51
|
Ashraf S, Santerre P, Kandel R. Induced senescence of healthy nucleus pulposus cells is mediated by paracrine signaling from TNF-α-activated cells. FASEB J 2021; 35:e21795. [PMID: 34403508 DOI: 10.1096/fj.202002201r] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 05/27/2021] [Accepted: 06/28/2021] [Indexed: 12/25/2022]
Abstract
Intervertebral disc degeneration is an irreversible process associated with accumulation of senescent nucleus pulposus (NP) cells. This study investigates the hypothesis that Tumor necrosis factor-α (TNF-α)-treated senescent NP cells propagate senescence of neighboring healthy cells via a paracrine effect that involves p-Stat3 signaling and the cytokine interleukin-6 (IL-6). NP cells isolated from bovine caudal intervertebral disc (IVD) were treated with TNF-α to induce senescence which was confirmed by demonstrating upregulation of senescence-associated β-galactosidase and p16. This was correlated with downregulation of NP-associated markers, Aggrecan, Col2A1, and Sox9. Direct contact and non-contact co-culture of healthy and senescent cells showed that TNF-α-treated cells increased the senescence in healthy cells via a paracrine effect. The senescent cells have a secretory phenotype as indicated by increased gene and protein levels of IL-6. Phosphorylated Signal Transducer and Activator of Transcription 3 (pStat3) levels were also high in treated cells and appeared to upregulate IL-6 as inhibition of Stat3 phosphorylation by StatticV downregulated IL-6 mRNA expression in cells and protein levels in the culture media. All trans retinoic acid, an IL-6 inhibitor, also decreased the secretion of IL-6 and reduced the paracrine effect of senescent cells on healthy cells. Decreased pStat3 levels and inhibition of IL-6 secretion did not fully restore NP gene expression of Col2A1 but importantly, appeared to cause senescent cells to undergo apoptosis and cell death. This study demonstrated the paracrine effect of senescent NP cells which involves Stat3 and IL-6 and may explain why senescent NP cells accumulate in IVD with age. The role of pSTAT3 and IL-6 in mediating NP senescence requires further study as it may be a novel strategy for modulating the senescent-inducing effects of TNF-α.
Collapse
Affiliation(s)
- Sajjad Ashraf
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
| | - Paul Santerre
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, ON, Canada
| | - Rita Kandel
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada.,Institute of Biomaterials and Biomedical Engineering, University of Toronto, ON, Canada.,Pathology and Laboratory Medicine, Sinai Health System, Toronto, ON, Canada.,Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
52
|
Chen H, Liu O, Chen S, Zhou Y. Aging and Mesenchymal Stem Cells: Therapeutic Opportunities and Challenges in the Older Group. Gerontology 2021; 68:339-352. [PMID: 34161948 DOI: 10.1159/000516668] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 04/07/2021] [Indexed: 11/19/2022] Open
Abstract
With aging, a portion of cells, including mesenchymal stem cells (MSCs), become senescent, and these senescent cells accumulate and promote various age-related diseases. Therefore, the older age group has become a major population for MSC therapy, which is aimed at improving tissue regeneration and function of the aged body. However, the application of MSC therapy is often unsatisfying in the aged group. One reasonable conjecture for this correlation is that aging microenvironment reduces the number and function of MSCs. Cellular senescence also plays an important role in MSC function impairment. Thus, it is necessary to explore the relationship between senescence and MSCs for improving the application of MSCs in the elderly. Here, we present the influence of aging on MSCs and the characteristics and functional changes of senescent MSCs. Furthermore, current therapeutic strategies for improving MSC therapy in the elderly group are also discussed.
Collapse
Affiliation(s)
- Huan Chen
- Hunan Key Laboratory of Oral Health Research, Hunan 3D Printing Engineering Research Center of Oral Care, Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital, and Xiangya School of Stomatology, Central South University, Changsha, China
| | - Ousheng Liu
- Hunan Key Laboratory of Oral Health Research, Hunan 3D Printing Engineering Research Center of Oral Care, Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital, and Xiangya School of Stomatology, Central South University, Changsha, China
| | - Sijia Chen
- Hunan Key Laboratory of Oral Health Research, Hunan 3D Printing Engineering Research Center of Oral Care, Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital, and Xiangya School of Stomatology, Central South University, Changsha, China
| | - Yueying Zhou
- Hunan Key Laboratory of Oral Health Research, Hunan 3D Printing Engineering Research Center of Oral Care, Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital, and Xiangya School of Stomatology, Central South University, Changsha, China
| |
Collapse
|
53
|
Wang XS, Yang JM, Ding RJ, Liu XZ, Jiang XB, Yang ZJ, Ling ZM, Hu TX, Wei FX. Fabrication of a Polylactide-Glycolide/Poly-ε-Caprolactone/Dextran/Plastrum Testudinis Extract Composite Anti-Inflammation Nanofiber Membrane via Electrospinning for Annulus Fibrosus Regeneration. J Biomed Nanotechnol 2021; 17:873-888. [PMID: 34082873 DOI: 10.1166/jbn.2021.3070] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Tissue engineering is a promising approach for the treatment of chronic lower back pain (LBP) caused by intervertebral disc degeneration (IDD) resulting from degeneration and inflammation of annulus fibrosus (AF) tissue. However, scaffold with an anti-inflammatory effect on AF cells has not been reported. In this study, we fabricated a polylactide-glycolide (PLGA)/poly-ε-caprolactone (PCL)Zdextran (DEX) composite membrane loaded with plastrum testudinis extract (PTE), a Traditional Chinese Medicine herbal extract, via electrospinning. The membranes were characterized by mechanical measurements and scanning electron microscopy (SEM). Using an in vitro inflammation model induced by interleukin (IL)-1β, the cytocompatibility and anti-inflammatory effects of the composites were investigated by CCK-8 assay and flow cytometry. Potential regulatory mechanisms were examined by RT-qPCR and Western blotting. The results showed that the P10P8D2 (PLGA 10 g, PCL 8 g, DEX 2 g) composite nanofiber membrane exhibited the most uniform diameter distribution, best mechanical properties, a moderate degradation rate, and the best cytocompatibility characteristics. The optimal concentration of PTE was 120 µg/mL. Importantly, P10P8D2 combined with PTE exhibited anti-inflammatory and cell proliferation promotion effects. Moreover, the NF-κBB/NLRP3/IL-β signaling pathway was inactivated. Our findings suggested that the nanofiber membrane composed of P10P8D2 and PTE has anti-inflammatory and pro-proliferation effects on AF cells. It may provide an effective strategy for AF tissue regeneration.
