Abstract
The incidence of thyroid cancer and breast cancer is increasing year by year, and the specific pathogenesis is unclear. Posttranslational modifications constitute an important regulatory mechanism that affects the function of almost all proteins, are essential for a diverse and well-functioning proteome and can integrate metabolism with physiological and pathological processes. In recent years, posttranslational modifications, which mainly include metabolic enzyme-mediated protein posttranslational modifications, such as methylation, phosphorylation, acetylation and succinylation, have become a research hotspot. Among these modifications, lysine succinylation is a newly discovered broad-spectrum, dynamic, non-enzymatic protein post-translational modification, and it plays an important regulatory role in a variety of tumors. Studies have shown that succinylation can affect the synthesis of thyroid hormones, and the regulation of this post-translational modification can inhibit the apoptosis and migration of thyroid cancer cell lines, and promote breast cancer cell proliferation, DNA damage repair and autophagy-related regulation. However, the specific regulatory mechanism of succinylation in thyroid cancer and breast cancer is currently unclear. Therefore, this article mainly reviews the research progress of succinylation modification in thyroid cancer and breast cancer. It is expected to provide new directions and targets for the prevention and treatment of thyroid cancer and breast cancer.
Keywords: Thyroid cancer, breast cancer, posttranslational modification, succinylation, succinyl coenzyme
Introduction
Thyroid cancer (TC) and breast cancer (BC) are the two most common malignant tumors in women, and their incidence is increasing each year [1-4]. Studies conducted over the past few years have found that patients with a previous incidence of BC or TC exhibit a significantly higher overall risk of TC or BC from second primary tumor, respectively [5-8], which indicates a two-way and potential causality relationship between BC and TC. In addition, the development of these two tumor types has common etiological characteristics [9], such as hormonal factors (both TC and BC are regulated by the hypothalamic-pituitary axis), genetic factors (e.g., mutations in the tumor suppressor gene PTEN cause Cowden syndrome (CS), which is associated with a high incidence of BC and TC) and environmental and treatment-related factors [10]. Moreover, most types of TC have a good prognosis, but some types of TC are resistant to surgery and radioactive iodine therapy (RAI) [11,12] and have a poor prognosis [13]. For example, poorly differentiated thyroid carcinoma (PDTC) shows poorly differentiated and higher metastatic tendency and the patient prognosis is poor [14,15]. This therapy resistance makes the treatment of TC difficult. Similarly, BC is not only the malignant tumor with the highest incidence in women in the world, but also the main cause of female cancer deaths [16]. Its recurrence and metastasis rates are high, and the prognosis for metastatic BC is poor [16-19]. Therefore, it is necessary to further explore new diagnosis and therapy targets for TC and BC.
Protein modification refers to chemical modification after protein biosynthesis, also known as posttranslational modification (PTM). Studies have shown that PTMs play important regulatory roles in biochemical reactions and that the modification of proteins in different states leads to different functions [20,21]. Hundreds of PTMs have been identified, and these include methylation, acetylation, phosphorylation, ubiquitination, glycosylation, and succinylation, etc. [22-24]. Among these PTMs, DNA methylation represents a basic epigenetic modification that can regulate chromatin structure and gene transcription [25]. Many diseases, including cancer, display abnormal methylation patterns [26]. DNA methylation inhibitors have been used to block methylation-dependent gene silencing for the treatment of hematopoietic tumors and restoration of the expression of silenced developmental genes [27]. Similarly, histone acetylation modifications (HAMs) affect cell function and are mainly mediated by histone acetyltransferase (HAT) and histone deacetylase (HDAC), and the HAMs are essential for the development and prognosis of BC and TC [28,29]. In addition, phosphorylation is also one of the most widely studied PTMs, and is associated with colorectal cancer (CRC) [30], glioblastoma (GBM) [31], lung cancer [32,33], BC [34,35]. Therefore, PTMs are essential for the regulation of biological protein functions and cancer progression.
Succinylation is a newly discovered protein PTM that is widely present in cells and can participate in a variety of life activities by regulating protease activity and gene expression [36,37]. Studies have shown that succinylation can change the processes of enzymes and metabolic pathways (particularly mitochondrial metabolic pathways) [38-40] and thus plays a variety of important roles in cell metabolism [41,42], such as in tricarboxylic acid circulation, the electron transport chain, glycolysis, ketone body formation, fatty acid oxidation, and the urea cycle [43,44]. Succinylation is also related to many diseases, such as diseases of the liver, heart, lung and other organs [21]. In addition, increasing evidence shows that succinylation modulators can promote or inhibit a variety of cancers by regulating the succinylation level of substrate targets [45-47]. For example, carnitine palmitoyl transferase 1A (CPT1A) promotes BC cell proliferation through succinylation enolase 1 [48], and promotes gastric cancer (GC) cell metastasis through the succinylation of S100A10 [46,49]. In addition, lysine acetyltransferase 2A (KAT2A) can upregulate 14-3-3ζ through its succinyltransferase activity to promote the proliferation, migration and invasion of human pancreatic ductal adenocarcinoma (PDAC) cells [50]. The complex of KAT2A and α-ketoglutarate dehydrogenase (α-KGDH) acts as a histone H3 succinyltransferase and promotes the proliferation and development of glioma cells [51]. In addition, succinylation regulators may promote the malignant progression of renal clear cell carcinoma (ccRCC) by regulating the infiltration of immune cells and the methylation of RNA N6-methyladenosine (m6A) [52]. In contrast, the downregulation of SIRT5 (a desuccinylase) is related to the succinylation of ACOX1, an increase in its activity and the oxidative DNA damage response in hepatocellular carcinoma (HCC) [45]. And in lung cancer recombinant human superoxide dismutase-1 (SOD1) inhibits the growth of lung tumor cells through a novel posttranslational regulation of succinylation and SIRT5-dependent desuccinylation [53]. In addition, in osteosarcoma cells, the inactivation of SIRT5 can cause serine hydroxymethyltransferase (SHMT2) to enzymatically downregulate and eliminate cell growth under metabolic stress [54].
In short, succinylation can promote the occurrence of BC, gastric cancer, renal clear cell carcinoma, glioma and other tumors, and has a tumor suppressor effect on liver cancer, lung cancer and osteosarcoma cells (see Table 1 for details). However, relatively few studies have investigated the role of succinylation in the thyroid and breast, but the available evidence shows that the regulation of succinylation can inhibit the apoptosis of TC cells and promote their migration. The PTM also has important functions in BC cells, such as promoting proliferation and mediating DNA damage repair. Therefore, studying the role of succinylation modification in the occurrence and development of TC and BC has also become a new focus of cancer research, but there are few reviews in this field. Therefore, we summarized the regulatory role of succinylation modification in these two tumors, in order to determine new targets for the treatment of TC and BC patients.
Table 1.
Succinylation expression in various tumors, gene symbol, impact, Ksucc sites, regulatory factors of succinylation and inhibitor
| Tumor | Gene Symbol | Impact in Tumors | Ksucc Sites | Regulatory Factors of Succinylation | Inhibitor | References |
|---|---|---|---|---|---|---|
| Gastric Cancer | S100A10 | Promote cancer cell invasion and metastasis | K47 | CPT1A/SIRT5 | CHX | [46] |
| Lung Cancer | SOD1 | Inhibits cancer cell growth | K123 | Mutation/SIRT5 | Hygromycin | [53] |
| Clear cell renal cell Carcinoma | AGER/IL20RB/SAA1 | Immune cell infiltration and m6A methylation | HNRNP (A2B1/C/G), LRPPRC/EIF3B | CPT1A/SIRT5/SIRT7/KAT2A | PD-1 | [52] |
| Glioma | KAT2A (Tyr645Ala) | Tumour cell proliferation and development | H3K79 | Succinyl-CoA/α-KGDH | IPTG | [51] |
| Hepatocellular Carcinoma | ACOX1 | Inhibit the progression of liver cancer | K89R/K437R/K488R/K500R/K537R/K637R | SIRT5 | Streptomyces hygroscopious | [45] |
| Breast Cancer | GLS | Breast cell proliferation and tumor formation | K164 | SIRT5 | Leupeptin | [48] |
| Osteosarcoma | SHMT2 | Inhibits the proliferation of cancer cells | K280 | SIRT5 | Nicotinamide | [54] |
| Thyroid Cancer | SDHx/PTEN | Inhibit thyroid cancer cell apoptosis and promote migration | SDHD-G12S/SDHD-H50R | SDH/Succinic acid | FAD/NAD | [100] |
Basic characteristics and function of succinylation
The succinylation of proteins involves succinyl group donors, such as succinyl-CoA [37,55,56], and refers to the process of the covalently bonding of a succinyl group to a lysine residue of a protein through enzymatic or nonenzymatic methods [57-59]. In 2004, succinylation was first discovered in Escherichia coli, followed by eukaryotes [37,58,60]. To date, approximately 20,000 succinylated gut microbial peptides have been identified in the human gut flora [61]. Succinylation is also a common PTM in prokaryotes and eukaryotes, and this process mainly occurs in the cytoplasm and nucleus [57,62].
In the cytoplasm, succinylation mainly occurs in mitochondria, and the mitochondrial enzyme a-ketoglutarate dehydrogenase complex can control succinylation. Succinylation, which is also the key mitochondrial process of energy production [63], directly couples the cyclic metabolism of tricarboxylic acid (TCA) with changes in the charge, structure and activity of a protein through succinyl-CoA to participate in different cellular processes and can link metabolism with protein function to regulate the body’s metabolic activities [38]. In addition, G protein-coupled receptors (GPCRs) also play an important role in the production of mitochondrial adenosine triphosphate (ATP) [64]. GPRG1 in the GPCR family is also known as succinate receptor 1 (SUCNR1), is extensively involved in human cell metabolism [65] and activates intracellular calcium mobilization and dendritic cytokine-dependent proinflammatory cytokines [66], and thereby activates immune cells and enhances inflammation [67].
