Next Article in Journal
Hsp60 in Skeletal Muscle Fiber Biogenesis and Homeostasis: From Physical Exercise to Skeletal Muscle Pathology
Next Article in Special Issue
A Disintegrin and Metalloprotease 15 is Expressed on Rheumatoid Arthritis Synovial Tissue Endothelial Cells and may Mediate Angiogenesis
Previous Article in Journal
Lack of LTβR Increases Susceptibility of IPEC-J2 Cells to Porcine Epidemic Diarrhea Virus
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Defective T-Cell Apoptosis and T-Regulatory Cell Dysfunction in Rheumatoid Arthritis

by
Charles J. Malemud
1,2
1
Department of Medicine, Division of Rheumatic Diseases, Case Western Reserve University School of Medicine, Foley Medical Building, 2061 Cornell Road, Suite 207, Cleveland, OH 44122-5076, USA
2
Department of Medicine, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
Submission received: 16 October 2018 / Revised: 13 November 2018 / Accepted: 20 November 2018 / Published: 22 November 2018
(This article belongs to the Special Issue The Molecular and Cellular Basis for Rheumatoid Arthritis)

Abstract

:
Rheumatoid arthritis (RA) is a chronic, progressive, systemic autoimmune disease that mostly affects small and large synovial joints. At the molecular level, RA is characterized by a profoundly defective innate and adaptive immune response that results in a chronic state of inflammation. Two of the most significant alterations in T-lymphocyte (T-cell) dysfunction in RA is the perpetual activation of T-cells that result in an abnormal proliferation state which also stimulate the proliferation of fibroblasts within the joint synovial tissue. This event results in what we have termed “apoptosis resistance”, which we believe is the leading cause of aberrant cell survival in RA. Finding therapies that will induce apoptosis under these conditions is one of the current goals of drug discovery. Over the past several years, a number of T-cell subsets have been identified. One of these T-cell subsets are the T-regulatory (Treg) cells. Under normal conditions Treg cells dictate the state of immune tolerance. However, in RA, the function of Treg cells become compromised resulting in Treg cell dysfunction. It has now been shown that several of the drugs employed in the medical therapy of RA can partially restore Treg cell function, which has also been associated with amelioration of the clinical symptoms of RA.

1. Introduction

Rheumatoid arthritis (RA) is a chronic, progressive inflammatory disease of synovial joints [1]. Chronic inflammation results from aberrant innate and adaptive immune responses that help drive RA to the point of joint failure [2,3,4]. Moreover, RA patients have a higher mortality than people in the general population [5] and importantly, the inflammation of RA is not confined to synovial joints [6,7] as manifested by pathologic changes also seen in the cardiovascular and pulmonary system, kidney and skin. In addition, the malfunctioning joints in RA severely limits the patient’s mobility with an associated decline in the quality of life [8].
Several recent reviews have focused on the pathophysiology of RA which emphasized that maladaptive immune responses in RA result from hyperactive T-cell and B-cell patterns of dysfunction [2,9,10,11,12,13]. These abnormalities encompass defective signal transduction [10], and an elevated level of transcription of pro-inflammatory cytokine genes [e.g., TNF-α, IL-1ß, IL-6, IFN-γ] to name only a few [12,14,15]. For example, after T-cells are activated they begin to secrete many of these cytokines. However, abnormal functioning of antigen-presenting cells (APCs) will also frequently accompany autoimmune activation. As such dendritic cells (DCs) acting as APCs also become chronically activated and in doing so function in a maximum antigen-presenting mode which is not their normal state. Therefore, this scenario can result in the imbalances in the patterns of T-cell cytokine secretion that further promotes chronic inflammation. Defective signal transduction also leads to an increased transcription of matrix metalloproteinase (MMP) genes [16]. Thus, MMPs, including MMP-1 (collagenase-1), MMP-2 (72kDa gelatinase), MMP-9 (92kDa gelatinase), MMP-3 (stromelysin-1) and MMP-14 (MT1-MMP) are the activated proteolytic enzymes responsible for the breakdown of extracellular matrix proteins. This component of RA pathology primarily relevant to altered articular cartilage structural integrity is often coupled to an overall impairment of the negative regulators of certain adaptive cellular immune responses [17].
Another facet of RA progression that involves T-cells resides within the two branches of effector CD4+ T-cells; the T follicular helper (TFH) cells that are programmed to act on B-cells that produce high-affinity class-switched antibodies [18], and the non-TFH effector cells, including the Th1, Th2 and Th17 T-cell subsets. These latter cells exit lymphoid tissues to drive adaptive and innate immune responses [19]. Although the exact operative mechanism that drives this distinction between the types of T-effector cells is not completely understood, a skewing of these T-cell subsets towards one type or the other would be expected to significantly alter the host immune response. In keeping with this, Niu et al. [20] recently suggested that enhanced IL-6/STAT3 signaling could be a mechanism underlying the formation of TFH cells by causing the ratio of TFH cells to T follicular regulator (TFR) cells towards the former as RA progresses. Furthermore, Wang et al. [21] confirmed that the ratio of TFR cells to TFH cells decreased as RA progressed and that this skewing of the TFR cells to TFH cells was correlated with changes in serum C-reactive protein, erythrocyte sedimentation rate, rheumatoid factor, anti-cyclic citrullinated peptide antibodies, IgG and the DAS-28, the latter a measure of disease activity.
This narrative review focuses on several aspects of abnormal adaptive cellular and humoral immunity that is characteristic of RA. This would center about some compelling data emerging from recent studies regarding changes occurring in T-cells that may be responsible for “apoptosis-resistance” and would include, the failure of T-cell costimulatory molecules (e.g., cytotoxic T-lymphocyte-associated antigen 4 [CTLA-4]) to regulate T-cell proliferation and the loss of T-regulatory (Treg) lymphocyte functions.
In fact, the drug abatacept is a fusion protein that selectively modulates T-cell activation including T-cell proliferation. Thus, abatacept works by binding to CD80 and CD86 receptors on APCs. In that regard, abatacept inhibits T cell activation via this mechanism by selectively interfering with a specific interaction between CD80/CD86 receptors and CD28 thus leading to inhibition of T-cell proliferation and B cell immunological response [22]. In addition, abatacept, which is employed in adult RA and is also approved for the treatment of juvenile idiopathic arthritis was shown to reduce the proliferation of T-helper cells from healthy donors in response to recall antigens as well as reducing the level of IFN-γ and TNF-α in vitro [23]. Of note, the skewing of the T-cell profile towards circulating CCR4+CXCR3T-helper subsets, Th2 and Th17 prior to initiating any RA therapy in patients diagnosed with “early” RA suggested that these cells may have a particular role in the beginning stages of RA [24]. In addition, on the B-cell side, abnormalities in IgG-antigen binding and IgG antigen-forming immune complexes as well as the negative effects of anti-idiotypic antibody production have now been implicated in the progression of RA.
Here we have focused on dysregulated T-cell proliferation and the loss of functional regulation of immune tolerance by Treg cells. These core elements are very critical to adaptive immune cell dysfunction in RA. Therefore it is likely that the attention paid to these abnormalities will further our understanding of how these components contribute to RA pathophysiology. It is also likely to inform us about how these crucial negative regulators of immune cell function become gradually ineffective in the setting of RA. Thus, these changes in immune cell function are mainly responsible for RA progression and promotion of synovial joint articular cartilage degradation and subchondral bone erosions. Furthermore, the attention paid to these abnormalities has resulted in published research related to the extent to which medical therapies for RA could restore control of T-cell proliferation via induction of apoptosis and functional Treg cells. In addition, the continued study of these aspects of RA pathology are proposed in order to stimulate the development of specific novel therapeutics for RA. Importantly, recent findings in this field of research have promoted an upsurge in designing novel drugs to restore T-cell homeostasis.

