1
|
Tian Z, Chen K, Shofer FS, Ciesielski B, Wang H, O'Brien WT, Qin L, Zhang Y. Gene Expression Changes Precede Elevated Mechanical Sensitivity in the Mouse Intervertebral Disc Injury Model. JOR Spine 2025; 8:e70049. [PMID: 39989623 PMCID: PMC11847628 DOI: 10.1002/jsp2.70049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 01/20/2025] [Accepted: 01/24/2025] [Indexed: 02/25/2025] Open
Abstract
Background Back pain after intervertebral disc (IVD) injury is a common clinical problem. Previous work examining early molecular changes post injury mainly used a candidate marker approach. Methods In this study, gene expression in the injured and intact mouse tail IVDs was determined with a nonbiased whole transcriptome approach and related to subsequent pain behavior. Mouse tail IVD injury was induced by a needle puncture. Whole murine transcriptome was determined by RNASeq. Transcriptomes of injured IVDs were compared with those of intact controls by bioinformatic methods. Mechanical allodynia was assessed by the Von Frey method. Results Among the 17,722 murine genes with meaningful expressions, 7242 genes were differentially expressed (P.adj ⟨ 0.01). Ontology study of upregulated genes revealed that leukocyte migration was the most enriched biological process, and network analysis showed that Tnfa had the most protein-protein interactions. The most enriched downregulated pathways were related to the pattern specification process. Mechanical allodynia persisted at the 4-week end point. Conclusion The RNASeq data revealed numerous early genes that participate in inflammation and repair processes post IVD injury. Mechanical allodynia followed these gene expression changes.
Collapse
Affiliation(s)
- Zuozhen Tian
- Department of Physical Medicine & RehabilitationPerelman School of Medicine, University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Ken Chen
- Department of OrthopedicsXiangya Hospital, Central South UniversityChangshaHunanPeople's Republic of China
| | - Frances S. Shofer
- Department of Emergency MedicinePerelman School of Medicine, University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Brianna Ciesielski
- Neurobehavior Testing Core, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Huan Wang
- Department of Orthopedic SurgeryPerelman School of Medicine, University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Department of Orthopedic SurgeryTongji Hospital, Huazhong University of Science and TechnologyWuhanPeople's Republic of China
| | - W. Timothy O'Brien
- Neurobehavior Testing Core, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Ling Qin
- Department of Orthopedic SurgeryPerelman School of Medicine, University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Yejia Zhang
- Department of Physical Medicine & RehabilitationPerelman School of Medicine, University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Section of Rehabilitation Medicine, Corporal Michael J. Crescenz Veterans Affairs Medical CenterPhiladelphiaPennsylvaniaUSA
| |
Collapse
|
2
|
Negrón-Piñeiro LJ, Wu Y, Mehta R, Maguire JE, Chou C, Lee J, Dahia CL, Di Gregorio A. Fine-Tuned Expression of Evolutionarily Conserved Signaling Molecules in the Ciona Notochord. Int J Mol Sci 2024; 25:13631. [PMID: 39769393 PMCID: PMC11728170 DOI: 10.3390/ijms252413631] [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: 10/23/2024] [Revised: 11/27/2024] [Accepted: 12/02/2024] [Indexed: 01/16/2025] Open
Abstract
The notochord is an axial structure required for the development of all chordate embryos, from sea squirts to humans. Over the course of more than half a billion years of chordate evolution, in addition to its structural function, the notochord has acquired increasingly relevant patterning roles for its surrounding tissues. This process has involved the co-option of signaling pathways and the acquisition of novel molecular mechanisms responsible for the precise timing and modalities of their deployment. To reconstruct this evolutionary route, we surveyed the expression of signaling molecules in the notochord of the tunicate Ciona, an experimentally amenable and informative chordate. We found that several genes encoding for candidate components of diverse signaling pathways are expressed during notochord development, and in some instances, display distinctive regionalized and/or lineage-specific patterns. We identified and deconstructed notochord enhancers associated with TGF-β and Ctgf, two evolutionarily conserved signaling genes that are expressed dishomogeneously in the Ciona notochord, and shed light on the cis-regulatory origins of their peculiar expression patterns.
Collapse
Affiliation(s)
- Lenny J. Negrón-Piñeiro
- Department of Molecular Pathobiology, New York University College of Dentistry, 345 E 24th Street, New York, NY 10010, USA
| | - Yushi Wu
- Department of Molecular Pathobiology, New York University College of Dentistry, 345 E 24th Street, New York, NY 10010, USA
| | - Ravij Mehta
- Orthopedic Soft Tissue Research Program, Hospital for Special Surgery, New York, NY 10021, USA
| | - Julie E. Maguire
- Department of Molecular Pathobiology, New York University College of Dentistry, 345 E 24th Street, New York, NY 10010, USA
| | - Cindy Chou
- Department of Molecular Pathobiology, New York University College of Dentistry, 345 E 24th Street, New York, NY 10010, USA
| | - Joyce Lee
- Department of Molecular Pathobiology, New York University College of Dentistry, 345 E 24th Street, New York, NY 10010, USA
| | - Chitra L. Dahia
- Orthopedic Soft Tissue Research Program, Hospital for Special Surgery, New York, NY 10021, USA
- Department of Cell and Developmental Biology, Weill Cornell Medicine, Graduate School of Medical Science, New York, NY 10065, USA
| | - Anna Di Gregorio
- Department of Molecular Pathobiology, New York University College of Dentistry, 345 E 24th Street, New York, NY 10010, USA
| |
Collapse
|
3
|
Ambrosio L, Schol J, Ruiz-Fernández C, Tamagawa S, Joyce K, Nomura A, de Rinaldis E, Sakai D, Papalia R, Vadalà G, Denaro V. Getting to the Core: Exploring the Embryonic Development from Notochord to Nucleus Pulposus. J Dev Biol 2024; 12:18. [PMID: 39051200 PMCID: PMC11270426 DOI: 10.3390/jdb12030018] [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: 05/20/2024] [Revised: 06/12/2024] [Accepted: 07/02/2024] [Indexed: 07/27/2024] Open
Abstract
The intervertebral disc (IVD) is the largest avascular organ of the human body and plays a fundamental role in providing the spine with its unique structural and biomechanical functions. The inner part of the IVD contains the nucleus pulposus (NP), a gel-like tissue characterized by a high content of type II collagen and proteoglycans, which is crucial for the disc's load-bearing and shock-absorbing properties. With aging and IVD degeneration (IDD), the NP gradually loses its physiological characteristics, leading to low back pain and additional sequelae. In contrast to surrounding spinal tissues, the NP presents a distinctive embryonic development since it directly derives from the notochord. This review aims to explore the embryology of the NP, emphasizing the pivotal roles of key transcription factors, which guide the differentiation and maintenance of the NP cellular components from the notochord and surrounding sclerotome. Through an understanding of NP development, we sought to investigate the implications of the critical developmental aspects in IVD-related pathologies, such as IDD and the rare malignant chordomas. Moreover, this review discusses the therapeutic strategies targeting these pathways, including the novel regenerative approaches leveraging insights from NP development and embryology to potentially guide future treatments.
Collapse
Affiliation(s)
- Luca Ambrosio
- Operative Research Unit of Orthopaedic and Trauma Surgery, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy; (L.A.); (R.P.); (V.D.)
- Research Unit of Orthopaedic and Trauma Surgery, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, 01128 Rome, Italy;
- Department of Orthopaedic Surgery, Tokai University School of Medicine, Isehara 259-1143, Japan; (J.S.); (C.R.-F.); (A.N.); (D.S.)
| | - Jordy Schol
- Department of Orthopaedic Surgery, Tokai University School of Medicine, Isehara 259-1143, Japan; (J.S.); (C.R.-F.); (A.N.); (D.S.)
| | - Clara Ruiz-Fernández
- Department of Orthopaedic Surgery, Tokai University School of Medicine, Isehara 259-1143, Japan; (J.S.); (C.R.-F.); (A.N.); (D.S.)
| | - Shota Tamagawa
- Department of Medicine for Orthopaedics and Motor Organ, Juntendo University Graduate School of Medicine, Tokyo 113-8421, Japan;
| | - Kieran Joyce
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, H91 W2TY Galway, Ireland;
- School of Medicine, University of Galway, H91 W2TY Galway, Ireland
| | - Akira Nomura
- Department of Orthopaedic Surgery, Tokai University School of Medicine, Isehara 259-1143, Japan; (J.S.); (C.R.-F.); (A.N.); (D.S.)
| | - Elisabetta de Rinaldis
- Research Unit of Orthopaedic and Trauma Surgery, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, 01128 Rome, Italy;
| | - Daisuke Sakai
- Department of Orthopaedic Surgery, Tokai University School of Medicine, Isehara 259-1143, Japan; (J.S.); (C.R.-F.); (A.N.); (D.S.)
| | - Rocco Papalia
- Operative Research Unit of Orthopaedic and Trauma Surgery, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy; (L.A.); (R.P.); (V.D.)
- Research Unit of Orthopaedic and Trauma Surgery, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, 01128 Rome, Italy;
| | - Gianluca Vadalà
- Operative Research Unit of Orthopaedic and Trauma Surgery, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy; (L.A.); (R.P.); (V.D.)
- Research Unit of Orthopaedic and Trauma Surgery, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, 01128 Rome, Italy;
| | - Vincenzo Denaro
- Operative Research Unit of Orthopaedic and Trauma Surgery, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy; (L.A.); (R.P.); (V.D.)
| |
Collapse
|
4
|
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
|
5
|
Warin J, Vedrenne N, Tam V, Zhu M, Yin D, Lin X, Guidoux-D’halluin B, Humeau A, Roseiro L, Paillat L, Chédeville C, Chariau C, Riemers F, Templin M, Guicheux J, Tryfonidou MA, Ho JW, David L, Chan D, Camus A. In vitro and in vivo models define a molecular signature reference for human embryonic notochordal cells. iScience 2024; 27:109018. [PMID: 38357665 PMCID: PMC10865399 DOI: 10.1016/j.isci.2024.109018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 11/13/2023] [Accepted: 01/22/2024] [Indexed: 02/16/2024] Open
Abstract
Understanding the emergence of human notochordal cells (NC) is essential for the development of regenerative approaches. We present a comprehensive investigation into the specification and generation of bona fide NC using a straightforward pluripotent stem cell (PSC)-based system benchmarked with human fetal notochord. By integrating in vitro and in vivo transcriptomic data at single-cell resolution, we establish an extended molecular signature and overcome the limitations associated with studying human notochordal lineage at early developmental stages. We show that TGF-β inhibition enhances the yield and homogeneity of notochordal lineage commitment in vitro. Furthermore, this study characterizes regulators of cell-fate decision and matrisome enriched in the notochordal niche. Importantly, we identify specific cell-surface markers opening avenues for differentiation refinement, NC purification, and functional studies. Altogether, this study provides a human notochord transcriptomic reference that will serve as a resource for notochord identification in human systems, diseased-tissues modeling, and facilitating future biomedical research.
Collapse
Affiliation(s)
- Julie Warin
- Nantes Université, Oniris, CHU Nantes, Inserm, Regenerative Medicine and Skeleton, RMeS, UMR 1229, 44000 Nantes, France
| | - Nicolas Vedrenne
- Nantes Université, Oniris, CHU Nantes, Inserm, Regenerative Medicine and Skeleton, RMeS, UMR 1229, 44000 Nantes, France
- Inserm, Univ. Limoges, Pharmacology & Transplantation, U1248, CHU Limoges, Service de Pharmacologie, toxicologie et pharmacovigilance, FHU SUPORT, 87000 Limoges, France
| | - Vivian Tam
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Mengxia Zhu
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Danqing Yin
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
- Laboratory of Data Discovery for Health Limited (D24H), Hong Kong Science Park, Hong Kong SAR, China
| | - Xinyi Lin
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
- Laboratory of Data Discovery for Health Limited (D24H), Hong Kong Science Park, Hong Kong SAR, China
| | - Bluwen Guidoux-D’halluin
- Nantes Université, Oniris, CHU Nantes, Inserm, Regenerative Medicine and Skeleton, RMeS, UMR 1229, 44000 Nantes, France
| | - Antoine Humeau
- Inserm, Univ. Limoges, Pharmacology & Transplantation, U1248, CHU Limoges, Service de Pharmacologie, toxicologie et pharmacovigilance, FHU SUPORT, 87000 Limoges, France
| | - Luce Roseiro
- Nantes Université, Oniris, CHU Nantes, Inserm, Regenerative Medicine and Skeleton, RMeS, UMR 1229, 44000 Nantes, France
| | - Lily Paillat
- Nantes Université, Oniris, CHU Nantes, Inserm, Regenerative Medicine and Skeleton, RMeS, UMR 1229, 44000 Nantes, France
| | - Claire Chédeville
- Nantes Université, Oniris, CHU Nantes, Inserm, Regenerative Medicine and Skeleton, RMeS, UMR 1229, 44000 Nantes, France
| | - Caroline Chariau
- Nantes Université, CHU Nantes, Inserm, CNRS, BioCore, 44000 Nantes, France
| | - Frank Riemers
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Markus Templin
- NMI Natural and Medical Sciences Institute, Markwiesenstraße 55, 72770 Reutlingen, Germany
| | - Jérôme Guicheux
- Nantes Université, Oniris, CHU Nantes, Inserm, Regenerative Medicine and Skeleton, RMeS, UMR 1229, 44000 Nantes, France
| | - Marianna A. Tryfonidou
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Joshua W.K. Ho
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
- Laboratory of Data Discovery for Health Limited (D24H), Hong Kong Science Park, Hong Kong SAR, China
| | - Laurent David
- Nantes Université, CHU Nantes, Inserm, CNRS, BioCore, 44000 Nantes, France
- Nantes Université, CHU Nantes, Inserm, CR2TI, 44000 Nantes, France
| | - Danny Chan
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Anne Camus
- Nantes Université, Oniris, CHU Nantes, Inserm, Regenerative Medicine and Skeleton, RMeS, UMR 1229, 44000 Nantes, France
| |
Collapse
|
6
|
Zhang L, Hu S, Xiu C, Li M, Zheng Y, Zhang R, Li B, Chen J. Intervertebral disc-intrinsic Hedgehog signaling maintains disc cell phenotypes and prevents disc degeneration through both cell autonomous and non-autonomous mechanisms. Cell Mol Life Sci 2024; 81:74. [PMID: 38308696 PMCID: PMC10838248 DOI: 10.1007/s00018-023-05106-x] [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: 11/13/2023] [Revised: 12/18/2023] [Accepted: 12/22/2023] [Indexed: 02/05/2024]
Abstract
Intervertebral disc degeneration is closely related to abnormal phenotypic changes in disc cells. However, the mechanism by which disc cell phenotypes are maintained remains poorly understood. Here, Hedgehog-responsive cells were found to be specifically localized in the inner annulus fibrosus and cartilaginous endplate of postnatal discs, likely activated by Indian Hedgehog. Global inhibition of Hedgehog signaling using a pharmacological inhibitor or Agc1-CreERT2-mediated deletion of Smo in disc cells of juvenile mice led to spontaneous degenerative changes in annulus fibrosus and cartilaginous endplate accompanied by aberrant disc cell differentiation in adult mice. In contrast, Krt19-CreER-mediated deletion of Smo specifically in nucleus pulposus cells led to healthy discs and normal disc cell phenotypes. Similarly, age-related degeneration of nucleus pulposus was accelerated by genetic inactivation of Hedgehog signaling in all disc cells, but not in nucleus pulposus cells. Furthermore, inactivation of Gli2 in disc cells resulted in partial loss of the vertebral growth plate but otherwise healthy discs, whereas deletion of Gli3 in disc cells largely corrected disc defects caused by Smo ablation in mice. Taken together, our findings not only revealed for the first time a direct role of Hedgehog-Gli3 signaling in maintaining homeostasis and cell phenotypes of annuls fibrosus and cartilaginous endplate, but also identified disc-intrinsic Hedgehog signaling as a novel non-cell-autonomous mechanism to regulate nucleus pulposus cell phenotype and protect mice from age-dependent nucleus pulposus degeneration. Thus, targeting Hedgehog signaling may represent a potential therapeutic strategy for the prevention and treatment of intervertebral disc degeneration.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Clinical Medicine, Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, 310015, Zhejiang, China
- Orthopedic Institute, Suzhou Medical College, Soochow University, Suzhou, 215006, Jiangsu, China
| | - Siyuan Hu
- Orthopedic Institute, Suzhou Medical College, Soochow University, Suzhou, 215006, Jiangsu, China
| | - Chunmei Xiu
- Department of Clinical Medicine, Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, 310015, Zhejiang, China
| | - Meng Li
- Orthopedic Institute, Suzhou Medical College, Soochow University, Suzhou, 215006, Jiangsu, China
| | - Yixin Zheng
- Orthopedic Institute, Suzhou Medical College, Soochow University, Suzhou, 215006, Jiangsu, China
| | - Rui Zhang
- Orthopedic Institute, Suzhou Medical College, Soochow University, Suzhou, 215006, Jiangsu, China
| | - Bin Li
- Department of Clinical Medicine, Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, 310015, Zhejiang, China.
