Next Article in Journal
Integrated Multi-Omic Characterization of the Detachment Process of Adherent Vero Cells with Animal-Based and Animal-Origin-Free Enzymes
Next Article in Special Issue
Activation of β2-Adrenergic Receptors in Microglia Alleviates Neuropathic Hypersensitivity in Mice
Previous Article in Journal
Whole Transcriptome Sequencing Reveals Cancer-Related, Prognostically Significant Transcripts and Tumor-Infiltrating Immunocytes in Mantle Cell Lymphoma
Previous Article in Special Issue
Prolonged Suppression of Neuropathic Hypersensitivity upon Neurostimulation of the Posterior Insula in Mice
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Role of SUMOylation in Neurodegenerative Diseases

Institute of Pharmacology, Medical Faculty Heidelberg, Heidelberg University, 69120 Heidelberg, Germany
*
Author to whom correspondence should be addressed.
Cells 2022, 11(21), 3395; https://doi.org/10.3390/cells11213395
Submission received: 8 August 2022 / Revised: 23 October 2022 / Accepted: 24 October 2022 / Published: 27 October 2022
(This article belongs to the Special Issue Cellular and Molecular Mechanisms Underlying Pain Chronicity)

Abstract

:
Neurodegenerative diseases (NDDs) are irreversible, progressive diseases with no effective treatment. The hallmark of NDDs is the aggregation of misfolded, modified proteins, which impair neuronal vulnerability and cause brain damage. The loss of synaptic connection and the progressive loss of neurons result in cognitive defects. Several dysregulated proteins and overlapping molecular mechanisms contribute to the pathophysiology of NDDs. Post-translational modifications (PTMs) are essential regulators of protein function, trafficking, and maintaining neuronal hemostasis. The conjugation of a small ubiquitin-like modifier (SUMO) is a reversible, dynamic PTM required for synaptic and cognitive function. The onset and progression of neurodegenerative diseases are associated with aberrant SUMOylation. In this review, we have summarized the role of SUMOylation in regulating critical proteins involved in the onset and progression of several NDDs.

1. Introduction

Neurodegenerative diseases (NDDs) impose a substantial medical and financial burden on society worldwide. NDDs are marked by neuronal dysfunction and death, causing progressive degeneration of the structure and function of the central and peripheral nervous system [1,2,3]. A longer life span results in an increased prevalence of age-dependent neurodegenerative diseases. Thus, a significant percentage of the population affected by NDDs is elderly. Inadequate knowledge and a lack of diagnostics tools cause NDDs to have limited options for treatment. Clinical interventions may help to reduce the progression of neurodegenerative diseases, but they fail to cure them permanently. NDDs have a devastating impact on individuals, family and society. The onset of NDDs causes mild symptoms that are easy to ignore.
The progressive loss of a large number of neurons and the miscommunication of neuronal circuits results in memory impairment, learning disabilities, deteriorated synaptic function and cognitive defects [1,4,5,6,7]. NDDs are associated with the transformation and accumulation of misfolded pathogenic proteins [8], mitochondrial dysfunction [9], oxidative stress [10,11], glutamate toxicity and neuroinflammation [12] in neuronal cells, leading to cellular dysfunction and neuronal death. Alteration in post-translational modifications (PTMs) could lead to dysregulation in intracellular processes. This could be because of compromised protein-quality control, altered protein–protein interactions, dysfunctional molecular chaperones, and the impairment of the ubiquitin–proteasome system and lysosomal degradation pathways [13,14]. These cause the accumulation of toxic aggregates and contribute to neuronal degeneration [15]. PTMs are covalent modifications of protein that operate by attaching different chemical entities at single or multiple amino acid residues. These modifications add to the complexity of proteins’ functions by regulating their activity, localization, interactions, life cycle and trafficking [16,17]. The PTMs are classified into several groups: acetylation, phosphorylation, nitration, methylation, ubiquitination and SUMOylation [17,18]. Several studies have demonstrated that aberrant PTM is the reason for pathogenicity associated with NDDs. Herein, we summarize the role of SUMOylation in the onset and progression of neurodegenerative diseases.

