Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2016 Aug 1.
Published in final edited form as: Exp Hematol. 2015 Jul 2;43(8):587–598. doi: 10.1016/j.exphem.2015.05.016

Deconstructing innate immune signaling in Myelodysplastic Syndromes

Melinda E Varney 1, Katelyn Melgar 1,2, Madeline Niederkorn 1,3,#, Molly Smith 1,3,#, Laura Barreyro 1,#, Daniel T Starczynowski 1,3
PMCID: PMC4635673  NIHMSID: NIHMS706877  PMID: 26143580

Abstract

Overexpression of immune-related genes is widely reported in Myelodysplastic syndromes (MDS), and chronic immune stimulation increases the risk for developing MDS. Aberrant innate immune activation, such as due to increased Toll-like receptor (TLR) signaling, in MDS can contribute to systemic effects on hematopoiesis in addition to cell-intrinsic defects on hematopoietic stem/progenitor cell (HSPC) function. This review will deconstruct aberrant function of TLR signaling mediators within MDS HSPC that may contribute to cell intrinsic consequences on hematopoiesis and disease pathogenesis. We will discuss the contribution of chronic TLR signaling to the pathogenesis of MDS based on evidence from patients and mouse genetic models.

Introduction

Myelodysplastic syndromes (MDS) are heterogeneous clonal hematopoietic stem cell (HSC) disorders1-6. MDS patients present with blood cytopenia, myeloid cell dysplasia, and ineffective hematopoiesis due to de novo mutations or prior chemotherapy7,8. As patients progress towards bone marrow failure (BMF), ensuing hematologic complications are fatal if untreated. Moreover, approximately one third of MDS patients also develop AML due to acquisition of additional mutations in the defective hematopoietic stem/progenitor cells (HSPC)9,10. MDS is comprised of distinct subtypes based on biological, genetic, and morphological features11. Regardless of subtype, MDS arises from a HSC that has acquired genetic and/or epigenetic abnormalities3-5. MDS are genetically defined by somatic mutations and chromosomal abnormalities affecting epigenetic plasticity, ribosome function, spliceosome function, and immune signaling. Overexpression of immune-related genes is widely reported in MDS12,13, and chronic inflammation and autoimmune disorders increase the risk of developing MDS14. Multiple independent observations suggest that hyperactivation of innate immune/Toll-like receptor (TLR) signaling is a feature of MDS: association with chronic inflammatory and autoimmune disorders, increased expression of inflammatory cytokines and chemokines, abnormal cellular immunity, and overexpression of TLR mediators, along with reduced expression of negative TLR regulators. The importance of innate immune signaling in primary MDS also is supported by the use of IRAK1/4 inhibitors in preclinical models as effective agents to suppress innate immune activation and the MDS clone15.

Aberrant innate immune activation in MDS can contribute to non-cell autonomous effects on hematopoiesis (Figure 1) in addition to cell-intrinsic defects within hematopoietic stem/progenitor cells (HSPC) (Figure 2). Several recent comprehensive reviews have described the systemic and non-cell autonomous consequences of chronic innate immune activation in MDS, including the effects of increased inflammation and abnormal adaptive and innate immunity on BM HSC16,17. Herein, we will systematically deconstruct aberrant function of TLR signaling mediators within MDS HSC that may contribute to the cell intrinsic consequences associated with ineffective hematopoiesis and disease pathogenesis (Figure 2).

Figure 1. Overview of non-cell autonomous effects of chronic innate immune activation in MDS.

Figure 1

The MDS bone marrow (BM) consists of MDS-derived hematopoietic cells and residual normal cells. The non-cell autonomous consequences of the MDS-derived BM include increased expression of cytokines, expansion of myeloid-derived suppressor cells (MDSC), and lymphocyte-mediated attack on hematopoietic cells. In addition, MDS HSC affect mesenchymal stromal cells (MSC), therefore altering the HSC niche and hematopoietic differentiation.

Figure 2. Overview of cell-intrinsic TLR activation in MDS HSPC cells.

Figure 2

Toll-like receptor (TLR) and IL-1 receptor (IL1R) on HSPC recruit MyD88 and IRAK4, which initiate the assembly of the Myddosome complex. TIRAP can also increase the efficiency of Myddosome assembly by binding MyD88. IRAK4, a serine/threonine kinase, activates IRAK2 and/or IRAK1 through IRAK4-dependent phosphorylation. IRAK1 associates with an E3 Ub ligase, TRAF6, which mediates activation of NF-kB (IKKα/β/γ complex) and MAPK (TAB2/TAB3/TAK1) through K63-linked Ub chains. TRAF6 can also regulate other proteins (indicated by “?”) that may contribute to immune signaling and MDS. miR-146a suppresses IRAK1 and TRAF6 protein expression by direct binding at 3′UTR sites within IRAK1 and TRAF6 mRNAs. A20 suppresses TRAF6 by de-ubiquitinating TRAF6 K63 linkages. Reduced levels of miR-146a or A20 result in increased IRAK1/TRAF6-mediated signaling, impaired HSPC function, and altered myeloid differentiation. The red boxes represent molecules that are overexpressed/activated in MDS. The green boxes represent molecules that are deleted/downregulated in MDS.

Mediators of Innate Immune Signaling in MDS

Toll-Like Receptors

The innate immune system recognizes foreign pathogens by cell surface pattern recognition receptors (PRRs). These receptors recognize foreign pathogen components, termed pathogen-associated molecular patterns (PAMPs), as well as host cellular by-products, referred to as damage-associated molecular patterns (DAMPs). Among the first PRRs to be identified were TLRs. Together with the Interleukin-1 receptor (IL1R), TLRs form the TLR/IL1R (TIR) superfamily, which share a common a TIR domain. TLRs consist of a single-pass transmembrane protein with a leucine-rich ectodomain. There are 10 human and 12 murine TLRs, which can be divided into two main groups based on subcellular location: extracellular (TLR1, TLR2, TLR4, TLR5, TLR6, and TLR11) and intracellular, or endosomal (TLR3, TLR7, TLR8, and TLR9). The location of the receptors, in turn, relates to their specific ligands. The intracellular receptors bind to features of pathogen nucleic acids, such as dsRNA, and unmethylated CpG DNA; whereas the extracellular receptors bind to pathogen membrane components, the best characterized being TLR4 binding to lipopolysaccharide (LPS) of gram-negative bacteria.

Binding of a TLR to its ligand results in recruitment of a TIR domain-containing adaptor protein. There are two main TLR adaptor proteins, Myeloid differentiation primary response (MyD88) and TIR-domain-containing adapter-inducing interferon-β (TRIF), which induce the activation of separate innate immune signaling pathways. All of the TLRs, with the exception of TLR3, use MyD88 (Figure 1). Through a series of mediators, signaling through this adaptor results in activation of MAPK and NF-κB pathways, leading to transcription of pro-inflammatory cytokines, such as tumor necrosis factor alpha (TNFα), interleukin-1 (IL-1), IL-8, and IL-6. Instead of MyD88, TLR3 recruits TRIF. Signaling through TRIF results in activation of IRF3 and MyD88-independent activation of NF-κB, leading to transcription of similar pro-inflammatory cytokines as the MyD88 pathway with the addition of type 1 interferons. TLR4 is the only receptor that utilizes both MyD88 and TRIF18-20.

Overactivation of TLRs have been associated with autoinflammatory disorders. More recently, TLR signaling has been implicated in HSC development21,22, suggesting that changes in TLR expression and function could lead to dysregulated hematopoiesis. Several mouse studies have shown that chronic administration of low levels of LPS in vivo, meant to model chronic infection, result in loss of HSC quiescence, increased HSCs, myeloid skewing, and decreased progenitor function23-25. Taken together, chronic TLR signaling impairs HSC function and alters normal hematopoiesis, which suggests a causal role in MDS.

Indeed, overexpression or gain-of-function mutations in a subset of TLR has been described in MDS. TLR4 is overexpressed at both the mRNA and protein levels in CD34+ cells isolated from the bone marrow (BM) of MDS patients26. MDS CD34+ cells with overexpressed TLR4 exhibited normal functional capacity as assessed by ICAM-1 expression after LPS stimulation. Interestingly, TNFα was shown to increase TLR4 expression in a dose-dependent manner, while depletion of TNFα with anti-TNF antibody treatment of MDS BM cells resulted in decreased TLR4 expression, suggesting that the TLR4 expression is TNFα dependent. Furthermore, Maratheftis et al. found a significant correlation between TLR4 expression and apoptosis in CD34+ MDS BM cells, providing correlative evidence for increased TLR4 expression and cell intrinsic defects associated with MDS HSPC. However, TLR4 expression within the total BM population from MDS patients did not differ from normal controls, nor was there a correlation between TNFα levels and TLR4 expression27. Kuninaka et al. described TLR9 overexpression in MDS BM cells, which positively correlated with TNFα levels. Interestingly, TLR9 expression decreased once the MDS transformed to leukemia in these patients, further supporting the link between TLR expression and balance of cell survival signals in MDS. Both of these studies also describe TLR2 overexpression in MDS HSPC, and through deep sequencing analysis, a more recent study identified a TLR2 gain-of-function variant (F217S) in the CD34+ BM cells of 11% of MDS patients examined28. Treatment with the TLR2 agonist MALP2 inhibited erythroid differentiation in healthy CD34+ BM cells28. Furthermore shRNA knockdown of TLR2 in MDS CD34+ BM cells increased erythroid progenitors in colony formation assays28.

