1
|
Chen S, Fu J, Long J, Liu C, Ai X, Long D, Leng X, Zhang Y, Liao Z, Li C, Zhou Y, Dong S, Huang B, Feng C. Bulk RNA-seq conjoined with ScRNA-seq analysis reveals the molecular characteristics of nucleus pulposus cell ferroptosis in rat aging intervertebral discs. Arthritis Res Ther 2025; 27:90. [PMID: 40247370 PMCID: PMC12004870 DOI: 10.1186/s13075-025-03550-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 03/27/2025] [Indexed: 04/19/2025] Open
Abstract
OBJECTIVE Recently, several studies have reported that nucleus pulposus (NP) cell ferroptosis plays a key role in IDD. However, the characteristics and molecular mechanisms of cell subsets involved remain unclear. We aimed to define the key factors driving ferroptosis, and the characteristics of ferroptotic NP cells subsets during IDD. METHODS The accumulation of iron ions in NP tissues of rats caudal intervertebral discs (IVDs) was determined by Prussian blue staining. Fluorescent probe Undecanoyl Boron Dipyrromethene (C11-BODIPY) and lipid peroxidation product 4-Hydroxynonenal (4-HNE) staining were performed to assess lipid peroxidation level of NP cells. The differentially expressed genes in NP tissues with aging were overlapped with FerrDB database to screen ferroptosis driving genes associated with aging-related IDD. In addition, single cell sequencing (ScRNA-seq) was used to map the NP cells, and further identify ferroptotic NP cell subsets, as well as their crucial drivers. Finally, cluster analysis was performed to identify the marker genes of ferroptotic NP cells. RESULTS Histological staining showed that, compared with 10 months old (10M-old) group, the accumulation of iron ions increased in NP tissues of 20 months old (20M-old) rats, and the level of lipid peroxidation was also enhanced. 15 ferroptosis driving factors related to IDD were selected by cross-enrichment. ScRNA-seq identified 14 subsets in NP tissue cells, among which the number and ratio of 5 subsets was reduced, and the intracellular ferroptosis related signaling pathways were significantly enriched, accompanied by enhanced cell lipid peroxidation. Notably, ranking the up-regulation fold of ferroptosis related genes, we found Atf3 was always present within TOP2 of these five cell subsets, suggests it is the key driving factor in NP cell ferroptosis. Finally, cluster cross-enrichment and fluorescence colocalization analysis revealed that Rps6 +/Cxcl1- was a common molecular feature among the 5 ferroptotic NP cell subsets. CONCLUSIONS This study reveals that ATF3 is a key driver of NP cell ferroptosis during IDD, and Rps6 +/Cxcl1- is a common molecular feature of ferroptotic NP cell subsets. These findings provide evidence and theoretical support for subsequent targeted intervention of NP cell ferroptosis, as well as provide directions for preventing and delaying IDD.
Collapse
Affiliation(s)
- Shipeng Chen
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, Chongqing, 400037, P. R. China
- Chongqing Municipal Health Commission Key Laboratory of Precise Orthopedics, Chongqing, China
| | - Jiawei Fu
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, Chongqing, 400037, P. R. China
- Chongqing Municipal Health Commission Key Laboratory of Precise Orthopedics, Chongqing, China
| | - Jiang Long
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, Chongqing, 400037, P. R. China
- Chongqing Municipal Health Commission Key Laboratory of Precise Orthopedics, Chongqing, China
| | - Chang Liu
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, Chongqing, 400037, P. R. China
- Chongqing Municipal Health Commission Key Laboratory of Precise Orthopedics, Chongqing, China
| | - Xuezheng Ai
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, Chongqing, 400037, P. R. China
- Chongqing Municipal Health Commission Key Laboratory of Precise Orthopedics, Chongqing, China
| | - Dan Long
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, Chongqing, 400037, P. R. China
- Chongqing Municipal Health Commission Key Laboratory of Precise Orthopedics, Chongqing, China
| | - Xue Leng
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, Chongqing, 400037, P. R. China
- Chongqing Municipal Health Commission Key Laboratory of Precise Orthopedics, Chongqing, China
| | - Yang Zhang
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, Chongqing, 400037, P. R. China
- Chongqing Municipal Health Commission Key Laboratory of Precise Orthopedics, Chongqing, China
| | - Zhengao Liao
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, Chongqing, 400037, P. R. China
- Chongqing Municipal Health Commission Key Laboratory of Precise Orthopedics, Chongqing, China
| | - Changqing Li
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, Chongqing, 400037, P. R. China
- Chongqing Municipal Health Commission Key Laboratory of Precise Orthopedics, Chongqing, China
| | - Yue Zhou
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, Chongqing, 400037, P. R. China
- Chongqing Municipal Health Commission Key Laboratory of Precise Orthopedics, Chongqing, China
| | - Shiwu Dong
- Department of Biomedical Materials Science, School of Biomedical Engineering, Army Medical University, Chongqing, China.
- State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University, Chongqing, China.
| | - Bo Huang
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, Chongqing, 400037, P. R. China.
- Chongqing Municipal Health Commission Key Laboratory of Precise Orthopedics, Chongqing, China.
| | - Chencheng Feng
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, Chongqing, 400037, P. R. China.
- Chongqing Municipal Health Commission Key Laboratory of Precise Orthopedics, Chongqing, China.
| |
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
|
Negrón-Piñeiro LJ, Di Gregorio A. Single-cell Transcriptomic Studies Unveil Potential Nodes of the Notochord Gene Regulatory Network. Integr Comp Biol 2024; 64:1194-1213. [PMID: 38914463 PMCID: PMC11579531 DOI: 10.1093/icb/icae084] [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: 03/31/2024] [Revised: 06/01/2024] [Accepted: 06/16/2024] [Indexed: 06/26/2024] Open
Abstract
Transcription factors (TFs) are DNA-binding proteins able to modulate the timing, location, and levels of gene expression by binding to regulatory DNA regions. Therefore, the repertoire of TFs present in the genome of a multicellular organism and the expression of variable constellations of TFs in different cellular cohorts determine the distinctive characteristics of developing tissues and organs. The information on tissue-specific assortments of TFs, their cross-regulatory interactions, and the genes/regulatory regions targeted by each TF is summarized in gene regulatory networks (GRNs), which provide genetic blueprints for the specification, development, and differentiation of multicellular structures. In this study, we review recent transcriptomic studies focused on the complement of TFs expressed in the notochord, a distinctive feature of all chordates. We analyzed notochord-specific datasets available from organisms representative of the three chordate subphyla, and highlighted lineage-specific variations in the suite of TFs expressed in their notochord. We framed the resulting findings within a provisional evolutionary scenario, which allows the formulation of hypotheses on the genetic/genomic changes that sculpted the structure and function of the notochord on an evolutionary scale.
Collapse
Affiliation(s)
- Lenny J Negrón-Piñeiro
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA
| | - Anna Di Gregorio
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA
| |
Collapse
|
4
|
Zhang Y, Liang C, Xu H, Li Y, Xia K, Wang L, Huang X, Chen J, Shu J, Cheng F, Shi K, Wang J, Tao Y, Wang S, Zhang Y, Li H, Feng S, Li F, Zhou X, Chen Q. Dedifferentiation-like reprogramming of degenerative nucleus pulposus cells into notochordal-like cells by defined factors. Mol Ther 2024; 32:2563-2583. [PMID: 38879755 PMCID: PMC11405157 DOI: 10.1016/j.ymthe.2024.06.018] [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: 02/06/2024] [Revised: 05/09/2024] [Accepted: 06/14/2024] [Indexed: 07/14/2024] Open
Abstract
The extensive degeneration of functional somatic cells and the depletion of endogenous stem/progenitor populations present significant challenges to tissue regeneration in degenerative diseases. Currently, a cellular reprogramming approach enabling directly generating corresponding progenitor populations from degenerative somatic cells remains elusive. The present study focused on intervertebral disc degeneration (IVDD) and identified a three-factor combination (OCT4, FOXA2, TBXT [OFT]) that could induce the dedifferentiation-like reprogramming of degenerative nucleus pulposus cells (dNPCs) toward induced notochordal-like cells (iNCs). Single-cell transcriptomics dissected the transitions of cell identity during reprogramming. Further, OCT4 was found to directly interact with bromodomain PHD-finger transcription factor to remodel the chromatin during the early phases, which was crucial for initiating this dedifferentiation-like reprogramming. In rat models, intradiscal injection of adeno-associated virus carrying OFT generated iNCs from in situ dNPCs and reversed IVDD. These results collectively present a proof-of-concept for dedifferentiation-like reprogramming of degenerated somatic cells into corresponding progenitors through the development of a factor-based strategy, providing a promising approach for regeneration in degenerative disc diseases.
Collapse
Affiliation(s)
- Yuang Zhang
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine; Hangzhou City, Zhejiang Province 310009, China; Orthopedics Research Institute of Zhejiang University; Hangzhou City, Zhejiang Province 310009, China
| | - Chengzhen Liang
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine; Hangzhou City, Zhejiang Province 310009, China; Orthopedics Research Institute of Zhejiang University; Hangzhou City, Zhejiang Province 310009, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province; Hangzhou City, Zhejiang Province 310009, China
| | - Haibin Xu
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine; Hangzhou City, Zhejiang Province 310009, China; Orthopedics Research Institute of Zhejiang University; Hangzhou City, Zhejiang Province 310009, China
| | - Yi Li
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine; Hangzhou City, Zhejiang Province 310009, China; Orthopedics Research Institute of Zhejiang University; Hangzhou City, Zhejiang Province 310009, China
| | - Kaishun Xia
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine; Hangzhou City, Zhejiang Province 310009, China; Orthopedics Research Institute of Zhejiang University; Hangzhou City, Zhejiang Province 310009, China
| | - Liyin Wang
- Department of Ophthalmology and Vision Science, Eye and ENT Hospital, Fudan University; Shanghai 200031, China
| | - Xianpeng Huang
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine; Hangzhou City, Zhejiang Province 310009, China; Orthopedics Research Institute of Zhejiang University; Hangzhou City, Zhejiang Province 310009, China
| | - Jiangjie Chen
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine; Hangzhou City, Zhejiang Province 310009, China; Orthopedics Research Institute of Zhejiang University; Hangzhou City, Zhejiang Province 310009, China
| | - Jiawei Shu
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine; Hangzhou City, Zhejiang Province 310009, China; Orthopedics Research Institute of Zhejiang University; Hangzhou City, Zhejiang Province 310009, China
| | - Feng Cheng
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine; Hangzhou City, Zhejiang Province 310009, China; Orthopedics Research Institute of Zhejiang University; Hangzhou City, Zhejiang Province 310009, China
| | - Kesi Shi
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine; Hangzhou City, Zhejiang Province 310009, China; Orthopedics Research Institute of Zhejiang University; Hangzhou City, Zhejiang Province 310009, China
| | - Jingkai Wang
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine; Hangzhou City, Zhejiang Province 310009, China; Orthopedics Research Institute of Zhejiang University; Hangzhou City, Zhejiang Province 310009, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province; Hangzhou City, Zhejiang Province 310009, China
| | - Yiqing Tao
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine; Hangzhou City, Zhejiang Province 310009, China; Orthopedics Research Institute of Zhejiang University; Hangzhou City, Zhejiang Province 310009, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province; Hangzhou City, Zhejiang Province 310009, China
| | - Shaoke Wang
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine; Hangzhou City, Zhejiang Province 310009, China; Orthopedics Research Institute of Zhejiang University; Hangzhou City, Zhejiang Province 310009, China
| | - Yongxiang Zhang
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine; Hangzhou City, Zhejiang Province 310009, China; Orthopedics Research Institute of Zhejiang University; Hangzhou City, Zhejiang Province 310009, China
| | - Hao Li
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine; Hangzhou City, Zhejiang Province 310009, China; Orthopedics Research Institute of Zhejiang University; Hangzhou City, Zhejiang Province 310009, China
| | - Shoumin Feng
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine; Hangzhou City, Zhejiang Province 310009, China; Orthopedics Research Institute of Zhejiang University; Hangzhou City, Zhejiang Province 310009, China
| | - Fangcai Li
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine; Hangzhou City, Zhejiang Province 310009, China; Orthopedics Research Institute of Zhejiang University; Hangzhou City, Zhejiang Province 310009, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province; Hangzhou City, Zhejiang Province 310009, China.