- Orthopedic Institute, Suzhou Medical College, Soochow University, Suzhou, 215006, Jiangsu, China.
| | - Jianquan Chen
- Department of Clinical Medicine, Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, 310015, Zhejiang, China.
- Orthopedic Institute, Suzhou Medical College, Soochow University, Suzhou, 215006, Jiangsu, China.
| |
Collapse
|
7
|
Chen K, Tian Z, Wang H, Qin L, Enomoto-Iwamoto M, Zhang Y. Gene Expression Profiles Perturbed by Injury to the Mouse Intervertebral Disc. Am J Phys Med Rehabil 2024; 104:45-50. [PMID: 38984547 PMCID: PMC11647451 DOI: 10.1097/phm.0000000000002541] [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: 07/11/2024]
Abstract
OBJECTIVES Back pain subsequent to intervertebral disc injury is a common clinical problem. Previous work examining early molecular changes post injury mainly used a candidate marker approach. In this study, gene expression in the injured and intact mouse tail intervertebral discs was determined with a nonbiased whole transcriptome approach. DESIGN Mouse tail intervertebral disc injury was induced by a needle puncture. Whole murine transcriptome was determined by RNASeq. Transcriptomes of injured intervertebral discs were compared with those of intact controls by bioinformatic methods. RESULTS Among the 18,078 murine genes examined, 592 genes were differentially expressed ( P.adj < 0.01). Novel genes upregulated in injured compared with intact intervertebral discs included Chl1, Lum , etc. Ontology study of upregulated genes revealed that leukocyte migration was the most enriched biological process, and network analysis showed that Tnfa had the most protein-protein interactions. Novel downregulated genes in the injured intervertebral discs included 4833412C05Rik , Myoc , etc . The most enriched downregulated pathways were related to cytoskeletal organization. CONCLUSIONS Novel genes highly regulated after disc injury were identified with an unbiased approach; they may serve as biomarkers of injury and response to treatments in future experiments. Enriched biological pathways and molecules with high numbers of connections may be targets for treatments after injury.
Collapse
Affiliation(s)
- Ken Chen
- Department of Orthopedic Surgery, University of
Pennsylvania, Philadelphia, PA, 19146
- Department of Orthopedics, Xiangya Hospital, Central South
University, Changsha, Hunan, P.R. China 410008
| | - Zuozhen Tian
- Department of Physical Medicine & Rehabilitation,
University of Pennsylvania, Philadelphia, PA, 19146
| | - Huan Wang
- Department of Orthopedic Surgery, University of
Pennsylvania, Philadelphia, PA, 19146
- Department of Orthopedics, Xiangya Hospital, Central South
University, Changsha, Hunan, P.R. China 410008
| | - Ling Qin
- Department of Orthopedic Surgery, University of
Pennsylvania, Philadelphia, PA, 19146
| | - Motomi Enomoto-Iwamoto
- Department of Orthopedics, University of Maryland School of
Medicine, Baltimore, MD 21201
| | - Yejia Zhang
- Department of Physical Medicine & Rehabilitation,
University of Pennsylvania, Philadelphia, PA, 19146
- Section of Rehabilitation Medicine, Corporal Michael J.
Crescenz Veterans Affairs Medical Center, Philadelphia, PA, 10104
| |
Collapse
|
8
|
Zhang C, Zhong L, Lau YK, Wu M, Yao L, Schaer TP, Mauck RL, Malhotra NR, Qin L, Smith LJ. Single cell RNA sequencing reveals emergent notochord-derived cell subpopulations in the postnatal nucleus pulposus. FASEB J 2024; 38:e23363. [PMID: 38085183 PMCID: PMC10757564 DOI: 10.1096/fj.202301217r] [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: 06/20/2023] [Revised: 11/08/2023] [Accepted: 11/27/2023] [Indexed: 12/18/2023]
Abstract
Intervertebral disc degeneration is a leading cause of chronic low back pain. Cell-based strategies that seek to treat disc degeneration by regenerating the central nucleus pulposus (NP) hold significant promise, but key challenges remain. One of these is the inability of therapeutic cells to effectively mimic the performance of native NP cells, which are unique amongst skeletal cell types in that they arise from the embryonic notochord. In this study, we use single cell RNA sequencing to demonstrate emergent heterogeneity amongst notochord-derived NP cells in the postnatal mouse disc. Specifically, we established the existence of progenitor and mature NP cells, corresponding to notochordal and chondrocyte-like cells, respectively. Mature NP cells exhibited significantly higher expression levels of extracellular matrix (ECM) genes including aggrecan, and collagens II and VI, along with elevated transforming growth factor-beta and phosphoinositide 3 kinase-protein kinase B signaling. Additionally, we identified Cd9 as a novel surface marker of mature NP cells, and demonstrated that these cells were localized to the NP periphery, increased in numbers with increasing postnatal age, and co-localized with emerging glycosaminoglycan-rich matrix. Finally, we used a goat model to show that Cd9+ NP cell numbers decrease with moderate severity disc degeneration, suggesting that these cells are associated with maintenance of the healthy NP ECM. Improved understanding of the developmental mechanisms underlying regulation of ECM deposition in the postnatal NP may inform improved regenerative strategies for disc degeneration and associated low back pain.
Collapse
Affiliation(s)
- Chenghao Zhang
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, 3450 Hamilton Walk, Philadelphia, PA, 19104 USA
| | - Leilei Zhong
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, 3450 Hamilton Walk, Philadelphia, PA, 19104 USA
| | - Yian Khai Lau
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, 3450 Hamilton Walk, Philadelphia, PA, 19104 USA
| | - Meilun Wu
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, 3450 Hamilton Walk, Philadelphia, PA, 19104 USA
| | - Lutian Yao
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, 3450 Hamilton Walk, Philadelphia, PA, 19104 USA
| | - Thomas P. Schaer
- Department of Clinical Studies, New Bolton Center, School of Veterinary Medicine, University of Pennsylvania, 382 W Street Rd, Kennett Square, PA, USA 19348
| | - Robert L. Mauck
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, 3450 Hamilton Walk, Philadelphia, PA, 19104 USA
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Philadelphia VA Medical Center, 3900 Woodland Avenue, Philadelphia, PA, USA 19104
| | - Neil R. Malhotra
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, 3450 Hamilton Walk, Philadelphia, PA, 19104 USA
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104 USA
| | - Ling Qin
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, 3450 Hamilton Walk, Philadelphia, PA, 19104 USA
| | - Lachlan J. Smith
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, 3450 Hamilton Walk, Philadelphia, PA, 19104 USA
- Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Philadelphia VA Medical Center, 3900 Woodland Avenue, Philadelphia, PA, USA 19104
- Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA 19104 USA
| |
Collapse
|
9
|
Popsuj S, Di Gregorio A, Swalla BJ, Stolfi A. Loss of collagen gene expression in the notochord of the tailless tunicate Molgula occulta. Integr Comp Biol 2023; 63:990-998. [PMID: 37403333 PMCID: PMC10714901 DOI: 10.1093/icb/icad071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 05/22/2023] [Accepted: 06/11/2023] [Indexed: 07/06/2023] Open
Abstract
In tunicates, several species in the Molgulidae family have convergently lost the tailed, swimming larval body plan, including the morphogenesis of the notochord, a major chordate-defining trait. Through the comparison of tailless M. occulta and a close relative, the tailed species M. oculata, we show that notochord-specific expression of the Collagen Type I/II Alpha (Col1/2a) gene appears to have been lost specifically in the tailless species. Using CRISPR/Cas9-mediated mutagenesis in the tailed laboratory model tunicate Ciona robusta, we demonstrate that Col1/2a plays a crucial role in the convergent extension of notochord cells during tail elongation. Our results suggest that the expression of Col1/2a in the notochord, although necessary for its morphogenesis in tailed species, is dispensable for tailless species. This loss is likely a result of the accumulation of cis-regulatory mutations in the absence of purifying selective pressure. More importantly, the gene itself is not lost, likely due to its roles in other developmental processes, including during the adult stage. Our study further confirms the Molgulidae as an interesting family in which to study the evolutionary loss of tissue-specific expression of indispensable genes.
Collapse
Affiliation(s)
- Sydney Popsuj
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | - Anna Di Gregorio
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA
| | - Billie J Swalla
- Department of Biology, University of Washington, Seattle, WA 98195, USA
| | - Alberto Stolfi
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA 30332, USA
| |
Collapse
|
10
|
Schifferl D, Scholze-Wittler M, Villaronga Luque A, Pustet M, Wittler L, Veenvliet JV, Koch F, Herrmann BG. Genome-wide identification of notochord enhancers comprising the regulatory landscape of the brachyury locus in mouse. Development 2023; 150:dev202111. [PMID: 37882764 PMCID: PMC10651091 DOI: 10.1242/dev.202111] [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: 06/22/2023] [Accepted: 10/17/2023] [Indexed: 10/27/2023]
Abstract
The node and notochord are important signaling centers organizing the dorso-ventral patterning of cells arising from neuro-mesodermal progenitors forming the embryonic body anlage. Owing to the scarcity of notochord progenitors and notochord cells, a comprehensive identification of regulatory elements driving notochord-specific gene expression has been lacking. Here, we have used ATAC-seq analysis of FACS-purified notochord cells from Theiler stage 12-13 mouse embryos to identify 8921 putative notochord enhancers. In addition, we established a new model for generating notochord-like cells in culture, and found 3728 of these enhancers occupied by the essential notochord control factors brachyury (T) and/or Foxa2. We describe the regulatory landscape of the T locus, comprising ten putative enhancers occupied by these factors, and confirmed the regulatory activity of three of these elements. Moreover, we characterized seven new elements by knockout analysis in embryos and identified one new notochord enhancer, termed TNE2. TNE2 cooperates with TNE in the trunk notochord, and is essential for notochord differentiation in the tail. Our data reveal an essential role of Foxa2 in directing T-expressing cells towards the notochord lineage.
