- Review
- Open access
- Published:
The many connections of UFMylation with Alzheimer’s disease: a comprehensive review
Molecular Neurodegeneration volume 20, Article number: 66 (2025)
Abstract
Alzheimer’s disease (AD) is a complex neurodegenerative disorder that is characterized by the accumulation of pathologic tau and beta-amyloid proteins. UFMylation is an emerging ubiquitin-like post-translational modification that is crucial for healthy brain development. The UFM1 cascade was recently identified as a major modifier of tau aggregation in vitro and in vivo. Moreover, post-mortem AD brain shows pronounced alterations of UFMylation that are significantly associated with pathological tau, suggesting UFM1 might indeed be a modifier of human disease. However, the link between AD and UFMylation is yet to be fully explored. Interestingly, the UFMylation cascade is known to play important roles for several pathways that are known to be altered in AD, such as the DNA damage response, ER homeostasis, autophagy and the immune response. This review discusses the many connections between UFMylation with AD pathogenesis, emphasizing the role of UFMylation in these pathways and their abnormalities in AD. Understanding these connections is important to elucidate molecular mechanisms how UFM1 may impact AD and to uncover novel therapeutic strategies targeting UFMylation pathways for disease modification.
Introduction
Alzheimer’s disease (AD) is a devastating neurodegenerative disorder that affects millions of people worldwide, causing progressive cognitive decline and memory loss [1, 2]. Despite extensive research, the underlying mechanisms of AD are not yet fully understood, and effective therapies remain elusive. Pathologic tau and amyloid-β (Aβ) deposits are the main neuropathological hallmarks of AD [3]. Their accumulation has been associated with abnormalities in various cellular processes, including DNA damage response (DDR) [4,5,6,7], endoplasmic reticulum (ER) homeostasis [8,9,10,11], autophagy functions [12,13,14,15], as well as the immune response [16,17,18,19]. These processes are critical for maintaining neuronal health, and their dysregulation has significant implications for the development and progression of AD.
Interestingly, many studies have highlighted the importance of UFMylation in these processes [20,21,22,23,24]. UFMylation is a ubiquitin-like (UBL) posttranslational modification that was discovered 20 years ago [24], but remains understudied. The UFM1 cascade is essential for proper brain development, as evidenced by the observation that complete loss of function in any component of the UFMylation pathway results in severe neurodevelopmental disorders [25,26,27,28,29]. Given this essential role of UFMylation for brain, this pathway may also play a significant role in the pathogenesis of age-related neurodegenerative diseases, such as AD. However, the role of UFMylation in AD is only beginning to emerge and remains to be fully explored.
In this review, we provide an overview of the current understanding of UFMylation and its role in brain development and neurodevelopmental disorders, and we summarize the latest findings with regards to neurodegenerative disease. Given the strong overlap between pathways with UFM1 function and abnormities in AD, we further discuss common specific cellular processes such as the DDR, ER homeostasis, autophagy, and the immune response. We further propose future research directions to advance our understanding of the potential link between UFMylation and AD and discuss potential therapeutic strategies targeting UFMylation for the treatment of AD (Fig. 1).
Key reported biological functions of UFMylation. The UFMylation pathway is known to play a role in various physiological processes including DNA damage response (DDR), endoplasmic reticulum (ER) stress response, regulating autophagy functions, and influencing the immune response. There is also evidence suggesting its involvement in mitochondrial and ribosome-associated protein quality controls. Dysregulation of UFMylation has been associated with developmental disorders, particularly neurodevelopmental disorders, various types of cancers, and it may also have a potential link with neurodegenerative diseases like Alzheimer’s Disease (AD). Reported proteins modified by UFM1 have been categorized based on the pathways they are involved in. Proteins labeled with “*” have been connected with AD
The ufmylation pathway
Ubiquitin fold modifier 1 (UFM1) is a compact protein with 85 amino acids, and only a 15% sequence homology to ubiquitin [24, 30]. Its three-dimensional structure, however, which is characterized by a beta-grasp fold, is reminiscent of ubiquitin [30]. The process by which UFM1 is conjugated to its target proteins, termed UFMylation, follows a cascade involving a UFM1-specific set of E1, E2, and E3 enzymes, in analogy to the ubiquitin system [24, 31, 32] (Fig. 2). The initiation of this process necessitates the maturation of the precursor form of UFM1, known as proUFM1. This maturation is mediated by specific cysteine proteases, UFM1-specific peptidase UFSP1 and UFSP2, which trim the C-terminal Ser-Cys dipeptide of proUFM1 [33,34,35]. This reveals a single glycine residue, a departure from the typical diglycine C-terminus observed in other ubiquitin-like proteins (UBLs) [33].
Cascade of the UFMylation and deUFMylation processes. The UFMylation pathway initiates with the precursor of UFM1 (pro-UFM). In the first step, UFM1-specific cysteine proteases, UFSP1 and UFSP2, enzymes cleave two amino acids from pro-UFM, resulting in mature UFM1 that exposes a glycine residue. Subsequently, UBA5 activates this mature UFM1. Once activated, UFM1 is handed over to UFC1. The critical step of ligating UFM1 to its target proteins is conducted by UFL1, aided by two adaptor proteins, DDRGK1 and CDK5RAP3, which expand the range of potential substrates for UFL1. The UFMylation process is reversible, with UFSP1/2 cleaving UFM1 from its substrates for recycling. Abbreviations: UFC1, ubiquitin fold-modifier conjugating enzyme 1; UFL1, ubiquitin fold-modifier specific ligase 1; UFM1, ubiquitin-fold modifier 1; VGSC: valine–glycine–serine–cysteine amino acids
Following its maturation, UFM1 undergoes activation via the pathway’s E1 enzyme, specifically referred to as ubiquitin activating enzyme 5 (UBA5) [24]. This activation occurs through an ATP-dependent mechanism, where UBA5 forms a high-energy thioester bond with the C-terminal glycine residue of UFM1 [36]. Next, UFM1 is transferred to the E2 conjugating enzyme in the cascade, Ubiquitin-fold modifier-conjugating enzyme 1 (UFC1) [24]. UFC1 also forms a thioester bond with UFM1, effectively accepting the activated UFM1 from UBA5 [37, 38]. This transfer is crucial for the subsequent ligation of UFM1 to its target protein, which is mediated by the E3 ligase in the cascade, UFM1-ligase 1 (UFL1) [31]. UFL1 plays a central role in determining the substrate specificity of the UFM1 conjugation system. It recognizes and binds to specific target proteins and facilitates the transfer of UFM1 from UFC1 to the target protein’s lysine residue [32]. This transfer results in the formation of an isopeptide bond between UFM1 and the target protein, effectively modifying the target protein and altering its function or stability. The exact molecular mechanisms and determinants of substrate recognition by UFL1 are not yet fully understood, and further research is needed to elucidate the principles governing substrate specificity in the UFM1 conjugation system [22, 25]. DDRGK1/UFBP1 and CDK5RAP3 are important components of the UFM1 conjugation system. DDRGK1 has been shown to form a large protein complex with UFL1, UFM1, and CDK5RAP3, suggesting a coordinated and spatially regulated role of these proteins in the UFM1 conjugation system [39]. DDRGK1 is mainly localized at the ER, and this might explain the outsized role of UFMylation for ER homeostasis detailed below [40].
The removal of UFM1 from target proteins is an essential process that allows for the recycling of UFM1 and the reversal of UFMylation-mediated protein modification. The deconjugation is mediated by the same two UFM1-specific proteases that mediate the maturation of UFM1 [33]. UFSP1 and UFSP2 recognize and cleave the isopeptide bond between UFM1 and the target protein, effectively releasing UFM1 and reverting the target protein back to its unmodified state. It is not clear whether UFSP1 and UFSP2 have entirely redundant functions. At least two studies have demonstrated that the deconjugation seems more reliant on UFSP2 compared to UFSP1. Knockdown of UFPS2 but not UFSP1 results in a significant increase in UFM1 conjugates [34, 35]. However, this could be cell-type specific. The dynamic process of conjugation and deconjugation is critical for maintaining cellular homeostasis and ensuring proper regulation of various cellular processes.
The balance between UFM1 conjugation and deconjugation is tightly regulated by multiple factors, including the availability of ATP, the expression levels of UFM1 system components, and the presence of cellular stress signals. Disruptions in this balance can lead to dysregulated protein function and have been implicated in various pathophysiological conditions [20,21,22, 25, 32] (Fig. 1). A better understanding of the molecular mechanisms governing the UFM1 conjugation and deconjugation processes, as well as the identification of novel target proteins and regulatory pathways, will be crucial for elucidating the biological functions of the UFM1 cascade in the brain under physiological and pathological conditions.
The UFM1 cascade regulates neurodevelopment
UFMylation pathway genes and proteins are widely expressed across various human tissues, as indicated by The Human Protein Atlas (https://www.proteinatlas.org/). This is consistent with a significant role of the UFMylation pathway for the development of mice, as several systems including the hematopoietic, liver, intestinal, skeletal, cardiac, renal, immune, and central nervous systems, have been shown to be affected upon knockout (KO) of UFM1 pathway components [25, 27, 41,42,43,44,45,46]. In humans, several genetic variants in the genes encoding the core components of the UFMylation pathway have been associated with severe neurological and bone developmental disorders [23, 25] (Table 1), indicating that the UFM1 cascade is especially important for these processes. However, it remains unclear in which cell types and brain regions the UFM1 protein is expressed and how this is regulated.
A three base pair homozygous deletion in the promoter region of the UFM1 gene has been identified in patients exhibiting hypomyelination and atrophy of the basal ganglia and cerebellum. It was shown that this mutation results in reduced UFM1 expression in neuroblastoma (SH-SY5Y) and astroglioma (U371) cell lines, but not in other tested cell lines (HeLa (cervical adenocarcinoma) or HOG-F2 (oligodendroglioma)) [47]. Moreover, a homozygous missense mutation in UFM1 (UFM1 p.R81C) has been observed in patients suffering from severe early-onset encephalopathy accompanied by progressive microcephaly. This specific missense mutation significantly hinders the formation of both UFM1-UBA5 and UFM1-UFC1 intermediates, leading to impaired UFMylation of cellular proteins [26]. Furthermore, eighteen UBA5 variants have been detected in patients with neurodevelopmental disorders such as hypomyelination with atrophy, early-onset encephalopathy, early myoclonic epilepsy and severe infantile-onset encephalopathy [27, 48,49,50,51,52,53]. Most UBA5 variants result in a loss of function by reducing UBA5 expression levels, decreasing protein half-life, attenuating UFM1 activation, or impairing the transfer of activated UFM1 to UFC1. Notably, the UBA5 (Arg246-to-Ter (R246X)) mutation exhibits increased nuclear localization; however, it also demonstrates loss of function due to reduced protein half-life and diminished UFM1 activation (Table 1). In addition, two UFC1 mutations have been found in patients with severe early-onset encephalopathy and progressive microcephaly [26]. These UFC1 mutations adversely affect the formation of UFM1-UFC1 intermediates, resulting in a widespread reduction in protein UFMylation. UFSP2 mutations have also been discovered in patients presenting with cerebral visual impairment [54]. In a recent study, a UFSP2 variant has been reported as the causative mutation for an autosomal recessive form of pediatric neurodevelopmental anomalies and epilepsy [28]. This UFSP2 mutation reduced the abundance of UFPS2 which was concomitant with an increased abundance of UFMylated targets, indicating the variant may impair de-UFMylation rather than UFMylation. The potential connection between neurodevelopmental and neurodegenerative diseases has garnered significant attention in recent research [55,56,57,58,59]. However, while the essential role of UFMylation for neurodevelopment disorders is evident, its role for degenerative diseases, such as AD, is just emerging.
UFM1 is a novel regulator of Tau aggregation and emerging AD modifier
Recently, the UFMylation cascade was identified as a novel key modifier of tau aggregation. Samelson et al.. conducted a genome-wide CRISPRi-based modifier screen in induced pluripotent stem cell (iPSC)-derived neurons to systematically identify cellular factors involved in the accumulation of tau aggregates in human neurons [60]. They discovered that repression of genes essential for UFMylation, such as UFM1, UFL1, and DDRGK1, led to a reduction in tau oligomer levels. Based on this, Parra Bravo et al. tested a subset of genes using a newly developed iPSC-derived 4R tauopathy model. This confirmed the UFMylation cascade as a major positive regulator of tau propagation. The authors further demonstrated that inhibition of the UFMylation pathway by knocking down the E1 enzyme UBA5 reduced tau spreading in the PS19 tau mouse model [61]. Together this data suggests that the UFM1 cascade is a powerful regulator of tau aggregation and spreading in vitro and in vivo. The mechanism of this regulation remains unknown. It had been speculated that the reduction of UFMylation might involve the reduction of endogenous tau protein [61], although the effect size on endogenous tau protein levels was much smaller compared to the effect on tau aggregation [60].