| | - Xiaopeng Zhou
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine; Hangzhou City, Zhejiang Province 310009, China; Orthopedics Research Institute of Zhejiang University; Hangzhou City, Zhejiang Province 310009, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province; Hangzhou City, Zhejiang Province 310009, China.
| | - Qixin Chen
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine; Hangzhou City, Zhejiang Province 310009, China; Orthopedics Research Institute of Zhejiang University; Hangzhou City, Zhejiang Province 310009, China; Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province; Hangzhou City, Zhejiang Province 310009, China.
| |
Collapse
|
5
|
Negrón-Piñeiro LJ, Wu Y, Popsuj S, José-Edwards DS, Stolfi A, Di Gregorio A. Cis-regulatory interfaces reveal the molecular mechanisms underlying the notochord gene regulatory network of Ciona. Nat Commun 2024; 15:3025. [PMID: 38589372 PMCID: PMC11001920 DOI: 10.1038/s41467-024-46850-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 03/12/2024] [Indexed: 04/10/2024] Open
Abstract
Tissue-specific gene expression is fundamental in development and evolution, and is mediated by transcription factors and by the cis-regulatory regions (enhancers) that they control. Transcription factors and their respective tissue-specific enhancers are essential components of gene regulatory networks responsible for the development of tissues and organs. Although numerous transcription factors have been characterized from different organisms, the knowledge of the enhancers responsible for their tissue-specific expression remains fragmentary. Here we use Ciona to study the enhancers associated with ten transcription factors expressed in the notochord, an evolutionary hallmark of the chordate phylum. Our results illustrate how two evolutionarily conserved transcription factors, Brachyury and Foxa2, coordinate the deployment of other notochord transcription factors. The results of these detailed cis-regulatory analyses delineate a high-resolution view of the essential notochord gene regulatory network of Ciona, and provide a reference for studies of transcription factors, enhancers, and their roles in development, disease, and evolution.
Collapse
Affiliation(s)
- Lenny J Negrón-Piñeiro
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY, 10010, USA
| | - Yushi Wu
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY, 10010, USA
| | - Sydney Popsuj
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Diana S José-Edwards
- Post-Baccalaureate Premedical Program, Washington University, St. Louis, MO, 63130, USA
| | - Alberto Stolfi
- 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.
| |
Collapse
|
6
|
Li Y, Zhang H, Zhu D, Yang F, Wang Z, Wei Z, Yang Z, Jia J, Kang X. Notochordal cells: A potential therapeutic option for intervertebral disc degeneration. Cell Prolif 2024; 57:e13541. [PMID: 37697480 PMCID: PMC10849793 DOI: 10.1111/cpr.13541] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 08/08/2023] [Accepted: 08/21/2023] [Indexed: 09/13/2023] Open
Abstract
Intervertebral disc degeneration (IDD) is a prevalent musculoskeletal degenerative disorder worldwide, and ~40% of chronic low back pain cases are associated with IDD. Although the pathogenesis of IDD remains unclear, the reduction in nucleus pulposus cells (NPCs) and degradation of the extracellular matrix (ECM) are critical factors contributing to IDD. Notochordal cells (NCs), derived from the notochord, which rapidly degrades after birth and is eventually replaced by NPCs, play a crucial role in maintaining ECM homeostasis and preventing NPCs apoptosis. Current treatments for IDD only provide symptomatic relief, while lacking the ability to inhibit or reverse its progression. However, NCs and their secretions possess anti-inflammatory properties and promote NPCs proliferation, leading to ECM formation. Therefore, in recent years, NCs therapy targeting the underlying cause of IDD has emerged as a novel treatment strategy. This article provides a comprehensive review of the latest research progress on NCs for IDD, covering their biological characteristics, specific markers, possible mechanisms involved in IDD and therapeutic effects. It also highlights significant future directions in this field to facilitate further exploration of the pathogenesis of IDD and the development of new therapies based on NCs strategies.
Collapse
Affiliation(s)
- Yanhu Li
- Lanzhou University Second HospitalLanzhouPeople's Republic of China
- Orthopaedics Key Laboratory of Gansu ProvinceLanzhouPeople's Republic of China
| | - Haijun Zhang
- Lanzhou University Second HospitalLanzhouPeople's Republic of China
- Orthopaedics Key Laboratory of Gansu ProvinceLanzhouPeople's Republic of China
- The Second People's Hospital of Gansu ProvinceLanzhouPeople's Republic of China
| | - Daxue Zhu
- Lanzhou University Second HospitalLanzhouPeople's Republic of China
- Orthopaedics Key Laboratory of Gansu ProvinceLanzhouPeople's Republic of China
| | - Fengguang Yang
- Lanzhou University Second HospitalLanzhouPeople's Republic of China
- Orthopaedics Key Laboratory of Gansu ProvinceLanzhouPeople's Republic of China
| | - Zhaoheng Wang
- Lanzhou University Second HospitalLanzhouPeople's Republic of China
- Orthopaedics Key Laboratory of Gansu ProvinceLanzhouPeople's Republic of China
| | - Ziyan Wei
- Lanzhou University Second HospitalLanzhouPeople's Republic of China
- Orthopaedics Key Laboratory of Gansu ProvinceLanzhouPeople's Republic of China
| | - Zhili Yang
- Lanzhou University Second HospitalLanzhouPeople's Republic of China
- Orthopaedics Key Laboratory of Gansu ProvinceLanzhouPeople's Republic of China
| | - Jingwen Jia
- Lanzhou University Second HospitalLanzhouPeople's Republic of China
- Orthopaedics Key Laboratory of Gansu ProvinceLanzhouPeople's Republic of China
| | - Xuewen Kang
- Lanzhou University Second HospitalLanzhouPeople's Republic of China
- Orthopaedics Key Laboratory of Gansu ProvinceLanzhouPeople's Republic of China
| |
Collapse
|
7
|
Mohd Isa IL, Zulkiflee I, Ogaili RH, Mohd Yusoff NH, Sahruddin NN, Sapri SR, Mohd Ramli ES, Fauzi MB, Mokhtar SA. Three-dimensional hydrogel with human Wharton jelly-derived mesenchymal stem cells towards nucleus pulposus niche. Front Bioeng Biotechnol 2023; 11:1296531. [PMID: 38149172 PMCID: PMC10749916 DOI: 10.3389/fbioe.2023.1296531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 11/29/2023] [Indexed: 12/28/2023] Open
Abstract
Introduction: A regenerative strategy employing extracellular matrix (ECM)-based biomaterials and stem cells provide a better approach to mimicking the three-dimensional (3D) microenvironment of intervertebral disc for endogenous tissue regeneration. However, there is currently limited understanding regarding the human Wharton Jelly derived-mesenchymal stem cells (hWJ-MSCs) towards nucleus pulposus (NP)-like cells. Our study focused on the development of 3D bioengineered hydrogel based on the predominant ECM of native NP, including type II collagen (COLII) and hyaluronic acid (HA), which aims to tailor the needs of the microenvironment in NP. Methods: We have fabricated a 3D hydrogel using from COLII enriched with HA by varying the biomacromolecule concentration and characterised it for degradation, stability and swelling properties. The WJ-MSC was then encapsulated in the hydrogel system to guide the cell differentiation into NP-like cells. Results: We successfully fabricated COLII hydrogel (2 mg/ml) and HA 10 mg/ml at a weight ratio of HA and COLII at 1:9 and 4.5:9, and both hydrogels physically maintained their 3D sphere-shaped structure after complete gelation. The higher composition of HA in the hydrogel system indicated a higher water intake capacity in the hydrogel with a higher amount of HA. All hydrogels showed over 60% hydrolytic stability over a month. The hydrogel showed an increase in degradation on day 14. The hWJ-MSCs encapsulated in hydrogel showed a round morphology shape that was homogenously distributed within the hydrogel of both groups. The viability study indicated a higher cell growth of hWJ-MSCs encapsulated in all hydrogel groups until day 14. Discussion: Overall, our findings demonstrate that HA/COLII hydrogel provides an optimal swelling capacity, stability, degradability, and non-cytotoxic, thus mimics the NP microenvironment in guiding hWJ-MSCs towards NP phenotype, which is potentially used as an advanced cell delivery system for intervertebral disc regeneration.
Collapse
Affiliation(s)
- Isma Liza Mohd Isa
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway, Ireland
- School of Medicine, University of Galway, Galway, Ireland
| | - Izzat Zulkiflee
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway, Ireland
| | - Raed H. Ogaili
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Nurul Huda Mohd Yusoff
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Natasya Nadia Sahruddin
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Shaiful Ridzwan Sapri
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Elvy Suhana Mohd Ramli
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Mh Busra Fauzi
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Sabarul Afian Mokhtar
- Department of Orthopaedics and Traumatology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
8
|
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
|
9
|
Damle SR, Krzyzanowska AK, Korsun MK, Morse KW, Gilbert S, Kim HJ, Boachie-Adjei O, Rawlins BA, van der Meulen MCH, Greenblatt MB, Hidaka C, Cunningham ME. Inducing Angiogenesis in the Nucleus Pulposus. Cells 2023; 12:2488. [PMID: 37887332 PMCID: PMC10605635 DOI: 10.3390/cells12202488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/22/2023] [Accepted: 09/26/2023] [Indexed: 10/28/2023] Open
Abstract
Bone morphogenetic protein (BMP) gene delivery to Lewis rat lumbar intervertebral discs (IVDs) drives bone formation anterior and external to the IVD, suggesting the IVD is inhospitable to osteogenesis. This study was designed to determine if IVD destruction with a proteoglycanase, and/or generating an IVD blood supply by gene delivery of an angiogenic growth factor, could render the IVD permissive to intra-discal BMP-driven osteogenesis and fusion. Surgical intra-discal delivery of naïve or gene-programmed cells (BMP2/BMP7 co-expressing or VEGF165 expressing) +/- purified chondroitinase-ABC (chABC) in all permutations was performed between lumbar 4/5 and L5/6 vertebrae, and radiographic, histology, and biomechanics endpoints were collected. Follow-up anti-sFlt Western blotting was performed. BMP and VEGF/BMP treatments had the highest stiffness, bone production and fusion. Bone was induced anterior to the IVD, and was not intra-discal from any treatment. chABC impaired BMP-driven osteogenesis, decreased histological staining for IVD proteoglycans, and made the IVD permissive to angiogenesis. A soluble fragment of VEGF Receptor-1 (sFlt) was liberated from the IVD matrix by incubation with chABC, suggesting dysregulation of the sFlt matrix attachment is a possible mechanism for the chABC-mediated IVD angiogenesis we observed. Based on these results, the IVD can be manipulated to foster vascular invasion, and by extension, possibly osteogenesis.