Taken together, these studies provide evidence for the role of HSPC-expressed TLRs in the pathogenesis of MDS. There is some discrepancy in which specific TLRs are most important in the context of MDS since they regulate many of the same pathways. The specific TLR, however, may not be as important to identify as the hematopoietic lineage exhibiting overexpression of the TLR and the downstream effects that result from TLR dysregulation. Inhibition of the induction of these receptors, particularly through TNFα, which was implicated in both TLR4 and TLR9 expression, may be an attractive therapeutic strategy in the treatment MDS.

MyD88

MyD88, one of two intracellular TLR adaptor proteins, functions as a central node for innate immune signaling. It contains three domains: a death domain (DD), an intermediate domain, and a TIR domain29,30. Upon TLR activation, TIR-TIR domain interaction recruits MyD88 to activated TLRs31. Following this interaction, IL-1 receptor associated kinase 4 (IRAK4), a serine-threonine kinase, complexes with MyD88 through the association of their respective DDs (Figure 2). IRAK family members (such as IRAK1 and IRAK2) are subsequently recruited and phosphorylated, forming a multiprotein complex referred to as the Myddosome32,33. Phosphorylated IRAK1 and IRAK2 can then interact with TNF receptor associated factor 6 (TRAF6), an E3 ubiquitin (Ub) ligase, which ubiquitinates and activates TAK134,35. Activated TAK1 then signals to the MAPK and NF-κB pathways35. Moreover, MyD88 knockout mice exhibit hematopoietic system, immune signaling, and cell survival defects. The immune system abnormalities in these animals result in increased susceptibility to bacterial and viral infections36.

Aberrant MyD88 expression and function is widely implicated in hematopoietic disease. Somatic mutation of Leucine-265 to Proline (L265P) within the TLR domain are observed in lymphoid malignancies and Waldenstrom macroglobulinemia, as well as in del(5q) MDS following gammopathy of undetermined significance37,38. L265P results in a gain of function MyD88 protein, as the mutated TIR domain of MyD88 displays an intrinsic propensity for augmented oligomerization and spontaneous formation of cytosolic Myddosome aggregates39. Although the level of MyD88-L265P expression in malignant lymphoid cells is comparable to wildtype MyD88 in normal tissues, its molecular consequences resemble that of MyD88 overexpression, indicating that L265P or overexpression of wildtype results in increased MyD88 signaling. Although MyD88 is not commonly mutated in myeloid malignancies, overexpression of wildtype MyD88 mRNA and protein is reported in MDS HSPC. Dimicoli et al. found that MyD88 mRNA is increased in the CD34+ BM cells of 40% of MDS patients examined. Furthermore, MDS patients with increased MyD88 expression in CD34+ BM cells were found to have shorter survival than patients with lower MyD88 expression, indicating that MyD88 expression has prognostic value40. Blocking MyD88 signaling with a 26-amino acid inhibitory peptide in CD34+ cells from lower risk MDS patients results in increased differentiation of the erythroid lineage, coinciding with upregulation of erythroid differentiation genes. Additionally, hematopoietic differentiation genes, including GATA1 and GATA2, were upregulated in these cells, with an increased GATA1/GATA2 ratio, supporting the positive effect of a MyD88 blockade on erythroid differentiation of MDS CD34+ cells. Similarly, inhibition of IL-8, a downstream inflammatory cytokine dependent on MyD88, results in increased erythroid differentiation of CD34+ cells from lower risk MDS patients40,41. Taken together, these findings implicate MyD88 activation and suggest a potential therapeutic role for inhibiting MyD88 and downstream effector signaling in MDS.

TIRAP

Myddosome assembly is also catalyzed by TIR domain containing adaptor protein (TIRAP) (Figure 2). TIRAP-deficient mice respond normally to TLR5, TLR7, and TLR9 ligands and to IL-1 and IL-18; however they exhibit defects in cytokine production and NF-κB and MAPK activation in response to LPS and bacterial lipoproteins42. TIRAP is composed of 3 domains: a TIR domain, a PEST domain, and an N-terminal PIP2 binding domain. TIRAP primarily functions as a molecular bridge between MyD88 and receptor complexes for TLR-2 and TLR-4 signaling. TIRAP recruits cytosolic MyD88 to interact with activated TIR domains of TLR dimers at the cell membrane, thus catalyzing formation of the Myddosome43-45.

TIRAP has MyD88-independent functions, such as activation of Rac1-PI3K-Akt signaling46. Additionally, TIRAP binds TRAF6 and recruits it to the plasma membrane, which is required for the serine phosphorylation of p65/RelA47. TLR2- and TLR4-induced NF-κB activation requires Bruton’s tyrosine kinase (Btk) phosphorylation of TIRAP. Also relevant to hematopoietic malignancies, TIRAP enhances cAMP responsive element protein (CREB) activation by signaling through TRAF6, Pellino3, p38 MAPK, and MK248.

The relevance of TIRAP to MDS was based on findings related to miR-145 expression, which targets TIRAP mRNA and is located within the commonly deleted region (CDR) on chromosome 5 (q33.1) in MDS49. Since TIRAP is a direct target of miR-145, MDS patients with deletion of miR-145 exhibit increase TIRAP mRNA and protein expression49. Using retroviral approaches, Ibrahim et al. demonstrated that TIRAP overexpression in HSPC results in BMF, independent of TRAF650. The contribution of TIRAP to MDS is supported by the genetic loss of miR-145 in mice as knockout of miR-145 results in phenotypically similar hematologic disease as TIRAP-overexpressing mice. Both mouse models exhibit defects in short-term repopulating HSC and progenitors of the myeloid lineage51.

IL-1 RAP

Another member of the TIR superfamily, interleukin receptor accessory protein (IL1RAP) is evolutionarily conserved among species and required for appropriate host responses to injury and infection. IL1RAP is expressed on cells required for the innate and adaptive immune response, including mast cells, monocytes, macrophages, neutrophils, basophils, NK cells, B cells, T helper type 2 cells (TH2), T helper 17 (TH17), and certain subsets of basal epithelial cells52-55. In humans, alternative splicing of the gene produces soluble (sIL1RAP) and membrane bound (mIL1RAP) isoforms. The membrane-bound isoform contains three extracellular immunoglobulin domains and a TIR domain in the cytoplasmic portion, and it was originally described as a necessary partner of the interleukin 1 receptor 1 (IL1R1)56-59 to initiate IL1 signaling. Binding of IL1α or IL1β to the IL1R1 recruits mIL1RAP, which increases the avidity of the ligand to IL1R1. The juxtaposition of the cytoplasmic TIR domains of mIL1RAP and IL1R1 anchors MYD88, IRAK4, TRAF6 and other signaling intermediates60-62, leading to the induction of NF-κB and MAPK pathways63-66. As a result, the expression of pro-inflammatory genes, IL6, TNFα, Interferon α/β, Tumor Growth Factor, and IL8, is upregulated. Active cytokines are then released from the cell. mIL1RAP interacts with other receptors including the interleukin-1 receptor 2 (IL1R2)67,68, the interleukin 33 receptor (ST2)69-71, the receptor tyrosine kinase KIT72 and the receptor for interleukin 36 family (IL1RL2)73. Through its cytoplasmic domain, mIL1RAP transiently associates with the p85 subunit of phosphatidylinositol 3-kinase (PI3K) upon IL1 stimulation and mediates IL1β-dependent activation of NF-κB p65/RelA subunit via Akt74. In murine cells, Ruhul Amin et al. reported that mIL1RAP complexes with the glycoprotein SIRPα1 to activate Akt and Erk in response to IL1β stimulation75. The soluble form of IL1RAP, lacking the transmembrane and cytoplasmic domains, counteracts IL-1 signaling by binding to the decoy receptor IL1R2 and increasing its affinity for IL1α or IL1β76. Similarly, in mast cells, sIL1RAP inhibits IL33 signaling71.

Dysregulated expression of IL1RAP has been observed in MDS77, AML77,78, and chronic Myeloid leukemia (CML)79-81 as well as during inflammation82-85. In MDS, mRNA and protein levels of mIL1RAP are increased in CD34+ or phenotypically defined HSC from high-risk MDS patients while protein levels in patients with low risk MDS are lower77, suggesting a role in disease progression. In AML patients with monosomy 7 and complex karyotype, IL1RAP is overexpressed in phenotypically defined hematopoietic stem and progenitor cells77,78. AML patients with normal karyotype present variable levels of mRNA expression of IL1RAP in HSC. In this subtype of AML, patients with higher levels of IL1RAP have lower overall survival than patients expressing lower levels of IL1RAP in BM cells77, indicating a prognostic value for IL1RAP. Inhibition of IL1RAP with short hairpin RNAs in AML cell lines and primary high-risk MDS cells leads to reduction of clonogenic capacity and cell death77,86. IL1RAP is also proposed as a stem cell marker in CML, where its expression is restricted to CD34+ cells that express BCR-ABL80. Further studies are warranted to determine the mechanistic role and therapeutic potential of IL1RAP in the pathogenesis of MDS.