Collapse
Affiliation(s)
- Xiao-Shuai Wang
- Department of Orthopaedics, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, P. R. China
| | - Jia-Ming Yang
- Department of Orthopaedics, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, P. R. China
| | - Ren-Jie Ding
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510030, P. R. China
| | - Xi-Zhe Liu
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510030, P. R. China.,Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology Orthopaedic Research Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510030, P. R. China
| | - Xiao-Bing Jiang
- Department of Spinal Surgery, The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510080, P. R. China
| | - Zhi-Jian Yang
- Department of Joint Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510030, P. R. China
| | - Ze-Min Ling
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510030, P. R. China.,Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology Orthopaedic Research Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510030, P. R. China
| | - Tian-Xue Hu
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510030, P. R. China
| | - Fu-Xin Wei
- Department of Orthopaedics, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, P. R. China
| |
Collapse
|
54
|
Zheng J, Chang L, Bao X, Zhang X, Li C, Deng L. TRIM21 drives intervertebral disc degeneration induced by oxidative stress via mediating HIF-1α degradation. Biochem Biophys Res Commun 2021; 555:46-53. [PMID: 33813275 DOI: 10.1016/j.bbrc.2021.03.088] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 03/16/2021] [Indexed: 12/20/2022]
Abstract
The onset and progression of intervertebral disc degeneration (IVDD) is strictly associated with oxidative stress. TRIM21 (Tripartite motif-containing protein 21), a ubiquitin E3 ligase, has been shown to play an essential role in liver redox homeostasis; however, whether TRIM21 is involved in IVDD, especially in oxidative stress-induced IVDD, is unknown. Here, we reported that TRIM21 was upregulated in nucleus pulposus (NPs) with increasing severity of IVDD, and that oxidative stress was a stimulator of TRIM21 expression. Furthermore, we found that TRIM21 deficiency significantly protected NP cells from degeneration induced by oxidative stress as well as ameliorated disc degeneration in aged mice. Mechanistically, TRIM21 facilitated NP cells degeneration induced by oxidative stress via HIF-1α. TRIM21 could physically interact with HIF-1α and facilitated its degradation via its ubiquitylating activity. Taken together, these findings revealed that TRIM21 drived IVDD induced by oxidative stress by increasing HIF-1α degradation. These findings implicates the potential of TRIM21 as a therapeutic target in IVDD, especially in oxidative stress-induced IVDD.
Collapse
Affiliation(s)
- Jiancheng Zheng
- Department of Orthopedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Leilei Chang
- Department of Orthopedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xiaogang Bao
- Department of Orthopedic Surgery, Spine Center, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Xingkai Zhang
- Department of Orthopedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| | - Changwei Li
- Department of Orthopedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| | - Lianfu Deng
- Department of Orthopedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopedics, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
55
|
Leon KE, Tangudu NK, Aird KM, Buj R. Loss of p16: A Bouncer of the Immunological Surveillance? Life (Basel) 2021; 11:309. [PMID: 33918220 PMCID: PMC8065641 DOI: 10.3390/life11040309] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 03/26/2021] [Accepted: 03/30/2021] [Indexed: 02/07/2023] Open
Abstract
p16INK4A (hereafter called p16) is an important tumor suppressor protein frequently suppressed in human cancer and highly upregulated in many types of senescence. Although its role as a cell cycle regulator is very well delineated, little is known about its other non-cell cycle-related roles. Importantly, recent correlative studies suggest that p16 may be a regulator of tissue immunological surveillance through the transcriptional regulation of different chemokines, interleukins and other factors secreted as part of the senescence-associated secretory phenotype (SASP). Here, we summarize the current evidence supporting the hypothesis that p16 is a regulator of tumor immunity.
Collapse
Affiliation(s)
- Kelly E. Leon
- UPMC Hillman Cancer Center, Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (K.E.L.); (N.K.T.); (K.M.A.)
- Biomedical Sciences Graduate Program, Penn State College of Medicine, Hershey, PA 15213, USA
| | - Naveen Kumar Tangudu
- UPMC Hillman Cancer Center, Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (K.E.L.); (N.K.T.); (K.M.A.)
| | - Katherine M. Aird
- UPMC Hillman Cancer Center, Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (K.E.L.); (N.K.T.); (K.M.A.)
| | - Raquel Buj
- UPMC Hillman Cancer Center, Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (K.E.L.); (N.K.T.); (K.M.A.)
| |
Collapse
|
56
|
Novais EJ, Choi H, Madhu V, Suyama K, Anjo SI, Manadas B, Shapiro IM, Salgado AJ, Risbud MV. Hypoxia and Hypoxia-Inducible Factor-1α Regulate Endoplasmic Reticulum Stress in Nucleus Pulposus Cells: Implications of Endoplasmic Reticulum Stress for Extracellular Matrix Secretion. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 191:487-502. [PMID: 33307037 PMCID: PMC7927276 DOI: 10.1016/j.ajpath.2020.11.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 11/03/2020] [Accepted: 11/24/2020] [Indexed: 12/20/2022]
Abstract
Endoplasmic reticulum (ER) stress is shown to promote nucleus pulposus (NP) cell apoptosis and intervertebral disc degeneration. However, little is known about ER stress regulation by the hypoxic disc microenvironment and its contribution to extracellular matrix homeostasis. NP cells were cultured under hypoxia (1% partial pressure of oxygen) to assess ER stress status, and gain-of-function and loss-of-function approaches were used to assess the role of hypoxia-inducible factor (HIF)-1α in this pathway. In addition, the contribution of ER stress induction on the NP cell secretome was assessed by a nontargeted quantitative proteomic analysis by sequential windowed data independent acquisition of the total high-resolution mass spectra-mass spectrometry. NP cells exhibited a lower ER stress burden under hypoxia. Knockdown of HIF-1α increased C/EBP homologous protein, protein kinase RNA-like endoplasmic reticulum kinase (PERK), and activating transcription factor 6 (ATF6) levels, whereas HIF-1α stabilization decreased the expression of ER stress markers Ddit3, Hsp5a, Atf6, and Eif2a. Interestingly, ER stress inducers tunicamycin and thapsigargin induced HIF-1α activity under hypoxia while promoting the unfolded protein response. NP cell secretome analysis demonstrated an impact of ER stress induction on extracellular matrix secretion, with decreases in collagens and cell adhesion-related proteins. Moreover, analysis of transcriptomic data of NP tissues from aged mice and degenerated human discs showed higher levels of unfolded protein response markers and decreased levels of matrix components. Our study shows, for the first time, that hypoxia and HIF-1α attenuate ER stress responses in NP cells, and ER stress promotes inefficient extracellular matrix secretion under hypoxia.
Collapse
Affiliation(s)
- Emanuel J Novais
- Department of Orthopedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania; Graduate Program in Cell Biology and Regenerative Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania; Life and Health Sciences Research Institute, School of Medicine, University of Minho, Braga, Portugal; Life and Health Sciences Research Institute/Biomaterials, Biodegradables and Biomimetics (ICVS/3B's) - PT Government Associate Laboratory, Braga, Portugal
| | - Hyowon Choi
- Department of Orthopedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania; Graduate Program in Cell Biology and Regenerative Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Vedavathi Madhu
- Department of Orthopedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania; Graduate Program in Cell Biology and Regenerative Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Kaori Suyama
- Department of Orthopedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania; Department of Anatomy and Cellular Biology, Tokai University School of Medicine, Isehara, Japan
| | - Sandra I Anjo
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Bruno Manadas
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Irving M Shapiro
- Department of Orthopedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania; Graduate Program in Cell Biology and Regenerative Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - António J Salgado
- Life and Health Sciences Research Institute, School of Medicine, University of Minho, Braga, Portugal; Life and Health Sciences Research Institute/Biomaterials, Biodegradables and Biomimetics (ICVS/3B's) - PT Government Associate Laboratory, Braga, Portugal
| | - Makarand V Risbud
- Department of Orthopedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania; Graduate Program in Cell Biology and Regenerative Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania.
