- Review
- Open access
- Published:
Histone demethylases in autophagy and inflammation
Cell Communication and Signaling volume 23, Article number: 24 (2025)
Abstract
Autophagy dysfunction is associated with changes in autophagy-related genes. Various factors are connected to autophagy, and the mechanism regulating autophagy is highly complicated. Epigenetic changes, such as aberrant expression of histone demethylase, are actively associated not only with oncogenesis but also with inflammatory responses. Among post-translational modifications, histone lysine methylation holds significant importance. There are over 30 members of histone lysine demethylases (KDMs), which act as epigenetic regulators in physiological processes and diseases. Importantly, KDMs are abnormally expressed in the regulation of cellular autophagy and inflammation, representing a crucial mechanism affecting inflammation-related diseases. This article reviewed the function of KDMs proteins in autophagy and inflammation. Specifically, It focused on the specific regulatory mechanisms underlying the activation or inhibition of autophagy, as well as their abnormal expression in inflammatory responses. By analyzing each KDM in epigenetic modification, this review provides a reliable theoretical basis for clinical decision marking regarding autophagy abnormalities and inflammatory diseases.
Introduction
Autophagy is a lysosomal degradation process in eukaryotic cells that plays a critical role in the renewal of cell components and the maintenance of cell homeostasis during states of nutrient or energy deficiency [1,2,3]. It can be adjusted by various autophagy-related genes and signaling pathways, involving the activation of innate and adaptive immunity, thereby promoting direct clearance of pathogens, and the processing and the presentation antigens [4,5,6]. Abnormal autophagy may lead to widespread inflammation and overactivation of immune responses [7,8,9,10]. There are several types of autophagy (Fig. 1), which can be divided into three classes based on their mechanisms: macroautophagy, microautophagy, and Chaperone-mediated autophagy (CMA) [11,12,13]. Among these, autophagy is the most widely studied process, involving de novo synthesis of a vesicle-like autophagosome using cellular membrane structure, followed by the translocation and fusion of the autophagosome with lysosomes to degrade the contents [14]. Autophagy is closely related to histone demethylase and inflammation [15,16,17,18].
Molecular mechanism of autophagy. A Macroautophagy: The process of autophagy includes the initiation of autophagy, nucleation of phagophore, membrane elongation, autophagosome maturation, autophagosome-lysosome fusion, and the final degradation and recycling of cellular content. B Chaperone-mediated autophagy (CMA): HSC70 specifically mediates protein transport into lysosomes through the LAMP2A receptor. C Microautophagy: It involves direct phagocytosis of aggregates by lysosomes
The process of autophagy can be broken down into four main steps: initiation, extension and maturation of the phagophore, fusion of the autophagosome and lysosome, as well as degradation of the substrate [19,20,21]. Autophagy contains two types non-selective autophagy and selective autophagy each with different degradation substrates (Fig. 2). Selective autophagy includes processes such as mitophagy, pexophagy, endoplasmic reticulum-phagy, and ribophagy and so on [22,23,24]. Autophagy-related genes (ATGs) participate in the formation of autophagy and are regulated by multiple signaling pathways [25, 26]. The initiation of autophagy requires the interaction of Unc-51 like kinase 1 (ULK1), ATG13, ATG101, and FIP2000, forming the ULK1 complex that induces phagophore formation. This process is mainly regulated by cellular nutrition and energy status [27,28,29]. Amino acids and oxygen can activate mechanistic target of rapamycin complex 1 (mTORC1), leading to the phosphorylation of ATG13, thereby inhibiting the synthesis of the ULK1 complex and preventing the initiation of autophagy [11, 30]. Under conditions of low glucose ATP levels, the AMPK-PKA pathway can become activated, promoting autophagy through the phosphorylation of ULK1 [31]. Subsequently, the class III phosphatidylinositol 3-kinase complex is formed and catalyzes the production of phosphatidylinositol-3-phosphate on the membranes [32, 33]. Simultaneously, WD-repeat protein interacting with phosphoInositides (WIPI) and ATG2 facilitate in the recruitment of ATG9A vesicles, jointly regulating the extension of phagocytic membranes [34]. Additionally, two crucial ubiquitin-like conjugation systems, ATG12-ATG4-ATG16L and light chain 3 (LC3)-II, are involved in the extension and maturation of autophagosomes [35, 36]. Firstly, the E1-like protein ATG7 and the E2-like protein ATG10 co-catalyze the assembly of the ATG12-ATG4-ATG16L1 complex, which functions as an E3-like enzyme. Along with ATG7, ATG3, and the cysteine protease ATG4B, it catalyzes the aggregation of LC3 to phosphatidylethanolamine, forming LC3-II thereby promoting the closure and maturation of autophagosomes [37,38,39,40]. Subsequently, the autophagosomes are transported to the lysosomes, where they form autolysosomes and are degraded by acidic hydrolases. The resulting materials are released into cytoplasm for reuse [41, 42]. This process is monitored by various proteins [43]. For example, microtubules are involved in the movement of autophagosomes, and inhibiting their polymerization or depolymerization will hinder autophagy [44,45,46]. Another example includes the soluble N-ethylmaleimide-sensitive factor attachment protein receptors (SNARE) complex, which facilitates membrane fusion between autophagosomes and lysosomes [47]. Additionally, LC3-II can mediate the processes of selective autophagy [48,49,50]. For instance, during the maturation of red blood cells, BNIP3L on the mitochondrial outer membrane can interact with LC3-II to promote mitochondrial autophagy [51].
Schematic of the autophagy pathway. A In Bulk: Cytoplasmic cargo is first trapped in double-membrane vesicles (autophagosomes), whose membranes are mediated by the autophagy-related protein LC3 with lipid phosphatidylethanolamine, along with other autophagy-related proteins (e.g., ATG5, ATG16, and ATG12). B Selective autophagy: Particular substrates, such as mitochondria, endoplasmic reticulum, aggregates, and lipid droplets, which are ubiquitinated, bind to the autophagy receptor, such as P62, which interact with LC3 on the phagosome membrane to transport the substrates into the vesicles. These specific substrates include mitochondria, endoplasmic reticulum, aggregates and lipid droplets. C Fusion of autophagosomes- lysosomes: autophagosomes fuse forms lysosomes, forming autolysosomes responsible for degrading the engulfed molecules
Chromatin is composed of DNA and surrounded by nucleosomes, forming a dynamic structure that continually changes depending on the external environment [52,53,54]. Each nucleosome consists of an octamer of four histones (H2A, H2B, H3, and H4) (Fig. 3), which affects chromatin compression and subsequently regulates transcription levels of different genes [55,56,57,58]. Histone modifications on specific residues include acetylation, methylation, phosphorylation, citrullination, ubiquitination, ADP-ribosylation, deamidation, formylation, O-linked N-acetylglucosamine glycosylation (O-GlcNAC), propionylation, butylation, crotonylation, and proline isomerization. These epigenetic modifications play a critical role in regulating gene expression during disease development [59,60,61,62,63]. Methylation of lysine residues on histones is considered a significant class of post-translational modifications. The methylation of lysine is catalyed by histone lysine methyltransferases (KMTs) [64,65,66], resulting in monomethylation, dimethylation, or trimethylation (Kme1, Kme2, and Kme3). These enzymes recognize specific methylation marks on histones to mediate their effects. The removal of lysine methylation is carried out by lysine demethylases (KDMs) [67,68,69]. Based on sequence homology and structural similarity, KDMs can be further classified into eight subfamilies (KDM1-8). According to the catalytic mechanism, KDM1 is categorized as a flavin adenine dinucleotide (FAD)-dependent amine oxidase (LSD family), whereas KDM2-8 (Fig. 4) are demethylases containing the JumonjiC (JmjC) domain (JMJD family) [68, 70, 71].
Histone post-translational modifications (PTMs). Histones can undergo various PTMs at different amino acid residues. PTMs influences the interactions of histone with DNA and other proteins, thereby either promoting or repressing gene transcription. Common PTMs include methylation (purple), acetylation (red), phosphorylation (orange), etc
Phylogenetic tree of the KDMs family. KDMs are classified into two major families based on substrates and reaction mechanisms: lysine-specific histone demethylases (LSD) and JmjC demethylases. Notably, LSD1 was the first demethylase to be identified. Additionally, KDM2-8 are JmjC domain-containing demethylase
Both types of KDMs catalyze N-methyl-lysine demethylation through oxidation mechanisms but in different ways [72, 73]. LSDs use FADs and electron transfer. Since the electron pairs required for demethylation are only present on monomethylated and dimethylated histones, LSDs cannot remove trimethylation from lysine residues [74, 75]. In contrast, the JmjC domain of the Jumonji C domain-containing (JMJD) family proteins contains 170 amino acids. The 2-oxoglutarate and O2 are used as the co-substrate, and Fe(II) is used as a cofactor to promote the enzyme’s oxygenase reaction and exert its activity [76, 77]. This means that JmjC demethylase can remove monomethyl, dimethyl, and trimethyl labels on histone lysine residues [78]. JMJD family members can be further categorized based on the molecular weight (> 100 kDa or < 100 kDa), lysine demethylation specificity, or the presence of functional domains [79]. In recent years, an increasing number of studies have shown that autophagy plays an essential role in inflammation by affecting the differentiation and maturation of inflammatory cells. Autophagy also participates in modulating gene transcription and the secretion of cytokines [80]. Autophagy and cytokines mutually regulate each other to maintain cellular homeostasis [81]. This review focused on the specific mechanisms by which histone demethylation could activated or inhibited autophagy regulation, providing a reliable theoretical basis for the clinical search for identifying efficient therapeutic targets for inflammatory diseases.
KDMs in autophagy
Increasing evidence has demonstrated that, in addition to traditional metabolic-related signals, epigenetic modifications are also key mechanisms in autophagy regulation, in which histone demethylases play an important role [82]. The KDM protein family consists of KDM1-KDM8 subfamilies. These subfamilies act on different histone methylation sites and exert different regulatory effects on gene expression and autophagy (Fig. 5).
KDMs in autophagy regulation. A KDM1A inhibits autophagy by depleting histone H3K4me2, thus suppressing SESN2 and p62 transcription. It is recruited by SHP to CREB target genes, removing H3K4me3 and repressing the expression of autophagy-related genes such as TFEB, ATG3, and ATG10. Additionally, KDM1A directly mediates the demethylation of K151me1 of ATG16L1, enhancing the phosphorylation of ATG16L1 and its binding to ATG12-ATG5, consequently promoting the extension and maturation of autophagosomes. In addition, knockdown of KDM1A can block PTEN ubiquitination and enhance its stability, thereby regulating mTOR activity and inhibiting autophagy. B NUPR1 binds to KDM3A, reducing H3K9me2 at the TFEB promoter and promoting TFEB-mediated autophagy; C KDM3B activates autophagy by inducing GABARAPL1 transcription. D Nutlin treatment promotes KDM4B expression, which activates ULK1 and ATG16L1 transcription by decreasing H3K9/K36me3, thereby promoting autophagy and inhibiting apoptosis. On the contrary, Nutlin treatment reduces KDM4B expression in MHM and SJSA1 cells by activating p53, thereby inhibiting autophagy and promoting apoptosis. KDM3B mediated H3K9me3 demethylation also inhibits the expression of LC3B and promotes autophagy. E KDM6A may interact with MLL3 to regulate the increase of H3K4me3 and promote the expression of autophagy genes. F KDM6B enhances autophagy by inhibiting H3K27me3 at the TFEB promoter. In addition, ecdysone recruits dUTX to regulate autophagy-related genes by managing H3K27me3 at the promoter. FGF21 activates KDM6B phosphorylation and promotes the expression of TFEB, ATG7, ULK1, and other genes by regulating H3K27me3, thereby enhancing autophagy. G KDM5C interacts with ULK1 to mediate the demethylation of R170me2. Hypoxia weakens the catalytic activity of KDM5C, activating ULK1 by enhancing R170me2 and promoting the initiation of autophagy. H KDM7B activates ATG17/FIP200 transcription by binding to its promoter region, thereby activating autophagy
KDM1
Current studies have identified KDM1A as one of the proteins potentially closely associated with the regulation of autophagy. KDM1A, also known as LSD1, belongs to the KDM1 subfamily Unlike the rest of the KDM2-KDM8 subfamily, KDM1A does not contain the JmjC domain, but has FAD-dependent amine oxidase activity [83]. KDM1A can form a co-suppressor complex with corepressor of repressor element-1 silencing transcription factor (CoREST) to remove histone modification of histone H3 lysine 4 dimethylation/trimethylation (H3K4me2/3), thereby inhibiting target gene transcription [84, 85]. Furthermore, in specific instances, KDM1A can act upon histone h3 lysine 9 monomethylation/dimethylation (H3K9me1/2) to activate target gene expression [86]. Predominantly, KDM1A exerts inhibitory effects on autophagy, and the modulation of the mTORC1 pathway is one of its pivotal mechanisms. Feng et al. [87]reported that KDM1A regulated the initiation of autophagy through regulating the protein kinase B (AKT)/mechanistic target of rapamycin (mTOR) pathway. Treatment with the KDM1A inhibitor S2101 reduced the levels of phosphorylated (p)-AKT, p-mTOR, and p-70S6K in ovarian cancer cells, thereby enhancing autophagy and inhibiting ovarian cancer cells proliferation. Subsequently, Ambrosio et al. [88] elucidated that the regulation of mTORC1 activity by KDM1A relied on the expression of Sestrin2 (SESN2). SESN2, an intracellular leucine receptor, interacted with GATOR2 to inhibit the activity of mTORC1. By suppressing of SESN2 expression, KDM1A increased mTORC1 activity, thereby inhibiting autophagy. Importantly, KDM1A bound to the transcriptional starting site of SESN2. Upon KDM1A inhibition or knockdown, the level of H3K4me2 significantly increased in this area, accompanied by enhanced histone H3 acetylation, reduced H3K27me3, and no obvious change in H3K9me2 levels. These findings suggest that KDM1A-mediated epigenetic changes play a key role in regulating mTOR. The KDM1A/SESN2/mTORC1 pathway contributes to modulating autophagy in various tumors and inflammatory diseases. A novel LSD1 inhibitor, ZY0511, was found to augment autophagy and apoptosis in diffuse large B-cell lymphoma cells by inhibiting mTORC1 activity, leading to suppressed tumor proliferation in mouse models [89]. Knockdown of KDM1A promoted autophagy in macrophages through the KDM1A/SESN2/mTORC1 pathway and inhibited oxidized low-density lipoprotein (ox-LDL)-induced NLRP3 inflammasome activation and the release of inflammatory cytokines [90]. In rats with kidney failure, the histone deacetylase histone deacetylase 1 (HDAC1) was shown to negatively regulated KDM1A transcription. Consequently, inhibition of KDM1A led to heightened activation of the SESN2 promoter and enhanced autophagy, and exacerbated vascular calcification [91]. In addition, Shi et al. [92] demonstrated that knocking down KDM1A could hinder phosphatase and tensin homolog (PTEN)ubiquitination, thereby promoting its stability. Given that PTEN is also an inhibitor of the AKT/mTORC1 pathway, the KDM1A/PTEN/mTORC1 axis represents another mechanism for regulating autophagy.
