Open Access

Studies on the functional role of UFMylation in cells (Review)

  • Authors:
    • Rong Qin
    • Yu Tang
    • Yuhang Yuan
    • Fangyu Meng
    • Kepu Zheng
    • Xingyu Yang
    • Jiumei Zhao
    • Chuanhua Yang
  • View Affiliations

  • Published online on: May 2, 2025     https://doi.org/10.3892/mmr.2025.13556
  • Article Number: 191
  • Copyright: © Qin et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

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


Abstract

Protein post‑translational modifications (PTMs) play crucial roles in various life activities and aberrant protein modifications are closely associated with numerous major human diseases. Ubiquitination, the first identified protein modification system, involves the covalent attachment of ubiquitin molecules to lysine residues of target proteins. UFMylation, a recently discovered ubiquitin‑like modification, shares similarities with ubiquitination. The precursor form of ubiquitin fold modifier 1 (UFM1) undergoes synthesis and cleavage by UFM1‑specific protease 1 or UFM1‑specific protease 2 to generate activated UFM1‑G83. Subsequently, UFM1‑G83 is activated by a specific E1‑like activase, UFM1‑activating enzyme 5. UFM1‑conjugating enzyme 1 and an E3‑like ligase, UFM1‑specific ligase 1, recognize the target protein and facilitate UFMylation, leading to the degradation of the target protein. Current knowledge regarding UFMylation remains limited. Previous studies have demonstrated that defects in the UFMylation pathway can result in embryonic lethality in mice and various human diseases, highlighting the critical biological functions of UFMylation. However, the precise mechanisms underlying UFMylation remain elusive. This present review aimed to summarize recent research advances in UFMylation, with the aim of providing novel insights and perspectives for future investigations into this essential protein modification system.

Introduction

Post-translational modifications (PTMs) refer to the covalent alterations of proteins that occur during or after their biosynthesis, typically catalyzed by enzymes (1). These modifications encompass the addition of functional groups (such as phosphorylation, methylation and acetylation), the conjugation of other proteins or peptides (such as ubiquitination), chemical modifications of amino acid (AA) residues (such as deamidation) and structural changes (such as disulfide bridge formation) (2,3). Ubiquitination, one of the most prevalent PTMs, involves a highly conserved protein composed of 76 amino acids with a molecular weight of approximately 8.5 kDa, which is ubiquitously present in all eukaryotic cells (4,5). Ubiquitin is conjugated to its target proteins through a complex enzymatic reaction cascade, consisting of an activating enzyme (E1), a conjugating enzyme (E2) and a specific ligase (E3), playing a pivotal role in the regulation of eukaryotic cellular processes (6,7).

Ubiquitin fold modifier 1 (UFM1), a member of the ubiquitin-like (UBL) protein family (8,9), is composed of 85 amino acids and has a molecular weight of 9.1 kDa. Despite limited sequence homology, UFM1 and ubiquitin exhibit analogous tertiary structures and conceptually similar enzymatic pathways, involving E1, E2 and E3 enzymes, which ultimately lead to the covalent attachment of UFM1 to the lysine residues of substrates through its C-terminal glycine. Akin to other UBLs, UFM1 is synthesized as a precursor protein that requires proteolytic cleavage to generate its active mature form (10).

UFM1 possesses the ability to form polymeric chains due to the presence of five lysine residues; however, these chains predominantly connect through lysine 69 (10). In contrast to ubiquitination, which involves numerous enzymes with overlapping functions, UFMylation is characterized by a more restricted set of cellular mechanisms, demonstrating high specificity. The maturation, activation, attachment and removal of UFM1 are facilitated by only a few ubiquitously expressed enzymes, specifically UBA5 (E1), UFC1 (E2), UFL1 (E3) and UFSP1 and UFSP2 (1014).

Several proteins associated with UFMylation serve various scaffolding and targeting functions, including UFM1 binding protein-1 (UFBP1), cyclin-dependent kinase 5 regulatory subnit-associated protein 3 and retinal nerve fibre layer/ODR4. These proteins ensure that enzymatic activities are accurately directed and compartmentalized within relevant cellular environments (15). Compared with the more complex and redundant mechanisms governing ubiquitination, this unique regulatory framework underscores the specialized role of UFM1 in cellular processes (16).

UFMylation, a post-translational modification identified a decade ago (17), remains incompletely understood and its biological significance warrants further investigation. Initial studies on UFM1 substrates identified UFBP1 and ASC1 through in vitro assays (18,19); however, the majority of UFM1-modified proteins remain uncharacterized. With the advent of advanced screening technologies and purification methods, new UFM1 substrates have been identified, providing molecular-level insights into the structure and function of UFMylation (9,20,21). These discoveries have expanded its mechanistic roles in key cellular processes, including the DNA damage response, endoplasmic reticulum homeostasis, ribosomal quality control and hematopoietic differentiation, establishing UFMylation as a crucial regulator of cellular homeostasis (21,22). Its dysregulation has been linked to the development of various human diseases, for example, severe anemia (18), skeletal diseases such as Sohat spondyloepiphyseal dysplasia (SEMD) (23) and neurological diseases such as early-onset cerebellar atrophy and early-onset intractable epilepsy (24,25). The present review refined the current understanding of the enzymatic reactions underlying UFMylation, elucidated its catalytic mechanisms and explored newly emerging regulatory pathways that contribute to cellular homeostasis. Furthermore, it comprehensively elucidated the specific roles and novel molecular mechanisms of the UFM1 conjugation system in various cancers and antitumor immunity, for instance, the loss of UFL1 in T cells inhibits the UFMylation of programmed cell death protein 1 (PD-1), thereby promoting the production of effector cytokines in CD8+ T cells and enhancing their antitumor efficacy. These findings may provide new strategies for identifying cancer diagnostic biomarkers and clinical therapeutic targets in the future.

Roles of different proteases in ubiquitin-like modifications

UFM1, like most UBL proteins, is expressed as a precursor that must be proteolytically cleaved to generate the active mature form (26). However, its precursor features a unique C-terminal Ser84-Cys85 dipeptide sequence, unlike the conserved C-terminal di-Gly (Gly-Gly) motif found in most UBLs. The proteases UFSP1 and UFSP2, purified from tissue extracts using His-GST-UFM1-Escontin as a substrate, have been identified as the enzymes responsible for processing the UFM1 precursor and facilitating the removal of UFM1 modifications (27).

UFSP1, an ~25 kDa family member protein, is present in flies, mice and humans but absent in plants and nematodes. By contrast, UFSP2, which is >40 kDa, is found in most multicellular organisms, including Caenorhabditis elegans and Arabidopsis thaliana (28). Despite sharing the same catalytic mechanism, UFSP1 and UFSP2 exhibit significant structural differences, particularly in the R-helix domain responsible for recognizing and binding the UFM1 precursor. In UFSP1, the R loop connecting β3 and β4, along with Trp98, is stabilized by interactions with water molecules and residues connecting the α6 helices and β7 strands. Conversely, in UFSP2, the R loop connecting β9, β10 and Trp342 shows no significant interactions apart from hydrophobic interactions between Trp342 and Val395 (29).

Thus, a crucial function of UFSPs is to proteolytically process the UFM1 precursor, producing mature UFM1. The removal of the C-terminal serine and cysteine residues exposes the C-terminal glycine of UFM1, enabling its maturation and subsequent binding to substrates (30,31).

Upon maturation, UFM1 undergoes activation through a trans-binding mechanism involving UBA5 dimerization. UBA5, the sole E1 enzyme for UFMylation, is primarily localized in the cytoplasm. Unbound UFM1-UBA5 dimers exhibit weak dimerization; however, UFM1 binding stabilizes the UBA5 dimer conformation and enhances its affinity for ATP (32). Specifically, two UFM1 molecules and two UBA5 molecules form an interlocking structure, with each UFM1 molecule interacting with one UFM1-interacting sequence (UIS) molecule at one end, while Gly83 at the other end forms a high-energy thioester bond with Cys250 of UBA5. ATP binding to the protomers in the UBA5 dimer enables the charging of UFM1, forming an activated complex carrying two UFM1 molecules (33). The dimeric UBA5 is essential not only for UFM1 activation but also for transferring UFM1 to UFC1. This highlights the critical interdependence of the two UBA5 monomers for adenylation and UIS domain structure (13,32).

Following activation, UFM1 is transferred from UBA5 to Cys116 of UFC1 through a trans-thiolation reaction, which involves interactions between the UBA5-UFM1 dimer and UFC1 (34). Unlike UBA5, UFC1 is primarily localized in the nucleus, with only partial cytoplasmic localization. Intriguingly, while the charging process of UBA5 requires ATP, the transfer of UFM1 from UBA5 to UFC1 does not (35). During this trans-thiolation reaction, one thioester bond (UBA5-UFM1) is cleaved and another (UFC1-UFM1) is formed, suggesting that energy transfer can occur bidirectionally, with both directions being energetically equivalent (36). The reaction direction is determined by the concentrations of UBA5, UFC1 and UFM1; overexpression of UBA5 can reverse the energy transfer from activated UFM1 in UFC1 back to UBA5, indicating the reversibility of E2 transfer. In the normative direction (E1-E2), UFC1 binds to a tetrameric complex composed of two UFM1 molecules and two UBA5 molecules, employing a similar trans-binding mechanism that necessitates interaction with one UBA5 monomer and the receipt of activated UFM1 from another (37).

The final step involves the transfer of UFM1 from UFC1 to covalently link to the substrate's lysine residue, mediated by the E3 enzyme UFL1 (38). UFL1 deficiency results in the loss of UFMylation and mice lacking UFL1 exhibit hematopoietic failure and embryonic lethality, suggesting its role as the primary or sole E3 ligase. Although UFL1′s role in UFMylation is well established, it does not directly mediate the process. Instead, it forms a functional heterodimeric E3 ligase complex with adapter proteins, such as UFBP1 and CDK5RAP3, classifying it as a scaffold-type E3 ligase (16,39). Scaffold-type E3 ligases recognize substrates and facilitate UFM1 transfer from UFC1 to the substrate. The E2 enzyme confers linkage specificity at K69 during dual UFM1 formation in the absence of E3 ligase, indicating that this specificity is determined by the E2 enzyme. The E2-E3 complex can exert various effects on the substrate, ranging from proteasome-dependent protein proteolysis to the regulation of protein function, structure, assembly, or localization (40,41). Following these processes, UFSP2 can recycle the UFM1 linked to the substrate, allowing it to enter the next UFMylation cycle (42) (Fig. 1).

The functions of UFMylation in cells

The UFMylation pathway has been demonstrated to regulate a wide range of cellular activities. These include DNA damage repair, endoplasmic reticulum stress response, ribosome modification, hematopoiesis, immune regulation (20) and neurodevelopment (4347).

UFMylation and the maintenance of genomic integrity in the cell nucleus

The genome of the cell is continually exposed to attacks from both exogenous and endogenous DNA-damaging factors, such as radiation, carcinogens and reactive free radicals. To maintain genomic stability, cells have evolved a complex DNA damage response (DDR) system. This system is responsible for sensing DNA damage, halting the cell cycle and initiating repair processes. Failure to detect or repair DNA damage can lead to genomic instability, a hallmark of tumorigenesis (30).

Double-strand breaks (DSBs) represent the most toxic form of DNA damage and their repair is primarily initiated by ATM kinase (48). The activation of ATM is mediated through the meiotic recombination 11 homolog (MRE11)-radiation sensitive 50-Nijmegen breakage syndrome 1 (NBS1) (MRN) complex, which phosphorylates Ser1981 and acetylates Lys3106. Once activated, the ATM kinase rapidly phosphorylates local chromatin, providing a scaffold for the assembly of higher-order complexes that facilitate DNA repair. Subsequent studies have revealed that the activation of ATM also involves the UFMylation pathway. Following DNA damage, MRE11 undergoes UFMylation at Lys282, a modification that is crucial for the assembly of the MRN complex. This UFMylation event is essential for optimal ATM activation, homologous recombination-mediated repair and the maintenance of genomic integrity (49).

DNA damage induces the UFMylation of MRE11, which facilitates the recruitment of the MRN complex to the damage site. This process helps relieve the self-inhibition of ATM kinase at DSBs, thereby promoting DSB repair and enhancing chromosomal stability (45,46). In addition to MRE11, the UFMylation of histone H4 has also been implicated in ATM activation. The MRN complex recruits UFL1 to DNA DSBs, where UFL1 catalyzes the release of histone H4 at Lys31, enhancing the recruitment of the SUV39H1 complex to DSBs (50). SUV39H1 induces H3K9me3 modification at DSBs, which can spread over thousands of bases, forming a temporary repressive heterochromatic domain (39). Subsequently, Tip60 is recruited to bind H3K9me3 and this interaction enhances its acetyltransferase activity. This leads to the acetylation of Tip60 and subsequent activation of ATM at DSBs (51,52).

Related studies have identified the nuclear-localized UFMylated protein P53, with endogenous UFMylation being detectable in both human cancer cells and primary mouse embryonic fibroblasts. In vitro UFMylation assays have demonstrated that the UFMylation components UBA5, UFC1, UFL1, UFM1 and DDRGK1 (also known as UFM1-binding protein 1, UFBP1) are all necessary for the modification of P53 (53). The absence of the amino (N) terminal region of P53 prevents its binding to UFL1, indicating that this region is crucial for UFL1 interaction. Notably, this region can also bind to mouse double minute 2 homologue (MDM2), the primary E3 ligase responsible for P53 degradation. Consequently, UFL1 competes with MDM2 for binding to P53, thus contributing to its stability (54,55). Notably, UFMylated P53 can be detected in cells lacking UBA5 overexpression, provided that UFC1, UFL1 and DDRGK1 are overexpressed. Conversely, UFMylated P53 levels decrease in cells where UFC1, UFL1 and DDRGK1 have been knocked out, while knocking out UBA5 has no significant effect. This suggests that while UBA5 is essential for P53 UFMylation, it is not a limiting factor in the cellular context (47).

