IMR Press / FBL / Volume 28 / Issue 11 / DOI: 10.31083/j.fbl2811294
Open Access Original Research
Ubiquitin C-Terminal Hydrolase L5 Plays an Essential Role in the Fly Innate Immune Defense against Bacterial Infection
Show Less
1 School of Life Sciences, Anhui Agricultural University, 230036 Hefei, Anhui, China
2 Faculty of Life Sciences, University of Strasbourg, 67000 Strasbourg, France
3 Zhangzhou Affiliated Hospital of Fujian Medical University, 363000 Zhangzhou, Fujian, China
*Correspondence: jism@ahau.edu.cn (Shanming Ji); yongzhihua1112@163.com (Yongzhi Hua)
These authors contributed equally.
Front. Biosci. (Landmark Ed) 2023, 28(11), 294; https://doi.org/10.31083/j.fbl2811294
Submitted: 21 March 2023 | Revised: 26 June 2023 | Accepted: 7 July 2023 | Published: 23 November 2023
Copyright: © 2023 The Author(s). Published by IMR Press.
This is an open access article under the CC BY 4.0 license.
Abstract

Background: Drosophila ubiquitin carboxy-terminal hydrolase L5 (Uch-L5) functions as a critical component of the 26S proteasome to mediate degradation of polyubiquitinated proteins. It was recently shown to modulate tissue/organ development by targeting the Smoothened protein in the hedgehog pathway. However, whether it plays a role in controlling organismal immune response remains largely unknown. Methods: Reverse transcription plus quantitative polymerase chain reaction (RT-qPCR), dual-luciferase, and Western blot assays were used to explore the potential function of Uch-L5 in the innate immune regulation in cultured Drosophila S2 cells. Further genetic manipulations and bacterial infections were conducted to confirm the findings in vivo. Results: Silencing of Uch-L5 antagonizes the immune deficiency (IMD) but not the Toll innate immune signaling both in vitro and in vivo. Moreover, Uch-L5 positively contributes to the Drosophila innate immune response via its N-terminal Uch domain, which is the catalytical triad executing its deubiquitinase activity. Conclusions: Our studies shed light on a novel function of the deubiquitinase Uch-L5 in governing the anti-microbial defense in Drosophila.

Keywords
Uch-L5
IMD signaling
innate immune response
Drosophila melanogaster
1. Introduction

Innate immunity reacts quickly and efficiently upon recognition of extracellular pathogenic invasion. This constitutes the first line of defense of most organisms [1]. Pattern recognition receptors (PRRs) have been suggested to be mainly responsible for recognizing pathogenic stimuli, in order to trigger downstream signaling transductions, thereby inducing expressions of various immune effectors [2, 3]. Several pioneering studies have demonstrated the pivotal roles of adaptor proteins and correlative modulators in the signal transductions from transmembrane and intracellular receptors to secreted effectors [reviewed in [1, 2, 3, 4, 5, 6]]. Identification and characterization of novel adaptors/modulators has always been one of the hotspots in the basic research of innate immunity.

Drosophila melanogaster (fruit fly) is an excellent animal model for uncovering novel modulators in the innate immune signaling pathways, due to its powerful genetic approaches and worldwide resources of Drosophila mutants and transgenes. The fly innate immune response is mainly governed by two signaling pathways, namely the Toll and the immune deficiency (IMD) pathways [7, 8, 9], which are similar to the mammalian Myd88 (myeloid differentiation factor 88)-dependent TLR (Toll-like receptor) and TNFR (tumor necrosis factor receptor) signaling pathways, respectively. Upon infection by most fungi or Gram-positive bacteria, the Toll receptor-involved signaling is activated in order to trigger the induction of downstream anti-microbial peptides (AMPs), for instance drosomycin (Drs) and metchnikowin (Mtk) [10]. On the other hand, the IMD signaling pathway is mostly activated by Gram-negative bacteria and some types of Gram-positive bacteria, resulting in secretion of another set of AMPs like attacins (Att), cecropin (Cec), and diptericin (Dpt) for the host immune defense [11].

The IMD signaling pathway is normally activated by the binding of bacteria-derived DAP (meso-diaminopimelic acid)-type PGN (peptidoglycan) to the PGN recognition proteins PGRP-LC (located on the cell membrane) and/or PGRP-LE (intracellular) [12]. This binding further leads to the recruitment of the adaptor protein, Imd, to the receptor, together with Fadd (Fas-associated death domain) and the caspase Dredd (death related ced3/nedd2-like caspase) [13]. Dredd participates in the cleavage of Imd and the activation of the downstream kinase complex Tak1 (TGF-β activated kinase 1)/Tab2 (Tak1-associated binding protein 2) [14, 15, 16]. Activated Tak1/Tab2 phosphorylates and activates the IKK (IκB kinase) complex, consisting of Ird5 (immune response deficient 5) and Key (kenny), which in turn phosphorylate the NF-κB (nuclear factor κB) family protein Rel (relish), leading to its cleavage [17, 18, 19, 20]. The cleaved N-terminal active domain of Rel (Rel-68) translocates to the nucleus and activates the transcription of targeted effector genes [20].

Uncontrolled IMD signaling is detrimental to flies [21]. The maintenance of IMD homeostasis requires a series of regulators, among which are the ubiquitin-related enzymes including E3 ligases and Dubs (deubiquitinases). For instance, E3 ligases including Dnr1 (defense repressor 1) [22], Faf (fat facets) [23], Posh (plenty of SH3s) [24], and SkpA (Skp1-related A) [25] negatively contribute to IMD signaling through mediating the ubiquitin assembly of targeted substrates. Additionally, a Dub Usp36 (ubiquitin-specific protease 36) associates with Imd to antagonize its K63-linked ubiquitination, thereby down-regulating the IMD signals [26]. Some other Dubs such as Cyld (cylindromatosis) [27], Trabid (Traf-binding domain) [28, 29], and Otu (ovarian tumor) [30] have been found to limit excessive activation of the IMD pathway under different physiological conditions. Recently, we noted that the ubiquitin carboxy-terminal hydrolase L5 (Uch-L5, also known as Uch37) functions as a typical Dub (belonging to the Uch sub-family of Dubs) to positively regulate Hh (hedgehog) signaling by restricting ubiquitination-mediated degradation of Smo (smoothened) [31]. However, little is known about the immunological role of Uch-L5 in the fly defense against microbial challenges.

In the present study, we focused mainly on exploring the potential role of Uch-L5 in regulating Drosophila innate immunity. We show that silencing of Uch-L5 results in markedly decreased IMD signaling, whereas ectopic expression of Uch-L5 behaves oppositely. The N-terminal Uch domain is both required and sufficient for Uch-L5 positively impacting on IMD signaling, suggesting that Uch-L5 executes its immune function relying on the Dub enzymatical activity. We further provide evidence displaying that Uch-L5 is essential for the immune defense of adult flies upon bacterial infections. Collectively, our results uncover a novel biological role of Uch-L5 in mediating the IMD innate immune reaction in Drosophila.

2. Materials and Methods
2.1 Drosophila Husbandry

Flies (Drosophila melanogaster) were raised at 25 °C with a 12 h of light/dark cycle and 65% humidity. Standard Drosophila medium (6.65% cornmeal, 7.15% dextrose, 5% yeast, 0.66% agar, 2.2% nipagin, and 3.4 mL/L propionic acid) was used to feed all flies. The w1118 strain was used as the wild-type control and all other flies were back-crossed with w1118 for isogenization (at least 5 generations). The detailed information of the flies used in this study are as follows: (1) keyc02831 (#11044) and Uch-L5j2B8(#12078) were purchased from Bloomington Drosophila Stock Center; (2) Uch-L5 RNAi #1 (#103481) was obtained from Vienna Drosophila Resource Center; (3) Uch-L5 RNAi #2 (#04757.N) was acquired from Tsinghua Fly Center; and (4) c564-gal4 was described previously [32].

2.2 dsRNA Synthesis and RNAi in S2 Cells

All dsRNAs were synthesized according to methods described previously [33]. In brief, DNA templates were amplified using specific primers (Supplementary Table 1). PCR products were then combined with 1/10 volume of 5 M NaCl and 2.5-fold volume of EtOH, followed by centrifugation for 10 min at high speed. The pellet was washed with 75% EtOH and dissolved in RNase/DNase-free water. The T7 in vitro transcription kit (Promega, Cat#P1300, Madison, Wisconson, USA) was used for dsRNA synthesis and purification according to the manufacture’s protocol. Purified dsRNAs were diluted in RNase/DNase-free water to a final concentration of 1 µg/µL. The quality of dsRNAs was confirmed by agarose gel electrophoresis (Supplementary Fig. 1A). For gene silencing in S2 cells, dsRNA at the amount of 3 µg was added directly into S2 cells for 48 h.

2.3 S2 Cell Transfection and Luciferase Reporter Assay

S2 cells were cultured in a 27 °C incubator using insect medium (Gibco) with 10% FBS (Hyclone). The MycoAlertTM kit (Lonza, Cat#LT07-318, Basel, Halbkanton, Switzerland) was used for mycoplasma detection and validation of the authenticity of S2 cells. LipofectamineTM 2000 (Thermo Fisher, Cat#11668019, Waltham, Massachusetts, USA) was used for plasmid transfection into S2 cells according to previous protocols [34]. The Att-Luc [35] and Drs-Luc [33] reporter systems were utilized to detect the relative activities of the IMD and Toll pathways, respectively. For the IMD pathway, we transfected S2 cells with the plasmid that express Imd, in order to induce the Att-Luc activity. As a positive control, we used the dsRNA targeting kenny, which is one of the pivotal genes of the IMD pathway. For the Toll pathway, the Myd88 expressing plasmid and the pelle dsRNA (positive control) were used. The methods for detecting the Firefly and Renilla luciferase activities were as follows: S2 cells were adequately lysed with 100 µL of passive lysis buffer (Promega, Cat#PR-E1941, Madison, Wisconson, USA). After centrifugation, 30 µL of the supernatant from each sample was added into a 96-well plate containing detection reagents. Firefly and Renilla luciferase activities were detected and finally the ratio of Firefly to Renilla was calculated. Results were analyzed based on data from 3 independent biological replications.

2.4 Reverse transcription plus quantitative polymerase chain reaction (RT-qPCR) Assay

S2 cells or fly samples were homogenized in Trizol Reagent (Invitrogen, Cat#15596026, Waltham, Massachusetts, USA). Chloroform solution (1/5 volume) was added into the sample, followed by intense votexing for 15 sec, and centrifugation at high speed for 15 min. The upper layer of the supernatant was transferred into a fresh tube and an equal volume of isopropanol was added. After centrifugation again at high speed for 10 min, the pellet was washed with 75% EtOH and diluted in RNase/DNase-free water. Samples were further incubated with DNase for 30 min, and the quality of RNA was assessed by examining the 260:280 ratio, and the pattern in the agarose gel electrophoresis assay. The first-strand cDNA synthesis kit (Transgen, Cat#AT341-01, Beijing, China) was used to reverse-transcribe RNA (1 µg) into cDNA. Quantitative PCR assays (in 3 technical repetitions) were performed using a SYBR Green Master Mix (Thermo Fisher, Cat#A46012, Waltham, Massachusetts, USA) in the Lightcycler 480 PCR platform (Roche, Cat#05015278001, Basel, Halbkanton, Switzerland). Rp49 was used as the internal control. Results were analyzed based on data from 7 independent biological replications. The primers used in RT-qPCR experiments are shown in Supplementary Table 1.

2.5 Infection, Fly Survival, and Bacterial Burden Assay

Infection experiments were performed as previously described [36]. In brief, overnight bacterial cultures were harvested and diluted in sterile PBS at a concentration of OD600 = 1. The Serratia marcescens strain (#1.1215) was obtained from China General Microbiological Culture Collection Center (CGMCC) and the Ecc15 was a kind gift from Dr. Dominique Ferrandon’s laboratory (Institut de Biologie Moleculaire et Cellulaire, France). For injection, male flies were collected and anesthetizsed on the fly pad with CO2. Diluted bacteria (4.6 nL), or the same volume of PBS, was injected into each fly with a nanoliter injector (Nanoject III, Drummond, Cat#3-000-207, Broomall, Pennsylvania, USA). Flies that died within 2 h after bacterial infection were not included in further studies. For survival analysis, flies were transferred to fresh vials and counted for death every day. For bacterial burden experiments, 10 flies were collected, dipped in 75% EtOH, and volatilized with EtOH on the fly pad for several minutes. The flies were then homogenized in 200 µL sterile PBS, followed by serial dilutions. Finally, 100 µL of each diluent was inoculated on an LB agar plate at 30 °C for 12 h before we counted the numbers of the colonies.

2.6 Statistical Analysis

All statistical analyses were performed by using GraphPad Prism 9 (v. 9.4.0, Dotmatics, Boston, MA, USA). Data were shown as means plus standard errors. Statistical significance was determined by using the ANOVA or Mann-Whitney tests except for survival assays, in which the Log-Rank test (Kaplan-Meier method) was used. The p value of less than 0.05 was considered statistically significant. * p < 0.05, ** p < 0.01, *** p < 0.001, ns (not significant) p > 0.05.

3. Results
3.1 Uch-L5 Positively Modulates IMD Signaling in Drosophila S2 Cells

We examined the potential role of Uch-L5, a previously described Dub modulating Hh signaling [31], in affecting the innate immune reaction in Drosophila. To do this, we first designed 3 types of dsRNAs that targeted different regions of the coding sequence of Uch-L5 (referred to as Uch-L5 dsRNAs #1, #2, and #3, respectively, Supplementary Fig. 1A). Cultured Drosophila S2 cells were treated with these Uch-L5 dsRNAs (dsRNA that targeted GFP was used as the control), and the knockdown efficiency was monitored by RT-qPCR assays (Supplementary Fig. 1B). As illustrated in Fig. 1A, silencing of Uch-L5 resulted in drastic decreases (by ~75% to ~80%) of the Att-Luc activities upon Imd over-expression, indicating that Uch-L5 is a potentially positive regulator in the IMD signaling pathway in cultured S2 cells. Further, we performed RT-qPCR assays to detect the endogenous inductions of the AMPs that are downstream of the IMD pathway, including AttA, Dpt, and CecA1. We obtained consistent results: down-regulation of Uch-L5 significantly prevented the Imd-driven transcription of several AMP genes (Fig. 1B–D).

Fig. 1.

Silencing of ubiquitin carboxy-terminal hydrolase L5 (Uch-L5) reduces immune deficiency (IMD) signaling in Drosophila S2 cells. (A) S2 cells were pretreated with indicated dsRNAs. 48 h later, cells were transfected with various combinations of expressing plasmids, followed by dual-luciferase assays. (B–D) S2 cells were pretreated with dsRNAs as in (A) for 48 h. Cells were then transfected with indicated expressing plasmids for 36 h and harvested for RT-qPCR assays to detect the mRNA levels of AttA (B), Dpt (C), and CecA1 (D). In (A–D), each dot represents one independent replicate and data are shown as means plus standard errors. * p < 0.05, ** p < 0.01, *** p < 0.001.

3.2 Uch-L5 is Dispensable for Affecting Toll Signaling

As mentioned in the Introduction, the Drosophila innate immune response is mostly governed by two signaling cascades, the Toll and the IMD pathways. We then conducted another luciferase reporter assay (Drs-Luc) in S2 cells, as previously reported, aiming to test whether Uch-L5 is also involved in affecting Toll signaling. However, we did not observe apparent alterations in the Myd88-driven Drs-Luc activities when Uch-L5 was knocked down, compared to that in the control (Supplementary Fig. 2A). Similar results were obtained when we looked at the transcript levels of Drs and Mtk, two well-known downstream AMP genes in the Toll pathway (Supplementary Fig. 2B,C). These data indicated that Drosophila Dub Uch-L5 is specifically involved in controlling the IMD other than the Toll innate immune signaling pathway in S2 cell cultures.

3.3 Uch-L5 Relies on the N-Terminal Uch Domain to Regulate IMD Signaling

We collected more evidence on the functional role of Uch-L5 in controlling IMD innate immunity by constructing a plasmid expressing Myc-tagged Uch-L5 in S2 cells. By again using the Att-Luc reporter system, we observed that ectopic expression of Myc-Uch-L5 enhanced IMD signaling in a dose-dependent manner (Fig. 2A). Accordingly, co-transfection of Uch-L5 expressing plasmid resulted in increased inductions (by ~32% to ~136%) of the IMD-downstream AMPs upon Imd over-expression (Fig. 2B–D), which indicated a positive role of Uch-L5 in modulating IMD signaling.

Fig. 2.

Uch-L5 affects IMD signaling in a Uch domain-dependent manner. (A–D) S2 cells were transfected with expressing plasmids as indicated, followed by dual-luciferase assays (A) or RT-qPCR assays to examine the expression levels of AttA (B), Dpt (C), and CecA1 (D). (E) Domain analysis of Uch-L5. (F) S2 cells were transfected with indicated combinations of expressing plasmids, followed by dual-luciferase assays. In (A–D,F), each dot represents one independent replicate and data are shown as means plus standard errors. * p < 0.05, ** p < 0.01, *** p < 0.001, ns p > 0.05.

A recent study that explored the biochemical characteristic of Uch-L5 demonstrated that Uch-L5 harbors a typical Uch domain at its N-terminus, which is the catalytical region for its Dub enzymatical activity [31]. We therefore constructed two kinds of plasmids that expressed truncated forms of Uch-L5, including Uch-L5UD (Uch domain) and Uch-L5CTD (C-terminal domain) (Fig. 2E), and transfected them into S2 cells for Att-Luc reporter assay. As illustrated in Fig. 2F, over-expression of Uch-L5UD, but not Uch-L5CTD, mimicked the positive contribution of Uch-L5FL (full-length Uch-L5) to IMD signaling. Taken together, these data suggested that the N-terminal Uch domain is both required and sufficient for Uch-L5 to benifit Drosophila IMD signaling.

3.4 Uch-L5 Mediates the IMD Innate Immune Defense in Adult Flies

We next sought to decipher the immune function of Uch-L5 in vivo. To this end, we collected w1118 flies (referred to as the wild-type control), keyc02831 flies (key loss-of-function mutant as the positive control), and Uch-L5j2B8 flies (Uch-L5 loss-of-function mutant, isogenized with w1118) for infection experiments using the Gram-negative bacteria S. marcescens (Serratia marcescens), to activate the host IMD innate immune defense. Six hours after infection, we detected decreases (by ~34% to ~56%) of AttA, Dpt, and CecA1 in the Uch-L5-defective flies relative to those in the wild-type control (Fig. 3A–C). To confirm our results, we subjected these flies to infection with another type of Gram-negative bacteria, Ecc15 (Erwinia carotovora carotovora 15). Similar reduction trends were observed when we compared the expression levels of AMPs in the Uch-L5 and key loss-of-function mutants to those in the wild-type flies (Fig. 3A–C). In addition, we noted comparable survival rates of these flies (w1118, keyc02831, and Uch-L5j2B8) after injection of sterile PBS buffer (Fig. 4A). However, the Uch-L5 loss-of-function mutant flies succumbed much faster than did the wild-type controls upon injection of either S. marcescens (Fig. 4B) or Ecc15 (Fig. 4C), indicating that Uch-L5 plays an essential role in the host defense against bacterial infections.

Fig. 3.

Loss of Uch-L5 antagonizes IMD downstream AMP expressions. (A–C) w1118 (wild-type control), keyc02831, and Uch-L5j2B8 males (6- to 7-day) were injected with PBS, S. marcescens, or Ecc15. 6 h later, flies were harvested for RT-qPCR assays to detect the expression levels of AttA (A), Dpt (B), and CecA1 (C). Each dot represents one independent replicate and data are shown as means plus standard errors. * p < 0.05, ** p < 0.01, *** p < 0.001.

Fig. 4.

Uch-L5 plays a critical role in the fly anti-microbial defense. (A–C) w1118 (wild-type control), keyc02831, and Uch-L5j2B8 males (6- to 7-day) were infected with PBS (A), S. marcescens (B), or Ecc15 (C), followed by survival analyses. The numbers of flies in each figure are as follows. In (A), w1118: 104, 103, 106; keyc02831: 105, 105, 102; Uch-L5j2B8: 102, 103, 104. In (B), w1118: 102, 104, 106; keyc02831: 104, 103, 105; Uch-L5j2B8: 105, 103, 103. In (C), w1118: 104, 107, 106; keyc02831: 106, 104, 103; Uch-L5j2B8: 104, 103, 106. (D,E) w1118 (wild-type control), keyc02831, and Uch-L5j2B8 males were infected with S. marcescens (D) or Ecc15 (E), followed by bacterial burden assays at indicated time points (0, 6, and 12 h). Each dot represents one independent replicate and data are shown as means plus standard errors. For each replicate, 10 male flies were collected. In (B–E), * p < 0.05, ** p < 0.01, *** p < 0.001.

To test the involvement of Uch-L5 in affecting the proliferation of injected pathogens (S. marcescens or Ecc15), we performed CFU (colony-forming-units) assays at different time points (0, 6, and 12 h) after injection. As shown in Fig. 4D,E, the burdens of S. marcescens and Ecc15 in Uch-L5j2B8 adults were significantly higher than those in the wild-type flies. In summary, our in vivo data strongly support the notion that Uch-L5 plays a critical role in the host defense against bacterial pathogens in Drosophila.

3.5 Silencing of Uch-L5 in Fat Body Leads to Immune Defects

Fat body is one of the main responsible immune tissues/organs during systemic infection in Drosophila. As the Uch-L5 transcript is relatively abundant in the fat body cells, according to the high-throughput sequencing or array data in the FlyBase website (https://flybase.org/reports/FBgn0011327), we sought to investigate the functional role of Uch-L5 in Drosophila fat body. Two different Uch-L5 RNAi (RNA interference) flies (Uch-L5 RNAi #1 and #2, for detailed information, see Materials and Methods) were crossed with the fat body-specific driver c564-gal4. The Tub-gal80𝑡𝑠 strain was used in order to allow gene RNAi at adult stage (Supplementary Fig. 3A). The progenies, including c564𝑡𝑠>Uch-L5 RNAi #1 and c564𝑡𝑠>Uch-L5 RNAi #2, were collected and subjected to S. marcescens or Ecc15 infection (c564𝑡𝑠>+ was used as the wild-type control). Six hours after S. marcescens or Ecc15 infection, the inductions of AttA, Dpt, and CecA1 in Uch-L5 RNAi flies were prominently decreased compared to those in the control group (Fig. 5A–C). These results suggested that Uch-L5 functions in fat body to modulate IMD signaling in response to bacterial challenge. Consistently, we observed that the Uch-L5 RNAi flies were less resistant to S. marcescens or Ecc15 infection: they displayed higher mortality than did the wild-type control (Fig. 5D–F). Moreover, the bacterial burdens at various time points after infection were remarkably elevated by the loss of Uch-L5 in fat body (Fig. 5G,H).

Fig. 5.

Silencing of Uch-L5 in fat body results in immune defects. (A–H) c564𝑡𝑠>+ (wild-type control), c564𝑡𝑠>Uch-L5 RNAi #1, and c564𝑡𝑠>Uch-L5 RNAi #2 males (6- to 7-day) were injected with PBS, S. marcescens, or Ecc15. Flies were collected for RT-qPCR assays to examine the inductions of AttA (A), Dpt (B), and CecA1 (C) 6h after infection, or subjected to survival analyses (D–F), or bacterial burden assays (G,H). In (A–C, G,H), each dot represents one independent replicate and data are shown as means plus standard errors. In (D), the numbers of flies are as follows. c564𝑡𝑠>+: 104, 105, 104; c564𝑡𝑠>Uch-L5 RNAi #1: 103, 102, 102; c564𝑡𝑠>Uch-L5 RNAi #2: 103, 106, 105. In (E), c564𝑡𝑠>+: 102, 103, 106; c564𝑡𝑠>Uch-L5 RNAi #1: 104, 105, 105; c564𝑡𝑠>Uch-L5 RNAi #2: 103, 104, 102. In (F), c564𝑡𝑠>+: 105, 104, 102; c564𝑡𝑠>Uch-L5 RNAi #1: 106, 103, 104; c564𝑡𝑠>Uch-L5 RNAi #2: 105, 105, 104. In (A–H), * p < 0.05, ** p < 0.01, *** p < 0.001.

4. Discussion

Drosophila Uch-L5 belongs to the Uch sub-family of Dubs [31, 37]. Dub-mediated cleavage of ubiquitin/poly-ubiquitin from ubiquitinated substrates has been widely studied and demonstrated to be involved in a broad range of cellular processes [reviewed in [38, 39, 40]]. However, our knowledge regarding the biological function of the fly Uch-L5 is incomplete. In the present study, we carried out a series of investigations using both in vitro and in vivo models. We showed that Uch-L5 behaves as a positive modulator in the fly IMD innate immune defense against bacterial stimuli. Our data shed light on a previously undescribed role of Uch-L5 in controlling organismal innate immunity.

How does Uch-L5 execute its essential role in regulating the IMD signaling pathway? A recent study by Zhou and colleagues illustrated that Uch-L5 depends on its N-terminal Uch domain to positively contribute to Hh signaling [31], highlighting the critical requirement of the Dub catalytical triad for Uch-L5 functioning. Indeed, when we utilized several truncated forms of Uch-L5 expressing plasmids and conducted Att-Luc reporter assays in cultured S2 cells, we observed that Uch-L5 without the Uch domain no longer promoted IMD signaling. It is of interest to note that the Dub enzymatical activity of Drosophila Uch-L5 is somehow autoinhibited by the CTD via oligomerization, which can be alleviated by the association of a co-factor such as Rpn13 [31, 37]. However, we would like to conclude that this is not the case for the regulatory role of Uch-L5 in innate immune regulation, based on the following reasons: (1) ectopic expression of Uch-L5 without CTD showed a similar contribution to the IMD signaling as that of the Uch-L5FL in S2 cells; and (2) co-expression of Rpn13 hardly affected the beneficial role of Uch-L5 in IMD signaling (Supplementary Fig. 4A). It seems that Uch-L5 relies on the same Uch domain to participate in different signaling cascades, but its CTD largely functions in distinct manners. Nevertheless, it would be worthwhile to generate corresponding Uch-L5 transgenic flies to confirm these hypotheses in vivo.

Recently, numerous efforts have been made to decipher the critical role of Dubs in regulating IMD signaling. To date, only Imd, Tak1, and Dredd in the canonical IMD pathway, have been clearly demonstrated to be modified by ubiquitination/deubiquitination. Whether the other key factors also involve a ubiquitin-mediated modulation remains a mystery. Uch-L5 might therefore target a factor, or some of these factors, to enhance IMD signaling. The 48th lysine (K48)-linked ubiquitination has been suggested to primarily mediate protein recognition and degradation by the 26S proteasome, whereas the 63rd lysine (K63)-linked ubiquitination commonly governs signal transduction [41]. According to our current knowledge and the results of the present study, we speculate that the Dub Uch-L5 may inhibit the K48-linked ubiquitination of its substrate(s), thus improving its (their) stability. Future biochemical approaches, for instance co-immunoprecipitation and ubiquitination assays, will greatly help us further understand the molecular mechanism by which Uch-L5 benefits the IMD innate immune defense in Drosophila.

On the other hand, it is somehow lagged off that no Dubs have been shown to play a role in the Toll innate immune defense of Drosophila. This may be due to the fact that ubiquitination modification in Toll signaling is not as important as that in IMD signaling. Even though we failed to observe in the present study any involvement of Uch-L5 in controlling the Toll pathway, we cannot exclude the possibility that the Toll pathway is regulated by other Dubs. One strategy would be to explore Dubs that can physically associate with MyD88, a pivotal Toll downstream adaptor protein that undergoes ubiquitination [33].

5. Conclusions

We have demonstrated a critical regulatory role of the Dub Uch-L5 in Drosophila IMD innate immunity. The present study has provided insight into the understanding of the precise dynamic modulation of IMD signaling in response to bacterial infection.

Availability of Data and Materials

The datasets used and analysed during the current study are available from the corresponding author on reasonable request.

Author Contributions

SJ and YH designed the research study. CZ, SZ, FK, YX, KS, YJ, JL, AQ, and XZ performed the research. CZ, SZ, FK, and YH analyzed the data. SJ and YH wrote the manuscript. All authors contributed to editorial changes in the manuscript. All authors read and approved the final manuscript.

Ethics Approval and Consent to Participate

Not applicable.

Acknowledgment

We are grateful for the Bloomington Drosophila Stock Center, the Vienna Drosophila Resource Center, and the Tsinghua Fly Center for fly resources.

Funding

This research was funded by the National Natural Science Foundation of China (32100702) and the Anhui Provincial Natural Science Foundation (2008085J14).

Conflict of Interest

The authors declare no conflict of interest.

References
[1]
Beutler B. Innate immunity: an overview. Molecular Immunology. 2004; 40: 845–859.
[2]
Taguchi T, Mukai K. Innate immunity signalling and membrane trafficking. Current Opinion in Cell Biology. 2019; 59: 1–7.
[3]
Yu S, Luo F, Xu Y, Zhang Y, Jin LH. Drosophila Innate Immunity Involves Multiple Signaling Pathways and Coordinated Communication Between Different Tissues. Frontiers in Immunology. 2022; 13: 905370.
[4]
Fitzgerald KA, Kagan JC. Toll-like Receptors and the Control of Immunity. Cell. 2020; 180: 1044–1066.
[5]
Akira S, Uematsu S, Takeuchi O. Pathogen recognition and innate immunity. Cell. 2006; 124: 783–801.
[6]
Akira S. Pathogen recognition by innate immunity and its signaling. Proceedings of the Japan Academy. Series B, Physical and Biological Sciences. 2009; 85: 143–156.
[7]
Hoffmann JA. The immune response of Drosophila. Nature. 2003; 426: 33–38.
[8]
Lemaitre B, Hoffmann J. The host defense of Drosophila melanogaster. Annual Review of Immunology. 2007; 25: 697–743.
[9]
Zhang ZQ, Ji SM, Yang LY, Mei XP. Triptolide Alleviates Neuropathic Pain by Inhibiting the Activation of Microglial Toll-Like Receptor 3. Journal of Integrative Neuroscience. 2022; 21: 150.
[10]
Valanne S, Wang JH, Rämet M. The Drosophila Toll signaling pathway. Journal of Immunology (Baltimore, Md.: 1950). 2011; 186: 649–656.
[11]
Kleino A, Silverman N. The Drosophila IMD pathway in the activation of the humoral immune response. Developmental and Comparative Immunology. 2014; 42: 25–35.
[12]
Kaneko T, Yano T, Aggarwal K, Lim JH, Ueda K, Oshima Y, et al. PGRP-LC and PGRP-LE have essential yet distinct functions in the drosophila immune response to monomeric DAP-type peptidoglycan. Nature Immunology. 2006; 7: 715–723.
[13]
Naitza S, Rossé C, Kappler C, Georgel P, Belvin M, Gubb D, et al. The Drosophila immune defense against gram-negative infection requires the death protein dFADD. Immunity. 2002; 17: 575–581.
[14]
Paquette N, Broemer M, Aggarwal K, Chen L, Husson M, Ertürk-Hasdemir D, et al. Caspase-mediated cleavage, IAP binding, and ubiquitination: linking three mechanisms crucial for Drosophila NF-kappaB signaling. Molecular Cell. 2010; 37: 172–182.
[15]
Vidal S, Khush RS, Leulier F, Tzou P, Nakamura M, Lemaitre B. Mutations in the Drosophila dTAK1 gene reveal a conserved function for MAPKKKs in the control of rel/NF-kappaB-dependent innate immune responses. Genes & Development. 2001; 15: 1900–1912.
[16]
Silverman N, Zhou R, Erlich RL, Hunter M, Bernstein E, Schneider D, et al. Immune activation of NF-kappaB and JNK requires Drosophila TAK1. The Journal of Biological Chemistry. 2003; 278: 48928–48934.
[17]
Rutschmann S, Jung AC, Zhou R, Silverman N, Hoffmann JA, Ferrandon D. Role of Drosophila IKK gamma in a toll-independent antibacterial immune response. Nature Immunology. 2000; 1: 342–347.
[18]
Silverman N, Zhou R, Stöven S, Pandey N, Hultmark D, Maniatis T. A Drosophila IkappaB kinase complex required for Relish cleavage and antibacterial immunity. Genes & Development. 2000; 14: 2461–2471.
[19]
Stoven S, Silverman N, Junell A, Hedengren-Olcott M, Erturk D, Engstrom Y, et al. Caspase-mediated processing of the Drosophila NF-kappaB factor Relish. Proceedings of the National Academy of Sciences of the United States of America. 2003; 100: 5991–5996.
[20]
Mishima Y, Quintin J, Aimanianda V, Kellenberger C, Coste F, Clavaud C, et al. The N-terminal domain of Drosophila Gram-negative binding protein 3 (GNBP3) defines a novel family of fungal pattern recognition receptors. The Journal of Biological Chemistry. 2009; 284: 28687–28697.
[21]
Myllymäki H, Valanne S, Rämet M. The Drosophila imd signaling pathway. Journal of Immunology (Baltimore, Md.: 1950). 2014; 192: 3455–3462.
[22]
Guntermann S, Primrose DA, Foley E. Dnr1-dependent regulation of the Drosophila immune deficiency signaling pathway. Developmental and Comparative Immunology. 2009; 33: 127–134.
[23]
Yagi Y, Lim YM, Tsuda L, Nishida Y. fat facets induces polyubiquitination of Imd and inhibits the innate immune response in Drosophila. Genes to Cells: Devoted to Molecular & Cellular Mechanisms. 2013; 18: 934–945.
[24]
Tsuda M, Langmann C, Harden N, Aigaki T. The RING-finger scaffold protein Plenty of SH3s targets TAK1 to control immunity signalling in Drosophila. EMBO Reports. 2005; 6: 1082–1087.
[25]
Khush RS, Cornwell WD, Uram JN, Lemaitre B. A ubiquitin-proteasome pathway represses the Drosophila immune deficiency signaling cascade. Current Biology: CB. 2002; 12: 1728–1737.
[26]
Thevenon D, Engel E, Avet-Rochex A, Gottar M, Bergeret E, Tricoire H, et al. The Drosophila ubiquitin-specific protease dUSP36/Scny targets IMD to prevent constitutive immune signaling. Cell Host & Microbe. 2009; 6: 309–320.
[27]
Tsichritzis T, Gaentzsch PC, Kosmidis S, Brown AE, Skoulakis EM, Ligoxygakis P, et al. A Drosophila ortholog of the human cylindromatosis tumor suppressor gene regulates triglyceride content and antibacterial defense. Development (Cambridge, England). 2007; 134: 2605–2614.
[28]
Fernando MDA, Kounatidis I, Ligoxygakis P. Loss of Trabid, a new negative regulator of the drosophila immune-deficiency pathway at the level of TAK1, reduces life span. PLoS Genetics. 2014; 10: e1004117.
[29]
Hua Y, Zhu Y, Hu Y, Kong F, Duan R, Zhang C, et al. A Feedback Regulatory Loop Involving dTrbd/dTak1 in Controlling IMD Signaling in Drosophila Melanogaster. Frontiers in Immunology. 2022; 13: 932268.
[30]
Ji S, Luo Y, Cai Q, Cao Z, Zhao Y, Mei J, et al. LC Domain-Mediated Coalescence Is Essential for Otu Enzymatic Activity to Extend Drosophila Lifespan. Molecular Cell. 2019; 74: 363–377.e5.
[31]
Zhou Z, Yao X, Pang S, Chen P, Jiang W, Shan Z, et al. The deubiquitinase UCHL5/UCH37 positively regulates Hedgehog signaling by deubiquitinating Smoothened. Journal of Molecular Cell Biology. 2018; 10: 243–257.
[32]
Cai Q, Guo H, Fang R, Hua Y, Zhu Y, Zheng X, et al. A Toll-dependent Bre1/Rad6-cact feedback loop in controlling host innate immune response. Cell Reports. 2022; 41: 111795.
[33]
Ji S, Sun M, Zheng X, Li L, Sun L, Chen D, et al. Cell-surface localization of Pellino antagonizes Toll-mediated innate immune signalling by controlling MyD88 turnover in Drosophila. Nature Communications. 2014; 5: 3458.
[34]
Ji S, Li C, Hu L, Liu K, Mei J, Luo Y, et al. Bam-dependent deubiquitinase complex can disrupt germ-line stem cell maintenance by targeting cyclin A. Proceedings of the National Academy of Sciences of the United States of America. 2017; 114: 6316–6321.
[35]
Zhu Y, Cai Q, Zheng X, Liu L, Hua Y, Du B, et al. Aspirin Positively Contributes to Drosophila Intestinal Homeostasis and Delays Aging through Targeting Imd. Aging and Disease. 2021; 12: 1821–1834.
[36]
Zhu Y, Liu L, Zhang C, Zhang C, Han T, Duan R, et al. Endoplasmic reticulum-associated protein degradation contributes to Toll innate immune defense in Drosophila melanogaster. Frontiers in Immunology. 2023; 13: 1099637.
[37]
Jiao L, Ouyang S, Shaw N, Song G, Feng Y, Niu F, et al. Mechanism of the Rpn13-induced activation of Uch37. Protein & Cell. 2014; 5: 616–630.
[38]
Amerik AY, Hochstrasser M. Mechanism and function of deubiquitinating enzymes. Biochimica et Biophysica Acta. 2004; 1695: 189–207.
[39]
Lange SM, Armstrong LA, Kulathu Y. Deubiquitinases: From mechanisms to their inhibition by small molecules. Molecular Cell. 2022; 82: 15–29.
[40]
Clague MJ, Urbé S, Komander D. Breaking the chains: deubiquitylating enzyme specificity begets function. Nature Reviews. Molecular Cell Biology. 2019; 20: 338–352.
[41]
Swatek KN, Komander D. Ubiquitin modifications. Cell Research. 2016; 26: 399–422.

Publisher’s Note: IMR Press stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share
Back to top