In contrast, in the nucleus, succinylation can occur in more than one-third of nucleosomes and in both histone and nonhistone lysine residues [68]. Lysine succinylation (Kcuss) provides the lysine group with two negative charges and thereby causes changes in protein properties. A succinyl group has a larger spatial structure than an acetyl group. Therefore, succinylation has a greater impact on the structure and function of a protein [37]; for example, the α-ketoglutarate dehydrogenase (α-KGDH) complex is located in the nucleus of human cell lines, interacts with lysine acetyltransferase 2A (KAT2A, also known as GCN5) by binding to the promoter region of the gene [51], and participates in gene transcription regulation [68]. KAT2A also acts as a succinyltransferase and succinylates histone H3 on lysine 79, with a maximum frequency around the transcription start sites of genes. Preventing the α-KGDH complex from entering the nucleus, or expression of KAT2A (Tyr645Ala), reduces gene expression and inhibits tumour cell proliferation and tumour growth [51].
Regulatory factors of succinylation
Succinylation is regulated by both nonenzymatic and enzymatic factors. It was initially thought that succinylation is a PTM of proteins controlled by nonenzymatic factors [69,70]. Similar to acetyl-CoA, succinyl-CoA is an inherently reactive short-chain coenzyme A thioester that can maintain the stability of the internal environment in the mitochondrial matrix over a low concentration range (0.1-0.6 mM). The mixing of succinyl-CoA with albumin or isocitrate dehydrogenase (ICDH) can increase succinylation in a PH and dose-dependent manner [39,57,58]. Protein acylation in mitochondria may be an alkaline chemical event promoted by high concentrations of active acyl-CoA in the mitochondrial matrix [39,69]. In summary, the nonenzymatic process of succinylation is mainly controlled by the concentration of succinyl-CoA in mitochondria, pH, and protein parameters [39,71,72].
In-depth research on succinylation has revealed that the level of succinylation is mainly regulated by succinyl donors, succinyltransferases and desuccinylases. The succinyl donor succinyl-CoA is mainly derived from mitochondria [58] and is produced by amino acid metabolism or the TCA cycle [73]. Succinyl-CoA is mainly produced by regulation of the TCA cycle polyprotein complex α-ketoglutarate dehydrogenase complex (KGDHC), which consists of three components, E1k [α-ketoglutarate dehydrogenase (KGDH) (EC 1.2.4.2)], E2k [dihydrolipoyl succinyltransferase (EC 2.3.1.61)] and E3 [dihydrolipoyl dehydrogenase (EC 1.8.1.4)] [73]. KGDHC can indirectly change the level of succinylation (such as regulating the level of succinyl-CoA) or directly participate in the succinylation process. For example, in yeast, the mutation and loss or activation of the E1k gene can change the level of succinylation [58]. Moreover, KGDHC can mediate succinylation under a variety of conditions. For example, the compound I inhibitor rotenone can inhibit respiration by inhibiting aconitase and reduce the ketoglutarate in KGDHC to reduce the concentration of succinyl-CoA in mitochondria [74]. Etelvino, et al., indicated that the amino acids are used for the synthesis of heme, an important source of succinyl-CoA, and this process is also mediated by KGDHC [75]. If the TCA cycle cannot provide sufficient succinyl-CoA, glutamine is deaminated to α-ketoglutarate and is converted by α-ketoglutarate diacid dehydrogenase (KDH) into succinyl-CoA for the synthesis of heme [76]. In addition, the succinic acid is a key regulator of hypoxia [77], which leads to tumor formation and congenital inflammatory diseases. Another transferase with lysine succinyltransferase activity in mammalian cells is carnitine palmitoyltransferase 1A (CPT1A) [48], as has been confirmed in gastric cancer (GC) [46]. CPT1A is a mitochondrial enzyme that uses succinyl-CoA as a substrate without changing the succinyl-CoA levels to increase lysine succinylation in cells [48]. In summary, the abovementioned succinyl donors and succinyltransferases, including the Ros-α-ketoglutarate dehydrogenase complex (ROS-KGDHC) [57,58] and carnitine palmitoyl glyceride 1A (CPT1A) [48], exert positive regulatory effects on succinylation [58].
In addition, desuccinylases, such as COBb (the first succinylase found in prokaryotes) [37] as well as SIRT5 [72] and SIRT7 [78] in the sirtuin family, exert inhibitory effects on succinylation modification. Previous studies have shown that the consumption of SIRT5 can impair the function of complex II (SDH) and fatty acid β-oxidation, which indicates that succinylation inhibits mitochondrial enzymes [40,79] and thereby affects mitochondrial metabolism. In summary, the replacement of factors during succinylation can help us understand the transformational processes of succinylation, which will benefit the diagnosis and treatment of diseases (Figure 1).
Figure 1.

The process of succinylation and its simple regulatory factors. This figure mainly shows that the succinyl donor succinyl-CoA in the protein participates in the succinylation modification process of the covalent binding of the lysine residues of the protein through enzymatic or non-enzymatic regulatory factors. Among them, succinyl-CoA is mainly derived from the TCA cycle, amino acid metabolism and down-regulation of SDH enzymes. The dynamic balance of succinylation modification adjustment mainly relies on enzyme control factors (succinylase α-KGDHC, CPT1A and desuccinylase COBb, SIRT5/7) and non-enzymatic control factors (succinyl-CoA, pH value and protein Parameters, etc.) regulation.
Role of succinylation in thyroid physiology and thyroid cancer
Regulatory effect of succinylation on thyroid physiology
The main functions of the thyroid gland, which is the body’s largest endocrine organ, are to synthesize thyroid hormone, maintain the body’s metabolism and function, and promote the growth and development of infants and young children [80-82]. The protein precursor of thyroid hormone is thyroglobulin (TG) [83,84]. Thyroid hormone is synthesized through the coupling of iodine and tyrosine and is completed by TG proteolysis [85]. Iodinated TG, which is a macromolecule, is stored in the follicular cavity of the thyroid, and this process is accompanied by the activation and release of thyroid-stimulating hormone (TSH) [86].
Tarutani O and Dunn JT et al. [87,88] showed that sodium dodecyl sulfate (SDS) treatment can decompose the iodine protein obtained by the human body into three subunits (S-19, S-27 and S-12). A large amount of 19S, 27S or 12S thyroglobulin is closely related to the ability of tumors to accumulate radioactive iodine; therefore, TG is used as a potential biomarker for the diagnosis of TC [89]. In addition, some scholars have studied the dissociation effect of SDS and succinic anhydride on thyroid 27S iodide, and the results have shown that the succinyl dissociation mode of 27S iodide is basically the same as that of SDS treatment [87], and the succinylation can affect the dissociation of iodoprotein subunits [90,91]. Tanini A et al. [90] showed that after extensive succinylation, the 26000-Da peptide isolated from the 19S protein in the human thyroid gland can unfold and disrupt noncovalent bond interactions and thereby affects the synthesis of thyroxine [90]. In addition, previous studies have found that extensive succinylation can induce the cleavage of 12S thyroglobulin, increase the affinity of thyroglobulin for membrane receptors [92], and thereby affect the interaction between hormones on the thyroid membrane and their specific receptors [93]. In summary, the effects of the chemical modification of succinyl on the thyroid mainly include regulating the synthesis of thyroid hormones and regulating the affinity of thyroglobulin for membrane receptors, but the specific mechanisms of action remain unclear and need to be further explored (Figure 2).
Figure 2.

The pathological and physiological effects of succinylation on thyroid. This figure shows that under physiological conditions, the protein precursor thyroglobulin (TG) of thyroid hormone is mainly regulated by TSH. Iodinated TG can be decomposed into three subunits, 19S, 12S and 27S. After succinylation, it can promote the synthesis of thyroxine and affect the interaction of hormones and their specific receptors on the thyroid cell membrane. Under pathological conditions, the presence of SDH enzyme will down-regulate the accumulation of succinic acid in mitochondria, which can transmit “carcinogenic” signals from mitochondria to the cytoplasm and promote the occurrence of thyroid cancer.
Roles of succinylation in thyroid cancer
Studies have shown that TC may be caused by abnormal TSH secretion caused by TG mutation. Moreover, the TG levels after thyroidectomy are significantly related to the prognosis of papillary thyroid carcinoma and follicular carcinoma and can predict tumor recurrence and metastasis [86]. Therefore, the factors that affect TG changes (such as succinylation) deserve our attention.
Previous studies have shown that SDH, also known as mitochondrial complex II (the SDH enzyme, also known as mitochondrial complex II), is a heterotetrameric protein composed of four subunits, namely, SDHA, SDHB, SDHC and SDHD, and participates in the electron transport chain and the tricarboxylic acid cycle [94,95]. In the TCA cycle, the biallelic loss of genes encoding the subunits of the SDH complex can lead to succinate and succinyl-CoA accumulation [96]. Therefore, SDH participates in the regulation process of succinylation. Succinic acid is a metabolite of the TCA cycle, and its accumulation is downregulated due to the presence of SDH and transmits “carcinogenic” signals from the mitochondria to the cytoplasm. Once in the cytoplasm, succinic acid inhibits HIF-α prolyl hydroxylase (PHD) and thereby stabilizes HIF-1α under normoxic conditions [97]. Thus, succinate can increase the expression of genes that facilitate angiogenesis, metastasis, and glycolysis, which ultimately leads to tumor progression.
CS is a Madelina autosomal dominant genetic disease, and patients with CS are prone to BC, TC and other cancers [98]. TC is one of the main components of CS, which is usually related to a germline mutation of PTEN. Germline variants of different subunit genes encoding SDH (SDHBB-D) are found in 10% of PTEN mutation-negative CS/CS-like (CSL) individuals [99,100]. Notably, in 2012, some scholars noted that SDHB-D mutations coexist with germline PTEN mutations, including PTEN (mut+) patients or PTEN and (mut+)/SDH (var+) carriers, in approximately 6-8% of CS/CSL individuals. In addition, individuals with SDH (var+) alone exhibit a higher prevalence of TC [99]. It has been established that germline SDHD variants and somatic SDHD/B changes are associated with hereditary and sporadic TC, respectively [101-103].