2. SUMOylation

SUMOylation involves the attachment of small ubiquitin-like modifier (SUMO) proteins to the lysine residues in the protein. Meluh and Koshland first identified it in 1995 [19]. SUMO proteins belong to the ubiquitin-like proteins family (Ubls), characterized by the presence of globular β-grasp fold and a C-terminus Gly-Gly motif in the mature peptide [20,21,22]. In late 1990, SUMO-encoding genes were identified in mammals encoding five different SUMO paralogs, namely, SUMO-1, -2, -3, -4 and -5. SUMO2 and SUMO3 share 97% peptide sequence homology, and differ only in the second and third amino acids and the missing asparagine in SUMO3 [22,23,24,25]. Mature SUMO2 and SUMO3 are 92 and 93 amino acids. The available antibodies cannot differentiate between SUMO2 and SUMO3, collectively referred to as SUMO2/3 [26]. The presence of internal SUMO acceptor lysine K11 in SUMO2/3 enables them to form uniform poly-SUMO chains [23,26]. The additional lysine residue in SUMO2/3 contributes to chain formation under different physiological conditions. SUMO1 is distinct and shares only 47% sequence identity with SUMO2/3. In contrast to SUMO1 and -2/3, SUMO 4 and SUMO5 have not been studied in detail. Guo et al. showed that SUMO4 conjugates to I kappa B alpha (IκB) and negatively regulates NK kappa B (NKκB) transcriptional factor activity [25]. Additional studies have shown the role of SUMO4 in oxidative stress in human placenta tissue [27]. SUMO1 and -2/3 are widely expressed in tissues. In contrast, SUMO4 expression is restricted to the heart, spleen, pancreatic tissues, kidney, brain and lymph node. SUMO5 is highly homologous to SUMO1 [28,29]. It is shown to express in primates’ testes and blood cells, but appears absent in mice. Promyelocytic leukaemia (PML) conjugate to polySUMO5 at lysine 160 residue, which facilitates the formation of promyelocytic leukaemia nuclear bodies (PML-NB) [30] that play a crucial role in regulating transcriptional activity, DNA repair, tumor suppression and apoptosis. Dysregulation in PML-NB generation or function causes polyglutamine repeat-induced neurodegenerative diseases [30].
SUMOylation is a dynamic process involving target protein switching between SUMOylated or deSUMOylated states. SUMOylation regulates target protein trafficking and subcellular localization, stability, solubility, activity structural conformations, and protein interactions [31]. In an unstressed cell, only a tiny fraction of the target protein is SUMOylated [32]. Stress or any other physiological signal could change the balance between the SUMO-conjugated and unconjugated protein pools. A SUMOylation burst may alter the modified protein’s solubility and structural conformation. As a result, it could instigate other PTMs, such as phosphorylation, ubiquitination, and acetylation [31]. These modifications can stabilize the substrate protein in the functional complex or regulate the subcellular localization, even after the deSUMOylation event. Alternatively, in response to intrinsic and external stimuli, the SUMOylated fraction of the protein could be trafficked to the target subcellular compartment, or recruited within the functional complex [33]. The emerging concept proposes that SUMOylation modifies multiple proteins within a biological process. SUMOylation may inhibit or activate different components of a biological pathway to integrate the biological process outcome [31,34]. For example, in Saccharomyces cerevisiae, multiple proteins involved in DNA repair pathways undergo SUMOylation in response to DNA damage signals. It expedites the DNA repair process [35]. Similarly, in diabetes induced-neuropathy, the SUMOylation of several metabolic enzymes regulates the bioenergetics of cells. This results in the regulation growth of tissue and limits the tissue damage pathways [36]. Although an analysis of individual SUMOylated target proteins is very much needed, new studies deciphering the impact of SUMOylation on different signaling cascades in human disease are a critical approach for the future.
SUMOylation is a reversible post-translational protein modification. Similar to ubiquitination, SUMO moieties form isopeptide bonds with the ε-amino group of acceptor target lysine residue [22,31,37]. SUMOylation is an energy-driven, enzymatic cascade that involves E1-activating enzymes, E2-conjugating enzymes and E3 SUMO ligases (a schematic overview is given in Figure 1). The expressed SUMO forms are C-terminally extended precursors. The SUMO precursors undergo C-terminal cleavage mediated by Sentrin/SUMO-specific protease (SENP) enzymes, which expose the C-terminal diglycine motif of SUMO. The mature SUMO protein binds to the heterodimeric E1 enzyme. The E1 enzyme consists of SUMO-activating enzyme subunits 1 (SAE1) and subunit 2 (SAE2, also known as Uba2). The activation of SUMO is an ATP-dependent two-step reaction. The first step involves the SAE1-mediated adenylation of the C-terminal diglycine motif of SUMO. The structural modulation expedites the binding of adenylated-SUMO molecules via a thioester bond to cysteine residues in the SAE2 enzyme. The SAE2-attached SUMO is then actively transferred to the cysteine residue of the E2-conjugating enzyme. UBE2I (ubiquitin conjugating enzyme E2I) is the human analogue of the E2-conjugating enzyme, and it is also known as UBC9 in yeast [38]. This single known enzyme ensures SUMO binding to the substrate lysine at the consensus site.