IRAK

The IRAK family of proteins consists of four members: IRAK1, IRAK2, IRAK4, and IRAK-M. These proteins are serine/threonine kinases involved in signaling downstream of TLR/IL1-R activation. As mentioned above, upon TLR activation and recruitment of MyD88, MyD88 forms a multi-unit complex with IRAK4 via DD interactions (Figure 2)33. Following its activation, IRAK4 phosphorylates IRAK1 at Threonine-209 followed by Threonine-387, which permits IRAK1 to form a complex with the Ub ligase TRAF6. Lysine 63 ubiquitination of IRAK1 by TRAF6 assembles a scaffold for recruitment of additional downstream proteins87. Ultimately, the IRAK-mediated signal results in the activation of NF-κB and MAPK pathways. In addition to activating IRAK1, phosphorylation of IRAK1 by IRAK4 has been shown to induce degradation of IRAK1, providing a negative feedback mechanism for TLR signaling88. Originally it was thought that both IRAK2 and IRAK-M lacked kinase activity; however recent studies have shown that IRAK2 can act as a kinase and plays a major role in TLR-induced NF-κB activation, particularly from TLR3, TLR4 and TLR889. Additionally, it has been proposed that IRAK-M may have kinase-independent activity regulating alternative NF-κB activation90.

IRAK4 deficiency has been reported in patients that show recurrent infections and poor inflammatory response89. Interestingly, in these patients, the rate of infection decreases with age91, presumably due to intact adaptive memory responses, though further investigation is needed to determine an exact mechanism. IRAK knockout mice for each of the four IRAK family members have been generated. Mice deficient for IRAK1, 2, and 4 exhibit poor response to both viral and bacterial infections, recapitulating the human IRAK4 deficiency syndrome92-95. Alternatively, IRAK-M knockout mice exhibit increased TLR-induced NF-κB and MAPK activity, highlighting the regulatory role of IRAK-M96.

Gene expression profiling of CD34+ BM cells from MDS patients show that IRAK mRNA is overexpressed in 10-30% of patient samples compared with normal CD34+ BM cells12,13,15,97. IRAK1 protein levels correlated with its mRNA overexpression in primary MDS patient BM samples and several MDS/AML cell lines. Not only was IRAK1 protein level elevated, but phosphorylation of IRAK1 at Threonine-209 was also increased. IRAK1 inhibition, either by RNAi-mediated knockdown or through the use of an IRAK1/4 inhibitor, suppressed an MDS-like disease and delayed mortality in a xenograft model utilizing an MDS patient-derived cell line (MDSL)15. Alternatively, IRAK4 knockdown caused a less dramatic reduction of MDSL progenitor function and did not suppress IRAK1 phosphorylation, suggesting that IRAK4 is not as essential to MDS pathogenesis as IRAK197. These results suggest that IRAK inhibition could present a useful therapeutic target in the treatment of MDS. Selective IRAK4 small molecule inhibitors are in development for the treatment of lymphoid-derived hematologic malignancies, particularly those with MyD88 gain-of-function mutations98, and thus present an exciting prospect for treatment of MDS. A comprehensive review of IRAK signaling in hematologic malignancies was recently published, emphasizing the potential importance of IRAK1 signaling and its inhibition in MDS99.

TRAF6

During normal TLR/IL-1R activation, interaction of IRAK family members with TRAF6 is a rate-limiting step for efficient innate immune activation. TRAF6 contains a RING domain, which functions as an Ub ligase by exclusively forming K63 linkages on protein substrate lysines (Figure 2). The RING domain of TRAF6 binds a specific Ub-conjugating enzyme, UBE2N (Ubc13), which catalyzes the synthesis of Ub chains100,101. Importantly, Ubc13 (and its non-enzymatic factor Uev1A) is essential for TRAF6-dependent IKK function101,102. Inhibition of Ubc13 activity with a small molecule inhibitor abolishes TRAF6 Ub ligase function and transfer of Ub103. Following formation of the IRAK1-IRAK4-MyD88 complex, TRAF6 ubiquitinates downstream signaling molecules necessary for of TLRs/IL-1R signaling104. In parallel, TRAF6 induces its polyautoubiquitination and subsequent oligomerization105, which further promotes its interaction with additional molecules, such as NEMO, TAB2/3, TAK1106, and Akt107. One of the key functions of TRAF6 is to activate NF-κB in response to proinflammatory cytokines. IRAK1 undergoes TRAF6-mediated K63 polyubiquitination, which initiates activation of IKK and the subsequent induction of pro-inflammatory cytokines108. In response to stimulation of TLR4, TRAF6 also links innate immune response to autophagy through the ubiquitination of Beclin-1109. While significant progress has been made in identifying pathway-versatile regulatory roles for TRAF6, its full potential in inflammatory disease pathogenesis remains to be discovered.

Given the critical role of TRAF6 in immune signaling, it is not surprising that TRAF6 function and expression is tightly regulated. A major regulatory node for intracellular inflammatory signaling occurs at the TRAF6 mRNA level, via miR-146a. As discussed below in more detail, miR-146a is an immune responsive target that is a feedback inhibitor by directly regulating TRAF6 (and also IRAK1). Moreover the dual ubiquitin-editing enzyme A20 antagonizes TRAF6 autoubiquitination by deubiquitinating the K63 linkages. TRAF6 function is also modulated by its other protein interactors, as a recent report suggests that protein phosphatase 1 (PP1-γ) directly associates with TRAF6, enhances its E3 ligase function, and promotes NF-κB signaling110. Meanwhile, Jiao et al. found that MST4 directly interacts with and phosphorylates TRAF6 to prevent its oligomerization and autoubiquitination, which in turn suppresses its activity111. Taken together, the balance of inhibitory and stimulatory effectors regulating TRAF6 plays a key role in fine-tuning cell-intrinsic response to inflammatory stimuli. Despite our understanding of how TRAF6 protein function is regulated, much less is known about transcriptional control of TRAF6.

While it is generally accepted that inflammation plays a key part in tumorigenesis, many intracellular mechanisms underlying this process remain to be elucidated. Significant progress has been made in ascribing TRAF6 as a key molecule for cell-intrinsic inflammatory signaling and as a result, corroborative data generated from patient samples, in vitro studies, and in vivo mouse model characterizations have appreciably implicated increased TRAF6 signaling in the pathogenesis of MDS. Evidence of increased TRAF6 function was first described by Hofmann et al12. Gene expression profiling of CD34+ BM cells from MDS patients revealed a 10-fold increase in TRAF6 mRNA as compared to normal CD34+ controls. This observation was further supported by derepression of TRAF6 mRNA and protein in del(5q) MDS as a consequence of miR-146a haploinsufficiency49. Conversely, knockdown of TRAF6 in MDS/AML cell lines or patient samples resulted in rapid apoptosis and impaired malignant hematopoietic stem/progenitor function112. Importantly in the context of elevated TRAF6 expression in MDS, overexpression of TRAF6 in human or mouse cells is sufficient to induce its oligomerization, autoubiquitination, and downstream pathway activation. Collectively, TRAF6 overexpression may explain immune pathway overactivation in the MDS-initiating HSPC.

Experimental mouse data supports the hypothesis that increased TRAF6 function has a causal role in the pathogenesis of MDS. Overexpression of TRAF6 in mouse HSPC using a retroviral approach resulted in elevated platelets, neutropenia, dysplasia, and myeloid leukemia in a subset of mice49. Some of the effects are mediated by IL-6 as overexpression of TRAF6 in IL-6 deficient HSPC restored platelets and neutrophil counts. However, the IL-6 deficiency did not delay BMF and AML. Consistent with these observations, Zhao et al. similarly reported a fatal BMF syndrome and associated cytopenias in TRAF6-overexpressing mice. Despite similarities between miR-146a-deficient and TRAF6-transduced HSPC (see comparison below), the level of TRAF6 expression in transduced HSPC is at least 10-fold higher than observed in miR-146a-deficient HSPC. Therefore, to better decipher the contribution of TRAF6 to the miR-146a-deficient HSPC phenotype, enforced expression levels of TRAF6 in transgenic mice need to mimic what is observed in miR-146a-deficient HSPC and in MDS patients. Despite our understanding of TRAF6 Ub-dependent activation of IKK, very few additional TRAF6 substrates have been identified. More importantly, constitutive NF-κB-dependent activation is not sufficient to induce MDS in mice113,114, suggesting additional pathways downstream of TRAF6 contribute to initiation of MDS while NF-κB activation may contribute to the maintenance of the diseased phenotype115. The consequences of TRAF6 activation and relevant substrates have not been evaluated in HSC. Given its potential role in MDS, an extensive investigation is necessary to elucidate the function of TRAF6 in HSC and MDS.

Negative Regulators of Innate Immune Signaling in MDS

miR-146a

miR-146a belongs to a family of microRNAs, which also includes miR-146b. Both miRNAs have an identical seed region and putative mRNA targets. The miR-146 family was initially described as TLR4/NF-κB target genes through a microarray study to identify miRNAs that were regulated upon LPS stimulation in THP1 cells116. Furthermore, a promoter analysis identified two functional and conserved NF-κB binding sites upstream of the miR-146a gene. Follow up studies have confirmed that miR-146a is a rheostat for innate immunity by targeting several components of the TLR signaling pathway, including TLR4, TRAF6, and IRAK1117.

miR-146a lies within chromosome 5q33.3 and its deletion occurs in 80% of all del(5q) MDS patients. Consistent with deletion of a single miR-146a allele, expression in BM HSPC from del(5q) MDS patients is reduced by greater than half49,118. miR-146a is also downregulated in non-del(5q) MDS patients suggesting that other mechanisms may contribute to its reduced expression in MDS119,120. In this non-del(5q) MDS patient cohort, miR-146a is part of a miRNA diagnostic signature that distinguishes MDS patients from age-matched controls119. Although miR-146a is consistently downregulated in del(5q) MDS, additional miRNAs residing on chr 5q (i.e., miR-145) may also contribute to aspects of MDS pathogenesis. Concomitant knockdown of miR-145 and miR-146a using a miRNA decoy approach in mouse HSPC resulted in elevated platelets, neutropenia, megakaryocytic dysplasia, and myeloid leukemia. The distinction between miR-145 and miR-146a’s contribution to the hematopoietic defects has been revealed by examination of the miR-146a-deficient mice.

