Abstract
RNA metabolism plays a central role in regulating of T cell-mediated immunity. RNA processing, modifications, and regulations of RNA decay influence the tight and rapid regulation of gene expression during T cell phase transition. Thymic selection, quiescence maintenance, activation, differentiation, and effector functions of T cells are dependent on selective RNA modulations. Recent technical improvements have unveiled the complex crosstalk between RNAs and T cells. Moreover, resting T cells contain large amounts of untranslated mRNAs, implying that the regulation of RNA metabolism might be a key step in controlling gene expression. Considering the immunological significance of T cells for disease treatment, an understanding of RNA metabolism in T cells could provide new directions in harnessing T cells for therapeutic implications.
Keywords: T-lymphocytes, RNA, RNA metabolism, Immunity, Cellular
INTRODUCTION
RNA metabolism refers to the processes that encompass the life cycle of RNA, including RNA synthesis, RNA splicing, RNA modification, RNA decay, and overall quality control. mRNA is transcribed from DNA as a single-stranded RNA template, and is processed simultaneously for 5'-capping, 3'-polyadenylation and alternative splicing (1,2,3). In addition, various modifications, such as RNA methylation, transform RNA structure to increase the accessibility of RNA-binding proteins or to promote degradation by recruiting the CNOT complex (4,5). In the cell, quality control of gene expression largely relies on RNA, therefore, RNA metabolism is elaborately controlled, particularly when drastic, temporal, or poised gene expression changes are involved. For instance, RNA metabolism is extremely important in T cells. During their life cycle, T cells undergo multiple selection processes and dramatic changes of cellular state. In thymocytes, β-chain rearrangement of TCRs occurs through V(D)J recombination to pair with pre-TCRα at the double-negative (DN, CD4-CD8-) stage, which is called β-selection (6). Subsequent double-positive (DP, CD4+CD8+) thymocytes are positively selected depending on the binding ability of TCR to MHC-peptide complexes (6). Then, late DP and single-positive (SP, CD4+ or CD8+) thymocytes that show strong affinity to self-peptide:MHC complexes are removed by negative selection to prevent autoimmunity (6) (Fig. 1). Matured naïve T cells maintain a quiescent state characterized by cell-cycle arrest and low metabolic activity, while they are poised for immediate response to antigen stimulation (7,8). Upon stimulation of both TCR and co-stimulatory molecules, naïve T cells become activated and increase in size with a duplication rate of 6-12 h, and reprogram their metabolic status (9). Together with TCR activation, various combinations of cytokines instruct activated T cells to differentiate into effector T cell subsets (Fig. 1). At the end of their effector function, the majority of T cells enter activation-induced programmed cell death, whereas some surviving effector T cells remain to comprise the memory pool of the adaptive immune system (10). Such a vast shift in the cellular status of T cells demands rapid and timely control of gene expression upon external cues. To this end, intrinsic regulation of gene expression by RNA metabolism is an effective strategy to achieve immediate ‘switch on’ or ‘switch off’ of target genes. Thus, understanding RNA metabolism in T cells may provide new insight into T cell biology.
Over the decades, a body of research has revealed how the complex processes of RNA metabolism are involved in a variety of physiological contexts. This review summarizes recent advances in RNA metabolism, with a particular focus on T cell-mediated immunity. Herein, we describe the molecular details of RNA metabolism in the T cell life cycle. In addition, we also briefly mention the pathological aspects of RNA dysregulation attributed to T cell-related disorders. The role of microRNA in T cells is excluded, as it has been previously reviewed elsewhere (11,12).
IMBALANCE OF TRANSCRIPTION AND TRANSLATIO IN T CELLS
The transition from naive to activated T cells triggers global upregulation of gene expression via rapid recruitment of RNA polymerase II (RNA Pol II) and simultaneously accelerates transcription rates (13). Mapping of de novo transcriptome and ribosome profiling with time-course tracking techniques has shown gene-specific discrepancies in approximately 95% of upregulated genes (13). The fastest response in transcription is for genes involved in basic cell physiology, such as ribosome biogenesis and translational machinery (13,14). Likewise, gene sets associated with production of cytokines and receptors exhibit a marked increase in transcriptional rates concomitant with translation (13). However, a large portion of genes show unmatched rates of transcription and translation, indicating high rates of protein synthesis relative to their de novo transcription rates (13). A study of the dynamics of mRNA translation using the pulsed SILAC technique has delineated an imbalance between transcription and translation rates in the early stage of T cell activation (15). Within the first 6 h after activation, the total mRNA copies showed only a mild (approximately 1.4-fold) increase (15). Notably, with negligible changes in ribosomal proteins and rRNA, the translation rate showed a 5-fold increase from 0.8/sec to 4.0/sec (15). This discrepancy can be explained by the presence of idling ribosomes, which are poised, and mRNAs are translationally repressed before activation (Fig. 2). Furthermore, similar evidence of idling translational machinery has been reported in the context of metabolic reprogramming during the naive-to-activation transition. Naive T cells accumulate mRNAs associated with glycolysis and fatty acid synthesis but stall translation until they are engaged by TCR stimulation (16). T cell activation leads to translation of the glucose transporter GLUT1 and acetyl-coenzyme A carboxylase 1, consequently completing metabolic reprogramming (16). Collectively, these studies indicate that a large portion of mRNAs remain untranslated in resting T cells, despite adequate translational machinery. This highlights the significance of RNA regulation combined with transcriptional and protein signaling regulation to control T cell homeostasis.
ROLE OF RNA SPLICING IN T CELL HOMEOSTASIS
Following transcription, primary transcripts, termed pre-mRNAs, undergo processing steps including 5'-capping, 3'-polyadenylation, and alternative splicing (1,2,3). RNA processing occurs concurrently with transcription because RNA pol II engages RNA processing machinery, including cap-binding proteins, polyadenylation complexes, and spliceosomes (3). 5'-capping and polyadenylation at the 3'-end influence mRNA stability, which is reviewed in the next section. Most pre-mRNAs in eukaryotic cells are alternatively spliced (17). 5' or 3'-splicing or exon skipping contributes to proteomic diversity by generating multiple isoforms from a limited number of genes (18). In addition, exon inclusion and intron retention determine the subsequent fates of mRNAs (19). As seen in other cell types, splicing events broadly regulate T cell homeostasis throughout their life cycle.
Global regulation of RNA splicing in T cells
A study using RNA-seq profiling of both human T cell lines and primary T cells found a comprehensive pattern of RNA splicing that characterized the isoform changes of 178 exons in 168 genes in response to stimulation (20). Transcription factors Foxa1 and Foxa2 (Foxa1/2) have recently been identified as global regulators of RNA splicing during the transition from DP to SP thymocytes (21). Double conditional knockout of Foxa1/2 (Foxa1fl/flFoxa2fl/flCD4cre) disrupted the development of SP population and caused aberrant RNA splicing (21). Combined analysis of human CD4+ T cells using RNA-immunoprecipitation and mass spectrometry has also identified U2 small nuclear RNA auxiliary factor 2 (U2AF2) as a central regulator of RNA splicing by recruiting spliceosome to target mRNA, suggesting that U2AF plays an indispensable role in the optimal expression of many effector molecules, such as CD25 and CD62L, and subset-specific cytokines in T cells (22). Similarly, another study suggested that serine/arginine-rich splicing factor 1 (SRSF1) has an impact in T cell development (23). Additionally, this study also identified many binding partners of SRSF1 related to T cell differentiation factors, implying that SRSF1 has multiple effects on T cell biology (23). Furthermore, an abnormal decrease in SRSF1 was observed in patients with systemic lupus erythematosus (SLE) with a hyperresponsive T cell population (24). Consistent with these findings, mice lacking SRSF1 exhibit aberrantly elevated mTOR activity and reduced expression of its repressor, phosphatase and tensin homolog (24). A recent study demonstrated that different RNA splicing patterns affect the maintenance of T cell quiescence and it highlighted the negative effects of transcript variants containing long interspersed nuclear element 1 (LINE1). Owing to the unique splicing pattern in naive CD4+ T cells, intronic LINE1 is inserted into the transcripts upregulated during T cell activation, and these variants act with nucleolin to repress gene expression in the chromatin in a cis-acting manner (25). The activation cue alters the splicing pattern, which reduces LINE1-containing transcripts via the splicing suppressor polypyrimidine-tract binding protein 1 (PTBP1), leading to the expression of the corresponding genes (25). Although PTBP1 regulates T cell activation and IL-2 production (26), its role in T cell biology remains unknown. Collectively, complicated patterns of RNA splicing for diverse gene sets globally orchestrate T cell homeostasis throughout the T cell life cycle.
