Skip to main content
Erschienen in: Arthritis Research & Therapy 1/2017

Open Access 01.12.2017 | Review

Molecular mechanisms underlying osteoarthritis development: Notch and NF-κB

verfasst von: Taku Saito, Sakae Tanaka

Erschienen in: Arthritis Research & Therapy | Ausgabe 1/2017

Abstract

Osteoarthritis (OA) is a multi-factorial and highly prevalent joint disorder worldwide. Since the establishment of murine surgical knee OA models in 2005, many of the key molecules and signalling pathways responsible for OA development have been identified. Here we review the roles of two multi-functional signalling pathways in OA development: Notch and nuclear factor kappa-light-chain-enhancer of activated B cells. Previous studies have identified various aspects of articular chondrocyte regulation by these pathways. However, comprehensive understanding of the molecular networks regulating articular cartilage homeostasis and OA pathogenesis is needed.
Abkürzungen
Adam
A disintegrin and metalloproteinase
Adamts5
A disintegrin and metallopeptidase with thrombospondin type 1 motif 5
CaMK2
Calcium/calmodulin-dependent protein kinase II
ChIP-seq
Chromatin immunoprecipitation and deep sequencing
Hes
Hairy and enhancer of split
Hey
Hairy/enhancer-of-split related with YRPW motif
HIF-2α
Hypoxia-inducible factor 2 alpha
ICAT
Inhibitor of β-catenin and T cell factor
ICD
Intracellular domain
IKK
IκB kinase
iNOS
Inducible nitric oxide synthase
IκB
Inhibitors of NF-κB
MAGP1
Microfibril-associated glycoprotein 1
Mmp13
Matrix metalloproteinase 13
NF-κB
Nuclear factor kappa-light-chain-enhancer of activated B cells
Nov
Nephroblastoma overexpressed gene
OA
Osteoarthritis
Prg4
Proteoglycan-4
Rela
V-rel reticuloendotheliosis viral oncogene homologue A
TGF-β
Transforming growth factor beta
Vegfa
Vascular endothelial growth factor A

Background

Osteoarthritis (OA) is the most prevalent joint disease worldwide, causing chronic disability in older people. Various factors are associated with its pathogenesis, including aging, obesity, joint instability, and joint inflammation [1]. Since the establishment of experimental murine models with surgically induced knee joint instability [2, 3], many studies have revealed the major molecules or signalling pathways responsible for OA, such as a disintegrin-like and metallopeptidase with thrombospondin type 1 motif 5 (Adamts 5), matrix metalloproteinase-13 (Mmp13), hedgehog signalling, syndecan-4, Wnt signalling, and hypoxia-inducible factor 2-alpha (HIF-2α) [2, 410]. More recently, an increasing number of papers have reported involvement of molecules or signalling pathways that regulate various biological phenomena, including oxidative stress, autophagy, epigenetic change, cellular senescence, circadian rhythm, and microRNAs [1117].
Adult articular chondrocytes have long been regarded as cycle-arrested cells that are unable to undergo proliferation. Hence, articular cartilage was previously referred to as “permanent cartilage” and was thought to lack the capability for self-restoration. Interestingly, a recent study using a Cre/loxP-based cell tracking technique has shown that proteoglycan-4 (Prg4)-expressing cells located in the superficial zone of adult articular cartilage expand to the middle zone above the tidemark, indicating that chondrocytes are slowly differentiating in adult articular cartilage [18]. Consequently, this novel finding is now innovating the concept of articular cartilage homeostasis.
Here, we focus on two multi-functional signalling pathways, Notch and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB). Previous studies have reported various and somewhat contradictory effects of these two signalling pathways in chondrocytes. We primarily select studies in which roles of Notch or NF-κB were investigated using murine experimental OA models, and discuss the roles of these signalling pathways in OA pathophysiology and articular cartilage homeostasis, considering recent findings in articular cartilage.