In normal mouse and human hematopoietic cells, miR-146a expression is not restricted to particular hematopoietic lineages. Evidence suggests a role for miR-146a throughout development of blood and lymphoid cells due to a significant increase in its expression as HSCs mature. A germ-line knock out mouse model of miR-146a suggests minimal requirement for miR-146a during steady state hematopoiesis. Conversely, a model of enforced miR-146a expression in mouse HSPC has marginal effects on hematopoiesis. However, miR-146a is essential during stressed and age-associated hematopoiesis. Knockout of miR-146a results in an early onset of myeloid expansion in the BM, immune dysfunction, and progression to more aggressive diseases such as lymphomas, BMF, and myeloid leukemia121-123. Furthermore, miR-146a-deficient HSPC produce higher levels of pro-inflammatory cytokines, such as IL-6, TNFα, GM-CSF and IL-1β, in response to TLR activation. Additional concepts and description of miR-146a-deficient mice pertaining to the pathogenesis of MDS has been recently summarized117.

TNFAIP3

TNFAIP3(A20) regulates TLR/NF-κB signaling through its effects on TRAF6. A20 is an anti-inflammatory signaling molecule that regulates several intracellular signaling cascades by modifying Ub chains on upstream adaptor proteins. Under normal cellular conditions, A20 is expressed at low levels. However, stimulation of PAMPs and pro-inflammatory cytokines acutely induce the expression of A20 in an NF-κB-dependent manner124. In response to several immune pathways including TNF receptor, IL-1R, and TLR signaling, A20 negatively regulates excessive NF-κB signaling by modifying Ub chains on the key intermediates TRAF6 and Receptor Interacting Protein (RIP1)125.

The most striking data to demonstrate the involvement of A20 in the innate immune system is that A20-deficient mice prematurely die of severe multiorgan inflammation126. A20−/− RAG1−/− mice, which lack T and B lymphocytes, present with a similar phenotype, suggesting A20 regulates innate immune homeostasis independent of the adaptive immune system127. Given the limitation of the perinatal lethality of A20-deficient mice, conditionally targeted mice have been generated for lineage specific deletions of A20, which uniquely mimic human autoimmune and inflammatory diseases that are associated with single nucleotide polymorphisms (SNPs) in the A20 genomic region125,128-133. Cell-specific deletions highlight A20’s role in maintaining tissue homeostasis and the prevention of inflammatory diseases. Mice with hematopoietic-specific deletions of A20 exhibited severe cytokine-mediated inflammation leading to premature death134,135. Myeloid cells from these mice proliferated excessively and B cells underwent apoptosis accompanied by anemia and lymphopenia134. Constitutive NF-κB activity was observed in A20-deficient HSCs, leading to increased cell cycle entry. Loss of A20 in HSCs diminishes quiescence and depletes the HSC pool135. Furthermore, transplants of these cells into recipient mice revealed impaired long-term HSC function134.

A20 is a central regulator of multiple signaling cascades activating NF-κB. As an NF-κB target gene, A20 negatively feeds back to dampen NF-κB signaling by several mechanisms: removal of polyubiquitin chains that participate in signaling activation, and attachment of polyubiquitin chains that facilitate the degradation of proteins involved in signaling complexes. Its deubiquitinase activity promotes the depolymerization of K63-linked polyubiquitin chains on RIP1. Subsequently, its E3 ligase activity covalently attaches K48-linked polyubiquitin chains on RIP1, which results in proteasomal degradation of RIP1, thereby regulating TNFα-induced NF-κB signaling136. K63-linked polyubiquitin chains are also deubiquitinated on TRAF6, while K48-linked polyubiquitin chains are ligated on the E2 enzyme Ubc13, resulting in the degradation of Ubc13 and the disruption of the E2 and E3 ligase interaction, and consequently terminating TLR4/ IL-1R1 signaling to NF-κB136.

The dual ubiquitin-editing functions of A20 are accomplished by two functional domains. The deubiquitination activity is mediated by the catalytic cysteine in the N-terminal ovarian tumor (OTU) domain while the C-terminal zinc fingers 4 and 7 mediate E3 ligase activity. Several studies have investigated the specific function of each domain in the molecular and cellular context. In Tnfaip3OTU/OTU cells, which are mutant for A20 DUB function, RIP1 immunoprecipitation experiments showed increased polyubiquitin chains, suggesting that the OTU domain is responsible for the removal of K48 and K63-linked polyubiquitin chains from RIP1137. Bosanac et al. found that A20 zinc finger 4 appears to be necessary for the modification of NF-κB signaling138. Zinc finger 4 recognizes K63-linked Ub chain and exhibits binding capabilities. By overexpressing zinc finger A20 mutants, Tokunaga and colleagues determined that zinc finger 7 regulates NF-κB activity through the recognition of linear polyubiquitin chains and also weakly interacts with K63-linked polyubiquitin chain139. Therefore, there is evidence that both the DUB activity in the OTU domain and E3 ligase activity in zinc fingers 4 and 7 participate in the modulation of Ub chains to control intracellular immune signaling cascades that limit excessive inflammation.

Expression of A20 mRNA is extremely high in lymphoid organs and regulation of A20 in lymphocytes differs from other cell types140. A20 has recently been identified as a recurrently mutated gene in lymphoid leukemias141. Since dysregulation of the NF-κB signaling pathway has been found to be involved in leukemogenesis and deletions and mutations of A20 have been observed in lymphomas, A20 is considered a tumor suppressor142. Certain autoimmune and inflammatory diseases linked with SNPs in A20 are associated with increased risk of lymphomas143.

Increasing evidence also suggests a connection between A20 expression and MDS pathogenesis. Consistent with the hypothesis that reduced A20 expression and increased innate immune function is associated with MDS144, empirical data revealed A20 mRNA is significantly reduced in CD34+ cells isolated from the BM of MDS patients145. Moreover, miR-125a, which targets A20, is overexpressed in MDS CD34+ cells16,146. As mentioned above, population-based study revealed that autoimmune disorders are a potential trigger for MDS development14. Interestingly, the MDS-associated autoimmune disorders, such as rheumatoid arthritis, systemic lupus erythematosus, and Sjogren’s syndrome are also genetically linked to A20 SNPs and observed in A20-deficient mice. Taken together, this suggests an impelling case for A20 as a relevant immune-related gene in MDS.