In addition to the mTORC1 pathway, KDM1A regulates autophagy through other targets. P62 is an important carrier that mediates the binding of autophagy substrates to autophagosomes. In various tumor cells, KDM1A binds to the p62 protein and reduced its stability by regulating ubiquitination, thereby inhibiting autophagy and promoting the occurrence of gynecological tumors. The regulatory impact of KDM1A on p62 depends on its catalytic activity [93]. Furthermore, KDM1A suppresses the transcription of p62 by eliminating H3K4me2, which contributes to the regulation of programmed oocyte death [94]. Byun et al. [95] observed an increase in hepatic fibroblast growth factor after food consumption. In this context, KDM1A was recruited by small heterodimer partner (SHP) to the cAMP response element binding protein (CREB) target gene locus, where it removed H3K4me2/3 and H3K9/14 acetylation modifications, while enhancing H3K9me2. It further resulted in the suppression of autophagy-related genes such as TFEB, ATG3, and ATG10, leading to a reduction in lipid autophagy within hepatocyte phagocytic vesicles. Moreover, KDM1A inhibits autophagy in diseases, such as acute myeloid leukemia, prostate cancer, hypopharyngeal cancer, and other diseases. Knockdown or inhibition of KDM1A often hinder the occurrence and progression of these tumors. However, the specific mechanism and clinical implications of KDM1A in these contexts need to be further clarified [96,97,98].
KDM1A can also promote autophagy through other mechanisms. For instance, in studies on Alzheimer’s disease (AD), KDM1A was found to interact with proteins associated with mRNA methylation, enhancing the autophagic degradation of p-Tau by increasing the activity of the transcription factor EB (TFEB). This process helped impede the progression of AD [99]. Additionally, KDM1A targeted the promoter of methyltransferase-like 3 (METTL3), resulting in a decrease in the levels of H3K9me3 [99]. METTL3 subsequently activated the ubiquitin ligase STUB1 through m6A methylation modification, which facilitated the degradation of inactivated p-TFEB while increasing activated TFEB levels. This, in turn, promoted the expression of autophagy-related genes [100]. Furthermore, KDM1A functions as non-histone demethylase by directly mediating the demethylation of ATG16L1 at the K151me1 site. This enhanced the phosphorylation of ATG16L1, thereby strengthening its binding to ATG12-ATG5 complex, ultimately promoting the extension and maturation of autophagosomes [101]. In hypoxia/reoxygenation-stressed cardiomyocytes, the expression of the demethylase KDM1A was up-regulated, while the methylase SET domain containing 7 (SETD7) was down-regulated. KDM1A and SETD7 antagonized each other, collectively regulating the methylation of ATG15L1 at the K151 site. This interaction promoted autophagy and protected the survival of cardiomyocytes under stress conditions [101].
JmjC proteins and autophagy
KDM2
The KDM2-KDM8 proteins are members of the JmjC family, whose catalytic activity depends on JmjC activity and requires the participation of cofactors such as α-ketoglutaric acid (α-KG) and Fe2+ [102]. Among them, KDM2A/KDM2B mainly target the methylation of H3K36 to activate the expression of target genes [103, 104]. In ovarian cancer, the high expression of KDM2A have been implicated in the ubiquitination-mediated degradation of Beclin-1. Knockdown of KDM2A enhances the stability of Beclin-1 and promotes the activation of autophagy [105]. Similarly, the high expression of KDM2B can be observed in gastric cancer or lung squamous cell carcinoma tissues. However, knocking down KDM2B leads to the reduced p-Akt/mTOR activity, increased the ratio of LC3-II/LC3-I, and decreased p62 protein levels, indicating enhanced autophagy and inhibited of tumor cell growth [106, 107]
KDM3
The KDM3 family mainly includes KDM3A and KDM3B. It acts on H3K9me1/2 to activate the transcription of target genes [108]. KDM3 proteins primarily promotes autophagy by activating autophagy-related genes. Nutrient deprivation, such as glycogen or amino acid starvation, is a common trigger for inducing autophagy. Kim et al. [109] found that starvation-induced autophagy depended on the up-regulation of KDM3A, which could bind to the promoters of autophagy-related genes such as Map1lc3b and Atp6v1c1 to reduce H3K9me2 levels, thereby promoting autophagy in mice liver. In glioma cells, hypoxia enhanced the expression of the promoting gene Nupr1, which bound to KDM3A to reduce H3K9me2 levels at the promoter of TFEB, promoting TFEB mediated autophagy and inducing tumor resistance to temozolomide [82]. Similarly, in colon cancer cells, starvation culture upregulated KDM3B protein, which promoted autophagy by regulating H3K9me2 at the promoters of ATG5 and ATG7 [110]. Furthermore, Song et al. [111] discovered that in acute myeloid leukemia (AML) cells, KDM3B can activated the transcription of GABA(A) receptor-associated protein like 1 (GABARAPL1). GABARAPL1, belonging to the ATG8 protein family along with LC3, plays a key role in autophagosome elongation. Thus, KDM3B-mediated transcriptional activation facilitates autophagy in AML cells. Overall, KDM3 proteins are activated by nutrient deficiency and play a pivotal role in promoting autophagy.
KDM4
The KDM4 family, consisting of KDM4A-KDM4D, targets inhibitory sites marked by H3K9me2/3 or H3K36me3 [112, 113]. Recent studies have established their close association with autophagy. KDM4A was highly expressed in glioma cells, and silencing KDM4A (siKDM4A) promoted autophagy, leading to decreased cell viability and increased apoptosis [114]. Duan et al. [115] revealed that Nutlin treatment stimulated the expression of KDM4B, activating the transcription of ULK1 and ATG16L1 by demethylating H3K9/K36me3 in A549 cells, thereby promoting autophagy and inhibiting apoptosis. Similarly, Tan et al. [116] demonstrated that in colon cancer cells, glycogen deprivation increased KDM4B expression and suppressed LC3B expression through demethylating H3K9me3 at its the promoter, thereby improving the level of autophagy and favoring cell survival. In castration-resistant prostate cancer, KDM4B promoted autophagy by activating the activity of the Wnt/β-catenin signaling pathway [117]. Additionally, KDM4C has been implicated in stress-induced autophagy. In a mouse model of kidney injury, knockdown of KDM4C inhibited autophagy, suggesting that KDM4C may positively regulate autophagy under stress conditions [118]. Moreover, KDM4D is involved in selective autophagy during autoimmune encephalomyelitis. Optineurin (OPTN) mediated the interaction of ubiquitinated-KDM4D interacting with LC3 in autophagosomes. TRIM14 inhibited the autophagic degradation of KDM4D by recruiting deubiquitination enzymes, resulting in increasing KDM4D protein levels, and enhanced expression of pro-inflammatory factors through H3K9me3 [119]. Overall, the KDM4 family proteins can either promote or inhibit autophagy in different contexts. Further studies are needed to confirm these mechanisms.
KDM5
KDM5 primarily functions as an H3K4 demethylase, with its members including KDM5A-C [120, 121]. Wang et al. [122] observed an increase in the promoter H3K4me3 level at the PIK3C3 gene promoter upon KDM5B knockdown in esophageal squamous cell cancer cells, consequently activating autophagy and apoptosis while enhancing the sensitivity of mouse models to radiotherapy. Li et al. [123] identified a distinctive demethylase activity of KDM5C, which targeted arginine residues within the autophagy protein ULK1, thereby inhibiting hypoxia-induced autophagy. In hypoxia-cultured tumor cells, the ULK1 protein was found to be symmetrically dimethylated at the arginine 170 site (R170me2s). Mutations at the R170 site inhibited hypoxia-induced autophagy. The demethylase KDM5C could interact with ULK1 to mediate the methylation of R170me2 through the catalytic activity of the JmjC domain. Under hypoxic conditions, the catalytic activity of KDM5C was diminished, leading to increased R170me2s of ULK1. This modification activated ULK1 by promoting its phosphorylation at T180, subsequently inducing the phosphorylation of ATG13 and Beclin-1, and activating downstream autophagy pathways. Furthermore, in LN229 glioma cells, hypoxia-induced autophagy promoted mitochondrial clearance and reduced oxygen consumption by inducing autophagy, ultimately favoring cell survival [123].
KDM6
The KDM6 family, including KDM6A (UTX), KDM6B (JMJD3), and KDM6C (UTY), demethylates H3K27me2/3 to activated the expression of related genes, thereby promoting autophagy [124,125,126]. Similarly, in Drosophila, demethylase UTX (dUTX), a homolog of KDM6A, is recruited by the steroid hormone ecdysone to regulate autophagy and apoptosis-related genes, including ATG 1, ATG 2, ATG 7, ATG 9 and ATG 18, by regulating H3K27me3 at the promoters. Therefore, dUTX is an important regulator of ecdysone-mediated programmed death in Drosophila salivary glands [127]. Yin et al. [128] observed a significant upregulation of KDM6B expression in dental pulp stimulated by tumor necrosis factor-alpha (TNF-α), thereby promoting autophagy. However, silencing KDM6B inhibited the expression of autophagy-related genes such as ATG5, LC3B, FIP200, and ATG12, by regulating H3K27me3 levels at their promoters. Wang et al. [129] further demonstrated that KDM6B enhanced autophagy in thyroid cancer cells by decreasing H3K27me3 levels at the TFEB promoter. KDM6B promoted TFEB transcription, thereby upregulating lysosomal-related gene expression and promoting autophagy. Additionally, KDM6B participated in lipid autophagy in the liver. The starvation-sensing hormone Fibroblast growth factor 21 (FGF21) mediated the activation of KDM6B via PKA-mediated phosphorylation, elevating the expression of TFEB, ATG7, and ULK1 by reducing H3K27me3. This enhanced liver autophagy, promoting the decomposition of triglycerides and β-oxidation of fatty acids [17]. Furthermore, KDM6A may regulate autophagy via another mechanism. During starvation, transcription factor E3 (TFE3) translocated to nucleus and recruited KDM6A to target gene promoters such as MAP1LC3B and WIPI2. This recruitment increased H3K4me3 levels, ultimately promoting autophagy and cell proliferation in renal cancer cells. Interestingly, mutant variants of KDM6A lacking catalytic activity still promoted autophagy, indicating its regulatory role on target genes is partially independent of its enzyme activity [130]. Knockout of KDM6A also reduced the recruitment of methyltransferase mixed-lineage leukemia 3 (MLL3) to the target gene promoters, suggesting a potential interaction between KDM6A and MLL3 in the regulation of autophagy [130].
KDM7
The KDM7 family members, including KDM7A, KDM7B, and KDM7C, target H3K9/K27 as their substrates [131]. They are also the only KDM proteins capable of demethylating histone H4. Particularly, KDM7B (PHF8) can positively regulate autophagy. In hepatocellular cancer (HCC) cells, KDM7B promotes autophagy by binding to the transcriptional initiation domain of ATG17/FIP200, thereby enhancing its transcription. This process leads to the degradation of E-cadherin, which in turn increases the invasion and migration of HCC cells. Moreover, KDM7B-mediated autophagy is implicated in Sorafenib resistance of HCC mice, further promoting tumor progression [132, 133]. However, Witucki et al.’s study on AD revealed that KDM7B inhibited autophagy by enhancing the H4K20me1 modification at the mTOR promoter, leading to increased activation of the mTOR pathway and inhibiting autophagy. Therefore, it contributes to increased Aβ protein aggregation in AD models [134].
KDMs in autophagy and inflammation
Histone modifications contribute to significant changes in genome structure and function. The KDMs family is involved in signaling pathways that regulate autophagy and inflammation (Fig. 6). Substantial evidence underscores that dysregulated expression of KDMs is closely associated with multiple inflammatory responses.
Mechanisms of KDMs in regulation of inflammatory factors. KDMs regulate the expression of inflammatory factors in response to LPS, PAMPs, and various other stimuli. 1) KDM1A is stimulated by ox-LDL, which suppresses the PI3K/mTOR inhibitor SESN2 and inhibits autophagy of NLRP3 inflammasome, resulting in elevated inflammatory cytokines. Additionally, KDM1A and KDM1B interact with the NF-κB pathway and modulate the expression of IL-6, CCL2, and IL-12β through histone demethylation. 2) KDM2A is driven by NF-κB and negatively regulates inflammation by demethylation and consequently inhibiting p65. 3) KDM3A interacts with MRTF-A, promoting inflammation by removing H3K9me2 of ITGB 2. 4) KDM4A is recruited to NF-κB binding sites and promotes the production of IL-1β, CCL2, and TNF by altering histone methylation. Additionally, KDM4B and KDM4D control the activation of NF-κB and the expression of cytokines such as IL-12β and IL-23. 5) KDM5A is recruited by p50 and suppresses the expression of Socs1 to activate NK cells. Upon LPS stimulation, KDM5A is enriched at the PTEN promoter and inhibits its expression. KDM5B can be recruited to the promoter of IL-6, IL-23a, and Th1 cytokines, including TNF, IL-1β, and IFN-β, inhibiting their production by H3K4me3 demethylation. 6) KDM6A removes the H3K27me3 of Lcn2 and IL-6. Additionally, KDM6A interacts with MLL4 to induce the expression of IFN-β by promoting H3K4me2. KDM6B regulates macrophage polarization by removing H3K27me3 of M2 transcriptional factors, such as STAT6, IRF4, and T-bet. KDM6A and KDM7A mediate the transcription of VCAM1, Cxcl1/2, IL-8, and p65 via H3K27me3 and H3K9me2, respectively.7) KDM4A/6B and KDM7B participate in the regulation of IL-6, IL-8, IFN-β, and others through histone methylation. KDM7C can interact with the CBP/p300 coactivator to promote TNF, CCL4, and IL-1β expression by H4K20 demethylation
KDM1
KDM1 primarily removes monomethylated and dimethylated modifications on histones, participating in the regulation of autophagy and inflammation through epigenetic modifications [135]. Zhou et al. [90] found that downregulation of KDM1A/LSD1 promoted autophagy by inhibiting SESN2-mediated activation of the mTOR pathway in atherosclerotic disease, which in turn reduced ox-LDL-stimulated activation of the NLRP3 inflammasome and cytokine production. However, Xie et al. [136] discovered in homocysteine upregulated the expression of SNF5 through KDM1A in a mouse model of atherosclerosis, regulated the monomethylated modification of histone H3K4 in macrophages, and then led to an increase in the interleukin (IL)−1β level, aggravating the inflammation of macrophages and ultimately promoting the formation of atherosclerosis. KDM1A inhibitors significantly reduced the protein expression level of aortic inflammatory factors [137]. In Addition, KDM1A has been found to positively regulate the expression of toll-like receptor 4 (TLR4) by catalyzing H3K9me2 in the promoter region of HBV-associated glomerulonephritis. Knockdown of KDM1A inhibited the TLR4-nuclear factor-kappaB (NF-κB)/ Jun N-terminal kinase (JNK) signaling axis in HBV-induced HK-2 cells, reducing the production of pro-inflammatory mediators [135]. Wang et al. [138] observed that inhibition of KDM1A activity by GSK-LSD1 led to elevated H3K4me2 and H3K9me2 modifications, which further inhibited the activation of NF-κB signaling, subsequently inhibiting the production of cytokines (TNF-α, IL-6, and IL-1β), thereby alleviating the inflammatory response of mastitis. In rheumatoid arthritis (RA), KDM1A has promoted the proliferation of CD4+T cells and the production of inflammatory cytokines IL-17 and immune interferon (IFN)-γ [139]. In lipopolysaccharide (LPS)-induced sepsis, phosphorylation of KDM1A has promoted the demethylation of p65 K314/315 to activate NF-κB targeted genes and recruited KDM1A and p65 to the NF-κB promoter, thus activating PKCα-KDM1A-NF-κB signaling to promote the inflammatory response in sepsis [140]. In the adipose tissue of obesity, knockout of KDM1A has promoted the binding of CCAAT/enhancer binding protein beta (C/EBPβ) and NF-κB at the IL-6 promoter, subsequently increasing the transcription of inflammatory genes including carbon tetrachloride (CCL)2, CCL20, IL-6, IL-15, and C/EBPβ [141]. KDM1B also regulates inflammation by interacting with the NF-κB protein through the connection between AO-N and AO-C subdomains. Disrupting the interaction prevents KDM1B recruitment to CCL22 and IL-12b, mitigating inflammatory responses [142].