The variants G12S and H50R are both located in the signal peptide of the SDHD preprotein. Previous studies have found that these two variants (G12S and H50R) may cause mislocalization of the SDH complex in the mitochondrial membrane or change the cleavage site of the signal peptide [104]. In papillary thyroid carcinoma and follicular carcinoma cell lines, SDHD-G12S and SDHD-H50R change the subcellular localization of PTEN and impair the function of PTEN, which results in inhibition of the apoptosis of TC cells and promotion of the migration of TC cells [105]. In addition, during the process of mitochondrial metabolism, the succinic acid that accumulates from the dysfunctional SDH complex can not only act as a second messenger to activate HIF signals but also drive the generation of intracellular ROS. The additional ROS stress accompanied by SDHx mutations can inhibit the apoptosis of TC cells and promote tumorigenesis [97,105,106], and the HIF signaling pathway can also be regulated by the AKT and mTOR signaling pathways downstream of PTEN to promote tumorigenesis [107]. In addition, NQO1 is a protein containing FAD, and the presence of excessive FAD can lead to activation of the AKT and MAPK signal transduction pathways [108]. In SDHx mutant cells, the combination of NQO1 and p53 leads to loss of NQO1 function, which leads to a reduction of p53 protein [99], and this reduction affects the occurrence of tumors. In summary, in CS/CSL tumorigenesis, the occurrence of TC may occur through the regulation of HIF1α, p53 and PTEN signal transduction by mitochondrial metabolism. The interaction between SDH and PTEN can lead to the activation of a variety of tumor-mediated signaling pathways (such as AKT, mTOR, and MAPK) and thereby promotes the occurrence of TC.
In contrast, in 2016, ED Accordi et al. conducted a study of a large Brazilian family and found that our common papillary thyroid carcinoma (PTC) is not associated with SDHx mutations [109], but because the author only studied one family, we believe that the insufficient sample size may have affected the final result. In 2017, further research conducted by W. Yu et al. revealed that SDHD-G12S and SDHD-H50R mutations can also increase the nuclear phosphorylation of AKT, which in turn affects the phosphorylation of the subsequent transcription factor FOXO3a (the main regulator of autophagy). This phosphorylation leads to the downregulation of autophagy [110], which suggests that SDHD plays a role in the pathogenesis of differentiated TC related to autophagy. The latest research has shown that missense mutations in the SDH complex are also considered the cause of differentiated TC. In addition, Compared with the more common multifocal paraganglioma found in carriers of SDHD mutation (PGL-1), carriers of SDHB mutation (PGL-4) are more likely to develop malignant diseases and extrahepatic tumors, including renal cell carcinoma and TC [111,112]. Therefore, the correlation between various types of thyroid and SDHx mutations remains controversial and needs to be further explored by researchers.
In addition, in 2017, Lai X [113] et al. found that succinyl-CoA ligase [GDP-forming] subunit beta is a protein biomarker that can be used for the diagnosis of thyroid follicular carcinoma and for distinguishing follicular carcinoma from follicular adenoma. Among the intermediate metabolic enzymes, isocitrate dehydrogenase (IDH1/2) mutations have been found in a variety of cancers, including TC [114], and these newly discovered carcinogenic metabolic lesions may be new anticancer treatments of selective targets. For example, in mice, by downregulating the mitogen-activated protein kinase (AMPK) pathway and inhibiting epithelial-mesenchymal transition, the SRC inhibitor SKI-606 (bosutinib) can significantly prevent the dedifferentiation, vascular invasion and lung metastasis of TC cells [115]. Therefore, oral SKI-606 can be considered for the clinical treatment of patients with refractory TC.
In summary, in TC, succinyl-CoA ligase [GDP-forming] subunit β can be used as a biodiagnostic marker for follicular TC, whereas SDH subunit mutations are related to various types of TC. Therefore, we speculate that each mutant protein subunit of SDH may be a biomarker for distinguishing different types of TC. However, relatively few studies have investigated the succinylation of goiter, and further research is needed (Figure 2).
Role of succinylation in the physiology of the mammary gland and breast cancer
Regulatory effect of succinylation on mammary gland physiology
The mammary gland is a symbolic organ of mammals, and its development mainly occurs after birth and consists of three consecutive stages: puberty, pregnancy and lactation. The main function of this gland is the production of milk [116,117]. The macronutrients and biologically active ingredients found in milk play an important role in the nutrition of breastfed infants and dairy products. These ingredients include some physiologically related compounds, such as vitamins, peptides, neuroactive compounds and hormones [118]. Among the components, succinyl-CoA and acetyl-CoA, among others, will increase linearly with increases in the pantothenic acid (RPP) supplement dose [119].
As early as 1958, researchers found that the activities of succinyl oxidase and cytochrome oxidase are increased in the mammary gland tissue of lactating rats, and both activities exhibited similar activity patterns: the activities gradually increase during pregnancy and childbirth and enter the stable phase during lactation, and this phase is followed by a significant increase in the activity of succinyl oxidase [120,121]. Based on these findings, succinylation may have a certain regulatory effect during pregnancy, childbirth and lactation in mammals. In addition, the proteins in cow’s milk (particularly casein) are widely regarded as a good carrier for the delivery of various biologically active compounds (including minerals). Succinylation is one of the most common chemical modification techniques for enhancing the mineral binding capacity of casein. The addition of minerals to a succinylated protein may change the physical, chemical and biochemical properties of the protein [122]. PH-adjusted succinylation can improve the storage properties of emulsions by increasing the surface charge density and amphiphilic balance. Therefore, succinylation has great potential to adjust the relationship between the structure and properties of protein-based products [123]. In addition, in milk, the prepared succinylated milk proteins can be used for preparation of the milk protein-vitamin A (Vit A) complex, and consumption of this type of milk can prevent the occurrence of Vit A deficiency [124]. In summary, the succinylation may play an important regulatory role in milk secretion and mammary physiology (Figure 3).
Figure 3.

The pathological and physiological effects of succinylation on breast. The figure shows that under physiological conditions, the mammary, as an organ for secretion of milk, is regulated by succinylation during pregnancy, childbirth and lactation. Moreover, succinyl-CoA in milk can be modified by succinylation to enhance the binding capacity of casein minerals. Under pathological conditions, succinylation promotes the occurrence of breast cancer by stimulating oxidative stress, inhibiting DNA damage repair, and promoting the proliferation and autophagy of breast cancer cells.
Pathological functions of succinylation in breast cancer
At present, with the rapid development of proteomics technology, BC proteomics and PTM research, including phosphorylation, acetylation, methylation, and succinylation, have gradually become a research hotspot. Liu C et al. studied the effect of lysine succinylation on BC for the first time. These researchers found multiple succinylation sites in BC and discovered a new mechanism, namely, the pentose phosphate pathway (PPP) and the endoplasmic reticulum protein processing pathway can be regulated by lysine succinylation modification of its core enzyme [125]. Previous studies have shown that lysine succinylation is involved in energy metabolism, and the PPP is the main pathway of glucose catabolism. It is known that the PPP is significantly regulated in cancer cells, and it has been determined that changes in the succinylation levels in this pathway follow a specific pattern. The inhibition of aerobic respiration enhances the glycolytic pathway. To meet the rapid growth of cancer cells, the PPP is activated, and the nicotinamide adenine dinucleotide (NADPH) content is increased [126]. Most of the related reactions in this process are reversible, which indicates that the PPP balance can be controlled by succinylation. In advanced cancers, the PPP proteins show a significant increase in succinylation, which indicates that the activation of lysine succinylation may be the main cause of changes in protein expression. In addition, the TCA cycle produces and consumes succinic acid (the intermediate substrate for succinylation of lysine), whereas the PPP uses the intermediate component of glycolysis to produce NADPH (a reducing substrate), which is essential for oxidative stress [125-127]. Therefore, it has been speculated that during the regulation of glucose metabolism activated by breast tumors, the balance between the production and consumption of succinic acid can lead to the complex regulation of succinylation and protein expression, which in turn affects the occurrence and development of BC.
Gao X et al. [128] systematically studied the succinylation and acetylation modification of all proteins in invasive ductal carcinoma using proteomics technology and found that the modification level of most proteins in BC tissue was significantly higher than that in normal paracancerous tissues. In addition, a bioinformatics analysis revealed that highly succinylated proteins are significantly enriched in histone H2A.X complex, and nucleophospholipid 1 (NPM1) may be a key determinant [128]. Yu, et al., showed that the H2A.X complex can form dynamic lesions at DNA break sites under different DNA damage conditions [129]. Damage to DNA activates H2A.X, and activated H2A.X recruits many other proteins to form protein foci for mediating DNA damage repair [130]. This process can be regulated by protein modification [131-133]. Therefore, the hyperacetylation and succinylation of proteins in the H2A.X complex may affect DNA damage repair by regulating the formation of protein foci in BC [128].
In addition, L. Polletta et al. found that the immunoprecipitation of SIRT5 (lysine desuccinase) and glutaminase and the inhibition of SIRT5 can lead to an increase in the succinylation of glutaminase and promote BC [134,135]. Glutamine, which is the most abundant nonessential amino acid, can be used as a stored form of glutamate and ammonia and is important for the tricarboxylic acid cycle [136]. Therefore, in tumor cells that use glucose mainly through glycolysis, the metabolism of glutamine is accelerated, and glutamine represents an important source of energy [137-139]. According to previous studies, SIRT5 can play a cancer-promoting effect in BC through the desuccinylation substrate glutaminase (GLS) [135], and SIRT5 can regulate ammonia by regulating the metabolism of glutamine in BC cells. The production and ammonia-induced autophagy [134] indicate that the metabolism of ammonia in BC is related to the regulation of succinylation. In addition, the latest study found that SIRT5 is highly expressed in human BC and is associated with a poor prognosis [140,141]. It is worth noting that SIRT5 can be also used as a biomarker for the response of triple-negative BC to anthracycline taxane neoadjuvant chemotherapy [141]. In addition, Fu XL et al. identified the expression level of succinylated proteins. The succinic acid in the endoplasmic reticulum (ER) pathway was significantly increased, which indicated that succinylation may occur in ER proteins, and the abnormal expression of ER-related proteins may be destroyed in BC tissues [125,142]. Other studies include that CPT1A can promote the proliferation of BC cells by the succinylation of enolase 1 [48].
