikovsky Y, Herashchenko S, Department of Histology, Cytology and Embryology, Ivano-Frankivsk National Medical University, Ivano-Frankivsk, Ukraine, Deltsova O, Department of Histology, Cytology and Embryology, Ivano-Frankivsk National Medical University, Ivano-Frankivsk, Ukraine. Problems and Perspectives of Using Stem Cells of Cartilage Tissues. PROBLEMS OF CRYOBIOLOGY AND CRYOMEDICINE 2019; 29:303-316. [DOI: 10.15407/cryo29.04.303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
19
|
Characterization of the microstructure of the intervertebral disc in patients with chronic low back pain by diffusion kurtosis imaging. 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 2019; 28:2517-2525. [PMID: 31410620 DOI: 10.1007/s00586-019-06095-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 06/19/2019] [Accepted: 08/04/2019] [Indexed: 10/26/2022]
Abstract
PURPOSE Multivariate analysis of T2-weighted signal, diffusion ADC, and DKI parameters and tractography were used to differentiate chronic non-specific low back pain (CLBP) patients and asymptomatic controls (AC). METHODS A total of 30 patients with CLBP and 23 AC underwent diffusion kurtosis imaging (DKI) of lumbar spine with a 3T MRI scanner to get the ADC values and seven parameters of DKI in the nucleus pulposus (NP) of the intervertebral disc. The tractography and the tract-related parameters as other parameters were also generated to indicate the intactness of annulus fibrosus (AF). T2-grades of the discs were also quantified based on an eight-grade degeneration grading system. ADC and T2-grades were compared with DKI parameters for the differentiation of CLBP and AC groups. RESULTS There was no difference in the T2 grades, ADC value, and multiple parameters in DKI of NP between CLBP and AC groups (P > 0.05). The average FA values in NP in AC group were found significantly higher than in the CLBP group (P < 0.05). The scores for the intactness of AF of the intervertebral discs were significantly different in CLBP and AC groups, with 90% of sensitivity and 70% specificity (P < 0.05). Additionally, there were significantly differences in the length and volume values of the AF in CLBP and AC groups (P < 0.05). CONCLUSION DKI is a good noninvasive method, and it might help to differentiate CLBP from AC. Particularly, the continuation of DKI tractography reflects the presence of annulus fibrosus fissures, an important character in the generation of the low back pain. These slides can be retrieved under Electronic Supplementary Material.
Collapse
|
20
|
Tessier S, Madhu V, Johnson ZI, Shapiro IM, Risbud MV. NFAT5/TonEBP controls early acquisition of notochord phenotypic markers, collagen composition, and sonic hedgehog signaling during mouse intervertebral disc embryogenesis. Dev Biol 2019; 455:369-381. [PMID: 31301300 DOI: 10.1016/j.ydbio.2019.07.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 06/12/2019] [Accepted: 07/09/2019] [Indexed: 12/21/2022]
Abstract
High osmolarity, bound water, and hydrostatic pressure contribute to notochord mechanics and its morphogenesis into the nucleus pulposus (NP) compartment of the intervertebral disc. Indeed, the osmoadaptive transcription factor, nuclear factor of activated T-cells 5 (NFAT5 aka TonEBP), is robustly expressed by resident cells of the notochord and NP. Nevertheless, the molecular mechanisms that drive notochord osmoregulation and the functions of NFAT5 in disc embryogenesis remain largely unexplored. In this study, we show that deletion of NFAT5 in mice results in delayed vertebral column development and a reduced NP aspect ratio in the caudal spine. This phenotype is associated with lower levels of the T-box transcription factor, Brachyury, delayed expression of notochord phenotypic markers, and decreased collagen II deposition in the perinotochordal sheath and condensing mesenchyme. In addition, NFAT5 mutants showed a stage-dependent dysregulation of sonic hedgehog (Shh) signaling with non-classical expression of Gli1. Generation of mice with notochord-specific deletion of IFT88 (ShhcreERT2;Ift88f/f) supported this mode of Gli1 regulation. Using isolated primary NP cells and bioinformatics approaches, we further show that Ptch1 and Smo expression is controlled by NFAT5 in a cell autonomous manner. Altogether, our results demonstrate that NFAT5 contributes to notochord and disc embryogenesis through its regulation of hallmark notochord phenotypic markers, extracellular matrix, and Shh signaling.
Collapse
Affiliation(s)
- Steven Tessier
- Graduate Program in Cell Biology and Regenerative Medicine, Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, PA, USA
| | - Vedavathi Madhu
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Zariel I Johnson
- Graduate Program in Cell Biology and Regenerative Medicine, Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, PA, USA
| | - Irving M Shapiro
- Graduate Program in Cell Biology and Regenerative Medicine, Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, PA, USA; Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Makarand V Risbud
- Graduate Program in Cell Biology and Regenerative Medicine, Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, PA, USA; Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
21
|
Zhao R, Liu W, Xia T, Yang L. Disordered Mechanical Stress and Tissue Engineering Therapies in Intervertebral Disc Degeneration. Polymers (Basel) 2019; 11:polym11071151. [PMID: 31284436 PMCID: PMC6680713 DOI: 10.3390/polym11071151] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 06/27/2019] [Accepted: 07/01/2019] [Indexed: 12/11/2022] Open
Abstract
Low back pain (LBP), commonly induced by intervertebral disc degeneration, is a lumbar disease with worldwide prevalence. However, the mechanism of degeneration remains unclear. The intervertebral disc is a nonvascular organ consisting of three components: Nucleus pulposus, annulus fibrosus, and endplate cartilages. The disc is structured to support our body motion and endure persistent external mechanical pressure. Thus, there is a close connection between force and intervertebral discs in LBP. It is well established that with aging, disordered mechanical stress profoundly influences the fate of nucleus pulposus and the alignment of collagen fibers in the annulus fibrosus. These support a new understanding that disordered mechanical stress plays an important role in the degeneration of the intervertebral discs. Tissue-engineered regenerative and reparative therapies are being developed for relieving disc degeneration and symptoms of lower back pain. In this paper, we will review the current literature available on the role of disordered mechanical stress in intervertebral disc degeneration, and evaluate the existing tissue engineering treatment strategies of the current therapies.
Collapse
Affiliation(s)
- Runze Zhao
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, Chongqing 400044, China
| | - Wanqian Liu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, Chongqing 400044, China
| | - Tingting Xia
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, Chongqing 400044, China.
| | - Li Yang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, Bioengineering College, Chongqing University, Chongqing 400044, China.
| |
Collapse
|
22
|
Clouet J, Fusellier M, Camus A, Le Visage C, Guicheux J. Intervertebral disc regeneration: From cell therapy to the development of novel bioinspired endogenous repair strategies. Adv Drug Deliv Rev 2019; 146:306-324. [PMID: 29705378 DOI: 10.1016/j.addr.2018.04.017] [Citation(s) in RCA: 144] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Revised: 03/29/2018] [Accepted: 04/24/2018] [Indexed: 12/15/2022]
Abstract
Low back pain (LBP), frequently associated with intervertebral disc (IVD) degeneration, is a major public health concern. LBP is currently managed by pharmacological treatments and, if unsuccessful, by invasive surgical procedures, which do not counteract the degenerative process. Considering that IVD cell depletion is critical in the degenerative process, the supplementation of IVD with reparative cells, associated or not with biomaterials, has been contemplated. Recently, the discovery of reparative stem/progenitor cells in the IVD has led to increased interest in the potential of endogenous repair strategies. Recruitment of these cells by specific signals might constitute an alternative strategy to cell transplantation. Here, we review the status of cell-based therapies for treating IVD degeneration and emphasize the current concept of endogenous repair as well as future perspectives. This review also highlights the challenges of the mobilization/differentiation of reparative progenitor cells through the delivery of biologics factors to stimulate IVD regeneration.