| |
Collapse
|
57
|
Shao Z, Ni L, Hu S, Xu T, Meftah Z, Yu Z, Tian N, Wu Y, Sun L, Wu A, Pan Z, Chen L, Gao W, Zhou Y, Zhang X, Wang X. RNA-binding protein HuR suppresses senescence through Atg7 mediated autophagy activation in diabetic intervertebral disc degeneration. Cell Prolif 2021; 54:e12975. [PMID: 33372336 PMCID: PMC7848958 DOI: 10.1111/cpr.12975] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 12/08/2020] [Accepted: 12/14/2020] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVES Diabetes is a risk factor for intervertebral disc degeneration (IVDD). Studies have demonstrated that diabetes may affect IVDD through transcriptional regulation; however, whether post-transcriptional regulation is involved in diabetic IVDD (DB-IVDD) is still unknown. This study was performed to illustrate the role of HuR, an RNA-binding protein, in DB-IVDD development and its mechanism. MATERIALS AND METHODS The expression of HuR was evaluated in nucleus pulposus (NP) tissues from diabetic IVDD patients and in high glucose-treated NP cells. Senescence and autophagy were assessed in HuR over-expressing and downregulation NP cells. The mRNAs that were regulated by HuR were screened, and immunoprecipitation was applied to confirm the regulation of HuR on targeted mRNAs. RESULTS The results showed that the expression of HuR was decreased in diabetic NP tissues and high glucose-treated NP cells. Downregulation of HuR may lead to increased senescence in high glucose-treated NP cells, while autophagy activation attenuates senescence in HuR deficient NP cells. Mechanistic study showed that HuR prompted Atg7 mRNA stability via binding to the AU-rich elements. Furthermore, overexpression of Atg7, but not HuR, may ameliorate DB-IVDD in rats in vivo. CONCLUSIONS In conclusion, HuR may suppress senescence through autophagy activation via stabilizing Atg7 in diabetic NP cells; while Atg7, but not HuR, may serve as a potential therapeutic target for DB-IVDD.
Collapse
|
58
|
Madhu V, Guntur AR, Risbud MV. Role of autophagy in intervertebral disc and cartilage function: implications in health and disease. Matrix Biol 2020; 100-101:207-220. [PMID: 33301899 DOI: 10.1016/j.matbio.2020.12.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 12/02/2020] [Accepted: 12/03/2020] [Indexed: 12/12/2022]
Abstract
The intervertebral disc and cartilage are specialized, extracellular matrix-rich tissues critical for absorbing mechanical loads, providing flexibility to the joints, and longitudinal growth in the case of growth plate cartilage. Specialized niche conditions in these tissues, such as hypoxia, are critical in regulating cellular activities including autophagy, a lysosomal degradation pathway that promotes cell survival. Mounting evidence suggests that dysregulation of autophagic pathways underscores many skeletal pathologies affecting the spinal column, articular and growth plate cartilages. Many lysosomal storage disorders characterized by the accumulation of partially degraded glycosaminoglycans (GAGs) due to the lysosomal dysfunction thus affect skeletal tissues and result in altered ECM structure. Likewise, pathologies that arise from mutations in genes encoding ECM proteins and ECM processing, folding, and post-translational modifications, result in accumulation of misfolded proteins in the ER, ER stress and autophagy dysregulation. These conditions evidence reduced secretion of ECM proteins and/or increased secretion of mutant proteins, thereby impairing matrix quality and the integrity of affected skeletal tissues and causing a lack of growth and degeneration. In this review, we discuss the role of autophagy and mechanisms of its regulation in the intervertebral disc and cartilages, as well as how dysregulation of autophagic pathways affects these skeletal tissues.
Collapse
Affiliation(s)
- Vedavathi Madhu
- Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, PA, USA
| | - Anyonya R Guntur
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, ME, USA; Tufts University School of Medicine, Tufts University, Boston, MA USA; Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, ME USA
| | - Makarand V Risbud
- Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, PA, USA; Cell Biology and Regenerative Medicine Graduate Program, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
59
|
Cellular senescence-mediated exacerbation of Duchenne muscular dystrophy. Sci Rep 2020; 10:16385. [PMID: 33046751 PMCID: PMC7550355 DOI: 10.1038/s41598-020-73315-6] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 09/14/2020] [Indexed: 01/10/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a progressive disease characterised by chronic muscle degeneration and inflammation. Our previously established DMD model rats (DMD rats) have a more severe disease phenotype than the broadly used mouse model. We aimed to investigate the role of senescence in DMD using DMD rats and patients. Senescence was induced in satellite cells and mesenchymal progenitor cells, owing to the increased expression of CDKN2A, p16- and p19-encoding gene. Genetic ablation of p16 in DMD rats dramatically restored body weight and muscle strength. Histological analysis showed a reduction of fibrotic and adipose tissues invading skeletal muscle, with increased muscle regeneration. Senolytic drug ABT263 prevented loss of body weight and muscle strength, and increased muscle regeneration in rats even at 8 months—the late stage of DMD. Moreover, senescence markers were highly expressed in the skeletal muscle of DMD patients. In situ hybridization of CDKN2A confirmed the expression of it in satellite cells and mesenchymal progenitor cells in patients with DMD. Collectively, these data provide new insights into the integral role of senescence in DMD progression.
Collapse
|
60
|
Tsingas M, Ottone OK, Haseeb A, Barve RA, Shapiro IM, Lefebvre V, Risbud MV. Sox9 deletion causes severe intervertebral disc degeneration characterized by apoptosis, matrix remodeling, and compartment-specific transcriptomic changes. Matrix Biol 2020; 94:110-133. [PMID: 33027692 DOI: 10.1016/j.matbio.2020.09.003] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 09/30/2020] [Accepted: 09/30/2020] [Indexed: 12/27/2022]
Abstract
SOX9 plays an important role in chondrocyte differentiation and, in the developing axial skeleton, maintains the notochord and the demarcation of intervertebral disc compartments. Diminished expression is linked to campomelic dysplasia, resulting in severe scoliosis and progressive disc degeneration. However, the specific functions of SOX9 in the adult spinal column and disc are largely unknown. Accordingly, employing a strategy to conditionally delete Sox9 in Acan-expressing cells (AcanCreERT2Sox9fl/fl), we delineated these functions in the adult intervertebral disc. AcanCreERT2Sox9fl/fl mice (Sox9cKO) showed extensive and progressive remodeling of the extracellular matrix in nucleus pulposus (NP) and annulus fibrosus (AF), consistent with human disc degeneration. Progressive degeneration of the cartilaginous endplates (EP) was also evident in Sox9cKO mice, and it preceded morphological changes seen in the NP and AF compartments. Fate mapping using tdTomato reporter, EdU chase, and quantitative immunohistological studies demonstrated that SOX9 is crucial for disc cell survival and phenotype maintenance. Microarray analysis showed that Sox9 regulated distinct compartment-specific transcriptomic landscapes, with prominent contributions to the ECM, cytoskeleton-related, and metabolic pathways in the NP and ion transport, the cell cycle, and signaling pathways in the AF. In summary, our work provides new insights into disc degeneration in Sox9cKO mice at the cellular, molecular, and transcriptional levels, underscoring tissue-specific roles of this transcription factor. Our findings may direct future cell therapies targeting SOX9 to mitigate disc degeneration.