Furthermore, UFMylation has been reported to play an unanticipated role in maintaining telomere length. UFMylation-modified MRE11 promotes the recruitment of PP1-α to the MRN complex and facilitates the dephosphorylation of NBS1. This process aids in releasing the MRN complex from telomeres and allows Apollo to bind to TRF2. In the absence of UFMylation-modified MRE11, NBS1 remains phosphorylated, leading to reduced MRN recruitment at the telomeres (56,57). The absence of MRN at the telomeres favors the formation of the TRF2/SNM1 complex, ultimately resulting in the shortening of the telomeric leading strand length (44).

As a co-activator of nuclear receptors, the UFMylation modification of ASC1 is a pivotal step in the transactivation of estrogen receptor α (ERα) by 17β-estradiol (E2). In the absence of E2, the UFM1-specific protease UFSP2 binds to ASC1, maintaining it in a non-UFMylated state (43,58). However, in the presence of E2, ERα binds to ASC1, displacing UFSP2 and leading to ASC1 activation. The subsequent poly-UFMylation of ASC1 enhances the recruitment of P300, SRC1 and ASC1 to the promoters of ERα target genes. Overexpression of ASC1 or knockdown of UFSP2 promotes ERα-mediated tumor formation in vivo, which can be attenuated by treatment with the anti-breast cancer drug tamoxifen (18,43). These findings underscore the critical role of UFMylation in regulating genomic stability and its potential implications in breast cancer progression.

UFMylation and endoplasmic reticulum homeostasis

To maintain endoplasmic reticulum (ER) homeostasis, cells have evolved a sophisticated protein quality control system comprising various pathways. These pathways include ribosome-associated quality control (RQC), endoplasmic reticulum-associated degradation (ERAD), the unfolded protein response (UPR) and ER-phagy (5961).

In eukaryotic cells, a primary cause of ribosome stalling due to erroneous translation is naturally occurring faulty mRNA. These aberrant translation products can interfere with functional proteins and are detrimental to the cell. The RQC pathway effectively eliminates these stalled products in the cytoplasm when ribosomes become inoperative (62,63). This pathway employs a series of coordinated factors that sense translation stalling, rescue stalled ribosomes and degrade abnormal nascent polypeptides. Specifically, ribosome stalling during protein translocation induces the attachment of the ubiquitin-like modifier UFM1 to two conserved lysine residues near the COOH terminus of the ER 60S ribosomal subunit RPL26 (uL24) (64,65). Notably, RPL26-UFMylation facilitates the degradation of stalled nascent chains. However, unlike the ERAD or previously established RQC mechanisms utilizing proteasomal degradation, ribosomal UFMylation directs translocation-stalled ER proteins for lysosomal degradation. In summary, UFMylation plays a critical role in regulating protein homeostasis within quality control pathways (66).

ERAD, a protein degradation system located in the ER, removes unfolded or misfolded proteins for proteasomal degradation (67). Proteins that fail to achieve their native conformation are targeted for degradation through the ERAD pathway via a series of closely coupled steps: substrate recognition, retrotranslocation to the cytosol and ubiquitination for proteasomal degradation. HRD1 (HMG-CoA reductase degradation protein, also known as SYVN1) serves as one of the key ubiquitin ligases for ERAD. HRD1 has been shown to target various proteins, including the tumor suppressor tumor protein p53, programmed death-ligand 1 (PD-L1), peroxisome proliferator-activated receptor 1β, inositol-requiring enzyme 1 alpha (IRE1α), cyclic amp-responsive element-binding, lipoprotein lipase, proopiomelanocortin, sigma non-opioid intracellular receptor 1, ATP citrate lyase, nuclear factor erythroid 2-related factor 2 and the rate-limiting acyltransferases glycerol-3-phosphate acyltransferase, monoacylglycerol and diacylglycerol O-acyltransferase 2 The regulation of these proteins by HRD1 affects cellular physiological and pathological processes through the degradation of misfolded proteins (68).

Under normal conditions, cells regulate the ERAD clearance of misfolded ER-resident proteins through the UFMylation modification of HRD1. However, during ER stress, the accumulation of misfolded proteins within the ER triggers stress signals that cause the dissociation of the DDRGK1-UFL1 complex from HRD1. This dissociation diminishes HRD1 UFMylation, thereby inhibiting its function and initiating the UPR to mitigate ER stress. In this model, the removal of UFL1, HRD1, or the expression of HRD1-K610R in HRD1 knockout cells facilitates the activation of the UPR, particularly the IRE1α-XBP1 signaling pathway, which fine-tunes the folding capacity of the ER (68).

Although ER-phagy is a form of selective autophagy, it involves additional selectivity in the degradation of specific ER proteins and subdomains (60). Misfolded proteins in the ER are typically targeted for degradation via the ERAD pathway; however, some misfolded proteins cannot enter this pathway due to their inability to bind to the ERAD machinery or their tightly packed structure that prevents unfolding. These proteins ultimately accumulate in the ER and are delivered as cargo to isolation vesicles known as autophagosomes, which subsequently fuse with lysosomes in a process termed ER-phagy (69,70). In summary, ER subdomains containing these abnormal proteins exhibit affinity for ATG8 family proteins or FIP200, ensuring selectivity for the autophagic degradation of the ER. These proteins are expressed in specific tissues and localized to different ER subdomains, such as sheet-like ER, tubular ER and three-way junctions, facilitating the formation of autophagosomes and undergoing ER-phagy (7173).

In macro-ER-phagy, isolation membranes or phagophores form along the ER marked for degradation (74). The localization of the E3 ligase complex at the ER is crucial for the autophagic degradation of the ER, with UFL1 and UFBP1 playing significant roles. The UFM1 substrate modification of NADH-cytochrome B5 reductase 3 (CYB5R3) is dependent on the E3 ligase complex UFL1 and UFBP1, which are located on the ER membrane (75,76). The UFM1 binding site, Lys214, is situated at the interface between the NADH and FAD domains of CYB5R3, resulting in the disruption of the FAD domain and rendering CYB5R3 inactive. UFMylated CYB5R3 is recognized by UFBP1, which is essential for further UFMylation of CYB5R3 and enhances the E3 ligase activity of the UFL1-UFBP1 complex towards CYB5R3. Ultimately, the interaction of the UFL1 complex with UFMylated CYB5R3 and UFBP1 leads to the autophagic degradation of ER subdomains (9,64,66,77).

In plasma cells, the IRE1α/XBP1 axis within the UPR pathway upregulates Ufbp1 expression, which is critical for the expansion of the ER network (75,78). Simultaneously, UFBP1 plays a pivotal role in suppressing the PERK pathway. This suppressive relationship is demonstrated by the observation that PERK deletion restores the defects in B-cell to plasma cell development caused by UFBP1 deficiency. Moreover, the expression of UFBP1 and other molecules involved in the UFMylation pathway in immature B cells is independent of IRE1α/XBP1 (75,79). Thus, UFBP1 markedly regulates different branches of the UPR pathway to promote plasma cell development and function. Specifically, the IRE1α/XBP1 axis upregulates UFBP1 and genes in the UFMylation pathway in plasma cells, while UFBP1 deficiency impairs ER expansion and hinders immunoglobulin production (53,80). Structural and functional analyses reveal that Lys267 in UFBP1 is a critical lysine residue. Although not essential for plasma cell development, it is vital for immunoglobulin production and stimulating ER expansion in IRE1α-deficient plasma cells. In summary, UFBP1 exerts differential impacts on the development and function of plasma cells by regulating distinct steps in the UPR pathway (79,81).

Relevant studies have combined genetic disruption of UFMylation and de-UFMylation with affinity capture liquid chromatography-tandem mass spectrometry (LC-MS/MS) techniques, applying three consecutive filtration steps: i) Low-confidence hits (<3 unique peptides in UFSP2 knockout), ii) proteins whose abundance did not increase after UFSP2 knockout and iii) proteins whose abundance increased following UBA5 knockout, to identify the conserved ribosomal protein RPL26. Subsequently, genetic manipulation using small interfering siRNA depletion or CRISPR-Cas9-mediated cell line models was employed to investigate the UFMylation process of RPL26 (65,77). Although the absence of UFL1 or its interaction with UFBP1 eliminates RPL26 UFMylation, the UFMylation of RPL26 at two distinct lysine residues (K132 and K134) is markedly increased in UFSP2-/- cell lines (77,82). This modification is continuous; overexpression of RPL26 K134R in UFSP2-/- cells abolishes both monomeric and dimeric RPL26, while monomeric RPL26 is still present in cells overexpressing RPL26 K132R. In conditions where UFMylation is not dominant, such as in UFSP haploinsufficient cell lines (UFSP1-/+/UFSP2-/-), only K134 modifications are observed, indicating that the modification of RPL26 is highly specific. Moreover, replacing endogenous RPL26 with a mutant allele lacking both residues (RPL26K132R/K134R) abolishes the modification of RPL26 (63,64,83). This further indicates that the ribosomal RPL26 undergoes a dynamic cycle of UFMylation and de-UFMylation catalyzed by enzymes attached to the cytoplasmic surface of the ER, providing a functional link between the UFMylation process and ER protein homeostasis (64,66).

In eukaryotes, secretory pathway proteins are primarily synthesized by ribosomes bound to the ER and inserted into the ER lumen or integrated into membranes via the Sec61 translocon (63,84,85). This process is highly sensitive to disturbances such as translation stalling or defects in protein modifications, folding and assembly, all of which can lead to defective polypeptides that clog the translocon. The clogging of translocons triggers ribosome filtration and activates transport-associated quality control (TAQC) to degrade the clogged substrates (86,87). Wang et al (88) identified a membrane protein named SAYSD1 through whole-genome CRISPR-Cas9 screening, which aids in the TAQC process by binding to SEC61. SAYSD1 also directly recognizes ribosomes and UFM1, associating with stalled nascent chains to ensure their transport to lysosomes for degradation via the TRAPP complex. Similar to UFM1 deficiency, depletion of SAYSD1 leads to the accumulation of proteins stalled in the ER, inducing ER stress. Notably, disrupting UFM1- and SAYSD1-dependent TAQC in Drosophila results in the accumulation of collagen that is stalled during translocation, leading to collagen deposition defects, abnormal basement membrane integrity and reduced stress tolerance (89). Thus, SAYSD1 functions as a sensor for UFM1, coordinating the ribosomal UFMylation of blocked translocon sites to maintain ER homeostasis during animal development (88) (Fig. 2).

UFMylation and hematopoiesis

Hematopoiesis, a complex process regulated by specific transcription factors and cytokines, supports the self-renewal, differentiation and survival of hematopoietic progenitor cells at various stages of maturation (90,91). The UFMylation system plays a pivotal role in modulating the expression levels and activation of transcription factors that facilitate erythropoiesis. It is crucial to identify UFM1 targets for understanding how UFM1 protein modification governs erythroid development, as defects in this process can lead to pathological conditions, such as leukemia and myelodysplastic syndromes.

Germline deletions of UBA5, UFL1, or UFBP1 in mice result in embryonic lethality and disrupted hematopoietic development (92,93). UBA5 and other genes involved in UFMylation are expressed in both primitive and definitive erythrocytes, with the highest levels of UBA5 found in primitive erythroid lineages. Mice deficient in UBA5 exhibit severe anemia due to defects in megakaryocyte and erythrocyte differentiation, ultimately resulting in death in utero. This deficiency impairs the differentiation of common bone marrow progenitors into megakaryocytes and erythroid progenitors. Notably, transgenic expression of UBA5 in the erythroid lineage can rescue embryos from anemia, extending their survival (92).

UFL1 is essential for embryonic development, hematopoietic stem cell (HSC) survival and erythroid differentiation. Knockout of UFL1 leads to reduced HSC survival rates and embryonic death due to severe anemia (94). UFL1 is considered to function as an E3 ligase for UFM1, promoting the UFMylation of DDRGK1 and ASC1. Notably, both UBA5 and UFL1 knockout mice display extensive phenotypic similarities, including defects in embryonic erythropoiesis and increased mortality (93). Specifically, UBA5 deficiency disrupts the development of megakaryocyte-erythroid progenitors (MEP) in the fetal liver without affecting granulocyte-monocyte progenitors (GMP). Conversely, conditional deletion of RCAD in adult mice causes defects in MEP progenitors but does not affect GMP. Additionally, a significant number of multinucleated erythroid cells are observed in the embryos of both UBA5 and RCAD-deficient mice, suggesting that UBA5 and RCAD may function in similar cellular processes or signaling pathways during erythroid development (90).

UFBP1 is also critical for embryonic development and hematopoiesis; its deficiency results in defects in erythroid development and embryonic death in mice. In adult mice, UFBP1 deficiency leads to impaired hematopoiesis, resulting in pancytopenia and increased mortality (18). At the cellular level, the absence of UFBP1 heightens endoplasmic reticulum stress and activates the UPR, causing cell death in hematopoietic progenitor/stem cells (19). Furthermore, UFBP1 deficiency suppresses the expression of erythroid transcription factors GATA-1 and KLF1, inhibiting the transition from colony-forming units-erythroid to proerythroblasts. Similar phenotypic defects are observed in UFBP1-deficient mice when compared with UFL1 and UBA5-deficient mice, indicating that UFBP1 serves as a crucial downstream effector in the regulation of hematopoietic development mediated by the UFM1 system through UFMylation. These findings underscore the significant role of UFMylation in erythroid development and differentiation (18) (Table I).