2. T-Cell and RA-Fibroblast-Like Synoviocytes: “Apoptosis-Resistance”

T-cell proliferation is dysregulated in RA. Thus, it was readily predictable that up-regulation of the immune check point proteins, programmed cell death protein 1 ligand 1 (PD-L1) and CTLA-4 would prevent T-cell activation [25] since these proteins represented likely targets for restoring normal T-cell proliferation in RA [26].
Programmed cell death also known as “controlled” cell death or apoptosis is one of the major contributors to maintaining tissue and organ homeostasis [27]. In that regard, RA is characterized, in part, by an imbalance conferred on RA-fibroblast-like synoviocyte (RA-FLS) proliferation by the presence of autoreactive T-cell and B-cell and APCs [28], which render RA-FLS unable to appropriately respond to endogenous mediators that regulate the balance between cell survival and apoptosis, skewing the balance towards cell survival [13]. We have termed this phenomenon “apoptosis resistance” [27,29]. Thus, a combination of events that significantly reduce RA-FLS apoptosis results from the elevated levels of several pro-inflammatory cytokines (e.g., TNF-α, IL-1ß, IL-6, IL-7, IL-8, IL-12/IL-23, IL-15, IL-17, IL-18, IL-32, IFN-γ] and growth factors, including fibroblast growth factor-2 (FGF-2) and vascular endothelial growth factor (VEGF), which cause an aberrant survival of RA-FLS [3,30,31,32,33,34,35]. Many of the downstream events that result in abnormal survival of RA-FLS are dysregulated by constitutive as well as cytokine and growth factor-induced activation of Janus Kinase-Signal Transducers and Activators of Transcription (JAKTAT) [36,37,38,39,40], Stress-Activated Protein Kinase/Mitogen-Activated Protein Kinase (SAPK/MAPK) [40,41,42,43] and PI3K/Akt/Phosphatase and tensin homolog deleted on chromosome 10 (PTEN)/mechanistic target of rapamycin (mTOR) signaling [10,44,45,46,47,48,49] signaling pathways which regulate the balance between cell survival and apoptosis. PTEN is especially important in this context due to the fact that under normal conditions PTEN is a tumor suppressor acting through its phosphatase activity and as such, PTEN regulates cell cycle progression. However, because dysregulated cell proliferation is common in RA, it is likely that abnormalities in PTEN play a critical role in aberrant nonimmune and immune-cell proliferation [49].
Several of the pro-inflammatory cytokines (e.g., TNF-α, IL-1ß) and other molecules (e.g., Fas, VEGF) implicated in the progression of RA activate the extrinsic apoptosis pathway involving the coupling of a protein ligand to a cognate receptor, whereas the intrinsic apoptosis cascade is initiated when cytochrome c is released from mitochondria. This latter event requires energy generated by ATP hydrolysis [50]. In the intrinsic apoptosis pathway, the capacity to release cytochrome c from mitochondria is a function of the balance between the Bcl-2 family of pro-apoptotic and anti-apoptotic proteins [51,52]. Thus, the Bcl-2 protein family, consisting of B-cell lymphoma-2 (Bcl-2), B-cell lymphoma-extra-Large (Bcl-xL), myeloid cell leukemia sequence-1 (Mcl-1), Bacterioides fragilis toxin-1 (Bft-1), Bcl-2 antagonist/killer (Bak), Bcl-2-like protein 11 (Bim), B-cell related ovarian killer (Bok), A1 protein, and B-cell lymphoma-w (Bcl-w) were identified as the main anti-apoptosis proteins whereas Bcl-2-associated x protein (Bax), Bcl-2 homologous antagonist killer (Bak) and B-cell lymphoma-xS (Bcl-xS) were characterized as the major pro-apoptotic proteins. Of these several apoptotic regulators, Bcl-2, Bcl-X, Bcl-xL, Mcl-1, Bim, and Bax have been most thoroughly studied in an attempt to determine their precise role in synovial tissue, myeloid DC and chondrocyte apoptosis [53,54,55,56,57,58]. Of note, the expression of Bim, particularly in macrophages was reduced in RA synovial tissue compared to controls [59], whereas Lee et al. [60] showed that IL-17-mediated Bcl-2 expression was dependent on phospho-STAT3 for promoting the survival of RA-FLS.
Therefore based on our understanding of the various components involved in regulating apoptosis we can envision what the main defects are in the T-cell apoptosis cascade that perpetuates the survival of activated nonimmune and immune cells in the RA milieu. To begin with, the results of several classical studies performed with RA cells had already linked defective Fas-signaling, defective Fas Associated Death Domain (FADD) protein synthesis and/or recruitment, over-activation of nuclear factor-κB (NF-κB), altered Signal Transducers and Activators of Transcription-3 (STAT3) signaling, BH3-only proteins, low expression of p53 [61,62,63], low FADD-like IL-1ß-Converting Enzyme (FLICE) Inhibitory Protein (FLIP) levels [61], over-expression of the anti-apoptosis protein, sentrin [64], aberrant PI3K signaling [49] via PTEN [65] to defective regulation of T-cell and RA-FLS apoptosis.
Additional strategies have been exploited in an attempt to induce apoptosis in RA cells. These modalities included treating RA-FLS with anti-Apo2L/TNF-related apoptosis-inducing ligand (TRAIL), also known as CD253 [66]. Of note, apoptosis could be initiated in normal FLS with recombinant TRAIL (rTRAIL) [67]. However, pretreatment of FLS with IFN-γ blunted rTRAIL-induced apoptosis suggesting that pro-inflammatory cytokines such as IFN-γ which activate JAK-STAT signaling [38] may be involved in suppressing rTRAIL-mediated apoptosis in T-cells and RA-FLS. Despite this finding Neve et al. [68] proposed that TRAIL be tested to determine its effectiveness in promoting apoptosis in synoviocytes and infiltrating lymphocytes collected from RA patients. Furthermore, specifically targeting autoreactive CD8+ cells to induce apoptosis in these cell types has also been considered as a preemptive measure designed to reduce the state of chronic inflammation in RA [69].