Collapse
Affiliation(s)
- Maria Tsingas
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA; Graduate Program in Cell Biology and Regenerative Medicine, Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Olivia K Ottone
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA; Graduate Program in Cell Biology and Regenerative Medicine, Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Abdul Haseeb
- Department of Surgery/Division of Orthopaedic Surgery, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Ruteja A Barve
- Department of Genetics, Genome Technology Access Centre at the McDonnell Genome Institute, Washington University, School of Medicine, St. Louis, MO 63110, USA
| | - Irving M Shapiro
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Véronique Lefebvre
- Department of Surgery/Division of Orthopaedic Surgery, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Makarand V Risbud
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA; Graduate Program in Cell Biology and Regenerative Medicine, Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| |
Collapse
|
61
|
Cell Senescence: A Nonnegligible Cell State under Survival Stress in Pathology of Intervertebral Disc Degeneration. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:9503562. [PMID: 32934764 PMCID: PMC7479476 DOI: 10.1155/2020/9503562] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 08/01/2020] [Accepted: 08/10/2020] [Indexed: 01/10/2023]
Abstract
The intervertebral disc degeneration (IDD) with increasing aging mainly manifests as low back pain (LBP) accompanied with a loss of physical ability. These pathological processes can be preliminarily interpreted as a series of changes at cellular level. In addition to cell death, disc cells enter into the stagnation with dysfunction and deteriorate tissue microenvironment in degenerative discs, which is recognized as cell senescence. During aging, many intrinsic and extrinsic factors have been proved to have strong connections with these cellular senescence phenomena. Growing evidences of these connections require us to gather up critical cues from potential risk factors to pathogenesis and relative interventions for retarding cell senescence and attenuating degenerative changes. In this paper, we try to clarify another important cell state apart from cell death in IDD and discuss senescence-associated changes in cells and extracellular microenvironment. Then, we emphasize the role of oxidative stress and epigenomic perturbations in linking risk factors to cell senescence in the onset of IDD. Further, we summarize the current interventions targeting senescent cells that may exert the benefits of antidegeneration in IDD.
Collapse
|
62
|
The Role of CDKs and CDKIs in Murine Development. Int J Mol Sci 2020; 21:ijms21155343. [PMID: 32731332 PMCID: PMC7432401 DOI: 10.3390/ijms21155343] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 07/22/2020] [Accepted: 07/26/2020] [Indexed: 02/07/2023] Open
Abstract
Cyclin-dependent kinases (CDKs) and their inhibitors (CDKIs) play pivotal roles in the regulation of the cell cycle. As a result of these functions, it may be extrapolated that they are essential for appropriate embryonic development. The twenty known mouse CDKs and eight CDKIs have been studied to varying degrees in the developing mouse, but only a handful of CDKs and a single CDKI have been shown to be absolutely required for murine embryonic development. What has become apparent, as more studies have shone light on these family members, is that in addition to their primary functional role in regulating the cell cycle, many of these genes are also controlling specific cell fates by directing differentiation in various tissues. Here we review the extensive mouse models that have been generated to study the functions of CDKs and CDKIs, and discuss their varying roles in murine embryonic development, with a particular focus on the brain, pancreas and fertility.
Collapse
|
63
|
Iozzo RV, Theocharis AD, Neill T, Karamanos NK. Complexity of matrix phenotypes. Matrix Biol Plus 2020; 6-7:100038. [PMID: 33543032 PMCID: PMC7852209 DOI: 10.1016/j.mbplus.2020.100038] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 05/11/2020] [Indexed: 02/06/2023] Open
Abstract
The extracellular matrix is engaged in an ever-evolving and elegant ballet of dynamic reciprocity that directly and bi-directionally regulates cell behavior. Homeostatic and pathophysiological changes in cell-matrix signaling cascades manifest as complex matrix phenotypes. Indeed, the extracellular matrix can be implicated in virtually every known human disease, thus, making it the most critical and dynamic "organ" in the human body. The overall goal of this Special Issue is to provide an accurate and inclusive functional definition that addresses the inherent complexity of matrix phenotypes. This goal is summarily achieved via a corpus of expertly written articles, reviews and original research, focused at answering this question empirically and fundamentally via state-of-the-art methods and research strategies.
Collapse
Key Words
- ADAM, a disintegrin and metalloproteinases
- AGE, advanced glycation end products
- Angiogenesis
- Cancer
- Collagen
- DDR1, discoidin domain receptor 1
- ECM, extracellular matrix
- EGF, epidermal growth factor
- EGFR, epidermal growth factor receptor
- EMILIN1, elastin microfibril interfacer 1
- EMILIN2, elastin microfibril interfacer 2
- EMT, epithelial-mesenchymal transition
- ERα, estrogen receptor α
- ERβ, estrogen receptor β
- GBM, glioblastoma
- HA, hyaluronan
- HAS2, hyaluronan synthase 2
- HAS2-AS1, HAS2 antisense 1
- HB-EGF, heparin binding EGF
- HMGA2, high-mobility group AT-Hook 2
- IBC, inflammatory breast cancer
- IGF-IR, insulin growth factor I receptor
- IR-A, insulin receptor A
- LEKTI, lympho-epithelial Kazal-type inhibitor
- LOX, lysyl oxidases
- LTBP, latent TGFβ-binding proteins
- MAGP, microfibril-associated glycoproteins
- MET, mesenchymal-epithelial transition
- MMP, matrix metalloproteinases
- Methodologies
- OB, osteoblast
- OI, osteogenesis imperfecta
- PARs, protease activated receptors
- PG, proteoglycans
- PLL, poly-l-lysine
- Proteoglycans
- ROS, reactive oxygen species
- RTK, receptor tyrosine kinase
- SLRP, small leucine rich proteoglycans
- SSR, solar-simulated radiation
- TGFβ, transforming growth factor β
- TNT, tunneling nanotubes
- UVR, ultraviolet radiation
- VEGF, vascular endothelial growth factor
- miR, microRNA
- tPA, tissue-type plasminogen activator
- uPA, urokinase-type plasminogen activator
Collapse
Affiliation(s)
- Renato V. Iozzo
- Department of Pathology, Anatomy and Cell Biology and the Cancer Cell Biology and Signaling Program, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States of America
| | - Achilleas D. Theocharis
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| | - Thomas Neill
- Department of Pathology, Anatomy and Cell Biology and the Cancer Cell Biology and Signaling Program, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, United States of America
| | - Nikos K. Karamanos
- Biochemistry, Biochemical Analysis and Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| |
Collapse
|
64
|
Novais EJ, Tran VA, Miao J, Slaver K, Sinensky A, Dyment NA, Addya S, Szeri F, Wetering K, Shapiro IM, Risbud MV. Comparison of inbred mouse strains shows diverse phenotypic outcomes of intervertebral disc aging. Aging Cell 2020; 19:e13148. [PMID: 32319726 PMCID: PMC7253061 DOI: 10.1111/acel.13148] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 02/29/2020] [Accepted: 03/11/2020] [Indexed: 12/27/2022] Open
Abstract
Intervertebral disc degeneration presents a wide spectrum of clinically degenerative disc phenotypes; however, the contribution of genetic background to the degenerative outcomes has not been established. We characterized the spinal phenotype of 3 mouse strains with varying cartilage-regenerative potential at 6 and 23 months: C57BL/6, LG/J and SM/J. All strains showed different aging phenotypes. Importantly, LG/J mice showed an increased prevalence of dystrophic disc calcification in caudal discs with aging. Quantitative-histological analyses of LG/J and SM/J caudal discs evidenced accelerated degeneration compared to BL6, with cellular disorganization and cell loss together with fibrosis of the NP, respectively. Along with the higher grades of disc degeneration, SM/J, at 6M, also differed the most in terms of NP gene expression compared to other strains. Moreover, although we found common DEGs between BL6 and LG/J aging, most of them were divergent between the strains. Noteworthy, the common DEGs altered in both LG/J and BL6 aging were associated with inflammatory processes, response to stress, cell differentiation, cell metabolism and cell division. Results suggested that disc calcification in LG/J resulted from a dystrophic calcification process likely aggravated by cell death, matrix remodelling, changes in calcium/phosphate homeostasis and cell transformation. Lastly, we report 7 distinct phenotypes of human disc degeneration based on transcriptomic profiles, that presented similar pathways and DEGs found in aging mouse strains. Together, our results suggest that disc aging and degeneration depends on the genetic background and involves changes in various molecular pathways, which might help to explain the diverse phenotypes seen during disc disease.
