191 results on '"SHMT2"'
Search Results
2. Mitochondrial outer membrane protein Samm50 protects against hypoxia-induced cardiac injury by interacting with Shmt2
- Author
-
Zhou, Yufei, Kang, Le, Xu, Ran, Zhao, Di, Wang, Jienan, Wu, Jiaying, Lin, Hong, Ding, Zhiwen, and Zou, Yunzeng
- Published
- 2024
- Full Text
- View/download PDF
3. Mild neurodevelopmental disorder due to reduced SHMT2 enzymatic activity caused by novel compound heterozygous variants: expanding the phenotypic spectrum.
- Author
-
Pan, Hu, He, Mei, Luo, Xuan, Hu, Juanli, Mao, Xiao, Cheng, Yong, and Liu, Zhen
- Abstract
Biallelic variants in SHMT2 cause neurodevelopmental disorders with cardiomyopathy, spasticity, and brain abnormalities (NEDCASB; OMIM: 619121). This recently described metabolic disorder are characterized by severe intellectual disability, microcephaly, spastic paraplegia, peripheral neuropathy, corpus callosum dysgenesis, facial and limb deformities, and progressive hypertrophic cardiomyopathy. Herein we describe the clinical characteristics of a 13 years old patient with novel compound heterozygous SHMT2 missense variants (c.1274G>A: p.R425Q and c.1042C>T: p.R348W), presenting with mild intellectual disability, corpus callosum dysgenesis, and speech delay. Different from previous cases, our patient represents the mildest phenotype reported to date, and expand the phenotypic spectrum of disease associated with SHMT2 variants. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
4. Elucidation of the Role of SHMT2 in L-Serine Homeostasis in Hypoxic Hepa1-6 Cells.
- Author
-
Zhang, Shuo, He, Ruoxu, Zhang, Mingsi, Zhang, Jingcheng, Wu, Mengting, Zhang, Guangji, and Jiang, Tao
- Subjects
- *
HEPATOCELLULAR carcinoma , *HOMEOSTASIS , *CATABOLISM , *CELL culture , *HYPOXEMIA - Abstract
Hypoxia is a characteristic feature of malignancy; however, its effect on metabolism remains unclear. In this study, Hepa1-6 cells were cultured under hypoxic conditions and their metabolites were analyzed. Elevated levels of L-serine along with increased glycolytic activity are prominent features of hypoxia. Transcriptome sequencing revealed the downregulation of genes involved in L-serine synthesis and metabolism, which was confirmed by PCR analysis and comparison with public databases. Further experimental evidence indicates that the accumulation of L-serine under hypoxic conditions is attributable not only to enhanced glycolysis but also to a reduction in the catabolism of L-serine mediated by serine hydroxymethyltransferase 2 (SHMT2). [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
5. Hypoxia-induced SHMT2 protein lactylation facilitates glycolysis and stemness of esophageal cancer cells.
- Author
-
Qiao, Zhe, Li, Yu, Li, Shaomin, Liu, Shiyuan, and Cheng, Yao
- Abstract
Esophageal cancer (EC) is a familiar digestive tract tumor with highly lethal. The hypoxic environment has been demonstrated to be a significant factor in modulating malignant tumor progression and is strongly associated with the abnormal energy metabolism of tumor cells. Serine hydroxymethyl transferase 2 (SHMT2) is one of the most frequently expressed metabolic enzymes in human malignancies. The study was designed to investigate the biological functions and regulation mechanisms of SHMT2 in EC under hypoxia. We conducted RT-qPCR to assess SHMT2 levels in EC tissues and cells (TE-1 and EC109). EC cells were incubated under normoxia and hypoxia, respectively, and altered SHMT2 expression was evaluated through RT-qPCR, western blot, and immunofluorescence. The biological functions of SHMT2 on EC cells were monitored by performing CCK-8, EdU, transwell, sphere formation, glucose uptake, and lactate production assays. The SHMT2 protein lactylation was measured by immunoprecipitation and western blot. In addition, SHMT2-interacting proteins were analyzed by bioinformatics and validated by rescue experiments. SHMT2 was notably upregulated in EC tissues and cells. Hypoxia elevated SHMT2 protein expression, augmenting EC cell proliferation, migration, invasion, stemness, and glycolysis. In addition, hypoxia triggered lactylation of the SHMT2 protein and enhanced its stability. SHMT2 knockdown impeded the malignant phenotype of EC cells. Further mechanistic studies disclosed that SHMT2 is involved in EC progression by interacting with MTHFD1L. Hypoxia-induced SHMT2 protein lactylation and upregulated its protein level, which in turn enhanced MTHFD1L expression and accelerated the malignant progression of EC cells. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
6. Multiomics analysis reveals serine catabolism as a potential therapeutic target for MELAS.
- Author
-
Liufu, Tongling, Zhao, Xutong, Yu, Meng, Xie, Zhiying, Meng, Lingchao, Lv, He, Zhang, Wei, Yuan, Yun, Xing, Guogang, Deng, Jianwen, and Wang, Zhaoxia
- Abstract
Mitochondrial disease is a devastating genetic disorder, with mitochondrial myopathy, encephalopathy, lactic acidosis, and stroke‐like episodes (MELAS) and m.3243A>G being the most common phenotype and genotype, respectively. The treatment for MELAS patients is still less effective. Here, we performed transcriptomic and proteomic analysis in muscle tissue of MELAS patients, and discovered that the expression of molecules involved in serine catabolism were significantly upregulated, and serine hydroxymethyltransferase 2 (SHMT2) increased significantly in both the mRNA and protein levels. The SHMT2 protein level was also increased in myoblasts with m.3243A>G mutation, which was transdifferentiated from patients derived fibroblasts, accompanying with the decreased nicotinamide adenine dinucleotide (NAD+)/reduced NAD+ (NADH) ratio and cell viability. After treating with SHMT2 inhibitor (SHIN1), the NAD+/NADH ratio and cell viability in MELAS myoblasts increased significantly. Taken together, our study indicates that enhanced serine catabolism plays an important role in the pathogenesis of MELAS and that SHIN1 can be a potential small molecule for the treatment of this disease. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
7. Phosphorylated SHMT2 Regulates Oncogenesis Through m6A Modification in Lung Adenocarcinoma.
- Author
-
Han, Tianyu, Wang, Yanan, Cheng, Minzhang, Hu, Qifan, Wan, Xiaorui, Huang, Menglin, Liu, Yuhan, Xun, Wenze, Xu, Jin, Wang, Lei, Luo, Ruiguang, Yuan, Yi, Wang, Keru, and Wang, Jianbin
- Subjects
- *
LUNGS , *MITOGEN-activated protein kinases , *RNA sequencing , *CARCINOGENESIS , *CHEMICAL libraries , *ADENOCARCINOMA - Abstract
Targeting cancer‐specific metabolic processes is a promising therapeutic strategy. Here, this work uses a compound library that directly inhibits metabolic enzymes to screen the potential metabolic targets in lung adenocarcinoma (LUAD). SHIN1, the specific inhibitor of serine hydroxymethyltransferase 1/2 (SHMT1/2), has a highly specific inhibitory effect on LUAD cells, and this effect depends mainly on the overexpression of SHMT2. This work clarifies that mitogen‐activated protein kinase 1 (MAPK1)‐mediated phosphorylation at Ser90 is the key mechanism underlying SHMT2 upregulation in LUAD and that this phosphorylation stabilizes SHMT2 by reducing STIP1 homology and U‐box containing protein 1 (STUB1)‐mediated ubiquitination and degradation. SHMT2‐Ser90 dephosphorylation decreases S‐adenosylmethionine levels in LUAD cells, resulting in reduced N6‐methyladenosine (m6A) levels in global RNAs without affecting total protein or DNA methylation. Methylated RNA immunoprecipitation sequencing (MeRIP‐Seq) and RNA sequencing (RNA‐Seq) analyses further demonstrate that SHMT2‐Ser90 dephosphorylation accelerates the RNA degradation of oncogenic genes by reducing m6A modification, leading to the inhibition of tumorigenesis. Overall, this study elucidates a new regulatory mechanism of SHMT2 during oncogenesis and provides a theoretical basis for targeting SHMT2 as a therapeutic target in LUAD. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
8. SHMT2 Mediates Small‐Molecule‐Induced Alleviation of Alzheimer Pathology Via the 5′UTR‐dependent ADAM10 Translation Initiation.
- Author
-
Song, Li, Pan, Qiu‐Ling, Zhou, Gui‐Feng, Liu, Sheng‐Wei, Zhu, Bing‐Lin, Lin, Pei‐Jia, Hu, Xiao‐Tong, Zha, Jing‐Si, Long, Yan, Luo, Biao, Chen, Jian, Tang, Ying, Tang, Jing, Xiang, Xiao‐Jiao, Xie, Xiao‐Yong, Deng, Xiao‐Juan, and Chen, Guo‐Jun
- Subjects
- *
RNA-binding proteins , *PEPTIDASE , *ALZHEIMER'S disease , *CHEMICAL biology - Abstract
It is long been suggested that one‐carbon metabolism (OCM) is associated with Alzheimer's disease (AD), whereas the potential mechanisms remain poorly understood. Taking advantage of chemical biology, that mitochondrial serine hydroxymethyltransferase (SHMT2) directly regulated the translation of ADAM metallopeptidase domain 10 (ADAM10), a therapeutic target for AD is reported. That the small‐molecule kenpaullone (KEN) promoted ADAM10 translation via the 5′ untranslated region (5′UTR) and improved cognitive functions in APP/PS1 mice is found. SHMT2, which is identified as a target gene of KEN and the 5′UTR‐interacting RNA binding protein (RBP), mediated KEN‐induced ADAM10 translation in vitro and in vivo. SHMT2 controls AD signaling pathways through binding to a large number of RNAs and enhances the 5′UTR activity of ADAM10 by direct interaction with GAGGG motif, whereas this motif affected ribosomal scanning of eukaryotic initiation factor 2 (eIF2) in the 5′UTR. Together, KEN exhibits therapeutic potential for AD by linking OCM with RNA processing, in which the metabolic enzyme SHMT2 "moonlighted" as RBP by binding to GAGGG motif and promoting the 5′UTR‐dependent ADAM10 translation initiation. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
9. Endoplasmic reticulum stress-dependent regulation of the expression of serine hydroxymethyltransferase 2 in glioblastoma cells
- Author
-
Minchenko Oleksandr H., Sliusar Myroslava Y., Khita Olena O., Viletska Yuliia M., Luzina Olha Y., Danilovskyi Serhiy V., and Minchenko Dmytro O.
- Subjects
ern1 knockdown ,hypoxia ,nutrient deprivation ,shmt2 ,gene expression ,u87mg cells ,Diseases of the endocrine glands. Clinical endocrinology ,RC648-665 - Abstract
Objective. Serine hydroxymethyltransferase (SHMT2) plays a multifunctional role in mitochondria (folate-dependent tRNA methylation, translation, and thymidylate synthesis). The endoplasmic reticulum stress, hypoxia, and glucose and glutamine supply are significant factors of malignant tumor growth including glioblastoma. Previous studies have shown that the knockdown of the endoplasmic reticulum to nucleus signaling 1 (ERN1) pathway of endoplasmic reticulum stress strongly suppressed glioblastoma cell proliferation and modified the sensitivity of these cells to hypoxia and glucose or glutamine deprivations. The present study aimed to investigate the regulation of the SHMT2 gene in U87MG glioblastoma cells by ERN1 knockdown, hypoxia, and glucose or glutamine deprivations with the intent to reveal the role of ERN1 signaling in sensitivity of this gene expression to hypoxia and nutrient supply.
- Published
- 2024
- Full Text
- View/download PDF
10. SHMT2 regulates esophageal cancer cell progression and immune Escape by mediating m6A modification of c-myc
- Author
-
Zhe Qiao, Yu Li, Yao Cheng, Shaomin Li, and Shiyuan Liu
- Subjects
SHMT2 ,EC ,m6A modification ,c-myc ,One carbon metabolism ,Biotechnology ,TP248.13-248.65 ,Biology (General) ,QH301-705.5 ,Biochemistry ,QD415-436 - Abstract
Abstract Background In recent years, the role of altered cellular metabolism in tumor progression has attracted widespread attention. Related metabolic enzymes have also been considered as potential cancer therapeutic targets. Serine hydroxymethyltransferase 2 (SHMT2) has been reported to be upregulated in several cancers and associated with poor prognosis. However, there are few studies of SHMT2 in esophageal cancer (EC), and the related functions and mechanisms also need to be further explored. Methods In this study, we first analyzed SHMT2 expression in EC by online database and clinical samples. Then, the biological functions of SHMT2 in EC were investigated by cell and animal experiments. The intracellular m6A methylation modification levels were also evaluated by MeRIP. Linked genes and mechanisms of SHMT2 were analyzed by bioinformatics and rescue experiments. Results We found that SHMT2 expression was abnormally upregulated in EC and associated with poor prognosis. Functionally, SHMT2 silencing suppressed c-myc expression in an m6A-dependent manner, thereby blocking the proliferation, migration, invasion and immune escape abilities of EC cells. Mechanistically, SHMT2 encouraged the accumulation of methyl donor SAM through a one-carbon metabolic network, thereby regulating the m6A modification and stability of c-myc mRNA in a METTL3/FTO/ALKBH5/IGF2BP2-dependent way. In vivo animal experiments also demonstrated that SHMT2 mediated MYC expression by m6A-methylation modification, thus boosting EC tumorigenesis. Conclusion In conclusion, our data illustrated that SHMT2 regulated malignant progression and immune escape of EC cell through c-myc m6A modification. These revealed mechanisms related to SHMT2 in EC and maybe offer promise for the development of new therapeutic approaches.
- Published
- 2023
- Full Text
- View/download PDF
11. SHMT2 Promotes Invasion and Migration of Breast Cancer Cells Through Binding to and Up-regulating HAX1
- Author
-
CHEN Qianzhi, SHEN Na, ZHANG Ning, and CHEN Yan
- Subjects
breast cancer ,shmt2 ,shmt2 inhibitor ,hax1 ,invasion ,migration ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Objective To explore the molecular mechanism of SHMT2 regulating the invasion and migration of breast cancer cells. Methods Bioinformatics analysis was used to verify the role of SHMT2 in breast cancer tissues. Transwell assay was used to detect the changes of invasion and migration abilities of breast cancer cells. Co-immunoprecipitation, knockdown plasmid transfection and Western blot were used to determine the regulatory relationship between different proteins. Results Bioinformatics analysis showed that the expression level of SHMT2 in invasive breast cancer tissues was significantly higher than that in adjacent normal tissues (P < 0.001). The 5-year disease-specific survival and overall survival in the SHMT2 high expression group were significantly lower than those in the SHMT2 low expression group (both P < 0.001). Transwell assay showed that SHMT2 knockdown significantly reduced the invasion ability (t=5.375, P=0.0058) and migration ability (t=6.274, P=0.0033) of MCF7 cells. Western blot showed that SHMT2 could combine to HAX1, and knockdown of SHMT2 reduced the protein level of HAX1. Transwell assay showed that the inhibitory effect of SHMT2 knockdown on the migration of MCF7 cells could be reversed by overexpression of HAX1 (t=6.274, P=0.0033; t=8.041, P=0.0013), while SHMT2 inhibitor (SHIN1, 10 nmol/L) significantly inhibited the migration of MCF7 cells induced by SHMT2 overexpression (t=10.16, P=0.0005; t=8.741, P=0.0009). Conclusion SHMT2 was closely related to the poor prognosis of breast cancer, and was a key factor in the invasion and migration of breast cancer cells. The mechanism was that SHMT2 increased the invasion and migration ability of breast cancer cells by binding to and up-regulating HAX1. It was verified that SHMT2 inhibitor could significantly reduce the migration ability of breast cancer cells. This study explored the therapeutic potential of SHMT2 inhibitor in metastatic breast cancer, and found potential intervention targets for its clinical treatment.
- Published
- 2023
- Full Text
- View/download PDF
12. The expression and clinical significance of serine hydroxymethyltransferase2 in gastric cancer
- Author
-
Yiming Shan, Dongdong Liu, Yingze Li, Chu Wu, and Yanwei Ye
- Subjects
SHMT2 ,Gastric cancer ,Prognosis ,Biomarker ,Medicine ,Biology (General) ,QH301-705.5 - Abstract
Background Gastric cancer (GC) is one of the most common malignant tumours in the digestive system. Serine hydroxymethyltransferase 2 (SHMT2) is one of the key enzymes associated with serine metabolism. However, the prognostic role of SHMT2 in GC carcinogenesis has yet to be studied. Methods The expression of SHMT2 in human tumors and normal tissues was detected by the Assistant for Clinical Bioinformatics and Immunohistochemistry (IHC). The relationship of the expression of SHMT2 with clinical characteristics and survival data was analysed by the chi-square test, survival analysis and online databases. Finally, the correlation between SHMT2 expression and associated signalling channels, and molecules was analysed by online databases. Results SHMT2 was strongly expressed in numerous human cancers. The expression rate of SHMT2 was 56.44% in GC (P = 0.018). The survival analysis indicated that patients with high expression of SHMT2 had the worse overall survival (OS; log-rank P = 0.007). The expression of SHMT2 was correlated with tumour size (P = 0.034) and, TNM stage (P = 0.042). In particular, SHMT2, vessel invasion and M stage were independent factors for OS in GC (P = 0.044, P < 0.001, P < 0.001). The SHMT2 gene was substantially correlated with cell signalling pathways. Conclusions SHMT2 is highly expressed in GC and is associated with a poor prognosis. The exploration of its mechanism may be related to tumour proliferation, DNA repair and replication. SHMT2 is an independent prognostic risk factor and a potential biomarker for the diagnosis and treatment of GC.
- Published
- 2024
- Full Text
- View/download PDF
13. SHMT2 regulates esophageal cancer cell progression and immune Escape by mediating m6A modification of c-myc.
- Author
-
Qiao, Zhe, Li, Yu, Cheng, Yao, Li, Shaomin, and Liu, Shiyuan
- Subjects
ESOPHAGEAL cancer ,CANCER invasiveness ,CANCER cells ,ANIMAL experimentation ,ONLINE databases - Abstract
Background: In recent years, the role of altered cellular metabolism in tumor progression has attracted widespread attention. Related metabolic enzymes have also been considered as potential cancer therapeutic targets. Serine hydroxymethyltransferase 2 (SHMT2) has been reported to be upregulated in several cancers and associated with poor prognosis. However, there are few studies of SHMT2 in esophageal cancer (EC), and the related functions and mechanisms also need to be further explored. Methods: In this study, we first analyzed SHMT2 expression in EC by online database and clinical samples. Then, the biological functions of SHMT2 in EC were investigated by cell and animal experiments. The intracellular m6A methylation modification levels were also evaluated by MeRIP. Linked genes and mechanisms of SHMT2 were analyzed by bioinformatics and rescue experiments. Results: We found that SHMT2 expression was abnormally upregulated in EC and associated with poor prognosis. Functionally, SHMT2 silencing suppressed c-myc expression in an m6A-dependent manner, thereby blocking the proliferation, migration, invasion and immune escape abilities of EC cells. Mechanistically, SHMT2 encouraged the accumulation of methyl donor SAM through a one-carbon metabolic network, thereby regulating the m6A modification and stability of c-myc mRNA in a METTL3/FTO/ALKBH5/IGF2BP2-dependent way. In vivo animal experiments also demonstrated that SHMT2 mediated MYC expression by m6A-methylation modification, thus boosting EC tumorigenesis. Conclusion: In conclusion, our data illustrated that SHMT2 regulated malignant progression and immune escape of EC cell through c-myc m6A modification. These revealed mechanisms related to SHMT2 in EC and maybe offer promise for the development of new therapeutic approaches. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
14. Vital role of SHMT2 in diverse disease.
- Author
-
Ma, Wenqi, Liu, Ronghan, Zhao, Kai, and Zhong, Jiangbo
- Subjects
- *
CANCER cell growth , *NUCLEOTIDE synthesis , *ENZYME metabolism , *CANCER cell proliferation , *CARCINOGENESIS - Abstract
One-carbon metabolism is essential for our human cells to carry out nucleotide synthesis, methylation, and reductive metabolism through one-carbon units, and these pathways ensure the high proliferation rate of cancer cells. Serine hydroxymethyltransferase 2 (SHMT2) is a key enzyme in one-carbon metabolism. This enzyme can convert serine into a one-carbon unit bound to tetrahydrofolate and glycine, ultimately supporting the synthesis of thymidine and purines and promoting the growth of cancer cells. Due to SHMT2's crucial role in the one-carbon cycle, it is ubiquitous in human cells and even in all organisms and highly conserved. Here, we summarize the impact of SHMT2 on the progression of various cancers to highlight its potential use in the development of cancer treatments. • SHMT2 is a relatively novel research goal in recent years. • SHMT2 is a key enzyme in one-carbon metabolism. • SHMT2 potential use in the development of cancer treatments. • This study aims to understand the progression of various cancers and explore long-term treatment options. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
15. SHMT2通过结合并上调HAX1促进乳腺癌 细胞的侵袭和迁移.
- Author
-
陈前智, 沈娜, 张宁, and 陈嫣
- Abstract
Copyright of Cancer Research on Prevention & Treatment is the property of Cancer Research on Prevention & Treatment Editorial Office and its content may not be copied or emailed to multiple sites or posted to a listserv without the copyright holder's express written permission. However, users may print, download, or email articles for individual use. This abstract may be abridged. No warranty is given about the accuracy of the copy. Users should refer to the original published version of the material for the full abstract. (Copyright applies to all Abstracts.)
- Published
- 2023
- Full Text
- View/download PDF
16. Serine Catabolism Feeds NADH when Respiration Is Impaired
- Author
-
Yang, Lifeng, Garcia Canaveras, Juan Carlos, Chen, Zihong, Wang, Lin, Liang, Lingfan, Jang, Cholsoon, Mayr, Johannes A, Zhang, Zhaoyue, Ghergurovich, Jonathan M, Zhan, Le, Joshi, Shilpy, Hu, Zhixian, McReynolds, Melanie R, Su, Xiaoyang, White, Eileen, Morscher, Raphael J, and Rabinowitz, Joshua D
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Animals ,Cell Hypoxia ,Cell Line ,Humans ,Mice ,Mice ,Inbred C57BL ,Mice ,Nude ,Mitochondria ,NAD ,Oxygen ,Serine ,MTHFD2 ,NADH ,SHMT2 ,complex I inhibitor ,hypoxia ,methylene tetrahydrofolate dehydrogenase ,mitochondrial disease ,redox ,respiration inhibition ,serine catabolism ,serine hydroxymethyltransferase ,Medical Biochemistry and Metabolomics ,Endocrinology & Metabolism ,Biochemistry and cell biology ,Medical biochemistry and metabolomics - Abstract
NADH provides electrons for aerobic ATP production. In cells deprived of oxygen or with impaired electron transport chain activity, NADH accumulation can be toxic. To minimize such toxicity, elevated NADH inhibits the classical NADH-producing pathways: glucose, glutamine, and fat oxidation. Here, through deuterium-tracing studies in cultured cells and mice, we show that folate-dependent serine catabolism also produces substantial NADH. Strikingly, when respiration is impaired, serine catabolism through methylene tetrahydrofolate dehydrogenase (MTHFD2) becomes a major NADH source. In cells whose respiration is slowed by hypoxia, metformin, or genetic lesions, mitochondrial serine catabolism inhibition partially normalizes NADH levels and facilitates cell growth. In mice with engineered mitochondrial complex I deficiency (NDUSF4-/-), serine's contribution to NADH is elevated, and progression of spasticity is modestly slowed by pharmacological blockade of serine degradation. Thus, when respiration is impaired, serine catabolism contributes to toxic NADH accumulation.
- Published
- 2020
17. Understanding The Regulatory Role of USP32 and SHMT2 in The Progression of Gastric Cancer
- Author
-
Jun Li, Yafei Bo, Bo Ding, and Lei Wang
- Subjects
cancer ,gastric cancer ,h. pylori ,shmt2 ,usp32 ,Medicine ,Science - Abstract
Objective: Gastric cancer is the fifth most common neoplasm and the fourth reason for mortality globally. Incidencerates are highly variable and dependent on risk factors, epidemiologic and carcinogenesis patterns. Previous studiesreported that Helicobacter pylori (H. pylori) infection is one the strongest known risk factor for gastric cancer. USP32 isa deubiquitinating enzyme identified as a potential factor associated with tumor progression and a key player in cancerdevelopment. On the other hand, SHMT2 is involved in serine-glycine metabolism to support cancer cell proliferation.Both USP32 and SHMT2 are reported to be upregulated in many cancer types, including gastric cancer, but its completemechanism is not fully explored yet. The present study explored possible mechanism of action of USP32 and SHMT2in the progression of gastric cancer.Materials and Methods: In this experimental study, Capsaicin (0.3 g/kg/day) and H. pylori infection combination wasused to successfully initiate gastric cancer conditions in mice. It was followed by 40 and 70 days of treatment toestablish initial and advanced conditions of gastric cancer.Results: Histopathology confirmed formation of signet ring cell and initiation of cellular proliferation in the initial gastriccancer. More proliferative cells were also observed. In addition, tissue hardening was confirmed in the advancedstage of gastric cancer. USP32 and SHMT2 showed progressive upregulated expression, as gastric cancer progress.Immunohistologically, it showed signals in abnormal cells and high-intensity signals in the advanced stage of cancer.In USP32 silenced tissue, expression of SHMT2 was completely blocked and reverted cancer development as evidentwith less abnormal cell in initial gastric cancer. Reduction of SHMT2 level to one-fourth was observed in the advancedgastric cancer stages of USP32 silenced tissue.Conclusion: USP32 had a direct role in regulating SHMT2 expression, which attracted therapeutic target for futuretreatment.
- Published
- 2023
- Full Text
- View/download PDF
18. Knockdown of SHMT2 enhances the sensitivity of gastric cancer cells to radiotherapy through the Wnt/β-catenin pathway
- Author
-
Mao Yu and Zhang Tiyong
- Subjects
gastric cancer ,shmt2 ,apoptosis ,radiosensitivity ,wnt/β-catenin pathway ,Biology (General) ,QH301-705.5 - Abstract
Gastric cancer (GC) is one of the most common malignant tumors. The mechanism of GC radioresistance and new radiosensitizers must be revealed and developed to treat GC. Serine hydroxymethyltransferase 2 (SHMT2) is responsible for encoding the mitochondrial form of the pyridoxal phosphate-dependent enzyme. SHMT2 plays a critical role in several types of cancers, while its possible effect on the radiological resistance in GC is still unclear. In this study, we investigated the role of SHMT2 in the radiological resistance of GC. Our data confirmed that SHMT2 was highly expressed in radiation-resistant GC cells. SHMT2 reduced the radiosensitivity of GC cells. In addition, SHMT2 is involved in radiation-induced GC cell apoptosis. Further, SHMT2 regulated the Wnt/β-catenin pathway, therefore reducing the radiosensitivity of GC cells in vivo. In conclusion, we revealed that depletion of SHMT2 enhanced the sensitivity of GC cells to interventional radiotherapy through the Wnt/β-catenin pathway.
- Published
- 2022
- Full Text
- View/download PDF
19. Pairing structural reconstruction with catalytic competence to evaluate the mechanisms of key enzymes in the folate‐mediated one‐carbon pathway.
- Author
-
Zhao, Li Na and Kaldis, Philipp
- Subjects
- *
NUCLEOTIDE synthesis , *ENZYMES , *OXIDATION states , *FOLIC acid , *CELL proliferation , *DRUG development - Abstract
Mammalian metabolism comprises a series of interlinking pathways that include two major cycles: the folate and methionine cycles. The folate‐mediated metabolic cycle uses several oxidation states of tetrahydrofolate to carry activated one‐carbon units to be readily used and interconverted within the cell. They are required for nucleotide synthesis, methylation and metabolism, and particularly for proliferation of cancer cells. Based on the latest progress in genome‐wide CRISPR loss‐of‐function viability screening of 789 cell lines, we focus on the most cancer‐dependent enzymes in this pathway, especially those that are hyperactivated in cancer, to provide new insight into the chemical basis for cancer drug development. Since the complete 3D structure of several of these enzymes of the one‐carbon pathway in their active form are not available, we used homology modelling integrated with the interpretation of the reaction mechanism. In addition, have reconstructed the most likely scenario for the reactions taking place paired with their catalytic competence that provides a testable framework for this pathway. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
20. SHMT2 Promotes Gastric Cancer Development through Regulation of HIF1α/VEGF/STAT3 Signaling.
- Author
-
Wang, Weida, Wang, Mingjin, Du, Tingting, Hou, Zhenyan, You, Shen, Zhang, Sen, Ji, Ming, Xue, Nina, and Chen, Xiaoguang
- Subjects
- *
STOMACH cancer , *CARCINOGENESIS , *NEOVASCULARIZATION , *INHIBITION of cellular proliferation , *REGULATOR genes , *CELL lines , *HOMEOSTASIS - Abstract
The metabolic enzymes involved in one-carbon metabolism are closely associated with tumor progression and could be potential targets for cancer therapy. Recent studies showed that serine hydroxymethyltransferase 2 (SHMT2), a crucial enzyme in the one-carbon metabolic pathway, plays a key role in tumor proliferation and development. However, the precise role and function of SHMT2 in gastric cancer (GC) remain poorly understood. In this study, we presented evidence that SHMT2 was necessary for hypoxia-inducible factor-1α (HIF1α) stability and contributed to GC cells' hypoxic adaptation. The analysis of datasets retrieved from The Cancer Genome Atlas and the experimentation with human cell lines revealed a marked increase in SHMT2 expression in GC. The SHMT2 knockdown in MGC803, SGC7901, and HGC27 cell lines inhibited cell proliferation, colony formation, invasion, and migration. Notably, SHMT2 depletion disrupted redox homeostasis and caused glycolytic function loss in GC cells under hypoxic circumstances. Mechanistically, we discovered SHMT2 modulated HIF1α stability, which acted as a master regulator of hypoxia-inducible genes under hypoxic conditions. This, in turn, regulated the downstream VEGF and STAT3 pathways. The in vivo xenograft experiments showed that SHMT2 knockdown markedly reduced GC growth. Our results elucidate the novel function of SHMT2 in stabilizing HIF1α under hypoxic conditions, thus providing a potential therapeutic strategy for GC treatment. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
21. Impairments in SHMT2 expression or cellular folate availability reduce oxidative phosphorylation and pyruvate kinase activity.
- Author
-
Fiddler, Joanna L., Blum, Jamie E., Heyden, Katarina E., Castillo, Luisa F., Thalacker-Mercer, Anna E., and Field, Martha S.
- Abstract
Background: Serine hydroxymethyltransferase 2 (SHMT2) catalyzes the reversible conversion of tetrahydrofolate (THF) and serine-producing THF-conjugated one-carbon units and glycine in the mitochondria. Biallelic SHMT2 variants were identified in humans and suggested to alter the protein's active site, potentially disrupting enzymatic function. SHMT2 expression has also been shown to decrease with aging in human fibroblasts. Immortalized cell models of total SHMT2 loss or folate deficiency exhibit decreased oxidative capacity and impaired mitochondrial complex I assembly and protein levels, suggesting folate-mediated one-carbon metabolism (FOCM) and the oxidative phosphorylation system are functionally coordinated. This study examined the role of SHMT2 and folate availability in regulating mitochondrial function, energy metabolism, and cellular proliferative capacity in both heterozygous and homozygous cell models of reduced SHMT2 expression. In this study, primary mouse embryonic fibroblasts (MEF) were isolated from a C57Bl/6J dam crossed with a heterozygous Shmt2
+/− male to generate Shmt2+/+ (wild-type) or Shmt2+/− (HET) MEF cells. In addition, haploid chronic myeloid leukemia cells (HAP1, wild-type) or HAP1 cells lacking SHMT2 expression (ΔSHMT2) were cultured for 4 doublings in either low-folate or folate-sufficient culture media. Cells were examined for proliferation, total folate levels, mtDNA content, protein levels of pyruvate kinase and PGC1α, pyruvate kinase enzyme activity, mitochondrial membrane potential, and mitochondrial function. Results: Homozygous loss of SHMT2 in HAP1 cells impaired cellular folate accumulation and altered mitochondrial DNA content, formate production, membrane potential, and basal respiration. Formate rescued proliferation in HAP1, but not ΔSHMT2, cells cultured in low-folate medium. Pyruvate kinase activity and protein levels were impaired in ΔSHMT2 cells and in MEF cells exposed to low-folate medium. Mitochondrial biogenesis protein levels were elevated in Shmt2+/− MEF cells, while mitochondrial mass was increased in both homozygous and heterozygous models of SHMT2 loss. Conclusions: The results from this study indicate disrupted mitochondrial FOCM impairs mitochondrial folate accumulation and respiration, mitochondrial formate production, glycolytic activity, and cellular proliferation. These changes persist even after a potentially compensatory increase in mitochondrial biogenesis as a result of decreased SHMT2 levels. [ABSTRACT FROM AUTHOR]- Published
- 2023
- Full Text
- View/download PDF
22. SHMT2 regulates CD8+ T cell senescence via the reactive oxygen species axis in HIV-1 infected patients on antiretroviral therapy.
- Author
-
Zhang QS, Wang JN, Yang TL, Li SY, Li JQ, Liu DN, Shang H, and Zhang ZN
- Abstract
Background: Although antiretroviral therapy (ART) effectively inhibits viral replication, it does not fully mitigate the immunosenescence instigated by HIV infection. Cellular metabolism regulates cellular differentiation, survival, and senescence. Serine hydroxymethyltransferase 2 (SHMT2) is the first key enzyme for the entry of serine into the mitochondria from the de novo synthesis pathway that orchestrates its conversion glutathione (GSH), a key molecule in neutralising ROS and ensuring the stability of the immune system. It remains incompletely understood whether SHMT2 is involved in the senescence of CD8+ T cells, crucial for immune vigilance against HIV., Methods: HIV-infected individuals receiving antiretroviral therapy were enrolled in our study. SHMT2-siRNA was electroporated into T cells to disrupt the gene expression of SHMT2, followed by the quantification of mRNA levels of crucial serine metabolism enzymes using real-time PCR. Immunophenotyping, proliferation, cellular and mitochondrial function, and senescence-associated signalling pathways were examined using flow cytometry in CD8+ T cell subsets., Findings: Our findings revealed that CD8+ T cells in HIV-infected subjects are inclined towards senescence, and we identified that SHMT2, a key enzyme in serine metabolism, plays a role in CD8+ T cell senescence. SHMT2 can regulate glutathione (GSH) synthesis and protect mitochondrial function, thus effectively controlling intracellular reactive oxygen species (ROS) levels. Moreover, SHMT2 significantly contributes to averting immunosenescence and sustaining CD8+ T cell competence by modulating downstream DNA damage and phosphorylation cascades in pathways intricately linked to cellular senescence. Additionally, our study identified glycine can ameliorate CD8+ T cell senescence in HIV-infected individuals., Interpretation: Decreased SHMT2 levels in HIV-infected CD8+ T cells affect ROS levels by altering mitochondrial function and GSH content. Increased ROS levels activate senescence-related signalling pathways in the nucleus. However, glycine supplementation counteracts these effects and moderates senescence., Funding: This study was supported by grants from the National Key R&D Program of China (2021YFC2301900-2021YFC2301901), National Natural Science Foundation of China (82372240), and Department of Science and Technology of Liaoning Province Project for the High-Quality Scientific and Technological Development of China Medical University (2022JH2/20200074)., Competing Interests: Declaration of interests All authors declare no conflicts of interest., (Copyright © 2025 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2025
- Full Text
- View/download PDF
23. SHMT as a Potential Therapeutic Target for Renal Cell Carcinoma
- Author
-
Yongli Situ, Juying Zhang, Wenyu Liao, Quanyan Liang, Lingling Lu, Qinying Xu, Jv Chen, Xiaoyong Lu, Yongshi Cui, Zheng Shao, and Li Deng
- Subjects
shmt1 ,shmt2 ,renal cell carcinoma ,target prediction ,gene regulation network ,Biochemistry ,QD415-436 ,Biology (General) ,QH301-705.5 - 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.
- Published
- 2023
- Full Text
- View/download PDF
24. SHMT2 Induces Stemness and Progression of Head and Neck Cancer.
- Author
-
Jin, Yanli, Jung, Seung-Nam, Lim, Mi Ae, Oh, Chan, Piao, Yudan, Kim, Hae Jong, Nguyena, QuocKhanh, Kang, Yea Eun, Chang, Jae Won, Won, Ho-Ryun, and Koo, Bon Seok
- Subjects
- *
HEAD & neck cancer , *CANCER cell proliferation , *CELLULAR control mechanisms , *LYMPHATIC metastasis , *NOTCH genes - Abstract
Various enzymes in the one-carbon metabolic pathway are closely related to the development of tumors, and they can all be potential targets for cancer therapy. Serine hydroxymethyltransferase2 (SHMT2), a key metabolic enzyme, is very important for the proliferation and growth of cancer cells. However, the function and mechanism of SHMT2 in head and neck cancer (HNC) are not clear. An analysis of The Cancer Genome Atlas (TCGA) data showed that the expression of SHMT2 was higher in tumor tissue than in normal tissue, and its expression was significantly associated with male sex, aggressive histological grade, lymph node metastasis, distant metastasis, advanced TNM stage, and lymphovascular invasion in HNC. SHMT2 knockdown in FADU and SNU1041 cell lines significantly inhibited cell proliferation, colony formation, migration, and invasion. Additionally, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses using TCGA data revealed that SHMT2 was closely related to cancer stem cell regulation and maintenance. Furthermore, we found that silencing SHMT2 inhibited the expression of stemness markers and tumor spheroid formation compared with a control group. On the contrary, stemness markers were significantly increased after SHMT2 overexpression in HEP-2 cells. Interestingly, we found that knocking down SHMT2 reduced the expression of genes related to the Notch and Wnt pathways. Finally, silencing SHMT2 significantly reduced tumor growth and decreased stemness markers in a xenograft model. Taken together, our study suggests that targeting SHMT2 may play an important role in inhibiting HNC progression. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
25. SHMT2 expression as a diagnostic and prognostic marker for thyroid cancer
- Author
-
Meihua Jin, Woo Kyung Lee, Mi-Hyeon You, Ahreum Jang, Sheue-yann Cheng, Won Gu Kim, Min Ji Jeon, and Yu-Mi Lee
- Subjects
thyroid cancer ,shmt2 ,diagnosis ,prognosis ,Diseases of the endocrine glands. Clinical endocrinology ,RC648-665 - Abstract
Background: Catabolism of serine via serine hydroxymethyltransferase2 (SHMT2) through the mitochondrial one-carbon unit pathway is important in tumorigenesis. Therefore, SHMT2 may play a role in thyroid cancer. Methods: Thyroid tissue samples and The Cancer Genome Atlas (TCGA) database were used to evaluate SHMT2 expression in thyroid tissues and the association with clinical outcomes. Results: SHMT2 protein expression was evaluated in thyroid tissues consisting of 52 benign nodules, 129 papillary thyroid carcinomas (PTC) and matched normal samples, and 20 anaplastic thyroid carcinomas (ATC). ATCs presented the highest (95.0%) positivity of SMHT2 protein expression. PTCs showed the second highest (73.6%) positivity of SHMT2 expression, which was significantly higher than that of be nign nodules (19.2%, P = 0.016) and normal thyroid tissues (0%, P < 0.001). Analysis of TCGA data showed that SHMT2 messenger RNA (mRNA) expression was significantly higher in tumo rs than in normal tissues (P < 0.001). When we classified thyroid cancer into high and low gr oups according to SHMT2 mRNA expression levels, the thyroid differentiation score for t he high SHMT2 group was significantly lower than that of the low SH MT2 group (P < 0.001). There was also a significant correlation between SHMT2 mRNA expression and the stemness index (r = 0.41, P < 0.001). The high SHMT2 group had more advanced TNM stages and shorter progression-free survival rates than the low SHMT2 group (P < 0.01 and P = 0.007, respectively). Conclusion: SHMT2 expression is higher in thyroid cancers than normal or benign tissues and is associated with de-differentiation and poor clinical outc omes. Thus, SHMT2 might be useful as a diagnostic and prognostic marker for thyroid cancer.
- Published
- 2021
- Full Text
- View/download PDF
26. SHMT2 regulates serine metabolism to promote the progression and immunosuppression of papillary renal cell carcinoma.
- Author
-
Weiyu Kong, Zhongyuan Wang, Nuoran Chen, Yiwen Mei, Yang Li, and Yulin Yue
- Subjects
RENAL cell carcinoma ,SERINE ,GENE expression profiling ,PROGNOSIS ,TUMOR microenvironment - Abstract
Recent research has demonstrated the diverse relationship between tumour metabolism and the tumour microenvironment (TME), for example, abnormal serine metabolism. This study investigated the role of serine metabolism in papillary renal cell carcinoma (pRCC) focusing on the prognostic value and regulatory mechanisms. Gene expression profiles and clinical data of patients with pRCC were obtained from The Cancer Genome Atlas (TCGA) database and Gene Expression Omnibus (GEO) database. Kaplan–Meier curves were used for survival analysis and consensus clustering for tumour serine metabolic signatures extraction. Functional analysis, including the Kyoto Encyclopedia of Genes and Genomes (KEGG) and gene set enrichment analysis (GSEA), was applied to explore the biological characteristics. The gene set variation analysis (GSVA), single-sample GSEA (ssGSEA), and Estimation of Stromal and Immune cells in Malignant Tumour tissues using Expression data (ESTIMATE) methods were utilised to estimate the immune infiltration in the various subtypes. Five serine metabolic genes (SMGs) were used to classify patients with pRCC, with four clusters identified with diverse prognoses and immune features based on these survival-related SMGs. Further analysis of the best and worst clusters (B and D clusters) revealed variations in survival, clinical progression, oncogenic pathways, and TME, which included immune infiltration scores, immunosuppressive cell infiltration, and expression of immune checkpoints. In addition, SMGs, especially SHMT2, exacerbated the carcinogenesis and immunosuppressive cells in pRCC, thus promoting tumour proliferation. In conclusion, higher SHMT2 gene expression and higher serine metabolism in tumour cells are associated with poorer clinical outcomes in pRCC. SHMT2 is a potential novel target gene for targeted therapy and immunotherapy in pRCC. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
27. Silencing SHMT2 inhibits the progression of tongue squamous cell carcinoma through cell cycle regulation
- Author
-
Yan Liao, Fang Wang, Yadong Zhang, Hongshi Cai, Fan Song, and Jinsong Hou
- Subjects
SHMT2 ,Tongue squamous cell carcinoma ,Survival analysis ,Cell cycle ,Weighted gene co-expression network analysis ,Gene set enrichment analysis ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 ,Cytology ,QH573-671 - Abstract
Abstract Background Serine hydroxymethyltransferase 2 (SHMT2) is a vital metabolic enzyme in one carbon metabolism catalyzing the conversion of serine to glycine, which has been reported to play a crucial role in the progression of tumors. However, its function in tongue squamous cell carcinoma (TSCC) remains unclear. Methods SHMT2 expression was analyzed using samples in online databases, and was assessed through immunohistochemistry staining of collected clinical specimens. The correlation between SHMT2 expression and the cell cycle was predicted through bioinformatic analysis, including weighted gene co-expression network analysis (WGCNA) and gene set enrichment analysis (GSEA). After transfection with siRNA, CCK8 assay, Edu staining, flow cytometry, trans-well assay, and wound healing experiments were performed to verify the functional role of SHMT2 in vitro. A stable cell line with SHMT2 silencing was established to detect the oncogenic function of SHMT2 in vivo. Results The expression of SHMT2 was up-regulated in TSCC tissues and cell lines compared with normal groups, and highly expressed SHMT2 significantly indicated a poorer clinical outcome for TSCC patients. Bioinformatic analysis found that high expression of SHMT2 was closely related with biologic process including cell cycle and cell cycle G1/S transition. Down regulating of SHMT2 significantly suppressed the proliferation, invasive and migrative ability of TSCC cells, and induced the prolongation of the G1 phase of the cell cycle in vitro. Furthermore, western blot showed that cell cycle-related regulators such as cyclin-dependent kinase 4 (CDK4) and cyclinD1 expression levels were decreased, while the expression levels of the cyclin-dependent kinase inhibitors p21Cip1 and p27Kip1 were increased after SHMT2 knockdown. Silencing SHMT2 in the HN6 cell line using short hairpin RNA also impeded tumor growth in vivo. Conclusions Overexpression of SHMT2 in TSCC indicated low survival rates, and was associated with aggressive behaviors of TSCC. It was also found to be involved in cell cycle regulation of TSCC cells. SHMT2 may serve as a novel prognostic indicator of TSCC.
- Published
- 2021
- Full Text
- View/download PDF
28. The role of SHMT2 in modulating lipid metabolism in hepatocytes via glycine-mediated mTOR activation.
- Author
-
Choi, You-Jin, Lee, Geunhye, Yun, Sung Ho, Lee, Wonseok, Yu, Jieun, Kim, Sang Kyum, and Lee, Byung-Hoon
- Subjects
- *
LIPID metabolism , *LIVER cells , *RNA metabolism , *FATTY liver , *FATTY acids , *METHYL groups - Abstract
Serine hydroxymethyltransferase 2 (SHMT2) converts serine into glycine in the mitochondrial matrix, transferring a methyl group to tetrahydrofolate. SHMT2 plays an important role in the maintenance of one-carbon metabolism. Previously, we found a negative correlation between the serine concentration and the progression of fatty liver disease (FLD). However, little is known about the role of SHMT2 in hepatic lipid metabolism. We established SHMT2 knockdown (KD) mouse primary hepatocytes using RNA interference to investigate the role of SHMT2 in lipid metabolism. SHMT2 KD hepatocytes showed decreased lipid accumulation with reduced glycine levels compared to the scramble cells, which was restored upon reintroducing SHMT2. SHMT2 KD hepatocytes showed downregulation of the mTOR/PPARɣ pathway with decreased gene expression related to lipogenesis and fatty acid uptake. Pharmacological activation of mTOR or PPARɣ overexpression blocked the inhibitory effect of SHMT2 KD on lipid accumulation. We also showed that glycine activated mTOR/PPARɣ signaling and identified glycine as a mediator of SHMT2-responsive lipid accumulation in hepatocytes. In conclusion, silencing SHMT2 in hepatocytes ameliorates lipid accumulation via the glycine-mediated mTOR/PPARɣ pathway. Our findings underscore the possibility of SHMT2 as a therapeutic target of FLD. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
29. Circ_0072995 Promotes Cell Carcinogenesis via Up-Regulating miR-149-5p-Mediated SHMT2 in Breast Cancer
- Author
-
Qi C, Qin X, Zhou Z, Wang Y, Yang Q, and Liao T
- Subjects
circ_0072995 ,mir-149-5p ,shmt2 ,breast cancer ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Chuang Qi,1,* Xianxiong Qin,2,* Zuozhi Zhou,1 Yan Wang,1 Qin Yang,1 Tianzhi Liao1 1Department of Oncology, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi 445000, Hubei, People’s Republic of China; 2Department of Breast Surgery, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi 445000, Hubei, People’s Republic of China*These authors contributed equally to this workCorrespondence: Tianzhi LiaoDepartment of Oncology, The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, No. 158 Wuyang Street, Enshi 445000, Hubei, People’s Republic of ChinaTel +86-718-8233580Email kzj3dgk@163.comBackground: Circ_0072995 is a novel identified circRNA and has been identified to involve in the metastasis of breast cancer. However, the detailed function and mechanism of circ_0072995 in the biological property of breast cancer cell remain vague.Materials and Methods: The expression of circ_0072995, microRNA (miR)-149-5p and serine hydroxymethyltransferase 2 (SHMT2) mRNA was detected using quantitative real-time polymerase chain reaction. Western blot was used to detect the protein levels of SHMT2, hexokinase-2 (HK-2), lactate dehydrogenase a chain (LDHA), and glucose transporter 1 (GLUT1). Cell proliferation, apoptosis, migration, and invasion were analyzed using cell counting kit-8 assay, flow cytometry, caspase-3 activity analysis, cell adhesion assay and transwell assay, respectively. Glucose metabolism was calculated by measuring glucose uptake, lactate production, and adenosine triphosphate (ATP) levels. The interaction between miR-149-5p and circ_0072995 or SHMT2 was confirmed by dual-luciferase reporter assay. In vivo tumorigenesis was performed using the murine xenograft model.Results: Circ_0072995 and SHMT2 were up-regulated in breast cancer tissues and cell lines, and knockdown of circ_0072995 or SHMT2 suppressed cell malignant properties and anaerobic glycolysis; importantly, SHMT2 overexpression attenuated the anticancer action of circ_0072995 knockdown in breast cancer. Besides, we also found circ_0072995 directly targeted miR-149-5p, thereby regulating its downstream gene SHMT2 by competitively binding to miR-149-5p. Additionally, xenograft analysis showed circ_0072995 silencing suppressed tumor growth via regulating SHMT2 and miR-149-5p in vivo.Conclusion: This study demonstrated that circ_0072995 promoted cell malignant phenotypes and anaerobic glycolysis in breast cancer via up-regulating SHMT2 through sponging miR-149-5p, indicating a promising molecular target for breast cancer treatment.Keywords: circ_0072995, miR-149-5p, SHMT2, breast cancer
- Published
- 2020
30. Evaluating the clinical significance of SHMT2 and its co-expressed gene in human kidney cancer
- Author
-
Huan Wang, Tie Chong, Bo-Yong Li, Xiao-San Chen, and Wen-Bo Zhen
- Subjects
SHMT2 ,NDUFA4L2 ,Prognosis ,Kidney cancer ,Bioinformatic analysis ,Biology (General) ,QH301-705.5 - Abstract
Abstract Background Kidney cancer is one of the most common cancers in the world. It is necessary to clarify its underlying mechanism and find its prognostic biomarkers. Current studies showed that SHMT2 may be participated in several kinds of cancer. Methods Our studies investigated the expression of SHMT2 in kidney cancer by Oncomine, Human Protein Atlas database and ULCAN database. Meanwhile, we found its co-expression gene by cBioPortal online tool and validated their relationship in A498 and ACHN cells by cell transfection, western blot and qRT-PCR. Besides these, we also explored their prognostic values via the Kaplan–Meier plotter database in different types of kidney cancer patients. Results SHMT2 was found to be increased in 7 kidney cancer datasets, compared to normal renal tissues. For the cancer stages, ages and races, there existed significant difference in the expression of SHMT2 among different groups by mining of the UALCAN database. High SHMT2 expression is associated with poor overall survival in patients with kidney cancer. Among all co-expressed genes, NDUFA4L2 and SHMT2 had a high co-expression efficient. SHMT2 overexpression led to the increased expression of NDUFA4L2 at both mRNA and protein levels. Like SHMT2, overexpressed NDUFA4L2 also was associated with worse overall survival in patients with kidney cancer. Conclusion Based on above results, overexpressed SHMT2 and its co-expressed gene NDUFA4L2 were all correlated with the prognosis in kidney cancer. The present study might be benefit for better understanding the clinical significance of SHMT2 and provided a potential therapeutic target for kidney cancer in future.
- Published
- 2020
- Full Text
- View/download PDF
31. Blocking the Metabolic Switch Toward Cytosolic 1C Flux: A Novel Therapeutic Approach for Tumors With Low SLC19A1 Expression
- Author
-
Zhe Chen, Hong Zhou, Haoliang Hu, and Linxi Chen
- Subjects
One-carbon units ,SHMT1 ,SHMT2 ,RPMI ,TSH ,MTHFD1 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 ,Pathology ,RB1-214 - Published
- 2022
- Full Text
- View/download PDF
32. Mild neurodevelopmental disorder due to reduced SHMT2 enzymatic activity caused by novel compound heterozygous variants: expanding the phenotypic spectrum.
- Author
-
Pan H, He M, Luo X, Hu J, Mao X, Cheng Y, and Liu Z
- Subjects
- Humans, Adolescent, Intellectual Disability genetics, Male, Female, Phenotype, Neurodevelopmental Disorders genetics, Heterozygote, Mutation, Missense, Glycine Hydroxymethyltransferase genetics
- Abstract
Biallelic variants in SHMT2 cause neurodevelopmental disorders with cardiomyopathy, spasticity, and brain abnormalities (NEDCASB; OMIM: 619121). This recently described metabolic disorder are characterized by severe intellectual disability, microcephaly, spastic paraplegia, peripheral neuropathy, corpus callosum dysgenesis, facial and limb deformities, and progressive hypertrophic cardiomyopathy. Herein we describe the clinical characteristics of a 13 years old patient with novel compound heterozygous SHMT2 missense variants (c.1274G>A: p.R425Q and c.1042C>T: p.R348W), presenting with mild intellectual disability, corpus callosum dysgenesis, and speech delay. Different from previous cases, our patient represents the mildest phenotype reported to date, and expand the phenotypic spectrum of disease associated with SHMT2 variants., Competing Interests: Declarations. Ethics approval and consent to participate: This study was conducted in accordance with the Declaration of Helsinki and received approval from the Institutional Review Board (IRB) of Hunan Provincial Maternal and Child Health Hospital. Written informed consent was obtained from the legal guardians of all participants involved. This consent encompasses all aspects of participation and data usage. To protect the privacy of both patients and control subjects, all personal identifiers have been removed from the research data. Consent for publication: We hereby clarify that written informed consent has been obtained from all participants involved in the study regarding the publication of their personal or clinical details, as well as any identifying images. For minor patients, written informed consent has been obtained from their parents. This consent covers the use of these details and images in this study and in any potential publications in scientific journals. Competing interests: The authors declare no competing interests., (© 2024. The Author(s), under exclusive licence to Springer-Verlag GmbH Germany, part of Springer Nature.)
- Published
- 2024
- Full Text
- View/download PDF
33. Knockdown of SHMT2 enhances the sensitivity of gastric cancer cells to radiotherapy through the Wnt/β-catenin pathway.
- Author
-
Yu Mao and Tiyong Zhang
- Abstract
Gastric cancer (GC) is one of the most common malignant tumors. The mechanism of GC radioresistance and new radiosensitizers must be revealed and developed to treat GC. Serine hydroxymethyltransferase 2 (SHMT2) is responsible for encoding the mitochondrial form of the pyridoxal phosphate-dependent enzyme. SHMT2 plays a critical role in several types of cancers, while its possible effect on the radiological resistance in GC is still unclear. In this study, we investigated the role of SHMT2 in the radiological resistance of GC. Our data confirmed that SHMT2 was highly expressed in radiation-resistant GC cells. SHMT2 reduced the radiosensitivity of GC cells. In addition, SHMT2 is involved in radiation-induced GC cell apoptosis. Further, SHMT2 regulated the Wnt/β-catenin pathway, therefore reducing the radiosensitivity of GC cells in vivo. In conclusion, we revealed that depletion of SHMT2 enhanced the sensitivity of GC cells to interventional radiotherapy through the Wnt/β-catenin pathway. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
34. Serine hydroxymethyltransferase 2: a novel target for human cancer therapy.
- Author
-
Xie, Min and Pei, Dong-Sheng
- Subjects
DISEASE progression ,CARCINOGENESIS ,CELL cycle ,TRANSFERASES ,SERINE ,CELL proliferation ,GENES ,TUMORS ,OXIDATION-reduction reaction - Abstract
Summary: Serine and glycine are the primary sources of one-carbon units that are vital for cell proliferation. Their abnormal metabolism is known to be associated with cancer progression. As the key enzyme of serine metabolism, Serine Hydroxymethyltransferase 2 (SHMT2) has been a research hotspot in recent years. SHMT2 is a PLP-dependent tetrameric enzyme that catalyzes the reversible transition from serine to glycine, thus promoting the production of one-carbon units that are indispensable for cell growth and regulation of the redox and epigenetic states of cells. Under a hypoxic environment, SHMT2 can be upregulated and could promote the generation of nicotinamide adenine dinucleotide phosphate (NADPH) and glutathione for maintaining the redox balance. Accumulating evidence confirmed that SHMT2 facilitates cell proliferation and tumor growth and is tightly associated with poor prognosis. In this review, we present insights into the function and research development of SHMT2 and summarize the possible molecular mechanisms of SHMT2 in promoting tumor growth, in the hope that it could provide clues to more effective clinical treatment of cancer. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
35. Reduced Shmt2 Expression Impairs Mitochondrial Folate Accumulation and Respiration, and Leads to Uracil Accumulation in Mouse Mitochondrial DNA.
- Author
-
Fiddler, Joanna L, Xiu, Yuwen, Blum, Jamie E, Lamarre, Simon G, Phinney, Whitney N, Stabler, Sally P, Brosnan, Margaret E, Brosnan, John T, Thalacker-Mercer, Anna E, and Field, Martha S
- Subjects
- *
RESEARCH , *FOLIC acid deficiency , *FIBROBLASTS , *DNA , *HETEROCYCLIC compounds , *ANIMAL experimentation , *RESEARCH methodology , *MEDICAL cooperation , *EVALUATION research , *MITOCHONDRIA , *COMPARATIVE studies , *FOLIC acid , *RESPIRATION , *MICE - Abstract
Background: Adequate cellular thymidylate (dTMP) pools are essential for preservation of nuclear and mitochondrial genome stability. Previous studies have indicated that disruption in nuclear dTMP synthesis leads to increased uracil misincorporation into DNA, affecting genome stability. To date, the effects of impaired mitochondrial dTMP synthesis in nontransformed tissues have been understudied.Objectives: This study aimed to determine the effects of decreased serine hydroxymethyltransferase 2 (Shmt2) expression and dietary folate deficiency on mitochondrial DNA (mtDNA) integrity and mitochondrial function in mouse tissues.Methods: Liver mtDNA content, and uracil content in liver mtDNA, were measured in Shmt2+/- and Shmt2+/+ mice weaned onto either a folate-sufficient control diet (2 mg/kg folic acid; C) or a modified diet lacking folic acid (0 mg/kg folic acid) for 7 wk. Shmt2+/- and Shmt2+/+ mouse embryonic fibroblast (MEF) cells were cultured in defined culture medium containing either 0 or 25 nM folate (6S-5-formyl-tetrahydrofolate, folinate) to assess proliferative capacity and mitochondrial function. Chi-square tests, linear mixed models, and 2-factor ANOVA with Tukey post hoc analyses were used to analyze data.Results: Shmt2 +/- mice exhibited a 48%-67% reduction in SHMT2 protein concentrations in tissues. Interestingly, Shmt2+/- mice consuming the folate-sufficient C diet exhibited a 25% reduction in total folate in liver mitochondria. There was also a >20-fold increase in uracil in liver mtDNA in Shmt2+/- mice consuming the C diet, and dietary folate deficiency also increased uracil content in mouse liver mtDNA from both Shmt2+/+ and Shmt2+/- mice. Furthermore, decreased Shmt2 expression in MEF cells reduced cell proliferation, mitochondrial membrane potential, and oxygen consumption rate.Conclusions: This study demonstrates that Shmt2 heterozygosity and dietary folate deficiency impair mitochondrial dTMP synthesis in mice, as evidenced by the increased uracil in mtDNA. In addition, Shmt2 heterozygosity impairs mitochondrial function in MEF cells. These findings suggest that elevated uracil in mtDNA may impair mitochondrial function. [ABSTRACT FROM AUTHOR]- Published
- 2021
- Full Text
- View/download PDF
36. Uncovering SIRT3 and SHMT2-dependent pathways as novel targets for apigenin in modulating colorectal cancer: In vitro and in vivo studies.
- Author
-
Abdelmaksoud, Nourhan M., Abulsoud, Ahmed I., Abdelghany, Tamer M., Elshaer, Shereen Saeid, Rizk, Sherine Maher, Senousy, Mahmoud A., and Maurice, Nadine W.
- Subjects
- *
APIGENIN , *COLORECTAL cancer , *METABOLIC reprogramming , *APOPTOSIS , *LINCRNA , *IN vivo studies , *ADENOMATOUS polyps - Abstract
Despite significant advances in the treatment of colorectal cancer (CRC), identification of novel targets and treatment options are imperative for improving its prognosis and survival rates. The mitochondrial SIRT3 and SHMT2 have key roles in metabolic reprogramming and cell proliferation. This study investigated the potential use of the natural product apigenin in CRC treatment employing both in vivo and in vitro models and explored the role of SIRT3 and SHMT2 in apigenin-induced CRC apoptosis. The role of SHMT2 in CRC patients' survival was verified using TCGA database. In vivo , apigenin treatment restored the normal colon appearance. On the molecular level, apigenin augmented the immunohistochemical expression of cleaved caspase-3 and attenuated SIRT3 and SHMT2 mRNA expression CRC patients with decreased SHMT2 expression had improved overall and disease-free survival rates. In vitro , apigenin reduced the cell viability in a time-dependent manner, induced G0/G1 cell cycle arrest, and increased the apoptotic cell population compared to the untreated control. Mechanistically, apigenin treatment mitigated the expression of SHMT2, SIRT3, and its upstream long intergenic noncoding RNA LINC01234 in CRC cells. Conclusively, apigenin induces caspase-3-dependent apoptosis in CRC through modulation of SIRT3-triggered mitochondrial pathway suggesting it as a promising therapeutic agent to improve patient outcomes. [Display omitted] [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
37. Overexpression of SHMT2 Predicts a Poor Prognosis and Promotes Tumor Cell Growth in Bladder Cancer
- Author
-
Peng Zhang and Qian Yang
- Subjects
SHMT2 ,bladder cancer ,prognosis ,cell cycle ,apoptosis ,Genetics ,QH426-470 - Abstract
SHMT2 was overexpressed in many tumors, however, the role of SHMT2 in bladder cancer (BLCA) remains unclear. We first analyzed the expression pattern of SHMT2 in BLCA using the TNMplot, Oncomine, the Cancer Genome Atlas (TCGA), and the Gene Expression Omnibus (GEO) databases. Next, the association between SHMT2 expression and overall survival (OS)/disease-free survival (DFS) in BLCA patients were analyzed using TCGA and PrognoScan database. The correlation between SHMT2 expression and clinicopathology was determined using TCGA database. Furthermore, the genes co-expressed with SHMT2 and their underlying molecular function in BLCA were explored based on the Oncomine database, Metascape and gene set enrichment analysis (GSEA). Finally, the effects of SHMT2 on cell proliferation, cell cycle, and apoptosis were assessed using in vitro experiments. As a results, SHMT2 was significantly overexpressed in BLCA tissues and cells compared to normal bladder tissues and cells. A high SHMT2 expression predicts a poor OS of BLCA patients. In addition, SHMT2 expression was higher in patients with a high tumor grade and in those who were older than 60 years. However, the expression of SHMT2 was not correlated with gender, tumor stage, lymph node stage, and distant metastasis stage. Finally, overexpression of SHMT2 promoted BLCA cell proliferation and suppressed apoptosis, the silencing of SHMT2 significantly inhibited BLCA cell proliferation by impairing the cell cycle, and promoting apoptosis. SHMT2 mediates BLCA cells growth by regulating STAT3 signaling. In summary, SHMT2 regulates the proliferation, cell cycle and apoptosis of BLCA cells, and may act as a candidate therapeutic target for BLCA.
- Published
- 2021
- Full Text
- View/download PDF
38. Overexpression of SHMT2 Predicts a Poor Prognosis and Promotes Tumor Cell Growth in Bladder Cancer.
- Author
-
Zhang, Peng and Yang, Qian
- Subjects
CANCER cell growth ,INHIBITION of cellular proliferation ,TUMOR growth ,OVERALL survival ,CELL cycle - Abstract
SHMT2 was overexpressed in many tumors, however, the role of SHMT2 in bladder cancer (BLCA) remains unclear. We first analyzed the expression pattern of SHMT2 in BLCA using the TNMplot, Oncomine, the Cancer Genome Atlas (TCGA), and the Gene Expression Omnibus (GEO) databases. Next, the association between SHMT2 expression and overall survival (OS)/disease-free survival (DFS) in BLCA patients were analyzed using TCGA and PrognoScan database. The correlation between SHMT2 expression and clinicopathology was determined using TCGA database. Furthermore, the genes co-expressed with SHMT2 and their underlying molecular function in BLCA were explored based on the Oncomine database, Metascape and gene set enrichment analysis (GSEA). Finally, the effects of SHMT2 on cell proliferation, cell cycle, and apoptosis were assessed using in vitro experiments. As a results, SHMT2 was significantly overexpressed in BLCA tissues and cells compared to normal bladder tissues and cells. A high SHMT2 expression predicts a poor OS of BLCA patients. In addition, SHMT2 expression was higher in patients with a high tumor grade and in those who were older than 60 years. However, the expression of SHMT2 was not correlated with gender, tumor stage, lymph node stage, and distant metastasis stage. Finally, overexpression of SHMT2 promoted BLCA cell proliferation and suppressed apoptosis, the silencing of SHMT2 significantly inhibited BLCA cell proliferation by impairing the cell cycle, and promoting apoptosis. SHMT2 mediates BLCA cells growth by regulating STAT3 signaling. In summary, SHMT2 regulates the proliferation, cell cycle and apoptosis of BLCA cells, and may act as a candidate therapeutic target for BLCA. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
39. A large QTL for fear and anxiety mapped using an F2 cross can be dissected into multiple smaller QTLs.
- Author
-
Parker, CC, Sokoloff, G, Leung, E, Kirkpatrick, SL, and Palmer, AA
- Subjects
Chromosomes ,Mammalian ,Animals ,Mice ,Inbred C57BL ,Mice ,Carrier Proteins ,Chromosome Mapping ,Crosses ,Genetic ,Anxiety ,Fear ,Haplotypes ,Quantitative Trait Loci ,F2 intercrosses ,QTL ,Rnf41 ,Shmt2 ,chromosome substitution strains ,conditioned fear ,congenic mice ,mouse behavioral genetics ,quantitative trait locus mapping ,Ubiquitin-Protein Ligases ,F-2 intercrosses ,Neurology & Neurosurgery ,Biological Sciences ,Medical and Health Sciences ,Psychology and Cognitive Sciences - Abstract
Using chromosome substitution strains (CSS), we previously identified a large quantitative trait locus (QTL) for conditioned fear (CF) on mouse chromosome 10. Here, we used an F2 cross between CSS-10 and C57BL/6J (B6) to localize that QTL to distal chromosome 10. That QTL accounted for all the difference between CSS-10 and B6. We then produced congenic strains to fine-map that interval. We identified two congenic strains that captured some or all the QTL. The larger congenic strain (Line 1: 122.387121-129.068 Mb; build 37) appeared to account for all the difference between CSS-10 and B6. The smaller congenic strain (Line 2: 127.277-129.068 Mb) was intermediate between CSS-10 and B6. We used haplotype mapping followed by quantitative polymerase chain reaction to identify one gene that was differentially expressed in both lines relative to B6 (Rnf41) and one that was differentially expressed between only Line 1 and B6 (Shmt2). These cis-eQTLs may cause the behavioral QTLs; however, further studies are required to validate these candidate genes. More generally, our observation that a large QTL mapped using CSS and F2 crosses can be dissected into multiple smaller QTLs shows a weaknesses of two-stage approaches that seek to use coarse mapping to identify large regions followed by fine-mapping. Indeed, additional dissection of these congenic strains might result in further subdivision of these QTL regions. Despite these limitations, we have successfully fine-mapped two QTLs to small regions and identified putative candidate genes, showing that the congenic approach can be effective for fine-mapping QTLs.
- Published
- 2013
40. Silencing SHMT2 inhibits the progression of tongue squamous cell carcinoma through cell cycle regulation.
- Author
-
Liao, Yan, Wang, Fang, Zhang, Yadong, Cai, Hongshi, Song, Fan, and Hou, Jinsong
- Subjects
CELL cycle regulation ,SQUAMOUS cell carcinoma ,CYCLIN-dependent kinase inhibitors ,CELL cycle ,GENE regulatory networks - Abstract
Background: Serine hydroxymethyltransferase 2 (SHMT2) is a vital metabolic enzyme in one carbon metabolism catalyzing the conversion of serine to glycine, which has been reported to play a crucial role in the progression of tumors. However, its function in tongue squamous cell carcinoma (TSCC) remains unclear. Methods: SHMT2 expression was analyzed using samples in online databases, and was assessed through immunohistochemistry staining of collected clinical specimens. The correlation between SHMT2 expression and the cell cycle was predicted through bioinformatic analysis, including weighted gene co-expression network analysis (WGCNA) and gene set enrichment analysis (GSEA). After transfection with siRNA, CCK8 assay, Edu staining, flow cytometry, trans-well assay, and wound healing experiments were performed to verify the functional role of SHMT2 in vitro. A stable cell line with SHMT2 silencing was established to detect the oncogenic function of SHMT2 in vivo. Results: The expression of SHMT2 was up-regulated in TSCC tissues and cell lines compared with normal groups, and highly expressed SHMT2 significantly indicated a poorer clinical outcome for TSCC patients. Bioinformatic analysis found that high expression of SHMT2 was closely related with biologic process including cell cycle and cell cycle G1/S transition. Down regulating of SHMT2 significantly suppressed the proliferation, invasive and migrative ability of TSCC cells, and induced the prolongation of the G1 phase of the cell cycle in vitro. Furthermore, western blot showed that cell cycle-related regulators such as cyclin-dependent kinase 4 (CDK4) and cyclinD1 expression levels were decreased, while the expression levels of the cyclin-dependent kinase inhibitors p21
Cip1 and p27Kip1 were increased after SHMT2 knockdown. Silencing SHMT2 in the HN6 cell line using short hairpin RNA also impeded tumor growth in vivo. Conclusions: Overexpression of SHMT2 in TSCC indicated low survival rates, and was associated with aggressive behaviors of TSCC. It was also found to be involved in cell cycle regulation of TSCC cells. SHMT2 may serve as a novel prognostic indicator of TSCC. [ABSTRACT FROM AUTHOR]- Published
- 2021
- Full Text
- View/download PDF
41. Increased Expression of SHMT2 Is Associated With Poor Prognosis and Advanced Pathological Grade in Oral Squamous Cell Carcinoma
- Author
-
Zhi-Zhong Wu, Shuo Wang, Qi-Chao Yang, Xiao-Long Wang, Lei-Lei Yang, Bing Liu, and Zhi-Jun Sun
- Subjects
SHMT2 ,oral squamous cell carcinoma ,prognosis ,TCGA ,tumor microenvironment ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
This study focused on the expression of mitochondrial serine hydroxymethyltransferase (SHMT2) in oral squamous cell carcinoma (OSCC) and its correlation with clinical traits and the prognosis of OSCC patients. Immunochemical staining and Western blotting were used to quantify the expression of SHMT2 and related immune markers in OSCC. Using OSCC microarrays and The Cancer Genome Atlas (TCGA) database, we evaluated the association between SHMT2 and various clinical traits. We found that increased expression of SHMT2 was detected in OSCC and correlated with advanced pathological grade and recurrence of OSCC. By a multivariate Cox proportional hazard model, high expression of SHMT2 was shown to indicate a negative prognosis. In addition, in the OSCC microenvironment, increasing the expression of SHMT2 was associated with high expression levels of programmed cell death-ligand 1 (PD-L1), CKLF-like MARVEL transmembrane domain containing 6 (CMTM6), V-type immunoglobulin domain-containing suppressor (VISTA), B7-H4, Slug, and CD317. In the future, more effort will be required to investigate the role of SHMT2 in the OSCC microenvironment.
- Published
- 2020
- Full Text
- View/download PDF
42. Identification of three new compounds that directly target human serine hydroxymethyltransferase 2.
- Author
-
Han, Yanfang, He, Liping, Qi, Yifei, Zhao, Yue, Pan, Yue, Fang, Bohuan, Li, Sha, Zhang, John Z. H., and Zhang, Lujia
- Subjects
- *
THYMIDYLATE synthase , *TETRAHYDROFOLATE dehydrogenase , *SERINE , *PROTEIN expression , *TARGETED drug delivery - Abstract
Mitochondrial serine hydroxymethyltransferase 2 (SHMT2) is an important drug target in the one‐carbon metabolic pathway, since its activity is critical for purine and pyrimidine biosynthesis. Additionally, it plays a prominent role during metabolic reprogramming of cancer cells, and SHMT2 inhibitors have proven useful as anticancer drugs. Compared to drugs targeting one‐carbon metabolic enzymes (mainly dihydrofolate reductase and thymidylate synthase) that have been used for clinical treatment of cancer, efficient SHMT2‐specific inhibitors are lacking. Therefore, we established a direct system for virtual screening, protein expression, and identification of inhibitors targeting SHMT2. First, 27 compounds qualifying as potential SHMT2 inhibitors were selected for biological activity verification through virtual screening of the 210 thousand compounds registered in the Specs database. Second, these 27 hits were subjected to quick screening by an in vitro non‐competitive kinetic assay of SHMT2 single‐enzyme catalysis. This allowed us to identify three compounds featuring medium‐strength and non‐competitive inhibition of SHMT2: AM‐807/42004511 (IC50 = 14.52 ± 4.1665 μM), AM‐807/40675298 (IC50 = 12.74 ± 5.8991 μM), and AM‐807/42004633 (IC50 = 9.43 ± 0.5646 μM). We describe a quick screening method for the identification of inhibitors targeting SHMT2, providing a basis for subsequent identification and screening of new inhibitors. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
43. Impairment of the mitochondrial one-carbon metabolism enzyme SHMT2 causes a novel brain and heart developmental syndrome.
- Author
-
García-Cazorla, Àngels, Verdura, Edgard, Juliá-Palacios, Natalia, Anderson, Eric N., Goicoechea, Leire, Planas-Serra, Laura, Tsogtbaatar, Enkhtuul, Dsouza, Nikita R., Schlüter, Agatha, Urreizti, Roser, Tarnowski, Jessica M., Gavrilova, Ralitza H., SHMT2 Working Group, Oyarzábal, Alfonso, Medina, Inés, Ormazábal, Aida, Muchart, Jordi, Carretero, Juan Manuel, Jou, Cristina, and del Toro, Mireia
- Subjects
- *
ENZYME metabolism , *EYELASHES , *LIPS , *AMINO acid metabolism - Abstract
Keywords: SHMT2; Mitochondrial one-carbon metabolism; Congenital microcephaly; Perisylvian polymicrogyria; Cardiomyopathy EN SHMT2 Mitochondrial one-carbon metabolism Congenital microcephaly Perisylvian polymicrogyria Cardiomyopathy 971 975 5 11/17/20 20201201 NES 201201 The members of SHMT2 Working Group are listed in the Acknowledgements section. SHMT2 performs the first step in a series of reactions that provide one-carbon units covalently bound to folate species in mitochondria: it transfers one-carbon units from serine to tetrahydrofolate (THF), generating glycine and 5,10-methylene-THF [[4], [11]]. Microcephaly associated with hypomyelination is also seen in patients with loss of PYCR2, an enzyme of proline synthesis which interacts with SHMT2, causing hyperglycinemia, underscoring the impact of dysregulated glycine/serine levels on neurodevelopment [[6]]. [Extracted from the article]
- Published
- 2020
- Full Text
- View/download PDF
44. Increased Expression of SHMT2 Is Associated With Poor Prognosis and Advanced Pathological Grade in Oral Squamous Cell Carcinoma.
- Author
-
Wu, Zhi-Zhong, Wang, Shuo, Yang, Qi-Chao, Wang, Xiao-Long, Yang, Lei-Lei, Liu, Bing, and Sun, Zhi-Jun
- Subjects
SQUAMOUS cell carcinoma ,PROGRAMMED death-ligand 1 ,PROPORTIONAL hazards models ,BIOMARKERS ,PROGNOSIS - Abstract
This study focused on the expression of mitochondrial serine hydroxymethyltransferase (SHMT2) in oral squamous cell carcinoma (OSCC) and its correlation with clinical traits and the prognosis of OSCC patients. Immunochemical staining and Western blotting were used to quantify the expression of SHMT2 and related immune markers in OSCC. Using OSCC microarrays and The Cancer Genome Atlas (TCGA) database, we evaluated the association between SHMT2 and various clinical traits. We found that increased expression of SHMT2 was detected in OSCC and correlated with advanced pathological grade and recurrence of OSCC. By a multivariate Cox proportional hazard model, high expression of SHMT2 was shown to indicate a negative prognosis. In addition, in the OSCC microenvironment, increasing the expression of SHMT2 was associated with high expression levels of programmed cell death-ligand 1 (PD-L1), CKLF-like MARVEL transmembrane domain containing 6 (CMTM6), V-type immunoglobulin domain-containing suppressor (VISTA), B7-H4, Slug, and CD317. In the future, more effort will be required to investigate the role of SHMT2 in the OSCC microenvironment. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
45. Proteomic profiling of breast cancer metabolism identifies SHMT2 and ASCT2 as prognostic factors
- Author
-
Stephan Bernhardt, Michaela Bayerlová, Martina Vetter, Astrid Wachter, Devina Mitra, Volker Hanf, Tilmann Lantzsch, Christoph Uleer, Susanne Peschel, Jutta John, Jörg Buchmann, Edith Weigert, Karl-Friedrich Bürrig, Christoph Thomssen, Ulrike Korf, Tim Beissbarth, Stefan Wiemann, and Eva Johanna Kantelhardt
- Subjects
Protein arrays ,Breast cancer ,Cancer metabolism ,SHMT2 ,SLC1A5 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Breast cancer tumors are known to be highly heterogeneous and differences in their metabolic phenotypes, especially at protein level, are less well-understood. Profiling of metabolism-related proteins harbors the potential to establish new patient stratification regimes and biomarkers promoting individualized therapy. In our study, we aimed to examine the relationship between metabolism-associated protein expression profiles and clinicopathological characteristics in a large cohort of breast cancer patients. Methods Breast cancer specimens from 801 consecutive patients, diagnosed between 2009 and 2011, were investigated using reverse phase protein arrays (RPPA). Patients were treated in accordance with national guidelines in five certified German breast centers. To obtain quantitative expression data, 37 antibodies detecting proteins relevant to cancer metabolism, were applied. Hierarchical cluster analysis and individual target characterization were performed. Clustering results and individual protein expression patterns were associated with clinical data. The Kaplan-Meier method was used to estimate survival functions. Univariate and multivariate Cox regression models were applied to assess the impact of protein expression and other clinicopathological features on survival. Results We identified three metabolic clusters of breast cancer, which do not reflect the receptor-defined subtypes, but are significantly correlated with overall survival (OS, p ≤ 0.03) and recurrence-free survival (RFS, p ≤ 0.01). Furthermore, univariate and multivariate analysis of individual protein expression profiles demonstrated the central role of serine hydroxymethyltransferase 2 (SHMT2) and amino acid transporter ASCT2 (SLC1A5) as independent prognostic factors in breast cancer patients. High SHMT2 protein expression was significantly correlated with poor OS (hazard ratio (HR) = 1.53, 95% confidence interval (CI) = 1.10–2.12, p ≤ 0.01) and RFS (HR = 1.54, 95% CI = 1.16–2.04, p ≤ 0.01). High protein expression of ASCT2 was significantly correlated with poor RFS (HR = 1.31, 95% CI = 1.01–1.71, p ≤ 0.05). Conclusions Our data confirm the heterogeneity of breast tumors at a functional proteomic level and dissects the relationship between metabolism-related proteins, pathological features and patient survival. These observations highlight the importance of SHMT2 and ASCT2 as valuable individual prognostic markers and potential targets for personalized breast cancer therapy. Trial registration ClinicalTrials.gov, NCT01592825 . Registered on 3 May 2012.
- Published
- 2017
- Full Text
- View/download PDF
46. MiR-383-5p inhibits the proliferation and migration of lung adenocarcinoma cells by targeting SHMT2.
- Author
-
Bi X, Wang L, Li H, Ma Y, Guo R, Yue J, Kong L, Gong X, Jiao F, Chinn E, and Hu J
- Abstract
Purpose: To explore the effects of miR-383-5p and serine hydroxymethyltransferase 2 (SHMT2) on the proliferation and migration of lung adenocarcinoma cells. Methods: SHMT2 expression in lung adenocarcinoma and normal tissues was investigated using The Cancer Genome Atlas database. Immunohistochemical analysis was performed to confirm SHMT2 expression in lung adenocarcinoma and adjacent normal lung tissues. Bioinformatics analysis and luciferase reporter assays were used to analyze the relationship between miR-383-5p and SHMT2 expression. The protein expression levels of SHMT2, vimentin, N-cadherin, E-cadherin, Bcl-2, and cyclinD1 were analyzed using western blotting. The reverse transcription-quantitative polymerase chain reaction was used to detect SHMT2 knockdown efficiency, miR-383-5p overexpression, and inhibition efficiency. The proliferative ability of cells was detected using the Cell Counting Kit-8 assay. The Transwell assay was used to detect the migration ability of cells. Results: SHMT2 expression was significantly increased in patients with lung adenocarcinoma compared to that in control patients; the higher the SHMT2 expression the worse the outcomes were in patients with lung adenocarcinoma. SHMT2 knockdown inhibited the proliferation, migration, and epithelial-mesenchymal transition of lung adenocarcinoma A549 and H1299 cells. MiR-383-5p directly targeted and downregulated SHMT2 in A549 and H1299 cells. The effects of miRNA-383-5p on the proliferation and migration of these cells differed from those of SHMT2. Exogenous overexpression of SHMT2 reversed the miR-383-5p-induced proliferation and migration inhibition in A549 and H1299 cells. Conclusion: MiR-383-5p inhibits the proliferation and migration of lung adenocarcinoma cells by targeting and downregulating SHMT2., Competing Interests: Competing Interests: The authors have declared that no competing interest exists., (© The author(s).)
- Published
- 2024
- Full Text
- View/download PDF
47. High expression of SHMT2 is correlated with tumor progression and predicts poor prognosis in gastrointestinal tumors.
- Author
-
LIU, Y., YIN, C., DENG, M.-M., WANG, Q., HE, X.-Q., LI, M.-T., LI, C.-P., and WU, H.
- Abstract
OBJECTIVE: Gastrointestinal tumors are malignant tumors with high morbidity. Mitochondrial serine hydroxymethyltransferase 2 (SHMT2) is a key enzyme in the synthesis of serine and glycine, which has prognostic and therapeutic value for many malignant tumors. However, the role of SHMT2 in gastric cancer (GC), esophageal cancer (ESCC), and colorectal cancer (CC) has not been clarified. PATIENTS AND METHODS: The expression of SHMT2 was detected in GC, ESCC, and CC by immunohistochemistry and reverse real time transcription-polymerase chain reaction. The relationships between SHMT2 expression and clinicopathologic characteristics, recurrence-free survival (RFS), and disease-specific survival (DSS) were analyzed by the survival analysis and correlation analysis. RESULTS: The positive expression rate of SHMT2 in GC, ESCC, and CC was 74.1%, 69.2%, and 71.7%, respectively. Patients with high expression of SHMT2 had a worse prognosis. In GC, high SHMT2 expression had positive correlation with lymph node metastasis (p=0.005) and histological grade (p=0.002). In ESCC, high SHMT2 expression had positive correlation with pT classification (p=0.033) and pM classification (p=0.029). In CC, high SHMT2 expression had positive correlation with tumor size (p=0.004), lymph node metastasis (p=0.035), TNM stage (p=0.007), and histological grade (p=0.020). Notably, SHMT2 expression was an independent prognostic factor for RFS and DSS in GC, ESCC, and CC (p<0.05). CONCLUSIONS: SHMT2 is upregulated in GC, ESCC, and CC. The high expression of SHMT2 is correlated with gastrointestinal tumors progression, and poor prognosis, which is a potential new target for the diagnosis and treatment of gastrointestinal tumors. [ABSTRACT FROM AUTHOR]
- Published
- 2019
48. HDAC11 regulates type I interferon signaling through defatty-acylation of SHMT2.
- Author
-
Ji Cao, Lei Sun, Aramsangtienchai, Pornpun, Spiegelman, Nicole A., Xiaoyu Zhang, Weishan Huang, Seto, Edward, and Lin, Hening
- Subjects
- *
DEACETYLASES , *INTERFERONS , *ACYLATION kinetics , *IMMUNE response , *CELL proliferation - Abstract
The smallest histone deacetylase (HDAC) and the only class IV HDAC member, HDAC11, is reported to regulate immune activation and tumorigenesis, yet its biochemical function is largely unknown. Here we identify HDAC11 as an efficient lysine defattyacylase that is >10,000-fold more efficient than its deacetylase activity. Through proteomics studies, we hypothesized and later biochemically validated SHMT2 as a defatty-acylation substrate of HDAC11. HDAC11-catalyzed defatty-acylation did not affect the enzymatic activity of SHMT2. Instead, it affects the ability of SHMT2 to regulate type I IFN receptor ubiquitination and cell surface level. Correspondingly, HDAC11 depletion increased type I IFN signaling in both cell culture and mice. This study not only demonstrates that HDAC11 has an activity that is much more efficient than the corresponding deacetylase activity, but also expands the physiological functions of HDAC11 and protein lysine fatty acylation, which opens up opportunities to develop HDAC11-specific inhibitors as therapeutics to modulate immune responses. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
49. BRCA1-A and BRISC: Multifunctional Molecular Machines for Ubiquitin Signaling
- Author
-
Julius Rabl
- Subjects
deubiquitination ,BRCA1 ,DNA repair ,immune regulation ,BRCC36 ,SHMT2 ,Microbiology ,QR1-502 - Abstract
The K63-linkage specific deubiquitinase BRCC36 forms the core of two multi-subunit deubiquitination complexes: BRCA1-A and BRISC. BRCA1-A is recruited to DNA repair foci, edits ubiquitin signals on chromatin, and sequesters BRCA1 away from the site of damage, suppressing homologous recombination by limiting resection. BRISC forms a complex with metabolic enzyme SHMT2 and regulates the immune response, mitosis, and hematopoiesis. Almost two decades of research have revealed how BRCA1-A and BRISC use the same core of subunits to perform very distinct biological tasks.
- Published
- 2020
- Full Text
- View/download PDF
50. Curcumin in Health and Disease.
- Author
-
Bachmeier, Beatrice
- Subjects
Akt/mTOR signaling ,Alzheimer's disease ,Crohn's disease ,Curcuma longa ,Helicobacter pylori ,IL-17 ,ImageJ ,SHMT2 ,STAT3 ,TLC bioautography ,TLC-MS ,Zingiberaceae ,ageing ,amino-acids ,amyloid ,amyloidosis ,anti-cancer ,anti-inflamation ,anti-tumor ,anticancer ,antimicrobial agents ,antioxidant activity ,antioxidants ,apoptosis ,autophagy ,brain ischemia ,cancer ,cancer treatment ,cell cycling ,cellular pathway ,centrifugal partition chromatography ,chaperone-mediated autophagy ,chitosan ,complementary medicine ,curcumin ,death receptor ,delivery system ,diet ,direct protein binding ,drug discovery ,gastric cancer ,gastric ulcer ,gastroprotection ,genes ,glioblastoma multiforme ,hydrostatic counter-current chromatography ,inflamm-aging ,inflammatory bowel disease ,macronutrients ,mechanism of action ,metabolic reprogramming ,microbiota ,micronutrients ,minerals ,mitophagy ,n/a ,nanoparticles ,necrotizing enterocolitis ,neurodegeneration ,nutrition ,oxidative metabolites ,protein aggregation ,protein misfolding ,reflux esophagitis ,renal cell cancer ,senescence ,senolytics ,silica ,structure activity relationship ,supportive care ,tau protein ,transmission electron microscopy ,transthyretin ,tumor growth ,tumor proliferation ,turmeric tuber ,ulcerative colitis ,vitamins ,wound ,wound healing - Abstract
Summary: The plant-derived polyphenol curcumin has been used in promoting health and combating disease for thousands of years. Its therapeutic effects have been successfully utilized in Ayurvedic and Traditional Chinese Medicine in order to treat inflammatory diseases. Current results from modern biomolecular research reveal the modulatory effects of curcumin on a variety of signal transduction pathways associated with inflammation and cancer. In this context, curcumin's antioxidant, anti-inflammatory, anti-tumorigenic, and even anti-metastatic activities are discussed. On the cellular level, the reduced activity of several transcription factors (such as NFkB or AP-1) and the suppression of inflammatory cytokines, matrix degrading enzymes, metastasis related genes and even microRNAs are reported. On functional levels, these molecular effects translate into reduced proliferative, invasive, and metastatic capacity, as well as induced tumor cell apoptosis. All these effects have been observed not only in vitro but also in animal models. In combination with anti-neoplastic drugs like Taxol, kinase inhibitors, and radiation therapy, curcumin potentiates the drugs' therapeutic power and can protect against undesired side effects. Natural plant-derived compounds like curcumin have one significant advantage: They do not usually cause side effects. This feature qualifies curcumin for primary prevention in healthy persons with a predisposition to cancer, arteriosclerosis, or chronic inflammatory diseases. Nonetheless, curcumin is considered safe, although potential toxic effects stemming from high dosages, long-term intake, and pharmacological interactions with other compounds have yet to be assessed. This Special Issue examines in detail and updates current research on the molecular targets, protective effects, and modes of action of natural plant-derived compounds and their roles in the prevention and treatment of human diseases.
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.