3. Treg Cell Function

Treg cells are a subset of CD4+CD25+ lymphocytes that express the forkhead box P3 (Foxp3) gene and are responsible for maintaining immune tolerance [70]. Treg cells accomplish this function through the release of IL-10 and transforming growth factor-ß (TGF-ß) resulting in immune suppression. Treg cells suppress self-reactive T-cells through cell-to-cell interaction that is mediated by membrane-bound molecules, such as CTLA-4 [71]. Thus, under normal conditions CTLA-4 blocks T-cell activation by binding to CD80/86 more avidly than CD28. Of note, Treg cells expressing Foxp3 can be distinguished from other T-cell subsets such as Tr1, Th3 and CD8+CD28+, which also exhibit suppressor function.
It has been postulated that the reduced numbers of Th17 cells in patients with established RA may have resulted from the shift of Th17 cells to either Th1 or Treg cells [72]. More recently, Kotake et al. [73] in discussing the plasticity of Th17 cells showed that in peripheral blood of early-onset RA patients, the ratio of CD161+ Th1 was elevated, which correlated with IFN-γ from (INF-γ)+Th17 cells and was inversely related to anti-CCP antibodies.
Indeed, functional defects in Treg cells have been conjectured to play a critical role in the pathogenesis of RA as well as other autoimmune diseases [74], although the majority of previous studies indicated that Treg cells in RA retained some suppressive activity [75,76]. However, when a functional deficiency in Treg cells was reported, the results of a recent study by Sun et al. [77] traced Treg cell functional deficiency in RA to the reduced expression of T-cell immunoglobulin and mucin-domain containing-3 protein (Tim3). Indeed, the proportion of Treg cells defined by both Foxp3 and CD25+ was lower in RA patients than in a control group [78]. However, the proportion of these cells was higher in synovial fluid than in peripheral blood in these RA patients necessitating the need to confirm or refute any differences in Treg cell function between the two compartments. Logically, an extension of these results designed to probe the functionality of Treg cells under various conditions would eventually lead to a spate of recent studies that explored the response of Treg cells to various medical therapies approved for RA. The results of these studies are summarized in Table 1.
Thus, the take-home message from the results of the studies shown in Table 1 is that the number of Treg cells as well as Treg function can be restored with medical therapies that are already approved for RA (e.g., methotrexate, adalimumab, tocilizumab) as well as by tregalizumab, a drug in development for RA. However, study results with abatacept on Treg cell levels were variable with one study indicating a loss of Foxp3-containing cells compared to Ror-γt-containing T-cells [81] whereas another study indicated that abatacept therapy resulted in a rise in Treg cells [82].
Additional recent study results have also illuminated several mechanisms that may be required for the restoration of Treg function in autoimmune arthritis. Thus, Klocke et al. [85] reported that CTLA-4, which contributes to altered Treg function in human RA did not have the same effect on autoreactive T-cells as CTLA-4 had on Treg cells from mice with collagen-induced arthritis (CIA). In the mouse study, the dominant collagen Type-II T-cell epitope was employed to induce arthritis, which was compared to the collagen Type-II epitope mutated at E266D in mouse cartilage. As expected, CTLA-4 expression was required to dampen arthritis severity but only conventional T-cells were required to dampen naïve autoreactive T-cells. However, CTLA-4 expressed on Treg cells prevented inflammation. Taken together the data from this study suggested a “window-of-opportunity” when CTLA-4 expression on Treg cells was likely to be most critical in having an effect tantamount to ameliorating the clinical symptoms of RA.
Another study has identified PTEN as a major contributor to Treg function. Thus, systemic infusion of PTEN to mice with CIA reduced the severity of arthritis while over-expression of PTEN decreased T-cell activation and also differentially modulated Th17 and Treg cell function [86]. Of note, in this study, a deficiency in p53 was accompanied by reduced PTEN gene expression, which also induced phosphorylation of STAT3 and exacerbated autoimmune arthritis. Therefore, this finding suggested that PTEN could potentially be exploited to modify Treg cell function.
Most recently, Safari et al. [87] reported that the genome editing technology known as Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) in combination with the CRISPR-associated (Cas) 9 system had the capacity to alter Treg cells. Thus CRISPR-Cas9 could eventually become useful for recruiting Treg cells ex vivo for use in a modality of RA personalized therapy.

