UHMK 1 Promotes Gastric Cancer Progression Through Reprogramming Nucleotide Metabolism

Xing Feng,Dong Ma,Jiabao Zhao,Yongxi Song,Yuekun Zhu,Qingxin Zhou,Fei Ma,Xing Liu,Mengya Zhong,Yu Liu,Yubo Xiong,Xingfeng Qiu,Zhen Zhang,Heng Zhang,Yongxiang Zhao,Kaiguang Zhang,Xuehui Hong,Zhiyong Zhang
DOI: https://doi.org/10.15252/embj.2019102541
2020-01-01
The EMBO Journal
Abstract:Article23 January 2020free access UHMK1 promotes gastric cancer progression through reprogramming nucleotide metabolism Xing Feng orcid.org/0000-0001-8226-3389 The Affiliated Hospital of Guilin Medical University, Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guangxi Neurological Diseases Clinical Research Center, Guilin, Guangxi, China Department of Immuobiology, Yale University School of Medicine, New Haven, CT, USA Search for more papers by this author Dong Ma Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China Search for more papers by this author Jiabao Zhao Department of Gastrointestinal Surgery, Zhongshan Hospital, School of Medicine, Xiamen University, Xiamen, China Search for more papers by this author Yongxi Song Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Shenyang, China Search for more papers by this author Yuekun Zhu Medical Center, Duke University, Durham, NC, USA Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China Search for more papers by this author Qingxin Zhou Department of Oncology, The Third Affiliated Hospital of Harbin Medical University, Harbin, China Search for more papers by this author Fei Ma Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China Search for more papers by this author Xing Liu Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China Search for more papers by this author Mengya Zhong Department of Gastrointestinal Surgery, Zhongshan Hospital, School of Medicine, Xiamen University, Xiamen, China Search for more papers by this author Yu Liu Department of Gastrointestinal Surgery, Zhongshan Hospital, School of Medicine, Xiamen University, Xiamen, China Search for more papers by this author Yubo Xiong Department of Gastrointestinal Surgery, Zhongshan Hospital, School of Medicine, Xiamen University, Xiamen, China Search for more papers by this author Xingfeng Qiu Department of Gastrointestinal Surgery, Zhongshan Hospital, School of Medicine, Xiamen University, Xiamen, China Search for more papers by this author Zhen Zhang Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Anhui, China Search for more papers by this author Heng Zhang Department of Histology and Embryology, Xiang Ya School of Medicine, Central South University, Changsha, China Search for more papers by this author Yongxiang Zhao National Center for International Research of Biological Targeting Diagnosis and Therapy (Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research), Guangxi Medical University, Nanning, China Search for more papers by this author Kaiguang Zhang Corresponding Author [email protected] orcid.org/0000-0001-9462-6335 Department of Digestive Disease, The First Affiliated Hospital of USTC, Anhui Provincial Hospital, University of Science and Technology of China, Anhui, China Search for more papers by this author Xuehui Hong Corresponding Author [email protected] [email protected] orcid.org/0000-0002-8853-7384 Department of Gastrointestinal Surgery, Zhongshan Hospital, School of Medicine, Xiamen University, Xiamen, China Search for more papers by this author Zhiyong Zhang Corresponding Author [email protected] [email protected] orcid.org/0000-0003-4061-7374 The Affiliated Hospital of Guilin Medical University, Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guangxi Neurological Diseases Clinical Research Center, Guilin, Guangxi, China Department of Surgery, Robert-Wood-Johnson Medical School University Hospital, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA Search for more papers by this author Xing Feng orcid.org/0000-0001-8226-3389 The Affiliated Hospital of Guilin Medical University, Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guangxi Neurological Diseases Clinical Research Center, Guilin, Guangxi, China Department of Immuobiology, Yale University School of Medicine, New Haven, CT, USA Search for more papers by this author Dong Ma Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China Search for more papers by this author Jiabao Zhao Department of Gastrointestinal Surgery, Zhongshan Hospital, School of Medicine, Xiamen University, Xiamen, China Search for more papers by this author Yongxi Song Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Shenyang, China Search for more papers by this author Yuekun Zhu Medical Center, Duke University, Durham, NC, USA Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China Search for more papers by this author Qingxin Zhou Department of Oncology, The Third Affiliated Hospital of Harbin Medical University, Harbin, China Search for more papers by this author Fei Ma Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China Search for more papers by this author Xing Liu Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China Search for more papers by this author Mengya Zhong Department of Gastrointestinal Surgery, Zhongshan Hospital, School of Medicine, Xiamen University, Xiamen, China Search for more papers by this author Yu Liu Department of Gastrointestinal Surgery, Zhongshan Hospital, School of Medicine, Xiamen University, Xiamen, China Search for more papers by this author Yubo Xiong Department of Gastrointestinal Surgery, Zhongshan Hospital, School of Medicine, Xiamen University, Xiamen, China Search for more papers by this author Xingfeng Qiu Department of Gastrointestinal Surgery, Zhongshan Hospital, School of Medicine, Xiamen University, Xiamen, China Search for more papers by this author Zhen Zhang Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Anhui, China Search for more papers by this author Heng Zhang Department of Histology and Embryology, Xiang Ya School of Medicine, Central South University, Changsha, China Search for more papers by this author Yongxiang Zhao National Center for International Research of Biological Targeting Diagnosis and Therapy (Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research), Guangxi Medical University, Nanning, China Search for more papers by this author Kaiguang Zhang Corresponding Author [email protected] orcid.org/0000-0001-9462-6335 Department of Digestive Disease, The First Affiliated Hospital of USTC, Anhui Provincial Hospital, University of Science and Technology of China, Anhui, China Search for more papers by this author Xuehui Hong Corresponding Author [email protected] [email protected] orcid.org/0000-0002-8853-7384 Department of Gastrointestinal Surgery, Zhongshan Hospital, School of Medicine, Xiamen University, Xiamen, China Search for more papers by this author Zhiyong Zhang Corresponding Author [email protected] [email protected] orcid.org/0000-0003-4061-7374 The Affiliated Hospital of Guilin Medical University, Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guangxi Neurological Diseases Clinical Research Center, Guilin, Guangxi, China Department of Surgery, Robert-Wood-Johnson Medical School University Hospital, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA Search for more papers by this author Author Information Xing Feng1,2,‡, Dong Ma3,‡, Jiabao Zhao4,‡, Yongxi Song5,‡, Yuekun Zhu6,7, Qingxin Zhou8, Fei Ma9, Xing Liu10, Mengya Zhong4, Yu Liu4, Yubo Xiong4, Xingfeng Qiu4, Zhen Zhang11, Heng Zhang12, Yongxiang Zhao13, Kaiguang Zhang *,14, Xuehui Hong *,*,4,‡ and Zhiyong Zhang *,*,1,15,‡ 1The Affiliated Hospital of Guilin Medical University, Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guangxi Neurological Diseases Clinical Research Center, Guilin, Guangxi, China 2Department of Immuobiology, Yale University School of Medicine, New Haven, CT, USA 3Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China 4Department of Gastrointestinal Surgery, Zhongshan Hospital, School of Medicine, Xiamen University, Xiamen, China 5Department of Surgical Oncology and General Surgery, The First Hospital of China Medical University, Shenyang, China 6Medical Center, Duke University, Durham, NC, USA 7Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China 8Department of Oncology, The Third Affiliated Hospital of Harbin Medical University, Harbin, China 9Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China 10Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China 11Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Anhui, China 12Department of Histology and Embryology, Xiang Ya School of Medicine, Central South University, Changsha, China 13National Center for International Research of Biological Targeting Diagnosis and Therapy (Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research), Guangxi Medical University, Nanning, China 14Department of Digestive Disease, The First Affiliated Hospital of USTC, Anhui Provincial Hospital, University of Science and Technology of China, Anhui, China 15Department of Surgery, Robert-Wood-Johnson Medical School University Hospital, Rutgers University, The State University of New Jersey, New Brunswick, NJ, USA ‡These authors contributed equally to this work ‡These authors contributed equally to this work as senior authors *Corresponding author. Tel: +86 551 62283114; Fax: +86 551 62283114; E-mail: [email protected] *Corresponding author. Tel: +86 592 2993182; Fax: +86 592 2993182; E-mails: [email protected]; [email protected] *Corresponding author. Tel: +1 732 2356853; Fax: +1 732 2356853; E-mails: [email protected]; [email protected] EMBO J (2020)39:e102541https://doi.org/10.15252/embj.2019102541 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 UHMK1 is a nuclear serine/threonine kinase recently implicated in carcinogenesis. However, the functions and action mechanisms of UHMK1 in the pathogenesis of human gastric cancer (GC) are unclear. Here, we observed that UHMK1 was markedly upregulated in GC. UHMK1 silencing strongly inhibited GC aggressiveness. Interestingly, UHMK1-induced GC progression was mediated primarily via enhancing de novo purine synthesis because inhibiting purine synthesis reversed the effects of UHMK1 overexpression. Mechanistically, UHMK1 activated ATF4, an important transcription factor in nucleotide synthesis, by phosphorylating NCOA3 at Ser (S) 1062 and Thr (T) 1067. This event significantly enhanced the binding of NCOA3 to ATF4 and the expression of purine metabolism-associated target genes. Conversely, deficient phosphorylation of NCOA3 at S1062/T1067 significantly abrogated the function of UHMK1 in GC development. Clinically, Helicobacter pylori and GC-associated UHMK1 mutation induced NCOA3-S1062/T1067 phosphorylation and enhanced the activity of ATF4 and UHMK1. Importantly, the level of UHMK1 was significantly correlated with the level of phospho-NCOA3 (S1062/T1067) in human GC specimens. Collectively, these results show that the UHMK1-activated de novo purine synthesis pathway significantly promotes GC development. Synopsis This study delineates a new role for the nuclear serine/threonine kinase UHMK1 as driver of gastric cancer (GC) via regulating purine metabolism and NCOA3/ATF4-mediated gene expression control. Levels of UHMK1 and NCOA3 phosphorylation are promising stratifiers for patient outcome. UHMK1 expression is significantly upregulated in GC and correlates with poor clinical outcome. UHMK1 directly phosphorylates NCOA3 at Ser1062 and Thr1067 residues. NCOA3-S1062/T1067 phosphorylation promotes ATF4 binding, expression of purine metabolic target genes, and GC progression. Helicobacter pylori infection and GC-related UHMK1 mutation (M134T and T217K) enhance NCOA3-S1062/T1067 phosphorylation, activity of ATF4 and UHMK1. Introduction Gastric cancer (GC) has been indicated to be one of the deadliest malignancies. Helicobacter pylori (H. pylori) infection, dietary habits, and other environmental risk agents contribute greatly to GC development (Wroblewski et al, 2010). Although surgical resection is a primary therapeutic option for GC patients, the 5-year survival rate remains approximately 30.6% but is only 5.2% for patients with advanced stages (Yusefi et al, 2018). Therefore, a better understanding of molecular aberrations involved in GC pathogenesis is necessary to improve the clinical outcomes of GC patients. U2AF homology motif kinase 1 (UHMK1) is a ubiquitously expressed nuclear serine (Ser, S)/threonine (Thr, T) kinase (Francone et al, 2010). It was initially identified to regulate the function of stathmin (Barbutti et al, 2017). Since then, UHMK1 has been indicated to bind a range of proteins, such as eEF1A, FAM64, cyclin-dependent kinase inhibitor (CDKI), p27KIP1, SF3b155, and CPEB1, suggesting the varied roles of UHMK1 in different cellular processes (Manceau et al, 2008; Cambray et al, 2009). Although UHMK1 dysregulation or mutation has been recently indicated to be a high-penetrant factor in different types of human tumors, such as pancreatic and ovarian cancer (Katchman et al, 2017; Grant et al, 2018; Wang et al, 2018), its effect and action mechanisms in most cancers including GC still were uncovered. Tumor cells reprogram glucose metabolism by switching from oxidative phosphorylation (OXPHOS) to a process termed the Warburg effect (Song et al, 2014). However, emerging evidence indicates that dysregulated purine metabolism is implicated in tumors (Nishimura et al, 2019). This finding is not surprising, because many metabolites from the Warburg effect or pentose phosphate pathway (PPP) are essential carbon sources for purine biosynthesis (Hong et al, 2014). Several kinases and transcription factors (TFs), for example, mechanistic target of rapamycin kinase (mTOR), activating transcription factor 4 (ATF4), microphthalmia-associated transcription factor (MITF), and c-Myc, dictate cancer-dependent purine biosynthesis (Ben-Sahra et al, 2016; Karigane et al, 2016). We recently reported that dual-specificity tyrosine-(Y)-phosphorylation-regulated kinase 3 (Dyrk3) inhibited liver cancer by downregulating purine synthesis (Ma et al, 2019). However, the effect of purine metabolism on GC is not clear. Herein, by analyzing data from The Cancer Genome Atlas (TCGA) and Oncomine database, we found that genomic DNA of UHMK1 was frequently amplified in GC. Therefore, we further investigated the role and mechanism of UHMK1 in GC. We found that upregulation of UHMK1 suggested poor prognosis of GC patients. We demonstrated that UHMK1 functioned as an oncogene mainly by promoting the purine synthesis pathway in GC. Therefore, UHMK1-enhanced nucleotide synthesis might be an important therapeutic target in GC. Results High-level UHMK1 expression is strongly correlated with GC aggressiveness We first examined the functions of UHMK1 in GC by investigating the relative levels of UHMK1 in five paired GC patient tissues and corresponding normal tissues. The amount of UHMK1 was significantly increased in GC tissues compared with control tissues (Fig 1A). In addition, the amount of UHMK1 in most GC cell lines was often higher than that in the GES-1 cell line, a normal gastric epithelial cell line (Fig 1B). Similar results were found in other studies of GC in TCGA and Oncomine datasets (Fig EV1A–F). The tissue array analysis results indicated that compared to normal tissues, GC tissues presented increased expression levels of UHMK1 (Fig 1C) and that the percentage of cells with UHMK1 expression in patients with stage I, II, III, and IV GC was 18.2, 36.4, 69, and 100, respectively (Fig 1D), further implying that the level of UHMK1 is strongly correlated with GC malignancy. Analysis of 100 patients with GC confirmed that the level of UHMK1 was strongly associated with tumor status, stage, and lymph node metastasis (Fig 1E and Appendix Table S1). Kaplan–Meier analysis of two independent cohorts indicated that GC patients with higher amounts of UHMK1 presented shorter disease-free survival (DFS) and overall survival (OS) times (Figs 1F and EV1G and H). Multivariate analyses identified UHMK1 as an independent prognostic factor in GC (Appendix Table S2). Collectively, our data reveal that UHMK1 may promote the pathogenesis of GC. Figure 1. High expression of UHMK1 predicts poor prognosis of GC patients A. UHMK1 levels in tumor tissues (T) and corresponding nontumor tissues (N) from five GC patients were analyzed using Western blotting and qRT–PCR. The relative mRNA expression of UHMK1 was presented as mean ± standard deviation from three replicates. Pairwise two-tailed statistical significance was assessed by Student's t-test. B. Immunoblotting and qRT–PCR were used to measure the levels of UHMK1 in GES-1 and GC cell lines. The relative mRNA expression of UHMK1 was presented as mean ± standard deviation from three replicates. Pairwise two-tailed statistical significance was assessed by Student's t-test. C. IHC analysis of UHMK1 expression in a GC patient tissue array (n = 102). Representative images are shown. Scale bars = 100 μm. D. The IHC signals were scored using the Allred score as described in Material and Methods. Data were shown as mean ± standard deviation from three independent experiments. E. UHMK1 expression in 100 GC patients was analyzed based on the following parameters: tumor stage, tumor status, and lymph node invasion status. Fisher's test was applied to calculate the exact P value. F. Patients with GC were stratified by the UHMK1 level (n = 100); OS and DFS were assessed by Kaplan–Meier analysis. Data information: *P < 0.05, **P < 0.01, ***P < 0.001. Download figure Download PowerPoint Click here to expand this figure. Figure EV1. TCGA and Oncomine analysis of UHMK1 expression in GC patients A. TCGA analyzing the levels of UHMK1 in GC patients. *P < 0.05. The central line stands for the median expression of UHMK1. Upper line stands for upper limit (Upper quartile +1.5 interquartile range). Lower line stands for lower limit (lower quartile –1.5 interquartile range). B–F. Analysis of UHMK1 expression from Oncomine database. The central line stands for the median expression of UHMK1. Upper line stands for upper limit (Upper quartile +1.5 interquartile range). Lower line stands for lower limit (lower quartile –1.5 interquartile range) (three biological replicates). G, H. Kaplan–Meier analysis from the second independent cohort of GC patients indicated that patients with higher levels of UHMK1 had shorter OS (G) and DFS (H). Survival curves were plotted according to Kaplan–Meier method, with the log-rank test applied for comparison. Download figure Download PowerPoint UHMK1 knockdown strongly inhibits the proliferation and invasion of GC cells, possibly via metabolic pathway reprogramming To elucidate the functions of UHMK1 in GC, three lentivirus-mediated UHMK1-specific short hairpin RNAs (shRNAs) were transfected to knock down endogenous UHMK1 in two GC cell lines. shUHMK1-#1 and shUHMK1-#2, with high inhibition rates, were selected for further experiments (Fig 2A). UHMK1 deficiency significantly reduced GC cell proliferation, colony formation, migration, and invasion relative to the corresponding levels in negative control cells (Figs 2B and C, and EV2A–F). However, reintroduction of a shRNA-resistant UHMK1 construct, UHMK1Δ, significantly reversed these phenotypes in SGC7901-shUHMK1-#1 or MGC803-shUHMK1-#1 cells (Figs 2B and C, and EV2A–F). Consistent with this result, UHMK1 overexpression in BGC823 and HGC27 cells markedly promoted their proliferative and invasive abilities (Fig EV2G–M). Figure 2. UHMK1 knockdown strongly suppresses the proliferative and invasive abilities of GC cells, possibly via metabolic pathway reprogramming A–C. (A) GC cells (SGC7901 and MGC803) were transfected with (A) shUHMK1-#1, shUHMK1-#2, shUHMK1-#3, or control shRNA lentiviral vector or (B) with a shRNA-resistant expression construct, UHMK1Δ. Western blotting was used to measure the levels of UHMK1. NIH ImageJ software was used to quantify the band intensity. Western blotting assay was conducted for three replicates. (B and C) SGC7901 cells were transfected with or without shUHMK1-#1 or the shRNA-resistant expression construct UHMK1Δ. CCK-8 and colony formation assays were conducted. Scale bars = 5 mm. D, E. The volumes of subcutaneous gastric tumors derived from SGC7901 cells in NOD/SCID mice were determined at different time points. Tumors and representative bioluminescence images are also shown. UHMK1 silencing in the mouse model was confirmed by Western blotting (three biological replicates). Data were presented as mean ± standard deviation from shcontrol and shUHMK1-#1 mice (n = 10 mice/group). Unpaired two-tailed statistical significance was assessed by Student's t-test. Scale bars = 1 cm. F, G. UHMK1 silencing (F) and overexpression (G) affected GC cell abdominal metastasis. The quantitative number of metastatic nodules (left panel) in nude mice (n = 10 mice/group). Representative images of abdominal metastases are also shown (right panel). Scale bars = 1 cm. H. Heatmap of the top 500 genes upregulated in GC patients with high UHMK1 expression. I. Gene ontology enrichment analysis was used to analyze the top 20 enriched biological processes among GC patients with high UHMK1 expression. Data information: **P < 0.01, ***P < 0.001, # marked no significance. Download figure Download PowerPoint Click here to expand this figure. Figure EV2. UHMK1 knockdown strongly suppresses GC cell proliferation and invasion A, B. SGC7901 cells were infected with or without shUHMK1-#1, or a shRNA-resistant expression construct, UHMK1Δ. We performed Transwell assays to measure GC cell migration and invasion, respectively. (Left panel) The quantitative number of migrated or invaded cells in shcontrol, shUHMK1-#1, shUHMK1-#1+Vector, and shUHMK1-#1+UHMK1Δ group. Data were presented as mean ± standard deviation from three replicates. Two-tailed statistical significance was assessed by Student's t-test. (Right panel) Representative images of migrated or invaded cells in each group are shown. Scale bars = 100 μm. C. MGC803 cells were transfected with or without shUHMK1-#1, or UHMK1Δ. CCK-8 assay was conducted to explore the biological function of UHMK1 on cell growth. The growth rate was evaluated from Day 1 to Day 6. Data were presented as mean ± standard deviation from three replicates. Two-tailed statistical significance was assessed by Student's t-test. D–F. MGC803 cells were transfected with or without shUHMK1-#1 or UHMK1Δ. Colony formation and Transwell assays were used to measure GC cell proliferation, invasion, and migration, respectively. (Left panel) The quantitative number of colonies, migrated or invaded cells in shcontrol, shUHMK1-#1, shUHMK1-#1+Vector, and shUHMK1-#1+UHMK1Δ group. Data were presented as mean ± standard deviation from three replicates. (Right panel) Representative images of colonies, migrated or invaded cells in each group are shown. Scale bars = 100 μm. G. Western blotting was performed to analyze UHMK1 expression in BGC823 cells transfected with or without UHMK1. CCK-8 assay was conducted to explore the biological function of UHMK1 on cell growth. The growth rate was evaluated from Day 1 to Day 6. Data were presented as mean ± standard deviation from three replicates. Two-tailed statistical significance was assessed by Student's t-test. H, I. BGC823 cells were transfected with or without UHMK1. Colony formation and Transwell assays were used to measure GC cell proliferation, invasion, and migration, respectively. (Left panel) The quantitative number of colonies, migrated or invaded cells in Vector and UHMK1 group. Data were presented as mean ± standard deviation from three replicates. (Right panel) Representative images of colonies, migrated or invaded cells in each group are shown. Scale bars = 100 μm. J. Western blotting was performed to analyze UHMK1 expression in HGC27 cells transfected with or without UHMK1. CCK-8 assay was conducted to explore the biological function of UHMK1 on cell growth. The growth rate was evaluated from Day 1 to Day 6. Data were presented as mean ± standard deviation from three replicates. Two-tailed statistical significance was assessed by Student's t-test. K–M. HGC27 cells were transfected with or without UHMK1. Colony formation and Transwell assays were used to measure GC cell proliferation, invasion, and migration, respectively. (Left panel) The quantitative number of colonies, migrated or invaded cells in Vector and UHMK1 group. Data were presented as mean ± standard deviation from three replicates. (Right panel) Representative images of colonies, migrated or invaded cells in each group are shown. Scale bars = 100 μm. N. NOD/SCID mice (n = 10 per group) were injected subcutaneously with 1.5 × 106 BGC823-vector and BGC823-UHMK1 into opposite flanks. At indicated time points, the xenograft tumors were collected and presented. Scale bars = 5 mm. Data information: Unpaired two-tailed statistical significance was assessed by Student's t-test. *P < 0.05, **P < 0.01, ***P < 0.001, # marked no significance. Download figure Download PowerPoint In vivo, knockdown of UHMK1 significantly decreased GC tumorigenesis and the number of abdominal metastatic nodules compared to these parameters in control mice (Fig 2D–F). However, UHMK1 overexpression significantly promoted GC growth and increased the number of metastatic nodules (Figs EV2N and 2G). To further uncover the function of UHMK1 in GC, we assessed the transcriptomes of GC patients in TCGA with varying levels of UHMK1 expression. And the top 500 differentially expressed genes were analyzed in UHMK1high versus UHMK1low patients (Fig 2H and Appendix Table S3). Gene ontology enrichment analysis indicated that high UHMK1 expression in GC patients might be associated mainly with tumor metabolism (Fig 2I). In summary, UHMK1 upregulation significantly promotes GC growth and metastasis possibly via metabolic pathway reprogramming in tumors. UHMK1 upregulation promotes the invasion and proliferation of GC cells by promoting the de novo purine synthesis pathway To examine whether UHMK1 reprograms GC-associated metabolic pathways, mass spectrometry (MS) was performed to assemble the collect metabolic profiles of SGC7901 cells with or without UHMK1 knockdown. Interestingly, UHMK1 silencing significantly reduced the intracellular pools of purine intermediates and a moderate decrease in pyrimidine metabolites (Appendix Table S4 and Fig 3A and B). Thus, silencing UHMK1 in GC cells primarily inhibits purine biosynthesis. Figure 3. UHMK1 upregulation significantly increases the proliferative and invasive abilities of GC cells by reprogramming purine metabolism A. Schematic representation of the main metabolic pathways. B. LC-MS/MS was used to examine the metabolites in SGC7901 cells with or without UHMK1 knockdown. The data are shown in the heatmap. C. (Upper panel) LC-MS/MS analysis was performed to measure 15N-glutamine-labeled purine synthesis intermediates in BGC823 cells transfected with or without the WT-UHMK1 or UHMK1-K54R constructs. (Lower panel) LC-MS/MS was used to analyze metabolites labeled with 13C-glycine in BGC823 cells transfected with or without the WT-UHMK1 or UHMK1-K54R constructs (three biological replicates). D. RNA and DNA with incorporated 14C-glycine in BGC823 cells transfected with or without the WT-UHMK1 or UHMK1-K54R constructs were examined using LC-MS/MS (three biological replicates). E. qRT–PCR assays were used to analyze the effects of WT-UHMK1 or UHMK1-K54R on the genes controlling purine metabolism. Data were presented as mean ± standard deviation from three replicates. Unpaired two-tailed statistical significance was assessed by Student's t-test. F. UHMK1 silencing significantly decreased SGC7901 cell proliferation, while ATIC overexpression or purine supplementation markedly reversed this inhibition. Data were presented as mean ± standard deviation from three replicates. Unpaired two-tailed statistical significance was assessed by Student's t-test. G. Treatment with the ATIC inhibitor significantly reversed the proliferation of BGC823 cells induced by UHMK1 overexpression. Data were presented as mean ± standard deviation from three replicates. Unpaired two-tailed statistical significance was assessed by Student's t-test. Data information: *P < 0.05, **P < 0.01, ***P < 0.001, # marked no significance. Download figure Download PowerPoint We then used stable isotope-labeled glutamine (amide-15N) to investigate the effect of UHMK1 on purine metabolism (Dayie & Thakur, 2010). Overexpression of wild-type (WT) UHMK1 but not the kinase-dead UHMK1-K54R mutant in BGC823 cells obviously increased the amounts of 15N-purine intermediates (IMP, AMP, and GMP) (Fig 3C upper panel), suggesting that the kinase activity o
What problem does this paper attempt to address?