Collapse
Affiliation(s)
- Sheela R. Damle
- HSS Research Institute, Hospital for Special Surgery, 515 E 71st Street, New York, NY 10021, USA
| | - Agata K. Krzyzanowska
- HSS Research Institute, Hospital for Special Surgery, 515 E 71st Street, New York, NY 10021, USA
| | - Maximilian K. Korsun
- HSS Research Institute, Hospital for Special Surgery, 515 E 71st Street, New York, NY 10021, USA
| | - Kyle W. Morse
- HSS Research Institute, Hospital for Special Surgery, 515 E 71st Street, New York, NY 10021, USA
| | - Susannah Gilbert
- HSS Research Institute, Hospital for Special Surgery, 515 E 71st Street, New York, NY 10021, USA
| | - Han Jo Kim
- HSS Research Institute, Hospital for Special Surgery, 515 E 71st Street, New York, NY 10021, USA
- Weill Cornell Medical College, Cornell University, New York, NY 10065, USA
| | - Oheneba Boachie-Adjei
- HSS Research Institute, Hospital for Special Surgery, 515 E 71st Street, New York, NY 10021, USA
- Weill Cornell Medical College, Cornell University, New York, NY 10065, USA
| | - Bernard A. Rawlins
- HSS Research Institute, Hospital for Special Surgery, 515 E 71st Street, New York, NY 10021, USA
- Weill Cornell Medical College, Cornell University, New York, NY 10065, USA
| | - Marjolein C. H. van der Meulen
- HSS Research Institute, Hospital for Special Surgery, 515 E 71st Street, New York, NY 10021, USA
- Meinig School of Biomedical Engineering and Sibley School of Mechanical & Aerospace Engineering, Cornell University, Ithaca, NY 14853, USA
| | | | - Chisa Hidaka
- HSS Research Institute, Hospital for Special Surgery, 515 E 71st Street, New York, NY 10021, USA
- Department of Genetic Medicine and Belfer Gene Therapy Core Facility, Weill Medical College of Cornell University, New York, NY 10065, USA
| | - Matthew E. Cunningham
- HSS Research Institute, Hospital for Special Surgery, 515 E 71st Street, New York, NY 10021, USA
- Weill Cornell Medical College, Cornell University, New York, NY 10065, USA
| |
Collapse
|
10
|
Lin P, Yan P, Zhu J, Huang S, Wang Z, Hu O, Jin H, Li Y, Zhang L, Zhao J, Chen L, Liu B, He J, Gan Y, Liu P. Spatially multicellular variability of intervertebral disc degeneration by comparative single-cell analysis. Cell Prolif 2023; 56:e13464. [PMID: 37025067 PMCID: PMC10542621 DOI: 10.1111/cpr.13464] [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: 02/14/2023] [Revised: 03/10/2023] [Accepted: 03/20/2023] [Indexed: 04/08/2023] Open
Abstract
Previous studies have revealed cellular heterogeneity in intervertebral discs (IVDs). However, the cellular and molecular alteration patterns of cell populations during degenerative progression remain to be fully elucidated. To illustrate the cellular and molecular alteration of cell populations in intervertebral disc degeneration (IDD), we perform single cell RNA sequencing on cells from four anatomic sites of healthy and degenerative goat IVDs. EGLN3+ StressCs, TGFBR3+ HomCs and GPRC5A+ RegCs exhibit the characteristics associated with resistance to stress, maintaining homeostasis and repairing, respectively. The frequencies and signatures of these cell clusters fluctuate with IDD. Notably, the chondrogenic differentiation programme of PROCR+ progenitor cells is altered by IDD, while notochord cells turn to stemness exhaustion. In addition, we characterise CAV1+ endothelial cells that communicate with chondrocytes through multiple signalling pathways in degenerative IVDs. Our comprehensive analysis identifies the variability of key cell clusters and critical regulatory networks responding to IDD, which will facilitate in-depth investigation of therapeutic strategies for IDD.
Collapse
Affiliation(s)
- Peng Lin
- Department of Spine Surgery, Center of Orthopedics, State Key Laboratory of Trauma, Burns and Combined Injury, Daping HospitalArmy Medical University (Third Military Medical University)Chongqing400042China
| | - Pulin Yan
- Department of Spine Surgery, Center of Orthopedics, State Key Laboratory of Trauma, Burns and Combined Injury, Daping HospitalArmy Medical University (Third Military Medical University)Chongqing400042China
| | - Jun Zhu
- Department of Spine Surgery, Center of Orthopedics, State Key Laboratory of Trauma, Burns and Combined Injury, Daping HospitalArmy Medical University (Third Military Medical University)Chongqing400042China
| | - Sha Huang
- Department of Spine Surgery, Center of Orthopedics, State Key Laboratory of Trauma, Burns and Combined Injury, Daping HospitalArmy Medical University (Third Military Medical University)Chongqing400042China
| | - Zhong Wang
- Department of Spine Surgery, Center of Orthopedics, State Key Laboratory of Trauma, Burns and Combined Injury, Daping HospitalArmy Medical University (Third Military Medical University)Chongqing400042China
| | - Ou Hu
- Department of Spine Surgery, Center of Orthopedics, State Key Laboratory of Trauma, Burns and Combined Injury, Daping HospitalArmy Medical University (Third Military Medical University)Chongqing400042China
| | - Huaijian Jin
- Department of Spine Surgery, Center of Orthopedics, State Key Laboratory of Trauma, Burns and Combined Injury, Daping HospitalArmy Medical University (Third Military Medical University)Chongqing400042China
| | - Yangyang Li
- Department of Spine Surgery, Center of Orthopedics, State Key Laboratory of Trauma, Burns and Combined Injury, Daping HospitalArmy Medical University (Third Military Medical University)Chongqing400042China
| | - Liang Zhang
- Department of Spine Surgery, Center of Orthopedics, State Key Laboratory of Trauma, Burns and Combined Injury, Daping HospitalArmy Medical University (Third Military Medical University)Chongqing400042China
| | - Jianhua Zhao
- Department of Spine Surgery, Center of Orthopedics, State Key Laboratory of Trauma, Burns and Combined Injury, Daping HospitalArmy Medical University (Third Military Medical University)Chongqing400042China
| | - Lin Chen
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Laboratory for the Prevention and Rehabilitation of Military Training Related Injuries, Daping HospitalArmy Medical University (Third Military Medical University)Chongqing400042China
| | - Bing Liu
- State Key Laboratory of Proteomics, Academy of Military Medical SciencesAcademy of Military SciencesBeijing100071China
- State Key Laboratory of Experimental Hematology, Institute of HematologyFifth Medical Center of Chinese PLA General HospitalBeijing100071China
- Key Laboratory for Regenerative Medicine of Ministry of EducationInstitute of Hematology, School of Medicine, Jinan UniversityGuangzhou510632China
- State Key Laboratory of Experimental HematologyInstitute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical SciencesTianjin300020China
| | - Jian He
- Department of Spine Surgery, Center of Orthopedics, State Key Laboratory of Trauma, Burns and Combined Injury, Daping HospitalArmy Medical University (Third Military Medical University)Chongqing400042China
- Laboratory of Basic MedicineThe General Hospital of Western Theater CommandChengdu610031China
| | - Yibo Gan
- Department of Spine Surgery, Center of Orthopedics, State Key Laboratory of Trauma, Burns and Combined Injury, Daping HospitalArmy Medical University (Third Military Medical University)Chongqing400042China
| | - Peng Liu
- Department of Spine Surgery, Center of Orthopedics, State Key Laboratory of Trauma, Burns and Combined Injury, Daping HospitalArmy Medical University (Third Military Medical University)Chongqing400042China
| |
Collapse
|
11
|
Xu H, Li J, Fei Q, Jiang L. Contribution of immune cells to intervertebral disc degeneration and the potential of immunotherapy. Connect Tissue Res 2023; 64:413-427. [PMID: 37161923 DOI: 10.1080/03008207.2023.2212051] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 05/02/2023] [Accepted: 05/03/2023] [Indexed: 05/11/2023]
Abstract
Substantial evidence supports that chronic low back pain is associated with intervertebral disc degeneration (IDD), which is accompanied by decreased cell activity and matrix degradation. The role of immune cells, especially macrophages, in a variety of diseases has been extensively studied; therefore, their role in IDD has naturally attracted widespread scholarly interest. The IVD is considered to be an immunologically-privileged site given the presence of physical and biological barriers that include an avascular microenvironment, a high proteoglycan concentration, high physical pressure, the presence of apoptosis inducers such as Fas ligand, and the presence of notochordal cells. However, during IDD, immune cells with distinct characteristics appear in the IVD. Some of these immune cells release factors that promote the inflammatory response and angiogenesis in the disc and are, therefore, important drivers of IDD. Although some studies have elucidated the role of immune cells, no specific strategies related to systemic immunotherapy have been proposed. Herein, we summarize current knowledge of the presence and role of immune cells in IDD and consider that immunotherapy targeting immune cells may be a novel strategy for alleviating IDD symptoms.
Collapse
Affiliation(s)
- Hao Xu
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Juan Li
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qinming Fei
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Libo Jiang
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, Xiamen, Fujian Province, China
| |
Collapse
|
12
|
Murphy K, Lufkin T, Kraus P. Development and Degeneration of the Intervertebral Disc-Insights from Across Species. Vet Sci 2023; 10:540. [PMID: 37756062 PMCID: PMC10534844 DOI: 10.3390/vetsci10090540] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 08/13/2023] [Accepted: 08/16/2023] [Indexed: 09/28/2023] Open
Abstract
Back pain caused by intervertebral disc (IVD) degeneration has a major socio-economic impact in humans, yet historically has received minimal attention in species other than humans, mice and dogs. However, a general growing interest in this unique organ prompted the expansion of IVD research in rats, rabbits, cats, horses, monkeys, and cows, further illuminating the complex nature of the organ in both healthy and degenerative states. Application of recent biotechnological advancements, including single cell RNA sequencing and complex data analysis methods has begun to explain the shifting inflammatory signaling, variation in cellular subpopulations, differential gene expression, mechanical loading, and metabolic stresses which contribute to age and stress related degeneration of the IVD. This increase in IVD research across species introduces a need for chronicling IVD advancements and tissue biomarkers both within and between species. Here we provide a comprehensive review of recent single cell RNA sequencing data alongside existing case reports and histo/morphological data to highlight the cellular complexity and metabolic challenges of this unique organ that is of structural importance for all vertebrates.
Collapse
Affiliation(s)
| | - Thomas Lufkin
- Department of Biology, Clarkson University, Potsdam, NY 13699, USA;
| | - Petra Kraus
- Department of Biology, Clarkson University, Potsdam, NY 13699, USA;
| |
Collapse
|
13
|
Günay B, Matthews E, Morgan J, Tryfonidou MA, Saldova R, Pandit A. An insight on the N-glycome of notochordal cell-rich porcine nucleus pulposus during maturation. FASEB Bioadv 2023; 5:321-335. [PMID: 37554546 PMCID: PMC10405234 DOI: 10.1096/fba.2023-00011] [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: 02/14/2023] [Revised: 04/25/2023] [Accepted: 05/17/2023] [Indexed: 08/10/2023] Open
Abstract
Degeneration of the intervertebral disc is an age-related condition. It also accompanies the disappearance of the notochordal cells, which are remnants of the developmental stages of the nucleus pulposus (NP). Molecular changes such as extracellular matrix catabolism, cellular phenotype, and glycosaminoglycan loss in the NP have been extensively studied. However, as one of the most significant co- and posttranslational modifications, glycosylation has been overlooked in cells in degeneration. Here, we aim to characterize the N-glycome of young and mature NP and identify patterns related to aging. Accordingly, we isolated N-glycans from notochordal cell-rich NP from porcine discs, characterized them using a combined approach of exoglycosidase digestions and analysis with hydrophilic interaction ultra-performance liquid chromatography and mass spectrometry. We have assigned over 300 individual N-glycans for each age group. Moreover, we observed a notable abundance of antennary structures, galactosylation, fucosylation, and sialylation in both age groups. In addition, as indicated from our results, increasing outer arm fucosylation and decreasing α(2,3)-linked sialylation with aging suggest that these traits are age-dependent. Lastly, we have focused on an extensive characterization of the N-glycome of the notochordal cell-rich NP in aging without inferred degeneration, describing glycosylation changes specific for aging only. Our findings in combination with those of other studies, suggest that the degeneration of the NP does not involve identical processes as aging.