4. Conclusions and Future Perspectives

The inability of T-cells to undergo apoptosis in response to appropriate signaling molecules, such as IL-1ß, TNF-α and Fas, which are capable of inducing cell death under normal conditions, is a hallmark of RA progression. In that regard, it is now recognized that several molecules involved in RA pathophysiology that should be involved in the induction of apoptosis, including CTLA-4, are not working properly. Thus, survival of activated T-cells ensures that both nonimmune cells such as FLS as well as immune cells, including B-cells, macrophages, DCs, mast cells and neutrophils continue to survive where they promote the chronic inflammatory milieu of RA. Therefore the search must continue to identify appropriate therapeutics that will induce T-cell apoptosis which would likely result in ameliorating the progression of damage to synovial tissue, articular cartilage and subchondral bone that are typically present in the synovial joints of RA patients. Another aspect consistent with this theme is the inability of Treg cells to adequately regulate immune responses in RA. Thus, a targeted approach to restoring aberrant Treg cell function may ameliorate the severity of arthritis when tested in experimental animal models of the disease. Moreover, several drugs that have demonstrated clinical efficacy in RA patients, including methotrexate, adalimumab and tocilizumab result in the production of increased numbers of Treg cells with a concomitant improvement in the functioning of these cells. Newer approaches for editing the genome of Treg cells using CRISPR-Cas 9 technology may eventually provide clinicians with the opportunity to treat RA patients with edited Treg cells for personalized medical therapy.

Acknowledgments

The results of studies from the Malemud research group at Case Western Reserve University were funded, in part, by grants from the NIH and Genentech/Roche Group.

Conflicts of Interest

The author declares no conflicts of interest.