Collapse
Affiliation(s)
- Johann Clouet
- INSERM, UMR 1229, RMeS, Regenerative Medicine and Skeleton, Université de Nantes, ONIRIS, Nantes F-44042, France; CHU Nantes, Pharmacie Centrale, PHU 11, Nantes F-44093, France; Université de Nantes, UFR Sciences Biologiques et Pharmaceutiques, Nantes F-44035, France; Université de Nantes, UFR Odontologie, Nantes F-44042, France
| | - Marion Fusellier
- INSERM, UMR 1229, RMeS, Regenerative Medicine and Skeleton, Université de Nantes, ONIRIS, Nantes F-44042, France; Department of Diagnostic Imaging, CRIP, National Veterinary School (ONIRIS), Nantes F-44307, France
| | - Anne Camus
- INSERM, UMR 1229, RMeS, Regenerative Medicine and Skeleton, Université de Nantes, ONIRIS, Nantes F-44042, France; Université de Nantes, UFR Odontologie, Nantes F-44042, France
| | - Catherine Le Visage
- INSERM, UMR 1229, RMeS, Regenerative Medicine and Skeleton, Université de Nantes, ONIRIS, Nantes F-44042, France; Université de Nantes, UFR Odontologie, Nantes F-44042, France
| | - Jérôme Guicheux
- INSERM, UMR 1229, RMeS, Regenerative Medicine and Skeleton, Université de Nantes, ONIRIS, Nantes F-44042, France; Université de Nantes, UFR Odontologie, Nantes F-44042, France; CHU Nantes, PHU4 OTONN, Nantes, F-44093, France.
| |
Collapse
|
23
|
Zhu J, Xia K, Yu W, Wang Y, Hua J, Liu B, Gong Z, Wang J, Xu A, You Z, Chen Q, Li F, Tao H, Liang C. Sustained release of GDF5 from a designed coacervate attenuates disc degeneration in a rat model. Acta Biomater 2019; 86:300-311. [PMID: 30660009 DOI: 10.1016/j.actbio.2019.01.028] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Revised: 12/23/2018] [Accepted: 01/14/2019] [Indexed: 12/17/2022]
Abstract
Low back pain is often caused by intervertebral disc degeneration, which is characterized by nucleus pulposus (NP) and extracellular matrix (ECM) degeneration. Human adipose-derived stem cells (hADSCs) induced by growth and differentiation factor-5 (GDF5) can differentiate into an NP-like phenotype. Although stem cell-based therapy with prolonged exposure to growth factors is regarded as a promising treatment, the efficacy of this approach in attenuating the disc degeneration process is limited by the short lifespan of growth factors. In our study, a unique growth factor delivery vehicle composed of heparin and the synthetic polycation poly(ethylene argininylaspartate diglyceride) (PEAD) was used to sustain GDF5 release. The results showed that sustained release of GDF5 by the PEAD:heparin delivery system promoted hADSC differentiation to an NP-like phenotype in vitro. After injection of the PEAD:heparin:GDF5 delivery platform and hADSCs into intervertebral spaces of coccygeal (Co) vertebrae Co7/Co8 and Co8/Co9 of the rat, the disc height, water content, and structure of the NPs decreased more slowly than other treatment groups. This new strategy may be used as an alternative treatment for attenuating intervertebral disc degeneration with hADSCs without the need for gene therapy. STATEMENT OF SIGNIFICANCE: Low back pain is often caused by intervertebral disc degeneration, which is characterized by nucleus pulposus (NP) and extracellular matrix (ECM) degeneration. Human adipose-derived stem cells (hADSCs) induced by growth and differentiation factor-5 (GDF-5) can differentiate into an NP-like phenotype. Although stem cell-based therapy with prolonged exposure to growth factor is regarded as a promising treatment, the efficacy of this approach in the disc regeneration process is limited by the short life of growth factors. In our study, a unique growth factor delivery vehicle comprised of heparin and the synthetic polycation poly(ethylene argininylaspartate diglyceride) (PEAD) was used to sustain the release of GDF-5. Numerous groups have explored IDD regeneration methods in vitro and in vivo. Our study differs in that GDF5 was incorporated into a vehicle through charge attraction and exhibited a sustained release profile. Moreover, GDF-5 seeded coacervate combined with hADSC injection could be a minimally invasive approach for tissue engineering that is suitable for clinical application. We investigated the stimulatory effects of our GDF-5 seeded coacervate on the differentiation of ADSCs in vitro and the reparative effect of the delivery system on degenerated NP in vivo.
Collapse
|
24
|
Wang F, Zhang C, Sinkemani A, Shi R, Xie ZY, Chen L, Mao L, Wu XT. A histocytological and radiological overview of the natural history of intervertebral disk: from embryonic formation to age-related degeneration. 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 2019; 28:633-648. [PMID: 30715648 DOI: 10.1007/s00586-019-05903-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 01/05/2019] [Accepted: 01/25/2019] [Indexed: 12/24/2022]
Abstract
PURPOSE To elucidate the natural history of intervertebral disk (IVD) and characterize its embryonic beginnings and age-related degeneration. METHODS Coronal sections of embryonic (E13.5-neonatal) and postnatal (4-60-week-old) Sprague-Dawley rat IVD were stained by a series of histological stainings (hematoxylin and eosin, Alcian blue, Picrosirius red, Masson, Periodic acid-Schiff). Growth kinetics within embryonic IVD were evaluated by immunohistochemical staining of Ki67 and proliferating cell nuclear antigen. Postnatal maturation and degeneration of IVD were visualized on radiology by X-ray, CT, and MR imaging. RESULTS During the formation of rat IVD, inner annulus fibrosus (AF) and cartilaginous endplate (CEP) shared similar cell density, extracellular matrix, and potential of growth kinetics; notochord provided increased and enlarged cytoplasmic vacuoles to generate nucleus pulposus (NP), part of which was retained within CEP. Postnatally, vacuolated notochord cells were reduced by devacuolation, while chondrocytic NP cells increased; cartilaginous layers of CEP were narrowed by vertebrae growth and secondary ossification; fibrotic portion of AF decreased as cartilaginous matrix accumulated and infiltrated outward. In aged and degenerated IVD, large longitudinal fissures were detected near the boundaries between inner and outer AF, whereas both reduced cellularity and accumulated cell clusters were evident within the dehydrated NP; only part of these histocytological changes could be reported on radiology. CONCLUSIONS By showing that the natural history of IVD is orchestrated by a dynamic histocytological regulation, our study may facilitate better understanding of the developmental defects, cellular heterogeneity, age-related degenerative mechanisms, and biological regeneration of IVD. These slides can be retrieved under Electronic Supplementary Material.
Collapse
Affiliation(s)
- Feng Wang
- Department of Spine Surgery, Zhongda Hospital, School of Medicine, Southeast University, 87# Dingjiaqiao Road, Nanjing, 210009, China.,Surgery Research Center, School of Medicine, Southeast University, 87# Dingjiaqiao Road, Nanjing, 210009, China
| | - Cong Zhang
- Department of Spine Surgery, Zhongda Hospital, School of Medicine, Southeast University, 87# Dingjiaqiao Road, Nanjing, 210009, China.,Surgery Research Center, School of Medicine, Southeast University, 87# Dingjiaqiao Road, Nanjing, 210009, China
| | - Arjun Sinkemani
- Department of Spine Surgery, Zhongda Hospital, School of Medicine, Southeast University, 87# Dingjiaqiao Road, Nanjing, 210009, China.,Surgery Research Center, School of Medicine, Southeast University, 87# Dingjiaqiao Road, Nanjing, 210009, China
| | - Rui Shi
- Department of Spine Surgery, Zhongda Hospital, School of Medicine, Southeast University, 87# Dingjiaqiao Road, Nanjing, 210009, China.,Surgery Research Center, School of Medicine, Southeast University, 87# Dingjiaqiao Road, Nanjing, 210009, China
| | - Zhi-Yang Xie
- Department of Spine Surgery, Zhongda Hospital, School of Medicine, Southeast University, 87# Dingjiaqiao Road, Nanjing, 210009, China.,Surgery Research Center, School of Medicine, Southeast University, 87# Dingjiaqiao Road, Nanjing, 210009, China
| | - Lu Chen
- Department of Spine Surgery, Zhongda Hospital, School of Medicine, Southeast University, 87# Dingjiaqiao Road, Nanjing, 210009, China.,Surgery Research Center, School of Medicine, Southeast University, 87# Dingjiaqiao Road, Nanjing, 210009, China
| | - Lu Mao
- Department of Spine Surgery, Zhongda Hospital, School of Medicine, Southeast University, 87# Dingjiaqiao Road, Nanjing, 210009, China.,Surgery Research Center, School of Medicine, Southeast University, 87# Dingjiaqiao Road, Nanjing, 210009, China
| | - Xiao-Tao Wu
- Department of Spine Surgery, Zhongda Hospital, School of Medicine, Southeast University, 87# Dingjiaqiao Road, Nanjing, 210009, China. .,Surgery Research Center, School of Medicine, Southeast University, 87# Dingjiaqiao Road, Nanjing, 210009, China.