Conclusions

In summary, chronic innate immune signaling and activation is widely reported in MDS. The consequences of increased TLR signaling in MDS can result in systemic effects on hematopoiesis, such as due to inflammation and abnormal adaptive and innate immunity on BM HSPC. In addition to these non-cell autonomous consequences of chronic innate immune activation, increased TLR signaling within HSPC from MDS is also widely reported, and thought to contribute to cell-intrinsic defects in HSC. Although mounting evidence implicates chronic innate immune signaling in the pathogenesis of MDS, there are many unresolved questions. How best to measure chronic innate immune signaling and activation in MDS? In addition to the evidence above, how else is the innate immune pathway activated in MDS, and in which hematopoietic cell lineage? Which MDS subtypes exhibit activation of innate immune signaling? Lastly, it is prudent to distinguish between therapies targeting the systemic effects of chronic innate immunity versus cell-intrinsic TLR signaling in MDS, as these treatments will likely have distinct consequences on disease outcome.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Tehranchi R, et al. Persistent malignant stem cells in del(5q) myelodysplasia in remission. N. Engl. J. Med. 2010;363:1025–1037. doi: 10.1056/NEJMoa0912228. [DOI] [PubMed] [Google Scholar]
  • 2.Will B, et al. Stem and progenitor cells in myelodysplastic syndromes show aberrant stage-specific expansion and harbor genetic and epigenetic alterations. Blood. 2012;120:2076–2086. doi: 10.1182/blood-2011-12-399683. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Woll PS, et al. Myelodysplastic syndromes are propagated by rare and distinct human cancer stem cells in vivo. Cancer Cell. 2014;25:794–808. doi: 10.1016/j.ccr.2014.03.036. [DOI] [PubMed] [Google Scholar]
  • 4.Nilsson L, et al. Isolation and characterization of hematopoietic progenitor/stem cells in 5q-deleted myelodysplastic syndromes: evidence for involvement at the hematopoietic stem cell level. Blood. 2000;96:2012–2021. [PubMed] [Google Scholar]
  • 5.Nilsson L, et al. The molecular signature of MDS stem cells supports a stem-cell origin of 5q-myelodysplastic syndromes. Blood. 2007;110:3005–3014. doi: 10.1182/blood-2007-03-079368. [DOI] [PubMed] [Google Scholar]
  • 6.Nilsson L, et al. Involvement and functional impairment of the CD34+CD38-Thy-1+ hematopoietic stem cell pool in myelodysplastic syndromes with trisomy 8. Blood. 2002;100:259–267. doi: 10.1182/blood-2001-12-0188. [DOI] [PubMed] [Google Scholar]
  • 7.Corey SJ, et al. Myelodysplastic syndromes: the complexity of stem-cell diseases. Nat. Rev. Cancer. 2007;7:118–129. doi: 10.1038/nrc2047. [DOI] [PubMed] [Google Scholar]
  • 8.Nimer SD. Myelodysplastic syndromes. Blood. 2008;111:4841–4851. doi: 10.1182/blood-2007-08-078139. [DOI] [PubMed] [Google Scholar]
  • 9.Greenberg P, et al. International scoring system for evaluating prognosis in myelodysplastic syndromes. Blood. 1997;89:2079–2088. [PubMed] [Google Scholar]
  • 10.Walter MJ, et al. Clonal architecture of secondary acute myeloid leukemia. N Engl J Med. 2012;366:1090–1098. doi: 10.1056/NEJMoa1106968. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Cazzola M, Malcovati L. Prognostic classification and risk assessment in myelodysplastic syndromes. Hematology/Oncology Clinics of North America. 2010;24:459–468. doi: 10.1016/j.hoc.2010.02.005. [DOI] [PubMed] [Google Scholar]
  • 12.Hofmann W-K, et al. Characterization of gene expression of CD34+ cells from normal and myelodysplastic bone marrow. Blood. 2002;100:3553–3560. doi: 10.1182/blood.V100.10.3553. [DOI] [PubMed] [Google Scholar]
  • 13.Pellagatti A, et al. Deregulated gene expression pathways in myelodysplastic syndrome hematopoietic stem cells. Leuk. Off. J. Leuk. Soc. Am. Leuk. Res. Fund, U.K. 2010;24:756–764. doi: 10.1038/leu.2010.31. [DOI] [PubMed] [Google Scholar]
  • 14.Kristinsson SY, et al. Chronic immune stimulation might act as a trigger for the development of acute myeloid leukemia or myelodysplastic syndromes. J. Clin. Oncol. 2011;29:2897–2903. doi: 10.1200/JCO.2011.34.8540. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Rhyasen GW, et al. Targeting IRAK1 as a Therapeutic Approach for Myelodysplastic Syndrome. Cancer Cell. 2013;24:90–104. doi: 10.1016/j.ccr.2013.05.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Gañán-Gómez I, et al. Overexpression of miR-125a in myelodysplastic syndrome CD34+ cells modulates NF-κB activation and enhances erythroid differentiation arrest. PLoS One. 2014;9 doi: 10.1371/journal.pone.0093404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Yang L, Qian Y, Eksioglu E, Epling-Burnette P, Wei S. The inflammatory microenvironment in MDS. Cell Mol Life Sci. 2015;72:1959–66. doi: 10.1007/s00018-015-1846-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Akira S, Takeda K. Toll-like receptor signalling. Nat. Rev. Immunol. 2004;4:499–511. doi: 10.1038/nri1391. [DOI] [PubMed] [Google Scholar]
  • 19.Kawai T, Akira S. The role of pattern-recognition receptors in innate immunity: update on Toll-like receptors. Nat. Immunol. 2010;11:373–384. doi: 10.1038/ni.1863. [DOI] [PubMed] [Google Scholar]
  • 20.Moresco EMY, LaVine D, Beutler B. Toll-like receptors. Curr. Biol. 2011;21:R488–R493. doi: 10.1016/j.cub.2011.05.039. [DOI] [PubMed] [Google Scholar]
  • 21.Boiko JR, Borghesi L. Hematopoiesis sculpted by pathogens: Toll-like receptors and inflammatory mediators directly activate stem cells. Cytokine. 2012;57:1–8. doi: 10.1016/j.cyto.2011.10.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Sioud M, Fløisand Y. TLR agonists induce the differentiation of human bone marrow CD34+ progenitors into CD11c+ CD80/86+ DC capable of inducing a Th1-type response. Eur. J. Immunol. 2007;37:2834–2846. doi: 10.1002/eji.200737112. [DOI] [PubMed] [Google Scholar]
  • 23.Takizawa H, Regoes RR, Boddupalli CS, Bonhoeffer S, Manz MG. Dynamic variation in cycling of hematopoietic stem cells in steady state and inflammation. J. Exp. Med. 2011;208:273–284. doi: 10.1084/jem.20101643. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Esplin BL, et al. Chronic exposure to a TLR ligand injures hematopoietic stem cells. J. Immunol. 2011;186:5367–5375. doi: 10.4049/jimmunol.1003438. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Zhao Y, Ling F, Wang HC, Sun XH. Chronic TLR Signaling Impairs the Long-Term Repopulating Potential of Hematopoietic Stem Cells of Wild Type but Not Id1 Deficient Mice. PLoS One. 2013;8 doi: 10.1371/journal.pone.0055552. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Maratheftis CI, Andreakos E, Moutsopoulos HM, Voulgarelis M. Toll-like receptor-4 is up-regulated in hematopoietic progenitor cells and contributes to increased apoptosis in myelodysplastic syndromes. Clin. Cancer Res. 2007;13:1154–1160. doi: 10.1158/1078-0432.CCR-06-2108. [DOI] [PubMed] [Google Scholar]
  • 27.Kuninaka N, et al. Expression of Toll-like receptor 9 in bone marrow cells of myelodysplastic syndromes is down-regulated during transformation to overt leukemia. Exp. Mol. Pathol. 2010;88:293–298. doi: 10.1016/j.yexmp.2010.01.009. [DOI] [PubMed] [Google Scholar]
  • 28.Wei Y, et al. Toll-like receptor alterations in myelodysplastic syndrome. Leukemia. 2013;27:1832–40. doi: 10.1038/leu.2013.180. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Nishiya T, Kajita E, Horinouchi T, Nishimoto A, Miwa S. Distinct roles of TIR and non-TIR regions in the subcellular localization and signaling properties of MyD88. FEBS Lett. 2007;581:3223–3229. doi: 10.1016/j.febslet.2007.06.008. [DOI] [PubMed] [Google Scholar]
  • 30.Wesche H, Henzel WJ, Shillinglaw W, Li S, Cao Z. MyD88: An adapter that recruits IRAK to the IL-1 receptor complex. Immunity. 1997;7:837–847. doi: 10.1016/s1074-7613(00)80402-1. [DOI] [PubMed] [Google Scholar]
  • 31.Loiarro M, et al. Mutational analysis identifies residues crucial for homodimerization of myeloid differentiation factor 88 (MyD88) and for its function in immune cells. J. Biol. Chem. 2013;288:30210–30222. doi: 10.1074/jbc.M113.490946. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Lin S-C, Lo Y-C, Wu H. Helical assembly in the MyD88-IRAK4-IRAK2 complex in TLR/IL-1R signalling. Nature. 2010;465:885–890. doi: 10.1038/nature09121. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Motshwene PG, et al. An Oligomeric Signaling Platform formed by the toll-like receptor signal transducers MyD88 and IRAK-4. J. Biol. Chem. 2009;284:25404–25411. doi: 10.1074/jbc.M109.022392. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Häcker H, et al. Specificity in Toll-like receptor signalling through distinct effector functions of TRAF3 and TRAF6. Nature. 2006;439:204–207. doi: 10.1038/nature04369. [DOI] [PubMed] [Google Scholar]