JmjC protein autophagy and inflammation
KDM2
KDM2A can either positively or negatively regulate inflammatory cytokines and inflammatory signaling pathways in different situations, while KDM2B positively regulates the expression of inflammatory cytokines [143]. High expression of KDM2A in ovarian carcinoma participates in the ubiquitination degradation of Beclin-1. Knockdown of KDM2A enhances the stability of Beclin-1 and promotes the activation of autophagy [105].KDM2A could demethylate the lysine K218 or K221 residues of p65 inhibit its expression. This process was antagonized by nuclear receptor binding SET domain protein 1 (NSD1), which serves as a methyltransferase to K218 or K221 residue of p65 to promote the transcriptional activity of NF-κB [144]. In psoriasis-like dermatitis, KDM2A reduced the inflammatory response of epidermal keratinocytes by suppressing the activity of NF-κB, while KDM2A inhibitors enhanced the expression of cytokines such as IL-8 and CCL20 induced by poly (I:C) [145]. In high-fat diet -treated mice, deletion of KDM2A significantly reduced the expression of inflammatory factors IL-6, IL-1β, and CCL2, thereby suppressing chronic inflammation in adipose tissue. Deletion of KDM2A has increased the H3K36me2 level at the peroxisome proliferator-activated receptor gamma (PPARγ) site, promoting the binding of STAT6 and mediating the polarization of M2 macrophages [146]. In osteoarthritis (OA) mice, the levels of KDM2A, IL-1β, IL-6, and TNF-α were significantly reduced, and overexpression of miR-31 has down-regulated the expression of KDM2A and activated the E2F1/PTTG1 axis, thereby reducing the expression levels of IL-1β and TNF-α in OA [147]. Hypoxia increased the expression of KDM2A in stem cells of apical papilla, and KDM2A has inhibited the transcription of secreted Frizzle-related protein 2 by reducing the methylation of histone H3K4 and H3K36 in its promoter, inhibiting the canonical Wnt/β-catenin pathway, and then inhibiting the NF-κB signaling pathway [148]. Zhou et al. found that KDM2B bound to the SWI/SNF complex containing Brg1 in macrophages and dendritic cells (DCs) to catalyze chromatin remodeling of the IL-6 promoter. In addition, KDM2B has directly engages RNA polymerase II to further initiate IL-6 transcription, thereby promoting IL-6 production in the inflammatory response [149].
KDM3
KDM3 regulates the inflammatory response by controlling proinflammatory factors and NF-κB pathways. In high glucose and hypoxia-treated human umbilical vein endothelial cells(ECs), KDM3A has increased inflammatory damage by up-regulating the expression of IL-6, IL-8, and monocyte chemoattractant protein-1 (MCP-1), and promoted oxidative stress progression by enhancing the levels of reactive oxygen species(ROS) and malondialdehyde (MDA) while reducing the activity of superoxide dismutase(SOD) [150]. Inhibition of KDM3A significantly reduced insulin-induced cytokines such as IL-6 and MCP-1 and suppressed the activation of phosphorylated mitogen-activated protein Kinases (p-MAPKs) and NF-κB/p65 pathways in vascular smooth muscle cells [151]. In glioma cells, hypoxia enhanced the expression of the promoting gene Nupr1, which bound to KDM3A to reduce H3K9me2 of TFEB promoter, promoting TFEB mediated autophagy and thus inducing tumor resistance to temozolomide [82]. Zhang et al. [152]have demonstrated that KDM3A enhanced the transcription of NF-κB/p65 to drive the continuous inflammatory response in diabetic myocardial injury. Liang et al. [153]found that leukocidin enhanced the transcriptional activity of transforming growth factor β inducible factor 1 by promoting the ubiquitination and degradation of KDM3A, which inhibited the activation of the TGF-β1/Smad2/3 signaling pathway and suppressed the inflammatory response in diabetic nephropathy. The interaction between KDM3A and myoglobin-associated transcription factor A (MRTF-A) might contribute to integrin beta 2(ITGB2) transcription, leading to macrophage adhesion to ECs, and cardiac inflammation was suppressed in macrophage MRTF-A conditional knockout mice [154]. In ECs, the collaborative action of KDM3A alongside Brg1 serves to initiate the transcription of colony-stimulating factor 1(CSF1), thereby facilitating the recruitment of macrophages, consequently perpetuating vascular inflammation [155]. In colon cancer cells, the KDM3A peptide was recruited to the 15-LOX-1 promoter, subsequently demethylating H3K9me2 and ameliorating carcinogenesis [156]. In KDM3C knockdown THP-1 cells, LPS induced the phosphorylation of NF-κB p65 and its nuclear translocation, and stimulated the expression of TNF-α, IL-1β, and IL-6. In contrast, overexpression of KDM3C inhibited NF-κB signaling and osteoclast generation, thereby enhancing the anti-inflammatory function of macrophages against oral bacterial infection [157].
KDM4
The KDM4 family predominantly assumes a proinflammatory role in regulating inflammatory responses. Various studies have highlighted the critical role of KDM4A in the inflammation cardiovascular system. In a study involving mice with kidney injury, it was found that knockdown of KDM4C inhibited autophagy under stress conditions, promoted the secretion of inflammatory cytokines [118]. The TRIM14-USP14-BRCC3 complex inhibits OPTN-mediated autophagic degradation of KDM4D by reducing K63 ubiquitination on KDM4D, thereby removing the H3K9me3 modification from the promoters of IL-12 and IL-23, promoting the expression of the pro-inflammatory cytokines IL-12 and IL-23, and enhancing the inflammatory response [158].Upon exposure to ox-LDL, KDM4A is upregulated in macrophages, provoking pro-inflammatory M1 polarization. Knockout of KDM4A significantly reduces the expression of M1 inflammatory factors such as TNF-α, IL-1β, and MCP-1, independent of NF-κB or HIF pathways activation, suggesting that KDM4A may exert its proinflammatory role through alternative pathways such as epigenetic modification [159]. In the LPS-induced inflammatory response of vascular ECs, KDM4A was recruited to the NF-κB target gene promoter, removing H3K9me3. Subsequently, methyltransferase SET1A was recruited, leading to an increase in H3K4me3, thereby activating the expression of IL-1β, CCL2, and TNF [160]. Compound Danshen dripping pills inhibited the activity of KDM4A and block the transcription of p65 in a mouse model of heart failure induced by a high-fat diet, consequently reducing the inflammatory response [161]. Moreover, KDM4A/4B is involved in nervous system inflammation. In a rat cerebral ischemia model, knockdown of KDM4A inhibited NF-κB signaling in microglia, reducing the neuroinflammatory response and promoting the recovery of stroke [162]. LPS-induced inflammation in nerve cells, and knockdown of KDM4B inhibited the expression of inflammatory factors upon KDM4B knockdown by recruiting inhibitory H3K9me3 to Notch1, IL-1β, and IL-2 promoters [163]. Choi et al. [164] found that KDM4B could bind to intercellular adhesion molecule 1 (ICAM1) promoter to reduce H3K9me2, mediating the expression of ICAM1 and inhibiting inflammatory-induced leukocyte extravasation, consequently suppressing nervous system inflammation. Furthermore, KDM4D is also involved in inflammatory responses. Jin et al. [165] showed that upon LPS stimulation activated TLR receptors in DCs and increased the stability of KDM4D protein through inducing the expression of deubiquitinase Trabid. KDM4D promotes the expression of IL-12a, IL-12b, and IL-23a by regulating their H3K9 methylation, inducing the differentiation of Th cells into T-helper 17 (Th17) subtypes and fostering autoimmune inflammation in mice. TRIM14 interacts with USP14 and BRCC3 to inhibit OPTN mediates the interaction of ubiquitinated KDM4D with LC3 in autophagosomes in experimental allergic encephalomyelitis (EAE) mice. TRIM14 inhibits KDM4D autophagic degradation by recruiting deubiquitinating enzymes, resulting in increased KDM4D protein and the expression of inflammatory cytokines such as IL-12 and IL-23 through the removal of H3K9me3, which leads to inflammation-related T cell differentiation [119]. TNF-α upregulates KDM4D expression in dextran sulfate sodium (DSS)-induced UC mice. KDM4D inhibits epithelial cell apoptosis and promotes colitis recovery by activating the Hedgehog signaling pathway [166].
KDM5
KDM5A/5C/5D positively regulates the inflammatory response. Zhao et al. [167] revealed that p50 recruited KDM5A and inhibited its transcription by removing H3K4me3 binding on the Socs1 promoter, which in turn stimulated NK cells to release a substantial quantity of IFN-γ through the JAK2-STAT4 signaling pathway. In contrast, the activation of NK cells in mice with KDM5A knockout was inhibited [167]. Liu. et al. [168] found that dexamethasone (DEX) could inhibit KDM5A activity, leading to increased H3K4me3 modification of inflammation-related genes such as TNF-α, nitric oxide synthase 2 (NOS2), and CCL2. Inhibiting KDM5A could weaken the inflammatory response in mice with renal injury. Qi et al. [169] demonstrated higher expression of KDM5C/6A in aged female microglia compared to males. They highlighted that KDM5C inhibited the expression of the IRF4 gene through H3K4 demethylation. The IRF4 signal mainly directed the production of anti-inflammatory cytokines. Consequently, knockout of KDM5C significantly reduced the expression of anti-inflammatory factors such as IL-4 and CD206/Arg1, and other anti-inflammatory factors, indicating that the poor prognosis of cerebral ischemia in elderly females may be related to the inflammatory regulation of KDM5C.In addition, LOXL1 antisense RNA 1 (LOXL1-AS1) was up-regulated in OA and enhances KDM5C expression via miR-423-5p, thereby intensifying the inflammatory response and apoptosis, ultimately promoting OA development [170]. Ebadi et al. [171] found that KDM5D was the most significantly up-regulated gene in patients with myocardial infarction and coronary artery disease (CAD) through bioinformatics analysis. Moreover, the expression of inflammatory genes such as CCL20, IL-1β, and IL-17 was also significantly enhanced.
Specifically, Wang et al. [122] observed an increase in the promoter H3K4me3 level of the PIK3C3 gene upon KDM5B knockdown in esophageal squamous cell cancer cells, consequently activating autophagy and apoptosis while enhancing the sensitivity of mouse models to radiotherapy. KDM5B may exert multiple effects on the regulation of the inflammatory response. In LPS-induced bone marrow-derived DCs (BMDMs), LPS induced KDM5B to bind to IL-6 and IL-23a promoters. KDM5B can inhibited NF-κB binding to them by reducing H3K4 trimethylation, thereby contributing to inflammation dissipation [172]. Moreover, KDM5B was closely associated with inflammatory damage caused by respiratory syncytial virus (RSV) infection. RSV infection upregulated the expression of KDM5B and inhibits the generation of Th1 antiviral cytokines, such as IFN-β, IL-6, and TNF-α by regulating H3K4me3 modification, consequently promoting the differentiation of CD4+ T cells to the Th2 type. However, in RSV-infected KDM5B knockout mice, the levels of Th2 inflammatory factors IL-4, IL-5, and IL-13 were reduced, and the secretion of mucus was less, alleviating lung injury [173]. In addition to histone demethylation, KDM5B also promotes protein translation by maintaining the length of the 3 ‘UTR and increasing mRNA stability. Vascone et al. [174] discovered that soluble epoxide hydrolase (SEH) plays a crucial role in Angiotensin (Ang) II-mediated vascular endothelial inflammation, and KDM5B can increase the stability of SEH mRNA through this mechanism. Conversely, knockdown or inhibition of KDM5B can reduce endothelial inflammatory damage.
KDM6
KDM6A/6B is widely implicated in the regulation of inflammatory genes and primarily plays a pro-inflammatory role [175]. Previous studies have indicated that TLR4 is activated in LPS-stimulated macrophages and induces the up-regulation of KDM6B in a manner dependent on the NF-κB. However, KDM6B does not affect the overall level of H3K27me3 but forms complexes with Wdr5, RbBP5, and Ash2L, as well as Polycomb group proteins, which selectively regulates H3K27me3 demethylation of target genes such as Bmp-2 to enhance their transcriptional activity [176].Approximately 70% of the genes activated by LPS were targeted by KDM6B, including CCL5, IL-12b, CXCL11, and CCL9 [177]. Knockdown of KDM6B affected the expression of various key genes in NF-κB, chemokine, and CD40 signaling in THP-1 cells. Wang et al. [129] demonstrated that KDM6B enhanced autophagy in thyroid cancer cells by inhibiting TFEB promoter H3K27me3. KDM6B promoted TFEB transcription, thus amplifying lysosomal-related gene expression and promoting autophagy. In fact, knockout of KDM6B can inhibit the expression of NF-κB-mediated inflammatory genes by recruiting the H3K27me3 to their promoters to inhibit the inflammatory response [178]. Li et al. [179] found that KDM6A demethylated the promoter through H3K27me3 to enhance the expression of IL-6 in macrophages, while KDM6A indirectly promoted the expression of IFN-β through the transcription of specific enhancer S-IRE1. Kruidenier et al. [180] developed a small molecule inhibitor GSK-J4 targeting the H3K27 demethylation activity of KDM6A/6B, enabling a more intuitive study of the effects of KDM6A/6B-mediated histone demethylation on inflammatory gene expression. Surprisingly, knockout of KDM6B alone did not result in a significant enhancement of H3K27me3 at the target gene. However, treatment with GSK-J4 or inhibition of both KDM6A and KDM6B by RNA interference suppressed TNF-α transcription and upregulated H3K27me3 at the transcription start region [177, 180]. Inhibition of KDM6B and KDM6A by GSK-J4 also antagonized the production and release of IL-1β in macrophages induced by zoledronic acid, consequently ameliorating the inflammatory response in mice [181]. Moreover, GSK-J4 treatment in NK cells reduced the secretion of IFN-γ, TNF-α, granulocyte–macrophage colony stimulating factor (GM-CSF), and IL-10 by up-regulating H3K27me3, although it did not affect the cytotoxic activity of NK cells against cancer cells [182]. This suggests that KDM6A and KDM6B may play similar proinflammatory functions in inflammation, and subsequent studies have revealed their involvement in various inflammatory diseases. Wang et al. [183] observed an upregulation of KDM6B expression in LPS-induced mastitis, whereas GSK-J1 alleviated mammary inflammation. It was further demonstrated that knockout of KDM6B reduced the expression of TLR4 by regulating H3K27me3, thereby inhibiting the downstream NF-κB proinflammatory signal transduction and inhibiting the production of TNF-α, IL-1β, and IL-6 in breast tissues. Additionally, Johnstone et al. [184] demonstrated the up-regulation of KDM6B in the prefrontal cortex and nucleus accumbens of alcohol-dependent mice, where it regulated H3K27me3 demethylation of multiple genes in the IL-6 pathway. Knockout of KDM6B significantly reduced IL-6 expression in macrophages, suggesting a close relationship between alcohol dependence and KDM6B-mediated inflammatory response.