In addition to UBC9-mediated target recognition potency, the binding affinity of UBC9 to the target sequence enhances substrate SUMOylation. Additional mechanisms further vary the pool of SUMOylated proteins in a given tissue type. These involve the external or internal stimuli-induced regulation of UBC9 expression. UBC9 activity is modulated via intrinsic post-translation modifications, such as self-SUMOylation at Lys 14 and Cdk1/Cyclin B-mediated phosphorylation [39]. Identifying a single E2-conjugating enzyme is perplexing, and a cause for a pathogen to ambush the SUMOylation pathway to facilitate proliferation in host cells. Mice lacking UBC9 die during embryonic development [40]. The E2-conjugated enzyme promotes the transfer of the SUMO molecule to the target lysine. The transfer generally requires SUMO E3 ligase activity, which stabilizes the interaction between target lysine and SUMO-charged E2-conjugated enzyme. The E3 ligase thereby reinforces the formation of a thioester bond between the target lysine and SUMO molecules [37]. This process does not require interaction between the E3 ligases and target lysine residue in the consensus motif of the substrate protein. SUMO E3 ligases promote quantitative target modification by expediting SUMO conjugation.
Based on molecular structure, the SUMO–E3 ligases can be classified mainly into the SP-RING domain family, the TRIM superfamily, SIM-containing SUMO E3 ligases and others [41,42,43,44] (list of E3 ligases, Table 1). The SP-RING domain in E3 ligases binds to UBC9 and stimulates SUMO ligation to target lysine residues. Protein inhibitors of activation STAT (PIAS) proteins were the first identified member of the SP-RING domain family of enzymes [45]. Furthermore, PIAS proteins contain a subsidiary SUMO-interaction motif (SIM) that promotes SUMO-UBC9-PIAS complex formation, increasing SUMOylation efficacy on the target lysine residues [43]. The TRIM superfamily of E3 ligases has approximately 100 members. They share the common tripartite motif (TRIM) consisting of the RING domain, B-box domains and coiled-coil region [41]. The RING domain commences UBC9 binding, whereas the B-box domain binds to the substrate, which enhances the SUMO transfer from UBC9 to target lysine [44]. SIM-containing E3 ligases form the third type of ligases in the SUMOylation pathway, where SIMs are the short stretch of hydrophobic residues with an acidic or polar residue at position 2 or 3. The SIM motif sequence forms a tetra core sequence of ψ X ψ ψ or ψ ψ X ψ. In the consensus sequence, “ψ” is the hydrophobic amino acid with a large non-polar side chain, typically Isoleucine (I), Valine (V) or Leucine (L), and X is any amino acid. The hydrophobic core is flanked on one side or the other by negative charged Aspartic acid (Asp), Glutamic acid (Glu) or Phosphoserine (Ser) residues. Hydrophobic sequences facilitate SUMO interaction. RanBP2, CBX4, SLX4 and ZNF451 are some of the more well-studied E3 ligases in this family [46,47,48,49]. Even though several E3 ligases have been identified, further investigation is required to understand their functionality in substrate structure–function recognition. SENPs can rapidly remove the SUMO attached to a lysine residue. This process is known as deSUMOylation.
Proteomic analysis revealed that most acceptor lysine residue is in the consensus motif. The classical consensus SUMOylation motif is ψKXE/D, where ψ represents hydrophobic residue, K is the acceptor lysine residue, X represents any amino acid, and E/D is glutamic or aspartic acid residues, respectively [75]. ψ contributes to the binding of UBC9, and its mutation abolishes complex formation and the SUMOylation of the target lysine. In addition, several other SUMOylation motifs were identified. They include the inverse consensus motif E/DXKψ [76], phosphorylation-dependent SUMOylation motif (PDSM; ψKXEXX(pS)P), where P is proline residue [77], hydrophobic consensus motif (ψψψKXE) [76] and negatively charged amino acid-dependent SUMOylation motif (NDSM; ψ KXEXXEEEE) [78]. These PDSMs or NDSMs represent an additional way to regulate the substrate SUMOylation in an inducible manner. The proximity of the phosphorylation site/negatively charged motif to the SUMOylation motif directly affects the substrate–UBC9 affinity and thus introduces a further regulatory steps [77,78]. Table 2 summarizes the different SUMOylation motifs.
Several studies have established the role of SUMOylation in the onset and progression of NDDs. Aberrant SUMOylation causes the aggregation of the toxic proteins, defects in protein trafficking, the modification of the ion channel properties and the release of neurotransmitters in neurons, oxidative stress and alteration in gene expression. Altered SUMOylation potentially modifies the functions of hundreds of proteins in many different pathways, and results in cognitive deficits associated with NDDs.