Table I.

Phenotypic comparison of Uba5, Ufl1 and Ufbp1 knockout mice.

Table I.

Phenotypic comparison of Uba5, Ufl1 and Ufbp1 knockout mice.

First author, yearGeneKO phenotypeCKO phenotypeBiochemical function(Refs.)
Tatsumi et al, 2011UBA5Embryo death and impaired hematopoietic development. Fewer erythroid progenitor cells. UFM1 E1enzyme(92)
Zhang et al, 2015UFL1Embryo death and impaired hematopoietic development. Fewer erythroid progenitor cells.Animal death around 3 weeks after initial injection of tamoxifen. Pancytopenia and severe anemia. Reduced number of erythroid progenitor cells in bone marrow. Increased percentage and number of GMPs in bone marrow. Impaired development from CFU-E to proerythroblast. Loss of HSC function.UFM1 E3 ligase(93)
Cai et al, 2015UFBP1Embryo death and impaired hematopoietic development. Impaired development of hematopoietic progenitor cells.Animal death around 3 weeks after initial injection of tamoxifen. Pancytopenia and severe anemia. Reduced number of erythroid progenitor cells in bone marrow. Increased percentage and number of GMPs in bone marrow. Impaired development from CFU-E to proerythroblast. Loss of HSC function.UFM1 specific protease(18)

[i] UBA5, ubiquitin-like modifier activating enzyme 5; UFL1, UFM1 specific ligase 1; UFBP1, UFM1-binding and PCI domain-containing protein1; UFM1, ubiquitin-fold modifier 1; GMP, guanine monophosphate synthase; CFU-E, colony forming unit-erythrocyte; HSC, hematopoietic stem cell.

UFMylation and skeletal development

Clinical studies have identified diseases genetically linked to components of the UFM1 pathway, emphasizing their crucial roles in development and tissue homeostasis. The majority of mutations in UFM1 pathway components are associated with a skeletal pathology known as SEMD (23,95). A clinical case study of affected Italian families reported a missense mutation, specifically UFSP2 D418A, in the UFSP2 gene. The patients exhibited systemic skeletal dysplasia with infantile onset, delayed skeletal development, demineralization, atrophy, restricted mobility and joint destruction (95). Subsequently, heterozygous variants of the UFSP2 gene, including Y290H, D426A and H428R, were identified through exome sequencing, with the SEMD phenotype observed in individuals harboring mutations in the catalytic histidine (H420R) and the tyrosine residue (Y290H) that form the oxocation pore (96). The Y282H mutation resulted in a markedly milder phenotype. This finding supports a new perspective that varying clinical outcomes may be associated with the loss of catalytic UFSP2 activity linked to different mutants, which has been confirmed in vitro (24).

Furthermore, loss-of-function mutations in DDRGK1 (UFBP1, C20orf116) have also been demonstrated to cause SEMD. DDRGK1-/- mice exhibit delayed mesenchymal condensation in limb buds and early embryonic lethality. To further investigate this, conditional knockout mice were generated by crossing Prx1-Cre transgenic mice with DDRGKfl/fl mice to delete DDRGK1 expression in limb mesenchymal cells. The mutant mice displayed progressively severe shortening of the limbs, joint abnormalities, disorganized growth plate tissue, reduced proliferation zones and enlarged hypertrophic zones. These data highlight the importance of DDRGK1 in the development of growth plates (97). By contrast, conditional knockout of DDRGK1 using osteoblast-specific osteocalcin-Cre and leptin receptor-Cre lines did not result in bone phenotypes, indicating that the effect on limb development is cartilage-specific. These findings suggest that DDRGK1 is necessary postnatally for the maintenance of normal growth plate morphology (98). Collectively, these discoveries underscore the physiological role of DDRGK1 in the development and maintenance of growth plate cartilage. Moreover, these genetic mouse models recapitulate the clinical phenotypes of short stature and joint abnormalities observed in patients with Sohat-type SEMD (98).

UFMylation and neurodevelopment

UFMylation plays a crucial role in neurodevelopment. Abnormalities in the UFMylation system can manifest as severe infantile-onset encephalopathy, with or without seizures, severe congenital neuropathies, or early-onset cerebellar atrophy accompanied by ataxia (17,99). As the disease progresses, non-specific findings such as mild myelination delay, hyperintense white matter signals, thinned thalami and corpus callosum, cerebellar hypoplasia, or generalized brain atrophy can be observed (24). Individuals with the most severe forms of the disease may die shortly after birth or in infancy, while those with milder symptoms can survive >20 years. Whole exome sequencing analysis has identified rare autosomal recessive variants in UBA5 in five children from four unrelated families, all of whom exhibited similar severe intellectual disabilities, microcephaly, motor disorders, or early-onset refractory epilepsy (25,100). To date, 24 variants in UBA5 have been linked to neurological disorders. Of these variants, ~67% are located in the adenylylation domain of the UBA5 protein, 19% in the UFC1-binding domain and C-terminus and 14% in the N-terminus (101). Most missense mutations lead to mild impairment of UBA5 function; however, two homozygous inherited N-terminal variants have resulted in severe disease, leading to the mortality of affected individuals within 16 days and 16 weeks, respectively (102,103). In zebrafish, UBA5 gene knockout at early stages exhibits peripheral nerve and cerebellar axonal damage, with mitochondrial damage identified in peripheral and central nervous systems as well as in skeletal muscles (104). In Caenorhabditis elegans, knockout of UBA5 and its human ortholog in the UFM1 cascade alters cholinergic neurotransmission without affecting glutamatergic transmission (105).

NCAM, a molecule that plays a role in neuronal development and synaptic plasticity in the adult brain, has been shown to interact with the UFMylation system. NCAM140, a subtype of NCAM, interacts with UFC1 through its cytoplasmic domain and co-localizes with UFC1 at the surface of B35 neuroblastoma cells (101). Overexpression of UFM1 also leads to increased endocytosis of NCAM140. Whole exome sequencing analysis revealed rare autosomal recessive variants in UBA5 in five children from four unrelated families, presenting similar severe intellectual disabilities, microcephaly, motor disorders, or early-onset refractory epilepsy (106). Biochemical and cellular studies of UBA5 mutant proteins in fibroblasts from affected individuals indicate that pathogenic mutations impair UFMylation, resulting in abnormal endoplasmic reticulum structure. In C. elegans, knockout of UBA5 and its human ortholog in the UFM1 cascade affects cholinergic neurotransmission without affecting glutamatergic neurotransmission (107). Furthermore, silencing of UBA5 in zebrafish induced abnormal movements while reducing locomotor ability, suggesting a link between UFMylation and seizures (105,108). These clinical, biochemical and experimental findings demonstrate that UBA5 mutations can lead to early-onset encephalopathy due to abnormal protein UFMylation (105). Collectively, these studies highlight the significant role of UFMylation in neurodevelopment (Table II).

Table II.

Summary of verified UFMylation substrates and enzymes.

Table II.

Summary of verified UFMylation substrates and enzymes.

First author, yearUFMylation substrateFunctional sitesFunction(Refs.)
Bakkenist and Kastan, 2003MRE11K282Promotes MRN complex formation and recruitment, maintains telomere length.(49)
Qin et al, 2020Histone 4K31Amplifies ATM activation and maintains genomic. integrity(50)
Liu et al, 2020P53K351, K357, K370, K373Maintains P53 stability and suppresses tumor growth.(47)
Wang et al, 2020ASC1K324, K325, K334, K367Promotes ERα transactivation and breast cancer progression.(165)
Luo et al, 2023HRD1K610Regulating the degradation of endoplasmic reticulum related proteins and clearing misfolded ER resident proteins.(68)
Ishimura et al, 2022CYB5R3K214Promotes the formation of autophagosomes and initiate endoplasmic reticulum phagocytosis.(76)
Zhu et al, 2019UFBP1K267Maintains ER homeostasis, promotes self-renewal and differentiation of hematopoietic progenitor cells.(75)
Liu et al, 2020RPL26K132, K134Maintains ER homeostasis.(47)
Wang et al, 2023SAYSD1 Maintains ER homeostasis.(88)
Tatsumi et al, 2011UBA5 Promotes self-renewal and differentiation of hematopoietic progenitor cells, promotes neuronal development and synaptic plasticity in the brain.(92)
Zhang et al, 2015UFL1 Promotes self-renewal and differentiation of hematopoietic progenitor cells.(93)
Di Rocco et al, 2018UFSP2Y290H, D426A, H428RMaintains normal bone shape.(95)

[i] MRE11, meiotic recombination 11 homolog; ASC1, activating signal cointegrator 1; HRD1, HMG-CoA reductase degradation protein 1; CYB5R3, cytochrome b5 reductase 3; UFBP1, UFM1 binding protein1; RPL26, ribosomal protein L26; SAYSD1, saysvfn motif-containing domain 1; UBA5, ubiquitin like modifier activating enzyme 5; UFL1, UFM1 specific ligase 1; UFSP2, UFM1 specific peptidase 2; UFBP1, UFM1 binding protein 1; ATM, ataxia-telangiectasia mutated.

UFMylation and immune regulation

The UFM1 conjugation family has emerged as a promising therapeutic target for modulating immune responses, either by enhancing antiviral defense or mitigating excessive inflammation in autoimmune diseases. Studies have revealed that upon RNA virus infection (109,110), UFL1 is recruited to the membrane-associated scaffold protein 14-3-3ε, where it undergoes UFMylation. This process leads to the activation of retinoic acid-inducible gene I, which in turn triggers mitochondrial antiviral signaling protein (MAVS)-mediated signaling cascades. These cascades ultimately culminate in the induction of type I and III interferons (IFNs) (110). In response to herpes simplex virus type 1 infection, UFL1 expression is rapidly downregulated at both the mRNA and protein levels in peritoneal macrophages. The conditional deletion of UFL1 in macrophages results in increased viral loads in the serum and peripheral immune cells. This increase is accompanied by a marked reduction in proinflammatory cytokines, including interleukin-6 (IL-6), tumor necrosis factor-α (TNF-α) and IFN-β. During Epstein-Barr virus (EBV) infection, the viral-encoded G protein-coupled receptor BILF1 hijacks UFL1 to mediate MAVS UFMylation. This process promotes MAVS mitochondrial mislocalization and subsequent lysosomal degradation, effectively suppressing EBV-induced activation of the NLRP3 inflammasome and thereby dampening host antiviral immune responses (109). These findings demonstrate that UFMylation plays a crucial role in the body's immune regulation.

The role of UFMylation in common malignancies

Numerous studies have reported the amplification, deletion, or mutation of genes encoding UFMylation factors (UBA5, UFC1, UFL1, UFSP2 and UFM1) in malignant tumors across various tissues. These findings indicate that UFMylation may either promote or inhibit tumorigenesis, depending on the cellular environment. The loss or mutation of components within the UFMylation pathway is associated with a range of diseases. Deepening our understanding of UFMylation's role in cancer may pave the way for the development of novel therapeutic strategies (43,53,103,107,111).

Breast cancer

Breast cancer ranks among the top three most common cancers worldwide (112,113). A significant body of literature indicates that estrogen receptor-positive (ER+) tumors constitute a large portion of cases. PTMs of ERα are critical in regulating its expression, subcellular localization and hormonal response sensitivity (114,115). UFMylation plays a crucial role in positively regulating ERα stability and transactivation, which is key in breast cancer development (58,116). By inhibiting ubiquitination, ERα stability is markedly enhanced, whereas silencing UBA5 decreases ERα stability. Lys171 and Lys180 of ERα have been identified as primary UFM1 receptor sites and substituting these lysine residues with arginine (2KR mutation) markedly reduces ERα stability (116). Furthermore, the 2KR mutation abolishes ERα's 17β-estradiol-induced transcriptional activity and the expression of downstream targets such as PS2, cyclin D1 and C-MYC, highlighting the essential role of ERα's transcriptional activation function. The 2KR mutation also prevents MCF7 cells from forming anchorage-independent colonies (48,116,117). Notably, UFM1 and its associated mechanisms (UBA5, UFC1, UFL1 and UFBP1) are markedly upregulated in ERα-positive breast cancer cell lines and tissues. In summary, these findings suggest that ERα enhances breast cancer development by stabilizing and promoting its transactivation function through UFMylation (116,118).

Metformin can induce ferroptosis in breast cancer cell lines, thereby inhibiting tumor growth independently of conventional AMP-activated protein kinase (AMPK) signaling (119). Mechanistically, metformin increases intracellular Fe2+ and lipid ROS levels. Specifically, metformin suppresses the UFMylation process of SLC7A11, a key regulator of iron metabolism, leading to decreased protein stability of SLC7A11 and consequently inhibiting cancer proliferation (120).

Recently, two additional UFMylation substrates, PLAC8 and PD-L1, have been identified as playing significant roles in the pathogenesis of breast cancer (121123). Mao et al (121) reported that PLAC8 is generally expressed at high levels in triple-negative breast cancer (TNBC). This is because it is modified by UFM1 UFMylation at Lys103, which maintains its protein stability. The stable PLAC8 interacts with glycosylated PD-L1, thereby upregulating PD-L1 levels, promoting cancer cell proliferation and inhibiting immune responses (123,124). Additionally, PD-L1 can be modified by various deubiquitinating and ubiquitinating proteins, such as USP22, CSN5 and ARIH1. Treatment of MDA-MB-231 and HCC-1937 cells with MG-132 and chloroquine demonstrated that MG-132 inhibited proteasome activity, while chloroquine inhibited lysosomal activity. Notably, chloroquine rescued the reduction of PD-L1 protein levels induced by PLAC8 knockout. Collectively, these data suggest that PLAC8 may also stabilize PD-L1 protein by regulating its ubiquitination (121).

Liver cancer

Hepatocellular carcinoma (HCC) is a leading cause of cancer-related deaths in a number of regions of the world. Risk factors for HCC include chronic hepatitis, alcohol addiction, metabolic liver diseases and exposure to dietary toxins such as aflatoxins and aristolochic acids. In most cases, acute hepatitis, chronic hepatitis and cirrhosis caused by chronic hepatitis B or C virus infections are significant contributors to the development of hepatocellular carcinoma (125). Reports indicate that the UFMylation system is associated with the occurrence and progression of HCC. In the livers of mice re-fed with dihydro-2,4,6-trimethy l-3,5-pyridinedicarboxylate (DDC) and in humans with alcoholic hepatitis (AH) and non-alcoholic steatohepatitis (NASH) characterized by the presence of Mallory-Denk bodies (MDB), the UFMylation pathway is downregulated, including protein quality control (126,127). Notably, feeding the methyl donor betaine alongside DDC markedly prevents the increase in UFMylation expression in DDC-pretreated mice (128,129). Betaine notably inhibited the transcriptional silencing of UFM1, UBA5 and UFSP1 associated with MDB formation and prevented the increase in FAT10 and LMP7 expression induced by DDC re-feeding. A similar downregulation of UFMylation was observed in multiple biopsies from patients with AH and NASH. Compared with normal subjects, patients with AH and NASH exhibited markedly elevated levels of DNA methylation in the promoter CpG regions of UFM1, UFC1 and UFSP1 (130132). The mRNA levels of DNA methyltransferase 1 (DNMT1) and DNA methyltransferase 3β (DNMT3B) were markedly upregulated in patients with AH and NASH, indicating that the maintenance of UFMylation methylation may be co-mediated by DNMT1 and DNMT3B (133,134).

It has been reported that an HCC suppressor known as B3GALT5-AS1 negatively regulates the proliferation, invasion and metastasis of HCC cells by modulating miR-934 and UFM1. This suppression of HCC cell proliferation, invasion and metastasis was observed when pGL3-UFM1-WT and pGL3-UFM1-MUT plasmids were co-transfected with miR-NC or miR-934 into HCC cell and compared with the co-transfection group with pGL3-UFM1-WT and NC (135), the co-transfection of the pGL3-UFM1-WT plasmid with miR-934 markedly reduced the luciferase activity of the reporter plasmid, thereby indirectly elucidating the interaction between UFM1 and miR-934 and confirming that UFM1 is a regulatory target of miR-934. Subsequently, the expression of UFM1 was measured in three groups (NC, miR-934 inhibitor and si-UFM1) after transfection, revealing that the UFM1 mRNA expression levels in the miR-934 inhibitor and si-UFM1 groups were markedly lower than those in the miR-934 inhibitor-only group. Importantly, it was determined that si-UFM1 could reverse the decreased cell proliferation and migration abilities induced by the miR-934 inhibitor (136).

Additionally, UFL1 has been identified as a tumor suppressor in HCC, playing a critical role in the pathogenesis of HCC by preventing cell invasion, inhibiting NF-κB signaling and increasing the stability of LZAP protein. However, the exact role of CDK5RAP3 in HCC remains controversial (39,137). One study reports that CDK5RAP3 may act as an oncogene, promoting the migratory and invasive characteristics of the SMMC-7721 and HEPG2 cell lines (138). By contrast, another group of studies indicated that CDK5RAP3 functions as a tumor suppressor in HEPG2 cells. The precise role, function and mechanism of CDK5RAP3 in HCC warrant further investigation (16,76).

Lung cancer

Lung cancer is one of the deadliest cancers and is becoming increasingly prevalent worldwide (139). Notably, the mortality rate of lung cancer exceeds that of other tumors. Lung cancer encompasses various subtypes, including lung adenocarcinoma (LUAD), lung squamous cell carcinoma (LUSC), large cell lung carcinoma and small cell lung carcinoma (140,141). Recently, UBA5 has been identified as playing a significant role in the growth of LUAD cells, resistance to cisplatin and promoting immune evasion while also participating in the macrophage M2 polarization process, thereby altering the tumor microenvironment and facilitating tumor progression (142). In LUAD, both UBA5 mRNA and protein levels are highly expressed. The pharmacological inhibition of UBA5 using DKM 2–93 effectively inhibited LUAD growth, indicating that the suppression of UBA5 hinders LUAD cell proliferation (143).

Additionally, UBA5 positively correlates with macrophage M2 polarization in LUAD. The knockdown of UBA5 directly suppressed the in vitro polarization of macrophages to M2, reduced the in vivo infiltration of macrophage M2 and decreased lactate production in LUAD. These results suggest that UBA5 regulates macrophage M2 polarization through lactate secretion, thereby altering the immune microenvironment and facilitating the escape of LUAD cells from immune surveillance (144).

Despite its tumor-suppressive role in hepatocellular carcinoma, UFL1 may act as an oncogene in lung adenocarcinoma. UFL1 is upregulated in early lung adenocarcinoma tissues and its overexpression promotes the proliferation of H1299 lung adenocarcinoma cells. UFL1 can bind to the regulatory domain of P120-catenin, inhibiting the ubiquitin-mediated proteasomal degradation of P120-catenin. Subsequently, P120-catenin promotes the proliferation of lung adenocarcinoma through its interaction with NLBP (145).

Colorectal cancer

In relevant studies on colorectal cancer, Zhou et al (146) observed that the knockdown of UFSP2 markedly promoted the growth rate of colorectal cancer cells HT29 and HCT116 and their anchorage-independent growth. Notably, the knockdown of UFSP2 expression markedly enhanced the growth of xenograft tumors derived from UFSP2-depleted HT29 cells. These results demonstrate that UFSP2 is a potential tumor suppressor in colorectal cancer.

Gastric cancer

Despite a decline in the incidence and mortality rates of primary cancers in recent decades, gastric cancer remains one of the three most common cancers worldwide. Most gastric cancer patients exhibit nonspecific early symptoms and are often diagnosed at advanced stages, which severely affects their prognosis. Therefore, identifying new biomarkers is essential for early diagnosing and treating gastric cancer (147,148). DDRGK1 is a target protein for UFMylation and a key component of the UFMylation modification system, playing a crucial role in cancer development (149). Shiwaku et al (150) found that the amino acid sequence of CDK5RAP3 contains a ubiquitin-protein ligase binding domain. Wu et al (151) revealed that CDK5RAP3 can interact with DDRGK1 and UFL1 (RCAD), regulating the stability of CDK5RAP2 and DDRGK1. Notably, patients with low expressions of CDK5RAP3 and DDRGK1 had the worst prognoses, while those with high expressions of these proteins exhibited the best prognoses, with other patients falling in between. The predictive accuracy of the combined expression of CDK5RAP3 and DDRGK1 was higher than using either CDK5RAP3 or DDRGK1 alone and their combined expression demonstrated superior predictive capability for overall survival in cancer patients (152). Xi et al (153) discovered that DDRGK1 interacts with IkBa and regulates its stability, thereby modulating the transcriptional activity of NF-κB and the expression of its target genes. Conversely, Wu et al found that the downregulation of CDK5RAP3 increased cellular invasiveness and enhanced the transcriptional activity of NF-κB (151). CDK5RAP3 binds to RelA to inhibit its phosphorylation, increasing the association of HDAC with RERA and thereby suppressing NF-κB transcriptional activity.

In summary, CDK5RAP3 and DDRGK1 interact and their roles in the NF-κB pathway are similar. Additionally, Lin et al (154) found that UFM1 was downregulated in gastric cancer tissues. Patients with low UFM1 expression levels had poor prognoses, while UFM1 exhibits inhibitory effects on oncogenicity, invasion and migration of gastric cancer cells. Mechanistically, UFM1 suppresses gastric cancer cells' epithelial-mesenchymal transition (EMT) by negatively regulating the PI3K/AKT signaling pathway and increasing the ubiquitination of PDK1, thereby exerting its tumor-suppressive function (155).

Renal cell carcinoma

In renal cell carcinoma (RCC), the expressions of UFL1 and UFBP1 are also downregulated and positively correlate with P53 levels, a protein closely associated with various cancers. It has been reported that P53 interacts with UFL1 and UfBP1 and is modified by UFM1. UFMylation mediated by UFL1 and UFBP1 stabilizes P53 by antagonizing MDM2-mediated ubiquitination and proteasomal degradation, inhibiting cellular growth and tumor formation in vivo (155).

In addition to analyzing RCC tissue microarrays from 40 paired patient samples, studies using mouse xenograft models indicate that UFL1 and UfBP1 can function as tumor suppressors by regulating P53 stability. These results suggest that UFMylation is a key post-translational modification for maintaining P53 stability and tumor-suppressive function, implicating UFMylation as a promising therapeutic target in cancer (47) (Table III).

Table III.

Function of UFMylation in various types of cancer.

Table III.

Function of UFMylation in various types of cancer.

First author, yearCancer typeGeneFunction(Refs.)
Yoo et al, 2022Breast cancerERaPromotes breast cancer development.(116)
Yang et al, 2021Breast cancerSLC7A11UFMylation stabilizes SALC7A11 and metformin reduces the protein stability of SLC7A11 by reducing UFM1.(120)
Mao et al, 2022Breast cancerPLAC8UFMylation of PLAC8 may influence tumor progression and immune response in triple negative breast cancer cells by reducing PD-L1 ubiquitination.(121)
Lim et al, 2016Breast cancerPD-L1UFMylation of PD-L1 destabilizes PD-L1 by acting synergistically to promote its ubiquitination.(124)
Chen et al, 2021Hepatocellular carcinomaUFM1B3GALT5-AS1 regulates miR-934 and UFM1 to achieve negative regulation of HCC cell proliferation, invasion, and metastasis.(136)
Liu et al, 2014Hepatocellular carcinomaUFSP1Alcoholic hepatitis and non-alcoholic steatohepatitis transcriptional down regulation of FATylation and UFMylation.(133)
Yang et al, 2019Hepatocellular carcinomaUFL1UFL1 act as gatekeepers to prevent liver fibrosis and subsequent steatohepatitis and Hepatocellular carcinoma development by inhibiting the mTOR pathway.(16)
Zhou et al, 2021Colon cancerUfSP2UFSP2 is a potential tumor suppressor in colon cancer.(146)
Lin et al, 2018Gastric cancerCDK5RAP3 and DDRGK1CDK5RAP3 interacting with DDRGK1 suppresses the development of gastric cancer by inhibiting the phosphorylation of AKT/GSK-3β and negatively regulating Wnt/β-catenin signaling.(152)
Lin et al, 2019Gastric cancerUFM1UFM1 has inhibitory effects on carcinogenicity, invasion, and migration of gastric cancer cells.(154)
Liu et al, 2020Renal cancerP53UFMylation stabilizes p53 by inhibiting its ubiquitination, which suppresses cell growth and tumor formation.(47)

[i] Era, estrogen receptor alpha; SLC7A11, solute carrier family member 11; PLAC8, placenta specific 8; PD-L1, programmed death ligand; UFM1, ubiquitin fold modifier 1; UFSP1, Ufm1 specific peptidase 1; UFL1, Ufm1 ligase 1; CDK5RAP3, cyclin-dependent kinase 5 regulatory subunit associated protein 3; DDRGK1, DNA damage-regulated autophagy modulator 1; UFM1, ubiquitin fold modifier 1.

UFMylation in immunotherapy

Immune cells are central to antiviral defense and antitumor immunity, yet the intricate tumor microenvironment enables malignant cells to evade immune-mediated elimination through diverse mechanisms (156). Among these, the upregulation of inhibitory immune checkpoint receptors, such as PD-1, serves as a key strategy to suppress T cell activation and cytotoxic function. However, the limited clinical efficacy of anti-PD-1/PD-L1 immunotherapy underscores the urgent need for more effective therapeutic strategies (21). Recent findings highlight PD-L1 UFMylation as a crucial regulator of PD-1/PD-L1 axis homeostasis in both human and murine tumor cells, with its dysregulation compromising immune evasion (157). Loss of UFL1 in T cells abrogates PD-1 UFMylation, thereby enhancing antitumor immunity. Specifically, UFL1 deletion in T cells diminishes PD-1 UFMylation, facilitates CD8+T cell-mediated tumor rejection and promotes K48-linked ubiquitination and subsequent proteasomal degradation of PD-1. In vitro and in vivo analyses both demonstrate that UFL1 deficiency markedly reduces PD-1 protein abundance while augmenting the production of effector cytokines, including IFNγ, TNF and granzyme B, in CD8+T cells.

Furthermore, AMPK phosphorylates UFL1 at T536, disrupting its interaction with PD-1 and attenuating PD-1 UFMylation (158). Notably, conditional T cell-specific knockout (cKO) of UFL1 enhances antitumor immunity. In murine models of Lewis lung carcinoma and MC38 colorectal cancer, UFL1 cKO markedly improves responsiveness to anti-CTLA-4 immunotherapy. However, UFL1 depletion in T cells diminishes the antitumor efficacy of AMPK activators, suggesting that the AMPK-UFL1 axis plays a pivotal role in regulating T cell-mediated antitumor immunity (159).