| |
Collapse
|
25
|
Levillain A, Rolfe R, Huang Y, Iatridis J, Nowlan N. Short-term foetal immobility temporally and progressively affects chick spinal curvature and anatomy and rib development. Eur Cell Mater 2019; 37:23-41. [PMID: 30644077 PMCID: PMC6505690 DOI: 10.22203/ecm.v037a03] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Congenital spine deformities may be influenced by movements in utero, but the effects of foetal immobility on spine and rib development remain unclear. The purpose of the present study was to determine (1) critical time-periods when rigid paralysis caused the most severe disruption in spine and rib development and (2) how the effects of an early, short-term immobilisation were propagated to the different features of spine and rib development. Chick embryos were immobilised once per single embryonic day (E) between E3 and E6 and harvested at E9. To assess the ontogenetic effects following single-day immobilisation, other embryos were immobilised at E4 and harvested daily between E5 and E9. Spinal curvature, vertebral shape and segmentation and rib development were analysed by optical projection tomography and histology. The results demonstrated that periods critical for movement varied for different aspects of spine and rib development. Single-day immobilisation at E3 or E4 resulted in the most pronounced spinal curvature abnormalities, multiple wedged vertebrae and segmentation defects, while single-day immobilisation at E5 led to the most severe rib abnormalities. Assessment of ontogenetic effects following single-day immobilisation at E4 revealed that vertebral segmentation defects were subsequent to earlier vertebral body shape and spinal curvature abnormalities, while rib formation (although delayed) was independent from thoracic vertebral shape or curvature changes. A day-long immobilisation in chicks severely affected spine and rib development, highlighting the importance of abnormal foetal movements at specific time-points and motivating targeted prenatal monitoring for early diagnosis of congenital scoliosis.
Collapse
Affiliation(s)
- A. Levillain
- Department of Bioengineering, Imperial College London, London, UK
| | - R.A. Rolfe
- Department of Bioengineering, Imperial College London, London, UK,Department of Zoology, Trinity College Dublin, Dublin, Ireland
| | - Y. Huang
- Department of Bioengineering, Imperial College London, London, UK
| | - J.C. Iatridis
- Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - N.C. Nowlan
- Department of Bioengineering, Imperial College London, London, UK,Address for correspondence: Niamh C. Nowlan, Department of Bioengineering, Imperial College London, London SW72AZ, UK. Telephone number: +44 2075945189
| |
Collapse
|
26
|
He Z, Jia M, Yu Y, Yuan C, Wang J. Roles of SDF-1/CXCR4 axis in cartilage endplate stem cells mediated promotion of nucleus pulposus cells proliferation. Biochem Biophys Res Commun 2018; 506:94-101. [PMID: 30340825 DOI: 10.1016/j.bbrc.2018.10.069] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 10/11/2018] [Indexed: 01/07/2023]
Abstract
Stem cells transplantation has shown considerable promise in intervertebral disc repair and low-back pain release. Cartilage endplate stem cells (CESCs) also showed potential for nucleus pulposus (NP) regeneration in a rabbit disc degeneration model, the precise mechanism was unclear. Here we investigated the effects of CESCs on NP cells (NPCs) proliferation and the mechanism in vitro. CESCs and NPCs were isolated from surgical specimens of degenerative human lumbar disc. NPCs were co-cultured with CESCs at a 1:1 ratio or cultured in CESCs conditioned medium (CESCs-CM). NPCs proliferation was evaluated by Ki-67 staining, CCK-8 assay and cell cycle analysis. Gene expressions were detected by qRT-PCR and activation of Akt and ERK1/2 was detected by western blot. CXCR4 antagonist AMD3100 was used to block SDF-1/CXCR4 axis. ERK1/2 and Akt inhibitors were used to block Akt and ERK1/2 activation. Results showed that NPCs proliferation was promoted by direct-contact co-culturing with CESCs as well as culturing in CESCs-CM. SDF-1 expression level in CESCs was significantly higher than that in NPCs, while CXCR4 was the opposite. Promotion of NPCs proliferation mediated by CESCs-CM could be partially attenuated by AMD3100. CESCs-CM activated both Akt and ERK1/2 in NPCs, while rhSDF-1 scarcely activated Akt but obviously activated ERK1/2. Akt and ERK1/2 inhibitors could partially inhibited CESCs-CM mediated promotion of NPCs proliferation and showed cumulative effect, while ERK1/2 inhibitor and AMD3100 could significantly abrogate SDF-1 mediated promotion of NPCs proliferation. Our results suggested that CESCs might promote NPCs proliferation in a paracrine pathway, which was partially mediated by SDF-1/CXCR4 axis via ERK1/2 signaling transduction pathway.
Collapse
Affiliation(s)
- Zhiliang He
- Department of Orthopaedics, Xinqiao Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Min Jia
- Department of Orthopaedics, Xinqiao Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Youjia Yu
- Department of Forensic Medicine, Nanjing Medical University, Nanjing, 211166, China
| | - Chao Yuan
- Department of Orthopaedics, Xinqiao Hospital, Third Military Medical University, Chongqing, 400038, China.
| | - Jian Wang
- Department of Orthopaedics, Xinqiao Hospital, Third Military Medical University, Chongqing, 400038, China.
| |
Collapse
|
27
|
Wang F, Zhang C, Shi R, Xie ZY, Chen L, Wang K, Wang YT, Xie XH, Wu XT. The embryonic and evolutionary boundaries between notochord and cartilage: a new look at nucleus pulposus-specific markers. Osteoarthritis Cartilage 2018; 26:1274-1282. [PMID: 29935307 DOI: 10.1016/j.joca.2018.05.022] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 05/16/2018] [Accepted: 05/23/2018] [Indexed: 02/02/2023]
Abstract
The adult nucleus pulposus (NP) and articular cartilage are similar in terms of their histocytological components and biomechanical functionalities, requiring a deep understanding of NP-specific markers to better evaluate stem-cell-based NP regeneration. Here, we seek to distinguish NP cells from articular chondrocytes (ACs), focusing on differences in their embryonic formation and evolutionary origin. Embryonically, NP cells are conservatively derived from the axial notochord, whereas ACs originate in a diversified manner from paraxial mesoderm and neural crest cells. Evolutionarily, although the origins of vertebrate NP and AC cells can be traced to similar structures within protostomia-like bilaterian ancestors, the distant phylogenetic relationship between the two groups of animals and the differences in the bodily origins of the tissues suggest that the tissues may in fact have undergone parallel evolution within the protostomia and deuterostomia. The numbers of supposedly NP-specific markers are increasing gradually as microarray studies proceed, but no final consensus has been attained on the specificity and physiology of "exclusive" NP markers because of innate variations among species; intrinsic expression of genes that destabilize the circadian clock; and cooperation by, and crosstalk among, different genes in terms of physiology-related phenotypes. We highlight the embryonic and evolutionary boundaries between NP and AC cells, to aid in recognition of the challenges associated with evaluation of the role played by nucleopulpogenic differentiation during stem-cell-based intervertebral disc regeneration.