3. Role of SUMOylation in Alzheimer’s Disease (AD)

AD is the dominant form of escalating dementia. AD primarily occurs in an ageing population, but hereditary or non-hereditary mutations, lifestyle and environment may contribute to the onset and progression of AD pathogenicity. Currently, there are more than 50 million AD patients, and by 2050 these numbers are predicted to reach 152 million [6]. Clinically, AD is diagnosed and marked by initial memory dysfunction and, in different advanced stages, is associated with multiple cognitive dysfunctions. AD is associated with the aggregation of extracellular amyloid plaques and intracellular neurofibrillary tangles (NFT), gliosis, and the extensive loss of neurons and synapses. Amyloid plaques are primarily composed of deposits of beta-amyloid protein (Aβ) formed by the proteolytic cleavage of the transmembrane amyloid precursor protein (APP) by beta-secretase (β-secretase) and gamma-secretase (γ-secretase). In the hippocampus, cortex and amygdala, aggregating these plaques engenders neurotoxicity, causes astrocytes and microglia stimulation, and results in neuronal dysfunction and degeneration [79,80,81,82]. NFTs are abnormally paired helical filaments of the hyperphosphorylated Tau proteins. Under physiological conditions, Tau protein binds to microtubules and assists in their stabilization and neuronal transport. However, the hyperphosphorylation of Tau causes microtubules disintegration. It results in the accumulation of NFT in the neuralperikaryal, cytoplasm, axons and dendrites, which causes neuronal devastation and degeneration. This highly complex interplay results in oxidative and mitochondrial stress that causes synaptic homeostasis loss and neuronal loss [83,84].
Cell culture-based in vitro systems and a transgenic mouse model of AD were used to study the impact of SUMOylation on AD pathology. Expression studies in the early-aged Tg2576 mouse model of AD revealed no changes in the global levels of SUMO-1 [85]. In contrast, SUMO-2/3 showed several disparities in their expression patterns. However, aged Tg2576 mice exhibit excessive levels of SUMO-1 conjugated proteins in cortical and hippocampal tissue compared to age-matched wildtype mice [85]. Electrophysiological studies in hippocampal neurons manifested that impaired SUMOylation contributes to deficits in learning and memory pathology associated with Alzheimer’s disease [86]. The overexpression of UBC9 rescued Aβ-mediated deficits in long-term potentiation (LTP) and hippocampal-dependent learning and memory in a mouse model of AD [86]. UBC9 is highly mobile in neurons [87]. The Aβ peptide facilitates the redistribution of the UBC9 protein from presynaptic to postsynaptic terminals. Extracellular vesicles (EV) are responsible for the release of UBC9 in the presynaptic neuron, and also may facilitate the synaptic gap transmission. The presence of the Aβ peptide results in Synaptosome Associated Protein 23 (SNAP-23) phosphorylation, facilitating the release of UBC9 from EVs, promoting the development of AD [88]. In addition, APP is SUMOylated at lysine 587 and 595 [89]. Proximity to the β-secretase cleavage site raises the possibility of the SUMOylation-mediated modulation of Aβ levels. Site-directed mutagenesis studies of APP SUMOylation sites evince that the deSUMOylation of APP promotes Aβ levels. Over-expressing genes encoding enzymes of SUMOylation pathways ensure raised APP SUMOylation and decreased Aβ aggregates [89,90]. In contrast, SUMOylation positively modulates the expression of APP-cleaving protease β-secretase-encoding gene [89]. This results in increased levels of Aβ aggregates [91]. These paradoxical findings enhance the understating of SUMOylation-mediated pathological changes in AD. A recently published in vitro study on cultured cortical neurons demonstrated that Aβ1-42 peptide downregulates the levels of SUMO-activating enzyme SAE2 and the SUMO E3 ligase PIAS1/2. This results in decreased levels of SUMO1 or SUMO2/3-conjugated proteins [92]. Moreover, the impact of SUMOylation on APP trafficking remains unexplored. The presence of phosphorylated Tau protein is associated with AD pathology. Biochemical studies revealed that the Tau protein lysine K340 shared the SUMOylation and ubiquitination modifications. SUMOylation at K340 blocks ubiquitin binding to Tau and, thereby, its proteasomal degradation [93]. The activity-based signal determines the balance between SUMOylation and ubiquitination, and thus, the fate of the Tau protein. The histopathology of AD mice revealed that SUMO1 colocalized with phosphorylated Tau aggregates [93]. Furthermore, increased SUMO1 immunoreactivity was detected in Tau aggregates in AD patients’ cerebral cortex [94]. Pharmacologically inhibiting proteasomal degradation manifests Tau SUMOylation, causing Tau phosphorylation and tangle formation. The results demonstrate that the SUMOylation of Tau protein endorses Tau hyperphosphorylation. It leads to reduced microtubule stability and neuronal dysfunction [94]. This is the preliminary evidence depicting the critical role of SUMOylation in the onset and progression of AD in humans and rodents. There are still several open questions concerning the role of SUMOylation in glia and astrocytes, which are known to modulate neuronal activity and can contribute to the progression of AD. Few studies indicate that SUMOylation is involved in redox imbalance in astrocytes. This redox imbalance may cause neuroinflammation and pathophysiological symptoms associated with AD [95]. The role of SUMOylation in the glia-mediated modulation of neuronal dysfunction is an emerging concept and needs further investigation. Figure 2 shows an overview of the different mechanisms by which SUMOylation plays a role in AD.

