Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 119
Filter
1.
Cell Commun Signal ; 22(1): 419, 2024 Aug 28.
Article in English | MEDLINE | ID: mdl-39192354

ABSTRACT

BACKGROUND: Intervertebral disc (IVD) degeneration is a multifactorial pathological process resulting in the dysregulation of IVD cell activity. The catabolic shift observed in IVD cells during degeneration leads to increased inflammation, extracellular matrix (ECM) degradation, aberrant intracellular signaling and cell loss. Importantly, these pathological processes are known to be interconnected and to collectively contribute to the progression of the disease. MicroRNAs (miRNAs) are known as strong post-transcriptional regulators, targeting multiple genes simultaneously and regulating numerous intracellular pathways. Specifically, miR-155-5p has been of particular interest since it is known as a pro-inflammatory mediator and contributing factor to diseases like cancer and osteoarthritis. This study investigated the role of miR-155-5p in IVD degeneration with a specific focus on inflammation and mechanosensing. METHODS: Gain- and loss-of-function studies were performed through transfection of human Nucleus pulposus (NP) and Annulus fibrosus (AF) cells isolated from degenerated IVDs with miR-155-5p mimics, inhibitors or their corresponding non-targeting control. Transfected cells were then subjected to an inflammatory environment or mechanical loading. Conditioned media and cell lysates were collected for phosphorylation and cytokine secretion arrays as well as gene expression analysis. RESULTS: Increased expression of miR-155-5p in AF cells resulted in significant upregulation of interleukin (IL)-8 cytokine secretion during cyclic stretching and a similar trend in IL-6 secretion during inflammation. Furthermore, miR-155-5p mimics increased the expression of the brain-derived neurotrophic factor (BDNF) in AF cells undergoing cyclic stretching. In NP cells, miR-155-5p gain-of-function resulted in the activation of the mitogen-activated protein kinase (MAPK) signaling pathway through increased phosphorylation of p38 and p53. Lastly, miR-155-5p inhibition caused a significant increase in the anti-inflammatory cytokine IL-10 in AF cells and the tissue inhibitor of metalloproteinases (TIMP)-4 in NP cells respectively. CONCLUSION: Overall, these results show that miR-155-5p contributes to IVD degeneration by enhancing inflammation through pro-inflammatory cytokines and MAPK signaling, as well as by promoting the catabolic shift of AF cells during mechanical loading. The inhibition of miR-155-5p may constitute a potential therapeutic approach for IVD degeneration and low back pain.


Subject(s)
Inflammation , Intervertebral Disc Degeneration , MicroRNAs , MicroRNAs/genetics , MicroRNAs/metabolism , Intervertebral Disc Degeneration/genetics , Intervertebral Disc Degeneration/metabolism , Intervertebral Disc Degeneration/pathology , Humans , Inflammation/genetics , Inflammation/pathology , Inflammation/metabolism , Nucleus Pulposus/metabolism , Nucleus Pulposus/pathology , Male , Weight-Bearing , Middle Aged , Female , Annulus Fibrosus/metabolism , Annulus Fibrosus/pathology
2.
J Cell Physiol ; 237(2): 1266-1284, 2022 02.
Article in English | MEDLINE | ID: mdl-34787318

ABSTRACT

Intervertebral disc degeneration (IDD) is a leading contributor to low back pain. The intervertebral disc (IVD) is composed of three tissue types: the central gelatinous nucleus pulposus (NP) tissue, the surrounding annulus fibrosus (AF) tissue, and the inferior and superior cartilage endplates. The IVD microenvironment is hypoxic, acidic, hyperosmotic, and low in nutrients because it is mostly avascular. The cellular processes that underlie IDD initiation and progression are still poorly understood. Specifically, a lack of understanding regarding NP cell metabolism and physiology hinders the development of effective therapeutics to treat IDD patients. Autophagy is a vital intracellular degradation process that removes damaged organelles, misfolded proteins, and intracellular pathogens and recycles the degraded components for cellular energy and function. NP cells have adapted to survive within their harsh tissue microenvironment using processes that are largely unknown, and we postulate autophagy is one of these undiscovered mechanisms. In this review, we describe unique features of the IVD tissue, review how physiological stressors impact autophagy in NP cells in vitro, survey the current understanding of autophagy regulation in the IVD, and assess the relationship between autophagy and IDD. Published studies confirm autophagy markers are present in IVD tissue, and IVD cells can regulate autophagy in response to cellular stressors in vitro. However, data are still lacking to determine the exact mechanisms regulating autophagy in IVD cells. More in-depth research is needed to establish whether autophagy is necessary to maintain IVD cell health and validate autophagy as a relevant therapeutic target for treating IDD.