Collapse
Affiliation(s)
- Emanuel J. Novais
- Department of Orthopedic Surgery Sidney Kimmel Medical College Philadelphia PA USA
- Graduate Program in Cell Biology and Regenerative Medicine Thomas Jefferson University Philadelphia PA USA
- Life and Health Sciences Research Institute (ICVS) School of Medicine University of Minho Braga Portugal
- ICVS/3B’s – PT Government Associate Laboratory Braga Portugal
| | - Victoria A. Tran
- Department of Orthopedic Surgery Sidney Kimmel Medical College Philadelphia PA USA
| | - Jingya Miao
- Department of Orthopedic Surgery Sidney Kimmel Medical College Philadelphia PA USA
| | - Katie Slaver
- Department of Orthopedic Surgery Sidney Kimmel Medical College Philadelphia PA USA
| | - Andrew Sinensky
- Department of Orthopedic Surgery Sidney Kimmel Medical College Philadelphia PA USA
| | - Nathaniel A. Dyment
- Department of Orthopaedic Surgery University of Pennsylvania Philadelphia PA USA
| | - Sankar Addya
- Sidney Kimmel Cancer Center Thomas Jefferson University Philadelphia PA USA
| | - Flora Szeri
- Department of Dermatology and Cutaneous Biology Sidney Kimmel Medical College Thomas Jefferson University Philadelphia PA USA
- The PXE International Center of Excellence in Research and Clinical Care Thomas Jefferson University Philadelphia PA USA
- The Jefferson Institute of Molecular Medicine Thomas Jefferson University Philadelphia PA USA
| | - Koen Wetering
- Department of Dermatology and Cutaneous Biology Sidney Kimmel Medical College Thomas Jefferson University Philadelphia PA USA
- The PXE International Center of Excellence in Research and Clinical Care Thomas Jefferson University Philadelphia PA USA
- The Jefferson Institute of Molecular Medicine Thomas Jefferson University Philadelphia PA USA
| | - Irving M. Shapiro
- Department of Orthopedic Surgery Sidney Kimmel Medical College Philadelphia PA USA
- Graduate Program in Cell Biology and Regenerative Medicine Thomas Jefferson University Philadelphia PA USA
| | - Makarand V. Risbud
- Department of Orthopedic Surgery Sidney Kimmel Medical College Philadelphia PA USA
- Graduate Program in Cell Biology and Regenerative Medicine Thomas Jefferson University Philadelphia PA USA
| |
Collapse
|
65
|
Ohnishi T, Novais EJ, Risbud MV. Alterations in ECM signature underscore multiple sub-phenotypes of intervertebral disc degeneration. Matrix Biol Plus 2020; 6-7:100036. [PMID: 33543030 PMCID: PMC7852332 DOI: 10.1016/j.mbplus.2020.100036] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 04/07/2020] [Accepted: 04/07/2020] [Indexed: 12/18/2022] Open
Abstract
The intervertebral disc is a specialized connective tissue critical for absorption of mechanical loads and providing flexibility to the spinal column. The disc ECM is complex and plays a vital role in imparting tissue its biomechanical function. The central NP is primarily composed of large aggregating proteoglycans (PGs) while surrounding AF is composed of fibrillar collagens, I and II. Aggrecan and versican in particular, due to their high concentration of sulfated GAG chains form large aggregates with hyaluronic acid (HA) and provide water binding capacity to the disc. Degradation of aggrecan core protein due to aggrecanase and MMP activity, SNPs that affect number of chondroitin sulfate (CS) substitutions and alteration in enzymes critical in synthesis of CS chains can impair the aggrecan functionality. Similarly, levels of many matrix and matrix-related molecules e.g. Col2, Col9, HAS2, ccn2 are dysregulated during disc degeneration and genetic animal models have helped establish causative link between their expression and disc health. In the degenerating and herniated discs, increased levels of inflammatory cytokines such as TNF-α, IL-1β and IL-6 are shown to promote matrix degradation through regulating expression and activity of critical proteases and stimulate immune cell activation. Recent studies of different mouse strains have better elucidated the broader impact of spontaneous degeneration on disc matrix homeostasis. SM/J mice showed an increased cell apoptosis, loss of cell phenotype, and cleavage of aggrecan during early stages followed by tissue fibrosis evident by enrichment of several collagens, SLRPs and fibronectin. In summary, while disc degeneration encompasses wide spectrum of degenerative phenotypes extensive matrix degradation and remodeling underscores all of them. The intervertebral disc absorbs loads and provides flexibility to the spine. The ECM is complex and vital for imparting tissue its biomechanical function. Numerous types of proteoglycans and collagens designate the quality of the disc. Many matrix and matrix-related molecules are dysregulated during disc degeneration. Matrix degradation and remodeling underscores wide spectrum of phenotype.
Collapse
Affiliation(s)
- Takashi Ohnishi
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA.,Department of Orthopaedic Surgery, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Emanuel J Novais
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA.,Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga, Portugal
| | - Makarand V Risbud
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
66
|
Gorth DJ, Ottone OK, Shapiro IM, Risbud MV. Differential Effect of Long-Term Systemic Exposure of TNFα on Health of the Annulus Fibrosus and Nucleus Pulposus of the Intervertebral Disc. J Bone Miner Res 2020; 35:725-737. [PMID: 31800975 PMCID: PMC7145745 DOI: 10.1002/jbmr.3931] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Revised: 11/07/2019] [Accepted: 11/22/2019] [Indexed: 02/03/2023]
Abstract
The inflammatory cytokine tumor necrosis factor alpha (TNFα) is considered to play a key role in the pathogenesis of intervertebral disc disease. To evaluate the importance of this cytokine we examined the inflammatory environment and spinal phenotype of 9-month-old human TNFα overexpressing transgenic (hTNFα-TG) mice. The mice evidenced increased circulating levels of interleukin-1β (IL-1β), IL-2, keratinocyte chemoattractant/human growth-regulated oncogene (KC/GRO), and monocyte chemoattractant protein-1 (MCP-1) along with thinning of the cortical and trabecular vertebral bone. Surprisingly, although the nucleus pulposus (NP) of these mice was intact and healthy, the caudal annulus fibrosus (AF) evidenced robust cell death and immune cell infiltration. Despite these differences, there were no obvious alterations in the collagen or aggrecan content in the NP and AF. However, there was a reduction in cartilage oligomeric matrix protein (COMP), suggesting destabilization of the AF matrix. Microarray analysis of the NP from hTNFα-TG mice cells revealed minimal changes in global gene expression. These findings lend support to the notion that NP tissue is isolated from systemic inflammation. In contrast, the severe AF phenotype suggests that systemic inflammation interferes with AF health, predisposing discs to herniation as opposed to directly causing NP degeneration. © 2020 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Deborah J Gorth
- Department of Orthopaedic Surgery and Graduate Program in Cell Biology and Regenerative Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Olivia K Ottone
- Department of Orthopaedic Surgery and Graduate Program in Cell Biology and Regenerative Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Irving M Shapiro
- Department of Orthopaedic Surgery and Graduate Program in Cell Biology and Regenerative Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Makarand V Risbud
- Department of Orthopaedic Surgery and Graduate Program in Cell Biology and Regenerative Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
67
|
Che H, Li J, Li Y, Ma C, Liu H, Qin J, Dong J, Zhang Z, Xian CJ, Miao D, Wang L, Ren Y. p16 deficiency attenuates intervertebral disc degeneration by adjusting oxidative stress and nucleus pulposus cell cycle. eLife 2020; 9:52570. [PMID: 32125276 PMCID: PMC7065909 DOI: 10.7554/elife.52570] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 02/12/2020] [Indexed: 12/20/2022] Open
Abstract
The cell cycle regulator p16 is known as a biomarker and an effector of aging. However, its function in intervertebral disc degeneration (IVDD) is unclear. In this study, p16 expression levels were found to be positively correlated with the severity of human IVDD. In a mouse tail suspension (TS)-induced IVDD model, lumbar intervertebral disc height index and matrix protein expression levels were reduced significantly were largely rescued by p16 deletion. In TS mouse discs, reactive oxygen species levels, proportions of senescent cells, and the senescence-associated secretory phenotype (SASP) were all increased, cell cycling was delayed, and expression was downregulated for Sirt1, superoxide dismutase 1/2, cyclin-dependent kinases 4/6, phosphorylated retinoblastoma protein, and transcription factor E2F1/2. However, these effects were rescued by p16 deletion. Our results demonstrate that p16 plays an important role in IVDD pathogenesis and that its deletion attenuates IVDD by promoting cell cycle and inhibiting SASP, cell senescence, and oxidative stress. Neck and shoulder pain, lower back pain and leg numbness are conditions that many people will encounter as years go by. This is because intervertebral discs, the padding structures that fit between the bones in the spine, degenerate with age: their cells enter a ‘senescent’, inactive state, and stop multiplying. A protein known as p16, an important regulator of cell growth and division, is known to accumulate in senescent cells. In fact, in mouse fat tissue, muscles or eyes, removing the cells that contain high levels of p16 delays aging-associated disorders. However, it was still unknown whether deactivating the gene that codes p16 in senescent cells could delay disc degeneration. Here, Che, Li et al. discovered that p16 is highly present in the senescent cells of severely degenerated human intervertebral discs. The cells in the nucleus pulposus, the jelly-like and most critical tissue in the intervertebral discs, were extracted and grown in the lab under conditions that replicate the early stages of damage to the spine. Drugs and genetic manipulations were then used to decrease the amount of p16 in these cells. The experiments showed that reducing the levels of p16 results in the senescent cells multiplying more and showing fewer signs of damage and aging. In addition, the discs of mice in which the gene that codes for p16 had been deleted were less prone to degeneration compared to ‘normal’ mice in similar conditions. Overall, the work by Che, Li et al. shows that inhibiting p16 in disc cells delays the aging process and reduces the degeneration of intervertebral discs. These findings may one day be applicable to people with intervertebral disc diseases who, for example, could potentially benefit from a gene therapy targeting the cells which produce p16.
Collapse
Affiliation(s)
- Hui Che
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,University Medical Center, Albert-Ludwigs-University, Freiburg, Germany
| | - Jie Li
- Department of Orthopaedics, Xuzhou Central Hospital, Xuzhou Clinical College of Nanjing Medical University, The Affiliated Xuzhou Hospital of Southeast University, Xuzhou, China
| | - You Li
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Cheng Ma
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Huan Liu
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,The Affiliated Huai'an No.1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Jingyi Qin
- Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, China
| | - Jianghui Dong
- Department of Hand Surgery, Department of Plastic Reconstructive Surgery, Ningbo, China.,School of Pharmacy and Medical Sciences and UniSA Cancer Research Institute, University of South Australia, Adelaide, Australia
| | - Zhen Zhang
- Department of Hand Surgery, Department of Plastic Reconstructive Surgery, Ningbo, China
| | - Cory J Xian
- School of Pharmacy and Medical Sciences and UniSA Cancer Research Institute, University of South Australia, Adelaide, Australia
| | - Dengshun Miao
- State Key Laboratory of Reproductive Medicine, The Research Center for Bone and Stem Cells, Department of Anatomy, Histology and Embryology, Nanjing Medical University, Nanjing, China
| | - Liping Wang
- Department of Hand Surgery, Department of Plastic Reconstructive Surgery, Ningbo, China.,School of Pharmacy and Medical Sciences and UniSA Cancer Research Institute, University of South Australia, Adelaide, Australia
| | - Yongxin Ren
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
68
|
Silagi ES, Novais EJ, Bisetto S, Telonis AG, Snuggs J, Le Maitre CL, Qiu Y, Kurland IJ, Shapiro IM, Philp NJ, Risbud MV. Lactate Efflux From Intervertebral Disc Cells Is Required for Maintenance of Spine Health. J Bone Miner Res 2020; 35:550-570. [PMID: 31692093 PMCID: PMC7064427 DOI: 10.1002/jbmr.3908] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 10/21/2019] [Accepted: 10/30/2019] [Indexed: 12/19/2022]
Abstract
Maintenance of glycolytic metabolism is postulated to be required for health of the spinal column. In the hypoxic tissues of the intervertebral disc and glycolytic cells of vertebral bone, glucose is metabolized into pyruvate for ATP generation and reduced to lactate to sustain redox balance. The rise in intracellular H+ /lactate concentrations are balanced by plasma-membrane monocarboxylate transporters (MCTs). Using MCT4 null mice and human tissue samples, complemented with genetic and metabolic approaches, we determine that H+ /lactate efflux is critical for maintenance of disc and vertebral bone health. Mechanistically, MCT4 maintains glycolytic and tricarboxylic acid (TCA) cycle flux and intracellular pH homeostasis in the nucleus pulposus compartment of the disc, where hypoxia-inducible factor 1α (HIF-1α) directly activates an intronic enhancer in SLC16A3. Ultimately, our results provide support for research into lactate as a diagnostic biomarker for chronic, painful, disc degeneration. © 2019 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Elizabeth S Silagi
- Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, PA, USA
| | - Emanuel J Novais
- Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, PA, USA
| | - Sara Bisetto
- Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Aristeidis G Telonis
- Computational Medicine Center, Sidney Kimmel Medical College Thomas Jefferson University, Philadelphia, PA, USA
| | - Joseph Snuggs
- Biomolecular Sciences Research Centre Sheffield Hallam University, Sheffield, UK
| | | | - Yunping Qiu
- Department of Medicine, Fleischer Institute for Diabetes and Metabolism Albert Einstein College of Medicine, Bronx, NY, USA
| | - Irwin J Kurland
- Department of Medicine, Fleischer Institute for Diabetes and Metabolism Albert Einstein College of Medicine, Bronx, NY, USA
| | - Irving M Shapiro
- Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, PA, USA
| | - Nancy J Philp
- Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Makarand V Risbud
- Department of Orthopaedic Surgery, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
69
|
Tessier S, Doolittle AC, Sao K, Rotty JD, Bear JE, Ulici V, Loeser RF, Shapiro IM, Diekman BO, Risbud MV. Arp2/3 inactivation causes intervertebral disc and cartilage degeneration with dysregulated TonEBP-mediated osmoadaptation. JCI Insight 2020; 5:131382. [PMID: 31961823 DOI: 10.1172/jci.insight.131382] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 01/15/2020] [Indexed: 01/01/2023] Open
Abstract
Extracellular matrix and osmolarity influence the development and homeostasis of skeletal tissues through Rho GTPase-mediated alteration of the actin cytoskeleton. This study investigated whether the actin-branching Arp2/3 complex, a downstream effector of the Rho GTPases Cdc42 and Rac1, plays a critical role in maintaining the health of matrix-rich and osmotically loaded intervertebral discs and cartilage. Mice with constitutive intervertebral disc- and cartilage-specific deletion of the critical Arp2/3 subunit Arpc2 (Col2-Cre; Arpc2fl/fl) developed chondrodysplasia and spinal defects. Since these mice did not survive to adulthood, we generated mice with inducible Arpc2 deletion in disc and cartilage (Acan-CreERT2; Arpc2fl/fl). Inactivation of Arp2/3 at skeletal maturity resulted in growth plate closure, loss of proteoglycan content in articular cartilage, and degenerative changes in the intervertebral disc at 1 year of age. Chondrocytes with Arpc2 deletion showed compromised cell spreading on both collagen and fibronectin. Pharmacological inhibition of Cdc42 and Arp2/3 prevented the osmoadaptive transcription factor TonEBP/NFAT5 from recruiting cofactors in response to a hyperosmolarity challenge. Together, these findings suggest that Arp2/3 plays a critical role in cartilaginous tissues through the regulation of cell-extracellular matrix interactions and modulation of TonEBP-mediated osmoadaptation.
Collapse
Affiliation(s)
- Steven Tessier
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College
| | - Alexandra C Doolittle
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College.,Graduate Program in Cell Biology and Regenerative Medicine, Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Kimheak Sao
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College.,Graduate Program in Cell Biology and Regenerative Medicine, Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Jeremy D Rotty
- Department of Biochemistry, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA.,Department of Cell Biology and Physiology.,UNC Lineberger Comprehensive Cancer Center
| | - James E Bear
- Department of Cell Biology and Physiology.,UNC Lineberger Comprehensive Cancer Center
| | - Veronica Ulici
- Thurston Arthritis Research Center, and.,Division of Rheumatology, Allergy, and Immunology, Department of Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Richard F Loeser
- Thurston Arthritis Research Center, and.,Division of Rheumatology, Allergy, and Immunology, Department of Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Irving M Shapiro
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College.,Graduate Program in Cell Biology and Regenerative Medicine, Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Brian O Diekman
- Division of Rheumatology, Allergy, and Immunology, Department of Medicine, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA.,Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Chapel Hill and Raleigh, North Carolina, USA
| | - Makarand V Risbud
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College.,Graduate Program in Cell Biology and Regenerative Medicine, Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
70
|
Tessier S, Tran VA, Ottone OK, Novais EJ, Doolittle A, DiMuzio MJ, Shapiro IM, Risbud MV. TonEBP-deficiency accelerates intervertebral disc degeneration underscored by matrix remodeling, cytoskeletal rearrangements, and changes in proinflammatory gene expression. Matrix Biol 2019; 87:94-111. [PMID: 31707045 DOI: 10.1016/j.matbio.2019.10.007] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 10/15/2019] [Accepted: 10/30/2019] [Indexed: 01/08/2023]
Abstract
The tonicity-responsive enhancer binding protein (TonEBP) plays an important role in intervertebral disc and axial skeleton embryogenesis. However, the contribution of this osmoregulatory transcription factor in postnatal intervertebral disc homeostasis is not known in vivo. Here, we show for the first time that TonEBP-deficient mice have pronounced age-related degeneration of the intervertebral disc with annular and endplate herniations. Using FTIR-imaging spectroscopy, quantitative immunohistochemistry, and tissue-specific transcriptomic analysis, we provide morphological and molecular evidence that the overall phenotype is driven by a replacement of water-binding proteoglycans with fibrocartilaginous matrix. Whereas TonEBP deficiency in the AF compartment caused tissue fibrosis associated with alterations in actin cytoskeleton and adhesion molecules, predominant changes in pro-inflammatory pathways were seen in the NP compartment of mutants, underscoring disc compartment-specific effects. Additionally, TonEBP-deficient mice presented with compromised trabecular bone properties of vertebrae. These results provide the first in vivo support to the long-held hypothesis that TonEBP is crucial for postnatal homeostasis of the spine and controls a multitude of functions in addition to cellular osmoadaptation.