Collapse
Affiliation(s)
- Büşra Günay
- CÚRAM SFI Research Centre for Medical DevicesUniversity of GalwayGalwayIreland
| | - Elizabeth Matthews
- NIBRT GlycoScience GroupNational Institute for Bioprocessing Research and Training (NIBRT)DublinIreland
| | - Jack Morgan
- NIBRT GlycoScience GroupNational Institute for Bioprocessing Research and Training (NIBRT)DublinIreland
| | - Marianna A. Tryfonidou
- Faculty of Veterinary Medicine, Department of Clinical SciencesUtrecht UniversityUtrechtThe Netherlands
| | - Radka Saldova
- NIBRT GlycoScience GroupNational Institute for Bioprocessing Research and Training (NIBRT)DublinIreland
- School of Medicine, College of Health and Agricultural ScienceUniversity College DublinDublinIreland
| | - Abhay Pandit
- CÚRAM SFI Research Centre for Medical DevicesUniversity of GalwayGalwayIreland
| |
Collapse
|
14
|
Rajasekaran S, Soundararajan DCR, Nayagam SM, Tangavel C, Raveendran M, Sri Vijay Anand KS, Shetty AP, Kanna RM. Novel Biomarkers of Health and Degeneration in Human Intervertebral Discs: In-depth Proteomic Analysis of Collagen Framework of Fetal, Healthy, Scoliotic, Degenerate, and Herniated Discs. Asian Spine J 2023; 17:17-29. [PMID: 35421910 PMCID: PMC9977988 DOI: 10.31616/asj.2021.0535] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 02/03/2022] [Indexed: 11/23/2022] Open
Abstract
STUDY DESIGN Profiling proteins expressed in the nucleus pulposus (NP) of intervertebral discs (IVDs) in five different biological states. PURPOSE To evaluate the molecular complexity of the collagen (COL) framework and its role in the health and disease of human IVDs. OVERVIEW OF LITERATURE Changes in COL composition have been linked to degenerative disk disease (DDD). Despite the fact that humans have 28 different types of COLs, most of the literature focuses solely on COL-1 and COL-2. This study used high-end proteomic technology to examine the entire COL composition of the human IVD across fetal (developmental-FD), normal (healthy-ND), scoliotic (early degeneration-SD), herniated (degenerate-DH), and degenerated (DD) disk phenotypes. METHODS Forty NP tissues were snap-frozen in liquid nitrogen (-196°C) immediately before being subjected to proteomic and bioinformatic analyses from five different disk phenotypes (eight each). RESULTS Tandem mass spectrometric analysis revealed a total of 1,050 proteins in FDs, 1,809 in ND, 1,487 in SD, 1,859 in DH, and 1,538 in the DD group. Of 28 major collagens reported in the human body, this study identified 24 different collagens with 34 subtypes in NP. Fibril-forming collagens (COL-1, 2, and 11A1) and fibril-associated collagens with interrupted triple helices (COL-9A1, 12A1, and 14A1) were abundantly expressed in FDs, representing their role in the development of NP. Multiplexin (COL-15), a hybrid proteoglycan-collagen molecule, was discovered only in FDs. Degeneration was associated with COL2A1 downregulation and COL-10A1 upregulation. CONCLUSIONS COL10 was discovered to be a new biomarker for disk degeneration. Besides COL-1 and 2, other important COLs (6, 9, 11, 12, 14, 15) with anabolic potential and abundant expression in the fetal phenotype could be investigated for tissue engineering and novel DDD therapy.
Collapse
Affiliation(s)
| | | | | | | | - Muthuraja Raveendran
- Department of Plant Biotechnology, Tamil Nadu Agricultural University, Coimbatore,
India
| | | | | | | |
Collapse
|
15
|
Jiang W, Glaeser JD, Salehi K, Kaneda G, Mathkar P, Wagner A, Ho R, Sheyn D. Single-cell atlas unveils cellular heterogeneity and novel markers in human neonatal and adult intervertebral discs. iScience 2022; 25:104504. [PMID: 35754733 PMCID: PMC9213722 DOI: 10.1016/j.isci.2022.104504] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 05/03/2022] [Accepted: 05/26/2022] [Indexed: 11/19/2022] Open
Abstract
The origin, composition, distribution, and function of cells in the human intervertebral disc (IVD) have not been fully understood. Here, cell atlases of both human neonatal and adult IVDs have been generated and further assessed by gene ontology pathway enrichment, pseudo-time trajectory, histology, and immunofluorescence. Comparison of cell atlases revealed the presence of two subpopulations of notochordal cells (NCs) and their associated markers in both the neonatal and adult IVDs. Developmental trajectories predicted 7 different cell states that describe the developmental process from neonatal to adult cells in IVD and analyzed the NC’s role in the IVD development. A high heterogeneity and gradual transition of annulus fibrosus cells (AFCs) in the neonatal IVD was detected and their potential relevance in IVD development assessed. Collectively, comparing single-cell atlases between neonatal and adult IVDs delineates the landscape of IVD cell biology and may help discover novel therapeutic targets for IVD degeneration. Compared scRNA-seq between human neonatal and adult IVD Identified two notochordal cell populations in adults and their novel markers Notochordal cells preserved their identity and functions into adulthood Unveiled heterogeneity of nucleus pulposus and annulus fibrosus cells in human IVD
Collapse
Affiliation(s)
- Wensen Jiang
- Orthopaedic Stem Cell Research Laboratory, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Juliane D. Glaeser
- Orthopaedic Stem Cell Research Laboratory, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Orthopedics, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Khosrowdad Salehi
- Orthopaedic Stem Cell Research Laboratory, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Giselle Kaneda
- Orthopaedic Stem Cell Research Laboratory, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Pranav Mathkar
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Anton Wagner
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Ritchie Ho
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Center for Neural Sciences and Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Dmitriy Sheyn
- Orthopaedic Stem Cell Research Laboratory, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Orthopedics, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Corresponding author
| |
Collapse
|
16
|
Mohd Isa IL, Mokhtar SA, Abbah SA, Fauzi MB, Devitt A, Pandit A. Intervertebral Disc Degeneration: Biomaterials and Tissue Engineering Strategies toward Precision Medicine. Adv Healthc Mater 2022; 11:e2102530. [PMID: 35373924 PMCID: PMC11469247 DOI: 10.1002/adhm.202102530] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 03/01/2022] [Indexed: 12/22/2022]
Abstract
Intervertebral disc degeneration is a common cause of discogenic low back pain resulting in significant disability. Current conservative or surgical intervention treatments do not reverse the underlying disc degeneration or regenerate the disc. Biomaterial-based tissue engineering strategies exhibit the potential to regenerate the disc due to their capacity to modulate local tissue responses, maintain the disc phenotype, attain biochemical homeostasis, promote anatomical tissue repair, and provide functional mechanical support. Despite preliminary positive results in preclinical models, these approaches have limited success in clinical trials as they fail to address discogenic pain. This review gives insights into the understanding of intervertebral disc pathology, the emerging concept of precision medicine, and the rationale of personalized biomaterial-based tissue engineering tailored to the severity of the disease targeting early, mild, or severe degeneration, thereby enhancing the efficacy of the treatment for disc regeneration and ultimately to alleviate discogenic pain. Further research is required to assess the relationship between disc degeneration and lower back pain for developing future clinically relevant therapeutic interventions targeted towards the subgroup of degenerative disc disease patients.
Collapse
Affiliation(s)
- Isma Liza Mohd Isa
- Department of AnatomyFaculty of MedicineUniversiti Kebangsaan MalaysiaKuala Lumpur56000Malaysia
- CÚRAMSFI Research Centre for Medical DevicesNational University of IrelandGalwayH91W2TYIreland
| | - Sabarul Afian Mokhtar
- Department of Orthopaedics and TraumatologyFaculty of MedicineUniversiti Kebangsaan MalaysiaKuala Lumpur56000Malaysia
| | - Sunny A. Abbah
- CÚRAMSFI Research Centre for Medical DevicesNational University of IrelandGalwayH91W2TYIreland
| | - Mh Busra Fauzi
- Centre for Tissue Engineering and Regenerative MedicineFaculty of MedicineUniversiti Kebangsaan MalaysiaKuala Lumpur56000Malaysia
| | - Aiden Devitt
- CÚRAMSFI Research Centre for Medical DevicesNational University of IrelandGalwayH91W2TYIreland
- Department of Orthopedic SurgeryUniversity Hospital GalwayGalwayH91YR71Ireland
| | - Abhay Pandit
- CÚRAMSFI Research Centre for Medical DevicesNational University of IrelandGalwayH91W2TYIreland
| |
Collapse
|
17
|
Importance of Matrix Cues on Intervertebral Disc Development, Degeneration, and Regeneration. Int J Mol Sci 2022; 23:ijms23136915. [PMID: 35805921 PMCID: PMC9266338 DOI: 10.3390/ijms23136915] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 06/17/2022] [Accepted: 06/20/2022] [Indexed: 01/25/2023] Open
Abstract
Back pain is one of the leading causes of disability worldwide and is frequently caused by degeneration of the intervertebral discs. The discs’ development, homeostasis, and degeneration are driven by a complex series of biochemical and physical extracellular matrix cues produced by and transmitted to native cells. Thus, understanding the roles of different cues is essential for designing effective cellular and regenerative therapies. Omics technologies have helped identify many new matrix cues; however, comparatively few matrix molecules have thus far been incorporated into tissue engineered models. These include collagen type I and type II, laminins, glycosaminoglycans, and their biomimetic analogues. Modern biofabrication techniques, such as 3D bioprinting, are also enabling the spatial patterning of matrix molecules and growth factors to direct regional effects. These techniques should now be applied to biochemically, physically, and structurally relevant disc models incorporating disc and stem cells to investigate the drivers of healthy cell phenotype and differentiation. Such research will inform the development of efficacious regenerative therapies and improved clinical outcomes.
Collapse
|
18
|
Fan Y, Zhao L, Lai Y, Lu K, Huang J. CRISPR-Cas9-mediated loss of function of β-catenin attenuates intervertebral disc degeneration. MOLECULAR THERAPY - NUCLEIC ACIDS 2022; 28:387-396. [PMID: 35505959 PMCID: PMC9035381 DOI: 10.1016/j.omtn.2022.03.024] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 03/27/2022] [Indexed: 11/06/2022]
Abstract
Intervertebral disc degeneration is a very common medical condition causing pain and disability, and it cannot be reversed by available treatment options. Here we report that targeting β-catenin, a pivotal factor associated with disc degeneration, ameliorates disc degeneration in a mouse model of disc injury. Degenerative changes in the disc in response to disc injury include decompression of nucleus pulposus (NP), replacement of notochordal cells in the NP by chondrocyte-like cells, and disorganization of annulus fibrosus (AF). Importantly, downregulation of β-catenin through intradiscal injection of CRISPR-Cas9-expressing adeno-associated virus significantly mitigated all these pathological changes, by preserving notochordal cells and attenuating chondro-osteogenesis in the NP, as well as maintaining the AF structure. Moreover, β-catenin loss-of-function decelerated the rapid induction of catabolic reactions in disc matrix and attenuated pain-related neural events during disc degeneration. Thus, our data demonstrate that targeting β-catenin in disc cells through CRISPR-Cas9 has multifaceted therapeutic effects on disc degeneration, and we suggest that β-catenin plays a fundamental role in the remodeling and degenerative processes of the disc. In addition, this study proposes that CRISPR-Cas9 is a useful tool for identifying new drug targets and developing therapeutic strategies for disc degeneration.