Collapse
Affiliation(s)
- F Wang
- Department of Spine Surgery, Zhongda Hospital, School of Medicine, Southeast University, 87# Dingjiaqiao Road, 210009 Nanjing, China; Surgery Research Center, School of Medicine, Southeast University, 87# Dingjiaqiao Road, 210009 Nanjing, China.
| | - C Zhang
- Department of Spine Surgery, Zhongda Hospital, School of Medicine, Southeast University, 87# Dingjiaqiao Road, 210009 Nanjing, China; Surgery Research Center, School of Medicine, Southeast University, 87# Dingjiaqiao Road, 210009 Nanjing, China.
| | - R Shi
- Department of Spine Surgery, Zhongda Hospital, School of Medicine, Southeast University, 87# Dingjiaqiao Road, 210009 Nanjing, China; Surgery Research Center, School of Medicine, Southeast University, 87# Dingjiaqiao Road, 210009 Nanjing, China.
| | - Z-Y Xie
- Department of Spine Surgery, Zhongda Hospital, School of Medicine, Southeast University, 87# Dingjiaqiao Road, 210009 Nanjing, China; Surgery Research Center, School of Medicine, Southeast University, 87# Dingjiaqiao Road, 210009 Nanjing, China.
| | - L Chen
- Department of Spine Surgery, Zhongda Hospital, School of Medicine, Southeast University, 87# Dingjiaqiao Road, 210009 Nanjing, China; Surgery Research Center, School of Medicine, Southeast University, 87# Dingjiaqiao Road, 210009 Nanjing, China.
| | - K Wang
- Department of Spine Surgery, Zhongda Hospital, School of Medicine, Southeast University, 87# Dingjiaqiao Road, 210009 Nanjing, China; Surgery Research Center, School of Medicine, Southeast University, 87# Dingjiaqiao Road, 210009 Nanjing, China.
| | - Y-T Wang
- Department of Spine Surgery, Zhongda Hospital, School of Medicine, Southeast University, 87# Dingjiaqiao Road, 210009 Nanjing, China; Surgery Research Center, School of Medicine, Southeast University, 87# Dingjiaqiao Road, 210009 Nanjing, China.
| | - X-H Xie
- Department of Spine Surgery, Zhongda Hospital, School of Medicine, Southeast University, 87# Dingjiaqiao Road, 210009 Nanjing, China; Surgery Research Center, School of Medicine, Southeast University, 87# Dingjiaqiao Road, 210009 Nanjing, China.
| | - X-T Wu
- Department of Spine Surgery, Zhongda Hospital, School of Medicine, Southeast University, 87# Dingjiaqiao Road, 210009 Nanjing, China; Surgery Research Center, School of Medicine, Southeast University, 87# Dingjiaqiao Road, 210009 Nanjing, China.
| |
Collapse
|
28
|
Torre OM, Mroz V, Bartelstein MK, Huang AH, Iatridis JC. Annulus fibrosus cell phenotypes in homeostasis and injury: implications for regenerative strategies. Ann N Y Acad Sci 2018; 1442:61-78. [PMID: 30604562 DOI: 10.1111/nyas.13964] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 08/05/2018] [Accepted: 08/15/2018] [Indexed: 12/11/2022]
Abstract
Despite considerable efforts to develop cellular, molecular, and structural repair strategies and restore intervertebral disk function after injury, the basic biology underlying intervertebral disk healing remains poorly understood. Remarkably, little is known about the origins of cell populations residing within the annulus fibrosus, or their phenotypes, heterogeneity, and roles during healing. This review focuses on recent literature highlighting the intrinsic and extrinsic cell types of the annulus fibrosus in the context of the injury and healing environment. Spatial, morphological, functional, and transcriptional signatures of annulus fibrosus cells are reviewed, including inner and outer annulus fibrosus cells, which we propose to be referred to as annulocytes. The annulus also contains peripheral cells, interlamellar cells, and potential resident stem/progenitor cells, as well as macrophages, T lymphocytes, and mast cells following injury. Phases of annulus fibrosus healing include inflammation and recruitment of immune cells, cell proliferation, granulation tissue formation, and matrix remodeling. However, annulus fibrosus healing commonly involves limited remodeling, with granulation tissues remaining, and the development of chronic inflammatory states. Identifying annulus fibrosus cell phenotypes during health, injury, and degeneration will inform reparative regeneration strategies aimed at improving annulus fibrosus healing.
Collapse
Affiliation(s)
- Olivia M Torre
- Leni and Peter W. May Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Victoria Mroz
- Leni and Peter W. May Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Meredith K Bartelstein
- Leni and Peter W. May Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Alice H Huang
- Leni and Peter W. May Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, New York
| | - James C Iatridis
- Leni and Peter W. May Department of Orthopaedics, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
29
|
Séguin CA, Chan D, Dahia CL, Gazit Z. Latest advances in intervertebral disc development and progenitor cells. JOR Spine 2018; 1:e1030. [PMID: 30687811 PMCID: PMC6338208 DOI: 10.1002/jsp2.1030] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 07/23/2018] [Accepted: 07/26/2018] [Indexed: 12/12/2022] Open
Abstract
This paper is a concise review aiming to assemble the most relevant topics presented by the authors at ORS-Philadelphia Spine Research Society Fourth International Spine Research Symposium. It centers on the latest advances in disc development, its main structural entities, and the populating cells, with emphasis on the advances in pivotal molecular pathways responsible for forming the intervertebral discs (IVD). The objective of finding and emphasizing pathways and mechanisms that function to control tissue formation is to identify and to explore modifications occurring during normal aging, disease, and tissue repair. Thus, to comprehend that the cellular and molecular basis of tissue degeneration are crucial in the study of the dynamic interplay that includes cell-cell communication, gene regulation, and growth factors required to form a healthy and functional tissue during normal development.
Collapse
Affiliation(s)
- Cheryle A Séguin
- Schulich School of Medicine and Dentistry Bone and Joint Institute, The University of Western Ontario London ON Canada
| | - Danny Chan
- School of Biomedical Sciences LKS Faculty of Medicine, The University of Hong Kong Hong Kong China
| | - Chitra L Dahia
- Hospital for Special Surgery Weill Cornell Medical College New York New York
| | - Zulma Gazit
- Department of Surgery Regenerative Medicine Institute, Cedars-Sinai Medical Center Los Angeles California
| |
Collapse
|
30
|
Schubert AK, Smink JJ, Arp M, Ringe J, Hegewald AA, Sittinger M. Quality Assessment of Surgical Disc Samples Discriminates Human Annulus Fibrosus and Nucleus Pulposus on Tissue and Molecular Level. Int J Mol Sci 2018; 19:ijms19061761. [PMID: 29899321 PMCID: PMC6032144 DOI: 10.3390/ijms19061761] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 06/11/2018] [Accepted: 06/11/2018] [Indexed: 01/07/2023] Open
Abstract
A discrimination of the highly specialised annulus fibrosus (AF) and nucleus pulposus (NP) cells in the mature human intervertebral disc (IVD) is thus far still not possible in a reliable way. The aim of this study was to identify molecular markers that distinguish AF and NP cells in human disc tissue using microarray analysis as a screening tool. AF and NP samples were obtained from 28 cervical discs. First, all samples underwent quality sorting using two novel scoring systems for small-sized disc tissue samples including macroscopic, haptic and histological evaluation. Subsequently, samples with clear disc characteristics of either AF or NP that were free from impurities of foreign tissue (IVD score) and with low signs of disc degeneration on cellular level (DD score) were selected for GeneChip analysis (HGU1332P). The 11 AF and 9 NP samples showed distinctly different genome-wide transcriptomes. The majority of differentially expressed genes (DEGs) could be specifically assigned to the AF, whereas no DEG was exclusively expressed in the NP. Nevertheless, we identified 11 novel marker genes that clearly distinguished AF and NP, as confirmed by quantitative gene expression analysis. The novel established scoring systems and molecular markers showed the identity of AF and NP in disc starting material and are thus of great importance in the quality assurance of cell-based therapeutics in regenerative treatment of disc degeneration.
Collapse
Affiliation(s)
- Ann-Kathrin Schubert
- Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Tissue Engineering Laboratory and Berlin-Brandenburg Center for Regenerative Therapies, 13353 Berlin, Germany.