Subject(s)
Annulus Fibrosus , Intervertebral Disc Degeneration , Intervertebral Disc , Nucleus Pulposus , Annulus Fibrosus/metabolism , Autophagy , Humans , Intervertebral Disc/metabolism , Intervertebral Disc Degeneration/metabolism , Nucleus Pulposus/metabolism
3.
FASEB J ; 35(11): e21919, 2021 11.
Article in English | MEDLINE | ID: mdl-34591994

ABSTRACT

Back and neck pain are significant healthcare burdens that are commonly associated with pathologies of the intervertebral disc (IVD). The poor understanding of the cellular heterogeneity within the IVD makes it difficult to develop regenerative IVD therapies. To address this gap, we developed an atlas of bovine (Bos taurus) caudal IVDs using single-cell RNA-sequencing (scRNA-seq). Unsupervised clustering resolved 15 unique clusters, which we grouped into the following annotated partitions: nucleus pulposus (NP), outer annulus fibrosus (oAF), inner AF (iAF), notochord, muscle, endothelial, and immune cells. Analyzing the pooled gene expression profiles of the NP, oAF, and iAF partitions allowed us to identify novel markers for NP (CP, S100B, H2AC18, SNORC, CRELD2, PDIA4, DNAJC3, CHCHD7, and RCN2), oAF (IGFBP6, CTSK, LGALS1, and CCN3), and iAF (MGP, COMP, SPP1, GSN, SOD2, DCN, FN1, TIMP3, WDR73, and GAL) cells. Network analysis on subpopulations of NP and oAF cells determined that clusters NP1, NP2, NP4, and oAF1 displayed gene expression profiles consistent with cell survival, suggesting these clusters may uniquely support viability under the physiological stresses of the IVD. Clusters NP3, NP5, oAF2, and oAF3 expressed various extracellular matrix (ECM)-associated genes, suggesting their role in maintaining IVD structure. Lastly, transcriptional entropy and pseudotime analyses found that clusters NP3 and NP1 had the most stem-like gene expression signatures of the NP partition, implying these clusters may contain IVD progenitor cells. Overall, results highlight cell type diversity within the IVD, and these novel cell phenotypes may enhance our understanding of IVD development, homeostasis, degeneration, and regeneration.


Subject(s)
Annulus Fibrosus/cytology , Annulus Fibrosus/metabolism , Genetic Heterogeneity , Homeostasis/genetics , Nucleus Pulposus/cytology , Nucleus Pulposus/metabolism , RNA-Seq/methods , Single-Cell Analysis/methods , Transcriptome , Animals , Biomarkers/metabolism , Cattle , Extracellular Matrix/metabolism , Female , Intervertebral Disc Degeneration/genetics , Intervertebral Disc Degeneration/metabolism , Phenotype , Stem Cells/metabolism
4.
Int J Mol Sci ; 23(7)2022 Apr 05.
Article in English | MEDLINE | ID: mdl-35409374

ABSTRACT

Previous research has identified an association between external radiation and disc degeneration, but the mechanism was poorly understood. This study explores the effects of ionizing radiation (IR) on inducing cellular senescence of annulus fibrosus (AF) in cell culture and in an in vivo mouse model. Exposure of AF cell culture to 10-15 Gy IR for 5 min followed by 5 days of culture incubation resulted in almost complete senescence induction as evidenced by SA-ßgal positive staining of cells and elevated mRNA expression of the p16 and p21 senescent markers. IR-induced senescent AF cells exhibited increased matrix catabolism, including elevated matrix metalloproteinase (MMP)-1 and -3 protein expression and aggrecanolysis. Analogous results were seen with whole body IR-exposed mice, demonstrating that genotoxic stress also drives disc cellular senescence and matrix catabolism in vivo. These results have important clinical implications in the potential adverse effects of ionizing radiation on spinal health.


Subject(s)
Annulus Fibrosus , Intervertebral Disc Degeneration , Intervertebral Disc , Animals , Annulus Fibrosus/metabolism , Cellular Senescence , Intervertebral Disc/metabolism , Intervertebral Disc Degeneration/genetics , Intervertebral Disc Degeneration/metabolism , Matrix Metalloproteinases/metabolism , Mice , Radiation, Ionizing
5.
Int J Mol Sci ; 24(1)2022 Dec 22.
Article in English | MEDLINE | ID: mdl-36613651

ABSTRACT

Intervertebral disc (IVD) degeneration is a major contributing factor for discogenic low back pain (LBP), causing a significant global disability. The IVD consists of an inner core proteoglycan-rich nucleus pulposus (NP) and outer lamellae collagen-rich annulus fibrosus (AF) and is confined by a cartilage end plate (CEP), providing structural support and shock absorption against mechanical loads. Changes to degenerative cascades in the IVD cause dysfunction and instability in the lumbar spine. Various treatments include pharmacological, rehabilitation or surgical interventions that aim to relieve pain; however, these modalities do not halt the pathologic events of disc degeneration or promote tissue regeneration. Loss of stem and progenitor markers, imbalance of the extracellular matrix (ECM), increase of inflammation, sensory hyperinnervation and vascularization, and associated signaling pathways have been identified as the onset and progression of disc degeneration. To better understand the pain originating from IVD, our review focuses on the anatomy of IVD and the pathophysiology of disc degeneration that contribute to the development of discogenic pain. We highlight the key mechanisms and associated signaling pathways underlying disc degeneration causing discogenic back pain, current clinical treatments, clinical perspective and directions of future therapies. Our review comprehensively provides a better understanding of healthy IVD and degenerative events of the IVD associated with discogenic pain, which helps to model painful disc degeneration as a therapeutic platform and to identify signaling pathways as therapeutic targets for the future treatment of discogenic pain.