Collapse
|
19
|
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
|
20
|
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
|
21
|
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
|
22
|
Li W, Zhang S, Wang D, Zhang H, Shi Q, Zhang Y, Wang M, Ding Z, Xu S, Gao B, Yan M. Exosomes Immunity Strategy: A Novel Approach for Ameliorating Intervertebral Disc Degeneration. Front Cell Dev Biol 2022; 9:822149. [PMID: 35223870 PMCID: PMC8870130 DOI: 10.3389/fcell.2021.822149] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 12/21/2021] [Indexed: 12/11/2022] Open
Abstract
Low back pain (LBP), which is one of the most severe medical and social problems globally, has affected nearly 80% of the population worldwide, and intervertebral disc degeneration (IDD) is a common musculoskeletal disorder that happens to be the primary trigger of LBP. The pathology of IDD is based on the impaired homeostasis of catabolism and anabolism in the extracellular matrix (ECM), uncontrolled activation of immunologic cascades, dysfunction, and loss of nucleus pulposus (NP) cells in addition to dynamic cellular and biochemical alterations in the microenvironment of intervertebral disc (IVD). Currently, the main therapeutic approach regarding IDD is surgical intervention, but it could not considerably cure IDD. Exosomes, extracellular vesicles with a diameter of 30–150 nm, are secreted by various kinds of cell types like stem cells, tumor cells, immune cells, and endothelial cells; the lipid bilayer of the exosomes protects them from ribonuclease degradation and helps improve their biological efficiency in recipient cells. Increasing lines of evidence have reported the promising applications of exosomes in immunological diseases, and regarded exosomes as a potential therapeutic source for IDD. This review focuses on clarifying novel therapies based on exosomes derived from different cell sources and the essential roles of exosomes in regulating IDD, especially the immunologic strategy.
Collapse
Affiliation(s)
- Weihang Li
- Department of Orthopedic Surgery, Xijing Hospital, Air Force Medical University, Xi’an, China
| | - Shilei Zhang
- Department of Orthopedic Surgery, Xijing Hospital, Air Force Medical University, Xi’an, China
| | - Dong Wang
- Department of Orthopedic Surgery, Xijing Hospital, Air Force Medical University, Xi’an, China
- Department of Orthopaedics, Affiliated Hospital of Yanan University, Yanan, China
| | - Huan Zhang
- Department of Orthopedic Surgery, Xijing Hospital, Air Force Medical University, Xi’an, China
| | - Quan Shi
- Department of Orthopedic Surgery, Xijing Hospital, Air Force Medical University, Xi’an, China
| | - Yuyuan Zhang
- Department of Critical Care Medicine, Xijing Hospital, Air Force Medical University, Xi’an, China
| | - Mo Wang
- The First Brigade of Basic Medical College, Air Force Military Medical University, Xi’an, China
| | - Ziyi Ding
- Department of Orthopedic Surgery, Xijing Hospital, Air Force Medical University, Xi’an, China
| | - Songjie Xu
- Beijing Luhe Hospital, Capital Medical University, Beijing, China
- *Correspondence: Songjie Xu, ; Bo Gao, ; Ming Yan,
| | - Bo Gao
- Department of Orthopedic Surgery, Xijing Hospital, Air Force Medical University, Xi’an, China
- *Correspondence: Songjie Xu, ; Bo Gao, ; Ming Yan,
| | - Ming Yan
- Department of Orthopedic Surgery, Xijing Hospital, Air Force Medical University, Xi’an, China
- *Correspondence: Songjie Xu, ; Bo Gao, ; Ming Yan,
| |
Collapse
|
23
|
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
|
24
|
Gan Y, He J, Zhu J, Xu Z, Wang Z, Yan J, Hu O, Bai Z, Chen L, Xie Y, Jin M, Huang S, Liu B, Liu P. Spatially defined single-cell transcriptional profiling characterizes diverse chondrocyte subtypes and nucleus pulposus progenitors in human intervertebral discs. Bone Res 2021; 9:37. [PMID: 34400611 PMCID: PMC8368097 DOI: 10.1038/s41413-021-00163-z] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 04/30/2021] [Accepted: 06/10/2021] [Indexed: 02/07/2023] Open
Abstract
A comprehensive understanding of the cellular heterogeneity and molecular mechanisms underlying the development, homeostasis, and disease of human intervertebral disks (IVDs) remains challenging. Here, the transcriptomic landscape of 108 108 IVD cells was mapped using single-cell RNA sequencing of three main compartments from young and adult healthy IVDs, including the nucleus pulposus (NP), annulus fibrosus, and cartilage endplate (CEP). The chondrocyte subclusters were classified based on their potential regulatory, homeostatic, and effector functions in extracellular matrix (ECM) homeostasis. Notably, in the NP, a PROCR+ resident progenitor population showed enriched colony-forming unit-fibroblast (CFU-F) activity and trilineage differentiation capacity. Finally, intercellular crosstalk based on signaling network analysis uncovered that the PDGF and TGF-β cascades are important cues in the NP microenvironment. In conclusion, a single-cell transcriptomic atlas that resolves spatially regulated cellular heterogeneity together with the critical signaling that underlies homeostasis will help to establish new therapeutic strategies for IVD degeneration in the clinic.
Collapse
Affiliation(s)
- Yibo Gan
- grid.410570.70000 0004 1760 6682Department of Spine Surgery, Center of Orthopedics, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China ,grid.410570.70000 0004 1760 6682State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University (Third Military Medical University), Chongqing, China
| | - Jian He
- grid.410740.60000 0004 1803 4911State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China
| | - Jun Zhu
- grid.410570.70000 0004 1760 6682Department of Spine Surgery, Center of Orthopedics, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Zhengyang Xu
- grid.410740.60000 0004 1803 4911State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China
| | - Zhong Wang
- grid.410570.70000 0004 1760 6682Department of Spine Surgery, Center of Orthopedics, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Jing Yan
- grid.410740.60000 0004 1803 4911State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China
| | - Ou Hu
- grid.410570.70000 0004 1760 6682Department of Spine Surgery, Center of Orthopedics, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Zhijie Bai
- grid.410740.60000 0004 1803 4911State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China
| | - Lin Chen
- grid.410570.70000 0004 1760 6682Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Laboratory for the Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yangli Xie
- grid.410570.70000 0004 1760 6682Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Laboratory for the Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Min Jin
- grid.410570.70000 0004 1760 6682Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Laboratory for the Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Shuo Huang
- grid.410570.70000 0004 1760 6682Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Laboratory for the Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Bing Liu
- grid.410740.60000 0004 1803 4911State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China ,grid.11135.370000 0001 2256 9319State Key Laboratory of Experimental Hematology, Institute of Hematology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China ,grid.258164.c0000 0004 1790 3548Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, China
| | - Peng Liu
- grid.410570.70000 0004 1760 6682Department of Spine Surgery, Center of Orthopedics, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China ,grid.410570.70000 0004 1760 6682State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University (Third Military Medical University), Chongqing, China
| |
Collapse
|
25
|
Gan Y, He J, Zhu J, Xu Z, Wang Z, Yan J, Hu O, Bai Z, Chen L, Xie Y, Jin M, Huang S, Liu B, Liu P. Spatially defined single-cell transcriptional profiling characterizes diverse chondrocyte subtypes and nucleus pulposus progenitors in human intervertebral discs. Bone Res 2021; 9:37. [PMID: 34400611 PMCID: PMC8368097 DOI: 10.1038/s41413-021-00163-z+10.1038/s41413-021-00163-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 04/30/2021] [Accepted: 06/10/2021] [Indexed: 01/21/2024] Open
Abstract
A comprehensive understanding of the cellular heterogeneity and molecular mechanisms underlying the development, homeostasis, and disease of human intervertebral disks (IVDs) remains challenging. Here, the transcriptomic landscape of 108 108 IVD cells was mapped using single-cell RNA sequencing of three main compartments from young and adult healthy IVDs, including the nucleus pulposus (NP), annulus fibrosus, and cartilage endplate (CEP). The chondrocyte subclusters were classified based on their potential regulatory, homeostatic, and effector functions in extracellular matrix (ECM) homeostasis. Notably, in the NP, a PROCR+ resident progenitor population showed enriched colony-forming unit-fibroblast (CFU-F) activity and trilineage differentiation capacity. Finally, intercellular crosstalk based on signaling network analysis uncovered that the PDGF and TGF-β cascades are important cues in the NP microenvironment. In conclusion, a single-cell transcriptomic atlas that resolves spatially regulated cellular heterogeneity together with the critical signaling that underlies homeostasis will help to establish new therapeutic strategies for IVD degeneration in the clinic.
Collapse
Affiliation(s)
- Yibo Gan
- Department of Spine Surgery, Center of Orthopedics, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University (Third Military Medical University), Chongqing, China
| | - Jian He
- State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China
| | - Jun Zhu
- Department of Spine Surgery, Center of Orthopedics, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Zhengyang Xu
- State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China
| | - Zhong Wang
- Department of Spine Surgery, Center of Orthopedics, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Jing Yan
- State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China
| | - Ou Hu
- Department of Spine Surgery, Center of Orthopedics, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Zhijie Bai
- State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China
| | - Lin Chen
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Laboratory for the Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yangli Xie
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Laboratory for the Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Min Jin
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Laboratory for the Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Shuo Huang
- Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Laboratory for the Prevention and Rehabilitation of Military Training Related Injuries, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Bing Liu
- State Key Laboratory of Proteomics, Academy of Military Medical Sciences, Academy of Military Sciences, Beijing, China.
- State Key Laboratory of Experimental Hematology, Institute of Hematology, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China.
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, China.
| | - Peng Liu
- Department of Spine Surgery, Center of Orthopedics, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, China.
- State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University (Third Military Medical University), Chongqing, China.
| |
Collapse
|
26
|
Spatially defined single-cell transcriptional profiling characterizes diverse chondrocyte subtypes and nucleus pulposus progenitors in human intervertebral discs. Bone Res 2021; 9:37. [PMID: 34400611 PMCID: PMC8368097 DOI: 10.1038/s41413-021-00163-z 10.1038/s41413-021-00163-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2023] Open
Abstract
A comprehensive understanding of the cellular heterogeneity and molecular mechanisms underlying the development, homeostasis, and disease of human intervertebral disks (IVDs) remains challenging. Here, the transcriptomic landscape of 108 108 IVD cells was mapped using single-cell RNA sequencing of three main compartments from young and adult healthy IVDs, including the nucleus pulposus (NP), annulus fibrosus, and cartilage endplate (CEP). The chondrocyte subclusters were classified based on their potential regulatory, homeostatic, and effector functions in extracellular matrix (ECM) homeostasis. Notably, in the NP, a PROCR+ resident progenitor population showed enriched colony-forming unit-fibroblast (CFU-F) activity and trilineage differentiation capacity. Finally, intercellular crosstalk based on signaling network analysis uncovered that the PDGF and TGF-β cascades are important cues in the NP microenvironment. In conclusion, a single-cell transcriptomic atlas that resolves spatially regulated cellular heterogeneity together with the critical signaling that underlies homeostasis will help to establish new therapeutic strategies for IVD degeneration in the clinic.