Collapse
Affiliation(s)
- Steven Tessier
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Victoria A Tran
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Olivia K Ottone
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA; Graduate Program in Cell Biology and Regenerative Medicine, Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, PA, USA
| | - Emanuel J Novais
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA; Graduate Program in Cell Biology and Regenerative Medicine, Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, PA, USA; Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal; ICVS/3B's-PT Government Associate Laboratory, Braga, Portugal
| | - Alexandra Doolittle
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA; Graduate Program in Cell Biology and Regenerative Medicine, Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, PA, USA
| | - Michael J DiMuzio
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Irving M Shapiro
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA; Graduate Program in Cell Biology and Regenerative Medicine, Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, PA, USA
| | - Makarand V Risbud
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA; Graduate Program in Cell Biology and Regenerative Medicine, Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
71
|
Sessions GA, Copp ME, Liu JY, Sinkler MA, D'Costa S, Diekman BO. Controlled induction and targeted elimination of p16 INK4a-expressing chondrocytes in cartilage explant culture. FASEB J 2019; 33:12364-12373. [PMID: 31408372 DOI: 10.1096/fj.201900815rr] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Cellular senescence is a phenotypic state that contributes to age-related diseases through the secretion of matrix-degrading and inflammatory molecules. An emerging therapeutic strategy for osteoarthritis (OA) is to selectively eliminate senescent cells by initiating apoptosis. This study establishes a cartilage explant model of senescence induction and senolytic clearance using p16Ink4a expression as a biomarker of senescence. Growth-factor stimulation of explants increased the expression of p16Ink4a at both the mRNA and protein levels. Applying this culture system to cartilage from p16tdTom reporter mice (a knockin allele with tdTomato fluorescent protein regulated by the endogenous p16Ink4a promoter) demonstrated the emergence of a p16-high population that was quantified using flow cytometry for tdTomato. Cell sorting was used to separate chondrocytes based on tdTomato fluorescence and p16-high cells showed higher senescence-associated β-galactosidase activity and increased gene expression of the senescence-associated secretory phenotype as compared with p16-low cells. The potential for effective senolysis within the cartilage extracellular matrix was assessed using navitoclax (ABT-263). Navitoclax treatment reduced the percentage of p16-high cells from 17.9 to 6.1% (mean of 13 matched pairs; P < 0.001) and increased cleaved caspase-3 confirmed apoptotic activity. Together, these findings establish a physiologically relevant cartilage explant model for testing the induction and elimination of senescent chondrocytes, which will support investigations of senolytic therapy for OA.-Sessions, G. A., Copp, M. E., Liu, J.-Y., Sinkler, M. A., D'Costa, S., Diekman, B. O. Controlled induction and targeted elimination of p16INK4a-expressing chondrocytes in cartilage explant culture.
Collapse
Affiliation(s)
- Garrett A Sessions
- Thurston Arthritis Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Michaela E Copp
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina-North Carolina State University, Raleigh, North Carolina, USA
| | - Jie-Yu Liu
- Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Margaret A Sinkler
- Thurston Arthritis Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA.,Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Susan D'Costa
- Thurston Arthritis Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Brian O Diekman
- Thurston Arthritis Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA.,Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina-North Carolina State University, Raleigh, North Carolina, USA.,Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
72
|
Gorth DJ, Shapiro IM, Risbud MV. A New Understanding of the Role of IL-1 in Age-Related Intervertebral Disc Degeneration in a Murine Model. J Bone Miner Res 2019; 34:1531-1542. [PMID: 30875127 PMCID: PMC6697204 DOI: 10.1002/jbmr.3714] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 03/05/2019] [Accepted: 03/10/2019] [Indexed: 12/25/2022]
Abstract
Increased cytokine expression, in particular interleukin-1β (IL-1β), is considered a hallmark of intervertebral disc degeneration. However, the causative relationship between IL-1 and age-dependent degeneration has not been established. To investigate the role of IL-1 in driving age-related disc degeneration, we studied the spine phenotype of global IL-1α/β double knockout (IL-1KO) mice at 12 and 20 months. Multiplex ELISA analysis of blood revealed significant reductions in the concentrations of IFN-γ, IL-5, IL-15, TNF-α, IP-10, and a trend of reduced concentrations of IL-10, macrophage inflammatory protein 1α (MIP-1α), keratinocyte chemoattractant/human growth-regulated oncogene (KC/GRO), and IL-6. However, the circulating level of MIP-2, a neutrophil chemoattractant, was increased in the IL-1KO. The alterations in systemic cytokine levels coincided with altered bone morphology-IL-1KO mice exhibited significantly thicker caudal cortical bone at 12 and 20 months. Despite these systemic inflammatory and bony changes, IL-1 deletion only minimally affected disc health. Both wild-type (WT) and IL-1KO mice showed age-dependent disc degeneration. Unexpectedly, rather than protecting the animals from degeneration, the aging phenotype was more pronounced in IL-1KO animals: knockout mice evidenced significantly more degenerative changes in the annulus fibrosis (AF) together with alterations in collagen type and maturity. At 20 months, there were no changes in nucleus pulposus (NP) extracellular matrix composition or cellular marker expression; however, the IL-1KO NP cells occupied a smaller proportion of the NP compartment that those of WT controls. Taken together, these results show that IL-1 deletion altered the systemic inflammatory environment and vertebral bone morphology. However, instead of protecting discs from age-related disc degeneration, global IL-1 deletion amplified the degenerative phenotype. © 2019 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Deborah J Gorth
- Department of Orthopaedic Surgery and Graduate Program in Cell Biology and Regenerative Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - Irving M Shapiro
- Department of Orthopaedic Surgery and Graduate Program in Cell Biology and Regenerative Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| | - Makarand V Risbud
- Department of Orthopaedic Surgery and Graduate Program in Cell Biology and Regenerative Medicine, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
73
|
Mohanty S, Pinelli R, Dahia CL. Characterization of Krt19 CreERT allele for targeting the nucleus pulposus cells in the postnatal mouse intervertebral disc. J Cell Physiol 2019; 235:128-140. [PMID: 31187500 PMCID: PMC6778700 DOI: 10.1002/jcp.28952] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 05/16/2019] [Accepted: 05/20/2019] [Indexed: 12/12/2022]
Abstract
Intervertebral disc degeneration and associated back pain are relatively common but sparsely understood conditions, affecting over 70% of the population during some point of life. Disc degeneration is often associated with a loss of nucleus pulposus (NP) cells. Genetic mouse models offer convenient avenues to understand the cellular and molecular regulation of the disc during its formation, growth, maintenance, and aging. However, due to the lack of inducible driver lines to precisely target NP cells in the postnatal mouse disc, progress in this area of research has been moderate. NP cells are known to express cytokeratin 19 (Krt19), and tamoxifen (Tam)‐inducible Krt19CreERT allele is available. The current study describes the characterization of Krt19CreERT allele to specifically and efficiently target NP cells in neonatal, skeletally mature, middle‐aged, and aged mice using two independent fluorescent reporter lines. The efficiency of recombination at all ages was validated by immunostaining for KRT19. Results show that following Tam induction, Krt19CreERT specifically drives recombination of NP cells in the spine of neonatal and aged mice, while no recombination was detected in the surrounding tissues. Knee joints from skeletally mature Tam‐treated Krt19CreERT/+; R26tdTOM mouse show the absence of recombination in all tissues and cells of the knee joint. Thus, this study provides evidence for the use of Krt19CreERT allele for genetic characterization of NP cells at different stages of the mouse life.
Collapse
Affiliation(s)
- Sarthak Mohanty
- Orthopaedic Soft Tissue Research, Hospital for Special Surgery, New York, New York
| | - Robert Pinelli
- Orthopaedic Soft Tissue Research, Hospital for Special Surgery, New York, New York
| | - Chitra Lekha Dahia
- Orthopaedic Soft Tissue Research, Hospital for Special Surgery, New York, New York.,Department of Cell and Developmental Biology, Weill Cornell Medicine, Graduate School of Medical Sciences, New York, New York
| |
Collapse
|