- CO.DON AG, 14513 Teltow, Germany.
| | | | - Mirko Arp
- Department of Neurosurgery, University Medical Center Mannheim, Heidelberg University, 68167 Mannheim, Germany.
| | - Jochen Ringe
- Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Tissue Engineering Laboratory and Berlin-Brandenburg Center for Regenerative Therapies, 13353 Berlin, Germany.
| | - Aldemar A Hegewald
- Department of Neurosurgery, University Medical Center Mannheim, Heidelberg University, 68167 Mannheim, Germany.
- Department of Neurosurgery and Spine Surgery, Helios Baltic Sea Hospital Damp, 24351 Damp, Germany.
| | - Michael Sittinger
- Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin and Berlin Institute of Health, Tissue Engineering Laboratory and Berlin-Brandenburg Center for Regenerative Therapies, 13353 Berlin, Germany.
| |
Collapse
|
31
|
Zhang Y, Xiong C, Kudelko M, Li Y, Wang C, Wong YL, Tam V, Rai MF, Cheverud J, Lawson HA, Sandell L, Chan WCW, Cheah KSE, Sham PC, Chan D. Early onset of disc degeneration in SM/J mice is associated with changes in ion transport systems and fibrotic events. Matrix Biol 2018; 70:123-139. [PMID: 29649547 DOI: 10.1016/j.matbio.2018.03.024] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 03/30/2018] [Accepted: 03/30/2018] [Indexed: 12/24/2022]
Abstract
Intervertebral disc degeneration (IDD) causes back pain and sciatica, affecting quality of life and resulting in high economic/social burden. The etiology of IDD is not well understood. Along with aging and environmental factors, genetic factors also influence the onset, progression and severity of IDD. Genetic studies of risk factors for IDD using human cohorts are limited by small sample size and low statistical power. Animal models amenable to genetic and functional studies of IDD provide desirable alternatives. Despite differences in size and cellular content as compared to human intervertebral discs (IVDs), the mouse is a powerful model for genetics and assessment of cellular changes relevant to human biology. Here, we provide evidence for early onset disc degeneration in SM/J relative to LG/J mice with poor and good tissue healing capacity respectively. In the first few months of life, LG/J mice maintain a relatively constant pool of notochordal-like cells in the nucleus pulposus (NP) of the IVD. In contrast, chondrogenic events are observed in SM/J mice beginning as early as one-week-old, with progressive fibrotic changes. Further, the extracellular matrix changes in the NP are consistent with IVD degeneration. Leveraging on the genomic data of two parental and two recombinant inbred lines, we assessed the genetic contribution to the NP changes and identified processes linked to the regulation of ion transport systems. Significantly, "transport" system is also in the top three gene ontology (GO) terms from a comparative proteomic analysis of the mouse NP. These findings support the potential of the SM/J, LG/J and their recombinant inbred lines for future genetic and biological analysis in mice and validation of candidate genes and biological relevance in human cohort studies. The proteomic data has been deposited to the ProteomeXchange Consortium via the PRIDE [1] partner repository with the dataset identifier PXD008784.
Collapse
Affiliation(s)
- Ying Zhang
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Chi Xiong
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Mateusz Kudelko
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Yan Li
- Centre for Genomic Sciences, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Cheng Wang
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Yuk Lun Wong
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Vivian Tam
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Muhammad Farooq Rai
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110, USA
| | - James Cheverud
- Department of Biology, Loyola University of Chicago, IL 60660, USA
| | - Heather A Lawson
- Department of Genetics, Washington University, St. Louis, MO 63110, USA
| | - Linda Sandell
- Department of Orthopaedic Surgery, Washington University, St. Louis, MO 63110, USA
| | - Wilson C W Chan
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong SAR, China; The University of Hong Kong - Shenzhen Institute of Research and Innovation (HKU-SIRI), Hi-Tech Industrial Park, Nanshan, Shenzhen, China
| | - Kathryn S E Cheah
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Pak C Sham
- Centre for Genomic Sciences, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Danny Chan
- School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong SAR, China; The University of Hong Kong - Shenzhen Institute of Research and Innovation (HKU-SIRI), Hi-Tech Industrial Park, Nanshan, Shenzhen, China.
| |
Collapse
|
32
|
Pimenta L, Marchi L, Oliveira L, Nogueira-Neto J, Coutinho E, Amaral R. Elastomeric Lumbar Total Disc Replacement: Clinical and Radiological Results With Minimum 84 Months Follow-Up. Int J Spine Surg 2018; 12:49-57. [PMID: 30280083 PMCID: PMC6162034 DOI: 10.14444/5009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Total lumbar disc replacement (TDR) devices have been designed to maintain motion, but both biomechanical and clinical data have indicated that a more controlled motion and additional load absorption in TDR would be beneficial. This work analyzed long-term results of an elastomeric disc (Physio-L) for degenerative lumbar conditions. MATERIAL AND METHODS This was a prospective, noncomparative, single-center clinical and radiological study. A total of 15 patients with predominant low back pain due degenerative disc disease received anterior total disc replacement with a Physio-L disc. Clinical outcomes were assessed both with a visual analog scale for pain and Oswestry Disability Index questionnaires. Radiological outcomes included implant failure, range of motion (ROM), facet degeneration, and adjacent level disease. Complication and reoperation rates were also recorded. The cases were assessed with a minimum follow-up of 84 months. RESULTS A total of 15 patients were enrolled (20 TDRs)-10 single-level cases (L5S1) and 5 two-level cases (L4L5/L5S1). After 84 months, clinical outcomes scores still demonstrated significant improvement compared with baseline (P < .001). Mean visual analog scale scores dropped from 7.1 to 2.9, and the Oswestry Disability Index improved from 50 to 16. No disc has experienced migration or breakage. The average range of motion value went from a baseline of 12.0° to 13.3° at 12 months, and at the final follow-up it decreased to 9.9°. Regarding the double-level cases, 3 of 5 (60%) had adverse events; just 1 single-level (10%) had adverse events. At final follow-up, radiological signs of facet degeneration were present in 7 of 15 patients (47%) but with only 1 of 15 (6.7%) symptomatic. Two patients (13%) required surgery at the adjacent level. At the 84-month follow-up, 16 of 18 prostheses (89%) were still active (2 revised to fusion and 2 were lost to follow-up). CONCLUSION The long-term follow-up data shows satisfactory clinical results for the use of Physio-L elastomeric TDR in the treatment of degenerative disc disease. Studies with bigger cohorts are needed to replicate results and add new information regarding other details.
Collapse
Affiliation(s)
- Luiz Pimenta
- Instituto de Patologia da Coluna, São Paulo, Brazil
- University of California San Diego, San Diego, California
| | - Luis Marchi
- Instituto de Patologia da Coluna, São Paulo, Brazil
| | | | | | | | | |
Collapse
|
33
|
Wang F, Gao ZX, Cai F, Sinkemani A, Xie ZY, Shi R, Wei JN, Wu XT. Formation, function, and exhaustion of notochordal cytoplasmic vacuoles within intervertebral disc: current understanding and speculation. Oncotarget 2017; 8:57800-57812. [PMID: 28915712 PMCID: PMC5593684 DOI: 10.18632/oncotarget.18101] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 05/01/2017] [Indexed: 01/08/2023] Open
Abstract
Notochord nucleus pulposus cells are characteristic of containing abundant and giant cytoplasmic vacuoles. This review explores the embryonic formation, biological function, and postnatal exhaustion of notochord vacuoles, aiming to characterize the signal network transforming the vacuolated nucleus pulposus cells into the vacuole-less chondrocytic cells. Embryonically, the cytoplasmic vacuoles within vertebrate notochord originate from an evolutionarily conserved vacuolation process during neurulation, which may continue to provide mechanical and signal support in constructing a mammalian intervertebral disc. For full vacuolation, a vacuolating specification from dorsal organizer cells, synchronized convergent extension, well-structured notochord sheath, and sufficient post-Golgi trafficking in notochord cells are required. Postnatally, age-related and species-specific exhaustion of vacuolated nucleus pulposus cells could be potentiated by Fas- and Fas ligand-induced apoptosis, intolerance to mechanical stress and nutrient deficiency, vacuole-mediated proliferation check, and gradual de-vacuolation within the avascular and compression-loaded intervertebral disc. These results suggest that the notochord vacuoles are active and versatile organelles for both embryonic notochord and postnatal nucleus pulposus, and may provide novel information on intervertebral disc degeneration to guide cell-based regeneration.