Collapse
|
27
|
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
|
28
|
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
|
29
|
Amorim JP, Gali-Macedo A, Marcelino H, Bordeira-Carriço R, Naranjo S, Rivero-Gil S, Teixeira J, Galhardo M, Marques J, Bessa J. A Conserved Notochord Enhancer Controls Pancreas Development in Vertebrates. Cell Rep 2021; 32:107862. [PMID: 32640228 PMCID: PMC7355232 DOI: 10.1016/j.celrep.2020.107862] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 04/06/2020] [Accepted: 06/09/2020] [Indexed: 12/31/2022] Open
Abstract
The notochord is an evolutionary novelty in vertebrates that functions as an important signaling center during development. Notochord ablation in chicken has demonstrated that it is crucial for pancreas development; however, the molecular mechanism has not been fully described. Here, we show that in zebrafish, the loss of function of nog2, a Bmp antagonist expressed in the notochord, impairs β cell differentiation, compatible with the antagonistic role of Bmp in β cell differentiation. In addition, we show that nog2 expression in the notochord is induced by at least one notochord enhancer and its loss of function reduces the number of pancreatic progenitors and impairs β cell differentiation. Tracing Nog2 diffusion, we show that Nog2 emanates from the notochord to the pancreas progenitor domain. Finally, we find a notochord enhancer in human and mice Nog genomic landscapes, suggesting that the acquisition of a Nog notochord enhancer occurred early in the vertebrate phylogeny and contributes to the development of complex organs like the pancreas.
Collapse
Affiliation(s)
- João Pedro Amorim
- i3S (Instituto de Investigação e Inovação em Saúde), Universidade do Porto, Porto, Portugal; IBMC (Instituto de Biologia Molecular e Celular), Universidade do Porto, Porto, Portugal
| | - Ana Gali-Macedo
- i3S (Instituto de Investigação e Inovação em Saúde), Universidade do Porto, Porto, Portugal; IBMC (Instituto de Biologia Molecular e Celular), Universidade do Porto, Porto, Portugal
| | - Hugo Marcelino
- i3S (Instituto de Investigação e Inovação em Saúde), Universidade do Porto, Porto, Portugal; IBMC (Instituto de Biologia Molecular e Celular), Universidade do Porto, Porto, Portugal
| | - Renata Bordeira-Carriço
- i3S (Instituto de Investigação e Inovação em Saúde), Universidade do Porto, Porto, Portugal; IBMC (Instituto de Biologia Molecular e Celular), Universidade do Porto, Porto, Portugal
| | - Silvia Naranjo
- CABD (Centro Andaluz de Biología del Desarrollo), Universidad Pablo de Olavide, Seville, Spain
| | - Solangel Rivero-Gil
- CABD (Centro Andaluz de Biología del Desarrollo), Universidad Pablo de Olavide, Seville, Spain
| | - Joana Teixeira
- i3S (Instituto de Investigação e Inovação em Saúde), Universidade do Porto, Porto, Portugal; IBMC (Instituto de Biologia Molecular e Celular), Universidade do Porto, Porto, Portugal
| | - Mafalda Galhardo
- i3S (Instituto de Investigação e Inovação em Saúde), Universidade do Porto, Porto, Portugal; CIBIO (Centro de Investigação em Biodiversidade e Recursos Genéticos), Universidade do Porto, Vairão, Portugal
| | - Joana Marques
- i3S (Instituto de Investigação e Inovação em Saúde), Universidade do Porto, Porto, Portugal; IBMC (Instituto de Biologia Molecular e Celular), Universidade do Porto, Porto, Portugal
| | - José Bessa
- i3S (Instituto de Investigação e Inovação em Saúde), Universidade do Porto, Porto, Portugal; IBMC (Instituto de Biologia Molecular e Celular), Universidade do Porto, Porto, Portugal.
| |
Collapse
|
30
|
|
31
|
Zhang Y, Zhang Z, Chen P, Ma CY, Li C, Au TYK, Tam V, Peng Y, Wu R, Cheung KMC, Sham PC, Tse HF, Chan D, Leung VY, Cheah KSE, Lian Q. Directed Differentiation of Notochord-like and Nucleus Pulposus-like Cells Using Human Pluripotent Stem Cells. Cell Rep 2021; 30:2791-2806.e5. [PMID: 32101752 DOI: 10.1016/j.celrep.2020.01.100] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 10/15/2019] [Accepted: 01/28/2020] [Indexed: 12/26/2022] Open
Abstract
Intervertebral disc degeneration might be amenable to stem cell therapy, but the required cells are scarce. Here, we report the development of a protocol for directed in vitro differentiation of human pluripotent stem cells (hPSCs) into notochord-like and nucleus pulposus (NP)-like cells of the disc. The first step combines enhancement of ACTIVIN/NODAL and WNT and inhibition of BMP pathways. By day 5 of differentiation, hPSC-derived cells express notochordal cell characteristic genes. After activating the TGF-β pathway for an additional 15 days, qPCR, immunostaining, and transcriptome data show that a wide array of NP markers are expressed. Transcriptomically, the in vitro-derived cells become more like in vivo adolescent human NP cells, driven by a set of influential genes enriched with motifs bound by BRACHYURY and FOXA2, consistent with an NP cell-like identity. Transplantation of these NP-like cells attenuates fibrotic changes in a rat disc injury model of disc degeneration.
Collapse
Affiliation(s)
- Yuelin Zhang
- Department of Medicine, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong; Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, Guangzhou, Guangdong 510080, China
| | - Zhao Zhang
- Department of Medicine, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong; Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, Guangzhou, Guangdong 510080, China
| | - Peikai Chen
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong
| | - Chui Yan Ma
- Department of Medicine, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong
| | - Cheng Li
- Department of Medicine, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong
| | - Tiffany Y K Au
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong
| | - Vivian Tam
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong
| | - Yan Peng
- Department of Orthopaedics and Traumatology, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong
| | - Ron Wu
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong
| | - Kenneth Man Chee Cheung
- Department of Orthopaedics and Traumatology, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong
| | - Pak C Sham
- Centre for PanorOmic Sciences, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong
| | - Hung-Fat Tse
- Department of Medicine, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong
| | - Danny Chan
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong
| | - Victor Y Leung
- Department of Orthopaedics and Traumatology, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong
| | - Kathryn S E Cheah
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong.
| | - Qizhou Lian
- Department of Medicine, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong; Guangzhou Women and Children's Medical Centre, Guangzhou Medical University, Guangzhou, Guangdong 510080, China; The State Key Laboratory of Pharmaceutical Biotechnology, the University of Hong Kong, Hong Kong.
| |
Collapse
|
32
|
Guerrero J, Häckel S, Croft AS, Albers CE, Gantenbein B. The effects of 3D culture on the expansion and maintenance of nucleus pulposus progenitor cell multipotency. JOR Spine 2021; 4:e1131. [PMID: 33778405 PMCID: PMC7984018 DOI: 10.1002/jsp2.1131] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 10/29/2020] [Accepted: 11/04/2020] [Indexed: 02/06/2023] Open
Abstract
INTRODUCTION Low back pain (LBP) is a global health concern. Increasing evidence implicates intervertebral disk (IVD) degeneration as a major contributor. In this respect, tissue-specific progenitors may play a crucial role in tissue regeneration, as these cells are perfectly adapted to their niche. Recently, a novel progenitor cell population was described in the nucleus pulposus (NP) that is positive for Tie2 marker. These cells have self-renewal capacity and in vitro multipotency potential. However, extremely low numbers of the NP progenitors limit the feasibility of cell therapy strategies. OBJECTIVE Here, we studied the influence of the culture method and of the microenvironment on the proliferation rate and the differentiation potential of human NP progenitors in vitro. METHOD Cells were obtained from human NP tissue from trauma patients. Briefly, the NP tissue cells were cultured in two-dimensional (2D) (monolayer) or three-dimensional (3D) (alginate beads) conditions. After 1 week, cells from 2D or 3D culture were expanded on fibronectin-coated flasks. Subsequently, expanded NP cells were then characterized by cytometry and tri-lineage differentiation, which was analyzed by qPCR and histology. Moreover, experiments using Tie2+ and Tie2- NP cells were also performed. RESULTS The present study aims to demonstrate that 3D expansion of NP cells better preserves the Tie2+ cell populations and increases the chondrogenic and osteogenic differentiation potential compared to 2D expansion. Moreover, the cell sorting experiments reveal that only Tie2+ cells were able to maintain the pluripotent gene expression if cultured in 3D within alginate beads. Therefore, our results highly suggest that the maintenance of the cell's multipotency is mainly, but not exclusively, due to the higher presence of Tie2+ cells due to 3D culture. CONCLUSION This project not only might have a scientific impact by evaluating the influence of a two-step expansion protocol on the functionality of NP progenitors, but it could also lead to an innovative clinical approach.
Collapse
Affiliation(s)
- Julien Guerrero
- Tissue Engineering for Orthopaedics & Mechanobiology, Department for BioMedical Research (DBMR) of the Faculty of Medicine of the University of BernUniversity of BernSwitzerland
| | - Sonja Häckel
- Department of Orthopaedic Surgery & Traumatology, InselspitalBern University HospitalBernSwitzerland
| | - Andreas S. Croft
- Tissue Engineering for Orthopaedics & Mechanobiology, Department for BioMedical Research (DBMR) of the Faculty of Medicine of the University of BernUniversity of BernSwitzerland
| | - Christoph E. Albers
- Department of Orthopaedic Surgery & Traumatology, InselspitalBern University HospitalBernSwitzerland
| | - Benjamin Gantenbein
- Tissue Engineering for Orthopaedics & Mechanobiology, Department for BioMedical Research (DBMR) of the Faculty of Medicine of the University of BernUniversity of BernSwitzerland
- Department of Orthopaedic Surgery & Traumatology, InselspitalBern University HospitalBernSwitzerland
| |
Collapse
|
33
|
Fearing BV, Speer JE, Jing L, Kalathil A, P. Kelly M, M. Buchowski J, P. Zebala L, Luhmann S, C. Gupta M, A. Setton L. Verteporfin treatment controls morphology, phenotype, and global gene expression for cells of the human nucleus pulposus. JOR Spine 2020; 3:e1111. [PMID: 33392449 PMCID: PMC7770208 DOI: 10.1002/jsp2.1111] [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: 02/14/2020] [Revised: 06/24/2020] [Accepted: 07/02/2020] [Indexed: 12/15/2022] Open
Abstract
Cells of the nucleus pulposus (NP) are essential contributors to extracellular matrix synthesis and function of the intervertebral disc. With age and degeneration, the NP becomes stiffer and more dehydrated, which is associated with a loss of phenotype and biosynthetic function for its resident NP cells. Also, with aging, the NP cell undergoes substantial morphological changes from a rounded shape with pronounced vacuoles in the neonate and juvenile, to one that is more flattened and spread with a loss of vacuoles. Here, we make use of the clinically relevant pharmacological treatment verteporfin (VP), previously identified as a disruptor of yes-associated protein-TEA domain family member-binding domain (TEAD) signaling, to promote morphological changes in adult human NP cells in order to study variations in gene expression related to differences in cell shape. Treatment of adult, degenerative human NP cells with VP caused a shift in morphology from a spread, fibroblastic-like shape to a rounded, clustered morphology with decreased transcriptional activity of TEAD and serum-response factor. These changes were accompanied by an increased expression of vacuoles, NP-specific gene markers, and biosynthetic activity. The contemporaneous observation of VP-induced changes in cell shape and prominent, time-dependent changes within the transcriptome of NP cells occurred over all timepoints in culture. Enriched gene sets with the transition to VP-induced cell rounding suggest a major role for cell adhesion, cytoskeletal remodeling, vacuolar lumen, and MAPK activity in the NP phenotypic and functional response to changes in cell shape.