Notch signalling

Notch is a single-pass transmembrane cell surface receptor that plays a crucial role in cell-fate determination by regulating differentiation and apoptosis during embryogenesis and post-natal development [19, 20]. In mammals, the Notch signalling pathway consists of several molecules, including Notch ligands (Delta-like 1, 3 and 4, or Jagged (Jag) 1 and 2), Notch receptors (Notch 1–4), the transcriptional effector recombination signal binding protein for Ig kappa J (Rbpj), and target transcription factors hairy and enhancer of split (Hes) and hairy/enhancer-of-split related with YRPW motif (Hey) [20]. Notch signalling is initiated when Notch ligands on the cell surface bind to Notch receptors on adjacent cells. Upon ligand binding, the Notch receptor is cleaved by Adam and subsequently by a γ-secretase complex. Notch-intracellular domain (ICD) then translocates to the nucleus and binds to Rbpj to form a transcriptional activator that induces the Hes/Hey family.
We have reported previously that the activation of Notch signalling in articular chondrocytes contributes to the development of OA [21, 22]. Notch 1 and 2 receptors are highly expressed in articular chondrocytes [21] and are localized at the cell surface in normal mouse and human articular cartilage, but are translocated into the nucleus in degenerated cartilage [21]. Inhibition of Notch signalling by Rbpj knockout in chondrocytes after skeletal development suppresses OA development in a murine surgical model, and injection of the γ-secretase inhibitor DAPT into the knee joints of the wild-type OA model mice results in a similar protective effect [21]. Mmp13 expression is increased by overexpression of Notch-ICD in mouse primary chondrocytes and chondrocyte cell lines, and is decreased in Rbpj-knockout cartilage [21].
Among canonical Notch ligands, Jag1 is abundantly expressed and markedly increased during murine OA development [21]. Although nephroblastoma overexpressed (Nov, also known as Ccn3) and microfibril-associated glycoprotein 1 (MAGP1) are reported as non-canonical Notch ligands and are expressed in normal articular cartilage, their levels do not increase with the development of OA [21]. Increased expression of Jag1 may be involved in OA; however, its role as an OA trigger is not proven. Considering that articular chondrocytes are not in close contact with each other, ligand-independent activation of Notch signalling or ligand secretion may be associated with OA pathogenesis.
With respect to downstream signalling molecules, Hes1, Hes5, Hes7, Hey1, Hey2, and HeyL are well known to be direct transcriptional targets and mediate the effects of Notch signalling. However, only Hes1 is highly expressed in articular chondrocytes [21, 22]. Hes1 overexpression induces Mmp13 expression as well as Notch-ICD, and knockdown of Hes1 by siRNA cancels the increase of Mmp13 by Notch-ICD [21, 22]. This indicates that the catabolic effect of Notch signalling is mediated by Hes1. Furthermore, Hes1 knockout in adult mouse chondrocytes suppresses surgically induced OA with decreased expression of Mmp13 [22]. Although Hes1 itself is a known transcriptional repressor, it functions as a transcriptional activator in cooperation with calcium/calmodulin-dependent protein kinase II (CaMK2) [23]. In articular cartilage, CaMK2δ is abundantly expressed and required for transcriptional induction of catabolic genes by Hes1 [22]. Microarray analysis and chromatin immunoprecipitation and deep sequencing (ChIP-seq) have revealed that Hes1 induces many inflammation-related molecules such as interleukin (IL)-6 and IL-1 receptor-like 1 (IL1RL1, a receptor of IL-33), in addition to Mmp13 and Adamts5 [22]. Induction of these molecules by the Notch–Hes1 axis seems to be consistent with amelioration of murine collagen-induced arthritis by Notch inhibition, as reported recently by other researchers [24, 25].
In contrast to the mouse surgical OA model data, recent reports have shown that inhibition of Notch signalling results in accelerated OA progression with aging [26, 27]. In one study, Rbpj knockout in limb mesenchymal progenitor cells using Prx1-Cre, or in adult chondrocytes using Col2a1-CreERT2, promoted OA development within 8 months without surgical induction [26]. Similar results have been reported by the same group using Acan-CreERT2 and Rbpj-flox mice [27]. Interestingly, fibrotic cells and high levels of Mmp13 are observed in the superficial zone of Rbpj-knockout cartilage [27].
It is difficult to interpret the contradictory results of OA regulation by Notch signalling. Notch signalling regulates organogenesis by suppressing stem or progenitor cell differentiation [20]. In bone homeostasis, Notch signalling maintains a pool of mesenchymal progenitors in the bone marrow by suppressing osteoblast differentiation [28]. Meanwhile, Notch1 is highly expressed in chondrocytes of the superficial layer, and inhibition of Notch signalling abolishes the colony-forming ability of these cells [29]. Taken together, and including the recent paper highlighting articular chondrocyte turnover [18], we believe Notch signalling contributes to maintain progenitors whilst suppressing the regulation of chondrocyte progenitor differentiation. Furthermore, its mis-activation in differentiated chondrocytes in the middle and deep zones may lead to degeneration (Fig. 1).
OA development is not only caused by abnormalities of the articular cartilage, but also in the synovium, meniscus, ligament, and subchondral bone. Gamma-secretase inhibitor treatment suppresses production of proinflammatory cytokines from the synoviocytes of rheumatoid arthritis patients [25], implying that Notch signalling may contribute to enhanced production of inflammation-related molecules in OA synoviocytes as well as in chondrocytes. Because Notch signalling regulates bone homeostasis through maintenance of mesenchymal progenitors [28], it may be also associated with pathological changes of the subchondral bone, despite Notch signalling in mature osteoblasts not being involved in OA [27].

NF-κB signalling

The NF-κB family has essential roles in a wide range of biological processes including cell survival, proliferation, differentiation, apoptosis, aging, inflammation, and immune responses [3032]. Its members are v-rel reticuloendotheliosis viral oncogene homologue A (RelA, also known as p65), RelB, Rel, p105/p50, and p100/p52, each of which includes a Rel homology domain that mediates DNA binding and dimerization. These proteins form heterodimers and work as transcriptional activators. Inhibitors of NF-κB (IκB) proteins, such as IκBα, IκBβ, IκBγ, IκBε, IκBζ, and Bcl-3, bind to NF-κB family members in the cytoplasm [33]. Activation of IκB kinases (IKKs) in response to several signals phosphorylates IκB proteins and causes their degradation, which enables free NF-κB complexes to translocate from the cytoplasm into the nucleus and trigger target gene transactivation [34, 35]. NF-κB signalling is widely involved in OA pathophysiology through various effects and is activated in OA chondrocytes during aging and inflammation [36]. The NF-κB pathway is essential to induce various inflammation-related factors, including Mmp proteins, inducible nitric oxide synthase (iNOS), IL-1β, and TNF-α, and these induced cytokines further activate the signalling cascade [36].
HIF-2α is a NF-κB-related molecule that is essential for OA development and similar to HIF-1α is a transcription factor of the HIF family. HIF-1α protein is stabilized only under hypoxic conditions, and exerts various effects necessary for cell survival and adaptation in hypovascular and hypoxic tissues. In contrast, HIF-2α is expressed in well-vascularized tissues [37, 38]. HIF proteins share ~50% amino acid homology [39] and accumulating evidence indicates their distinct functions [3943]. HIF-2α expression is increased in the middle and deep zones of articular cartilage [9] and surgically induced OA progression is markedly suppressed in HIF-2α hetero-knockout mice. Furthermore, various catabolic factors such as Mmp13, Mmp9, and vascular endothelial growth factor A (Vegfa) are down-regulated by HIF-2α haploinsufficiency, whilst others are directly induced by HIF-2α [9, 10]. HIF-2α is induced by pro-inflammatory cytokines IL-1β and TNF-α, and its induction is suppressed by treatment with IKK inhibitor [9, 44]. These data, combined with our promoter analyses, indicate that HIF-2α is a direct transcriptional target of NF-κB [9].
In addition to its catabolic effects in chondrocytes, NF-κB signalling plays an essential role in cell survival. We next examined the physiological and pathophysiological roles of NF-κB in articular cartilage. RelA is predominantly localized in the cytoplasm of normal articular chondrocytes and is translocated into the nucleus of OA chondrocytes, accompanied with phosphorylated IκBα [44]. In-vivo analyses using Col2a1-CreERT and Rela-flox mice have revealed that RelA homozygous knockout in chondrocytes after skeletal growth leads to marked acceleration of OA, whereas RelA heterozygous knockout suppresses OA progression [44]. Apoptotic cells are increased in homozygous-knockout cartilage, and anti-apoptotic genes such as Traf2, c-IAP1, and c-IAP2 are down-regulated by RelA deficiency [44]. Notably, HIF-2α expression is suppressed in both homozygous and heterozygous knockout cartilage, but the anti-apoptotic genes are not down-regulated in heterozygous knockout cartilage. Although the anti-apoptotic genes and HIF-2α are transcriptional targets of RelA, the former factors are induced by a smaller amount of RelA than the latter [44]. Thus, NF-κB signalling in chondrocytes is critical for cartilage homeostasis and OA development (Fig. 2).
Numerous studies have revealed that NF-κB signalling is involved in inflammatory responses. Various pro-inflammatory factors secreted from articular chondrocytes and synovial cells modulate OA development, and NF-κB signalling plays essential roles in mediating their effects [45]. In addition, NF-κB signalling regulates some responses to mechanical loading [46]. Dynamic compressive strain enhances phosphorylation of IκBα and subsequent intra-nuclear translocation of RelA in a magnitude-dependent manner without IL-1β treatment, whereas low magnitude strain suppresses this with IL-1β treatment [47]. Considering that excessive mechanical loading is the major risk factor for OA [1], elucidation of the molecular mechanisms underlying the mechano-responses regulated by NF-κB signalling may lead to a more comprehensive understanding of OA pathophysiology. Furthermore, the NF-κB–HIF-2α axis may be a potent therapeutic target because haploinsufficient RelA or a low dose of an IKK inhibitor can suppress HIF-2α expression without significant effect on cell survival [44].

Prospects

Many cells and tissues are regulated by complex multi-functional signalling pathways. In addition to Notch and NF-κB, regulation of articular cartilage by the canonical Wnt signalling pathway remains contradictory. In articular chondrocytes, OA development is enhanced by activation of canonical Wnt signalling in Col2a1-CreERT2;β-cateninfx(ex3)/wt mice [8]; however, it is also enhanced by inhibition of Wnt signalling using Col2a1-ICAT transgenic mice, in which inhibitor of β-catenin and T cell factor (ICAT) is overexpressed under the Col2a1 promoter [4]. In contrast to these data in adult mice, activation of canonical Wnt signalling in chondrocytes enhances superficial zone thickness during the skeletal growth period, while conditional ablation of β-catenin causes the opposite result [48]. These data imply that canonical Wnt signalling exerts various age-dependent and locus-dependent effects. However, the in-depth processes of chondrocyte regulation by the Notch, NF-κB, and Wnt pathways remain ambiguous, and appear to be context dependent. Although these pathways are known to be involved in transcriptional induction of Mmp13 and Adamts5, understanding how they regulate each other is important. Because transcription of Jag1 is enhanced by NF-κB in endothelial cells [49], activation of NF-κB signalling may precede that of Notch signalling. In rheumatoid arthritis model mice, Notch is activated by TNF and inhibits osteoblast differentiation in cooperation with the non-canonical NF-κB proteins p52 and RELB [50]. These findings imply that mutual interactions occur between these pathways during OA development, although the exact mechanisms remain unidentified (Fig. 3). Determination of the signalling networks that regulate maintenance of the progenitors, their differentiation to mature chondrocytes, and turnover of articular chondrocytes will be necessary to better understand OA pathophysiology.
In addition to these findings obtained from molecular biology experiments using murine models, we should address another essential issue: how these signalling pathways regulate human articular cartilage and OA. Many experimental studies using human samples have shown that activation of NF-κB signalling in chondrocytes and synovial cells is closely involved in enhanced production of inflammatory cytokines or catabolic enzymes [51]. Similarly, previous studies have shown enhanced expression of Notch-related molecules in human OA cartilage [52]. These data imply involvement of these signalling pathways in the pathophysiology of both human and murine OA. To date, however, NF-κB-related or Notch-related molecules have not been identified as OA susceptibility genes by previous genome-wide association analyses.

Conclusion

The Notch and NF-κB signalling pathways regulate articular chondrocyte homeostasis and OA development in various ways. Comprehensive understanding of the molecular networks including articular chondrocyte differentiation is needed for further elucidation of OA pathogenesis.

Acknowledgements

Not applicable.

Funding

Not applicable.

Authors’ contributions

TS and ST wrote the manuscript. All authors read and approved the final manuscript.

Availability of data and materials

Not applicable.

Competing interests

The authors declare that they have no competing interests.
Not applicable.
Not applicable.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat Moskowitz RW. The burden of osteoarthritis: clinical and quality-of-life issues. Am J Manag Care. 2009;15(8 Suppl):S223–9.PubMed Moskowitz RW. The burden of osteoarthritis: clinical and quality-of-life issues. Am J Manag Care. 2009;15(8 Suppl):S223–9.PubMed
2.
Zurück zum Zitat Glasson SS, Askew R, Sheppard B, Carito B, Blanchet T, Ma HL, Flannery CR, Peluso D, Kanki K, Yang Z, et al. Deletion of active ADAMTS5 prevents cartilage degradation in a murine model of osteoarthritis. Nature. 2005;434(7033):644–8.CrossRefPubMed Glasson SS, Askew R, Sheppard B, Carito B, Blanchet T, Ma HL, Flannery CR, Peluso D, Kanki K, Yang Z, et al. Deletion of active ADAMTS5 prevents cartilage degradation in a murine model of osteoarthritis. Nature. 2005;434(7033):644–8.CrossRefPubMed
3.
Zurück zum Zitat Kamekura S, Hoshi K, Shimoaka T, Chung U, Chikuda H, Yamada T, Uchida M, Ogata N, Seichi A, Nakamura K, et al. Osteoarthritis development in novel experimental mouse models induced by knee joint instability. Osteoarthritis Cartilage. 2005;13(7):632–41.CrossRefPubMed Kamekura S, Hoshi K, Shimoaka T, Chung U, Chikuda H, Yamada T, Uchida M, Ogata N, Seichi A, Nakamura K, et al. Osteoarthritis development in novel experimental mouse models induced by knee joint instability. Osteoarthritis Cartilage. 2005;13(7):632–41.CrossRefPubMed
4.
Zurück zum Zitat Zhu M, Chen M, Zuscik M, Wu Q, Wang YJ, Rosier RN, O’Keefe RJ, Chen D. Inhibition of beta-catenin signaling in articular chondrocytes results in articular cartilage destruction. Arthritis Rheum. 2008;58(7):2053–64.CrossRefPubMedPubMedCentral Zhu M, Chen M, Zuscik M, Wu Q, Wang YJ, Rosier RN, O’Keefe RJ, Chen D. Inhibition of beta-catenin signaling in articular chondrocytes results in articular cartilage destruction. Arthritis Rheum. 2008;58(7):2053–64.CrossRefPubMedPubMedCentral
5.
Zurück zum Zitat Echtermeyer F, Bertrand J, Dreier R, Meinecke I, Neugebauer K, Fuerst M, Lee YJ, Song YW, Herzog C, Theilmeier G, et al. Syndecan-4 regulates ADAMTS-5 activation and cartilage breakdown in osteoarthritis. Nat Med. 2009;15(9):1072–6.CrossRefPubMed Echtermeyer F, Bertrand J, Dreier R, Meinecke I, Neugebauer K, Fuerst M, Lee YJ, Song YW, Herzog C, Theilmeier G, et al. Syndecan-4 regulates ADAMTS-5 activation and cartilage breakdown in osteoarthritis. Nat Med. 2009;15(9):1072–6.CrossRefPubMed
6.
Zurück zum Zitat Lin AC, Seeto BL, Bartoszko JM, Khoury MA, Whetstone H, Ho L, Hsu C, Ali SA, Alman BA. Modulating hedgehog signaling can attenuate the severity of osteoarthritis. Nat Med. 2009;15(12):1421–5.CrossRefPubMed Lin AC, Seeto BL, Bartoszko JM, Khoury MA, Whetstone H, Ho L, Hsu C, Ali SA, Alman BA. Modulating hedgehog signaling can attenuate the severity of osteoarthritis. Nat Med. 2009;15(12):1421–5.CrossRefPubMed
7.
Zurück zum Zitat Little CB, Barai A, Burkhardt D, Smith SM, Fosang AJ, Werb Z, Shah M, Thompson EW. Matrix metalloproteinase 13-deficient mice are resistant to osteoarthritic cartilage erosion but not chondrocyte hypertrophy or osteophyte development. Arthritis Rheum. 2009;60(12):3723–33.CrossRefPubMedPubMedCentral Little CB, Barai A, Burkhardt D, Smith SM, Fosang AJ, Werb Z, Shah M, Thompson EW. Matrix metalloproteinase 13-deficient mice are resistant to osteoarthritic cartilage erosion but not chondrocyte hypertrophy or osteophyte development. Arthritis Rheum. 2009;60(12):3723–33.CrossRefPubMedPubMedCentral
8.
Zurück zum Zitat Zhu M, Tang D, Wu Q, Hao S, Chen M, Xie C, Rosier RN, O’Keefe RJ, Zuscik M, Chen D. Activation of beta-catenin signaling in articular chondrocytes leads to osteoarthritis-like phenotype in adult beta-catenin conditional activation mice. J Bone Miner Res. 2009;24(1):12–21.CrossRefPubMed Zhu M, Tang D, Wu Q, Hao S, Chen M, Xie C, Rosier RN, O’Keefe RJ, Zuscik M, Chen D. Activation of beta-catenin signaling in articular chondrocytes leads to osteoarthritis-like phenotype in adult beta-catenin conditional activation mice. J Bone Miner Res. 2009;24(1):12–21.CrossRefPubMed
9.
Zurück zum Zitat Saito T, Fukai A, Mabuchi A, Ikeda T, Yano F, Ohba S, Nishida N, Akune T, Yoshimura N, Nakagawa T, et al. Transcriptional regulation of endochondral ossification by HIF-2alpha during skeletal growth and osteoarthritis development. Nat Med. 2010;16(6):678–86.CrossRefPubMed Saito T, Fukai A, Mabuchi A, Ikeda T, Yano F, Ohba S, Nishida N, Akune T, Yoshimura N, Nakagawa T, et al. Transcriptional regulation of endochondral ossification by HIF-2alpha during skeletal growth and osteoarthritis development. Nat Med. 2010;16(6):678–86.CrossRefPubMed
10.
Zurück zum Zitat Yang S, Kim J, Ryu JH, Oh H, Chun CH, Kim BJ, Min BH, Chun JS. Hypoxia-inducible factor-2alpha is a catabolic regulator of osteoarthritic cartilage destruction. Nat Med. 2010;16(6):687–93.CrossRefPubMed Yang S, Kim J, Ryu JH, Oh H, Chun CH, Kim BJ, Min BH, Chun JS. Hypoxia-inducible factor-2alpha is a catabolic regulator of osteoarthritic cartilage destruction. Nat Med. 2010;16(6):687–93.CrossRefPubMed
12.
Zurück zum Zitat Gossan N, Boot-Handford R, Meng QJ. Ageing and osteoarthritis: a circadian rhythm connection. Biogerontology. 2015;16(2):209–19.CrossRefPubMed Gossan N, Boot-Handford R, Meng QJ. Ageing and osteoarthritis: a circadian rhythm connection. Biogerontology. 2015;16(2):209–19.CrossRefPubMed
13.
14.
Zurück zum Zitat Mobasheri A, Matta C, Zakany R, Musumeci G. Chondrosenescence: definition, hallmarks and potential role in the pathogenesis of osteoarthritis. Maturitas. 2015;80(3):237–44.CrossRefPubMed Mobasheri A, Matta C, Zakany R, Musumeci G. Chondrosenescence: definition, hallmarks and potential role in the pathogenesis of osteoarthritis. Maturitas. 2015;80(3):237–44.CrossRefPubMed
15.
Zurück zum Zitat Asahara H. Current status and strategy of microRNA research for cartilage development and osteoarthritis pathogenesis. J Bone Metab. 2016;23(3):121–7.CrossRefPubMedPubMedCentral Asahara H. Current status and strategy of microRNA research for cartilage development and osteoarthritis pathogenesis. J Bone Metab. 2016;23(3):121–7.CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat Bouderlique T, Vuppalapati KK, Newton PT, Li L, Barenius B, Chagin AS. Targeted deletion of Atg5 in chondrocytes promotes age-related osteoarthritis. Ann Rheum Dis. 2016;75(3):627–31.CrossRefPubMed Bouderlique T, Vuppalapati KK, Newton PT, Li L, Barenius B, Chagin AS. Targeted deletion of Atg5 in chondrocytes promotes age-related osteoarthritis. Ann Rheum Dis. 2016;75(3):627–31.CrossRefPubMed
18.
Zurück zum Zitat Kozhemyakina E, Zhang M, Ionescu A, Ayturk UM, Ono N, Kobayashi A, Kronenberg H, Warman ML, Lassar AB. Identification of a Prg4-expressing articular cartilage progenitor cell population in mice. Arthritis Rheumatol. 2015;67(5):1261–73.CrossRefPubMedPubMedCentral Kozhemyakina E, Zhang M, Ionescu A, Ayturk UM, Ono N, Kobayashi A, Kronenberg H, Warman ML, Lassar AB. Identification of a Prg4-expressing articular cartilage progenitor cell population in mice. Arthritis Rheumatol. 2015;67(5):1261–73.CrossRefPubMedPubMedCentral
19.
Zurück zum Zitat Yoon K, Gaiano N. Notch signaling in the mammalian central nervous system: insights from mouse mutants. Nat Neurosci. 2005;8(6):709–15.CrossRefPubMed Yoon K, Gaiano N. Notch signaling in the mammalian central nervous system: insights from mouse mutants. Nat Neurosci. 2005;8(6):709–15.CrossRefPubMed
21.
Zurück zum Zitat Hosaka Y, Saito T, Sugita S, Hikata T, Kobayashi H, Fukai A, Taniguchi Y, Hirata M, Akiyama H, Chung UI, et al. Notch signaling in chondrocytes modulates endochondral ossification and osteoarthritis development. Proc Natl Acad Sci U S A. 2013;110(5):1875–80.CrossRefPubMedPubMedCentral Hosaka Y, Saito T, Sugita S, Hikata T, Kobayashi H, Fukai A, Taniguchi Y, Hirata M, Akiyama H, Chung UI, et al. Notch signaling in chondrocytes modulates endochondral ossification and osteoarthritis development. Proc Natl Acad Sci U S A. 2013;110(5):1875–80.CrossRefPubMedPubMedCentral
22.
Zurück zum Zitat Sugita S, Hosaka Y, Okada K, Mori D, Yano F, Kobayashi H, Taniguchi Y, Mori Y, Okuma T, Chang SH, et al. Transcription factor Hes1 modulates osteoarthritis development in cooperation with calcium/calmodulin-dependent protein kinase 2. Proc Natl Acad Sci U S A. 2015;112(10):3080–5.CrossRefPubMedPubMedCentral Sugita S, Hosaka Y, Okada K, Mori D, Yano F, Kobayashi H, Taniguchi Y, Mori Y, Okuma T, Chang SH, et al. Transcription factor Hes1 modulates osteoarthritis development in cooperation with calcium/calmodulin-dependent protein kinase 2. Proc Natl Acad Sci U S A. 2015;112(10):3080–5.CrossRefPubMedPubMedCentral
23.
Zurück zum Zitat Ju BG, Solum D, Song EJ, Lee KJ, Rose DW, Glass CK, Rosenfeld MG. Activating the PARP-1 sensor component of the groucho/TLE1 corepressor complex mediates a CaMKinase IIdelta-dependent neurogenic gene activation pathway. Cell. 2004;119(6):815–29.CrossRefPubMed Ju BG, Solum D, Song EJ, Lee KJ, Rose DW, Glass CK, Rosenfeld MG. Activating the PARP-1 sensor component of the groucho/TLE1 corepressor complex mediates a CaMKinase IIdelta-dependent neurogenic gene activation pathway. Cell. 2004;119(6):815–29.CrossRefPubMed
24.
Zurück zum Zitat Jiao Z, Wang W, Hua S, Liu M, Wang H, Wang X, Chen Y, Xu H, Lu L. Blockade of Notch signaling ameliorates murine collagen-induced arthritis via suppressing Th1 and Th17 cell responses. Am J Pathol. 2014;184(4):1085–93.CrossRefPubMed Jiao Z, Wang W, Hua S, Liu M, Wang H, Wang X, Chen Y, Xu H, Lu L. Blockade of Notch signaling ameliorates murine collagen-induced arthritis via suppressing Th1 and Th17 cell responses. Am J Pathol. 2014;184(4):1085–93.CrossRefPubMed
25.
Zurück zum Zitat Park JS, Kim SH, Kim K, Jin CH, Choi KY, Jang J, Choi Y, Gwon AR, Baik SH, Yun UJ, et al. Inhibition of notch signalling ameliorates experimental inflammatory arthritis. Ann Rheum Dis. 2015;74(1):267–74.CrossRefPubMed Park JS, Kim SH, Kim K, Jin CH, Choi KY, Jang J, Choi Y, Gwon AR, Baik SH, Yun UJ, et al. Inhibition of notch signalling ameliorates experimental inflammatory arthritis. Ann Rheum Dis. 2015;74(1):267–74.CrossRefPubMed
26.
Zurück zum Zitat Mirando AJ, Liu Z, Moore T, Lang A, Kohn A, Osinski AM, O’Keefe RJ, Mooney RA, Zuscik MJ, Hilton MJ. RBP-Jkappa-dependent Notch signaling is required for murine articular cartilage and joint maintenance. Arthritis Rheum. 2013;65(10):2623–33.PubMedPubMedCentral Mirando AJ, Liu Z, Moore T, Lang A, Kohn A, Osinski AM, O’Keefe RJ, Mooney RA, Zuscik MJ, Hilton MJ. RBP-Jkappa-dependent Notch signaling is required for murine articular cartilage and joint maintenance. Arthritis Rheum. 2013;65(10):2623–33.PubMedPubMedCentral
27.
Zurück zum Zitat Liu Z, Ren Y, Mirando AJ, Wang C, Zuscik MJ, O’Keefe RJ, Hilton MJ. Notch signaling in postnatal joint chondrocytes, but not subchondral osteoblasts, is required for articular cartilage and joint maintenance. Osteoarthritis Cartilage. 2016;24(4):740–51.CrossRefPubMed Liu Z, Ren Y, Mirando AJ, Wang C, Zuscik MJ, O’Keefe RJ, Hilton MJ. Notch signaling in postnatal joint chondrocytes, but not subchondral osteoblasts, is required for articular cartilage and joint maintenance. Osteoarthritis Cartilage. 2016;24(4):740–51.CrossRefPubMed
28.
Zurück zum Zitat Hilton MJ, Tu X, Wu X, Bai S, Zhao H, Kobayashi T, Kronenberg HM, Teitelbaum SL, Ross FP, Kopan R, et al. Notch signaling maintains bone marrow mesenchymal progenitors by suppressing osteoblast differentiation. Nat Med. 2008;14(3):306–14.CrossRefPubMedPubMedCentral Hilton MJ, Tu X, Wu X, Bai S, Zhao H, Kobayashi T, Kronenberg HM, Teitelbaum SL, Ross FP, Kopan R, et al. Notch signaling maintains bone marrow mesenchymal progenitors by suppressing osteoblast differentiation. Nat Med. 2008;14(3):306–14.CrossRefPubMedPubMedCentral
29.
Zurück zum Zitat Dowthwaite GP, Bishop JC, Redman SN, Khan IM, Rooney P, Evans DJ, Haughton L, Bayram Z, Boyer S, Thomson B, et al. The surface of articular cartilage contains a progenitor cell population. J Cell Sci. 2004;117(Pt 6):889–97.CrossRefPubMed Dowthwaite GP, Bishop JC, Redman SN, Khan IM, Rooney P, Evans DJ, Haughton L, Bayram Z, Boyer S, Thomson B, et al. The surface of articular cartilage contains a progenitor cell population. J Cell Sci. 2004;117(Pt 6):889–97.CrossRefPubMed
30.
Zurück zum Zitat Bonizzi G, Karin M. The two NF-kappaB activation pathways and their role in innate and adaptive immunity. Trends Immunol. 2004;25(6):280–8.CrossRefPubMed Bonizzi G, Karin M. The two NF-kappaB activation pathways and their role in innate and adaptive immunity. Trends Immunol. 2004;25(6):280–8.CrossRefPubMed
31.
Zurück zum Zitat Li Q, Withoff S, Verma IM. Inflammation-associated cancer: NF-kappaB is the lynchpin. Trends Immunol. 2005;26(6):318–25.CrossRefPubMed Li Q, Withoff S, Verma IM. Inflammation-associated cancer: NF-kappaB is the lynchpin. Trends Immunol. 2005;26(6):318–25.CrossRefPubMed
32.
Zurück zum Zitat Meffert MK, Baltimore D. Physiological functions for brain NF-kappaB. Trends Neurosci. 2005;28(1):37–43.CrossRefPubMed Meffert MK, Baltimore D. Physiological functions for brain NF-kappaB. Trends Neurosci. 2005;28(1):37–43.CrossRefPubMed
33.
Zurück zum Zitat Neumann M, Naumann M. Beyond IkappaBs: alternative regulation of NF-kappaB activity. FASEB J. 2007;21(11):2642–54.CrossRefPubMed Neumann M, Naumann M. Beyond IkappaBs: alternative regulation of NF-kappaB activity. FASEB J. 2007;21(11):2642–54.CrossRefPubMed
34.
Zurück zum Zitat Chen LF, Greene WC. Shaping the nuclear action of NF-kappaB. Nat Rev Mol Cell Biol. 2004;5(5):392–401.CrossRefPubMed Chen LF, Greene WC. Shaping the nuclear action of NF-kappaB. Nat Rev Mol Cell Biol. 2004;5(5):392–401.CrossRefPubMed
36.
Zurück zum Zitat Marcu KB, Otero M, Olivotto E, Borzi RM, Goldring MB. NF-kappaB signaling: multiple angles to target OA. Curr Drug Targets. 2010;11(5):599–613.CrossRefPubMedPubMedCentral Marcu KB, Otero M, Olivotto E, Borzi RM, Goldring MB. NF-kappaB signaling: multiple angles to target OA. Curr Drug Targets. 2010;11(5):599–613.CrossRefPubMedPubMedCentral
37.
Zurück zum Zitat Semenza GL. HIF-1 and human disease: one highly involved factor. Genes Dev. 2000;14(16):1983–91.PubMed Semenza GL. HIF-1 and human disease: one highly involved factor. Genes Dev. 2000;14(16):1983–91.PubMed
38.
Zurück zum Zitat Stewart AJ, Houston B, Farquharson C. Elevated expression of hypoxia inducible factor-2alpha in terminally differentiating growth plate chondrocytes. J Cell Physiol. 2006;206(2):435–40.CrossRefPubMed Stewart AJ, Houston B, Farquharson C. Elevated expression of hypoxia inducible factor-2alpha in terminally differentiating growth plate chondrocytes. J Cell Physiol. 2006;206(2):435–40.CrossRefPubMed
39.
Zurück zum Zitat Tian H, Hammer RE, Matsumoto AM, Russell DW, McKnight SL. The hypoxia-responsive transcription factor EPAS1 is essential for catecholamine homeostasis and protection against heart failure during embryonic development. Genes Dev. 1998;12(21):3320–4.CrossRefPubMedPubMedCentral Tian H, Hammer RE, Matsumoto AM, Russell DW, McKnight SL. The hypoxia-responsive transcription factor EPAS1 is essential for catecholamine homeostasis and protection against heart failure during embryonic development. Genes Dev. 1998;12(21):3320–4.CrossRefPubMedPubMedCentral
40.
Zurück zum Zitat Jain S, Maltepe E, Lu MM, Simon C, Bradfield CA. Expression of ARNT, ARNT2, HIF1 alpha, HIF2 alpha and Ah receptor mRNAs in the developing mouse. Mech Dev. 1998;73(1):117–23.CrossRefPubMed Jain S, Maltepe E, Lu MM, Simon C, Bradfield CA. Expression of ARNT, ARNT2, HIF1 alpha, HIF2 alpha and Ah receptor mRNAs in the developing mouse. Mech Dev. 1998;73(1):117–23.CrossRefPubMed
41.
Zurück zum Zitat O’Rourke JF, Tian YM, Ratcliffe PJ, Pugh CW. Oxygen-regulated and transactivating domains in endothelial PAS protein 1: comparison with hypoxia-inducible factor-1alpha. J Biol Chem. 1999;274(4):2060–71.CrossRefPubMed O’Rourke JF, Tian YM, Ratcliffe PJ, Pugh CW. Oxygen-regulated and transactivating domains in endothelial PAS protein 1: comparison with hypoxia-inducible factor-1alpha. J Biol Chem. 1999;274(4):2060–71.CrossRefPubMed
42.
Zurück zum Zitat Scortegagna M, Ding K, Oktay Y, Gaur A, Thurmond F, Yan LJ, Marck BT, Matsumoto AM, Shelton JM, Richardson JA, et al. Multiple organ pathology, metabolic abnormalities and impaired homeostasis of reactive oxygen species in Epas1-/- mice. Nat Genet. 2003;35(4):331–40.CrossRefPubMed Scortegagna M, Ding K, Oktay Y, Gaur A, Thurmond F, Yan LJ, Marck BT, Matsumoto AM, Shelton JM, Richardson JA, et al. Multiple organ pathology, metabolic abnormalities and impaired homeostasis of reactive oxygen species in Epas1-/- mice. Nat Genet. 2003;35(4):331–40.CrossRefPubMed
44.
Zurück zum Zitat Kobayashi H, Chang SH, Mori D, Itoh S, Hirata M, Hosaka Y, Taniguchi Y, Okada K, Mori Y, Yano F, et al. Biphasic regulation of chondrocytes by Rela through induction of anti-apoptotic and catabolic target genes. Nat Commun. 2016;7:13336.CrossRefPubMedPubMedCentral Kobayashi H, Chang SH, Mori D, Itoh S, Hirata M, Hosaka Y, Taniguchi Y, Okada K, Mori Y, Yano F, et al. Biphasic regulation of chondrocytes by Rela through induction of anti-apoptotic and catabolic target genes. Nat Commun. 2016;7:13336.CrossRefPubMedPubMedCentral
47.
Zurück zum Zitat Nam J, Aguda BD, Rath B, Agarwal S. Biomechanical thresholds regulate inflammation through the NF-kappaB pathway: experiments and modeling. PLoS One. 2009;4(4), e5262.CrossRefPubMedPubMedCentral Nam J, Aguda BD, Rath B, Agarwal S. Biomechanical thresholds regulate inflammation through the NF-kappaB pathway: experiments and modeling. PLoS One. 2009;4(4), e5262.CrossRefPubMedPubMedCentral
48.
Zurück zum Zitat Yasuhara R, Ohta Y, Yuasa T, Kondo N, Hoang T, Addya S, Fortina P, Pacifici M, Iwamoto M, Enomoto-Iwamoto M. Roles of beta-catenin signaling in phenotypic expression and proliferation of articular cartilage superficial zone cells. Lab Invest. 2011;91(12):1739–52.CrossRefPubMedPubMedCentral Yasuhara R, Ohta Y, Yuasa T, Kondo N, Hoang T, Addya S, Fortina P, Pacifici M, Iwamoto M, Enomoto-Iwamoto M. Roles of beta-catenin signaling in phenotypic expression and proliferation of articular cartilage superficial zone cells. Lab Invest. 2011;91(12):1739–52.CrossRefPubMedPubMedCentral
50.
Zurück zum Zitat Zhang H, Hilton MJ, Anolik JH, Welle SL, Zhao C, Yao Z, Li X, Wang Z, Boyce BF, Xing L. NOTCH inhibits osteoblast formation in inflammatory arthritis via noncanonical NF-kappaB. J Clin Invest. 2014;124(7):3200–14.CrossRefPubMedPubMedCentral Zhang H, Hilton MJ, Anolik JH, Welle SL, Zhao C, Yao Z, Li X, Wang Z, Boyce BF, Xing L. NOTCH inhibits osteoblast formation in inflammatory arthritis via noncanonical NF-kappaB. J Clin Invest. 2014;124(7):3200–14.CrossRefPubMedPubMedCentral
51.
Zurück zum Zitat Olivotto E, Otero M, Marcu KB, Goldring MB. Pathophysiology of osteoarthritis: canonical NF-kappaB/IKKbeta-dependent and kinase-independent effects of IKKalpha in cartilage degradation and chondrocyte differentiation. RMD Open. 2015;1 Suppl 1:e000061.CrossRefPubMedPubMedCentral Olivotto E, Otero M, Marcu KB, Goldring MB. Pathophysiology of osteoarthritis: canonical NF-kappaB/IKKbeta-dependent and kinase-independent effects of IKKalpha in cartilage degradation and chondrocyte differentiation. RMD Open. 2015;1 Suppl 1:e000061.CrossRefPubMedPubMedCentral
52.
Zurück zum Zitat Sassi N, Laadhar L, Driss M, Kallel-Sellami M, Sellami S, Makni S. The role of the Notch pathway in healthy and osteoarthritic articular cartilage: from experimental models to ex vivo studies. Arthritis Res Ther. 2011;13(2):208.CrossRefPubMedPubMedCentral Sassi N, Laadhar L, Driss M, Kallel-Sellami M, Sellami S, Makni S. The role of the Notch pathway in healthy and osteoarthritic articular cartilage: from experimental models to ex vivo studies. Arthritis Res Ther. 2011;13(2):208.CrossRefPubMedPubMedCentral
Metadaten
Titel
Molecular mechanisms underlying osteoarthritis development: Notch and NF-κB
verfasst von
Taku Saito
Sakae Tanaka
Publikationsdatum
01.12.2017
Verlag
BioMed Central
Erschienen in
Arthritis Research & Therapy / Ausgabe 1/2017
Elektronische ISSN: 1478-6362
DOI
https://doi.org/10.1186/s13075-017-1296-y

Weitere Artikel der Ausgabe 1/2017

Arthritis Research & Therapy 1/2017 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.