KDM6 plays a crucial role in the regulation of macrophage polarization, in which KDM6B mainly promotes anti-inflammatory M2-type polarization, while KDM6A favors pro-inflammatory M1-type polarization. Ishii et al. [185] discovered that IL-4-stimulated macrophages induced KDM6B expression through the activation of the STAT6, which then targeted H3K27me2/3 at the promoter of M2 marker genes such as Chitinase-like 3 (Chi3l3), Resistin-like alpha (Retnla), and Arginase 1 (Arg1) and activated their expression, thus mediating IL-4-induced M2 polarization. Similarly, Satoh et al. [186] demonstrated that KDM6B was essential for M2 polarization. In chitin-stimulated or helminth-infected mouse peritoneal macrophages, KDM6B inhibited H3K27me3 at the IRF4 promoter. Conversely, knockdown of KDM6B enhanced H3K27me3 and suppressed IRF4 expression, consequently impeding the generation of M2 macrophages. Moreover, Salmonella Typhimurium infection led to increased host KDM6B expression and reduced the H3K27me3 mark. KDM6B was recruited to the promoter of WNT pathway genes, including PPARδ, and activated its transcription, thereby promoting M2 polarization of macrophages, leading to the prolonged survival of S. typhi in the intestinal tract and the formation of chronic infection [187].Additionally, KDM6B also participated in the polarization of microglial cells. In Parkinson’s disease, IL-4 induced the production of KDM6B and was associated with the microglia phenotype. KDM6B enhanced the expression of Arg 1 by modifying histone H3K27me3. It further promoted the anti-inflammatory M2-type polarization of microglia and antagonized the pro-inflammatory M1-type response, ultimately alleviating the death of dopaminergic neurons [188]. Alexaki et al. [189] observed that during the process of dehydroepiandrosterone (DHEA) inhibiting nervous system inflammation, DHEA bound to the receptor of tropomyosin receptor kinase A (TrkA) and activated Akt1/Akt2 and cAMP, inducing the expression of KDM6B, thereby regulating the polarization of microglia and suppressing the expression of M1 pro-inflammatory genes.
In contrast, KDM6A promotes M1 polarization and inhibits M2 polarization. Chen et al. [190] discovered that LPS or IL-4 stimulation did not change the expression level of KDM6A in BMDM cells. However, knocking out KDM6A significantly impeded the M1 polarization induced by LPS while facilitating the M2 polarization induced by IL-4. KDM6A was found to promote the expression of M1 markers including Nos2 and IL-6 while inhibiting the expression of M2 markers such as Arg1 and Retnla [190]. Similarly, knocking out KDM6A can enhance the expression of miR-467b-3p in ECs, which subsequently induce M2-type polarization of macrophages after spinal cord injury (SCI) through the PI3K/AKT/mTOR pathway [191].
Moreover, in a mouse model of bladder cancer, the knockout of KDM6A enhanced the secretion of pro-inflammatory factors IL-6 and CCL-2, inducing the polarization of M2 macrophages, thereby promoting bladder cancer cell proliferation by activating inflammatory pathways such as STAT3 [192]. In the periodontitis model, the expression of KDM6A protein increased in macrophages infiltrating periodontal tissue, and the disruption of circadian rhythm further upregulated KDM6A. Knockdown of KDM6A in macrophages reduced the expression of M1 cytokines including IL-6, IL-1β, and TNF-α, but did not affect the expression of M2 cytokines, indicating that KDM6A may aggravate periodontitis by inducing pro-inflammatory M1 polarization [193]. Additionally, in diabetic retinal microglia and macrophages, KDM6A promoted Lipocalin-2 (Lcn2) expression by removing H3K27me3, thereby regulating metabolic status and inhibiting the inflammatory response. However, the knockout of KDM6A reduced retinal inflammatory factors, such as TNF-α, NOS2, and inhibited M1 polarization [194]. Furthermore, Qi et al. [169] observed that the expression of KDM5C/6A in microglia in aged females was higher compare to that in males and was associated with a poor prognosis of cerebral ischemia. KDM6A promoted the expression of the IRF5 gene through H3K27 demethylation, and IRF5 signaling further induced the transcription of pro-inflammatory cytokines. Knockout of KDM6A significantly reduced the expression of inflammatory factors such as TNFα and NOS2.
KDM6A/6B can also regulate T helper cell function and contribute to the pathogenesis of autoimmune diseases. Doña et al. [195] demonstrated that inhibiting KDM6B with GSK-J4 induced immune tolerance in DC cells. The expression of co-stimulatory molecules and secretion of pro-inflammatory cytokines (IL-6, IFN-γ, and TNF-α) were reduced in GSK-J4-treated DC cells, while the secretion of anti-inflammatory mediators such as TGF-β1 was enhanced, leading to an increase in regulatory T cell (Treg) generation. Transfer of GSK-J4-treated DCs into EAE mice significantly suppressed the degree of inflammatory CD4+ T cell infiltration in the central nervous system. Similarly, knockout of KDM6A in CD4+ T cells induced Th2 differentiation and substantially reduced the neuroinflammatory response in mice by inhibiting CD44 expression through H3K27me3 demethylation. In the CD4+ T cells of EAE mice, KDM6A down-regulated the expressions of IL-2, IFN-γ, and IL-17A, but upregulated the expression of IL-5, indicating that KDM6A can participate in Th2 differentiation during autoimmune inflammation [196]. Cribbs et al. [197] found that GSK-J4-induced inhibition of KDM6A and KDM6B reduced differentiation and proliferation of Th17 and decreased the release of Th17-type proinflammatory cytokines including IL-17 and IFN-γ. GSK-J4 treatment increased the repressive H3K27me3 modification, leading to the reduced expression of vital Th17-type TFs such as MYC, PPARγ, and peroxisome proliferator-activated receptor gamma coactivator 1-related protein 1 (PPRC1), which ultimately diminished the inflammatory response in ankylotic arthritis. Additionally, KDM6 is associated with autoimmune diseases such as colitis, RA, and OA. Doñas et al. [198] showed that GSK-J4 could reduce the secretion of IL-6 and IL-17, weaken the response of Th17, thereby alleviating inflammatory colitis. Inhibition of KDM6B by GSK-J4 enhanced H3K27 methylation at the Nrf2 gene promoter and downregulated Nrf2 expression in macrophages, which is essential for NLRP3 inflammasome activation. Consequently, GSK-J4 reduced NLRP3-mediated inflammatory response and mitigated intestinal injury in mice with colitis [199]. The expression of KDM6B was upregulated in RA patients. In mice with collagen-induced arthritis(CIA), GSK-J4 could reduce the levels of pro-inflammatory factors and like IL-1β inflammatory cell infiltration by inhibiting KDM6B, thereby reducing the severity of arthritis in CIA mice [200]. In OA, KDM6B is overexpressed. GSK-J4 inhibited the NF-κB signaling pathway activated by IL-1β, reducing the production of pro-inflammatory factors such as IL-6 and IL-8, and consequently decreasing cartilage damage [201].
KDM7
The KDM7 family might also contribute to promoting inflammation, although current research in this area is limited. KDM7A was rapidly recruited to the NF-κB binding site in TNF-α-stimulated human ECs and can promote the expression of downstream inflammatory factors such as vascular cell adhesion molecule 1 (VCAM1) and selectin E (SELE) by regulating H3K9me2 [202]. In human brain microvascular ECs, KDM7A increased the stability of ICAM1 protein by regulating TFEB-mediated lysosomal activity and upregulated the expression of ICAM1, which was closely related to leukocyte migration and adsorption, thus promoting brain inflammation [203]. In hepatocellular cancer cells, KDM7B triggers ATG17/FIP200 transcription by binding with the startup domain, thereby activating autophagy. In LPS-stimulated macrophages, KDM7B may facilitate the activation of NF-κB by mediating H3K9me2 demethylation of NF-κB, promoting the expression of inflammatory factors and enhancing the activation and proliferation of T cells [204]. Furthermore, KDM7C is involved in TLR4-stimulated inflammatory responses. TLR4 signaling and the downstream NF-κB pathway can lead to the activation of KDM7C, which in turn triggers the expression of inflammatory factors such as TNF and CXCL10 in macrophages by removing the inhibitory histone H4 lysine 20 trimethylation (H4K20me3) [205].
KDMs as therapeutic targets
Autophagy is closely related with inflammatory diseases [8, 18]. On one hand, it constitutes an important component of the immune response. Through xenophagy, pathogens such as bacteria entering cells are recognized by selective autophagy receptors and transported to lysosomes for degradation, thereby playing an anti-infective role and restraining excessive inflammatory responses. On the other hand, autophagy often functions as an anti-inflammatory mechanism. Mitophagy can degrade the aged or damaged mitochondria and inhibits the activation of the NLRP3 inflammasome, thus playing an anti-inflammatory role in diseases such as chronic obstructive pulmonary disease, and inflammatory bowel diseases, and other conditions [206]. Autophagy also interrupts inflammatory pathways activated by viral DNA or RNA and negatively regulates type I interferon (IFN-I) production. Moreover, it exerts anti-apoptotic effects, inhibits pyroptosis, and promotes the clearance of necrotic cells, thereby preventing the overactivation of the inflammatory response [207]. However, effective strategies to regulate autophagy are still lacking, and the potential regulation of autophagy through epigenetic modifications is holds promise for drug development.
Evidence shown that inhibitors of KDMs, particularly KDM1A/LSD1 inhibitors, can regulate the autophagy process. Tranylcypromine (TCP, 2-PCPA) is a monoamine oxidase inhibitor and the first identified KDM1A inhibitor. It antagonizes the demethylase activity of KDM1A [75].Subsequently, selective inhibitors of KDM1A, such as SP2509 [208], GSK-lysine-specific demethylase 1 (GSK-LSD 1) [209], and S2101 [210], have been developed, with many novel inhibitors continually discovered. TCP activates the SESN2 promoter by regulating modifications such as H3K4me2, leading to the inactivation of mTOR pathway and promoting autophagy, thereby impeding the growth of cells such as neuroblastoma and ovarian cancer [211, 212]. Treatment with SP2509 promotes p62 transcription and enhances p62 protein stability, thereby activating cellular autophagy, inducing apoptosis of uterine serous cell carcinoma ARK 2 cells, and inhibiting tumor growth in mice [94, 213]. Addition of S2101 to ovarian cancer serous cystadenocarcinoma ovarian cell line 3 (SKOV3) cells inhibits the p-AKT-mTOR pathway, promoting cellular autophagy, subsequently inducing apoptosis and hindering SKOV3 cell growth [87]. In addition, novel inhibitors, such as NCL1, JL1037, and ZY0511, were all capable of inhibiting tumor growth in mouse models by promoting autophagy and apoptosis [89, 97, 214].
Within the JmjC family proteins, JIB04 is a widely used JmjC inhibitor [215]. JIB04 inhibited KDM4C, aggravates apoptosis of HEK29 cells after oxidative stress, and aggravated kidney injury caused by ischemia–reperfusion in mice [118]. Additionally, 5-Carboxy-8-Hydroxyquinoline (IOX-1) acts as a small-molecule 2-oxoglutarate (2-OG) dependent oxygenase inhibitor antagonizing the catalytic activity of JmjC proteins [216]. Using IOX-1 or si-KDM4B, inhibiting the expression of ULK1 and ATG16L1 by increasing H3K9/H3K36 levels and then inhibiting autophagy could induce apoptosis of glioma A549 cells and exert a unique anti-tumor effect [115]. However, GSK-J1/4 showed selective inhibition of KDM6 family proteins. In TNF-α-stimulated dental pulp cells, GSK-J4 inhibited the expression of autophagy genes such as LC3B, ATG5, and ATG12 and cellular autophagy in pulpitis [128].
Inhibitors of KDMs play pivotal regulatory roles in multiple inflammatory diseases. In hepatitis B (HBV)-induced glomerulonephritis, KDM1A regulated H3K9me1/2 and promoted TLR4 transcription. However, the KDM1A inhibitor TCP alleviated inflammation by downregulating TNF-α, IL-1β, and IL-6 expression in the kidney cortex by inhibiting TLR4/NF-κB signaling in the HBV mouse model [135]. For instance, the KDM6 protein inhibitor GSK-J1/4 could downregulate the inflammatory response in conditions such as inflammatory bowel disease, RA, multiple sclerosis, and mastitis [182, 183, 195, 198, 217]. KDM6B-mediated H3K27me3 modification is critical for Th17/Treg cell differentiation in ulcerative colitis mice. However, Treg cells were increased and Th17 was decreased in colon tissues following GSK-J1 treatment, leading to reduced secretion of inflammatory factors [218]. In the context of inflammatory diseases, inhibitors of KDMs have the potential to modulate inflammatory responses by regulating autophagy. SESN2 activation in si-LSD1 treated macrophages promotes autophagy and inhibits the activation of ox-LDL-induced NLRP3 inflammasome and the release of proinflammatory factors such as IL-1β, IL-6, and TNF-α [90].Furthermore, KDM4D can be regulated by OPTN-mediated selective autophagy in macrophages in experimental allergic encephalomyelitis (EAE) mice, while IOX-1 mitigates inflammatory symptoms in KMD4D by increasing H3K9me3 modification in IL-12b, IL-12a, and IL-23a promoters, thereby inhibiting EAE in mice [119].
Therefore, the potential clinical applications of KDMs inhibitors in regulating autophagy and inflammation are evident. Nevertheless, several issues remain to be addressed. Firstly, our understanding of the roles and mechanisms of autophagy in inflammation is not yet complete. Secondly, existing evidence indicates that the regulatory effects of KDMs on autophagy and inflammation vary across different disease states, tissues, and organs. Further trials are necessary to explore the specific conditions under which KDMs inhibitors are most suitable for use. In addition, some KDM1A inhibitors have been used in clinical studies of tumors and hematological diseases [219, 220]. However, additional clinical trials are need to validate the effectiveness and safety of other KDM inhibitors.