  • 35.Xia Z-P, et al. Direct activation of protein kinases by unanchored polyubiquitin chains. Nature. 2009;461:114–119. doi: 10.1038/nature08247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Hou B, Reizis B, DeFranco AL. Toll-like receptor-mediated dendritic cell-dependent and -independent stimulation of innate and adaptive immunity. 2008;29:272–282. doi: 10.1016/j.immuni.2008.05.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Xu L, et al. MYD88 L265P in Waldenstrom’s Macroglobulinemia, IgM Monoclonal Gammopathy, and other B-cell Lymphoproliferative Disorders using Conventional and Quantitative Allele-Specific PCR. Blood. 2013:2051–2058. doi: 10.1182/blood-2012-09-454355. doi:10.1182/blood-2012-09-454355. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Zagaria A, et al. Myelodysplastic syndrome with 5q deletion following IgM monoclonal gammopathy, showing gene mutation MYD88 L265P. Blood Cells, Molecules, and Diseases. 2015;54:51–52. doi: 10.1016/j.bcmd.2014.07.022. [DOI] [PubMed] [Google Scholar]
  • 39.Avbelj M, et al. Activation of lymphoma-associated MyD88 mutations via allostery-induced TIR-domain oligomerization. Blood. 2014;124:3896–904. doi: 10.1182/blood-2014-05-573188. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Dimicoli S, et al. Overexpression of the Toll-Like Receptor (TLR) Signaling Adaptor MYD88, but Lack of Genetic Mutation, in Myelodysplastic Syndromes. PLoS One. 2013;8 doi: 10.1371/journal.pone.0071120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Schinke C, et al. IL-8-CXCR2 pathway inhibition as a therapeutic strategy against MDS and AML stem cells. Blood. 2015;125:3144–52. doi: 10.1182/blood-2015-01-621631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Horng T, Barton GM, Flavell RA, Medzhitov R. The adaptor molecule TIRAP provides signalling specificity for Toll-like receptors. Nature. 2002;420:329–333. doi: 10.1038/nature01180. [DOI] [PubMed] [Google Scholar]
  • 43.Fitzgerald KA, et al. Mal (MyD88-adapter-like) is required for Toll-like receptor-4 signal transduction. Nature. 2001;413:78–83. doi: 10.1038/35092578. [DOI] [PubMed] [Google Scholar]
  • 44.Horng T, Barton GM, Medzhitov R. TIRAP: an adapter molecule in the Toll signaling pathway. Nat. Immunol. 2001;2:835–841. doi: 10.1038/ni0901-835. [DOI] [PubMed] [Google Scholar]
  • 45.Bonham KS, et al. A promiscuous lipid-binding protein diversifies the subcellular sites of toll-like receptor signal transduction. Cell. 2014;156:705–716. doi: 10.1016/j.cell.2014.01.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Equils O, et al. Rac1 and Toll-IL-1 receptor domain-containing adapter protein mediate Toll-like receptor 4 induction of HIV-long terminal repeat. J. Immunol. 2004;172:7642–7646. doi: 10.4049/jimmunol.172.12.7642. [DOI] [PubMed] [Google Scholar]
  • 47.Verstak B, et al. MyD88 adapter-like (Mal)/TIRAP interaction with TRAF6 is critical for TLR2- and TLR4-mediated NF-κB proinflammatory responses. J. Biol. Chem. 2009;284:24192–24203. doi: 10.1074/jbc.M109.023044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Mellett M, Atzei P, Jackson R, O’Neill LA, Moynagh PN. Mal mediates TLR-induced activation of CREB and expression of IL-10. J. Immunol. 2011;186:4925–4935. doi: 10.4049/jimmunol.1002739. [DOI] [PubMed] [Google Scholar]
  • 49.Starczynowski DT, et al. Identification of miR-145 and miR-146a as mediators of the 5q-syndrome phenotype. Nat. Med. 2010;16:49–58. doi: 10.1038/nm.2054. [DOI] [PubMed] [Google Scholar]
  • 50.Ibrahim R, et al. Activation of Non-canonical Immune Sigaling Pathways Drives Bone Marrow Failure in Murine Model of MDS; 56th Am. Soc. Hematol. Annu. Meet.; Session. 2014. [Google Scholar]
  • 51.Lam J, et al. Loss of MiR-143 and MiR-145 Inhibits Hematopoietic Stem Cell Self-Renewal through Dysregulated TGFβ Signaling; 56th American Society of Hematology Annual Meeting; 2014. [Google Scholar]
  • 52.Dinarello CH. Biologic basis for interleukin-1 in disease. Blood. 1996;87:2095–2147. [PubMed] [Google Scholar]
  • 53.Dinarello CA. Immunological and inflammatory functions of the interleukin-1 family. Annu. Rev. Immunol. 2009;27:519–550. doi: 10.1146/annurev.immunol.021908.132612. [DOI] [PubMed] [Google Scholar]
  • 54.Smith DE. IL-33: A tissue derived cytokine pathway involved in allergic inflammation and asthma. Clinical and Experimental Allergy. 2010;40:200–208. doi: 10.1111/j.1365-2222.2009.03384.x. [DOI] [PubMed] [Google Scholar]
  • 55.Gabay C, McInnes IB. The biological and clinical importance of the ‘new generation’ cytokines in rheumatic diseases. Arthritis Res. Ther. 2009;11:230. doi: 10.1186/ar2680. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Cullinan EB, et al. IL-1 receptor accessory protein is an essential component of the IL-1 receptor. J. Immunol. 1998;161:5614–5620. [PubMed] [Google Scholar]
  • 57.Greenfeder SA, et al. Molecular cloning and characterization of a second subunit of the interleukin 1 receptor complex. J. Biol. Chem. 1995;270:13757–13765. doi: 10.1074/jbc.270.23.13757. [DOI] [PubMed] [Google Scholar]
  • 58.Hofmeister R, et al. Activation of acid sphingomyelinase by interleukin-1 (IL-1) requires the IL-1 receptor accessory protein. Journal of Biological Chemistry. 1997;272:27730–27736. doi: 10.1074/jbc.272.44.27730. [DOI] [PubMed] [Google Scholar]
  • 59.Korherr C, Hofmeister R, Wesche H, Falk W. A critical role for interleukin-1 receptor accessory protein in interleukin-1 signaling. Eur. J. Immunol. 1997;27:262–267. doi: 10.1002/eji.1830270139. [DOI] [PubMed] [Google Scholar]
  • 60.Huang J, Gao X, Li S, Cao Z. Recruitment of IRAK to the interleukin 1 receptor complex requires interleukin 1 receptor accessory protein. Proc. Natl. Acad. Sci. U. S. A. 1997;94:12829–12832. doi: 10.1073/pnas.94.24.12829. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Jefferies C, et al. Transactivation by the p65 subunit of NF-kappaB in response to interleukin-1 (IL-1) involves MyD88, IL-1 receptor-associated kinase 1, TRAF-6, and Rac1. Molecular and cellular biology. 2001;21:4544–4552. doi: 10.1128/MCB.21.14.4544-4552.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Brikos C, Wait R, Begum S, O’Neill LAJ, Saklatvala J. Mass spectrometric analysis of the endogenous type I interleukin-1 (IL-1) receptor signaling complex formed after IL-1 binding identifies IL-1RAcP, MyD88, and IRAK-4 as the stable components. Mol. Cell. Proteomics. 2007;6:1551–1559. doi: 10.1074/mcp.M600455-MCP200. [DOI] [PubMed] [Google Scholar]
  • 63.Barnes PJ, Karin M. Nuclear factor-kappa B--a pivotal transcription factor in chronic inflammatory diseases. N. Engl. J. Med. 1997;336:1066–1071. doi: 10.1056/NEJM199704103361506. [DOI] [PubMed] [Google Scholar]
  • 64.O’Neill L. a J. Towards an understanding of the signal transduction pathways for interleukin 1. BiochimBiophysActa. 1995;1266:31–44. doi: 10.1016/0167-4889(94)00217-3. [DOI] [PubMed] [Google Scholar]
  • 65.O’Neill L. a. Molecular mechanisms underlying the actions of the pro-inflammatory cytokine interleukin 1. Royal Irish Academy Medal Lecture. Biochemical Society transactions. 1997;25:295–302. doi: 10.1042/bst0250295. [DOI] [PubMed] [Google Scholar]
  • 66.O’Neill L. a J. The interleukin-1 receptor/Toll-like receptor superfamily: 10 Years of progress. Immunological Reviews. 2008;226:10–18. doi: 10.1111/j.1600-065X.2008.00701.x. [DOI] [PubMed] [Google Scholar]
  • 67.Lang D, et al. The type II IL-1 receptor interacts with the IL-1 receptor accessory protein: a novel mechanism of regulation of IL-1 responsiveness. Journal of immunology (Baltimore, Md. : 1950) 1998;161:6871–6877. [PubMed] [Google Scholar]
  • 68.Malinowsky D, Lundkvist J, Layé S, Bartfai T. Interleukin-1 receptor accessory protein interacts with the type II interleukin-1 receptor. FEBS Lett. 1998;429:299–302. doi: 10.1016/s0014-5793(98)00467-0. [DOI] [PubMed] [Google Scholar]
  • 69.Ali S, et al. IL-1 receptor accessory protein is essential for IL-33-induced activation of T lymphocytes and mast cells. Proc. Natl. Acad. Sci. U. S. A. 2007;104:18660–18665. doi: 10.1073/pnas.0705939104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Chackerian AA, et al. IL-1 receptor accessory protein and ST2 comprise the IL-33 receptor complex. J. Immunol. 2007;179:2551–2555. doi: 10.4049/jimmunol.179.4.2551. [DOI] [PubMed] [Google Scholar]
  • 71.Palmer G, et al. The IL-1 receptor accessory protein (AcP) is required for IL-33 signaling and soluble AcP enhances the ability of soluble ST2 to inhibit IL-33. Cytokine. 2008;42:358–364. doi: 10.1016/j.cyto.2008.03.008. [DOI] [PubMed] [Google Scholar]
  • 72.Drube S, et al. The receptor tyrosine kinase c-Kit controls IL-33 receptor signaling in mast cells. Blood. 2010;115:3899–3906. doi: 10.1182/blood-2009-10-247411. [DOI] [PubMed] [Google Scholar]