Subject(s)
Annulus Fibrosus , Intervertebral Disc Degeneration , Intervertebral Disc , Low Back Pain , Humans , Intervertebral Disc Degeneration/metabolism , Low Back Pain/etiology , Low Back Pain/therapy , Annulus Fibrosus/metabolism , Cartilage/metabolism , Back Pain/pathology , Intervertebral Disc/metabolism
6.
Int J Mol Sci ; 23(14)2022 Jul 19.
Article in English | MEDLINE | ID: mdl-35887297

ABSTRACT

Excessive mechanical loading is a major cause of spinal degeneration, typically originating from a tear in the annulus fibrosus (AF). Endoplasmic reticulum (ER) stress and NLRP3 (NOD-, LRR- and pyrin domain-containing protein 3) inflammasome have been implicated in the pathogenesis of intervertebral disc (IVD) degeneration. However, the causal relationship between the mechanical stretching of AF cells and the NLRP3 inflammasome response associated with ER stress remains scarce. To elucidate the pathogenesis and regulatory mechanisms of mechanical stretch-induced IVD degeneration, human AF cell lines were subjected to different degrees of cyclic stretching to simulate daily spinal movements. Our results indicated that 15% high cyclic stretch (HCS) induced the expression of NLRP3 and interleukin-1 beta (IL-1ß) and was also responsible for the increased expression of NADPH (nicotinamide adenine dinucleotide phosphate) oxidase 2 (NOX2) and reactive oxygen species (ROS) in human AF cells. In addition, HCS increased the expression of glucose-regulated protein 78 (GRP78), an ER stress chaperone, which was neutralized with tauroursodeoxycholic acid (TUDCA), an ER stress inhibitor. In addition, HCS was found to induce thioredoxin-interacting protein (TXNIP) expression and NLRP3 inflammasome activation, which can be suppressed by si-NOX2 or the NOX2 inhibitor GSK2795039. Consequently, HCS upregulated ER stress and ROS production, leading to increased NLRP3 and IL-1ß expression in human AF cells, and may further accelerate IVD degeneration.


Subject(s)
Annulus Fibrosus , Intervertebral Disc Degeneration , Annulus Fibrosus/metabolism , Endoplasmic Reticulum Stress/physiology , Humans , Inflammasomes/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Intervertebral Disc Degeneration/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Reactive Oxygen Species/metabolism
7.
J Cell Physiol ; 236(1): 677-687, 2021 01.
Article in English | MEDLINE | ID: mdl-32654176

ABSTRACT

Cadmium (Cd), a type of heavy metal that accumulates in the body because of smoking, mediates the toxic effect of smoking in many diseases, such as cardiovascular disease, osteoarthritis, and osteoporosis. However, the toxic effect of Cd on intervertebral disc tissues have not been reported. In the current study, we demonstrated that Cd induced the apoptosis of annulus fibrosus (AF) cells, which contributed to intervertebral disc degeneration (IVDD). Specifically, Cd induced the nuclear translocation of FoxO1a, which drives AF cells apoptosis through mitochondrial-related pathway. Phosphatidylinositol 3-kinase/protein kinase B (PI3K/AKT) signal pathway is also involved in this process. The combined use of LY29002, an inhibitor of PI3K, and small interfering RNA-targeting FoxO1a confirmed the relationship between the PI3K/AKT signal pathway and FoxO1a. In summary, present research explores the mechanism behind the contribution of smoking to IVDD and finds a new feasible target for preventing IVDD in smoking.


Subject(s)
Annulus Fibrosus/metabolism , Intervertebral Disc Degeneration/metabolism , Nerve Tissue Proteins/metabolism , Smoking/adverse effects , Animals , Apoptosis/drug effects , Cadmium/pharmacology , Intervertebral Disc/metabolism , Mitochondria/metabolism , Rats, Sprague-Dawley , Signal Transduction/drug effects , Signal Transduction/physiology
8.
J Cell Physiol ; 236(4): 2725-2739, 2021 04.
Article in English | MEDLINE | ID: mdl-32892384

ABSTRACT

Ferroptosis is a necrotic form of regulated cell death that was associated with lipid peroxidation and free iron-mediated Fenton reactions. It has been reported that iron deficiency had been implicated in the pathogenesis of intervertebral disc degeneration (IVDD) by activating apoptosis. However, the role of ferroptosis in the process of IVDD has not been illuminated. Here, we demonstrate the involvement of ferroptosis in IVDD pathogenesis. Our in vitro models show the changes in protein levels of ferroptosis marker and enhanced lipid peroxidation level during oxidative stress. Safranin O staining, hematoxylin-eosin staining, and immunohistochemical were used to assess the IVDD after 8 weeks of surgical procedure in vivo. Treatment with ferrostatin-1, deferoxamine, and RSL3 demonstrate the role of ferroptosis in tert-butyl hydroperoxide (TBHP)-treated annulus fibrosus cells (AFCs) and nucleus pulposus cells (NPCs). Ferritinophagy, nuclear receptor coactivator 4 (NCOA4)-mediated ferritin selective autophagy, is originated during the process of ferroptosis in response to TBHP treatment. Knockdown and overexpression NCOA4 further prove TBHP may induce ferroptosis of AFCs and NPCs in an autophagy-dependent way. These findings support a role for oxidative stress-induced ferroptosis in the pathogenesis of IVDD.


