Open access peer-reviewed chapter

Pharmacologic Interventions for Preventing Chondrocyte Apoptosis in Rheumatoid Arthritis and Osteoarthritis

Written By

Charles J. Malemud

Submitted: 11 July 2017 Reviewed: 15 December 2017 Published: 19 September 2018

DOI: 10.5772/intechopen.73174

From the Edited Volume

Drug Discovery - Concepts to Market

Edited by Varaprasad Bobbarala

Chapter metrics overview

1,203 Chapter Downloads

View Full Metrics

Abstract

Chronic inflammation drives the progression of rheumatoid arthritis (RA) and osteoarthritis (OA) to synovial joint failure. The inflammatory state in both musculoskeletal diseases is associated with significantly elevated levels of pro-inflammatory cytokines in joint synovial fluid, which is best exemplified by increases in interleukin-1β (IL-1β), IL-6, IL-17, tumor necrosis factor-α, among others, as well as increased activity of soluble mediators such as nitric oxide and certain growth factors including vascular endothelial growth factor and fibroblast growth factor. The multitude of these factors activate chondrocyte signal transduction pathways resulting in programmed cell death, otherwise known as apoptosis as well as compromising chondrocyte autophagy. Importantly, chondrocyte apoptosis causes a loss of articular cartilage vitality which dampens cartilage repair mechanisms because at present, the possibility that chondrocyte progenitor cells could replace those differentiated chondrocytes lost via apoptosis remains debatable. Certain pharmacologic interventions which have been proven to induce apoptosis in various cancer cell studies in vitro suggest the possibility that drugs could be developed to specifically suppress or completely inhibit chondrocyte apoptosis in RA and OA cartilage. This review supports that contention and indicates that apoptosis can be inhibited by identifying novel cellular targets which promote apoptosis and autophagy.

Keywords

  • apoptosis
  • chondrocyte
  • osteoarthritis
  • rheumatoid arthritis
  • signal transduction

1. Introduction

Controlled cell death otherwise known as programmed cell death or apoptosis constitutes a critical event which is germane to the normal development of the immune system, the appropriate integration of cells within tissues and organs and organ homeostasis [1, 2, 3, 4, 5, 6, 7]. However, the aberrant frequency of apoptotic cells can compromise normal tissue architecture and, in doing so, contribute to the loss of cell vitality [8, 9, 10]. This is especially the case in explaining the loss of chondrocyte viability in arthritic conditions of synovial joints, such as rheumatoid arthritis (RA) and osteoarthritis (OA).

Significant progress has been achieved over the previous decade or so in furthering our understanding of the cellular and molecular events that trigger the increased frequency of chondrocyte apoptosis in RA and OA [11, 12, 13, 14, 15, 16]. These advances include (1) an appreciation that the significant increase in the levels of pro-inflammatory cytokines in synovial fluid from RA and OA patients also can induce chondrocyte apoptosis in vitro [17, 18, 19, 20, 21]; (2) that the elevated frequency of chondrocyte apoptosis by these cytokines is deregulated by altered signal transduction which can involve continuous activation of stress-activated/mitogen-activated protein kinases (SAPK/MAPK) [22, 23, 24, 25], the Janus kinase/Signal Transducers and Activators of Transcription (JAK/STAT) pathway [26, 27, 28, 29, 30, 31], the phosphatidylinositide-3-kinase/Akt/mammalian target of rapamycin (PI3K/Akt/mTOR) pathway [32, 33, 34, 35, 36], and other protein kinase pathways [37, 38, 39, 40]; (3) that specific co-factors are capable of regulating the activation of these signaling pathways [41, 42, 43, 44]; and (4) that micro-RNAs (mi-RNAs) can regulate the activity of these co-factors and, in this manner, control the induction of apoptosis via these signaling pathways [45, 46, 47, 48].

Furthermore, the increased frequency of chondrocyte apoptosis now confirmed in guinea pig OA cartilage [49] as well as human RA and OA articular cartilage [50, 51, 52, 53] presents a particularly onerous scenario for the survival of synovial joints under these conditions. Although a population of chondroprogenitor cells was identified in several studies of adult normal and diseased articular cartilage [54, 55, 56], significant repair of damaged articular cartilage in RA and OA by these cells is effete. This effect on cartilage repair may result from elevated levels of pro-inflammatory cytokines such as IL-17, which was recently shown to inhibit the chondrocyte maturation lineage emanating from progenitor cells in RA [57]. Therefore, even if chondrocyte precursor cells exist in adult articular cartilage which could potentially become authentic chondrocytes lost from articular cartilage via apoptosis or chondrocytes lost via diffusion of cartilage extracellular matrix fragments into synovial fluid, the reduction in chondrocyte vitality via apoptosis would be a challenging event to overcome, especially in a synovial joint microenvironment replete with pro-inflammatory cytokines.

In this chapter, we have systematically examined the mechanistic underpinning for identifying novel targets in order to suppress or even inhibit chondrocyte apoptosis in RA and OA. However, the scenario in RA, in particular, is even more complex than in OA because in the context of devising therapeutic strategies designed to inhibit chondrocyte apoptosis in RA, one must also take into account the fact that in the hyperplastic RA synovial tissue, comprised of activated synoviocytes, immune cells, macrophages and other inflammatory cells are generally considered to be relatively “apoptosis-resistant” [58, 59]. Thus, this characteristic of the RA joint ensures a plentiful source of immune-mediated cells and non-immune inflammatory cells which drive the progression of RA. It is also likely that at some time during the course of the progression of OA, immune-mediated inflammation may also cause a similar chronic inflammatory microenvironment, as found in RA, to arise and persist in OA synovial tissue [55] resulting, in part, in an increased frequency of apoptotic chondrocytes [60].

Advertisement

2. Compelling evidence that many factors relevant to RA and OA promote or induce chondrocyte apoptosis in vitro

Analyses of synovial fluids and sera from RA and OA patients with active disease showed that these samples contained significantly elevated levels of various pro-inflammatory cytokines and growth factors when compared to a control group [52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68]. Of note, incubation of rat [69], non-arthritic or human chondrocytes from OA cartilage [70, 71, 72, 73, 74, 75, 76] or immortalized lines of human chondrocytes [77] with physiological levels of these cytokines, growth factors (e.g., VEGF and FGF) or additional soluble mediators (e.g., nitric oxide) were shown to induce apoptosis, which was accompanied by activation of SAPK/MAPK, JAK/STAT or PI3K/Akt/mTor signaling in these cells [78, 79, 80, 81]. In addition, mediators of inflammation, including prostaglandin E2 and neuropeptides (e.g., Substance P), are also implicated in perpetuating chronic inflammation [38]. The induction of apoptosis was also shown to be related to altered levels of various down-regulators of apoptosis. These included BCL-2-like protein-11 (Bim) [18], B-cell lymphoma-2 (Bcl-2) [75], cell-derived inhibitors of apoptosis proteins (IAPs) [81, 82, 83, 84] and Suppressor of Cytokine Synthesis (SOCS) [85, 86]. Furthermore, alterations in the functions of mitochondria [87] and endoplasmic reticulum (ER) [88, 89, 90, 91] related to cell stress, the generation of reactive oxygen species [92] and the recently described advanced oxidation protein products [93] with respect to their capacity to induce apoptosis were reported as well. Taken together, these results provided compelling evidence that pro-inflammatory cytokines, growth factors and soluble mediators germane to the progression of RA and OA are responsible for inducing chondrocyte apoptosis in these conditions.

Advertisement

3. The relationship between apoptosis and autophagy

A recent review of RA pathogenesis by Angelotti et al. [94] emphasized the view that numerous cells comprising the innate immune system, including macrophages, dendritic cells, mast cells, natural killer cells and neutrophils as well as T-cells and B-cells, regulators of adaptive immunity, are primarily responsible for perpetuating the state of chronic inflammation through their capacity to alter the survival of resident synovial fibroblasts. It is also likely that such cell combinations are also involved in the progression of OA as well, as evidenced by the skewing of cartilage homeostasis toward catabolism, and the loss of chondrocyte vitality via the induction of apoptosis [95, 96, 97]. However, it was previously shown that the classical apoptosis cascade can be activated by various pro-inflammatory cytokines, and certain growth factors, chemokines, chemokine receptors (Table 1) and other interleukins, including IL-8 (CXCL8) and adhesion molecules [64]. Thus, these factors are likely to be the most influential in inducing apoptosis pathway RA. However, a non-classical form of apoptosis, termed, “chondroptosis” is just as likely to be activated in OA. In that regard, “chondroptosis” involves an increase in the number of ER and Golgi apparatus reflecting an increase in protein synthesis that accompanies the loss of viable chondrocytes [98].