References

  1. Firestein, G.S.; McInnes, I.B. Immunopathogenesis of rheumatoid arthritis. Immunity 2017, 46, 183–196. [Google Scholar] [CrossRef] [PubMed]
  2. Toh, M.L.; Moissec, P. The role of T cells in rheumatoid arthritis: New subsets and new targets. Curr. Opin. Rheumatol. 2007, 19, 284–288. [Google Scholar] [CrossRef] [PubMed]
  3. Karouzakis, E.; Neidhart, M.; Gay, R.E.; Gay, S. Molecular and cellular basis of rheumatoid arthritis destruction. Immunol. Lett. 2006, 106, 8–13. [Google Scholar] [CrossRef] [PubMed]
  4. Imboden, J.B. The immunopathogenesis of rheumatoid arthritis. Ann. Rev. Path. 2009, 4, 417–434. [Google Scholar] [CrossRef] [PubMed]
  5. Dadoun, S.; Zeboulon-Ktorza, N.; Combescure, C.; Elhai, M.; Rozenberg, S.; Gossec, L.; Fautrel, B. Mortality in rheumatoid arthritis over the past fifty years: Systematic review and meta-analysis. Joint Bone Spine 2013, 80, 29–33. [Google Scholar] [CrossRef] [PubMed]
  6. Hollan, I.; Meroni, P.L.; Ahearn, J.M.; Cohen Tervaert, J.W.; Curran, S.; Goodyear, C.S.; Hestad, K.A.; Kahaleh, B.; Riggio, M.; Shields, K.; et al. Cardiovascular disease in autoimmune rheumatic diseases. Autoimmun. Rev. 2013, 12, 1004–1015. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Mattar, M.; Jandali, B.; Malemud, C.J.; Askari, A.D. Atherosclerosis and rheumatic diseases. Rheumatology (Sunnyvale) 2015, 5, 147. [Google Scholar]
  8. Matcham, F.; Scott, I.C.; Rayner, L.; Hotopf, M.; Kingsley, G.H.; Norton, S.; Scott, D.L.; Steer, S. The impact of rheumatoid arthritis on quality-of-life assessed using the SF-36: A systematic review and meta-analysis. Semin. Arthritis Rheum. 2014, 44, 123–130. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Bugatti, S.; Codullo, V.; Caporali, R.; Montecucco, C. B cells in rheumatoid arthritis. Autoimmun. Rev. 2007, 6, 482–487. [Google Scholar] [CrossRef] [PubMed]
  10. Choy, E. Understanding the dynamics: Pathways involved in the pathogenesis of rheumatoid arthritis. Rheumatology (Oxford) 2012, 51, v3–v11. [Google Scholar] [CrossRef] [PubMed]
  11. Luu, V.P.; Vazquez, M.I.; Zlotnik, A. B cells participate in tolerance and autoimmunity through cytokine production. Autoimmunity 2014, 47, 1–12. [Google Scholar] [CrossRef] [PubMed]
  12. Veale, D.J.; Orr, C.; Fearon, U. Cellular and molecular perspectives in rheumatoid arthritis. Semin. Immunopathol. 2017, 39, 343–354. [Google Scholar] [CrossRef] [PubMed]
  13. Burmeister, G.R.; Feist, E.; Dörner, T. Emerging cell and cytokine targets in rheumatoid arthritis. Nat. Rev. Rheumatol. 2014, 10, 77–88. [Google Scholar] [CrossRef] [PubMed]
  14. Asquith, D.L.; McInnes, I.B. Emerging cytokine targets in rheumatoid arthritis. Curr. Opin. Rheumatol. 2007, 19, 246–251. [Google Scholar] [CrossRef] [PubMed]
  15. Mateen, S.; Zafar, A.; Moin, S.; Khan, A.Q.; Zubair, S. Understanding the role of cytokines in the pathogenesis of rheumatoid arthritis. Clin. Chim. Acta 2016, 455, 161–171. [Google Scholar] [CrossRef] [PubMed]
  16. Malemud, C.J. Matrix metalloproteases and synovial joint pathology. In Matrix Metalloproteinases and Tissue Remodeling in Health and Disease Part II: Target Tissues and Therapy; Khalil, R.A., Ed.; Progress in Molecular Biology and Translational Science; Elsevier: Amsterdam, The Netherlands, 2017; pp. 306–325. [Google Scholar]
  17. Malemud, C.J. Suppressor of cytokine signaling and rheumatoid arthritis. Integr. Mol. Med. 2016, 3, 17–20. [Google Scholar]
  18. Gensous, N.; Charrier, M.; Duluc, D.; Contin-Bordes, C.; Truchetet, M.E.; Lazaro, E.; Duffau, P.; Blanco, P.; Richez, C. T follicular helper cells in autoimmune diseases. Front. Immunol. 2018, 9, 1637. [Google Scholar] [CrossRef] [PubMed]
  19. DiToro, D.; Winstead, C.J.; Pham, D.; Witte, S.; Andargachew, R.; Singer, J.R.; Wilson, C.G.; Zindl, C.L.; Luther, R.J.; Silberger, D.J.; et al. Differential IL-2 expression defines developmental fates of follicular versus nonfollicular helper T cells. Science 2018, 361, 1086. [Google Scholar] [CrossRef] [PubMed]
  20. Niu, Q.; Huang, Z.C.; Wu, X.J.; Jin, Y.X.; An, Y.F.; Li, Y.M.; Xu, H.; Yang, B.; Wang, L. Enhanced IL-6/phosphorylated STAT3 signaling is related to the imbalance of circulating T follicular helper/T follicular regulatory cells in patients with rheumatoid arthritis. Arthritis Res. Ther. 2018, 20, 200. [Google Scholar] [CrossRef] [PubMed]
  21. Wang, X.; Yang, C.; Xu, F.; Qi, L.; Wang, J.; Yang, P. Imbalance of circulating Tfr/Tfh in patients with rheumatoid arthritis. Clin. Exp. Med. 2018. [Google Scholar] [CrossRef] [PubMed]
  22. Herrero-Beaumont, G.; Martinez Calatrava, M.J.; Castañeda, S. Abatacept mechanism in action: Coincidence with its clinical profile. Rheumatol. Clin. 2012, 8, 78–83. [Google Scholar] [CrossRef]
  23. Maggi, L.; Cimaz, R.; Capone, M.; Santarlasci, V.; Rossi, M.C.; Mazzoni, A.; Montaini, G.; Pagnini, I.; Giani, T.; Simonini, G.; et al. Immunosuppressive activity of abatacept on circulating T helper lymphocytes from juvenile idiopathic arthritis patients. Int. Arch. Allergy Immunol. 2016, 171, 45–53. [Google Scholar] [CrossRef] [PubMed]