Collapse
Affiliation(s)
- Feng Wang
- Department of Spine Surgery, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu Province, China
- Surgery Research Center, School of Medicine, Southeast University, Nanjing, Jiangsu Province, China
| | - Zeng-Xin Gao
- Department of Spine Surgery, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu Province, China
- Surgery Research Center, School of Medicine, Southeast University, Nanjing, Jiangsu Province, China
| | - Feng Cai
- Department of Orthopedic Surgery, The First Affiliated Hospital of Soochow University, Soochow, Jiangsu Province, China
| | - Arjun Sinkemani
- Department of Spine Surgery, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu Province, China
- Surgery Research Center, School of Medicine, Southeast University, Nanjing, Jiangsu Province, China
| | - Zhi-Yang Xie
- Department of Spine Surgery, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu Province, China
- Surgery Research Center, School of Medicine, Southeast University, Nanjing, Jiangsu Province, China
| | - Rui Shi
- Department of Spine Surgery, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu Province, China
- Surgery Research Center, School of Medicine, Southeast University, Nanjing, Jiangsu Province, China
| | - Ji-Nan Wei
- Surgery Research Center, School of Medicine, Southeast University, Nanjing, Jiangsu Province, China
- Department of Orthopedics, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu Province, China
| | - Xiao-Tao Wu
- Department of Spine Surgery, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu Province, China
- Surgery Research Center, School of Medicine, Southeast University, Nanjing, Jiangsu Province, China
| |
Collapse
|
34
|
Chen S, Fu P, Wu H, Pei M. Meniscus, articular cartilage and nucleus pulposus: a comparative review of cartilage-like tissues in anatomy, development and function. Cell Tissue Res 2017; 370:53-70. [PMID: 28413859 DOI: 10.1007/s00441-017-2613-0] [Citation(s) in RCA: 126] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 03/17/2017] [Indexed: 01/07/2023]
Abstract
The degradation of cartilage in the human body is impacted by aging, disease, genetic predisposition and continued insults resulting from daily activity. The burden of cartilage defects (osteoarthritis, rheumatoid arthritis, intervertebral disc damage, knee replacement surgeries, etc.) is daunting in light of substantial economic and social stresses. This review strives to broaden the scope of regenerative medicine and tissue engineering approaches used for cartilage repair by comparing and contrasting the anatomical and functional nature of the meniscus, articular cartilage (AC) and nucleus pulposus (NP). Many review papers have provided detailed evaluations of these cartilages and cartilage-like tissues individually but none have comprehensively examined the parallels and inconsistencies in signaling, genetic expression and extracellular matrix composition between tissues. For the first time, this review outlines the importance of understanding these three tissues as unique entities, providing a comparative analysis of anatomy, ultrastructure, biochemistry and function for each tissue. This novel approach highlights the similarities and differences between tissues, progressing research toward an understanding of what defines each tissue as distinctive. The goal of this paper is to provide researchers with the fundamental knowledge to correctly engineer the meniscus, AC and NP without inadvertently developing the wrong tissue function or biochemistry.
Collapse
Affiliation(s)
- Song Chen
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics and Division of Exercise Physiology, West Virginia University, One Medical Center Drive, PO Box 9196, Morgantown, WV, 26506-9196, USA
- Department of Orthopaedics, Changzheng Hospital, Second Military Medical University, Shanghai, 200003, People's Republic of China
| | - Peiliang Fu
- Department of Orthopaedics, Changzheng Hospital, Second Military Medical University, Shanghai, 200003, People's Republic of China
| | - Haishan Wu
- Department of Orthopaedics, Changzheng Hospital, Second Military Medical University, Shanghai, 200003, People's Republic of China
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics and Division of Exercise Physiology, West Virginia University, One Medical Center Drive, PO Box 9196, Morgantown, WV, 26506-9196, USA.
| |
Collapse
|
35
|
Tavakoli J. Tissue Engineering of the Intervertebral Disc's Annulus Fibrosus: A Scaffold-Based Review Study. Tissue Eng Regen Med 2017; 14:81-91. [PMID: 30603465 PMCID: PMC6171584 DOI: 10.1007/s13770-017-0024-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 04/10/2016] [Accepted: 06/08/2016] [Indexed: 12/11/2022] Open
Abstract
Tissue engineering as a high technology solution for treating disc's problem has been the focus of some researches recently; however, the upcoming successful results in this area depends on understanding the complexities of biology and engineering interface. Whereas the major responsibility of the nucleus pulposus is to provide a sustainable hydrated environment within the disc, the function of the annulus fibrosus (AF) is more mechanical, facilitating joint mobility and preventing radial bulging by confining of the central part, which makes the AF reconstruction important. Although the body of knowledge regarding the AF tissue engineering has grown rapidly, the opportunities to improve current understanding of how artificial scaffolds are able to mimic the AF concentric structure-including inter-lamellar matrix and cross-bridges-addressed unresolved research questions. The aim of this literature review was to collect and discuss, from the international scientific literature, information about tissue engineering of the AF based on scaffold fabrication and material properties, useful for developing new strategies in disc tissue engineering. The key parameter of this research was understanding if role of cross-bridges and inter-lamellar matrix has been considered on tissue engineering of the AF.
Collapse
Affiliation(s)
- Javad Tavakoli
- Medical Device Research Institute, School of Computer Science, Engineering and Mathematics, Flinders University, Adelaide, SA 5042 Australia
| |
Collapse
|
36
|
Ghannam M, Jumah F, Mansour S, Samara A, Alkhdour S, Alzuabi MA, Aker L, Adeeb N, Massengale J, Oskouian RJ, Tubbs RS. Surgical anatomy, radiological features, and molecular biology of the lumbar intervertebral discs. Clin Anat 2017; 30:251-266. [PMID: 27997062 DOI: 10.1002/ca.22822] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 12/04/2016] [Indexed: 01/07/2023]
Abstract
The intervertebral disc (IVD) is a joint unique in structure and functions. Lying between adjacent vertebrae, it provides both the primary support and the elasticity required for the spine to move stably. Various aspects of the IVD have long been studied by researchers seeking a better understanding of its dynamics, aging, and subsequent disorders. In this article, we review the surgical anatomy, imaging modalities, and molecular biology of the lumbar IVD. Clin. Anat. 30:251-266, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Malik Ghannam
- An-Najah National University Hospital, Nablus, Palestine
| | - Fareed Jumah
- An-Najah National University Hospital, Nablus, Palestine
| | - Shaden Mansour
- An-Najah National University Hospital, Nablus, Palestine
| | - Amjad Samara
- An-Najah National University Hospital, Nablus, Palestine
| | - Saja Alkhdour
- An-Najah National University Hospital, Nablus, Palestine
| | | | - Loai Aker
- An-Najah National University Hospital, Nablus, Palestine
| | - Nimer Adeeb
- Department of Neurosurgery, Boston Medical Center, Boston University, Massachusetts
| | - Justin Massengale
- Department of Neurosurgery, Boston Medical Center, Boston University, Massachusetts
| | | | - R Shane Tubbs
- Department of Anatomical Sciences, St. George's University, Grenada.,Seattle Science Foundation, Seattle, Washington
| |
Collapse
|
37
|
Wang F, Cai F, Shi R, Wei JN, Wu XT. Hypoxia regulates sumoylation pathways in intervertebral disc cells: implications for hypoxic adaptations. Osteoarthritis Cartilage 2016; 24:1113-24. [PMID: 26826302 DOI: 10.1016/j.joca.2016.01.134] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 01/13/2016] [Accepted: 01/19/2016] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To explore the hypoxic regulation of sumoylation pathways and cell viability in nucleus pulposus (NP) and annulus fibrosus (AF) cells. DESIGN Expression of small ubiquitin-like modifier (SUMO) molecules, SUMO E1 activating enzymes SAE1 and SAE2, SUMO E2 conjugating enzyme UBC9, and de-sumoylation enzyme sentrin/SUMO-specific proteases (SENP)1 was immunolocalized in rat intervertebral disc (IVD) cells. NP and AF cells were cultured in hypoxia and cell viability was evaluated by quantifying cell proliferation, cellular senescence, apoptosis, and cell cycle distribution. Hypoxic regulation of sumoylation pathways was studied by analyzing the transcription and expression of SUMO molecules and sumoylation enzymes. Loss of function study using SENP1 siRNA was performed to investigate the regulatory role of sumoylation on the function of hypoxia inducible factor 1α (HIF-1α) and the hypoxic tolerance of IVD cells. RESULTS Sumoylation pathways were expressed in IVD cells and localized predominantly in nuclei. Both NP and AF cells maintained viability under hypoxia and upregulated the expression of SENP1. In NP cells hypoxia transiently increased the expression of SUMO-1, SUMO-2/3, SAE2, and UBC9, whereas SUMO-1 was elevated while SUMO-2/3, SAE1, SAE2, and UBC9 were reduced by low oxygen tensions in AF cells. Although downregulation of SENP1 decreased the transcriptional activity of HIF-1α, the viability of disc cells showed no significant loss under hypoxia. CONCLUSIONS NP and AF cells equally tolerate oxygen deficiency, but differently regulate the sumoylation pathways under hypoxia. The distinct sumoylation dynamics may help extend our understanding of the cell-specific regulation of the molecular basis that promotes cell survival in the hypoxic IVD.