Cytokine, chemokine, chemokine receptors and growth factorsReference
IL-17, IL-18BP[62]
IL-17, IL-20, IL-21[63]
IL-7[64]
IL-12/IL-23[64]
IL-15/IL-16[64]
IL-17/IL-18[64]
IL-19/-20/-21[64]
IL-32[64]
VEGF1; TGF-β12; Leptin; FGF3[64, 65, 66]
CXCR3; CXCR4,-5; CXCR-1,-5,-6; IL-8; MIP-1α4; GRO-α5(CXCL1); GRO-βγ6; MCP-17; RANTES8; Eotaxin-1[65, 68]
IL-6[64, 67, 68]
TNF-α[64, 67, 69]
IL-1β[64]

Table 1.

Cytokines, chemokines, chemokine receptors and growth factors in RA and OA.

Vascular endothelial growth factor.


Transforming growth factor-β1.


Fibroblast growth factor.


Macrophage inflammatory protein-1 (CCL3).


Growth-related oncogene-α.


GRO-βγ.


Monocyte chemotactic protein-1.


Regulated on activation, normal T-cell expressed and secreted.


Furthermore, changes in the number of viable articular chondrocytes in experimentally induced arthritis [99], human RA [100], and OA are almost certainly associated with the autophagic-mediated cell death of chondrocytes [101, 102, 103, 104, 105], which occurs in concert with the activation of the extrinsic apoptosis pathway, the latter mediated by Tumor necrosis Factor-Related Apoptosis-Inducing Ligand (TRAIL), Death Receptor-5 (DR5) and caspase-3 [106, 107]. In that regard, it was noteworthy that Huang et al. [108] reported that exogenous leptin promoted chondrocyte apoptosis while inhibiting chondrocyte autophagy via the up-regulation of lysyl oxidase-like 3 (LOXL3). Thus, in their study [108], overexpression of LOXL3 inhibited chondrocyte autophagy by activating mechanistic target of rapamycin complex-1 (mTORC1) [36]. In contrast, cartilage-specific deletion of mTOR resulted in the up-regulation of autophagy [102]. Autophagy protected mouse cartilage from degeneration [109]. Autophagy was also shown to protect chondrocytes from glucocorticoid-induced apoptosis via reactive oxygen species, Akt and FOXO3 signaling [110] as well as from advanced glycation end-product-induced apoptosis which was accompanied by lower levels of MMP-3 and MMP-13 in rat chondrocyte cultures [111].

Advertisement

4. Do micro-RNAs play a critical role in chondrocyte apoptosis?

Micro-RNAs (miRs) are critical mediators of mRNA degradation as well acting as repressors of translation. MiRs have been implicated in the development of skeletal long bones via their multiple effects on osteogenesis [112]. However, recent evidence has also improved the recognized role of miRs in RA pathology as a result of evidence that many miRs including miR-16, miR-146a/b, miR-150, miR-155 and miR-223 are over-expressed both in the peripheral circulation of RA patients and in the RA synovial joint [113], although other miRs relevant to RA, such as miR-21, miR-125a, miR-223, and miR-451 are principally found to be at elevated levels in the plasma and sera of RA patients.

Additional evidence has been presented to show that several of these miRs also regulate apoptosis. For example, the level of the pro-apoptotic protein, Bim, was increased when activated T-cells were incubated with a repressor of miR-148a resulting in an increase in the Th1 apoptotic response [114]. Thus, accumulating evidence showed that both the immune cells and chondrocyte apoptotic and autophagic response can be manipulated by either experimental overexpression or repression of various miRs which are relevant to OA [115] and RA [116] pathophysiology.

The results of numerous studies have confirmed the role of specific miRs as directly affecting apoptosis or indirectly affecting apoptosis through their activity on other molecules that regulate chondrocyte apoptosis. For example, mIR-146a was reported to be over-expressed in OA [117]. In that regard, mIR-146a was shown to promote human OA chondrocyte proliferation and to inhibit apoptosis by targeting tumor necrosis factor receptor-associated factor 6 (TRAF6) via NF-κB [118, 119]. By contrast, mIR-146a was also shown to target IL-1β, to induce VEGF production, and to promote rat chondrocyte apoptosis via Smad4 [120]. On the other hand, silencing of miR-34a inhibited rat chondrocyte apoptosis [121], whereas overexpression of mIR-34a promoted apoptosis in normal human chondrocytes by targeting SIRT1/p53 signaling [122], although other evidences indicated that miR-34a was increased in intervertebral disc degeneration and was associated with an elevated frequency of apoptotic cartilage end plate chondrocytes [123]. In another study, glycerol-3-phosphate dehydrogenase 1-like protein was shown to be a target for mIR-181a, which is deregulated in OA wherein human chondrocyte apoptosis was increased [124]. In yet another study, phosphatase and tensin homolog deleted on chromosome 10 (PTEN) [36] was identified as the target for mIR-181 whereby mIR-181 up-regulated the expression of proteins associated with apoptosis, including caspase-3 and PARP. However, mIR-181 also up-regulated MMP-2 (gelatinase A; 72 kDa gelatinase) and MMP-9 (gelatinase B; 92 kDa gelatinase) [125] which are two MMPs directly relevant to cartilage matrix protein degradation in arthritis [126].

Of note, several miRs were identified as potential targets for inducing chondrocyte survival and therefore could be considered anti-apoptosis factors; these included mIR-98 [127, 128, 129], mIR-9 [130], mIR-15a-5p [131], mIR-142-3p [132] and mIR-502-5p [133]. Additional mIRs, exemplified by miR-195 [134], mIR-139 [135], mIR-29b-3p [136], mIR-488-3p [137] and mIR-203 [138] could very well be included in this group. For example, by employing the C28/I2 line of immortalized human chondrocytes, Zhao et al. [138] showed that knockdown of mIR-203 targeting the myeloid cell leukemia-1 (MCL-1) protein activated Wnt/β-catenin and JAK/STAT signaling promoted chondrocyte survival.

To summarize this section, gaining a further understanding of how to manipulate specific mIRs to achieve increased or decreased synthesis of specific targets known to influence chondrocyte apoptosis may signal the next major advance in targeted OA and RA therapy designed to promote chondrocyte survival.

Advertisement

5. Pharmacologic interventions designed to specifically inhibit chondrocyte apoptosis

5.1. Signal transduction pathways

Lewis and Malemud [82] previously reviewed several potential pharmacologic strategies designed to limit the loss of chondrocyte vitality via apoptosis. These included, targeting x-linked inhibitor of apoptosis (XIAP), tumor necrosis factor-like weak inducer of apoptosis (TWEAK), TRAIL, decoy-receptor-3 (DcR3), tumor necrosis factor receptor protein-like molecules, p53 up-regulated modulator of apoptosis (PUMA), and apoptosis-signal-regulating kinases. In that regard, we proposed several interventional strategies which we acknowledged involved developing a deeper understanding of which signal transduction pathways were altered in RA and OA chondrocytes [19, 28, 36, 77, 82, 106]. For example, XIAP, an inhibitor of activated caspase-9, and caspases-3 and -7 [82] was also shown to interact with mitogen-activated protein kinase kinase 2 (MEKK2) [139]. The interaction between XIAP and MEKK2 resulted in a biphasic activation of NF-κB, a known downstream effector of TNF-α-mediated apoptosis. This finding is relevant to the regulation of chondrocyte apoptosis in both RA and OA primarily because XIAP is a well-known inhibitor of apoptosis protein-3 (IAP3) [140]. In fact, we had previously shown that recombinant human TNF-α (rhTNF-α) induced human chondrocyte apoptosis via activation of p38, JNK1/2 and STAT3 [72], whereas apoptosis of the immortalized human chondrocyte line, C-28/I2 induced by rhTNF-α, but not by rhIL-6, was dependent on upstream MEK1/2 [77]. Therefore, we posit that in order to consider using potential drug interventions that alter the activation of various signaling pathways it will be useful to consider what we know about how alterations in receptor-mediated signaling pathways (reviewed in [19]) may influence apoptosis.

Activation of SAPK/MAPK signaling is most often associated with induction of chondrocyte apoptosis. Thus, induction of rabbit articular chondrocyte apoptosis by the nitric oxide (NO) donor sodium nitroprusside (SNP) was linked to inhibition of c-Jun-amino-terminal kinase (JNK) by virtue of the finding that the JNK small molecule inhibitor (SMI) SP600125, reduced the frequency of apoptotic chondrocytes along with NO-induced NF-κB, p53 and caspase-3 [141]. IL-1β, another potent inducer of chondrocyte apoptosis, was also shown to be JNK-dependent as both chemical inhibitors of JNK as well as RNA interference with Bim, the latter up-regulated by IL-1β, were shown to be phosphorylated-JNK-dependent [142]. In other studies, chondrocyte apoptosis was again linked to IL-1β-induced activation of p38 kinase [143, 144, 145], along with JNK [145], and MMP-3 gene expression with IL-1β negatively regulating chondrocyte autophagy [145]. Of note, AG490, a pan-JAK SMI significantly reduced leptin-induced chondrocyte apoptosis in vitro as well as reducing STAT3 phosphorylation, reactive oxygen species, MMP-13 and B-cell lymphoma 2-associated X protein [146]. Interestingly, Li et al. [147] showed that the PI3K/NF-κB pathway was activated by TNF-α in human chondrocytes. However, the effect of leptin did not involve mTOR, suggesting that newly developed small molecule mTOR inhibitors (reviewed in [36]) might not be useful for neutralizing activated PI3K/NF-κB in response to leptin-induced apoptosis.