Collapse
Affiliation(s)
- Bailey V. Fearing
- Department of Biomedical EngineeringWashington University in St. LouisSt. LouisMissouriUSA
- Department of Orthopaedic SurgeryAtrium Health Musculoskeletal InstituteCharlotteNorth CarolinaUSA
| | - Julie E. Speer
- Department of Biomedical EngineeringWashington University in St. LouisSt. LouisMissouriUSA
| | - Liufang Jing
- Department of Biomedical EngineeringWashington University in St. LouisSt. LouisMissouriUSA
| | - Aravind Kalathil
- Department of Biomedical EngineeringWashington University in St. LouisSt. LouisMissouriUSA
| | - Michael P. Kelly
- Department of Orthopaedic SurgeryWashington University in St. LouisSt. LouisMissouriUSA
| | - Jacob M. Buchowski
- Department of Orthopaedic SurgeryWashington University in St. LouisSt. LouisMissouriUSA
| | - Lukas P. Zebala
- Department of Orthopaedic SurgeryWashington University in St. LouisSt. LouisMissouriUSA
| | - Scott Luhmann
- Department of Orthopaedic SurgeryWashington University in St. LouisSt. LouisMissouriUSA
| | - Munish C. Gupta
- Department of Orthopaedic SurgeryWashington University in St. LouisSt. LouisMissouriUSA
| | - Lori A. Setton
- Department of Biomedical EngineeringWashington University in St. LouisSt. LouisMissouriUSA
- Department of Orthopaedic SurgeryWashington University in St. LouisSt. LouisMissouriUSA
| |
Collapse
|
34
|
Notochordal-Cell-Derived Exosomes Induced by Compressive Load Inhibit Angiogenesis via the miR-140-5p/Wnt/β-Catenin Axis. MOLECULAR THERAPY. NUCLEIC ACIDS 2020; 22:1092-1106. [PMID: 33294295 PMCID: PMC7691158 DOI: 10.1016/j.omtn.2020.10.021] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 10/13/2020] [Indexed: 12/16/2022]
Abstract
Angiogenesis is a pathological signature of intervertebral disc degeneration (IDD). Accumulating evidence has shown that notochordal cells (NCs) play an essential role in maintaining intervertebral disc development and homeostasis with inhibitive effect on blood vessel in-growth. However, the anti-angiogenesis mechanism of NCs is still unclear. In the current study, we, for the first time, isolated NC-derived exosomes (NC-exos) and showed their increased concentration following compressive load cultures. We further found that NC-exos from 0.5 MPa compressive load cultures (0.5 MPa/NC-exos) inhibit angiogenesis via transferring high expressed microRNA (miR)-140-5p to endothelial cells and regulating the downstream Wnt/β-catenin pathway. Clinical evidence showed that exosomal miR-140-5p expression of the nucleus pulposus is negatively correlated with angiogenesis in IDD. Finally, 0.5 MPa/NC-exos were demonstrated to have a therapeutical impact on the degenerated disc with an anti-angiogenesis effect in an IDD model. Consequently, our present findings provide insights into the anti-angiogenesis mechanism of NC-exos, indicating their therapeutic potential for IDD.
Collapse
|
35
|
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
|
36
|
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
|
37
|
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
|
38
|
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
|
39
|
Ashraf S, Chatoor K, Chong J, Pilliar R, Santerre P, Kandel R. Transforming Growth Factor β Enhances Tissue Formation by Passaged Nucleus Pulposus Cells In Vitro. J Orthop Res 2020; 38:438-449. [PMID: 31529713 DOI: 10.1002/jor.24476] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 09/10/2019] [Indexed: 02/04/2023]
Abstract
The nucleus pulposus (NP) is composed of NP and notochord cell. It is a paucicellular tissue and if it is to be used as a source of cells for tissue engineering the cell number will have to be expanded by cell passaging. The hypothesis of this study is that passaged NP and notochordal cells grown in three-dimensional (3D) culture in the presence of transforming growth factor β (TGFβ) will show enhanced NP tissue formation compared with cells grown in the absence of this growth factor. Bovine NP cells isolated by sequential enzymatic digestion from caudal intervertebral discs were either placed directly in 3D culture (P0) or serially passaged up to passage 3 (P3) prior to placement in 3D culture. Serial cell passage in monolayer culture led to de-differentiation, increased senescence and oxidative stress and decreases in the gene expression of NP and notochordal associated markers and increases in de-differentiation markers. The NP tissue regeneration capacity of cells in 3D culture decreases with passaging as indicated by diminished tissue thickness and total collagen content when compared with tissues formed by P0 cells. Immunohistochemical studies showed that type II collagen accumulation appeared to decrease. TGFβ1 or TGFβ3 treatment enhanced the ability of cells at each passage to form tissue, in part by decreasing cell death. However, neither TGFβ1 nor TGFβ3 were able to restore the notochordal phenotype. Although TGFβ1/3 recovered NP tissue formation by passaged cells, to generate NP in vitro that resembles the native tissue will require identification of conditions facilitating retention of notochordal cell differentiation. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 38:438-449, 2020.
Collapse
Affiliation(s)
- Sajjad Ashraf
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada
| | - Kenny Chatoor
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada.,Institute of Biomaterials and Biomedical Engineering, University of Toronto, Ontario, Canada
| | - Jasmine Chong
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada.,Institute of Biomaterials and Biomedical Engineering, University of Toronto, Ontario, Canada
| | - Robert Pilliar
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Ontario, Canada
| | - Paul Santerre
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Ontario, Canada
| | - Rita Kandel
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Ontario, Canada.,Pathology and Laboratory Medicine, Sinai Health System and Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
40
|
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
|
41
|
Borem R, Madeline A, Bowman M, Gill S, Tokish J, Mercuri J. Differential Effector Response of Amnion- and Adipose-Derived Mesenchymal Stem Cells to Inflammation; Implications for Intradiscal Therapy. J Orthop Res 2019; 37:2445-2456. [PMID: 31287173 DOI: 10.1002/jor.24412] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 06/25/2019] [Indexed: 02/04/2023]
Abstract
Intervertebral disc degeneration (IVDD) is a progressive condition marked by tissue destruction and inflammation. The therapeutic effector functions of mesenchymal stem cells (MSCs) makes them an attractive therapy for patients with IVDD. While several sources of MSCs exist, the optimal choice for use in the inflamed IVD remains a significant question. Adipose (AD)- and amnion (AM)-derived MSCs have several advantages compared with other sources, however, no study has directly compared the impact of IVDD inflammation on their effector functions. Human MSCs were cultured in media with or without supplementation of interleukin-1β (IL-1β) and tumor necrosis factor-α at concentrations reportedly produced by IVDD cells. MSC proliferation and production of pro- and anti-inflammatory cytokines were quantified following 24 and 48 h of culture. Additionally, the osteogenic and chondrogenic potential of AD- and AM-MSCs was characterized via histology and biochemical analysis following 28 days of culture. In inflammatory culture, AM-MSCs produced significantly more anti-inflammatory IL-10 (14.47 ± 2.39 pg/ml; p = 0.004) and larger chondrogenic pellets (5.67 ± 0.26 mm2 ; p = 0.04) with greater percent area staining positively for glycosaminoglycan (82.03 ± 3.26%; p < 0.001) compared with AD-MSCs (0.00 ± 0.00 pg/ml; 2.76 ± 0.18 mm2 ; 34.75 ± 2.49%; respectively). Conversely, AD-MSCs proliferated more resulting in higher cell numbers (221,000 ± 8,021 cells; p = 0.048) and produced higher concentrations of pro-inflammatory cytokines prostaglandin E2 (1,118.30 ± 115.56 pg/ml; p = 0.030) and IL-1β (185.40 ± 7.63 pg/ml; p = 0.010) compared with AM-MSCs (109,667 ± 5,696 cells; 1,291.40 ± 78.47 pg/ml; 144.10 ± 4.57 pg/ml; respectively). AD-MSCs produced more mineralized extracellular matrix (3.34 ± 0.05 relative absorbance units [RAU]; p < 0.001) compared with AM-MSCs (1.08 ± 0.06 RAU). Under identical inflammatory conditions, a different effector response was observed with AM-MSCs producing more anti-inflammatories and demonstrating enhanced chondrogenesis compared with AD-MSCs, which produced more pro-inflammatory cytokines and demonstrated enhanced osteogenesis. These findings may begin to help inform researchers which MSC source may be optimal for IVD regeneration. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 37:2445-2456, 2019.
Collapse
Affiliation(s)
- Ryan Borem
- Department of Bioengineering, Laboratory of Orthopaedic Tissue Regeneration & Orthobiologics, Clemson University, Clemson, South Carolina, 29634
| | - Allison Madeline
- Department of Bioengineering, Laboratory of Orthopaedic Tissue Regeneration & Orthobiologics, Clemson University, Clemson, South Carolina, 29634
| | - Mackenzie Bowman
- Department of Bioengineering, Laboratory of Orthopaedic Tissue Regeneration & Orthobiologics, Clemson University, Clemson, South Carolina, 29634
| | - Sanjitpal Gill
- Department of Bioengineering, Laboratory of Orthopaedic Tissue Regeneration & Orthobiologics, Clemson University, Clemson, South Carolina, 29634.,Department of Orthopaedic Surgery, Medical Group of the Carolinas-Pelham, Spartanburg Regional Healthcare System, Greer, South Carolina, 29651
| | - John Tokish
- Department of Bioengineering, Laboratory of Orthopaedic Tissue Regeneration & Orthobiologics, Clemson University, Clemson, South Carolina, 29634.,Department of Orthopaedic Surgery, Mayo Clinic, Phoenix, Arizona, 85054
| | - Jeremy Mercuri
- Department of Bioengineering, Laboratory of Orthopaedic Tissue Regeneration & Orthobiologics, Clemson University, Clemson, South Carolina, 29634
| |
Collapse
|
42
|
Frauchiger DA, Tekari A, May RD, Džafo E, Chan SCW, Stoyanov J, Bertolo A, Zhang X, Guerrero J, Sakai D, Schol J, Grad S, Tryfonidou M, Benneker LM, Gantenbein B. Fluorescence-Activated Cell Sorting Is More Potent to Fish Intervertebral Disk Progenitor Cells Than Magnetic and Beads-Based Methods. Tissue Eng Part C Methods 2019; 25:571-580. [PMID: 31154900 DOI: 10.1089/ten.tec.2018.0375] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Low back pain related to intervertebral disk (IVD) degeneration has a major socioeconomic impact on our aging society. Therefore, stem cell therapy to activate self-repair of the IVD remains an exciting treatment strategy. In this respect, tissue-specific progenitors may play a crucial role in IVD regeneration, as these cells are perfectly adapted to this niche. Such a rare progenitor cell population residing in the nucleus pulposus (NP) (NP progenitor cells [NPPCs]) was found positive for the angiopoietin-1 receptor (Tie2+), and was demonstrated to possess self-renewal capacity and in vitro multipotency. Here, we compared three sorting protocols; that is, fluorescence-activated cell sorting (FACS), magnetic-activated cell sorting (MACS), and a mesh-based label-free cell sorting system (pluriSelect), with respect to cell yield, potential to form colonies (colony-forming units), and in vitro functional differentiation assays for tripotency. The aim of this study was to demonstrate the efficiency of three widespread cell sorting methods for picking rare cells (<5%) and how these isolated cells then behave in downstream functional differentiation in adipogenesis, osteogenesis, and chondrogenesis. The cell yields among the isolation methods differed widely, with FACS presenting the highest yield (5.0% ± 4.0%), followed by MACS (1.6% ± 2.9%) and pluriSelect (1.1% ± 1.0%). The number of colonies formed was not significantly different between Tie2+ and Tie2- NPPCs. Only FACS was able to separate into two functionally different populations that showed trilineage multipotency, while MACS and pluriSelect failed to maintain a clear separation between Tie2+ and Tie2- populations in differentiation assays. To conclude, the isolation of NPPCs was possible with all three sorting methods, while FACS was the preferred technique for separation of functional Tie2+ cells. Impact Statement Tissue-specific progenitor cells such as nucleus pulposus progenitor cells of the IVD could become an ultimate cell source for tissue engineering strategies as these cells are presumably best adapted to the tissue's microenvironment. Fluorescence-activated cell sorting seemed to outcompete magnetic-activated cell sorting and pluriSelect concerning selecting a rare cell population from IVD tissue as could be demonstrated by improved cell yield and functional differentiation assays.