Collapse
Affiliation(s)
- F Wang
- Department of Spine Surgery, Zhongda Hospital, School of Medicine, Southeast University, 87# Dingjiaqiao Road, 210009 Nanjing, China; Surgery Research Center, School of Medicine, Southeast University, 87# Dingjiaqiao Road, 210009 Nanjing, China.
| | - F Cai
- Department of Spine Surgery, Zhongda Hospital, School of Medicine, Southeast University, 87# Dingjiaqiao Road, 210009 Nanjing, China; Surgery Research Center, School of Medicine, Southeast University, 87# Dingjiaqiao Road, 210009 Nanjing, China.
| | - R Shi
- Department of Spine Surgery, Zhongda Hospital, School of Medicine, Southeast University, 87# Dingjiaqiao Road, 210009 Nanjing, China; Surgery Research Center, School of Medicine, Southeast University, 87# Dingjiaqiao Road, 210009 Nanjing, China.
| | - J-N Wei
- Surgery Research Center, School of Medicine, Southeast University, 87# Dingjiaqiao Road, 210009 Nanjing, China; Department of Orthopedics, Zhongda Hospital, School of Medicine, Southeast University, 87# Dingjiaqiao Road, 210009 Nanjing, China.
| | - X-T Wu
- Department of Spine Surgery, Zhongda Hospital, School of Medicine, Southeast University, 87# Dingjiaqiao Road, 210009 Nanjing, China; Surgery Research Center, School of Medicine, Southeast University, 87# Dingjiaqiao Road, 210009 Nanjing, China.
| |
Collapse
|
38
|
Decoding the intervertebral disc: Unravelling the complexities of cell phenotypes and pathways associated with degeneration and mechanotransduction. Semin Cell Dev Biol 2016; 62:94-103. [PMID: 27208724 DOI: 10.1016/j.semcdb.2016.05.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 05/17/2016] [Indexed: 12/20/2022]
Abstract
Back pain is the most common cause of pain and disability worldwide. While its etiology remains unknown, it is typically associated with intervertebral disc (IVD) degeneration. Despite the prevalence of back pain, relatively little is known about the specific cellular pathways and mechanisms that contribute to the development, function and degeneration of the IVD. Consequently, current treatments for back pain are largely limited to symptomatic interventions. However, major progress is being made in multiple research directions to unravel the biology and pathology of the IVD, raising hope that effective disease-modifying interventions will soon be developed. In this review, we will discuss our current knowledge and gaps in knowledge on the developmental origin of the IVD, the phenotype of the distinct cell types found within the IVD tissues, molecular targets in IVD degeneration identified using bioinformatics strategies, and mechanotransduction pathways that influence IVD cell fate and function.
Collapse
|
39
|
Komla-Ebri D, Dambroise E, Kramer I, Benoist-Lasselin C, Kaci N, Le Gall C, Martin L, Busca P, Barbault F, Graus-Porta D, Munnich A, Kneissel M, Di Rocco F, Biosse-Duplan M, Legeai-Mallet L. Tyrosine kinase inhibitor NVP-BGJ398 functionally improves FGFR3-related dwarfism in mouse model. J Clin Invest 2016; 126:1871-84. [PMID: 27064282 DOI: 10.1172/jci83926] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 02/25/2016] [Indexed: 01/08/2023] Open
Abstract
Achondroplasia (ACH) is the most frequent form of dwarfism and is caused by gain-of-function mutations in the fibroblast growth factor receptor 3-encoding (FGFR3-encoding) gene. Although potential therapeutic strategies for ACH, which aim to reduce excessive FGFR3 activation, have emerged over many years, the use of tyrosine kinase inhibitor (TKI) to counteract FGFR3 hyperactivity has yet to be evaluated. Here, we have reported that the pan-FGFR TKI, NVP-BGJ398, reduces FGFR3 phosphorylation and corrects the abnormal femoral growth plate and calvaria in organ cultures from embryos of the Fgfr3Y367C/+ mouse model of ACH. Moreover, we demonstrated that a low dose of NVP-BGJ398, injected subcutaneously, was able to penetrate into the growth plate of Fgfr3Y367C/+ mice and modify its organization. Improvements to the axial and appendicular skeletons were noticeable after 10 days of treatment and were more extensive after 15 days of treatment that started from postnatal day 1. Low-dose NVP-BGJ398 treatment reduced intervertebral disc defects of lumbar vertebrae, loss of synchondroses, and foramen-magnum shape anomalies. NVP-BGJ398 inhibited FGFR3 downstream signaling pathways, including MAPK, SOX9, STAT1, and PLCγ, in the growth plates of Fgfr3Y367C/+ mice and in cultured chondrocyte models of ACH. Together, our data demonstrate that NVP-BGJ398 corrects pathological hallmarks of ACH and support TKIs as a potential therapeutic approach for ACH.
Collapse
|
40
|
Wei A, Shen B, Williams LA, Bhargav D, Gulati T, Fang Z, Pathmanandavel S, Diwan AD. Expression of growth differentiation factor 6 in the human developing fetal spine retreats from vertebral ossifying regions and is restricted to cartilaginous tissues. J Orthop Res 2016; 34:279-89. [PMID: 26184900 DOI: 10.1002/jor.22983] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2015] [Accepted: 07/14/2015] [Indexed: 02/04/2023]
Abstract
During embryogenesis vertebral segmentation is initiated by sclerotomal cell migration and condensation around the notochord, forming anlagen of vertebral bodies and intervertebral discs. The factors that govern the segmentation are not clear. Previous research demonstrated that mutations in growth differentiation factor 6 resulted in congenital vertebral fusion, suggesting this factor plays a role in development of vertebral column. In this study, we detected expression and localization of growth differentiation factor 6 in human fetal spinal column, especially in the period of early ossification of vertebrae and the developing intervertebral discs. The extracellular matrix proteins were also examined. Results showed that high levels of growth differentiation factor 6 were expressed in the nucleus pulposus of intervertebral discs and the hypertrophic chondrocytes adjacent to the ossification centre in vertebral bodies, where strong expression of proteoglycan and collagens was also detected. As fetal age increased, the expression of growth differentiation factor 6 was decreased correspondingly with the progress of ossification in vertebral bodies and restricted to cartilaginous regions. This expression pattern and the genetic link to vertebral fusion suggest that growth differentiation factor 6 may play an important role in suppression of ossification to ensure proper vertebral segmentation during spinal development.