Several attempts to employ various compounds and/or natural products to inhibit chondrocyte apoptosis have taken advantage of various findings related to the role of these compounds and natural products in many of the aforementioned signal transduction pathways. For example, IL-1β-induced chondrocyte apoptosis was inhibited by oligomeric proanthocyanidin, a water-soluble plant polyphenolic compound [148]. Thus, the over-expression of peroxiredoxin 4 (PRDX4), a member of the PRDX family (a molecule essential for scavenging free radicals and reducing reactive oxygen species) reduced IL-1β-induced rat chondrocyte apoptosis [149]. Importantly, AZD5363, an inhibitor of Akt activation also reduced the apoptosis protective effect of PRDX4. In another aspect, chondrocyte apoptosis induced by IL-1β not only involved reduced Bcl-2 levels, activated (i.e., phosphorylated) Akt, and activated PRAS40, a proline-rich 40 kDa Akt substrate and an inhibitor of mTORC1 kinase activity, but was also linked to increasing the levels of Bax, and activated caspase-3/-9 [149].

Shikonin, a compound with anti-tumor, anti-inflammatory, anti-viral and pharmacological efficacy significantly inhibited apoptosis by decreasing IL-1β, TNF-α and inducible NO synthase (iNOS) in rats with experimentally induced OA [150]. The effect of shikonin in this animal model of OA was accompanied not only by reduced caspase-3 and cyclooxygenase-2 activity but also by increased activation of Akt, indicating a prominent role for PI3K/Akt signaling in this rat model of OA. Finally, a few novel targets, including protein kinase R-like endoplasmic reticulum kinase and activating transcription factor 6, were identified as potent regulators of chondrocyte apoptosis in vitro and in vivo [151], although the precise signaling mechanisms attributed to these molecules have yet to be completely established.

5.2. Additional potential targets related to apoptosis

Several experimental studies, of note, have focused on several cellular components which may eventually become suitable pharmacologic targets for altering chondrocyte apoptosis and/or autophagy in osteoarthritis (Table 2). However, the results of these mainly in vitro studies point out why it will be necessary to determine the underlying mechanisms regarding how these factors work to inhibit apoptosis. Thus, only after successful evaluation in animal models of RA and OA, can we envision that these targets could eventually be employed in a clinical setting.

FactorTargetReference
HIF-1α/HIF-2α1HIF-1α—SOX9; HIF-2α—Fas[152]
Integrin-β1G1T12[153]
IGFBP-33Nur774[154]
SGBT5Caspase-3/Hsp706[155]
UCP47ROS8[156]
DEL19Caspase-3/Caspase-7[157]
Rela10Pik3r111[158]
Beclin12Bcl-2; Bcl-2 associated X[159]
AST13LC3-II/I14; P62/SQSTM115[160]
AQP-116Caspase-3[161]
Mt1/Mt217ROS8[162]
Sirt118Bcl-2; Bax[163]

Table 2.

Cellular factors that regulate chondrocyte apoptosis and/or autophagy in vitro.

Hypoxia-inducible factor-1α/-2α.


G-protein-coupled receptor kinase interacting protein-1.


Insulin-like growth factor-1 binding protein-3.


Nerve growth factor 1B.


Small glutamine-rich tetratricopeptide repeat-containing β.


Heat shock protein-70.


Uncoupling protein-4.


Reactive oxygen species.


Developmental endothelial locus-1.


RelA/p65 of NF-κB complex.


Pik3r1 encodes a p85α regulatory protein that is a subunit of phosphatidylinositol 3-kinase (PI3K).


Beclin-1, a product of the BECN1 gene is a mammalian ortholog of the yeast autophagy-related gene 6 (Atg6) and BEC-1 in the C. elegans nematode.


Astragaloside IV.


Microtubule-associated protein 1A/1B-light chain-I/II.


P62/sequestosome-1.


Aquaporin-1.


Metallothionein-1/metallothionein-2.


Silent information regulation of transcription 1.


Advertisement

6. Conclusions and future perspectives

It is now generally agreed upon by many investigators that a chronic state of inflammation is, in part, responsible for driving and perpetuating the progression of RA (reviewed in [94]) and OA (reviewed in [58, 164]). Moreover, additional recent evidence has indicated that both apoptosis and an altered state of autophagy are critical events in chronic musculoskeletal disorders, such as RA and OA [60, 102, 103, 104, 105, 164, 165, 166, 167]. Although the loss of chondrocyte vitality is a common pathological finding in RA and OA, cellular mechanisms that result in synovial fibroblast and immune-cell-mediated “apoptosis-resistance,” such as those that were found to underlie B-cell activity in RA [168, 169, 170, 171], aids in distinguishing between the fundamental underpinning responsible for bony abnormalities in the two conditions. However, equally important is that the increased frequency of apoptotic chondrocytes in both RA and OA constitutes a major contributor to inefficient cartilage repair and synovial joint failure. We contend that any pharmacologic strategies designed to simultaneously target the “apoptosis-resistance” in the RA hyperplastic synovial tissue and the elevated frequency of chondrocyte apoptosis would be an onerous undertaking. So for the present, concentrating on developing agents that suppress or inhibit chondrocyte apoptosis might be the initial way to proceed. For example, the current literature on this subject has already indicated that certain drugs used in the medical therapy of RA suppress chondrocyte apoptosis in vitro. In that regard, the drug sulphasalazine, commonly employed in combination with methotrexate, for treating RA [172] was found to inhibit rabbit chondrocyte apoptosis induced by SNP [173]. In this study, the reduced frequency of chondrocyte apoptosis was accompanied by an increase in phosphorylated p38 kinase and ERK1/2 expression compared to treatment of chondrocytes with SNP alone.

Another area worthy of consideration is to employ in vitro studies to, in effect, rule out for further considerations for developing drugs for treating potential targets of chondrocyte apoptosis. A suitable example of this strategy was a recent finding by Nasi et al. [174] that the NALP3 inflammasome (reviewed in [175]) was not involved in chondrocyte apoptosis characteristic of several alterations in cartilage characteristically found in a murine menisectomy model of OA. Although alterations in the structure of articular cartilage such as cartilage destruction, synovial inflammation, cell death and calcification were seen in this OA animal model, a deficiency in IL-1α had no impact on these features. Importantly, deficiencies in IL-1β and NALP3 actually resulted in an enhancement of cartilage damage in this OA animal model.

The suppression or more favorably the complete inhibition of chondrocyte apoptosis in RA and OA using pharmacologic interventional strategies would be a laudable achievement in the continuing search for novel disease-modifying-anti-rheumatic drugs. A variety of potential novel targets have now been identified during the previous 3 years that at least employing cancer cells induces the frequency of apoptosis. Thus, targets have been identified to induce apoptosis in these cancers and therefore, may be eventually exploited for blocking chondrocyte apoptosis. For example, the activity of 2,5-dihydroxy-3-undecyl-1,4 benzoquinone, 6 g, also known as embelin was reviewed [82] as an inducer of apoptosis in inflammatory breast cancer cells and pancreatic cancer cells. Embelin was also shown to block the transcription of several gene products relevant to tumor cell survival, proliferation, invasiveness and metastatic cancer cell proliferation. Of note, treatment with non-toxic concentrations of embelin could also sensitize cultured malignant glioma to TRAIL-induced apoptosis [176]. Lewis and Malemud [82] reviewed the findings showing that embelin blocked the activation of NF-κB, RANKL and, STAT3, the latter finding demonstrating “proof-of-principle” that STAT3-activated transcription could also be employed to probe the extent to which this signaling pathway was required for maintaining chondrocyte survival in vitro [177]. In that regard, these pre-clinical results may provide a suitable platform for exploiting the overall objective of preventing synovial joint failure in RA and OA through the maintenance of normal articular chondrocyte viability and cartilage integrity.

Advertisement

Acknowledgments

The results of experimental studies cited in Refs. [70, 72, 77, 80] were supported in part by grants from the National Institutes of Health, Takeda Pharmaceuticals of North America and Genentech/Roche Group.