Other tumors

Beyond the aforementioned tumor types, the UFM1 conjugation system has also been implicated in other human malignancies. Sarcomas, a heterogeneous group of mesenchymal-derived malignant neoplasms of connective tissue, are broadly classified into soft tissue sarcomas and primary bone sarcomas. These categories encompass a wide array of subtypes, contributing to the remarkable diversity of this tumor group (160). Among them, osteosarcoma is the most prevalent primary bone malignancy in children, adolescents and young adults (161). Notably, studies have reported that UFBP1 suppresses the proliferation, migration and invasion of human osteosarcoma cells (15,16,162). Mechanistically, as a component of the E3 ligase complex in the UFM1 conjugation system, UFBP1 directly interacts with IκBα, regulating its stability and attenuating NF-κB transcriptional activity (16). Notably, UFBP1 primarily binds to the N-terminal domain of IκBα (1–106 aa), which harbors critical phosphorylation sites (Ser32 and Ser36) and ubiquitination sites (Lys21 and Lys22). By regulating the phosphorylation and stability of IκBα, UFBP1 modulates NF-κB transcriptional activity and the expression of its downstream target genes. Furthermore, UFBP1 depletion via siRNA markedly alters the expression of NF-κB target genes, including cytokines, chemokines, adhesion molecules and receptors, as well as enzymes involved in proliferation, differentiation, apoptosis and stress responses. In summary, UFBP1 may promote the UFMylation-mediated degradation of IκBα, thereby downregulating the expression of NF-κB pathway target genes and suppressing osteosarcoma cell proliferation, migration and invasion (153). In addition, RPL10 UFMylation has been identified in pancreatic cancer tissues and cell lines (163), with this modification being catalyzed by UFL1 at specific sites and reversed by UFSP2-mediated de-UFMylation (164). Reduced UFMylation of RPL10 inhibits pancreatic cancer cell proliferation and stemness. Moreover, transcription factor KLF4 positively regulates the relationship between RPL10 inactivation and cellular stemness. Loss of RPL10 is closely associated with tumorigenesis, primarily by enhancing the expression of stemness-associated surface markers and upregulating KLF4. Notably, mutations at key UFMylation sites in RPL10 markedly impair pancreatic cancer cell proliferation and stemness, further highlighting the functional relevance of this post-translational modification in tumor development (163).

Conclusion

Protein PTM is a standard physiological process in cells. Ubiquitination is a special modification of protein post-translational modification, which plays a vital role in cell life activities. UFM1 is a class ubiquitin modifier that attaches to lysine residues on substrates after translation by a dedicated enzyme system conserved in most eukaryotes. Despite structural similarities between UFM1 and ubiquitin, the UFMylation machinery employs unique mechanisms to ensure fidelity. Although the physiological triggers and consequences of UFMylation are not fully understood, its biological importance is reflected in frequent mutations in the UFMylation pathway in human pathophysiology, including musculoskeletal and neurodevelopmental disorders. Some of these diseases can be explained by the increased endoplasmic reticulum (ER) stress and disruption of translational homeostasis observed upon loss of UFMylation. The role of UFM1 in these processes may stem from its function in the ER, where ribosomes are UFM1-glycosylated due to translational arrest. In addition, UFMylation has been implicated in other cellular processes, including DNA damage response and telomere maintenance.

To date, certain studies have shown that UFMylation modification is also closely related to the development of tumors (43,146,164,165). UFMylation modification seems related to the stable expression of specific tumor suppressor genes. For example, in clear cell renal cell carcinoma, UFMylation of p53 promotes its stability, inhibiting tumor cell proliferation.

UFMylation also plays a pivotal role in antitumor immunity. Depletion of UFL1 in CD8+ T cells suppresses PD-L1 expression, leading to the upregulation of downstream effector cytokines such as IFN-γ and enhancing T-cell cytotoxicity. As a potential therapeutic target, inhibition of UFMylation has shown promise in cancer treatment. Notably, metformin disrupts the UFMylation of ferroptosis-related proteases, destabilizing SLC7A11 and promoting ferroptosis, thereby suppressing tumor growth through a mechanism independent of conventional AMPK signaling. However, does UFMylation have a stabilizing effect on other tumor suppressor proteins? At the same time, does UFMylation also participate in the degradation of tumor-related genes and inhibit the occurrence and development of tumors? In addition, is UFMylation also associated with activation/inhibition of tumor-related signaling pathways? None of this has been reported in much research. The revelation of these mechanisms may expand the field of UFMylation and human diseases.

In summary, studies of the mechanisms and biological functions of UFMylation-related signaling pathways will reveal insights into basic cell biology and may provide new therapeutic opportunities for human health. However, this study also has certain limitations, e.g. numerous tumor-related tumor suppressor proteins or oncoproteins may have UFMylation, which seriously affects the occurrence and development of various major human diseases; however, these proteins were not extensively described in this study. Therefore, such correlation studies should be further analyzed in the future.

Acknowledgements

Not applicable.

Funding

The present study was supported by program for the grants from the Scientific Research project of Education Department of Yunnan Province (grant no. 2023Y0787), Science and Technology Plan of Yunnan Provincial Department of Science and Technology, 202401AY070001-283 and The Kunming Health Science and Technology Personnel Training Project (grant no. 2022-SW-10).

Availability of data and materials

Not applicable.

Authors' contributions

YT and CY designed and revised the article. RQ, JZ, YY, FM, XY and KZ wrote the first draft of this review. Data authentication is not applicable. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Millar AH, Heazlewood JL, Giglione C, Holdsworth MJ, Bachmair A and Schulze WX: The scope, functions, and dynamics of posttranslational protein modifications. Annu Rev Plant Biol. 70:119–151. 2019. View Article : Google Scholar : PubMed/NCBI

2 

Hsu YC, Hsieh YH, Liao CC, Chong LW, Lee CY, Yu YL and Chou RH: Targeting post-translational modifications of histones for cancer therapy. Cell Mol Biol (Noisy-le-grand). 30:69–84. 2015.

3 

Hitosugi T and Chen J: Post-translational modifications and the Warburg effect. Oncogene. 33:4279–4285. 2013. View Article : Google Scholar : PubMed/NCBI

4 

Herhaus L and Dikic I: Expanding the ubiquitin code through post-translational modification. EMBO Rep. 16:1071–1083. 2015. View Article : Google Scholar : PubMed/NCBI

5 

Hochstrasser M: Origin and function of ubiquitin-like proteins. Nature. 458:422–429. 2009. View Article : Google Scholar : PubMed/NCBI

6 

Sahtoe DD and Sixma TK: Layers of DUB regulation. Trends Biochem Sci. 40:456–467. 2015. View Article : Google Scholar : PubMed/NCBI

7 

Karpiyevich M and Artavanis-Tsakonas K: Ubiquitin-like modifiers: Emerging regulators of protozoan parasites. Biomolecules. 10:14032020. View Article : Google Scholar : PubMed/NCBI

8 

Komatsu M, Chiba T, Tatsumi K, Iemura S, Tanida I, Okazaki N, Ueno T, Kominami E, Natsume T and Tanaka K: A novel protein-conjugating system for Ufm1, a ubiquitin-fold modifier. EMBO J. 23:1977–1986. 2004. View Article : Google Scholar : PubMed/NCBI

9 

Gerakis Y, Quintero M, Li H and Hetz C: The UFMylation system in proteostasis and beyond. Trends Cell Biol. 29:974–986. 2019. View Article : Google Scholar : PubMed/NCBI

10 

Banerjee S, Kumar M and Wiener R: Decrypting UFMylation: How proteins are modified with UFM1. Biomolecules. 10:14422020. View Article : Google Scholar : PubMed/NCBI

11 

Witting KF, van der Heden van Noort GJ, Kofoed C, Ormeno CT, El Atmioui D, Mulder MPC and Ovaa H: Generation of the UFM1 toolkit for profiling UFM1-specific proteases and ligases. Angew Chem Int Ed Engl. 57:14164–14168. 2018. View Article : Google Scholar : PubMed/NCBI

12 

Lorenz S, Cantor AJ, Rape M and Kuriyan J: Macromolecular juggling by ubiquitylation enzymes. BMC Biol. 11:652013. View Article : Google Scholar : PubMed/NCBI

13 

Jing Y, Mao Z and Chen F: UFMylation system: An emerging player in tumorigenesis. Cancers (Basel). 14:35012022. View Article : Google Scholar : PubMed/NCBI

14 

Clague MJ, Urbe S and Komander D: Breaking the chains: Deubiquitylating enzyme specificity begets function. Nat Rev Mol Cell Biol. 20:338–352. 2019. View Article : Google Scholar : PubMed/NCBI

15 

Lin M, Zheng X and Jin J: Nontraditional translation is the key to UFMylation and beyond. J Biol Chem. 298:1024312022. View Article : Google Scholar : PubMed/NCBI

16 

Yang R, Wang H, Kang B, Chen B, Shi Y, Yang S, Sun L, Liu Y, Xiao W, Zhang T, et al: CDK5RAP3, a UFL1 substrate adaptor, is crucial for liver development. Development. 146:dev1692352019. View Article : Google Scholar : PubMed/NCBI

17 

Yang S, Moy N and Yang R: The UFM1 conjugation system in mammalian development. Dev Dyn. 252:976–985. 2023. View Article : Google Scholar : PubMed/NCBI

18 

Cai Y, Pi W, Sivaprakasam S, Zhu X, Zhang M, Chen J, Makala L, Lu C, Wu J, Teng Y, et al: UFBP1, a key component of the ufm1 conjugation system, is essential for ufmylation-mediated regulation of erythroid development. PLoS Genet. 11:e10056432015. View Article : Google Scholar : PubMed/NCBI

19 

Tandra V, Anderson T, Ayala JD, Weintraub NL, Singh N, Li H and Li J: Ufmylation of UFBP1 is dispensable for endoplasmic reticulum stress response, embryonic development, and cardiac and intestinal homeostasis. Cells. 12:19232023. View Article : Google Scholar : PubMed/NCBI

20 

Wang X, Lv X, Ma J and Xu G: UFMylation: An integral post-translational modification for the regulation of proteostasis and cellular functions. Pharmacol Ther. 260:1086802024. View Article : Google Scholar : PubMed/NCBI

21 

Ding LJ, Jiang X, Li T and Wang S: Role of UFMylation in tumorigenesis and cancer immunotherapy. Front Immunol. 15:14548232024. View Article : Google Scholar : PubMed/NCBI

22 

Wang X, Xu X and Wang Z: The post-translational role of UFMylation in physiology and disease. Cells. 12:25432023. View Article : Google Scholar : PubMed/NCBI

23 

Yang X, Zhou T, Wang X, Xia Y, Cao X, Cheng X, Cao Y, Ma P, Ma H, Qin A and Zhao J: Loss of DDRGK1 impairs IRE1α UFMylation in spondyloepiphyseal dysplasia. Int J Biol Sci. 19:4709–4725. 2023. View Article : Google Scholar : PubMed/NCBI

24 

Zhang G, Tang S, Wang H, Pan H, Zhang W, Huang Y, Kong J and Wang Y, Gu J and Wang Y: UFSP2-related spondyloepimetaphyseal dysplasia: A confirmatory report. Eur J Med Genet. 63:1040212020. View Article : Google Scholar : PubMed/NCBI

25 

Colin E, Daniel J, Ziegler A, Wakim J, Scrivo A, Haack TB, Khiati S, Denommé AS, Amati-Bonneau P, Charif M, et al: Biallelic variants in UBA5 reveal that disruption of the UFM1 cascade can result in early-onset encephalopathy. Am J Hum Genet. 99:695–703. 2016. View Article : Google Scholar : PubMed/NCBI

26 

Sasakawa H, Sakata E, Yamaguchi Y, Komatsu M, Tatsumi K, Kominami E, Tanaka K and Kato K: Solution structure and dynamics of Ufm1, a ubiquitin-fold modifier 1. Biochem Biophys Res Commun. 343:21–26. 2006. View Article : Google Scholar : PubMed/NCBI

27 

Millrine D, Cummings T, Matthews SP, Peter JJ, Magnussen HM, Lange SM, Macartney T, Lamoliatte F, Knebel A and Kulathu Y: Human UFSP1 is an active protease that regulates UFM1 maturation and UFMylation. Cell Rep. 40:1111682022. View Article : Google Scholar : PubMed/NCBI

28 

Kang SH, Kim GR, Seong M, Baek SH, Seol JH, Bang OS, Ovaa H, Tatsumi K, Komatsu M, Tanaka K and Chung CH: Two novel ubiquitin-fold modifier 1 (Ufm1)-specific proteases, UfSP1 and UfSP2. J Biol Chem. 282:5256–5262. 2007. View Article : Google Scholar : PubMed/NCBI

29 

Ha BH, Jeon YJ, Shin SC, Tatsumi K, Komatsu M, Tanaka K, Watson CM, Wallis G, Chung CH and Kim EE: Structure of ubiquitin-fold modifier 1-specific protease UfSP2. J Biol Chem. 286:10248–10257. 2011. View Article : Google Scholar : PubMed/NCBI

30 

Millrine D, Peter JJ and Kulathu Y: A guide to UFMylation, an emerging posttranslational modification. FEBS J. 290:5040–5056. 2023. View Article : Google Scholar : PubMed/NCBI

31 

Liang Q, Jin Y, Xu S, Zhou J, Mao J, Ma X, Wang M and Cong YS: Human UFSP1 translated from an upstream near-cognate initiation codon functions as an active UFM1-specific protease. J Biol Chem. 298:1020162022. View Article : Google Scholar : PubMed/NCBI

32 

Mashahreh B, Hassouna F, Soudah N, Cohen-Kfir E, Strulovich R, Haitin Y and Wiener R: Trans-binding of UFM1 to UBA5 stimulates UBA5 homodimerization and ATP binding. FASEB J. 32:2794–2802. 2018. View Article : Google Scholar : PubMed/NCBI

33 

Soudah N, Padala P, Hassouna F, Kumar M, Mashahreh B, Lebedev AA, Isupov MN, Cohen-Kfir E and Wiener R: An N-terminal extension to UBA5 adenylation domain boosts UFM1 activation: isoform-specific differences in ubiquitin-like protein activation. J Mol Biol. 431:463–478. 2018. View Article : Google Scholar : PubMed/NCBI

34 

Kumar M, Padala P, Fahoum J, Hassouna F, Tsaban T, Zoltsman G, Banerjee S, Cohen-Kfir E, Dessau M, Rosenzweig R, et al: Structural basis for UFM1 transfer from UBA5 to UFC1. Nat Commun. 12:57082021. View Article : Google Scholar : PubMed/NCBI

35 

Tolmachova KA, Farnung J, Liang JR, Corn JE and Bode JW: Facile preparation of UFMylation activity-based probes by chemoselective installation of electrophiles at the C-terminus of recombinant UFM1. ACS Cent Sci. 8:756–762. 2022. View Article : Google Scholar : PubMed/NCBI

36 

Kumari S, Banerjee S, Kumar M, Hayashi A, Solaimuthu B, Cohen-Kfir E, Shaul YD, Rouvinski A and Wiener R: Overexpression of UBA5 in cells mimics the phenotype of cells lacking UBA5. Int J Mol Sci. 23:74452022. View Article : Google Scholar : PubMed/NCBI

37 

Liess AKL, Kucerova A, Schweimer K, Yu L, Roumeliotis TI, Diebold M, Dybkov O, Sotriffer C, Urlaub H, Choudhary JS, et al: Autoinhibition mechanism of the ubiquitin-conjugating enzyme UBE2S by autoubiquitination. Structure. 27:1195–1210. e72019. View Article : Google Scholar : PubMed/NCBI

38 

Tatsumi K, Sou YS, Tada N, Nakamura E, Iemura S, Natsume T, Kang SH, Chung CH, Kasahara M, Kominami E, et al: A novel type of E3 ligase for the Ufm1 conjugation system. J Biol Chem. 285:5417–5427. 2009. View Article : Google Scholar : PubMed/NCBI

39 

Peter JJ, Magnussen HM, DaRosa PA, Millrine D, Matthews SP, Lamoliatte F, Sundaramoorthy R, Kopito RR and Kulathu Y: A non-canonical scaffold-type E3 ligase complex mediates protein UFMylation. EMBO J. 41:e1110152022. View Article : Google Scholar : PubMed/NCBI

40 

Deshaies RJ and Joazeiro CA: RING domain E3 ubiquitin ligases. Annu Rev Biochem. 78:399–434. 2009. View Article : Google Scholar : PubMed/NCBI

41 

Deol KK, Lorenz S and Strieter ER: Enzymatic logic of ubiquitin chain assembly. Front Physiol. 10:8352019. View Article : Google Scholar : PubMed/NCBI

42 

Jiang Q, Wang Y, Xiang M, Hua J, Zhou T, Chen F, Lv X, Huang J and Cai Y: UFL1, a UFMylation E3 ligase, plays a crucial role in multiple cellular stress responses. Front Endocrinol (Lausanne). 14:11231242023. View Article : Google Scholar : PubMed/NCBI

43 

Yoo HM, Kang SH, Kim JY, Lee JE, Seong MW, Lee SW, Ka SH, Sou YS, Komatsu M, Tanaka K, et al: Modification of ASC1 by UFM1 is crucial for ERα transactivation and breast cancer development. Mol Cell. 56:261–274. 2014. View Article : Google Scholar : PubMed/NCBI

44 

Lee L, Oliva AB, Martinez-Balsalobre E, Churikov D, Peter J, Rahmouni D, Audoly G, Azzoni V, Audebert S, Camoin L, et al: UFMylation of MRE11 is essential for telomere length maintenance and hematopoietic stem cell survival. Sci Adv. 7:eabc73712021. View Article : Google Scholar : PubMed/NCBI

45 

Qin B, Yu J, Nowsheen S, Wang M, Tu X, Liu T, Li H, Wang L and Lou Z: UFL1 promotes histone H4 ufmylation and ATM activation. Nat Commun. 10:12422019. View Article : Google Scholar : PubMed/NCBI

46 

Wang Z, Gong Y, Peng B, Shi R, Fan D, Zhao H, Zhu M, Zhang H, Lou Z, Zhou J, et al: MRE11 UFMylation promotes ATM activation. Nucleic Acids Res. 47:4124–4135. 2019. View Article : Google Scholar : PubMed/NCBI

47 

Liu J, Guan D, Dong M, Yang J, Wei H, Liang Q, Song L, Xu L, Bai J, Liu C, et al: UFMylation maintains tumour suppressor p53 stability by antagonizing its ubiquitination. Nat Cell Biol. 22:1056–1063. 2020. View Article : Google Scholar : PubMed/NCBI

48 

Lee JH and Paull TT: Activation and regulation of ATM kinase activity in response to DNA double-strand breaks. Oncogene. 26:7741–7748. 2007. View Article : Google Scholar : PubMed/NCBI

49 

Bakkenist CJ and Kastan MB: DNA damage activates ATM through intermolecular autophosphorylation and dimer dissociation. Nature. 421:499–506. 2003. View Article : Google Scholar : PubMed/NCBI

50 

Qin B, Yu J, Nowsheen S, Zhao F, Wang L and Lou Z: STK38 promotes ATM activation by acting as a reader of histone H4 ufmylation. Sci Adv. 6:eaax82142020. View Article : Google Scholar : PubMed/NCBI

51 

Fang Z and Pan Z: Essential role of ubiquitin-fold modifier 1 conjugation in DNA damage response. DNA Cell Biol. 38:1030–1039. 2019. View Article : Google Scholar : PubMed/NCBI

52 

Sun Y, Jiang X, Chen S, Fernandes N and Price BD: A role for the Tip60 histone acetyltransferase in the acetylation and activation of ATM. Proc Natl Acad Sci USA. 102:13182–13187. 2005. View Article : Google Scholar : PubMed/NCBI

53 

Liu J, Wang Y, Song L, Zeng L, Yi W, Liu T, Chen H, Wang M, Ju Z and Cong YS: A critical role of DDRGK1 in endoplasmic reticulum homoeostasis via regulation of IRE1α stability. Nat Commun. 8:141862017. View Article : Google Scholar : PubMed/NCBI

54 

Rusiecki R, Witkowski J and Jaszczewska-Adamczak J: MDM2-p53 interaction inhibitors: The current state-of-art and updated patent review (2010-Present). Recent Pat Anticancer Drug Discov. 14:324–369. 2019. View Article : Google Scholar : PubMed/NCBI

55 

Wang S, Zhao Y, Aguilar A, Bernard D and Yang CY: Targeting the MDM2-p53 protein-protein interaction for new cancer therapy: Progress and challenges. Cold Spring Harb Perspect Med. 7:a0262452017. View Article : Google Scholar : PubMed/NCBI

56 

Adams KE, Medhurst AL, Dart DA and Lakin ND: Recruitment of ATR to sites of ionising radiation-induced DNA damage requires ATM and components of the MRN protein complex. Oncogene. 25:3894–3904. 2006. View Article : Google Scholar : PubMed/NCBI

57 

Vazifehmand R, Ali DS, Homaie FM, Jalalvand FM, Othman Z, Deming C, Stanslas J and Sekawi Z: Effects of HSV-G47Delta oncolytic virus on telomerase and telomere length alterations in glioblastoma multiforme cancer stem cells under hypoxia and normoxia conditions. Curr Cancer Drug Targets. 24:1262–1274. 2024. View Article : Google Scholar : PubMed/NCBI

58 

Ufmylation of ASC1 is essential for breast cancer development. Cancer Discov. 4:OF102014. View Article : Google Scholar

59 

Chen Q, Xiao Y, Chai P, Zheng P, Teng J and Chen J: ATL3 is a tubular ER-Phagy receptor for GABARAP-mediated selective autophagy. Curr Biol. 29:846–855. e62019. View Article : Google Scholar : PubMed/NCBI

60 

Stephani M, Picchianti L, Gajic A, Beveridge R, Skarwan E, de Medina Hernandez V, Mohseni A, Clavel M, Zeng Y, Naumann C, et al: A cross-kingdom conserved ER-phagy receptor maintains endoplasmic reticulum homeostasis during stress. Elife. 9:e583962020. View Article : Google Scholar : PubMed/NCBI

61 

Simsek D, Tiu GC, Flynn RA, Byeon GW, Leppek K, Xu AF, Chang HY and Barna M: The mammalian Ribo-interactome reveals ribosome functional diversity and heterogeneity. Cell. 169:1051–1065. e182017. View Article : Google Scholar : PubMed/NCBI

62 

Komatsu M, Inada T and Noda NN: The UFM1 system: Working principles, cellular functions, and pathophysiology. Mol Cell. 84:156–169. 2023. View Article : Google Scholar : PubMed/NCBI

63 

Ishimura R, Ito S, Mao G, Komatsu-Hirota S, Inada T, Noda NN and Komatsu M: Mechanistic insights into the roles of the UFM1 E3 ligase complex in ufmylation and ribosome-associated protein quality control. Sci Adv. 9:eadh36352023. View Article : Google Scholar : PubMed/NCBI

64 

Walczak CP, Leto DE, Zhang L, Riepe C, Muller RY, DaRosa PA, Ingolia NT, Elias JE and Kopito RR: Ribosomal protein RPL26 is the principal target of UFMylation. Proc Natl Acad Sci USA. 116:1299–1308. 2019. View Article : Google Scholar : PubMed/NCBI

65 

Scavone F, Gumbin SC, Da Rosa PA and Kopito RR: RPL26/uL24 UFMylation is essential for ribosome-associated quality control at the endoplasmic reticulum. Proc Natl Acad Sci USA. 120:e22203401202023. View Article : Google Scholar : PubMed/NCBI

66 

Wang L, Xu Y, Rogers H, Saidi L, Noguchi CT, Li H, Yewdell JW, Guydosh NR and Ye Y: UFMylation of RPL26 links translocation-associated quality control to endoplasmic reticulum protein homeostasis. Cell Res. 30:5–20. 2019. View Article : Google Scholar : PubMed/NCBI

67 

French SW, Masouminia M, Samadzadeh S, Tillman BC, Mendoza A and French BA: Role of protein quality control failure in alcoholic hepatitis pathogenesis. Biomolecules. 7:112017. View Article : Google Scholar : PubMed/NCBI

68 

Luo H, Jiao QB, Shen CB, Gong WY, Yuan JH, Liu YY, Chen Z, Liu J, Xu XL, Cong YS and Zhang XW: UFMylation of HRD1 regulates endoplasmic reticulum homeostasis. FASEB J. 37:e232212023. View Article : Google Scholar : PubMed/NCBI

69 

Wilkinson S: Emerging principles of selective ER autophagy. J Mol Biol. 432:185–205. 2019. View Article : Google Scholar : PubMed/NCBI

70 

Chino H and Mizushima N: ER-Phagy: Quality control and turnover of endoplasmic reticulum. Trends Cell Biol. 30:384–398. 2020. View Article : Google Scholar : PubMed/NCBI

71 

Picchianti L, de Medina Hernandez V, Zhan N, Irwin NA, Groh R, Stephani M, Hornegger H, Beveridge R, Sawa-Makarska J, Lendl T, et al: Shuffled ATG8 interacting motifs form an ancestral bridge between UFMylation and autophagy. EMBO J. 42:e1120532023. View Article : Google Scholar : PubMed/NCBI

72 

Stephani M, Picchianti L and Dagdas Y: C53 is a cross-kingdom conserved reticulophagy receptor that bridges the gap betweenselective autophagy and ribosome stalling at the endoplasmic reticulum. Autophagy. 17:586–587. 2020. View Article : Google Scholar : PubMed/NCBI

73 

Klebanovych A, Vinopal S, Draberova E, Sladkova V, Sulimenko T, Sulimenko V, Vosecká V, Macůrek L, Legido A and Dráber P: C53 Interacting with UFM1-protein ligase 1 regulates microtubule nucleation in response to ER stress. Cells. 11:5552022. View Article : Google Scholar : PubMed/NCBI

74 

Reggiori F and Molinari M: ER-phagy: Mechanisms, regulation, and diseases connected to the lysosomal clearance of the endoplasmic reticulum. Physiol Rev. 102:1393–1448. 2022. View Article : Google Scholar : PubMed/NCBI

75 

Zhu H, Bhatt B, Sivaprakasam S, Cai Y, Liu S, Kodeboyina SK, Patel N, Savage NM, Sharma A, Kaufman RJ, et al: Ufbp1 promotes plasma cell development and ER expansion by modulating distinct branches of UPR. Nat Commun. 10:10842019. View Article : Google Scholar : PubMed/NCBI

76 

Ishimura R, El-Gowily AH, Noshiro D, Komatsu-Hirota S, Ono Y, Shindo M, Hatta T, Abe M, Uemura T, Lee-Okada HC, et al: The UFM1 system regulates ER-phagy through the ufmylation of CYB5R3. Nat Commun. 13:78572022. View Article : Google Scholar : PubMed/NCBI

77 

Liang JR, Lingeman E, Luong T, Ahmed S, Muhar M, Nguyen T, Olzmann JA and Corn JE: A genome-wide ER-phagy screen highlights key roles of mitochondrial metabolism and ER-resident UFMylation. Cell. 180:1160–1177. e202020. View Article : Google Scholar : PubMed/NCBI

78 

Raymundo DP, Doultsinos D, Guillory X, Carlesso A, Eriksson LA and Chevet E: Pharmacological targeting of IRE1 in cancer. Trends Cancer. 6:1018–1030. 2020. View Article : Google Scholar : PubMed/NCBI

79 

Cai Y, Zhu G, Liu S, Pan Z, Quintero M, Poole CJ, Lu C, Zhu H, Islam B, Riggelen JV, et al: Indispensable role of the Ubiquitin-fold modifier 1-specific E3 ligase in maintaining intestinal homeostasis and controlling gut inflammation. Cell Discov. 5:72019. View Article : Google Scholar : PubMed/NCBI

80 

Hwang J and Qi L: Quality control in the endoplasmic reticulum: Crosstalk between ERAD and UPR pathways. Trends Biochem Sci. 43:593–605. 2018. View Article : Google Scholar : PubMed/NCBI

81 

Lemaire K, Moura RF, Granvik M, Igoillo-Esteve M, Hohmeier HE, Hendrickx N, Newgard CB, Waelkens E, Cnop M and Schuit F: Ubiquitin fold modifier 1 (UFM1) and its target UFBP1 protect pancreatic beta cells from ER stress-induced apoptosis. PLoS One. 6:e185172011. View Article : Google Scholar : PubMed/NCBI

82 

DaRosa PA, Penchev I, Gumbin SC, Scavone F, Wachalska M, Paulo JA, Ordureau A, Peter JJ, Kulathu Y, Harper JW, et al: UFM1 E3 ligase promotes recycling of 60S ribosomal subunits from the ER. Nature. 627:445–452. 2024. View Article : Google Scholar : PubMed/NCBI

83 

Makhlouf L, Peter JJ, Magnussen HM, Thakur R, Millrine D, Minshull TC, Harrison G, Varghese J, Lamoliatte F, Foglizzo M, et al: The UFM1 E3 ligase recognizes and releases 60S ribosomes from ER translocons. Nature. 627:437–444. 2024. View Article : Google Scholar : PubMed/NCBI

84 

von der Malsburg K, Shao S and Hegde RS: The ribosome quality control pathway can access nascent polypeptides stalled at the Sec61 translocon. Mol Biol Cell. 26:2168–2180. 2015. View Article : Google Scholar : PubMed/NCBI

85 

Brandman O and Hegde RS: Ribosome-associated protein quality control. Nat Struct Mol Biol. 23:7–15. 2016. View Article : Google Scholar : PubMed/NCBI

86 

Wan L, Juszkiewicz S, Blears D, Bajpe PK, Han Z, Faull P, Mitter R, Stewart A, Snijders AP, Hegde RS and Svejstrup JQ: Translation stress and collided ribosomes are co-activators of cGAS. Mol Cell. 81:2808–2822. e102021. View Article : Google Scholar : PubMed/NCBI

87 

Inada T: Quality controls induced by aberrant translation. Nucleic Acids Res. 48:1084–1096. 2020. View Article : Google Scholar : PubMed/NCBI

88 

Wang L, Xu Y, Yun S, Yuan Q, Satpute-Krishnan P and Ye Y: SAYSD1 senses UFMylated ribosome to safeguard co-translational protein translocation at the endoplasmic reticulum. Cell Rep. 42:1120282023. View Article : Google Scholar : PubMed/NCBI

89 

Joazeiro CAP: Mechanisms and functions of ribosome-associated protein quality control. Nat Rev Mol Cell Biol. 20:368–383. 2019. View Article : Google Scholar : PubMed/NCBI

90 

Cai Y, Singh N and Li H: Essential role of Ufm1 conjugation in the hematopoietic system. Exp Hematol. 44:442–446. 2016. View Article : Google Scholar : PubMed/NCBI

91 

Hattangadi SM, Wong P, Zhang L, Flygare J and Lodish HF: From stem cell to red cell: Regulation of erythropoiesis at multiple levels by multiple proteins, RNAs, and chromatin modifications. Blood. 118:6258–6268. 2011. View Article : Google Scholar : PubMed/NCBI

92 

Tatsumi K, Yamamoto-Mukai H, Shimizu R, Waguri S, Sou YS, Sakamoto A, Taya C, Shitara H, Hara T, Chung CH, et al: The Ufm1-activating enzyme Uba5 is indispensable for erythroid differentiation in mice. Nat Commun. 2:1812011. View Article : Google Scholar : PubMed/NCBI

93 

Zhang M, Zhu X, Zhang Y, Cai Y, Chen J, Sivaprakasam S, Gurav A, Pi W, Makala L, Wu J, et al: RCAD/Ufl1, a Ufm1 E3 ligase, is essential for hematopoietic stem cell function and murine hematopoiesis. Cell Death Differ. 22:1922–1934. 2015. View Article : Google Scholar : PubMed/NCBI

94 

Zhang Y, Zhang M, Wu J, Lei G and Li H: Transcriptional regulation of the Ufm1 conjugation system in response to disturbance of the endoplasmic reticulum homeostasis and inhibition of vesicle trafficking. PLoS One. 7:e485872012. View Article : Google Scholar : PubMed/NCBI

95 

Di Rocco M, Rusmini M, Caroli F, Madeo A, Bertamino M, Marre-Brunenghi G and Ceccherini I: Novel spondyloepimetaphyseal dysplasia due to UFSP2 gene mutation. Clin Genet. 93:671–674. 2017. View Article : Google Scholar : PubMed/NCBI

96 

Zhang G, Tang S, Wang H, Pan H, Zhang W, Huang Y, Kong J and Wang Y, Gu J and Wang Y: Corrigendum to UFSP2-related spondyloepimetaphyseal dysplasia: A confirmatory report. Eur J Med Genet. 63:1040212020. View Article : Google Scholar : PubMed/NCBI

97 

Franceschi R, Iascone M, Maitz S, Marchetti D, Mariani M, Selicorni A, Soffiati M and Maines E: A missense mutation in DDRGK1 gene associated to Shohat-type spondyloepimetaphyseal dysplasia: Two case reports and a review of literature. Am J Med Genet A. 188:2434–2437. 2022. View Article : Google Scholar : PubMed/NCBI

98 

Weisz-Hubshman M, Egunsula AT, Dawson B, Castellon A, Jiang MM, Chen-Evenson Y, Zhiyin Y, Lee B and Bae Y: DDRGK1 is required for the proper development and maintenance of the growth plate cartilage. Hum Mol Genet. 31:2820–2830. 2022. View Article : Google Scholar : PubMed/NCBI

99 

Ni M, Afroze B, Xing C, Pan C, Shao Y, Cai L, Cantarel BL, Pei J, Grishin NV, Hewson S, et al: A pathogenic UFSP2 variant in an autosomal recessive form of pediatric neurodevelopmental anomalies and epilepsy. Genet Med. 23:900–908. 2021. View Article : Google Scholar : PubMed/NCBI

100 

Muona M, Ishimura R, Laari A, Ichimura Y, Linnankivi T, Keski-Filppula R, Herva R, Rantala H, Paetau A, Pöyhönen M, et al: Biallelic variants in UBA5 link dysfunctional ufm1 ubiquitin-like modifier pathway to severe infantile-onset encephalopathy. Am J Hum Genet. 99:683–694. 2016. View Article : Google Scholar : PubMed/NCBI

101 

Zhang J, Zhu H, Liu S, Quintero M, Zhu T, Xu R, Cai Y, Han Y and Li H: Deficiency of murine UFM1-Specific E3 ligase causes microcephaly and inflammation. Mol Neurobiol. 59:6363–6372. 2022. View Article : Google Scholar : PubMed/NCBI

102 

Arnadottir GA, Jensson BO, Marelsson SE, Sulem G, Oddsson A, Kristjansson RP, Benonisdottir S, Gudjonsson SA, Masson G, Thorisson GA, et al: Compound heterozygous mutations in UBA5 causing early-onset epileptic encephalopathy in two sisters. BMC Med Genet. 18:1032017. View Article : Google Scholar : PubMed/NCBI

103 

Nahorski MS, Maddirevula S, Ishimura R, Alsahli S, Brady AF, Begemann A, Mizushima T, Guzmán-Vega FJ, Obata M, Ichimura Y, et al: Biallelic UFM1 and UFC1 mutations expand the essential role of ufmylation in brain development. Brain. 141:1934–1945. 2018. View Article : Google Scholar : PubMed/NCBI

104 

Serrano RJ, Oorschot V, Palipana D, Calcinotto V, Sonntag C, Ramm G and Bryson-Richardson RJ: Genetic model of UBA5 deficiency highlights the involvement of both peripheral and central nervous systems and identifies widespread mitochondrial abnormalities. Brain Commun. 5:fcad3172023. View Article : Google Scholar : PubMed/NCBI

105 

Cabrera-Serrano M, Coote DJ, Azmanov D, Goullee H, Andersen E, McLean C, Davis M, Ishimura R, Stark Z, Vallat JM, et al: A homozygous UBA5 pathogenic variant causes a fatal congenital neuropathy. J Med Genet. 57:835–842. 2020. View Article : Google Scholar : PubMed/NCBI

106 

Mignon-Ravix C, Milh M, Kaiser CS, Daniel J, Riccardi F, Cacciagli P, Nagara M, Busa T, Liebau E and Villard L: Abnormal function of the UBA5 protein in a case of early developmental and epileptic encephalopathy with suppression-burst. Hum Mutat. 39:934–938. 2018. View Article : Google Scholar : PubMed/NCBI

107 

Duan R, Shi Y, Yu L, Zhang G, Li J, Lin Y, Guo J, Wang J, Shen L, Jiang H, et al: UBA5 mutations cause a new form of autosomal recessive cerebellar ataxia. PLoS One. 11:e01490392016. View Article : Google Scholar : PubMed/NCBI

108 

Al-Saady ML, Kaiser CS, Wakasuqui F, Korenke GC, Waisfisz Q, Polstra A, Pouwels PJW, Bugiani M, van der Knaap MS, Lunsing RJ, et al: Homozygous UBA5 variant leads to hypomyelination with thalamic involvement and axonal neuropathy. Neuropediatrics. 52:489–494. 2021. View Article : Google Scholar : PubMed/NCBI

109 

Yiu SPT, Zerbe C, Vanderwall D, Huttlin EL, Weekes MP and Gewurz BE: An Epstein-Barr virus protein interaction map reveals NLRP3 inflammasome evasion via MAVS UFMylation. Mol Cell. 83:2367–2386. e152023. View Article : Google Scholar : PubMed/NCBI

110 

Snider DL, Park M, Murphy KA, Beachboard DC and Horner SM: Signaling from the RNA sensor RIG-I is regulated by ufmylation. Proc Natl Acad Sci USA. 119:e21195311192022. View Article : Google Scholar : PubMed/NCBI

111 

Xie Z, Fang Z and Pan Z: Ufl1/RCAD, a Ufm1 E3 ligase, has an intricate connection with ER stress. Int J Biol Macromol. 135:760–767. 2019. View Article : Google Scholar : PubMed/NCBI

112 

DeSantis CE, Ma J, Gaudet MM, Newman LA, Miller KD, Sauer A, Jemal A and Siegel RL: Breast cancer statistics, 2019. CA Cancer J Clin. 69:438–451. 2019. View Article : Google Scholar : PubMed/NCBI

113 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2020. CA Cancer J Clin. 70:7–30. 2020. View Article : Google Scholar : PubMed/NCBI

114 

Li Z, Wei H, Li S, Wu P and Mao X: The role of progesterone receptors in breast cancer. Drug Des Devel Ther. 16:305–314. 2022. View Article : Google Scholar : PubMed/NCBI

115 

Jozwik KM and Carroll JS: Pioneer factors in hormone-dependent cancers. Nat Rev Cancer. 12:381–385. 2012. View Article : Google Scholar : PubMed/NCBI

116 

Yoo HM, Park JH, Kim JY and Chung CH: Modification of ERα by UFM1 increases its stability and transactivity for breast cancer development. Mol Cells. 45:425–434. 2022. View Article : Google Scholar : PubMed/NCBI

117 

Yoo HM, Park JH, Jeon YJ and Chung CH: Ubiquitin-fold modifier 1 acts as a positive regulator of breast cancer. Front Endocrinol (Lausanne). 6:362015. View Article : Google Scholar : PubMed/NCBI

118 

Le Romancer M, Poulard C, Cohen P, Sentis S, Renoir JM and Corbo L: Cracking the estrogen receptor's posttranslational code in breast tumors. Endocr Rev. 32:597–622. 2011. View Article : Google Scholar : PubMed/NCBI

119 

Schulten HJ: Pleiotropic effects of metformin on cancer. Int J Mol Sci. 19:28502018. View Article : Google Scholar : PubMed/NCBI

120 

Yang J, Zhou Y, Xie S, Wang J, Li Z, Chen L, Mao M, Chen C, Huang A, Chen Y, et al: Metformin induces ferroptosis by inhibiting UFMylation of SLC7A11 in breast cancer. J Exp Clin Cancer Res. 40:2062021. View Article : Google Scholar : PubMed/NCBI

121 

Mao M, Chen Y, Yang J, Cheng Y, Xu L, Ji F, Zhou J, Zhang X, Li Z, Chen C, et al: Modification of PLAC8 by UFM1 affects tumorous proliferation and immune response by impacting PD-L1 levels in triple-negative breast cancer. J Immunother Cancer. 10:e0056682022. View Article : Google Scholar : PubMed/NCBI

122 

Feng X, Wei Z, Tao X, Du Y, Wu J, Yu Y, Yu H and Zhao H: PLAC8 promotes the autophagic activity and improves the growth priority of human trophoblast cells. FASEB J. 35:e213512021. View Article : Google Scholar : PubMed/NCBI

123 

Hsu JM, Li CW, Lai YJ and Hung MC: Posttranslational modifications of PD-L1 and their applications in cancer therapy. Cancer Res. 78:6349–6353. 2018. View Article : Google Scholar : PubMed/NCBI

124 

Lim SO, Li CW, Xia W, Cha JH, Chan LC, Wu Y, Chang SS, Lin WC, Hsu JM, Hsu YH, et al: Deubiquitination and stabilization of PD-L1 by CSN5. Cancer Cell. 30:925–939. 2016. View Article : Google Scholar : PubMed/NCBI

125 

Kulsuptrakul J, Wang R, Meyers NL, Ott M and Puschnik AS: A genome-wide CRISPR screen identifies UFMylation and TRAMP-like complexes as host factors required for hepatitis A virus infection. Cell Rep. 34:1088592021. View Article : Google Scholar : PubMed/NCBI

126 

French SW, Bardag-Gorce F, Li J, French BA and Oliva J: Mallory-Denk body pathogenesis revisited. World J Hepatol. 2:295–301. 2010. View Article : Google Scholar : PubMed/NCBI

127 

Bardag-Gorce F, Oliva J, Villegas J, Fraley S, Amidi F, Li J, Dedes J, French B and French SW: Epigenetic mechanisms regulate Mallory Denk body formation in the livers of drug-primed mice. Exp Mol Pathol. 84:113–121. 2008. View Article : Google Scholar : PubMed/NCBI

128 

Li J, Li XM, Caudill M, Malysheva O, Bardag-Gorce F, Oliva J, French BA, Gorce E, Morgan K, Kathirvel E, et al: Betaine feeding prevents the blood alcohol cycle in rats fed alcohol continuously for 1 month using the rat intragastric tube feeding model. Exp Mol Pathol. 91:540–547. 2011. View Article : Google Scholar : PubMed/NCBI

129 

Oliva J, Bardag-Gorce F, Li J, French BA, Nguyen SK, Lu SC and French SW: Betaine prevents Mallory-Denk body formation in drug-primed mice by epigenetic mechanisms. Exp Mol Pathol. 86:77–86. 2008. View Article : Google Scholar : PubMed/NCBI

130 

Liu H, Gong M, French BA, Li J, Tillman B and French SW: Mallory-Denk body (MDB) formation modulates Ufmylation expression epigenetically in alcoholic hepatitis (AH) and non-alcoholic steatohepatitis (NASH). Exp Mol Pathol. 97:477–483. 2014. View Article : Google Scholar : PubMed/NCBI

131 

Dong Y and Wang A: Aberrant DNA methylation in hepatocellular carcinoma tumor suppression (Review). Oncol Lett. 8:963–968. 2014. View Article : Google Scholar : PubMed/NCBI

132 

Esteller M, Corn PG, Baylin SB and Herman JG: A gene hypermethylation profile of human cancer. Cancer Res. 61:3225–3229. 2001.PubMed/NCBI

133 

Liu H, Li J, Tillman B, French BA and French SW: Ufmylation and FATylation pathways are downregulated in human alcoholic and nonalcoholic steatohepatitis, and mice fed DDC, where Mallory-Denk bodies (MDBs) form. Exp Mol Pathol. 97:81–88. 2014. View Article : Google Scholar : PubMed/NCBI

134 

Li H, Rauch T, Chen ZX, Szabo PE, Riggs AD and Pfeifer GP: The histone methyltransferase SETDB1 and the DNA methyltransferase DNMT3A interact directly and localize to promoters silenced in cancer cells. J Biol Chem. 281:19489–19500. 2006. View Article : Google Scholar : PubMed/NCBI

135 

Fagerberg L, Hallstrom BM, Oksvold P, Kampf C, Djureinovic D, Odeberg J, Habuka M, Tahmasebpoor S, Danielsson A, Edlund K, et al: Analysis of the human tissue-specific expression by genome-wide integration of transcriptomics and antibody-based proteomics. Mol Cell Proteomics. 13:397–406. 2014. View Article : Google Scholar : PubMed/NCBI

136 

Chen E, Zhou B, Bian S, Ni W and Chen Z: The lncRNA B3GALT5-AS1 functions as an HCC suppressor by regulating the miR-934/UFM1 axis. J Oncol. 2021:17764322021. View Article : Google Scholar : PubMed/NCBI

137 

Yang S, Yang R, Wang H, Huang Y and Jia Y: CDK5RAP3 deficiency restrains liver regeneration after partial hepatectomy triggering endoplasmic reticulum stress. Am J Pathol. 190:2403–2416. 2020. View Article : Google Scholar : PubMed/NCBI

138 

Sheng L, Li J, Rao S, Yang Z and Huang Y: Cyclin-dependent kinase 5 regulatory subunit associated protein 3: Potential functions and implications for development and disease. Front Oncol. 11:7604292021. View Article : Google Scholar : PubMed/NCBI

139 

Bade BC and Cruz CS: Lung cancer 2020: Epidemiology, etiology, and prevention. Clin Chest Med. 41:1–24. 2020. View Article : Google Scholar : PubMed/NCBI

140 

Xia C, Dong X, Li H, Cao M, Sun D, He S, Yang F, Yan X, Zhang S, Li N and Chen W: Cancer statistics in China and United States, 2022: Profiles, trends, and determinants. Chin Med J (Engl). 135:584–590. 2022. View Article : Google Scholar : PubMed/NCBI

141 

Nabet BY, Hamidi H, Lee MC, Banchereau R, Morris S, Adler L, Gayevskiy V, Elhossiny AM, Srivastava MK, Patil NS, et al: Immune heterogeneity in small-cell lung cancer and vulnerability to immune checkpoint blockade. Cancer Cell. 42:429–443. e42024. View Article : Google Scholar : PubMed/NCBI

142 

Wang X, Wu Y, Gu J and Xu J: Tumor-associated macrophages in lung carcinoma: From mechanism to therapy. Pathol Res Pract. 229:1537472022. View Article : Google Scholar : PubMed/NCBI

143 

Hu J, Zhang L, Xia H, Yan Y, Zhu X, Sun F, Sun L, Li S, Li D, Wang J, et al: Tumor microenvironment remodeling after neoadjuvant immunotherapy in non-small cell lung cancer revealed by single-cell RNA sequencing. Genome Med. 15:142023. View Article : Google Scholar : PubMed/NCBI

144 

Xu D, Zhang D, Wei W and Zhang C: UBA5 inhibition restricts lung adenocarcinoma via blocking macrophage M2 polarization and cisplatin resistance. Exp Cell Res. 440:1141482024. View Article : Google Scholar : PubMed/NCBI

145 

Kim CH, Nam HS, Lee EH, Han SH, Cho HJ, Chung HJ, Lee NS, Choi SJ, Kim H, Ryu JS, et al: Overexpression of a novel regulator of p120 catenin, NLBP, promotes lung adenocarcinoma proliferation. Cell Cycle. 12:2443–2453. 2013. View Article : Google Scholar : PubMed/NCBI

146 

Zhou J, Ma X, Xu L, Liang Q, Mao J, Liu J, Wang M, Yuan J and Cong YS: Genomic profiling of the UFMylation family genes identifies UFSP2 as a potential tumour suppressor in colon cancer. Clin Transl Med. 11:e6422021. View Article : Google Scholar : PubMed/NCBI

147 

Smyth EC, Nilsson M, Grabsch HI, van Grieken NC and Lordick F: Gastric cancer. Lancet. 396:635–648. 2020. View Article : Google Scholar : PubMed/NCBI

148 

Karimi P, Islami F, Anandasabapathy S, Freedman ND and Kamangar F: Gastric cancer: Descriptive epidemiology, risk factors, screening, and prevention. Cancer Epidemiol Biomarkers Prev. 23:700–713. 2014. View Article : Google Scholar : PubMed/NCBI

149 

Lin M, Lian NZ, Cao LL, Huang CM, Zheng CH, Li P, Xie JW, Wang JB, Lu J, Chen QY, et al: Down-regulated expression of CDK5RAP3 and UFM1 suggests a poor prognosis in gastric cancer patients. Front Oncol. 12:9277512022. View Article : Google Scholar : PubMed/NCBI

150 

Shiwaku H, Yoshimura N, Tamura T, Sone M, Ogishima S, Watase K, Tagawa K and Okazawa H: Suppression of the novel ER protein Maxer by mutant ataxin-1 in Bergman glia contributes to non-cell-autonomous toxicity. EMBO J. 29:2446–2460. 2010. View Article : Google Scholar : PubMed/NCBI

151 

Wu J, Lei G, Mei M, Tang Y and Li H: A novel C53/LZAP-interacting protein regulates stability of C53/LZAP and DDRGK domain-containing Protein 1 (DDRGK1) and modulates NF-kappaB signaling. J Biol Chem. 285:15126–15136. 2010. View Article : Google Scholar : PubMed/NCBI

152 

Lin JX, Xie XS, Weng XF, Zheng CH, Xie JW, Wang JB, Lu J, Chen QY, Cao LL, Lin M, et al: Low expression of CDK5RAP3 and DDRGK1 indicates a poor prognosis in patients with gastric cancer. World J Gastroenterol. 24:3898–3907. 2018. View Article : Google Scholar : PubMed/NCBI

153 

Xi P, Ding D, Zhou J, Wang M and Cong YS: DDRGK1 regulates NF-κB activity by modulating IκBα stability. PLoS One. 8:e642312013. View Article : Google Scholar : PubMed/NCBI

154 

Lin JX, Xie XS, Weng XF, Qiu SL, Yoon C, Lian NZ, Xie JW, Wang JB, Lu J, Chen QY, et al: UFM1 suppresses invasive activities of gastric cancer cells by attenuating the expres7sion of PDK1 through PI3K/AKT signaling. J Exp Clin Cancer Res. 38:4102019. View Article : Google Scholar : PubMed/NCBI

155 

Zhou Y, Ye X, Zhang C, Wang J, Guan Z, Yan J, Xu L, Wang K, Guan D, Liang Q, et al: Ufl1 deficiency causes kidney atrophy associated with disruption of endoplasmic reticulum homeostasis. J Genet Genomics. 48:403–410. 2021. View Article : Google Scholar : PubMed/NCBI

156 

Momayyezi P, Bilev E, Ljunggren HG and Hammer Q: Viral escape from NK-cell-mediated immunosurveillance: A lesson for cancer immunotherapy? Eur J Immunol. 53:e23504652023. View Article : Google Scholar : PubMed/NCBI

157 

Zhou J, Ma X, He X, Chen B, Yuan J, Jin Z, Li L, Wang Z, Xiao Q, Cai Y, et al: Dysregulation of PD-L1 by UFMylation imparts tumor immune evasion and identified as a potential therapeutic target. Proc Natl Acad Sci USA. 120:e22157321202023. View Article : Google Scholar : PubMed/NCBI

158 

Ma EH, Poffenberger MC, Wong AH and Jones RG: The role of AMPK in T cell metabolism and function. Curr Opin Immunol. 46:45–52. 2017. View Article : Google Scholar : PubMed/NCBI

159 

He C, Xing X, Chen HY, Gao M, Shi J, Xiang B, Xiao X, Sun Y, Yu H, Xu G, et al: UFL1 ablation in T cells suppresses PD-1 UFMylation to enhance anti-tumor immunity. Mol Cell. 84:1120–1138. e82024. View Article : Google Scholar : PubMed/NCBI

160 

Zhu MMT, Shenasa E and Nielsen TO: Sarcomas: Immune biomarker expression and checkpoint inhibitor trials. Cancer Treat Rev. 91:1021152020. View Article : Google Scholar : PubMed/NCBI

161 

Brown HK, Schiavone K, Gouin F, Heymann MF and Heymann D: Biology of bone sarcomas and new therapeutic developments. Calcif Tissue Int. 102:174–195. 2017. View Article : Google Scholar : PubMed/NCBI

162 

Meng H, Ai H, Li D, Jiang X, Zhang H, Xu J and Huang S: Bombyx mori UFBP1 regulates apoptosis and promotes BmNPV proliferation by affecting the expression of ER chaperone BmBIP. Int J Biol Macromol. 283:1376812024. View Article : Google Scholar : PubMed/NCBI

163 

Wang K, Chen S, Wu Y, Wang Y, Lu Y, Sun Y and Chen Y: The ufmylation modification of ribosomal protein L10 in the development of pancreatic adenocarcinoma. Cell Death Dis. 14:3502023. View Article : Google Scholar : PubMed/NCBI

164 

MacLeod G, Bozek DA, Rajakulendran N, Monteiro V, Ahmadi M, Steinhart Z, Kushida MM, Yu H, Coutinho FJ, Cavalli FMG, et al: Genome-wide CRISPR-Cas9 screens expose genetic vulnerabilities and mechanisms of temozolomide sensitivity in glioblastoma stem cells. Cell Rep. 27:971–986.e9. 2019. View Article : Google Scholar : PubMed/NCBI

165 

Wang S, Jia M, Su M, Hu X, Li J, Xu Y and Qiu W: Ufmylation is activated in renal cancer and is not associated with von hippel-lindau mutation. DNA Cell Biol. 39:654–660. 2020. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2025
Volume 32 Issue 1

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Qin R, Tang Y, Yuan Y, Meng F, Zheng K, Yang X, Zhao J and Yang C: Studies on the functional role of UFMylation in cells (Review). Mol Med Rep 32: 191, 2025.
APA
Qin, R., Tang, Y., Yuan, Y., Meng, F., Zheng, K., Yang, X. ... Yang, C. (2025). Studies on the functional role of UFMylation in cells (Review). Molecular Medicine Reports, 32, 191. https://doi.org/10.3892/mmr.2025.13556
MLA
Qin, R., Tang, Y., Yuan, Y., Meng, F., Zheng, K., Yang, X., Zhao, J., Yang, C."Studies on the functional role of UFMylation in cells (Review)". Molecular Medicine Reports 32.1 (2025): 191.
Chicago
Qin, R., Tang, Y., Yuan, Y., Meng, F., Zheng, K., Yang, X., Zhao, J., Yang, C."Studies on the functional role of UFMylation in cells (Review)". Molecular Medicine Reports 32, no. 1 (2025): 191. https://doi.org/10.3892/mmr.2025.13556