IMR Press / FBL / Volume 28 / Issue 9 / DOI: 10.31083/j.fbl2809196
Open Access Original Research
SHMT as a Potential Therapeutic Target for Renal Cell Carcinoma
Show Less
1 Department of Parasitology, Guangdong Medical University, 524023 Zhanjiang, Guangdong, China
2 Department of Pharmacology, Guangdong Medical University, 524023 Zhanjiang, Guangdong, China
3 Department of Pharmacy, Affiliated Hospital of Guangdong Medical University, 524001 Zhanjiang, Guangdong, China
4 School of Medical Technology, Guangdong Medical University, 523808 Dongguan, Guangdong, China
*Correspondence: shaozheng@gdmu.edu.cn (Zheng Shao); dengli@gdmu.edu.cn (Li Deng)
Front. Biosci. (Landmark Ed) 2023, 28(9), 196; https://doi.org/10.31083/j.fbl2809196
Submitted: 27 November 2022 | Revised: 22 January 2023 | Accepted: 10 February 2023 | Published: 12 September 2023
Copyright: © 2023 The Author(s). Published by IMR Press.
This is an open access article under the CC BY 4.0 license.
Abstract

Background: Serine hydroxymethyltransferase (SHMT) is a serine-glycine-one-carbon metabolic enzyme in which SHMT1 and SHMT2 encode the cytoplasmic and mitochondrial isoenzymes, respectively. SHMT1 and SHMT2 are key players in cancer metabolic reprogramming, and thus are attractive targets for cancer therapy. However, the role of SHMT in patients with renal cell carcinoma (RCC) has not been fully elucidated. We aimed to systematically analyze the expression, gene regulatory network, prognostic value, and target prediction of SHMT1 and SHMT2 in patients with kidney chromophobe (KICH), kidney renal clear cell carcinoma (KIRC), and kidney renal papillary cell carcinoma (KIRP); elucidate the association between SHMT expression and RCC; and identify potential new targets for clinical RCC treatment. Methods: Several online databases were used for the analysis, including cBioPortal, TRRUST, GeneMANIA, GEPIA, Metascape, UALCAN, LinkedOmics, and TIMER. Results: SHMT1 and SHMT2 transcript levels were significantly down- and upregulated, respectively, in patients with KICH, KIRC, and KIRP, based on sample type, individual cancer stage, sex, and patient age. Compared to men, women with KIRC and KIRP showed significantly up- and downregulated SHMT1 transcript levels, respectively. However, SHMT2 transcript levels were significantly upregulated in the patients mentioned above. KIRC and KIRP patients with high SHMT1 expression had longer survival periods than those with low SHMT1 expression. In patients with KIRC, the findings were similar to those mentioned above. However, in KICH patients, the findings were the opposite regarding SHMT2 expression. SHMT1 versus SHMT2 were altered by 9% versus 3% (n = 66 KICH patients), 4% versus 4% (n = 446 KIRC patients), and 6% versus 7% (n = 280 KIRP patients). SHMT1 versus SHMT2 promoter methylation levels were significantly up- and downregulated in patients with KIRP versus KIRC and KIRP, respectively. SHMT1, SHMT2, and their neighboring genes (NG) formed a complex network of interactions. The molecular functions of SHMT1 and its NG in patients with KICH, KIRC, and KIRP, included clathrin adaptor, metalloendopeptidase, and GTPase regulator activities; lipid binding, active transmembrane transporter activity, and lipid transporter activity; and type I interferon receptor binding, integrin binding, and protein heterodimerization, respectively. Their respective Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways were involved in lysosome activity, human immunodeficiency virus 1 infection, and endocytosis; coronavirus disease 2019 and neurodegeneration pathways (multiple diseases); and RIG-I-like receptor signaling pathway, cell cycle, and actin cytoskeleton regulation. The molecular functions of SHMT2 and its NG in patients with KICH, KIRC, and KIRP included cell adhesion molecule binding and phospholipid binding; protein domain-specific binding, enzyme inhibitor activity, and endopeptidase activity; and hormone activity, integrin binding, and protein kinase regulator activity, respectively. For patients with KIRC versus KIRP, the KEGG pathways were involved in cAMP and calcium signaling pathways versus microRNAs (MiRNAs) in cancer cells and neuroactive ligand-receptor interactions, respectively. We identified the key transcription factors of SHMT1 and its NG. Conclusions: SHMT1 and SHMT2 expression levels were different in patients with RCC. SHMT1 and SHMT2 may be potential therapeutic and prognostic biomarkers in these patients. Transcription factor (MYC, STAT1, PPARG, AR, SREBF2, and SP3) and miRNA (miR-17-5P, miR-422, miR-492, miR-137, miR-30A-3P, and miR-493) regulations may be important strategies for RCC treatment.

Keywords
SHMT1
SHMT2
renal cell carcinoma
target prediction
gene regulation network
1. Introduction

Renal cell carcinoma (RCC) is the most common kidney malignancy, accounting for approximately 90% of all renal malignancies [1]. An estimated 431,288 new RCC cases and 179,368 cancer-related deaths were recorded globally in 2020 [2]. The three main pathological types of RCC are kidney renal clear cell carcinoma (KIRC), kidney renal papillary cell carcinoma (KIRP), and kidney chromophobe (KICH). KIRC is the most common type of RCC, accounting for 75–80% of RCC cases, followed by KIRP, accounting for 10–15% of cases [3]. In RCC, KICH accounts for approximately 4–5% of the cases [4]. RCC is the second most lethal urological malignancy [5]. In advanced RCC, the 5-year survival rate is only 11.7% [6]. Although alternative treatments exist, surgery is the gold standard for RCC treatment [7, 8]. However, 20–40% of patients with RCC experience relapse [9]. In addition, 90% of RCC recur within five years after surgery [10]. Cancer subtypes have different origins, histology, genetics, and epigenetic changes, and thus should be classified and treated as distinct cancers [11]. Hence, the systematic analysis of the targeted predictive and prognostic markers of patients with different RCC subtypes helps achieve precise treatment and improve the survival rates of these patients.

Serine hydroxymethyltransferase (SHMT) is a serine-glycine-one-carbon-metabolizing enzyme. SHMT catalyzes the conversion of serine to glycine, which transfers a one-carbon unit to tetrahydrofolate to produce 5,10-methylenetetrahydrofolate. This enzyme has cytoplasmic (SHMT1) and mitochondrial (SHMT2) isoforms [12], which are highly upregulated in cancer cells. Metabolic reprogramming mediated by SHMT isoenzymes is a hallmark of cancer that supports tumor growth, survival, and chemotherapy resistance [13]. Although normal cells rely on extracellular serine and glycine supplies, a substantial proportion of cancer patients are dependent to intracellular serine/glycine synthesis, thereby providing an attractive action target. Furthermore, one-carbon metabolism mediated by the folate co-factor promotes cancer growth and proliferation by supporting purine and pyrimidine biosynthesis, amino acid homeostasis, epigenetic maintenance, and redox defense [14, 15, 16, 17, 18]. Hence, modulating the expression of SHMT may be an effective cancer treatment strategy; this modulation is expected to be a target for blocking tumor progression and improving survival.

The role of SHMT in RCC pathogenesis is not well understood. Therefore, this study systematically analyzed the expression, gene regulatory network, prognostic value, and target prediction of SHMT1 and SHMT2 in patients with KICH, KIRC, and KIRP; elucidated the association between SHMT and RCC; and identified new potential targets for RCC therapy.

2. Materials and Methods
2.1 UALCAN Analysis

UALCAN (http://ualcan.path.uab.edu/analysis.html) is an online professional database for analyzing tumor gene expression and methylation levels. We used the UALCAN database to analyze the expression and methylation levels of SHMT1 and SHMT2 in healthy individuals and patients with KICH, KIRC, and KIRP. Furthermore, we used the Student’s t-test for comparative analysis, and the difference was considered significant at a p-value < 0.05 [19, 20].

2.2 GEPIA Analysis

Gene Expression Profiling (GEPIA) (http://gepia.cancer-pku.cn/index.html) is a free online platform for analyzing the correlation between gene expression levels and the pathological tumor stage and prognostic value. We used the GEPIA database to analyze the correlation between pathological tumor stage and the prognostic value of SHMT1 and SHMT2 expression levels in patients with KICH, KIRC, and KIRP. Comparative analysis was performed using Student’s t-tests, with a p-value less than 0.05 considered significant [19, 20].

2.3 cBioPortal Analysis

cBioPortal (http://cbioportal.org) is an online professional database used to analyze genetic alterations in tumors. We used the cBioPortal database to analyze genetic alterations in SHMT1, SHMT2, and their neighboring genes (NG). A total of 66, 446, and 280 KICH, KIRC, and KIRP samples were analyzed, respectively, and mRNA expression z-scores were obtained relative to all samples (log RNA Seq V2 RSEM) using a z-score threshold of ±2.0 [19, 20].

2.4 STRING Analysis

STRING (https://string-db.org/cgi/input.pl) is an online professional database for analyzing protein-protein interactions (PPI). We used the STRING database to build a low-confidence level (0.150) PPI network and screening criteria for species defined as humans. Finally, K-means cluster analysis was performed on SHMT1, SHMT2, and their NG [19, 20].

2.5 GeneMANIA Analysis

GeneMANIA (http://www.genemania.org) is a free professional tool for analyzing gene interactions. We used the GeneMANIA database to explore the interactions between SHMT1, SHMT2, and their altered NG [19, 20].

2.6 Metascape Analysis

Metascape (https://metascape.org) is a professional-free tool for analyzing gene ontology (GO) functions and the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment. We used the Metascape database to analyze the GO function and KEGG pathway enrichment of SHMT1, SHMT2, and their altered NG in patients with KICH, KIRC, and KIRP [19, 20].

2.7 TRRUST Analysis

TRRUST (https://www.grnpedia.org/trrust/) is an online professional database that analyzes transcriptional gene regulators. We used the TRRUST database to analyze the transcriptional regulators of SHMT1, SHMT2, and their altered NG in patients with RCC.

2.8 LinkedOmics Analysis

LinkedOmics (http://www.linkedomics.org/) is a free online platform for analyzing microRNA (miRNA) target enrichment and differentially expressed genes associated with tumor genes. We used the LinkedOmics database to analyze the miRNA target enrichment and differentially expressed genes associated with SHMT1 and SHMT2 [19, 20].

2.9 TIMER Analysis

TIMER (https://cistrome.shinyapps.io/timer/) is a specialized database for systematically analyzing tumor genes associated with infiltrating immune cells. We used the TIMER database to analyze the correlation between SHMT1 and SHMT2 expression and immune cell infiltration levels [19, 20].

3. Results
3.1 SHMT Expression in RCC

We compared the SHMT1 and SHMT2 expression levels based on sample type, individual cancer stage, sex, and patient age among patients with RCC. Results showed that SHMT1 transcript levels were significantly downregulated in patients with KICH, KIRC, and KIRP, based on sample type, individual cancer stage, sex, and patient age (p < 0.05; Fig. 1). The transcript level of SHMT1 in stage 4 cancer was significantly lower than in stage 1 among patients with KIRC and KIRP (p < 0.05; Fig. 1F,J). In patients with KIRC, SHMT1 transcript levels were significantly upregulated in females compared to males (p < 0.001; Fig. 1G). However, SHMT1 transcript levels in patients with KIRP were significantly downregulated in females compared to that in males (p < 0.001; Fig. 1K). In addition, results indicated that SHMT2 transcript levels were significantly upregulated in patients with KICH, KIRC, and KIRP, based on sample type, individual cancer stage, sex, and patient age (p < 0.05; Fig. 2). In patients with KIRP, SHMT2 transcript levels were significantly upregulated in females compared to that in males (p < 0.05; Fig. 2K).

Fig. 1.

Expression of SHMT1 in RCC (UALCAN). (A) The expression of SHMT1 in KICH based on sample types. (B) The expression of SHMT1 in KICH based on individual cancer stage. (C) The expression of SHMT1 in KICH based on patient’s gender. (D) The expression of SHMT1 in KICH based on patient’s age. (E) The expression of SHMT1 in KIRC based on sample types. (F) The expression of SHMT1 in KIRC based on individual cancer stage. (G) The expression of SHMT1 in KIRC based on patient’s gender. (H) The expression of SHMT1 in KIRC based on patient’s age. (I) The expression of SHMT1 in KIRP based on sample types. (J) The expression of SHMT1 in KIRP based on individual cancer stage. (K) The expression of SHMT1 in KIRP based on patient’s gender. (L) The expression of SHMT1 in KIRP based on patient’s age; Data are expressed as mean ± SE. *p < 0.05; **p < 0.01; ***p < 0.001.

Fig. 2.

Expression of SHMT2 in renal cell carcinoma (RCC) (UALCAN). (A) The expression of SHMT2 in KICH based on sample types. (B) The expression of SHMT2 in KICH based on individual cancer stage. (C) The expression of SHMT2 in KICH based on patient’s gender. (D) The expression of SHMT2 in KICH based on patient’s age. (E) The expression of SHMT2 in KIRC based on sample types. (F) The expression of SHMT2 in KIRC based on individual cancer stage. (G) The expression of SHMT2 in KIRC based on patient’s gender. (H) The expression of SHMT2 in KIRC based on patient’s age. (I) The expression of SHMT2 in KIRP based on sample types. (J) The expression of SHMT2 in KIRP based on individual cancer stage. (K) The expression of SHMT2 in KIRP based on patient’s gender. (L) The expression of SHMT2 in KIRP based on patient’s age; Data are expressed as mean ± SE. *p < 0.05; **p < 0.01; ***p < 0.001.

Moreover, we evaluated the correlation between the differential expression levels of SHMT1 and SHMT2 and the pathological stage of RCC. A significant correlation was found between the expression of SHMT1 (p = 0.0142; Fig. 3B) and pathological stage of KIRC. The expression of SHMT1 (p = 0.00326; Fig. 3C), SHMT2 (p = 7.32 × 10-8; Fig. 3F), and the pathological stage of KIRP also had a significant correlation. Moreover, results showed a significant correlation between the expression of SHMT2 (p = 0.000135; Fig. 3D) and pathological stage of KICH.

Fig. 3.

Correlation between the expression of SHMT and the pathological stage of RCC (GEPIA). (A) SHMT1 in patients with KICH. (B) SHMT1 in patients with KIRC. (C) SHMT1 in patients with KIRP. (D) SHMT2 in patients with KICH. (E) SHMT2 in patients with KIRC. (F) SHMT2 in patients with KIRP.

Finally, the GEPIA database was used to assess the prognostic value of SHMT1 and SHMT2 expression in patients with RCC. Results indicated that patients with KIRC, and those with KIRP, with high SHMT1 expression had a longer overall and disease-free survival rate than those with low SHMT1 expression (p < 0.05; Fig. 4C–F). In contrast, patients with KICH with low SHMT2 expression had longer overall and disease-free survival times than those with high SHMT2 expression (p < 0.05; Fig. 5A,B). However, patients with KIRC with high SHMT2 expression had longer overall survival times than those with low SHMT2 expression (p < 0.05; Fig. 5C).

Fig. 4.

Prognostic value of SHMT1 in RCC (GEPIA). (A) The overall survival curve for patients with KICH based on SHMT1 expression levels. (B) The disease-free survival cure for patients with KICH based on SHMT1 expression levels. (C) The overall survival curve for patients with KIRC based on SHMT1 expression levels. (D) The disease-free survival cure for patients with KIRC based on SHMT1 expression levels. (E) The overall survival curve for patients with KIRP based on SHMT1 expression levels. (F) The disease-free survival cure for patients with KIRP based on SHMT1 expression levels.

Fig. 5.

Prognostic value of SHMT2 in RCC (GEPIA). (A) The overall survival curve for patients with KICH based on SHMT2 expression levels. (B) The disease-free survival cure for patients with KICH based on SHMT2 expression levels. (C) The overall survival curve for patients with KIRC based on SHMT2 expression levels. (D) The disease-free survival cure for patients with KIRC based on SHMT2 expression levels. (E) The overall survival curve for patients with KIRP based on SHMT2 expression levels. (F) The disease-free survival cure for patients with KIRP based on SHMT2 expression levels.

3.2 Genetic Alteration and Promoter Methylation of SHMT in RCC

The genetic alterations in SHMT1 and SHMT2 were assessed in 66, 446, and 280 patients with KICH, KIRC, and KIRP, respectively, using the TCGA database. Results indicated that SHMT1 expression was altered by 9% in patients with KICH, and the types of genetic alterations mainly included high- and low RNA levels (Fig. 6A). In addition, SHMT1 and SHMT2 expression levels were altered by 4% in patients with KIRC. The types of genetic alterations mainly included missense and truncating mutations as well as high- and low RNA levels (Fig. 6E and Fig. 7E). Meanwhile, among patients with KIRP the SHMT1 expression was altered by 6%, and the types of genetic alterations mainly included missense mutations, deep deletions, and low RNA levels (Fig. 6J). SHMT2 expression was altered by 3% in patients with KICH, and the type of genetic alteration mainly included high RNA levels (Fig. 7A). Meanwhile, the expression of SHMT2 was altered by 7% among patients with KIRP, and the types of genetic alterations mainly included missense mutations, amplification, and high and low RNA levels (Fig. 7J).

Fig. 6.

Genetic alteration and promoter methylation of SHMT1 in RCC (UALCAN). (A) Genetic alteration of SHMT1 in patients with KICH. (B) Promoter methylation of SHMT1 in KICH based on individual cancer stage. (C) Promoter methylation of SHMT1 in KICH based on patient’s gender. (D) Promoter methylation of SHMT1 in KICH based on patient’s age. (E) Genetic alteration of SHMT1 in patients with KIRC. (F) Promoter methylation of SHMT1 in KIRC based on sample types. (G) Promoter methylation of SHMT1 in KIRC based on individual cancer stage. (H) Promoter methylation of SHMT1 in KIRC based on patient’s gender. (I) Promoter methylation of SHMT1 in KIRC based on patient’s age. (J) Genetic alteration of SHMT1 in patients with KIRP. (K) Promoter methylation of SHMT1 in KIRP based on sample types. (L) Promoter methylation of SHMT1 in KIRP based on individual cancer stage. (M) Promoter methylation of SHMT1 in KIRP based on patient’s gender. (N) Promoter methylation of SHMT1 in KIRP based on patient’s age; Data are expressed as mean ± SE. *p < 0.05; **p < 0.01; ***p < 0.001.

Fig. 7.

Genetic alteration and promoter methylation of SHMT2 in RCC (UALCAN). (A) Genetic alteration of SHMT2 in patients with KICH. (B) Promoter methylation of SHMT2 in KICH based on individual cancer stage. (C) Promoter methylation of SHMT2 in KICH based on patient’s gender. (D) Promoter methylation of SHMT2 in KICH based on patient’s age. (E) Genetic alteration of SHMT2 in patients with KIRC. (F) Promoter methylation of SHMT2 in KIRC based on sample types. (G) Promoter methylation of SHMT2 in KIRC based on individual cancer stage. (H) Promoter methylation of SHMT2 in KIRC based on patient’s gender. (I) Promoter methylation of SHMT2 in KIRC based on patient’s age. (J) Genetic alteration of SHMT2 in patients with KIRP. (K) Promoter methylation of SHMT2 in KIRP based on sample types. (L) Promoter methylation of SHMT2 in KIRP based on individual cancer stage. (M) Promoter methylation of SHMT2 in KIRP based on patient’s gender. (N) Promoter methylation of SHMT2 in KIRP based on patient’s age; Data are expressed as mean ± SE. *p < 0.05; **p < 0.01; ***p < 0.001.

Subsequently, we assessed the promoter methylation levels of SHMT1 and SHMT2 in patients with RCC using the UALCAN database. Results indicate that SHMT1 promoter methylation levels were significantly upregulated in patients with KIRP, based on sample type, individual cancer stage (stages 1 and 3), and patient age (41–60 years and 61–80 years) (p < 0.05; Fig. 6K,L,N). SHMT1 promoter methylation levels were significantly upregulated in male patients with KIRP (p < 0.001; Fig. 6M). However, SHMT2 promoter methylation levels were significantly downregulated in patients with KIRC and KIRP based on sample type, individual cancer stage, sex, and patient age (p < 0.05; Fig. 7F–N).

3.3 NG Alteration and SHMT Interaction Network in RCC

We evaluated alterations in the NG of SHMT1 and SHMT2 in RCC using the cBioPortal database. Results showed gene alteration frequencies 16.67%, 16.67%, and 11.76% for the 50 most frequently altered NG of SHMT1 in patients with KICH, KIRC, and KIRP, respectively (Tables 1,2,3). Furthermore, in patients with KICH, KIRC, and KIRP, the 50 most frequently altered NG of SHMT2 showed 100.00%, 15.79%, and 10.00%, respectively (Tables 4,5,6). In patients with KICH, KIRC, and KIRP, the most frequently altered NG of SHMT1 were AP5Z1 (33.33%), MUC16 (20.00%), and STAG2 (23.53%), respectively (Tables 1,2,3). However, AARD (100.00%), PAPPA2 (31.58%), and CDKN2A (55.00%) were the most frequently altered NG of SHMT2 among patients with KICH, KIRC, and KIRP, respectively (Tables 4,5,6).

Table 1.The top 50 SHMT1 NG alterations in KICH (cBioPortal).
Gene Altered group Unaltered group
AP5Z1 2 (33.33%) 0 (0.00%)
BIRC3 2 (33.33%) 0 (0.00%)
DNAJC13 2 (33.33%) 0 (0.00%)
ENPP6 2 (33.33%) 0 (0.00%)
FAM47C 2 (33.33%) 0 (0.00%)
MTOR 2 (33.33%) 0 (0.00%)
MYO18B 2 (33.33%) 0 (0.00%)
OTOA 2 (33.33%) 0 (0.00%)
PACS1 2 (33.33%) 0 (0.00%)
PRODH 2 (33.33%) 0 (0.00%)
CNTN5 2 (33.33%) 1 (1.67%)
DMXL1 2 (33.33%) 1 (1.67%)
NLGN4X 2 (33.33%) 1 (1.67%)
SECISBP2L 2 (33.33%) 1 (1.67%)
ATRX 2 (33.33%) 2 (3.33%)
DNAH1 2 (33.33%) 2 (3.33%)
ZNF799 2 (33.33%) 2 (3.33%)
ABCC10 1 (16.67%) 0 (0.00%)
ABHD17A 1 (16.67%) 0 (0.00%)
ABTB2 1 (16.67%) 0 (0.00%)
ACP1 1 (16.67%) 0 (0.00%)
ACTBL2 1 (16.67%) 0 (0.00%)
ADAM17 1 (16.67%) 0 (0.00%)
ADAM19 1 (16.67%) 0 (0.00%)
ADAMTS16 1 (16.67%) 0 (0.00%)
ADGRB2 1 (16.67%) 0 (0.00%)
ADGRL1 1 (16.67%) 0 (0.00%)
ADH1C 1 (16.67%) 0 (0.00%)
ADI1 1 (16.67%) 0 (0.00%)
ADORA2A 1 (16.67%) 0 (0.00%)
AGAP6 1 (16.67%) 0 (0.00%)
AGO3 1 (16.67%) 0 (0.00%)
AKAP9 1 (16.67%) 0 (0.00%)
ALKAL2 1 (16.67%) 0 (0.00%)
ALPK3 1 (16.67%) 0 (0.00%)
ALPL 1 (16.67%) 0 (0.00%)
ANKRD52 1 (16.67%) 0 (0.00%)
ANKRD66 1 (16.67%) 0 (0.00%)
AP1M1 1 (16.67%) 0 (0.00%)
AP1M2 1 (16.67%) 0 (0.00%)
AP2A1 1 (16.67%) 0 (0.00%)
AP3M2 1 (16.67%) 0 (0.00%)
APOBEC3G 1 (16.67%) 0 (0.00%)
ARHGEF39 1 (16.67%) 0 (0.00%)
ARHGEF40 1 (16.67%) 0 (0.00%)
ARRB1 1 (16.67%) 0 (0.00%)
ASAP2 1 (16.67%) 0 (0.00%)
ASIC4 1 (16.67%) 0 (0.00%)
ASPHD1 1 (16.67%) 0 (0.00%)
ATP11A 1 (16.67%) 0 (0.00%)
Table 2.The top 50 SHMT1 NG alterations in KIRC (cBioPortal).
Gene Altered group Unaltered group
MUC16 9 (50.00%) 50 (11.68%)
OVGP1 7 (38.89%) 14 (3.27%)
OR4N5 6 (33.33%) 7 (1.64%)
TLR3 6 (33.33%) 8 (1.87%)
BPIFC 6 (33.33%) 9 (2.10%)
BTRC 6 (33.33%) 9 (2.10%)
SLPI 6 (33.33%) 11 (2.57%)
TAS2R3 6 (33.33%) 11 (2.57%)
HEBP1 6 (33.33%) 15 (3.50%)
TSKS 6 (33.33%) 15 (3.50%)
CD4 6 (33.33%) 16 (3.74%)
MAP1B 6 (33.33%) 16 (3.74%)
PHACTR1 6 (33.33%) 16 (3.74%)
PDZD2 6 (33.33%) 17 (3.97%)
SSX3 6 (33.33%) 17 (3.97%)
ABCA6 6 (33.33%) 18 (4.21%)
TSHZ3 6 (33.33%) 20 (4.67%)
FAP 5 (27.78%) 3 (0.70%)
IL32 5 (27.78%) 3 (0.70%)
CCS 5 (27.78%) 7 (1.64%)
OSBPL3 5 (27.78%) 7 (1.64%)
NUMA1 5 (27.78%) 8 (1.87%)
OSBPL2 5 (27.78%) 9 (2.10%)
ZNF462 5 (27.78%) 9 (2.10%)
FYB1 5 (27.78%) 10 (2.34%)
LRP4 5 (27.78%) 10 (2.34%)
SLC8B1 5 (27.78%) 10 (2.34%)
C6 5 (27.78%) 11 (2.57%)
FAM104A 5 (27.78%) 11 (2.57%)
STAT2 5 (27.78%) 12 (2.80%)
NOMO1 5 (27.78%) 13 (3.04%)
NRSN2 5 (27.78%) 13 (3.04%)
CFAP53 4 (22.22%) 3 (0.70%)
EPG5 4 (22.22%) 3 (0.70%)
CWH43 4 (22.22%) 4 (0.93%)
FRYL 4 (22.22%) 4 (0.93%)
JAG1 4 (22.22%) 4 (0.93%)
ARFGEF2 4 (22.22%) 5 (1.17%)
KIF5A 4 (22.22%) 5 (1.17%)
PABIR3 4 (22.22%) 5 (1.17%)
SDHAF2 4 (22.22%) 5 (1.17%)
CCDC18 4 (22.22%) 6 (1.40%)
LGI2 4 (22.22%) 6 (1.40%)
NKD1 4 (22.22%) 6 (1.40%)
SLC12A9 4 (22.22%) 6 (1.40%)
MT-ND6 3 (16.67%) 0 (0.00%)
SLC4A11 3 (16.67%) 0 (0.00%)
STIM2 3 (16.67%) 0 (0.00%)
DIDO1 3 (16.67%) 1 (0.23%)
PDYN 3 (16.67%) 1 (0.23%)
Table 3.The top 50 SHMT1 NG alterations in KIRP (cBioPortal).
Gene Altered group Unaltered group
STAG2 4 (23.53%) 4 (1.52%)
FH 3 (17.65%) 1 (0.38%)
FGFR3 3 (17.65%) 2 (0.76%)
CRISP3 2 (11.76%) 0 (0.00%)
LLGL1 2 (11.76%) 0 (0.00%)
MINK1 2 (11.76%) 0 (0.00%)
SAYSD1 2 (11.76%) 0 (0.00%)
SNRPE 2 (11.76%) 0 (0.00%)
TRPV2 2 (11.76%) 0 (0.00%)
TP53 3 (17.65%) 5 (1.90%)
CACNG2 2 (11.76%) 1 (0.38%)
CCNL2 2 (11.76%) 1 (0.38%)
DNAJA2 2 (11.76%) 1 (0.38%)
ITGAE 2 (11.76%) 1 (0.38%)
ITGAL 2 (11.76%) 1 (0.38%)
KIAA0753 2 (11.76%) 1 (0.38%)
NDC80 2 (11.76%) 1 (0.38%)
NUP88 2 (11.76%) 1 (0.38%)
PABPN1 2 (11.76%) 1 (0.38%)
SLC29A1 2 (11.76%) 1 (0.38%)
SLC2A4 2 (11.76%) 1 (0.38%)
TMEM212 2 (11.76%) 1 (0.38%)
TXNIP 2 (11.76%) 1 (0.38%)
WDR59 2 (11.76%) 1 (0.38%)
ZNF689 2 (11.76%) 1 (0.38%)
ATP1B1 3 (17.65%) 6 (2.28%)
ALYREF 2 (11.76%) 2 (0.76%)
ANAPC11 2 (11.76%) 2 (0.76%)
ARHGDIA 2 (11.76%) 2 (0.76%)
ARL16 2 (11.76%) 2 (0.76%)
CCDC137 2 (11.76%) 2 (0.76%)
DEFB110 2 (11.76%) 2 (0.76%)
GCGR 2 (11.76%) 2 (0.76%)
HSDL2 2 (11.76%) 2 (0.76%)
IFNA10 2 (11.76%) 2 (0.76%)
IFNA14 2 (11.76%) 2 (0.76%)
IFNA16 2 (11.76%) 2 (0.76%)
IFNA17 2 (11.76%) 2 (0.76%)
IFNA21 2 (11.76%) 2 (0.76%)
IFNA4 2 (11.76%) 2 (0.76%)
IFNA7 2 (11.76%) 2 (0.76%)
IFNW1 2 (11.76%) 2 (0.76%)
KIF19 2 (11.76%) 2 (0.76%)
MAFG 2 (11.76%) 2 (0.76%)
MCRIP1 2 (11.76%) 2 (0.76%)
MEP1A 2 (11.76%) 2 (0.76%)
MRPL12 2 (11.76%) 2 (0.76%)
MYADML2 2 (11.76%) 2 (0.76%)
NPB 2 (11.76%) 2 (0.76%)
P4HB 2 (11.76%) 2 (0.76%)
Table 4.The top 50 SHMT2 NG alterations in KICH (cBioPortal).
Gene Altered group Unaltered group
AARD 2 (100.00%) 0 (0.00%)
ASAP1 2 (100.00%) 0 (0.00%)
ATAD2 2 (100.00%) 0 (0.00%)
C8ORF76 2 (100.00%) 0 (0.00%)
CASC8 2 (100.00%) 0 (0.00%)
CCAT1 2 (100.00%) 0 (0.00%)
CCDC26 2 (100.00%) 0 (0.00%)
CCN3 2 (100.00%) 0 (0.00%)
CCN4 2 (100.00%) 0 (0.00%)
COLEC10 2 (100.00%) 0 (0.00%)
CYRIB 2 (100.00%) 0 (0.00%)
DEPTOR 2 (100.00%) 0 (0.00%)
DERL1 2 (100.00%) 0 (0.00%)
DSCC1 2 (100.00%) 0 (0.00%)
EFR3A 2 (100.00%) 0 (0.00%)
EIF3H 2 (100.00%) 0 (0.00%)
EXT1 2 (100.00%) 0 (0.00%)
FAM83A 2 (100.00%) 0 (0.00%)
FAM91A1 2 (100.00%) 0 (0.00%)
FBXO32 2 (100.00%) 0 (0.00%)
FER1L6 2 (100.00%) 0 (0.00%)
GSDMC 2 (100.00%) 0 (0.00%)
HHLA1 2 (100.00%) 0 (0.00%)
HPYR1 2 (100.00%) 0 (0.00%)
KCNQ3 2 (100.00%) 0 (0.00%)
KHDRBS3 2 (100.00%) 0 (0.00%)
KLHL38 2 (100.00%) 0 (0.00%)
LINC00861 2 (100.00%) 0 (0.00%)
LINC00964 2 (100.00%) 0 (0.00%)
LINC00977 2 (100.00%) 0 (0.00%)
LINC02912 2 (100.00%) 0 (0.00%)
LRATD2 2 (100.00%) 0 (0.00%)
MED30 2 (100.00%) 0 (0.00%)
MRPL13 2 (100.00%) 0 (0.00%)
MTSS1 2 (100.00%) 0 (0.00%)
NDRG1 2 (100.00%) 0 (0.00%)
NDUFB9 2 (100.00%) 0 (0.00%)
NSMCE2 2 (100.00%) 0 (0.00%)
NTAQ1 2 (100.00%) 0 (0.00%)
OC90 2 (100.00%) 0 (0.00%)
PCAT1 2 (100.00%) 0 (0.00%)
PCAT2 2 (100.00%) 0 (0.00%)
PLEC 2 (100.00%) 0 (0.00%)
POU5F1B 2 (100.00%) 0 (0.00%)
PVT1 2 (100.00%) 0 (0.00%)
RAD21 2 (100.00%) 0 (0.00%)
RN7SKP153 2 (100.00%) 0 (0.00%)
RN7SKP155 2 (100.00%) 0 (0.00%)
RN7SKP206 2 (100.00%) 0 (0.00%)
RN7SKP226 2 (100.00%) 0 (0.00%)
Table 5.The top 50 SHMT2 NG alterations in KIRC (cBioPortal).
Gene Altered group Unaltered group
PAPPA2 6 (31.58%) 17 (3.98%)
C3 6 (31.58%) 15 (3.51%)
PREP 5 (26.32%) 6 (1.41%)
EFNB3 5 (26.32%) 7 (1.64%)
CST4 5 (26.32%) 8 (1.87%)
ALOX12 5 (26.32%) 11 (2.58%)
GLI1 5 (26.32%) 12 (2.81%)
KLK1 5 (26.32%) 12 (2.81%)
MRPL24 5 (26.32%) 13 (3.04%)
KIAA0408 5 (26.32%) 14 (3.28%)
ATP2B4 5 (26.32%) 15 (3.51%)
CACNA1S 5 (26.32%) 16 (3.75%)
DDB1 5 (26.32%) 16 (3.75%)
HEBP1 5 (26.32%) 16 (3.75%)
LRP1 5 (26.32%) 16 (3.75%)
ABCA1 5 (26.32%) 17 (3.98%)
ARFGEF1 5 (26.32%) 17 (3.98%)
CD4 5 (26.32%) 17 (3.98%)
MAP1B 5 (26.32%) 17 (3.98%)
MUC17 5 (26.32%) 17 (3.98%)
CNTROB 4 (21.05%) 2 (0.47%)
NETO2 4 (21.05%) 3 (0.70%)
OR10G2 4 (21.05%) 3 (0.70%)
SENP6 4 (21.05%) 4 (0.94%)
ARHGEF40 4 (21.05%) 5 (1.17%)
ATN1 4 (21.05%) 5 (1.17%)
PPP1R10 4 (21.05%) 5 (1.17%)
TMCC3 4 (21.05%) 5 (1.17%)
OR52B6 4 (21.05%) 7 (1.64%)
DGKA 4 (21.05%) 8 (1.87%)
PKD2 4 (21.05%) 8 (1.87%)
ACSM2A 4 (21.05%) 9 (2.11%)
ADAMTS8 4 (21.05%) 9 (2.11%)
ASH1L 4 (21.05%) 9 (2.11%)
DUOX1 4 (21.05%) 9 (2.11%)
HMGCLL1 3 (15.79%) 0 (0.00%)
ATRN 3 (15.79%) 1 (0.23%)
C9ORF152 3 (15.79%) 1 (0.23%)
TMEM30A 3 (15.79%) 1 (0.23%)
WDR27 3 (15.79%) 1 (0.23%)
CCN2 3 (15.79%) 2 (0.47%)
CD109 3 (15.79%) 2 (0.47%)
FILIP1 3 (15.79%) 2 (0.47%)
MFSD4B 3 (15.79%) 2 (0.47%)
PATJ 3 (15.79%) 2 (0.47%)
PTGER3 3 (15.79%) 2 (0.47%)
SLITRK2 3 (15.79%) 2 (0.47%)
ERN2 3 (15.79%) 3 (0.70%)
SIRPA 3 (15.79%) 3 (0.70%)
TROAP 3 (15.79%) 3 (0.70%)
Table 6.The top 50 SHMT2 NG alterations in KIRP (cBioPortal).
Gene Altered group Unaltered group
CDKN2A 5 (25.00%) 10 (3.85%)
SIRPA 3 (15.00%) 0 (0.00%)
ARHGAP21 3 (15.00%) 1 (0.38%)
PAK5 3 (15.00%) 1 (0.38%)
CPXM1 3 (15.00%) 2 (0.77%)
GRAMD1A 3 (15.00%) 2 (0.77%)
SRD5A1 3 (15.00%) 2 (0.77%)
ADAM33 2 (10.00%) 0 (0.00%)
ADRA1D 2 (10.00%) 0 (0.00%)
ANGPT4 2 (10.00%) 0 (0.00%)
AP5S1 2 (10.00%) 0 (0.00%)
ATRN 2 (10.00%) 0 (0.00%)
ATXN3L 2 (10.00%) 0 (0.00%)
AVP 2 (10.00%) 0 (0.00%)
BMI1 2 (10.00%) 0 (0.00%)
BMP2 2 (10.00%) 0 (0.00%)
C20ORF141 2 (10.00%) 0 (0.00%)
C20ORF202 2 (10.00%) 0 (0.00%)
C20ORF27 2 (10.00%) 0 (0.00%)
C20ORF96 2 (10.00%) 0 (0.00%)
CDC25B 2 (10.00%) 0 (0.00%)
CDS2 2 (10.00%) 0 (0.00%)
CHGB 2 (10.00%) 0 (0.00%)
COL20A1 2 (10.00%) 0 (0.00%)
CRLS1 2 (10.00%) 0 (0.00%)
DDRGK1 2 (10.00%) 0 (0.00%)
DEFB125 2 (10.00%) 0 (0.00%)
DEFB126 2 (10.00%) 0 (0.00%)
DEFB127 2 (10.00%) 0 (0.00%)
DEFB128 2 (10.00%) 0 (0.00%)
DEFB129 2 (10.00%) 0 (0.00%)
DEFB132 2 (10.00%) 0 (0.00%)
EBF4 2 (10.00%) 0 (0.00%)
ENKUR 2 (10.00%) 0 (0.00%)
FAM110A 2 (10.00%) 0 (0.00%)
FAM47B 2 (10.00%) 0 (0.00%)
FASTKD5 2 (10.00%) 0 (0.00%)
FERMT1 2 (10.00%) 0 (0.00%)
FKBP1A 2 (10.00%) 0 (0.00%)
FRMD5 2 (10.00%) 0 (0.00%)
GFRA4 2 (10.00%) 0 (0.00%)
GNRH2 2 (10.00%) 0 (0.00%)
GPR174 2 (10.00%) 0 (0.00%)
GRIPAP1 2 (10.00%) 0 (0.00%)
HACD1 2 (10.00%) 0 (0.00%)
HAUS6 2 (10.00%) 0 (0.00%)
HSPA12B 2 (10.00%) 0 (0.00%)
ITGA8 2 (10.00%) 0 (0.00%)
ITPA 2 (10.00%) 0 (0.00%)
LAMP5 2 (10.00%) 0 (0.00%)

We further evaluated the potential interactions between SHMT1, SHMT2, and their NG. Results indicated that 42 nodes, 146 edges, and 3 clusters were obtained in the constructed PPI network of SHMT1 and its NG among patients with KICH (Fig. 8A). SHMT1 and its NG were linked to a complex interaction network (71 genes and 267 edges) through physical interactions, co-expression, pathways, shared protein domains, and predictions (Fig. 8B). Furthermore, we found that 40 nodes, 106 edges, and 3 clusters were obtained in the constructed PPI network of SHMT1 and its NG among patients with KIRC (Fig. 8C). SHMT1 and its NG were linked to a complex interaction network (70 genes and 389 edges) through co-expression, pathway, genetic interactions, co-localization, and shared protein domains (Fig. 8D). Furthermore, 44 nodes, 198 edges, and 3 clusters were obtained in the constructed PPI network of SHMT1 and its NG among patients with KIRP (Fig. 8E). SHMT1 and its NG were linked to a complex interaction network (71 genes and 361 edges) through co-expression, physical interactions, shared protein domains, co-localization, and prediction (Fig. 8F). Conversely, 34 nodes, 206 edges, and 3 clusters were obtained in the constructed PPI network of SHMT2 and its NG in KICH (Fig. 9A). SHMT2 and its NG were linked to a complex interaction network (56 genes and 154 edges) through co-expression, physical interactions, and shared protein domains (Fig. 9B). In addition, 38 nodes, 146 edges, and 3 clusters were obtained in the constructed PPI network of SHMT2 and its NG in KIRC (Fig. 9C). SHMT2 and its NG were linked to a complex interaction network (71 genes and 151 edges) through co-expression, co-localization, physical interactions, shared protein domains, and prediction (Fig. 9D). A total of 43 nodes, 148 edges, and 3 clusters were obtained in the constructed PPI network of SHMT2 and its NG among patients with KIRP (Fig. 9E). SHMT2 and its NG were linked to a complex interaction network (71 genes and 219 edges) through co-expression, physical interactions, co-localization, shared protein domains, and prediction (Fig. 9F).

Fig. 8.

Interaction analyses of SHMT1 and their NG in RCC (STRING and GeneMANIA). (A) PPI network of SHMT1 and its NG in KICH. (B) Network analyses of SHMT1 and its NG in KICH. (C) PPI network of SHMT1 and its NG in KIRC. (D) Network analyses of SHMT1 and its NG in KIRC. (E) PPI network of SHMT1 and its NG in KIRP. (F) Network analyses of SHMT1 and its NG in KIRP.

Fig. 9.

Interaction analyses of SHMT2 and their NG in RCC (STRING and GeneMANIA). (A) PPI network of SHMT2 and its NG in KICH. (B) Network analyses of SHMT2 and its NG in KICH. (C) PPI network of SHMT2 and its NG in KIRC. (D) Network analyses of SHMT2 and its NG in KIRC. (E) PPI network of SHMT2 and its NG in KIRP. (F) Network analyses of SHMT2 and its NG in KIRP.

3.4 GO and KEGG Pathway Enrichment Analyses

We further performed GO and KEGG pathway enrichment analyses of SHMT1, SHMT2, and their top 50 altered NG in patients with RCC using the Metascape database. Results indicated that biological processes related to SHMT1 and its NG among patients with KICH mainly included cardiac muscle cell development, pyrimidine-containing compound metabolic processes, and multicellular organismal signaling (Fig. 10A). Their cellular components included the AP-type membrane coat adaptor complex, anchored membrane component, and synaptic membrane (Fig. 10B). Clathrin adaptor-, metalloendopeptidase-, and GTPase regulator activities were involved to their molecular functions (Fig. 10C). KEGG pathway analysis revealed that lysosome activity, human immunodeficiency virus 1 infection, and endocytosis were enriched (Fig. 10D). However, our results showed that synapse assembly, canonical Wnt signaling pathway regulation, and myeloid cell differentiation regulation were the biological processes related to SHMT1 and its NG among patients with KIRC (Fig. 10E). Neuronal cell bodies, sites of polarized growth, and extrinsic membrane components were related to their cellular components (Fig. 10F). Lipid binding and active transmembrane transporter- and lipid transporter activity were related to their main molecular functions (Fig. 10G). KEGG pathway analysis revealed that SHMT1 and its NG were associated with the coronavirus disease 2019 and pathways of neurodegeneration (Fig. 10H). Furthermore, biological processes related to SHMT1 and its NG among patients with KIRP mainly included a positive regulation of peptidyl-serine phosphorylation of the STAT protein, nucleocytoplasmic transport, and cellular response to hypoxia (Fig. 10I). The cellular components were the external side of the plasma membrane, axon, and chromosomal region (Fig. 10J). Type I interferon receptor binding, integrin binding, and protein heterodimerization were associated with their main molecular functions (Fig. 10K). RIG-I-like receptor signaling pathway, cell cycle, and actin cytoskeleton regulation were enriched according to the KEGG pathway analysis (Fig. 10L).

Fig. 10.

GO function and KEGG pathways enrichment analyses of SHMT1 and their NG in RCC (Metascape). (A) Biological processes of SHMT1 and its NG in KICH. (B) Cellular components of SHMT1 and its NG in KICH. (C) Molecular functions of SHMT1 and its NG in KICH. (D) KEGG pathway analysis of SHMT1 and its NG in KICH. (E) Biological processes of SHMT1 and its NG in KIRC. (F) Cellular components of SHMT1 and its NG in KIRC. (G) Molecular functions of SHMT1 and its NG in KIRC. (H) KEGG pathway analysis of SHMT1 and its NG in KIRC. (I) Biological processes of SHMT1 and its NG in KIRP. (J) Cellular components of SHMT1 and its NG in KIRP. (K) Molecular functions of SHMT1 and its NG in KIRP. (L) KEGG pathway analysis of SHMT1 and its NG in KIRP.

The biological processes related to SHMT2 and its NG among patients with KICH mainly included plasma membrane organization, cell-cell junction organization, and positive regulation of organelle organization (Fig. 11A). Their cellular components included the ubiquitin ligase complex, chromosomal region, and mitochondrial membrane (Fig. 11B). Cell adhesion molecule and phospholipid bindings were involved to their molecular functions (Fig. 11C). However, we found that calcium ion transmembrane import into the cytosol, positive regulation of lipid localization, and regulation of acute inflammatory response were biological processes associated to SHMT2 and its NG among patients with KIRC (Fig. 11D). The apical plasma membrane, perinuclear region of the cytoplasm, and sarcolemma were related to their cellular components (Fig. 11E). Protein domain-specific binding, enzyme inhibitor activity, and endopeptidase activity were associated to their main molecular functions (Fig. 11F). KEGG pathway analysis revealed that SHMT2 and its NG were associated with cAMP and calcium signaling pathways (Fig. 11G). Furthermore, biological processes related to SHMT2 and its NG among patients with KIRP mainly included the regulation of the transforming growth factor-beta receptor signaling pathway, defense response to Gram-negative bacteria, and positive binding regulation (Fig. 11H). Their cellular components included the leading-edge membrane, extrinsic membrane component, and spindle (Fig. 11I). Hormone activity, integrin, and protein kinase regulatory activity was related to their main molecular functions (Fig. 11J). MiRNAs involved in cancer and neuroactive ligand-receptor interactions were enriched according to KEGG pathway analysis (Fig. 11K).

Fig. 11.

GO function and KEGG pathways enrichment analyses of SHMT2 and their NG in RCC (Metascape). (A) Biological processes of SHMT2 and its NG in KICH. (B) Cellular components of SHMT2 and its NG in KICH. (C) Molecular functions of SHMT2 and its NG in KICH. (D) Biological processes of SHMT2 and its NG in KIRC. (E) Cellular components of SHMT2 and its NG in KIRC. (F) Molecular functions of SHMT2 and its NG in KIRC. (G) KEGG pathway analysis of SHMT2 and its NG in KIRC. (H) Biological processes of SHMT2 and its NG in KIRP. (I) Cellular components of SHMT2 and its NG in KIRP. (J) Molecular functions of SHMT2 and its NG in KIRP. (K) KEGG pathway analysis of SHMT2 and its NG in KIRP.

3.5 Transcription Factor and miRNA Targets of SHMT in RCC

We used the TRRUST database to analyze the key regulatory factors of SHMT1 and SHMT2 in patients with RCC. We found that MYC was the critical transcription factor of SHMT1 and its NG among patients with KICH (p < 0.05) (Table 7). PRODH and SHMT1 were the primary regulatory genes of MYC. In addition, STAT1 was the critical transcription factor of SHMT1 and its NG among patients with KIRC (p < 0.05) (Table 7). STAT1 regulated the functions of STAT2 and TLR3. Furthermore, PPARG, NR3C1, HDAC1, STAT1, and MYC were the critical transcription factors of SHMT1 and its NG among patients with KIRP (p < 0.05) (Table 7). SLC2A4, TP53, and TXNIP were the primary regulatory genes of PPARG. NR3C1 regulated the functions of ATP1B1 and TP53. Moreover, TP53 and TXNIP were the primary regulatory genes of HDAC1. STAT1 regulated the functions of FGFR3 and TP53. SHMT1 and TP53 were the central regulatory genes of MYC. However, we found that AR was the key transcription factor of SHMT2 and its NG among patients with KICH (p < 0.01) (Table 8). ATAD2 and NDRG1 were the primary regulatory genes of AR. In addition, SREBF2, MYCN, and SP3 were the key transcription factors of SHMT2 and its NG among patients with KIRC (p < 0.05) (Table 8). SREBF2 regulated the functions of ABCA1 and LRP1. DDB1 and EFNB3were the central regulatory genes of MYCN. SP3 regulated the functions of ABCA1 and ALOX12. Furthermore, FOXM1, DNMT1, REST, and E2F1 were the key transcription factors of SHMT2 and its NG in KIRP (p < 0.05) (Table 8). CDC25B and CDKN2A were the central regulatory genes of FOXM1. DNMT1 regulated the functions of BMP2 and CDKN2A. Moreover, AVP and CHGB were the primary regulatory genes of REST. E2F1 regulated the functions of BMI1 and CDKN2A.

Table 7.Key regulated factor of SHMT1 and the top 50 NG in renal cell carcinoma (TRRUST).
Type Key TF Description Regulated gene p-value
KICH MYC v-myc myelocytomatosis viral oncogene homolog (avian) PRODH, SHMT1 0.03
KIRC STAT1 signal transducer and activator of transcription 1 STAT2, TLR3 0.0193
KIRP PPARG peroxisome proliferator-activated receptor gamma SLC2A4, TP53, TXNIP 0.000755
NR3C1 nuclear receptor subfamily 3, group C, member 1 ATP1B1, TP53 0.00473
HDAC1 histone deacetylase 1 TP53, TXNIP 0.0158
STAT1 signal transducer and activator of transcription 1 FGFR3, TP53 0.0217
MYC v-myc myelocytomatosis viral oncogene homolog (avian) SHMT1, TP53 0.03
Table 8.Key regulated factor of SHMT2 and the top 50 NG in renal cell carcinoma (TRRUST).
Type Key TF Description Regulated gene p-value
KICH AR androgen receptor ATAD2, NDRG1 0.00895
KIRC SREBF2 sterol regulatory element binding transcription factor 2 ABCA1, LRP1 0.00122
MYCN v-myc myelocytomatosis viral related oncogene, neuroblastoma derived (avian) DDB1, EFNB3 0.00609
SP3 Sp3 transcription factor ABCA1, ALOX12 0.0348
KIRP FOXM1 forkhead box M1 CDC25B, CDKN2A 0.000806
DNMT1 DNA (cytosine-5-)-methyltransferase 1 BMP2, CDKN2A 0.0027
REST RE1-silencing transcription factor AVP, CHGB 0.00287
E2F1 E2F transcription factor 1 BMI1, CDKN2A 0.044

miRNA targets of SHMT1 and SHMT2 were analyzed using the LinkedOmics database. We found that (GCACTTT) miR-17-5P, miR-20A, miR-106A, miR-106B, miR-20B, and miR-519D; (TTTGCAC) miR-19A and miR-19B; and (AAGCCAT) miR-135A and miR-135B were the top three miRNA targets of SHMT1 among patients with KICH (p < 0.001; Table 9). In addition, the top three miRNA targets of SHMT1 among patients with KIRC were (AAGTCCA) miR-422B and miR-422A, (GCTTGAA) miR-498, and (GGGGCCC) miR-296 (p < 0.001; Table 9). Furthermore, (CAGGTCC) miR-492, (ACTGAAA) miR-30A-3P and miR-30E-3P, and (CTTTGTA) miR-524 were the top three miRNA targets of SHMT1 in KIRP (p < 0.001; Table 9). However, (AAGCAAT) miR-137, (GGGCATT) miR-365, and (AACATTC) miR-409-3P were the top three miRNA targets of SHMT2 in KICH (p < 0.001; Table 10). The top three miRNA targets of SHMT2 in KIRC were (ACTGAAA) miR-30A-3P and miR-30E-3P, (CATGTAA) miR-496, and (AGCATTA) miR-155 (p < 0.001; Table 10). Moreover, (ATGTACA) miR-493, (GGGCATT) miR-365, and (CTTTGCA) miR-527 were the top three miRNA targets of SHMT2 in KIRP (p < 0.001; Table 10).

Table 9.The top three miRNA targets of SHMT1 in renal cell carcinoma (LinkedOmics).
Type Enriched category Gene set Leading edge number p-value
KICH miRNA Target GCACTTT, miR-17-5P, miR-20A, miR-106A, miR-106B, miR-20B, miR-519D 111 0.001
TTTGCAC, miR-19A, miR-19B 96 0.001
AAGCCAT, miR-135A, miR-135B 58 0.001
KIRC miRNA Target AAGTCCA, miR-422B, miR-422A 23 0.001
GCTTGAA, miR-498 26 0.001
GGGGCCC, miR-296 11 0.001
KIRP miRNA Target CAGGTCC, miR-492 22 0.001
ACTGAAA, miR-30A-3P, miR-30E-3P 68 0.001
CTTTGTA, miR-524 137 0.001
Table 10.The top three miRNA targets of SHMT2 in renal cell carcinoma (LinkedOmics).
Type Enriched category Gene set Leading edge number p-value
KICH miRNA Target AAGCAAT, miR-137 48 0.001
GGGCATT, miR-365 22 0.001
AACATTC, miR-409-3P 31 0.001
KIRC miRNA Target ACTGAAA, miR-30A-3P, miR-30E-3P 72 0.001
CATGTAA, miR-496 62 0.001
AGCATTA, miR-155 48 0.001
KIRP miRNA Target ATGTACA, miR-493 101 0.001
GGGCATT, miR-365 46 0.001
CTTTGCA, miR-527 66 0.001
3.6 Correlation of Differentially Expressed Genes and SHMT Expression in RCC

The mRNA sequencing data from patients with KICH (n = 66), KIRC (n = 533), and KIRP (n = 290), using the LinkedOmics database, were analyzed. Results indicated that the expression of 19,216 genes was correlated with SHMT1 expression in KICH. The 10,757 and 8459 genes were positively and negatively correlated with SHMT1 expression, respectively (Fig. 12A). We screened 50 genes whose expression levels were significantly correlated with SHMT1 expression in KICH (p < 0.05; Fig. 12B,C). PRODH (Pearson correlation coefficient (PCO) = 0.7353, p = 2.05 × 10-12; Fig. 13A), SH3KBP1 (PCO = 0.7242, p = 6.321 × 10-12; Fig. 13B), and EML6 (PCO = 0.7169, p = 1.288 × 10-11; Fig. 13C) were the top three genes whose expression levels were positively correlated with SHMT1 expression. In addition, the expression levels of 20,158 genes were correlated with SHMT1 expression in KIRC. A total of 9036 and 11,122 genes were positively and negatively correlated with SHMT1 expression, respectively (Fig. 12D). We screened 50 genes whose expression levels were significantly correlated with SHMT1 expression in KIRC (p < 0.05; Fig. 12E,F). Among the screened genes, ACSM2B (PCO = 0.7968, p = 2.754 × 10-118; Fig. 13D), ACSM2A (PCO = 0.792, p = 7.393 × 10-116; Fig. 13E), and PDZK1 (PCO = 0.7817, p = 5.842 × 10-111; Fig. 13F) were the top three genes whose expression levels were positively correlated with SHMT1 expression. Furthermore, the expression levels of 20,023 genes were correlated with SHMT1 expression in KIRP, of which 8091 and 11,932 genes were positively and negatively correlated with SHMT1 expression, respectively (Fig. 12G). In addition, we screened 50 genes whose expression levels were significantly positively and negatively correlated with SHMT1 expression in KIRP (p < 0.05; Fig. 12H,I). Among the screened genes, SMTNL2 (PCO = 0.8186, p = 2.592 × 10-71; Fig. 13G), ACSM2A (PCO = 0.7928, p = 6.781 × 10-64; Fig. 13H), and ACSM5 (PCO = 0.7911, p = 1.956 × 10-63; Fig. 13I) were the top three genes whose expression levels were positively correlated with SHMT1 expression.

Fig. 12.

Genes differentially expressed in correlation with SHMT1 expression in RCC (LinkedOmics). (A) A Pearson test was used to analyze correlations between SHMT1 expression and genes differentially expressed in patients with KICH. (B,C) Heatmaps showing positively and negatively expressed genes correlated with SHMT1 expression in patients with KICH. (D) A Pearson test was used to analyze correlations between SHMT1 expression and genes differentially expressed in patients with KIRC. (E,F) Heatmaps showing genes whose expression positively and negatively correlated with SHMT1 expression in patients with KIRC. (G) A Pearson test was used to analyze correlations between SHMT1 expression and genes differentially expressed in patients with KIRP. (H,I) Heatmaps showing genes whose expression positively and negatively correlated with SHMT1 expression in patients with KIRP.

Fig. 13.

Pearson correlation analysis of gene expression and SHMT1 in RCC (LinkedOmics). (A) PRODH in patients with KICH. (B) SH3KBP1 in patients with KICH. (C) EML6 in patients with KICH. (D) ACSM2B in patients with KIRC. (E) ACSM2A in patients with KIRC. (F) PDZK1 in patients with KIRC. (G) SMTNL2 in patients with KIRP. (H) ACSM2A in patients with KIRP. (I) ACSM5 in patients with KIRP.

Our results showed that the expression levels of 19,216 genes were correlated with SHMT2 expression in KICH. Moreover, 10,436 and 8780 genes were positively and negatively correlated with SHMT2 expression, respectively (Fig. 14A). We screened 50 genes whose expression levels were significantly positively and negatively correlated with SHMT2 expression in KICH (p < 0.05; Fig. 14B,C). PSAT1 (PCO = 0.7665, p = 6.356 × 10-14; Fig. 15A), ASNS (PCO = 0.7594, p = 1.458 × 10-13; Fig. 15B), and YARS (PCO = 0.7573, p = 1.871 × 10-13; Fig. 15C) were the top three genes whose expression levels were positively correlated with SHMT2 expression. However, the expression levels of 20,158 genes were correlated with SHMT2 expression among patients with KIRC; of which 10,443 and 9715 genes were positively and negatively correlated with SHMT2 expression, respectively (Fig. 14D). Additionally, we screened 50 genes whose expression levels were significantly positively and negatively correlated with SHMT2 expression among patients with KIRC (p < 0.05; Fig. 14E,F). NDUFA4L2 (PCO = 0.6471, p = 1.481 × 10-64; Fig. 15D), RALGPS1 (PCO = –0.5957, p = 1.684 × 10-52; Fig. 15E), and NOL3 (PCO = 0.5789, p = 5.235 × 10-49; Fig. 15F) were the top three genes whose expression levels were positively or negatively correlated with SHMT2 expression. Furthermore, the expression levels of 20,023 genes were correlated with SHMT2 expression among patients with KIRP; of which 9759 and 10,264 genes were positively and negatively correlated with SHMT2 expression, respectively (Fig. 14G). We also screened 50 genes whose expression levels were significantly positively and negatively correlated with SHMT2 expression among patients with KIRP (p < 0.05; Fig. 14H,I). CDK4 (PCO = 0.6734, p = 1.154 × 10-39; Fig. 15G), GK5 (PCO = –0.6554, p = 5.696 × 10-37; Fig. 15H), and C12orf52 (PCO = 0.6466, p = 9.859 × 10-36; Fig. 15I) were the top three genes whose expression levels were positively or negatively correlated with SHMT2 expression.

Fig. 14.

Genes differentially expressed in correlation with SHMT2 expression in RCC (LinkedOmics). (A) A Pearson test was used to analyze correlations between SHMT2 expression and genes differentially expressed in patients with KICH. (B,C) Heat maps showing genes whose expression positively and negatively correlated with SHMT2 expression in patients with KICH. (D) A Pearson test was used to analyze correlations between SHMT2 expression and genes differentially expressed in patients with KIRC. (E,F) Heat maps showing genes whose expression positively and negatively correlated with SHMT2 expression in patients with KIRC. (G) A Pearson test was used to analyze correlations between SHMT2 expression and genes differentially expressed in patients with KIRP. (H,I) Heat maps showing genes whose expression positively and negatively correlated with SHMT2 expression in patients with KIRP.

Fig. 15.

Pearson correlation analysis of gene expression and SHMT2 in RCC (LinkedOmics). (A) PSAT1 in patients with KICH. (B) ASNS in patients with KICH. (C) YARS in patients with KICH. (D) NDUFA4L2 in patients with KIRC. (E) RALGPS1 in patients with KIRC. (F) NOL3 in patients with KIRC. (G) CDK4 in patients with KIRP. (H) GK5 in patients with KIRP. (I) C12orf52 in patients with KIRP.

3.7 Immune Cell Infiltration and SHMT Expression in RCC

The TIMER database was used to evaluate the relationship of immune cell infiltration levels with SHMT1 and SHMT2 expression among patients with RCC. Results showed that SHMT1 expression was positively correlated with B-cell (Cor = 0.308, p = 1.25 × 10-2) and dendritic cell infiltration levels (Cor = 0.421, p = 4.82 × 10-4; Fig. 16A) in KICH. The SHMT1 expression was positively correlated with B-cell infiltration level in KIRC (Cor = 0.161, p = 5.42 × 10-4; Fig. 16B). In addition, SHMT1 expression was positively correlated with macrophage infiltration levels in KIRP (Cor = 0.257, p = 3.97 × 10-5; Fig. 16C). However, B-cell (Cor = –0.227, p = 2.56 × 10-4; Fig. 16C) and CD8+ T-cell (Cor = –0.226, p = 2.58 × 10-4; Fig. 16C) infiltration levels were negatively correlated with SHMT1 expression in KIRP. The SHMT2 expression was positively correlated with CD4+ T-cell (Cor = 0.363, p = 2.98 × 10-3), neutrophil (Cor = 0.397, p = 1.07 × 10-3), and dendritic cell (Cor = 0.312, p = 1.14 × 10-2) infiltration levels in KICH (Fig. 16D). SHMT2 expression was positively correlated with B-cell (Cor = 0.229, p = 2.19 × 10-4; Fig. 16F) and CD8+ T-cell (Cor = 0.319, p = 1.69 × 10-7) infiltration levels in KIRP (Fig. 16F).

Fig. 16.

Correlation between SHMT expression and immune cell infiltration levels in RCC (TIMER). (A) SHMT1 in patients with KICH. (B) SHMT1 in patients with KIRC. (C) SHMT1 in patients with KIRP. (D) SHMT2 in patients with KICH. (E) SHMT2 in patients with KIRC. (F) SHMT2 in patients with KIRP.

4. Discussion

Metabolic reprogramming is an essential hallmark of cancer development. Normal cells typically rely on the uptake of serine/glycine from the environment. However, some cancer subtypes synthesize their own serine/glycine intracellularly and become dependent to synthesis [21, 22] by relying on intracellular serine synthesis to meet the high demands of cancer cells, synthesize purines, control redox homeostasis, regulate DNA methylation, and support lipid metabolism [22]. As a result, tumor growth, differentiation, and metastasis are promoted. SHMT is a central enzyme for metabolic reprogramming in cancer cells, which activates a carbon unit in serine-glycine one-carbon metabolism. Previous studies have shown that SHMT1 and SHMT2 are highly upregulated in some cancers [23]. However, the expression, prognosis, gene regulation network, and regulatory targets of SHMT1 and SHMT2 have not been reported in patients with RCC.

The expression levels of SHMT1 and SHMT2 were compared in patients with RCC based on sample type, individual cancer stage, sex, and patient age. This study found that SHMT2 transcript levels were significantly upregulated in patients with KICH, KIRC, and KIRP. Contrary to previous findings [23], SHMT1 expression was significantly downregulated in these patients. Our results showed that SHMT1 transcript levels in patients with KIRC were significantly upregulated in females compared to that in males. However, SHMT1 transcript levels were significantly downregulated and SHMT2 transcript levels were upregulated in female patients with KIRP compared to those in the corresponding male patients. Nevertheless, this finding has not been previously reported. The differential expression of SHMT1 and SHMT2 between the sexes merits increased attention. This may be an essential basis for the differential treatment of SHMT targets in male and female patients with RCC.

Furthermore, a significant positive correlation between SHMT1 expression and the pathological stages of KIRC and KIRP was found. Similarly, there was a significant positive correlation between SHMT2 expression and the pathological stages of KICH and KIRP. SHMT1 versus SHMT2 were altered by 9% vs 3% (n = 66 patients with KICH), 4% vs 4% (n = 446 patients with KIRC), and 6% vs 7% (n = 280 patients with KIRP), respectively. Moreover, SHMT1 promoter methylation levels were significantly upregulated in patients with KIRP. In contrast, SHMT2 promoter methylation levels were significantly downregulated in patients with KIRC and KIRP. Genetic changes and promoter methylation often result in abnormal gene expression and function. Abnormal SHMT1 and SHMT2 expression caused by genetic changes and promoter methylation may also be an essential factor. In addition, the most frequently altered NG of SHMT1 and SHMT2 in patients with KICH, KIRP, and KIRC were AP5Z1, MUC16, STAG2, AARD, PAPPA2, and CDKN2A. These genes promote cancer cell growth, differentiation, and invasion [24, 25, 26, 27]. Therefore, these gene alterations may be involved, to different degrees and via different pathways, in the occurrence and development of RCC.

Finally, we assessed the prognostic value of SHMT expression in patients with RCC. Patients with KIRC or KIRP with high SHMT1 expression had longer survival period than those with low SHMT1 expression. In contrast, KICH patients with low SHMT2 expression had longer survival period than those with high SHMT2 expression. Interestingly, we found that patients with KIRC with high SHMT2 expression had longer survival period than those with low SHMT2 expression. This contradicts the previous findings that SHMT2 expression levels were significantly upregulated in patients with KIRC. High SHMT2 expression may have other vital functions in patients with KIRC, which requires further investigation. In summary, SHMT1 and SHMT2 may be potential prognostic markers in patients with RCC.

We further evaluated the potential interactions between SHMT and its NG. SHMT1, SHMT2, and their NG were linked to a complex interaction network through co-expression, physical interactions, and shared protein domains. Subsequently, we evaluated the functions of SHMT and its NG. The molecular functions of SHMT1 and its NG in KICH were tumor-associated metalloendopeptidase- and GTPase regulatory activity. For example, membrane metalloendopeptidase inhibits prostate cancer development by attenuating the effects of gastrin-releasing peptides on stem/progenitor cells [28]. Additionally, the activation of Rap1 GTPase mediated by junctional adhesion molecule-A modulates the migration of breast cancer cells [29]. The KEGG pathway was involved in lysosome activity and endocytosis. Multidrug resistance in tumor cells reduces the efficacy of chemotherapy by preventing drug accumulation in cells through drug efflux pumps and lysosomal sequestration/exocytosis. To overcome this anti-cancer resistance, targeting lysosomes is considered an effective strategy for effective and selective anti-cancer therapy [30]. Enhancing receptor-mediated endocytosis is an essential strategy for the treatment of cancer and the reduction of side effects and multidrug resistance [31].

Furthermore, we found that active transmembrane transporter activity is related to tumor resistance, which is the molecular function of SHMT1 and its NG among patients with KIRC [32]. The main molecular functions associated with SHMT1 and its NG among patients with KIRP include type I interferon receptor binding, integrin binding, and protein heterodimerization related to tumor growth. Interferons are the most widely used human proteins for treating various cancers. Cell surface receptors are composed of two subunits, IFNAR1 and IFNAR2, which bind to all type I interferons and inhibit tumor growth [33]. Integrins are cell adhesion receptors (usually transmembrane glycoproteins) linked to the extracellular matrix. Enhanced integrin binding promotes cancer cell invasion and metastasis [34]. Protein heterodimerization is closely associated with tumor growth and migration. For example, HER2 and HER3, two members of the human epidermal receptor family of tyrosine kinase receptors, are associated with poor survival in patients with colorectal cancer. HER2/HER3 heterodimerization promotes the growth of colorectal cancer cells [35]. Moreover, CXCL12-CXCL4 heterodimerization prevents CXCL12-driven migration of breast cancer cells [36]. Their KEGG pathway involved the RIG-I-like receptor signaling pathway, cell cycle, and regulation of the actin cytoskeleton. Upregulation of the RIG-I-like receptor signaling pathway can promote the invasion and migration of non-small cell lung cancer cells [37]. Mitosis, the process by which cells divide, is regulated by cell cycle checkpoints. Interference with the cell cycle is vital in cancer treatment [38]. The actin cytoskeleton is an essential component of the eukaryotic cytoskeleton and plays a vital role in cancer metastasis [39].

Our results showed that the molecular functions of SHMT2 and its NG among patients with KICH were tumor-associated cell adhesion molecule- and phospholipid binding. For example, tumor cell migration depends on the interaction between adhesion proteins and the extracellular matrix. Lutheran/basal cell adhesion molecule promotes tumor cell migration by binding to the sub-basal adhesin α5 chains of laminin 511 and 521 [40]. Phospholipids, major components of plasma membranes, may induce multidrug resistance and act as therapeutic targets in cancer cells [41]. In this study, protein domain-specific binding, enzyme inhibitor activity, and endopeptidase activity were the molecular functions of SHMT2 and its NG among patients with KIRC. These functions may be necessary for cancer cell growth inhibition. Deleted-in-liver cancer 1 plays an inhibitory role in cancer growth suppression by binding to the Rho-GTPase-activating protein domain [42]. An inhibition of tartrate-resistant acid phosphatase activity can prevent the migration and invasion of breast cancer cells [43]. Asparagine endopeptidase (AEP) is highly expressed in various solid tumors and promotes cancer cells’ invasion, migration, and metastasis. Blocking AEP can inhibit cancer metastasis [44]. Their KEGG pathway was involved in the cAMP and calcium signaling pathways, which are closely related to cancer development [45, 46]. In addition, the molecular functions associated with SHMT2 and its NG in patients with KIRP mainly included hormone activity, integrin binding, and protein kinase regulatory activity. These factors are closely related to the occurrence and development of RCC. Epidemiological, clinical, biochemical, and genetic studies have shown that RCC etiology is hormone-related. Hormone signaling pathways may be therapeutic targets for RCC [47].

Targeting mitogen-activated protein kinases in patients with RCC can inhibit the proliferation and migration of cancer cells [48]. Their KEGG pathway involves miRNAs in cancer. In recent years, miRNAs have become essential factors in cancer-related cachexia occurrence and participate in the mutual regulatory network of pro-inflammatory signaling pathways [49]. Hence, regulating the functions of SHMT and its NG may be an essential strategy for RCC treatment.

Subsequently, we explored the transcription factors and miRNA targets of SHMT in RCC. We found that MYC was a key transcription factor for SHMT1 and its NG in KICH. Moreover, c-MYC promotes the growth of RCC cells by inhibiting PLA2R1 transcription [50]. Our results also showed that STAT1, which inhibits the growth of A498 RCC cells [51], was the key transcription factor of SHMT1 and its NG in KIRC. Furthermore, we found that PPARG, NR3C1, HDAC1, STAT1, and MYC were the key transcription factors of SHMT1 and its NG among patients with KIRP. Recent studies have shown that these transcription factors affect the progression of multiple tumors [52, 53, 54]. Furthermore, this study indicated that AR, a potential target for Xp11.2 translocation RCC therapy [55], was the key transcription factor of SHMT2 and its NG among patients with KICH. We found that SREBF2, MYCN, and SP3 were the key transcription factors of SHMT2 and its NG among patients with KIRC. SREBF2, MYCN, and SP3 regulate the proliferation and migration of multiple cancer cells [56, 57, 58]. Additionally, FOXM1, DNMT1, REST, and E2F1 were the key transcription factors of SHMT2 and its NG among patients with KIRP. These transcription factors are major drivers of tumorigenesis and progression of multiple cancers, and thus are potential targets for anti-cancer therapy [59, 60]. miRNAs can be potentially use as diagnostic markers and therapeutic targets in cancer clinics. Therefore, we explored the miRNA targets of SHMT in patients with RCC. Recent studies have shown that these miRNAs regulate the occurrence and development of cancer. For example, miR-106 overexpression can promote proliferation and inhibit apoptosis of endometrial carcinoma RL95-2 cells [61]. Another study suggested that targeting miR-19 can inhibit the proliferation and invasion of breast cancer [62]. Moreover, miR-492 promotes cancer progression by targeting GJB4, a novel bladder cancer biomarker [63]. These miRNA targets are also involved in the regulation of tumor growth. For example, miR-137-3p inhibits colorectal cancer cell migration by regulating KDM1A-dependent epithelial-mesenchymal transition [64]. In addition, miR-496 inhibits the proliferation of gastric cancer cells through the AKT/mTOR signaling pathway targeting LYN [65]. Further, miR-493 induces the proliferation, migration, and invasion of esophageal cancer cells in vivo and in vitro by regulating the c-Jun target Wnt5a/PD-L1 [66]. Therefore, the transcription factors and miRNA targets of SHMT are associated with tumor cell proliferation, migration, invasion, and drug resistance. Hence, these transcription factors and miRNA of SHMT may be promising targets for cancer therapy. However, their relationship with RCC has not been fully elucidated. These results suggest that the transcription factors and miRNAs of SHMT may be potential RCC therapeutic targets.

We examined the correlation between differentially expressed genes and SHMT expression in patients with RCC. We found that in patients with KICH, KIRC, and KIRP, PRODH, SH3KBP1, and EML6; ACSM2B, ACSM2A, and PDZK1; and SMTNL2, ACSM2A, and ACSM5 were the top three genes, respectively, whose expression levels were positively associated with SHMT1 expression. These genetic abnormalities play essential roles in the development of multiple cancers. For example, PRODH regulates many pathophysiological processes, including cancer cell apoptosis and metastasis [67]. A loss of PDZK1 expression in patients with gastric cancer promotes the proliferation of gastric cancer cells by activating PI3K/AKT signaling through PTEN phosphorylation [68]. However, our results showed that in patients with KICH, KIRC, and KIRP, PSAT1, ASNS, and YARS; NDUFA4L2, RALGPS1, and NOL3; and CDK4, GK5, and C12orf52 were the top three genes, respectively, whose expression levels were positively or negatively correlated with SHMT2 expression. These genetic abnormalities can promote multiple cancer cells’ proliferation, differentiation, and metastasis. For example, PSAT1 may act as an oncogene that plays a vital role in various cancers. PSAT1 upregulation is involved in the growth of cervical cancer cells and cisplatin resistance via PI3K/AKT signaling pathway upregulation [69]. NDUFA4L2 promotes trastuzumab resistance in HER2-positive breast cancer cells [70]. CDK4 is a crucial cell cycle regulator frequently dysregulated in human malignancies. CDK4 inhibitor use can effectively suppress tumor growth and improve the efficacy of cisplatin treatment in patients with breast and ovarian cancers [71]. Therefore, targeting these genes may serve as adjuvant therapy for RCC.

Immunotherapy is a novel clinical treatment for cancer patients. Tumor immune infiltration is closely related to clinical prognosis. As expected, SHMT expression in patients with RCC was positively or negatively correlated with immune cell infiltration levels. Dendritic cells are the most effective antigen-presenting cells, whereas T-cells are highly effective antitumor-effector cells. The lack of interaction between these cells is a principal mechanism of tumor tolerance [72]. B-cell infiltration plays an essential role in tumor progression and prognosis. B-cells are considered the primary effector cells involved in humoral immunity and inhibit tumor progression by secreting immunoglobulins, promoting T-cell response, and directly killing cancer cells [73]. In recent years, the complex role of neutrophils in tumorigenesis and cancer progression has attracted much attention. Neutrophils kill tumor cells through direct cytotoxic and indirect effects by activating adaptive immune responses. In contrast, the pre-tumor phenotype of neutrophils may be related to cell proliferation, angiogenesis, and immunosuppression in the tumor microenvironment. Neutrophils have recently been identified as potential targets for cancer therapy [74]. Our results showed that SHMT1 expression was positively (B-cells and dendritic cells in KICH, B-cells in KIRC, and macrophages in KIRP) and negatively (B-cells and CD8+ T-cells in KIRP) correlated with immune cell infiltration levels. However, SHMT2 expression was positively correlated with immune cell infiltration levels (CD4+ T-cells, neutrophils, and dendritic cells in KICH and B-cells and CD8+ T-cells in KIRP). Therefore, the aim for SHMT or SHMT-related regulatory targets may be feasible for regulating immune cell infiltration in patients with RCC.

5. Conclusions

This study systematically analyzed the expression, gene regulatory network, prognostic value, and target prediction of SHMT in patients with RCC. SHMT1 and SHMT2 expression levels were different in patients with RCC. Therefore, different patients with RCC should be treated differently. These enzymes may be potential therapeutic and prognostic biomarkers in these patients and an essential strategy for accurately treating patients with RCC. Transcription factor regulation may be an essential strategy for RCC treatment. During this study, we successfully elucidated the relationship between SHMT1, SHMT2, and RCC and provided new insights for RCC treatment. However, further research is required to evaluate the pathological significance of the differential expression of these two SHMT isoenzymes in patients with RCC.

Availability of Data and Materials

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

Author Contributions

YS, ZS and LD designed the research study. YS performed the research. YS, ZS, LD, JZ, WL, QL, LL, QX, JC, XL, and YC analyzed the data. All authors contributed to editorial changes in the manuscript. All authors read and approved the final manuscript. All authors have participated sufficiently in the work and agreed to be accountable for all aspects of the work.

Ethics Approval and Consent to Participate

Not applicable.

Acknowledgment

We thank Guangdong Medical University for their project support.

Funding

This work was supported by the project of financial fund science and technology special competitive allocation of Zhanjiang (Zhanke [2010]174) and postdoctoral Foundation of Guangdong Medical University (4SG22292G).

Conflict of Interest

The authors declare no conflict of interest.

References
[1]
Lobo J, Ohashi R, Amin MB, Berney DM, Compérat EM, Cree IA, et al. WHO 2022 landscape of papillary and chromophobe renal cell carcinoma. Histopathology. 2022; 81: 426–438.
[2]
Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA: A Cancer Journal for Clinicians. 2021; 71: 209–249.
[3]
Zhang T, Gong J, Maia MC, Pal SK. Systemic Therapy for Non-Clear Cell Renal Cell Carcinoma. American Society of Clinical Oncology Educational Book. American Society of Clinical Oncology. Annual Meeting. 2017; 37: 337–342.
[4]
Delahunt B, Cheville JC, Martignoni G, Humphrey PA, Magi-Galluzzi C, McKenney J, et al. The International Society of Urological Pathology (ISUP) grading system for renal cell carcinoma and other prognostic parameters. The American Journal of Surgical Pathology. 2013; 37: 1490–1504.
[5]
Liu X, Zhang M, Liu X, Sun H, Guo Z, Tang X, et al. Urine Metabolomics for Renal Cell Carcinoma (RCC) Prediction: Tryptophan Metabolism as an Important Pathway in RCC. Frontiers in Oncology. 2019; 9: 663.
[6]
Jemal A, Siegel R, Xu J, Ward E. Cancer statistics, 2010. CA: A Cancer Journal for Clinicians. 2010; 60: 277–300.
[7]
Ljungberg B, Cowan NC, Hanbury DC, Hora M, Kuczyk MA, Merseburger AS, et al. EAU guidelines on renal cell carcinoma: the 2010 update. European Urology. 2010; 58: 398–406.
[8]
Campbell SC, Novick AC, Belldegrun A, Blute ML, Chow GK, Derweesh IH, et al. Guideline for management of the clinical T1 renal mass. The Journal of Urology. 2009; 182: 1271–1279.
[9]
Hollingsworth JM, Miller DC, Daignault S, Hollenbeck BK. Five-year survival after surgical treatment for kidney cancer: a population-based competing risk analysis. Cancer. 2007; 109: 1763–1768.
[10]
Fujii Y, Ikeda M, Kurosawa K, Tabata M, Kamigaito T, Hosoda C, et al. Different clinicopathological features between patients who developed early and late recurrence following surgery for renal cell carcinoma. International Journal of Clinical Oncology. 2015; 20: 802–807.
[11]
Relli V, Trerotola M, Guerra E, Alberti S. Abandoning the Notion of Non-Small Cell Lung Cancer. Trends in Molecular Medicine. 2019; 25: 585–594.
[12]
Tramonti A, Nardella C, di Salvo ML, Barile A, Cutruzzolà F, Contestabile R. Human Cytosolic and Mitochondrial Serine Hydroxymethyltransferase Isoforms in Comparison: Full Kinetic Characterization and Substrate Inhibition Properties. Biochemistry. 2018; 57: 6984–6996.
[13]
Geeraerts SL, Kampen KR, Rinaldi G, Gupta P, Planque M, Louros N, et al. Repurposing the Antidepressant Sertraline as SHMT Inhibitor to Suppress Serine/Glycine Synthesis-Addicted Breast Tumor Growth. Molecular Cancer Therapeutics. 2021; 20: 50–63.
[14]
Ducker GS, Rabinowitz JD. One-Carbon Metabolism in Health and Disease. Cell Metabolism. 2017; 25: 27–42.
[15]
Maddocks ODK, Labuschagne CF, Adams PD, Vousden KH. Serine Metabolism Supports the Methionine Cycle and DNA/RNA Methylation through De Novo ATP Synthesis in Cancer Cells. Molecular Cell. 2016; 61: 210–221.
[16]
Ducker GS, Chen L, Morscher RJ, Ghergurovich JM, Esposito M, Teng X, et al. Reversal of Cytosolic One-Carbon Flux Compensates for Loss of the Mitochondrial Folate Pathway. Cell Metabolism. 2016; 23: 1140–1153.
[17]
Yang M, Vousden KH. Serine and one-carbon metabolism in cancer. Nature Reviews. Cancer. 2016; 16: 650–662.
[18]
Li AM, Ye J. Reprogramming of serine, glycine and one-carbon metabolism in cancer. Biochimica et Biophysica Acta. Molecular Basis of Disease. 2020; 1866: 165841.
[19]
Situ Y, Xu Q, Deng L, Zhu Y, Gao R, Lei L, et al. System analysis of VEGFA in renal cell carcinoma: The expression, prognosis, gene regulation network and regulation targets. The International Journal of Biological Markers. 2022; 37: 90–101.
[20]
Situ Y, Gao R, Lei L, Deng L, Xu Q, Shao Z. System analysis of FHIT in LUAD and LUSC: The expression, prognosis, gene regulation network, and regulation targets. The International Journal of Biological Markers. 2022; 37: 158–169.
[21]
Locasale JW. Serine, glycine and one-carbon units: cancer metabolism in full circle. Nature Reviews. Cancer. 2013; 13: 572–583.
[22]
Pan S, Fan M, Liu Z, Li X, Wang H. Serine, glycine and one carbon metabolism in cancer (Review). International Journal of Oncology. 2021; 58: 158–170.
[23]
García-Cañaveras JC, Lancho O, Ducker GS, Ghergurovich JM, Xu X, da Silva-Diz V, et al. SHMT inhibition is effective and synergizes with methotrexate in T-cell acute lymphoblastic leukemia. Leukemia. 2021; 35: 377–388.
[24]
Rajesh C, Sagar S, Rathinavel AK, Chemparathy DT, Peng XL, Yeh JJ, et al. Truncated O-Glycan-Bearing MUC16 Enhances Pancreatic Cancer Cells Aggressiveness via α4β1 Integrin Complexes and FAK Signaling. International Journal of Molecular Sciences. 2022; 23: 5459.
[25]
Gordon NS, Humayun-Zakaria N, Goel A, Abbotts B, Zeegers MP, Cheng KK, et al. STAG2 Protein Expression in Non-muscle-invasive Bladder Cancer: Associations with Sex, Genomic and Transcriptomic Changes, and Clinical Outcomes. European Urology Open Science. 2022; 38: 88–95.
[26]
Dong Y, Zhao L, Duan J, Bai H, Chen D, Li S, et al. PAPPA2 mutation as a novel indicator stratifying beneficiaries of immune checkpoint inhibitors in skin cutaneous melanoma and non-small cell lung cancer. Cell Proliferation. 2022; 55: e13283.
[27]
Padhi SS, Roy S, Kar M, Saha A, Roy S, Adhya A, et al. Role of CDKN2A/p16 expression in the prognostication of oral squamous cell carcinoma. Oral Oncology. 2017; 73: 27–35.
[28]
Cheng CY, Zhou Z, Stone M, Lu B, Flesken-Nikitin A, Nanus DM, et al. Membrane metalloendopeptidase suppresses prostate carcinogenesis by attenuating effects of gastrin-releasing peptide on stem/progenitor cells. Oncogenesis. 2020; 9: 38.
[29]
McSherry EA, Brennan K, Hudson L, Hill ADK, Hopkins AM. Breast cancer cell migration is regulated through junctional adhesion molecule-A-mediated activation of Rap1 GTPase. Breast Cancer Research. 2011; 13: R31.
[30]
Keum C, Hong J, Kim D, Lee SY, Kim H. Lysosome-Instructed Self-Assembly of Amino-Acid-Functionalized Perylene Diimide for Multidrug-Resistant Cancer Cells. ACS Applied Materials & Interfaces. 2021; 13: 14866–14874.
[31]
Tashima T. Effective cancer therapy based on selective drug delivery into cells across their membrane using receptor-mediated endocytosis. Bioorganic & Medicinal Chemistry Letters. 2018; 28: 3015–3024.
[32]
Puri S, Juvale K. Monocarboxylate transporter 1 and 4 inhibitors as potential therapeutics for treating solid tumours: A review with structure-activity relationship insights. European Journal of Medicinal Chemistry. 2020; 199: 112393.
[33]
Quadt-Akabayov SR, Chill JH, Levy R, Kessler N, Anglister J. Determination of the human type I interferon receptor binding site on human interferon-alpha2 by cross saturation and an NMR-based model of the complex. Protein Science. 2006; 15: 2656–2668.
[34]
Yousefi H, Vatanmakanian M, Mahdiannasser M, Mashouri L, Alahari NV, Monjezi MR, et al. Understanding the role of integrins in breast cancer invasion, metastasis, angiogenesis, and drug resistance. Oncogene. 2021; 40: 1043–1063.
[35]
Zhang BY, Zhang L, Chen YM, Qiao X, Zhao SL, Li P, et al. Corosolic acid inhibits colorectal cancer cells growth as a novel HER2/HER3 heterodimerization inhibitor. British Journal of Pharmacology. 2021; 178: 1475–1491.
[36]
Nguyen KTP, Druhan LJ, Avalos BR, Zhai L, Rauova L, Nesmelova IV, et al. CXCL12-CXCL4 heterodimerization prevents CXCL12-driven breast cancer cell migration. Cellular Signalling. 2020; 66: 109488.
[37]
Tang XD, Zhang DD, Jia L, Ji W, Zhao YS. lncRNA AFAP1-AS1 Promotes Migration and Invasion of Non-Small Cell Lung Cancer via Up-Regulating IRF7 and the RIG-I-Like Receptor Signaling Pathway. Cellular Physiology and Biochemistry. 2018; 50: 179–195.
[38]
Vakili-Samiani S, Khanghah OJ, Gholipour E, Najafi F, Zeinalzadeh E, Samadi P, et al. Cell cycle involvement in cancer therapy; WEE1 kinase, a potential target as therapeutic strategy. Mutation Research. 2022; 824: 111776.
[39]
Ma X, Dang Y, Shao X, Chen X, Wu F, Li Y. Ubiquitination and Long Non-coding RNAs Regulate Actin Cytoskeleton Regulators in Cancer Progression. International Journal of Molecular Sciences. 2019; 20: 2997.
[40]
Guadall A, Cochet S, Renaud O, Colin Y, Le Van Kim C, de Brevern AG, et al. Dimerization and phosphorylation of Lutheran/basal cell adhesion molecule are critical for its function in cell migration on laminin. The Journal of Biological Chemistry. 2019; 294: 14911–14921.
[41]
Kopecka J, Trouillas P, Gašparović AČ, Gazzano E, Assaraf YG, Riganti C. Phospholipids and cholesterol: Inducers of cancer multidrug resistance and therapeutic targets. Drug Resistance Updates. 2020; 49: 100670.
[42]
Joshi R, Qin L, Cao X, Zhong S, Voss C, Min W, et al. DLC1 SAM domain-binding peptides inhibit cancer cell growth and migration by inactivating RhoA. The Journal of Biological Chemistry. 2020; 295: 645–656.
[43]
Krumpel M, Reithmeier A, Senge T, Baeumler TA, Frank M, Nyholm PG, et al. The small chemical enzyme inhibitor 5-phenylnicotinic acid/CD13 inhibits cell migration and invasion of tartrate-resistant acid phosphatase/ACP5-overexpressing MDA-MB-231 breast cancer cells. Experimental Cell Research. 2015; 339: 154–162.
[44]
Qi Q, Obianyo O, Du Y, Fu H, Li S, Ye K. Blockade of Asparagine Endopeptidase Inhibits Cancer Metastasis. Journal of Medicinal Chemistry. 2017; 60: 7244–7255.
[45]
Ahmed MB, Alghamdi AAA, Islam SU, Lee JS, Lee YS. cAMP Signaling in Cancer: A PKA-CREB and EPAC-Centric Approach. Cells. 2022; 11: 2020.
[46]
Wang XX, Xiao FH, Li QG, Liu J, He YH, Kong QP. Large-scale DNA methylation expression analysis across 12 solid cancers reveals hypermethylation in the calcium-signaling pathway. Oncotarget. 2017; 8: 11868–11876.
[47]
Czarnecka AM, Niedzwiedzka M, Porta C, Szczylik C. Hormone signaling pathways as treatment targets in renal cell cancer (Review). International Journal of Oncology. 2016; 48: 2221–2235.
[48]
Su Z, Chen D, Zhang E, Li Y, Yu Z, Shi M, et al. MicroRNA-509-3p inhibits cancer cell proliferation and migration by targeting the mitogen-activated protein kinase kinase kinase 8 oncogene in renal cell carcinoma. Molecular Medicine Reports. 2015; 12: 1535–1543.
[49]
Santos JMO, Peixoto da Silva S, Bastos MMSM, Oliveira PA, Gil da Costa RM, Medeiros R. Decoding the role of inflammation-related microRNAs in cancer cachexia: a study using HPV16-transgenic mice and in silico approaches. Journal of Physiology and Biochemistry. 2022; 78: 439–455.
[50]
Vindrieux D, Devailly G, Augert A, Le Calvé B, Ferrand M, Pigny P, et al. Repression of PLA2R1 by c-MYC and HIF-2alpha promotes cancer growth. Oncotarget. 2014; 5: 1004–1013.
[51]
Zhang F, Shang D, Zhang Y, Tian Y. Interleukin-22 suppresses the growth of A498 renal cell carcinoma cells via regulation of STAT1 pathway. PLoS ONE. 2011; 6: e20382.
[52]
Ding X, Han X, Yuan H, Zhang Y, Gao Y. The Impact of PPARD and PPARG Polymorphisms on Glioma Risk and Prognosis. Scientific Reports. 2020; 10: 5140.
[53]
Xiong H, Chen Z, Lin B, Xie B, Liu X, Chen C, et al. Naringenin Regulates FKBP4/NR3C1/NRF2 Axis in Autophagy and Proliferation of Breast Cancer and Differentiation and Maturation of Dendritic Cell. Frontiers in Immunology. 2022; 12: 745111.
[54]
Xu Y, Feng Y, Sun Z, Li Q. RNF168 promotes RHOC degradation by ubiquitination to restrain gastric cancer progression via decreasing HDAC1 expression. Biochemical and Biophysical Research Communications. 2021; 557: 135–142.
[55]
Liu N, Chen Y, Yang L, Shi Q, Lu Y, Ma W, et al. Both SUMOylation and ubiquitination of TFE3 fusion protein regulated by androgen receptor are the potential target in the therapy of Xp11.2 translocation renal cell carcinoma. Clinical and Translational Medicine. 2022; 12: e797.
[56]
Cai C, Zhang Y, Peng X. Knocking down Sterol regulatory element binding protein 2 (SREBF2) inhibits the Serine Protease 8 (PRSS8) /sodium channel epithelial 1alpha subunit (SCNN1A) axis to reduce the cell proliferation, migration and epithelial-mesenchymal transformation of ovarian cancer. Bioengineered. 2021; 12: 9390–9400.
[57]
Shen Z, Sun S. CircPTCH1 Promotes Migration in Lung Cancer by Regulating MYCN Expression Through miR-34c-5p. OncoTargets and Therapy. 2021; 14: 4779–4789.
[58]
Mansour MA. SP3 is associated with migration, invasion, and Akt/PKB signalling in MDA-MB-231 breast cancer cells. Journal of Biochemical and Molecular Toxicology. 2021; 35: e22657.
[59]
Wang K, Dai X, Yu A, Feng C, Liu K, Huang L. Peptide-based PROTAC degrader of FOXM1 suppresses cancer and decreases GLUT1 and PD-L1 expression. Journal of Experimental & Clinical Cancer Research. 2022; 41: 289.
[60]
Ma X, Li Y, Zhao B. Ribosomal protein L5 (RPL5)/ E2F transcription factor 1 (E2F1) signaling suppresses breast cancer progression via regulating endoplasmic reticulum stress and autophagy. Bioengineered. 2022; 13: 8076–8086.
[61]
Li X, Yi X, Bie C, Wang Z. Expression of miR-106 in endometrial carcinoma RL95-2 cells and effect on proliferation and invasion of cancer cells. Oncology Letters. 2018; 16: 2251–2254.
[62]
Liu AX, Yang F, Huang L, Zhang LY, Zhang JR, Zheng RN. Long non-coding RNA Tubulin Alpha 4B (TUBA4B) inhibited breast cancer proliferation and invasion by directly targeting miR-19. European Review for Medical and Pharmacological Sciences. 2019; 23: 708–715.
[63]
Wang K, Lü H, Qu H, Xie Q, Sun T, Gan O, et al. miR-492 Promotes Cancer Progression by Targeting GJB4 and Is a Novel Biomarker for Bladder Cancer. OncoTargets and Therapy. 2019; 12: 11453–11464.
[64]
Ding X, Zhang J, Feng Z, Tang Q, Zhou X. MiR-137-3p Inhibits Colorectal Cancer Cell Migration by Regulating a KDM1A-Dependent Epithelial-Mesenchymal Transition. Digestive Diseases and Sciences. 2021; 66: 2272–2282.
[65]
Su R, Zhao E, Zhang J. miR-496 inhibits proliferation via LYN and AKT pathway in gastric cancer. Open Medicine. 2021; 16: 1206–1214.
[66]
Bian W, Li Y, Zhu H, Gao S, Niu R, Wang C, et al. miR-493 by regulating of c-Jun targets Wnt5a/PD-L1-inducing esophageal cancer cell development. Thoracic Cancer. 2021; 12: 1579–1588.
[67]
Liu Y, Mao C, Liu S, Xiao D, Shi Y, Tao Y. Proline dehydrogenase in cancer: apoptosis, autophagy, nutrient dependency and cancer therapy. Amino Acids. 2021; 53: 1891–1902.
[68]
Zhao C, Tao T, Yang L, Qin Q, Wang Y, Liu H, et al. Loss of PDZK1 expression activates PI3K/AKT signaling via PTEN phosphorylation in gastric cancer. Cancer Letters. 2019; 453: 107–121.
[69]
Duan W, Liu X. PSAT1 Upregulation Contributes to Cell Growth and Cisplatin Resistance in Cervical Cancer Cells via Regulating PI3K/AKT Signaling Pathway. Annals of Clinical and Laboratory Science. 2020; 50: 512–518.
[70]
Yuan Y, Gao H, Zhuang Y, Wei L, Yu J, Zhang Z, et al. NDUFA4L2 promotes trastuzumab resistance in HER2-positive breast cancer. Therapeutic Advances in Medical Oncology. 2021; 13: 17588359211027836.
[71]
Tian C, Wei Y, Li J, Huang Z, Wang Q, Lin Y, et al. A Novel CDK4/6 and PARP Dual Inhibitor ZC-22 Effectively Suppresses Tumor Growth and Improves the Response to Cisplatin Treatment in Breast and Ovarian Cancer. International Journal of Molecular Sciences. 2022; 23: 2892.
[72]
Lurje I, Hammerich L, Tacke F. Dendritic Cell and T Cell Crosstalk in Liver Fibrogenesis and Hepatocarcinogenesis: Implications for Prevention and Therapy of Liver Cancer. International Journal of Molecular Sciences. 2020; 21: 7378.
[73]
Tokunaga R, Naseem M, Lo JH, Battaglin F, Soni S, Puccini A, et al. B cell and B cell-related pathways for novel cancer treatments. Cancer Treatment Reviews. 2019; 73: 10–19.
[74]
Que H, Fu Q, Lan T, Tian X, Wei X. Tumor-associated neutrophils and neutrophil-targeted cancer therapies. Biochimica et Biophysica Acta. Reviews on Cancer. 2022; 1877: 188762.

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

Share
Back to top