4. Role of SUMOylation in Parkinson’s Disease (PD)

PD is a multifactorial neurodegenerative disease. It is manifested by the progressive loss of dopaminergic neurons projecting from the substantia nigra compacta to the striatum. PD-associated neurological disorders include motor and nonmotor symptoms. The early pathological symptoms are rapid eye movement, sleep behavioral disorder and decreased smell. The progression of the disease is associated with the onset of cognitive impairment and hallucinations. The hallmark of PD progression is motor dysfunction. It includes tremors, rigidity, bradykinesia, gait stiffness, hypokinesia and imbalance [96,97,98]. PD prevalence is increasing rapidly. In 2016 there were 6.1 million PD patients globally. Currently, this number has increased to more than 10 million. The incidence of PD increases with age, peaking between 85 and 89 years. However, 4% of people with PD are diagnosed before 50. PD is 1.4 times more prevalent in males than females [99,100].
PD is pathological and primarily marked by the cytoplasmic deposition of Lewy bodies, a neuronal inclusion consisting of α-synuclein protein aggregations. The intracellular aggregation of α-synuclein causes decreased membrane potential. Furthermore, it results in oxidative stress, excitotoxicity, neuroinflammation and finally, apoptosis of the neuron [96]. The sequence of molecular alterations is not well delineated. However, it may lead to dysregulation of DJ-1 (protein deglycase DJ-1, encoded by PARK7 gene) transcriptional factors, an impaired ubiquitin-proteasome system, aberrant splicing events, mitochondrial dysfunction and neuronal oxidative stress. These could potentially lead to the deposition of Lewy bodies in neurons. Lewy bodies are the eosinophilic cytoplasmic inclusions of 5-30 µm spherical masses with a dense core surrounded by radiating fibrils. Lewy bodies are strongly stained for α-synuclein protein [101]. In addition, Lewy bodies consist of neurofilament protein, ubiquitin-binding protein p62, tubulin and synphilin-1. The abundance of α-synuclein in Lewy bodies establishes its importance in the pathogenesis of PD. α-Synuclein is a 140 amino acid protein abundant in presynaptic nerve terminals. In a monomeric state, it is involved in the vesicular transport and storage, release and recycling of neurotransmitters [102]. SCNA gene encodes the α-synuclein protein. Autosomal dominant missense mutations in the SNCA gene lead to the early onset of PD [103]. In addition, SCNA gene multiplication (duplication and triplication of SCNA gene) increases the dose of wildtype proteins and can cause autosomal dominant parkinsonism. A significant number of genetic, transgenic and viral-based studies in rodents and primates, using either wildtype or mutant α-synuclein protein, clearly demonstrate an α-synuclein association with the progressive degeneration of neurons, and thus its crucial role in the pathogenesis of PD. α-Synuclein oligomerization promotes its binding to the mitochondrial outer membrane translocase (TOM20) and the voltage-dependent anion channel (VDAC). It results in the inhibition of the mitochondrial import system [104]. This eventually leads to disrupted mitochondrial–ER interaction, decreased mitochondrial ATP production, mitochondrial swelling and depolarization, consequently causing oxidative stress and accelerated cytochrome C release in neurons and, lastly, neuronal apoptosis [105]. α-Synuclein is colocalized with ubiquitin and 20S proteasome in Lewy bodies. The direct interaction of α-synuclein and 20S proteasome causes the inhibition of the chymotrypsin-like proteasome and a dysfunctional ubiquitin–proteasome system (UPS) [106]. Compromised UPS results in the accumulation of misfolded proteins and builds toxic aggregates [107,108]. NDDs begin with the alteration at synaptic transmission. Wildtype α-synuclein promotes SNARE-complex assembly. SNARE (soluble N-ethylmaleimide-sensitive factor attachment protein receptor) mediates membrane fusion at synaptic terminals. SNARE proteins are present on vesicles (v-SNARE) or target membranes (t-SNARE). The C-terminus of α-synuclein binds to