Subject(s)
Annulus Fibrosus/metabolism , Ferroptosis , Intervertebral Disc Degeneration/metabolism , Nucleus Pulposus/metabolism , Oxidative Stress , Animals , Annulus Fibrosus/drug effects , Annulus Fibrosus/ultrastructure , Autophagy , Carbolines/toxicity , Case-Control Studies , Cells, Cultured , Deferoxamine/pharmacology , Disease Models, Animal , Ferroptosis/drug effects , Humans , Intervertebral Disc Degeneration/genetics , Intervertebral Disc Degeneration/pathology , Intervertebral Disc Degeneration/prevention & control , Lipid Peroxidation , Male , Nuclear Receptor Coactivators/genetics , Nuclear Receptor Coactivators/metabolism , Nucleus Pulposus/drug effects , Nucleus Pulposus/ultrastructure , Oxidative Stress/drug effects , Rats, Sprague-Dawley , Siderophores/pharmacology , Signal Transduction , tert-Butylhydroperoxide/toxicity
9.
Osteoarthritis Cartilage ; 29(3): 389-401, 2021 03.
Article in English | MEDLINE | ID: mdl-33338640

ABSTRACT

OBJECTIVE: Rat intervertebral disc (IVD) is one of the most commonly used and cost-effective alternative models for human IVD. Many IVD related clinical studies need to be pre-tested on rat IVDs. However, studies on the heterogeneous cell clusters of the rat IVD are inadequate, and a further understanding of the marker genes and cell phenotypes of healthy mature IVD cells is essential. METHODS: In this study, we used the 10X Genomics technology to analyze the single-cell transcriptome of purified wild-type rat IVDs. RESULTS: We identified potentially new gene markers of IVDs via single-cell sequencing. Based on the unsupervised cluster analysis of 13,578 single-cell transcripts, 3 known IVD cell types were identified. We provided a complete single-cell gene expression map of the IVD. Immunohistochemical and immunofluorescence images of rat disc sections confirmed the new marker genes of all cell types. One group of heterologous cell groups expressed multi-functional stem cell (MSC)-specific genes, indicating the stem cell potential of IVD cells. CONCLUSION: We provided the phenotype and marker genes of IVD cells at the single-cell level, reconfirmed existing data, and proposed new marker genes, including MSC marker genes. By identifying more accurate target cells and genes, our results pave the way for further study of the response of individual disc cells to disease states and provide the basis for future disc regeneration therapies.


Subject(s)
Annulus Fibrosus/metabolism , Biomarkers/metabolism , Gene Expression Profiling , Intervertebral Disc/metabolism , Nucleus Pulposus/metabolism , RNA, Messenger/metabolism , Stem Cells/metabolism , Animals , Annulus Fibrosus/cytology , Fluorescent Antibody Technique , Immunohistochemistry , Intervertebral Disc/cytology , Nucleus Pulposus/cytology , RNA-Seq , Rats , Single-Cell Analysis
10.
Eur Cell Mater ; 42: 1-19, 2021 07 08.
Article in English | MEDLINE | ID: mdl-34235715

ABSTRACT

Mesenchymal stem/stromal cell (MSC)-based therapies have been proposed for back pain and disc degeneration, despite limited knowledge on their mechanism of action. The impact of MSCs/their secretome on annulus fibrosus (AF) cells and tissue was analysed in bovine AF organ cultures (AF-OCs) exposed to upper-physiological cyclic tensile strain (CTS, 9 %, 1 Hz, 3 h/d) and interleukin (IL)-1ß in a custom-made device. A 4 d treatment of the CTS + IL-1ß-stimulated AF-OCs with MSC secretome downregulated the expression of inflammation markers [IL-6, IL-8, prostaglandin-endoperoxide synthase 2 (PTGS2)], complement system regulators [cluster of differentiation (CD)46, CD55, CD59] and matrix metalloproteinase 1 but also of tissue inhibitors of metalloproteinases (TIMP-1, TIMP-2) and collagen type I. At the protein level, it was confirmed that IL-6, MMP-3 and collagen content was decreased in AF-OCs treated with the MSC secretome compared to the CTS + IL-1ß stimulation alone. 9 d after treatment, a biomechanical peel-force test showed that the annular adhesive strength was significantly decreased by the MSC secretome treatment. Overall, MSC secretome had a stronger impact on AF tissue than MSCs in co-culture. The secretome contributed to a decrease in the inflammatory and catabolic status of AF cells activated by CTS + IL-1ß and played a role in the regulation of the complement system. However, it also contributed to a decrease in collagen at the gene/protein level and in AF mechanical strength compared to the CTS + IL-1ß stimulation alone. Therefore, the use of MSC secretome requires further investigation regarding its influence on disc matrix properties.