  • 73.Towne JE, Garka KE, Renshaw BR, Virca GD, Sims JE. Interleukin (IL)-1F6, IL-1F8, and IL-1F9 Signal Through IL-1Rrp2 and IL-1RAcP to Activate the Pathway Leading to NF-kB and MAPKs. Journal of Biological Chemistry. 2004;279:13677–13688. doi: 10.1074/jbc.M400117200. [DOI] [PubMed] [Google Scholar]
  • 74.Sizemore N, Leung S, Stark GR. Activation of phosphatidylinositol 3-kinase in response to interleukin-1 leads to phosphorylation and activation of the NF-kappaB p65/RelA subunit. Molecular and cellular biology. 1999;19:4798–4805. doi: 10.1128/mcb.19.7.4798. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Ruhul Amin ARM, et al. A role for SHPS-1/SIRPalpha1 in IL-1beta- and TNFalpha-dependent signaling. Oncogene. 2002;21:8871–8878. doi: 10.1038/sj.onc.1206018. [DOI] [PubMed] [Google Scholar]
  • 76.Smith DE, et al. The Soluble Form of IL-1 Receptor Accessory Protein Enhances the Ability of Soluble Type II IL-1 Receptor to Inhibit IL-1 Action. Immunity. 2003;18:87–96. doi: 10.1016/s1074-7613(02)00514-9. [DOI] [PubMed] [Google Scholar]
  • 77.Barreyro L, et al. Overexpression of IL-1 receptor accessory protein in stem and progenitor cells and outcome correlation in AML and MDS. Blood. 2012;120:1290–1298. doi: 10.1182/blood-2012-01-404699. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Askmyr M, et al. Selective killing of candidate AML stem cells by antibody targeting of IL1RAP. Blood. 2013;121:3709–3713. doi: 10.1182/blood-2012-09-458935. [DOI] [PubMed] [Google Scholar]
  • 79.Gerber JM, et al. Genome-wide comparison of the transcriptomes of highly enriched normal and chronic myeloid leukemia stem and progenitor cell populations. Oncotarget. 2013;4:715–28. doi: 10.18632/oncotarget.990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Järås M, et al. Isolation and killing of candidate chronic myeloid leukemia stem cells by antibody targeting of IL-1 receptor accessory protein. Proc. Natl. Acad. Sci. U. S. A. 2010;107:16280–16285. doi: 10.1073/pnas.1004408107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Zhao K, et al. IL1RAP as a surface marker for leukemia stem cells is related to clinical phase of chronic myeloid leukemia patients. Int J Clin Exp Med. 2014;7:4787–98. [PMC free article] [PubMed] [Google Scholar]
  • 82.Nishida A, et al. Expression of interleukin 1-like cytokine interleukin 33 and its receptor complex (ST2L and IL1RAcP) in human pancreatic myofibroblasts. Gut. 2010;59:531–541. doi: 10.1136/gut.2009.193599. [DOI] [PubMed] [Google Scholar]
  • 83.Michaud N, Al-Akoum M, Akoum A. Blood soluble interleukin 1 receptor accessory protein levels are consistently low throughout the menstrual cycle of women with endometriosis. Reprod. Biol. Endocrinol. 2014;12:51. doi: 10.1186/1477-7827-12-51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Bozaoglu K, et al. Plasma levels of soluble Interleukin 1 Receptor Accessory Protein are Reduced in Obesity. J. Clin. Endocrinol. Metab. 2014 doi: 10.1210/jc.2013-4475. jc20134475. doi:10.1210/jc.2013-4475. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Smallridge RC, et al. RNA sequencing identifies multiple fusion transcripts, differentially expressed genes, and reduced expression of immune function genes in BRAF (V600E) mutant vs BRAF wild-type papillary thyroid carcinoma. J. Clin. Endocrinol. Metab. 2014;99 doi: 10.1210/jc.2013-2792. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Barreyro L, et al. Abstarct C225: IL1RAP as functionally relevant target for stem-cell directed therapy in acute myeloid leukemia (AML) and myelodysplastic syndromes (MDS) Mol. Cancer Ther. 2013;12 [Google Scholar]
  • 87.Windheim M, Stafford M, Peggie M, Cohen P. Interleukin-1 (IL-1) induces the Lys63-linked polyubiquitination of IL-1 receptor-associated kinase 1 to facilitate NEMO binding and the activation of IkappaBalpha kinase. Mol. Cell. Biol. 2008;28:1783–1791. doi: 10.1128/MCB.02380-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Kubo-Murai M, et al. IRAK-4-dependent degradation of IRAK-1 is a negative feedback signal for TLR-mediated NF-KB activation. Journal of Biochemistry. 2008;143:295–302. doi: 10.1093/jb/mvm234. [DOI] [PubMed] [Google Scholar]
  • 89.Keating SE, Maloney GM, Moran EM, Bowie AG. IRAK-2 participates in multiple Toll-like receptor signaling pathways to NF??B via activation of TRAF6 ubiquitination. J. Biol. Chem. 2007;282:33435–33443. doi: 10.1074/jbc.M705266200. [DOI] [PubMed] [Google Scholar]
  • 90.Su J, Zhang T, Tyson J, Li L. The interleukin-1 receptor-associated kinase M selectively inhibits the alternative, instead of the classical NF??B pathway. J. Innate Immun. 2009;1:164–174. doi: 10.1159/000158541. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Picard C, et al. Clinical Features and Outcome of Patients With IRAK-4 and MyD88 Deficiency. Pediatrics. 2010;89:403–25. doi: 10.1097/MD.0b013e3181fd8ec3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Thomas JA, et al. Impaired cytokine signaling in mice lacking the IL-1 receptor-associated kinase. J. Immunol. 1999;163:978–984. [PubMed] [Google Scholar]
  • 93.Kawagoe T, et al. Sequential control of Toll-like receptor-dependent responses by IRAK1 and IRAK2. Nat. Immunol. 2008;9:684–691. doi: 10.1038/ni.1606. [DOI] [PubMed] [Google Scholar]
  • 94.Suzuki N, et al. Severe impairment of interleukin-1 and Toll-like receptor signalling in mice lacking IRAK-4. Nature. 2002;416:750–756. doi: 10.1038/nature736. [DOI] [PubMed] [Google Scholar]
  • 95.Meylan E, Tschopp J. IRAK2 takes its place in TLR signaling. Nature immunology. 2008;9:581–582. doi: 10.1038/ni0608-581. [DOI] [PubMed] [Google Scholar]
  • 96.Seki M, et al. Critical role of IL-1 receptor-associated kinase-M in regulating chemokine-dependent deleterious inflammation in murine influenza pneumonia. Journal of immunology (Baltimore, Md. : 1950) 2010;184:1410–1418. doi: 10.4049/jimmunol.0901709. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Rhyasen GW, Bolanos L, Starczynowski DT. Differential IRAK signaling in hematologic malignancies. Experimental Hematology. 2013;41:1005–1007. doi: 10.1016/j.exphem.2013.09.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Chaudhary D, Robinson S, Romero D. Recent Advances in the Discovery of Small Molecule Inhibitors of Interleukin 1 Receptor-Associated Kinase 4 (IRAK4) as a Therapeutic Target for In flammation and Oncology Disorders. J. Med. Chem. 2015;58:96–110. doi: 10.1021/jm5016044. [DOI] [PubMed] [Google Scholar]
  • 99.Rhyasen G, Starczynowski DT. IRAK signalling in cancer. Br J Cancer. 2015;112:232–7. doi: 10.1038/bjc.2014.513. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Hofmann RM, Pickart CM. Noncanonical MMS2-encoded ubiquitin-conjugating enzyme functions in assembly of novel polyubiquitin chains for DNA repair. Cell. 1999;96:645–653. doi: 10.1016/s0092-8674(00)80575-9. [DOI] [PubMed] [Google Scholar]
  • 101.Deng L, et al. Activation of the IkappaB kinase complex by TRAF6 requires a dimeric ubiquitin-conjugating enzyme complex and a unique polyubiquitin chain. Cell. 2000;103:351–361. doi: 10.1016/s0092-8674(00)00126-4. [DOI] [PubMed] [Google Scholar]
  • 102.Fukushima T, et al. Ubiquitin-conjugating enzyme Ubc13 is a critical component of TNF receptor-associated factor (TRAF)-mediated inflammatory responses. Proc. Natl. Acad. Sci. U. S. A. 2007;104:6371–6376. doi: 10.1073/pnas.0700548104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Pulvino M, et al. Inhibition of proliferation and survival of diffuse large B-cell lymphoma cells by a small-molecule inhibitor of the ubiquitin-conjugating enzyme Ubc13-Uev1A. Blood. 2012;120:1668–1677. doi: 10.1182/blood-2012-02-406074. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Zhang X, Zhang J, Zhang L, van Dam H, ten Dijke P. UBE2O negatively regulates TRAF6-mediated NF-κB activation by inhibiting TRAF6 polyubiquitination. Cell Res. 2013;23:366–77. doi: 10.1038/cr.2013.21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Muroi M, Tanamoto K. TRAF6 distinctively mediates MyD88- and IRAK-1-induced activation of NF-kappaB. Journal of leukocyte biology. 2008;83:702–707. doi: 10.1189/jlb.0907629. [DOI] [PubMed] [Google Scholar]
  • 106.Wang C, et al. TAK1 is a ubiquitin-dependent kinase of MKK and IKK. Nature. 2001;412:346–351. doi: 10.1038/35085597. [DOI] [PubMed] [Google Scholar]