Collapse
Affiliation(s)
- Dennis Schifferl
- Max Planck Institute for Molecular Genetics, Department Developmental Genetics, Ihnestr. 63-73, 14195 Berlin, Germany
| | - Manuela Scholze-Wittler
- Max Planck Institute for Molecular Genetics, Department Developmental Genetics, Ihnestr. 63-73, 14195 Berlin, Germany
| | - Alba Villaronga Luque
- Max Planck Institute for Molecular Genetics, Department Developmental Genetics, Ihnestr. 63-73, 14195 Berlin, Germany
| | - Milena Pustet
- Max Planck Institute for Molecular Genetics, Department Developmental Genetics, Ihnestr. 63-73, 14195 Berlin, Germany
| | - Lars Wittler
- Max Planck Institute for Molecular Genetics, Department Developmental Genetics, Ihnestr. 63-73, 14195 Berlin, Germany
| | - Jesse V Veenvliet
- Max Planck Institute for Molecular Genetics, Department Developmental Genetics, Ihnestr. 63-73, 14195 Berlin, Germany
| | - Frederic Koch
- Max Planck Institute for Molecular Genetics, Department Developmental Genetics, Ihnestr. 63-73, 14195 Berlin, Germany
| | - Bernhard G Herrmann
- Max Planck Institute for Molecular Genetics, Department Developmental Genetics, Ihnestr. 63-73, 14195 Berlin, Germany
| |
Collapse
|
11
|
Raghavan R, Coppola U, Wu Y, Ihewulezi C, Negrón-Piñeiro LJ, Maguire JE, Hong J, Cunningham M, Kim HJ, Albert TJ, Ali AM, Saint-Jeannet JP, Ristoratore F, Dahia CL, Di Gregorio A. Gene expression in notochord and nuclei pulposi: a study of gene families across the chordate phylum. BMC Ecol Evol 2023; 23:63. [PMID: 37891482 PMCID: PMC10605842 DOI: 10.1186/s12862-023-02167-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 08/08/2023] [Indexed: 10/29/2023] Open
Abstract
The transition from notochord to vertebral column is a crucial milestone in chordate evolution and in prenatal development of all vertebrates. As ossification of the vertebral bodies proceeds, involutions of residual notochord cells into the intervertebral discs form the nuclei pulposi, shock-absorbing structures that confer flexibility to the spine. Numerous studies have outlined the developmental and evolutionary relationship between notochord and nuclei pulposi. However, the knowledge of the similarities and differences in the genetic repertoires of these two structures remains limited, also because comparative studies of notochord and nuclei pulposi across chordates are complicated by the gene/genome duplication events that led to extant vertebrates. Here we show the results of a pilot study aimed at bridging the information on these two structures. We have followed in different vertebrates the evolutionary trajectory of notochord genes identified in the invertebrate chordate Ciona, and we have evaluated the extent of conservation of their expression in notochord cells. Our results have uncovered evolutionarily conserved markers of both notochord development and aging/degeneration of the nuclei pulposi.
Collapse
Affiliation(s)
- Rahul Raghavan
- Hospital for Special Surgery, Orthopedic Soft Tissue Research Program, New York, NY, 10021, USA
| | - Ugo Coppola
- Stazione Zoologica 'A. Dohrn', Villa Comunale 1, 80121, Naples, Italy
- Present Address: Molecular Cardiovascular Biology Division and Heart Institute, Cincinnati Children's Research Foundation, Cincinnati, OH, 45229, USA
| | - Yushi Wu
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY, 10010, USA
| | - Chibuike Ihewulezi
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY, 10010, USA
| | - Lenny J Negrón-Piñeiro
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY, 10010, USA
| | - Julie E Maguire
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY, 10010, USA
| | - Justin Hong
- Hospital for Special Surgery, Orthopedic Soft Tissue Research Program, New York, NY, 10021, USA
| | - Matthew Cunningham
- Hospital for Special Surgery, New York, NY, 10021, USA
- Weill Cornell Medical College, New York, NY, 10065, USA
| | - Han Jo Kim
- Hospital for Special Surgery, New York, NY, 10021, USA
- Weill Cornell Medical College, New York, NY, 10065, USA
| | - Todd J Albert
- Hospital for Special Surgery, New York, NY, 10021, USA
- Weill Cornell Medical College, New York, NY, 10065, USA
| | - Abdullah M Ali
- Department of Medicine, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Jean-Pierre Saint-Jeannet
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY, 10010, USA
| | | | - Chitra L Dahia
- Hospital for Special Surgery, Orthopedic Soft Tissue Research Program, New York, NY, 10021, USA.
- Department of Cell and Developmental Biology, Weill Cornell Medicine, Graduate School of Medical Science, New York, NY, 10065, USA.
| | - Anna Di Gregorio
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY, 10010, USA.
| |
Collapse
|
12
|
Kemmler CL, Smolikova J, Moran HR, Mannion BJ, Knapp D, Lim F, Czarkwiani A, Hermosilla Aguayo V, Rapp V, Fitch OE, Bötschi S, Selleri L, Farley E, Braasch I, Yun M, Visel A, Osterwalder M, Mosimann C, Kozmik Z, Burger A. Conserved enhancers control notochord expression of vertebrate Brachyury. Nat Commun 2023; 14:6594. [PMID: 37852970 PMCID: PMC10584899 DOI: 10.1038/s41467-023-42151-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 09/29/2023] [Indexed: 10/20/2023] Open
Abstract
The cell type-specific expression of key transcription factors is central to development and disease. Brachyury/T/TBXT is a major transcription factor for gastrulation, tailbud patterning, and notochord formation; however, how its expression is controlled in the mammalian notochord has remained elusive. Here, we identify the complement of notochord-specific enhancers in the mammalian Brachyury/T/TBXT gene. Using transgenic assays in zebrafish, axolotl, and mouse, we discover three conserved Brachyury-controlling notochord enhancers, T3, C, and I, in human, mouse, and marsupial genomes. Acting as Brachyury-responsive, auto-regulatory shadow enhancers, in cis deletion of all three enhancers in mouse abolishes Brachyury/T/Tbxt expression selectively in the notochord, causing specific trunk and neural tube defects without gastrulation or tailbud defects. The three Brachyury-driving notochord enhancers are conserved beyond mammals in the brachyury/tbxtb loci of fishes, dating their origin to the last common ancestor of jawed vertebrates. Our data define the vertebrate enhancers for Brachyury/T/TBXTB notochord expression through an auto-regulatory mechanism that conveys robustness and adaptability as ancient basis for axis development.
Collapse
Affiliation(s)
- Cassie L Kemmler
- Section of Developmental Biology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jana Smolikova
- Institute of Molecular Genetics of the ASCR, v. v. i., Prague, Czech Republic
| | - Hannah R Moran
- Section of Developmental Biology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Brandon J Mannion
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- Comparative Biochemistry Program, University of California, Berkeley, CA, 94720, USA
| | - Dunja Knapp
- Technische Universität Dresden, CRTD Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Fabian Lim
- Department of Medicine, Health Sciences, University of California San Diego, La Jolla, CA, USA
- Department of Molecular Biology, Biological Sciences, University of California San Diego, La Jolla, CA, USA
- Biological Sciences Graduate Program, University of California San Diego, La Jolla, CA, USA
| | - Anna Czarkwiani
- Technische Universität Dresden, CRTD Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Viviana Hermosilla Aguayo
- Program in Craniofacial Biology, University of California San Francisco, San Francisco, CA, USA
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA
- Department of Orofacial Sciences, University of California San Francisco, San Francisco, CA, USA
- Department of Anatomy, University of California San Francisco, San Francisco, CA, USA
| | - Vincent Rapp
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Olivia E Fitch
- Department of Integrative Biology and Ecology, Evolution and Behavior Program, Michigan State University, East Lansing, MI, USA
| | - Seraina Bötschi
- Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Licia Selleri
- Program in Craniofacial Biology, University of California San Francisco, San Francisco, CA, USA
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA
- Department of Orofacial Sciences, University of California San Francisco, San Francisco, CA, USA
- Department of Anatomy, University of California San Francisco, San Francisco, CA, USA
| | - Emma Farley
- Department of Medicine, Health Sciences, University of California San Diego, La Jolla, CA, USA
- Department of Molecular Biology, Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | - Ingo Braasch
- Department of Integrative Biology and Ecology, Evolution and Behavior Program, Michigan State University, East Lansing, MI, USA
| | - Maximina Yun
- Technische Universität Dresden, CRTD Center for Regenerative Therapies Dresden, Dresden, Germany
- Max Planck Institute for Molecular Cell Biology and Genetics, Dresden, Germany
- Cluster of Excellence Physics of Life, Technische Universität Dresden, Dresden, Germany
| | - Axel Visel
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- US Department of Energy Joint Genome Institute, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- School of Natural Sciences, University of California Merced, Merced, CA, USA
| | - Marco Osterwalder
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Department of Cardiology, Bern University Hospital, Bern, Switzerland
| | - Christian Mosimann
- Section of Developmental Biology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Zbynek Kozmik
- Institute of Molecular Genetics of the ASCR, v. v. i., Prague, Czech Republic.
| | - Alexa Burger
- Section of Developmental Biology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
13
|
Zhang C, Zhong L, Lau YK, Wu M, Yao L, Schaer TP, Mauck RL, Malhotra NR, Qin L, Smith LJ. Single Cell RNA Sequencing Reveals Emergent Notochord-Derived Cell Subpopulations in the Postnatal Nucleus Pulposus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.21.541589. [PMID: 37292597 PMCID: PMC10245831 DOI: 10.1101/2023.05.21.541589] [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
Intervertebral disc degeneration is a leading cause of chronic low back pain. Cell-based strategies that seek to treat disc degeneration by regenerating the central nucleus pulposus hold significant promise, but key challenges remain. One of these is the inability of therapeutic cells to effectively mimic the performance of native nucleus pulposus cells, which are unique amongst skeletal cell types in that they arise from the embryonic notochord. In this study we use single cell RNA sequencing to demonstrate emergent heterogeneity amongst notochord-derived nucleus pulposus cells in the postnatal mouse disc. Specifically, we established the existence of early and late stage nucleus pulposus cells, corresponding to notochordal progenitor and mature cells, respectively. Late stage cells exhibited significantly higher expression levels of extracellular matrix genes including aggrecan, and collagens II and VI, along with elevated TGF-β and PI3K-Akt signaling. Additionally, we identified Cd9 as a novel surface marker of late stage nucleus pulposus cells, and demonstrated that these cells were localized to the nucleus pulposus periphery, increased in numbers with increasing postnatal age, and co-localized with emerging glycosaminoglycan-rich matrix. Finally, we used a goat model to show the Cd9+ nucleus pulposus cell numbers decrease with moderate severity disc degeneration, suggesting that these cells are associated with maintenance of the healthy nucleus pulposus extracellular matrix. Improved understanding of the developmental mechanisms underlying regulation of ECM deposition in the postnatal NP may inform improved regenerative strategies for disc degeneration and associated low back pain.
Collapse
|
14
|
Johnston SN, Silagi ES, Madhu V, Nguyen DH, Shapiro IM, Risbud MV. GLUT1 is redundant in hypoxic and glycolytic nucleus pulposus cells of the intervertebral disc. JCI Insight 2023; 8:e164883. [PMID: 36917198 PMCID: PMC10243741 DOI: 10.1172/jci.insight.164883] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 03/08/2023] [Indexed: 03/15/2023] Open
Abstract
Glycolysis is central to homeostasis of nucleus pulposus (NP) cells in the avascular intervertebral disc. Since the glucose transporter, GLUT1, is a highly enriched phenotypic marker of NP cells, we hypothesized that it is vital for the development and postnatal maintenance of the disc. Surprisingly, primary NP cells treated with 2 well-characterized GLUT1 inhibitors maintained normal rates of glycolysis and ATP production, indicating intrinsic compensatory mechanisms. We showed in vitro that NP cells mitigated GLUT1 loss by rewiring glucose import through GLUT3. Of note, we demonstrated that substrates, such as glutamine and palmitate, did not compensate for glucose restriction resulting from dual inhibition of GLUT1/3, and inhibition compromised long-term cell viability. To investigate the redundancy of GLUT1 function in NP, we generated 2 NP-specific knockout mice: Krt19CreERT Glut1fl/fl and Foxa2Cre Glut1fl/fl. There were no apparent defects in postnatal disc health or development and maturation in mutant mice. Microarray analysis verified that GLUT1 loss did not cause transcriptomic alterations in the NP, supporting that cells are refractory to GLUT1 loss. These observations provide the first evidence to our knowledge of functional redundancy in GLUT transporters in the physiologically hypoxic intervertebral disc and underscore the importance of glucose as the indispensable substrate for NP cells.
Collapse
Affiliation(s)
- Shira N. Johnston
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, and
- Graduate Program in Cell Biology and Regenerative Medicine, College of Life Sciences, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Elizabeth S. Silagi
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, and
- Graduate Program in Cell Biology and Regenerative Medicine, College of Life Sciences, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Vedavathi Madhu
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, and
| | - Duc H. Nguyen
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, and
- Graduate Program in Cell Biology and Regenerative Medicine, College of Life Sciences, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Irving M. Shapiro
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, and
- Graduate Program in Cell Biology and Regenerative Medicine, College of Life Sciences, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Makarand V. Risbud
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, and
- Graduate Program in Cell Biology and Regenerative Medicine, College of Life Sciences, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
15
|
Gómez AE, Addish S, Alvarado K, Boatemaa P, Onyali AC, Ramirez EG, Rojas MF, Rai J, Reynolds KA, Tang WJ, Kwon RY. Multiple Mechanisms Explain Genetic Effects at the CPED1-WNT16 Bone Mineral Density Locus. Curr Osteoporos Rep 2023; 21:173-183. [PMID: 36943599 PMCID: PMC10202127 DOI: 10.1007/s11914-023-00783-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/07/2023] [Indexed: 03/23/2023]
Abstract
PURPOSE OF REVIEW Chromosome region 7q31.31, also known as the CPED1-WNT16 locus, is robustly associated with BMD and fracture risk. The aim of the review is to highlight experimental studies examining the function of genes at the CPED1-WNT16 locus. RECENT FINDINGS Genes that reside at the CPED1-WNT16 locus include WNT16, FAM3C, ING3, CPED1, and TSPAN12. Experimental studies in mice strongly support the notion that Wnt16 is necessary for bone mass and strength. In addition, roles for Fam3c and Ing3 in regulating bone morphology in vivo and/or osteoblast differentiation in vitro have been identified. Finally, a role for wnt16 in dually influencing bone and muscle morphogenesis in zebrafish has recently been discovered, which has brought forth new questions related to whether the influence of WNT16 in muscle may conspire with its influence in bone to alter BMD and fracture risk.
