SIRT 2‐dependent IDH 1 Deacetylation Inhibits Colorectal Cancer and Liver Metastases
Bo Wang,Yingjiang Ye,Xin Yang,Boya Liu,Zhe Wang,Shuaiyi Chen,Kewei Jiang,Wei Zhang,Hongpeng Jiang,Harri Mustonen,Pauli Puolakkainen,Shan Wang,Jianyuan Luo,Zhanlong Shen
DOI: https://doi.org/10.15252/embr.201948183
IF: 9.071
2020-01-01
EMBO Reports
Abstract:Article5 March 2020free access Transparent process SIRT2-dependent IDH1 deacetylation inhibits colorectal cancer and liver metastases Bo Wang Department of Gastroenterological Surgery, Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People's Hospital, Beijing, China Search for more papers by this author Yingjiang Ye Department of Gastroenterological Surgery, Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People's Hospital, Beijing, China Search for more papers by this author Xin Yang Department of Medical Genetics, Peking University Health Science Center, Beijing, China Search for more papers by this author Boya Liu Department of Medical Genetics, Peking University Health Science Center, Beijing, China Search for more papers by this author Zhe Wang Department of Medical Genetics, Peking University Health Science Center, Beijing, China Search for more papers by this author Shuaiyi Chen Department of Medical Genetics, Peking University Health Science Center, Beijing, China Search for more papers by this author Kewei Jiang Department of Gastroenterological Surgery, Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People's Hospital, Beijing, China Search for more papers by this author Wei Zhang orcid.org/0000-0002-2001-1052 Department of Gastroenterological Surgery, Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People's Hospital, Beijing, China Search for more papers by this author Hongpeng Jiang Department of Gastroenterological Surgery, Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People's Hospital, Beijing, China Search for more papers by this author Harri Mustonen Department of Surgery, Helsinki University Central Hospital, University of Helsinki, Helsinki, Finland Search for more papers by this author Pauli Puolakkainen Department of Surgery, Helsinki University Central Hospital, University of Helsinki, Helsinki, Finland Search for more papers by this author Shan Wang Corresponding Author [email protected] orcid.org/0000-0003-3396-5936 Department of Gastroenterological Surgery, Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People's Hospital, Beijing, China Search for more papers by this author Jianyuan Luo Corresponding Author [email protected] orcid.org/0000-0001-6057-2914 Department of Medical Genetics, Peking University Health Science Center, Beijing, China Search for more papers by this author Zhanlong Shen Corresponding Author [email protected] orcid.org/0000-0002-8717-2686 Department of Gastroenterological Surgery, Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People's Hospital, Beijing, China Search for more papers by this author Bo Wang Department of Gastroenterological Surgery, Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People's Hospital, Beijing, China Search for more papers by this author Yingjiang Ye Department of Gastroenterological Surgery, Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People's Hospital, Beijing, China Search for more papers by this author Xin Yang Department of Medical Genetics, Peking University Health Science Center, Beijing, China Search for more papers by this author Boya Liu Department of Medical Genetics, Peking University Health Science Center, Beijing, China Search for more papers by this author Zhe Wang Department of Medical Genetics, Peking University Health Science Center, Beijing, China Search for more papers by this author Shuaiyi Chen Department of Medical Genetics, Peking University Health Science Center, Beijing, China Search for more papers by this author Kewei Jiang Department of Gastroenterological Surgery, Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People's Hospital, Beijing, China Search for more papers by this author Wei Zhang orcid.org/0000-0002-2001-1052 Department of Gastroenterological Surgery, Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People's Hospital, Beijing, China Search for more papers by this author Hongpeng Jiang Department of Gastroenterological Surgery, Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People's Hospital, Beijing, China Search for more papers by this author Harri Mustonen Department of Surgery, Helsinki University Central Hospital, University of Helsinki, Helsinki, Finland Search for more papers by this author Pauli Puolakkainen Department of Surgery, Helsinki University Central Hospital, University of Helsinki, Helsinki, Finland Search for more papers by this author Shan Wang Corresponding Author [email protected] orcid.org/0000-0003-3396-5936 Department of Gastroenterological Surgery, Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People's Hospital, Beijing, China Search for more papers by this author Jianyuan Luo Corresponding Author [email protected] orcid.org/0000-0001-6057-2914 Department of Medical Genetics, Peking University Health Science Center, Beijing, China Search for more papers by this author Zhanlong Shen Corresponding Author [email protected] orcid.org/0000-0002-8717-2686 Department of Gastroenterological Surgery, Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People's Hospital, Beijing, China Search for more papers by this author Author Information Bo Wang1,‡, Yingjiang Ye1,‡, Xin Yang2, Boya Liu2, Zhe Wang2, Shuaiyi Chen2, Kewei Jiang1, Wei Zhang1, Hongpeng Jiang1, Harri Mustonen3, Pauli Puolakkainen3, Shan Wang *,1, Jianyuan Luo *,2 and Zhanlong Shen *,1 1Department of Gastroenterological Surgery, Laboratory of Surgical Oncology, Beijing Key Laboratory of Colorectal Cancer Diagnosis and Treatment Research, Peking University People's Hospital, Beijing, China 2Department of Medical Genetics, Peking University Health Science Center, Beijing, China 3Department of Surgery, Helsinki University Central Hospital, University of Helsinki, Helsinki, Finland ‡These authors contributed equally to this work *Corresponding author. Tel: +86 1088324479; Fax: +86 1088324479; E-mail: [email protected] *Corresponding author. Tel: +86 82805861; Fax: +86 62015582; E-mail: [email protected] *Corresponding author. Tel: +86 1088324479; Fax: +86 1088324479; E-mail: [email protected] EMBO Rep (2020)21:e48183https://doi.org/10.15252/embr.201948183 PDFDownload PDF of article text and main figures. Peer ReviewDownload a summary of the editorial decision process including editorial decision letters, reviewer comments and author responses to feedback. ToolsAdd to favoritesDownload CitationsTrack CitationsPermissions ShareFacebookTwitterLinked InMendeleyWechatReddit Figures & Info Abstract Protein lysine acetylation affects colorectal cancer (CRC) distant metastasis through multiple pathways. In a previous proteomics screen, we found that isocitrate dehydrogenase 1 (IDH1) is hyperacetylated in CRC primary tumors and liver metastases. Here, we further investigate the function of IDH1 hyperacetylation at lysine 224 in CRC progression. We find that IDH1 K224 deacetylation promotes its enzymatic activity and the production of α-KG, and we identify sirtuin-2 (SIRT2) as a major deacetylase for IDH1. SIRT2 overexpression significantly inhibits CRC cell proliferation, migration, and invasion. IDH1 acetylation is modulated in response to intracellular metabolite concentration and regulates cellular redox hemostasis. Moreover, IDH1 acetylation reversely regulates HIF1α-dependent SRC transcription which in turn controls CRC progression. Physiologically, our data indicate that IDH1 deacetylation represses CRC cell invasion and migration in vitro and in vivo, while the hyperacetylation of IDH1 on K224 is significantly correlated to distant metastasis and poor survival of colorectal cancer patients. In summary, our study uncovers a novel mechanism through which SIRT2-dependent IDH1 deacetylation regulates cellular metabolism and inhibits liver metastasis of colorectal cancer. Synopsis SIRT2 inhibits colorectal cancer cell invasion and metastasis by regulating IDH1 acetylation on K224. SIRT2 