VAMP2 (vesicle-associated membrane protein 2) and promotes SNARE complex formation. α-synuclein overexpression or aggregates inhibit v-SNARE binding to t-SNARE, which impairs neurotransmitter release and thus leads to synaptic dysfunction [109,110]. α-synuclein binds to microtubules’ tubulin α2β2 tetramer. It stabilizes the α-synuclein helical structure, which promotes microtubule nucleation and growth. Mutant α-synuclein does not undergo tubulin-induced structural change. It results in tubulin aggregation, rather than polymerization [111,112]. Inhibited axonal transport and synaptic dysfunction further drivie neuronal degeneration [113,114].
α-Synuclein post-translational modification (PTM) may influence its toxicity and aggregation. Phosphorylation, nitration, acetylation, ubiquitination, O-GlcNAcylation and SUMOylation extensively modify α-synuclein [115,116,117,118]. The PTM of α-synuclein modifies its membrane binding, protein interaction, solubility and aggregation properties. PD pathogenicity is linked to α-synuclein phosphorylation at serine 87 and 129 [116,119]. In contrast, SUMOylation prevents α-synuclein toxicity. α-Synuclein is SUMOylated at multiple lysine residues. Among them, lysine 96 and 102 were shown to be critical in regulating α-synuclein toxicity [85,120]. Substitutions of lysine 96 and 102 to arginine impaired α-synuclein SUMOylation and reduced its solubility. This results in enhanced aggregation and toxicity in HEK cells and the substantia nigra neurons in a rat PD model [85]. Similar observations were made in transgenic mice expressing His-tagged SUMO2 [85]. SUMOylation preferentially and effectively occurs on α-synuclein lysine102 residue. In vitro studies have demonstrated that the SUMOylation of only 10% of α-synuclein delayed its aggregation [85]. α-Synuclein contains two SIM motifs located between the amino acids 33 and 58. The synthetic SUMO1-derived fragment SUMO1(15-55) can bind to both SIMs, and it prefers SIM-binding defective mutants that only possess one SIM. SUMO1(15-55) inhibits α-synuclein oligomerization by modulating the monomeric state and preventing aggregation in SH-SY5Y neuronal cells. It was also shown that the expression of SUMO1(15-55) in a transgenic α-synuclein Drosophila model exerts neuroprotective effects [121]. Lewy bodies are also associated with the parkin protein (encoded by the PARK2 gene) [122]. Mutations in PARK2 are the cause of autosomal recessive juvenile parkinsonism [123]. Parkin is a ubiquitin E3 ligase that regulates UPS and lysosomal degradation pathways. It plays a crucial role in degrading misfolded proteins, clearing reactive oxygen species, mitophagy, and promoting cellular survival [124]. Parkin directly and selectively non-covalently interacts with SUMO1 in vitro and in vivo. The SUMOylation of parkin promotes its nuclear transport and self-ubiquitination, which causes a loss of its E3 ligase activity and decreases levels in the cytoplasm. In contrast, SUMOylation promotes parkin transcriptional factor function, resulting in the expression of neuroprotective genes [125]. Neurons deprived of parkin protein are susceptible to oxidative stress, mitochondrial dysfunction and impaired proteasome-mediated protein degradation. Thus, parkin is critical for cellular function and viability [126]. In addition to parkin, the DJ-1 (protein deglycase DJ-1, encoded by PARK7 gene) protein contributes to detecting and modulating neuronal oxidative stress. It functions as a regulator of mitochondrial biogenesis and a stress regulator. Mutations in DJ-1 are identified in PD patients [127,128]. The Leucine166 Proline substitution causes aberrant SUMOylation in vitro, and in vivo studies identified that DJ-1 is SUMOylated at lysine 130 residue [129]. The SUMOylation of DJ-1 promotes its activity, and thus its ability to affect oxidative stress, mitochondrial function and cell survival [129,130].
It could be hypothesized that the SUMOylation of multiple key proteins involved in mitochondrial biogenesis reinforces cell survival, whereas deSUMOylation causes PD pathogenesis. Figure 3 depicts how SUMOylation can affect Parkinson’s disease.

5. Role of SUMOylation in Huntington’s Disease (HD)

Huntington’s disease is a rare, incurable, neurodegenerative, monogenetic, dominantly inherited disease. It is a polyglutamine (poly Q) disorder of Huntingtin protein (HTT). It is caused by the expansion of a CAG repeat (encoding glutamine) at the exon 1 of the Huntingtin (HTT) gene [131,132,133]. The onset and progression of HD are associated with the presence of more than 36 CAG triplet repeats encoding polyQ tract in the HTT protein. Patient data showed that 40–50 repeats cause the onset of disease in adults, whereas 50–120 repeats lead to juvenile HD [133,134,135,136]. The HTT gene encodes the 348 kDa Huntingtin (HTT) protein found in the cytoplasm and nucleus of neurons. The HTT protein plays a critical role in microtubule-based axon trafficking, which involves nuclear–cytoplasmic protein trafficking and antero- and retrograde axonal transport [137,138]. The HTT competence of bidirectional switching between antero/retrograde axonal transport is phosphorylation-dependent. The phosphorylation of Serine 421 residue of the Huntingtin protein promotes anterograde transport, whereas nonphosphorylated Huntingtin is likely to undergo retrograde transport [139]. In addition, HTT mitigates mitochondria transport along neurites [140]. Homozygous HTT mice are embryonically lethal [141,142]. Conditional HTT knockout mice showed impaired vesicular and mitochondrial trafficking in both directions, and progressive brain degeneration [143]. The HTT gene with the abnormally high numbers of CAG triplet repeats (polyQ) is termed mutant HTT (mHTT). mHTT mitigates the HD disease pathology mainly in striatal medium-sized spiny neurons, cortical pyramidal neurons and thalamic neurons. However, mHTT is susceptible to aggregation, which causes neuronal dysfunction and death [144,145]. HD is an incurable neurodegenerative disease that usually appears in a subject’s 40s. HD is associated with motor, cognitive and psychiatric disorders [146,147,148,149].
The motor symptoms include involuntary movements, impaired voluntary movements, speech difficulties and unusual eye movements, whereas cognitive symptoms include learning deficit and lack of impulse control. The neuropsychiatric symptoms include depression, insomnia, frequent fatigue and suicidal tendencies. Several post-translational modifications modify Huntingtin’s protein. Ubiquitination and SUMOylation antagonistically modify the same lysine residue [108,150,151,152,153]. The ubiquitination of HTT causes its proteasome-mediated degradation, whereas the SUMOylation of the identical lysine residues protects it from degradation. SUMOylated HTT may be responsible for toxic accumulation. In contrast, the SUMOylation of the protein fragment encoded by exon 1 of the HTT gene (HTTex1) has a paradoxical effect. It leads to a reduced ability to form aggregates and increased solubility, and promotes its ability to repress transcription [154]. In addition, the SUMOylation of HTTex1 inhibits its interaction with lipids mediated by the lipid-binding domain to prevent aggregation [155]. SUMO E3 ligase PIAS1 mediates the SUMOylation of HTT. The downregulation of PIAS1 in the striatum reduces HTT SUMOylation and improves motor impairment independent of mutant HTT presence [156,157]. PIAS1, through interaction with HTT, is also part of the transcription-coupled repair complex (TCR) and polynucleotide kinase 3′-phosphatase (PNKP). PIAS1 can modulate the activity of PNKP in the mouse brain as well as in neurons derived from HD patients [156]. Rhes (Ras-homolog enriched in the striatum) is a multifunction protein. In striatal neurons, Rhes acts as a SUMO E3 ligase, and mitigates the SUMOylation of the mHTT protein and associated cytotoxicity [66,153]. Rhes can transit from neuron to neuron and facilitate the spreading of mHTT [158]. Rhes knockout mice are neuroprotective, indicating that Rhes-mediated SUMOylation is a critical event in mHTT-driven neuronal pathology [159,160,161,162]. mHtt also impairs mitochondrial function [163,164]. A rodent model of HD and human brains affected by HD demonstrated aggregated mHTT proteins in mitochondria [159,165]. Biochemical experiments provided evidence that mHTT interacts with the mitochondrial translocase of inner membrane 23 (TIM23), and mitochondrial fission proteins dynamin-related protein 1 (DRP1) and fission 1 (FIS1) [165,166,167,168,169]. mHTT–TIM23 interaction resulted in a mitochondrial protein import defect in in vitro and in vivo studies [165,168,170]. Moreover, mHTT’s interaction with DRP1/FIS1 promotes mitochondrial fission via the increased expression of mitochondrial fission genes DRP1 and FIS1. In addition, mHTT–DRP1 interaction is also associated with the increased enzymatic activity of GTPase DRP1 [166].
Collectively, mHTT directly contributes to mitochondrial dysfunction and neuronal death. However, SUMOylation has a dual effect on mHTT-mediated effects. Depending upon the SUMO paralog attached to mHTT, it could either improve the cellular clearance of mHTT or enhance its toxicity by stabilizing the aggregates. Thus, SUMOylation plays a critical role in the pathophysiology of HD. A summary of the role of SUMOylation can be seen in Figure 4.