  24. Pandya, J.M.; Lundell, A.C.; Hallström, M.; Andersson, K.; Nordström, I.; Rudin, A. Circulating T helper and T regulatory subsets in untreated early rheumatoid arthritis and healthy control subjects. J. Leukoc. Biol. 2016, 100, 823–833. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Zamani, M.R.; Aslani, S.; Salmaninejad, A.; Javan, M.B.; Razaei, N. PD-1/PD-L and autoimmunity: A growing relationship. Cell. Immunol. 2016, 310, 27–41. [Google Scholar] [CrossRef] [PubMed]
  26. Schiotis, R.E.; Buzoianu, A.D.; Muresanu, D.E.; Suciu, S. New pharmacological strategies in rheumatic diseases. J. Med. Life 2016, 9, 227–234. [Google Scholar] [PubMed]
  27. Malemud, C.J.; Gillespie, H.J. The role of apoptosis in arthritis. Curr. Rheum. Rev. 2005, 1, 131–142. [Google Scholar] [CrossRef]
  28. Fox, D.A.; Gizinski, A.; Morgan, B.; Lundy, S.K. Cell-cell interactions in rheumatoid arthritis synovium. Rheum. Dis. Clin. N. Am. 2010, 36, 311–323. [Google Scholar] [CrossRef] [PubMed]
  29. Malemud, C.J. Apoptosis resistance in rheumatoid arthritis synovial tissue. J. Clin. Cell. Immunol. 2012, S3, 6. [Google Scholar] [CrossRef]
  30. Malemud, C.J.; Reddy, S.K. Targeting cytokines, chemokines and adhesion molecules in rheumatoid arthritis. Curr. Rheum. Rev. 2008, 4, 219–234. [Google Scholar] [CrossRef]
  31. Szekanecz, Z.; Koch, A.E. Macrophages and their products in rheumatoid arthritis. Curr. Opin. Rheumatol. 2007, 19, 482–487. [Google Scholar] [CrossRef] [PubMed]
  32. Petrovic-Rackov, P.; Pejnovic, N. Clinical significance of IL-18, IL-15, IL-12 and TNF-α measurement in rheumatoid arthritis. Clin. Rheumatol. 2006, 25, 448–452. [Google Scholar] [CrossRef] [PubMed]
  33. Malemud, C.J. Molecular mechanisms in rheumatic diseases. Rationale for novel drug development—Introduction. Anti-Inflamm. Anti-Allergy Agents Med. Chem. 2011, 10, 73–77. [Google Scholar] [CrossRef]
  34. Sarkar, S.; Fox, D.A. Targeting IL-17 and Th17 cells in rheumatoid arthritis. Rheum. Dis. Clin. N. Am. 2010, 36, 345–366. [Google Scholar] [CrossRef] [PubMed]
  35. Malemud, C.J. Growth hormone, VEGF and FGF. Involvement in rheumatoid arthritis. Clin. Chim. Acta 2007, 375, 10–19. [Google Scholar] [CrossRef] [PubMed]
  36. Yokota, A.; Narazaki, N.; Shima, Y.; Murata, N.; Tanaka, T.; Suemura, M.; Yoshizaki, K.; Fujiwara, H.; Tsuyuguchi, I.; Kishimoto, T. Preferential and persistent activation of the STAT1 pathway in rheumatoid synovial fluid cells. J. Rheumatol. 2001, 28, 1952–1959. [Google Scholar] [PubMed]
  37. Kasperkovitz, P.V.; Verbeet, N.L.; Smeets, T.J. Activation of the STAT1 pathway in rheumatoid arthritis. Ann. Rheum. Dis. 2006, 65, 149–156. [Google Scholar] [CrossRef]
  38. Malemud, C.J.; Pearlman, E. Targeting JAK/STAT signaling pathway in inflammatory diseases. Curr. Signal. Transduct. Ther. 2009, 4, 201–221. [Google Scholar] [CrossRef]
  39. Malemud, C.J. The role of the JAK/STAT signal pathway in rheumatoid arthritis. Ther. Adv. Musculoskel. Dis. 2018, 10, 117–127. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Mertens, M.; Singh, J.A. Anakinra for rheumatoid arthritis: A systematic review. J. Rheumatol. 2009, 36, 1118–1125. [Google Scholar] [CrossRef] [PubMed]
  41. Malemud, C.J. Small molecular weight inhibitors of stress-activated and mitogen-activated protein kinases. Mini-Rev. Med. Chem. 2006, 6, 689–698. [Google Scholar] [CrossRef] [PubMed]
  42. Korb, A.; Tohidast-Akrad, M.; Cetin, E.; Axman, R.; Smolen, J.; Schett, G. Differential tissue expression and activation of p38 MAPK α, ß, γ, δ isoforms in rheumatoid arthritis. Arthritis Rheum. 2006, 54, 2745–2756. [Google Scholar] [CrossRef] [PubMed]
  43. Malemud, C.J. Intracellular signaling pathways in rheumatoid arthritis. J. Clin. Cell. Immunol. 2013, 4, 160. [Google Scholar] [CrossRef] [PubMed]
  44. Ramadani, F.; Bolland, D.J.; Garcon, F.; Emery, J.L.; Vanhaesebroeck, B.; Corcoran, A.E.; Okkenhaug, K. The PI3K isoforms p110α and p108δ are essential for pre-B cell receptor signaling and B cell development. Sci. Signal. 2010, 3, Ra60. [Google Scholar] [CrossRef] [PubMed]
  45. Benham-Hall, E.; Clatworthy, M.R.; Okkenhaug, K. The therapeutic potential for PI3K inhibitors in autoimmune rheumatic diseases. Open Rheumatol. J. 2012, 6, 245–258. [Google Scholar] [CrossRef] [PubMed]
  46. Safina, B.S.; Baker, S.; Baumgardner, M.; Blaney, P.M.; Chan, B.K.; Chen, Y.H.; Cartwright, M.W.; Castanedo, G.; Chabot, C.; Cheguillaume, A.J.; et al. Discovery of novel 3K δ specific inhibitors for the treatment of rheumatoid arthritis: Taming CYP3A4 time-dependent inhibition. J. Med. Chem. 2012, 55, 5587–5900. [Google Scholar] [CrossRef] [PubMed]
  47. Bartok, B.; Boyle, D.L.; Liu, Y.; Ren, P.; Ball, S.T.; Bugbee, W.D.; Rommel, C.; Firestein, G.S. PI3 kinase δ is a key regulator of synoviocyte function in rheumatoid arthritis. Am. J. Pathol. 2012, 180, 1906–1916. [Google Scholar] [CrossRef] [PubMed]
  48. Haruta, K.; Mori, S.; Tamura, N.; Sasaki, A.; Nagamine, M.; Yaguchi, S.; Kamachi, F.; Enami, J.; Kobayashi, S.; Yamori, T.; et al. Inhibitory effects of ZSTK474, a phosphatidylinositol-3-kinase inhibitor, on adjuvant-induced arthritis in rats. Inflamm. Res. 2012, 61, 551–562. [Google Scholar] [CrossRef] [PubMed]
  49. Malemud, C.J. PI3K/Akt/PTEN/mTOR signaling: A fruitful target for inducing cell death in rheumatoid arthritis? Future Med. Chem. 2015, 7, 1137–1147. [Google Scholar] [CrossRef] [PubMed]
  50. Reed, J.C. Mechanisms of apoptosis. Am. J. Pathol. 2000, 157, 1415–1430. [Google Scholar] [CrossRef]