deacetylates IDH1 on Lys224 in colorectal cancer. IDH1 K224 acetylation activates HIF1α/SRC axis to promote colorectal cancer metastasis. IDH1 acetylation predicts poor prognosis in colorectal cancer. Introduction Colorectal cancer (CRC) is one of the most common malignancies and one of the major causes of morbidity and mortality worldwide. Approximately 50% of patients with CRC develop liver metastases during their course of disease 1. Despite remarkable progress in cancer therapy, the prognosis for CRC patients with distant metastases remains unsatisfactory 2, 3. In addition, the molecular mechanisms underlying the progression and liver metastasis of CRC still remain unclear. Protein post-translational modification (PTM), especially lysine acetylation, has been reported to be essential in cancer progression. Protein acetylation regulates cancer proliferation through controlling multiple cellular pathways, such as cell cycle, apoptosis, and different metabolism pathways. Metabolism reprogramming is a hallmark for tumors maintaining proliferation rates accompanying the malignant phenotype. The acetylation of key oncogenic enzymes affects the enzymatic and non-enzymatic activity and subsequently promotes tumor progression. IDH1 is a NADP+-dependent enzyme located in the cytoplasm that converts isocitrate to α-ketoglutarate (α-KG). Its mutant form converts α-KG to the oncometabolite 2-hydroxyglutarate (2-HG) and induces epigenetic and cell signaling alterations, thus causing tumorigenesis 4, 5. Compared to primary gliomas, isocitrate IDH1 mutations have been detected in more than 70% of secondary gliomas; among those, arginine 132 has been found in nearly all cases 6, 7. IDH1 has recently received great attention because of its involvement in glutamine metabolism in cancer cells. Despite that rare IDH1 mutations are found in colon cancer samples, the glutamine addiction of colon cancer cells suggests that IDH1 may also have an important role in CRC progression. Nevertheless, IDH1 acetylation modification and its function in tumorigenesis have not yet been reported. Sirtuin-2 (SIRT2) is a cytoplasmic NAD-dependent protein deacetylase, which can serve as a histone deacetylase with a preference for histone H4 lysine 16 (H4K16Ac), and can affect chromosomal condensation during cell mitosis 8, 9. SIRT2 deacetylates several notable metabolic enzymes or transcription factors, including glucose-6-phosphate 1-dehydrogenase (G6PD), phosphoglycerate mutase (PGAM), ATP-citrate synthase (ACLY), forkhead box protein O3 (FOXO3), and peroxiredoxin-1 (Prdx-1) 10-14, which affects cell function. In addition, preclinical studies have shown that SIRT2 deficit mice may develop mammary tumors, hepatocellular carcinomas, and gastrointestinal tumors 15, 16, which suggests that SIRT2 could be a target for cancer therapeutic strategy. Yet, other studies have reported on oncogenic characteristic of SIRT2 in leukemia and breast cancer 17, 18. Therefore, the precise role of SIRT2 acting as oncogene or tumor suppressor remains elusive and is probably regulated by its downstream targets. In our previous study, we used mass spectrometry-based quantitative proteomics to quantify dynamic changes of non-histone protein acetylation between matched primary colorectal cancer and liver metastatic tumor specimens. We found aberrant acetylation levels in several cellular metabolism enzymes during colorectal cancer progression and liver metastasis 19. In the present study, we further investigated the function of IDH1 hyperacetylation at lysine 224 in colorectal cancer progression and liver metastases. We discovered that SIRT2 could deacetylate IDH1 at K224 and exhibit tumor suppression function in colon cancer cell model through IDH1 enzymatic activities and HIF1α-SRC transcription axis. Deacetylation of IDH1 K224 by SIRT2 significantly inhibited the malignant behaviors of CRC cells in vitro and in vivo and predicted poor survival in colorectal cancer samples. These findings further elucidate SIRT2-dependent IDH1 acetylation treatment of liver metastasis in CRC. Results Identification of functional acetylation sites in IDH1 In our previous study 19, we found eight proteins with eight acetylated sites which showed more than 2.5-fold changes at acetylation levels, indicating the potential function of these acetylated proteins in colorectal cancer progression and liver metastases (Table 1). In this study, we further evaluated the acetylation levels of these proteins in CRC cell lines. Briefly, we found decreased acetylation levels of K224R on IDH1 or K112R on CSRP1 (Fig EV1A). Furthermore, after performing a cell invasion assay, we found that the overexpression of IDH1 K224R was able to inhibit cell invasive behavior, while no effect was observed for CSRP1 K112R (Fig EV1B). Hence, IDH1 K224 acetylation was selected for further investigation. Table 1. Differentially expressed acetylation sites obtained in 3 paired samples according to acetylation |Diff| of metastases vs. tumor ≥ 2.5 ProtDesc Position Meta/Tumor Acetylation |Diff| Total protein |Diff| TPM2 152 Down 4.031 1.188 ADH1B 331 Up 3.807 1.077 GLUD1 84 Up 3.711 1.098 ASS1 58 Up 3.067 1.153 CSRP1 112 Down 2.973 1.126 TAGLN 21 Down 2.946 1.219 IDH1 224 Up 2.911 1.055 VTN 275 Up 2.603 1.162 ProtDesc: protein name; Position: acetylation site; Meta/Tumor: liver metastases/colorectal primary tumor; |Diff|: absolute value of the differences. Click here to expand this figure. Figure EV1. IDH1 K224 is confirmed as the object for further investigation A. Acetylation levels of protein candidates were assessed by Western Blot. K224R on IDH1 or K112R on CSRP1 showed decreased acetylation levels comparing to others (Acetylation level of ADH1B or VTN was failed to be detected, data not shown). B. Transwell assay showed IDH1 K224R could inhibit cell invasion but CSRP1 K112R had no such effect. Results were expressed as mean ± SD of three independent experiments (n ≥ 3 per experimental condition). Differences between groups were evaluated with a two-tailed unpaired Student's t-test. **P < 0.01. C. IDH1 K224 is evolutionarily conserved from C. elegans to humans. The sequences of IDH1 were aligned of five species. Acetylation lysine 224 identified by proteomic study was highlighted in red. D. IDH1 protein levels after knockout or treated with empty vector, wild-type, K224R and K224Q. Download figure Download PowerPoint To confirm IDH1 acetylation, we treated HEK293T cells with trichostatin A (TSA) and nicotinamide (NAM), the inhibitors of HDAC class I and II or sirtuins, respectively. The immunoprecipitated acetylated IDH1 was increased under treatment of NAM, suggesting that IDH1 acetylation was controlled by sirtuins (Fig 1A). We mutated all the candidate acetylated lysine sites of IDH1 (from UniProt database) to arginine (R) which mimicked the deacetylated states of protein. As shown in Fig 1B, only K224R showed significant lower acetylation levels, indicating that K224 is the major site for IDH1 acetylation. Figure 1. SIRT2 deacetylates IDH1 on K224 A. IDH1 acetylation levels upon treatment with NAM or TSA. Flag-tagged IDH1 was ectopically expressed in HEK293T cells treated with NAM (5 mM) and/or TSA (0.5 mM) for the indicated time period. B. Five putative lysine residues were mutated. Acetylation levels of Flag-bead-purified IDH1 were determined by Western blot analysis using a pan-anti-acetyl lysine antibody. Relative IDH1 acetylation ratios were calculated after normalizing against Flag. C. Mutation of IDH1 K224R and K224Q resulted in altered acetylation levels using Western blot. D. IDH1 catalytic activity of IDH1 K224 mutants in vitro. Wild-type IDH1 and K224R, K224Q mutants were expressed in HEK293 cells. Proteins were purified by immunoprecipitation (IP), IDH1 levels were normalized for protein, and activity assays were performed. E, F. Steady-state kinetic analysis of IDH1 and variants in HEK293T cells. Comparison of IDH1 WT and K224R and K224Q showed that the K224 site was important in catalysis. G. IDH1 directly interacted with SIRT2 but not SIRT1. SIRT2 deacetylated IDH1 effectively. Flag-tagged IDH1 and HA-tagged SIRT1 or SIRT2 were co-transfected into cells. The acetylation level of Flag-bead-purified IDH1 and the protein association between IDH1 and SIRT1 or SIRT2 were determined by Western blot. H. Co-immunoprecipitation showed endogenous proteins of IDH1 and SIRT2 interacted with each other. I. SIRT2 interacted with IDH1 in vitro. GST pull-down assay with GST-fused SIRT2 and Flag-tagged IDH1 proteins. J. Acetylation level of endogenous IDH1 K224 when treated with SIRT1 or SIRT2 by Western blot analysis. K. IDH1 K224 deacetylation was dependent on SIRT2 catalytic activity. SIRT2 H187Y was a catalytically inactive mutant. Data information: All results were expressed as mean ± SD of three independent experiments (n ≥ 3 per experimental condition). For multiple comparisons in (D), a one-way ANOVA with Newman–Keuls post hoc test was used to test for statistical significance. For (E, F), statistical significance was determined using the two-way ANOVA followed by Tukey's post hoc test. *P < 0.05 and **P < 0.01. Download figure Download PowerPoint Then, we determined the in vitro enzymatic activities of IDH1 mutants, K224R and K224Q, expressed in HEK293T cells and found that IDH1 K224Q mutants had a greater than 40% reduction in activity as compared with the wild-type (WT) IDH1 whereas IDH1 K224R mutants had an enhancement (Fig 1C and D). To investigate the mechanism by which K224 acetylation might affect IDH1 activity, we recombinantly expressed and purified human wild-type IDH1 and the K224R and K224Q mutants from E. coli. Steady-state kinetic analysis of IDH1 and variants indicated that substitution of K224 with arginine (R) or glutamine (Q) could alter the Km values for isocitrate and NADP+ and exhibited a change in Vmax when compared with WT (Fig 1E and F, Table 2). Table 2. Steady-state kinetic analysis of IDH1 WT and mutants NADP+ Isocitrate WT Km(μM) 4.7 ± 0.9 5.2 ± 0.6 Vmax (μmol/min/mg) 41.1 ± 2.4 56.5 ± 2.0 K224R Km(μM) 3.9 ± 0.7 3.9 ± 0.5 Vmax(μmol/min/mg) 44.0 ± 2.2 63.3 ± 2.1 K224Q Km(μM) 8.6 ± 1.1 7.5 ± 1.0 Vmax(μmol/min/mg) 16.0 ± 1.5 16.4 ± 1.6 SIRT2 deacetylates IDH1 on K224 SIRT1, SIRT2, SIRT6, and SIRT7 in sirtuins family might regulate cytoplasmic protein acetylation. As IDH1 is located in cytoplasm, we explored the relationship between IDH1 acetylation and sirtuins mentioned above. After co-transfection of Flag-IDH1 and HA-SIRT1 or HA-SIRT2 or HA-SIRT6 or HA-SIRT7 in HEK293T cells, we found that IDH1 directly interacted with SIRT2, but not with SIRT1 (Fig 1G), SIRT6 and SIRT7 (Fig EV2B); in addition, IDH1 acetylation levels were downregulated by SIRT2 as well. Consistently, endogenous IDH1 interacted with SIRT2 in HCT116 cells other than SIRT1 (Fig 1H). We also performed the GST pull-down assay and confirmed that SIRT2 can interact with IDH1 in vitro (Fig 1I). Click here to expand this figure. Figure EV2. IDH1 deacetylation is specifically regulated by SIRT2 A. CRC cells were treated with five different KATs. IDH1 K224 was acetylated by CBP or P300. B. IDH1 interacts with SIRT2, but not SIRT6 and SIRT7. Flag-tagged IDH1 was ectopically expressed in HEK293T cells together with the individual HA-tagged SIRT as indicated. C. Knockdown of SIRT2 resulted in upregulation of IDH1 K224 acetylation level. D. K224 acetylation-specific antibody was generated through rabbit immunization. No. 1 or 2 refers to the acetyl-K224 peptide while No. 3 stands for the unmodified one as negative control. E. The purified antibody reacted strongly with the 47 KD IDH1 protein on extract from mouse liver tissue and HeLa cells. F. Transwell assay evaluating the invasion abilities of HCT116 cells after transfection of SIRT2 or with IDH1 K224Q. G. Cell migration ability was evaluated by wound healing assay in HCT116 cells. H. Colony formation ability after overexpression of SIRT2 or with IDH1 K224Q. Data information: All results were expressed as mean ± SD of three independent experiments (n ≥ 3 per experimental condition). For (F, G, and H), differences between groups were evaluated with a two-tailed unpaired Student's t-test. *P < 0.05 and **P < 0.01. Download figure Download PowerPoint To further investigate the role of K224 acetylation on IDH1, we constructed a K224-specific antibody (Figs EV2D and EV3A). As expected, SIRT2, but not SIRT1, greatly decreased the acetylation levels on K224 site (Fig 1J). HEK293T cells were co-transfected with IDH1 and wild-type (WT) SIRT2 or enzymatically defective mutant SIRT2 H187Y; compared with SIRT2 WT, SIRT2 H187Y lost IDH1 K224 deacetylation ability (Fig 1K). Taken together, IDH1 K224 acetylation may have an important role in colorectal cancer progression and liver metastases and SIRT2 is the major deacetylase responsible for the regulation of IDH1 K224 acetylation. Click here to expand this figure. Figure EV3. IDH1 K224Ac, HIF1α, or SRC protein levels are detected by immunohistochemistry methods in clinical samples A. Acetyl-IDH1 K224 antibody was used in clinical samples. After treatment with corresponding peptides, the signal of IDH1 K224Ac was significantly diminished, indicating that IDH1 K224 peptides successfully blocked the acetylation antibody. B. HIF1α or SRC protein expression in CRC and the adjacent normal tissues. Download figure Download PowerPoint SIRT2 is a tumor suppressor in CRC To investigate the function of SIRT2 in CRC tumorigenesis, we performed a number of cellular tumor biological assays. We found that the overexpression of SIRT2 could dramatically decrease CRC cell proliferation, migration, and cell invasive behavior (Fig 2A–C). Moreover, overexpression of SIRT2 induced cell cycle S phase arrest phenotype (Fig 2D). However, after treated with AGK2 (exclusive SIRT2 inhibitor) or SIRT2 siRNA in HCT116 cells, significantly increased cell invasion, migration, and colony formation were detected. All these experiments demonstrated that SIRT2 inhibits colorectal cancer cell proliferation and invasion. Figure 2. SIRT2 is a tumor suppressor in CRC A. Colony formation ability after transfection of SIRT2 plasmids, AGK2, and SIRT2 siRNA. B. Transwell assay was performed to evaluate the invasion abilities of HCT116 cells after overexpression or downregulation of SIRT2 or treated with AGK2. Stained cells in the lower chambers were quantified. C. Cell migration ability was assessed by wound healing assay in HCT116 cells. D. Cell cycle distribution was evaluated flow cytometrically after treatment with SIRT2 plasmids, SIRT2 siRNA, or AGK2. E. Immunohistochemical analysis of SIRT2 protein in representative colorectal tissue specimens is shown (magnification ×100). Weak cytoplasmic staining for SIRT2 was observed in CRC tissues and metastatic tissues, whereas strong cytoplasmic staining was observed in non-cancerous tissues. Data information: All results were expressed as mean ± SD of three independent experiments (n ≥ 3 per experimental condition). For (A, B, and D), statistical significance was assessed with the one-way ANOVA with Newman–Keuls post hoc test. For (C), statistical significance was assessed with the two-way ANOVA followed by Tukey's post hoc test. *P < 0.05 and **P < 0.01. Download figure Download PowerPoint Next, SIRT2 antibodies were used to detect the expression levels of SIRT2 in CRC primary tissues and their corresponding adjacent normal tissues, as well as liver metastatic tumors. As shown in Fig 2E, SIRT2 expression was significantly decreased in CRC tissues or liver metastases than in corresponding colorectal normal tissues. These data suggest that the downregulation of SIRT2 could lead to CRC progression and liver metastases. IDH1 acetylation senses metabolite concentration and regulate cellular redox hemostasis The benefit of IDH1 for rapidly growing tumorigenic cells is believed to result from a decreased IDH1 activity. To further define the physiologic functions of the SIRT2-IDH1 regulation, we first examined the effect of IDH1 K224 acetylation and enzymatic activities under different metabolite concentrations. Given that glucose is the major carbon and energy source for cancer cell proliferation, we treated HCT116 cells with high glucose levels. High glucose concentration resulted in increased K224 acetylation levels of IDH1 with reduced IDH1 activity in a dose-dependent manner (Figs 3A and B, and EV4A and B). Recent studies have underlined that cancer cells survive for their glutamine addiction 20, 21. Consistent with these data, IDH1 K224 acetylation was increased with high glutamine in a dose-dependent manner with decreased IDH1 activity (Figs 3C and D, and EV4C). Thus, IDH1 acetylation sensed the metabolite concentration and tightly controlled IDH1 enzymatic activities. We further investigated the function of SIRT2 in this regulation process. Knockdown of SIRT2 in HCT116 cells abrogated the effect of changing the K224 acetylation levels under high glucose or glutamine treatment (Fig 3E and F), suggesting the pivotal regulating role of SIRT2 in IDH1 enzymatic activities. Figure 3. IDH1 acetylation senses metabolite concentration and regulate cellular redox hemostasis A, B. IDH1 K224 acetylation level was enhanced with increasing glucose concentration. Flag-IDH1 was overexpressed in cells treated with increased concentrations of glucose for 6 h. IDH1 proteins were purified by Flag beads, and then, IDH1 K224 acetylation level was determined by Western blot and IDH1 catalytic activity was assessed. C, D. IDH1 K224 acetylation level was upregulated, and IDH1 activity was weakened with increasing glutamine concentration. Flag-IDH1 was overexpressed in cells treated with increased glutamine for 6 h. IDH1 proteins were purified by Flag beads, and the K224 acetylation level of IDH1 was determined by Western blot analysis, and then, IDH1 catalytic activity was evaluated. E, F. Downregulation of SIRT2 diminished the effect of glucose or glutamine on changing IDH1 K224 acetylation. Flag-tagged IDH1 was overexpressed in HCT116 cells with or without transient SIRT2 knockdown. The cells were treated with different concentrations of glucose (E) and glutamine (F). G. IDH1 K224 deacetylation regulated cellular NADPH/NADP+ redox in cells. In HCT116 stable cells with IDH1 knockout and re-expressing the indicated proteins, the ratio of NADPH/NADP+ in cells was measured as followed by manufacturer's instruction. H. IDH1 K224 deacetylation promoted GSH production in HCT116 cells. In HCT116 stable cells with IDH1 knockout and re-expressing the indicated proteins, the ratio of GSH/GSSG was assessed as followed by manufacturer's instruction. I. IDH1 K224 deacetylation suppressed cellular ROS levels in HCT116 cells. ROS was determined in cells under non-stressed condition or exposed to menadione. *P < 0.05 and **P < 0.01. Data information: All results were expressed as mean ± SD of three independent experiments (n ≥ 3 per experimental condition). For (B, D, G, and H), statistical significance was assessed with the one-way ANOVA with Newman–Keuls post hoc test. For (I), statistical significance was assessed with the two-way ANOVA followed by Tukey's post hoc test. *P < 0.05 and **P < 0.01. n.s. = not significant. Download figure Download PowerPoint Click here to expand this figure. Figure EV4. IDH1 acetylation level regulates α-KG and metabolite concentration A. Cellular α-KG levels decreased with increasing IDH1 K224Q expression. The α-KG level in cells transfected with empty vector was set as 100%. B, C. The endogenous IDH1 K224 acetylation level was enhanced with increasing glucose or glutamine. D. Knocking out of IDH1 inhibited NADPH production in HCT116 cells. E. Knocking out of IDH1 repressed GSH production in HCT116 cells. **P < 0.01. Data information: All results were expressed as mean ± SD of three independent experiments (n ≥ 3 per experimental condition). For (A), statistical significance was assessed with a one-way ANOVA with Newman–Keuls post hoc test. For (D and E), differences between groups were evaluated with a two-tailed unpaired Student's t-test. **P < 0.01. Download figure Download PowerPoint IDH1 and its enzymatic reaction product α-KG are indispensable for transition between NADP+ and NADPH, whose ratio displays cellular redox balance 22. To investigate the role of IDH1 K224 acetylation in this process, we knocked down IDH1 in HCT116 cells by CRISPR/Cas9 and generated fo