References

  1. 1. Reed JC. Bcl-2 and the regulation of programmed cell death. Journal of Cell Biology. 1994;124:1-6. PMCID: PMC2119888
  2. 2. Ashkenazi A, Dixit M. Death receptors: Signaling and modulation. Science. 1998;281:1305-1308. PMID: 9721089
  3. 3. Opferman JT, Letia A, Beard C, et al. Development and maintenance of B and T lymphocytes requires antiapoptotic MCL-1. Nature. 2003;426:671-676. DOI: 10.1038/nature02067
  4. 4. Schultz DR, Harrington WJ Jr. Apoptosis: Programmed cell death at a molecular level. Seminars in Arthritis & Rheumatism. 2003;32:345-369. DOI: 10.1053/sarh.2003.50005
  5. 5. Starr TK, Jameson SC, Hogquist KA. Positive and negative selection of T cells. Annual Review of Immunology. 2003;21:139-176. DOI: 10.1146/annurev.immunol.21.120601.141107
  6. 6. Hernandez JB, Newton RH, Walsh CM. Life and death in the thymus—Cell death signaling during T cell development. Current Opinion in Cell Biology. 2010;22:865-871. DOI: 10.1016/j.ceb.2010.08.003
  7. 7. Malemud CJ. Immunotherapies and rheumatoid arthritis-introduction. Journal of Clinical and Cellular Immunology. 2013;S6:e001
  8. 8. Wolfer A, Wilson A, Nemir M, et al. Inactivation of Notch1 impairs VDJβ rearrangement and allows pre-TCR-independent survival of early αβ lineage thymocytes. Immunity. 2002;16:869-879. PMID: 12121668
  9. 9. Schultzke JD, Bojarski C, Zeissig S, et al. Disrupted barrier function through epithelial cell apoptosis. Annals of the New York Academy of Sciences. 2006;1072:288-299. DOI: 10.1196/annals.1326.027
  10. 10. Beug ST, Cheung HH, LaCasse EC, et al. Modulation of immune signaling by inhibitors of apoptosis. Trends in Immunology. 2012;33:535-545. DOI: 10.1016/j.it.2012.06.004
  11. 11. Liu H, Pope RM. Apoptosis in rheumatoid arthritis: Friend or foe. Rheumatic Disease Clinics of North America. 2004;30:603-625. DOI: 10.1016/j.rdc.2004.04.010
  12. 12. Malemud CJ, Gillespie HJ. The role of apoptosis in arthritis. Current Rheumatology Reviews. 2005;1:131-142
  13. 13. Korb A, Pavenstädt H, Pap T. Cell death in rheumatoid arthritis. Apoptosis. 2009;14:447-454. DOI: 10.1007/s10495-009-0317-y
  14. 14. Kim HA, Blanco FJ. Cell death and apoptosis in osteoarthritis. Current Drug Targets. 2007;8:333-345. PMID: 17305511
  15. 15. Del Carlo M Jr, Loesser RF. Cell death in osteoarthritis. Current Rheumatology Reports. 2008;10:37-42. PMID: 18457610
  16. 16. Zamli Z, Sharif M. Chondrocyte apoptosis: A cause or consequence of osteoarthritis. International Journal of Rheumatic Diseases. 2011;14:159-166. DOI: 10.1111/j.1756-185X.2011.01618.x
  17. 17. Cope AP, Schultze-Koops H, Aringer AM. The central role of T cells in rheumatoid arthritis. Clinical and Experimental Rheumatology. 2007;5(Suppl 46):S4-S11. PMID: 17977483
  18. 18. Hutcheson J, Perlman H. Apoptotic regulators and RA. Current Rheumatology Reviews. 2008;4:254-258
  19. 19. Malemud CJ. Recent advances in neutralizing the IL-6 pathway in arthritis. Open Access Rheumatology: Research and Reviews. 2009;1:133-150. PMID: 27789987
  20. 20. Charlier E, Relic B, Deroyer C, et al. Insights on the mechanisms of chondrocyte death in osteoarthritis. International Journal of Molecular Sciences. 2016;17. pii. E2146. DOI: 10.3390.ijms17122146
  21. 21. Wu L, Liu Z. The molecular mechanisms of preventing apoptosis of cartilage chondrocyte to target osteoarthritis. Future Medicinal Chemistry. 2017;9:537-540. DOI: 10.4155/fmc-2017-0034
  22. 22. Kühn K, Shikhman AR, Lotz M. Role of nitric acid, reactive oxygen species, and 38 MAP kinase in the regulation of human chondrocyte apoptosis. Journal of Cellular Physiology. 2003;197:379-387. DOI: 10.1002/jcp.10372
  23. 23. Deng Y, Ren X, Yang L, et al. A JNK-dependent pathway is required for TNF-α-induced apoptosis. Cell. 2003;115:61-70 14532003
  24. 24. Asquith DL, McInnes IB. Emerging cytokine targets in rheumatoid arthritis. Current Opinion in Rheumatology. 2007;19:246-251. DOI: 10.1097/BOR.0b013e3280eed78c
  25. 25. Wisler BA, Dennis JE, Malemud CJ. New organ-specific pharmacological strategies interfering with signal pathways in inflammatory disorders/autoimmune disorders. Current Signal Transduction Therapy. 2011;6:279-291
  26. 26. Ivashkiv LB, Xu X. Signaling by STATs. Arthritis Research & Therapy. 2004;6:159-168. DOI: 10.1186/ar1197
  27. 27. Kaplan MH. STAT4: A critical regulator of inflammation in vivo. Immunologic Research. 2005;31:231-242. DOI: 10.1385/IR:31:3:231
  28. 28. Malemud CJ, Pearlman E. Targeting JAK/STAT signaling pathway in inflammatory diseases. Current Signal Transduction Therapy. 2009;4:201-221
  29. 29. Stark GR, Darnell JE Jr. The JAK-STAT pathway at 20. Immunity. 2012;36:503-514. DOI: 10.1016/j.immuni.2012.03.013
  30. 30. Rani A, Murphy JJ. STAT5 in cancer and immunity. Journal of Interferon Cytokine Research. 2016;36:226-237. DOI: 10.1089/jir.2015.0054
  31. 31. Malemud CJ. Negative regulators of JAK/STAT signaling in rheumatoid arthritis and osteoarthritis. International Journal of Molecular Sciences. 2017;18. pii: E484. DOI: 10.3390/ijms18030484
  32. 32. Choy E. Understanding the dynamics: Pathways involved in the pathogenesis of rheumatoid arthritis. Rheumatology (Oxford, England). 2012;51(Suppl 5):v3-v11. DOI: 10.1093/rheumatology/kes113
  33. 33. Banham-Hall E, Clatworthy MR, Okkenhaug K. The therapeutic potential for PI3K inhibitors in autoimmune rheumatic diseases. Open Rheumatology. 2012;6:245-258. DOI: 10.2174/1874312901206010245
  34. 34. Nagai S, Kurebayashi Y, Koyasu S. Role of PI3K/Akt and mTOR complexes in Th17 differentiation. Annals of the New York Academy of Sciences. 2013;1280:30-34. DOI: 10.1111/nyas.12059
  35. 35. Alghasham A, Rasheed Z. Therapeutic targets for rheumatoid arthritis: Progress and promises. Autoimmunity. 2014;47:77-94. DOI: 10.3109/08916934.2013.873413
  36. 36. Malemud CJ. The PI3K/Akt/PTEN/mTOR pathway: A fruitful target for inducing cell death in rheumatoid arthritis? Future Medicinal Chemistry. 2015;7:1137-1147. DOI: 10.4155/fmc.15.55
  37. 37. Tristano AG. Tyrosine kinases as targets in rheumatoid arthritis. International Immunopharmacology. 2009;9:1-9. DOI: 10.1016/j.intimp.2008.09.010
  38. 38. Malemud CJ. The discovery of novel experimental therapies for inflammatory arthritis. Mediators of Inflammation. 2009;2009:698769. DOI: 10.1155/2009/698769
  39. 39. Tan SL, Liao C, Lucas MC, et al. Targeting the SyK-BTK axis for the treatment of immunological and hematological disorders: Recent progress and therapeutic perspectives. Pharmacologic Therapy. 2013;138:294-309. DOI: 10.1016/j.pharmthera.2013.02.001
  40. 40. Malemud CJ. Investigative drugs for rheumatoid arthritis: Novel targets. Journal of Autoimmune Disorders. 2016;2:12
  41. 41. Wu L, Huang X, Li L, et al. Insights on the biology and pathology of HIF-1α/-2α, TGFβ/BMP. Wnt/β-catenin, and NF-κB pathways in osteoarthritis. Current Pharmaceutical Design. 2012;18:3293-3312. PMID: 22646092
  42. 42. Husa M, Petursson F, Lotz M, et al. C/EBP homologous protein drives pro-catabolic responses in chondrocytes. Arthritis Research & Therapy. 2013;15:R218. DOI: 10.1186/ar4415
  43. 43. Wang X, Xu Z, Wang J, et al. DUSP19, a downstream effector of leptin, inhibits chondrocyte apoptosis via dephosphorylating JNK during osteoarthritis pathogenesis. Molecular BioSystems. 2016;12:721-728. DOI: 10.1039/c5mb00776c