  51. Gupta, S. Molecular signaling in death receptors and mitochondrial pathways of apoptosis. Int. J. Oncol. 2003, 22, 15–20. [Google Scholar] [CrossRef] [PubMed]
  52. Shultz, D.R.; Harrington, W.J., Jr. Apoptosis: Programmed cell death at a molecular level. Semin. Arthritis Rheum. 2003, 32, 345–369. [Google Scholar] [CrossRef] [PubMed]
  53. Mountz, J.D.; Zhang, H.G. Regulation of apoptosis of synovial fibroblasts. Curr. Dir. Autoimmun. 2001, 3, 216–239. [Google Scholar] [PubMed]
  54. Islam, N.; Haqqi, T.M.; Jepsen, K.J.; Kraay, M.; Welter, J.F.; Goldberg, V.M.; Malemud, C.J. 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. J. Cell. Biochem. 2002, 87, 266–278. [Google Scholar] [CrossRef] [PubMed]
  55. Liu, H.; Eksarko, P.; Temkin, V.; Haines, G.K., 3rd; Perlman, H.; Koch, A.E.; Thimmapaya, R.; Pope, R.M. Mcl-1 is essential for the survival of synovial fibroblasts in rheumatoid arthritis. J. Immunol. 2005, 175, 8337–8345. [Google Scholar] [CrossRef] [PubMed]
  56. Lomonosova, E.; Chinnadurai, G. BH3-only proteins in apoptosis and beyond: An overview. Oncogene 2008, 27 (Suppl. 1), S2–S19. [Google Scholar] [CrossRef]
  57. Hutcheson, J.; Perlman, H. Apoptotic regulators and RA. Curr. Rheum. Rev. 2008, 4, 254–258. [Google Scholar] [CrossRef]
  58. Olsson Åkefeldt, S.; Ismail, M.B.; Valentin, H.; Aricò, M.; Henter, J.I.; Delprat, C. Targeting BCL-2 family in human myeloid dendritic cells: A challenge to cure diseases with chronic inflammation associated with bone loss. Clin. Dev. Immunol. 2013, 2013, 701305. [Google Scholar] [CrossRef] [PubMed]
  59. Scatizzi, C.; Hutcheson, J.; Pope, R.M.; Firestein, G.S.; Koch, A.E.; Mavers, M.; Smason, A.; Agarwal, H.; Haines, G.K., 3rd; Chandel, N.S.; et al. Bim-Bcl-2 homology mimetic therapy is effective in suppressing inflammatory arthritis through the activation of myeloid cell apoptosis. Arthritis Rheum. 2010, 62, 441–451. [Google Scholar] [CrossRef] [PubMed]
  60. Lee, S.Y.; Kwok, S.K.; Son, H.J.; Ryu, J.G.; Kim, E.K.; Oh, H.J.; Cho, M.L.; Ju, J.H.; Park, S.H.; Kim, H.Y. IL-17-mediated Bcl-2 expression regulates survival of fibroblast-like synoviocytes in rheumatoid arthritis through STAT3 activation. Arthritis Res. Ther. 2013, 15, R31. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. Baier, A.; Meineckel, I.; Gay, S.; Pap, T. Apoptosis in rheumatoid arthritis. Curr. Opin. Rheumatol. 2003, 15, 274–279. [Google Scholar] [CrossRef] [PubMed]
  62. Liu, H.; Pope, R.M. The role of apoptosis in rheumatoid arthritis. Curr. Opin. Pharmacol. 2003, 3, 317–322. [Google Scholar] [CrossRef]
  63. Hutcheson, J.; Perlman, H. BH3-only proteins in rheumatoid arthritis: Potential targets for therapeutic intervention. Oncogene 2008, 27, 168S. [Google Scholar] [CrossRef] [PubMed]
  64. Franz, J.K.; Pap, T.; Hummel, K.M.; Nawrath, M.; Aicher, W.K.; Shigeyama, Y.; Müller-Ladner, U.; Gay, R.E.; Gay, S. Expression of sentrin, a novel antiapoptotic molecule, at sites of synovial invasion, in rheumatoid arthritis. Arthritis Rheum. 2000, 43, 599–607. [Google Scholar] [CrossRef]
  65. Pap, T.; Franz, J.K.; Hummel, K.M.; Jeisy, E.; Gay, R.; Gay, S. Activation of synovial fibroblasts in rheumatoid arthritis: Lack of expression of the tumour suppressor PTEN at sites of invasive growth and destruction. Arthritis Res. 2000, 2, 59–64. [Google Scholar] [CrossRef] [PubMed]
  66. Tsokos, G.C.; Tsokos, M. The TRAIL to arthritis. J. Clin. Investig. 2003, 112, 1315–1317. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Tamai, M.; Kawakami, A.; Tanaka, F.; Miyashita, T.; Nakamura, H.; Iwanaga, N.; Izumi, Y.; Arima, K.; Aratake, K.; Huang, M.; et al. Significant inhibition of TRAIL-mediated fibroblast-like synovial cells apoptosis by IFN-γ through JAK/STAT pathway by translational regulation. J. Lab. Clin. Med. 2006, 147, 182–190. [Google Scholar] [CrossRef] [PubMed]
  68. Neve, A.; Corrado, A.; Cantatore, F.P. TNF-related apoptosis-inducing ligand (TRAIL) in rheumatoid arthritis: What’s new? Clin. Exp. Med. 2014, 14, 115–120. [Google Scholar] [CrossRef] [PubMed]
  69. Citro, A.; Barnaba, V.; Martini, H. From T cell apoptosis to chronic immune activation in inflammatory diseases. Int. Arch. Allergy Immunol. 2014, 164, 140–146. [Google Scholar] [CrossRef] [PubMed]
  70. Tao, J.H.; Cheng, M.; Tang, J.P.; Liu, Q.; Pan, F.; Li, X.P. Foxp3, regulatory T cell and autoimmune diseases. Inflammation 2017, 40, 328–339. [Google Scholar] [CrossRef] [PubMed]
  71. Langdon, K.; Haleagrahara, N. Regulatory T-cell dynamics with abatacept treatment in rheumatoid arthritis. Int. Rev. Immunol. 2018, 37, 206–214. [Google Scholar] [CrossRef] [PubMed]
  72. Kotake, S.; Nanke, Y.; Yago, T.; Kawamoto, M.; Kobashigawa, T.; Yamanaka, H. Elevated ratio of Th17 cell-derived Th1 cells (CD161+Th1 cells) to CD161+Th17 cells in peripheral blood of early-onset rheumatoid arthritis patients. Biomed. Res. Int. 2016, 2016, 418602. [Google Scholar] [CrossRef] [PubMed]
  73. Kotake, S.; Yago, T.; Kobashigawa, T.; Nanke, Y. The plasticity of Th17 cells in the pathogenesis of rheumatoid arthritis. J. Clin. Med. 2017, 6, 67. [Google Scholar] [CrossRef] [PubMed]
  74. Miao, J.; Zhu, P. Functional defects of Treg cells: New targets in rheumatic diseases, including ankylosing spondylitis. Curr. Rheumatol. Rep. 2018, 20, 30. [Google Scholar] [CrossRef] [PubMed]