Collapse
Affiliation(s)
- Arianna Ericka Gómez
- Department of Orthopaedics and Sports Medicine, University of Washington School of Medicine, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Sumaya Addish
- Department of Orthopaedics and Sports Medicine, University of Washington School of Medicine, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Kurtis Alvarado
- Department of Orthopaedics and Sports Medicine, University of Washington School of Medicine, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Priscilla Boatemaa
- Department of Orthopaedics and Sports Medicine, University of Washington School of Medicine, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Anne C Onyali
- Department of Orthopaedics and Sports Medicine, University of Washington School of Medicine, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Emily G Ramirez
- Department of Orthopaedics and Sports Medicine, University of Washington School of Medicine, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Maria F Rojas
- Department of Orthopaedics and Sports Medicine, University of Washington School of Medicine, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Jyoti Rai
- Department of Orthopaedics and Sports Medicine, University of Washington School of Medicine, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Kiana A Reynolds
- Department of Orthopaedics and Sports Medicine, University of Washington School of Medicine, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - W Joyce Tang
- Department of Orthopaedics and Sports Medicine, University of Washington School of Medicine, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Ronald Young Kwon
- Department of Orthopaedics and Sports Medicine, University of Washington School of Medicine, Seattle, WA, USA.
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA.
| |
Collapse
|
16
|
Lu Z, Chen P, Xu Q, Li B, Jiang S, Jiang L, Zheng X. Constitutive and conditional gene knockout mice for the study of intervertebral disc degeneration: Current status, decision considerations, and future possibilities. JOR Spine 2023; 6:e1242. [PMID: 36994464 PMCID: PMC10041386 DOI: 10.1002/jsp2.1242] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 12/13/2022] [Accepted: 12/22/2022] [Indexed: 01/09/2023] Open
Abstract
There have been an increasing number of patients with degenerative disc diseases due to the aging population. In light of this, studies on the pathogenesis of intervertebral disc degeneration have become a hot topic, and gene knockout mice have become a valuable tool in this field of research. With the development of science and technology, constitutive gene knockout mice can be constructed using homologous recombination, zinc finger nuclease, transcription activator-like effector nuclease technology and clustered regularly interspaced short palindromic repeats/Cas9 (CRISPR/Cas9) system, and conditional gene knockout mice can be constructed using the Cre/LoxP system. The gene-edited mice using these techniques have been widely used in the studies on disc degeneration. This paper reviews the development process and principles of these technologies, functions of the edited genes in disc degeneration, advantages, and disadvantages of different methods and possible targets of the specific Cre recombinase in intervertebral discs. Recommendations for the choice of suitable gene-edited model mice are presented. At the same time, possible technological improvements in the future are also discussed.
Collapse
Affiliation(s)
- Ze‐Yu Lu
- Spine CenterXinhua Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Peng‐Bo Chen
- Spine CenterXinhua Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Qing‐Yin Xu
- Spine CenterXinhua Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Bo Li
- Spine CenterXinhua Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Sheng‐Dan Jiang
- Spine CenterXinhua Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Lei‐Sheng Jiang
- Spine CenterXinhua Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xin‐Feng Zheng
- Spine CenterXinhua Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
17
|
Johnston SN, Madhu V, Shapiro IM, Risbud MV. Conditional Deletion of HIF-2α in Mouse Nucleus Pulposus Reduces Fibrosis and Provides Mild and Transient Protection From Age-Dependent Structural Changes in Intervertebral Disc. J Bone Miner Res 2022; 37:2512-2530. [PMID: 36117450 PMCID: PMC9772060 DOI: 10.1002/jbmr.4707] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 07/05/2022] [Accepted: 07/29/2022] [Indexed: 01/19/2023]
Abstract
Hypoxia-inducible factors (HIFs) are critical to the development and homeostasis of hypoxic tissues. Although HIF-2α, one of the main HIF-α isoforms, is expressed in nucleus pulposus (NP) cells, its functions remain unknown. We deleted HIF-2α in the NP tissue using a notochord-specific FoxA2Cre allele to study HIF-2α function in the adult intervertebral disc. Unlike observations in HIF-1αcKO mice, fate mapping studies using Rosa26-mTmG reporter showed that HIF-2α loss in NP did not negatively impact cell survival or affect compartment development. Rather, loss of HIF-2α resulted in slightly better attributes of NP morphology in 14-month-old HIF-2αcKO mice as evident from lower scores of degeneration. These 14-month-old HIF-2αcKO mice also exhibited significant reduction in NP tissue fibrosis and lower collagen turnover in the annulus fibrosis (AF) compartment. Imaging-Fourier transform-infrared (FTIR) analyses showed decreased collagen and protein content in the NP and maintained chondroitin sulfate levels in 14-month-old HIF-2αcKO . Mechanistically, global transcriptomic analysis showed enrichment of differentially expressed genes with Gene Ontology (GO) terms related to metabolic processes and cell development, molecular functions concerned with histone and protein binding, and associated pathways, including oxidative stress. Noteworthy, these morphological differences were not apparent in 24-month-old HIF-2αcKO , indicating that aging is the dominant factor in governing disc health. Together these data suggest that loss of HIF-2α in the NP compartment is not detrimental to the intervertebral disc development but rather mitigates NP tissue fibrosis and offers mild but transient protection from age-dependent early degenerative changes. © 2022 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Shira N. Johnston
- Graduate Program in Cell Biology and Regenerative Medicine, Thomas Jefferson University, Philadelphia, PA USA
- Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, PA USA
| | - Vedavathi Madhu
- Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, PA USA
| | - Irving M. Shapiro
- Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, PA USA
| | - Makarand V. Risbud
- Graduate Program in Cell Biology and Regenerative Medicine, Thomas Jefferson University, Philadelphia, PA USA
- Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, PA USA
| |
Collapse
|
18
|
Watson CJ, Tang WJ, Rojas MF, Fiedler IAK, Morfin Montes de Oca E, Cronrath AR, Callies LK, Swearer AA, Ahmed AR, Sethuraman V, Addish S, Farr GH, Gómez AE, Rai J, Monstad-Rios AT, Gardiner EM, Karasik D, Maves L, Busse B, Hsu YH, Kwon RY. wnt16 regulates spine and muscle morphogenesis through parallel signals from notochord and dermomyotome. PLoS Genet 2022; 18:e1010496. [PMID: 36346812 PMCID: PMC9674140 DOI: 10.1371/journal.pgen.1010496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 11/18/2022] [Accepted: 10/24/2022] [Indexed: 11/09/2022] Open
Abstract
Bone and muscle are coupled through developmental, mechanical, paracrine, and autocrine signals. Genetic variants at the CPED1-WNT16 locus are dually associated with bone- and muscle-related traits. While Wnt16 is necessary for bone mass and strength, this fails to explain pleiotropy at this locus. Here, we show wnt16 is required for spine and muscle morphogenesis in zebrafish. In embryos, wnt16 is expressed in dermomyotome and developing notochord, and contributes to larval myotome morphology and notochord elongation. Later, wnt16 is expressed at the ventral midline of the notochord sheath, and contributes to spine mineralization and osteoblast recruitment. Morphological changes in wnt16 mutant larvae are mirrored in adults, indicating that wnt16 impacts bone and muscle morphology throughout the lifespan. Finally, we show that wnt16 is a gene of major effect on lean mass at the CPED1-WNT16 locus. Our findings indicate that Wnt16 is secreted in structures adjacent to developing bone (notochord) and muscle (dermomyotome) where it affects the morphogenesis of each tissue, thereby rendering wnt16 expression into dual effects on bone and muscle morphology. This work expands our understanding of wnt16 in musculoskeletal development and supports the potential for variants to act through WNT16 to influence bone and muscle via parallel morphogenetic processes.
Collapse
Affiliation(s)
- Claire J. Watson
- Department of Orthopaedics and Sports Medicine, University of Washington School of Medicine, Seattle, Washington, United States of America
- Insitute for Stem Cell and Regenerative Medicines, University of Washington, Seattle Washington, United States of America
| | - W. Joyce Tang
- Department of Orthopaedics and Sports Medicine, University of Washington School of Medicine, Seattle, Washington, United States of America
- Insitute for Stem Cell and Regenerative Medicines, University of Washington, Seattle Washington, United States of America
| | - Maria F. Rojas
- Department of Orthopaedics and Sports Medicine, University of Washington School of Medicine, Seattle, Washington, United States of America
- Insitute for Stem Cell and Regenerative Medicines, University of Washington, Seattle Washington, United States of America
| | - Imke A. K. Fiedler
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ernesto Morfin Montes de Oca
- Department of Orthopaedics and Sports Medicine, University of Washington School of Medicine, Seattle, Washington, United States of America
- Insitute for Stem Cell and Regenerative Medicines, University of Washington, Seattle Washington, United States of America
| | - Andrea R. Cronrath
- Department of Orthopaedics and Sports Medicine, University of Washington School of Medicine, Seattle, Washington, United States of America
- Insitute for Stem Cell and Regenerative Medicines, University of Washington, Seattle Washington, United States of America
| | - Lulu K. Callies
- Department of Orthopaedics and Sports Medicine, University of Washington School of Medicine, Seattle, Washington, United States of America
- Insitute for Stem Cell and Regenerative Medicines, University of Washington, Seattle Washington, United States of America
| | - Avery Angell Swearer
- Department of Orthopaedics and Sports Medicine, University of Washington School of Medicine, Seattle, Washington, United States of America
- Insitute for Stem Cell and Regenerative Medicines, University of Washington, Seattle Washington, United States of America
| | - Ali R. Ahmed
- Department of Orthopaedics and Sports Medicine, University of Washington School of Medicine, Seattle, Washington, United States of America
- Insitute for Stem Cell and Regenerative Medicines, University of Washington, Seattle Washington, United States of America
| | - Visali Sethuraman
- Department of Orthopaedics and Sports Medicine, University of Washington School of Medicine, Seattle, Washington, United States of America
- Insitute for Stem Cell and Regenerative Medicines, University of Washington, Seattle Washington, United States of America
| | - Sumaya Addish
- Department of Orthopaedics and Sports Medicine, University of Washington School of Medicine, Seattle, Washington, United States of America
- Insitute for Stem Cell and Regenerative Medicines, University of Washington, Seattle Washington, United States of America
| | - Gist H. Farr
- Center for Developmental Biology and Regenerative Medicine, Seattle Children’s Research Institute, Seattle, Washington, United States of America
| | - Arianna Ericka Gómez
- Department of Orthopaedics and Sports Medicine, University of Washington School of Medicine, Seattle, Washington, United States of America
- Insitute for Stem Cell and Regenerative Medicines, University of Washington, Seattle Washington, United States of America
| | - Jyoti Rai
- Department of Orthopaedics and Sports Medicine, University of Washington School of Medicine, Seattle, Washington, United States of America
- Insitute for Stem Cell and Regenerative Medicines, University of Washington, Seattle Washington, United States of America
| | - Adrian T. Monstad-Rios
- Department of Orthopaedics and Sports Medicine, University of Washington School of Medicine, Seattle, Washington, United States of America
- Insitute for Stem Cell and Regenerative Medicines, University of Washington, Seattle Washington, United States of America
| | - Edith M. Gardiner
- Department of Orthopaedics and Sports Medicine, University of Washington School of Medicine, Seattle, Washington, United States of America
- Insitute for Stem Cell and Regenerative Medicines, University of Washington, Seattle Washington, United States of America
| | - David Karasik
- Institute for Aging Research, Hebrew SeniorLife, Boston, Massachusetts, United States of America
| | - Lisa Maves
- Center for Developmental Biology and Regenerative Medicine, Seattle Children’s Research Institute, Seattle, Washington, United States of America
- Department of Pediatrics, Division of Cardiology, University of Washington School of Medicine, Seattle, Washington, United States of America
| | - Bjorn Busse
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Yi-Hsiang Hsu
- Institute for Aging Research, Hebrew SeniorLife, Boston, Massachusetts, United States of America
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, United States of America
- Broad Institute of Harvard and Massachusetts Institute of Technology, Boston, Massachusetts, United States of America
| | - Ronald Young Kwon
- Department of Orthopaedics and Sports Medicine, University of Washington School of Medicine, Seattle, Washington, United States of America
- Insitute for Stem Cell and Regenerative Medicines, University of Washington, Seattle Washington, United States of America
| |
Collapse
|
19
|
Chen X, Ji Y, Feng F, Liu Z, Qian L, Shen H, Lao L. C-type lectin domain-containing protein CLEC3A regulates proliferation, regeneration and maintenance of nucleus pulposus cells. Cell Mol Life Sci 2022; 79:435. [PMID: 35864364 PMCID: PMC11071857 DOI: 10.1007/s00018-022-04477-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 07/02/2022] [Accepted: 07/06/2022] [Indexed: 11/03/2022]
Abstract
It is widely assumed that as connective tissue, the intervertebral disc (IVD) plays a crucial role in providing flexibility for the spinal column. The disc is comprised of three distinct tissues: the nucleus pulposus (NP), ligamentous annulus fibrous (AF) that surrounds the NP, and the hyaline cartilaginous endplates (CEP). Nucleus pulposus, composed of chondrocyte-like NP cells and its secreted gelatinous matrix, is critical for disc health and function. The NP matrix underwent dehydration accompanied by increasing fibrosis with age. The degeneration of matrix is almost impossible to repair, with the consequence of matrix stiffness and senescence of NP cells and intervertebral disc, suggesting the value of glycoproteins in extracellular matrix (ECM). Here, via database excavation and biological function screening, we investigated a C-type lectin protein, CLEC3A, which could support differentiation of chondrocytes as well as maintenance of NP cells and was essential to intervertebral disc homeostasis. Furthermore, mechanistic analysis revealed that CLEC3A could stimulate PI3K-AKT pathway to accelerate cell proliferation to further play part in NP cell regeneration.
Collapse
Affiliation(s)
- Xiuyuan Chen
- Department of Spine Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Yucheng Ji
- Department of Spine Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Fan Feng
- Department of Spine Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Zude Liu
- Department of Spine Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Lie Qian
- Department of Spine Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Hongxing Shen
- Department of Spine Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Lifeng Lao
- Department of Spine Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
| |
Collapse
|
20
|
Single-Cell RNA-Seq Analysis of Cells from Degenerating and Non-Degenerating Intervertebral Discs from the Same Individual Reveals New Biomarkers for Intervertebral Disc Degeneration. Int J Mol Sci 2022; 23:ijms23073993. [PMID: 35409356 PMCID: PMC8999935 DOI: 10.3390/ijms23073993] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 03/31/2022] [Accepted: 04/01/2022] [Indexed: 02/07/2023] Open
Abstract
In this study, we used single-cell transcriptomic analysis to identify new specific biomarkers for nucleus pulposus (NP) and inner annulus fibrosis (iAF) cells, and to define cell populations within non-degenerating (nD) and degenerating (D) human intervertebral discs (IVD) of the same individual. Cluster analysis based on differential gene expression delineated 14 cell clusters. Gene expression profiles at single-cell resolution revealed the potential functional differences linked to degeneration, and among NP and iAF subpopulations. GO and KEGG analyses discovered molecular functions, biological processes, and transcription factors linked to cell type and degeneration state. We propose two lists of biomarkers, one as specific cell type, including C2orf40, MGP, MSMP, CD44, EIF1, LGALS1, RGCC, EPYC, HILPDA, ACAN, MT1F, CHI3L1, ID1, ID3 and TMED2. The second list proposes predictive IVD degeneration genes, including MT1G, SPP1, HMGA1, FN1, FBXO2, SPARC, VIM, CTGF, MGST1, TAF1D, CAPS, SPTSSB, S100A1, CHI3L2, PLA2G2A, TNRSF11B, FGFBP2, MGP, SLPI, DCN, MT-ND2, MTCYB, ADIRF, FRZB, CLEC3A, UPP1, S100A2, PRG4, COL2A1, SOD2 and MT2A. Protein and mRNA expression of MGST1, vimentin, SOD2 and SYF2 (p29) genes validated our scRNA-seq findings. Our data provide new insights into disc cells phenotypes and biomarkers of IVD degeneration that could improve diagnostic and therapeutic options.
Collapse
|
21
|
Kamatani T, Hagizawa H, Yarimitsu S, Morioka M, Koyamatsu S, Sugimoto M, Kodama J, Yamane J, Ishiguro H, Shichino S, Abe K, Fujibuchi W, Fujie H, Kaito T, Tsumaki N. Human iPS cell-derived cartilaginous tissue spatially and functionally replaces nucleus pulposus. Biomaterials 2022; 284:121491. [PMID: 35395453 DOI: 10.1016/j.biomaterials.2022.121491] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 03/23/2022] [Accepted: 03/26/2022] [Indexed: 12/12/2022]
Abstract
The loss of nucleus pulposus (NP) precedes the intervertebral disk (IVD) degeneration that causes back pain. Here, we demonstrate that the implantation of human iPS cell-derived cartilaginous tissue (hiPS-Cart) restores this loss by replacing lost NP spatially and functionally. NP cells consist of notochordal NP cells and chondrocyte-like NP cells. Single cell RNA sequencing (scRNA-seq) analysis revealed that cells in hiPS-Cart corresponded to chondrocyte-like NP cells but not to notochordal NP cells. The implantation of hiPS-Cart into a nuclectomized space of IVD in nude rats prevented the degeneration of the IVD and preserved its mechanical properties. hiPS-Cart survived and occupied the nuclectomized space for at least six months after implantation, indicating spatial and functional replacement of lost NP by hiPS-Cart. Further scRNA-seq analysis revealed that hiPS-Cart cells changed their profile after implantation, differentiating into two lineages that are metabolically distinct from each other. However, post-implanted hiPS-Cart cells corresponded to chondrocyte-like NP cells only and did not develop into notochordal NP cells, suggesting that chondrocyte-like NP cells are nearly sufficient for NP function. The data collectively indicate that hiPS-Cart is a candidate implant for regenerating NP spatially and functionally and preventing IVD degeneration.
Collapse
Affiliation(s)
- Takashi Kamatani
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan; Department of Orthopaedic Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Hiroki Hagizawa
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan; Department of Orthopaedic Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan; Department of Tissue Biochemistry, Graduate School of Medicine and Frontier Biosciences, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Seido Yarimitsu
- Department of Mechanical Systems Engineering, Faculty of Systems Design, Tokyo Metropolitan University, 1-1 Minami-Osawa, Hachioji, Tokyo, 192-0397, Japan
| | - Miho Morioka
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan; Department of Tissue Biochemistry, Graduate School of Medicine and Frontier Biosciences, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Saeko Koyamatsu
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan; Department of Tissue Biochemistry, Graduate School of Medicine and Frontier Biosciences, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Michihiko Sugimoto
- Technology and Development Team for Mammalian Genome Dynamics, RIKEN BioResource Research Center, 3-1-1 Koyadai, Tsukuba, Ibaraki, 305-0074, Japan
| | - Joe Kodama
- Department of Orthopaedic Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Junko Yamane
- Department of Life Science Frontiers, Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Hiroyuki Ishiguro
- Department of Orthopaedic Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Shigeyuki Shichino
- Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute of Biomedical Sciences, Tokyo University of Science, Building 17 Second Floor, 2641, Yamasaki, Noda, Chiba, 278-0042, Japan
| | - Kuniya Abe
- Technology and Development Team for Mammalian Genome Dynamics, RIKEN BioResource Research Center, 3-1-1 Koyadai, Tsukuba, Ibaraki, 305-0074, Japan
| | - Wataru Fujibuchi
- Department of Life Science Frontiers, Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Hiromichi Fujie
- Department of Mechanical Systems Engineering, Faculty of Systems Design, Tokyo Metropolitan University, 1-1 Minami-Osawa, Hachioji, Tokyo, 192-0397, Japan
| | - Takashi Kaito
- Department of Orthopaedic Surgery, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Noriyuki Tsumaki
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, 53 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan; Department of Tissue Biochemistry, Graduate School of Medicine and Frontier Biosciences, Osaka University, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
22
|
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
|
23
|
Hickman TT, Rathan-Kumar S, Peck SH. Development, Pathogenesis, and Regeneration of the Intervertebral Disc: Current and Future Insights Spanning Traditional to Omics Methods. Front Cell Dev Biol 2022; 10:841831. [PMID: 35359439 PMCID: PMC8963184 DOI: 10.3389/fcell.2022.841831] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 02/09/2022] [Indexed: 02/06/2023] Open
Abstract
The intervertebral disc (IVD) is the fibrocartilaginous joint located between each vertebral body that confers flexibility and weight bearing capabilities to the spine. The IVD plays an important role in absorbing shock and stress applied to the spine, which helps to protect not only the vertebral bones, but also the brain and the rest of the central nervous system. Degeneration of the IVD is correlated with back pain, which can be debilitating and severely affects quality of life. Indeed, back pain results in substantial socioeconomic losses and healthcare costs globally each year, with about 85% of the world population experiencing back pain at some point in their lifetimes. Currently, therapeutic strategies for treating IVD degeneration are limited, and as such, there is great interest in advancing treatments for back pain. Ideally, treatments for back pain would restore native structure and thereby function to the degenerated IVD. However, the complex developmental origin and tissue composition of the IVD along with the avascular nature of the mature disc makes regeneration of the IVD a uniquely challenging task. Investigators across the field of IVD research have been working to elucidate the mechanisms behind the formation of this multifaceted structure, which may identify new therapeutic targets and inform development of novel regenerative strategies. This review summarizes current knowledge base on IVD development, degeneration, and regenerative strategies taken from traditional genetic approaches and omics studies and discusses the future landscape of investigations in IVD research and advancement of clinical therapies.
Collapse
Affiliation(s)
- Tara T. Hickman
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
- Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, United States
- Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Sudiksha Rathan-Kumar
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
- Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, United States
- Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Sun H. Peck
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
- Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, United States
- Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, Nashville, TN, United States
- *Correspondence: Sun H. Peck,
| |
Collapse
|
24
|
Xiong Y, Yang YL, Gao YS, Wang XM, Yu X. Histological Changes of Cervical Disc Tissue in Patients with Degenerative Ossification. J Korean Neurosurg Soc 2022; 65:186-195. [PMID: 35108774 PMCID: PMC8918244 DOI: 10.3340/jkns.2021.0082] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 06/20/2021] [Indexed: 12/02/2022] Open
Abstract
Objective To explore the histological feature of the cervical disc degeneration in patients with degenerative ossification (DO) and its potential mechanisms.
Methods A total of 96 surgical segments, from cervical disc degenerative disease patients with surgical treatment, were divided into ossification group (group O, n=46) and non-ossification group (group NO, n=50) based on preoperative radiological exams. Samples of disc tissues and osteophytes were harvested during the decompression operation. The hematoxylin-eosin staining, Masson trichrome staining and Safranin O-fast green staining were used to compare the histological differences between the two groups. And the distribution and content of transforming growth factor (TGF)-β1, p-Smad2 and p-Smad3 between the two groups were compared by a semi-quantitative immunohistochemistry (IHC) method.
Results For all the disc tissues, the content of disc cells and collagen fibers decreased gradually from the outer annulus fibrosus (OAF) to the central nucleus pulposus (NP). Compared with group NO, the number of disc cells in group O increased significantly. But for proteoglycan in the inner annulus fibrosus (IAF) and NP, the content in group O decreased significantly. IHC analysis showed that TGF-β1, p-Smad2, and p-Smad3 were detected in all tissues. For group O, the content of TGF-β1 in the OAF and NP was significantly higher than that in group NO. For p-Smad2 in IAF and p-Smad3 in OAF, the content in group O were significantly higher than group NO.
Conclusion Histologically, cervical disc degeneration in patients with DO is more severe than that without DO. Local higher content of TGF-β1, p-Smad2, and p-Smad3 are involved in the disc degeneration with DO. Further studies with multi-approach analyses are needed to better understand the role of TGF-β/Smads signaling pathway in the disc degeneration with DO.
Collapse
Affiliation(s)
- Yang Xiong
- School of Materials Science and Engineering, Tsinghua University, Beijing, China.,Department of Orthopedics, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Ying-Li Yang
- Department of Traditional Chinese Medicine, Peking Union Medical College Hospital, Beijing, China
| | - Yu-Shan Gao
- School of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xiu-Mei Wang
- School of Materials Science and Engineering, Tsinghua University, Beijing, China
| | - Xing Yu
- Department of Orthopedics, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
25
|
Intervertebral disc repair and regeneration: Insights from the notochord. Semin Cell Dev Biol 2021; 127:3-9. [PMID: 34865989 DOI: 10.1016/j.semcdb.2021.11.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 11/05/2021] [Accepted: 11/11/2021] [Indexed: 12/25/2022]
Abstract
The vertebrate notochord plays an essential role in patterning multiple structures during embryonic development. In the early 2000s, descendants of notochord cells were demonstrated to form the entire nucleus pulposus of the intervertebral disc in addition to their key role in embryonic patterning. The nucleus pulposus undergoes degeneration during postnatal life, which can lead to back pain. Recently, gene and protein profiles of notochord and nucleus pulposus cells have been identified. These datasets, coupled with the ability to differentiate human induced pluripotent stem cells (iPSCs) into cells that resemble nucleus pulposus cells, provide the possibility of generating a cell-based therapy to halt and/or reverse disc degeneration.
Collapse
|
26
|
Fukunaga K, Tanji M, Hanzawa N, Kuroda H, Inui M. Protocadherin-1 is expressed in the notochord of mouse embryo but is dispensable for its formation. Biochem Biophys Rep 2021; 27:101047. [PMID: 34189280 PMCID: PMC8219654 DOI: 10.1016/j.bbrep.2021.101047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 05/25/2021] [Accepted: 06/06/2021] [Indexed: 11/18/2022] Open
Abstract
Notochord is an embryonic midline structure that serves as mechanical support for axis elongation and the signaling center for the surrounding tissues. Precursors of notochord are initially induced in the dorsal most mesoderm region in gastrulating embryo and separate from the surrounding mesoderm/endoderm tissue to form an elongated rod-like structure, suggesting that cell adhesion molecules may play an important role in this step. In Xenopus embryo, axial protocadherin (AXPC), an orthologue of mammalian Protocadherin-1 (PCDH1), is indispensable for the assembly and separation from the surrounding tissue of the notochord cells. However, the role of PCDH1 in mammalian notochord remains unknown. We herein report that PCDH1 is expressed in the notochord of mouse embryo and that PCDH1-deficient mice form notochord normally. First, we examined the temporal expression pattern of pcdh1 and found that pcdh1 mRNA was expressed from embryonic day (E) 7.5, prior to the stage when notochord cells detach from the surrounding endoderm tissue. Second, we found that PCDH1 protein is expressed in the notochord of mouse embryos in addition to the previously reported expression in endothelial cells. To further investigate the role of PCDH1 in embryonic development, we generated PCDH1-deficient mice using the CRISPR-Cas9 system. In PCDH1-deficient embryos, notochord formation and separation from the surrounding tissue were normal. Structure and marker gene expression of notochord were also unaffected by loss of PCDH1. Major vascular patterns in PCDH1-deficient embryo were essentially normal. These results suggest that PCDH1 is dispensable for notochord formation, including the tissue separation process, in mammalian embryos. We successfully identified the evolutionary conserved expression of PCDH1 in notochord, but its function may differ among species.
Collapse
Affiliation(s)
- Kanako Fukunaga
- Systems Biology Program, Graduate School of Media and Governance, Keio University, Kanagawa, 252-0882, Japan
- Institute for Advanced Biosciences, Keio University, Kanagawa, 252-0882, Japan
- Laboratory of Animal Regeneration Systemology, Department of Life Sciences, School of Agriculture, Meiji University, Kanagawa, 214-8571, Japan
| | - Masafumi Tanji
- Laboratory of Animal Regeneration Systemology, Department of Life Sciences, School of Agriculture, Meiji University, Kanagawa, 214-8571, Japan
| | - Nana Hanzawa
- Laboratory of Animal Regeneration Systemology, Department of Life Sciences, School of Agriculture, Meiji University, Kanagawa, 214-8571, Japan
| | - Hiroki Kuroda
- Institute for Advanced Biosciences, Keio University, Kanagawa, 252-0882, Japan
- Faculty of Environment and Information Studies, Keio University, Kanagawa, 252-0882, Japan
| | - Masafumi Inui
- Laboratory of Animal Regeneration Systemology, Department of Life Sciences, School of Agriculture, Meiji University, Kanagawa, 214-8571, Japan
- Department of Systems BioMedicine, National Institute for Child Health and Development, Tokyo, 157-8535, Japan
- Corresponding author. Laboratory of Animal Regeneration Systemology, Department of Life Science, School of Agriculture, Meiji University, Kanagawa, 214-8571, Japan.
| |
Collapse
|
27
|
Speer JE, Barcellona MN, Lu MY, Zha Z, Jing L, Gupta MC, Buchowski JM, Kelly MP, Setton LA. Development of a library of laminin-mimetic peptide hydrogels for control of nucleus pulposus cell behaviors. J Tissue Eng 2021; 12:20417314211021220. [PMID: 34188794 PMCID: PMC8211742 DOI: 10.1177/20417314211021220] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 05/12/2021] [Indexed: 12/15/2022] Open
Abstract
The nucleus pulposus (NP) of the intervertebral disc plays a critical role in
distributing mechanical loads to the axial skeleton. Alterations in NP cells and,
consequently, NP matrix are some of the earliest changes in the development of disc
degeneration. Previous studies demonstrated a role for laminin-presenting biomaterials in
promoting a healthy phenotype for human NP cells from degenerated tissue. Here we
investigate the use of laminin-mimetic peptides presented individually or in combination
on a poly(ethylene) glycol hydrogel as a platform to modulate the behaviors of
degenerative human NP cells. Data confirm that NP cells attach to select laminin-mimetic
peptides that results in cell signaling downstream of integrin and syndecan binding.