To investigate the role of the UFMylation pathway for human disease, we meta-analyzed a single-cell nuclear transcriptomic dataset of normal and AD brains [62, 63]. We assessed the expression of UFMylation pathway components across six primary cell types and found specific changes in excitatory neurons: five out of eight UFMylation pathway components (UFSP1, UFSP2, UFC1, UFL1, and DDRGK1) showed significant reduction in AD compared to controls [63]. Given the vulnerability of excitatory neurons to neurodegeneration, these results suggest that disruptions in the UFMylation pathway may play a role in the pathogenesis of AD. In fact, using a biochemical analysis of frozen brain tissue, we further discovered profound changes in UFSP2 and UFM1 protein levels in the frontal and temporal AD cortex of individuals with AD [63]. We found that conjugated UFM1 levels were increased in AD, while soluble UFSP2 levels were decreased in these affected regions but were not changed in the unaffected cerebellum. Importantly, in human brain UFM1 levels positively correlated with pathological tau levels (including insoluble total tau, soluble phosphorylated tau (p-tau), and insoluble p-tau) but not with total tau in the temporal and frontal cortex [63], supporting a role of UFM1 as tau aggregation modifier in AD.
Based on these findings, the UFM1 cascade seems an attractive pathway to target tau aggregation. However, given the unknown mechanism of how UFM regulates tau and which substrates it is conjugated to in AD brain, more work is needed validate the UFM1 pathway as a potential therapeutic avenue. Moreover, given the broad roles of UFM1 in a variety of stress-related pathways, abnormal UFMylation might have far-reaching and unwanted consequences. As detailed below there are a multitude of pathways known to be influenced by UFM1 that play a role for AD pathogenesis. It will be important to validate these as potential candidate pathways in order to further develop UFMylation modifying treatments into a viable therapeutic strategy.
Functional pathways that connect UFMylation and AD
The DNA damage response (DDR)
Overview and role in AD
DNA damage, an alteration in the chemical structure of DNA, is a key feature in post-mitotic neurons, which rely heavily on robust repair mechanisms to maintain genomic stability [64]. Neuronal DNA undergoes continuous, non-random breakage and repair. Recent studies mapped sites of DNA repair synthesis by sequencing in post-mitotic neurons have identified repair hotspots across the genome [65, 66]. These hotspots are enriched with histone H2A isoforms, RNA-binding proteins, and evolutionarily conserved elements of the human genome [66]. Notably, they are predominantly linked to single-strand breaks at CpG-methylated neuronal enhancers [65]. Among various types of DNA damage, double-strand breaks (DSBs) are most lethal. DSBs are primarily repaired through homologous recombination (HR) and non-homologous end-joining (NHEJ) [67, 68]. HR, an error-free pathway active in the post-S phase, uses sister chromatids and proteins like CtIP and RAD51, with DNA synthesis by polymerases. In contrast, NHEJ is error-prone but operates throughout the cell cycle, utilizing KU70/80, DNA-PK, and ligation by LIG4, XRCC4, and XLF. In neurons, NHEJ is the dominant repair pathway due to their post-mitotic state, but HR also plays a significant role [69]. The MRN complex (MRE11, RAD50, and NBS1) is crucial for HR, recruiting repair proteins and being activated by ATM kinase, while also supporting NHEJ [70,71,72,73]. The choice between HR and NHEJ depends on factors like DNA damage type, cell cycle phase, and competitive binding at the DSB site by BRCA1 (favoring HR) and 53BP1 (favoring NHEJ) [74, 75].
DNA damage and impaired DNA repair are key characteristics of aging and various neurodegenerative disorders, including AD [76,77,78,79,80,81,82]. Studies on postmortem human brain samples have demonstrated a significant increase in γH2AX, a widely accepted marker of DSBs [83], in neurons and astrocytes of AD brain, indicating elevated DNA damage in these cells [7, 80, 84]. Additionally, high γH2AX signals have been detected in oligodendrocytes within the white matter of AD brains [6, 85]. Although these γH2AX signals were not significantly different between controls and AD, they were markedly elevated in other dementias [85], and oxidative DNA damage in oligodendrocytes has been identified as a characteristic feature of white matter lesion pathogenesis in the aging human brain [86]. The accumulation of DSBs in AD is largely attributed to increased oxidative stress, driven by the pathogenesis of Aβ and tau [87,88,89,90]. In line with the accumulation of γH2AX signal, increased levels of the other DNA damage sensors such as 53BP1, and phospho-Ser1981 ATM (p-ATM) have also been reported [6, 79, 80, 91], although total levels of ATM and MRN proteins seem reduced in AD cortical neurons, which could further contribute to the accumulation of DNA damage [92, 93]. Studies with both human AD samples and animal models further indicate that the NHEJ DNA repair pathway is compromised in AD [4, 94]. The compromised ability to repair DSBs in AD has been suggested to cause genomic instability, tau hyperphosphorylation, and apoptosis, all of which exacerbate neurodegeneration [6, 76, 89, 95].
The role of ufmylation and connections with AD
UFMylation plays a crucial role in the DDR. Loss of UFL1 increases DDR in the absence of exogenous stress, leads to p53 activation, and enhanced cell death [46]. The formation of the MRN complex, its immediate recruitment to DNA damage sites, and proper ATM activation for initiating DNA repair all rely on UFL1-mediated UFMylation of MRE11 on lysine 282 [96]. ATM phosphorylates UFL1 before the activation and recruitment of the MRN complex to DSBs, thereby enhancing the ligase activity of UFL1 and leading to the UFMylation of histone H4 in a Tip60-dependent manner. This creates a positive feedback loop for ATM activation [97]. This process is counteracted by ATM induced de-UFMylation. Upon ATM-mediated phosphorylation, UFSP2 relocates from the MRN complex to double-strand breaks, where it de-UFMylates H4 [98]. Interestingly, our work showed that knocking out UFSP2 enhanced survival against DNA damage in a UFM1-dependent mechanism in neural progenitors that underwent neuronal differentiation, but not in naive progenitors [63]. The reason for this difference remains unclear and needs to be explored further. The UFMylation cascade has also been implicated in the stabilization of the replication fork. In BRCA1/2-deficient cells, UFL1 localizes to stalled replication forks and facilitates the UFMylation of PTIP, a component of the MLL3/4 methyltransferase complex [99]. This modification promotes the assembly of the PTIP-MLL3/4 complex, leading to increased H3K4me1 and H3K4me3 levels at stalled replication forks and the subsequent recruitment of the MRE11 nuclease.
In addition to MRE11, two other of the five reported UFM1-modified DDR substrates, p53 and poly (ADP-ribose) polymerase-1 (PARP1), have been linked to AD. UFMylation of the tumor suppressor p53, particularly at four lysine residues in its C-terminal domain, is crucial for its stabilization and enhanced transcriptional activity. This promotes genomic integrity and suppresses tumorigenesis. UFMylation of p53 impedes MDM2 ligase recruitment, reducing ubiquitylation and subsequent degradation of p53 [100]. PARP1, a key sensor of DNA damage and replication stress [101,102,103] known to play a critical role in genome integrity is also modified by UFM1 [104]. An in vitro study showed that PARP1 is UFMylated at K548, which enhances its catalytic activity. PARP UFMylation also supports CHK1 activation, which stabilizes replication forks under conditions of stress, thereby contributing to the preservation of genomic stability [104].
Both cell-based and animal studies suggest a protective role of normal tau protein in the repair of DSBs [6, 105,106,107]. Knockdown of endogenous tau in neurons leads to elevated levels of DNA damage, indicated by increased γH2AX [6]. Furthermore, neurons from tau KO mice show higher levels of DNA damage compared to WT neurons [107]. Tau translocates to the nucleus under heat or oxidative stress [106] and upon treatment with DNA damage-inducing agents such as etoposide [6]. The phosphorylation of tau that has been observed under such conditions might be facilitated by checkpoint kinases that are known to regulate DNA replication and the cell cycle in response to DNA damage [108]. However, pathological tau disrupts the neuronal DNA repair system, leading to the accumulation of DNA damage [88, 109]. In addition to exacerbating oxidative stress, abnormal tau interacts with and sequesters critical DNA repair proteins, hindering their ability to function effectively at sites of DNA damage [88, 110, 111]. In AD and other tauopathies, hyperphosphorylated and aggregated tau interferes with nuclear transport, mislocalizing critical repair proteins such as 53BP1 and BRCA1 [110]. This disruption may stem from tau’s interaction with nucleoporins like Nup98, which compromises nucleus-cytoplasm trafficking and contributes to tauopathy [111]. Moreover, tau oligomers bind p53 in the cytoplasm, impairing its nuclear relocation and DDR, ultimately leading to neuronal death [88]. Pathological tau also disrupts microtubule organization, further hindering the trafficking of DNA repair proteins [6]. These observations suggest a role of tau in DDR pathways and genomic stability. However, it is unclear whether this function of tau plays a major role in the pathogenesis of AD. Nevertheless, UFMylation induced tau-dependent or -independent effects on the DNA damage response might contribute to the genomic instability associated with AD.
Given the higher levels of conjugated UFM1 in AD brains [63], it is possible several substrates of UFM1 involved in the DDR are hyperUFMylated. Higher UFMylation of MRE11 might improve DNA damage repair while hyperUFMylation of p53 might contribute to the p53 accumulation that is observed in AD brain [112], which is further associated with p53 aggregation and excessive DNA damage [88]. Moreover, hyperUFMylation of PARP1 could enhance its activity, potentially contributing to the accumulation of ADP-ribose polymers and NAD + depletion observed in human AD brains [113, 114]. Additionally, the interplay between UFMylation and other cellular processes implicated in AD, such as autophagy and ER stress, may further impact the DDR. Further research is needed to elucidate the precise molecular mechanisms underlying the potential link between AD and UFMylation in the context of DDR and determine the therapeutic implications of targeting UFMylation to improve DDR in AD.
ER stress response
Overview and role in AD
The ER is a critical cellular organelle involved in protein synthesis. When the protein-folding capacity of the ER is overwhelmed, the accumulation of misfolded or unfolded proteins leads to a condition known as ER stress. This triggers the activation of the unfolded protein response (UPR), a signaling pathway that increases the production of ER-resident chaperones and foldases to assist with protein folding [115, 116]. The UPR also reduces the synthesis of new and promotes the degradation of misfolded proteins. There are three main ER transmembrane sensors: inositol-requiring enzyme 1 (IRE1), protein kinase RNA-like ER kinase (PERK), and activating transcription factor 6 (ATF6), which are kept in an inactive state by binding to the ER chaperone glucose-regulated protein 78 (GRP78), also known as BiP [117]. Accumulation of misfolded proteins leads to a lower availability of GRP78 to bind and inhibit ER transmembrane sensors, leading to their activation.
ER stress and the UPR play a significant role in AD [8, 118,119,120,121,122]. The accumulation of misfolded tau and Aβ causes ER stress and leads to the activation of the UPR [9,10,11]. Chronic activation of the UPR, however, can result in sustained ER stress, which has been associated with the development and progression of AD [9, 118, 123]. It is interesting to note that IRE1 phosphorylation, triggered by ER stress, correlates well with Braak tau stages, which classify the progression of tau pathology in the brain [123]. Conditional deletion of the RNase domain of IRE1 in the nervous system significantly reduced amyloid load and soluble and insoluble Aβ species in an AD mouse model by regulating amyloid precursor protein (APP) degradation at the ER through IRE1/XBP1 signaling [123]. ER stress further accelerates pathological tau phosphorylation via PERK-eIF2α activation in rTg4510 mice and it was shown that treatment with a PERK inhibitor effectively reduces levels of phospho-tau [124]. Moreover, ER stress is connected with several other pathological features of AD, such as synaptic dysfunction and neuroinflammation [11, 125, 126], although the molecular mechanisms remain enigmatic. A deeper understanding of how ER homeostasis and the UPR relate to the pathogenesis of AD will offer insights into the molecular mechanisms of AD and might reveal new therapeutic targets.
The role of ufmylation and connections with AD
The UFM1 cascade plays an essential role in preserving ER balance and is involved in the UPR during ER stress [20,21,22, 25, 32, 127, 128]. Numerous studies have found a direct connection between UFMylation and ER stress, with the UFM1 system being transcriptionally upregulated in various situations such as ischemic heart disease, diabetic mouse macrophages, exocrine pancreas, gut inflammation and kidney atrophy [42, 44, 129,130,131] and upon ER stress induction in cells, potentially by being direct targets of the XBP1 transcription factor [132]. Knockdown of DDRGK1 increased cell death in a beta-cell line upon ER stress [39], increased ER stress and activation of the UPR along with the cell death program in intestinal epithelial cells [42]. Loss of UFL1 was linked to elevated ER stress and UPR induction in bone marrow cells and to kidney atrophy due to dysfunctional ER homeostasis [44]. Mechanistically, it has been shown that the depletion of DDRGK1 inhibits IRE1α-XBP1 signaling and activates the PERK-eIF2α-CHOP apoptotic pathway through targeting the ER stress sensor IRE1α [40], while UFL1 regulates PERK signaling to prevent cardiomyocyte cell death [43].