  44. 44. Huang Y, Huang Q, Su H, et al. TAR DNA-binding protein 43 inhibits inflammatory responses and protects chondrocyte function by modulating RACK1 expression in osteoarthritis. Biomedicine & Pharmacotherapy. 2017;85:362-371. DOI: 10.1016/j.biopha.2016.11.037
  45. 45. Yu C, Chen WP, Wang XH. MicroRNA and osteoarthritis. Journal of International Medical Research. 2011;39:1-9. DOI: 10.1177/147323001103900101
  46. 46. Diaz-Prado S, Cicione C, Mulños-López E, et al. Characterization of microRNA expression in normal and osteoarthritic human chondrocytes. BMC Musculoskeletal Disorders. 2012;13:144. DOI: 10.1186/1471-2474-13-144
  47. 47. Genemaras AA, Ennis H, Kaplan L. Inflammatory cytokines induce specific time- and concentration-dependent MicroRNA release by chondrocytes, synoviocytes and meniscus cells. Journal of Orthopaedic Research. 2016;34:779-790. DOI: 10.1002/jor.23086
  48. 48. Cong L, Zhu Y, Tu G. A bioinformatics analysis of microRNAs in osteoarthritis. Osteoarthritis and Cartilage. 2017;25:1362-1371. DOI: 10.1016/j.joca.2017.03.012
  49. 49. Zamli Z, Adams MA, Tarlton JF, et al. Increased chondrocyte apoptosis is associated with the progression of osteoarthritis in spontaneous Guinea pig models of the disease. Inte-rnational Journal of Molecular Sciences. 2013;14:17729-17743. DOI: 10.3390/ijms140917729
  50. 50. Karouzakis E, Neidhart M, Gay RE, et al. Molecular and cellular basis of rheumatoid joint destruction. Immunology Letters. 2006;106:8-13. DOI: 10.1016/j.imlet.2006.04.011
  51. 51. Huber LC, Distler O, Tarner I, et al. Synovial fibroblasts: Key player in rheumatoid arthritis. Rheumatology (Oxford, England). 2006;45:669-675. DOI: 10.1093/rheumatology/kel065
  52. 52. Johnson EQ, Charchandi A, Babis GC, et al. Apoptosis in osteoarthritis: Morphology, mechanisms, and potential means for therapeutic intervention. Journal of Surgery Orthopedic Advances. 2008;17:147-152. PMID: 18851798
  53. 53. Thomas CM, Fuller CJ, Whittles CE, et al. Chondrocyte death by apoptosis is associated with the initiation and severity of articular cartilage degradation. International Journal of Rheumatic Diseases. 2011;14:191-198. DOI: 10.1111/j.1756-185X.2010.01578.x
  54. 54. Dowthwaite GP, Bishop JC, Redman SN, et al. The surface of articular cartilage contains a progenitor cell population. Journal of Cell Science. 2004;117:889-897. DOI: 10.1242/jcs.00912
  55. 55. Alsalameh S, Amin R, Gemba T, et al. Identification of mesenchymal progenitor cells in normal and osteoarthritic human articular cartilage. Arthritis and Rheumatism. 2004;50:1522-1532. DOI: 10.1002/art.20269
  56. 56. Koeling S, Kruegel J, Imer M, et al. Migratory chondrogenic progenitor cells from repair tissue during the later stages of human osteoarthritis. Cell Stem Cell. 2009;4:324-335. DOI: 10.1016/j.stem.2009.01.015
  57. 57. Schimke B, Trautman S, Mai B, et al. Interleukin 17 inhibits progenitor cells in rheumatoid arthritis cartilage. European Journal of Immunology. 2016;46:440-445. DOI: 10.1002/eji.201545910
  58. 58. Malemud CJ. Apoptosis resistance in rheumatoid arthritis synovial tissue. Journal of Clinical and Cellular Immunology. 2011;S3:006
  59. 59. Malemud CJ, Mobasheri A, editors. International Journal of Medical Sciences. Special Issue: Apoptotic Chondrocytes and Osteoarthritis
  60. 60. Liu-Bryan R, Terkeltaub R. Emerging regulators of the inflammatory process in osteoarthritis. Nature Reviews. Rheumatology. 2015;11:35-44. DOI: 10.1038/nrrheum.2014.162
  61. 61. Malemud CJ. Biologic basis of osteoarthritis: State of the evidence. Current Opinion in Rheumatology. 2015;27:289-294. DOI: 10.1097/BOR.0000000000000162
  62. 62. Miossec P. An update on the cytokine network in rheumatoid arthritis. Current Opinion in Rheumatology. 2004;16:218-222. DOI: 10.1097/BOR.0000000000000162
  63. 63. Brennan F, Beech J. Update on cytokines in rheumatoid arthritis. Current Opinion in Rheumatology. 2007;19:296-301. DOI: 10.1097/BOR.0b013e32805e87f1
  64. 64. Malemud CJ, Reddy SK. Targeting cytokines, chemokines and adhesion molecules in rheumatoid arthritis. Current Rheumatology Reviews. 2008;4:219-234
  65. 65. Houard X, Goldring MB, Berenbaum F. Homeostatic mechanisms in articular cartilage and role of inflammation in osteoarthritis. Current Rheumatology Reports. 2013;15:375. DOI: 10.1007/s11926-013-0375-6
  66. 66. Malemud CJ. Growth hormone, VEGF and FGF: Involvement in rheumatoid arthritis. Clinica Chimica Acta. 2007;375:10-19. DOI: 10.1016/j.cca.2006.06.033
  67. 67. Stannus O, Jones G, Cicuttini F, et al. Circulating levels of IL-6 and TNF-α are associated with knee radiographic osteoarthritis and knee cartilage loss in older adults. Osteoarthritis and Cartilage. 2010;18:1441-1447. DOI: 10.1016/j.joca.2010.08.016
  68. 68. Abe H, Sakai T, Ando W, et al. Synovial fluid cytokine levels in hip disease. Rheumatology (Oxford, England). 2014;53:165-172. DOI: 10.1093/rheumatology/ket334
  69. 69. Zan PF, Yao J, Wu Z, et al. Cyclin D1 gene silencing promotes IL-1β-induced apoptosis in rat chondrocytes. Journal of Cellular Biochemistry. 2018;119:290-299. DOI: 10.1002/jcb.26172
  70. 70. Islam N, Haqqi TM, Jepsen KJ, et al. Hydrostatic pressure induces apoptosis in human chondrocytes from osteoarthritic cartilage through up-regulation of tumor necrosis factor-α, inducible nitric oxide synthase, p53, c-myc and bax-α and suppression of bcl-2. Journal of Cellular Biochemistry. 2002;87:266-278. DOI: 10.1002/jcb.10317
  71. 71. Caramés B, López-Armada MJ, Cillero-Pastor B, et al. Differential effects of tumor necrosis factor-α and interleukin-1-β on cell death in human articular chondrocytes. Osteoarthritis and Cartilage. 2008;16:715-722. DOI: 10.1016/j.joca.2007.10.006
  72. 72. Malemud CJ, Sun Y, Pearlman E, et al. Monosodium urate and tumor necrosis factor-α increase apoptosis in human chondrocyte cultures. Rheumatology (Sunnyvale). 2012;2:113. DOI: 10.4172/2161-1149.1000113
  73. 73. Wang Y, Li de L, Zhang XB, et al. Increase of TNFα-stimulated osteoarthritic chondrocytes apoptosis and decrease of matrix metalloproteinase-9 by NF-κB inhibition. Biomedical and Environmental Sciences. 2013;26:277-283. DOI: 10.3967/0895-3988.2013.04.006
  74. 74. Cheleschi S, Cantarini L, Pascarelli NA, et al. Possible chondroprotective of canakinumab: An in vitro study of human osteoarthritic chondrocytes. Cytokine. 2015;71:165-172. DOI: 10.1016/j.cyto.2014.10.023
  75. 75. Lu H, Zeng C, Chen M, et al. Lentiviral vector-mediated over-expression of Sox9 protected chondrocytes from IL-1β induced degeneration and apoptosis. International Journal of Clinical and Experimental Pathology. 2015;8:10038-10049. PMCID: PMC4637526
  76. 76. Shi X, Ye H, Yao X, et al. The involvement and possible mechanism of NR4A1 in chondrocyte apoptosis during osteoarthritis. American Journal of Translational Research. 2017;9:746-754. PMCID: PMC5340710
  77. 77. Malemud CJ, Lewis AC, Wylie MA, et al. U0126, an inhibitor of MEK1/2, increases tumor necrosis factor-α-induced apoptosis, but not interleukin-6-induced apoptosis in C-28/I2 human chondrocytes. Journal of Autoimmune Disorders. 2015;1:4. PMCID: PMC4739803
  78. 78. Malemud CJ. Anticytokine therapy for osteoarthritis. Evidence to date. Drugs & Aging. 2010;27:95-115. DOI: 10.2165/11319950-000000000-00000