  • 107.Yang W-L, et al. The E3 ligase TRAF6 regulates Akt ubiquitination and activation. Science. 2009;325:1134–1138. doi: 10.1126/science.1175065. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Conze DB, Wu C-J, Thomas JA, Landstrom A, Ashwell JD. Lys63-linked polyubiquitination of IRAK-1 is required for interleukin-1 receptor- and toll-like receptor-mediated NF-kappaB activation. Mol. Cell. Biol. 2008;28:3538–3547. doi: 10.1128/MCB.02098-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Shi C-S, Kehrl JH. TRAF6 and A20 regulate lysine 63-linked ubiquitination of Beclin-1 to control TLR4-induced autophagy. Science signaling. 2010;3:ra42. doi: 10.1126/scisignal.2000751. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Opaluch AM, et al. Positive regulation of TRAF6-dependent innate immune responses by protein phosphatase PP1-γ. PLoS One. 2014;9 doi: 10.1371/journal.pone.0089284. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Shi J, et al. The kinase MST4 limits inflammatory responses through direct phosphorylation of the adaptor TRAF6. Nat. Immunol. 2015;16:246–257. doi: 10.1038/ni.3097. [DOI] [PubMed] [Google Scholar]
  • 112.Fang J, et al. Cytotoxic effects of bortezomib in myelodysplastic syndrome/acute myeloid leukemia depend on autophagy-mediated lysosomal degradation of TRAF6 and repression of PSMA 1. Blood. 2012;120:858–867. doi: 10.1182/blood-2012-02-407999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Beg AA, Sha WC, Bronson RT, Baltimore D. Constitutive NF-kappa B activation, enhanced granulopoiesis, and neonatal lethality in I kappa B alpha-deficient mice. Genes Dev. 1995;9:2736–2746. doi: 10.1101/gad.9.22.2736. [DOI] [PubMed] [Google Scholar]
  • 114.Rupec RA, et al. Stroma-mediated dysregulation of myelopoiesis in mice lacking IκBα. Immunity. 2005;22:479–491. doi: 10.1016/j.immuni.2005.02.009. [DOI] [PubMed] [Google Scholar]
  • 115.Breccia M, Alimena G. NF-(kappa)B as a potential therapeutic target in myelodysplastic syndromes and acute myeloid leukemia. Expert Opin. Ther. Targets. 2010;14:1157–1176. doi: 10.1517/14728222.2010.522570. [DOI] [PubMed] [Google Scholar]
  • 116.Taganov KD, Boldin MP, Chang K-J, Baltimore D. NF-kappaB-dependent induction of microRNA miR-146, an inhibitor targeted to signaling proteins of innate immune responses. Proc. Natl. Acad. Sci. U. S. A. 2006;103:12481–12486. doi: 10.1073/pnas.0605298103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Zhao JL, Starczynowski DT. Role of microRNA-146a in normal and malignant hematopoietic stem cell function. Front. Genet. 2014;5:219. doi: 10.3389/fgene.2014.00219. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Votavova H, et al. Differential expression of microRNAs in CD34+ cells of 5q-syndrome. J. Hematol. Oncol. 2011;4:1. doi: 10.1186/1756-8722-4-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Sokol L, et al. Identification of a risk dependent microRNA expression signature in myelodysplastic syndromes. Br. J. Haematol. 2011;153:24–32. doi: 10.1111/j.1365-2141.2011.08581.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Zhao JL, Rao DS, O’Connell RM, Garcia-Flores Y, Baltimore D. MicroRNA-146a acts as a guardian of the quality and longevity of hematopoietic stem cells in mice. eLife. 2013;2013 doi: 10.7554/eLife.00537. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Lu LF, et al. Function of miR-146a in Controlling Treg Cell-Mediated Regulation of Th1 Responses. Cell. 2010;142:914–929. doi: 10.1016/j.cell.2010.08.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Boldin MP, et al. miR-146a is a significant brake on autoimmunity, myeloproliferation, and cancer in mice. J. Exp. Med. 2011;208:1189–1201. doi: 10.1084/jem.20101823. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Zhao JL, et al. NF-kappaB dysregulation in microRNA-146a-deficient mice drives the development of myeloid malignancies. Proc. Natl. Acad. Sci. U. S. A. 2011;108:9184–9189. doi: 10.1073/pnas.1105398108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.De A, Dainichi T, Rathinam CV, Ghosh S. The deubiquitinase activity of A20 is dispensable for NF-κB signaling. EMBO reports. 2014;15:775–83. doi: 10.15252/embr.201338305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Kool M, et al. The ubiquitin-editing protein a20 prevents dendritic cell activation, recognition of apoptotic cells, and systemic autoimmunity. Immunity. 2011;35:82–96. doi: 10.1016/j.immuni.2011.05.013. [DOI] [PubMed] [Google Scholar]
  • 126.Lee EG, et al. Failure to regulate TNF-induced NF-kappaB and cell death responses in A20-deficient mice. Science. 2000;289:2350–2354. doi: 10.1126/science.289.5488.2350. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Boone DL, et al. The ubiquitin-modifying enzyme A20 is required for termination of Toll-like receptor responses. Nat. Immunol. 2004;5:1052–1060. doi: 10.1038/ni1110. [DOI] [PubMed] [Google Scholar]
  • 128.Vereecke L, Beyaert R, van Loo G. The ubiquitin-editing enzyme A20 (TNFAIP3) is a central regulator of immunopathology. Trends in Immunology. 2009;30:383–391. doi: 10.1016/j.it.2009.05.007. [DOI] [PubMed] [Google Scholar]
  • 129.Hövelmeyer N, et al. A20 deficiency in B cells enhances B-cell proliferation and results in the development of autoantibodies. Eur. J. Immunol. 2011;41:595–601. doi: 10.1002/eji.201041313. [DOI] [PubMed] [Google Scholar]
  • 130.Tavares RM, et al. The Ubiquitin Modifying Enzyme A20 Restricts B Cell Survival and Prevents Autoimmunity. Immunity. 2010;33:181–191. doi: 10.1016/j.immuni.2010.07.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Chu Y, et al. B cells lacking the tumor suppressor TNFAIP3/A20 display impaired differentiation and hyperactivation and cause inflammation and autoimmunity in aged mice. Blood. 2011;117:2227–2236. doi: 10.1182/blood-2010-09-306019. [DOI] [PubMed] [Google Scholar]
  • 132.Hammer GE, et al. Expression of A20 by dendritic cells preserves immune homeostasis and prevents colitis and spondyloarthritis. Nature Immunology. 2011;12:1184–1193. doi: 10.1038/ni.2135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Matmati M, et al. A20 (TNFAIP3) deficiency in myeloid cells triggers erosive polyarthritis resembling rheumatoid arthritis. Nat. Genet. 2011;43:908–912. doi: 10.1038/ng.874. [DOI] [PubMed] [Google Scholar]
  • 134.Nagamachi A, et al. Acquired deficiency of A20 results in rapid apoptosis, systemic inflammation, and abnormal hematopoietic stem cell function. PLoS One. 2014;9 doi: 10.1371/journal.pone.0087425. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Nakagawa M, Thummar K, Mandelbaum J, Pasqualucci L, Rathinam C. Lack of the ubiquitin-editing enzyme A20 results in loss of hematopoietic stem cell quiescence. J Exp Med. 2015;212:203–16. doi: 10.1084/jem.20132544. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Wertz IE, et al. De-ubiquitination and ubiquitin ligase domains of A20 downregulate NF-kappaB signalling. Nature. 2004;430:694–699. doi: 10.1038/nature02794. [DOI] [PubMed] [Google Scholar]
  • 137.Lu T, et al. Dimerization and Ubiquitin Mediated Recruitment of A20, a Complex Deubiquitinating Enzyme. Immunity. 2013;38:896–905. doi: 10.1016/j.immuni.2013.03.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Bosanac I, et al. Ubiquitin Binding to A20 ZnF4 Is Required for Modulation of NF-??B Signaling. Mol. Cell. 2010;40:548–557. doi: 10.1016/j.molcel.2010.10.009. [DOI] [PubMed] [Google Scholar]
  • 139.Tokunaga F, et al. Specific recognition of linear polyubiquitin by A20 zinc finger 7 is involved in NF-κB regulation. The EMBO Journal. 2012;31:3856–3870. doi: 10.1038/emboj.2012.241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Tewari M, et al. Lymphoid expression and regulation of A20, an inhibitor of programmed cell death. J. Immunol. 1995;154:1699–1706. [PubMed] [Google Scholar]
  • 141.Jaiswal S, et al. Age Related Clonal Hematopoiesis Associated with Adverse Outcomes. N. Engl. J. Med. 2014;371:2488–98. doi: 10.1056/NEJMoa1408617. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Honma K, et al. TNFAIP3/A20 functions as a novel tumor suppressor gene in several subtypes of non-Hodgkin lymphomas. Blood. 2009;114:2467–2475. doi: 10.1182/blood-2008-12-194852. [DOI] [PubMed] [Google Scholar]
  • 143.Nocturne G, et al. Germline and somatic genetic variations of TNFAIP3 in lymphoma complicating primary Sjogren’s syndrome. Blood. 2013;122:4068–76. doi: 10.1182/blood-2013-05-503383. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Wei Y, et al. Global H3K4me3 genome mapping reveals alterations of innate immunity signaling and overexpression of JMJD3 in human myelodysplastic syndrome CD34+ cells. Leukemia. 2013;27:2177–86. doi: 10.1038/leu.2013.91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Bar M, et al. Gene expression pattern in myelodyplasia underline the role of apoptosis and differentiation in disease initiation and progression. Translational Oncogenomics. 2008;2008:137–149. [PMC free article] [PubMed] [Google Scholar]
  • 146.Kim S-W, et al. MicroRNAs miR-125a and miR-125b constitutively activate the NF-B pathway by targeting the tumor necrosis factor alpha-induced protein 3 (TNFAIP3, A20) Proceedings of the National Academy of Sciences. 2012;109:7865–7870. doi: 10.1073/pnas.1200081109. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES