Abstract
Solid tumours often endure nutrient insufficiency during progression. How tumour cells adapt to temporal and spatial nutrient insufficiency remains unclear. We previously identified STC2 as one of the most upregulated genes in cells exposed to nutrient insufficiency by transcriptome screening, indicating the potential of STC2 in cellular adaptation to nutrient insufficiency. However, the molecular mechanisms underlying STC2 induction by nutrient insufficiency and subsequent adaptation remain elusive. Here, we report that STC2 protein is dramatically increased and secreted into the culture media by Gln-/Glc- deprivation. STC2 promoter contains cis-elements that are activated by ATF4 and p65/RelA, two transcription factors activated by a variety of cellular stress. Biologically, STC2 induction and secretion promote cell survival but attenuate cell proliferation during nutrient insufficiency, thus switching the priority of cancer cells from proliferation to survival. Loss of STC2 impairs tumour growth by inducing both apoptosis and necrosis in mouse xenografts. Mechanistically, under nutrient insufficient conditions, cells have increased levels of reactive oxygen species (ROS), and lack of STC2 further elevates ROS levels that lead to increased apoptosis. RNA-Seq analyses reveal STC2 induction suppresses the expression of monoamine oxidase B (MAOB), a mitochondrial membrane enzyme that produces ROS. Moreover, a negative correlation between STC2 and MAOB levels is also identified in human tumour samples. Importantly, the administration of recombinant STC2 to the culture media effectively suppresses MAOB expression as well as apoptosis, suggesting STC2 functions in an autocrine/paracrine manner. Taken together, our findings indicate that nutrient insufficiency induces STC2 expression, which in turn governs the adaptation of cancer cells to nutrient insufficiency through the maintenance of redox homoeostasis, highlighting the potential of STC2 as a therapeutic target for cancer treatment.
Similar content being viewed by others
Introduction
The major characteristics of tumour metabolism include the Warburg effect, active glutaminolysis and hypoxia [1,2,3]; as such, tumour cells have increased demand of glucose (Glc), glutamine (Gln) and oxygen to keep cell survival and proliferation [4,5,6]. However, solid tumours often have poor circulation that leads to an insufficient supply of Glc, Gln and oxygen; therefore, to investigate the mechanisms of how tumour cells respond to nutrient insufficiency could provide promising therapeutic targets. Our previous study identified Stanniocalcin 2 (STC2) as one of the most upregulated genes upon Gln-/Glc- deprivation and hypoxia exposure [1, 7], suggesting a critical role of STC2 in the cellular response to nutrient insufficiency.
STC2 belongs to the secreted glycoprotein hormone family that was reported to lower intracellular calcium concentrations by attenuating Ca2+ influx [8, 9]. Under physiological conditions, STC2 is highly expressed in skeletal muscle, heart, testis and pancreas [8, 10]; while under pathological conditions, STC2 is upregulated in a plethora of human tumours, including hepatocellular carcinoma (HCC), cervical cancer, nasopharyngeal carcinoma, colorectal cancer, gastric cancer, oesophageal squamous cell carcinoma, prostate cancer, renal cell carcinoma, etc. [7, 8, 11, 12]. Increased STC2 levels are strongly correlated with tumour development, progression and poor prognosis for most human tumours except breast cancer [13]. The expression of STC2 is regulated at both transcriptional and post-transcriptional levels, particularly under stress conditions, including ER stress, hypoxia, nutrient insufficiency and radiation-mediated stress [1, 7, 8, 11, 14]. Biologically, STC2 is involved in many biological processes in human tumours, such as cell survival, proliferation, migration and immune escape [8, 15,16,17]. However, the precise mechanisms underlying the stress-induced STC2 induction remain unclear.
Previous reports reveal hydrogen peroxide (H2O2) induces STC2 expression, and loss of STC2 impairs the tolerability of cells to oxidative stress [18,19,20], suggesting a potential mechanism that links nutrient insufficiency to STC2 induction, redox homoeostasis and cellular adaption. Current understanding is that aberrant metabolic activities of tumour cells result in an elevation of reactive oxygen species (ROS) [21]. Low to moderate levels of ROS may serve as a signalling mechanism to maintain cellular physiology; increased levels of ROS facilitate tumorigenesis by causing genetic alteration and oncogenic transformation; however, long-term and/or excessive ROS may trigger apoptosis [22]. In general, the glutathione (GSH) and nicotinamide adenine dinucleotide phosphate (NADPH) systems work coordinatively to maintain intracellular redox homoeostasis [23, 24]. As for tumour cell metabolism, Gln serves as the major source of glutamate that functions as an important substrate for de novo GSH biosynthesis; hence, Gln-deprivation results in decreased GSH levels and ROS accumulation [25, 26]. Glc insufficiency may trigger oxidative stress via impaired NADPH production, because Glc is the substrate for the pentose phosphate pathway, the major pathway to generate NADPH [27]. Moreover, hypoxia impairs the electron transport chain (ETC) activity and also enhances ROS production [28, 29]. Therefore, deprivation of each of these major nutrients may trigger oxidative stress, and cells must overcome the oxidative stress to maintain survival. A better understanding of how STC2 regulates the adaptation of tumour cells to nutrient insufficiency and the associated oxidative stress may provide new therapeutic targets.
In this study, we first confirm that STC2 protein is induced and secreted into the culture media by nutrient insufficiency. We next present data to define the cis-elements in the STC2 promoter that physically interact with activating transcription factor 4 (ATF4) and nuclear factor-kappa B p65 (p65/RelA) during nutrient insufficiency. We show that STC2 induction inhibits apoptosis but attenuates tumour cell proliferation during nutrient insufficiency. In addition, loss of STC2 leads to elevated ROS levels accompanied by apoptosis during nutrient insufficiency. We finally show that STC2 induction suppresses the expression of monoamine oxidase B (MAOB), a mitochondrial membrane-associated enzyme that produces ROS, and the administration of recombinant human STC2 to culture media is sufficient to suppress MAOB expression. Taken together, our findings indicate that STC2 serves as a key regulator of redox homoeostasis that governs the survival of tumour cells during nutrient insufficiency.
Materials and methods
Ethical approval
Animal studies were performed according to the protocol approved by the Laboratory Animal Ethics Committee at the Tianjin Medical University Cancer Institute & Hospital (TMUCIH) (#: AE-2022036). This study has complied with all relevant ethical regulations for animal experiments. All efforts were made to minimise the numbers and suffering of experimental animals.
Human HCC tissue microarray (TMA) was purchased from the Shanghai Outdo Biotechnology (China) with an Internal Review Board approval (YB M-05-02). The clinical samples (23 HCC specimens) were collected from the Department of Pathology at the TMUCIH from January 1, 2018 to December 31, 2018. All patients were provided with informed consent and the study was conducted according to a protocol approved by the Internal Review Board at the TMUCIH (bc2021308).
Cell culture
Hep3B, HeLa, MCF-7, SK-BR-3, MDA-MB-453, MDA-MB-468 and HEK293T (ATCC) cells were maintained in DMEM with 10% foetal bovine serum (FBS) supplemented with Pen/Strep. All cells were cultured in a humidified incubator at 37 °C with 5% CO2. MM01 cells were established through the extended culture of Hep3B cells in Gln-free DMEM supplemented with 10% dialysed FBS (Atlantic Biologicals, dialysed 10 kDa), 0.8 mM ammonia and Pen/Strep [30]. The culture media for MM01 cells were changed every 2 days, and the cells were passed once reaching ~90% confluency. All cells were tested routinely to ensure mycoplasma-free using Polymerase Chain Reaction (PCR) methods.
Tumour xenografts
The NTG and athymic nude, nu/nu mice were purchased from SPF Biotechnology Co. (Beijing, China). For Hep3B xenografts, 24 NTG mice (male, age: 4 weeks old) were randomly allocated into three groups (6 per group). For HeLa xenografts, 24 athymic nude, nu/nu mice (female, age: 4 weeks old) were randomly allocated into three groups (6 per group). In general, 5 × 106 Hep3B or HeLa cells were subcutaneously injected into each side of both flank regions. The xenografts were measured every three days to the completion of experiments. The growth curves were plotted with tumour sizes (calculated using the formula VT = 0.5 × L × W2, mm3). After euthanization, dissected xenografts were weighted and recorded for further analysis. The mice were euthanized if the xenograft size became ≥20 mm in the long diameter, and the mice died prior to the completion of experiments were excluded for analysis.
Statistical analysis
Statistical analyses were performed using SPSS 27.0 software (IBM SPSS Statistics). The results of qRT-PCR, tumour weight, IHC staining score, and luciferase analysis were presented as average ± standard deviation; Student t test or one-way ANOVA was performed to compare these results. The curves for cell proliferation and xenograft growth were compared using two-way ANOVA. Linear correlation was assessed using Spearman’s correlation analysis. Survival analysis was performed using the log rank test to estimate the survival probability, and P value less than 0.05 was set as statistically significant.
Addition information is provided in Supplementary Materials and Methods, and the original western blot data are included in the Original Data file.
Results
Nutrient insufficiency triggers STC2 induction and secretion
In previous work, we cultured Hep3B cells for the long term in Gln-free media supplemented with NH4+ as an alternative nitrogen source and established a long-term Gln-deprived cell line, MM01, which maintains a slow proliferation phenotype [30]. In addition, we cultured Hep3B cells in Gln-/Glc- free media for short term that served as a model for acute nutrient insufficiency. cDNA microarray analysis revealed that STC2 was among the most upregulated 58 genes shared by cells enduring acute Gln-/Glc- deprivation as well as long-term Gln-deprived condition (Fig. 1A, B, Supplementary Fig. S1A and Supplementary Table 1), indicating the potential of STC2 in mediating cellular adaptation to nutrient insufficiency. Further functional analyses suggested that most of these 58 genes were related to amino acid metabolism (Supplementary Fig. S1B). Moreover, our prior study found STC2 was also upregulated by hypoxia [7], suggesting STC2 induction is a common phenomenon when tumour cells exposed to nutrient insufficiency. In accordance, elevated STC2 protein levels were confirmed in MM01 cells relative to that in Hep3B cells (Fig. 1C), and STC2 was remarkably induced by acute Gln-deprivation (Fig. 1D–F and Supplementary Fig. S1C), whereas re-supplementation with Gln rapidly suppressed STC2 expression (Supplementary Fig. S1D). Similar results were observed when Gln was added back to MM01 cells (Supplementary Fig. S1E). To address whether STC2 upregulation is a common phenomenon in different tumour types enduring nutrient insufficiency, we exposed breast cancer cells to Gln-deprivation as well. We observed that STC2 was similarly induced in MCF-7, SK-BR-3, MDA-MB-453 and MDA-MB-468 cells by Gln-free media (Fig. 1G, H). In consistent with the cDNA microarray, Glc-deprivation also increased STC2 protein levels (Fig. 1I).
To test whether STC2 is induced by the lack of Gln itself or by a lack of metabolic derivatives of Gln, we administered the glutaminase inhibitor, DON, to Hep3B cells. We observed that DON treatment efficiently induced STC2 expression in Gln-rich media (Supplementary Fig. S1F, G), indicating that lack of Gln-derived metabolites is sufficient to trigger STC2 induction as well. As an inhibitor of protein secretion, BFA increased intracellular STC2 levels while decreased STC2 levels in the culture media (Supplementary Fig. S1H), confirming STC2 as a secreted protein. To address whether increased STC2 levels in cells is the result of secretion inhibition, we analysed STC2 levels in cell lysate and their corresponding culture media. We observed that increased STC2 levels in both cell lysate and culture media in a proportional manner upon Gln-/Glc- deprivation, suggesting that increased intracellular STC2 levels are not a consequence of secretion inhibition (Fig. 1J–M). Taken together, these data suggest that nutrient insufficiency triggers STC2 induction and secretion, which represents a converging consequence in response to different nutrient-insufficient conditions.
ATF4 and p65/RelA transcriptionally induce STC2 expression during nutrient insufficiency
Our previous studies indicated that STC2 mRNA levels were increased in cells under Gln-/Glc- deprived or hypoxic conditions [1, 8], suggesting STC2 is transcriptionally upregulated by nutrient insufficiency. To further delineate the underlying mechanisms, we used ActD and CHX to block gene transcription and protein translation, respectively. ActD and CHX blocked STC2 expression at both mRNA and protein levels (Supplementary Fig. S2A,B), demonstrating that STC2 transcription depends on de novo synthesis of a transcription factor. In addition, to address whether Gln-deprivation affects STC2 protein stability, we performed CHX chase assay and found that after CHX treatment, STC2 showed a similar decay rate regardless of Gln levels (Supplementary Fig. S2C,D), ruling out the possibility that Gln-deprivation stabilises STC2 protein.
STC2 was reported to be induced by ER stress that is correlated with ATF4 activation. ATF4 is an important component of the integrated stress response system. Amino acid insufficiency leads to the accumulation of non-aminoacylated tRNAs, which promotes the activation of general control non-derepressible-2 (GCN2) and the phosphorylation of eIF2α, thereafter triggering the selective translation of ATF4 [31, 32]. To determine whether STC2 gene is a direct transcription target of ATF4, we performed bioinformatic analysis and found STC2 promoter contains multiple copies of putative ATF4, NF-κB and HIF-1 binding sites (HRE) (Fig. 2A). Next, we confirmed that Gln-deprivation enhanced eIF2α phosphorylation and ATF4 induction in our experimental setting (Supplementary Fig. S2E,F). In addition, ATF4 knockdown effectively abolished STC2 induction in cells cultured in Gln-free media (Fig. 2B). Indeed, ER stress inducers like MG132 which blocks proteosome-dependent degradation of proteins, or BFA which suppresses protein secretory pathway, induced STC2 expression as well; while ActD or CHX effectively blocked this induction (Supplementary Fig. S2G–J), suggesting ER stress-triggered STC2 induction depends on de novo transcription and translation. Finally, glutaminase inhibitor, DON or Glc-deprivation also induced a co-upregulation of ATF4 and STC2 (Supplementary Fig. S2K, L).
In consistence with the existence of putative NF-κB binding sites in the STC2 promoter, GSEA analyses highlighted the upregulation of NF-κB targets under Gln-insufficient conditions (Supplementary Fig. S3A and Supplementary Table 2). To evaluate the role of NF-κB in STC2 induction, we performed cell fractionation analysis and immunofluorescent staining and observed that Gln-deprivation triggered p65/RelA accumulation in the nuclei (Fig. 2C, D), demonstrating the activation of p65/RelA signalling in Gln-deprived cells. Next, we applied chemical inhibitors or activators to test the importance of p65/RelA in mediating STC2 expression upon Gln-deprivation. We observed that NF-κB inhibitors, BAY 11-7082 or curcumin, efficiently blocked STC2 upregulation (Supplementary Fig. S3B–G). Furthermore, genetically suppressing p65/RelA by siRNA or IκBαSR attenuated STC2 induction by Gln-deprivation (Fig. 2E and Supplementary Fig. S3H). On the other hand, broadly used NF-κB activators, PMA or TNF-α, induced STC2 that could be blocked by curcumin (Fig. 2F, G). Taken together, these data demonstrate that p65/RelA functions as another transcription activator for STC2 induction.
To define the cis-elements that regulate STC2 expression in response to Gln-deprivation, we cloned two promoter fragments into the pGL3-Luciferase reporter with a length of 2026 bp (P2.0) and 1412 bp (P1.4), respectively (Fig. 2A). Guided by the identified putative binding sites, a series of deletions were constructed and cloned into the pGL3-Luciferase reporter. Luciferase assays indicated a DNA region included in STC2-P2.0 but not in STC2-P1.4 is responsive to Gln-/Glc- deprivation or hypoxia (Supplementary Fig. S4A–C). Further deletion assays narrowed down to Deletion 4 (Del4) region that is indispensable for the response of STC2 promoter to nutrient insufficiency (Fig. 2H and Supplementary Fig. S4D). Mutation of either ATF4 or NF-κB binding sites in this region decreased the stress response (Fig. 2I and Supplementary Fig. S4E), further confirming that these binding sites are functionally important in regulating STC2 induction. Finally, ChIP assays confirmed the direct binding of ATF4 and p65/RelA to this region of STC2 promoter (Fig. 2J).
STC2 induction compromises tumour cell proliferation under nutrient-insufficient conditions
Our previous study revealed that Gln-/Glc- deprivation caused cell cycle arrest, hence attenuating cell proliferation [1]. Since STC2 is induced by nutrient insufficiency, it is intriguing to investigate the role of STC2 in regulating cell proliferation under such conditions. We next focused on Gln-deprivation as a model to investigate the role of STC2 in regulating cell proliferation. In regular media, STC2 knockdown impaired the optimal cell proliferation (Fig. 3A, C and Supplementary Fig. S5A), while in Gln-free media, STC2 knockdown cells showed higher proliferation rates (Fig. 3B, D and Supplementary Fig. S5B), indicating that basal levels of STC2 are required to maintain the optimal proliferation when cells cultured in nutrient-rich media, whereas under nutrient insufficient conditions, elevated STC2 levels can suppress cell proliferation. Interestingly, overexpression of STC2 did not affect cell proliferation of cells cultured in either regular media or nutrient-insufficient media (Fig. 3E–H and Supplementary Fig. S5C, D). Furthermore, supplementing exogenous recombinant STC2 (rSTC2) to media did not affect cell proliferation under either conditions (Fig. 3I, J and Supplementary Fig. S5E). Taken together, these data demonstrate that basal levels of STC2 are required to support cell proliferation under nutrient-rich conditions; while endogenous STC2 gene can be induced by nutrient insufficiency, thereby attenuating cell proliferation and facilitating cellular adaptation to stress conditions.
STC2 induction and secretion promote tumour cell survival during nutrient insufficiency
One common consequence of nutrient insufficiency and associated metabolic stress is apoptosis. We next focused on Gln-deprivation as a model to investigate whether STC2 induction played a role in maintaining cell survival during nutrient insufficiency. Western blot revealed STC2 knockdown increased PARP cleavage, a biomarker of apoptosis, under Gln-deprived conditions (Fig. 4A, B and Supplementary Fig. S6A). In accordance, flow cytometry analysis also supported that cells with STC2 knockdown were more sensitive to apoptosis triggered by Gln-deprivation (Fig. 4C, D and Supplementary Fig. S6C). On the other hand, overexpression of STC2 decreased apoptosis of cells exposed to Gln-free media (Fig. 4E–H and Supplementary Fig. S6B, D). As a secretory protein, STC2 has been proposed to function as a signalling molecule or to function intracellularly as a calcium regulator [9]. To test whether STC2 protects cells from apoptosis triggered by Gln-deprivation in an autocrine/paracrine manner, we added rSTC2 to cell culture media and found that rSTC2 efficiently suppressed PARP cleavage as well as apoptosis of cells cultured in Gln-free media (Fig. 4I, J and Supplementary Figs. S6E, F and S7). These data support that STC2 may function in an autocrine/paracrine manner to promote cell survival under nutrient insufficient conditions.
STC2 knockdown retards the growth of mouse xenografts
To evaluate the role of STC2 in a physiological context, we first run a large-scale Kaplan-Meier Plotter analysis and found that elevated STC2 levels were associated with poor prognosis in patients with a variety of tumours including HCC, cervical squamous cell carcinoma, thymoma, oesophageal squamous cell carcinoma, renal papillary cell carcinoma, stomach adenocarcinoma, sarcoma, head and neck squamous cell carcinoma, bladder cancer and lung squamous cell carcinoma (Supplementary Fig. S8). To confirm the role of STC2 induction in tumour progression in vivo, we established mouse xenografts by injecting 5 × 106 Hep3B or HeLa cells with STC2 knockdown or control shRNAs independently. We found that STC2 knockdown tumours grew slower than control ones (Fig. 5A, B, G, H). Moreover, STC2 knockdown xenografts overall had lighter tumour weights compared to control counterparts (Fig. 5C, I). To further determine the role of STC2 in cell survival and proliferation in vivo, we examined the expression of cleaved caspase-3 and Ki-67 in the dissected xenografts by IHC staining as markers of apoptosis and proliferation, respectively. We found that loss of STC2 led to increased caspase-3 cleavage, suggesting STC2 induction is a favourable factor for cell survival in vivo (Fig. 5D, J and Supplementary Fig. S9A), while there was no obvious difference in cell proliferation rates evaluated by Ki-67 IHC staining (Fig. 5E, K and Supplementary Fig. S9B). More importantly, larger necrotic areas were observed in STC2 knockdown xenografts compared to control ones (Fig. 5F, L), suggesting a failed adaptation to insufficient blood supply during tumour progression, which is consistent with a previous report that STC2 was involved in tumour angiogenesis [33]. These data highlight that STC2 plays a critical role in preventing cell death during tumour progression in vivo.
STC2 knockdown results in ROS elevation in cells under nutrient-insufficient conditions
Next, we asked how STC2 protects tumour cells from nutrient insufficiency-induced apoptosis. To address this question, we first performed RNA-Seq analysis to search for alterations of signalling pathways in STC2 knockdown versus control cells cultured in Gln-free media (Fig. 6A). The analysis of the RNA-Seq data identified an alteration of genes related to “hallmark gene sets” (Supplementary Fig. S10); particularly, both shRNAs targeting STC2 resulted in an elevation of genes related to redox homoeostasis under Gln-deprived conditions (Fig. 6B). We next determined how STC2 status affected ROS levels in cells exposed to Gln-deprivation. Using dihydroethidium (DHE) staining combined with flow cytometry analyses, we observed that Gln-deprivation moderately increased ROS levels, and STC2 knockdown significantly intensified ROS accumulation (Fig. 6C). Moreover, the administration of rSTC2 to culture media obviously decreased ROS levels in STC2 knockdown cells (Fig. 6D). Taken together, these data indicate that STC2 induction and secretion are required to ameliorate ROS elevation triggered by nutrient insufficiency.
STC2 knockdown leads to MAOB dysregulation and redox imbalance during nutrient insufficiency
To confirm a causative relationship between ROS accumulation and apoptosis in our experimental model, we administered NAC or GSH to STC2 knockdown cells. Not surprisingly, both NAC and GSH suppressed apoptosis of STC2 knockdown cells enduring Gln-deprivation (Fig. 7A, B and Supplementary Figs. S7 and S11), indicating increased ROS is a bona fide mediator of apoptosis in our experimental setting. To explore the molecular mechanism underlying the protective effects of upregulated STC2 in response to nutrient insufficiency, we mined the RNA-Seq data and found that MAOB, a mitochondrial membrane-associated enzyme that directly participates in ROS production, was among the genes most upregulated by STC2 knockdown (Fig. 7C and Supplementary Table 3). Subsequent qRT-PCR and western blot assays further confirmed that STC2 knockdown increased MAOB expression under Gln-deprived conditions (Fig. 7D, E), whereas no significant alteration of expression levels of other genes related to ROS generation was found in two independent STC2 knockdown cell lines. Particularly, no signal was detected for NADPH oxidase (NOX) family genes in both STC2 knockdown cell lines (Supplementary Fig. S12). In accordance, we did not find a significant change in mitochondrial ROS in STC2 knockdown cells (Supplementary Fig. S13).
To further explore the regulatory relationship between STC2 and MAOB, we applied rSTC2 to cells with STC2 knockdown. We observed that rSTC2 effectively decreased MAOB expression in STC2 knockdown cells in Gln-free media (Supplementary Fig. S14). To substantiate the biological importance of MAOB expression to ROS levels and cell survival, we used siRNA pool to knockdown MAOB, or specific inhibitors to suppress MAOB activity in STC2 knockdown cells. We observed that MAOB knockdown efficiently decreased ROS levels in STC2 knockdown cells cultured in Gln-free media (Fig. 7F, G and Supplementary Fig. S15A, B). In addition, MAOB knockdown not only suppressed apoptosis but also rescued cell proliferation (Fig. 7H, I and Supplementary Fig. S15C, D). To further evaluate whether MAOB is associated with STC2 loss, we employed two specific MAOB inhibitors, Rasagiline and Selegiline. Consistently, both inhibitors effectively attenuated ROS production and apoptosis in STC2 knockdown cells cultured in Gln-free media (Supplementary Fig. S16A–D), thereby rescuing cell proliferation (Supplementary Fig. S16E, F). As a metabolic precursor or substrate, the lack of Gln showed a detrimental effect on GSH levels. MAOB inhibitors, Rasagiline and Selegiline, slightly increased GSH levels, but in some experiments, these changes barely meet the criteria of statistical significance. Similarly, MAOB knockdown did not significantly change GSH levels (unpublished data), indicating MAOB is more likely a fine tuner of the GSH/GSSG ratio, which is reflected by the alteration of intracellular ROS levels and controls the survival or death of cells enduring oxidative stress. Taken together, these findings indicate that under nutrient-insufficient conditions, STC2 induction maintains redox homoeostasis mainly by suppressing MAOB expression, thus reducing intracellular ROS levels and maintaining cell survival.
The expression levels of STC2 and MAOB are negatively correlated, and elevated STC2 is associated with tumour progression in vivo
To address whether the observation in cell culture is relevant to tumour progression in vivo, we first performed IHC staining and confirmed the elevated expression of MAOB in STC2 knockdown xenografts (Fig. 8A–C). Furthermore, DepMap analyses suggested a reverse correlation between STC2 and MAOB expression in a series of human tumour cells (Supplementary Fig. S17A). Other bioinformatic analyses also revealed a negative correlation between STC2 and MAOB expression in human tumours (Supplementary Fig. S17B–F).
To investigate the potential of using the reciprocal STC2-MAOB expression as a prognostic indicator, we first performed immunofluorescent staining to examine their expression correlation in human HCC specimens. In consistency with cell culture findings, we confirmed an obviously negative correlation between STC2 and MAOB levels in the same HCC samples (Fig. 8D). IHC staining also verified a reverse correlation between STC2 and MAOB expression in human HCC tissues but not in normal counterparts (Fig. 8E–H and Supplementary Fig. S18). Importantly, clinicopathological analyses revealed that elevated STC2 expression was significantly associated with poor prognosis in patients with HCC, whereas increased MAOB levels were likely correlated with a favourable prognosis with a log-rank P value of 0.062 (Fig. 8I, J). Taken together, these data strongly indicate that STC2 negatively regulates MAOB expression via an un-characterised mechanism (Fig. 8K, L), and a combined analysis of both STC2 and MAOB levels is a potential approach in assessing the prognosis of patients with HCC.
Discussion
The rapid proliferation of tumour cells demands an increased supply of nutrients to support proliferation and active biosynthesis, while a defective blood vessel system often fails to support solid tumour cells with sufficient blood supply, leading to nutrient insufficiency and, subsequently, oxidative stress during tumour progression. Our previous study revealed that STC2 is one of the most upregulated genes under Gln-/Glc- deprived and hypoxic conditions [1]. With these data in context, our study focuses on addressing how STC2 is upregulated by nutrient insufficiency, and the biological roles of STC2 in the adaptation of tumour cells to nutrient insufficiency.
We presented data to conclude that ATF4 and p65/RelA are activated by nutrient insufficiency that in turn transactivates STC2 gene. Previous studies reported that STC2 was upregulated upon ATF4 activation [8, 18], but it was unclear whether ATF4 directly transactivates STC2 gene, or indirectly regulates its expression. Our current study identifies the cis-elements of the STC2 gene and demonstrates that these elements are indispensable for STC2 induction upon nutrient deprivation. It is well known that eIF2α-ATF4 axis represents the core of an integrated stress response mechanism [34]. Under various stress conditions, cells utilise multiple kinases to phosphorylate eIF2α. Phosphorylation of eIF2α results in a global translation inhibition but a selective translation of stress-related genes exemplified by ATF4. As a stress-responsive transcription factor, ATF4 transcribes several functional groups of genes, including ER chaperones that are involved in protein folding, ER-associated degradation of terminally damaged proteins, and apoptosis. In addition to ATF4, NF-κB is another transcription factor ubiquitously involved in cellular stress response. It was suggested that Gln-deprivation activated NF-κB signalling [35]. Consistent with that report, our findings demonstrate that p65/RelA, a member of NF-κB family, directly transactivates STC2 gene by a physical interaction with identified cis-elements upon activation. Considering NF-κB is activated by various cell injury, pathogen infection and inflammatory signalling, we deduce that STC2 could also be induced by a variety of stress, including cell injury and inflammatory factors.
Elevated ROS is a common result of many cellular stress and disease status, and is a major contributor to oxidative stress imbalance. ROS is mainly generated in the mitochondria as a byproduct of oxidative phosphorylation or in the cytosol from reactions catalysed by oxidoreductases such as NOX [36]. To some extent, ROS possesses beneficial effects for tumour cells, driving cell proliferation, genetic instability and metastasis [37]. However, excessive and/or long-term ROS may lead to the damage of macromolecules, and eventually cell death. From the nutrient point of view, Gln and Glc play critical roles in the intracellular counter-oxidative stress processes [25, 26]. Gln is the direct precursor of glutamate; both Gln and glutamate play central roles as a nitrogen donor in the biosynthesis of non-essential amino acids, including cysteine and glycine. Glutamate, cysteine and glycine are the direct substrates for GSH biosynthesis. Indeed, we observed that Gln-/Glc- deprivation altered the amino acid metabolic pathway (Supplementary Fig. S1B), reflecting a feedback response to global amino acid insufficiency that directly impair the biosynthesis of GSH and possibly other antioxidants. Our data revealed that tumour cells exposed to Gln-free media have elevated ROS levels, and STC2 knockdown further increases ROS levels, suggesting that STC2 plays a role in maintaining redox homoeostasis. RNA-Seq analyses also revealed that the change of ROS metabolic signature is one of the most affected pathways in STC2 knockdown cells upon Gln-deprivation, further providing additional evidence to our hypothesis.
Previous studies indicated a connection between oxidative stress and STC2 induction. For example, H2O2 treatment leads to STC2 upregulation, and loss of STC2 causes cell death when challenged with oxidative stress [18,19,20]. We note that a variety of cell stress, including nutrient insufficiency, may trigger oxidative stress and ER stress. Accordingly, as a primary or a secondary alteration, oxidative stress itself may be sufficient to independently activate ATF4 and NF-kB, hence upregulating STC2.
Whereas STC1 has been reported to regulate redox homoeostasis by promoting the expression of uncoupling protein 2, thus compromising the ETC-related ROS production [38, 39], our findings in this study suggest that STC2 utilises a novel mechanism to maintain the cellular redox homoeostasis during nutrient insufficiency, i.e., STC2 induction by nutrient insufficiency suppresses MAOB expression to decrease ROS production. MAOB belongs to the family of monoamine oxidase (MAO) enzymes that localise in the outer mitochondrial membrane to catalyse the oxidation of various monoamines [40]. MAOB is mainly expressed in liver, heart, duodenum, lung and brain [41]. Under physiological conditions, there is a clear increase of MAOB expression in aging or inflammatory brains [42, 43]. The reaction catalysed by MAOB is regarded as a source of H2O2 that is involved in ROS-mediated cellular injury in neurological and cardiac disorders [44,45,46,47]. In consistent with the literature, our data indicate that STC2 suppresses MAOB expression, whereas loss of STC2 leads to MAOB induction, which coincides with elevated ROS levels and cell death upon Gln-deprivation, highlighting the role of MAOB as a redox regulator during nutrient insufficiency.
Besides its functions in catalysing monoamine, it remains unclear whether MAOB directly affects the activity of ETC, the major source of ROS production. A previous study reported that MAOA, another member of the MAO family, is involved in regulating mitochondrial bioenergetics, and the MAOA-specific inhibitor, clorgyline, enhances mitochondrial ROS production at high concentrations by increasing cellular respiration [48].
Importantly, this study found the administration of rSTC2 can efficiently block Gln-deprivation-induced apoptosis, suggesting STC2 may perform its biological functions in an autocrine/paracrine manner. Till now, the majority of investigations have focused on utilising the genetically overexpressing systems to investigate the biological functions of STC2 to establish its role in regulating cell proliferation, migration, invasion and apoptosis [8]. But as a secreted glycoprotein, overexpression of STC2 leads to increased secretion when it has its own signal peptide (our unpublished data). Therefore, it’s unreasonable to simply attribute the biological effects of STC2 overexpression to its intracellular functions [49]. Our study found rSTC2 efficiently blocks MAOB induction, resulting in reduced ROS production and apoptosis under Gln-deprived conditions. These findings strongly suggest STC2 works in an autocrine/paracrine manner to regulate cellular processes. A recent study found the administration of rSTC2 promotes epithelial-mesenchymal transition by inducing the expression of snail family transcription repressor 2 and matrix metalloproteinases [16]. Recently, several clinicopathological studies have identified serum STC2 levels are elevated in patients with HCC, gastric cancer and colorectal cancer, and particularly high serum STC2 levels are associated with poor prognosis [20, 50,51,52], indicating secreted STC2 could perform its biological functions through an endocrine mechanism at organism levels. It has been reported that the cation-independent mannose-6-phosphate receptor/IGF2R interacts with STC1 and may serve as a receptor [53]. Whether STC2 shares the same receptor or uses a different surface protein as a receptor remains to be investigated.
Although a plethora of clinicopathological investigations indicate that STC2 expression is correlated with tumour size, disease stage and prognosis of various human tumours, that is also demonstrated by the bioinformatic analyses of the TCGA data in our study [8], it remains unclear how STC2 induction could biologically benefit tumour growth. This study provides new insights into our understanding of the role of STC2: serving as a critical regulator to coordinate cell survival and proliferation under nutrient-insufficient conditions. Specifically, STC2 is required to support optimal cell proliferation under normal conditions, indicating basal levels of intracellular STC2 facilitate cell proliferation, which might be attributed to its function in maintaining intracellular calcium homoeostasis (Fig. 8K, L). Under nutrient-insufficient conditions, STC2 slows down cell proliferation which might be more beneficial for tumour cells by allocating limited resources to fulfil their basic demands, such as maintaining cell survival. A potential explanation might be that various types of cellular stress triggers STC2 induction and secretion into the culture media; and the secreted STC2 serves as a ligand to activate downstream signalling, which subsequently slows down cell proliferation (Fig. 8K, L). Therefore, STC2 serves as a switch of cell priority: under normal conditions, basal levels of STC2 facilitate maintaining normal cell biology and functions; under stress conditions, STC2 is upregulated and secreted, thereby triggering a stress response that shifts cellular priority to survival.
It is an interesting question regarding the roles of STC2 in non-cancer cells. After careful reviewing of available literature and our own data, we propose that STC2 functions as a critical regulator of cellular adaptation to intracellular/extracellular metabolic state. Under normal conditions, the basal level of STC2 is likely to be an ER resident protein that supports specialised normal cell function via regulating calcium homoeostasis and dynamics. Under various stress conditions, transcriptionally induced STC2 is predominantly secreted as a signalling molecule to suppress non-survival related cellular functions to save resources, thus shifting the cellular priority to survival. This should confer the survival advantage of normal cells during microenvironmental challenges, but in cancers, it may facilitate therapeutical resistance by slowing down proliferation and promoting cell survival. Detailed physiological functions and signalling pathways of STC2 deserve further investigation.
In summary, as a secreted glycoprotein, STC2 is quickly and remarkably transactivated by ATF4 and p65/RelA in cells enduring nutrient insufficiency. As a signalling molecule, secreted STC2 functions as a ligand in an autocrine/paracrine manner to promote cell survival by alleviating oxidative stress. One possible mechanism is that STC2 suppresses MAOB expression at the transcriptional level, thus decreasing ROS production. Although the receptor of STC2 and its downstream signalling remain to be dissected, our current findings support a model that STC2 switches cancer cells’ priority from proliferation to survival, hence facilitating cellular adaptation to nutrient insufficiency. Therefore, STC2 can be regarded as a prognostic biomarker and a promising therapeutic target for cancer treatment.
Data availability
All datasets generated are included in the main text and its Supplementary Information files. Additional data are available from SQ (shuoqie@tmu.edu.cn) upon reasonable request.
References
Qie S, Liang D, Yin C, Gu W, Meng M, Wang C, et al. Glutamine depletion and glucose depletion trigger growth inhibition via distinctive gene expression reprogramming. Cell Cycle. 2012;11:3679–90.
Qie S, Chu C, Li W, Wang C, Sang N. ErbB2 activation upregulates glutaminase 1 expression which promotes breast cancer cell proliferation. J Cell Biochem. 2014;115:498–509.
Qie S, Yoshida A, Parnham S, Oleinik N, Beeson GC, Beeson CC, et al. Targeting glutamine-addiction and overcoming CDK4/6 inhibitor resistance in human esophageal squamous cell carcinoma. Nat Commun. 2019;10:1296.
Qie S, Diehl JA. Glutamine addiction: an Achilles heel in esophageal cancers with dysregulation of CDK4/6. Mol Cell Oncol. 2019;6:1610257.
Qie S, He D, Sang N. Overview of glutamine dependency and metabolic rescue protocols. Methods Mol Biol. 2019;1928:427–39.
Wu S, Fukumoto T, Lin J, Nacarelli T, Wang Y, Ong D, et al. Targeting glutamine dependence through GLS1 inhibition suppresses ARID1A-inactivated clear cell ovarian carcinoma. Nat Cancer. 2021;2:189–200.
He H, Qie S, Guo Q, Chen S, Zou C, Lu T, et al. Stanniocalcin 2 (STC2) expression promotes post-radiation survival, migration and invasion of nasopharyngeal carcinoma cells. Cancer Manag Res. 2019;11:6411–24.
Qie S, Sang N. Stanniocalcin 2 (STC2): a universal tumour biomarker and a potential therapeutical target. J Exp Clin Cancer Res. 2022;41:161.
Zeiger W, Ito D, Swetlik C, Oh-hora M, Villereal ML, Thinakaran G. Stanniocalcin 2 is a negative modulator of store-operated calcium entry. Mol Cell Biol. 2011;31:3710–22.
Hernandez Cordero AI, Gonzales NM, Parker CC, Sokolof G, Vandenbergh DJ, Cheng R, et al. Genome-wide associations reveal human-mouse genetic convergence and modifiers of myogenesis, CPNE1 and STC2. Am J Hum Genet. 2019;105:1222–36.
Lin S, Guo Q, Wen J, Li C, Lin J, Cui X, et al. Survival analyses correlate stanniocalcin 2 overexpression to poor prognosis of nasopharyngeal carcinomas. J Exp Clin Cancer Res. 2014;33:26.
Bu Q, Deng Y, Wang Q, Deng R, Hu S, Pei Z, et al. STC2 is a potential biomarker of hepatocellular carcinoma with its expression being upregulated in Nrf1alpha-deficient cells, but downregulated in Nrf2-deficient cells. Int J Biol Macromol. 2023;253:127575.
Chatterji S, Krzoska E, Thoroughgood CW, Saganty J, Liu P, Elsberger B, et al. Defining genomic, transcriptomic, proteomic, epigenetic, and phenotypic biomarkers with prognostic capability in male breast cancer: a systematic review. Lancet Oncol. 2023;24:e74–e85.
Jiang K, Yin X, Zhang Q, Yin J, Tang Q, Xu M, et al. STC2 activates PRMT5 to induce radioresistance through DNA damage repair and ferroptosis pathways in esophageal squamous cell carcinoma. Redox Biol. 2023;60:102626.
Zhong Q, Chu H, Wang G, Zhang C, Li R, Guo F, et al. Structural insights into the covalent regulation of PAPP-A activity by proMBP and STC2. Cell Discov. 2022;8:137.
Yun EJ, Kim D, Hsieh JT, Baek ST. Stanniocalcin 2 drives malignant transformation of human glioblastoma cells by targeting SNAI2 and matrix metalloproteinases. Cell Death Discov. 2022;8:308.
Chen X, Liu Q, Huang W, Cai C, Xia W, Peng Y, et al. Stanniocalcin-2 contributes to mesenchymal stromal cells attenuating murine contact hypersensitivity mainly via reducing CD8(+) Tc1 cells. Cell Death Dis. 2018;9:548.
Ito D, Walker JR, Thompson CS, Moroz I, Lin W, Veselits ML, et al. Characterization of stanniocalcin 2, a novel target of the mammalian unfolded protein response with cytoprotective properties. Mol Cell Biol. 2004;24:9456–69.
Joshi AD, Carter DE, Harper TA Jr, Elferink CJ. Aryl hydrocarbon receptor-dependent stanniocalcin 2 induction by cinnabarinic acid provides cytoprotection against endoplasmic reticulum and oxidative stress. J Pharm Exp Ther. 2015;353:201–12.
Huang FY, Wong DK, Mak LY, Cheung TT, Seto WK, Yuen MF. Hepatitis B virus X protein promotes hepatocarcinogenesis via the activation of HMGA2/STC2 signaling to counteract oxidative stress-induced cell death. Carcinogenesis. 2022;43:671–81.
Sies H, Belousov VV, Chandel NS, Davies MJ, Jones DP, Mann GE, et al. Defining roles of specific reactive oxygen species (ROS) in cell biology and physiology. Nat Rev Mol Cell Biol. 2022;23:499–515.
Badgley MA, Kremer DM, Maurer HC, DelGiorno KE, Lee HJ, Purohit V, et al. Cysteine depletion induces pancreatic tumor ferroptosis in mice. Science. 2020;368:85–89.
Son J, Lyssiotis CA, Ying H, Wang X, Hua S, Ligorio M, et al. Glutamine supports pancreatic cancer growth through a KRAS-regulated metabolic pathway. Nature. 2013;496:101–5.
Hayes JD, Dinkova-Kostova AT, Tew KD. Oxidative stress in cancer. Cancer Cell. 2020;38:167–97.
Jiang B, Zhang J, Zhao G, Liu M, Hu J, Lin F, et al. Filamentous GLS1 promotes ROS-induced apoptosis upon glutamine deprivation via insufficient asparagine synthesis. Mol Cell. 2022;82:1821–35.
Li M, Thorne RF, Shi R, Zhang XD, Li J, Li J, et al. DDIT3 directs a dual mechanism to balance glycolysis and oxidative phosphorylation during glutamine deprivation. Adv Sci. 2021;8:e2003732.
Yang C, Li B. Maintaining proton homeostasis is an essential role of the Warburg effect in proliferating cells. Cancer Biol Med. 2019;16:391–5.
Lee P, Chandel NS, Simon MC. Cellular adaptation to hypoxia through hypoxia inducible factors and beyond. Nat Rev Mol Cell Biol. 2020;21:268–83.
Martinez-Reyes I, Diebold LP, Kong H, Schieber M, Huang H, Hensley CT, et al. TCA cycle and mitochondrial membrane potential are necessary for diverse biological functions. Mol Cell. 2016;61:199–209.
Meng M, Chen S, Lao T, Liang D, Sang N. Nitrogen anabolism underlies the importance of glutaminolysis in proliferating cells. Cell Cycle. 2010;9:3921–32.
Bader DA, Hartig SM, Putluri V, Foley C, Hamilton MP, Smith EA, et al. Mitochondrial pyruvate import is a metabolic vulnerability in androgen receptor-driven prostate cancer. Nat Metab. 2019;1:70–85.
Tameire F, Verginadis II, Leli NM, Polte C, Conn CS, Ojha R, et al. ATF4 couples MYC-dependent translational activity to bioenergetic demands during tumour progression. Nat Cell Biol. 2019;21:889–99.
Law AY, Wong CK. Stanniocalcin-1 and -2 promote angiogenic sprouting in HUVECs via VEGF/VEGFR2 and angiopoietin signaling pathways. Mol Cell Endocrinol. 2013;374:73–81.
Kartha N, Gianopulos JE, Schrank Z, Cavender SM, Dobersch S, Kynnap BD, et al. Sirtuin 6 is required for the integrated stress response and resistance to inhibition of transcriptional cyclin-dependent kinases. Sci Transl Med. 2023;15:eabn9674.
Bobrovnikova-Marjon EV, Marjon PL, Barbash O, Jagt DLV, Abcouwer SF. Expression of angiogenic factors vascular endothelial growth factor and interleukin-8/CXCL8 is highly responsive to ambient glutamine availability: role of nuclear Factor-kappa B and activating protein-1. Cancer Res. 2004;64:4858–69.
Kong H, Reczek CR, McElroy GS, Steinert EM, Wang T, Sabatini DM, et al. Metabolic determinants of cellular fitness dependent on mitochondrial reactive oxygen species. Sci Adv. 2020;6:eabb7272.
Kong N, Zhang H, Feng C, Liu C, Xiao Y, Zhang X, et al. Arsenene-mediated multiple independently targeted reactive oxygen species burst for cancer therapy. Nat Commun. 2021;12:4777.
Ohkouchi S, Block GJ, Katsha AM, Kanehira M, Ebina M, Kikuchi T, et al. Mesenchymal stromal cells protect cancer cells from ROS-induced apoptosis and enhance the Warburg effect by secreting STC1. Mol Ther. 2012;20:417–23.
Ono M, Ohkouchi S, Kanehira M, Tode N, Kobayashi M, Ebina M, et al. Mesenchymal stem cells correct inappropriate epithelial-mesenchyme relation in pulmonary fibrosis using stanniocalcin-1. Mol Ther. 2015;23:549–60.
Tripathi RKP, Ayyannan SR. Monoamine oxidase-B inhibitors as potential neurotherapeutic agents: An overview and update. Med Res Rev. 2019;39:1603–706.
Costa-Machado LF, Garcia-Dominguez E, McIntyre RL, Lopez-Aceituno JL, Ballesteros-Gonzalez A, Tapia-Gonzalez A, et al. Peripheral modulation of antidepressant targets MAO-B and GABAAR by harmol induces mitohormesis and delays aging in preclinical models. Nat Commun. 2023;14:2779.
Clarke LE, Liddelow SA, Chakraborty C, Munch AE, Heiman M, Barres BA. Normal aging induces A1-like astrocyte reactivity. Proc Natl Acad Sci USA. 2018;115:E1896–E1905.
Park JH, Ju YH, Choi JW, Song HJ, Jang BK, Woo J, et al. Newly developed reversible MAO-B inhibitor circumvents the shortcomings of irreversible inhibitors in Alzheimer’s disease. Sci Adv. 2019;5:eaav0316.
Kaludercic N, Carpi A, Nagayama T, Sivakumaran V, Zhu G, Lai EW, et al. Monoamine oxidase B prompts mitochondrial and cardiac dysfunction in pressure overloaded hearts. Antioxid Redox Signal. 2014;20:267–80.
Wu Z, Xia Y, Wang Z, Su Kang S, Lei K, Liu X, et al. C/EBPbeta/delta-secretase signaling mediates Parkinson’s disease pathogenesis via regulating transcription and proteolytic cleavage of alpha-synuclein and MAOB. Mol Psychiatry. 2021;26:568–85.
Gabilondo AM, Hostalot C, Garibi JM, Meana JJ, Callado LF. Monoamine oxidase B activity is increased in human gliomas. Neurochem Int. 2008;52:230–4.
Yang YC, Chien MH, Lai TC, Su CY, Jan YH, Hsiao M, et al. Monoamine oxidase B expression correlates with a poor prognosis in colorectal cancer patients and is significantly associated with epithelial-to-mesenchymal transition-related gene signatures. Int J Mol Sci. 2020;21:2813.
Kalimon OJ, Vekaria HJ, Gerhardt GA, Sullivan PG. Inhibition of monoamine oxidase-a increases respiration in isolated mouse cortical mitochondria. Exp Neurol. 2023;363:114356.
Jepsen MR, Kloverpris S, Mikkelsen JH, Pedersen JH, Fuchtbauer EM, Laursen LS, et al. Stanniocalcin-2 inhibits mammalian growth by proteolytic inhibition of the insulin-like growth factor axis. J Biol Chem. 2015;290:3430–9.
Wu Z, Cheng H, Liu J, Zhang S, Zhang M, Liu F, et al. The oncogenic and diagnostic potential of stanniocalcin 2 in hepatocellular carcinoma. J Hepatocell Carcinoma. 2022;9:141–55.
Fang Z, Tian Z, Luo K, Song H, Yi J. Clinical significance of stanniocalcin expression in tissue and serum of gastric cancer patients. Chin J Cancer Res. 2014;26:602–10.
Li H, Zhou X, Zhang H, Jiang J, Fu H, Wang F. Combined efficacy of CXCL5, STC2, and CHI3L1 in the diagnosis of colorectal cancer. J Oncol. 2022;2022:7271514.
Wan HT, Ng AH, Lee WK, Shi F, Wong CK. Identification and characterization of a membrane receptor that binds to human STC1. Life Sci Alliance. 2022;5:e202201497.
Acknowledgements
The authors thank Drs. J Jiang and E Fisher (Drexel University) for their kind assistance in flow cytometry analysis. The authors also thank Dr. F Elefant for the helpful discussion on chromatin immunoprecipitation assay. Thanks for the assistance from all other members in Dr. Qie laboratory.
Funding
This work is partly supported by grant R01-CA129494 (NS) from NIH/NCI and a research contract fund from Lipogen (NS), the National Natural Science Foundation of China (SQ, 82373360), the Tianjin Municipal Education Commission Key Project Program (SQ, 2022ZD065), the Tianjin Medical University Cancer Institute and Hospital Talent Program and Start-up Grant for Doctors (SQ, B2102), the Start-up funding from the Tianjin Medical University Cancer Institute and Hospital (SQ, 2021-1-7), and the Tianjin Key Medical Discipline (Pathology) Construction Project (TJYXZDXK-012A).
Author information
Authors and Affiliations
Contributions
SQ and NS proposed and designed the experiments in this manuscript. SQ, HX, YL, CY, YW, LT and CW performed experiments and collected data. SQ, NS, CW, LT, HX, YL, CY and YW analysed and interpreted the data. SQ, HX, YL and NS wrote and edited the manuscript.
Corresponding authors
Ethics declarations
Competing interests
The authors declare no competing interests.
Ethics statement
The protocol was approved by the Laboratory Animal Ethics Committee at the Tianjin Medical University Cancer Institute & Hospital (TMUCIH) (#: AE-2022036). This study has complied with all relevant ethical regulations for animal experiments. Every effort was made to minimise the numbers and suffering of the included animals. Human HCC tissue microarray (TMA) was purchased from the Shanghai Outdo Biotechnology with an Internal Review Board approval (YB M-05-02). Twenty-three cases of HCC specimens were also collected from the Department of Pathology at the TMUCIH. The inpatient time was from January 1, 2018, to December 31, 2018. All patients provided informed consent and this study was conducted according to the guidelines of the Internal Review Board at the TMUCIH (bc2021308).
Additional information
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Edited by Massimiliano Agostini
Supplementary information
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.
About this article
Cite this article
Qie, S., Xiong, H., Liu, Y. et al. Stanniocalcin 2 governs cancer cell adaptation to nutrient insufficiency through alleviation of oxidative stress. Cell Death Dis 15, 567 (2024). https://doi.org/10.1038/s41419-024-06961-7
Received:
Revised:
Accepted:
Published:
DOI: https://doi.org/10.1038/s41419-024-06961-7