Furthermore, the peptide-functionalized hydrogels demonstrate an ability to promote cell
behaviors that mimic that of full-length laminins. These results identify a set of
peptides that can be used to regulate NP cell behaviors toward a regenerative engineering
strategy.
Collapse
Affiliation(s)
- Julie E Speer
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Marcos N Barcellona
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Michael Y Lu
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Zizhen Zha
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Liufang Jing
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Munish C Gupta
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Jacob M Buchowski
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Michael P Kelly
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Lori A Setton
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA.,Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
28
|
The Cellular Composition of Bovine Coccygeal Intervertebral Discs: A Comprehensive Single-Cell RNAseq Analysis. Int J Mol Sci 2021. [DOI: 10.3390/ijms22094917
expr 996488947 + 961598850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Abstract
Intervertebral disc (IVD) degeneration and its medical consequences is still one of the leading causes of morbidity worldwide. To support potential regenerative treatments for degenerated IVDs, we sought to deconvolute the cell composition of the nucleus pulposus (NP) and the annulus fibrosus (AF) of bovine intervertebral discs. Bovine calf tails have been extensively used in intervertebral disc research as a readily available source of NP and AF material from healthy and young IVDs. We used single-cell RNA sequencing (scRNAseq) coupled to bulk RNA sequencing (RNAseq) to unravel the cell populations in these two structures and analyze developmental changes across the rostrocaudal axis. By integrating the scRNAseq data with the bulk RNAseq data to stabilize the clustering results of our study, we identified 27 NP structure/tissue specific genes and 24 AF structure/tissue specific genes. From our scRNAseq results, we could deconvolute the heterogeneous cell populations in both the NP and the AF. In the NP, we detected a notochordal-like cell cluster and a progenitor stem cell cluster. In the AF, we detected a stem cell-like cluster, a cluster with a predominantly fibroblast-like phenotype and a potential endothelial progenitor cluster. Taken together, our results illustrate the cell phenotypic complexity of the AF and NP in the young bovine IVDs.
Collapse
|
29
|
Calió M, Gantenbein B, Egli M, Poveda L, Ille F. The Cellular Composition of Bovine Coccygeal Intervertebral Discs: A Comprehensive Single-Cell RNAseq Analysis. Int J Mol Sci 2021; 22:ijms22094917. [PMID: 34066404 PMCID: PMC8124861 DOI: 10.3390/ijms22094917] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/26/2021] [Accepted: 04/27/2021] [Indexed: 02/07/2023] Open
Abstract
Intervertebral disc (IVD) degeneration and its medical consequences is still one of the leading causes of morbidity worldwide. To support potential regenerative treatments for degenerated IVDs, we sought to deconvolute the cell composition of the nucleus pulposus (NP) and the annulus fibrosus (AF) of bovine intervertebral discs. Bovine calf tails have been extensively used in intervertebral disc research as a readily available source of NP and AF material from healthy and young IVDs. We used single-cell RNA sequencing (scRNAseq) coupled to bulk RNA sequencing (RNAseq) to unravel the cell populations in these two structures and analyze developmental changes across the rostrocaudal axis. By integrating the scRNAseq data with the bulk RNAseq data to stabilize the clustering results of our study, we identified 27 NP structure/tissue specific genes and 24 AF structure/tissue specific genes. From our scRNAseq results, we could deconvolute the heterogeneous cell populations in both the NP and the AF. In the NP, we detected a notochordal-like cell cluster and a progenitor stem cell cluster. In the AF, we detected a stem cell-like cluster, a cluster with a predominantly fibroblast-like phenotype and a potential endothelial progenitor cluster. Taken together, our results illustrate the cell phenotypic complexity of the AF and NP in the young bovine IVDs.
Collapse
Affiliation(s)
- Martina Calió
- Tissue Engineering for Orthopaedics & Mechanobiology (TOM), Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland; (M.C.); (B.G.)
- Department of Orthopaedic Surgery and Traumatology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland
- Space Biology Group, Institute of Medical Engineering, School of Engineering and Architecture, Lucerne University of Applied Sciences and Arts, 6052 Hergiswil, Switzerland;
| | - Benjamin Gantenbein
- Tissue Engineering for Orthopaedics & Mechanobiology (TOM), Department for BioMedical Research (DBMR), University of Bern, 3008 Bern, Switzerland; (M.C.); (B.G.)
- Department of Orthopaedic Surgery and Traumatology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland
| | - Marcel Egli
- Space Biology Group, Institute of Medical Engineering, School of Engineering and Architecture, Lucerne University of Applied Sciences and Arts, 6052 Hergiswil, Switzerland;
| | - Lucy Poveda
- Functional Genomics Center Zurich, Swiss Federal Institute of Technology, University of Zurich, 8057 Zurich, Switzerland;
| | - Fabian Ille
- Space Biology Group, Institute of Medical Engineering, School of Engineering and Architecture, Lucerne University of Applied Sciences and Arts, 6052 Hergiswil, Switzerland;
- Correspondence: ; Tel.: +41-41-349-36-15
| |
Collapse
|
30
|
Li Y, Liu S, Pan D, Xu B, Xing X, Zhou H, Zhang B, Zhou S, Ning G, Feng S. The potential role and trend of HIF‑1α in intervertebral disc degeneration: Friend or foe? (Review). Mol Med Rep 2021; 23:239. [PMID: 33537810 PMCID: PMC7893690 DOI: 10.3892/mmr.2021.11878] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 11/08/2019] [Indexed: 12/11/2022] Open
Abstract
Lower back pain (LBP) is one of the most common reasons for seeking medical advice in orthopedic clinics. Increasingly, research has shown that symptomatic intervertebral disc degeneration (IDD) is mostly related to LBP. This review first outlines the research and findings of studies into IDD, from the physiological structure of the intervertebral disc (IVD) to various pathological cascades. The vicious cycles of IDD are re-described in relation to the analysis of the relationship among the pathological mechanisms involved in IDD. Interestingly, a ‘chief molecule’ was found, hypoxia-inducible factor-1α (HIF-1α), that may regulate all other mechanisms involved in IDD. When the vicious cycle is established, the low oxygen tension activates the expression of HIF-1α, which subsequently enters into the hypoxia-induced HIF pathways. The HIF pathways are dichotomized as friend and foe pathways according to the oxygen tension of the IVD microenvironment. Combined with clinical outcomes and previous research, the trend of IDD development has been predicted in this paper. Lastly, an early precautionary diagnosis and treatment method is proposed whereby nucleus pulposus tissue for biopsy can be obtained through IVD puncture guided by B-ultrasound when the patient is showing symptoms but MRI imaging shows negative results. The assessment criteria for biopsy and the feasibility, superiority and challenges of this approach have been discussed. Overall, it is clear that HIF-1α is an indispensable reference indicator for the accurate diagnosis and treatment of IDD.
Collapse
Affiliation(s)
- Yongjin Li
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Shen Liu
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Dayu Pan
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Baoshan Xu
- Department of Spine Surgery, Tianjin Hospital, Tianjin 300000, P.R. China
| | - Xuewu Xing
- Department of Orthopedic Surgery, First Central Clinical of Tianjin Medical University, Tianjin 300052, P.R. China
| | - Hengxing Zhou
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Bin Zhang
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Suzhe Zhou
- Department of Orthopedics, The Affiliated Zhongshan Hospital of Fudan University, Shanghai 200034, P.R. China
| | - Guangzhi Ning
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Shiqing Feng
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| |
Collapse
|
31
|
Deal KK, Rosebrock JC, Eeds AM, DeKeyser JML, Musser MA, Ireland SJ, May-Zhang AA, Buehler DP, Southard-Smith EM. Sox10-cre BAC transgenes reveal temporal restriction of mesenchymal cranial neural crest and identify glandular Sox10 expression. Dev Biol 2020; 471:119-137. [PMID: 33316258 DOI: 10.1016/j.ydbio.2020.12.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 12/02/2020] [Accepted: 12/07/2020] [Indexed: 12/29/2022]
Abstract
Diversity of neural crest derivatives has been studied with a variety of approaches during embryonic development. In mammals Cre-LoxP lineage tracing is a robust means to fate map neural crest relying on cre driven from regulatory elements of early neural crest genes. Sox10 is an essential transcription factor for normal neural crest development. A variety of efforts have been made to label neural crest derivatives using partial Sox10 regulatory elements to drive cre expression. To date published Sox10-cre lines have focused primarily on lineage tracing in specific tissues or during early fetal development. We describe two new Sox10-cre BAC transgenes, constitutive (cre) and inducible (cre/ERT2), that contain the complete repertoire of Sox10 regulatory elements. We present a thorough expression profile of each, identifying a few novel sites of Sox10 expression not captured by other neural crest cre drivers. Comparative mapping of expression patterns between the Sox10-cre and Sox10-cre/ERT2 transgenes identified a narrow temporal window in which Sox10 expression is present in mesenchymal derivatives prior to becoming restricted to neural elements during embryogenesis. In more caudal structures, such as the intestine and lower urinary tract, our Sox10-cre BAC transgene appears to be more efficient in labeling neural crest-derived cell types than Wnt1-cre. The analysis reveals consistent expression of Sox10 in non-neural crest derived glandular epithelium, including salivary, mammary, and urethral glands of adult mice. These Sox10-cre and Sox10-cre/ERT2 transgenic lines are verified tools that will enable refined temporal and cell-type specific lineage analysis of neural crest derivatives as well as glandular tissues that rely on Sox10 for proper development and function.
Collapse
Affiliation(s)
- Karen K Deal
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Jennifer C Rosebrock
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Angela M Eeds
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Jean-Marc L DeKeyser
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA; Present address: Northwestern University, Dept. of Pharmacology, USA
| | - Melissa A Musser
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA; Present address: Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA, USA
| | - Sara J Ireland
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Aaron A May-Zhang
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Dennis P Buehler
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - E Michelle Southard-Smith
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA.
| |
Collapse
|
32
|
Abstract
The vertebrate body plan is characterized by the presence of a segmented spine along its main axis. Here, we examine the current understanding of how the axial tissues that are formed during embryonic development give rise to the adult spine and summarize recent advances in the field, largely focused on recent studies in zebrafish, with comparisons to amniotes where appropriate. We discuss recent work illuminating the genetics and biological mechanisms mediating extension and straightening of the body axis during development, and highlight open questions. We specifically focus on the processes of notochord development and cerebrospinal fluid physiology, and how defects in those processes may lead to scoliosis.
Collapse
Affiliation(s)
- Michel Bagnat
- Department of Cell Biology, Duke University, Durham, NC, 27710, USA
| | - Ryan S Gray
- Department of Nutritional Sciences, University of Texas at Austin, Dell Pediatrics Research Institute, Austin, TX, 78723, USA
| |
Collapse
|
33
|
Fernandes LM, Khan NM, Trochez CM, Duan M, Diaz-Hernandez ME, Presciutti SM, Gibson G, Drissi H. Single-cell RNA-seq identifies unique transcriptional landscapes of human nucleus pulposus and annulus fibrosus cells. Sci Rep 2020; 10:15263. [PMID: 32943704 PMCID: PMC7499307 DOI: 10.1038/s41598-020-72261-7] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 08/19/2020] [Indexed: 12/29/2022] Open
Abstract
Intervertebral disc (IVD) disease (IDD) is a complex, multifactorial disease. While various aspects of IDD progression have been reported, the underlying molecular pathways and transcriptional networks that govern the maintenance of healthy nucleus pulposus (NP) and annulus fibrosus (AF) have not been fully elucidated. We defined the transcriptome map of healthy human IVD by performing single-cell RNA-sequencing (scRNA-seq) in primary AF and NP cells isolated from non-degenerated lumbar disc. Our systematic and comprehensive analyses revealed distinct genetic architecture of human NP and AF compartments and identified 2,196 differentially expressed genes. Gene enrichment analysis showed that SFRP1, BIRC5, CYTL1, ESM1 and CCNB2 genes were highly expressed in the AF cells; whereas, COL2A1, DSC3, COL9A3, COL11A1, and ANGPTL7 were mostly expressed in the NP cells. Further, functional annotation clustering analysis revealed the enrichment of receptor signaling pathways genes in AF cells, while NP cells showed high expression of genes related to the protein synthesis machinery. Subsequent interaction network analysis revealed a structured network of extracellular matrix genes in NP compartments. Our regulatory network analysis identified FOXM1 and KDM4E as signature transcription factor of AF and NP respectively, which might be involved in the regulation of core genes of AF and NP transcriptome.
