Weisong
Lv†
ab,
Zheng
Wang†
ab,
Can
Zhang
b,
Taorui
Yang
a,
Tao
Liu
b,
Jia
Li
ab,
Xiaohui
Fan
*ab and
Xin
Li
*ab
aCollege of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Street, Hangzhou 310058, China. E-mail: fanxh@zju.edu.cn; lixin81@zju.edu.cn
bState Key Laboratory of Chinese Medicine Modernization, Innovation Center of Yangtze River Delta, Zhejiang University, Jiashan 314100, China
First published on 20th August 2025
Detecting enzyme activity that catalyzes subtle functional group transformations in live cells remains a major challenge. We introduce a conditional metabolic labeling strategy for enzymatic activity detection (cMLEAD), which harnesses cellular metabolic pathways to deliver indirect yet reliable activity readouts. Unlike traditional metabolic labeling approaches relying on nonspecific incorporation of tagged biomolecules, cMLEAD employs a tagged precursor whose metabolic incorporation is strictly dependent on specific enzymatic activity, effectively transforming a metabolic labeling event into an enzyme-activity measurement. Using aldehyde dehydrogenase 2 (ALDH2) as a proof of concept, we demonstrate the robustness of the strategy. cMLEAD for ALDH2 employs azido-tagged acetaldehyde, metabolized by ALDH2 into azidoacetate, which feeds into the acetyl-CoA biosynthetic pathway and is incorporated into lysine acylation, enabling fluorescence-based detection via click chemistry. The assay reliably reports ALDH2 activity, as validated through genetic and pharmacological modulation. cMLEAD further revealed suppressed ALDH2 activity under cellular senescence and oxidative stress, with direct inhibition by H2O2 likely contributing in part. Notably, cMLEAD is complementary to conventional in vitro assays and advantageous in preserving the native enzyme context. Leveraging this advantage, we developed a screening platform that identified sennoside A as a candidate ALDH2 activator, which alleviated light-induced retinal degeneration in mice. This study establishes cMLEAD as a robust and versatile platform for probing ALDH2 activity in pathophysiologically relevant contexts and facilitating therapeutic discovery. We envision the conceptual framework of cMLEAD may be adapted to other enzymes whose catalytic products feed into detectable metabolic incorporation.
Recent advancements have introduced activity-based small-molecule probes for measuring enzyme activity in live cells.5–8 These probes typically consist of organic dyes conjugated to natural substrates, with activatable signals generated by enzyme-catalyzed cleavage reactions. For instance, fluorophore–peptide conjugates detect protease activity;9 phosphorylated fluorophores monitor phosphatases;10 and fluorophore–saccharide conjugates track glycosidases.11 These enzymes generally tolerate significant structural changes in their substrates, and their cleavage-based reactions facilitate robust activatable signals. While these designs have yielded practical tools for detecting enzyme activities in intact cells, their efficacy diminishes when applied to enzymes that catalyze subtle chemical transformations, particularly those whose native substrates allow only minimal structural modification. The key challenges lie in maintaining efficient enzymatic turnover and in generating distinguishable fluorescence signals for reliable readouts.
Aldehyde dehydrogenase 2 (ALDH2) exemplifies this challenge. ALDH2 catalyzes the oxidation of aldehydes into carboxylic acids, with acetaldehyde as its primary substrate.12 Impaired ALDH2 function leads to the accumulation of toxic aldehydes, which are associated with numerous health risks, including alcohol-related disorders,13 cardiovascular diseases,14 neurodegenerative disorders,15 and cancers.16 Importantly, ALDH2 activation has shown therapeutic potential, offering protection against cardiac ischemia,17 cognitive deficits in Alzheimer's models,18 and septic acute respiratory distress syndrome.19 While ALDH2 is implicated in these pathologies, the mechanisms regulating its activity under various stress conditions remain poorly understood. Reliable methods for monitoring ALDH2 activity are therefore critical for elucidating its biological functions, uncovering its regulatory mechanisms, and discovering potential ALDH2 activators with therapeutic implications. ALDH2 activity is typically assessed using a standard in vitro spectrophotometric assay that monitors the conversion of NAD+ to NADH (Fig. 1A).17 However, this approach lacks the native cellular context and does not capture its dynamic regulation in live cells. While aldehyde-based fluorescent probes have been successfully developed for some other ALDH isoforms (Fig. 1B),20,21 designing selective probes for ALDH2 remains difficult, partly due to structural constraints. ALDH2's substrate-binding pocket is optimized for small-sized aldehydes and imposes steric limitations that restrict the incorporation of bulky fluorescent groups.22–24
Herein, we address this challenge by introducing a strategy that links ALDH2 activity to detectable metabolic incorporation of azide groups onto proteins that can be subsequently labeled with fluorophores. This approach, conditional metabolic labeling for enzymic activity detection (cMLEAD), leverages ALDH2's catalytic oxidation of azido-tagged acetaldehyde to azidoacetate, which is then funneled through the cellular metabolic machinery to produce azido-acetyl-CoA and subsequently azido-acetylated proteins.25,26 As a result, azidoacetaldehyde-derived protein azido-acetylation can serve as downstream proxy for ALDH2 activity (Fig. 1C). By coupling enzyme-specific substrate conversion with the metabolic labeling framework, cMLEAD enables the live-cell monitoring of enzyme activities that tolerate only minimal substrate modifications. We validated this concept using azido-tagged acetaldehyde and fluorescent click chemistry to monitor protein labeling. This assay reliably measured ALDH2 activity, as confirmed by genetic modulation of ALDH2 expression and pharmacological perturbations using specific activators and inhibitors. Furthermore, cMLEAD revealed decreased cellular ALDH2 activity under glucose deprivation and senescence, with direct inhibition by H2O2 likely contributing in part. We also developed a screening platform using cMLEAD and identified a candidate ALDH2 activator. These findings highlight the utility of cMLEAD for mechanistic investigation and therapeutic discovery. While this study focuses on ALDH2, the conceptual framework of cMLEAD may be extended to other enzymes, provided that their catalytic products feed into metabolic incorporation.
The feasibility of this approach hinges on fulfilling the following four key criteria. First, AAN3 must serve as an ALDH2 substrate. Previous studies support the feasibility of AAN3 serving as a substrate for ALDH2 because larger aldehydes have been shown to be processed as substrates of this enzyme.23,24 To evaluate this, we measured kinetic parameters of recombinant human ALDH2 to AAN3versus the native substrate acetaldehyde (AA), by quantifying NAD+ to NADH conversion. Both substrates were successfully converted by ALDH2, confirming AAN3's compatibility. However, AAN3 (80 μM) exhibited almost 40-fold higher Km than AA (1.8 μM) (Fig. 2B), underscoring ALDH2's limited tolerance for substrate modifications and supporting the need for a metabolic labeling-based detection method. Fortunately, the catalytic efficiency (Kcat/Km) towards AAN3 (1.6 min−1 μM−1) was only 10-fold compromised compared to AA (17 min−1 μM−1), warranting further study.
Second, AAN3 should not significantly alter endogenous protein acetylation levels, thereby ensuring an accurate readout of ALDH2 activity. To assess this, we examined cellular lysine acetylation using a pan-acetyl lysine antibody. At concentrations up to 0.5 mM, AAN3 had a negligible impact on global acetylation levels under the assay conditions; however, 1.0 mM AAN3 led to a noticeable decrease (Fig. 2C and S1), suggesting that working concentrations under 0.5 mM may be tolerable.
Third, AAN3 should exhibit low cytotoxicity. An MTT assay in Li-7 human liver cancer cells revealed no significant toxicity at concentrations up to 0.5 mM after 8 hours of incubation. In contrast, concentrations above 1.0 mM resulted in a marked decrease in cell viability (Fig. 2D), reinforcing 0.5 mM AAN3 as a physiologically compatible dose.
Fourth, the AAN3-derived azidoacetate must be recognized by endogenous acetyl-CoA synthetases and further processed by acetyltransferases to generate azido-labeled acylated proteins. To validate this, Li-7 cells pretreated with AAN3 were subjected to a classical CuAAC condition,33 labelled with an alkyne-Rhodamine (RHO) synthesized according to literature procedures.34 Confocal imaging showed an AAN3 dose- (Fig. 2E and S2) and incubation time-dependent increase in cellular fluorescence (Fig. 2F and G), with AAN3 at 0.5 mM and an incubation time of 8 h giving a statistically significant increase of cellular RHO fluorescence. This suggests the proposed metabolic incorporation of the azido group into proteins as shown in Fig. 2A. To exclude the possibility of direct covalent crosslinking of AAN3 with proteins or nucleic acids, we treated the samples with methoxyamine prior to CuAAC, a procedure generally employed to release aldehyde-induced crosslinking in chemoproteomics.35 No significant difference in cellular fluorescence intensity was observed between the samples with and without methoxyamine treatment (Fig. S3). This suggests minimal interference from aldehyde-induced conjugation effects, likely eliminated by the stringent CuAAC washing steps. Notably, background fluorescence was detected in cells without AAN3 pretreatment, indicating nonspecific binding of the RHO fluorophore. This nonspecific binding is also found in a commercial alkynyl Rhodamine (Fig. S4). Together, these results confirm that AAN3-derived metabolites are successfully incorporated into protein acetylation, providing a robust foundation for imaging ALDH2 activity using the cMLEAD strategy.
In CuAAC, a chelating ligand is necessary to protect the copper cation from generating excessive reactive oxygen species and to minimize side reactions.36 The catalytic Cu(I) species is generated by reducing Cu(II) in situ to catalyze the cycloaddition between an azide and an alkyne.37 According to this mechanism, both the reducing agent, Cu(II) dose, and the chelating ligand should be critical to determine the reaction efficiency. By systematic optimization on these three parameters (Fig. S5–S7), the final CuAAC reaction conditions were set at 0.5 mM CuSO4, 2.5 mM Vc, and 600 μM THPTA, resulting in a 35-fold increase in cellular fluorescence intensity compared to the initial conditions. Further optimization on the click reaction time and RHO concentration revealed 2.5 μM RHO and a 60-min reaction time as optimal (Fig. S8 and S9).
To further validate the reliability of the strategy, we pharmacologically modulated ALDH2 activity using well-characterized small molecules. HepG2 cells were treated with Alda-1, a known ALDH2 activator,17 or dyclonine, an ALDH2 inhibitor,38 prior to cMLEAD labeling. Alda-1 induced a dose-dependent increase in cMLEAD fluorescence intensity, with an EC50 of 11 μM (Fig. 3D, E and S11), consistent with reported values.17 Conversely, dyclonine treatment caused a dose-dependent decrease in fluorescence (Fig. 3D, F and S12). While the IC50 of dyclonine could not be accurately determined due to cytotoxicity at higher concentrations, the trend supports the dependence of cMLEAD signal on ALDH2 activity. Notably, neither Alda-1 nor dyclonine altered the expression levels of ALDH2 (Fig. S13), indicating that the observed fluorescence changes were due to functional modulation of enzymatic activity rather than changes in enzyme abundance. To check if the pharmacological treatments affected protein acetylation, we performed immunofluorescence staining with the pan acetyl lysine monoclonal antibody. No significant global changes were observed across treatment groups under the assay condition (Fig. S14), supporting ALDH2-dependence of the cMLEAD readout. In addition, ALDH2 activity was independently evaluated using a conventional NADH-based assay after cell lysis, and the resulting trends mirrored those seen with cMLEAD labeling (Fig. 3C), reinforcing the assay's reliability. Collectively, these results establish cMLEAD as an ALDH2-dependent live-cell imaging platform for quantifying its activity with desirable sensitivity. The consistency across genetic and pharmacological manipulations underscores its utility not only for biological investigations but also for screening small-molecule modulators of ALDH2 in cellular settings.
Multiplex imaging revealed that the RHO signal predominantly localized to the nucleus (Fig. 4A). This result suggests that acetate-derived protein acylation mainly takes place in the nucleus, consistent with the dominant nuclear distribution of acetate-dependent acetyl-CoA synthetase 2 (ACSS2), the primary enzyme generating acetyl-CoA from acetate for histone acetylation.25,39,40 Noteworthy, the Pearson's correlation coefficient between the RHO signal and acetylated histone H3 signal was 0.62 (Fig. 4B and C), suggesting that ALDH2-generated azidoacetate works as a mimic of native acetate to acylate histones. Additionally, moderate colocalization was observed between the RHO and α-tubulin signals (Pearson's coefficient = 0.63) (Fig. 4D and E), consistent with previous findings that chronic ethanol exposure enhances tubulin acetylation.41 This result provides the first direct evidence that upregulated acetylation of α-tubulin is at least partially derived from the carbon source provided by ingested alcohol. All these results confirm the mechanism of cMLEAD, which integrates ALDH2's catalytic conversion of aldehydes to acetate with the cellular metabolic machinery of utilizing acetate for protein acetylation. This highlights the strategy's unique capability to exploit native cellular processes for interrogating enzyme activity.
With the cMLEAD strategy established, we applied it to investigate how ALDH2 activity responds to cellular stress conditions. We focused on bleomycin-induced senescence and glucose deprivation-induced metabolic stress in HepG2 cells.42,43 Cells treated with increasing doses of bleomycin for 48 h exhibited a dose-dependent senescent phenotype, confirmed by β-galactosidase (β-Gal) staining (Fig. 5A and C). cMLEAD labeling revealed a clear inverse correlation between RHO fluorescence intensity and bleomycin concentration (Fig. 5A and D), indicating a progressive reduction in ALDH2 activity during senescence. To validate that this observation was not confounded by global changes in protein acetylation, we performed immunofluorescence staining using the pan-acetyl lysine antibody. No significant difference in protein acetylation levels was observed across treatment groups under the assay conditions (Fig. S15), supporting the ALDH2 activity-dependence of the cMLEAD signal. Moreover, western blot analysis revealed no significant change in ALDH2 protein expression level upon bleomycin exposure (40 μM, 48 h) (Fig. S13). NADH-based ALDH2 assay in cell lysates confirmed the decreasing trend in enzymatic activity, in agreement with the cMLEAD results (Fig. 5E). These findings align with previous reports linking reduced or dysfunctional ALDH2 activity to aging-related pathologies,44 and further highlight the utility of cMLEAD for monitoring stress-induced changes in enzyme function in live cells.
Glucose deprivation similarly led to a reduction in ALDH2 activity in HepG2 cells, as revealed by cMLEAD fluorescence imaging (Fig. 5B and F). Importantly, western blot analysis showed that ALDH2 protein expression remained largely unchanged under low-glucose conditions (0 g L−1, 8 h) (Fig. S13), indicating that the observed decrease in activity is not due to altered expression. Given the well-established link between glucose deprivation and oxidative stress, we next assessed intracellular reactive oxygen species (ROS) levels using the 2′,7′-dichlorodihydrofluorescein diacetate (DCFH-DA) probe. A marked increase in DCFH-DA fluorescence was detected following glucose deprivation, confirming elevated oxidative stress (Fig. 5B and G). Notably, the extent of ROS accumulation was inversely correlated with ALDH2 activity as measured by cMLEAD, suggesting that oxidative stress may impair ALDH2 enzymatic function. Protein acetylation immunostaining (Fig. S16) and NADH-based ALDH2 activity assay in cell lysates (Fig. 5H) further confirmed the selective suppression of ALDH2 activity without significant disruption of cellular protein acetylation levels. These findings reinforce the conclusion that oxidative stress compromises ALDH2 function, which is detectable by cMLEAD, consistent with previous reports linking oxidative stress to aldehyde accumulation and metabolic dysfunction.45–47
To shed light on the mechanism underlying the compromised ALDH2 activity under these stress conditions, we investigated the direct effects of typical oxidative stress-associated metabolites on ALDH2 enzymatic function. Specifically, we assessed the impact of hydrogen peroxide (H2O2), which accumulates during oxidative stress, using a recombinant ALDH2-based NADH assay. H2O2 was found to inhibit ALDH2 activity in a dose-dependent manner, with IC50 of 40 μM (Fig. S17). While the precise mechanism underlying the observed reduction in ALDH2 activity under oxidative and senescence-related stress remains to be elucidated, our results suggest that ALDH2 is functionally sensitive to redox modulation. Importantly, cMLEAD enables the detection of such dynamic enzymatic changes in live-cell contexts, providing a valuable tool for monitoring stress-induced alterations in enzyme activity.
Initially, an in silico docking-based screening was performed on a natural compound library of 2784 structures to prioritize those with the most promising binding to ALDH2 (Table S1). The top 100 candidates were then evaluated using ADMETlab 2.0 to filter out those with unfavorable pharmacokinetic properties,48 resulting in 41 promising structures (Table S2). HepG2 cells were incubated with these compounds, followed by cMLEAD labeling (Fig. 6A). Cellular fluorescence was used as a readout of ALDH2 activity (Fig. S19). Setting the vehicle-treated group as the baseline (Fig. 6B), we identified three activators and three inhibitors (Fig. S20–S25). Among these, sennoside A exhibited the most robust activation (Fig. 6C), with cellular fluorescence intensity 10-fold higher than the vehicle group. Further evaluation with cMLEAD determined an EC50 of 2.2 μM for sennoside A (Fig. 6D, E and S20). Moreover, NADH-based ALDH2 activity in cell lysates after sennoside A-pretreatment also verified its activating effect (Fig. 6F). We also confirmed that sennoside A didn't affect the ALDH2's expression levels (Fig. S13), nor did it affect cellular acetylation levels under the assay conditions (Fig. S14). These findings suggest cMLEAD's applicability for screening, and also propose sennoside A as a candidate ALDH2 activator with the exact mode of action to be explored.
A series of tests assessed the protective effects of sennoside A. Elevated plasma interleukin-6 (IL-6) levels (Fig. 7A) and malondialdehyde (MDA) levels (Fig. 7B) in the model group confirmed significant inflammation and oxidative stress induced by light exposure. Notably, high-dose sennoside A markedly alleviated these effects, comparable to the positive control Lutein, suggesting its protective potential. Although the total antioxidant capacity (T-AOC) assay didn't reach statistical significance, a dose-dependent protective trend was observed (Fig. S26). Since sennoside A is known to exhibit multiple cellular activities, including antioxidant and anti-inflammatory effects,50,51 we interrogated the involvement of ALDH2 in the observed protective effects. Immunofluorescence staining revealed increased ALDH2 expression in retinal tissues from sennoside A-treated group relative to the model group (Fig. 7C and S27). While short-term sennoside A treatment in HepG2 cells (12 h) did not result in a significant change in ALDH2 protein expression (Fig. S13), chronic administration in vivo (daily for 10 days) was associated with an upward trend in ALDH2 levels. This observation raises the possibility of a positive feedback mechanism, whereby prolonged ALDH2 activation by sennoside A may reduce oxidative stress and inflammation, which in turn could stabilize ALDH2 and reduce its degradation. However, further investigation is needed to elucidate the underlying regulatory pathways.
Histopathological evaluation using H&E staining revealed light-induced retinal damage in the model group, characterized by reduced thickness and structural looseness of the outer and inner nuclear layers (Fig. 7D). Both low- and high-dose sennoside A mitigated this damage, preserving retinal morphology and reducing the thinning of the nuclear layers (Fig. 7F and G). TUNEL staining further confirmed retinal cell apoptosis caused by light exposure, indicated by increased red fluorescence in the model group (Fig. 7E). Sennoside A treatment at both doses significantly reduced apoptosis, with effects comparable to Lutein (Fig. 7H). These results validate the protective effects of sennoside A in mitigating inflammation, oxidative stress, and apoptosis in this light-induced retinal degeneration model. Since sennoside A has been verified to boost cellular ALDH2 activity (Fig. 6E and F), this study underscores the potential of the cMLEAD strategy for identifying candidate ALDH2 activators.
Nonetheless, several limitations of the cMLEAD strategy should be acknowledged. First, cMLEAD provides an indirect readout of enzyme activity that depends not only on ALDH2-mediated transformation but also on subsequent metabolic incorporation steps. As such, appropriate controls and, when possible, orthogonal validation methods are necessary to confirm enzymatic specificity. Second, the assay exhibits a relatively high background signal, which may limit sensitivity. Further optimization, such as reducing nonspecific interactions with the alkyne-functionalized fluorophore, could improve signal-to-noise ratios. Third, while cMLEAD enabled a live-cell screening platform for identifying candidate ALDH2 modulators, additional mechanistic studies are required to validate whether these hits act through direct or indirect modulation of ALDH2 activity.
Footnote |
† These authors contributed equally to this work. |
This journal is © The Royal Society of Chemistry 2025 |