Conclusion and future perspectives
Under different external conditions, the KDMs family affects multiple signaling pathways through the demethylation modification on histones, thereby regulating autophagy and the secretion of inflammatory factors. Accumulating studies have shown that KDMs play a significant role in the development of various diseases, thus small molecule inhibitors related to KDMs, aimed at delaying disease progression and treating diseases, have gained increasing recognition. Substantial evidence has elucidated the mechanisms by which KDMs regulate autophagy in different diseases. In addition, KDMs can also regulate inflammation-related genes to influence the of inflammatory progression. Current studies have provided a solid theoretical basis for the treatment of KDMs protein inhibitors in inflammatory diseases. However, there are some obstacles that need to be overcome before their extensive application in clinical practice. Firstly, the altered expression of KDMs in cells, whether increased or decreased, can serve as a marker for the activation of autophagy or inflammation activation. For example, KDM1A can inhibit autophagy and promote gynecological tumors. However, it can also enhance the autophagic degradation of p-Tau, therefore slowing down the progression of AD. However, KDM6A can both positively and negatively regulate the inflammatory response. The behavior of KDMs varies across different cells. Therefore, understanding the unique efficiency of each KDM protein in autophagy and inflammation under different conditions is crucial for the targeted therapy of inflammatory diseases. Secondly, although the various active domains of the KDMs family have been elucidated, there are still obstacles to the successful construction of effective JMJD inhibitors due to the high polarity of the 2-OG binding pocket of the JMJD family. The efficacy of some developed JMJD inhibitors that have been developed so far is not very high. Furthermore, with the continuous development of research on KDMs inhibitors, there have been no inhibitors that can be universally applied for autophagy regulation in diverse cells or for various inflammatory diseases currently (Table 1). The impact of interactions between various KDMs proteins on disease makes the development of effective targeted drugs more challenging. For example, JMJD2 inhibitors have been used in cancer treatments, but only one drug is currently undergoing clinical evaluation. To screen for the interference of non-selective target interference and improve the precision of targeted therapy with KDMs inhibitors, further investigation into the structural and functional information of KDMs proteins and protein interactions is needed.
Data availability
No datasets were generated or analysed during the current study.
Abbreviations
- KDMs:
-
Histone lysine demethylases
- ATG:
-
Autophagy-related genes
- ULK1:
-
Unc-51 like kinase 1
- LSDs:
-
Flavin adenine dinucleotide-dependent amine oxidase
- JmjC:
-
JumonjiC
- JMJD:
-
JumonjiC domain
- SESN2:
-
Sestrin2
- ox-LDL:
-
Oxidized low-density lipoprotein
- PTEN:
-
Phosphatase and tensin homolog
- CREB:
-
CAMP response element binding protein
- TFEB:
-
Transcription factor EB
- METTL3:
-
Methyltransferase-like 3
- GABARAPL1:
-
GABA(A) receptor-associated protein like 1
- OPEN:
-
Optineurin
- PIK3C3:
-
Class III phosphatidylinositol 3-kinase complex
- R170me2s:
-
Arginine 170 site
- MLL3:
-
Mixed-lineage leukemia 3
- DCs:
-
Dendritic cells
- MRTF-A:
-
Myoglobin associated transcription factor A
- MCP-1:
-
Monocyte chemoattractant protein-1
- ICAM1:
-
Intercellular adhesion molecule 1
- RSV:
-
Respiratory syncytial virus
- SHE:
-
Soluble epoxide hydrolase
- DHEA:
-
Dehydroepiandrosterone
- TCP:
-
Tranylcypromine
- EAE:
-
Encephalomyelitis
- PTMs:
-
Post-translation modifications
References
Levine B, Kroemer G. Autophagy in the pathogenesis of disease. Cell. 2008;132:27–42.
Choi I, Heaton GR, Lee YK, Yue Z. Regulation of alpha-synuclein homeostasis and inflammasome activation by microglial autophagy. Sci Adv. 2022;8:eabn1298.
Boya P, Esteban-Martinez L, Serrano-Puebla A, Gomez-Sintes R, Villarejo-Zori B. Autophagy in the eye: Development, degeneration, and aging. Prog Retin Eye Res. 2016;55:206–45.
Metur SP, Klionsky DJ. Adaptive immunity at the crossroads of autophagy and metabolism. Cell Mol Immunol. 2021;18:1096–105.
Gao Y, Zheng X, Chang B, Lin Y, Huang X, Wang W, Ding S, Zhan W, Wang S, Xiao B, et al. Intercellular transfer of activated STING triggered by RAB22A-mediated non-canonical autophagy promotes antitumor immunity. Cell Res. 2022;32:1086–104.
Ma Z, Bai J, Jiang C, Zhu H, Liu D, Pan M, Wang X, Pi J, Jiang P, Liu X. Tegument protein UL21 of alpha-herpesvirus inhibits the innate immunity by triggering CGAS degradation through TOLLIP-mediated selective autophagy. Autophagy. 2023;19:1512–32.
Chung C, Seo W, Silwal P, Jo EK. Crosstalks between inflammasome and autophagy in cancer. J Hematol Oncol. 2020;13:100.
Deretic V. Autophagy in inflammation, infection, and immunometabolism. Immunity. 2021;54:437–53.
Pena-Martinez C, Rickman AD, Heckmann BL. Beyond autophagy: LC3-associated phagocytosis and endocytosis. Sci Adv. 2022;8:eabn1702.
Zhang R, Kang R, Tang D. The STING1 network regulates autophagy and cell death. Signal Transduct Target Ther. 2021;6:208.
Parzych KR, Klionsky DJ. An overview of autophagy: morphology, mechanism, and regulation. Antioxid Redox Signal. 2014;20:460–73.
Mizushima N. A brief history of autophagy from cell biology to physiology and disease. Nat Cell Biol. 2018;20:521–7.
Larabi A, Barnich N, Nguyen HTT. New insights into the interplay between autophagy, gut microbiota and inflammatory responses in IBD. Autophagy. 2020;16:38–51.
Yang Z, Klionsky DJ. Eaten alive: a history of macroautophagy. Nat Cell Biol. 2010;12:814–22.
Ambrosio S, Ballabio A, Majello B. Histone methyl-transferases and demethylases in the autophagy regulatory network: the emerging role of KDM1A/LSD1 demethylase. Autophagy. 2019;15:187–96.
Ma T, Li A, Guo Y, Li S, Li M, Feng S, Liu H. KDM1A/LSD1 as a promising target in various diseases treatment by regulating autophagy network. Biomed Pharmacother. 2022;148:112762.
Byun S, Seok S, Kim YC, Zhang Y, Yau P, Iwamori N, Xu HE, Ma J, Kemper B, Kemper JK. Fasting-induced FGF21 signaling activates hepatic autophagy and lipid degradation via JMJD3 histone demethylase. Nat Commun. 2020;11:807.
Matsuzawa-Ishimoto Y, Hwang S, Cadwell K. Autophagy and Inflammation. Annu Rev Immunol. 2018;36:73–101.
Li L, Tong M, Fu Y, Chen F, Zhang S, Chen H, Ma X, Li D, Liu X, Zhong Q. Lipids and membrane-associated proteins in autophagy. Protein Cell. 2021;12:520–44.
Chen T, Tu S, Ding L, Jin M, Chen H, Zhou H. The role of autophagy in viral infections. J Biomed Sci. 2023;30:5.
Liu S, Yao S, Yang H, Liu S, Wang Y. Autophagy: Regulator of cell death. Cell Death Dis. 2023;14:648.
Lamark T, Johansen T. Mechanisms of Selective Autophagy. Annu Rev Cell Dev Biol. 2021;37:143–69.
Onishi M, Yamano K, Sato M, Matsuda N, Okamoto K. Molecular mechanisms and physiological functions of mitophagy. EMBO J. 2021;40:e104705.
Li W, He P, Huang Y, Li YF, Lu J, Li M, Kurihara H, Luo Z, Meng T, Onishi M, et al. Selective autophagy of intracellular organelles: recent research advances. Theranostics. 2021;11:222–56.
Li X, He S, Ma B. Autophagy and autophagy-related proteins in cancer. Mol Cancer. 2020;19:12.
Kola L, Kohrt BA, Hanlon C, Naslund JA, Sikander S, Balaji M, Benjet C, Cheung EYL, Eaton J, Gonsalves P, et al. COVID-19 mental health impact and responses in low-income and middle-income countries: reimagining global mental health. Lancet Psychiatr. 2021;8:535–50.
Park JM, Lee DH, Kim DH. Redefining the role of AMPK in autophagy and the energy stress response. Nat Commun. 2023;14:2994.
Backe SJ, Sager RA, Heritz JA, Wengert LA, Meluni KA, Aran-Guiu X, Panaretou B, Woodford MR, Prodromou C, Bourboulia D, Mollapour M. Activation of autophagy depends on Atg1/Ulk1-mediated phosphorylation and inhibition of the Hsp90 chaperone machinery. Cell Rep. 2023;42:112807.
Wang L, Zhang S, Yi S, Ho MS. A new regulator of autophagy initiation in glia. Autophagy. 2024;20:207–9.
Thomas HE, Mercer CA, Carnevalli LS, Park J, Andersen JB, Conner EA, Tanaka K, Matsutani T, Iwanami A, Aronow BJ, et al. mTOR inhibitors synergize on regression, reversal of gene expression, and autophagy in hepatocellular carcinoma. Sci Transl Med. 2012;4:139ra184.
Kim J, Kundu M, Viollet B, Guan KL. AMPK and mTOR regulate autophagy through direct phosphorylation of Ulk1. Nat Cell Biol. 2011;13:132–41.
Boukhalfa A, Nascimbeni AC, Ramel D, Dupont N, Hirsch E, Gayral S, Laffargue M, Codogno P, Morel E. PI3KC2alpha-dependent and VPS34-independent generation of PI3P controls primary cilium-mediated autophagy in response to shear stress. Nat Commun. 2020;11:294.
Ye H, Gao J, Liang Z, Lin Y, Yu Q, Huang S, Jiang L. Arabidopsis ORP2A mediates ER-autophagosomal membrane contact sites and regulates PI3P in plant autophagy. Proc Natl Acad Sci U S A. 2022;119:e2205314119.
Yamamoto H, Kakuta S, Watanabe TM, Kitamura A, Sekito T, Kondo-Kakuta C, Ichikawa R, Kinjo M, Ohsumi Y. Atg9 vesicles are an important membrane source during early steps of autophagosome formation. J Cell Biol. 2012;198:219–33.
Nakatogawa H. Mechanisms governing autophagosome biogenesis. Nat Rev Mol Cell Biol. 2020;21:439–58.
Zhou C, Wu Z, Du W, Que H, Wang Y, Ouyang Q, Jian F, Yuan W, Zhao Y, Tian R, et al. Recycling of autophagosomal components from autolysosomes by the recycler complex. Nat Cell Biol. 2022;24:497–512.
Wollert T. Autophagy. Curr Biol. 2019;29:R671–7.
Quiles JM, Najor RH, Gonzalez E, Jeung M, Liang W, Burbach SM, Zumaya EA, Diao RY, Lampert MA, Gustafsson AB. Deciphering functional roles and interplay between Beclin1 and Beclin2 in autophagosome formation and mitophagy. Sci Signal. 2023;16:eabo4457.
Lai LTF, Ye H, Zhang W, Jiang L, Lau WCY. Structural biology and electron microscopy of the autophagy molecular machinery. Cells. 2019;8(12):1627.
Priem D, Huyghe J, Bertrand MJ. LC3-independent autophagy is vital to prevent TNF cytotoxicity. Autophagy. 2023;19:2585–9.
Feng Y, He D, Yao Z, Klionsky DJ. The machinery of macroautophagy. Cell Res. 2014;24:24–41.
Marshall RS, Vierstra RD. Autophagy: The Master of Bulk and Selective Recycling. Annu Rev Plant Biol. 2018;69:173–208.
Zhang P, Cheng S, Sheng X, Dai H, He K, Du Y. The role of autophagy in regulating metabolism in the tumor microenvironment. Genes Dis. 2023;10:447–56.
Kast DJ, Dominguez R. The Cytoskeleton-Autophagy Connection. Curr Biol. 2017;27:R318–26.
He M, Ding Y, Chu C, Tang J, Xiao Q, Luo ZG. Autophagy induction stabilizes microtubules and promotes axon regeneration after spinal cord injury. Proc Natl Acad Sci U S A. 2016;113:11324–9.
Orhon I, Dupont N, Pampliega O, Cuervo AM, Codogno P. Autophagy and regulation of cilia function and assembly. Cell Death Differ. 2015;22:389–97.
Itakura E, Kishi-Itakura C, Mizushima N. The hairpin-type tail-anchored SNARE syntaxin 17 targets to autophagosomes for fusion with endosomes/lysosomes. Cell. 2012;151:1256–69.
Le Guerroue F, Bunker EN, Rosencrans WM, Nguyen JT, Basar MA, Werner A, Chou TF, Wang C, Youle RJ. TNIP1 inhibits selective autophagy via bipartite interaction with LC3/GABARAP and TAX1BP1. Mol Cell. 2023;83(927–941):e928.
Tedesco B, Vendredy L, Timmerman V, Poletti A. The chaperone-assisted selective autophagy complex dynamics and dysfunctions. Autophagy. 2023;19:1619–41.
Shaid S, Brandts CH, Serve H, Dikic I. Ubiquitination and selective autophagy. Cell Death Differ. 2013;20:21–30.
Liu L, Sakakibara K, Chen Q, Okamoto K. Receptor-mediated mitophagy in yeast and mammalian systems. Cell Res. 2014;24:787–95.
Fyodorov DV, Zhou BR, Skoultchi AI, Bai Y. Emerging roles of linker histones in regulating chromatin structure and function. Nat Rev Mol Cell Biol. 2018;19:192–206.
Filipovski M, Soffers JHM, Vos SM, Farnung L. Structural basis of nucleosome retention during transcription elongation. Science. 2022;376:1313–6.
Strohkendl I, Saifuddin FA, Gibson BA, Rosen MK, Russell R, Finkelstein IJ. Inhibition of CRISPR-Cas12a DNA targeting by nucleosomes and chromatin. Sci Adv. 2021;7(11):eabd6030.
Weintraub H, Palter K, Van Lente F. Histones H2a, H2b, H3, and H4 form a tetrameric complex in solutions of high salt. Cell. 1975;6:85–110.
Foroozani M, Holder DH, Deal RB. Histone Variants in the Specialization of Plant Chromatin. Annu Rev Plant Biol. 2022;73:149–72.
Flury V, Reveron-Gomez N, Alcaraz N, Stewart-Morgan KR, Wenger A, Klose RJ, Groth A. Recycling of modified H2A–H2B provides short-term memory of chromatin states. Cell. 2023;186(1050–1065):e1019.
Nozawa K, Takizawa Y, Pierrakeas L, Sogawa-Fujiwara C, Saikusa K, Akashi S, Luk E, Kurumizaka H. Cryo-electron microscopy structure of the H3–H4 octasome: A nucleosome-like particle without histones H2A and H2B. Proc Natl Acad Sci U S A. 2022;119:e2206542119.
Jain K, Marunde MR, Burg JM, Gloor SL, Joseph FM, Poncha KF, Gillespie ZB, Rodriguez KL, Popova IK, Hall NW, et al. An acetylation-mediated chromatin switch governs H3K4 methylation read-write capability. Elife. 2023;12:e82596.
Smith R, Zentout S, Rother M, Bigot N, Chapuis C, Mihut A, Zobel FF, Ahel I, van Attikum H, Timinszky G, Huet S. HPF1-dependent histone ADP-ribosylation triggers chromatin relaxation to promote the recruitment of repair factors at sites of DNA damage. Nat Struct Mol Biol. 2023;30:678–91.
Kronlage M, Dewenter M, Grosso J, Fleming T, Oehl U, Lehmann LH, Falcao-Pires I, Leite-Moreira AF, Volk N, Grone HJ, et al. O-GlcNAcylation of Histone Deacetylase 4 Protects the Diabetic Heart From Failure. Circulation. 2019;140:580–94.
Nelson CJ, Santos-Rosa H, Kouzarides T. Proline isomerization of histone H3 regulates lysine methylation and gene expression. Cell. 2006;126:905–16.
Xiao P, Li M, Zhou M, Zhao X, Wang C, Qiu J, Fang Q, Jiang H, Dong H, Zhou R. TTP protects against acute liver failure by regulating CCL2 and CCL5 through m6A RNA methylation. JCI Insight. 2021;6(23):e149276.
McGrath J, Trojer P. Targeting histone lysine methylation in cancer. Pharmacol Ther. 2015;150:1–22.
Liao Q, Yang J, Ge S, Chai P, Fan J, Jia R. Novel insights into histone lysine methyltransferases in cancer therapy: From epigenetic regulation to selective drugs. J Pharm Anal. 2023;13:127–41.
Temimi A, Reddy YV, White PB, Guo H, Qian P, Mecinovic J. Lysine Possesses the Optimal Chain Length for Histone Lysine Methyltransferase Catalysis. Sci Rep. 2017;7:16148.
Black JC, Van Rechem C, Whetstine JR. Histone lysine methylation dynamics: establishment, regulation, and biological impact. Mol Cell. 2012;48:491–507.
Song YQ, Yang GJ, Ma DL, Wang W, Leung CH. The role and prospect of lysine-specific demethylases in cancer chemoresistance. Med Res Rev. 2023;43:1438–69.
Taylor-Papadimitriou J, Burchell JM. Histone methylases and demethylases regulating antagonistic methyl marks: Changes occurring in cancer. Cells. 2022;11(7):1113.
Hong S, Cho YW, Yu LR, Yu H, Veenstra TD, Ge K. Identification of JmjC domain-containing UTX and JMJD3 as histone H3 lysine 27 demethylases. Proc Natl Acad Sci U S A. 2007;104:18439–44.
Cui L, Fan Q, Cui L, Miao J. Histone lysine methyltransferases and demethylases in Plasmodium falciparum. Int J Parasitol. 2008;38:1083–97.
Markolovic S, Leissing TM, Chowdhury R, Wilkins SE, Lu X, Schofield CJ. Structure-function relationships of human JmjC oxygenases-demethylases versus hydroxylases. Curr Opin Struct Biol. 2016;41:62–72.
Forneris F, Battaglioli E, Mattevi A, Binda C. New roles of flavoproteins in molecular cell biology: histone demethylase LSD1 and chromatin. FEBS J. 2009;276:4304–12.
Song Y, Wang S, Yu B. Structural and Functional Landscape of FAD-Dependent Histone Lysine Demethylases for New Drug Discovery. J Med Chem. 2023;66:71–94.
Binda C, Valente S, Romanenghi M, Pilotto S, Cirilli R, Karytinos A, Ciossani G, Botrugno OA, Forneris F, Tardugno M, et al. Biochemical, structural, and biological evaluation of tranylcypromine derivatives as inhibitors of histone demethylases LSD1 and LSD2. J Am Chem Soc. 2010;132:6827–33.
Ramanan R, Chaturvedi SS, Lehnert N, Schofield CJ, Karabencheva-Christova TG, Christov CZ. Catalysis by the JmjC histone demethylase KDM4A integrates substrate dynamics, correlated motions and molecular orbital control. Chem Sci. 2020;11:9950–61.
Rotili D, Tomassi S, Conte M, Benedetti R, Tortorici M, Ciossani G, Valente S, Marrocco B, Labella D, Novellino E, et al. Pan-histone demethylase inhibitors simultaneously targeting Jumonji C and lysine-specific demethylases display high anticancer activities. J Med Chem. 2014;57:42–55.
Shen J, Xiang X, Chen L, Wang H, Wu L, Sun Y, Ma L, Gu X, Liu H, Wang L, et al. JMJD5 cleaves monomethylated histone H3 N-tail under DNA damaging stress. EMBO Rep. 2017;18:2131–43.
Oh S, Shin S, Janknecht R. The small members of the JMJD protein family: Enzymatic jewels or jinxes? Biochim Biophys Acta Rev Cancer. 2019;1871:406–18.
Bharath LP, Agrawal M, McCambridge G, Nicholas DA, Hasturk H, Liu J, Jiang K, Liu R, Guo Z, Deeney J, et al. Metformin Enhances Autophagy and Normalizes Mitochondrial Function to Alleviate Aging-Associated Inflammation. Cell Metab. 2020;32(44–55):e46.
Qian M, Fang X, Wang X. Autophagy and inflammation. Clin Transl Med. 2017;6:24.
Li SX, Li ZY, Ji JF. Surveys on diagnosis and treatment of esophagogastric junction adenocarcinoma by the Chinese Laparoscopic Gastrointestinal Surgery Study Group-10 Research Team. Zhonghua Wei Chang Wai Ke Za Zhi. 2023;26:773–9.
Suzuki T, Ozasa H, Itoh Y, Zhan P, Sawada H, Mino K, Walport L, Ohkubo R, Kawamura A, Yonezawa M, et al. Identification of the KDM2/7 histone lysine demethylase subfamily inhibitor and its antiproliferative activity. J Med Chem. 2013;56:7222–31.
Shi Y, Lan F, Matson C, Mulligan P, Whetstine JR, Cole PA, Casero RA, Shi Y. Histone demethylation mediated by the nuclear amine oxidase homolog LSD1. Cell. 2004;119:941–53.
Kim SA, Zhu J, Yennawar N, Eek P, Tan S. Crystal Structure of the LSD1/CoREST Histone Demethylase Bound to Its Nucleosome Substrate. Mol Cell. 2020;78(903–914):e904.
Metzger E, Wissmann M, Yin N, Muller JM, Schneider R, Peters AH, Gunther T, Buettner R, Schule R. LSD1 demethylates repressive histone marks to promote androgen-receptor-dependent transcription. Nature. 2005;437:436–9.
Feng S, Jin Y, Cui M, Zheng J. Lysine-Specific Demethylase 1 (LSD1) Inhibitor S2101 Induces Autophagy via the AKT/mTOR Pathway in SKOV3 Ovarian Cancer Cells. Med Sci Monit. 2016;22:4742–8.
Chantranupong L, Wolfson RL, Orozco JM, Saxton RA, Scaria SM, Bar-Peled L, Spooner E, Isasa M, Gygi SP, Sabatini DM. The Sestrins interact with GATOR2 to negatively regulate the amino-acid-sensing pathway upstream of mTORC1. Cell Rep. 2014;9:1–8.
Liu H, Wei J, Sang N, Zhong X, Zhou X, Yang X, Zhang J, Zuo Z, Zhou Y, Yang S, et al. The novel LSD1 inhibitor ZY0511 suppresses diffuse large B-cell lymphoma proliferation by inducing apoptosis and autophagy. Med Oncol. 2021;38:124.
Zhuo X, Wu Y, Yang Y, Gao L, Qiao X, Chen T. Knockdown of LSD1 meliorates Ox-LDL-stimulated NLRP3 activation and inflammation by promoting autophagy via SESN2-mesiated PI3K/Akt/mTOR signaling pathway. Life Sci. 2019;233:116696.
Zhou J, Zhou H, Liu C, Huang L, Lu D, Gao C. HDAC1-mediated deacetylation of LSD1 regulates vascular calcification by promoting autophagy in chronic renal failure. J Cell Mol Med. 2020;24:8636–49.
Shi YX, He YJ, Zhou Y, Li HK, Yang D, Li RY, Deng ZL, Gao YF. LSD1 negatively regulates autophagy in myoblast cells by driving PTEN degradation. Biochem Biophys Res Commun. 2020;522:924–30.
Lin CY, Chang CB, Wu RC, Chao A, Lee YS, Tsai CN, Chen CH, Yen CF, Tsai CL. Glucose Activates Lysine-Specific Demethylase 1 through the KEAP1/p62 Pathway. Antioxidants (Basel). 2021;10(12):1898.
He M, Zhang T, Zhu Z, Qin S, Wang H, Zhao L, Zhang X, Hu J, Wen J, Cai H, et al. LSD1 contributes to programmed oocyte death by regulating the transcription of autophagy adaptor SQSTM1/p62. Aging Cell. 2020;19:e13102.
Byun S, Kim YC, Zhang Y, Kong B, Guo G, Sadoshima J, Ma J, Kemper B, Kemper JK. A postprandial FGF19-SHP-LSD1 regulatory axis mediates epigenetic repression of hepatic autophagy. EMBO J. 2017;36:1755–69.
Wang Z, Long QY, Chen L, Fan JD, Wang ZN, Li LY, Wu M, Chen X. Inhibition of H3K4 demethylation induces autophagy in cancer cell lines. Biochim Biophys Acta Mol Cell Res. 2017;1864:2428–37.
Etani T, Naiki T, Naiki-Ito A, Suzuki T, Iida K, Nozaki S, Kato H, Nagayasu Y, Suzuki S, Kawai N, et al. NCL1, a highly selective lysine-specific demethylase 1 inhibitor, suppresses castration-resistant prostate cancer growth via regulation of apoptosis and autophagy. J Clin Med. 2019;8(4):442.
Wang H, Liu F. LSD1 silencing inhibits the proliferation, migration, invasion, and epithelial-to-mesenchymal transition of hypopharyngeal cancer cells by inducing autophagy and pyroptosis. Chin J Physiol. 2023;66:162–70.
Tang Z, Cao J, Yao J, Fan X, Zhao J, Zhao M, Duan Q, Han B, Duan S. KDM1A-mediated upregulation of METTL3 ameliorates Alzheimer’s disease via enhancing autophagic clearance of p-Tau through m6A-dependent regulation of STUB1. Free Radic Biol Med. 2023;195:343–58.
Perez-Pepe M, Desotell AW, Li H, Li W, Han B, Lin Q, Klein DE, Liu Y, Goodarzi H, Alarcon CR. 7SK methylation by METTL3 promotes transcriptional activity. Sci Adv. 2023;9:eade7500.
Song H, Feng X, Zhang M, Jin X, Xu X, Wang L, Ding X, Luo Y, Lin F, Wu Q, et al. Crosstalk between lysine methylation and phosphorylation of ATG16L1 dictates the apoptosis of hypoxia/reoxygenation-induced cardiomyocytes. Autophagy. 2018;14:825–44.
Intlekofer AM, Dematteo RG, Venneti S, Finley LW, Lu C, Judkins AR, Rustenburg AS, Grinaway PB, Chodera JD, Cross JR, Thompson CB. Hypoxia Induces Production of L-2-Hydroxyglutarate. Cell Metab. 2015;22:304–11.
Tzatsos A, Paskaleva P, Ferrari F, Deshpande V, Stoykova S, Contino G, Wong KK, Lan F, Trojer P, Park PJ, Bardeesy N. KDM2B promotes pancreatic cancer via Polycomb-dependent and -independent transcriptional programs. J Clin Invest. 2013;123:727–39.
Kang JY, Kim JY, Kim KB, Park JW, Cho H, Hahm JY, Chae YC, Kim D, Kook H, Rhee S, et al. KDM2B is a histone H3K79 demethylase and induces transcriptional repression via sirtuin-1-mediated chromatin silencing. FASEB J. 2018;32:5737–50.
Song W, Zeng Z, Zhang Y, Li H, Cheng H, Wang J, Wu F. CircRNF144B/miR-342-3p/FBXL11 axis reduced autophagy and promoted the progression of ovarian cancer by increasing the ubiquitination of Beclin-1. Cell Death Dis. 2022;13:857.
Zhao E, Tang C, Jiang X, Weng X, Zhong X, Zhang D, Hou J, Wang F, Huang M, Cui H. Inhibition of cell proliferation and induction of autophagy by KDM2B/FBXL10 knockdown in gastric cancer cells. Cell Signal. 2017;36:222–9.
Xie Z, Li H, Zang J. Knockdown of lysine (K)-specific demethylase 2B KDM2B inhibits glycolysis and induces autophagy in lung squamous cell carcinoma cells by regulating the phosphatidylinositol 3-kinase/AKT/mammalian target of rapamycin pathway. Bioengineered. 2021;12:12227–35.
Brauchle M, Yao Z, Arora R, Thigale S, Clay I, Inverardi B, Fletcher J, Taslimi P, Acker MG, Gerrits B, et al. Protein complex interactor analysis and differential activity of KDM3 subfamily members towards H3K9 methylation. PLoS ONE. 2013;8:e60549.
Kim J, Choi SA, Kim J, Kim H, Baek SH. Lysine-specific demethylase 3A is important for autophagic occurrence. Biochem Biophys Res Commun. 2020;526:176–83.
Jung H, Seo SB. Histone lysine demethylase 3B (KDM3B) regulates the propagation of autophagy via transcriptional activation of autophagy-related genes. PLoS ONE. 2020;15:e0236403.
Song Y, Zhang J, Wang H, Wang H, Liu Y, Hu Z. Histone lysine demethylase 3B regulates autophagy via transcriptional regulation of GABARAPL1 in acute myeloid leukemia cells. Int J Oncol. 2023;63(1):87.
Young LC, Hendzel MJ. The oncogenic potential of Jumonji D2 (JMJD2/KDM4) histone demethylase overexpression. Biochem Cell Biol. 2013;91:369–77.
Colmenares SU, Swenson JM, Langley SA, Kennedy C, Costes SV, Karpen GH. Drosophila Histone Demethylase KDM4A Has Enzymatic and Non-enzymatic Roles in Controlling Heterochromatin Integrity. Dev Cell. 2017;42(156–169):e155.
Wang B, Fan X, Ma C, Lei H, Long Q, Chai Y. Downregulation of KDM4A Suppresses the Survival of Glioma Cells by Promoting Autophagy. J Mol Neurosci. 2016;60:137–44.
Duan L, Perez RE, Lai X, Chen L, Maki CG. The histone demethylase JMJD2B is critical for p53-mediated autophagy and survival in Nutlin-treated cancer cells. J Biol Chem. 2019;294:9186–97.
Tan J, Wang HL, Yang J, Liu QQ, Li CM, Wang YQ, Fu LN, Gao QY, Chen YX, Fang JY. JMJD2B-induced amino acid alterations enhance the survival of colorectal cancer cells under glucose-deprivation via autophagy. Theranostics. 2020;10:5763–77.
Sha J, Han Q, Chi C, Zhu Y, Pan J, Dong B, Huang Y, Xia W, Xue W. Upregulated KDM4B promotes prostate cancer cell proliferation by activating autophagy. J Cell Physiol. 2020;235:2129–38.
Pan HC, Chen YH, Fang WC, Wu VC, Sun CY. Essential roles of the histone demethylase KDM4C in renal development and acute kidney injury. Int J Mol Sci. 2022,23(16):9318.
Liu D, Zhao Z, She Y, Zhang L, Chen X, Ma L, Cui J. TRIM14 inhibits OPTN-mediated autophagic degradation of KDM4D to epigenetically regulate inflammation. Proc Natl Acad Sci U S A. 2022;119(7):e2113454119.
Han D, Schaffner SH, Davies JP, Benton ML, Plate L, Nordman JT. BRWD3 promotes KDM5 degradation to maintain H3K4 methylation levels. Proc Natl Acad Sci U S A. 2023;120:e2305092120.
Plch J, Hrabeta J, Eckschlager T. KDM5 demethylases and their role in cancer cell chemoresistance. Int J Cancer. 2019;144:221–31.
Wang X, Gu M, Ju Y, Zhou J. Overcoming radio-resistance in esophageal squamous cell carcinoma via hypermethylation of PIK3C3 promoter region mediated by KDM5B loss. J Radiat Res. 2022;63:331–41.
Li J, Zhang T, Ren T, Liao X, Hao Y, Lim JS, Lee JH, Li M, Shao J, Liu R. Oxygen-sensitive methylation of ULK1 is required for hypoxia-induced autophagy. Nat Commun. 2022;13:1172.
Liu L, Cui J, Zhao Y, Liu X, Chen L, Xia Y, Wang Y, Chen S, Sun S, Shi B, Zou Y. KDM6A-ARHGDIB axis blocks metastasis of bladder cancer by inhibiting Rac1. Mol Cancer. 2021;20:77.
Zhang X, Liu L, Yuan X, Wei Y, Wei X. JMJD3 in the regulation of human diseases. Protein Cell. 2019;10:864–82.
Schulz WA, Lang A, Koch J, Greife A. The histone demethylase UTX/KDM6A in cancer: Progress and puzzles. Int J Cancer. 2019;145:614–20.
Denton D, Aung-Htut MT, Lorensuhewa N, Nicolson S, Zhu W, Mills K, Cakouros D, Bergmann A, Kumar S. UTX coordinates steroid hormone-mediated autophagy and cell death. Nat Commun. 2013;4:2916.
Yin B, Ma Q, Zhao L, Song C, Wang C, Yu F, Shi Y, Ye L. Epigenetic Control of Autophagy Related Genes Transcription in Pulpitis via JMJD3. Front Cell Dev Biol. 2021;9:654958.
Wang X, Zhang C, Dong N, Xu H, Zhou Y, Hou D. E2F1-driven histone demethylase KDM6B enhances thyroid malignancy via manipulating TFEB-dependent autophagy axis. Exp Cell Res. 2023;431:113742.
Song T, Lv S, Ma X, Zhao X, Fan L, Zou Q, Li N, Yan Y, Zhang W, Sun L. TRIM28 represses renal cell carcinoma cell proliferation by inhibiting TFE3/KDM6A-regulated autophagy. J Biol Chem. 2023;299:104621.
Tsukada Y, Ishitani T, Nakayama KI. KDM7 is a dual demethylase for histone H3 Lys 9 and Lys 27 and functions in brain development. Genes Dev. 2010;24:432–7.
Zhou W, Gong L, Wu Q, Xing C, Wei B, Chen T, Zhou Y, Yin S, Jiang B, Xie H, et al. PHF8 upregulation contributes to autophagic degradation of E-cadherin, epithelial-mesenchymal transition and metastasis in hepatocellular carcinoma. J Exp Clin Cancer Res. 2018;37:215.
Tao H, Zhang Y, Li J, Liu J, Yuan T, Wang W, Liang H, Zhang E, Huang Z. Oncogenic lncRNA BBOX1-AS1 promotes PHF8-mediated autophagy and elicits sorafenib resistance in hepatocellular carcinoma. Mol Ther Oncolytics. 2023;28:88–103.
Witucki L, Jakubowski H. Homocysteine metabolites inhibit autophagy, elevate amyloid beta, and induce neuropathy by impairing Phf8/H4K20me1-dependent epigenetic regulation of mTOR in cystathionine beta-synthase-deficient mice. J Inherit Metab Dis. 2023;46:1114–30.
Yang YT, Wang X, Zhang YY, Yuan WJ. The histone demethylase LSD1 promotes renal inflammation by mediating TLR4 signaling in hepatitis B virus-associated glomerulonephritis. Cell Death Dis. 2019;10:278.
Xie L, Ding N, Zhang H, Liu K, Xiong J, Ma S, Yang A, Zhang H, Jiang Y. SNF5 promotes IL-1beta expression via H3K4me1 in atherosclerosis induced by homocysteine. Int J Biochem Cell Biol. 2021;135:105974.
Manea SA, Vlad ML, Lazar AG, Muresian H, Simionescu M, Manea A. Pharmacological Inhibition of Lysine-Specific Demethylase 1A Reduces Atherosclerotic Lesion Formation in Apolipoprotein E-Deficient Mice by a Mechanism Involving Decreased Oxidative Stress and Inflammation; Potential Implications in Human Atherosclerosis. Antioxidants (Basel). 2022;11(12):2382.
Jingjing W, Zhikai W, Xingyi Z, Peixuan L, Yiwu F, Xia W, Youpeng S, Ershun Z, Zhengtao Y. Lysine-specific demethylase 1 (LSD1) serves as an potential epigenetic determinant to regulate inflammatory responses in mastitis. Int Immunopharmacol. 2021;91:107324.
Liu W, Fan JB, Xu DW, Zhu XH, Yi H, Cui SY, Zhang J, Cui ZM. Knockdown of LSD1 ameliorates the severity of rheumatoid arthritis and decreases the function of CD4 T cells in mouse models. Int J Clin Exp Pathol. 2018;11:333–41.
Kim D, Nam HJ, Lee W, Yim HY, Ahn JY, Park SW, Shin HR, Yu R, Won KJ, Bae JS, et al. PKCalpha-LSD1-NF-kappaB-Signaling Cascade Is Crucial for Epigenetic Control of the Inflammatory Response. Mol Cell. 2018;69(398–411):e396.
Hanzu FA, Musri MM, Sanchez-Herrero A, Claret M, Esteban Y, Kaliman P, Gomis R, Parrizas M. Histone demethylase KDM1A represses inflammatory gene expression in preadipocytes. Obesity (Silver Spring). 2013;21:E616-625.
van Essen D, Zhu Y, Saccani S. A feed-forward circuit controlling inducible NF-kappaB target gene activation by promoter histone demethylation. Mol Cell. 2010;39:750–60.
Liu CC, Sun C, Zheng X, Zhao MQ, Kong F, Xu FL, Chen XJ, Wang XX, Zhang M, Xia M. Regulation of KDM2B and Brg1 on Inflammatory Response of Nasal Mucosa in CRSwNP. Inflammation. 2019;42:1389–400.
Lu T, Jackson MW, Wang B, Yang M, Chance MR, Miyagi M, Gudkov AV, Stark GR. Regulation of NF-kappaB by NSD1/FBXL11-dependent reversible lysine methylation of p65. Proc Natl Acad Sci U S A. 2010;107:46–51.
Kim DH, Choi MR, Lee JK, Hong DK, Jung KE, Choi CW, Lee Y, Kim CD, Seo YJ, Lee JH. Possible Role of Lysine Demethylase 2A in the Pathophysiology of Psoriasis. Ann Dermatol. 2020;32:481–6.
Chen L, Zhang J, Zou Y, Wang F, Li J, Sun F, Luo X, Zhang M, Guo Y, Yu Q, et al. Kdm2a deficiency in macrophages enhances thermogenesis to protect mice against HFD-induced obesity by enhancing H3K36me2 at the Pparg locus. Cell Death Differ. 2021;28:1880–99.
Wang K, Li F, Yuan Y, Shan L, Cui Y, Qu J, Lian F. Synovial Mesenchymal Stem Cell-Derived EV-Packaged miR-31 Downregulates Histone Demethylase KDM2A to Prevent Knee Osteoarthritis. Mol Ther Nucleic Acids. 2020;22:1078–91.
Yang H, Li G, Han N, Zhang X, Cao Y, Cao Y, Fan Z. Secreted frizzled-related protein 2 promotes the osteo/odontogenic differentiation and paracrine potentials of stem cells from apical papilla under inflammation and hypoxia conditions. Cell Prolif. 2020;53:e12694.
Zhou Q, Zhang Y, Wang B, Zhou W, Bi Y, Huai W, Chen X, Chen Y, Liu Z, Liu X, Zhan Z. KDM2B promotes IL-6 production and inflammatory responses through Brg1-mediated chromatin remodeling. Cell Mol Immunol. 2020;17:834–42.
Zhao M, Wang S, Zuo A, Zhang J, Wen W, Jiang W, Chen H, Liang D, Sun J, Wang M. HIF-1alpha/JMJD1A signaling regulates inflammation and oxidative stress following hyperglycemia and hypoxia-induced vascular cell injury. Cell Mol Biol Lett. 2021;26:40.
Zhang BF, Jiang H, Chen J, Guo X, Hu Q, Yang S. KDM3A inhibition attenuates high concentration insulin-induced vascular smooth muscle cell injury by suppressing MAPK/NF-kappaB pathways. Int J Mol Med. 2018;41:1265–74.
Zhang B, Zhang J, Liu G, Guo X, Liu X, Chen J. KDM3A Inhibition Ameliorates Hyperglycemia-Mediated Myocardial Injury by Epigenetic Modulation of Nuclear Factor Kappa-B/P65. Front Cardiovasc Med. 2022;9:870999.
Chang L, Wang Q, Ju J, Li Y, Cai Q, Hao L, Zhou Y. Magnoflorine Ameliorates Inflammation and Fibrosis in Rats With Diabetic Nephropathy by Mediating the Stability of Lysine-Specific Demethylase 3A. Front Physiol. 2020;11:580406.
Liu L, Zhao Q, Kong M, Mao L, Yang Y, Xu Y. Myocardin-related transcription factor A regulates integrin beta 2 transcription to promote macrophage infiltration and cardiac hypertrophy in mice. Cardiovasc Res. 2022;118:844–58.
Zhang X, Liu S, Weng X, Wu T, Yu L, Xu Y, Guo J. Brg1 trans-activates endothelium-derived colony stimulating factor to promote calcium chloride induced abdominal aortic aneurysm in mice. J Mol Cell Cardiol. 2018;125:6–17.
Zuo X, Morris JS, Shureiqi I. Chromatin modification requirements for 15-lipoxygenase-1 transcriptional reactivation in colon cancer cells. J Biol Chem. 2008;283:31341–7.
Lee JY, Mehrazarin S, Alshaikh A, Kim S, Chen W, Lux R, Gwack Y, Kim RH, Kang MK. Histone Lys demethylase KDM3C demonstrates anti-inflammatory effects by suppressing NF-kappaB signaling and osteoclastogenesis. FASEB J. 2019;33:10515–27.
Liu D, Jin S, Cui J. The TRIM14-USP14-BRCC3 complex epigenetically regulates inflammation through inhibiting OPTN-mediated autophagic degradation of KDM4D. Autophagy. 2022;18:2001–2.
Wang X, Wang S, Yao G, Yu D, Chen K, Tong Q, Ye L, Wu C, Sun Y, Li H, et al. Identification of the histone lysine demethylase KDM4A/JMJD2A as a novel epigenetic target in M1 macrophage polarization induced by oxidized LDL. Oncotarget. 2017;8:114442–56.
Zhang Y, Yuan Y, Li Z, Chen H, Fang M, Xiao P, Xu Y. An interaction between BRG1 and histone modifying enzymes mediates lipopolysaccharide-induced proinflammatory cytokines in vascular endothelial cells. J Cell Biochem. 2019;120:13216–25.
Yang Y, Feng K, Yuan L, Liu Y, Zhang M, Guo K, Yin Z, Wang W, Zhou S, Sun H, et al. Compound Danshen Dripping Pill inhibits hypercholesterolemia/atherosclerosis-induced heart failure in ApoE and LDLR dual deficient mice via multiple mechanisms. Acta Pharm Sin B. 2023;13:1036–52.
Ma LL, Liu HM, Liu XM, Yuan XY, Xu C, Wang F, Lin JZ, Xu RC, Zhang DK. Screening S protein - ACE2 blockers from natural products: Strategies and advances in the discovery of potential inhibitors of COVID-19. Eur J Med Chem. 2021;226:113857.
Das ND, Choi MR, Jung KH, Park JH, Lee HT, Das A, Kim SH, Chai YG. Functional analysis of histone demethylase Jmjd2b on lipopolysaccharide-treated murine neural stem cells (NSCs). Neurotox Res. 2013;23:154–65.
Choi JY, Yoon SS, Kim SE, Ahn Jo S. KDM4B histone demethylase and G9a regulate expression of vascular adhesion proteins in cerebral microvessels. Sci Rep. 2017;7:45005.
Jin J, Xie X, Xiao Y, Hu H, Zou Q, Cheng X, Sun SC. Epigenetic regulation of the expression of Il12 and Il23 and autoimmune inflammation by the deubiquitinase Trabid. Nat Immunol. 2016;17:259–68.
Zhuo M, Chen W, Shang S, Guo P, Peng K, Li M, Mo P, Zhang Y, Qiu X, Li W, Yu C. Inflammation-induced JMJD2D promotes colitis recovery and colon tumorigenesis by activating Hedgehog signaling. Oncogene. 2020;39:3336–53.
Zhao D, Zhang Q, Liu Y, Li X, Zhao K, Ding Y, Li Z, Shen Q, Wang C, Li N, Cao X. H3K4me3 Demethylase Kdm5a Is Required for NK Cell Activation by Associating with p50 to Suppress SOCS1. Cell Rep. 2016;15:288–99.
Liu Y, Yu Y, Zhang J, Wang C. The therapeutic effect of dexmedetomidine on protection from renal failure via inhibiting KDM5A in lipopolysaccharide-induced sepsis of mice. Life Sci. 2019;239:116868.
Qi S, Al Mamun A, Ngwa C, Romana S, Ritzel R, Arnold AP, McCullough LD, Liu F. X chromosome escapee genes are involved in ischemic sexual dimorphism through epigenetic modification of inflammatory signals. J Neuroinflammation. 2021;18:70.
Chen K, Fang H, Xu N. LncRNA LOXL1-AS1 is transcriptionally activated by JUND and contributes to osteoarthritis progression via targeting the miR-423-5p/KDM5C axis. Life Sci. 2020;258:118095.
Ebadi N, Arefizadeh R, Nasrollahzadeh Sabet M, Goodarzi N. Identification of Key Genes and Biological Pathways Related to Myocardial Infarction through Integrated Bioinformatics Analysis. Iran J Med Sci. 2023;48:35–42.
Zhao Z, Su Z, Liang P, Liu D, Yang S, Wu Y, Ma L, Feng J, Zhang X, Wu C, et al. USP38 Couples Histone Ubiquitination and Methylation via KDM5B to Resolve Inflammation. Adv Sci (Weinh). 2020;7:2002680.
Ptaschinski C, Mukherjee S, Moore ML, Albert M, Helin K, Kunkel SL, Lukacs NW. RSV-Induced H3K4 Demethylase KDM5B Leads to Regulation of Dendritic Cell-Derived Innate Cytokines and Exacerbates Pathogenesis In Vivo. PLoS Pathog. 2015;11:e1004978.
Vasconez AE, Janetzko P, Oo JA, Pfluger-Muller B, Ratiu C, Gu L, Helin K, Geisslinger G, Fleming I, Schroder K, et al. The histone demethylase Jarid1b mediates angiotensin II-induced endothelial dysfunction by controlling the 3’UTR of soluble epoxide hydrolase. Acta Physiol (Oxf). 2019;225:e13168.
Salminen A, Kaarniranta K, Hiltunen M, Kauppinen A. Histone demethylase Jumonji D3 (JMJD3/KDM6B) at the nexus of epigenetic regulation of inflammation and the aging process. J Mol Med (Berl). 2014;92:1035–43.
De Santa F, Totaro MG, Prosperini E, Notarbartolo S, Testa G, Natoli G. The histone H3 lysine-27 demethylase Jmjd3 links inflammation to inhibition of polycomb-mediated gene silencing. Cell. 2007;130:1083–94.
De Santa F, Narang V, Yap ZH, Tusi BK, Burgold T, Austenaa L, Bucci G, Caganova M, Notarbartolo S, Casola S, et al. Jmjd3 contributes to the control of gene expression in LPS-activated macrophages. EMBO J. 2009;28:3341–52.
Das ND, Jung KH, Choi MR, Yoon HS, Kim SH, Chai YG. Gene networking and inflammatory pathway analysis in a JMJD3 knockdown human monocytic cell line. Cell Biochem Funct. 2012;30:224–32.
Li X, Zhang Q, Shi Q, Liu Y, Zhao K, Shen Q, Shi Y, Liu X, Wang C, Li N, et al. Demethylase Kdm6a epigenetically promotes IL-6 and IFN-beta production in macrophages. J Autoimmun. 2017;80:85–94.
Kruidenier L, Chung CW, Cheng Z, Liddle J, Che K, Joberty G, Bantscheff M, Bountra C, Bridges A, Diallo H, et al. A selective jumonji H3K27 demethylase inhibitor modulates the proinflammatory macrophage response. Nature. 2012;488:404–8.
Yang X, Xu X, Chen J, Wang Q, Wang G, Ai X, Wang X, Pan J. Zoledronic acid regulates the synthesis and secretion of IL-1beta through Histone methylation in macrophages. Cell Death Discov. 2020;6:47.
Cribbs A, Hookway ES, Wells G, Lindow M, Obad S, Oerum H, Prinjha RK, Athanasou N, Sowman A, Philpott M, et al. Inhibition of histone H3K27 demethylases selectively modulates inflammatory phenotypes of natural killer cells. J Biol Chem. 2018;293:2422–37.
Wang JJ, Wang X, Xian YE, Chen ZQ, Sun YP, Fu YW, Wu ZK, Li PX, Zhou ES, Yang ZT. The JMJD3 histone demethylase inhibitor GSK-J1 ameliorates lipopolysaccharide-induced inflammation in a mastitis model. J Biol Chem. 2022;298:102017.
Johnstone AL, Andrade NS, Barbier E, Khomtchouk BB, Rienas CA, Lowe K, Van Booven DJ, Domi E, Esanov R, Vilca S, et al. Dysregulation of the histone demethylase KDM6B in alcohol dependence is associated with epigenetic regulation of inflammatory signaling pathways. Addict Biol. 2021;26:e12816.
Ishii M, Wen H, Corsa CA, Liu T, Coelho AL, Allen RM, Carson WFt, Cavassani KA, Li X, Lukacs NW, et al. Epigenetic regulation of the alternatively activated macrophage phenotype. Blood. 2009;114(15):3244-54.
Satoh T, Takeuchi O, Vandenbon A, Yasuda K, Tanaka Y, Kumagai Y, Miyake T, Matsushita K, Okazaki T, Saitoh T, et al. The Jmjd3-Irf4 axis regulates M2 macrophage polarization and host responses against helminth infection. Nat Immunol. 2010;11:936–44.
Rana S, Maurya S, Mohapatra G, Singh S, Babar R, Chandrasekhar H, Chamoli G, Rathore D, Kshetrapal P, Srikanth CV. Activation of epigenetic regulator KDM6B by Salmonella Typhimurium enables chronic infections. Gut Microbes. 2021;13:1986665.
Tang Y, Li T, Li J, Yang J, Liu H, Zhang XJ, Le W. Jmjd3 is essential for the epigenetic modulation of microglia phenotypes in the immune pathogenesis of Parkinson’s disease. Cell Death Differ. 2014;21:369–80.
Alexaki VI, Fodelianaki G, Neuwirth A, Mund C, Kourgiantaki A, Ieronimaki E, Lyroni K, Troullinaki M, Fujii C, Kanczkowski W, et al. DHEA inhibits acute microglia-mediated inflammation through activation of the TrkA-Akt1/2-CREB-Jmjd3 pathway. Mol Psychiatry. 2018;23:1410–20.
Chen J, Xu X, Li Y, Li F, Zhang J, Xu Q, Chen W, Wei Y, Wang X. Kdm6a suppresses the alternative activation of macrophages and impairs energy expenditure in obesity. Cell Death Differ. 2021;28:1688–704.
Peng W, Xie Y, Luo Z, Liu Y, Xu J, Li C, Qin T, Lu H, Hu J. UTX deletion promotes M2 macrophage polarization by epigenetically regulating endothelial cell-macrophage crosstalk after spinal cord injury. J Nanobiotechnology. 2023;21:225.
Kobatake K, Ikeda KI, Nakata Y, Yamasaki N, Ueda T, Kanai A, Sentani K, Sera Y, Hayashi T, Koizumi M, et al. Kdm6a Deficiency Activates Inflammatory Pathways, Promotes M2 Macrophage Polarization, and Causes Bladder Cancer in Cooperation with p53 Dysfunction. Clin Cancer Res. 2020;26:2065–79.
Ma X, Chen X, Duan Z, Wu Y, Shu J, Wu P, Zhao Y, Wang X, Wang Y. Circadian rhythm disruption exacerbates the progression of macrophage dysfunction and alveolar bone loss in periodontitis. Int Immunopharmacol. 2023;116:109796.
Wen Y, Chen X, Feng H, Wang X, Kang X, Zhao P, Zhao C, Wei Y. Kdm6a deficiency in microglia/macrophages epigenetically silences Lcn2 expression and reduces photoreceptor dysfunction in diabetic retinopathy. Metabolism. 2022;136:155293.
Donas C, Carrasco M, Fritz M, Prado C, Tejon G, Osorio-Barrios F, Manriquez V, Reyes P, Pacheco R, Bono MR, et al. The histone demethylase inhibitor GSK-J4 limits inflammation through the induction of a tolerogenic phenotype on DCs. J Autoimmun. 2016;75:105–17.
Itoh Y, Golden LC, Itoh N, Matsukawa MA, Ren E, Tse V, Arnold AP, Voskuhl RR. The X-linked histone demethylase Kdm6a in CD4+ T lymphocytes modulates autoimmunity. J Clin Invest. 2019;129:3852–63.
Cribbs AP, Terlecki-Zaniewicz S, Philpott M, Baardman J, Ahern D, Lindow M, Obad S, Oerum H, Sampey B, Mander PK, et al. Histone H3K27me3 demethylases regulate human Th17 cell development and effector functions by impacting on metabolism. Proc Natl Acad Sci U S A. 2020;117:6056–66.
Donas C, Neira J, Osorio-Barrios F, Carrasco M, Fernandez D, Prado C, Loyola A, Pacheco R, Rosemblatt M. The demethylase inhibitor GSK-J4 limits inflammatory colitis by promoting de novo synthesis of retinoic acid in dendritic cells. Sci Rep. 2021;11:1342.
Huang M, Wang Q, Long F, Di Y, Wang J, Zhun Zhu Y, Liu X. Jmjd3 regulates inflammasome activation and aggravates DSS-induced colitis in mice. FASEB J. 2020;34:4107–19.
Jia W, Wu W, Yang D, Xiao C, Su Z, Huang Z, Li Z, Qin M, Huang M, Liu S, et al. Histone demethylase JMJD3 regulates fibroblast-like synoviocyte-mediated proliferation and joint destruction in rheumatoid arthritis. FASEB J. 2018;32:4031–42.
Jun Z, Xinmeng J, Yue L, Zhi W, Yan Z, Tieyi Y, Jiangan T. Jumonji domain containing-3 (JMJD3) inhibition attenuates IL-1beta-induced chondrocytes damage in vitro and protects osteoarthritis cartilage in vivo. Inflamm Res. 2020;69:657–66.
Higashijima Y, Matsui Y, Shimamura T, Nakaki R, Nagai N, Tsutsumi S, Abe Y, Link VM, Osaka M, Yoshida M, et al. Coordinated demethylation of H3K9 and H3K27 is required for rapid inflammatory responses of endothelial cells. EMBO J. 2020;39:e103949.
Choi JY, Jo SA. KDM7A histone demethylase mediates TNF-alpha-induced ICAM1 protein upregulation by modulating lysosomal activity. Biochem Biophys Res Commun. 2016;478:1355–62.
Erdogan O, Xie L, Wang L, Wu B, Kong Q, Wan Y, Chen X. Proteomic dissection of LPS-inducible, PHF8-dependent secretome reveals novel roles of PHF8 in TLR4-induced acute inflammation and T cell proliferation. Sci Rep. 2016;6:24833.
Stender JD, Pascual G, Liu W, Kaikkonen MU, Do K, Spann NJ, Boutros M, Perrimon N, Rosenfeld MG, Glass CK. Control of proinflammatory gene programs by regulated trimethylation and demethylation of histone H4K20. Mol Cell. 2012;48:28–38.
Saitoh T, Fujita N, Jang MH, Uematsu S, Yang BG, Satoh T, Omori H, Noda T, Yamamoto N, Komatsu M, et al. Loss of the autophagy protein Atg16L1 enhances endotoxin-induced IL-1beta production. Nature. 2008;456:264–8.
Maiuri MC, Zalckvar E, Kimchi A, Kroemer G. Self-eating and self-killing: crosstalk between autophagy and apoptosis. Nat Rev Mol Cell Biol. 2007;8:741–52.
Fiskus W, Sharma S, Shah B, Portier BP, Devaraj SG, Liu K, Iyer SP, Bearss D, Bhalla KN. Highly effective combination of LSD1 (KDM1A) antagonist and pan-histone deacetylase inhibitor against human AML cells. Leukemia. 2014;28:2155–64.
Mohammad HP, Smitheman KN, Kamat CD, Soong D, Federowicz KE, Van Aller GS, Schneck JL, Carson JD, Liu Y, Butticello M, et al. A DNA Hypomethylation Signature Predicts Antitumor Activity of LSD1 Inhibitors in SCLC. Cancer Cell. 2015;28:57–69.
Mimasu S, Umezawa N, Sato S, Higuchi T, Umehara T, Yokoyama S. Structurally designed trans-2-phenylcyclopropylamine derivatives potently inhibit histone demethylase LSD1/KDM1. Biochemistry. 2010;49:6494–503.
Ambrosio S, Sacca CD, Amente S, Paladino S, Lania L, Majello B. Lysine-specific demethylase LSD1 regulates autophagy in neuroblastoma through SESN2-dependent pathway. Oncogene. 2017;36:6701–11.
Wei Y, Han T, Wang R, Wei J, Peng K, Lin Q, Shao G. LSD1 negatively regulates autophagy through the mTOR signaling pathway in ovarian cancer cells. Oncol Rep. 2018;40:425–33.
Chao A, Lin CY, Chao AN, Tsai CL, Chen MY, Lee LY, Chang TC, Wang TH, Lai CH, Wang HS. Lysine-specific demethylase 1 (LSD1) destabilizes p62 and inhibits autophagy in gynecologic malignancies. Oncotarget. 2017;8:74434–50.
Liu S, Lu W, Li S, Li S, Liu J, Xing Y, Zhang S, Zhou JZ, Xing H, Xu Y, et al. Identification of JL1037 as a novel, specific, reversible lysine-specific demethylase 1 inhibitor that induce apoptosis and autophagy of AML cells. Oncotarget. 2017;8:31901–14.
Wang L, Chang J, Varghese D, Dellinger M, Kumar S, Best AM, Ruiz J, Bruick R, Pena-Llopis S, Xu J, et al. A small molecule modulates Jumonji histone demethylase activity and selectively inhibits cancer growth. Nat Commun. 2013;4:2035.
Schiller R, Scozzafava G, Tumber A, Wickens JR, Bush JT, Rai G, Lejeune C, Choi H, Yeh TL, Chan MC, et al. A cell-permeable ester derivative of the JmjC histone demethylase inhibitor IOX1. ChemMedChem. 2014;9:566–71.
Abu-Hanna J, Patel JA, Anastasakis E, Cohen R, Clapp LH, Loizidou M, Eddama MMR. Therapeutic potential of inhibiting histone 3 lysine 27 demethylases: a review of the literature. Clin Epigenetics. 2022;14:98.
Leng XY, Yang J, Fan H, Chen QY, Cheng BJ, He HX, Gao F, Zhu F, Yu T, Liu YJ. JMJD3/H3K27me3 epigenetic modification regulates Th17/Treg cell differentiation in ulcerative colitis. Int Immunopharmacol. 2022;110:109000.
Fang Y, Liao G, Yu B. LSD1/KDM1A inhibitors in clinical trials: advances and prospects. J Hematol Oncol. 2019;12:129.
Antonijoan RM, Ferrero-Cafiero JM, Coimbra J, Puntes M, Martinez-Colomer J, Arevalo MI, Mascaro C, Molinero C, Buesa C, Maes T. First-in-Human Randomized Trial to Assess Safety, Tolerability, Pharmacokinetics and Pharmacodynamics of the KDM1A Inhibitor Vafidemstat. CNS Drugs. 2021;35:331–44.
Acknowledgements
Not applicable.
Funding
This article was supported by the “National Natural Science Foundation of China” (No.82172301), “Hubei University of Science and Technology Development Fund Project” (No. BK202442), Foundation of Hubei University of Science and Technology Science “Special Project on Diabetes and Angiopathy” (No.2024TNB05), “Natural Science Foundation of Hubei Province, China” (No.2024AFB517, No.2024AFB505) “Innovation Team of Hubei University of Science and Technology” (2023T13), and Guidance Project of Hubei Provincial Education Department (B2022189).
Author information
Authors and Affiliations
Contributions
LHQ designed and supervised the manuscript. YG, WTL, TY and YYM were writing the manuscript and depicting the figures and tables. YJB, ZQL, CC, WJY, JYF, WBQ, RLT, YTS and SGS reviewed the manuscript and revised the references. LHQ, HFD and YFB revised and edited the manuscript. YG, WTL, TY and YYM contributed equally to this manuscript. All authors have read and approved the final manuscript.
Corresponding authors
Ethics declarations
Ethics approval and consent to participate
Not applicable.
Consent for publication
Not applicable.
Competing interests
The authors declare no competing interests.
Additional information
Publisher’s Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.
About this article
Cite this article
Ma, Y., Lv, W., Guo, Y. et al. Histone demethylases in autophagy and inflammation. Cell Commun Signal 23, 24 (2025). https://doi.org/10.1186/s12964-024-02006-w
Received:
Accepted:
Published:
DOI: https://doi.org/10.1186/s12964-024-02006-w