Collapse
Affiliation(s)
- Aiqun Wei
- Department of Orthopedic Research, Orthopedic Research Institute, St George Hospital, University of New South Wales, Sydney, Australia
| | - Bojiang Shen
- Department of Orthopedic Research, Orthopedic Research Institute, St George Hospital, University of New South Wales, Sydney, Australia
| | - Lisa A Williams
- Department of Orthopedic Research, Orthopedic Research Institute, St George Hospital, University of New South Wales, Sydney, Australia
| | - Divya Bhargav
- Department of Orthopedic Research, Orthopedic Research Institute, St George Hospital, University of New South Wales, Sydney, Australia
| | - Twishi Gulati
- Department of Orthopedic Research, Orthopedic Research Institute, St George Hospital, University of New South Wales, Sydney, Australia
| | - Zhimin Fang
- Human Molecular Genetics, St George Hospital, University of New South Wales, Sydney, Australia
| | - Sarennya Pathmanandavel
- Department of Orthopedic Research, Orthopedic Research Institute, St George Hospital, University of New South Wales, Sydney, Australia
| | - Ashish D Diwan
- Department of Orthopedic Research, Orthopedic Research Institute, St George Hospital, University of New South Wales, Sydney, Australia
| |
Collapse
|
41
|
Early development of the vertebral column. Semin Cell Dev Biol 2016; 49:83-91. [DOI: 10.1016/j.semcdb.2015.11.003] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 11/05/2015] [Indexed: 11/20/2022]
|
42
|
Serth K, Beckers A, Schuster-Gossler K, Pavlova MN, Müller J, Paul MC, Reinhardt R, Gossler A. Generation of an 870 kb deletion encompassing the Skt/Etl4 locus by combination of inter- and intra-chromosomal recombination. BMC Genet 2015; 16:143. [PMID: 26678520 PMCID: PMC4683868 DOI: 10.1186/s12863-015-0302-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 12/08/2015] [Indexed: 11/13/2022] Open
Abstract
Background Etl4lacZ (Enhancer trap locus 4) and SktGt (Sickle tail) are lacZ reporter gene integrations into the same locus on mouse chromosome 2 targeting a gene that is expressed in the notochord of early embryos and in multiple epithelia during later development. Both insertions caused recessive mutations that resulted exclusively in mild defects in the caudal vertebral column. Since notochord-derived signals are essential for formation of the vertebral column the phenotypes suggested that the lacZ insertions interfered with some notochord-dependent aspect of vertebral development. As both insertions occurred in introns it was unclear whether they represent hypomorphic alleles or abolish gene function. Here, we have generated a definitive null allele of the Skt/Etl4 gene and analysed homozygous mutants. Results We have introduced loxP sites into three positions of the gene based on additional upstream exons that we identified, and deleted approximately 870 kb of the locus by a combination of inter- and intra-chromosomal Cre-mediated recombinations in the female germ line of mice. This deletion removes about 90 % of the coding region and results in the loss of the SKT/ETL4 protein. Similar to the Etl4lacZ and SktGt alleles our deletion mutants are viable and fertile and show only mild defects in caudal vertebrae due to abnormal intervertebral disc development, although with higher penetrance. No other tissue with Skt/Etl4 expression that we analysed showed obvious defects. Conclusion The complete loss of Skt/Etl4 function affects only development of caudal notochord derivatives and is compensated for in its other expression domains. Electronic supplementary material The online version of this article (doi:10.1186/s12863-015-0302-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Katrin Serth
- Institut für Molekularbiologie OE5250, Medizinische Hochschule Hannover, Carl-Neuberg-Str.1, 30625, Hannover, Germany.
| | - Anja Beckers
- Institut für Molekularbiologie OE5250, Medizinische Hochschule Hannover, Carl-Neuberg-Str.1, 30625, Hannover, Germany.
| | - Karin Schuster-Gossler
- Institut für Molekularbiologie OE5250, Medizinische Hochschule Hannover, Carl-Neuberg-Str.1, 30625, Hannover, Germany.
| | - Maria N Pavlova
- Institut für Molekularbiologie OE5250, Medizinische Hochschule Hannover, Carl-Neuberg-Str.1, 30625, Hannover, Germany. .,Department of Anaesthesiology and Pain Medicine, University of Washington, Seattle, WA, 98001, USA.
| | - Julia Müller
- Institut für Molekularbiologie OE5250, Medizinische Hochschule Hannover, Carl-Neuberg-Str.1, 30625, Hannover, Germany. .,, Gasteiner Str. 31, 10717, Berlin, Germany.
| | - Mariel C Paul
- Institut für Molekularbiologie OE5250, Medizinische Hochschule Hannover, Carl-Neuberg-Str.1, 30625, Hannover, Germany. .,Department of Medicine I, Institute of Cancer Research, Medical University of Vienna, Borschkegasse 8a, 1090, Vienna, Austria.
| | - Richard Reinhardt
- Max Planck-Genome-Centre Cologne, Carl-von-Linné-Weg 10, D-50829, Köln, Germany.
| | - Achim Gossler
- Institut für Molekularbiologie OE5250, Medizinische Hochschule Hannover, Carl-Neuberg-Str.1, 30625, Hannover, Germany.
| |
Collapse
|
43
|
Wang F, Shi R, Cai F, Wang YT, Wu XT. Stem Cell Approaches to Intervertebral Disc Regeneration: Obstacles from the Disc Microenvironment. Stem Cells Dev 2015; 24:2479-95. [PMID: 26228642 DOI: 10.1089/scd.2015.0158] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Intervertebral disc (IVD) degeneration results in segmental instability and irritates neural compressive symptoms, such as low back pain and motor deficiency. The transplanting of stem cell into degenerative discs has attracted increasing clinical attention, as a new and proven approach to alleviating disc degeneration and to relieving discogenic pains. Aside from supplementation with stem cells, the IVD itself already contains a pool of stem and progenitor cells. Since the resident disc stem cells are incapable of reversing the pathologic changes that occur during aging and disc degeneration, it has been debated as to whether transplanted stem cells are capable of providing an efficient and durable therapeutic effect, even though there have been positive outcomes in both animal models and in clinical trials. This review aims to decipher the interactions between the stem cell and the disc microenvironment. Within their new niches in the IVD, the exogenous stem cell shows metabolic adaptation to the low-glucose supply, hypoxia, and compressive loadings, but demonstrates little tolerance to the disc-like acidity and hypertonicity. Similarly, the survival of endogenous stem cells is threatened as well by the harsh disc microenvironment, which may exhaust the stem cell resources and restrict the self-repair capacity of a degenerating IVD. To eliminate the intrinsic obstacles within the stressful disc niches, stem cells should be delivered with an injectable scaffold that provides both survival and mechanical support. Quick healing or concretion of the injection injuries, which minimizes stem cell leakage and disturbance to disc homeostasis, is of equal importance toward achieving efficient stem cell-based disc regeneration.
Collapse
Affiliation(s)
- Feng Wang
- 1 Department of Spine Surgery, Zhongda Hospital, Southeast University , Nanjing, China .,2 Surgery Research Center, Medical School of Southeast University , Nanjing, China
| | - Rui Shi
- 1 Department of Spine Surgery, Zhongda Hospital, Southeast University , Nanjing, China .,2 Surgery Research Center, Medical School of Southeast University , Nanjing, China
| | - Feng Cai
- 1 Department of Spine Surgery, Zhongda Hospital, Southeast University , Nanjing, China .,2 Surgery Research Center, Medical School of Southeast University , Nanjing, China
| | - Yun-Tao Wang
- 1 Department of Spine Surgery, Zhongda Hospital, Southeast University , Nanjing, China .,2 Surgery Research Center, Medical School of Southeast University , Nanjing, China
| | - Xiao-Tao Wu
- 1 Department of Spine Surgery, Zhongda Hospital, Southeast University , Nanjing, China .,2 Surgery Research Center, Medical School of Southeast University , Nanjing, China
| |
Collapse
|