In summary, there are relatively few studies on succinylation in BC. However, current studies reveal that succinylation can be a cancer-promoting factor in BC: succinylation not only promotes BC cell proliferation but also affects the repair of DNA damage and its energy metabolism process through its regulators. Therefore, we can reasonably assume that the specific regulatory role of succinylation in BC will become a new research direction in targeted therapy for BC (Figure 3).
Summary and prospects
Succinylation participates in a variety of physiological and pathological processes, as well as involved in the occurrence and development of a variety of tumors. Despite extensive work has been performed to investigate the expression pattern, functional diversity, regulatory mechanism and pathophysiology of succinylation in the breast and thyroid organs, the systematic review is rare in this field. Therefore, this review focuses on basic characteristics and function of succinylation, regulatory factors of succinylation, and pathological role of succinylation in thyroid and breast cancer (Figures 1, 2 and 3). We hope that we can provide a basic, systemic and summarized knowledge to this field, advocating researchers play more attention on the pathophysiological role of succinylation in the development and progression of thyroid and breast cancer, which may provide novel targets for the clinical diagnosis and treatment of these diseases.
Acknowledgements
We thank Guorong Wen, Hai Jin, Jiaxing An and Jiaxing Zhu, who provided us with suggestions for the paper and support for daily experiments. This research was supported by the National Natural Science Foundation of China (82160505 to Taolang Li, 81860103 and 82070536 to Xuemei Liu and 82073087 to Biguang Tuo); Guizhou Province Science Plan Program (Qian Ke He Foundation-ZK [2021] General 461) (2021 to Taolang Li) and 15851 talent projects of Zunyi City (2018 to Taolang Li).
Disclosure of conflict of interest
None.
Abbreviations
- ACOX1
acyl-CoA oxidase 1
- α-KGDH
α-ketoglutarate dehydrogenase
- ATP
Adenosine triphosphate
- BC
Breast cancer
- BRCA
breast cancer
- CRC
Colorectal cancer
- CPT1A
Carnitine palmitoyltransferase 1A
- CS
Cowden syndrome
- ccRCC
clear cell renal cell carcinoma
- CHX
cycloheximide
- ER
Endoplasmic reticulum
- FAD
flavin adenine dinucleotide
- GBM
Glioblastoma
- GPCRs
G protein-coupled receptors
- GC
Gastric cancer
- GLS
glutaminase
- HAMs
Histone acetylation modifications
- HAT
Histone acetyltransferase
- HDAC
Histone deacetylase
- HCC
hepatocellular carcinoma
- ICDH
Isocitrate dehydrogenase
- KGDHC
Ketoglutarate dehydrogenase complex
- Ksucc
Lysine succinylation
- KAT2A/GCN5
Lysine acetyltransferase 2A
- KGDHC
α-ketoglutarate dehydrogenase complex
- KGDH
α-ketoglutarate dehydrogenase
- LC
lung cancer
- KDH
α-ketoglutarate diacid dehydrogenase
- NADPH
Nicotinamide adenine dinucleotide
- NPM1
Nucleophospholipid 1
- NAD
Nicotinamide adenine dinucleotide
- PDTC
Poorly differentiated thyroid carcinoma
- PTM
Posttranslational modification
- PGL-1
SDHD mutation carriers
- PGL-4
SDHB mutation carriers
- PPP
Pentose phosphate pathway
- PTEN
phosphatase and tensin homolog
- TC
Thyroid cancer
- RAI
Radioactive iodine therapy
- ROS-KGDHC
Ros-α-ketoglutarate dehydrogenase complex
- RPP
Pantothenic acid
- SUCNR1
Succinate receptor 1
- SCS
Succinyl-CoA synthetase
- SHMT2
Serine hydroxymethyltransferase
- SDH
Succinate dehydrogenase
- SIRT5
Silent information regulator 5
- SIRT7
Silent information regulator 7
- SDS
Sodium dodecyl sulfate
- SDHx
Succinate dehydrogenase genes
- SDHA
Succinate dehydrogenase A
- SDHB
Succinate dehydrogenase B
- SDHC
Succinate dehydrogenase C
- SDHD
Succinate dehydrogenase D
- TCA
Tricarboxylic acid cycle
- TG
Thyroglobulin
- TSH
Thyroid stimulating hormone
- Vit A
Vitamin A
References
- 1.Akram M, Iqbal M, Daniyal M, Khan AU. Awareness and current knowledge of breast cancer. Biol Res. 2017;50:33. doi: 10.1186/s40659-017-0140-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Pellegriti G, Frasca F, Regalbuto C, Squatrito S, Vigneri R. Worldwide increasing incidence of thyroid cancer: update on epidemiology and risk factors. J Cancer Epidemiol. 2013;2013:965212. doi: 10.1155/2013/965212. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Siegel RL, Miller KD, Jemal A. Cancer statistics, 2017. CA Cancer J Clin. 2017;67:7–30. doi: 10.3322/caac.21387. [DOI] [PubMed] [Google Scholar]
- 4.Anastasiadi Z, Lianos GD, Ignatiadou E, Harissis HV, Mitsis M. Breast cancer in young women: an overview. Updates Surg. 2017;69:313–317. doi: 10.1007/s13304-017-0424-1. [DOI] [PubMed] [Google Scholar]
- 5.McTiernan A, Weiss NS, Daling JR. Incidence of thyroid cancer in women in relation to known or suspected risk factors for breast cancer. Cancer Res. 1987;47:292–295. [PubMed] [Google Scholar]
- 6.Brown AP, Chen J, Hitchcock YJ, Szabo A, Shrieve DC, Tward JD. The risk of second primary malignancies up to three decades after the treatment of differentiated thyroid cancer. J Clin Endocrinol Metab. 2008;93:504–515. doi: 10.1210/jc.2007-1154. [DOI] [PubMed] [Google Scholar]
- 7.Subramanian S, Goldstein DP, Parlea L, Thabane L, Ezzat S, Ibrahim-Zada I, Straus S, Brierley JD, Tsang RW, Gafni A, Rotstein L, Sawka AM. Second primary malignancy risk in thyroid cancer survivors: a systematic review and meta-analysis. Thyroid. 2007;17:1277–1288. doi: 10.1089/thy.2007.0171. [DOI] [PubMed] [Google Scholar]
- 8.Tanaka H, Tsukuma H, Koyama H, Kinoshita Y, Kinoshita N, Oshima A. Second primary cancers following breast cancer in the Japanese female population. Jpn J Cancer Res. 2001;92:1–8. doi: 10.1111/j.1349-7006.2001.tb01040.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.An JH, Hwangbo Y, Ahn HY, Keam B, Lee KE, Han W, Park DJ, Park IA, Noh DY, Youn YK, Cho BY, Im SA, Park YJ. A possible association between thyroid cancer and breast cancer. Thyroid. 2015;25:1330–1338. doi: 10.1089/thy.2014.0561. [DOI] [PubMed] [Google Scholar]
- 10.Dong L, Lu J, Zhao B, Wang W, Zhao Y. Review of the possible association between thyroid and breast carcinoma. World J Surg Oncol. 2018;16:130. doi: 10.1186/s12957-018-1436-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Mirian C, Grønhøj C, Jensen DH, Jakobsen KK, Karnov K, Jensen JS, Hahn CH, Klitmøller TA, Bentzen J, von Buchwald C. Trends in thyroid cancer: retrospective analysis of incidence and survival in Denmark 1980-2014. Cancer Epidemiol. 2018;55:81–87. doi: 10.1016/j.canep.2018.05.009. [DOI] [PubMed] [Google Scholar]
- 12.Ancker OV, Krüger M, Wehland M, Infanger M, Grimm D. Multikinase inhibitor treatment in thyroid cancer. Int J Mol Sci. 2019;21:10. doi: 10.3390/ijms21010010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Kapiteijn E, Schneider TC, Morreau H, Gelderblom H, Nortier JWR, Smit JWA. New treatment modalities in advanced thyroid cancer. Ann Oncol. 2012;23:10–18. doi: 10.1093/annonc/mdr117. [DOI] [PubMed] [Google Scholar]
- 14.Cabanillas ME, McFadden DG, Durante C. Thyroid cancer. Lancet. 2016;388:2783–2795. doi: 10.1016/S0140-6736(16)30172-6. [DOI] [PubMed] [Google Scholar]
- 15.Volante M, Collini P, Nikiforov YE, Sakamoto A, Kakudo K, Katoh R, Lloyd RV, LiVolsi VA, Papotti M, Sobrinho-Simoes M, Bussolati G, Rosai J. Poorly differentiated thyroid carcinoma: the Turin proposal for the use of uniform diagnostic criteria and an algorithmic diagnostic approach. Am J Surg Pathol. 2007;31:1256–1264. doi: 10.1097/PAS.0b013e3180309e6a. [DOI] [PubMed] [Google Scholar]
- 16.Winters S, Martin C, Murphy D, Shokar NK. Breast cancer epidemiology, prevention, and screening. Prog Mol Biol Transl Sci. 2017;151:1–32. doi: 10.1016/bs.pmbts.2017.07.002. [DOI] [PubMed] [Google Scholar]
- 17.Harbeck N, Gnant M. Breast cancer. Lancet. 2017;389:1134–1150. doi: 10.1016/S0140-6736(16)31891-8. [DOI] [PubMed] [Google Scholar]
- 18.Yeo SK, Guan JL. Breast cancer: multiple subtypes within a tumor? Trends Cancer. 2017;3:753–760. doi: 10.1016/j.trecan.2017.09.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Fahad Ullah M. Breast cancer: current perspectives on the disease status. Adv Exp Med Biol. 2019;1152:51–64. doi: 10.1007/978-3-030-20301-6_4. [DOI] [PubMed] [Google Scholar]
- 20.Wang R, Wang G. Protein modification and autophagy activation. Adv Exp Med Biol. 2019;1206:237–259. doi: 10.1007/978-981-15-0602-4_12. [DOI] [PubMed] [Google Scholar]