Subject(s)
Annulus Fibrosus , Mesenchymal Stem Cells , Animals , Annulus Fibrosus/metabolism , Cattle , Cells, Cultured , Organ Culture Techniques , Secretome
11.
Eur Cell Mater ; 41: 558-575, 2021 05 22.
Article in English | MEDLINE | ID: mdl-34021906

ABSTRACT

Embryonic muscle forces are necessary for normal vertebral development and spinal curvature, but their involvement in intervertebral disc (IVD) development remains unclear. The aim of the current study was to determine how muscle contractions affect (1) notochord involution and vertebral segmentation, and (2) IVD development including the mechanical properties and morphology, as well as collagen fibre alignment in the annulus fibrosus. Muscular dysgenesis (mdg) mice were harvested at three prenatal stages: at Theiler Stage (TS)22 when notochord involution starts, at TS24 when involution is complete, and at TS27 when the IVD is formed. Vertebral and IVD development were characterised using histology, immunofluorescence, and indentation testing. The results revealed that notochord involution and vertebral segmentation occurred independently of muscle contractions between TS22 and TS24. However, in the absence of muscle contractions, we found vertebral fusion in the cervical region at TS27, along with (i) a displacement of the nucleus pulposus towards the dorsal side, (ii) a disruption of the structural arrangement of collagen in the annulus fibrosus, and (iii) an increase in viscous behaviour of the annulus fibrosus. These findings emphasise the important role of mechanical forces during IVD development, and demonstrate a critical role of muscle loading during development to enable proper annulus fibrosus formation. They further suggest a need for mechanical loading in the creation of fibre-reinforced tissue engineering replacement IVDs as a therapy for IVD degeneration.


Subject(s)
Intervertebral Disc/physiology , Muscles/physiology , Notochord/physiology , Animals , Annulus Fibrosus/metabolism , Annulus Fibrosus/physiology , Collagen/metabolism , Extracellular Matrix/metabolism , Extracellular Matrix/physiology , Female , Intervertebral Disc/metabolism , Intervertebral Disc Degeneration/metabolism , Intervertebral Disc Degeneration/physiopathology , Male , Mice , Mice, Inbred C57BL , Muscles/metabolism , Notochord/metabolism , Nucleus Pulposus/metabolism , Nucleus Pulposus/physiology
12.
Proc Natl Acad Sci U S A ; 115(3): 477-482, 2018 01 16.
Article in English | MEDLINE | ID: mdl-29282316

ABSTRACT

Recapitulation of the form and function of complex tissue organization using appropriate biomaterials impacts success in tissue engineering endeavors. The annulus fibrosus (AF) represents a complex, multilamellar, hierarchical structure consisting of collagen, proteoglycans, and elastic fibers. To mimic the intricacy of AF anatomy, a silk protein-based multilayered, disc-like angle-ply construct was fabricated, consisting of concentric layers of lamellar sheets. Scanning electron microscopy and fluorescence image analysis revealed cross-aligned and lamellar characteristics of the construct, mimicking the native hierarchical architecture of the AF. Induction of secondary structure in the silk constructs was confirmed by infrared spectroscopy and X-ray diffraction. The constructs showed a compressive modulus of 499.18 ± 86.45 kPa. Constructs seeded with porcine AF cells and human mesenchymal stem cells (hMSCs) showed ∼2.2-fold and ∼1.7-fold increases in proliferation on day 14, respectively, compared with initial seeding. Biochemical analysis, histology, and immunohistochemistry results showed the deposition of AF-specific extracellular matrix (sulfated glycosaminoglycan and collagen type I), indicating a favorable environment for both cell types, which was further validated by the expression of AF tissue-specific genes. The constructs seeded with porcine AF cells showed ∼11-, ∼5.1-, and ∼6.7-fold increases in col Iα 1, sox 9, and aggrecan genes, respectively. The differentiation of hMSCs to AF-like tissue was evident from the enhanced expression of the AF-specific genes. Overall, the constructs supported cell proliferation, differentiation, and ECM deposition resulting in AF-like tissue features based on ECM deposition and morphology, indicating potential for future studies related to intervertebral disc replacement therapy.


Subject(s)
Annulus Fibrosus/cytology , Intervertebral Disc/cytology , Silk/chemistry , Tissue Engineering/instrumentation , Animals , Annulus Fibrosus/chemistry , Annulus Fibrosus/metabolism , Biomechanical Phenomena , Cell Proliferation , Cells, Cultured , Collagen/metabolism , Extracellular Matrix/metabolism , Humans , Intervertebral Disc/chemistry , Intervertebral Disc/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Swine , Tissue Scaffolds/chemistry
13.
Int J Mol Sci ; 22(10)2021 May 17.
Article in English | MEDLINE | ID: mdl-34067899

ABSTRACT

The intervertebral disc (IVD) is a complex joint structure comprising three primary components-namely, nucleus pulposus (NP), annulus fibrosus (AF), and cartilaginous endplate (CEP). The IVD retrieves oxygen from the surrounding vertebral body through CEP by diffusion and likely generates ATP via anaerobic glycolysis. IVD degeneration is characterized by a cascade of cellular, compositional, structural changes. With advanced age, pronounced changes occur in the composition of the disc extracellular matrix (ECM). NP and AF cells in the IVD possess poor regenerative capacity compared with that of other tissues. Hypoxia-inducible factor (HIF) is a master transcription factor that initiates a coordinated cellular cascade in response to a low oxygen tension environment, including the regulation of numerous enzymes in response to hypoxia. HIF-1α is essential for NP development and homeostasis and is involved in various processes of IVD degeneration process, promotes ECM in NP, maintains the metabolic activities of NP, and regulates dystrophic mineralization of NP, as well as angiogenesis, autophagy, and apoptosis during IVD degeneration. HIF-1α may, therefore, represent a diagnostic tool for early IVD degeneration and a therapeutic target for inhibiting IVD degeneration.