  75. Walter, G.J.; Fleskins, V.; Frederiksen, K.S.; Rajasekhar, M.; Menon, B.; Gerwien, J.G.; Evans, H.G.; Taams, L.S. Phenotypic, functional, and gene expression profiling of peripheral CD45RA+ and CD45RO+ CD4+CD25+CD127low Treg cells in patients with chronic rheumatoid arthritis. Arthritis Rheumatol. 2016, 68, 103–116. [Google Scholar] [CrossRef] [PubMed]
  76. Zaragoza, R.; Chen, X.; Oppenheim, J.J.; Baevens, A.; Gregoire, S.; Chader, D.; Gorochov, G.; Miyara, M.; Salomon, B.L. Suppressive activity of human regulatory T cells is maintained in the presence of TNF. Nat. Med. 2016, 22, 16–17. [Google Scholar] [CrossRef] [PubMed]
  77. Sun, H.; Gao, W.; Pan, W.; Zhang, Q.; Wang, G.; Feng, D.; Geng, X.; Yan, X.; Li, S. Tim3+ Foxp3+ Treg cells are potent inhibitors of effector T cells and are suppressed in rheumatoid arthritis. Inflammation 2017, 40, 1342–1350. [Google Scholar] [CrossRef] [PubMed]
  78. Morita, T.; Shima, Y.; Wing, J.B.; Sakaguchi, S.; Ogata, A.; Kumanogoh, A. The proportion of regulatory T cells in patients with rheumatoid arthritis: A. meta-analysis. PLoS ONE 2016, 11, e0162306. [Google Scholar] [CrossRef] [PubMed]
  79. Gupta, V.; Katiyar, S.; Singh, A.; Misra, R.; Aggarwal, A. CD39 positive regulatory T cell frequency as a biomarker of treatment response to methotrexate in rheumatoid arthritis. Int. J. Rheum. Dis. 2018, 21, 1548–1556. [Google Scholar] [CrossRef] [PubMed]
  80. Cribbs, A.P.; Kennedy, A.; Penn, H.; Amjadi, P.; Green, P.; Read, J.E.; Brennan, F.; Gregory, B.; Williams, R.O. Methotrexate restores regulatory T cell function through demethylation of the FoxP3 upstream enhancer in patients with rheumatoid arthritis. Arthritis Rheumatol. 2015, 67, 1182–1192. [Google Scholar] [CrossRef] [PubMed]
  81. Tada, Y.; Ono, N.; Suematsu, R.; Tashiro, S.; Sadanaga, Y.; Tokuda, Y.; Ono, Y.; Nakao, Y.; Maruyama, A.; Ohta, A.; et al. The balance between Foxp3 and ROR-γt expression in peripheral blood by tocilizumab and abatacept in patients with rheumatoid arthritis. BMC Musculoskelet. Disord. 2016, 17, 290. [Google Scholar] [CrossRef] [PubMed]
  82. Bonelli, M.; Göschl, L.; Blümi, S.; Karonitsch, T.; Hirahara, K.; Ferner, E.; Steiner, C.W.; Steiner, G.; Smolen, J.S.; Scheinecker, C. Abatacept (CTLA-4Ig) treatment reduces T cell apoptosis and regulatory T cell suppression in patients with rheumatoid arthritis. Rheumatology (Oxford) 2016, 55, 710–720. [Google Scholar] [CrossRef] [PubMed]
  83. König, M.; Rharbaoui, F.; Aigner, S.; Dälken, B.; Schüttrumpf, J. Tregalizumab—A monoclonal antibody to target regulatory T cells. Front. Immunol. 2016, 7, 11. [Google Scholar] [CrossRef] [PubMed]
  84. Nguyen, D.X.; Cotton, A.; Attipoe, L.; Ciurtin, C.; Doré, C.J.; Ehrenstein, M.R. Regulatory T cells as a biomarker for response to adalimumab in rheumatoid arthritis. J. Allergy Clin. Immunol. 2018, 142, 978–980.e9. [Google Scholar] [CrossRef] [PubMed]
  85. Klocke, K.; Holmdahl, R.; Wing, K. CTLA-4 expressed by FOXP3 regulatory T cells prevents inflammatory tissue attack and not T-cell priming in arthritis. Immunology 2017, 152, 125–137. [Google Scholar] [CrossRef] [PubMed]
  86. Lee, S.H.; Park, J.-S.; Byun, J.-K.; Jhun, J.; Jung, K.; Seo, H.-B.; Moon, Y.-B.; Kim, H.-Y.; Park, S.-H.; Cho, M.-L. PTEN ameliorates autoimmune arthritis through down-regulating STAT3 activation with reciprocal balance of Th17 and Tregs. Sci. Rep. 2016, 6, 34617. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Safari, F.; Farajnia, S.; Arya, M.; Zarredar, H.; Nasrolahi, A. CRISPR and personalized Treg therapy: New insights into the treatment of rheumatoid arthritis. Immunopharmacol. Immunotoxicol. 2018, 40, 201–211. [Google Scholar] [CrossRef] [PubMed]
Table 1. The Effect of RA Drug Therapies on Treg Cells.
Table 1. The Effect of RA Drug Therapies on Treg Cells.
Drug Therapy & TargetEffect(s) on Treg CellsReference
Methotrexate (General Immunosuppressant)↑ Frequency of CD39+ 1 and CD4+ CD25+ CD39+ 1 Treg Cells[79]
MethotrexateRestored Treg cell function via demethylation of FOXP3 locus[80]
Abatacept (Target: CTLA-4;CD80/86-CD28 Blockade)↓ Foxp3+/Ror-γt 2[81]
Abatacept↑ Treg cells; Diminished suppression of responder T-cell proliferation in RA[82]
Tocilizumab (Target: membrane and soluble IL-6R)↑ Foxp3+/Ror-γt 2[81]
Tregalizumab 3 (Target: CD39)Induced Treg Cell Activation[83]
Adalimumab (Target: TNF-α)↑ Treg cells in RA patients who responded favorably to treatment [84]
1 CD39 is an ectonucleotidase highly expressed on Treg Cells. 2 A transcription factor that characterizes Th17 cells; 3 humanized CD4-specific monoclonal antibody.

Share and Cite

MDPI and ACS Style

Malemud, C.J. Defective T-Cell Apoptosis and T-Regulatory Cell Dysfunction in Rheumatoid Arthritis. Cells 2018, 7, 223. https://0-doi-org.brum.beds.ac.uk/10.3390/cells7120223

AMA Style

Malemud CJ. Defective T-Cell Apoptosis and T-Regulatory Cell Dysfunction in Rheumatoid Arthritis. Cells. 2018; 7(12):223. https://0-doi-org.brum.beds.ac.uk/10.3390/cells7120223

Chicago/Turabian Style

Malemud, Charles J. 2018. "Defective T-Cell Apoptosis and T-Regulatory Cell Dysfunction in Rheumatoid Arthritis" Cells 7, no. 12: 223. https://0-doi-org.brum.beds.ac.uk/10.3390/cells7120223

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop