Open Access

IDH2: A novel biomarker for environmental exposure in blood circulatory system disorders (Review)

  • Authors:
    • Ya Qi Gong
    • Shuang Wei
    • Yuan Yun Wei
    • Yong Lin Chen
    • Jian Cui
    • Yue Qiu Yu
    • Xiang Lin
    • Hong Xia Yan
    • Hui Qin
    • Lan Yi
  • View Affiliations

  • Published online on: June 24, 2022     https://doi.org/10.3892/ol.2022.13398
  • Article Number: 278
  • Copyright: © Gong et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

As the risk of harmful environmental exposure is increasing, it is important to find suitable targets for the diagnosis and treatment of the diseases caused. Isocitrate dehydrogenase 2 (IDH2) is an enzyme located in the mitochondria; it plays an important role in numerous cell processes, including maintaining redox homeostasis, participating in the tricarboxylic acid cycle and indirectly taking part in the transmission of the oxidative respiratory chain. IDH2 mutations promote progression in acute myeloid leukemia, glioma and other diseases. The present review mainly summarizes the role and mechanism of IDH2 with regard to the biological effects, such as the mitophagy and apoptosis of animal or human cells, caused by environmental pollution such as radiation, heavy metals and other environmental exposure factors. The possible mechanisms of these biological effects are described in terms of IDH2 expression, reduced nicotine adenine dinucleotide phosphate content and reactive oxygen species level, among other variables. The impact of environmental pollution on human health is increasingly attracting attention. IDH2 may therefore become useful as a potential diagnostic and therapeutic target for environmental exposure‑induced diseases.

Introduction

Isocitrate dehydrogenase (IDH) is a key enzyme in the tricarboxylic acid cycle (1) and plays an important role in various metabolic functions and epigenetic cellular processes (2,3). There are three isoenzymes of IDH. The IDH1 and IDH2 genes are located in chromosomes 2q33 and 16q26, respectively (4). At the same time, the IDH3 protein is encoded by the IDH3A, IDH3B and IDH3G genes, which are located in chromosomes 15q25, 20p13 and Xq28 respectively. Structurally, IDH1 and IDH2 proteins are composed of a large domain, a clasp domain and a small domain (5). IDH mutations result in gain-of-function and reduction of α-ketoglutarate (α-KG) to 2-hydroxyglutarate (2-HG) (6). Mutated IDH may lead to the development of cancer, especially acute myeloid leukemia (AML) (7).

The expression of IDH2 is affected by exposure factors such as ionizing radiation. IDH2 plays a crucial role in disorders caused by exposure factors (8,9). Therefore, the present study provides an overview of the effect of IDH2 on circulatory system diseases and other system diseases with regard to exposure factors. IDH2 is a mitochondrial enzyme that catalyzes the oxidative decarboxylation of isocitrate to α-KG (10) and protects the body from oxidative stress by converting nicotinamide adenine dinucleotide phosphate (NADP+) to reduced NADP (NADPH) (11). Multiple internal and external factors may contribute to mutations of the IDH2 gene, which may lead to the loss of enzyme activity or obtaining new enzymatic functions (change-of-function) (12). The mutant IDH2 consumes a-KG and NADPH, and produces 2-HG and NADP+ (12) (Fig. 1). Mutations in the IDH2 potein often occur in arginine residues R140 and R172, and are closely associated with the development and progression of AML and glioma (13). Moreover, one study has revealed the effect of wild-type IDH2 on the molecular mechanism of Epstein Barr virus-mediated disease. EBV can cause nasopharyngeal carcinoma in humans, and wild-type IDH2 promotes the survival of nasopharyngeal carcinoma cells (14). Details on the different functions of IDH2 are expounded in the following sections.

IDH2 mutations that result in 2-HG accumulation promote the development of hematopoietic diseases

Over past decades, studies have shown that IDH2 is associated with hematological tumorigenesis or disorders, including AML, myelodysplastic syndrome (MDS) and chronic myelomonocytic leukemia (CMML). The frequencies of IDH2 mutations in diseases of the hematopoietic system, such as AML (1517), MDS (18,19) and chronic mononuclear leukemia, are 8.0–19, 4–4.6 and 7.8–8.8%, respectively (18,20). Although the frequency of diseases with an IDH2 gene mutation is low, it is crucial to understand the association between IDH2 mutations and disease occurrence. Therefore, the following sections will review the role of IDH2 in various physiological or pathological processes.

Obstructed hematopoietic differentiation caused by IDH2 gene mutations triggers development of hematopoietic neoplasms

IDH2 gene mutations contribute to hematopoietic neoplasms, such as AML and CMML (21,22). AML is a hazardous, enervating and invasive disease with a poor prognosis (23,24). IDH2 and PHD finger protein 6 mutations exert synergistic effects on leukemia formation through excessive production of 2-HG and damage to DNA repair (25). The mutated IDH2 enzyme exhibits gain-of-function activity and catalyzes the conversion of α-KG to 2-HG, which is involved in the pathogenesis of AML (26,27). The 2-HG metabolite is oncogenic and closely associated with the hypermethylation of DNA and histones, which alters the expression of different mRNAs and hematopoietic cell differentiation (28,29). IDH2 gene mutations are crucial for the maintenance of AML progenitor cells, but these mutations may not be the key preliminary step for AML development (30,31). Therefore, further studies are needed to confirm the role of IDH2 mutations in the occurrence of AML. IDH2-mutated cells have significantly increased sensitivity to IL-1β signaling, which may be a potential therapeutic target (32). Furthermore, there are drugs in various stages of clinical development for the treatment of AML with mutated IDH2 proteins, such as AG-221 (enasidenib) (33), AGI-6780 (34), CP-17 (34), TQ05310 (35) and AG-881 (36). AG-221 is an oral, valid, selective inhibitor of mutated IDH2 that has been approved by the US Food and Drug Administration for the treatment of relapsed or refractory leukemia with IDH2 gene mutations. AG-221 binds to the IDH2 dimer interface and blocks the production and accumulation of 2-HG, thereby allowing hematopoietic cells to differentiate from terminal or ancestral mutant clones (37,38). For patients who are unable to benefit from treatment with AG-221 alone, the addition of all-trans-retinoic acid may enhance the response rate of AG-221 therapy (39). Additionally, AG-221 in combination with azacitidine is more effective than azacitidine alone, and is a tolerated and effective treatment option for relapsed or refractory AML that promotes cell differentiation and overall response rates (33,40). There are usually no clinical signs of concurrent infection or clinical features of IDH inhibitor-associated differentiation syndrome, which is characterized by dyspnea, hypoxia, fluid retention and weight gain (41). AG-221 was found to exhibit a poor inhibitory effect on the IDH2R140Q mutation. However, AGI-6780, a preclinical inhibitor, is a selective inhibitor of the IDH2R140Q protein and may be an effective targeted drug therapy for this mutation (34,42,43). In addition, SH1573 is a potential inhibitor of IDH2R140Q, and has been demonstrated to be novel, safe and effective, and is currently in clinical trials (44). Furthermore, CP-17 acts as a potent inhibitor of the IDH2R140Q mutant and may be a lead compound for developing drugs against AML (34). Furthermore, TQ05310 showed selective specificity that targeted both IDH2R140Q and IDH2R172K mutant enzymes but had no effect on wild-type IDH or mutated IDH1 (35). AG-881 has lower specificity than the aforementioned drugs and may be useful as an inhibitor of IDH1 and IDH2 mutations for treating associated diseases (37). It is important to note that prognosis can be judged according to the IDH2 mutation site and whether other genes are mutated (2). Moreover, it has been reported that an IDH2 mutation is a marker for poor prognosis in patients with AML, and the overall survival (OS) time of the patients with wild-type IDH2 was greater than that of patients with an IDH2 mutation (45). However, another study found that patients with IDH2R172K mutations have improved relapse-free survival (RFS) and OS times compared with patients with the wild-type (46). Therefore, this topic remains controversial, and more experimental data are needed to confirm which view is correct. CMML is a myelodysplastic/myeloproliferative neoplasm. IDH2 can be used as an indicator of poor prognosis in CMML patients to a lesser extent. One study indicated that patients with IDH2 mutations in CMML had inferior OS rates than wild-type IDH2 patients (17,47).

IDH2 mutations promote the progression of other diseases in the hematopoietic system

IDH2 status not only plays an important role in hematopoietic neoplasms, but also regulates the development of other diseases in the hematopoietic system. Previous studies have discovered that a small number of patients with MDS have IDH2 mutations. The most common IDH2 mutant subtype in MDS is IDH2R140Q, and as aforementioned, this mutation results in enzymatic gain-of-function and inhibition of hemopoietic cell differentiation (48,49). The IDH2 mutation rate in patients with advanced MDS is higher than in those with early MDS (14). Mutations in IDH1 and IDH2 are mutually exclusive (50). Similarly, analysis of patients with MDS showed IDH2 and tet methylcytosine dioxygenase 2 (TET2) gene mutations do not exist simultaneously (51). IDH2 mutations may be involved in the development but not the progression of MDS (52). Moreover, high 2-HG serum levels predict the presence of IDH2 mutations; thus, mutated IDH2 proteins can be used as targets for pre-transplantation and post-transplantation treatment of MDS (53,54). For effective treatment of MDS with IDH2 gene mutations, the mutated protein can be specifically targeted (19). For example, AG-221 is an effective drug for the treatment of MDS with IDH2 mutations, including use in those patients for whom the use of hypomethylating agents has been unsatisfactory (55). There is a poor OS time among patients with MDS and IDH2 mutations under low-risk stratification in the International Prognostic Scoring System (53,56). In summary, IDH2 plays an extremely important role in the generation and progression of some diseases of the hematopoietic system, and can be used as a molecular marker and target for therapy.

Associations between IDH2 gene status and other pathological processes

IDH2 is closely associated with therapeutic effectiveness and sensitivity. IDH2 is not only associated with blood circulation disorders, but also with other diseases, such as glioma (57), non-small cell lung cancer (58), solid papillary carcinoma with reverse polarity (SPCRP) (59), angioimmunoblastic T-cell lymphoma (AITL) (60), high-grade chondrosarcoma (61) and undifferentiated sinus carcinoma (62).

IDH2 gene mutations participate in nervous system diseases, especially in gliomas

IDH2 mutations play an important role in some diseases of the nervous system, such as gliomas and 2-hydroxyglutaric aciduria. Studies have shown that IDH2 mutations are a driving factor in gliomas, especially in low-grade gliomas and glioblastomas (57,63). Glioma subtypes with IDH2 gene mutations have unique clinical characteristics. For instance, IDH2 mutations are more common in younger individuals and are more likely to occur in low-risk surgical areas (64). Furthermore, IDH2 can be used as a biomarker with diagnostic, prognostic and predictive implications (65,66). In gliomas, IDH2 mutations were found to be mutually exclusive with IDH1, phosphatase and tensin homolog, cellular tumor antigen p53 and α-thalassemia retardation syndrome X-linked mutations (63). Injecting mutated IDH2 into glioma mice accelerated tumor growth and increased mortality rate compared with that in mice with wild-type IDH2 (67). Moreover, IDH2 mutations are significantly correlated with the incidence of preoperative glioma-related epilepsy, and affect the surgical resectability of gliomas (68,69). IDH2 mutations may also trigger the development of 2-hydroxyglutaric aciduria and Parkinson's disease (70,71).

Tumor cell growth is promoted by wild-type IDH2

In addition to playing an important role in the progression of neurological diseases, IDH2 gene status promotes the development of certain respiratory diseases. Studies have shown that the IDH2 protein plays a role in specific lung cancer types; for example, IDH2 plays an important role in non-small cell lung cancer (58). The expression level of IDH2 in the serum of patients with non-small cell lung cancer is higher than that of the normal population, and after treatment, the expression of serum IDH2 in patients with non-small cell lung cancer decreases (58). Notably, the IDH2 rs11540478 genetic variant represents a novel susceptibility locus for lung cancer; it can change the level of IDH2 protein, and affect cancer cell viability and disease evolution (72). Compared with patients with lung cancer, healthy individuals have lower levels of IDH2 mRNA in peripheral blood lymphocytes (66). Wild-type IDH2 promotes the development of lung cancer, and an increased level of IDH2 protein is a characteristic of poor survival (73). Overall, IDH2 may represent a target for lung cancer treatment. Furthermore, IDH2 is important for the progression of lung injury. IDH2 has different effects depending on the cause of the lung injury. IDH2 can reduce acrolein-induced lung injury by providing NADPH (74), and as a provider of the pro-inflammatory metabolite α-KG, IDH2 is indirectly involved in lipopolysaccharide-induced acute lung injury. Therefore, targeting the IDH2 enzyme may be a treatment strategy for systemic inflammatory response syndrome (75).

Status of the IDH2 gene affects peripheral T-cell lymphoma

AITL is a subtype of peripheral T-cell lymphoma that is affected by IDH2 gene status. It has been reported that IDH2 expression is significantly upregulated in patients with myeloproliferative neoplasia (MPN)-AITL compared with that in AITL patients without MPN (76). Meanwhile, it has also been reported that mutated IDH2 and TET2 proteins usually occur together in AITL (60,77). In one study, AITL with the IDH2R172 mutation displayed a limited gene expression profile that correlated with cellular differentiation, and genes associated with interleukin-12 stimulation were significantly enriched (78). Moreover, an AITL case with mutated IDH2 protein presented with high intracellular levels of 2-HG without an increase in circulating 2-HG. This case suggests that levels of circulating 2-HG may not accurately reflect the presence of IDH2 gene mutations (79). In 20–30% of patients with AITL, IDH2 has diagnostic value and is a potential therapeutic target. Next-generation sequencing technology and allele-specific quantitative polymerase chain reaction show good sensitivity for the detection and diagnosis of IDH2 mutations (80). As expected, compared with the TET2 gene mutation alone, TET2/IDH2 co-mutations conferred longer progression-free survival times to affected patients (81).

Course of other diseases is also affected by IDH2 gene status

IDH2 status is important in other diseases, such as SPCRP, prostate and colon cancer. A previous clinical study determined that 77% of SPCRP cases had an IDH2 hot spot mutation at amino acid site 172 (59). One study indicated that the IDH2R172 mutation was highly specific for SPCRP in various subtypes of breast cancer, and that it may be a suitable diagnostic marker for SPCRP and guide effective treatment (82). Furthermore, high expression levels of IDH2 mRNA or protein were associated with a poor outcome in patients with invasive breast cancer (83). Enzymatic malfunction of IDH2 in prostate cancer cells disrupted oxidative bioenergetics, enhanced reactive oxygen species (ROS) generation and increased mitochondrial dynamics (84). Frequent IDH2 gene mutations have recently found in central chondrosarcomas, which resulted in longer RFS and metastasis-free survival times in high-grade chondrosarcomas; however, no association with OS was observed (61). Undifferentiated sinus carcinoma is an infrequent, aggressive, highly malignant tumor with finite therapy choices (85,86), and women with this disease have a higher frequency of IDH2 mutations compared with men with the disease (87). IDH2 mutated undifferentiated sinus carcinoma subtypes have high levels of DNA methylation and a poor prognosis (62). As a prime component in anti-oxidative damage, wild-type IDH2 protects cochlear hair cells from ROS and prevents age-related hearing loss by providing NADPH (88,89). Moreover, IDH2 is vital for conditions that develop from ROS exposure or mechanical damage (90). For example, IDH2 has a protective effect against ultraviolet B radiation-induced skin damage and is involved in skin wound healing (90,91). Downregulation of IDH2 protein is likely one of the mechanisms underlying 5-hydroxymethylcytosine loss in melanoma (92). Studies have shown that IDH2 knockout can inhibit the growth of colon cancer cell lines. Thus, the IDH2 enzyme has an important impact on the generation and progression of colon carcinoma (93,94). IDH2 may be a target to treat skin pigmentation, as IDH2 deficiency can cause skin pigmentation (95). Additionally, IDH2 may be a therapeutic target for adipose inflammation (96,97). In summary, IDH2 gene status drives the process of multiple diseases and may serve as a biomarker and/or therapeutic target (Table I).

Table I.

Associations between status of IDH2 and multiple diseases.

Table I.

Associations between status of IDH2 and multiple diseases.

A, Mutation

DiseaseEffect of pathological progressionPrognosis(Refs.)
AMLMaintain tumor cells and promote disease progressionNegative prognostic marker(30,45)
CMML Unfavorable molecular prognostic factor(17,47)
MDSParticipating in what happens does not promote progressPoor(48,53)
GliomaDriving factorsLonger OS and PFS times(66,67)
AITLPromote disease progressionLonger PFS time(84)
SPCRPDriving factorsPatients with high expression of IDH2 have poor outcome in IBC(59,83)
High-grade chondrosarcoma Longer relapse- and metastasis-free survival times(61)
Undifferentiated sinus carcinoma Higher survival rate(62,87)

B, Upregulated expression of wild-type IDH2

DiseaseEffect of pathological progression Prognosis(Refs.)

Lung cancerContribute to cancer cell growth and survivalPoor(58,73)

[i] IDH2, isocitrate dehydrogenase 2; AML, acute myeloid leukemia; CMML, chronic myelomonocytic leukemia; MDS, myelodysplastic syndrome; AITL, angioimmunoblastic T-cell lymphoma; SPCRP, solid papillary carcinoma with reverse polarity; OS, overall survival; PFS, progression-free survival.

IDH2 level is affected by exposure to environmental hazards

IDH2 protects cells from radiation damage by producing NADPH

IDH2 not only greatly contributes to the progression and treatment of diseases, but it is also involved in the maintenance of cellular metabolism under conditions of environmentally induced damage. IDH2 can protect cells from damage caused by excessive ROS production after cells are exposed to ionizing radiation (98). ROS are natural products of mitochondrial metabolism and can trigger oxidative damage (99). The protective effect of IDH2 on cells is based on the reduction of NADP+ to NADPH, which is a precondition for some cellular defense systems to reduce oxidative damage (100,101). Imbalance between mitochondrial oxidation and reduction reactions causes cell death in organisms exposed to ionizing radiation. When organisms are exposed to ionizing radiation, the level of ROS increases and the NADPH regulated by IDH2 to the antioxidant system decreases (102,103). Wild-type IDH2 protects cells against γ-irradiation by maintaining NADPH levels that buffer against radiation-induced ROS and protect cells from apoptosis. The latest studies and clinical data from the European Organization for Research on Treatment of Cancer trial have verified that gliomas with IDH2 gene mutations are extremely sensitive to radiotherapy (104106). Furthermore, IDH2 not only protects the cells from ionizing radiation, but also protects the cells from non-ionizing radiation. Under the non-ionizing radiation environment, IDH2 has a protective effect on ultraviolet B-induced skin damage (90).

IDH2 activity is regulated by binding to heavy metal ions and toxic substances

Environmental heavy metals affect the enzymatic activity of IDH2. When organisms are exposed to heavy metals, such as cadmium (Cd2+) (107), CoCl2 (108), mercury (109) or copper (8), the expression of IDH2 may be altered. Cd2+ ions have a dual effect on cells. Cd2+ activates IDH2, and activated IDH2 provides NADPH for cellular defense, as aforementioned. However, Cd2+ ions have a high affinity for thiols and contribute to the combination between Cd2+ ions and cysteine residues, which trigger IDH2 inactivation. Therefore, cells exposed to cadmium are prone to apoptosis (107,110). Removing cadmium from the environment with microorganisms is difficult due to its toxicity. The yeast Pichia kudriavzevii has been used as a model organism for cadmium removal, and acid stress can reduce the toxicity of Cd2+ and upregulate genes associated with ATP synthesis, such as IDH2 (111). CoCl2 can indirectly affect IDH2 expression. Hypoxia was induced by CoCl2 treatment increasing the expression of IDH2 in breast cancer cells after radiation (108). Jejunal epithelial cells induce mitochondrial dysfunction and mitosis through the Mitomir-1285-IDH2 axis during copper exposure Mitomir-1285 aggravates copper-induced mitochondrial dysfunction by inhibiting IDH2 expression (8) (Fig. 2). Exposure to arsenic in pregnant rats indirectly induces anxiety-like behavior in adult offspring through downregulation of IDH2 expression in the fetal brain (9). Moreover, some lower animals, such as bivalves and corals, may also be affected by heavy metals, resulting in a change in IDH2 activity. For example, bivalves exposed to mercury alone showed a reduction in IDH2 activity and a subsequent alteration in cellular energy production (109). When corals were exposed to copper, IDH2 activity was inhibited, and aerobic and oxidative metabolism was reduced (112) (Fig. 2) (Table II).

Table II.

Effect of different exposure environments on IDH2.

Table II.

Effect of different exposure environments on IDH2.

Exposure environmentRelated effectModel(Refs.)
Ionizing radiation
  γ-rayIDH2 protects cells from oxidative stressMouse, NIH3T3 cells(100)
IDH2 knockdown triggers radiation-reduced metabolism disorder in mitochondriaEsophageal squamous cell carcinomacell lines(98)
IDH2 expression is negatively correlated with radiation therapy sensitivityMouse(102)
Non-ionizing radiation
  UVBUVB-induced apoptosis and inflammation in the skin of IDH2-deficient miceMouse(90)
Heavy metals and toxic substances
  Cd2+Dual effect: Activates IDH2 by providing two ions, and inactivates IDH2 via high affinity for thiolsEscherichia coli(107,110)
  CoCl2Can enhance IDH2 expressionBreast cell line MCF-7(108)
  ArsenicDownregulation of IDH2 expressionPregnant rats(9)
  MercurySuppresses IDH2 expressionBivalves(109)
  CopperSuppresses IDH2 expressionCoral(112)

[i] IDH2, isocitrate dehydrogenase 2; UVB, ultra violet B; Cd2+, cadmium.

Regulation of IDH2 level is a multi-factor and multi-pathway process

IDH2 is involved in protecting cells from oxidative stress-induced by ROS (113). Deficiency causes abnormal mitochondrial function, which increases the production of ROS, promotes cell senescence by inducing cell-cycle arrest, impairs cell function and stimulates development of age-related diseases (89,114). As IDH2 makes a significant contribution to homeostasis and cellular differentiation, the following sections will discuss the factors and associated mechanisms that regulate IDH2 expression and activity.

Sirtuin3 (SIRT3) expression is positively associated with IDH2 activity

IDH2 is regulated by SIRT3 and other factors. SIRT3 is regulated by acetylation, which is a post-translational modification that alters its activity (115,116). IDH2 activity is increased significantly after deacetylation by SIRT3, which is a deacetylase in mitochondria. A decrease in SIRT3 protein reduces IDH2 enzymatic activity by decreasing IDH2 dimer formation (117,118) (Fig. 2). For example, in one study, during therapy for multiple myeloma, the combination of carfilzomib with SIRT3 inhibitors decreased IDH2 activity and increased multiple myeloma cell death (118). Overexpression of nicotinamide mononucleotide adenylate transferase 3 in bone marrow mesenchymal stem cells enhanced the ability of specific antioxidant stress by enhancing SIRT3 activity and decreasing IDH2 acetylation level (119).

Other moderating factors that regulate levels and activity of IDH2

To avoid damage caused by oxidative stress, the antioxidant activity of IDH2 can be enhanced by SUMOylation (120). A previous study showed that the activity of IDH2 is affected by post-translational modification. Under oxidative stress, IDH2 ubiquitination deficient cells had more apoptosis than normal cells, suggesting that ubiquitination is an important means of regulating IDH2 activity (120). Furthermore, IDH2R140Q and cytoplasmic nucleophosmin mutation increase myeloid ecotropic viral integration site 1 and homeobox A9 gene expression, respectively, which activates the hypoxia pathway in AML cells (121). Cells expressing the mutant IDH2 protein are deficient in their capacity for reductive carboxylation and the ability to produce acetyl-CoA may be impaired under hypoxic conditions. Acetyl-CoA is involved in certain metabolic processes in the body, such as cholesterol synthesis, fatty acid generation and glucose metabolism (122). In addition, decreased IDH2 expression may promote ferroptosis through the coordination of erastin (ferroptosis inhibitor) with the NADPH-glutathione-glutathione peroxidase 4 (GPX4) axis (Fig. 2) (123). Taken together, these factors provide new insight into the treatment of diseases involving the IDH2 gene.

Prospects

The present review summarizes the existing knowledge relating to various aspects of IDH2 expression and activity. IDH2 and its associated diseases, mechanisms of action and alterations after exposure to radiation and heavy metals are described. A number of environmental exposure factors can inhibit IDH2 expression or activity, indirectly leading to decreased GPX4 expression, which may lead to ferroptosis, but the specific mechanism is not clear. Further research may provide therapeutic methods for some diseases so that patients can obtain more accurate and effective treatment plans. In addition, it is possible to find other factors that regulate normal levels of IDH2 and maintain homeostasis in response to radiation exposure, a strategy that may have great application value. Furthermore, IDH2 is promising as a marker of environmental exposure for circulatory diseases. On the basis of the important role of IDH2 in blood circulatory system disorders, it is feasible to recommend IDH2 as a novel biomarker for environmental exposure in blood circulatory system disorders both theoretically and practically.

Acknowledgements

Not applicable.

Funding

This study was supported by the Hunan Natural Science Foundation (grant no. 2019JJ40238), the Key Scientific Research Project of Hunan Health Commission (grant no. 202102051816), the Hunan Health Commission (grant no. C2019096), the Project of Hengyang Science and Technology Bureau (grant no. 2020jh042) and the Hunan Provincial Natural Science Foundation of China (grant no. 2019JJ50509).

Availability of data and materials

Not applicable.

Authors' contributions

YQG and SW completed the writing and proofreading of the manuscript. YYW, YLC, and JC revised the manuscirpt critically for intellectual content and created the figures and tables. YQY, XL, HXY, and HQ made corrections to the original manuscript and also performed literature searches. LY was involved in the conception, directed the writing of the article, and made partial revisions. All authors have read and approved the final manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

2-HG

2-hydroxyglutarate

α-KG

α-ketoglutarate

AITL

angioimmunoblastic T-cell lymphoma

AML

acute myeloid leukemia

CMML

chronic myelomonocytic leukemia

GPX4

glutathione peroxidase 4

IDH

isocitrate dehydrogenase

MDS

myelodysplastic syndrome

MPN

myeloproliferative neoplasia

NADP+

nicotinamide adenine dinucleotide phosphate

NADPH

reduced NADP

OS

overall survival

RFS

relapse-free survival

ROS

reactive oxygen species

SIRT3

sirtuin 3

SPCRP

solid papillary carcinoma with reverse polarity

TET2

tet methylcytosine dioxygenase 2

References

1 

Dang L, Yen K and Attar EC: IDH mutations in cancer and progress toward development of targeted therapeutics. Ann Oncol. 27:599–608. 2016. View Article : Google Scholar : PubMed/NCBI

2 

Montalban-Bravo G and DiNardo CD: The role of IDH mutations in acute myeloid leukemia. Future Oncol. 14:979–993. 2018. View Article : Google Scholar : PubMed/NCBI

3 

Sharma H: Development of novel therapeutics targeting isocitrate dehydrogenase mutations in cancer. Curr Top Med Chem. 18:505–524. 2018. View Article : Google Scholar : PubMed/NCBI

4 

Willander K, Falk IJ, Chaireti R, Paul E, Hermansson M, Gréen H, Lotfi K and Söderkvist P: Mutations in the isocitrate dehydrogenase 2 gene and IDH1 SNP 105C > T have a prognostic value in acute myeloid leukemia. Biomark Res. 2:182014. View Article : Google Scholar : PubMed/NCBI

5 

Xu X, Zhao J, Xu Z, Peng B, Huang Q, Arnold E and Ding J: Structures of human cytosolic NADP-dependent isocitrate dehydrogenase reveal a novel self-regulatory mechanism of activity. J Biol Chem. 279:33946–33957. 2004. View Article : Google Scholar : PubMed/NCBI

6 

Medeiros BC, Fathi AT, DiNardo CD, Pollyea DA, Chan SM and Swords R: Isocitrate dehydrogenase mutations in myeloid malignancies. Leukemia. 31:272–281. 2017. View Article : Google Scholar : PubMed/NCBI

7 

Clark O, Yen K and Mellinghoff IK: Molecular pathways: Isocitrate dehydrogenase mutations in cancer. Clin Cancer Res. 22:1837–1842. 2016. View Article : Google Scholar : PubMed/NCBI

8 

Liao J, Li Q, Hu Z, Yu W, Zhang K, Ma F, Han Q, Zhang H, Guo J, Hu L, et al: Mitochondrial miR-1285 regulates copper-induced mitochondrial dysfunction and mitophagy by impairing IDH2 in pig jejunal epithelial cells. J Hazard Mater. 422:1268992022. View Article : Google Scholar : PubMed/NCBI

9 

Lv JW, Song YP, Zhang ZC, Fan YJ, Xu FX, Gao L, Zhang XY, Zhang C, Wang H and Xu DZ: Gestational arsenic exposure induces anxiety-like behaviors in adult offspring by reducing DNA hydroxymethylation in the developing brain. Ecotoxicol Environ Saf. 227:1129012021. View Article : Google Scholar : PubMed/NCBI

10 

Bergaggio E, Riganti C, Garaffo G, Vitale N, Mereu E, Bandini C, Pellegrino E, Pullano V, Omedè P, Todoerti K, et al: IDH2 inhibition enhances proteasome inhibitor responsiveness in hematological malignancies. Blood. 133:156–167. 2019. View Article : Google Scholar : PubMed/NCBI

11 

Park JB, Nagar H, Choi S, Jung SB, Kim HW, Kang SK, Lee JW, Lee JH, Park JW, Irani K, et al: IDH2 deficiency impairs mitochondrial function in endothelial cells and endothelium-dependent vasomotor function. Free Radic Biol Med. 94:36–46. 2016. View Article : Google Scholar : PubMed/NCBI

12 

Lang F, Jha A, Meuter L, Pacak K and Yang C: Identification of isocitrate dehydrogenase 2 (IDH2) mutation in carotid body paraganglioma. Front Endocrinol (Lausanne). 12:7310962021. View Article : Google Scholar : PubMed/NCBI

13 

Cairns RA and Mak TW: Oncogenic isocitrate dehydrogenase mutations: Mechanisms, models, and clinical opportunities. Cancer Discov. 3:730–741. 2013. View Article : Google Scholar : PubMed/NCBI

14 

Shi F, He Y, Li J, Tang M, Li Y, Xie L, Zhao L, Hu J, Luo X, Zhou M, et al: Wild-type IDH2 contributes to Epstein-Barr virus-dependent metabolic alterations and tumorigenesis. Mol Metab. 36:1009662020. View Article : Google Scholar : PubMed/NCBI

15 

Cerchione C, Romano A, Daver N, DiNardo C, Jabbour EJ, Konopleva M, Ravandi-Kashani F, Kadia T, Martelli MP, Isidori A, et al: IDH1/IDH2 inhibition in acute myeloid leukemia. Front Oncol. 11:6393872021. View Article : Google Scholar : PubMed/NCBI

16 

Stein EM, DiNardo CD, Fathi AT, Pollyea DA, Stone RM, Altman JK, Roboz GJ, Patel MR, Collins R, Flinn IW, et al: Molecular remission and response patterns in patients with mutant-IDH2 acute myeloid leukemia treated with enasidenib. Blood. 133:676–687. 2019. View Article : Google Scholar : PubMed/NCBI

17 

Abou Dalle I and DiNardo CD: The role of enasidenib in the treatment of mutant IDH2 acute myeloid leukemia. Ther Adv Hematol. 9:163–173. 2018. View Article : Google Scholar : PubMed/NCBI

18 

Rocquain J, Carbuccia N, Trouplin V, Raynaud S, Murati A, Nezri M, Tadrist Z, Olschwang S, Vey N, Birnbaum D, et al: Combined mutations of ASXL1, CBL, FLT3, IDH1, IDH2, JAK2, KRAS, NPM1, NRAS, RUNX1, TET2 and WT1 genes in myelodysplastic syndromes and acute myeloid leukemias. BMC Cancer. 10:4012010. View Article : Google Scholar : PubMed/NCBI

19 

Montoro J, Yerlikaya A, Ali A and Raza A: Improving treatment for myelodysplastic syndromes patients. Curr Treat Options Oncol. 19:662018. View Article : Google Scholar : PubMed/NCBI

20 

Gelsi-Boyer V, Trouplin V, Roquain J, Adélaïde J, Carbuccia N, Esterni B, Finetti P, Murati A, Arnoulet C, Zerazhi H, et al: ASXL1 mutation is associated with poor prognosis and acute transformation in chronic myelomonocytic leukaemia. Br J Haematol. 151:365–375. 2010. View Article : Google Scholar : PubMed/NCBI

21 

Woods BA and Levine RL: The role of mutations in epigenetic regulators in myeloid malignancies. Immunol Rev. 263:22–35. 2015. View Article : Google Scholar : PubMed/NCBI

22 

Molenaar RJ, Thota S, Nagata Y, Patel B, Clemente M, Przychodzen B, Hirsh C, Viny AD, Hosano N, Bleeker FE, et al: Clinical and biological implications of ancestral and non-ancestral IDH1 and IDH2 mutations in myeloid neoplasms. Leukemia. 29:2134–2142. 2015. View Article : Google Scholar : PubMed/NCBI

23 

Buege MJ, DiPippo AJ and DiNardo CD: Evolving treatment strategies for elderly leukemia patients with IDH mutations. Cancers (Basel). 10:1872018. View Article : Google Scholar : PubMed/NCBI

24 

Amaya ML and Pollyea DA: Targeting the IDH2 pathway in acute myeloid leukemia. Clin Cancer Res. 24:4931–4936. 2018. View Article : Google Scholar : PubMed/NCBI

25 

Chen TC, Yao CY, Chen YR, Yuan CT, Lin CC, Hsu YC, Chuang PH, Kao CJ, Li YH, Hou HA, et al: Oncogenesis induced by combined Phf6 and Idh2 mutations through increased oncometabolites and impaired DNA repair. Oncogene. 41:1576–1588. 2022. View Article : Google Scholar : PubMed/NCBI

26 

Gross S, Cairns RA, Minden MD, Driggers EM, Bittinger MA, Jang HG, Sasaki M, Jin S, Schenkein DP, Su SM, et al: Cancer-associated metabolite 2-hydroxyglutarate accumulates in acute myelogenous leukemia with isocitrate dehydrogenase 1 and 2 mutations. J Exp Med. 207:339–344. 2010. View Article : Google Scholar : PubMed/NCBI

27 

Chen C, Liu Y, Lu C, Cross JR, Morris JP IV, Shroff AS, Ward PS, Bradner JE, Thompson C and Lowe SW: Cancer-associated IDH2 mutants drive an acute myeloid leukemia that is susceptible to Brd4 inhibition. Genes Dev. 27:1974–1985. 2013. View Article : Google Scholar : PubMed/NCBI

28 

Waitkus MS, Diplas BH and Yan H: Biological role and therapeutic potential of IDH mutations in cancer. Cancer Cell. 34:186–195. 2018. View Article : Google Scholar : PubMed/NCBI

29 

Upadhyay VA, Brunner AM and Fathi AT: Isocitrate dehydrogenase (IDH) inhibition as treatment of myeloid malignancies: Progress and future directions. Pharmacol Ther. 177:123–128. 2017. View Article : Google Scholar : PubMed/NCBI

30 

Kats LM, Reschke M, Taulli R, Pozdnyakova O, Burgess K, Bhargava P, Straley K, Karnik R, Meissner A, Small D, et al: Proto-oncogenic role of mutant IDH2 in leukemia initiation and maintenance. Cell Stem Cell. 14:329–341. 2014. View Article : Google Scholar : PubMed/NCBI

31 

Chotirat S, Thongnoppakhun W, Wanachiwanawin W and Auewarakul CU: Acquired somatic mutations of isocitrate dehydrogenases 1 and 2 (IDH1 and IDH2) in preleukemic disorders. Blood Cells Mol Dis. 54:286–291. 2015. View Article : Google Scholar : PubMed/NCBI

32 

Sunthankar KI, Jenkins MT, Cote CH, Patel SB, Welner RS and Ferrell PB: Isocitrate dehydrogenase mutations are associated with altered IL-1β responses in acute myeloid leukemia. Leukemia. 36:923–934. 2022. View Article : Google Scholar : PubMed/NCBI

33 

Venugopal S, Takahashi K, Daver N, Maiti A, Borthakur G, Loghavi S, Short NJ, Ohanian M, Masarova L, Issa G, et al: Efficacy and safety of enasidenib and azacitidine combination in patients with IDH2 mutated acute myeloid leukemia and not eligible for intensive chemotherapy. Blood Cancer J. 12:102022. View Article : Google Scholar : PubMed/NCBI

34 

Chen J, Yang J, Wei Q, Weng L, Wu F, Shi Y, Cheng X, Cai X, Hu C and Cao P: Identification of a selective inhibitor of IDH2/R140Q enzyme that induces cellular differentiation in leukemia cells. Cell Commun Signal. 18:552020. View Article : Google Scholar : PubMed/NCBI

35 

Gao M, Zhu H, Fu L, Li Y, Bao X, Fu H, Quan H, Wang L and Lou L: Pharmacological characterization of TQ05310, a potent inhibitor of isocitrate dehydrogenase 2 R140Q and R172K mutants. Cancer Sci. 110:3306–3314. 2019. View Article : Google Scholar : PubMed/NCBI

36 

Ma R and Yun CH: Crystal structures of pan-IDH inhibitor AG-881 in complex with mutant human IDH1 and IDH2. Biochem Biophys Res Commun. 503:2912–2917. 2018. View Article : Google Scholar : PubMed/NCBI

37 

Yen K, Travins J, Wang F, David MD, Artin E, Straley K, Padyana A, Gross S, DeLaBarre B, Tobin E, et al: AG-221, a first-in-class therapy targeting acute myeloid leukemia harboring oncogenic IDH2 mutations. Cancer Discov. 7:478–493. 2017. View Article : Google Scholar : PubMed/NCBI

38 

Quek L, David MD, Kennedy A, Metzner M, Amatangelo M, Shih A, Stoilova B, Quivoron C, Heiblig M, Willekens C, et al: Clonal heterogeneity of acute myeloid leukemia treated with the IDH2 inhibitor enasidenib. Nat Med. 24:1167–1177. 2018. View Article : Google Scholar : PubMed/NCBI

39 

Kim Y, Jeung HK, Cheong JW, Song J, Bae SH, Lee JI and Min YH: All-trans retinoic acid synergizes with enasidenib to induce differentiation of IDH2-mutant acute myeloid leukemia cells. Yonsei Med J. 61:762–773. 2020. View Article : Google Scholar : PubMed/NCBI

40 

DiNardo CD, Schuh AC, Stein EM, Montesinos P, Wei AH, de Botton S, Zeidan AM, Fathi AT, Kantarjian HM, Bennett JM, et al: Enasidenib plus azacitidine versus azacitidine alone in patients with newly diagnosed, mutant-IDH2 acute myeloid leukaemia (AG221-AML-005): A single-arm, phase 1b and randomised, phase 2 trial. Lancet Oncol. 22:1597–1608. 2021. View Article : Google Scholar : PubMed/NCBI

41 

Stein EM, DiNardo CD, Pollyea DA, Fathi AT, Roboz GJ, Altman JK, Stone RM, DeAngelo DJ, Levine RL, Flinn IW, et al: Enasidenib in mutant-IDH2 relapsed or refractory acute myeloid leukemia. Blood. 130:722–731. 2017. View Article : Google Scholar : PubMed/NCBI

42 

Wang F, Travins J, DeLaBarre B, Penard-Lacronique V, Schalm S, Hansen E, Straley K, Kernytsky A, Liu W, Gliser C, et al: Targeted inhibition of mutant IDH2 in leukemia cells induces cellular differentiation. Science. 340:622–626. 2013. View Article : Google Scholar : PubMed/NCBI

43 

Chen J, Yang J, Sun X, Wang Z, Cheng X, Lu W, Cai X, Hu C, Shen X and Cao P: Allosteric inhibitor remotely modulates the conformation of the orthestric pockets in mutant IDH2/R140Q. Sci Rep. 7:164582017. View Article : Google Scholar : PubMed/NCBI

44 

Wang Z, Zhang Z, Li Y, Sun L, Peng D, Du D, Zhang X, Han L, Zhao L, Lu L, et al: Preclinical efficacy against acute myeloid leukaemia of SH1573, a novel mutant IDH2 inhibitor approved for clinical trials in China. Acta Pharm Sin B. 11:1526–1540. 2021. View Article : Google Scholar : PubMed/NCBI

45 

Aref S, Kamel Areida el S, Abdel Aaal MF, Adam OM, El-Ghonemy MS, El-Baiomy MA and Zeid TA: Prevalence and clinical effect of IDH1 and IDH2 mutations among cytogenetically normal acute myeloid leukemia patients. Clin Lymphoma Myeloma Leuk. 15:550–555. 2015. View Article : Google Scholar : PubMed/NCBI

46 

Middeke JM, Metzeler KH, Röllig C, Krämer M, Eckardt JN, Stasik S, Greif PA, Spiekermann K, Rothenberg-Thurley M, Krug U, et al: Differential impact of IDH1/2 mutational subclasses on outcome in adult AML: Results from a large multicenter study. Blood Adv. 6:1394–1405. 2022. View Article : Google Scholar : PubMed/NCBI

47 

Itzykson R, Kosmider O, Renneville A, Gelsi-Boyer V, Meggendorfer M, Morabito M, Berthon C, Adès L, Fenaux P, Beyne-Rauzy O, et al: Prognostic score including gene mutations in chronic myelomonocytic leukemia. J Clin Oncol. 31:2428–2436. 2013. View Article : Google Scholar : PubMed/NCBI

48 

Willekens C, Rahme R, Duchmann M, Vidal V, Saada V, Broutin S, Delahousse J, Renneville A, Marceau A, Clappier E, et al: Effects of azacitidine in 93 patients with IDH1/2 mutated acute myeloid leukemia/myelodysplastic syndromes: A French retrospective multicenter study. Leuk Lymphoma. 62:438–445. 2021. View Article : Google Scholar : PubMed/NCBI

49 

Hosono N: Genetic abnormalities and pathophysiology of MDS. Int J Clin Oncol. 24:885–892. 2019. View Article : Google Scholar : PubMed/NCBI

50 

Lin J, Yao DM, Qian J, Chen Q, Qian W, Li Y, Yang J, Wang CZ, Chai HY, Qian Z, et al: IDH1 and IDH2 mutation analysis in Chinese patients with acute myeloid leukemia and myelodysplastic syndrome. Ann Hematol. 91:519–525. 2012. View Article : Google Scholar : PubMed/NCBI