Prolyl 4-hydroxylase beta (P4HB), a chaperone that aids in correct disulfide bond formation during protein folding, plays a crucial role for ER stress [133]. UFMylation of P4HB at Lys69/114/130 is essential for maintaining its stability and the loss of UFMylation at these sites lead to its degradation via the ubiquitin-proteasome pathway, resulting in increased oxidative and ER stress [134]. The UFMylation of HRD1, an ER-associated protein degradation ubiquitin ligase, at Lys610, causes activation and leads to enhanced degradation of misfolded proteins, thereby maintaining ER homeostasis [135]. It has been suggested that HRD1 plays a key role in AD by targeting misfolded proteins for degradation [136]. HRD1 deficiency leads to the accumulation of APP, which is cleaved into Aβ [137]. Additionally, it has been shown that HRD1 targets tau and abnormal p-tau for proteasomal degradation [138]. Significantly reduced HRD1 protein levels have been identified in the cerebral cortex of AD patients [137], contributing to impaired protein degradation and the progression of AD pathology.
The association between the UFM1 system and ER stress is widespread and consistent phenomenon, with UFM1 modification potentially shielding cells from ER stress-induced apoptosis by preserving ER homeostasis. However, in our hands, KO of UFSP2 also caused the induction of the UPR under basal conditions and led to enhanced UPR and neuronal death upon ER stress [63], suggesting that both the reduction as well as the abnormal accumulation of UFMylated proteins can exacerbate the UPR. Whether more conjugated UFM1 as observed in AD also induces the UPR remains unknown. Investigating the molecular connections between UFMylation, ER homeostasis, and UPR in AD will help understand how the UPR and ER stress contribute to the pathogenesis of AD and might unveil potential therapeutic strategies to counteract the harmful effects of ER stress and UPR activation in the disease.
Autophagy
Overview and role in AD
Autophagy serves as a crucial cellular process that maintains metabolic homeostasis by breaking down and recycling cellular components [139,140,141]. The mammalian target of rapamycin complex 1 (mTORC1) holds a pivotal role in the biogenesis of autophagy, as it regulates the process in response to nutritional conditions [142,143,144]. Neurons exhibit a high degree of polarization, with autophagosomes predominantly forming at axonal tips and subsequently maturing during retrograde transport to the cell soma [145,146,147]. The biogenesis of autophagosomes entails the recruitment of Atg13 and Atg5 to Double-FYVE Containing Protein 1 (DFCP-1), an ER structure enriched in Phosphatidylinositol 3-Phosphate (PI3P). Following this step, lipidated LC3 is incorporated into the developing autophagosomes [148]. Notably, autophagy induction occurs more efficiently in younger neurons as the expression of several essential autophagy genes declines with age [14, 146, 149, 150]. Reduction of autophagy has been suggested to contribute to various age-related neurodegenerative diseases, including AD, highlighting the importance of understanding the intricate mechanisms of autophagy in neuronal cells.
Dysregulation of autophagy in AD is evident in both animal models and human patients [14, 146, 151,152,153,154,155]. Dystrophic neurites, abnormal neuronal processes that are a common feature in AD brain, show a large number of immature autophagic vacuoles, suggesting widespread deficiency of autophagy in AD brain [12, 154, 156, 157]. Moreover, several autophagy-related proteins were found downregulated during the progression of AD [155, 158, 159]. Autophagy malfunction has been linked to Apolipoprotein E4 as well as APP, presenilin-1 (PS-1), and presenilin-2 (PS-2) and therefore seems to play a role for sporadic and familial forms of AD [12, 160,161,162,163,164]. However, the mechanism(s) of how these genes affect autophagy remains largely elusive. Biochemical analyses reveal a dramatic increase in Atg8/LC3 in postmortem AD brains, with co-localization to hyperphosphorylated tau, and a significant elevation in APP/PS1 mice [154, 165]. It is noteworthy that the central elements of AD pathology, Aβ and tau, both have a complex relationship with autophagy [15, 166, 167]. In the early stages of AD, the accumulation of Aβ activates autophagy as a compensatory mechanism to promote their clearance [168,169,170,171]. Autophagy also regulates APP metabolism by inhibiting its proteolytic processing and enhancing its degradation through Atg5-dependent pathways, thereby reducing Aβ production [172]. Additionally, autophagy plays a critical role in the secretion of both Aβ and tau. For instance, autophagy deficiency caused by Atg7 knockdown impairs the secretion of Aβ into the extracellular space [173], while disrupted autophagic flux, induced by p300/CBP hyperactivation or bafilomycin A1 treatment, has been shown to increase tau secretion in neurons [174]. As AD progresses, the continuous production and accumulation of pathological Aβ and tau lead to significant disruption of the autophagy–lysosomal pathway [173, 175,176,177,178]. This dysfunction causes further aggregation of Aβ and tau, which in turn exacerbates the impairment of the autophagy–lysosomal system. This vicious cycle ultimately drives the formation of Aβ plaques and neurofibrillary tangles, contributing to the neurodegeneration observed in AD. However, enhancing autophagy has been shown to reduce Aβ levels and facilitate the degradation of pathological tau [167, 178,179,180], highlighting its potential as a therapeutic target for AD.
The role of UFMylation and connections with AD
UFMylation has been identified as a novel regulator of SQSTM1/p62, an essential protein in autophagy that is decorated by ubiquitin as well as several other UBLs [181]. However, the effect of UFM1 on p62 seemed to occur indirectly, through an elevated cell type-specific ER stress response, which led to p62 accumulation in the cytosol and was not observed in all investigated cell types. It is interesting that p62 has been implicated in the degradation of tau protein [182, 183], but it remains unknown whether this or other mechanisms are associated with the regulation of tau by the UFM1 cascade. In bone marrow cells, upregulation of p62 was also observed upon loss of UFL1 but here, loss of UFMylation also caused an increase of LC3-II and blocked autophagic degradation [46]. In line with a role for autophagic degradation, Atg9, which plays a role in autophagosome expansion, was identified as a conserved target of UFM1. The UFMylation of Atg9 is essential for maintaining Atg9A protein levels to coordinate autophagy [184]. In both 5XFAD and APP/PS1 mouse models, Atg9A accumulates extensively in dystrophic neurites surrounding amyloid plaques [185], but it is unclear whether this could be related to hyperUFMylation. In addition to degradation, a role of UFMylation for autophagy initiation has been suggested via UFMylation of the VCP/p97 protein at lysine 109 (K109). This was reported to enhance the stabilization of BECN1 and the assembly of the PtdIns3K complex [186]. VCP has been shown to mediate disaggregation of ubiquitylated tau fibrils [187].
UFMylation further plays crucial role in the selective autophagy process known as ER-phagy [184, 188, 189]. This process is essential for maintaining ER homeostasis under various stress conditions, cellular development, and environmental challenges [190,191,192,193,194]. It was shown that UFM1 conjugates to specific ER proteins via DDRGK1 and the UFL1 ligase [188]. This facilitates the degradation of ER sheets similar to PINK1-PRKN induced mitophagy [195]. UFL1 is recruited to the ER by DDRGK1 and there conjugates UFM1 to the substrate ribophorin I (RPN1) and ribosomal protein L26 (RPL26), which facilitates ER-phagy [188]. Interestingly, RPN1 is elevated in brain capillaries of AD patients [196]. Furthermore, RPL26 immunoreactivity is reduced in AD neurons, and it has been identified as a potential biomarker for AD pathogenesis [197, 198]. UFMylation of NADH-cytochrome b5 reductase 3 (CYB5R3) inactivates this protein and also induces ER-phagy [189]. The degradation of UFMylated CYB5R3 in lysosomes depends on the autophagy-related protein Atg7 and the autophagy-adaptor protein CDK5RAP3, which possesses non-canonical shuffled Atg8 interacting motifs essential for Atg8 interaction and autophagy initiation [199]. A crucial role of ER-phagy for AD is not established. However, it has been found CYB5R3 levels are reduced in the cerebrospinal fluid of 5XFAD mice [200]. Together with the above-mentioned changes in RPN1 and RPL26, this suggests that ER-phagy might be dysregulated in AD. In line with this, a recent study showed impaired ER-phagy in a Drosophila model expressing human AAP. In this model, moderate upregulation of ER-phagy in the brain was sufficient to drive APP degradation and to significantly mitigate the associated phenotypes [201]. It is interesting that mutations in the RTN3 gene, an ER-phagy receptor, have been detected in several early and late onset AD cases [202]. While the pathogenicity remains to be determined, RTN3 appears to play a role in regulating the production of Aβ by inhibiting BACE1 activity to produce amyloid beta-protein [203, 204]. These findings highlight the potential of targeting ER-phagy as a therapeutic strategy for treating diseases linked to ER protein aggregation.
In summary, UFMylation plays a multifaceted role in autophagy regulation, specifically ER-phagy, by modulating key proteins and pathways essential for maintaining cellular homeostasis, development, and stress response. While dysfunction of autophagy has been widely reported in AD, it is noteworthy that all identified autophagy-related UFM1 substrates—VCP, RPN1, CYB5R3, Atg8/LC3, Atg9A, and RPL26—have demonstrated links to AD. These findings highlight the significance of UFMylation in regulating autophagy across diverse biological contexts and organisms. Investigating the precise molecular mechanisms through which UFMylation might influence autophagy dysfunction in AD could offer insights into the disease’s pathogenesis and highlight novel therapeutic targets.
Immune response
Overview and role in AD
An immune response is fundamentally defined as the body’s reaction to foreign substances [205]. Within the central nervous system, this immune response is termed neuroinflammation, reflecting a mechanism vital for maintaining homeostasis and guarding against pathogenic insults [16]. Neuroinflammation can be triggered by a variety of pathological conditions, including infections, traumatic injuries, ischemic events, or exposure to toxins and is mostly mediated by microglia and astrocytes although other cell types also participate depending on the context [16, 19, 206]. The resulting cascade involves the secretion of pro-inflammatory cytokines such as interleukins, tumor necrosis factor (TNF), and chemokines.
Abnormal neuroinflammation is one of the major hallmarks of AD and linked to disease pathogenesis. Chronic activation of microglia and astrocytes fosters a persistent inflammatory state marked by excessive production of cytokines, synaptic pruning, and neuronal damage [16, 207,208,209,210]. This maladaptive immune response is further exacerbated by dysregulated transcription factors such as toll-like receptor 4 (TLR4)/nuclear factor kappa B (NF-κB), which amplify pro-inflammatory signaling pathways, and the activation of inflammasomes, including NLRP3, which drive excessive inflammatory cytokine release [211,212,213,214]. Immune checkpoints, such as Programmed death receptor 1/programmed death receptor-ligand 1 (PD-1/PD-L1), which normally maintain immune balance by modulating microglial activation and limiting excessive inflammation, also become dysfunctional in AD, further perpetuating neuroinflammation [215, 216]. Additionally, viral infections, including those caused by herpes simplex virus (HSV-1), Hepatitis C Virus (HCV) and Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), have been linked to the activation of innate immune responses, contributing to the inflammatory burden in AD [217,218,219]. Together, these elements contribute to a self-perpetuating cycle of neuroinflammation and neurodegeneration in AD, underscoring the potential therapeutic value of targeting immune pathways. Given the strong neuroinflammatory phenotypes in AD, it is not surprising that many AD-associated risk genes identified through genome-wide association studies, e.g. CD33, INPP5D, CLU, CR1, SPI1, ABCA7, EPHA1, the MS4A gene cluster, and TREM2 have been implicated in the adaptive and innate immune systems [220,221,222,223].
The role of UFMylation and connections with AD
Neuron-specific deletion of UFL1 or DDRGK1 both lead to elevated inflammatory responses in mouse brain [224], indicating that the UFM1 cascade is very important for the immune response in the nervous system. Consistently, recent studies have demonstrated the involvement of UFMylation in several aspects of the immune response, including the regulation of transcription factors, inflammasome, immune checkpoints and virus infections [21, 22, 25, 127, 225].