Subject(s)
Intervertebral Disc Degeneration/therapy , Intervertebral Disc/metabolism , Regeneration/physiology , Annulus Fibrosus/metabolism , Extracellular Matrix/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Nucleus Pulposus/metabolism
14.
Int J Mol Sci ; 22(11)2021 May 26.
Article in English | MEDLINE | ID: mdl-34073333

ABSTRACT

The intervertebral disc is the largest avascular low-nutrient organ in the body. Thus, resident cells may utilize autophagy, a stress-response survival mechanism, by self-digesting and recycling damaged components. Our objective was to elucidate the involvement of autophagy in rat experimental disc degeneration. In vitro, the comparison between human and rat disc nucleus pulposus (NP) and annulus fibrosus (AF) cells found increased autophagic flux under serum deprivation rather in humans than in rats and in NP cells than in AF cells of rats (n = 6). In vivo, time-course Western blotting showed more distinct basal autophagy in rat tail disc NP tissues than in AF tissues; however, both decreased under sustained static compression (n = 24). Then, immunohistochemistry displayed abundant autophagy-related protein expression in large vacuolated disc NP notochordal cells of sham rats. Under temporary static compression (n = 18), multi-color immunofluorescence further identified rapidly decreased brachyury-positive notochordal cells with robust expression of autophagic microtubule-associated protein 1 light chain 3 (LC3) and transiently increased brachyury-negative non-notochordal cells with weaker LC3 expression. Notably, terminal deoxynucleotidyl transferase dUTP nick end labeling-positive apoptotic death was predominant in brachyury-negative non-notochordal cells. Based on the observed notochordal cell autophagy impairment and non-notochordal cell apoptosis induction under unphysiological mechanical loading, further investigation is warranted to clarify possible autophagy-induced protection against notochordal cell disappearance, the earliest sign of disc degeneration, through limiting apoptosis.


Subject(s)
Annulus Fibrosus/metabolism , Autophagy , Intervertebral Disc Degeneration/metabolism , Nucleus Pulposus/metabolism , Animals , Annulus Fibrosus/pathology , Humans , Intervertebral Disc Degeneration/pathology , Male , Nucleus Pulposus/pathology , Rats , Rats, Sprague-Dawley
15.
Connect Tissue Res ; 61(1): 63-81, 2020 01.
Article in English | MEDLINE | ID: mdl-31597481

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.


Subject(s)
Aging/metabolism , Annulus Fibrosus/metabolism , Gene Expression Profiling , Gene Expression Regulation , Intervertebral Disc Degeneration/metabolism , Nucleus Pulposus/metabolism , Aging/genetics , Animals , Intervertebral Disc Degeneration/genetics , Mice , Mice, Transgenic
16.
Int J Mol Sci ; 21(6)2020 Mar 21.
Article in English | MEDLINE | ID: mdl-32245213

ABSTRACT

Intervertebral disc (IVD) herniation and degeneration is a major source of back pain. In order to regenerate a herniated and degenerated disc, closure of the anulus fibrosus (AF) is of crucial importance. For molecular characterization of AF, genome-wide Affymetrix HG-U133plus2.0 microarrays of native AF and cultured cells were investigated. To evaluate if cells derived from degenerated AF are able to initiate gene expression of a regenerative pattern of extracellular matrix (ECM) molecules, cultivated cells were stimulated with bone morphogenetic protein 2 (BMP2), transforming growth factor ß1 (TGFß1) or tumor necrosis factor-α (TNFα) for 24 h. Comparative microarray analysis of native AF tissues showed 788 genes with a significantly different gene expression with 213 genes more highly expressed in mild and 575 genes in severe degenerated AF tissue. Mild degenerated native AF tissues showed a higher gene expression of common cartilage ECM genes, whereas severe degenerated AF tissues expressed genes known from degenerative processes, including matrix metalloproteinases (MMP) and bone associated genes. During monolayer cultivation, only 164 differentially expressed genes were found. The cells dedifferentiated and altered their gene expression profile. RTD-PCR analyses of BMP2- and TGFß1-stimulated cells from mild and severe degenerated AF tissue after 24 h showed an increased expression of cartilage associated genes. TNFα stimulation increased MMP1, 3, and 13 expression. Cells derived from mild and severe degenerated tissues could be stimulated to a comparable extent. These results give hope that regeneration of mildly but also strongly degenerated disc tissue is possible.