Collapse
Affiliation(s)
- Daniela A Frauchiger
- Tissue Engineering, Orthopeadic Research & Mechanobiology, Department for BioMedical Research (DBMR), Medical Faculty, University of Bern, Bern, Switzerland
| | - Adel Tekari
- Laboratory of Molecular and Cellular Screening Processes, Centre of Biotechnology of Sfax, University of Sfax, Sfax, Tunisia
| | - Rahel D May
- Tissue Engineering, Orthopeadic Research & Mechanobiology, Department for BioMedical Research (DBMR), Medical Faculty, University of Bern, Bern, Switzerland
| | - Emina Džafo
- Tissue Engineering, Orthopeadic Research & Mechanobiology, Department for BioMedical Research (DBMR), Medical Faculty, University of Bern, Bern, Switzerland
| | - Samantha C W Chan
- Tissue Engineering, Orthopeadic Research & Mechanobiology, Department for BioMedical Research (DBMR), Medical Faculty, University of Bern, Bern, Switzerland
| | | | | | - Xingshuo Zhang
- Tissue Engineering, Orthopeadic Research & Mechanobiology, Department for BioMedical Research (DBMR), Medical Faculty, University of Bern, Bern, Switzerland
| | - Julien Guerrero
- Tissue Engineering, Orthopeadic Research & Mechanobiology, Department for BioMedical Research (DBMR), Medical Faculty, University of Bern, Bern, Switzerland.,Department of Orthopaedic Surgery and Traumatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Daisuke Sakai
- Department for Orthopaedic Surgery, Tokai University School of Medicine, Isehara, Japan
| | - Jordy Schol
- Department for Orthopaedic Surgery, Tokai University School of Medicine, Isehara, Japan
| | | | - Marianna Tryfonidou
- Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Lorin M Benneker
- Department of Orthopaedic Surgery and Traumatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Benjamin Gantenbein
- Tissue Engineering, Orthopeadic Research & Mechanobiology, Department for BioMedical Research (DBMR), Medical Faculty, University of Bern, Bern, Switzerland.,Department of Orthopaedic Surgery and Traumatology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
43
|
D'Agati G, Cabello EM, Frontzek K, Rushing EJ, Klemm R, Robinson MD, White RM, Mosimann C, Burger A. Active receptor tyrosine kinases, but not Brachyury, are sufficient to trigger chordoma in zebrafish. Dis Model Mech 2019; 12:dmm.039545. [PMID: 31221659 PMCID: PMC6679381 DOI: 10.1242/dmm.039545] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Accepted: 06/13/2019] [Indexed: 01/09/2023] Open
Abstract
The aberrant activation of developmental processes triggers diverse cancer types. Chordoma is a rare, aggressive tumor arising from transformed notochord remnants. Several potentially oncogenic factors have been found to be deregulated in chordoma, yet causation remains uncertain. In particular, sustained expression of TBXT – encoding the notochord regulator protein brachyury – is hypothesized as a key driver of chordoma, yet experimental evidence is absent. Here, we employ a zebrafish chordoma model to identify the notochord-transforming potential of implicated genes in vivo. We find that Brachyury, including a form with augmented transcriptional activity, is insufficient to initiate notochord hyperplasia. In contrast, the chordoma-implicated receptor tyrosine kinases (RTKs) EGFR and Kdr/VEGFR2 are sufficient to transform notochord cells. Aberrant activation of RTK/Ras signaling attenuates processes required for notochord differentiation, including the unfolded protein response and endoplasmic reticulum stress pathways. Our results provide the first in vivo evidence against a tumor-initiating potential of Brachyury in the notochord, and imply activated RTK signaling as a possible initiating event in chordoma. Furthermore, our work points at modulating endoplasmic reticulum and protein stress pathways as possible therapeutic avenues against chordoma. Summary: An injection-based chordoma model in zebrafish shows that the hypothesized chordoma oncogene brachyury is insufficient, whereas EGFR and VEGFR2 are sufficient, to trigger notochord hyperplasia in our model.
Collapse
Affiliation(s)
- Gianluca D'Agati
- Institute of Molecular Life Sciences, University of Zürich, 8057 Zürich, Switzerland
| | - Elena María Cabello
- Institute of Molecular Life Sciences, University of Zürich, 8057 Zürich, Switzerland
| | - Karl Frontzek
- Institute of Neuropathology, University Hospital Zürich, 8091 Zürich, Switzerland
| | - Elisabeth J Rushing
- Institute of Neuropathology, University Hospital Zürich, 8091 Zürich, Switzerland
| | - Robin Klemm
- Institute of Molecular Life Sciences, University of Zürich, 8057 Zürich, Switzerland
| | - Mark D Robinson
- Institute of Molecular Life Sciences, University of Zürich, 8057 Zürich, Switzerland.,SIB Swiss Institute of Bioinformatics, University of Zürich, 8057 Zürich, Switzerland
| | - Richard M White
- Cancer Biology & Genetics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.,Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Christian Mosimann
- Institute of Molecular Life Sciences, University of Zürich, 8057 Zürich, Switzerland
| | - Alexa Burger
- Institute of Molecular Life Sciences, University of Zürich, 8057 Zürich, Switzerland
| |
Collapse
|
44
|
Quantitative Single-Cell Transcript Assessment of Biomarkers Supports Cellular Heterogeneity in the Bovine IVD. Vet Sci 2019; 6:vetsci6020042. [PMID: 31083612 PMCID: PMC6631975 DOI: 10.3390/vetsci6020042] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 05/05/2019] [Accepted: 05/09/2019] [Indexed: 02/06/2023] Open
Abstract
Severe and chronic low back pain is often associated with intervertebral disc (IVD) degeneration. While imposing a considerable socio-economic burden worldwide, IVD degeneration is also severely impacting on the quality of life of affected individuals. Cell-based regenerative medicine approaches have moved into clinical trials, yet IVD cell identities in the mature disc remain to be fully elucidated and tissue heterogeneity exists, requiring a better characterization of IVD cells. The bovine coccygeal IVD is an accepted research model to study IVD mechano-biology and disc homeostasis. Recently, we identified novel IVD biomarkers in the outer annulus fibrosus (AF) and nucleus pulposus (NP) of the mature bovine coccygeal IVD through RNA in situ hybridization (AP-RISH) and z-proportion test. Here we follow up on Lam1, Thy1, Gli1, Gli3, Noto, Ptprc, Scx, Sox2 and Zscan10 with fluorescent RNA in situ hybridization (FL-RISH) and confocal microscopy. This permits sub-cellular transcript localization and the addition of quantitative single-cell derived values of mRNA expression levels to our previous analysis. Lastly, we used a Gaussian mixture modeling approach for the exploratory analysis of IVD cells. This work complements our earlier cell population proportion-based study, confirms the previously proposed biomarkers and indicates even further heterogeneity of cells in the outer AF and NP of a mature IVD.
Collapse
|
45
|
Qiu X, Zhuang M, Lu Z, Liu Z, Cheng D, Zhu C, Liu J. RIPK1 suppresses apoptosis mediated by TNF and caspase-3 in intervertebral discs. J Transl Med 2019; 17:135. [PMID: 31029152 PMCID: PMC6487042 DOI: 10.1186/s12967-019-1886-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 04/16/2019] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Low back pain has become a serious social and economic burden and the leading cause of disability worldwide. Among a variety of pathophysiological triggers, intervertebral disc (IVD) degeneration plays a primary underlying role in causing such pain. Specifically, multiple independent endplate changes have been implicated in the initiation and progression of IVD degeneration. METHODS In this study, we built a signaling network comprising both well-characterized IVD pathology-associated proteins as well as some potentially correlated proteins that have been associated with one or more of the currently known pathology-associated proteins. We then screened for the potential IVD degeneration-associated proteins using patients' normal and degenerative endplate specimens. Short hairpin RNAs for receptor interacting serine/threonine kinase 1 (RIPK1) were constructed to examine the effects of RIPK1 knockdown in primary chondrocyte cells and in animal models of caudal vertebra intervertebral disc degeneration in vivo. RESULTS RIPK1 was identified as a potential IVD degeneration-associated protein based on IVD pathology-associated signaling networks and the patients' degenerated endplate specimens. Construction of the short hairpin RNAs was successful, with short-term RIPK1 knockdown triggering inflammation in the primary chondrocytes, while long-term knockdown triggered apoptosis through cleavage of the caspase 3 pathway, down-regulated NF-κB and mitogen-activating protein kinase (MAPK)s cascades, and decreased cell survival and inflammation. Animal models of caudal vertebra intervertebral disc degeneration further demonstrated that apoptosis was induced by up-regulation of tumor necrosis factor (TNF) accompanied by down-regulation of NF-κB and MAPKs cascades that are dependent on caspase and RIPK1. CONCLUSIONS These results provide proof-of-concept for developing novel therapies to combat IVD degeneration through interfering with RIPK1-mediated apoptosis signaling pathways especially in patients with RIPK1 abnormality.
Collapse
Affiliation(s)
- Xubin Qiu
- Department of Spine, The Third Affiliated Hospital of Soochow University, 185 Juqian Street, Tianning District, Changzhou, 213003 Jiangsu China
| | - Ming Zhuang
- Department of Spine, The Third Affiliated Hospital of Soochow University, 185 Juqian Street, Tianning District, Changzhou, 213003 Jiangsu China
| | - Ziwen Lu
- School of Pharmacy, Jiangsu University, Zhenjiang, 212013 Jiangsu China
| | - Zhiwei Liu
- Department of Spine, The Third Affiliated Hospital of Soochow University, 185 Juqian Street, Tianning District, Changzhou, 213003 Jiangsu China
| | - Dong Cheng
- Department of Spine, The Third Affiliated Hospital of Soochow University, 185 Juqian Street, Tianning District, Changzhou, 213003 Jiangsu China
| | - Chenlei Zhu
- Department of Spine, The Third Affiliated Hospital of Soochow University, 185 Juqian Street, Tianning District, Changzhou, 213003 Jiangsu China
| | - Jinbo Liu
- Department of Spine, The Third Affiliated Hospital of Soochow University, 185 Juqian Street, Tianning District, Changzhou, 213003 Jiangsu China
| |
Collapse
|
46
|
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
|