One critical immune regulator that is regulated by UFMylation is the transcription factor NF-κB [226,227,228], which is known to induce pro-inflammatory genes in both innate and adaptive immune cells [229,230,231]. While the role of UFM1 in the context of NF-κB for neurons or in brain remains unclear, data from other cell types indicate complex and cell-type specific functions of UFM1. In human umbilical vein endothelial cells, UFM1 inhibits lipopolysaccharide (LPS) induced inflammation via NF-κB [232]. In goat endometrial epithelial cells, overexpression of UFM1 suppresses LPS-induced activation of the TLR4/NF-κB pathway [233]. Similarly, in human osteoarthritis chondrocytes, UFL1 has been shown to inhibit NF-κB activation triggered by IL-1β [234]. UFL1 was found to modulate NLRP3 inflammasome activation and protect against pyroptosis in LPS-stimulated bovine mammary epithelial cells. This protective effect was partly mediated through the inhibition of NF-κB signaling [235]. However, in the context of diabetes, UFMylation has been linked to enhanced inflammatory responses. UFM1 expression is elevated in macrophages from db/db (diabetic) mice and in LPS-stimulated macrophages, leading to increased pro-inflammatory cytokine production (e.g., TNF-α, IL-1β, IL-6) via activation of the NF-κB pathway through reduced LZAP expression [227]. Additionally, UFM1 promoted NF-κB p65 nuclear translocation in LPS-treated RAW264.7 cells and peritoneal macrophages in db/db mice by increasing the ubiquitination and degradation of IκBα [226]. In summary, the UFMylation pathway critically regulates the TLR4/NF-κB pathway, exerting a context- and cell-type-specific influence on inflammation by modulating NF-κB activation and pro-inflammatory cytokine production. However, its role in regulating the TLR4/NF-κB pathway during neuroinflammation remains unclear. In AD, activation of NF-κB is correlated with the production of pro-inflammatory cytokines, neuroinflammation, augmented Aβ production, and tau hyperphosphorylation, and neuronal death, which constitute key pathological hallmarks of the disease [17, 18, 212, 213]. A meta-analysis further identified NF-κB signaling as one of the most significantly disrupted pathways in late-onset AD brains [236]. In line with a role for pathogenesis, targeted inhibition of microglial NF-κB activation provided protection against Aβ toxicity in glial-neuron co-cultures [237]. TLR4 is also implicated in neuroinflammation in AD. Increased TLR4 expression has been observed in the brains of AD patients and familial AD mouse models [238]. In addition, single nucleotide polymorphisms (SNPs) in TLR4 have been associated with variations in AD risk [239, 240]. Hyperactivation of TLR4 has been shown to contribute to neuroinflammation in AD, with evidence indicating that Aβ can bind to TLR4, leading to immune activation and subsequent neuroinflammatory responses [238, 241,242,243]. Given the central role of the TLR4/NF-κB pathway in AD progression and the strong link between UFMylation and this signaling axis, targeting UFMylation alongside TLR4 and NF-κB signaling pathways may present novel therapeutic strategies to mitigate neuroinflammation and its detrimental effects in AD.
PD-1 and PD-L1 are key immune checkpoint molecules that regulate immune responses and are involved in the immune evasion of tumors [244, 245]. Both proteins can be UFMylated, but this modification has opposing effects depending on the context. In liver tumors, the UFMylation of PD-L1 facilitates its proteasome-mediated degradation, thereby suppressing tumor growth [246]. Conversely, UFL1 enhances the UFMylation of PD-1, stabilizing PD-1 and hindering the activation of CD8 + T cells, which promotes immune evasion [247]. In triple-negative breast cancer, UFM1 upregulates PD-L1 expression indirectly by stabilizing PLAC8, a protein highly expressed in this cancer type. This stabilization enhances PD-L1 expression through the modulation of its ubiquitination, contributing to tumor proliferation and immune modulation [248]. While a role of UFM1 for PD-L1 in the nervous system remains unknown, emerging evidence suggests that the PD-L1/PD-1 immune checkpoint pathway plays a critical role in modulating neuroinflammation, microglial function, amyloid-β clearance and p-tau clearance in AD [215, 216, 249]. Michal Schwartz’s team demonstrated that targeting PD-1 or its ligand PD-L1 with systemic injections of blocking antibodies could alter AD pathology in multiple mouse models. These include models of amyloid pathology (APP/PS1 and 5XFAD) and tau pathology (DM-hTAU, expressing human-tau with frontotemporal dementia mutations) [215, 216]. Blocking PD-1 signaling recruited monocyte-derived macrophages to the brain and facilitated the clearance of Aβ plaques and p-tau while improving cognitive performance. However, in contrast, a study by Kummer et al. found that PD-1 deficiency in APP/PS1 mice increased Aβ plaque accumulation and decreased microglial Aβ uptake, exacerbating neuroinflammation [249]. They also identified increased PD-L1 expression in astrocytes near Aβ plaques and higher PD-1 levels in microglia, suggesting a protective role for PD-1/PD-L1 signaling in AD. These findings highlight the dual role of PD-1/PD-L1 in AD, suggesting systemic PD-1 blockade may offer therapeutic potential, while local PD-1 deficiency might exacerbate neuroinflammation and disease severity. Whether changes in UFMylation alleviate or exacerbate microglial responses, Aβ clearance, and tau pathology by regulating PD-1/PD-L1 remains to be further explored.
Recent research has highlighted a crucial role of the UFM1 cascade for antiviral innate immunity [250, 251]. In the context of DNA virus infection, such as with HSV-1 or vaccinia virus (VACV), UFL1 protein levels are significantly reduced in peritoneal macrophages, suggesting involvement in promoting antiviral immunity. UFL1 was shown to regulate the cGAS-STING pathway by stabilizing STING protein, through competitive binding that prevents Lys48-linked ubiquitination, thereby enhancing antiviral responses [250], in a UFM1-independent fashion. It is interesting to note that HSV-1 has been associated with an increased risk of AD, especially in individuals carrying the APOE ɛ4 allele [218, 219] and it was shown that HSV-1 infection leads to the accumulation of Aβ and p-tau in cell cultures and 3D human brain models [252,253,254]. In RNA virus infections, UFL1 plays a critical role by dynamically relocating to mitochondrial-associated ER membranes, where it UFMylates mitochondrial antiviral signaling protein (MAVS), promoting its dislocation, incorporation into mitochondrial-derived vesicles (MDVs), and lysosomal degradation [255, 256]. This process is essential for RIG-I signaling, a key RNA virus sensor [251]. UFL1 also interacts with 14-3-3ε and RIG-I, leading to 14-3-3ε UFMylation, enhanced RIG-I activation, MAVS signaling, and interferon (IFN) production [251]. Additionally, UFL1-mediated UFMylation of RPL26 influences hepatitis A virus (HAV) RNA translation and replication [257]. While HAV infection has not been associated with AD, other RNA viruses, such as HCV and SARS-CoV-2, have been linked to AD [217, 225, 258, 259]. Furthermore, it has been shown that the level of the UFMylation substrate 14-3-3ε is significantly reduced in the frontal cortex of postmortem AD patients [260]. Investigating whether UFMylation plays a role in AD-related antiviral immunity could provide valuable insights. Further investigation is warranted to explore whether modulating UFMylation could offer a therapeutic avenue for reducing the risk of AD linked to viral infections.
The role of UFMylation for immune regulation is very complex and dynamic and the current body of evidence suggests that the UFM1 cascade is neither universally suppressive or promotive but rather tailored to the specific physiological or pathological context. Future studies are needed to elucidate the cell-type- and context-specific roles of UFMylation specifically for the nervous system and in the context of AD.
Other pathways
In addition to the pathways previously mentioned, several studies have suggested that the UFMylation pathway may also be involved in other AD-related pathways, including:
Mitochondrial function
Mitochondrial impairment is a key feature of AD [261,262,263] and there are some links between the UFMylation pathway and mitochondrial function as well. Impairment in the UFMylation process leads to decreased mitochondrial density in aged brains via Atg9, mirroring the previously reported mitochondrial abnormalities observed in the indirect flight muscles of flies with Atg9 knockdown [184]. In addition, P4HB, the target for UFMylation that triggers ER stress also has mitochondrial effects. Ineffective UFMylation of P4HB leads to mitochondrial dysfunction and heightened oxidative stress [134]. Thus, UFMylation pathway may be linked to AD through its involvement in mitochondria function regulation.
Ribosome-associated protein quality control
Ribosome dysfunction is an early event in AD [264]. This impairment in ribosome function leads to reduced protein synthesis rate and capacity, a decline in ribosomal RNA and tRNA levels, and an increase in RNA oxidation [265, 266]. Certain ribosome proteins, such as uS3, uS10, and RPL26, have been recognized as targets of UFMylation [267]. Specifically, UFMylation of RPL26 increases during ribosome stalling, signaling translation errors and activating the ribosome-associated quality control mechanism during protein synthesis [267, 268]. Therefore, the UFMylation pathway may be connected to AD through its role in ribosome quality control.
Ferroptosis
Ferroptosis is an iron-dependent lipid peroxidation-driven programmed cell death and recent research has indicated a significant association between ferroptosis and AD [269,270,271]. However, the precise underlying mechanism remains elusive. SLC7A11, a key regulator of ferroptosis, is stabilized by UFM1 modification [272]. Knockdown of UFM1 reduces SLC7A11 expression by decreasing its protein stability without affecting transcription. Metformin induces ferroptosis by downregulating SLC7A11 protein stability through inhibition of its UFMylation [272]. Thus, the UFMylation pathway may be involved in the dysregulation of ferroptosis in AD.
Conclusions and future perspectives
In this review, we have explored the many connections between AD and UFMylation, a critical but understudied ubiquitin-like post-translational modification. Given these many links, it is conceivable that the dysregulation of UFMylation could contribute to risk, onset or progression of AD and offer new therapeutic possibilities for intervention. A deeper understanding of the interplay between AD and UFMylation is needed to understand the multifaceted cellular processes and potential therapeutic outcomes. However, it is also important to note that dysfunctions in DDR, ER homeostasis, autophagy and immune responses are not exclusive to AD but are also prevalent in various other neurodegenerative disorders. These include Parkinson’s disease, Frontotemporal dementia, Amyotrophic Lateral Sclerosis, progressive multiple sclerosis and Huntington’s disease [119, 273,274,275,276,277,278]. This commonality suggests that deregulation of the UFMylation pathway could be a contributing factor in the pathogenesis of several of these neurodegenerative conditions.
One major shortfall in order to understand the role of UFM1 for neurological disease is that UFM1 function has not been sufficiently studied in brain. While there is a clear genetic link between UFMylation and neurodevelopmental disease, the majority of UFMylation research up to this point has focused mostly on non-neuronal cells. Neuronal effects remain understudied and nervous system-specific targets of UFM1 have not been identified yet, Therefore, it will be important to shift the focus and intensify research on brain-centric cells, including neurons, microglia, oligodendrocytes, and astrocytes. Such studies are essential to unravel the physiological and potentially pathological roles of UFMylation in the nervous system, which will be pivotal to understand the role of UFM1 plays for neuronal biology and its pathological impact on conditions such as neurodevelopmental and neurodegenerative diseases.
Given that the reduction of UFMylation has been shown to reduce the propagation of tau induced by seeding in vitro and in vivo [61], it is conceivable that UFM1 plays an important role for the pathogenesis of tauopathies. It will be important to shed light onto the mechanism of how UFM1 influences tau. It has recently been shown that the UFM1 cascade also regulates α-synuclein levels [279] and it was shown that mono-UFMylation of α-synuclein modulates its secretion, which then affects the spreading of aggregated synuclein. It remains to be seen whether a similar mechanism could also account for other misfolded proteins, such as tau, as suggested by the authors [280]. More research into the molecular mechanism and exploring the impact of UFMylation on biological and pathological levels of tau (Aβ, and α-synuclein), remains paramount. Understanding how UFMylation modifies these proteins and affects their aggregation, toxicity, clearance, and transmission will provide critical insights into the pathogenesis and progression of neurodegenerative diseases. It is currently unclear whether UFM1 is directly conjugated to tau in neurons or in brain. A thorough characterization of the UFMylation proteome in neuronal cells and human brain specimens is needed to identify specific substrates exhibiting increased UFMylation in AD-affected brains. These substrates may play a critical role in the development or progression of AD - offering a clearer understanding of the disease at a molecular level; might provide new biomarkers for early detection, monitoring, and prognosis; and help to identify tractable therapeutic targets for people AD.
New tools are needed to understand UFMylation on the molecular level. This includes high-quality antibodies, relevant animal models, gene edited human stem cell derived neurons, and pharmacological compounds to modulate this pathway, including antagonists and agonists. Since UFMylation involves a set of UFM1-specific enzymes that catalyze its addition and deconjugation, such compounds should be highly specific for just UFM1 and not affect other UBL modifications such as SUMOylation, NEDDylation or ISG15ylation. These tools will be important to pave the way for a more profound exploration of the UFMylation pathway, its druggability and therapeutic potential. While the molecular intricacies connecting AD, neurodevelopmental disorders, and UFMylation remain to be fully elucidated, we advocate for intensified research endeavors aimed at pinpointing UFMylation targets in these diseases.