Collapse
Affiliation(s)
- Lorenzo M Fernandes
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA, 30033, USA.,Atlanta VA Medical Center, Decatur, GA, USA
| | - Nazir M Khan
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA, 30033, USA.,Atlanta VA Medical Center, Decatur, GA, USA
| | - Camila M Trochez
- Center for Integrative Genomics, Georgia Institute of Technology, Atlanta, GA, USA
| | - Meixue Duan
- Center for Integrative Genomics, Georgia Institute of Technology, Atlanta, GA, USA
| | - Martha E Diaz-Hernandez
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA, 30033, USA.,Atlanta VA Medical Center, Decatur, GA, USA
| | - Steven M Presciutti
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA, 30033, USA.,Atlanta VA Medical Center, Decatur, GA, USA
| | - Greg Gibson
- Center for Integrative Genomics, Georgia Institute of Technology, Atlanta, GA, USA
| | - Hicham Drissi
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, GA, 30033, USA. .,Atlanta VA Medical Center, Decatur, GA, USA.
| |
Collapse
|
34
|
Piprode V, Mohanty S, Bonavita R, Loh S, Anbazhagan R, Saini C, Pinelli R, Pricop P, Dahia CL. An optimized step-by-step protocol for isolation of nucleus pulposus, annulus fibrosus, and end plate cells from the mouse intervertebral discs and subsequent preparation of high-quality intact total RNA. JOR Spine 2020; 3:e1108. [PMID: 33015579 PMCID: PMC7524240 DOI: 10.1002/jsp2.1108] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 05/27/2020] [Accepted: 06/29/2020] [Indexed: 12/14/2022] Open
Abstract
Intervertebral disc degeneration is the most significant, and least understood, cause of chronic back pain, affecting almost one in seven individuals at some point of time. Each intervertebral disc has three components; central nucleus pulposus (NP), concentric layers of annulus fibrosus (AF), and a pair of end plate (EP) that connects the disc to the vertebral bodies. Understanding the molecular and cellular basis of intervertebral disc growth, health, and aging will generate significant information for developing therapeutic approaches. Rapid and efficient preparations of homogeneous and pure cells are crucial for meaningful and rigorous downstream analysis at the cellular, molecular, and biochemical level. Cross-sample contamination may influence the interpretation of the results. In addition to altering gene expression, slow or delayed isolation procedures will also cause the degradation of cells and biomolecules that create a bias in the outcomes of the study. The mouse model system is extensively used to understand the intervertebral disc biology. Here we describe two protocols: (a) for efficient isolation of pure NP, AF, and EP cells from mouse lumbar intervertebral disc. We validated the purity of the NP and AF cells using Shh Cre/+ ; R26 mT/mG/+ dual-fluorescent reporter mice where all NP cells are GPF+ve, and by the sensitive approach of qPCR analysis using TaqMan probes for Shh, and Brachyury as NP-specific markers, Tenomodulin as AF-specific marker, and Osteocalcin as bone-specific marker. (b) For isolation of high-quality intact RNA with RIN of 9.3 to 10 from disc cells. These methods will be useful for the rigorous analysis of NP and AF cells, and improve our understanding of intervertebral disc biology.
Collapse
Affiliation(s)
| | | | | | - Sarah Loh
- Hospital for Special Surgery New York New York USA
| | | | | | | | - Paul Pricop
- Hospital for Special Surgery New York New York USA
| | - Chitra L Dahia
- Hospital for Special Surgery New York New York USA
- Department of Cell and Developmental Biology, Weill Cornell Medicine Graduate School of Medical Sciences New York New York USA
| |
Collapse
|
35
|
Veras MA, Lim YJ, Kuljanin M, Lajoie GA, Urquhart BL, Séguin CA. Protocol for parallel proteomic and metabolomic analysis of mouse intervertebral disc tissues. JOR Spine 2020; 3:e1099. [PMID: 33015574 PMCID: PMC7524214 DOI: 10.1002/jsp2.1099] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 04/25/2020] [Accepted: 05/14/2020] [Indexed: 01/07/2023] Open
Abstract
The comprehensiveness of data collected by "omics" modalities has demonstrated the ability to drastically transform our understanding of the molecular mechanisms of chronic, complex diseases such as musculoskeletal pathologies, how biomarkers are identified, and how therapeutic targets are developed. Standardization of protocols will enable comparisons between findings reported by multiple research groups and move the application of these technologies forward. Herein, we describe a protocol for parallel proteomic and metabolomic analysis of mouse intervertebral disc (IVD) tissues, building from the combined expertise of our collaborative team. This protocol covers dissection of murine IVD tissues, sample isolation, and data analysis for both proteomics and metabolomics applications. The protocol presented below was optimized to maximize the utility of a mouse model for "omics" applications, accounting for the challenges associated with the small starting quantity of sample due to small tissue size as well as the extracellular matrix-rich nature of the tissue.
Collapse
Affiliation(s)
- Matthew A Veras
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry The University of Western Ontario London Ontario Canada
- Bone and Joint Institute The University of Western Ontario London Ontario Canada
| | - Yong J Lim
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry The University of Western Ontario London Ontario Canada
| | - Miljan Kuljanin
- Department of Cell Biology Harvard Medical School Boston Massachusetts USA
| | - Gilles A Lajoie
- Department of Biochemistry, Schulich School of Medicine & Dentistry The University of Western Ontario London Ontario Canada
| | - Bradley L Urquhart
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry The University of Western Ontario London Ontario Canada
| | - Cheryle A Séguin
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry The University of Western Ontario London Ontario Canada
- Bone and Joint Institute The University of Western Ontario London Ontario Canada
| |
Collapse
|
36
|
Qu Z, Zhang F, Chen W, Lin T, Sun Y. High-dose TGF-β1 degrades human nucleus pulposus cells via ALK1-Smad1/5/8 activation. Exp Ther Med 2020; 20:3661-3668. [PMID: 32855718 PMCID: PMC7444386 DOI: 10.3892/etm.2020.9088] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 07/01/2020] [Indexed: 01/07/2023] Open
Abstract
Transforming growth factor β1 (TGF-β1) can promote the proliferation and differentiation of intervertebral disc cells and participates in its repair process. However, whether TGF-β1 engages in the process of disc degeneration has not yet been fully elucidated. The present study aimed to investigate the function of high-dose TGF-β1 on the metabolism of nucleus pulposus cells (NPCs). TGF-β1 levels in human degenerative intervertebral disc tissues and tumor necrosis factor (TNF)-α-induced degenerative NPCs were analyzed. Furthermore, NPCs were treated with TGF-β1 and inhibitors of TGF-β1 receptors [ALK tyrosine kinase receptor (ALK) 1 and ALK5] to determine the effect of the receptors in the mediation of NPC degeneration. The NPC state was determined by the components of secretory collagen I/II, tissue inhibitor of metalloproteinase-3 (TIMP-3) and matrix metalloproteinase (MMP)-13. The mRNA expression of Smad1/2/3/5/8, the downstream gene of TGF-β1 mediated by ALK, was also measured. Results showed that TGF-β1 and ALK1 were positively associated with the degree of degeneration of NP or NPCs in vitro, but negatively associated with ALK5. Furthermore, high-doses of TGF-β1 suppressed collagen II, but enhanced collagen I, TIMP-3, MMP-13, ALK1/5 and Smad1/2/3/5/8 expression. ALK5 inhibition induced the suppression of Smad2/3 and aggravated high-dose TGF-β1-induced NPC degeneration, as shown by the reduction in collagen II and increase in collagen I, TIMP-3 and MMP-13. By contrast, ALK1 inhibition resulted in Smad1/5/8 suppression and alleviated high-dose TGF-β1-induced NPC degeneration. Taken together, it was concluded that high-doses of TGF-β1 contributed to the degeneration of NPCs via the upregulation of ALK1 and Smad1/5/8.
Collapse
Affiliation(s)
- Zhiqiang Qu
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China.,Department of Orthopedics, Tongliao City Hospital, Tongliao, Inner Mongolia 028000, P.R. China
| | - Fengxiang Zhang
- Department of General Surgery, Tongliao City Hospital, Tongliao, Inner Mongolia 028000, P.R. China
| | - Weiwei Chen
- Department of Disinfecting Supply Division, Tongliao City Hospital, Tongliao, Inner Mongolia 028000, P.R. China
| | - Tao Lin
- Department of Disinfecting Supply Division, Tongliao City Hospital, Tongliao, Inner Mongolia 028000, P.R. China
| | - Yongming Sun
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, P.R. China
| |
Collapse
|
37
|
Dickinson PJ, Bannasch DL. Current Understanding of the Genetics of Intervertebral Disc Degeneration. Front Vet Sci 2020; 7:431. [PMID: 32793650 PMCID: PMC7393939 DOI: 10.3389/fvets.2020.00431] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 06/15/2020] [Indexed: 11/13/2022] Open
Abstract
Premature degeneration of the intervertebral disc and its association with specific chondrodystrophic dog breeds has been recognized for over a century. Several lines of evidence including disease breed predisposition, studies suggesting heritability of premature intervertebral disc degeneration (IVDD) and association of a dog chromosome 12 (CFA 12) locus with intervertebral disc calcification have strongly supported a genetic component in IVDD in dogs. Recent studies documenting association of IVDD with an overexpressing FGF4 retrogene on CFA 12 have opened up new areas of investigation to further define the pathophysiology of premature IVDD. While preliminary data from studies investigating FGF4 retrogenes in IVDD implicate FGF4 overexpression as a major disease factor, they have also highlighted knowledge gaps in our understanding of intervertebral disc herniation which is a complex and multifactorial disease process.
Collapse
Affiliation(s)
- Peter J Dickinson
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| | - Danika L Bannasch
- Department of Population Health and Reproduction, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States
| |
Collapse
|
38
|
Tessier S, Risbud MV. Understanding embryonic development for cell-based therapies of intervertebral disc degeneration: Toward an effort to treat disc degeneration subphenotypes. Dev Dyn 2020; 250:302-317. [PMID: 32564440 DOI: 10.1002/dvdy.217] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 06/09/2020] [Accepted: 06/14/2020] [Indexed: 12/19/2022] Open
Abstract
Chronic low back and neck pain are associated with intervertebral disc degeneration and are major contributors to the global burden of disability. New evidence now suggests that disc degeneration comprises a spectrum of subphenotypes influenced by genetic background, age, and environmental factors, which may be contributing to the mixed outcomes seen in clinical trials of cell-based therapies that aim to treat disc degeneration. This problem is further compounded by the fact that disc degeneration and aging coincide with an exhaustion of endogenous progenitor cells, imposing limitations on the regenerative capacity of the disc. At the bench-side, current work is focused on applying our knowledge of embryonic disc development to direct and refine differentiation of adult and human-induced pluripotent stem cells into notochord-like and nucleus pulposus-like cells for use in novel cell-based therapies. Accordingly, this review presents the salient features of intervertebral disc development, post-natal maintenance, and regeneration, with emphasis on recent advancements. We also discuss how a stratified approach can be undertaken for the development of future cell-based therapies to bring emerging subphenotypes into consideration.
Collapse
Affiliation(s)
- Steven Tessier
- Lewis Katz School of Medicine, Temple University, Philadelphia, Pennsylvania, USA.,Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Makarand V Risbud
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA.,Graduate Program in Cell Biology and Regenerative Medicine, Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
39
|
Nakamichi R, Asahara H. The transcription factors regulating intervertebral disc development. JOR Spine 2020; 3:e1081. [PMID: 32211592 PMCID: PMC7084052 DOI: 10.1002/jsp2.1081] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 01/22/2020] [Accepted: 02/01/2020] [Indexed: 01/06/2023] Open
Abstract
Damage to the intervertebral discs (IVDs) occurs due to aging or excessive mechanical stress, causing a series of IVD-related degenerative diseases, such as spinal disc herniation and spondylosis. These IVD-related diseases are difficult to cure, partially because the regeneration ability of IVDs is not sufficient. As a novel strategy for treatment of IVD-related diseases, mesenchymal stem cell transplantation to the damaged discs has been reported in animal studies. To further develop and improve this approach, it is necessary to gain a better understanding of the molecular network regulating IVD development by critical transcription factors. Recent findings reveal that during IVD development, nucleus pulposus and annuls fibrosus differentiation is coordinated by a series of transcription factors, such as Mkx, Pax1, 9, Shh, Foxa1, 2, T-Brachyury, and Sox5, 6, 9. The combination of mesenchymal stem cell transplantation with the regulation of these molecules may provide a novel strategy for treatment of degenerative disc diseases.
Collapse
Affiliation(s)
- Ryo Nakamichi
- Department of Molecular and Experimental MedicineThe Scripps Research InstituteLa JollaCalifornia
- Department of Orthopaedic SurgeryOkayama University Graduate School of Medicine, Dentistry, and Pharmaceutical SciencesOkayamaJapan
| | - Hiroshi Asahara
- Department of Molecular and Experimental MedicineThe Scripps Research InstituteLa JollaCalifornia
- Department of Systems BiomedicineTokyo Medical and Dental UniversityTokyoJapan
| |
Collapse
|
40
|
NOTO Transcription Factor Directs Human Induced Pluripotent Stem Cell-Derived Mesendoderm Progenitors to a Notochordal Fate. Cells 2020; 9:cells9020509. [PMID: 32102328 PMCID: PMC7072849 DOI: 10.3390/cells9020509] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 02/18/2020] [Accepted: 02/19/2020] [Indexed: 12/11/2022] Open
Abstract
The founder cells of the Nucleus pulposus, the centre of the intervertebral disc, originate in the embryonic notochord. After birth, mature notochordal cells (NC) are identified as key regulators of disc homeostasis. Better understanding of their biology has great potential in delaying the onset of disc degeneration or as a regenerative-cell source for disc repair. Using human pluripotent stem cells, we developed a two-step method to generate a stable NC-like population with a distinct molecular signature. Time-course analysis of lineage-specific markers shows that WNT pathway activation and transfection of the notochord-related transcription factor NOTO are sufficient to induce high levels of mesendoderm progenitors and favour their commitment toward the notochordal lineage instead of paraxial and lateral mesodermal or endodermal lineages. This study results in the identification of NOTO-regulated genes including some that are found expressed in human healthy disc tissue and highlights NOTO function in coordinating the gene network to human notochord differentiation.
Collapse
|
41
|
Veras MA, McCann MR, Tenn NA, Séguin CA. Transcriptional profiling of the murine intervertebral disc and age-associated changes in the nucleus pulposus. Connect Tissue Res 2020; 61:63-81. [PMID: 31597481 DOI: 10.1080/03008207.2019.1665034] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Purpose/Aim: The intervertebral disc (IVD) is composed of cell types whose subtle phenotypic differences allow for the formation of distinct tissues. The role of the nucleus pulposus (NP) in the initiation and progression of IVD degeneration is well established; however, the genes and pathways associated with NP degeneration are poorly characterized.Materials and Methods: Using a genetic strategy for IVD lineage-specific fluorescent reporter expression to isolate cells, gene expression and bioinformatic analysis was conducted on the murine NP at 2.5, 6, and 21 months-of-age and the annulus fibrosus (AF) at 2.5 and 6 months-of-age. A subset of differentially regulated genes was validated by qRT-PCR.Results: Transcriptome analysis identified distinct profiles of NP and AF gene expression that were remarkably consistent at 2.5 and 6 months-of-age. Prg4, Cilp, Ibsp and Comp were increased >50-fold in the AF relative to NP. The most highly enriched NP genes included Dsc3 and Cdh6, members of the cadherin superfamily, and microRNAs mir218-1 and mir490. Changes in the NP between 2.5 and 6 months-of-age were associated with up-regulation of molecular functions linked to laminin and Bmp receptor binding (including up-regulation of Bmp5 & 7), with the most up-regulated genes being Mir703, Shh, and Sfrp5. NP degeneration was associated with molecular functions linked to alpha-actinin binding (including up-regulation of Ttn & Myot) and cytoskeletal protein binding, with the overall most up-regulated genes being Rnu3a, Snora2b and Mir669h.Conclusions: This study provided insight into the phenotypes of NP and AF cells, and identified candidate pathways that may regulate degeneration.
Collapse
Affiliation(s)
- Matthew A Veras
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, The Bone and Joint Institute, The University of Western Ontario, London, Canada
| | - Matthew R McCann
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, The Bone and Joint Institute, The University of Western Ontario, London, Canada.,Sydney Medical School, University of Sydney, Sydney, Australia
| | - Neil A Tenn
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, The Bone and Joint Institute, The University of Western Ontario, London, Canada
| | - Cheryle A Séguin
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, The Bone and Joint Institute, The University of Western Ontario, London, Canada
| |
Collapse
|
42
|
Bratsman A, Couasnay G, Elefteriou F. A step-by-step protocol for isolation of murine nucleus pulposus cells. JOR Spine 2019; 2:e1073. [PMID: 31891122 PMCID: PMC6920701 DOI: 10.1002/jsp2.1073] [Citation(s) in RCA: 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] [Received: 08/21/2019] [Revised: 10/28/2019] [Accepted: 11/07/2019] [Indexed: 02/06/2023] Open
Abstract
The intervertebral disc (IVD) is composed of three separate tissues with distinct origins and properties. Elucidating changes occurring in these tissues in response to injury or age is paramount to identify new therapies to better manage disc and spine degenerative conditions, including low back pain. Despite their small size and different mechanical load pattern compared to higher species, the use of mouse models represents a cost-effective and powerful approach to better understand the formation, maintenance, and degeneration of the IVD. However, the isolation of the different compartments of the IVD is complicated by their diminutive size. Here, we describe a simple, step-by-step protocol for the isolation of the nucleus pulposus (NP) tissues that can then be processed for further analyses. Analysis from mouse NP tissues shows sufficient quantities of RNAs, purity of the NP fraction, and overall RNA quality for gene expression studies, and reveals no increase in expression of disc degeneration markers, including TNFa, IL1b, and Mmp1 up to 15 months of age in C57BL6 wildtype mice.
Collapse
Affiliation(s)
- Andrew Bratsman
- Department of Orthopedic SurgeryBaylor College of MedicineHoustonTexas
| | - Greig Couasnay
- Department of Orthopedic SurgeryBaylor College of MedicineHoustonTexas
| | - Florent Elefteriou
- Department of Orthopedic SurgeryBaylor College of MedicineHoustonTexas
- Department of Molecular and Human GeneticsBaylor College of MedicineHoustonTexas
| |
Collapse
|
43
|
Chen S, Liu S, Ma K, Zhao L, Lin H, Shao Z. TGF-β signaling in intervertebral disc health and disease. Osteoarthritis Cartilage 2019; 27:1109-1117. [PMID: 31132405 DOI: 10.1016/j.joca.2019.05.005] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 05/02/2019] [Accepted: 05/14/2019] [Indexed: 02/02/2023]
Abstract
OBJECTIVE This paper aims to provide a comprehensive review of the changing role of transforming growth factor-β (TGF-β) signaling in intervertebral disc (IVD) health and disease. METHODS A comprehensive literature search was performed using PubMed terms 'TGF-β' and 'IVD'. RESULTS TGF-β signaling is necessary for the development and growth of IVD, and can play a protective role in the restoration of IVD tissues by stimulating matrix synthesis, inhibiting matrix catabolism, inflammatory response and cell loss. However, excessive activation of TGF-β signaling is detrimental to the IVD, and inhibition of the aberrant TGF-β signaling can delay IVD degeneration. CONCLUSIONS Activation of TGF-β signaling has a promising treatment prospect for IVD degeneration, while excessive activation of TGF-β signaling may contribute to the progression of IVD degeneration. Studies aimed at elucidating the changing role of TGF-β signaling in IVD at different pathophysiological stages and its specific molecular mechanisms are needed, and these studies will contribute to safe and effective TGF-β signaling-based treatments for IVD degeneration.
Collapse
Affiliation(s)
- S Chen
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - S Liu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - K Ma
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - L Zhao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - H Lin
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Z Shao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
44
|
Gorth DJ, Shapiro IM, Risbud MV. A New Understanding of the Role of IL-1 in Age-Related Intervertebral Disc Degeneration in a Murine Model. J Bone Miner Res 2019; 34:1531-1542. [PMID: 30875127 PMCID: PMC6697204 DOI: 10.1002/jbmr.3714] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 03/05/2019] [Accepted: 03/10/2019] [Indexed: 12/25/2022]
Abstract
Increased cytokine expression, in particular interleukin-1β (IL-1β), is considered a hallmark of intervertebral disc degeneration. However, the causative relationship between IL-1 and age-dependent degeneration has not been established. To investigate the role of IL-1 in driving age-related disc degeneration, we studied the spine phenotype of global IL-1α/β double knockout (IL-1KO) mice at 12 and 20 months. Multiplex ELISA analysis of blood revealed significant reductions in the concentrations of IFN-γ, IL-5, IL-15, TNF-α, IP-10, and a trend of reduced concentrations of IL-10, macrophage inflammatory protein 1α (MIP-1α), keratinocyte chemoattractant/human growth-regulated oncogene (KC/GRO), and IL-6. However, the circulating level of MIP-2, a neutrophil chemoattractant, was increased in the IL-1KO. The alterations in systemic cytokine levels coincided with altered bone morphology-IL-1KO mice exhibited significantly thicker caudal cortical bone at 12 and 20 months. Despite these systemic inflammatory and bony changes, IL-1 deletion only minimally affected disc health. Both wild-type (WT) and IL-1KO mice showed age-dependent disc degeneration. Unexpectedly, rather than protecting the animals from degeneration, the aging phenotype was more pronounced in IL-1KO animals: knockout mice evidenced significantly more degenerative changes in the annulus fibrosis (AF) together with alterations in collagen type and maturity. At 20 months, there were no changes in nucleus pulposus (NP) extracellular matrix composition or cellular marker expression; however, the IL-1KO NP cells occupied a smaller proportion of the NP compartment that those of WT controls. Taken together, these results show that IL-1 deletion altered the systemic inflammatory environment and vertebral bone morphology. However, instead of protecting discs from age-related disc degeneration, global IL-1 deletion amplified the degenerative phenotype. © 2019 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Deborah J Gorth
- Department of Orthopaedic Surgery and Graduate Program in Cell Biology and Regenerative Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - Irving M Shapiro
- Department of Orthopaedic Surgery and Graduate Program in Cell Biology and Regenerative Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - Makarand V Risbud
- Department of Orthopaedic Surgery and Graduate Program in Cell Biology and Regenerative Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
45
|
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
|
46
|
Hamilton K, Rebsamen S, Salamat S, Ahmed R. Pediatric extraosseous sacral chordoma: case report and literature review of embryonic derivation and clinical implications. J Neurosurg Pediatr 2019; 23:628-633. [PMID: 30797212 DOI: 10.3171/2018.12.peds18544] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Accepted: 12/04/2018] [Indexed: 01/03/2023]
Abstract
An extraosseous intradural presentation for a sacral chordoma in the pediatric age group has not been reported to date. This is a report on an 11-year-old boy who presented with an extraosseous, intradural sacral chordoma. He underwent gross-total resection and received adjuvant proton beam therapy. Neoplastic transformation of the notochord is reviewed to illustrate the developmental basis for the surgical anatomy and pathogenesis of the classic chordoma variant. Clinical and pathological features are reviewed to differentiate this chordoma presentation from classic osseous chordomas and ecchordosis physaliphora, a related benign developmental notochordal lesion. Finally, the role of developmental signaling in the pathogenesis of chordomas from postembryonic notochordal tissue is discussed.
Collapse
Affiliation(s)
| | | | - Shahriar Salamat
- Departments of1Neurological Surgery
- 3Pathology, University of Wisconsin, Madison, Wisconsin
| | | |
Collapse
|
47
|
Abstract
Development of the axial skeleton is a complex, stepwise process that relies on intricate signaling and coordinated cellular differentiation. Disruptions to this process can result in a myriad of skeletal malformations that range in severity. The notochord and the sclerotome are embryonic tissues that give rise to the major components of the intervertebral discs and the vertebral bodies of the spinal column. Through a number of mouse models and characterization of congenital abnormalities in human patients, various growth factors, transcription factors, and other signaling proteins have been demonstrated to have critical roles in the development of the axial skeleton. Balance between opposing growth factors as well as other environmental cues allows for cell fate specification and divergence of tissue types during development. Furthermore, characterization of progenitor cells for specific cell lineages has furthered the understanding of specific spatiotemporal cues that cells need in order to initiate and complete development of distinct tissues. Identifying specific marker genes that can distinguish between the various embryonic and mature cell types is also of importance. Clinically, understanding developmental clues can aid in the generation of therapeutics for musculoskeletal disease through the process of developmental engineering. Studies into potential stem cell therapies are based on knowledge of the normal processes that occur in the embryo, which can then be applied to stepwise tissue engineering strategies.
Collapse
Affiliation(s)
| | | | - Rosa Serra
- Department of Cell Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, United States.
| |
Collapse
|
48
|
Liu Z, Zheng Z, Qi J, Wang J, Zhou Q, Hu F, Liang J, Li C, Zhang W, Zhang X. CD24 identifies nucleus pulposus progenitors/notochordal cells for disc regeneration. J Biol Eng 2018; 12:35. [PMID: 30598696 PMCID: PMC6303933 DOI: 10.1186/s13036-018-0129-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Accepted: 12/09/2018] [Indexed: 12/19/2022] Open
Abstract
Background Cell-based therapy by transplantation of nucleus pulposus (NP) progenitor/notochordal cells has been proposed as a promising way to halt and reverse the progression of disc degeneration. Although some studies have provided a broad panel of potential markers associated with the phenotype of notochordal cells, suitability of these markers for isolation of notochordal cells for the treatment of disc degeneration is unclear. Results Here, we found that the number of CD24-positive NP cells significantly decreased with increasing severity of disc degeneration. In addition, CD24-positive NP cells were shown to maintain their multipotent differentiation and self-renewal potential in vitro and to abundantly express brachyury, SHH, and GLUT-1, suggesting that CD24-positive NP cells are the progenitor/notochordal cells in the NP. Moreover, our in vivo experiments revealed that transplantation of CD24-positive NP cells enables the recovery of degenerate discs, as evidenced by increased disc height, restored magnetic resonance imaging T2-weighted signal intensity, and NP structure. In terms of the mechanism, HIF-1α-Notch1 pathway activation was essential for the maintenance of CD24-positive NP cells. Conclusion Our studies identify that CD24-positive NP cells are the resident progenitor/notochordal cells in disc regeneration and elucidate a crucial role of HIF-1α-Notch1 pathway in the phenotypic maintenance of CD24-positive NP cells.
Collapse
Affiliation(s)
- Zhuochao Liu
- 1Department of Orthopedics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025 China.,2Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhiyong Zheng
- 3Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Medicine, Jinan, China
| | - Jin Qi
- 1Department of Orthopedics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025 China.,2Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jun Wang
- 1Department of Orthopedics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025 China.,2Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Qi Zhou
- 1Department of Orthopedics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025 China.,2Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Fangqiong Hu
- 1Department of Orthopedics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025 China.,2Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jing Liang
- 1Department of Orthopedics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025 China.,2Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Changwei Li
- 1Department of Orthopedics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025 China.,2Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Weibin Zhang
- 1Department of Orthopedics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025 China.,2Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xingkai Zhang
- 1Department of Orthopedics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025 China.,2Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases with Integrated Chinese-Western Medicine, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
49
|
Role of Sonic Hedgehog Signaling Pathway in Intervertebral Disc Formation and Maintenance. ACTA ACUST UNITED AC 2018; 4:173-179. [PMID: 30687592 DOI: 10.1007/s40610-018-0107-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
a Purpose of Review The intervertebral discs (IVD) are an essential component of the spine. Degeneration of the discs, commonly due to age or injury, is a leading cause of chronic lower back pain. Despite its high prevalence, there is no effective treatment for disc disease due to limited understanding of disc at the cellular and molecular level. b Recent Findings Recent research has demonstrated the importance of the intracellular developmental pathway sonic hedgehog (Shh) during the formation and postnatal maintenance of the IVD. Recent studies corroborate that the down-regulation of SHH expression is associated with pathological changes in the IVDs and demonstrate the reactivation of the hedgehog pathway as a promising avenue for rescuing health disc structure and function. c Summary Understanding the role of developmental signaling pathways that regulate disc formation and maintenance may help develop strategies to recapitulate the same mechanism for disc treatment and hence improve the quality and longevity of patient lives.
Collapse
|
50
|
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
|