51 

Kosmider O, Gelsi-Boyer V, Slama L, Dreyfus F, Beyne-Rauzy O, Quesnel B, Hunault-Berger M, Slama B, Vey N, Lacombe C, et al: Mutations of IDH1 and IDH2 genes in early and accelerated phases of myelodysplastic syndromes and MDS/myeloproliferative neoplasms. Leukemia. 24:1094–1096. 2010. View Article : Google Scholar : PubMed/NCBI

52 

Lin CC, Hou HA, Chou WC, Kuo YY, Liu CY, Chen CY, Lai YJ, Tseng MH, Huang CF, Chiang YC, et al: IDH mutations are closely associated with mutations of DNMT3A, ASXL1 and SRSF2 in patients with myelodysplastic syndromes and are stable during disease evolution. Am J Hematol. 89:137–144. 2014. View Article : Google Scholar : PubMed/NCBI

53 

Lin P, Luo Y, Zhu S, Maggio D, Yang H, Hu C, Wang J, Zhang H, Ren Y, Zhou X, et al: Isocitrate dehydrogenase 2 mutations correlate with leukemic transformation and are predicted by 2-hydroxyglutarate in myelodysplastic syndromes. J Cancer Res Clin Oncol. 144:1037–1047. 2018. View Article : Google Scholar : PubMed/NCBI

54 

Kharfan-Dabaja MA, Komrokji RS, Zhang Q, Kumar A, Tsalatsanis A, Perkins J, Nishihori T, Field T, Al Ali N, Mishra A, et al: TP53 and IDH2 somatic mutations are associated with inferior overall survival after allogeneic hematopoietic cell transplantation for myelodysplastic syndrome. Clin Lymphoma Myeloma Leuk. 17:753–758. 2017. View Article : Google Scholar : PubMed/NCBI

55 

Stein EM, Fathi AT, DiNardo CD, Pollyea DA, Roboz GJ, Collins R, Sekeres MA, Stone RM, Attar EC, Frattini MG, et al: Enasidenib in patients with mutant IDH2 myelodysplastic syndromes: A phase 1 subgroup analysis of the multicentre, AG221-C-001 trial. Lancet Haematol. 7:e309–e319. 2020. View Article : Google Scholar : PubMed/NCBI

56 

Greenberg P, Cox C, LeBeau MM, Fenaux P, Morel P, Sanz G, Sanz M, Vallespi T, Hamblin T, Oscier D, et al: International scoring system for evaluating prognosis in myelodysplastic syndromes. Blood. 89:2079–2088. 1997. View Article : Google Scholar : PubMed/NCBI

57 

Turkalp Z, Karamchandani J and Das S: IDH mutation in glioma: New insights and promises for the future. JAMA Neurol. 71:1319–1325. 2014. View Article : Google Scholar : PubMed/NCBI

58 

Li JJ, Li R, Wang W, Zhang B, Song X, Zhang C, Gao Y, Liao Q, He Y, You S, et al: IDH2 is a novel diagnostic and prognostic serum biomarker for non-small-cell lung cancer. Mol Oncol. 12:602–610. 2018. View Article : Google Scholar : PubMed/NCBI

59 

Chiang S, Weigelt B, Wen HC, Pareja F, Raghavendra A, Martelotto LG, Burke KA, Basili T, Li A, Geyer FC, et al: IDH2 mutations define a unique subtype of breast cancer with altered nuclear polarity. Cancer Res. 76:7118–7129. 2016. View Article : Google Scholar : PubMed/NCBI

60 

Steinhilber J, Mederake M, Bonzheim I, Serinsöz-Linke E, Müller I, Fallier-Becker P, Lemonnier F, Gaulard P, Fend F and Quintanilla-Martinez L: The pathological features of angioimmunoblastic T-cell lymphomas with IDH2R172 mutations. Mod Pathol. 32:1123–1134. 2019. View Article : Google Scholar : PubMed/NCBI

61 

Zhu GG, Nafa K, Agaram N, Zehir A, Benayed R, Sadowska J, Borsu L, Kelly C, Tap WD, Fabbri N, et al: Genomic profiling identifies association of IDH1/IDH2 mutation with longer relapse-free and metastasis-free survival in high-grade chondrosarcoma. Clin Cancer Res. 26:419–427. 2020. View Article : Google Scholar : PubMed/NCBI

62 

Libera L, Ottini G, Sahnane N, Pettenon F, Turri-Zanoni M, Lambertoni A, Chiaravalli AM, Leone F, Battaglia P, Castelnuovo P, et al: Methylation drivers and prognostic implications in sinonasal poorly differentiated carcinomas. Cancers (Basel). 13:50302021. View Article : Google Scholar : PubMed/NCBI

63 

Miller JJ, Shih HA, Andronesi OC and Cahill DP: Isocitrate dehydrogenase-mutant glioma: Evolving clinical and therapeutic implications. Cancer. 123:4535–4546. 2017. View Article : Google Scholar : PubMed/NCBI

64 

Qi S, Yu L, Li H, Ou Y, Qiu X, Ding Y, Han H and Zhang X: Isocitrate dehydrogenase mutation is associated with tumor location and magnetic resonance imaging characteristics in astrocytic neoplasms. Oncol Lett. 7:1895–1902. 2014. View Article : Google Scholar : PubMed/NCBI

65 

Picca A, Berzero G, Di Stefano AL and Sanson M: The clinical use of IDH1 and IDH2 mutations in gliomas. Expert Rev Mol Diagn. 18:1041–1051. 2018. View Article : Google Scholar : PubMed/NCBI

66 

Gusyatiner O and Hegi ME: Glioma epigenetics: From subclassification to novel treatment options. Semin Cancer Biol. 51:50–58. 2018. View Article : Google Scholar : PubMed/NCBI

67 

Ludwig N, Rao A, Sandlesh P, Yerneni SS, Swain AD, Bullock KM, Hansen KM, Zhang X, Jaman E, Allen J, et al: Characterization of systemic immunosuppression by IDH mutant glioma small extracellular vesicles. Neuro Oncol. 24:197–209. 2022. View Article : Google Scholar : PubMed/NCBI

68 

Phan K, Ng W, Lu VM, McDonald KL, Fairhall J, Reddy R and Wilson P: Association between IDH1 and IDH2 mutations and preoperative seizures in patients with low-grade versus high-grade glioma: A systematic review and meta-analysis. World Neurosurg. 111:e539–e545. 2018. View Article : Google Scholar : PubMed/NCBI

69 

Chen R, Ravindra VM, Cohen AL, Jensen RL, Salzman KL, Prescot AP and Colman H: Molecular features assisting in diagnosis, surgery, and treatment decision making in low-grade gliomas. Neurosurg Focus. 38:E22015. View Article : Google Scholar : PubMed/NCBI

70 

Thirumal Kumar D, Jerushah Emerald L, George Priya Doss C, Sneha P, Siva R, Charles Emmanuel Jebaraj W and Zayed H: Computational approach to unravel the impact of missense mutations of proteins (D2HGDH and IDH2) causing D-2-hydroxyglutaric aciduria 2. Metab Brain Dis. 33:1699–1710. 2018. View Article : Google Scholar : PubMed/NCBI

71 

Kim H, Kim SH, Cha H, Kim SR, Lee JH and Park JW: IDH2 deficiency promotes mitochondrial dysfunction and dopaminergic neurotoxicity: Implications for Parkinson's disease. Free Radic Res. 50:853–860. 2016. View Article : Google Scholar : PubMed/NCBI

72 

Li J, Lu J, He Y, Wu Y, Wu Y, Song X, Jiang Y, Tang M, Weng X, Yi W, et al: A new functional IDH2 genetic variant is associated with the risk of lung cancer. Mol Carcinog. 56:1082–1087. 2017. View Article : Google Scholar : PubMed/NCBI

73 

Li J, He Y, Tan Z, Lu J, Li L, Song X, Shi F, Xie L, You S, Luo X, et al: Wild-type IDH2 promotes the Warburg effect and tumor growth through HIF1α in lung cancer. Theranostics. 8:4050–4061. 2018. View Article : Google Scholar : PubMed/NCBI

74 

Park JH, Ku HJ, Lee JH and Park JW: Idh2 deficiency exacerbates acrolein-induced lung injury through mitochondrial redox environment deterioration. Oxid Med Cell Longev. 2017:15951032017. View Article : Google Scholar : PubMed/NCBI

75 

Park JH, Ku HJ, Lee JH and Park JW: Disruption of IDH2 attenuates lipopolysaccharide-induced inflammation and lung injury in an α-ketoglutarate-dependent manner. Biochem Biophys Res Commun. 503:798–802. 2018. View Article : Google Scholar : PubMed/NCBI