In conclusion, while there has been limited exploration into the role of UFMylation in neurodegenerative diseases, the emerging research, especially in the context of AD, is promising. Future in-depth studies in this domain have the potential to not only deepen our understanding but also pave the way for innovative therapeutic strategies for affected individuals. Dissecting the molecular functions of UFMylation especially with regards to AD might offer new and exciting opportunities for disease intervention.
Data availability
No datasets were generated or analysed during the current study.
Abbreviations
- AD:
-
Alzheimer’s disease
- Aβ:
-
Beta-amyloid
- APP:
-
Amyloid precursor protein
- DDR:
-
DNA damage response
- DSBs:
-
Double-strand breaks
- ER:
-
Endoplasmic reticulum
- HAV:
-
Hepatitis a virus
- HR:
-
Homologous recombination
- HSV-1:
-
Herpes simplex virus
- IFN:
-
Interferon
- iPSC:
-
Induced pluripotent stem cell
- KO:
-
Knock out
- LPS:
-
Lipopolysaccharide
- MAVS:
-
Mitochondrial antiviral signaling protein
- MDVs:
-
Mitochondrial-derived vesicles
- NHEJ:
-
Non-homologous end joining
- p-ATM:
-
Phospho-ser1981 ATM
- ROS:
-
Reactive oxygen species
- SARS‑CoV‑2:
-
Severe acute respiratory syndrome coronavirus 2
- SNPs:
-
Single nucleotide polymorphisms
- TNF:
-
Tumor necrosis factor
- UBL:
-
Ubiquitin-like
- UPR:
-
Unfolded protein response
- VACV:
-
Vaccinia virus
References
Scheltens P, et al. Alzheimer’s disease. Lancet. 2021;397(10284):1577–90.
Masters CL, et al. Alzheimer’s disease. Nat Reviews Disease Primers. 2015;1(1):15056.
Serrano-Pozo A, et al. Neuropathological alterations in alzheimer disease. Cold Spring Harbor Perspectives in Medicine; 2011;1(1).
Shackelford DA. DNA end joining activity is reduced in alzheimer’s disease. Neurobiol Aging. 2006;27(4):596–605.
Mullaart E, et al. Increased levels of DNA breaks in cerebral cortex of alzheimer’s disease patients. Neurobiol Aging. 1990;11(3):169–73.
Asada-Utsugi M, et al. Failure of DNA double-strand break repair by Tau mediates alzheimer’s disease pathology in vitro. Commun Biology. 2022;5(1):358.
Thadathil N, et al. DNA double-strand break accumulation in alzheimer’s disease: evidence from experimental models and postmortem human brains. Mol Neurobiol. 2021;58(1):118–31.
Hoozemans JJM, et al. The unfolded protein response is activated in alzheimer’s disease. Acta Neuropathol. 2005;110(2):165–72.
Yoon SO, et al. JNK3 perpetuates metabolic stress induced by Aβ peptides. Neuron. 2012;75(5):824–37.
Abisambra JF, et al. Tau accumulation activates the unfolded protein response by impairing Endoplasmic Reticulum-Associated degradation. J Neurosci. 2013;33(22):9498–507.
Ajoolabady A, et al. ER stress and UPR in alzheimer’s disease: mechanisms, pathogenesis, treatments. Cell Death Dis. 2022;13(8):706.
Nixon RA, et al. Extensive involvement of autophagy in alzheimer disease: an Immuno-Electron microscopy study. J Neuropathology Experimental Neurol. 2005;64(2):113–22.
Yu WH, et al. Macroautophagy—a novel β-amyloid peptide-generating pathway activated in alzheimer’s disease. J Cell Biol. 2005;171(1):87–98.
Lipinski MM et al. Genome-wide analysis reveals mechanisms modulating autophagy in normal brain aging and in Alzheimer’s disease. Proceedings of the National Academy of Sciences, 2010;107(32):14164–14169.
Reddy PH, Oliver DM. Amyloid Beta and phosphorylated Tau-Induced defective autophagy and mitophagy in alzheimer’s disease. Cells. 2019;8(5):488.
Leng F, Edison P. Neuroinflammation and microglial activation in alzheimer disease: where do we go from here? Nat Reviews Neurol. 2021;17(3):157–72.
Terai K, Matsuo A, McGeer PL. Enhancement of immunoreactivity for NF-κB in the hippocampal formation and cerebral cortex of alzheimer’s disease. Brain Res. 1996;735(1):159–68.
Kaltschmidt B, et al. Transcription factor NF-κB is activated in primary neurons by amyloid β peptides and in neurons surrounding early plaques from patients with alzheimer disease. Proc Natl Acad Sci. 1997;94(6):2642–7.
Heneka MT, Kummer MP, Latz E. Innate immune activation in neurodegenerative disease. Nat Rev Immunol. 2014;14(7):463–77.
Gerakis Y, et al. The ufmylation system in proteostasis and beyond. Trends Cell Biol. 2019;29(12):974–86.
Witting KF, Mulder MPC. Highly specialized ubiquitin-like modifications: shedding light into the UFM1 enigma. Biomolecules. 2021;11(2):255.
Millrine D, Peter JJ, Kulathu Y. A guide to ufmylation, an emerging posttranslational modification. FEBS J. 2023;290(21):5040–56.
Zhou X, et al. UFMylation: a ubiquitin-like modification. Trends Biochem Sci. 2023;49(1):52–67.
Komatsu M, et al. A novel protein-conjugating system for Ufm1, a ubiquitin-fold modifier. EMBO J. 2004;23(9):1977–86.
Yang S, Moy N, Yang R. The UFM1 conjugation system in mammalian development. Dev Dyn. 2023;252(7):976–85.
Nahorski MS, et al. Biallelic UFM1 and UFC1 mutations expand the essential role of ufmylation in brain development. Brain. 2018;141(7):1934–45.
Muona M, et al. Biallelic variants in UBA5 link dysfunctional UFM1 ubiquitin-like modifier pathway to severe infantile-onset encephalopathy. Am J Hum Genet. 2016;99(3):683–94.
Ni M, et al. A pathogenic UFSP2 variant in an autosomal recessive form of pediatric neurodevelopmental anomalies and epilepsy. Genet Sci. 2021;23:900–8.
Wang X, Xu X, Wang Z. The post-translational role of ufmylation in physiology and disease. Cells. 2023;12(21):2543.
Sasakawa H, et al. Solution structure and dynamics of Ufm1, a ubiquitin-fold modifier 1. Biochem Biophys Res Commun. 2006;343(1):21–6.
Tatsumi K, et al. A novel type of E3 ligase for the Ufm1 conjugation system. J Biol Chem. 2010;285(8):5417–27.
Banerjee S, Kumar M, Wiener R. Decrypting ufmylation: how proteins are modified with UFM1. Biomolecules. 2020;10(10):1442.
Kang SH, et al. Two novel ubiquitin-fold modifier 1 (Ufm1)-specific proteases, UfSP1 and UfSP2. J Biol Chem. 2007;282(8):5256–62.
Liang Q et al. Human UFSP1 translated from an upstream near-cognate initiation codon functions as an active UFM1-specific protease. Journal of Biological Chemistry, 2022;102016.
Millrine D, et al. Human UFSP1 is an active protease that regulates UFM1 maturation and ufmylation. Cell Rep. 2022;40(5):111168.
Bacik J-P, et al. Crystal structure of the human ubiquitin-activating enzyme 5 (UBA5) bound to ATP: Mechanistic insights into a Minimalistic e1 Enzyme. J Biol Chem. 2010;285(26):20273–80.
Habisov S, et al. Structural and functional analysis of a novel interaction motif within UFM1-activating enzyme 5 (UBA5) required for binding to Ubiquitin-like proteins and ufmylation**. J Biol Chem. 2016;291(17):9025–41.
Mizushima T, et al. Crystal structure of Ufc1, the Ufm1-conjugating enzyme. Biochem Biophys Res Commun. 2007;362(4):1079–84.
Lemaire K, et al. Ubiquitin fold modifier 1 (UFM1) and its target UFBP1 protect pancreatic beta cells from ER stress-induced apoptosis. PLoS ONE. 2011;6(4):e18517.
Liu J, et al. A critical role of DDRGK1 in Endoplasmic reticulum homoeostasis via regulation of IRE1α stability. Nat Commun. 2017;8(1):14186.
Tatsumi K, et al. The Ufm1-activating enzyme Uba5 is indispensable for erythroid differentiation in mice. Nat Commun. 2011;2(1):181.
Cai Y, et al. Indispensable role of the Ubiquitin-fold modifier 1-specific E3 ligase in maintaining intestinal homeostasis and controlling gut inflammation. Cell Discovery. 2019;5(1):7.
Li J, et al. Ufm1-specific ligase Ufl1 regulates Endoplasmic reticulum homeostasis and protects against heart failure. Circulation: Heart Fail. 2018;11(10):e004917.
Zhou Y, et al. Ufl1 deficiency causes kidney atrophy associated with disruption of Endoplasmic reticulum homeostasis. J Genet Genomics. 2021;48(5):403–10.
Egunsola AT, et al. Loss of DDRGK1 modulates SOX9 ubiquitination in spondyloepimetaphyseal dysplasia. J Clin Investig. 2017;127(4):1475–84.
Zhang M, et al. RCAD/Ufl1, a Ufm1 E3 ligase, is essential for hematopoietic stem cell function and murine hematopoiesis. Cell Death Differ. 2015;22(12):1922–34.
Hamilton EMC, et al. UFM1 founder mutation in the Roma population causes recessive variant of H-ABC. Neurology. 2017;89(17):1821–8.
Colin E, et al. Biallelic variants in UBA5 reveal that disruption of the UFM1 cascade can result in early-onset encephalopathy. Am J Hum Genet. 2016;99(3):695–703.
Duan R, et al. UBA5 mutations cause a new form of autosomal recessive cerebellar ataxia. PLoS ONE. 2016;11(2):e0149039.
Mignon-Ravix C, et al. Abnormal function of the UBA5 protein in a case of early developmental and epileptic encephalopathy with suppression-burst. Hum Mutat. 2018;39(7):934–8.
Al-Saady ML, et al. Homozygous UBA5 variant leads to hypomyelination with thalamic involvement and axonal neuropathy. Neuropediatrics. 2021;52(06):489–94.
Briere LC, et al. A description of novel variants and review of phenotypic spectrum in UBA5-related early epileptic encephalopathy. Mol Case Stud. 2021;7(3):a005827.
Cabrera-Serrano M, et al. A homozygous UBA5 pathogenic variant causes a fatal congenital neuropathy. J Med Genet. 2020;57(12):835–42.
Bosch DG, et al. Novel genetic causes for cerebral visual impairment. Eur J Hum Genet. 2016;24(5):660–5.
Lule D, Ludolph AC, Ludolph AG. Neurodevelopmental and neurodegenerative diseases– Is there a pathophysiological link? Attention-deficit/hyperactivity disorder and amyotrophic lateral sclerosis as examples. Med Hypotheses. 2008;70(6):1133–8.
Arendt T, Stieler J, Ueberham U. Is sporadic alzheimer′s disease a developmental disorder? J Neurochem. 2017;143(4):396–408.
Ouellette J, Lacoste B. From neurodevelopmental to neurodegenerative disorders: the vascular continuum. Front Aging Neurosci. 2021;13:749026.
Magnin E, et al. Neurodevelopmental disorders modify neurodegenerative diseases: a medical history to screen in memory clinic. Psychol Neuropsychiatr Vieil. 2021;19(2):181–90.
Magnin E. Neurodevelopmental and neurodegenerative similarities and interactions: A point of view about lifelong neurocognitive trajectories. J Alzheimers Dis. 2021;79:1397–407.
Samelson AJ et al. CRISPR screens in iPSC-derived neurons reveal principles of tau proteostasis. bioRxiv, 2023;2023.06.16.545386.
Parra Bravo C, et al. Human iPSC 4R Tauopathy model uncovers modifiers of Tau propagation. Cell. 2024;187(10):2446–e246422.
Mathys H et al. Single-cell transcriptomic analysis of Alzheimer’s disease. Nature. 2019;570:332–337.
Yan T, et al. The ufmylation pathway is impaired in alzheimer’s disease. Mol Neurodegeneration. 2024;19(1):97.
McKinnon PJ. Genome integrity and disease prevention in the nervous system. Genes Dev. 2017;31(12):1180–94.
Wu W, et al. Neuronal enhancers are hotspots for DNA single-strand break repair. Nature. 2021;593(7859):440–4.
Reid DA, et al. Incorporation of a nucleoside analog maps genome repair sites in postmitotic human neurons. Science. 2021;372(6537):91–4.
Swift ML, et al. DSB repair pathway choice is regulated by recruitment of 53BP1 through cell cycle-dependent regulation of Sp1. Cell Rep. 2021;34(11):108840.
Shrivastav M, De Haro LP, Nickoloff JA. Regulation of DNA double-strand break repair pathway choice. Cell Res. 2008;18(1):134–47.
Merlo D, et al. DNA repair in post-mitotic neurons: a gene-trapping strategy. Cell Death Differ. 2005;12(3):307–9.
Bian L, et al. MRE11-RAD50-NBS1 complex alterations and DNA damage response: implications for cancer treatment. Mol Cancer. 2019;18(1):169.
Reginato G, Cejka P. The MRE11 complex: A versatile toolkit for the repair of broken DNA. DNA Repair. 2020;91–92:102869.
Huang J, Dynan WS. Reconstitution of the mammalian DNA double-strand break end-joining reaction reveals a requirement for an Mre11/Rad50/NBS1-containing fraction. Nucleic Acids Res. 2002;30(3):667–74.
Hopfner K-P, et al. The Rad50 zinc-hook is a structure joining Mre11 complexes in DNA recombination and repair. Nature. 2002;418(6897):562–6.
Shibata A. Regulation of repair pathway choice at two-ended DNA double-strand breaks. Mutat Research/Fundamental Mol Mech Mutagen. 2017;803–805:51–5.
Escribano-Díaz C, et al. A cell Cycle-Dependent regulatory circuit composed of 53BP1-RIF1 and BRCA1-CtIP controls DNA repair pathway choice. Mol Cell. 2013;49(5):872–83.
Welch G, Tsai L-H. Mechanisms of DNA damage-mediated neurotoxicity in neurodegenerative disease. EMBO Rep. 2022;23(6):e54217.
Lin X, et al. Contributions of DNA damage to alzheimer’s disease. Int J Mol Sci. 2020;21(5):1666.
Davydov V, Hansen LA, Shackelford DA. Is DNA repair compromised in alzheimer’s disease? Neurobiol Aging. 2003;24(7):953–68.
Chen J, et al. DNA damage and cell cycle events implicate cerebellar dentate nucleus neurons as targets of alzheimer’s disease. Mol Neurodegener. 2010;5:60.
Shanbhag NM, et al. Early neuronal accumulation of DNA double strand breaks in alzheimer’s disease. Acta Neuropathol Commun. 2019;7(1):77.
Su JH, Deng G, Cotman CW. Neuronal DNA damage precedes tangle formation and is associated with up-regulation of Nitrotyrosine in alzheimer’s disease brain. Brain Res. 1997;774(1):193–9.
Miller MB, et al. Somatic genomic changes in single alzheimer’s disease neurons. Nature. 2022;604(7907):714–22.
Mah LJ, El-Osta A, Karagiannis TC. γH2AX: a sensitive molecular marker of DNA damage and repair. Leukemia. 2010;24(4):679–86.
Myung N-H, et al. Evidence of DNA damage in alzheimer disease: phosphorylation of histone H2AX in astrocytes. AGE. 2008;30(4):209–15.
Tse K-H, et al. DNA damage-associated oligodendrocyte degeneration precedes amyloid pathology and contributes to Alzheimer’s disease and dementia. Volume 14. Alzheimer’s & Dementia; 2018;664–79. 5.
Al-Mashhadi S, et al. Oxidative glial cell damage associated with white matter lesions in the aging human brain. Brain Pathol. 2015;25(5):565–74.
Li Y, et al. Alzheimer’s amyloid-β accelerates human neuronal cell senescence which could be rescued by Sirtuin-1 and aspirin. Front Cell Neurosci. 2022;16:906270.
Farmer KM, et al. P53 aggregation, interactions with tau, and impaired DNA damage response in alzheimer’s disease. Acta Neuropathol Commun. 2020;8(1):132.
Ranganathan R, et al. Recent developments in the role of DNA damage response and Understanding its implications for new therapeutic approaches in alzheimer’s disease. Translational Med Aging. 2023;7:52–65.
Foret MK, et al. Early oxidative stress and DNA damage in Aβ-burdened hippocampal neurons in an Alzheimer’s-like Transgenic rat model. Commun Biology. 2024;7(1):861.
Silva ART, et al. Repair of oxidative DNA damage, cell-cycle regulation and neuronal death May influence the clinical manifestation of alzheimer’s disease. PLoS ONE. 2014;9(6):e99897.
Shen X et al. Neurons in vulnerable regions of the alzheimer’s disease brain display reduced ATM signaling. Eneuro, 2016;3(1):ENEURO.0124-15.2016.
Jacobsen E, et al. Deficiency of the Mre11 DNA repair complex in alzheimer’s disease brains. Mol Brain Res. 2004;128(1):1–7.
Kanungo J. DNA-PK deficiency in alzheimer’s disease. J Neurol Neuromedicine. 2016;1(3):17–22.
Welty S, Thathiah A, Levine AS. DNA damage increases secreted Aβ 40 and Aβ 42 in neuronal progenitor cells: relevance to alzheimer’s disease. J Alzheimers Dis. 2022;88:177–90.
Wang Z, et al. MRE11 ufmylation promotes ATM activation. Nucleic Acids Res. 2019;47(8):4124–35.
Qin B, et al. UFL1 promotes histone H4 ufmylation and ATM activation. Nat Commun. 2019;10(1):1242.
Qin B, et al. Dynamic recruitment of UFM1-specific peptidase 2 to the DNA double-strand breaks regulated by WIP1. Volume 3. Genome Instability & Disease 2022;217–26. 4.
Tian T, et al. UFL1 triggers replication fork degradation by MRE11 in BRCA1/2-deficient cells. Nat Chem Biol. 2024;20:1650–61.
Liu J, et al. UFMylation maintains tumour suppressor p53 stability by antagonizing its ubiquitination. Nat Cell Biol. 2020;22(9):1056–63.
Ahel D, et al. Poly(ADP-ribose)–Dependent regulation of DNA repair by the chromatin remodeling enzyme ALC1. Science. 2009;325(5945):1240–3.
Maya-Mendoza A, et al. High speed of fork progression induces DNA replication stress and genomic instability. Nature. 2018;559(7713):279–84.
Ray Chaudhuri A, Nussenzweig A. The multifaceted roles of PARP1 in DNA repair and chromatin remodelling. Nat Rev Mol Cell Biol. 2017;18(10):610–21.
Gong Y, et al. PARP1 ufmylation ensures the stability of stalled replication forks. Proc Natl Acad Sci. 2024;121(18):pe2322520121.
Colnaghi L et al. Tau and DNA damage in neurodegeneration. Brain Sci, 2020;10(12).
Sultan A, et al. Nuclear tau, a key player in neuronal DNA protection. J Biol Chem. 2011;286(6):4566–75.
Violet M, et al. A major role for Tau in neuronal DNA and RNA protection in vivo under physiological and hyperthermic conditions. Front Cell Neurosci. 2014;8:84.
Iijima-Ando K, et al. A DNA damage-activated checkpoint kinase phosphorylates Tau and enhances Tau-induced neurodegeneration. Hum Mol Genet. 2010;19(10):1930–8.
Park JS, et al. Brain somatic mutations observed in alzheimer’s disease associated with aging and dysregulation of Tau phosphorylation. Nat Commun. 2019;10(1):3090.
Nakamura M, et al. Aberrant accumulation of BRCA1 in alzheimer disease and other tauopathies. J Neuropathology Experimental Neurol. 2019;79(1):22–33.
Eftekharzadeh B, et al. Tau protein disrupts nucleocytoplasmic transport in alzheimer’s disease. Neuron. 2018;99(5):925–e9407.
Hooper C, et al. 53 is upregulated in alzheimer’s disease and induces Tau phosphorylation in HEK293a cells. Neurosci Lett. 2007;418(1):34–7.
Love S, Barber R, Wilcock GK. Increased poly(ADP-ribosyl)ation of nuclear proteins in alzheimer’s disease. Brain. 1999;122(2):247–53.
Mao K, Zhang G. The role of PARP1 in neurodegenerative diseases and aging. FEBS J. 2022;289(8):2013–24.
Hetz C, Zhang K, Kaufman RJ. Mechanisms, regulation and functions of the unfolded protein response. Nat Rev Mol Cell Biol. 2020;21(8):421–38.
Almanza A, et al. Endoplasmic reticulum stress signalling– from basic mechanisms to clinical applications. FEBS J. 2019;286(2):241–78.
Bertolotti A, et al. Dynamic interaction of bip and ER stress transducers in the unfolded-protein response. Nat Cell Biol. 2000;2(6):326–32.
Scheper W, Hoozemans JJM. The unfolded protein response in neurodegenerative diseases: a neuropathological perspective. Acta Neuropathol. 2015;130(3):315–31.
Hetz C, Saxena S. ER stress and the unfolded protein response in neurodegeneration. Nat Reviews Neurol. 2017;13(8):477–91.
Wodrich APK, et al. The unfolded protein responses in health, aging, and neurodegeneration: recent advances and future considerations. Front Mol Neurosci. 2022;15:831116.
van der Harg JM, et al. The unfolded protein response mediates reversible Tau phosphorylation induced by metabolic stress. Cell Death Dis. 2014;5(8):e1393–1393.
Hoozemans JJM, et al. The unfolded protein response is activated in pretangle neurons in alzheimer’s disease Hippocampus. Am J Pathol. 2009;174(4):1241–51.
Duran-Aniotz C, et al. IRE1 signaling exacerbates alzheimer’s disease pathogenesis. Acta Neuropathol. 2017;134(3):489–506.
Radford H, et al. PERK Inhibition prevents tau-mediated neurodegeneration in a mouse model of frontotemporal dementia. Acta Neuropathol. 2015;130(5):633–42.
Uddin MS, Yu WS, Lim LW. Exploring ER stress response in cellular aging and neuroinflammation in alzheimer’s disease. Ageing Res Rev. 2021;70:101417.
Ma T, et al. Suppression of eIF2α kinases alleviates alzheimer’s disease–related plasticity and memory deficits. Nat Neurosci. 2013;16(9):1299–305.
Chung CH, Yoo HM. Emerging role of protein modification by UFM1 in cancer. Biochem Biophys Res Commun. 2022;633:61–3.
Jing Y, Mao Z, Chen F. UFMylation system: an emerging player in tumorigenesis. Cancers. 2022;14(14):3501.
Hu X, et al. Ubiquitin-fold modifier 1 inhibits apoptosis by suppressing the Endoplasmic reticulum stress response in Raw264.7 cells. Int J Mol Med. 2014;33(6):1539–46.
Miller C, et al. RCAD/BiP pathway is necessary for the proper synthesis of digestive enzymes and secretory function of the exocrine pancreas. Am J Physiology-Gastrointestinal Liver Physiol. 2017;312(3):G314–26.
Azfer A, et al. Activation of Endoplasmic reticulum stress response during the development of ischemic heart disease. Am J Physiol Heart Circ Physiol. 2006;291(3):H1411–20.
Zhang Y, et al. Transcriptional regulation of the Ufm1 conjugation system in response to disturbance of the Endoplasmic reticulum homeostasis and Inhibition of vesicle trafficking. PLoS ONE. 2012;7(11):e48587.
Jang I, et al. PDIA1/P4HB is required for efficient proinsulin maturation and ß cell health in response to diet induced obesity. eLife. 2019;8:e44528.
Zhu J, et al. P4HB ufmylation regulates mitochondrial function and oxidative stress. Free Radic Biol Med. 2022;188:277–86.
Luo H, et al. UFMylation of HRD1 regulates Endoplasmic reticulum homeostasis. FASEB J. 2023;37(11):e23221.
Kaneko M, Okuma Y, Nomura Y. Molecular approaches to the treatment, prophylaxis, and diagnosis of alzheimer’s disease: possible involvement of HRD1, a novel molecule related to Endoplasmic reticulum stress, in alzheimer’s disease. J Pharmacol Sci. 2012;118(3):325–30.
Kaneko M, et al. Loss of HRD1-Mediated protein degradation causes amyloid precursor protein accumulation and amyloid-β generation. J Neurosci. 2010;30(11):3924–32.
Shen YX, et al. Hrd1 facilitates Tau degradation and promotes neuron survival. Curr Mol Med. 2012;12(2):138–52.
Klionsky DJ, et al. Guidelines for the use and interpretation of assays for monitoring autophagy (3rd edition). Autophagy. 2016;12(1):1–222.
Bento CF, et al. Mammalian autophagy: how does it work?? Annu Rev Biochem. 2016;85(1):685–713.
Dikic I, Elazar Z. Mechanism and medical implications of mammalian autophagy. Nat Rev Mol Cell Biol. 2018;19(6):349–64.
Saxton RA, Sabatini DM. mTOR signaling in growth, metabolism, and disease. Cell. 2017;168(6):960–76.
Kim YC, Guan K-L. mTOR: a Pharmacologic target for autophagy regulation. J Clin Investig. 2015;125(1):25–32.
Hosokawa N, et al. Nutrient-dependent mTORC1 association with the ULK1–Atg13–FIP200 complex required for autophagy. Mol Biol Cell. 2009;20(7):1981–91.
Kuijpers M, et al. Neuronal autophagy regulates presynaptic neurotransmission by controlling the axonal Endoplasmic reticulum. Neuron. 2021;109(2):299–e3139.
Boland B, et al. Autophagy induction and autophagosome clearance in neurons: relationship to autophagic pathology in alzheimer’s disease. J Neurosci. 2008;28(27):6926–37.
Kulkarni VV, et al. Retrograde axonal autophagy and endocytic pathways are parallel and separate in neurons. J Neurosci. 2022;42(45):8524–41.
Melia TJ, Lystad AH, Simonsen A. Autophagosome biogenesis: from membrane growth to closure. J Cell Biol, 2020;219(6).
Shibata M, et al. Regulation of intracellular accumulation of mutant Huntingtin by Beclin 1 *. J Biol Chem. 2006;281(20):14474–85.
Tsong H, Holzbaur ELF, Stavoe AKH. Aging Differentially Affects Axonal Autophagosome Formation Maturation Autophagy. 2023;19(12):3079–95.
Nixon RA. The role of autophagy in neurodegenerative disease. Nat Med. 2013;19(8):983–97.
Frake RA, et al. Autophagy and neurodegeneration. J Clin Investig. 2015;125(1):65–74.
Menzies FM, Fleming A, Rubinsztein DC. Compromised autophagy and neurodegenerative diseases. Nat Rev Neurosci. 2015;16(6):345–57.
Long Z, et al. Dynamic changes of autophagic flux induced by Abeta in the brain of postmortem alzheimer’s disease patients, animal models and cell models. Aging. 2020;12(11):10912–30.
Lachance V, et al. Autophagy protein NRBF2 has reduced expression in alzheimer’s brains and modulates memory and amyloid-beta homeostasis in mice. Mol Neurodegeneration. 2019;14(1):43.
Nixon RA, Yang D-S. Autophagy failure in alzheimer’s disease—locating the primary defect. Neurobiol Dis. 2011;43(1):38–45.
Suzuki K, Terry RD. Fine structural localization of acid phosphatase in senile plaques in alzheimer’s presenile dementia. Acta Neuropathol. 1967;8(3):276–84.
Pickford F, et al. The autophagy-related protein Beclin 1 shows reduced expression in early alzheimer disease and regulates amyloid β accumulation in mice. J Clin Investig. 2008;118(6):2190–9.
Heckmann BL, et al. Noncanonical function of an autophagy protein prevents spontaneous alzheimer’s disease. Sci Adv. 2020;6(33):eabb9036.
Lee J-H, et al. Lysosomal proteolysis and autophagy require presenilin 1 and are disrupted by Alzheimer-Related PS1 mutations. Cell. 2010;141(7):1146–58.
Belinson H, et al. Activation of the amyloid cascade in Apolipoprotein E4 Transgenic mice induces lysosomal activation and neurodegeneration resulting in marked cognitive deficits. J Neurosci. 2008;28(18):4690–701.
Sohn H-Y, et al. ApoE4 attenuates autophagy via FoxO3a repression in the brain. Sci Rep. 2021;11(1):17604.
Parcon PA, et al. Apolipoprotein E4 inhibits autophagy gene products through direct, specific binding to CLEAR motifs. Alzheimer’s Dement. 2018;14(2):230–42.
Simonovitch S, et al. Impaired autophagy in APOE4 astrocytes. J Alzheimers Dis. 2016;51:915–27.
Piras A, et al. Autophagic and lysosomal defects in human tauopathies: analysis of post-mortem brain from patients with Familial alzheimer disease, corticobasal degeneration and progressive supranuclear palsy. Acta Neuropathol Commun. 2016;4(1):22.
Zhang Z, et al. Autophagy in alzheimer’s disease pathogenesis: therapeutic potential and future perspectives. Ageing Res Rev. 2021;72:101464.
Fang EF, et al. Mitophagy inhibits amyloid-β and Tau pathology and reverses cognitive deficits in models of alzheimer’s disease. Nat Neurosci. 2019;22(3):401–12.
Lafay-Chebassier C, et al. mTOR/p70S6k signalling alteration by Aβ exposure as well as in APP-PS1 Transgenic models and in patients with alzheimer’s disease. J Neurochem. 2005;94(1):215–25.
Fan S, et al. PI3K/AKT/mTOR/p70S6K pathway is involved in Aβ25-35-Induced autophagy. Biomed Res Int. 2015;2015(1):161020.
Li W, et al. Autophagy is involved in oligodendroglial precursor-mediated clearance of amyloid peptide. Mol Neurodegeneration. 2013;8(1):27.
Kuang H, et al. Exploring the bi-directional relationship between autophagy and alzheimer’s disease. CNS Neurosci Ther. 2020;26(2):155–66.
Cavieres VA, et al. Tetrahydrohyperforin inhibits the proteolytic processing of amyloid precursor protein and enhances its degradation by Atg5-Dependent autophagy. PLoS ONE. 2015;10(8):e0136313.
Nilsson P, et al. Aβ secretion and plaque formation depend on autophagy. Cell Rep. 2013;5(1):61–9.
Chen X, et al. Promoting Tau secretion and propagation by hyperactive p300/CBP via autophagy-lysosomal pathway in Tauopathy. Mol Neurodegeneration. 2020;15(1):2.
Caccamo A, et al. Naturally secreted Amyloid-β increases mammalian target of Rapamycin (mTOR) activity via a PRAS40-mediated mechanism**. J Biol Chem. 2011;286(11):8924–32.
Sanchez-Varo R, et al. Abnormal accumulation of autophagic vesicles correlates with axonal and synaptic pathology in young alzheimer’s mice hippocampus. Acta Neuropathol. 2012;123(1):53–70.
Feng Q, et al. MAPT/Tau accumulation represses autophagy flux by disrupting IST1-regulated ESCRT-III complex formation: a vicious cycle in alzheimer neurodegeneration. Autophagy. 2020;16(4):641–58.
Zhang W, et al. Impairment of the autophagy–lysosomal pathway in alzheimer’s diseases: pathogenic mechanisms and therapeutic potential. Acta Pharm Sinica B. 2022;12(3):1019–40.
Xiao Q, et al. Neuronal-Targeted TFEB accelerates lysosomal degradation of APP, reducing Aβ generation and amyloid plaque pathogenesis. J Neurosci. 2015;35(35):12137–51.
Ozcelik S, et al. Rapamycin attenuates the progression of Tau pathology in P301S Tau Transgenic mice. PLoS ONE. 2013;8(5):e62459.
DeJesus R, et al. Functional CRISPR screening identifies the ufmylation pathway as a regulator of SQSTM1/p62. eLife. 2016;5:e17290.
Blaudin de Thé F-X, et al. P62 accumulates through neuroanatomical circuits in response to tauopathy propagation. Acta Neuropathol Commun. 2021;9(1):177.
Ferrari L, et al. Tau fibrils evade autophagy by excessive p62 coating and TAX1BP1 exclusion. Sci Adv. 2024;10(24):eadm8449.
Li H, et al. A neuroprotective role of ufmylation through Atg9 in the aging brain of Drosophila. Cell Mol Life Sci. 2023;80(5):129.
Sharoar MG, et al. Sequential formation of different layers of dystrophic neurites in alzheimer’s brains. Mol Psychiatry. 2019;24(9):1369–82.
Wang Z, et al. VCP/p97 ufmylation stabilizes BECN1 and facilitates the initiation of autophagy. Autophagy. 2024;26:1–14.
Saha I, et al. The AAA + chaperone VCP disaggregates Tau fibrils and generates aggregate seeds in a cellular system. Nat Commun. 2023;14(1):560.
Liang JR, et al. A Genome-wide ER-phagy screen highlights key roles of mitochondrial metabolism and ER-Resident ufmylation. Cell. 2020;180(6):1160–e117720.
Ishimura R, et al. The UFM1 system regulates ER-phagy through the ufmylation of CYB5R3. Nat Commun. 2022;13(1):7857.
Khaminets A, et al. Regulation of Endoplasmic reticulum turnover by selective autophagy. Nature. 2015;522(7556):354–8.
Chino H, Mizushima N. ER-Phagy: quality control and turnover of Endoplasmic reticulum. Trends Cell Biol. 2020;30(5):384–98.
Hübner CA, Dikic I. ER-phagy and human diseases. Cell Death Differ. 2020;27(3):833–42.
González A, et al. Ubiquitination regulates ER-phagy and remodelling of Endoplasmic reticulum. Nature. 2023;618(7964):394–401.
Öztürk Z, O’Kane CJ, Pérez-Moreno JJ. Axonal Endoplasmic reticulum dynamics and its roles in neurodegeneration. Front NeuroSci, 2020;14(48).
Pickrell AM, Youle RJ. The roles of PINK1, parkin, and mitochondrial fidelity in parkinson’s disease. Neuron. 2015;85(2):257–73.
Suzuki M, et al. Upregulation of ribosome complexes at the blood-brain barrier in alzheimer’s disease patients. J Cereb Blood Flow Metabolism. 2022;42(11):2134–50.
Wang H, Han X, Gao S. Identification of potential biomarkers for pathogenesis of alzheimer’s disease. Hereditas. 2021;158(1):23.
Mastroeni D, et al. Epigenetic changes in alzheimer’s disease: decrements in DNA methylation. Neurobiol Aging. 2010;31(12):2025–37.
Picchianti L, et al. Shuffled ATG8 interacting motifs form an ancestral Bridge between ufmylation and autophagy. EMBO J. 2023;42(10):e112053.
Wang H, et al. Integrated analysis of ultra-deep proteomes in cortex, cerebrospinal fluid and serum reveals a mitochondrial signature in alzheimer’s disease. Mol Neurodegeneration. 2020;15(1):43.
Mou W et al. Upregulation of neuronal ER-phagy improves organismal fitness and alleviates APP toxicity. Cell Rep, 2024;43(5).
Zou Y, et al. Identification of rare RTN3 variants in alzheimer’s disease in Han Chinese. Hum Genet. 2018;137(2):141–50.
Shi Q, et al. Reduced amyloid deposition in mice overexpressing RTN3 is adversely affected by preformed dystrophic neurites. J Neurosci. 2009;29(29):9163–73.
Murayama KS, et al. Reticulons RTN3 and RTN4-B/C interact with BACE1 and inhibit its ability to produce amyloid β-protein. Eur J Neurosci. 2006;24(5):1237–44.
Chaplin D.D. Overview of the immune response. J Allergy Clin Immunol. 2010;125(2, Supplement 2):S3–23.
DiSabato DJ, Quan N, Godbout JP. Neuroinflammation: the devil is in the details. J Neurochem. 2016;139(S2):136–53.
Haage V, De Jager PL. Neuroimmune contributions to alzheimer’s disease: a focus on human data. Mol Psychiatry. 2022;27(8):3164–81.
Jorfi M, Maaser-Hecker A, Tanzi RE. The neuroimmune axis of alzheimer’s disease. Genome Med. 2023;15(1):6.
Funato H, et al. Astrocytes containing amyloid beta-protein (Abeta)-positive granules are associated with Abeta40-positive diffuse plaques in the aged human brain. Am J Pathol. 1998;152(4):983–92.
Thal DR, et al. Amyloid β-protein (Aβ)-containing astrocytes are located preferentially near N-terminal-truncated Aβ deposits in the human entorhinal cortex. Acta Neuropathol. 2000;100(6):608–17.
Zhou Y, et al. TLR4 targeting as a promising therapeutic strategy for alzheimer disease treatment. Front NeuroSci. 2020;14:602508.
Sun E, et al. The pivotal role of NF-kB in the pathogenesis and therapeutics of alzheimer’s disease. Int J Mol Sci. 2022;23(16):8972.
Chen C-H, et al. Increased NF-κB signalling up-regulates BACE1 expression and its therapeutic potential in alzheimer’s disease. Int J Neuropsychopharmacol. 2012;15(1):77–90.
Zhong Z, et al. NF-κB restricts inflammasome activation via elimination of damaged mitochondria. Cell. 2016;164(5):896–910.
Baruch K, et al. PD-1 immune checkpoint Blockade reduces pathology and improves memory in mouse models of alzheimer’s disease. Nat Med. 2016;22(2):135–7.
Rosenzweig N, et al. PD-1/PD-L1 checkpoint Blockade harnesses monocyte-derived macrophages to combat cognitive impairment in a tauopathy mouse model. Nat Commun. 2019;10(1):465.
Onisiforou A, Zanos P. From viral infections to alzheimer’s disease: unveiling the mechanistic links through systems bioinformatics. J Infect Dis. 2024;230(Supplement2):S128–40.
Itzhaki RF, et al. Herpes simplex virus type 1 in brain and risk of alzheimer’s disease. Lancet. 1997;349(9047):241–4.
Wozniak M, Mee A, Itzhaki R. Herpes simplex virus type 1 DNA is located within alzheimer’s disease amyloid plaques. J Pathol. 2009;217(1):131–8.
Bertram L, et al. Genome-wide association analysis reveals putative alzheimer’s disease susceptibility loci in addition to APOE. Am J Hum Genet. 2008;83(5):623–32.
Hollingworth P, et al. Common variants at ABCA7, MS4A6A/MS4A4E, EPHA1, CD33 and CD2AP are associated with alzheimer’s disease. Nat Genet. 2011;43(5):429–35.
Naj AC, et al. Common variants at MS4A4/MS4A6E, CD2AP, CD33 and EPHA1 are associated with late-onset alzheimer’s disease. Nat Genet. 2011;43(5):436–41.
Guerreiro R, et al. TREM2 variants in alzheimer’s disease. N Engl J Med. 2013;368(2):117–27.
Zhang J, et al. Deficiency of murine UFM1-Specific E3 ligase causes microcephaly and inflammation. Mol Neurobiol. 2022;59(10):6363–72.
Bruno F, et al. Alzheimer’s disease as a viral disease: revisiting the infectious hypothesis. Ageing Res Rev. 2023;91:102068.
Hu X, et al. Ubiquitin fold modifier 1 activates NF-κB pathway by down-regulating LZAP expression in the macrophage of diabetic mouse model. Biosci Rep. 2020;40(1):BSR20191672.
Hu X, et al. Ubiquitin-fold modifier-1 participates in the diabetic inflammatory response by regulating NF-κB p65 nuclear translocation and the ubiquitination and degradation of IκBα. Drug Design, Development and Therapy 2020;795–810.
Xi P, et al. DDRGK1 regulates NF-κB activity by modulating IκBα stability. PLoS ONE. 2013;8(5):e64231.
Hayden MS, Ghosh S. Signaling to NF-κB. Genes Dev. 2004;18(18):2195–224.
Liu T, et al. NF-κB signaling in inflammation. Signal Transduct Target Therapy. 2017;2(1):17023.
Yu H, et al. Targeting NF-κB pathway for the therapy of diseases: mechanism and clinical study. Signal Transduct Target Therapy. 2020;5(1):209.
Li Y-Y, et al. Ufm1 inhibits LPS-induced endothelial cell inflammatory responses through the NF-κB signaling pathway. Int J Mol Med. 2017;39(5):1119–26.
Wang Z, et al. UFM1 inhibits the activation of the pyroptosis in LPS-induced goat endometritis. Theriogenology. 2023;196:50–8.
Yang G, et al. UFL1 attenuates IL-1β-induced inflammatory response in human osteoarthritis chondrocytes. Int Immunopharmacol. 2020;81:106278.
Li C, et al. UFL1 modulates NLRP3 inflammasome activation and protects against pyroptosis in LPS-stimulated bovine mammary epithelial cells. Mol Immunol. 2019;112:1–9.
Li X, et al. Integrated genomic approaches identify major pathways and upstream regulators in late onset alzheimer’s disease. Sci Rep. 2015;5(1):12393.
Chen J, et al. SIRT1 protects against Microglia-dependent Amyloid-β toxicity through inhibiting NF-κB signaling. J Biol Chem. 2005;280(48):40364–74.
Miron J, et al. TLR4 gene expression and Pro-Inflammatory cytokines in alzheimer’s disease and in response to hippocampal deafferentation in rodents. J Alzheimers Dis. 2018;63:1547–56.
Chen Y-C, et al. Sequence variants of toll like receptor 4 and Late-Onset alzheimer’s disease. PLoS ONE. 2012;7(12):e50771.
Miron J, et al. Association of TLR4 with alzheimer’s disease risk and presymptomatic biomarkers of inflammation. Alzheimer’s Dement. 2019;15(7):951–60.
Walter S, et al. Role of the Toll-Like receptor 4 in neuroinflammation in alzheimer’s disease. Cell Physiol Biochem. 2007;20(6):947–56.
Capiralla H, et al. Resveratrol mitigates lipopolysaccharide- and Aβ-mediated microglial inflammation by inhibiting the TLR4/NF-κB/STAT signaling cascade. J Neurochem. 2012;120(3):461–72.
Liu Y, et al. Beta-amyloid activates NLRP3 inflammasome via TLR4 in mouse microglia. Neurosci Lett. 2020;736:135279.
Chen G, et al. Exosomal PD-L1 contributes to immunosuppression and is associated with anti-PD-1 response. Nature. 2018;560(7718):382–6.
Juneja VR, et al. PD-L1 on tumor cells is sufficient for immune evasion in Immunogenic tumors and inhibits CD8 T cell cytotoxicity. J Exp Med. 2017;214(4):895–904.
Zhou J, et al. Dysregulation of PD-L1 by ufmylation imparts tumor immune evasion and identified as a potential therapeutic target. Proc Natl Acad Sci. 2023;120(11):e2215732120.
He C, et al. UFL1 ablation in T cells suppresses PD-1 ufmylation to enhance anti-tumor immunity. Mol Cell. 2024;84(6):1120–e11388.
Mao M, 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. 2022;10(12):e005668.
Kummer MP, et al. Microglial PD-1 stimulation by astrocytic PD‐L1 suppresses neuroinflammation and alzheimer’s disease pathology. EMBO J. 2021;40(24):e108662.
Tao Y, et al. UFL1 promotes antiviral immune response by maintaining STING stability independent of ufmylation. Cell Death Differ. 2023;30(1):16–26.
Snider DL et al. Signaling from the RNA sensor RIG-I is regulated by ufmylation. Proceedings of the National Academy of Sciences, 2022;119(15):e2119531119.
Wozniak MA, et al. Herpes simplex virus infection causes cellular β-amyloid accumulation and secretase upregulation. Neurosci Lett. 2007;429(2):95–100.
Wozniak MA, Frost AL, Itzhaki RF. Alzheimer’s Disease-Specific Tau phosphorylation is induced by herpes simplex virus type 1. J Alzheimers Dis. 2009;16:341–50.
Cairns DM, et al. A 3D human brain–like tissue model of herpes-induced alzheimer’s disease. Sci Adv. 2020;6(19):eaay8828.
Horner SM, et al. Proteomic analysis of mitochondrial-associated ER membranes (MAM) during RNA virus infection reveals dynamic changes in protein and organelle trafficking. PLoS ONE. 2015;10(3):e0117963.
Yiu SPT, et al. An Epstein-Barr virus protein interaction map reveals NLRP3 inflammasome evasion via MAVS ufmylation. Mol Cell. 2023;83(13):2367–e238615.
Kulsuptrakul J, et al. A genome-wide CRISPR screen identifies ufmylation and TRAMP-like complexes as host factors required for hepatitis A virus infection. Cell Rep. 2021;34(11):108859.
Lin H-C, et al. Association of alzhemier’s disease with hepatitis C among patients with bipolar disorder. PLoS ONE. 2017;12(6):e0179312.
Green, R., et al., SARS-CoV-2 infection increases the gene expression profile for Alzheimer’s disease risk. Molecular Therapy Methods & Clinical Development, 2022;27:217–229.
Gu Q, et al. Downregulation of 14-3-3 proteins in alzheimer’s disease. Mol Neurobiol. 2020;57(1):32–40.
Macdonald R, et al. Mitochondrial abnormalities in parkinson’s disease and alzheimer’s disease: can mitochondria be targeted therapeutically? Biochem Soc Trans. 2018;46(4):891–909.
Mary A, et al. Mitophagy in alzheimer’s disease: molecular defects and therapeutic approaches. Mol Psychiatry. 2023;28(1):202–16.
Moreira PI, et al. Mitochondrial abnormalities and oxidative imbalance in alzheimer disease. J Alzheimer’s Disease. 2006;9(2):147–53.
Ding Q, et al. Ribosome dysfunction is an early event in alzheimer’s disease. J Neurosci. 2005;25(40):9171–5.
Lu B. Translational regulation by ribosome-associated quality control in neurodegenerative disease, cancer, and viral infection. Front Cell Dev Biology. 2022;10:970654.
Simsek D, et al. The mammalian ribo-interactome reveals ribosome functional diversity and heterogeneity. Cell. 2017;169(6):1051–e106518.
Walczak CP, et al. Ribosomal protein RPL26 is the principal target of ufmylation. Proc Natl Acad Sci. 2019;116(4):1299–308.
Wang L, et al. UFMylation of RPL26 links translocation-associated quality control to Endoplasmic reticulum protein homeostasis. Cell Res. 2020;30(1):5–20.
Dixon SJ, et al. Ferroptosis: an Iron-Dependent form of nonapoptotic cell death. Cell. 2012;149(5):1060–72.
Chen K, et al. Ferroptosis, a potential therapeutic target in alzheimer’s disease. Front Cell Dev Biology. 2021;9:704298.
Majerníková N, et al. The link between amyloid β and ferroptosis pathway in alzheimer’s disease progression. Cell Death Dis. 2024;15(10):782.
Yang J, et al. Metformin induces ferroptosis by inhibiting ufmylation of SLC7A11 in breast cancer. J Experimental Clin Cancer Res. 2021;40(1):206.
Coppedè F, Migliore L. DNA damage in neurodegenerative diseases. Mutat Research/Fundamental Mol Mech Mutagen. 2015;776:84–97.
Qin N, Geng A, Xue R. Activated or impaired: an overview of DNA repair in neurodegenerative diseases. Aging Disease. 2022;13(4):987–1004.
Fleming A, et al. The different autophagy degradation pathways and neurodegeneration. Neuron. 2022;110(6):935–66.
Stephenson J, et al. Inflammation in CNS neurodegenerative diseases. Immunology. 2018;154(2):204–19.
Filipek B. T. Poplawski. Elevated level of DNA damage and impaired repair of DNA oxidative damage in multiple sclerosis patients (P7-6.010). 2024;Neurology 102 17_supplement_1 p7185.
Misrielal C, et al. Autophagy in multiple sclerosis: two sides of the same coin. Front Cell Neurosci. 2020;14:603710.
Santhosh Kumar S, et al. Sequential CRISPR screening reveals partial NatB Inhibition as a strategy to mitigate alpha-synuclein levels in human neurons. Sci Adv. 2024;10(6):eadj4767.
Wang L, et al. Mono-UFMylation promotes misfolding-associated secretion of α-synuclein. Sci Adv. 2024;10(11):eadk2542.
Watson CM, et al. Identification of a mutation in the ubiquitin-fold modifier 1-specific peptidase 2 gene, UFSP2, in an extended South African family with Beukes hip dysplasia. South Afr Med J. 2015;105(7):558–63.
Di Rocco M, et al. Novel spondyloepimetaphyseal dysplasia due to UFSP2 gene mutation. Clin Genet. 2018;93(3):671–4.
Zhang G, et al. UFSP2-related spondyloepimetaphyseal dysplasia: A confirmatory report. Eur J Med Genet. 2020;63(11):104021.
Acknowledgements
We thank all patients and their families who participate in research studies.
Funding
This work was supported by a Florida Department of Health Ed and Ethel Moore Alzheimer’s disease research program grant 22A07 (to F.C.F.), and a fellowship from Alzheimer’s Association AARF-22-974258 (to T.Y.). F.C.F. is supported by the Mayo Clinic Office of Research Equity, Inclusion and Diversity and the Center for Biomedical Discovery. B.D.C. is supported by a Career Development Award in Neurodegenerative Disease Research from the Dominium Foundation. W.S. is supported in part by the National Institutes of Health (NIH)/National Institute of Neurological Disorders and Stroke (NINDS) [RF1 NS085070, R01 NS110085, and U54 NS110435], National Institute on Aging (NIA) [R56 AG062556], the Department of Defense Congressionally Directed Medical Research Programs (CDMRP) [W81XWH-17-1-0248], Mayo Clinic Foundation, Mayo Clinic Center for Biomedical Discovery, Mayo Clinic Robert and Arlene Kogod Center on Aging, the Michael J. Fox Foundation for Parkinson’s Research, an American Parkinson Disease Association Center for Advanced Research, and the Ted Nash Long Life Foundation.
Author information
Authors and Affiliations
Contributions
T.Y. and F.C.F. conceptualized, wrote, and revised the manuscript, B.D.C., Z.L. and W.S. reviewed and revised the manuscript. All authors read and approved the final manuscript.
Corresponding author
Ethics declarations
Ethics approval and consent to participate
Not applicable.
Consent for publication
Not applicable.
Competing interests
The authors declare no competing interests.
Additional information
Publisher’s note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.
About this article
Cite this article
Yan, T., Clarkson, B.D., Lou, Z. et al. The many connections of UFMylation with Alzheimer’s disease: a comprehensive review. Mol Neurodegeneration 20, 66 (2025). https://doi.org/10.1186/s13024-025-00855-8
Received:
Accepted:
Published:
DOI: https://doi.org/10.1186/s13024-025-00855-8