Exon inclusion and skipping
The RNA splicing factor CUGBP Elav-like family member 2 (CELF2) has been described for its multiple roles in the regulation of metabolism, alternative splicing, and translation (27). T cell activation increases the mRNA stability of CELF2, which is responsible for widespread alternative splicing in T cells (28). It controls alternative splicing of one-third of the genes associated with TCR signaling and T cell development via either exon skipping or inclusion (28). The best-known targets of CELF2-dependent RNA regulation in T cells are MAPK kinase 7 (MKK 7) and lymphoid enhancer-binding factor 1 (LEF1). Optimal production of effector cytokines by activated T cells requires JNK activity (29). MKK7 activates JNK, leading to activation of the transcription factor c-Jun (30). The complete form of MKK 7 is incapable of interacting with JNK, as exon 2 in the MKK 7 transcript is inserted within the JNK docking site and prevents binding (31). Enhanced CELF2 activity upon T cell activation causes exon skipping of introns flanking exon 2 in MKK 7, forming an intact docking site for JNK (31). Conversely, CELF2 directs exon inclusion, generating the full length of LEF-1, which contains exon 6 (32). LEF-1 coordinates the expression of TCRα in developing thymocytes; thus, it is critical for DN-to-DP transition during T cell development (32). Exon 6 in LEF-1 encodes a context-dependent regulatory domain that enables LEF-1 to interact with the co-activators Aly and Ets for TCRα expression (32,33). The splicing factor SRSF1 also plays a key role in exon inclusion. Phenotypically, the loss of SRSF1 in developing T cells (Srsf1fl/fl Lckcre) resulted in impaired maturation in late stage T cell development (23). Genome-wide analysis of SRSF1 binding targets has shown that it binds to the pre-mRNA of Il-27ra and IFN regulatory factor 7 (Irf-7) in response to type I IFN signaling (23), which is critical for late T cell development and survival (34). In the absence of SRSF1, defective splicing of these genes, such as skipping of exon 3 in Il-27ra mRNA, or retention of intron 5 in Irf-7 mRNA, introduced a premature termination codon that lead to mRNA decay, resulting in increased levels of pro-apoptotic factors, Bax and Bcl2l11 (23) and compromised IL-27 signaling. Likewise, SRSF1 promoted the inclusion of exon 5 in the mRNA encoding the T cell surface glycoprotein CD6 (35). Downregulation of SRSF1 by T cell activation increases exon 5 skipping in CD6 mRNA (35). Although it has not been determined whether CD6 has a stimulatory or an inhibitory effect, skipping exon 5 in CD6 might limit its function because it encodes a ligand-binding domain (35). Splicing patterns of TCR-related molecules controlled by heterogeneous nuclear ribonucleoprotein (HNRNP) L and A1 determine the fate of CD4+ T cells (36). Strong TCR signaling combined with cytokine signals induces the development of effector T cell subsets, whereas tonic TCR stimulation induces Treg differentiation (37). Weak TCR stimulation alters several substrates of Akt, including splicing factors that favor the activation of HNRNP L and HNRNP A1 (36). In conditions favoring the development of Tregs, distinct patterns in alternative splicing induce the expression of other variants of CD3ζ, CD3η and CD3, which lack the immunoreceptor tyrosine-based activation motif, and favor CD45RO instead of CD45RB by skipping exon 5, contributing to Treg generation (36). Similarly, the splicing pattern of mucosa-associated lymphoid tissue lymphoma translocation protein 1 (MALT1) differs depending on whether T cells are resting or activated. In resting T cells, the splicing activity of HNRNP U generates the exon-excluded MALT1B isoform, whereas TCR stimulation relieves the suppressive effect of HNRNP U and allows the isoform, MALT1A, which contains exon 7, with enhanced binding ability to TRAF6 (38). From a disease perspective, distinct splicing events may drive pathological outcomes. For example, although the membrane-bound IL-7 receptor (IL-7R) plays an essential role in T cell survival and maintenance of homeostasis (39), aberrant skipping of exon 6 generates a secreted form of IL-7R, which is associated with an increased risk of multiple sclerosis (MS), as MS patients often possess a single nucleotide polymorphism in Il-7r exon 6 (40,41). On the contrary, RNA helicase DEAD box polypeptide 39 B is responsible for the formation of the complete form of IL-7R by preventing skipping of exon 6 (41).
Intron retention
The intron, a non-coding region of pre-mRNA, has a substantial impact on RNA regulation in T cells, as intron retention is one of the mechanisms regulating the number of transcripts linked to cell fate decisions (42). For example, selective induction of intron retention occurs in exons 4 to 6 of Ptprc mRNA (encoding CD45) by the RNA-binding protein HNRNPLL in T cells (43). Like exon splicing, global changes in intron retention are observed upon T cell activation, although the precise mechanism of intronic control of gene expression is still poorly understood. A study using strand-specific RNA-seq showed intronic sequences were favorably inserted in polyadenylated transcripts in resting CD4+ T cells, displaying distinctive patterns of intron retention in T cells (44). Furthermore, intron retention was observed in 185 genes that were most frequently upregulated during T cell activation, and retained introns were dramatically decreased upon T cell stimulation (44). Collectively, this emphasizes that gene expression is dominantly controlled by splicing rather than transcriptional regulation.
EPIGENETIC MODIFICATION OF RNA IN T CELLS
In addition to RNA processing steps, internal modification of RNA is another important step to determine mRNA fate and gene expression. Chemical modification or addition of certain molecules to RNAs represents most of the internal modifications encompassing methylation (m6A and m5C), ribose modification (2'-O-methylation; 2'-OMe), adenosine (A)-to-inosine (I) modification, 5-hydroxymethylcytosine, and uridylation and isomerization of uridine (45). These modifications alter the characteristics of RNAs depending on the marked molecules, subsequently affecting pre-mRNA processing, stability, and mRNA translation. Advances in analytical chemistry and high-throughput sequencing methods have given rise to a new concept of “epitranscriptome,” which has enabled the consideration of the molecular dynamics of chemically modified RNA. Correspondingly, many studies have actively been deciphering the role of RNA modification in T cell biology.
RNA methylation in T cells
Methylation of RNA includes modifications such as N6-methyladenosin (m6A), N1-mthyladenosine, and N5-methylcytosine (m5C), which are modulated by several proteins referred to as “writers”, “erasers”, and “readers” (46,47). “Writers” transfer methyl residues on the RNA molecule, whereas “erasers” remove them by demethylase activity, and “readers” recognize methylated marks on RNAs. m6A is one of the most abundant modifications and is enriched in 3'-untranslated region (UTR), long internal exons, and near the stop codons of RNAs (48). Methyltransferases such as METTL3 and METTL14 act as writers of m6A modifications (49). An m6A mark on RNAs has a multifunctional role by recruiting reader proteins that bind to the m6A structure. Given that various RNA-binding proteins, such as RNA stabilizers or splicing factors, can recognize m6A, changing its accessibility ultimately determines mRNA half-life and gene expression (50). For example, the binding of YTH N6-methyladenosine RNA-binding protein (YTHDF) 1, a reader protein, accelerates the translation of target mRNA by recruiting translation initiation factors (51). In addition, YTHDF2, another reader protein, recognizes m6A-marked mRNA and induces decay by recruiting the CCR4-NOT complex (52). The ablation of YTHDF1 enhances anti-tumor immunity by facilitating cross-presentation of CD8+ T cells from dendritic cells, suggesting that YTHDF1 has an indirect effect on T cells (4). The role of m6A modification in T cell homeostasis has also been well described. Deletion of METTL3, which acts as an m6A writer, disrupts IL-7-JAK/STAT5 signaling in T cell homeostasis and differentiation (53). Mettl3-deficient T cells (Mettl3fl/flCd4cre) adoptively transferred into RAG knockout mice failed to induce colitis due to their retained resting status (53). Upon IL-7 stimulation, METTL3-mediated m6A marking selectively targeted suppressor of cytokine signaling-1 (Socs1), Socs3, and cytokine-inducible SH2-containing protein which are inhibitory proteins for STAT signaling, and induced their degradation, consequently activating IL-7-JAK/STAT5 signaling (53). In line with this, the same group has reported that Tregs lose their suppressive ability in the absence of METTL3 (Mettl3fl/flFoxp3cre) due to the lack of m6A modification of the inhibitory Socs gene family (5), as reported previously (53). However, the studies that describe the role of METTL3 in follicular helper T cells (TFH) are controversial. Knockdown of METTL3 or METTL14 with shRNA promotes TFH differentiation upon lymphocytic choriomeningitis virus (LCMV) infection (54). In contrast, another study showed that METTL3 deficiency disrupted TFH differentiation due to the lack of stabilized transcription factor 7 via METTL3-mediated m6A (55). Another contributing factor to the complexity of m6A modifications in T cells is the demethylation of m6A by the eraser protein such as AlkB homolog 5 RNA demethylase (ALKBH5) known to involve to CD4+ T cell activation (56). TCR stimulation upregulated ALKBH5 expression, and the loss of ALKBH5 in T cells (Alkbh5fl/flCd4cre) resulted in enhanced m6A modification of IFN-γ and CXCL2 mRNAs, increasing their vulnerability to decay and reducing their protein expression (56). Consistently, the adoptive transfer of ALKBH5-deficient T cells into RAG knockout mice failed to induce autoimmune colitis, and Alkbh5fl/flCd4cre mice exhibited resistance to experimental autoimmune encephalomyelitis (EAE) induction (56). Compared to m6A modifications, the effects of m5C and m1A on T cells are unknown. The only known enzyme that catalyzes m5C modification is NOP2/Sun RNA methyltransferase 2 (NSUN2). The CD4+ T cells of patients with SLE exhibit a decreased pattern of m5C and significantly reduced NSUN2 expression (57). Meanwhile, m1A modification is mainly found in tRNAs (58). A recent study that screened for dynamic changes in the tRNA pool in T cells showed that the m1A level in tRNA did not change during T cell proliferation and differentiation (59). However, the same study has identified 2 unique modifications, namely, wybutosine and ms2t6A, which occur in tRNAs bearing slippery codons, and prevents ribosomal frameshifting (59). These modifications were markedly reduced in activated T cells, and such modifications are likely to increase proteome-wide frameshifting (59).
Adenosine-to-inosine (A-to-I) modification during T cell development
Apart from mRNA modification, A-to-I modification of RNA is uniquely exploited during T cell development. Developing thymocytes migrate from the cortex into the thymic medulla, which contains abundant type I IFN, and IRF-7 is constitutively expressed in thymocytes (60). Dependency on IFN signaling in the absence of infection may cause unnecessary activation through increased expression of genes responsible for antiviral immunity, such as melanoma differentiation-associated protein 5, a sensor of ds RNA (61). The RNA-editing molecule adenosine deaminase acting on RNA1 (ADAR1) plays a compensatory role. ADAR1 catalyzes A-to-I modification upon recognition of endogenous dsRNA, preventing unwanted sensing of dsRNA by relieving the dsRNA structure (62). ADAR1 appears to have a similar role in developing thymocytes, as the expression of ADAR1 is upregulated during the late stage of thymic T cell maturation (63). Indeed, ADAR1-deficient thymocytes fail to fully mature and remain in the semi-mature stage, defined by a CD69-MHC I+ phenotype ( 63).
Effects of RNA uridylation on T cell homeostasis
Although terminal uridylation of microRNA has been reported to be involved in its stability in CD4+ T cells (64), little is known about the direct impact of RNA uridylation in T cells. In the RAW 264.7 murine macrophage cell line, terminal uridyltransferase 7 (TUT7) positively regulates the expression of IL-6 by controlling the mRNA stability of Regnase-1 (also known as Zc3h12a). TUT7 expression requires p38 MAPK signaling and uridylated Regnase-1 mRNA by binding to the stem-loop in its 3’-UTR, ultimately facilitating its decay. Given the underlying mechanism of TUT7 shown in RAW264.7, it gives rise to the potential role of TUT7-mediated RNA uridylation in T cells indirectly because it shares similar signaling pathways upon TCR stimulation, and REGNASE-1 has a broad effect on T cell homeostasis by controlling mRNA decay. Thus, it may be an interesting area of research to explore in future studies.
Collectively, the crosstalk between chemically modified RNAs and T cell biology is an important mechanism of RNA-mediated T cell regulation. Given the importance of RNA modification in T cell regulation, it is worthwhile to explore the potential role of these mechanisms in controlling T cell function.
REGULATION OF RNA DECAY IN T CELLS
Many processes of RNA metabolism described above often closely correlate to RNA decay. Because of their unstable nature, typical mRNAs are protected by a 5'-7-methylguanosine cap (5'-cap) and 3' poly-A (poly(A)) tail (65,66). Degradation of mRNA occurs through multiple pathways. Most mRNAs undergo exonuclease-dependent mRNA decay that relies on poly(A) tail degradation in the 3' region of mRNA by de-adenylation complexes such as CCR4-NOT, exposing mRNA to an unfavorable environment (67). Subsequent decapping by the decapping complex Dcp1/Dcp2 removes the 5’-cap, increasing the vulnerability of mRNA (68,69). Lastly, exonucleases catalyze mRNA degradation. Endonuclease-dependent mRNA decay is achieved by endonucleases such as REGNASE-1 (70). Without chemical removal on each side of the mRNA, endonucleases cause exposed 5’- and 3’- ends to be susceptible to exonucleases (71). For rapid and tight regulation of gene expression in response to extracellular stimuli, T cells often harness the mechanisms that enhance or abrogate mRNA stability.
Exonuclease-dependent mRNA decay
The role of the CCR4-NOT de-adenylase complex in silencing gene expression has been extensively reported in previous descriptions of T cell biology. For example, the expression of CNOT3, a regulatory component required for the integrity of the CCR4-NOT complex, is upregulated in DP thymocytes (72), implying the involvement of mRNA de-adenylation during T cell development. T cell-specific deletion of Cnot3 (Cnot3fl/fl-CD4cre) has been shown to result in the failure of mature CD4+ and CD8+ T cell production with upregulated pro-apoptotic genes, including Dap2ip and Bbc3 mRNA, in the early and late stages of positive selection, respectively (72). The RNA-binding proteins ZFP36L1 and ZFP36L2 (ZFP36L1/2) also utilize the CCR4-NOT complex to guide target mRNA degradation across the T cell life cycle. Mechanistically, ZFP36L1/2 belong to the ZFP36 family, which contain a CCCH zinc finger domain that can bind to conserved sequences in the AU-rich element (ARE) of the 3'-UTR (73). The ZFP36L family, including ZFP36L1/2, can interact with CCR4, leading to the recruitment of NOT4 and the final assembly of the CCR4-NOT de-adenylase complex, which limits target gene expression by promoting mRNA degradation (74). In the early developmental stages, ZFP36L1/2 downregulates the expression of Notch-1, which is crucial for thymocyte proliferation. Despite its role in cell growth (75), uncontrolled signaling from Notch-1 induces thymocyte proliferation without proper maturation steps; therefore, it should be tightly regulated depending on the developmental stage. One study showed that mice with selective deficiency of ZFP36L1/2 in early thymocytes (Zfp36l1fl/flZfp36l2fl/fl-Cd2cre) showed T cell acute lymphoblastic leukemia with aberrant accumulation of T cells bypassing β-selection (76). ZFP36L1/2 interacted with the highly conserved ARE region of Notch-1 mRNA, although the mechanism of Notch-1 mRNA decay is not clear (76). In subsequent works, it was determined that ZFP36L1/2 was also associated with the genome-wide suppression of cell cycle genes activating the E2F pathway and DNA-damage responses (77,78). Overall, ZFP36L1/2 mediated mRNA decay via CCR4-NOT seems to secure β-selection of early thymocytes by delaying cell proliferation and inhibiting DNA repair genes for uninterrupted V(D)J recombination. RNA regulation by ZFP36 family proteins influences the effector functions of mature T cells. ZFP36L2 limits the translation of IFN-γ by binding to AREs in the 3'-UTR of Ifng mRNA (79). Consistently, ZFP36 deficient mice were more resistant to antiviral response against LCMV infection with not only a dramatic increase of IFN-γ-expressing T cells, but also enhanced expression of activation and proliferation markers (80). RNA-binding proteins belonging to the ROQUIN family, including ROQUIN-1 and ROQUIN-2 (ROQUIN1/2), share a CCR4-NOT-mediated mechanism to regulate target mRNA availability. Although ROQUIN1/2 have a CCCH-type zinc finger domain, the ROQ domain is majorly responsible for their binding specificity, and these domains act synergistically (81,82). Through this domain, ROQUIN1/2 bind to the 3'-UTR (100-200 bp downstream of the stop codon) of the inducible T cell co-stimulator (ICOS) and limit the abundance of Icos mRNA by recruiting the CCR4-NOT complex (81,83). Since ICOS is essential for the regulation of T cell immunity, particularly in TFH and germinal center (GC) responses (84), disrupted activity of ROQUIN1/2 spontaneously causes severe autoimmunity resembling SLE characterized by splenomegaly and abnormally increased GC formation, as seen in Sanroque strain mice which contain the M199R mutation in Rc3h1 (encoding ROQUIN1) (85,86). Consistently, similar phenotypes have been observed in the T cell-specific deletion of ROQUIN1/2 (Rc3h1/2fl/flCD4cre) (82). Recent work has also determined a role for CCR4-NOT-mediated mRNA decay in the quiescence maintenance of naive T cells. A study has identified B-cell translocation gene 1 (BTG1) and BTG2 (BTG1/2) as novel regulators of T cell quiescence by inducing global mRNA deadenylation and degradation (87). Remarkably, BTG1/2 were mainly expressed in quiescent T cells and swiftly turned off upon activation cues. BTG1/2-deficient T cells (BTG1/2fl/fl-CD4Cre) exhibited a global increase in mRNA abundance, leading to a compromised naive status, accompanied by the aberrant accumulation of effector memory population in steady-state conditions (87). In quiescent T cells, BTG1/2 can physically bind with poly(A) tail binding protein and the CCR4-NOT complex, even though they do not bear the direct RNA cognition domain, thereby facilitating constant deadenylation by inducing global mRNA decay machinery (87). Such a seemingly inefficient process is advantageous for a rapid response upon stimulation from the retained availability of pre-made mRNAs. Although the regulation of RNA metabolism in T cell quiescence has received less attention thus far, there is extensive evidence to elucidate how RNA metabolism is involved in T cell quiescence.
Endonuclease-dependent mRNA decay
REGNASE-1 acts as an endonuclease that harbors a PIN-like RNase domain and specifically cleaves mRNA upon recognition of the stem-loop structure cooperatively with helicase upstream frameshift 1 (88). In the immune system, it is known to destabilize mRNAs encoding the inflammatory cytokines IL-6 and IL-12 (89). Mice with homozygous knockout of REGNASE-1 (Zc3h12a-/-) succumbed within 12 weeks after birth due to severe anemia and showed significantly high effector/memory splenic T cells (89). Similarly, another study has shown that the loss of REGNASE-1 induced spontaneous autoimmune phenotype marked by the increased expression of T cell activation markers and inflammatory cytokines (90). In contrast, mice with a mutant form of REGNASE-1 with enhanced stability exhibited lymphopenia and were unresponsive to skin allografts due to disrupted thymic T cell development (91), indicating that REGNASE-1 controls T cell homeostasis in diverse aspects. In addition to its role in exonuclease-dependent mRNA decay, ROQUIN also cooperates with REGNASE-1 and induces target mRNA degradation as they share target mRNAs associated with inflammatory responses and Th17 differentiation (92). In contrast, upon TCR stimulation, REGNASE-1 is degraded in a MALT-1-dependent manner, relieving T cells from suppressive conditions (90).
Mechanisms protecting of mRNA from decay
As mentioned previously, ELAVL1 is a stabilizer of mRNA. Similar to other molecules contributing to mRNA decay, ELAVL1 interacts with introns and ARE regions in the 3'-UTR of mRNA, but it increases the mRNA lifespan by interfering with exonuclease access and destabilizing degrading complexes (93).
A study using Jurkat human T cell line has shown that long 3'-UTR is favorably targeted by ELAVL1 and is important for target protein localization (94). During T cell development, thymocytes deficient in ELAVL1 (Elavl1fl/flLckcre) exhibited defective β-selection and a failure of thymus egression concomitant with hyperproliferation of thymocytes (95). While the study did not clearly address the molecular mechanism of ELAVL1, it suggested that ELAVL1 is a stabilizer of p53, ultimately inducing cell cycle arrest of DN thymocytes (95). Recently, it was shown that ELAVL1 regulates the translation of Ccnd3 mRNA (encoding cyclin D3) via direct interaction (96). However, further studies are required to understand the multifunctional role of ELAVL1 in cell-cycle genes during T cell development.
RNA stabilizing activity of ELAVL1 regulates broad aspects of T cell function. The mRNA encoding IL-2 receptor α chain (CD25) requires ELAVL1 interaction for optimal translation, as the loss of ELAVL1 in CD4+ T cells (HuRfl/flLckcre) has shown defective IL-2 homeostasis (97). In effector T cell subsets, ELAVL1 stabilizes mRNA encoding IL-4, IL-13, and IL-17A. Deletion of ELAVL1 in activated T cells (HuRfl/flOX40cre) abrogated Th17 responses as they failed to induce EAE, while Th2 responses were unchanged (98,99). Moreover, transcriptome-wide analysis revealed potential targets of ELAVL1 associated with T cell activation pathways, further suggesting a comprehensive role of ELAVL1 in T cell function.
CONCLUSION
To date, T cell studies have mainly focused on their functions and cellular interactions from a macroscopic view, while the intrinsic activities of T cells have been poorly understood. Although clinical techniques involving T cells currently has improved medical therapeutics, there are still many limitations with utilizing T cells as an effective drug target. Therefore, more research is needed to gain a fundamental understanding of T cell biology. Recent breakthroughs in technical and analytical tools have enabled us to scrutinize the RNA world across T cell life cycles. A large portion of mRNAs remain untranslated in resting T cells and diverse RNA metabolism is intrinsically involved in T cell functions (summarized in Fig. 3). Several studies have also implicated that dysregulation of RNA metabolism in T cells is associated with pathological outcomes in both human and animal models (Table 1). Given the central role of RNA and the underlying mechanisms of T cell regulation, reconsidering the significance of RNA metabolism in T cells may open new paradigms for future therapeutic strategies.
Table 1. Effect of RNA metabolism on disease and pathological phenotypes.
RNA metabolism | Associated factor | Impact on T cells | Species | Related phenotype/Diseases | References |
---|---|---|---|---|---|
RNA splicing | FOXA1/2 | Positive selection DP thymocytes | Mouse | T cell developmental defects | (21) |
SRSF1 | Exon inclusion of Il-27ra and Irf-7 | Mouse | T cell developmental defects | (23) | |
Enhanced mTOR signaling | Human | SLE | (24) | ||
Reduced PTEN activity | |||||
DDX39B | Prevention of IL-7R exon 6 skipping | Human | MS | (40,41) | |
RNA modification | METTL3 | m6A writer; promoting IL-7-JAK-STAT5 signaling pathway | Mouse | T cell activation and effector function | (53) |
m6A writer; unknown | Mouse | TFH differentiation upon LCMV infection | (54) | ||
ALKBH5 | m6A eraser; inducing degradation of Ifng and Cxcl2 mRNA | Mouse | T cell activation and effector function | (56) | |
NSUN2 | m5C modification; unknown | Human | SLE | (57) | |
RNA decay | ZFP36L1/2 | β-selection of thymocytes | Mouse | T-ALL | (76,79,80) |
Limiting the abundance of Ifng mRNA | T cell activation and effector function | ||||
ROQUIN1/2 | Inducing decay of Icos mRNA | Mouse | TFH differentiation | (82,85,86) | |
Spontaneous SLE-like autoimmunity | |||||
BTG1/2 | Global mRNA decay in resting T cell | Mouse | T cell quiescence | (87) | |
REGNASE-1 | Inflammatory mRNAs decay | Mouse | Severe anemia | (89,90,91) | |
Spontaneous autoimmunity | |||||
T cell developmental defects |
m5C, N5-methylcytosine; m6A, N6-methyladenosin; PTEN, phosphatase and tensin homolog deleted on Chromosome 10; T-ALL, T-cell acute lymphoblastic leukemia.
ACKOWLEDGEMENTS
We thank all members of the Department of Biochemistry and Molecular Biology of Yonsei University College of Medicine for administrative support. This work was supported by the Brain Korea 21 FOUR Project for Medical Science, Yonsei University College of Medicine. This work of SSH was supported by the National Research Foundation of Korea (NRF) grant funded by the Korea government (MSIT) (NRF-2021R1A2C2093640 and 2018R1A5A2025079) and a faculty research grant (6-2021-0156) and a new faculty research seed money grant (2021-32-0055) of Yonsei University College of Medicine.
Abbreviations
- ADAR1
adenosine deaminase acting on RNA1
- ALKBH5
AlkB homolog 5 RNA demethylase
- ARE
AU-rich element
- A-to-I
adenosine-to-inosine
- BTG
B-cell translocation gene
- CELF
CUGBP Elav-like family member
- DN
double-negative
- DP
double-positive
- EAE
experimental autoimmune encephalomyelitis
- GC
germinal center
- ICOS
inducible T cell co-stimulator
- IL-7R
IL-7 receptor
- Irf-7
interferon regulatory factor 7
- LCMV
lymphocytic choriomeningitis virus
- LEF-1
lymphoid enhancer-binding factor 1
- LINE
long interspersed nuclear element
- MALT1
mucosa-associated lymphoid tissue lymphoma translocation protein 1
- MKK
MAPK kinase
- MS
multiple sclerosis
- NSUN
NOP2/Sun RNA methyltransferase
- poly(A)
poly-A
- PTBP
polypyrimidine-tract binding protein
- RNA Pol II
RNA polymerase II
- SLE
systemic lupus erythematosus
- Socs
suppressor of cytokine signaling
- SRSF
serine/arginine-rich splicing factor
- SP
single-positive
- TFH
follicular helper T cell
- TUT
terminal uridyltransferase
- UTR
untranslated region
- U2AF
U2 small nuclear RNA auxiliary factor
- YTHDF
YTH N6-methyladenosine RNA-binding protein
Footnotes
Conflict of Interest: The authors declare no potential conflicts of interest.
- Conceptualization: Hwang SS.
- Supervision: Hwang ss.
- Writing – original draft: Choi JO, Ham JH.
- Writing – review & editing: Choi JO, Ham JH, Hwang SS.
References
- 1.Beyer AL, Osheim YN. Splice site selection, rate of splicing, and alternative splicing on nascent transcripts. Genes Dev. 1988;2:754–765. doi: 10.1101/gad.2.6.754. [DOI] [PubMed] [Google Scholar]
- 2.Bird G, Zorio DA, Bentley DL. RNA polymerase II carboxy-terminal domain phosphorylation is required for cotranscriptional pre-mRNA splicing and 3′-end formation. Mol Cell Biol. 2004;24:8963–8969. doi: 10.1128/MCB.24.20.8963-8969.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Nagaike T, Logan C, Hotta I, Rozenblatt-Rosen O, Meyerson M, Manley JL. Transcriptional activators enhance polyadenylation of mRNA precursors. Mol Cell. 2011;41:409–418. doi: 10.1016/j.molcel.2011.01.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Han D, Liu J, Chen C, Dong L, Liu Y, Chang R, Huang X, Liu Y, Wang J, Dougherty U, et al. Anti-tumour immunity controlled through mRNA m6A methylation and YTHDF1 in dendritic cells. Nature. 2019;566:270–274. doi: 10.1038/s41586-019-0916-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Tong J, Cao G, Zhang T, Sefik E, Amezcua Vesely MC, Broughton JP, Zhu S, Li H, Li B, Chen L, et al. m6A mRNA methylation sustains Treg suppressive functions. Cell Res. 2018;28:253–256. doi: 10.1038/cr.2018.7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Love PE, Bhandoola A. Signal integration and crosstalk during thymocyte migration and emigration. Nat Rev Immunol. 2011;11:469–477. doi: 10.1038/nri2989. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Chapman NM, Boothby MR, Chi H. Metabolic coordination of T cell quiescence and activation. Nat Rev Immunol. 2020;20:55–70. doi: 10.1038/s41577-019-0203-y. [DOI] [PubMed] [Google Scholar]
- 8.Chapman NM, Chi H. Hallmarks of t-cell exit from quiescence. Cancer Immunol Res. 2018;6:502–508. doi: 10.1158/2326-6066.CIR-17-0605. [DOI] [PubMed] [Google Scholar]
- 9.Hwang JR, Byeon Y, Kim D, Park SG. Recent insights of T cell receptor-mediated signaling pathways for T cell activation and development. Exp Mol Med. 2020;52:750–761. doi: 10.1038/s12276-020-0435-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Kaech SM, Wherry EJ, Ahmed R. Effector and memory T-cell differentiation: implications for vaccine development. Nat Rev Immunol. 2002;2:251–262. doi: 10.1038/nri778. [DOI] [PubMed] [Google Scholar]
- 11.Gebert LF, MacRae IJ. Regulation of microRNA function in animals. Nat Rev Mol Cell Biol. 2019;20:21–37. doi: 10.1038/s41580-018-0045-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Baumjohann D, Ansel KM. MicroRNA-mediated regulation of T helper cell differentiation and plasticity. Nat Rev Immunol. 2013;13:666–678. doi: 10.1038/nri3494. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Davari K, Lichti J, Gallus C, Greulich F, Uhlenhaut NH, Heinig M, Friedel CC, Glasmacher E. Rapid genome-wide recruitment of RNA polymerase ii drives transcription, splicing, and translation events during t cell responses. Cell Reports. 2017;19:643–654. doi: 10.1016/j.celrep.2017.03.069. [DOI] [PubMed] [Google Scholar]
- 14.Araki K, Morita M, Bederman AG, Konieczny BT, Kissick HT, Sonenberg N, Ahmed R. Translation is actively regulated during the differentiation of CD8+ effector T cells. Nat Immunol. 2017;18:1046–1057. doi: 10.1038/ni.3795. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Wolf T, Jin W, Zoppi G, Vogel IA, Akhmedov M, Bleck CK, Beltraminelli T, Rieckmann JC, Ramirez NJ, Benevento M, et al. Dynamics in protein translation sustaining T cell preparedness. Nat Immunol. 2020;21:927–937. doi: 10.1038/s41590-020-0714-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Ricciardi S, Manfrini N, Alfieri R, Calamita P, Crosti MC, Gallo S, Müller R, Pagani M, Abrignani S, Biffo S. The translational machinery of human CD4+ T cells is poised for activation and controls the switch from quiescence to metabolic remodeling. Cell Metab. 2018;28:895–906.e5. doi: 10.1016/j.cmet.2018.08.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Pan Q, Shai O, Lee LJ, Frey BJ, Blencowe BJ. Deep surveying of alternative splicing complexity in the human transcriptome by high-throughput sequencing. Nat Genet. 2008;40:1413–1415. doi: 10.1038/ng.259. [DOI] [PubMed] [Google Scholar]
- 18.Wang ET, Sandberg R, Luo S, Khrebtukova I, Zhang L, Mayr C, Kingsmore SF, Schroth GP, Burge CB. Alternative isoform regulation in human tissue transcriptomes. Nature. 2008;456:470–476. doi: 10.1038/nature07509. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Jacob AG, Smith CW. Intron retention as a component of regulated gene expression programs. Hum Genet. 2017;136:1043–1057. doi: 10.1007/s00439-017-1791-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Martinez NM, Pan Q, Cole BS, Yarosh CA, Babcock GA, Heyd F, Zhu W, Ajith S, Blencowe BJ, Lynch KW. Alternative splicing networks regulated by signaling in human T cells. RNA. 2012;18:1029–1040. doi: 10.1261/rna.032243.112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Lau CI, Rowell J, Yanez DC, Solanki A, Ross S, Ono M, Crompton T. The pioneer transcription factors Foxa1 and Foxa2 regulate alternative RNA splicing during thymocyte positive selection. Development. 2021;148:dev199754. doi: 10.1242/dev.199754. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Whisenant TC, Peralta ER, Aarreberg LD, Gao NJ, Head SR, Ordoukhanian P, Williamson JR, Salomon DR. The activation-induced assembly of an RNA/protein interactome centered on the splicing factor U2AF2 regulates gene expression in human CD4 T cells. PLoS One. 2015;10:e0144409. doi: 10.1371/journal.pone.0144409. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Qi Z, Wang F, Yu G, Wang D, Yao Y, You M, Liu J, Liu J, Sun Z, Ji C, et al. SRSF1 serves as a critical posttranscriptional regulator at the late stage of thymocyte development. Sci Adv. 2021;7:eabf0753. doi: 10.1126/sciadv.abf0753. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Katsuyama T, Li H, Comte D, Tsokos GC, Moulton VR. Splicing factor SRSF1 controls T cell hyperactivity and systemic autoimmunity. J Clin Invest. 2019;129:5411–5423. doi: 10.1172/JCI127949. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Marasca F, Sinha S, Vadalà R, Polimeni B, Ranzani V, Paraboschi EM, Burattin FV, Ghilotti M, Crosti M, Negri ML, et al. LINE1 are spliced in non-canonical transcript variants to regulate T cell quiescence and exhaustion. Nat Genet. 2022;54:180–193. doi: 10.1038/s41588-021-00989-7. [DOI] [PubMed] [Google Scholar]
- 26.La Porta J, Matus-Nicodemos R, Valentín-Acevedo A, Covey LR. The rna-binding protein, polypyrimidine tract-binding protein 1 (PTBP1) is a key regulator of CD4 T cell activation. PLoS One. 2016;11:e0158708. doi: 10.1371/journal.pone.0158708. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Dasgupta T, Ladd AN. The importance of CELF control: molecular and biological roles of the CUG-BP, Elav-like family of RNA-binding proteins. Wiley Interdiscip Rev RNA. 2012;3:104–121. doi: 10.1002/wrna.107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Mallory MJ, Allon SJ, Qiu J, Gazzara MR, Tapescu I, Martinez NM, Fu XD, Lynch KW. Induced transcription and stability of CELF2 mRNA drives widespread alternative splicing during T-cell signaling. Proc Natl Acad Sci U S A. 2015;112:E2139–E2148. doi: 10.1073/pnas.1423695112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Dong C, Yang DD, Tournier C, Whitmarsh AJ, Xu J, Davis RJ, Flavell RA. JNK is required for effector T-cell function but not for T-cell activation. Nature. 2000;405:91–94. doi: 10.1038/35011091. [DOI] [PubMed] [Google Scholar]
- 30.Matsuda S, Moriguchi T, Koyasu S, Nishida E. T lymphocyte activation signals for interleukin-2 production involve activation of MKK6-p38 and MKK7-SAPK/JNK signaling pathways sensitive to cyclosporin A. J Biol Chem. 1998;273:12378–12382. doi: 10.1074/jbc.273.20.12378. [DOI] [PubMed] [Google Scholar]
- 31.Martinez NM, Agosto L, Qiu J, Mallory MJ, Gazzara MR, Barash Y, Fu XD, Lynch KW. Widespread JNK-dependent alternative splicing induces a positive feedback loop through CELF2-mediated regulation of MKK7 during T-cell activation. Genes Dev. 2015;29:2054–2066. doi: 10.1101/gad.267245.115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Mallory MJ, Jackson J, Weber B, Chi A, Heyd F, Lynch KW. Signal- and development-dependent alternative splicing of LEF1 in T cells is controlled by CELF2. Mol Cell Biol. 2011;31:2184–2195. doi: 10.1128/MCB.05170-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Bruhn L, Munnerlyn A, Grosschedl R. ALY, a context-dependent coactivator of LEF-1 and AML-1, is required for TCRalpha enhancer function. Genes Dev. 1997;11:640–653. doi: 10.1101/gad.11.5.640. [DOI] [PubMed] [Google Scholar]
- 34.Yoshida H, Hunter CA. The immunobiology of interleukin-27. Annu Rev Immunol. 2015;33:417–443. doi: 10.1146/annurev-immunol-032414-112134. [DOI] [PubMed] [Google Scholar]
- 35.da Glória VG, Martins de Araújo M, Mafalda Santos A, Leal R, de Almeida SF, Carmo AM, Moreira A. T cell activation regulates CD6 alternative splicing by transcription dynamics and SRSF1. J Immunol. 2014;193:391–399. doi: 10.4049/jimmunol.1400038. [DOI] [PubMed] [Google Scholar]
- 36.Hawse WF, Boggess WC, Morel PA. TCR signal strength regulates Akt substrate specificity to induce alternate murine Th and T regulatory cell differentiation programs. J Immunol. 2017;199:589–597. doi: 10.4049/jimmunol.1700369. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Bhattacharyya ND, Feng CG. Regulation of T helper cell fate by TCR signal strength. Front Immunol. 2020;11:624. doi: 10.3389/fimmu.2020.00624. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Meininger I, Griesbach RA, Hu D, Gehring T, Seeholzer T, Bertossi A, Kranich J, Oeckinghaus A, Eitelhuber AC, Greczmiel U, et al. Alternative splicing of MALT1 controls signalling and activation of CD4+ T cells. Nat Commun. 2016;7:11292. doi: 10.1038/ncomms11292. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Fry TJ, Mackall CL. The many faces of IL-7: from lymphopoiesis to peripheral T cell maintenance. J Immunol. 2005;174:6571–6576. doi: 10.4049/jimmunol.174.11.6571. [DOI] [PubMed] [Google Scholar]
- 40.Gregory SG, Schmidt S, Seth P, Oksenberg JR, Hart J, Prokop A, Caillier SJ, Ban M, Goris A, Barcellos LF, et al. Interleukin 7 receptor alpha chain (IL7R) shows allelic and functional association with multiple sclerosis. Nat Genet. 2007;39:1083–1091. doi: 10.1038/ng2103. [DOI] [PubMed] [Google Scholar]
- 41.Galarza-Muñoz G, Briggs FB, Evsyukova I, Schott-Lerner G, Kennedy EM, Nyanhete T, Wang L, Bergamaschi L, Widen SG, Tomaras GD, et al. Human epistatic interaction controls IL7R splicing and increases multiple sclerosis risk. Cell. 2017;169:72–84.e13. doi: 10.1016/j.cell.2017.03.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Middleton R, Gao D, Thomas A, Singh B, Au A, Wong JJ, Bomane A, Cosson B, Eyras E, Rasko JE, et al. IRFinder: assessing the impact of intron retention on mammalian gene expression. Genome Biol. 2017;18:51. doi: 10.1186/s13059-017-1184-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Cho V, Mei Y, Sanny A, Chan S, Enders A, Bertram EM, Tan A, Goodnow CC, Andrews TD. The RNA-binding protein hnRNPLL induces a T cell alternative splicing program delineated by differential intron retention in polyadenylated RNA. Genome Biol. 2014;15:R26. doi: 10.1186/gb-2014-15-1-r26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Ni T, Yang W, Han M, Zhang Y, Shen T, Nie H, Zhou Z, Dai Y, Yang Y, Liu P, et al. Global intron retention mediated gene regulation during CD4+ T cell activation. Nucleic Acids Res. 2016;44:6817–6829. doi: 10.1093/nar/gkw591. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Zhao BS, Roundtree IA, He C. Post-transcriptional gene regulation by mRNA modifications. Nat Rev Mol Cell Biol. 2017;18:31–42. doi: 10.1038/nrm.2016.132. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Cao G, Li HB, Yin Z, Flavell RA. Recent advances in dynamic m6A RNA modification. Open Biol. 2016;6:160003. doi: 10.1098/rsob.160003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Fu Y, Dominissini D, Rechavi G, He C. Gene expression regulation mediated through reversible m6A RNA methylation. Nat Rev Genet. 2014;15:293–306. doi: 10.1038/nrg3724. [DOI] [PubMed] [Google Scholar]
- 48.Ke S, Alemu EA, Mertens C, Gantman EC, Fak JJ, Mele A, Haripal B, Zucker-Scharff I, Moore MJ, Park CY, et al. A majority of m6A residues are in the last exons, allowing the potential for 3′ UTR regulation. Genes Dev. 2015;29:2037–2053. doi: 10.1101/gad.269415.115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Liu J, Yue Y, Han D, Wang X, Fu Y, Zhang L, Jia G, Yu M, Lu Z, Deng X, et al. A METTL3-METTL14 complex mediates mammalian nuclear RNA N6-adenosine methylation. Nat Chem Biol. 2014;10:93–95. doi: 10.1038/nchembio.1432. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Liu N, Dai Q, Zheng G, He C, Parisien M, Pan T. N6-methyladenosine-dependent RNA structural switches regulate RNA-protein interactions. Nature. 2015;518:560–564. doi: 10.1038/nature14234. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Wang X, Zhao BS, Roundtree IA, Lu Z, Han D, Ma H, Weng X, Chen K, Shi H, He C. N6-methyladenosine modulates messenger RNA translation efficiency. Cell. 2015;161:1388–1399. doi: 10.1016/j.cell.2015.05.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Du H, Zhao Y, He J, Zhang Y, Xi H, Liu M, Ma J, Wu L. YTHDF2 destabilizes m6A-containing RNA through direct recruitment of the CCR4-NOT deadenylase complex. Nat Commun. 2016;7:12626. doi: 10.1038/ncomms12626. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Li HB, Tong J, Zhu S, Batista PJ, Duffy EE, Zhao J, Bailis W, Cao G, Kroehling L, Chen Y, et al. m6A mRNA methylation controls T cell homeostasis by targeting the IL-7/STAT5/SOCS pathways. Nature. 2017;548:338–342. doi: 10.1038/nature23450. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Zhu Y, Zhao Y, Zou L, Zhang D, Aki D, Liu YC. The E3 ligase VHL promotes follicular helper T cell differentiation via glycolytic-epigenetic control. J Exp Med. 2019;216:1664–1681. doi: 10.1084/jem.20190337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Yao Y, Yang Y, Guo W, Xu L, You M, Zhang YC, Sun Z, Cui X, Yu G, Qi Z, et al. METTL3-dependent m6A modification programs T follicular helper cell differentiation. Nat Commun. 2021;12:1333. doi: 10.1038/s41467-021-21594-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Zhou J, Zhang X, Hu J, Qu R, Yu Z, Xu H, Chen H, Yan L, Ding C, Zou Q, et al. M6A demethylase ALKBH5 controls CD4+ T cell pathogenicity and promotes autoimmunity. Sci Adv. 2021;7:eabg0470. doi: 10.1126/sciadv.abg0470. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Zhang Y, Zhao M, Sawalha AH, Richardson B, Lu Q. Impaired DNA methylation and its mechanisms in CD4+ T cells of systemic lupus erythematosus. J Autoimmun. 2013;41:92–99. doi: 10.1016/j.jaut.2013.01.005. [DOI] [PubMed] [Google Scholar]
- 58.Dominissini D, Nachtergaele S, Moshitch-Moshkovitz S, Peer E, Kol N, Ben-Haim MS, Dai Q, Di Segni A, Salmon-Divon M, Clark WC, et al. The dynamic N1-methyladenosine methylome in eukaryotic messenger RNA. Nature. 2016;530:441–446. doi: 10.1038/nature16998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Rak R, Polonsky M, Eizenberg-Magar I, Mo Y, Sakaguchi Y, Mizrahi O, Nachshon A, Reich-Zeliger S, Stern-Ginossar N, Dahan O, et al. Dynamic changes in tRNA modifications and abundance during T cell activation. Proc Natl Acad Sci U S A. 2021;118:118. doi: 10.1073/pnas.2106556118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Otero DC, Baker DP, David M. IRF7-dependent IFN-β production in response to RANKL promotes medullary thymic epithelial cell development. J Immunol. 2013;190:3289–3298. doi: 10.4049/jimmunol.1203086. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Wu B, Peisley A, Richards C, Yao H, Zeng X, Lin C, Chu F, Walz T, Hur S. Structural basis for dsRNA recognition, filament formation, and antiviral signal activation by MDA5. Cell. 2013;152:276–289. doi: 10.1016/j.cell.2012.11.048. [DOI] [PubMed] [Google Scholar]
- 62.Liddicoat BJ, Piskol R, Chalk AM, Ramaswami G, Higuchi M, Hartner JC, Li JB, Seeburg PH, Walkley CR. RNA editing by ADAR1 prevents MDA5 sensing of endogenous dsRNA as nonself. Science. 2015;349:1115–1120. doi: 10.1126/science.aac7049. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Nakahama T, Kato Y, Kim JI, Vongpipatana T, Suzuki Y, Walkley CR, Kawahara Y. ADAR1-mediated RNA editing is required for thymic self-tolerance and inhibition of autoimmunity. EMBO Rep. 2018;19:e46303. doi: 10.15252/embr.201846303. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Gutiérrez-Vázquez C, Enright AJ, Rodríguez-Galán A, Pérez-García A, Collier P, Jones MR, Benes V, Mizgerd JP, Mittelbrunn M, Ramiro AR, et al. 3′ Uridylation controls mature microRNA turnover during CD4 T-cell activation. RNA. 2017;23:882–891. doi: 10.1261/rna.060095.116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Furuichi Y, LaFiandra A, Shatkin AJ. 5′-Terminal structure and mRNA stability. Nature. 1977;266:235–239. doi: 10.1038/266235a0. [DOI] [PubMed] [Google Scholar]
- 66.Bernstein P, Ross J. Poly(A), poly(A) binding protein and the regulation of mRNA stability. Trends Biochem Sci. 1989;14:373–377. doi: 10.1016/0968-0004(89)90011-x. [DOI] [PubMed] [Google Scholar]
- 67.Doidge R, Mittal S, Aslam A, Winkler GS. Deadenylation of cytoplasmic mRNA by the mammalian CCR4-NOT complex. Biochem Soc Trans. 2012;40:896–901. doi: 10.1042/BST20120074. [DOI] [PubMed] [Google Scholar]
- 68.Wang Z, Jiao X, Carr-Schmid A, Kiledjian M. The hDcp2 protein is a mammalian mRNA decapping enzyme. Proc Natl Acad Sci U S A. 2002;99:12663–12668. doi: 10.1073/pnas.192445599. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Beelman CA, Stevens A, Caponigro G, LaGrandeur TE, Hatfield L, Fortner DM, Parker R. An essential component of the decapping enzyme required for normal rates of mRNA turnover. Nature. 1996;382:642–646. doi: 10.1038/382642a0. [DOI] [PubMed] [Google Scholar]
- 70.Mao R, Yang R, Chen X, Harhaj EW, Wang X, Fan Y. Regnase-1, a rapid response ribonuclease regulating inflammation and stress responses. Cell Mol Immunol. 2017;14:412–422. doi: 10.1038/cmi.2016.70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Yang F, Peng Y, Schoenberg DR. Endonuclease-mediated mRNA decay requires tyrosine phosphorylation of polysomal ribonuclease 1 (PMR1) for the targeting and degradation of polyribosome-bound substrate mRNA. J Biol Chem. 2004;279:48993–49002. doi: 10.1074/jbc.M409776200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Ito-Kureha T, Miyao T, Nishijima S, Suzuki T, Koizumi SI, Villar-Briones A, Takahashi A, Akiyama N, Morita M, Naguro I, et al. The CCR4-NOT deadenylase complex safeguards thymic positive selection by down-regulating aberrant pro-apoptotic gene expression. Nat Commun. 2020;11:6169. doi: 10.1038/s41467-020-19975-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Brooks SA, Blackshear PJ. Tristetraprolin (TTP): interactions with mRNA and proteins, and current thoughts on mechanisms of action. Biochim Biophys Acta. 2013;1829:666–679. doi: 10.1016/j.bbagrm.2013.02.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Sandler H, Kreth J, Timmers HT, Stoecklin G. Not1 mediates recruitment of the deadenylase Caf1 to mRNAs targeted for degradation by tristetraprolin. Nucleic Acids Res. 2011;39:4373–4386. doi: 10.1093/nar/gkr011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Ciofani M, Knowles GC, Wiest DL, von Boehmer H, Zúñiga-Pflücker JC. Stage-specific and differential notch dependency at the αβ and γδ T lineage bifurcation. Immunity. 2006;25:105–116. doi: 10.1016/j.immuni.2006.05.010. [DOI] [PubMed] [Google Scholar]
- 76.Hodson DJ, Janas ML, Galloway A, Bell SE, Andrews S, Li CM, Pannell R, Siebel CW, MacDonald HR, De Keersmaecker K, et al. Deletion of the RNA-binding proteins ZFP36L1 and ZFP36L2 leads to perturbed thymic development and T lymphoblastic leukemia. Nat Immunol. 2010;11:717–724. doi: 10.1038/ni.1901. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Vogel KU, Bell LS, Galloway A, Ahlfors H, Turner M. The RNA-binding proteins zfp36l1 and zfp36l2 enforce the thymic beta-selection checkpoint by limiting DNA damage response signaling and cell cycle progression. J Immunol. 2016;197:2673–2685. doi: 10.4049/jimmunol.1600854. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Galloway A, Turner M. Cell cycle RNA regulons coordinating early lymphocyte development. Wiley Interdiscip Rev RNA. 2017;8:e1419. doi: 10.1002/wrna.1419. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Salerno F, Engels S, van den Biggelaar M, van Alphen FP, Guislain A, Zhao W, Hodge DL, Bell SE, Medema JP, von Lindern M, et al. Translational repression of pre-formed cytokine-encoding mRNA prevents chronic activation of memory T cells. Nat Immunol. 2018;19:828–837. doi: 10.1038/s41590-018-0155-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Moore MJ, Blachere NE, Fak JJ, Park CY, Sawicka K, Parveen S, Zucker-Scharff I, Moltedo B, Rudensky AY, Darnell RB. ZFP36 RNA-binding proteins restrain T cell activation and anti-viral immunity. Elife. 2018;7:e33057. doi: 10.7554/eLife.33057. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Heissmeyer V, Vogel KU. Molecular control of Tfh-cell differentiation by Roquin family proteins. Immunol Rev. 2013;253:273–289. doi: 10.1111/imr.12056. [DOI] [PubMed] [Google Scholar]
- 82.Vogel KU, Edelmann SL, Jeltsch KM, Bertossi A, Heger K, Heinz GA, Zöller J, Warth SC, Hoefig KP, Lohs C, et al. Roquin paralogs 1 and 2 redundantly repress the Icos and Ox40 costimulator mRNAs and control follicular helper T cell differentiation. Immunity. 2013;38:655–668. doi: 10.1016/j.immuni.2012.12.004. [DOI] [PubMed] [Google Scholar]
- 83.Glasmacher E, Hoefig KP, Vogel KU, Rath N, Du L, Wolf C, Kremmer E, Wang X, Heissmeyer V. Roquin binds inducible costimulator mRNA and effectors of mRNA decay to induce microRNA-independent post-transcriptional repression. Nat Immunol. 2010;11:725–733. doi: 10.1038/ni.1902. [DOI] [PubMed] [Google Scholar]
- 84.Choi YS, Kageyama R, Eto D, Escobar TC, Johnston RJ, Monticelli L, Lao C, Crotty S. ICOS receptor instructs T follicular helper cell versus effector cell differentiation via induction of the transcriptional repressor Bcl6. Immunity. 2011;34:932–946. doi: 10.1016/j.immuni.2011.03.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Vinuesa CG, Cook MC, Angelucci C, Athanasopoulos V, Rui L, Hill KM, Yu D, Domaschenz H, Whittle B, Lambe T, et al. A RING-type ubiquitin ligase family member required to repress follicular helper T cells and autoimmunity. Nature. 2005;435:452–458. doi: 10.1038/nature03555. [DOI] [PubMed] [Google Scholar]
- 86.Yu D, Tan AH, Hu X, Athanasopoulos V, Simpson N, Silva DG, Hutloff A, Giles KM, Leedman PJ, Lam KP, et al. Roquin represses autoimmunity by limiting inducible T-cell co-stimulator messenger RNA. Nature. 2007;450:299–303. doi: 10.1038/nature06253. [DOI] [PubMed] [Google Scholar]
- 87.Hwang SS, Lim J, Yu Z, Kong P, Sefik E, Xu H, Harman CC, Kim LK, Lee GR, Li HB, et al. mRNA destabilization by BTG1 and BTG2 maintains T cell quiescence. Science. 2020;367:1255–1260. doi: 10.1126/science.aax0194. [DOI] [PubMed] [Google Scholar]
- 88.Mino T, Murakawa Y, Fukao A, Vandenbon A, Wessels HH, Ori D, Uehata T, Tartey S, Akira S, Suzuki Y, et al. Regnase-1 and roquin regulate a common element in inflammatory mRNAs by spatiotemporally distinct mechanisms. Cell. 2015;161:1058–1073. doi: 10.1016/j.cell.2015.04.029. [DOI] [PubMed] [Google Scholar]
- 89.Matsushita K, Takeuchi O, Standley DM, Kumagai Y, Kawagoe T, Miyake T, Satoh T, Kato H, Tsujimura T, Nakamura H, et al. Zc3h12a is an RNase essential for controlling immune responses by regulating mRNA decay. Nature. 2009;458:1185–1190. doi: 10.1038/nature07924. [DOI] [PubMed] [Google Scholar]
- 90.Uehata T, Iwasaki H, Vandenbon A, Matsushita K, Hernandez-Cuellar E, Kuniyoshi K, Satoh T, Mino T, Suzuki Y, Standley DM, et al. Malt1-induced cleavage of regnase-1 in CD4+ helper T cells regulates immune activation. Cell. 2013;153:1036–1049. doi: 10.1016/j.cell.2013.04.034. [DOI] [PubMed] [Google Scholar]
- 91.Kong G, Dou Y, Xiao X, Wang Y, Ming Y, Li XC. Transgenic expression of a mutant ribonuclease regnase-1 in T cells disturbs T cell development and functions. Front Immunol. 2021;12:682220. doi: 10.3389/fimmu.2021.682220. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Jeltsch KM, Hu D, Brenner S, Zöller J, Heinz GA, Nagel D, Vogel KU, Rehage N, Warth SC, Edelmann SL, et al. Cleavage of roquin and regnase-1 by the paracaspase MALT1 releases their cooperatively repressed targets to promote TH17 differentiation. Nat Immunol. 2014;15:1079–1089. doi: 10.1038/ni.3008. [DOI] [PubMed] [Google Scholar]
- 93.Lisy S, Rothamel K, Ascano M. Rna binding proteins as pioneer determinants of infection: protective, proviral, or both? Viruses. 2021;13:2172. doi: 10.3390/v13112172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Berkovits BD, Mayr C. Alternative 3′ UTRs act as scaffolds to regulate membrane protein localization. Nature. 2015;522:363–367. doi: 10.1038/nature14321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Papadaki O, Milatos S, Grammenoudi S, Mukherjee N, Keene JD, Kontoyiannis DL. Control of thymic T cell maturation, deletion and egress by the RNA-binding protein HuR. J Immunol. 2009;182:6779–6788. doi: 10.4049/jimmunol.0900377. [DOI] [PubMed] [Google Scholar]
- 96.DeMicco A, Reich T, Arya R, Rivera-Reyes A, Fisher MR, Bassing CH. Lymphocyte lineage-specific and developmental stage specific mechanisms suppress cyclin D3 expression in response to DNA double strand breaks. Cell Cycle. 2016;15:2882–2894. doi: 10.1080/15384101.2016.1198861. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Techasintana P, Ellis JS, Glascock J, Gubin MM, Ridenhour SE, Magee JD, Hart ML, Yao P, Zhou H, Whitney MS, et al. The RNA-binding protein HuR posttranscriptionally regulates IL-2 homeostasis and CD4+ Th2 differentiation. Immunohorizons. 2017;1:109–123. doi: 10.4049/immunohorizons.1700017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Gubin MM, Techasintana P, Magee JD, Dahm GM, Calaluce R, Martindale JL, Whitney MS, Franklin CL, Besch-Williford C, Hollingsworth JW, et al. Conditional knockout of the RNA-binding protein HuR in CD4+ T cells reveals a gene dosage effect on cytokine production. Mol Med. 2014;20:93–108. doi: 10.2119/molmed.2013.00127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Chen J, Cascio J, Magee JD, Techasintana P, Gubin MM, Dahm GM, Calaluce R, Yu S, Atasoy U. Posttranscriptional gene regulation of IL-17 by the RNA-binding protein HuR is required for initiation of experimental autoimmune encephalomyelitis. J Immunol. 2013;191:5441–5450. doi: 10.4049/jimmunol.1301188. [DOI] [PMC free article] [PubMed] [Google Scholar]