76 

Holst JM, Enemark MB, Pedersen MB, Lauridsen KL, Hybel TE, Clausen MR, Frederiksen H, Møller MB, Nørgaard P, Plesner TL, et al: Proteomic profiling differentiates lymphoma patients with and without concurrent myeloproliferative neoplasia. Cancers (Basel). 13:55262021. View Article : Google Scholar : PubMed/NCBI

77 

Lemonnier F, Cairns RA, Inoue S, Li WY, Dupuy A, Broutin S, Martin N, Fataccioli V, Pelletier R, Wakeham A, et al: The IDH2 R172K mutation associated with angioimmunoblastic T-cell lymphoma produces 2HG in T cells and impacts lymphoid development. Proc Natl Acad Sci USA. 113:15084–15089. 2016. View Article : Google Scholar : PubMed/NCBI

78 

Wang C, McKeithan TW, Gong Q, Zhang W, Bouska A, Rosenwald A, Gascoyne RD, Wu X, Wang J, Muhammad Z, et al: IDH2R172 mutations define a unique subgroup of patients with angioimmunoblastic T-cell lymphoma. Blood. 126:1741–1752. 2015. View Article : Google Scholar : PubMed/NCBI

79 

Churchill H, Naina H, Boriack R, Rakheja D and Chen W: Discordant intracellular and plasma D-2-hydroxyglutarate levels in a patient with IDH2 mutated angioimmunoblastic T-cell lymphoma. Int J Clin Exp Pathol. 8:11753–11759. 2015.PubMed/NCBI

80 

Dupuy A, Lemonnier F, Fataccioli V, Martin-Garcia N, Robe C, Pelletier R, Poullot E, Moktefi A, Mokhtari K, Rousselet MC, et al: Multiple ways to detect IDH2 mutations in angioimmunoblastic T-cell lymphoma from immunohistochemistry to next-generation sequencing. J Mol Diagn. 20:677–685. 2018. View Article : Google Scholar : PubMed/NCBI

81 

Ye Y, Ding N, Mi L, Shi Y, Liu W, Song Y, Shu S and Zhu J: Correlation of mutational landscape and survival outcome of peripheral T-cell lymphomas. Exp Hematol Oncol. 10:92021. View Article : Google Scholar : PubMed/NCBI

82 

Pareja F, da Silva EM, Frosina D, Geyer FC, Lozada JR, Basili T, Da Cruz Paula A, Zhong E, Derakhshan F, D'Alfonso T, et al: Immunohistochemical analysis of IDH2 R172 hotspot mutations in breast papillary neoplasms: Applications in the diagnosis of tall cell carcinoma with reverse polarity. Mod Pathol. 33:1056–1064. 2020. View Article : Google Scholar : PubMed/NCBI

83 

Aljohani AI, Toss MS, Kurozumi S, Joseph C, Aleskandarany MA, Miligy IM, Ansari RE, Mongan NP, Ellis IO, Green AR and Rakha EA: The prognostic significance of wild-type isocitrate dehydrogenase 2 (IDH2) in breast cancer. Breast Cancer Res Treat. 179:79–90. 2020. View Article : Google Scholar : PubMed/NCBI

84 

Wang Y, Agarwal E, Bertolini I, Ghosh JC, Seo JH and Altieri DC: IDH2 reprograms mitochondrial dynamics in cancer through a HIF-1α-regulated pseudohypoxic state. FASEB J. 33:13398–13411. 2019. View Article : Google Scholar : PubMed/NCBI

85 

Jo VY, Chau NG, Hornick JL, Krane JF and Sholl LM: Recurrent IDH2 R172X mutations in sinonasal undifferentiated carcinoma. Mod Pathol. 30:650–659. 2017. View Article : Google Scholar : PubMed/NCBI

86 

Dogan S, Chute DJ, Xu B, Ptashkin RN, Chandramohan R, Casanova-Murphy J, Nafa K, Bishop JA, Chiosea SI, Stelow EB, et al: Frequent IDH2 R172 mutations in undifferentiated and poorly-differentiated sinonasal carcinomas. J Pathol. 242:400–408. 2017. View Article : Google Scholar : PubMed/NCBI

87 

Riobello C, López-Hernández A, Cabal VN, García-Marín R, Suárez-Fernández L, Sánchez-Fernández P, Vivanco B, Blanco V, López F, Franchi A, et al: IDH2 mutation analysis in undifferentiated and poorly differentiated sinonasal carcinomas for diagnosis and clinical management. Am J Surg Pathol. 44:396–405. 2020. View Article : Google Scholar : PubMed/NCBI

88 

Kim YR, Kim KH, Lee S, Oh SK, Park JW, Lee KY, Baek JI and Kim UK: Expression patterns of members of the isocitrate dehydrogenase gene family in murine inner ear. Biotech Histochem. 92:536–544. 2017. View Article : Google Scholar : PubMed/NCBI

89 

White K, Kim MJ, Han C, Park HJ, Ding D, Boyd K, Walker L, Linser P, Meneses Z, Slade C, et al: Loss of IDH2 accelerates age-related hearing loss in male mice. Sci Rep. 8:50392018. View Article : Google Scholar : PubMed/NCBI

90 

Ku HJ, Park JH, Kim SH and Park JW: Isocitrate dehydrogenase 2 deficiency exacerbates dermis damage by ultraviolet-B via ΔNp63 downregulation. Biochim Biophys Acta Mol Basis Dis. 1864:1138–1147. 2018. View Article : Google Scholar : PubMed/NCBI

91 

Kim SH and Park JW: IDH2 deficiency impairs cutaneous wound healing via ROS-dependent apoptosis. Biochim Biophys Acta Mol Basis Dis. 1865:1655232019. View Article : Google Scholar : PubMed/NCBI

92 

Lian CG, Xu Y, Ceol C, Wu F, Larson A, Dresser K, Xu W, Tan L, Hu Y, Zhan Q, et al: Loss of 5-hydroxymethylcytosine is an epigenetic hallmark of melanoma. Cell. 150:1135–1146. 2012. View Article : Google Scholar : PubMed/NCBI

93 

Lv Q, Xing S, Li Z, Li J, Gong P, Xu X, Chang L, Jin X, Gao F, Li W, et al: Altered expression levels of IDH2 are involved in the development of colon cancer. Exp Ther Med. 4:801–806. 2012. View Article : Google Scholar : PubMed/NCBI

94 

Teicher BA, Linehan WM and Helman LJ: Targeting cancer metabolism. Clin Cancer Res. 18:5537–5545. 2012. View Article : Google Scholar : PubMed/NCBI

95 

Park JH, Ku HJ, Lee JH and Park JW: IDH2 deficiency accelerates skin pigmentation in mice via enhancing melanogenesis. Redox Biol. 17:16–24. 2018. View Article : Google Scholar : PubMed/NCBI

96 

Liu Z, Gan L, Zhang T, Ren Q and Sun C: Melatonin alleviates adipose inflammation through elevating α-ketoglutarate and diverting adipose-derived exosomes to macrophages in mice. J Pineal Res. 64:e124552018. View Article : Google Scholar

97 

Gong F, Gao L and Ding T: IDH2 protects against nonalcoholic steatohepatitis by alleviating dyslipidemia regulated by oxidative stress. Biochem Biophys Res Commun. 514:593–600. 2019. View Article : Google Scholar : PubMed/NCBI

98 

Chen X, Zhuo S, Xu W, Chen X, Huang D, Sun X and Cheng Y: Isocitrate dehydrogenase 2 contributes to radiation resistance of oesophageal squamous cell carcinoma via regulating mitochondrial function and ROS/pAKT signalling. Br J Cancer. 123:126–136. 2020. View Article : Google Scholar : PubMed/NCBI

99 

Lee SJ, Cha H, Lee S, Kim H, Ku HJ, Kim SH, Park JH, Lee JH, Park KM and Park JW: Idh2 deficiency accelerates renal dysfunction in aged mice. Biochem Biophys Res Commun. 493:34–39. 2017. View Article : Google Scholar : PubMed/NCBI

100 

Lee SH, Jo SH, Lee SM, Koh HJ, Song H, Park JW, Lee WH and Huh TL: Role of NADP+-dependent isocitrate dehydrogenase (NADP+-ICDH) on cellular defence against oxidative injury by gamma-rays. Int J Radiat Biol. 80:635–642. 2004. View Article : Google Scholar : PubMed/NCBI

101 

Lee JH, Go Y, Kim DY, Lee SH, Kim OH, Jeon YH, Kwon TK, Bae JH, Song DK, Rhyu IJ, et al: Isocitrate dehydrogenase 2 protects mice from high-fat diet-induced metabolic stress by limiting oxidative damage to the mitochondria from brown adipose tissue. Exp Mol Med. 52:238–252. 2020. View Article : Google Scholar : PubMed/NCBI

102 

Lee JH, Kim SY, Kil IS and Park JW: Regulation of ionizing radiation-induced apoptosis by mitochondrial NADP+-dependent isocitrate dehydrogenase. J Biol Chem. 282:13385–13394. 2007. View Article : Google Scholar : PubMed/NCBI

103 

Kim SY, Yoo YH and Park JW: Silencing of mitochondrial NADP(+)-dependent isocitrate dehydrogenase gene enhances glioma radiosensitivity. Biochem Biophys Res Commun. 433:260–265. 2013. View Article : Google Scholar : PubMed/NCBI

104 

Li S, Chou AP, Chen W, Chen R, Deng Y, Phillips HS, Selfridge J, Zurayk M, Lou JJ, Everson RG, et al: Overexpression of isocitrate dehydrogenase mutant proteins renders glioma cells more sensitive to radiation. Neuro Oncol. 15:57–68. 2013. View Article : Google Scholar : PubMed/NCBI

105 

van den Bent MJ, Dubbink HJ, Marie Y, Brandes AA, Taphoorn MJ, Wesseling P, Frenay M, Tijssen CC, Lacombe D, Idbaih A, et al: IDH1 and IDH2 mutations are prognostic but not predictive for outcome in anaplastic oligodendroglial tumors: A report of the European organization for research and treatment of cancer brain tumor group. Clin Cancer Res. 16:1597–1604. 2010. View Article : Google Scholar : PubMed/NCBI

106 

Bleeker FE, Atai NA, Lamba S, Jonker A, Rijkeboer D, Bosch KS, Tigchelaar W, Troost D, Vandertop WP, Bardelli A and Van Noorden CJ: The prognostic IDH1 (R132) mutation is associated with reduced NADP+-dependent IDH activity in glioblastoma. Acta Neuropathol. 119:487–494. 2010. View Article : Google Scholar : PubMed/NCBI

107 

Cho HJ, Cho HY, Park JW, Kwon OS, Lee HS, Huh TL and Kang BS: NADP+-dependent cytosolic isocitrate dehydrogenase provides NADPH in the presence of cadmium due to the moderate chelating effect of glutathione. J Biol Inorg Chem. 23:849–860. 2018. View Article : Google Scholar : PubMed/NCBI

108 

Zhao CB, Shi L, Pu HH and Zhang QY: The promoting effect of radiation on glucose metabolism in breast cancer cells under the treatment of cobalt chloride. Pathol Oncol Res. 23:47–53. 2017. View Article : Google Scholar : PubMed/NCBI

109 

Oliveira P, Barboza LGA, Branco V, Figueiredo N, Carvalho C and Guilhermino L: Effects of microplastics and mercury in the freshwater bivalve corbicula fluminea (Müller, 1774): Filtration rate, biochemical biomarkers and mercury bioconcentration. Ecotoxicol Environ Saf. 164:155–163. 2018. View Article : Google Scholar : PubMed/NCBI

110 

Kil IS, Shin SW, Yeo HS, Lee YS and Park JW: Mitochondrial NADP+-dependent isocitrate dehydrogenase protects cadmium-induced apoptosis. Mol Pharmacol. 70:1053–1061. 2006. View Article : Google Scholar : PubMed/NCBI

111 

Li C, Xu Y, Li L, Yang X and Wang Y: Acid stress induces cross-protection for cadmium tolerance of multi-stress-tolerant Pichia kudriavzevii by regulating cadmium transport and antioxidant defense system. J Hazard Mater. 366:151–159. 2019. View Article : Google Scholar : PubMed/NCBI

112 

da Silva Fonseca J, de Barros Marangoni LF, Marques JA and Bianchini A: Energy metabolism enzymes inhibition by the combined effects of increasing temperature and copper exposure in the coral mussismilia harttii. Chemosphere. 236:1244202019. View Article : Google Scholar : PubMed/NCBI

113 

Pan JH, Kim HS, Beane KE, Montalbano AM, Lee JH, Kim YJ, Kim JH, Kong BC, Kim S, Park JW, et al: IDH2 deficiency aggravates fructose-induced NAFLD by modulating hepatic fatty acid metabolism and activating inflammatory signaling in female mice. Nutrients. 10:6792018. View Article : Google Scholar : PubMed/NCBI

114 

Chae U, Park JW, Lee SR, Lee HJ, Lee HS and Lee DS: Reactive oxygen species-mediated senescence is accelerated by inhibiting Cdk2 in Idh2-deficient conditions. Aging (Albany NY). 11:7242–7256. 2019. View Article : Google Scholar : PubMed/NCBI

115 

Xu Y, Liu L, Nakamura A, Someya S, Miyakawa T and Tanokura M: Studies on the regulatory mechanism of isocitrate dehydrogenase 2 using acetylation mimics. Sci Rep. 7:97852017. View Article : Google Scholar : PubMed/NCBI

116 

Yu W, Dittenhafer-Reed KE and Denu JM: SIRT3 protein deacetylates isocitrate dehydrogenase 2 (IDH2) and regulates mitochondrial redox status. J Biol Chem. 287:14078–14086. 2012. View Article : Google Scholar : PubMed/NCBI

117 

Zou X, Zhu Y, Park SH, Liu G, O'Brien J, Jiang H and Gius D: SIRT3-mediated dimerization of IDH2 directs cancer cell metabolism and tumor growth. Cancer Res. 77:3990–3999. 2017. View Article : Google Scholar : PubMed/NCBI

118 

Smolková K, Špačková J, Gotvaldová K, Dvořák A, Křenková A, Hubálek M, Holendová B, Vítek L and Ježek P: SIRT3 and GCN5L regulation of NADP+- and NADPH-driven reactions of mitochondrial isocitrate dehydrogenase IDH2. Sci Rep. 10:86772020. View Article : Google Scholar : PubMed/NCBI

119 

Wang T, Zhang F, Peng W, Wang L, Zhang J, Dong W, Tian X, Ye C, Li Y and Gong Y: Overexpression of NMNAT3 improves mitochondrial function and enhances antioxidative stress capacity of bone marrow mesenchymal stem cells via the NAD+-Sirt3 pathway. Biosci Rep. 42:BSR202110052022. View Article : Google Scholar : PubMed/NCBI

120 

Yu Y, Chen Y, Liu K, Cheng J and Tu J: SUMOylation enhances the activity of IDH2 under oxidative stress. Biochem Biophys Res Commun. 532:591–597. 2020. View Article : Google Scholar : PubMed/NCBI

121 

Ogawara Y, Katsumoto T, Aikawa Y, Shima Y, Kagiyama Y, Soga T, Matsunaga H, Seki T, Araki K and Kitabayashi I: IDH2 and NPM1 mutations cooperate to activate Hoxa9/Meis1 and hypoxia pathways in acute myeloid Leukemia. Cancer Res. 75:2005–2016. 2015. View Article : Google Scholar : PubMed/NCBI

122 

Leonardi R, Subramanian C, Jackowski S and Rock CO: Cancer-associated isocitrate dehydrogenase mutations inactivate NADPH-dependent reductive carboxylation. J Biol Chem. 287:14615–14620. 2012. View Article : Google Scholar : PubMed/NCBI

123 

Kim H, Lee JH and Park JW: Down-regulation of IDH2 sensitizes cancer cells to erastin-induced ferroptosis. Biochem Biophys Res Commun. 525:366–371. 2020. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2022
Volume 24 Issue 2

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Gong YQ, Wei S, Wei YY, Chen YL, Cui J, Yu YQ, Lin X, Yan HX, Qin H, Yi L, Yi L, et al: IDH2: A novel biomarker for environmental exposure in blood circulatory system disorders (Review). Oncol Lett 24: 278, 2022
APA
Gong, Y.Q., Wei, S., Wei, Y.Y., Chen, Y.L., Cui, J., Yu, Y.Q. ... Yi, L. (2022). IDH2: A novel biomarker for environmental exposure in blood circulatory system disorders (Review). Oncology Letters, 24, 278. https://doi.org/10.3892/ol.2022.13398
MLA
Gong, Y. Q., Wei, S., Wei, Y. Y., Chen, Y. L., Cui, J., Yu, Y. Q., Lin, X., Yan, H. X., Qin, H., Yi, L."IDH2: A novel biomarker for environmental exposure in blood circulatory system disorders (Review)". Oncology Letters 24.2 (2022): 278.
Chicago
Gong, Y. Q., Wei, S., Wei, Y. Y., Chen, Y. L., Cui, J., Yu, Y. Q., Lin, X., Yan, H. X., Qin, H., Yi, L."IDH2: A novel biomarker for environmental exposure in blood circulatory system disorders (Review)". Oncology Letters 24, no. 2 (2022): 278. https://doi.org/10.3892/ol.2022.13398