Subject(s)
Annulus Fibrosus/metabolism , Extracellular Matrix/metabolism , Gene Expression Regulation/genetics , Intervertebral Disc Degeneration/metabolism , Intervertebral Disc Displacement/metabolism , Intervertebral Disc/metabolism , Annulus Fibrosus/pathology , Bone Morphogenetic Protein 2/pharmacology , Cells, Cultured , Extracellular Matrix/genetics , Gene Expression Profiling , Gene Expression Regulation/drug effects , Humans , Intervertebral Disc/pathology , Intervertebral Disc Degeneration/genetics , Intervertebral Disc Degeneration/pathology , Intervertebral Disc Displacement/genetics , Intervertebral Disc Displacement/pathology , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 1/metabolism , Matrix Metalloproteinase 13/genetics , Matrix Metalloproteinase 13/metabolism , Matrix Metalloproteinase 3/genetics , Matrix Metalloproteinase 3/metabolism , Oligonucleotide Array Sequence Analysis , Regeneration/drug effects , Regeneration/genetics , Transforming Growth Factor beta1/pharmacology , Tumor Necrosis Factor-alpha/pharmacology
17.
J Cell Physiol ; 234(8): 13705-13719, 2019 08.
Article in English | MEDLINE | ID: mdl-31010267

ABSTRACT

Mice lacking equilibrative nucleoside transporter 1 (ENT1 -/- ) demonstrate progressive calcification of spinal tissues including the annulus fibrosus (AF) of the intervertebral disc (IVD). We previously established ENT1 as the primary nucleoside transporter in the AF and demonstrated dysregulation of biomineralization pathways. To identify cellular pathways altered by loss of ENT1, we conducted microarray analysis of AF tissue from wild-type (WT) and ENT1 -/- mice before calcification (2 months of age) and associated with calcification (6 months of age). Bioinformatic analyses identified cell cycle dysregulation in ENT1 -/- AF tissues and implicated the E2f family of transcription factors as potential effectors. Quantitative polymerase chain reaction analysis confirmed increased expression of multiple E2f transcription factors and E2f interacting proteins ( Rb1 and Cdk2) in ENT1 -/- AF cells compared with WT at 6 months of age. At this time point, ENT1 -/- AF tissues showed increased JNK MAPK pathway activation, CDK1, minichromosome maintenance complex component 5 (Mcm5), and proliferating cell nuclear antigen (PCNA) protein expression, and PCNA-positive proliferating cells compared with WT controls. The current study demonstrates that loss of ENT1-mediated adenosine transport leads to increased cell proliferation in the AF of the IVD.


Subject(s)
Annulus Fibrosus/metabolism , Annulus Fibrosus/pathology , Equilibrative Nucleoside Transporter 1/metabolism , Adenosine/metabolism , Animals , Calcinosis/metabolism , Cell Proliferation/physiology , Mice , Mice, Knockout
18.
J Cell Physiol ; 234(11): 19977-19989, 2019 11.
Article in English | MEDLINE | ID: mdl-30945295

ABSTRACT

Inflammation is one of the major causes of intervertebral disc degeneration (IDD). Emerging evidence has revealed that increase in the levels of pro-inflammatory cytokines, such as interleukin 6 (IL-6) and tumor necrosis factor alpha (TNF-α), can activate a variety of signaling pathways, eventually resulting in IDD. Here, we show that the two cullin family genes, CUL4A and CUL4B, but not other cullins, are specifically overexpressed in IDD samples compared with healthy controls, and the CUL4A and CUL4B levels are positively correlated with the severity of IDD. In vitro analyses in human osteoblast cells (hFOB1.19), nucleus pulposus cells (hNPCs), and annulus fibrosus cells (hAFCs) indicated that treatment with IL-6 and TNF-α can increase CUL4A and CUL4B levels. By performing a microRNA-based microarray analysis, we found a set of microRNAs (miRNAs) that were differentially expressed in IDD samples compared with samples from healthy controls. Of these miRNAs, miR-194-5p, was significantly downregulated in IDD samples and could bind to the three prime untranslated regions (3'-UTRs) of both CUL4A and CUL4B, thereby downregulating their expression. The in vitro overexpression or downregulation of miR-194-5p, with a miR-194-5p-mimic or with anti-miR-194-5p, can cause the repression or induction of both CUL4A and CUL4B, respectively. Interestingly, treatment with IL-6 and TNF-α inhibitors in primary hNPCs and hAFCs that were isolated from patients with IDD led to the downregulation of CUL4A and CUL4B. Together, these findings provide insight into how the inflammation-dependent downregulation of miR-194-5p contributes to the pathogenesis of IDD, which may aid in the development of new therapeutic approaches for IDD by directly targeting miR-194-5p or CUL4A and CUL4B.


Subject(s)
Cullin Proteins/metabolism , Down-Regulation/genetics , Inflammation/complications , Inflammation/genetics , Intervertebral Disc Degeneration/complications , Intervertebral Disc Degeneration/genetics , MicroRNAs/genetics , Annulus Fibrosus/metabolism , Cell Line , Cells, Cultured , Cullin Proteins/genetics , Cytokines/metabolism , Humans , Inflammation Mediators/metabolism , Interleukin-6/pharmacology , Intervertebral Disc Degeneration/pathology , MicroRNAs/metabolism , Models, Biological , Nucleus Pulposus/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Up-Regulation/genetics
19.
Osteoarthritis Cartilage ; 27(12): 1860-1869, 2019 12.
Article in English | MEDLINE | ID: mdl-31419488

ABSTRACT

OBJECTIVES: The objective of this study was to perform a quantitative analysis of the structural and functional alterations in the intervertebral disc during in vivo degeneration, using emerging tools that enable rigorous assessment from the microscale to the macroscale, as well as to correlate these outcomes with noninvasive, clinically relevant imaging parameters. DESIGN: Degeneration was induced in a rabbit model by puncturing the annulus fibrosus (AF) with a 16-gauge needle. 2, 4, 8, and 12 weeks following puncture, degenerative changes in the discs were evaluated via magnetic resonance imaging (MRI), whole motion segment biomechanics, atomic force microscopy, histology and polarized light microscopy, immunohistochemistry, biochemical content, and second harmonic generation imaging. RESULTS: Following puncture, degeneration was evident through marked changes in whole disc structure and mechanics. Puncture acutely compromised disc macro and microscale mechanics, followed by progressive stiffening and remodeling. Histological analysis showed substantial anterior fibrotic remodeling and osteophyte formation, as well as an overall reduction in disc height, and disorganization and infolding of the AF lamellae into the NP space. Increases in NP collagen content and aggrecan breakdown products were also noted within 4 weeks. On MRI, NP T2 was reduced at all post-puncture time points and correlated significantly with microscale indentation modulus. CONCLUSION: This study defined the time dependent changes in disc structure-function relationships during IVD degeneration in a rabbit annular injury model and correlated degeneration severity with clinical imaging parameters. Our findings identified AF infolding and occupancy of the space as a principle mechanism of disc degeneration in response to needle puncture, and provide new insights to direct the development of novel therapeutics.


Subject(s)
Annulus Fibrosus/diagnostic imaging , Intervertebral Disc Degeneration/diagnostic imaging , Nucleus Pulposus/diagnostic imaging , Aggrecans/metabolism , Animals , Annulus Fibrosus/metabolism , Annulus Fibrosus/pathology , Annulus Fibrosus/physiopathology , Biomechanical Phenomena , Collagen/metabolism , Disease Models, Animal , Disease Progression , Immunohistochemistry , Intervertebral Disc/diagnostic imaging , Intervertebral Disc/metabolism , Intervertebral Disc/pathology , Intervertebral Disc/physiopathology , Intervertebral Disc Degeneration/metabolism , Intervertebral Disc Degeneration/pathology , Intervertebral Disc Degeneration/physiopathology , Magnetic Resonance Imaging , Microscopy, Atomic Force , Microscopy, Polarization , Nucleus Pulposus/metabolism , Nucleus Pulposus/pathology , Nucleus Pulposus/physiopathology , Punctures , Rabbits , Second Harmonic Generation Microscopy
20.
J Anat ; 234(1): 16-32, 2019 01.
Article in English | MEDLINE | ID: mdl-30450595

ABSTRACT

Intervertebral disc (IVD) degeneration and trauma is a major socio-economic burden and the focus of cell-based regenerative medicine approaches. Despite numerous ongoing clinical trials attempting to replace ailing IVD cells with mesenchymal stem cells, a solid understanding of the identity and nature of cells in a healthy mature IVD is still in need of refinement. Although anatomically simple, the IVD is comprised of heterogeneous cell populations. Therefore, methods involving cell pooling for RNA profiling could be misleading. Here, by using RNA in situ hybridization and z proportion test, we have identified potential novel biomarkers through single cell assessment. We quantified the proportion of RNA transcribing cells for 50 genetic loci in the outer annulus fibrosus (AF) and nucleus pulposus (NP) in coccygeal bovine discs isolated from tails of four skeletally mature animals. Our data reconfirm existing data and suggest 10 novel markers such as Lam1 and Thy1 in the outer AF and Gli1, Gli3, Noto, Scx, Ptprc, Sox2, Zscan10 and LOC101904175 in the NP, including pluripotency markers, that indicate stemness potential of IVD cells. These markers could be added to existing biomarker panels for cell type characterization. Furthermore, our data once more demonstrate heterogeneity in cells of the AF and NP, indicating the need for single cell assessment by methods such as RNA in situ hybridization. Our work refines the molecular identity of outer AF and NP cells, which can benefit future regenerative medicine and tissue engineering strategies in humans.


Subject(s)
Annulus Fibrosus/metabolism , In Situ Hybridization/methods , Intervertebral Disc/metabolism , Nucleus Pulposus/metabolism , RNA/metabolism , Aggrecans/genetics , Aggrecans/metabolism , Animals , Annulus Fibrosus/cytology , Biomarkers/metabolism , Cattle , Intervertebral Disc/cytology , Intervertebral Disc Degeneration/genetics , Intervertebral Disc Degeneration/metabolism , Intervertebral Disc Degeneration/therapy , Laminin/genetics , Laminin/metabolism , Nucleus Pulposus/cytology , RNA/genetics
SELECTION OF CITATIONS
SEARCH DETAIL