  79. 79. Malemud CJ. Suppression of pro-inflammatory cytokines via targeting of STAT-responsive genes. In: El-Shemy H, editor. Drug Discovery. Rijeka: InTech Publishing; 2013. pp. 373-411
  80. 80. Meszaros EC, Malemud CJ. Phosphorylation of STAT proteins by recombinant human IL-6 in immortalized human chondrocyte cell lines, T/C28a2 and C28/I2. Journal of Inflammation Research. 2017;10:1-8. DOI: 10.2147.JIR.593797
  81. 81. Arora V, Cheung HH, Plenchette S, et al. Degradation of survivin by the X-linked inhibitor of apoptosis (XIAP)-XAF1 complex. The Journal of Biological Chemistry. 2007;282:26202-26209. DOI: 10.1074/jbc.M700776200
  82. 82. Lewis AC, Malemud CJ. Correction of dysfunctional apoptosis in arthritis by pharmacologic interventions: Focus on altering the activity of inhibitor of apoptosis protein. In: Pandalai SG, editor. Recent Research Developments in Pharmacology. Kerala: Research Signpost; 2011. pp. 69-84
  83. 83. Smolewski P, Robak T. Inhibitors of apoptosis proteins (IAPs) as potential molecular targets for therapy of hematological disorders. Current Molecular Medicine. 2011;11:633-649. PMID: 21902653
  84. 84. Saleem M, Qadir MI, Perveen N, et al. Inhibitors of apoptosis proteins: New targets for anticancer therapy. Chemical Biology & Drug Design. 2013;82:243-251. DOI: 10.1111/cbdd. 12176
  85. 85. Malemud CJ. Suppressor of cytokine signaling and rheumatoid arthritis. Integrative Molecular Medicine. 2016;3:17-20
  86. 86. He Q, Sun C, Lei W, et al. SOCS1 regulates apoptosis and inflammation by inhibiting IL-4 signaling in IL-1β-stimulated human osteoarthritic chondrocytes. BioMed Research International. 2017;2017:4601959. DOI: 10.1055/2017/4601959
  87. 87. Blanco FJ, Rego I, Ruiz-Romero C. The role of mitochondria in osteoarthritis. Nature Reviews Rheumatology. 2011;7:161-169. DOI: 10.1038/nrrheum.2010.213
  88. 88. Takada K, Hirose J, Yamabe S, et al. Endoplasmic reticulum stress mediates nitric-oxide-induced chondrocyte apoptosis. Biomedical Reports. 2013;1:315-319. DOI: 10.3892/br.2013.52
  89. 89. Uehara Y, Hirose J, Yamabe S, et al. Endoplasmic reticulum-stress-induced apoptosis contributes to articular cartilage degeneration via C/EBP homologous protein. Osteoarthritis and Cartilage. 2014;22:1007-1017. DOI: 10.1016/j.joca.2014.04.025
  90. 90. Hughes A, Oxford AE, Tawara K, et al. Endoplasmic reticulum stress and unfolded protein response in cartilage pathophysiology: Contributing factors to apoptosis and osteoarthritis. International Journal of Molecular Sciences. 2017;18. pii: E665. DOI: 10.3390/ijms18030665
  91. 91. Li XF, Zhang Z, Chen ZK, et al. Piezo protein induces the apoptosis of human osteoarthritis-derived chondrocytes by activating caspase-12, the signaling marker of ER stress. International Journal of Molecular Medicine. 2017;40:845-853. DOI: 10.3892/ijmm.2017.3075
  92. 92. Li D, Xie G, Wang W. Reactive oxygen species: The 2-edged sword of osteoarthritis. The American Journal of Medical Sciences. 2012;344:486-490. DOI: 10.1097/MAJ.0b013e3182579dc6
  93. 93. Wu Q, Zhong ZM, Zhu SY, et al. Advanced oxidation protein products induce chondrocyte apoptosis via receptor for advanced glycation end products-mediated, redox-dependent intrinsic apoptosis pathway. Apoptosis. 2016;21:36-50. DOI: 10.1007/s10495-015-1191-4
  94. 94. Angelotti F, Parma A, Cafaro G, et al. One year in review 2017: Pathogenesis of rheumatoid arthritis. Clinical and Experimental Rheumatology. 2017;35:368-378. PMID: 28631608
  95. 95. Battistelli M, Salucci S, Olivotto E, et al. Cell death in human articular chondrocytes. A morpho-functional study in micromass model. Apoptosis. 2014;19:1471-1483. DOI: 10.1007/s10495-014-1017-9
  96. 96. Gu YY, Chen J, Meng ZL, et al. Research progress on osteoarthritis mechanisms. Biomedical Pharmacotherapy. 2017;93:1246-1252. DOI: 10.1016/j.biopha.2017.07.034
  97. 97. Hwang HS, Kim HA. Chondrocyte apoptosis in the pathogenesis of osteoarthritis. International Journal of Molecular Sciences. 2015;16:26035-26054. DOI: 10.3390/ijms161125943
  98. 98. Roach HI, Aigner T, Kouri JB. Chondropotosis: A variant of apoptotic cell death in osteoarthritis. Apoptosis. 2004;9:265-277 15258458
  99. 99. Lin NY, Beyer C, Giessl A, et al. Autophagy regulates TNFα-mediated joint destruction in experimental arthritis. Annals of the Rheumatic Diseases. 2013;72:761-768. DOI: 10.1136/annrheumdis-2012-201671
  100. 100. Dai Y, Hu S. Recent insights into the role of autophagy in the pathogenesis of rheumatoid arthritis. Rheumatology (Oxford, England). 2016;55:403-410. DOI: 10.1093/rheumatology/kev337
  101. 101. Speziali A, Delcogliano M, Tei M, et al. Chondropenia: Current concept review. Musculoskeletal Surgery. 2015;99:189-200. DOI: 10.1007/s12306-015-0377-9
  102. 102. Chang J, Wang W, Zhang H, et al. The dual role of autophagy in chondrocyte responses in the pathogenesis of articular cartilage degeneration in osteoarthritis. International Journal of Molecular Medicine. 2013;32:1311-1318. DOI: 10.3892/ijmm.2013.1520
  103. 103. Musumeci G, Castrogiovanni P, Trovato FM, et al. Biomarkers of chondrocyte apoptosis and autophagy in osteoarthritis. International Journal of Molecular Sciences. 2015;16:20560-20575. DOI: 10.3390/ijms160920560
  104. 104. Jeon H, Im GI. Autophagy in osteoarthritis. Connective Tissue Research. 2017;58:497-508. DOI: 10.1080/03008207.2016.1240790
  105. 105. Li YS, Zhang FJ, Zeng C, et al. Autophagy in osteoarthritis. Joint, Bone, Spine. 2016;83:143-148. DOI: 10.1016/j.jbspin.2015.06.009
  106. 106. Wylie MA, Malemud CJ. Perspective: Deregulation of apoptosis in arthritis by altered signal transduction. International Journal of Clinical Rheumatology. 2013;8:483-490. DOI: 10.2217/IJR.13.31
  107. 107. Musumeci G, Loreto C, Carnazza M, et al. Characterization of apoptosis in articular cartilage derived from the knee joints of patients with osteoarthritis. Knee Surgery, Sports Traumatology, Arthroscopy. 2011;19:307-313. DOI: 10.1007/s00167-010-1215-0
  108. 108. Huang ZM, Du SH, Huang LG, et al. Leptin promotes apoptosis and inhibits autophagy of chondrocytes through upregulating lysyl oxidase-like 3 during osteoarthritis pathogenesis. Osteoarthritis and Cartilage. 2016;24:1246-1253. DOI: 10.1016/j.joca.2016.02.009
  109. 109. Zhang Y, Vasheghani F, Li YH, et al. Cartilage-specific deletion of mTOR upregulates autophagy and protects mice from osteoarthritis. Annals of Rheumatic Diseases. 2015;74:1432-1440. DOI: 10.1136/annrheumdis-2013-204599
  110. 110. Shen C, Cai GQ, Peng JP, et al. Autophagy protects chondrocytes from glucocorticoid-induced apoptosis via ROS/Akt/FOXO3 signaling. Osteoarthritis and Cartilage. 2015;23:2279-2287. DOI: 10.1016/j.joca.2015.06.020
  111. 111. Huang W, Ao P, Li J, et al. Autophagy protects advanced glycation end product-induced apoptosis and expression of MMP-3 and MMP-13 in rat chondrocytes. Biomedical Research International. 2017;2017:6341919. DOI: 10.1155/2017/6341919
  112. 112. Sera SR, Zur Nieden NI. microRNA regulation of skeletal development. Current Osteoporosis Reports. 2017;15:353-366. DOI: 10.1007/s11914-017-0379-7
  113. 113. Churov AV, Oleinik EK, Knip M. MicroRNAs in rheumatoid arthritis: Altered expression and diagnostic potential. Autoimmune Reviews. 2015;14:1029-1037. DOI: 10.1016/j.autrev.2015.07.005