- 21.Alleyn M, Breitzig M, Lockey R, Kolliputi N. The dawn of succinylation: a posttranslational modification. Am J Physiol Cell Physiol. 2018;314:C228–C232. doi: 10.1152/ajpcell.00148.2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Zhao S, Zhang X, Li H. Beyond histone acetylation-writing and erasing histone acylations. Curr Opin Struct Biol. 2018;53:169–177. doi: 10.1016/j.sbi.2018.10.001. [DOI] [PubMed] [Google Scholar]
- 23.Kouzarides T. Chromatin modifications and their function. Cell. 2007;128:693–705. doi: 10.1016/j.cell.2007.02.005. [DOI] [PubMed] [Google Scholar]
- 24.Tan M, Luo H, Lee S, Jin F, Yang JS, Montellier E, Buchou T, Cheng Z, Rousseaux S, Rajagopal N, Lu Z, Ye Z, Zhu Q, Wysocka J, Ye Y, Khochbin S, Ren B, Zhao Y. Identification of 67 histone marks and histone lysine crotonylation as a new type of histone modification. Cell. 2011;146:1016–1028. doi: 10.1016/j.cell.2011.08.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Zhu H, Wang G, Qian J. Transcription factors as readers and effectors of DNA methylation. Nat Rev Genet. 2016;17:551–565. doi: 10.1038/nrg.2016.83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Traube FR, Carell T. The chemistries and consequences of DNA and RNA methylation and demethylation. RNA Biol. 2017;14:1099–1107. doi: 10.1080/15476286.2017.1318241. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Ginder GD, Williams DC Jr. Readers of DNA methylation, the MBD family as potential therapeutic targets. Pharmacol Ther. 2018;184:98–111. doi: 10.1016/j.pharmthera.2017.11.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Guo P, Chen W, Li H, Li M, Li L. The histone acetylation modifications of breast cancer and their therapeutic implications. Pathol Oncol Res. 2018;24:807–813. doi: 10.1007/s12253-018-0433-5. [DOI] [PubMed] [Google Scholar]
- 29.Valvo V, Iesato A, Kavanagh TR, Priolo C, Zsengeller Z, Pontecorvi A, Stillman IE, Burke SD, Liu X, Nucera C. Fine-tuning lipid metabolism by targeting mitochondria-associated Acetyl-CoA-Carboxylase 2 in BRAF(V600E) papillary thyroid carcinoma. Thyroid. 2021;31:1335–1358. doi: 10.1089/thy.2020.0311. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Ni W, Yao S, Zhou Y, Liu Y, Huang P, Zhou A, Liu J, Che L, Li J. Long noncoding RNA GAS5 inhibits progression of colorectal cancer by interacting with and triggering YAP phosphorylation and degradation and is negatively regulated by the m6A reader YTHDF3. Mol Cancer. 2019;18:143. doi: 10.1186/s12943-019-1079-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Huang T, Kim CK, Alvarez AA, Pangeni RP, Wan X, Song X, Shi T, Yang Y, Sastry N, Horbinski CM, Lu S, Stupp R, Kessler JA, Nishikawa R, Nakano I, Sulman EP, Lu X, James CD, Yin XM, Hu B, Cheng SY. MST4 phosphorylation of ATG4B regulates autophagic activity, tumorigenicity, and radioresistance in glioblastoma. Cancer Cell. 2017;32:840–855. e848. doi: 10.1016/j.ccell.2017.11.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Malaney P, Palumbo E, Semidey-Hurtado J, Hardee J, Stanford K, Kathiriya JJ, Patel D, Tian Z, Allen-Gipson D, Davé V. PTEN physically interacts with and regulates E2F1-mediated transcription in lung cancer. Cell Cycle. 2018;17:947–962. doi: 10.1080/15384101.2017.1388970. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Xie J, Li Y, Jiang K, Hu K, Zhang S, Dong X, Dai X, Liu L, Zhang T, Yang K, Huang K, Chen J, Shi S, Zhang Y, Wu G, Xu S. CDK16 phosphorylates and degrades p53 to promote radioresistance and predicts prognosis in lung cancer. Theranostics. 2018;8:650–662. doi: 10.7150/thno.21963. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Li Z, Hou P, Fan D, Dong M, Ma M, Li H, Yao R, Li Y, Wang G, Geng P, Mihretab A, Liu D, Zhang Y, Huang B, Lu J. The degradation of EZH2 mediated by lncRNA ANCR attenuated the invasion and metastasis of breast cancer. Cell Death Differ. 2017;24:59–71. doi: 10.1038/cdd.2016.95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Yuan J, Yang Y, Gao Z, Wang Z, Ji W, Song W, Zhang F, Niu R. Tyr23 phosphorylation of Anxa2 enhances STAT3 activation and promotes proliferation and invasion of breast cancer cells. Breast Cancer Res Treat. 2017;164:327–340. doi: 10.1007/s10549-017-4271-z. [DOI] [PubMed] [Google Scholar]
- 36.Xie Z, Dai J, Dai L, Tan M, Cheng Z, Wu Y, Boeke JD, Zhao Y. Lysine succinylation and lysine malonylation in histones. Mol Cell Proteomics. 2012;11:100–107. doi: 10.1074/mcp.M111.015875. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Zhang Z, Tan M, Xie Z, Dai L, Chen Y, Zhao Y. Identification of lysine succinylation as a new post-translational modification. Nat Chem Biol. 2011;7:58–63. doi: 10.1038/nchembio.495. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Yang Y, Gibson GE. Succinylation links metabolism to protein functions. Neurochem Res. 2019;44:2346–2359. doi: 10.1007/s11064-019-02780-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Wagner GR, Payne RM. Widespread and enzyme-independent Nε-acetylation and Nε-succinylation of proteins in the chemical conditions of the mitochondrial matrix. J Biol Chem. 2013;288:29036–29045. doi: 10.1074/jbc.M113.486753. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Park J, Chen Y, Tishkoff DX, Peng C, Tan M, Dai L, Xie Z, Zhang Y, Zwaans BM, Skinner ME, Lombard DB, Zhao Y. SIRT5-mediated lysine desuccinylation impacts diverse metabolic pathways. Mol Cell. 2013;50:919–930. doi: 10.1016/j.molcel.2013.06.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Liu J, Qian C, Cao X. Post-translational modification control of innate immunity. Immunity. 2016;45:15–30. doi: 10.1016/j.immuni.2016.06.020. [DOI] [PubMed] [Google Scholar]
- 42.Hasan MM, Khatun MS, Kurata H. Large-scale assessment of bioinformatics tools for lysine succinylation sites. Cells. 2019;8:95. doi: 10.3390/cells8020095. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Zhang Y, Bharathi SS, Rardin MJ, Lu J, Maringer KV, Sims-Lucas S, Prochownik EV, Gibson BW, Goetzman ES. Lysine desuccinylase SIRT5 binds to cardiolipin and regulates the electron transport chain. J Biol Chem. 2017;292:10239–10249. doi: 10.1074/jbc.M117.785022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Wang X, Chen X, Li J, Zhou X, Liu Y, Zhong L, Tang Y, Zheng H, Liu J, Zhan R, Chen L. Global analysis of lysine succinylation in patchouli plant leaves. Hortic Res. 2019;6:133. doi: 10.1038/s41438-019-0216-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Chen XF, Tian MX, Sun RQ, Zhang ML, Zhou LS, Jin L, Chen LL, Zhou WJ, Duan KL, Chen YJ, Gao C, Cheng ZL, Wang F, Zhang JY, Sun YP, Yu HX, Zhao YZ, Yang Y, Liu WR, Shi YH, Xiong Y, Guan KL, Ye D. SIRT5 inhibits peroxisomal ACOX1 to prevent oxidative damage and is downregulated in liver cancer. EMBO Rep. 2018;19:e45124. doi: 10.15252/embr.201745124. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Wang C, Zhang C, Li X, Shen J, Xu Y, Shi H, Mu X, Pan J, Zhao T, Li M, Geng B, Xu C, Wen H, You Q. CPT1A-mediated succinylation of S100A10 increases human gastric cancer invasion. J Cell Mol Med. 2019;23:293–305. doi: 10.1111/jcmm.13920. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Bringman-Rodenbarger LR, Guo AH, Lyssiotis CA, Lombard DB. Emerging roles for SIRT5 in metabolism and cancer. Antioxid Redox Signal. 2018;28:677–690. doi: 10.1089/ars.2017.7264. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Kurmi K, Hitosugi S, Wiese EK, Boakye-Agyeman F, Gonsalves WI, Lou Z, Karnitz LM, Goetz MP, Hitosugi T. Carnitine palmitoyltransferase 1A has a lysine succinyltransferase activity. Cell Rep. 2018;22:1365–1373. doi: 10.1016/j.celrep.2018.01.030. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Song Y, Wang J, Cheng Z, Gao P, Sun J, Chen X, Chen C, Wang Y, Wang Z. Quantitative global proteome and lysine succinylome analyses provide insights into metabolic regulation and lymph node metastasis in gastric cancer. Sci Rep. 2017;7:42053. doi: 10.1038/srep42053. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Tong Y, Guo D, Yan D, Ma C, Shao F, Wang Y, Luo S, Lin L, Tao J, Jiang Y, Lu Z, Xing D. KAT2A succinyltransferase activity-mediated 14-3-3ζ upregulation promotes β-catenin stabilization-dependent glycolysis and proliferation of pancreatic carcinoma cells. Cancer Lett. 2020;469:1–10. doi: 10.1016/j.canlet.2019.09.015. [DOI] [PubMed] [Google Scholar]
- 51.Wang Y, Guo YR, Liu K, Yin Z, Liu R, Xia Y, Tan L, Yang P, Lee JH, Li XJ, Hawke D, Zheng Y, Qian X, Lyu J, He J, Xing D, Tao YJ, Lu Z. KAT2A coupled with the α-KGDH complex acts as a histone H3 succinyltransferase. Nature. 2017;552:273–277. doi: 10.1038/nature25003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Lu W, Che X, Qu X, Zheng C, Yang X, Bao B, Li Z, Wang D, Jin Y, Wang Y, Xiao J, Qi J, Liu Y. Succinylation regulators promote clear cell renal cell carcinoma by immune regulation and RNA N6-methyladenosine methylation. Front Cell Dev Biol. 2021;9:622198. doi: 10.3389/fcell.2021.622198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Lin ZF, Xu HB, Wang JY, Lin Q, Ruan Z, Liu FB, Jin W, Huang HH, Chen X. SIRT5 desuccinylates and activates SOD1 to eliminate ROS. Biochem Biophys Res Commun. 2013;441:191–195. doi: 10.1016/j.bbrc.2013.10.033. [DOI] [PubMed] [Google Scholar]
- 54.Yang X, Wang Z, Li X, Liu B, Liu M, Liu L, Chen S, Ren M, Wang Y, Yu M, Wang B, Zou J, Zhu WG, Yin Y, Gu W, Luo J. SHMT2 desuccinylation by SIRT5 drives cancer cell proliferation. Cancer Res. 2018;78:372–386. doi: 10.1158/0008-5472.CAN-17-1912. [DOI] [PubMed] [Google Scholar]
- 55.Peng C, Lu Z, Xie Z, Cheng Z, Chen Y, Tan M, Luo H, Zhang Y, He W, Yang K, Zwaans BM, Tishkoff D, Ho L, Lombard D, He TC, Dai J, Verdin E, Ye Y, Zhao Y. The first identification of lysine malonylation substrates and its regulatory enzyme. Mol Cell Proteomics. 2011;10 doi: 10.1074/mcp.M111.012658. M111.012658. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Sabari BR, Zhang D, Allis CD, Zhao Y. Metabolic regulation of gene expression through histone acylations. Nat Rev Mol Cell Biol. 2017;18:90–101. doi: 10.1038/nrm.2016.140. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Gibson GE, Xu H, Chen HL, Chen W, Denton TT, Zhang S. Alpha-ketoglutarate dehydrogenase complex-dependent succinylation of proteins in neurons and neuronal cell lines. J Neurochem. 2015;134:86–96. doi: 10.1111/jnc.13096. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Weinert BT, Schölz C, Wagner SA, Iesmantavicius V, Su D, Daniel JA, Choudhary C. Lysine succinylation is a frequently occurring modification in prokaryotes and eukaryotes and extensively overlaps with acetylation. Cell Rep. 2013;4:842–851. doi: 10.1016/j.celrep.2013.07.024. [DOI] [PubMed] [Google Scholar]
- 59.Papanicolaou KN, O’Rourke B, Foster DB. Metabolism leaves its mark on the powerhouse: recent progress in post-translational modifications of lysine in mitochondria. Front Physiol. 2014;5:301. doi: 10.3389/fphys.2014.00301. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Rosen R, Becher D, Büttner K, Biran D, Hecker M, Ron EZ. Probing the active site of homoserine trans-succinylase. FEBS Lett. 2004;577:386–392. doi: 10.1016/j.febslet.2004.10.037. [DOI] [PubMed] [Google Scholar]
- 61.Zhang X, Cheng K, Ning Z, Mayne J, Walker K, Chi H, Farnsworth CL, Lee K, Figeys D. Exploring the microbiome-wide lysine acetylation, succinylation, and propionylation in human gut microbiota. Anal Chem. 2021;93:6594–6598. doi: 10.1021/acs.analchem.1c00962. [DOI] [PubMed] [Google Scholar]
- 62.Chen H, Xu H, Potash S, Starkov A, Belousov VV, Bilan DS, Denton TT, Gibson GE. Mild metabolic perturbations alter succinylation of mitochondrial proteins. J Neurosci Res. 2017;95:2244–2252. doi: 10.1002/jnr.24103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Wang G, Meyer JG, Cai W, Softic S, Li ME, Verdin E, Newgard C, Schilling B, Kahn CR. Regulation of UCP1 and mitochondrial metabolism in brown adipose tissue by reversible succinylation. Mol Cell. 2019;74:844–857. e847. doi: 10.1016/j.molcel.2019.03.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Mills E, O’Neill LA. Succinate: a metabolic signal in inflammation. Trends Cell Biol. 2014;24:313–320. doi: 10.1016/j.tcb.2013.11.008. [DOI] [PubMed] [Google Scholar]
- 65.Cho EH. Succinate as a regulator of hepatic stellate cells in liver fibrosis. Front Endocrinol (Lausanne) 2018;9:455. doi: 10.3389/fendo.2018.00455. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Rubic T, Lametschwandtner G, Jost S, Hinteregger S, Kund J, Carballido-Perrig N, Schwärzler C, Junt T, Voshol H, Meingassner JG, Mao X, Werner G, Rot A, Carballido JM. Triggering the succinate receptor GPR91 on dendritic cells enhances immunity. Nat Immunol. 2008;9:1261–1269. doi: 10.1038/ni.1657. [DOI] [PubMed] [Google Scholar]
- 67.Connors J, Dawe N, Van Limbergen J. The role of succinate in the regulation of intestinal inflammation. Nutrients. 2018;11:25. doi: 10.3390/nu11010025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Choi S, Pfleger J, Jeon YH, Yang Z, He M, Shin H, Sayed D, Astrof S, Abdellatif M. Oxoglutarate dehydrogenase and acetyl-CoA acyltransferase 2 selectively associate with H2A. Z-occupied promoters and are required for histone modifications. Biochim Biophys Acta Gene Regul Mech. 2019;1862:194436. doi: 10.1016/j.bbagrm.2019.194436. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Wagner GR, Bhatt DP, O’Connell TM, Thompson JW, Dubois LG, Backos DS, Yang H, Mitchell GA, Ilkayeva OR, Stevens RD, Grimsrud PA, Hirschey MD. A class of reactive Acyl-CoA species reveals the non-enzymatic origins of protein acylation. Cell Metab. 2017;25:823–837. e828. doi: 10.1016/j.cmet.2017.03.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Simithy J, Sidoli S, Yuan ZF, Coradin M, Bhanu NV, Marchione DM, Klein BJ, Bazilevsky GA, McCullough CE, Magin RS, Kutateladze TG, Snyder NW, Marmorstein R, Garcia BA. Characterization of histone acylations links chromatin modifications with metabolism. Nat Commun. 2017;8:1141. doi: 10.1038/s41467-017-01384-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Philp A, Rowland T, Perez-Schindler J, Schenk S. Understanding the acetylome: translating targeted proteomics into meaningful physiology. Am J Physiol Cell Physiol. 2014;307:C763–773. doi: 10.1152/ajpcell.00399.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Nishida Y, Rardin MJ, Carrico C, He W, Sahu AK, Gut P, Najjar R, Fitch M, Hellerstein M, Gibson BW, Verdin E. SIRT5 regulates both cytosolic and mitochondrial protein malonylation with glycolysis as a major target. Mol Cell. 2015;59:321–332. doi: 10.1016/j.molcel.2015.05.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Frank RA, Price AJ, Northrop FD, Perham RN, Luisi BF. Crystal structure of the E1 component of the Escherichia coli 2-oxoglutarate dehydrogenase multienzyme complex. J Mol Biol. 2007;368:639–651. doi: 10.1016/j.jmb.2007.01.080. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Basu SS, Blair IA. Rotenone-mediated changes in intracellular coenzyme A thioester levels: implications for mitochondrial dysfunction. Chem Res Toxicol. 2011;24:1630–1632. doi: 10.1021/tx200366j. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Bechara EJ, Dutra F, Cardoso VE, Sartori A, Olympio KP, Penatti CA, Adhikari A, Assunção NA. The dual face of endogenous alpha-aminoketones: pro-oxidizing metabolic weapons. Comp Biochem Physiol C Toxicol Pharmacol. 2007;146:88–110. doi: 10.1016/j.cbpc.2006.07.004. [DOI] [PubMed] [Google Scholar]
- 76.Burch JS, Marcero JR, Maschek JA, Cox JE, Jackson LK, Medlock AE, Phillips JD, Dailey HA Jr. Glutamine via α-ketoglutarate dehydrogenase provides succinyl-CoA for heme synthesis during erythropoiesis. Blood. 2018;132:987–998. doi: 10.1182/blood-2018-01-829036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Masson N, Ratcliffe PJ. Hypoxia signaling pathways in cancer metabolism: the importance of co-selecting interconnected physiological pathways. Cancer Metab. 2014;2:3. doi: 10.1186/2049-3002-2-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Li L, Shi L, Yang S, Yan R, Zhang D, Yang J, He L, Li W, Yi X, Sun L, Liang J, Cheng Z, Shi L, Shang Y, Yu W. SIRT7 is a histone desuccinylase that functionally links to chromatin compaction and genome stability. Nat Commun. 2016;7:12235. doi: 10.1038/ncomms12235. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Rardin MJ, He W, Nishida Y, Newman JC, Carrico C, Danielson SR, Guo A, Gut P, Sahu AK, Li B, Uppala R, Fitch M, Riiff T, Zhu L, Zhou J, Mulhern D, Stevens RD, Ilkayeva OR, Newgard CB, Jacobson MP, Hellerstein M, Goetzman ES, Gibson BW, Verdin E. SIRT5 regulates the mitochondrial lysine succinylome and metabolic networks. Cell Metab. 2013;18:920–933. doi: 10.1016/j.cmet.2013.11.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Ortiga-Carvalho TM, Chiamolera MI, Pazos-Moura CC, Wondisford FE. Hypothalamus-pituitary-thyroid axis. Compr Physiol. 2016;6:1387–1428. doi: 10.1002/cphy.c150027. [DOI] [PubMed] [Google Scholar]
- 81.Sterrett M. Maternal and fetal thyroid physiology. Clin Obstet Gynecol. 2019;62:302–307. doi: 10.1097/GRF.0000000000000439. [DOI] [PubMed] [Google Scholar]
- 82.Mondal S, Raja K, Schweizer U, Mugesh G. Chemistry and biology in the biosynthesis and action of thyroid hormones. Angew Chem Int Ed Engl. 2016;55:7606–7630. doi: 10.1002/anie.201601116. [DOI] [PubMed] [Google Scholar]
- 83.Di Jeso B, Arvan P. Thyroglobulin from molecular and cellular biology to clinical endocrinology. Endocr Rev. 2016;37:2–36. doi: 10.1210/er.2015-1090. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Citterio CE, Targovnik HM, Arvan P. The role of thyroglobulin in thyroid hormonogenesis. Nat Rev Endocrinol. 2019;15:323–338. doi: 10.1038/s41574-019-0184-8. [DOI] [PubMed] [Google Scholar]
- 85.Carvalho DP, Dupuy C. Thyroid hormone biosynthesis and release. Mol Cell Endocrinol. 2017;458:6–15. doi: 10.1016/j.mce.2017.01.038. [DOI] [PubMed] [Google Scholar]
- 86.Lin JD. Thyroglobulin and human thyroid cancer. Clin Chim Acta. 2008;388:15–21. doi: 10.1016/j.cca.2007.11.002. [DOI] [PubMed] [Google Scholar]
- 87.Tarutani O, Kondo T, Smith DJ, Shulman S. Subunit structure of 27 S thyroid iodoprotein. Endocrinol Jpn. 1977;23:305–311. [PubMed] [Google Scholar]
- 88.Dunn JT, Ray SC. Variations in the structure of thyroglobulins from normal and goitrous human thyroids. J Clin Endocrinol Metab. 1978;47:861–869. doi: 10.1210/jcem-47-4-861. [DOI] [PubMed] [Google Scholar]
- 89.Valenta LJ, Michel-Béchet M. Ultrastructure and biochemistry of thyroid carcinoma. Cancer. 1977;40:284–300. doi: 10.1002/1097-0142(197707)40:1<284::aid-cncr2820400141>3.0.co;2-c. [DOI] [PubMed] [Google Scholar]
- 90.Tanini A, Shifrin S. Noncovalent interactions of a 26,000-dalton peptide with 19 S human thyroglobulin. J Biol Chem. 1983;258:12553–12557. [PubMed] [Google Scholar]
- 91.Dunn JT, Kim PS, Dunn AD. Favored sites for thyroid hormone formation on the peptide chains of human thyroglobulin. J Biol Chem. 1982;257:88–94. [PubMed] [Google Scholar]
- 92.Shifrin S, Kohn LD. Binding of thyroglobulin to bovine thyroid membranes. Role of specific amino acids in receptor recognition. J Biol Chem. 1981;256:10600–10605. [PubMed] [Google Scholar]
- 93.Consiglio E, Salvatore G, Rall JE, Kohn LD. Thyroglobulin interactions with thyroid plasma membranes. The existence of specific receptors and their potential role. J Biol Chem. 1979;254:5065–5076. [PubMed] [Google Scholar]
- 94.Gill AJ. Succinate dehydrogenase (SDH) and mitochondrial driven neoplasia. Pathology. 2012;44:285–292. doi: 10.1097/PAT.0b013e3283539932. [DOI] [PubMed] [Google Scholar]
- 95.Eng C, Kiuru M, Fernandez MJ, Aaltonen LA. A role for mitochondrial enzymes in inherited neoplasia and beyond. Nat Rev Cancer. 2003;3:193–202. doi: 10.1038/nrc1013. [DOI] [PubMed] [Google Scholar]
- 96.Smestad J, Erber L, Chen Y, Maher LJ 3rd. Chromatin succinylation correlates with active gene expression and is perturbed by defective TCA cycle metabolism. iScience. 2018;2:63–75. doi: 10.1016/j.isci.2018.03.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Selak MA, Armour SM, MacKenzie ED, Boulahbel H, Watson DG, Mansfield KD, Pan Y, Simon MC, Thompson CB, Gottlieb E. Succinate links TCA cycle dysfunction to oncogenesis by inhibiting HIF-alpha prolyl hydroxylase. Cancer Cell. 2005;7:77–85. doi: 10.1016/j.ccr.2004.11.022. [DOI] [PubMed] [Google Scholar]
- 98.Eng C. PTEN: one gene, many syndromes. Hum Mutat. 2003;22:183–198. doi: 10.1002/humu.10257. [DOI] [PubMed] [Google Scholar]
- 99.Ni Y, He X, Chen J, Moline J, Mester J, Orloff MS, Ringel MD, Eng C. Germline SDHx variants modify breast and thyroid cancer risks in Cowden and Cowden-like syndrome via FAD/NAD-dependant destabilization of p53. Hum Mol Genet. 2012;21:300–310. doi: 10.1093/hmg/ddr459. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Ni Y, Zbuk KM, Sadler T, Patocs A, Lobo G, Edelman E, Platzer P, Orloff MS, Waite KA, Eng C. Germline mutations and variants in the succinate dehydrogenase genes in Cowden and Cowden-like syndromes. Am J Hum Genet. 2008;83:261–268. doi: 10.1016/j.ajhg.2008.07.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.von Dobschuetz E, Leijon H, Schalin-Jäntti C, Schiavi F, Brauckhoff M, Peczkowska M, Spiazzi G, Demattè S, Cecchini ME, Sartorato P, Krajewska J, Hasse-Lazar K, Roszkowska-Purska K, Taschin E, Malinoc A, Akslen LA, Arola J, Lange D, Fassina A, Pennelli G, Barbareschi M, Luettges J, Prejbisz A, Januszewicz A, Strate T, Bausch B, Castinetti F, Jarzab B, Opocher G, Eng C, Neumann HP. A registry-based study of thyroid paraganglioma: histological and genetic characteristics. Endocr Relat Cancer. 2015;22:191–204. doi: 10.1530/ERC-14-0558. [DOI] [PubMed] [Google Scholar]
- 102.Opocher G, Schiavi F, Iacobone M, Toniato A, Sattarova S, Erlic Z, Martella M, Mian C, Merante Boschin I, Zambonin L, De Lazzari P, Murgia A, Pelizzo MR, Favia G, Mantero F. Familial nonsyndromic pheochromocytoma. Ann N Y Acad Sci. 2006;1073:149–155. doi: 10.1196/annals.1353.015. [DOI] [PubMed] [Google Scholar]
- 103.Ni Y, Seballos S, Ganapathi S, Gurin D, Fletcher B, Ngeow J, Nagy R, Kloos RT, Ringel MD, LaFramboise T, Eng C. Germline and somatic SDHx alterations in apparently sporadic differentiated thyroid cancer. Endocr Relat Cancer. 2015;22:121–130. doi: 10.1530/ERC-14-0537. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Gimenez-Roqueplo AP, Favier J, Rustin P, Mourad JJ, Plouin PF, Corvol P, Rötig A, Jeunemaitre X. The R22X mutation of the SDHD gene in hereditary paraganglioma abolishes the enzymatic activity of complex II in the mitochondrial respiratory chain and activates the hypoxia pathway. Am J Hum Genet. 2001;69:1186–1197. doi: 10.1086/324413. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Yu W, He X, Ni Y, Ngeow J, Eng C. Cowden syndrome-associated germline SDHD variants alter PTEN nuclear translocation through SRC-induced PTEN oxidation. Hum Mol Genet. 2015;24:142–153. doi: 10.1093/hmg/ddu425. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Ishii T, Yasuda K, Akatsuka A, Hino O, Hartman PS, Ishii N. A mutation in the SDHC gene of complex II increases oxidative stress, resulting in apoptosis and tumorigenesis. Cancer Res. 2005;65:203–209. [PubMed] [Google Scholar]
- 107.Zundel W, Schindler C, Haas-Kogan D, Koong A, Kaper F, Chen E, Gottschalk AR, Ryan HE, Johnson RS, Jefferson AB, Stokoe D, Giaccia AJ. Loss of PTEN facilitates HIF-1-mediated gene expression. Genes Dev. 2000;14:391–396. [PMC free article] [PubMed] [Google Scholar]
- 108.Tsvetkov P, Reuven N, Shaul Y. Ubiquitin-independent p53 proteasomal degradation. Cell Death Differ. 2010;17:103–108. doi: 10.1038/cdd.2009.67. [DOI] [PubMed] [Google Scholar]
- 109.Accordi ED, Xekouki P, Azevedo B, de Alexandre RB, Frasson C, Gantzel SM, Papadakis GZ, Angelousi A, Stratakis CA, Sotomaior VS, Faucz FR. Familiar papillary thyroid carcinoma in a large brazilian family is not associated with succinate dehydrogenase defects. Eur Thyroid J. 2016;5:94–99. doi: 10.1159/000444522. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Yu W, Ni Y, Saji M, Ringel MD, Jaini R, Eng C. Cowden syndrome-associated germline succinate dehydrogenase complex subunit D (SDHD) variants cause PTEN-mediated down-regulation of autophagy in thyroid cancer cells. Hum Mol Genet. 2017;26:1365–1375. doi: 10.1093/hmg/ddx037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Neumann HP, Pawlu C, Peczkowska M, Bausch B, McWhinney SR, Muresan M, Buchta M, Franke G, Klisch J, Bley TA, Hoegerle S, Boedeker CC, Opocher G, Schipper J, Januszewicz A, Eng C European-American Paraganglioma Study Group. Distinct clinical features of paraganglioma syndromes associated with SDHB and SDHD gene mutations. JAMA. 2004;292:943–951. doi: 10.1001/jama.292.8.943. [DOI] [PubMed] [Google Scholar]
- 112.Xekouki P, Stratakis CA. Succinate dehydrogenase (SDHx) mutations in pituitary tumors: could this be a new role for mitochondrial complex II and/or Krebs cycle defects? Endocr Relat Cancer. 2012;19:C33–40. doi: 10.1530/ERC-12-0118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Lai X, Umbricht CB, Fisher K, Bishop J, Shi Q, Chen S. Identification of novel biomarker and therapeutic target candidates for diagnosis and treatment of follicular carcinoma. J Proteomics. 2017;166:59–67. doi: 10.1016/j.jprot.2017.07.003. [DOI] [PubMed] [Google Scholar]
- 114.Teicher BA, Linehan WM, Helman LJ. Targeting cancer metabolism. Clin Cancer Res. 2012;18:5537–5545. doi: 10.1158/1078-0432.CCR-12-2587. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Kim WG, Guigon CJ, Fozzatti L, Park JW, Lu C, Willingham MC, Cheng SY. SKI-606, an Src inhibitor, reduces tumor growth, invasion, and distant metastasis in a mouse model of thyroid cancer. Clin Cancer Res. 2012;18:1281–1290. doi: 10.1158/1078-0432.CCR-11-2892. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Macias H, Hinck L. Mammary gland development. Wiley Interdiscip Rev Dev Biol. 2012;1:533–557. doi: 10.1002/wdev.35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Sumbal J, Chiche A, Charifou E, Koledova Z, Li H. Primary mammary organoid model of lactation and involution. Front Cell Dev Biol. 2020;8:68. doi: 10.3389/fcell.2020.00068. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.García-Lino AM, Álvarez-Fernández I, Blanco-Paniagua E, Merino G, Álvarez AI. Transporters in the mammary gland-contribution to presence of nutrients and drugs into milk. Nutrients. 2019;11:2372. doi: 10.3390/nu11102372. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Wang C, Liu Q, Li HQ, Wu XX, Guo G, Huo WJ, Pei CX, Zhang YL, Zhang SL. Effects of rumen-protected pantothenate supplementation on lactation performance, ruminal fermentation, nutrient digestion and blood metabolites in dairy cows. J Sci Food Agric. 2018;98:2098–2104. doi: 10.1002/jsfa.8691. [DOI] [PubMed] [Google Scholar]
- 120.Tuba J, Orr PF, Wiberg GS. Cytochrome oxidase and succinic oxidase activity in mammary gland mitochondria of lactating rats. Can J Biochem Physiol. 1955;33:904–908. [PubMed] [Google Scholar]
- 121.Smith TC, Richterich B. Patterns of succinoxidase, cytochrome oxidase, nucleic acids and other constituents in mammary glands of pregnant and lactating rats. Arch Biochem Biophys. 1958;74:398–407. doi: 10.1016/0003-9861(58)90010-9. [DOI] [PubMed] [Google Scholar]
- 122.Shilpashree BG, Arora S, Kapila S, Sharma V. Physicochemical characterization of mineral (iron/zinc) bound caseinate and their mineral uptake in Caco-2 cells. Food Chem. 2018;257:101–111. doi: 10.1016/j.foodchem.2018.02.157. [DOI] [PubMed] [Google Scholar]
- 123.Lu Y, Pan D, Xia Q, Cao J, Zhou C, He J, Sun Y, Xu S. Impact of pH-dependent succinylation on the structural features and emulsifying properties of chicken liver protein. Food Chem. 2021;358:129868. doi: 10.1016/j.foodchem.2021.129868. [DOI] [PubMed] [Google Scholar]
- 124.Gupta C, Arora S, Syama MA, Sharma A. Preparation of milk protein-vitamin A complexes and their evaluation for vitamin A binding ability. Food Chem. 2017;237:141–149. doi: 10.1016/j.foodchem.2017.05.106. [DOI] [PubMed] [Google Scholar]
- 125.Liu C, Liu Y, Chen L, Zhang M, Li W, Cheng H, Zhang B. Quantitative proteome and lysine succinylome analyses provide insights into metabolic regulation in breast cancer. Breast Cancer. 2019;26:93–105. doi: 10.1007/s12282-018-0893-1. [DOI] [PubMed] [Google Scholar]
- 126.Jiang P, Du W, Wu M. Regulation of the pentose phosphate pathway in cancer. Protein Cell. 2014;5:592–602. doi: 10.1007/s13238-014-0082-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Diaz-Moralli S, Aguilar E, Marin S, Coy JF, Dewerchin M, Antoniewicz MR, Meca-Cortés O, Notebaert L, Ghesquière B, Eelen G, Thomson TM, Carmeliet P, Cascante M. A key role for transketolase-like 1 in tumor metabolic reprogramming. Oncotarget. 2016;7:51875–51897. doi: 10.18632/oncotarget.10429. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Gao X, Bao H, Liu L, Zhu W, Zhang L, Yue L. Systematic analysis of lysine acetylome and succinylome reveals the correlation between modification of H2A. X complexes and DNA damage response in breast cancer. Oncol Rep. 2020;43:1819–1830. doi: 10.3892/or.2020.7554. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Du YC, Gu S, Zhou J, Wang T, Cai H, Macinnes MA, Bradbury EM, Chen X. The dynamic alterations of H2AX complex during DNA repair detected by a proteomic approach reveal the critical roles of Ca(2+)/calmodulin in the ionizing radiation-induced cell cycle arrest. Mol Cell Proteomics. 2006;5:1033–1044. doi: 10.1074/mcp.M500327-MCP200. [DOI] [PubMed] [Google Scholar]
- 130.Palla VV, Karaolanis G, Katafigiotis I, Anastasiou I, Patapis P, Dimitroulis D, Perrea D. gamma-H2AX: can it be established as a classical cancer prognostic factor? Tumour Biol. 2017;39:1010428317695931. doi: 10.1177/1010428317695931. [DOI] [PubMed] [Google Scholar]
- 131.Polo SE, Jackson SP. Dynamics of DNA damage response proteins at DNA breaks: a focus on protein modifications. Genes Dev. 2011;25:409–433. doi: 10.1101/gad.2021311. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Sharma GG, So S, Gupta A, Kumar R, Cayrou C, Avvakumov N, Bhadra U, Pandita RK, Porteus MH, Chen DJ, Cote J, Pandita TK. MOF and histone H4 acetylation at lysine 16 are critical for DNA damage response and double-strand break repair. Mol Cell Biol. 2010;30:3582–3595. doi: 10.1128/MCB.01476-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Murr R, Loizou JI, Yang YG, Cuenin C, Li H, Wang ZQ, Herceg Z. Histone acetylation by Trrap-Tip60 modulates loading of repair proteins and repair of DNA double-strand breaks. Nat Cell Biol. 2006;8:91–99. doi: 10.1038/ncb1343. [DOI] [PubMed] [Google Scholar]
- 134.Polletta L, Vernucci E, Carnevale I, Arcangeli T, Rotili D, Palmerio S, Steegborn C, Nowak T, Schutkowski M, Pellegrini L, Sansone L, Villanova L, Runci A, Pucci B, Morgante E, Fini M, Mai A, Russo MA, Tafani M. SIRT5 regulation of ammonia-induced autophagy and mitophagy. Autophagy. 2015;11:253–270. doi: 10.1080/15548627.2015.1009778. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Greene KS, Lukey MJ, Wang X, Blank B, Druso JE, Lin MJ, Stalnecker CA, Zhang C, Negrón Abril Y, Erickson JW, Wilson KF, Lin H, Weiss RS, Cerione RA. SIRT5 stabilizes mitochondrial glutaminase and supports breast cancer tumorigenesis. Proc Natl Acad Sci U S A. 2019;116:26625–26632. doi: 10.1073/pnas.1911954116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Newsholme P, Procopio J, Lima MM, Pithon-Curi TC, Curi R. Glutamine and glutamate--their central role in cell metabolism and function. Cell Biochem Funct. 2003;21:1–9. doi: 10.1002/cbf.1003. [DOI] [PubMed] [Google Scholar]
- 137.Szeliga M, Obara-Michlewska M. Glutamine in neoplastic cells: focus on the expression and roles of glutaminases. Neurochem Int. 2009;55:71–75. doi: 10.1016/j.neuint.2009.01.008. [DOI] [PubMed] [Google Scholar]
- 138.Medina MA, Sánchez-Jiménez F, Márquez J, Rodríguez Quesada A, Núñez de Castro I. Relevance of glutamine metabolism to tumor cell growth. Mol Cell Biochem. 1992;113:1–15. doi: 10.1007/BF00230880. [DOI] [PubMed] [Google Scholar]
- 139.Medina MA, Núñez de Castro I. Glutaminolysis and glycolysis interactions in proliferant cells. Int J Biochem. 1990;22:681–683. doi: 10.1016/0020-711x(90)90001-j. [DOI] [PubMed] [Google Scholar]
- 140.Abril YLN, Fernandez IR, Hong JY, Chiang YL, Kutateladze DA, Zhao Q, Yang M, Hu J, Sadhukhan S, Li B, He B, Remick B, Bai JJ, Mullmann J, Wang F, Maymi V, Dhawan R, Auwerx J, Southard T, Cerione RA, Lin H, Weiss RS. Pharmacological and genetic perturbation establish SIRT5 as a promising target in breast cancer. Oncogene. 2021;40:1644–1658. doi: 10.1038/s41388-020-01637-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Xu L, Che X, Wu Y, Song N, Shi S, Wang S, Li C, Zhang L, Zhang X, Qu X, Teng Y. SIRT5 as a biomarker for response to anthracycline-taxane-based neoadjuvant chemotherapy in triple-negative breast cancer. Oncol Rep. 2018;39:2315–2323. doi: 10.3892/or.2018.6319. [DOI] [PubMed] [Google Scholar]
- 142.Fu XL, Gao DS. Endoplasmic reticulum proteins quality control and the unfolded protein response: the regulative mechanism of organisms against stress injuries. Biofactors. 2014;40:569–585. doi: 10.1002/biof.1194. [DOI] [PubMed] [Google Scholar]
