An asymmetrically dimethylarginated nuclear 90 kDa protein (p90aDMA) induced by interleukin (IL)-2, IL-4 or IL-6 in the tumor microenvironment is selectively degraded by autophagy

  • Authors:
    • Lei Sun
    • Wu-Yan Xia
    • Shao-Hua Zhao
    • Ning Liu
    • Shan-Shan Liu
    • Peng Xiu
    • Lin-Feng Li
    • Xue-Lei Cao
    • Jian-Xin Gao
  • View Affiliations

  • Published online on: March 23, 2016     https://doi.org/10.3892/ijo.2016.3450
  • Pages: 2461-2471
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Protein arginine methylation is a common posttranslational modification resulting in the generation of asymmetric dimethylarginine (aDMA) and symmetric dimethylarginine (sDMA). Currently, the regulation of aDMA or sDMA by hypoxia, nutrient stavation or cytokines in the tumor microenvironment remains largely unknown. Here we show that p90aDMA, p70aDMA and p90sDMA, endogenous proteins containing aDMA or sDMA with mass 70 or 90 kDa, were widely and dominantly expressed in breast cancer cell lines. Notably, it was p90aDMA rather than p90sDMA that accumulated in the nucleus upon stimulation of cancer cells with interleukin (IL)-2, IL-4, IL-6 but not IL-8. In addition, the p90aDMA accumulation could be inhibited after treatment with a global methyltrasferase inhibitor, adenosine-2',3'-dialdehyde (AdOx). It seemed that some endogenous proteins in cancer cells were asymmetrically arginine-methylated upon exposure to some cytokines.. Furthermore, endogenous proteins of aDMA, such as p90aDMA and p70aDMA, were degraded in response to hypoxia, nutrient starvation and rapamycin treatment in breast and cervical cancer cells. IL-2/4/6 slightly increased basal autophagy but slightly decreased the rapamycin‑induced autophagy in cancer cells, suggesting that IL-2/4/6 and autophagy inducers play distinct roles in the regulation of aDMA of proteins. Conversely, rapamycin accumulated p90sDMA in MDA-MB‑231 and MCF-7 cells. Taken together, our results add a new dimension to the complexity of arginine methylated regulation in response to various stimuli and provide the first evidence that aDMA serves as one specific degradation signal of selective autophagy.

Introduction

The tumor microenvironment consists of extracellular matrix, stromal cells (for example, endothelial cells, fibroblasts, myofibroblasts and leukocytes), intratumoral hypoxia and nutrient starvation, and cytokines, chemokines and proteins secreted by epithelial, cancer or stromal cells (1,2). Cytokines, chemokines and growth factors, including interleukin (IL) superfamily, form complex immune signaling networks and have important roles in various aspectes of cancer initiation and progression (24). Several cytokines including IL-2, IL-12, TNF-α, type I IFNs and GM-CSF possess anticancer potential via intratumoral delivery (2,4,5). IL-2, which has antitumor functions that include the activation of natural killer (NK) and cytotoxic T cells, can induce durable remissions in 5–10% of patients with metastatic melanoma and renal cell carcinoma, malignancies with poor prognoses (6). During an immune response, cancer cells encounter extrinsic and intrinsic factors, including oxidative stress, nutrient availability, and inflammation, that can modulate their capacity to activate, proliferate, and survive.

Autophagy occurs at a constitutive basal level, but it can be enhanced in response to various types of stress, mainly including oxygen tension, nutrient deprivation and chemicals such as rapamycin, all of which can inhibit mechanistic target of rapamycin (mTOR) pathway and thus initiate autophagy (79). Steps in the autophagy pathway involve nucleation of targeted macromolecules on the ER membrane, trafficking of autophagosomes to lysosomes and, finally, fusion of the autophagosome-lysosome, resulting in targeted protein degradation (10,11). This process is controlled by the products of numerous autophagy-specific genes (ATGs). A key regulator of autophagy is microtubule-associated protein 1 light chain 3 (LC3, a mammalian homolog of yeast Atg8), which controls major steps in the autophagic pathway including the growth of autophagic membranes, recognition of autophagic cargoes, and the fusion of autophagosomes with lysosomes (12). Recent studies demonstrate that the phosphorylation of LC3 at threonine 50 (Thr50) plays a critical role in mediating fusion of autophagosomes with lysosomes (13).

Increasing evidence shows that the posttranslational modifications (PTMs), including phosphorylation, acetylation, methylation, and ubiquitination, are important for regulating the autophagy process by providing structural and functional diversity among proteins (12,1416). The majority of research is dedicated to the PTMs of key autophagy-related molecules containing autophagy receptors, providing a layer of regulation for the specificity and efficiency of selective autophagy. Selective autophagy refers to the selective degradation of, for instance, organelles (mitophagy and pexophagy), bacteria (xenophagy), ribosomes, macromolecular structures, specific proteins and protein aggregates (aggrephagy) by autophagy (17). Optineurin, an autophagy receptor, is phosphorylated by the protein kinase TBK1 (TANK binding kinase 1) at serine 177, which enhances the LC3 binding affinity and autophagic clearance of cytosolic Salmonella enterica (18). However, little is known about the PTMs of substrates determining their specific recognition by autophagy receptors. NDP52, which is an autophagy adaptor that contains an LC3-interacting region (LIR) motif (17), is induced by Nrf2 and specifically directs phosphorylated tau to the autophagic degradative pathway (19). Mad1, a member of the Myc/Max/Mad family, can also be phosphorylated at serine 145 and introduced into the autophagic degradation (20). Interestingly, EPG-11/PRMT-1 directly methylates arginines in the RGG domains of PGL-1 and PGL-3 and promotes their autophagic removal in C. elegans during embryogenesis (21). However, the link between arginine methylation and selective autophagy has not been clearly demonstrated in cancer.

Arginine methylation is one common PTM mainly of nuclear proteins in eukaryotic cells, and is catalyzed by a family of enzymes termed protein arginine methyltransferases (PRMTs) (22,23). Three main forms of methylarginine have been identified in eukaryotes: NG-monomethylarginine (MMA), NGNG (asymmetric) dimethylarginine (aDMA), and NGNG (symmetric) dimethylarginine (sDMA) (24). In humans, PRMTs are classified into type I (PRMT1, PRMT2, PRMT3, PRMT4 and PRMT6), type II (PRMT5 and PRMT7) and type III (PRMT7) methyltransferases, based on their corresponding aDMA, sDMA and MMA activities, respectively. PRMT1 and PRMT5 are the major asymmetric and symmetric arginine methyltransferases, respectively (25). Arginine methylation has received increasing attention over the last years as several recent reports have illustrated a novel role for this posttranslational modification in regulating protein-protein interaction and transcriptional induction (26,27), and is often deregulated in cancer (25). However, how arginine methylation could be regulated by interleukins or autophagy in the context of the tumor microenvironment has not yet been investigated.

In this study, we identified for the first time that p90aDMA, which is a 90-kDa protein of aDMA in the nucleus and accumulated by IL-2, IL-4 and IL-6, can serve as a unique substrate for selective autophagy. Conversely, p90sDMA was a 90-kDa protein of sDMA and accumulated in a dose-dependent manner in response to rapamycin treatment. Taken together, our study provides evidence for immunity regulation through crosstalk between arginine methylation and selective autophagy in the tumor microenvironment by using in vitro models.

Materials and methods

Chemicals and reagents

Recombinant human IL-2, IL-4, IL-6 and IL-8 were purchased from Peprotech (NJ, USA). They were dissolved in water to a concentration of 5 μg/ml. 3-Methyladenine (3-MA) was purchased from Sigma (MO, USA) using as an inhibitor of autophagy. 3-MA (100 mg) was dissolved in phosphate-buffered saline (PBS) to make a 100-mM stock solution. Adenosine-2,3 dialdehyde (AdOx) was purchased from Sigma and used as an inhibitor of methyltransferase. AdOx (5 mg) was dissolved in 0.2 M HCl to make a 10-mM stock solution. Rapamycin was also purchased from Sigma.

Cell culture

Human cancer cell lines including MDA-MB-231, MDA-MB-468, MCF-7, SKBR3, T47D and HeLa cells were obtained from American Type Tissue Culture Collection (ATCC, USA). The cells were grown in Dulbecco's modified Eagle's medium (DMEM, Gibco, CA, USA) containing 10% fetal bovine serum (FBS, CA, USA) and 100 U/ml penicillin-streptomycin (Gibco) in a humidified incubator of 5% CO2 at 37°C. The standard hypoxic conditions were 1% O2 and 5% CO2. Hypoxia was done in a multi-gas incubator chamber with a compact gas oxygen controller (MCO-5M, Sanyo, Osaka-SHI, Japan) to maintain oxygen concentration at 1% by injecting a gas mixture of 95% N2 and 5% CO2.

Small interfering RNA (siRNA) transfection

siRNA targeting ATG5 (5′-CCAUCAAUCGGAAACUCAUTT-3′) and negative control siRNA (5′-UUCUCCGAACGUGUCACGUTT-3′), which was used for normalisation, were synthesised by Genepharma (Shang Hai, China). For transfection, MCF-7 cells were seeded in 6-well plates at a density of 2×105 cells/well. After 24 h, cells were transfected with siRNA (50 pmol) using Lipofectamine® 2000 Reagent (Invitrogen, CA, USA) according to the instructions of the manufacturer. After 24 h of transfection, cells were treated with 100 nM rapamycin for another 24 h. Then the cells were harvested and the knockdown of ATG5 was confirmed by western blot analysis.

Western blot analysis

Monolayer cultures of respective cell lines at an 80–90% confluence were prepared with pre-cold RIPA lysis and extraction buffer (Thermo Scientific, CA, USA) containing protease inhibitor cocktail (Roche, Basel, Switzerland) on ice. The total cell lysate was centrifuged and the supernatant was denatured by boiling. Protein concentrations of supernatants were analyzed by bicinchoninic acid (BCA) assay kit (Beyotime, Nan Tong, China). Equivalent amounts of total proteins (20 μg) were subjected to 8–12% sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and then transferred to a 0.45-μM PVDF membrane (Millipore, MD, USA). The membranes were blocked for 2 h in 5% bovine serum albumin (BSA) at room temperature and incubated with specific primary antibodies at 4°C overnight. A list of the primary antibodies used for western blot analysis are characterized in Table I. Further incubation was performed with the corresponding horseradish peroxidase-coupled secondary antibodies (1:10,000, cat. nos. sc-2004 and sc-2005; Santa Cruz, CA, USA) at room temperature for 2 h. Then the bands were detected using Super Signal® West Pico Chemiluminescent Substrate kit (Thermo Scientific), and the results were recorded using the ChemiDox™ XRS+ system. Relative protein expression was normalized with β-actin.

Table I

Primary antibodies used in western blot analysis.

Table I

Primary antibodies used in western blot analysis.

TargetSourceHostDilutionCatalog
SYM11Merck Millipore, MA, USARabbit1:1,00007-413
ASYM24Merck Millipore, MA, USARabbit1:50007-414
LC3BCell Signaling Technology, Inc. (CST), MA, USARabbit1:1,000#3868
ATG5Cell Signaling Technology, Inc. (CST), MA, USARabbit1:1,000#12994
TBPProteintech, Wu Han, ChinaRabbit1:50022006-1-AP
β-actinZen Bioscience, Cheng Du, ChinaMouse1:20,00070068
Nuclear cytoplasmic fractionation

Nuclear and cytoplasmic extracts were prepared using NE-PER Nuclear and Cytoplasmic Extraction reagents (Pierce, Thermo Fisher Scientific, CA, USA). The quality of nuclear and cytoplasmic extracts was verified by immunoblotting with protein differentially enriched in the nucleus (TBP) or the cytoplasm (β-actin).

Results

Comparison of basal sDMA or aDMA levels in breast cancer cell lines

Breast cancer is a heterogeneous group of diseases with different histological prognostic and clinical aspects (28). To investigate the preference for the substrate methylation state, we collected the lysates of five human breast cancer cell lines and evaluated the basal aDMA and sDMA using immublotting. Endogenous proteins of aDMA and sDMA can be specifically recognized by antibodies ASYM24 and SYM11, respectively. Both SYM11 and ASYM24 antibodies recognized unknown endogenous proteins containing methylated arginine in breast cancer cells with molecular mass of 90, 70, 55 and 34 kDa. Proteins of 90 and 70 kDa recognized by SYM11 were termed p90sDMA and p70sDMA. Proteins of 90, 70 and 34 kDa recognized by ASYM24 were termed p90aDMA, p70aDMA and p34aDMA. p90sDMA, p90aDMA and p70aDMA were widely and dominantly expressed in breast cancer cells but at relatively higher levels in MCF-7 cells and lower levels in the MDA-MB-231 cells (Fig. 1). Interestingly, the p90sDMA and p70aDMA proteins were predominantly expressed in breast cancer cells at relatively higher levels in the MCF-7, MDA-MB-468, T47D and SKBR3 cells but lower levels in the MDA-MB-231 cells (Fig. 1). In contrast, p90aDMA and p70sDMA proteins were constitutively expressed at a lower level in the breast cancer cell lines (Fig. 1). As a result, the ratio of p90sDMA to p70sDMA was higher than the ratio of p90aDMA to p70aDMA in each line (Fig. 1).

Expression of p90aDMA is specifically enhanced by IL-2, IL-4 or IL-6 but not by IL-8 in cancer cells

To determine whether the cancer immune microenvironment enriched with cytokines could mobilize arginine methylation, we treated MDA-MB-231 cells with IL-2, IL-4, IL-6 or IL-8 for 24 h because of their action on cancer cell proliferation (29). As a result, IL-2, IL-4, IL-6 but not IL-8 specifically enhanced expression of p90aDMA in the MDA-MB-231 cells (Fig. 2C and D). In accordance with Fig. 2B and D, we observed that 90aDMA was increased in a dose-dependent manner upon simulation with IL-2/IL-6 in MDA-MB-231 cells (Fig. 3A and B). However, these cytokines had no effect on the expressions of p90sDMA, p70sDMA and p70aDMA (Fig. 2).

To confirm that the methylation signals detected by the ASYM24 antibody were specific, similar experiments were performed in IL-2 or IL-6 treated MDA-MB-231 cells incubated with or without adenosine-2′, 3′-dialdehyde (AdOx), a methyltransferase inhibitor. It was found that basal or IL-2/6 induced aDMA was almost blocked in the presence of AdOx (Fig. 3C and D). Of note, IL-2 or IL-6 did not alter the expression of p70aDMA (Fig. 3C and D). Therefore, it could be considered that IL-2 and IL-6 specifically induced the accumulation of p90aDMA in MDA-MB-231 cells. The reason for the specific increase in p90aDMA is unclear.

Rapamycin specifically inhibits p90aDMA expression in cancer cells independent of cytokine stimulation

Arginine methylation has been considered to be an irreversible post-translational modification until recently. However, a recent study suggested that autophagy may regulate arginine methylation (30). Then, rapamycin, an autophagy enhancer, was administrated in control or IL-2/4/6/8 treated MDA-MB-231 cells. It was the aDMA levels rather than the sDMA levels that were significantly reduced regardless of the interleukin stimulation (Fig. 2). On the contrary, p90sDMA was increased in rapamycin treated MDA-MB-231 cells (Fig. 2A and C). These results drew our attention to the possibility that aDMA serves as a novel and specific signaling molecule that provokes selective autophagic degradation.

To determine if the increased nuclear p90aDMA resulting from IL-2, IL-4 or IL-6 exposure could be due to autophagy inhibition, we monitored whether it affects the ratio of LC3-II/I, a canonical hallmark of autophagy. MDA-MB-231 cells were exposed to IL-2, IL-4, IL-6 or IL-8 for 24 h, lysed and subjected to western blotting for detecting the abundance of autophagic markers. As shown in Fig. 4A and B, IL-2, IL-4, IL-6 but not IL-8 slightly increased the ratio of LC3-II/I. To determine the dose curve of IL-2/IL-6 exposure for the expression of autophagic markers, MDA-MB-231 cells were exposed to varying doses of IL-2 or IL-6 (5, 25, 100 and 500 μM) for 24 h and assessed for expression of autophagic markers by western blotting. The ratio of LC3-II/I exhibited a similar dose-dependent trend in response to IL-2 or IL-6 with a slight increase of 20% at 25 ng/ml IL-2 and 100 ng/ml IL-6 exposure, respectively (Fig. 4C and D). We next determined whether IL-2, IL-4, IL-6 or IL-8 affects autophagy induction. We monitored rapamycin-induced autophagy by assessing conversion of LC3-I to LC3-II. IL-2, IL-4, IL-6 but not IL-8 slightly decreased conversion of LC3-I to LC3-II induced by rapamycin (Fig. 4A and B). Taken together, these findings demonstrated that IL-2, IL-4, IL-6 but not IL-8 slightly increased the basal autophagy but slightly decreased the rapamycin-induced autophagy in MDA-MB-231 cells, indicating that IL-2/IL-4/IL-6 accumulated p90aDMA in an autophagy-independent pathway.

The aDMA proteins are localized in the nucleus of cancer cells

Extensive studies have focused on autophagic turnover of cytoplasmic materials, little is known about the role of autophagy in degrading nuclear components (31). To determine the cellular localization of proteins containing aDMA, we examined p90aDMA and p70aDMA in the cytoplasm and nucleus through cellular fractionation experiments followed by western blot analysis in the IL-2 or IL-6 treated MDA-MB-231 cells, and found that both p90aDMA and p70aDMA distributed and accumulated predominantly in the nucleus regardless of cytokine stimulation, suggesting that the aDMA proteins were constitutively localized in the nucleus and might play a critical role (Fig. 5A). To monitor cytoplasmic contamination of the nuclear extracts, immunoblotting was performed using the anti-β-actin antibody and TBP (TATA box binding protein) antibody as the internal controls. The dramatic reduction of β-actin or the presence of TBP only in the nuclear fractions confirmed that cytoplasmic contamination did not occur (Fig. 5A).

Hypoxia and nutrient starvation disablizes nuclear p90aDMA and p70aDMA in cancer cells

Tumor cells are continually subjected to diverse stress conditions of the tumor microenvironment, including hypoxia, nutrient deprivation, oxidative or genotoxic stress (1). Among the stresses, hypoxia or starvation is a classical autophagy inducing stimulus (32,33). As shown in Fig. 5, two WB bands representing 70 kDa (p70aDMA) and 90 kDa (p90aDMA) nuclear-localized proteins, respectively, were detected in breast cancer MDA-MB-231 cells and cervical cancer HeLa cells. In accordance with the deacetylated nuclear LC3 being transported into the cytoplasm to carry out PE conjugation to pre-autophagic membranes (34), we observed that LC3 in the nucleus vanished under hypoxia or starvation (Fig. 5C and D) and aDMA proteins dominantly expressed in the nucleus were diminished synchronously (Fig. 5B, C and D).

As is well known, LC3 proteins play a key role in the selective recruitment of autophagic cargoes into autophagosomes, and serve as docking sites for adaptor proteins (12,31). Therefore, it is conceivable that proteins of aDMA including p90aDMA and p70aDMA as LC3 cargo substrates were translocated from the nucleus to the cytoplasm and tethered to the site of engulfing autophagosomes. The hypothesis needs further investigation under various stress conditions in our future work.

Autophagy inhibition reverses the degradation of p90aDMA and p70aDMA proteins in cancer cells

We first examined the effects of rapamycin treatment on LC3 protein levels in MCF-7 cells. Immunoblot analyses showed a concentration-dependent increase in the ratio of LC3-II/I, representing mounting activation of autophagy (Fig. 6A). Concomitantly with the activation of autophagy, a 90-kDa protein (p90sDMA) showed a marked dose-dependent increase in sDMA levels (Fig. 6B). Whereas the effects of rapamycin treatment on cellular aDMA in MCF-7 cells were tested by immunoblot using ASYM24 antibody, and exhibited to a concentration-dependent decrease in the intensity of multiple bands, including p90aDMA, p70aDMA and p34aDMA (Fig. 6C).

To determine if p90aDMA and p70aDMA degradation in rapamycin-treated MCF-7 cells was mediated by autophagy, we blocked the early stage of autophagy with 3-MA, a class III PI3K inhibitor (35), and examined its effect on p90aDMA and p70aDMA expression levels. One-hour pretreatment with 3-MA effectively blocked the rapamycin-induced decrease in the expression of p90aDMA and p70aDMA (Fig. 6D). Similarly, siRNA-mediated knockdown of ATG5 prevented the rapamycin mediated decrease in p90aDMA and p70aDMA expression levels, whereas the non-targeting control siRNA had no effect (Fig. 6E). Therefore, we conclude that rapamycin triggers p90aDMA and p70aDMA degradation through the autophagy pathway and aDMA serves as a specific degradation signal for autophagy.

Discussion

We demonstrate for the first time that IL-2, IL-4 and IL-6 specifically promote nuclear accumulation of p90aDMA, which is abrogated by AdOx, confirming that IL-2/4/6 treatment can effectively increase asymmetric dimethylarginine proteins (Figs. 2, 3 and 5A). In addition, the interleukins slightly increased basal autophagy but had an opposite effect on rapamycin-induced autophagy, which may decrease p90aDMA expression (Figs. 3 and 4). It can be concluded that IL-2/4/6 stimulated p90aDMA expression is not caused by inhibiting autophagy. Moreover, indirect evidence suggests that p90aDMA, p70aDMA and even p34aDMA could be degraded by LC3-mediated selective autophagy in response to hypoxia, starvation and rapamycin (Figs. 2B and D, 5B–D and 6C), whereas rapamycin can accumulate p90sDMA in a dose-dependent manner (Fig. 6B). It is obvious that the aDMA induced by IL-2, IL-4 or IL-6 determines a highly processive initial reaction on the substrate for specific autophagic degradation. Our findings reveal a novel role for selective autophagy in the regulation of immunologic responses and highlight the role of the posttranslational modification of aDMA in controlling nuclear protein aggregation induced by interleukins and selective autophagy of ubiquitinated proteins (Fig. 7). One fundamental question not addressed here is which endogenous proteins are methylated at arginine sites and translocated into the nucleus in response to IL-2/4/6 treatment.

Our results raise the fundamental questions how IL-2/IL-4/IL-6 regulates the biogenesis of p90aDMA and whether aDMA takes place in the cytoplasm or the nucleus of cancer cells (Figs. 2B and D and 3A and B). Protein arginine methylation is a common posttranslational modification in higher eukaryotes, but its precise role in providing structural and functional diversity among proteins is not well understood. Arginine methylation has been shown to affect several cellular processes, including intracellular localization, protein-protein interactions as well as transcription (36,37). Signal transducers and activators of transcription (STAT) proteins, for example, are a family of latent cytoplasmic transcription factors which mediate interferons (IFNs), interleukins, and some growth factors and peptide hormone signaling in cells (38). Once tyrosine phosphorylated, STAT proteins form homo- or heterodimers, which are actively imported into the nucleus and bind to DNA consensus motifs to elicit specific transcriptional responses (3840). However, arginine 31 methylation of STAT1 enhances its DNA binding by reducing association with the specific inhibitor PIAS1, thus intensifying the growth-restraining activities of the interferons (41). In addition, arginine methylation of STAT1 controls the rate of STAT1 dephosphorylation by modulating its interaction with PIAS1 and the nuclear tyrosine phosphatase TcPTP (42). STAT1 and STAT2 have a structural arginine/lysine-rich element involved in IFN-induced nuclear import (43). Considering: i) interleukins mainly activate STAT signaling pathways (39,40), and ii) p90aDMA derived from endogenous proteins has a similar molecular mass to the 90 kDa and nuclear location of activated STAT proteins (39,40), it can be deduced that p90aDMA may be one or several proteins of the STATs family that could be dimethylated by IL-2/4/6 commonly or differentially and promote its or their translocation into the nucleus. Further research involves the use of ASYM24 antibody to proteins of 90 kDa containing aDMA for immunoprecipitation (IP) experiments. The purified proteins are then digested and subjected to immublotting or mass spectrometric analysis to confirm our hypothesis.

Selective autophagy is a degradative pathway that controls the quality and abundance of proteins and cellular organelles and is mediated by autophagy receptors that simultaneously bind the designated target and LC3/GABARAP proteins on autophagosomal membranes (11). The autophagic/signaling adaptor LC3 is known to exert its functions through multiple domains containing a ubiquitin core with two α helices, α1 and α2 attached at its N-terminus, which is utilized as the interaction site with its target proteins (44). Physical linkages between autophagy adaptor proteins via polyubiquitin chains are required for autophagy flux. A recent research demonstrates that the deacetylated nuclear LC3 is transported into the cytoplasm to carry out PE conjugation to pre-autophagic membranes by sequential interaction with Atg7 and Atg3 (34). Especially, nuclear lamina protein lamin B1 degradation is achieved by nucleus to cytoplasm transport degradation that delivers lamin B1 to the lysosome via LC3-lamin B1 interaction in the nucleus (31). The reduction in p90aDMA, p70aDMA and p34aDMA levels implicates the specificity and apparent affinity of aDMA for autophagy receptors (Figs. 5B–D and 6C–E). Based on the above reports and results that p90aDMA and p70aDMA are dominantly expressed in the nucleus, we may deduce that selective autophagy via LC3 is required for the translocation to the autophagosomes and degradation of p90aDMA and p70aDMA. Once autophagy is stimulated in response to stress such as hypoxia, starvation or rapamycin, proteins undergoing aDMA can be transported to autophagosomes by deacetylated LC3 and suffer from autophagic degradation specifically. Based on our findings that p90aDMA and p70aDMA, dominantly expressed in the nucleus, are recognized and bound by LC3 translocating to the cytoplasm (Figs. 2, 3, 5 and 6A and C), we conclude that aDMA as a signal is required for LC3-mediatad selective autophagy traffic.

As a stress integrator pathway, autophagy is a major mechanism that mediates protein and organelle degradation in response to external and internal signals. Additionally, autophagy has been verified in different contexts to regulate immune responses to various stimuli (4550). In addition to the role of p62, NDP52 and optineurin as adaptors in selective autophagy, these proteins have also recently been shown to regulate innate immunity signaling pathways and, thus, were suggested to represent a new class of pattern recognition receptors, the sequestosome-1-like receptors (SLRs) (51,52). When IL-2 and IL-6 accumulate p90aDMA, which may be a STAT protein, to activate some signaling pathways or transcription, external stresses such as hypoxia, starvation or rapamycin antagonize the signaling transduction via autophagic degradation of p90aDMA (Fig. 2B and D). A growing body of evidence indicates that similar elimination of signaling molecules play key roles in autophagy-regulated immune responses (5357). For instance, microglial autophagy plays an important role in the clearance of extracellular Aβ (β-amyloid) fibrils and the regulation of Aβ-induced inflammation, thereby affecting neuronal viability (53). One the other hand, emerging data have suggested that additional mechanisms involved in cancer-related inflammation (CRI) are induction of angiogenesis, metastasis, invasion of surrounding tissues and genetic instability by inflammatory mediators, leading to accumulation of random genetic alterations in cancer cells (29,58,59). Based on the above, the possibility is raised that a negative feedback via the targeted regulation of p90aDMA is established in our study bridging immune microenvironment and selective autophagy that may have a potentially pivotal role in shaping the oncogenesis, immunogenic cell death and even heterogeneity in response to dynamic changes in a cancer cell metabolic, environmental, or developmental status.

In conclusion, our data support that a model for the link between arginine methylation and selective autophagy in the immune microenvironment could be proposed for further investigation. Additionally, it remains unclear how aDMA (or sDMA) is regulated by interleukins (or rapamycin) and their function need to be further explored. Collectively, our study expands what is known about the tumor microenvironment and supports the idea of the regulation of arginine methylation as a new immune-therapeutic method in the future.

Acknowledgements

This study was supported by the National Natural Science Foundation of China (J.X.G., nos. 81171940 and 81372188; L.F.L., no. 81402287); Science and Technology Support Program, Science and Technology Commission of Shanghai Municipality (12431900704 to J.X.G.); the Special Fund for Innovation and Development of Science and Technology and Cultivation Fund for Major Projects and Innovative Team (J.X.G., 2014), Shanghai Jiao Tong University, China; the State Key Laboratory of Oncogenes and Related Genes in China (J.X.G., no. 90-14-06); the University Doctorate Research Fund for Freshly Recruited Teachers (L.F.L., no. 20130073120010), Ministry of National Education, China; and Startup Funds (J.X.G.) from Renji Hospital and School of Medicine, Shanghai Jiao Tong University, China; the Fund for Key Disciplines and Specialties, Shanghai Health and Family Planning Committee, China (J.X.G.), and Shandong Outstanding Young and Middle-aged Scientists Research Award Fund (2014BSE27021 to Shao-Hua Zhao).

Abbreviations:

IL

interleukin

ATG

autophagy-related gene

LC3

microtubule-associated protein 1 light chain 3

PTMs

posttranslational modifications

sDMA

symmetric dimethylarginine

aDMA

asymmetric dimethylarginine

AdOx

adenosine-2′,3′-dialdehyde

HBSS

Hank′s balanced salt solution

References

1 

Semenza GL: The hypoxic tumor microenvironment: A driving force for breast cancer progression. Biochim Biophys Acta. S0167-4889(15)00192-5. 2015.PubMed/NCBI

2 

Johansson A, Hamzah J and Ganss R: More than a scaffold: Stromal modulation of tumor immunity. Biochim Biophys Acta. S0304-419X(15)00044-X. 2015.PubMed/NCBI

3 

Pitt LA, Tikhonova AN, Hu H, Trimarchi T, King B, Gong Y, Sanchez-Martin M, Tsirigos A, Littman DR, Ferrando AA, et al: CXCL12-producing vascular endothelial niches control acute T cell leukemia maintenance. Cancer Cell. 27:755–768. 2015. View Article : Google Scholar : PubMed/NCBI

4 

Dranoff G: Cytokines in cancer pathogenesis and cancer therapy. Nat Rev Cancer. 4:11–22. 2004. View Article : Google Scholar : PubMed/NCBI

5 

Van der Jeught K, Bialkowski L, Daszkiewicz L, Broos K, Goyvaerts C, Renmans D, Van Lint S, Heirman C, Thielemans K and Breckpot K: Targeting the tumor microenvironment to enhance antitumor immune responses. Oncotarget. 6:1359–1381. 2015. View Article : Google Scholar : PubMed/NCBI

6 

Rosenberg SA: IL-2: The first effective immunotherapy for human cancer. J Immunol. 192:5451–5458. 2014. View Article : Google Scholar : PubMed/NCBI

7 

Blagosklonny MV: Hypoxia, MTOR and autophagy: Converging on senescence or quiescence. Autophagy. 9:260–262. 2013. View Article : Google Scholar :

8 

Kim YC and Guan K-L: mTOR: A pharmacologic target for autophagy regulation. J Clin Invest. 125:25–32. 2015. View Article : Google Scholar : PubMed/NCBI

9 

Sun L, Li T, Wei Q, Zhang Y, Jia X, Wan Z and Han L: Upregulation of BNIP3 mediated by ERK/HIF-1α pathway induces autophagy and contributes to anoikis resistance of hepatocellular carcinoma cells. Future Oncol. 10:1387–1398. 2014. View Article : Google Scholar : PubMed/NCBI

10 

Mochida K, Oikawa Y, Kimura Y, Kirisako H, Hirano H, Ohsumi Y and Nakatogawa H: Receptor-mediated selective autophagy degrades the endoplasmic reticulum and the nucleus. Nature. 522:359–362. 2015. View Article : Google Scholar : PubMed/NCBI

11 

Stolz A, Ernst A and Dikic I: Cargo recognition and trafficking in selective autophagy. Nat Cell Biol. 16:495–501. 2014. View Article : Google Scholar : PubMed/NCBI

12 

Wild P, McEwan DG and Dikic I: The LC3 interactome at a glance. J Cell Sci. 127:3–9. 2014. View Article : Google Scholar

13 

Wilkinson DS, Jariwala JS, Anderson E, Mitra K, Meisenhelder J, Chang JT, Ideker T, Hunter T, Nizet V, Dillin A, et al: Phosphorylation of LC3 by the Hippo kinases STK3/STK4 is essential for autophagy. Mol Cell. 57:55–68. 2015. View Article : Google Scholar :

14 

Xie Y, Kang R, Sun X, Zhong M, Huang J, Klionsky DJ and Tang D: Posttranslational modification of autophagy-related proteins in macroautophagy. Autophagy. 11:28–45. 2015. View Article : Google Scholar :

15 

Hunter T: The age of crosstalk: Phosphorylation, ubiquitination, and beyond. Mol Cell. 28:730–738. 2007. View Article : Google Scholar : PubMed/NCBI

16 

Lazarou M, Sliter DA, Kane LA, Sarraf SA, Wang C, Burman JL, Sideris DP, Fogel AI and Youle RJ: The ubiquitin kinase PINK1 recruits autophagy receptors to induce mitophagy. Nature. 524:309–314. 2015. View Article : Google Scholar : PubMed/NCBI

17 

Birgisdottir AB, Lamark T and Johansen T: The LIR motif - crucial for selective autophagy. J Cell Sci. 126:3237–3247. 2013.PubMed/NCBI

18 

Wild P, Farhan H, McEwan DG, Wagner S, Rogov VV, Brady NR, Richter B, Korac J, Waidmann O, Choudhary C, et al: Phosphorylation of the autophagy receptor optineurin restricts Salmonella growth. Science. 333:228–233. 2011. View Article : Google Scholar : PubMed/NCBI

19 

Jo C, Gundemir S, Pritchard S, Jin YN, Rahman I and Johnson GV: Nrf2 reduces levels of phosphorylated tau protein by inducing autophagy adaptor protein NDP52. Nat Commun. 5:34962014. View Article : Google Scholar : PubMed/NCBI

20 

Zhu J, Blenis J and Yuan J: Activation of PI3K/Akt and MAPK pathways regulates Myc-mediated transcription by phosphorylating and promoting the degradation of Mad1. Proc Natl Acad Sci USA. 105:6584–6589. 2008. View Article : Google Scholar : PubMed/NCBI

21 

Li S, Yang P, Tian E and Zhang H: Arginine methylation modulates autophagic degradation of PGL granules in C. elegans. Mol Cell. 52:421–433. 2013. View Article : Google Scholar : PubMed/NCBI

22 

Pahlich S, Zakaryan RP and Gehring H: Protein arginine methylation: Cellular functions and methods of analysis. Biochim Biophys Acta. 1764:1890–1903. 2006. View Article : Google Scholar : PubMed/NCBI

23 

Yagoub D, Hart-Smith G, Moecking J, Erce MA and Wilkins MR: Yeast proteins Gar1p, Nop1p, Npl3p, Nsr1p, and Rps2p are natively methylated and are substrates of the arginine methyltransferase Hmt1p. Proteomics. 15:3209–3218. 2015. View Article : Google Scholar : PubMed/NCBI

24 

McBride AE and Silver PA: State of the arg: Protein methylation at arginine comes of age. Cell. 106:5–8. 2001. View Article : Google Scholar : PubMed/NCBI

25 

Yang Y and Bedford MT: Protein arginine methyltransferases and cancer. Nat Rev Cancer. 13:37–50. 2013. View Article : Google Scholar

26 

Bedford MT and Clarke SG: Protein arginine methylation in mammals: Who, what, and why. Mol Cell. 33:1–13. 2009. View Article : Google Scholar : PubMed/NCBI

27 

Gao WW, Xiao RQ, Peng BL, Xu HT, Shen HF, Huang MF, Shi TT, Yi J, Zhang WJ, Wu XN, et al: Arginine methylation of HSP70 regulates retinoid acid-mediated RARβ2 gene activation. Proc Natl Acad Sci USA. 112:E3327–E3336. 2015. View Article : Google Scholar

28 

Kao J, Salari K, Bocanegra M, Choi YL, Girard L, Gandhi J, Kwei KA, Hernandez-Boussard T, Wang P, Gazdar AF, et al: Molecular profiling of breast cancer cell lines defines relevant tumor models and provides a resource for cancer gene discovery. PLoS One. 4:e61462009. View Article : Google Scholar : PubMed/NCBI

29 

Lin WW and Karin M: A cytokine-mediated link between innate immunity, inflammation, and cancer. J Clin Invest. 117:1175–1183. 2007. View Article : Google Scholar : PubMed/NCBI

30 

Shirakawa T, Kako K, Shimada T, Nagashima Y, Nakamura A, Ishida J and Fukamizu A: Production of free methylarginines via the proteasome and autophagy pathways in cultured cells. Mol Med Rep. 4:615–620. 2011.PubMed/NCBI

31 

Dou Z, Xu C, Donahue G, Shimi T, Pan JA, Zhu J, Ivanov A, Capell BC, Drake AM, Shah PP, et al: Autophagy mediates degradation of nuclear lamina. Nature. 527:105–109. 2015. View Article : Google Scholar : PubMed/NCBI

32 

Klionsky DJ, Abdalla FC, Abeliovich H, Abraham RT, Acevedo-Arozena A, Adeli K, Agholme L, Agnello M, Agostinis P, Aguirre-Ghiso JA, et al: Guidelines for the use and interpretation of assays for monitoring autophagy. Autophagy. 8:445–544. 2012. View Article : Google Scholar : PubMed/NCBI

33 

Sun L, Liu N, Liu S, Xia W, Liu M, Li L and Gao J: Beclin-1-independent autophagy mediates programmed cancer cell death through interplays with endoplasmic reticulum and/or mitochondria in colbat chloride-induced hypoxia. Am J Cancer Res. 5:2626–2642. 2015.PubMed/NCBI

34 

Huang R, Xu Y, Wan W, Shou X, Qian J, You Z, Liu B, Chang C, Zhou T, Lippincott-Schwartz J, et al: Deacetylation of nuclear LC3 drives autophagy initiation under starvation. Mol Cell. 57:456–466. 2015. View Article : Google Scholar : PubMed/NCBI

35 

Blommaart EF, Krause U, Schellens JP, Vreeling-Sindelarova H and Meijer AJ: The phosphatidylinositol 3-kinase inhibitors wortmannin and LY294002 inhibit autophagy in isolated rat hepatocytes. Eur J Biochem. 243:240–246. 1997. View Article : Google Scholar : PubMed/NCBI

36 

Chang B, Chen Y, Zhao Y and Bruick RK: JMJD6 is a histone arginine demethylase. Science. 318:444–447. 2007. View Article : Google Scholar : PubMed/NCBI

37 

Wang H, Huang ZQ, Xia L, Feng Q, Erdjument-Bromage H, Strahl BD, Briggs SD, Allis CD, Wong J, Tempst P, et al: Methylation of histone H4 at arginine 3 facilitating transcriptional activation by nuclear hormone receptor. Science. 293:853–857. 2001. View Article : Google Scholar : PubMed/NCBI

38 

Buchert M, Burns CJ and Ernst M: Targeting JAK kinase in solid tumors: Emerging opportunities and challenges. Oncogene. May 18–2015.(Epub ahead of print). View Article : Google Scholar : 2015. PubMed/NCBI

39 

Bowman T, Garcia R, Turkson J and Jove R: STATs in oncogenesis. Oncogene. 19:2474–2488. 2000. View Article : Google Scholar : PubMed/NCBI

40 

Hirahara K, Onodera A, Villarino AV, Bonelli M, Sciumè G, Laurence A, Sun HW, Brooks SR, Vahedi G, Shih HY, et al: Asymmetric action of STAT transcription factors drives transcriptional outputs and cytokine specificity. Immunity. 42:877–889. 2015. View Article : Google Scholar : PubMed/NCBI

41 

Mowen KA, Tang J, Zhu W, Schurter BT, Shuai K, Herschman HR and David M: Arginine methylation of STAT1 modulates IFNalpha/beta-induced transcription. Cell. 104:731–741. 2001. View Article : Google Scholar : PubMed/NCBI

42 

Zhu W, Mustelin T and David M: Arginine methylation of STAT1 regulates its dephosphorylation by T cell protein tyrosine phosphatase. J Biol Chem. 277:35787–35790. 2002. View Article : Google Scholar : PubMed/NCBI

43 

Melen K, Kinnunen L and Julkunen I: Arginine/lysine-rich structural element is involved in interferon-induced nuclear import of STATs. J Biol Chem. 276:16447–16455. 2001. View Article : Google Scholar : PubMed/NCBI

44 

Sugawara K, Suzuki NN, Fujioka Y, Mizushima N, Ohsumi Y and Inagaki F: The crystal structure of microtubule-associated protein light chain 3, a mammalian homologue of Saccharomyces cerevisiae Atg8. Genes Cells. 9:611–618. 2004. View Article : Google Scholar : PubMed/NCBI

45 

Pei B, Zhao M, Miller BC, Véla JL, Bruinsma MW, Virgin HW and Kronenberg M: Invariant NKT cells require autophagy to coordinate proliferation and survival signals during differentiation. J Immunol. 194:5872–5884. 2015. View Article : Google Scholar : PubMed/NCBI

46 

Kanayama M, He YW and Shinohara ML: The lung is protected from spontaneous inflammation by autophagy in myeloid cells. J Immunol. 194:5465–5471. 2015. View Article : Google Scholar : PubMed/NCBI

47 

Schlie K, Westerback A, DeVorkin L, Hughson LR, Brandon JM, MacPherson S, Gadawski I, Townsend KN, Poon VI, Elrick MA, et al: Survival of effector CD8+ T cells during influenza infection is dependent on autophagy. J Immunol. 194:4277–4286. 2015. View Article : Google Scholar : PubMed/NCBI

48 

Michaud M, Martins I, Sukkurwala AQ, Adjemian S, Ma Y, Pellegatti P, Shen S, Kepp O, Scoazec M, Mignot G, et al: Autophagy-dependent anticancer immune responses induced by chemotherapeutic agents in mice. Science. 334:1573–1577. 2011. View Article : Google Scholar : PubMed/NCBI

49 

Konno H, Konno K and Barber GN: Cyclic dinucleotides trigger ULK1 (ATG1) phosphorylation of STING to prevent sustained innate immune signaling. Cell. 155:688–698. 2013. View Article : Google Scholar : PubMed/NCBI

50 

Guo ML, Liao K, Periyasamy P, Yang L, Cai Y, Callen SE and Buch S: Cocaine mediated microglial activation involves the ER stress-autophagy axis. Autophagy. 11:995–1009. 2015. View Article : Google Scholar

51 

Deretic V: Autophagy as an innate immunity paradigm: Expanding the scope and repertoire of pattern recognition receptors. Curr Opin Immunol. 24:21–31. 2012. View Article : Google Scholar :

52 

Chang KH, Sengupta A, Nayak RC, Duran A, Lee SJ, Pratt RG, Wellendorf AM, Hill SE, Watkins M, Gonzalez-Nieto D, et al: p62 is required for stem cell/progenitor retention through inhibition of IKK/NF-κB/Ccl4 signaling at the bone marrow macrophage-osteoblast niche. Cell Rep. 9:2084–2097. 2014. View Article : Google Scholar : PubMed/NCBI

53 

Cho MH, Cho K, Kang HJ, Jeon EY, Kim HS, Kwon HJ, Kim HM, Kim DH and Yoon SY: Autophagy in microglia degrades extracellular β-amyloid fibrils and regulates the NLRP3 inflammasome. Autophagy. 10:1761–1775. 2014. View Article : Google Scholar : PubMed/NCBI

54 

Liu L, Yang M, Kang R, Dai Y, Yu Y, Gao F, Wang H, Sun X, Li X, Li J, et al: HMGB1-DNA complex-induced autophagy limits AIM2 inflammasome activation through RAGE. Biochem Biophys Res Commun. 450:851–856. 2014. View Article : Google Scholar : PubMed/NCBI

55 

Meunier E, Dick MS, Dreier RF, Schürmann N, Kenzelmann Broz D, Warming S, Roose-Girma M, Bumann D, Kayagaki N, Takeda K, et al: Caspase-11 activation requires lysis of pathogen-containing vacuoles by IFN-induced GTPases. Nature. 509:366–370. 2014. View Article : Google Scholar : PubMed/NCBI

56 

Wildenberg ME, Vos AC, Wolfkamp SC, Duijvestein M, Verhaar AP, Te Velde AA, van den Brink GR and Hommes DW: Autophagy attenuates the adaptive immune response by destabilizing the immunologic synapse. Gastroenterology. 142:1493–1503 e1496. 2012. View Article : Google Scholar : PubMed/NCBI

57 

Lévy J, Cacheux W, Bara MA, L'Hermitte A, Lepage P, Fraudeau M, Trentesaux C, Lemarchand J, Durand A, Crain AM, et al: Intestinal inhibition of Atg7 prevents tumour initiation through a microbiome-influenced immune response and suppresses tumour growth. Nat Cell Biol. 17:1062–1073. 2015. View Article : Google Scholar : PubMed/NCBI

58 

Colotta F, Allavena P, Sica A, Garlanda C and Mantovani A: Cancer-related inflammation, the seventh hallmark of cancer: Links to genetic instability. Carcinogenesis. 30:1073–1081. 2009. View Article : Google Scholar : PubMed/NCBI

59 

Mantovani A, Allavena P, Sica A and Balkwill F: Cancer-related inflammation. Nature. 454:436–444. 2008. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

June-2016
Volume 48 Issue 6

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Sun L, Xia W, Zhao S, Liu N, Liu S, Xiu P, Li L, Cao X and Gao J: An asymmetrically dimethylarginated nuclear 90 kDa protein (p90aDMA) induced by interleukin (IL)-2, IL-4 or IL-6 in the tumor microenvironment is selectively degraded by autophagy. Int J Oncol 48: 2461-2471, 2016
APA
Sun, L., Xia, W., Zhao, S., Liu, N., Liu, S., Xiu, P. ... Gao, J. (2016). An asymmetrically dimethylarginated nuclear 90 kDa protein (p90aDMA) induced by interleukin (IL)-2, IL-4 or IL-6 in the tumor microenvironment is selectively degraded by autophagy. International Journal of Oncology, 48, 2461-2471. https://doi.org/10.3892/ijo.2016.3450
MLA
Sun, L., Xia, W., Zhao, S., Liu, N., Liu, S., Xiu, P., Li, L., Cao, X., Gao, J."An asymmetrically dimethylarginated nuclear 90 kDa protein (p90aDMA) induced by interleukin (IL)-2, IL-4 or IL-6 in the tumor microenvironment is selectively degraded by autophagy". International Journal of Oncology 48.6 (2016): 2461-2471.
Chicago
Sun, L., Xia, W., Zhao, S., Liu, N., Liu, S., Xiu, P., Li, L., Cao, X., Gao, J."An asymmetrically dimethylarginated nuclear 90 kDa protein (p90aDMA) induced by interleukin (IL)-2, IL-4 or IL-6 in the tumor microenvironment is selectively degraded by autophagy". International Journal of Oncology 48, no. 6 (2016): 2461-2471. https://doi.org/10.3892/ijo.2016.3450