  114. 114. Haftmann C, Stittrich AB, Zimmermann J, et al. miR-148a is upregulated by Twist1 and t-bet and promotes Th1-survival by regulating the proapoptotic gene Bim. European Journal of Immunology. 2015;45:1192-1205. DOI: 10.1002/eji.201444633
  115. 115. D’Adamo S, Cetrullo S, Minguzzi M, et al. MicroRNAs and autophagy: Fine players in the control of chondrocyte homeostatic activities in osteoarthritis. Oxidative Medicine and Cellular Longevity. 2017;2017:3720128. DOI: 10.1155/2017/3720128
  116. 116. Malemud CJ. Chondrocyte apoptosis in rheumatoid arthritis: Is preventive therapy possible? Immunotherapy (Los Angel). 2015;1. pii: 102. PMID: 26878072
  117. 117. Jin L, Zhao J, Jing W, et al. Role of miR-146a in human chondrocyte apoptosis in response to mechanical pressure injury in vitro. International Journal of Molecular Medicine. 2014;34:451-463. DOI: 10.3892/ijmm.2014.1808
  118. 118. Zhong JH, Li J, Liu CF, et al. Effects of microRNA-146a on the proliferation and apoptosis of human osteoarthritis chondrocytes by targeting TRAF6 through the NF-κB signaling pathway. BioScience Reports. 2017;37. pii: BSR20160578. DOI: 10.1042/BSR20160578
  119. 119. West C, McDermott MF. Effects of microRNA-146a on the proliferation and apoptosis of human osteochondrocytes by targeting TRAF6 through the NF-κB signalling pathway. BioScience Reports. 2017;37:BSR20170180. DOI: 10.1042/BSR20170180
  120. 120. Li J, Huang J, Dai L, et al. miR-146a, an IL-1β responsive miRNA, induces vascular endothelial growth factor and chondrocyte apoptosis by targeting Smad4. Arthritis Research & Therapy. 2012;14:R75. DOI: 10.1187/ar3798
  121. 121. Abouheif MM, Nakasa T, Shibuya H, et al. Silencing microRNA-34a inhibits chondrocyte apoptosis in a rat osteoarthritis model in vitro. Rheumatology (Oxford, England). 2010;49:2054-2060. DOI: 10.1093/rheumatology/keg247
  122. 122. Yan S, Wang M, Zhao J, et al. Micro-RNA-34a affects chondrocyte apoptosis and proliferation by targeting the SIRT1/p53 signaling pathway during the pathogenesis of osteoarthritis. International Journal of Molecular Medicine. 2016;38:201-209. DOI: 10.3892/ijmm.2016.2618
  123. 123. Chen H, Wang J, Hu B, et al. MiR-34a promotes Fas-mediated cartilage endplate chondrocyte apoptosis by targeting Bcl-2. Molecular and Cellular Biochemistry. 2015;406:21-30. DOI: 10.1007/s11010-015-2420-4
  124. 124. Zhai X, Meng R, Li H, et al. miR-181a modulates chondrocyte apoptosis by targeting glycerol-3-phosphate dehydrogenase 1-like protein (GPD1L) in osteoarthritis. Medical Science Monitor. 2017;23:1224-1231. PMCID: PMC5360418
  125. 125. Wu XF, Zhou ZH, Zou J. MicroRNA-181 inhibits proliferation and promotes apoptosis of chondrocytes in osteoarthritis by targeting PTEN. Biochemistry and Cell Biology. 2017;95:437-444. DOI: 10.1139/bcb-2016-0078
  126. 126. Malemud CJ. Matrix metalloproteases and synovial joint pathology. In: Khalil RA, editor. Progress in Molecular Biology and Translational Science. Vol. 148. Burlington: Academic Press; 2017. pp. 306-325
  127. 127. Wang GL, Wu YB, Liu JT, et al. Upregulation of miR-98 inhibits apoptosis in cartilage cells in osteoarthritis. Genetic Testing and Molecular Biomarkers. 2016;20:645-653. DOI: 10.1089/gtmb/2016.0011
  128. 128. Wang J, Chen L, Jin S, et al. MiR-98 promotes chondrocyte apoptosis by decreasing Bcl-2 expression in a rat model of osteoarthritis. Acta Biochimica Biophysica Sin (Shanghai). 2016;48:923-929. DOI: 10.1093/abbs/gmw084
  129. 129. Wang J, Chen L, Jin S, et al. Altered expression of microRNA-98 in IL-1β-induced cartilage degradation and its role in chondrocyte apoptosis. Molecular Medicine Reports. 2017;16:3208-3216. DOI: 10.3892/mmr.2017.7028
  130. 130. Song J, Kim D, Chun CH, et al. MicroRNA-9 regulates survival of chondroblasts and cartilage integrity by targeting protogenin. Cell Communication and Signaling. 2013;11:66. DOI: 10.1186/1478-811X-11-66
  131. 131. Chen H, Tian Y. MiR-15a-5p regulates viability and matrix degradation of human osteoarthritis chondrocytes via targeting VEGFA. Bioscience Trends. 2017;10:482-488. DOI: 10.5582/bst.2016.01187
  132. 132. Wang X, Guo Y, Wang C, et al. MicroRNA-142-3p inhibits chondrocyte apoptosis and inflammation in osteoarthritis by targeting HMGB1. Inflammation. 2016;39:1718-1728. DOI: 10.1007/s10753-016-0406-3
  133. 133. Zhang G, Sun Y, Wang Y, et al. MiR-502-5p inhibits IL-1β-induced chondrocyte injury by targeting TRAF2. Cellular Immunology. 2016;302:50-57. DOI: 10.1016/j.cellimm.2016.01.007
  134. 134. Bai R, Zhao AQ, Zhao ZQ, et al. MicroRNA-195 induced apoptosis in hypoxic chondrocytes by targeting hypoxia-inducible factor 1 alpha. European Review for Medical and Pharmacological Sciences. 2015;19:545-551. PMID: 25753868
  135. 135. Makki MS, Haqqi TM. miR-139 modulates MCPIP1/IL-6 expression and induces apoptosis in human chondrocytes. Experimental & Molecular Medicine. 2015;47:e189. DOI: 10.1038/emm.2015.66
  136. 136. Chen L, Li Q, Wang J, et al. MiR-29b-3p promotes chondrocyte apoptosis and facilitates the occurrence and development of osteoarthritis by targeting PRGN. Journal of Cellular and Molecular Medicine. 2017;21:3347-3359. DOI: 10.1111/jcmm.13237
  137. 137. Li Y, Li S, Luo Y, et al. LncRNA PVT1 regulates chondrocyte apoptosis in osteoarthritis by acting as a sponge for miR-488-3p. DNA Cell Biology. 2017;36:571-580. DOI: 10.1089/dna.2017.3678
  138. 138. Zhao C, Wang Y, Jin H, et al. Knockdown of microRNA-203 alleviates LPS-induced injury by targeting MCL-1 in C28/I2 chondrocytes. Experimental Cell Research. 2017;359:171-178. DOI: 10.1016/j.yexcr.2017.07.034
  139. 139. Winsauer G, Resch U, Hofer-Warbinek R, et al. XIAP regulates bi-phasic NF-κB induction involving physical interaction and ubiquitination of MEKK2. Cell Signaling. 2008;11:2107-2112. DOI: 10.1016/j.cellsig.2008.08.004
  140. 140. Rajalingam K, Dikic I. Inhibitors of apoptosis catch ubiquitin. Biochemical Journal. 2009;417:e1-e3. DOI: 10.1042/BJ20082215
  141. 141. Chen Q, Gao Y, Kao X, et al. SNP-induced apoptosis may be mediated with caspase inhibitor by JNK signaling pathways in rabbit articular chondrocytes. The Journal of Toxicological Sciences. 2012;37:157-167. PMID: 22293420
  142. 142. Ye Z, Chen Y, Zhang R, et al. C-Jun N-terminal kinase—C-Jun pathway transactivates Bim to promote osteoarthritis. Canadian Journal of Physiology and Pharmacology. 2014;92:132-139. DOI: 10.1139/cipp-2013-0228
  143. 143. Xu L, Zhai L, Ge Q, et al. Vacuolar protein sorting 4B (VPS4B) regulates apoptosis of chondrocytes via p38 mitogen-activated protein kinases (MAPK) in osteoarthritis. Inflammation. DOI: 10.1007/s10753-017-0633-2
  144. 144. Sun HY, Hu KZ, Yin ZS. Inhibition of p38-MAPK signaling pathway suppresses the apoptosis and expression of proinflammatory cytokines in human osteoarthritis chondrocytes. Cytokine. 2017;90:135-143. DOI: 10.1016/j.cyto.2016.11.002
  145. 145. Shi J, Zhang C, Yi Z, et al. Explore the variation of MMP3, JNK, p38 MAPKs, and autophagy at the early stages of osteoarthritis. IUBMB. 2016;68:293-302. DOI: 10.1002/jub.1482
  146. 146. Zhang ZM, Shen C, Li H, et al. Leptin induces the apoptosis of chondrocytes in an in vitro model of osteoarthritis via the JAK2-STAT3 signaling pathway. Molecular Medicine Reports. 2016;13:3684-3690. DOI: 10.3892/mmr.2016.4970
  147. 147. Li D, Wu Z, Duan Y, et al. TNFα-mediated apoptosis in human osteoarthritic chondrocytes sensitized by PI3K-NFκB inhibitor, not mTOR inhibitor. Rheumatology International. 2012;32:2017-2022. DOI: 10.1007/s00296-011-1929-4
  148. 148. Yin MH, Wang YT, Li Q, et al. Oligomeric proanthocyanidins inhibit apoptosis of chondrocytes induced by interleukin-1β. Molecular Medicine Reports. . DOI: 10.3892/mmr.2017.7124
  149. 149. Rao Z, Wang S, Wang J. Peroxiredoxin 4 inhibits IL-1β-induced chondrocyte apoptosis via PI3K/AKT signaling. Biomedicine & Pharmacotherapy. 2017;90:414-420. DOI: 10.1016/j.biopha.2017.03.075
  150. 150. Fu D, Shang X, Ni Z, et al. Shikonin inhibits inflammation and chondrocyte apoptosis by regulation of the PI3K/Akt signaling pathway in a rat model of osteoarthritis. Experimental and Therapeutic Medicine. 2016;12:2735-2740. DOI: 10.3892/etm.2016.3642
  151. 151. Wu Z, Li M, Zheng W, et al. Silencing of both ATF4 and PERK inhibits cell cycle progression and promotes the apoptosis of differentiating chondrocytes. International Journal of Molecular Medicine. 2017;40:101-111. DOI: 10.3892/ijmm.2017.2985
  152. 152. Zhang FJ, Luo W, Lei GH. Role of HIF-1α and HIF-2α in osteoarthritis. Joint, Bone, Spine. 2015;82:144-147. DOI: 10.1016/j.ibspin.2014.10.003
  153. 153. Zhang LO, Zhao GZ, Xu XY, et al. Integrin-β1 regulates chondrocyte proliferation and apoptosis through the upregulation of G1T1 expression. International Journal of Molecular Medicine. 2015;35:1074-1080. DOI: 10.3892/ijmm.2015.2114
  154. 154. Wei Z, Li HH. IGFBP-1 may trigger osteoarthritis by inducing apoptosis of chondrocytes through Nur77 translocation. International Journal of Clinical and Experimental Pathology. 2015;8:15599-15610. PMCID: PMC4730042
  155. 155. Bao G, Xu L, Xu X, et al. SGBT promotes the caspase-dependent apoptosis in chondrocytes of osteoarthritis. Inflammation. 2016;39:601-610. DOI: 10.1007/s10753-015-0285-z
  156. 156. Huang Z, Li J, Du S, et al. Effects of UCP4 on the proliferation and apoptosis of chondrocytes: Its possible involvement and regulation in osteoarthritis. PLoS One. 2016;11:e0150684. DOI: 10.1371/journal.pone.0150684
  157. 157. Wang Z, Tran C, Bhatia NJ, et al. Del1 knockout mice developed more severe osteoarthritis associated with increased susceptibility of chondrocytes to apoptosis. PLoS One. 2016;11:e0160684. DOI: 10.1371/journal.pone.0160684
  158. 158. Kobayashi H, Chang SH, Mori D, et al. Biphasic regulation of chondrocytes by Rela through induction of anti-apoptotic and catabolic target genes. Nature Communications. 2016;7:13336. DOI: 10.1038/ncomms13336
  159. 159. Song B, Song H, Wang W, et al. Beclin 1 overexpression inhibits chondrocyte apoptosis and downregulates extracellular matrix metabolism in osteoarthritis. Molecular Medicine Reports. 2017;16:3958-3964. DOI: 10.3892/mmr.2017.7064
  160. 160. Liu J, Meng Q, Jing H, et al. Astragaloside IV protects against apoptosis in human degenerative chondrocytes through autophagy activation. Molecular Medicine Reports. 2017;16:3269-3275. DOI: 10.3892/mmr.2017.6980
  161. 161. Gao H, Gui J, Wang L, et al. Aquaporin 1 contributes to chondrocyte apoptosis in a rat model of osteoarthritis. International Journal of Molecular Medicine. 2016;38:1752-1758. DOI: 10.3892/ijmm.2016.2785
  162. 162. Won Y, Shin Y, Chun CH, et al. Pleiotropic roles of metallothioneins as regulators of chondrocyte apoptosis and catabolic and anabolic pathways during osteoarthritis pathogenesis. Annals of the Rheumatic Diseases. 2016;75:2045-2052. DOI: 10.1136/annrheumdis-2015-208406
  163. 163. He Ds HXJ, Yan YQ, et al. Underlying mechanism of Sirt1 on apoptosis and extracellular matrix degradation of osteoarthritis chondrocytes. Molecular Medicine Reports. 2017;16:845-850. DOI: 10.3892/mmr.2017.6659
  164. 164. Berenbaum F, Griffin TM, Liu-Bryan R. 2017; review: Metabolic regulation of inflammation in osteoarthritis. Arthritis & Rheumatology. 2017;69:9-21. DOI: 10.1002/art.39842
  165. 165. Sorice M, Iannuccelli C, Manganelli V, et al. Autophagy generates citrullinated peptides in human synoviocytes: A possible trigger for anti-citrullinated peptide antibodies. Rheumatology (Oxford, England). 2016;55:1374-1385. DOI: 10.1093/rheumatology/kew178
  166. 166. Ireland JM, Unanue ER. Autophagy in antigen-presenting cells results in presentation of citrullinated peptides to CD4 T cells. Journal of Experimental Medicine. 2011;208:2625-2632. DOI: 10.1084/jem.20110640
  167. 167. Van Loosdregt J, Rossetti M, Spreafico R, et al. Increased autophagy in CD4+ T cells of rheumatoid arthritis patients results in T-cell hyperactivation and apoptosis resistance. European Journal of Immunology. 2016;46:2862-2870. DOI: 10.1002/eji.201646375
  168. 168. Dalbeth N, Smith T, Gray S, Doyle A, Antill P, Lobo M, et al. Cellular characterisation of magnetic resonance imaging bone oedema in rheumatoid arthritis; implications for pathogenesis of erosive disease. Annals of the Rheumatic Diseases. 2009;68:279-282. DOI: 10.1136/ard.2008.096024
  169. 169. Yang J, Zhao S, Yang X, et al. Inhibition of B-cell apoptosis is mediated through increased expression of Bcl-2 in patients with rheumatoid arthritis. International Journal of Rheumatic Diseases. 2016;19:134-140. DOI: 10.1111/1756-185X.12706
  170. 170. Störch H, Zimmerman B, Resch B, et al. Activated human B cells induce inflammatory fibroblasts with cartilage-destructive properties and become functionally suppressed in return. Annals of the Rheumatic Diseases. 2016;75:924-932. DOI: 10.1136/annrheumdis-2014-206965
  171. 171. Meednu N, Zhang H, Owen T, et al. Production of RANKL by memory B cells: A link between B cells and bone erosion and rheumatoid arthritis. Arthritis & Rheumatology. 2016;68:805-816. DOI: 10.1002/art.39489
  172. 172. Taherian E, Rao A, Malemud CJ, et al. The biological and clinical activity of anti-malarial drugs in autoimmune disorders. Current Rheumatology Reviews. 2013;9:45-62. PMID: 25198367
  173. 173. Lee WK, Kang JS. Modulation of apoptosis and differentiation by the treatment of sulfasalazine in rabbit articular chondrocytes. Toxicology Research. 2016;32:115-121. DOI: 10.5487/TR.2016/32.2.115
  174. 174. Nasi S, Ea HK, So A, et al. Revisiting the role of interleukin-1 pathway in osteoarthritis: Interleukin-1α and -1β and NALP3 inflammasome are not involved in pathological features of the murine meniscectomy model of osteoarthritis. Frontiers in Pharmacology. 2017;8:282. DOI: 10.3389/fphar.2017.00282
  175. 175. Campbell L, Raheem I, Malemud CJ, et al. The relationship between NALP3 and autoinflammatory syndromes. International Journal of Molecular Sciences. 2016;17:725. DOI: 10.3390/ijms17050725
  176. 176. Siegelin MD, Gaiser T, Siegelin Y. The XIAP inhibitor Embelin enhances TRAIL-mediated apoptosis in malignant glioma cells by down-regulation of the short isoform of FLIP. Neurochemistry International. 2009;55:423-430. DOI: 10.1016/j.neuint.2009.04.011
  177. 177. Malemud CJ. The small molecular weight inhibitor of protein kinase revolution for the treatment of rheumatoid arthritis. In: Vallisuta O, Olimat S, editors. Drug Discovery and Development—From Molecules to Medicine. Rijeka: InTech Publishing; 2015. p. 163-179. DOI: http://dx.doi.org/10.5772/59639

Written By

Charles J. Malemud

Submitted: 11 July 2017 Reviewed: 15 December 2017 Published: 19 September 2018