6. Role of SUMOylation in Diabetic Peripheral Neuropathy (DPN)

Diabetic peripheral neuropathy is a chronic complication caused by prolonged hyperglycemia in type 1 and 2 diabetes. It is a slowly progressing, inevitable diabetic complication. More than 50% of patients who have diabetes develop neuropathy, and it is diagnosed late [171,172]. Clinically, DPN is characterized by lower limb complications such as foot ulcers, paresthesia, numbness and neuropathic pain [173]. Depending upon the affected neuron type, it could be sensory, motor or autonomic neuropathy. In addition, DPN leads to axonal degeneration, a reduction in intraepidermal nerve fibers, diminished conduction velocities, and demyelination [36,174,175,176,177]. These symptoms may be sporadic or constant, and can be associated with painful or painless DPN.
Typically, glucose is metabolized through glycolysis to pyruvate. Pyruvate then undergoes oxidative decarboxylation to produce acetyl coenzyme in mitochondria. Acetyl coenzyme is further oxidized to carbon dioxide and water through the tricarboxylic acid cycle. The glucose breakdown yields 4 ATP and 12 molecules of NADH. The generated NADH combines with oxygen by passing electrons through the electron transport chain to form 34 ATP molecules [178]. Under hyperglycemic conditions, the excess glucose is shuttled to the toxic aldose reductase polyol pathway, protein kinase C activation, and generates toxic secondary metabolites and advanced glycation end products (AGEs) [36,179,180,181]. The PTMs, such as ubiquitination, phosphorylation, acetylation, and ribosylation, were shown to regulate the glycolytic enzymes involved in complex glucose metabolism [182]. For example, ubiquitination inhibits biphosphate aldolase (ALDOA) and glyceraldehyde-3-phosphate dehydrogenase (GAPDH) levels and activity. The deubiquitination of glycolytic enzymes stabilizes them and promotes glycolysis [183]. Similarly, the acetylation of lysine residues of the glycolytic enzyme blocks substrate binding and impairs metabolic pathways [184]. ]. In contrast, the role of SUMOylation is not well-defined. Only a few studies have attempted to delineate the role of SUMOylation in DPN. The LC-MS-based screening in dorsal root ganglia (DRG) neurons identified several critical enzymes in the glycolysis and Krebs cycle as targets for SUMOylation [36]. The activities of GAPH, citrate synthase, transketolase-like protein-1 (TKTL1) and isocitrate dehydrogenase 2 (IDH2) were shown to be modulated by SUMOylation [185]. In addition, detoxifying enzymes such as aldehyde dehydrogenase isoform α-3A1 (ALDH3A1) are SUMOylation targets [36]. The human sciatic nerve analysis showed that prolonged diabetes causes reduced levels of SUMOylated GAPDH and citrate synthase enzymes involved in glucose metabolism. These findings illustrate the importance of SUMOylation as a neuron-protective mechanism in DPN. Similarly, Hou et al. depicted the role of SUMOE3 ligases in the development of DPN. PIAS1 mediates Peroxisome Proliferator-Activated Receptor γ (PPAR-g) SUMOylation, and stabilizes it. This results in miR-124 upregulation, which inhibits STAT3 (Signal transducer and activator of transcription 3) activity and thus inhibits Schwann cells’ apoptosis and the progression of DPN [186]. In type 1 or 2 diabetes, prolonged hyperglycemia downregulates SUMOylation in neurons and Schwann cells [186]. These findings indicate that the SUMOylation of multiple proteins in several metabolic pathways is essential for endogenous protection from metabolic damage. Tissue-specific genetic ablation studies in rodents deleting the UBC9 gene abolish the SUMOylation of proteins in dorsal root ganglia neurons. These neurons were shown to be more susceptible to hyperglycemia-induced damage. Thus, these mutant mice demonstrate expeditious peripheral neuropathic pathological symptoms. These findings demonstrate that SUMOylation is a critical protective mechanism, the loss of which promotes hyperglycemia-induced tissue damage.
In addition, different studies have identified several ion channels, such as sodium channels 1.7 and 1.2 (Nav1.7 and Nav1.2), Transient receptor potential V1 (TRPV1), two-pore potassium channel (K2P1), and multiple voltage-gated potassium channels (Kv1.5, Kv2.1, Kv4.2, Kv7.1, Kv7.2 and Kv11.1), as SUMOylation targets [187,188,189,190,191]. Among these, TRP channels are the most studied ion channels under the pathological condition of DPN. TRPV1 is a cation channel and acts as a thermosensor. It is known to be activated by capsaicin, low pH and noxious heat > 42 °C [192]. In animal models for diabetic neuropathy, TRPV1 levels and function reduction were reported, and linked to the DPN pathophysiology [193]. Recently published studies reported TRPV1 SUMOylation at lysine 324 [36]. Electrophysiological recording and calcium imaging data elucidate the role of SUMOylation in TRPV1 channel function modulation in in vitro and in vivo conditions. In diabetic patient nerve samples, the levels of sumoylated-TRPV1 were shown to be diminished. En masse, SUMOylation increases neuronal excitability, but the differential regulation of individual ion channels under diabetic conditions has not yet been studied.
Prolonged chronic hyperglycemia is associated with excessive cellular oxidative stress and impaired mitochondrial function [194]. In vitro studies on cultured DRG neurons exposed to elevated levels of glucose exhibit increased levels of reactive oxygen free radicals (ROS) and hydrogen peroxide (H2O2) [174]. ROS/H2O2 attack the iron–sulfur centers of enzymes involved in glucose metabolism, such as complex I–III in mitochondria, and consequently inhibit their function [195]. This leads to bioenergetic crises, decreased biological activity, and impaired cellular signaling and transport. All these together cause neuronal dysfunction and neurodegeneration. SUMOylation and oxidative stress are bidirectional regulatory mechanisms. SUMOylation activates a signal cascade and transcriptional factors to increase detoxifying protein levels, inhibiting oxidative stress. In beta-pancreatic tissue, oxidative stress activates transcription factor NF-κB via the increased phosphorylation and subsequent degradation of IkB. Similarly, oxidative stress activates the JNK/c-Jun and Maf/Nrf2 pathways in different tissue types [196]. Prolonged chronic hyperglycemia generates H2O2, which transiently inactivates SUMO E1 and E2 enzymes. It induces the formation of a disulfide bond between their catalytic cysteines [197], which may lead to reduced levels of SUMOylated proteins in the neurons. It is hypothesized that enhanced oxidative stress impaired the SUMOylation-mediated cellular protective mechanism, which could lead to peripheral neuron dysfunction and degeneration. Although a more detailed analysis is required at different levels of disease progression, as yet unknown regulation mechanisms via SUMOylation should be explored as new drug targets to impede the progression of DPN. Figure 5 shows how SUMOylation can modulate DPN.

Author Contributions

Conceptualization, N.M. and N.A.; writing—original draft preparation, all the authors; writing—review and editing, N.M. and N.A. All authors have read and agreed to the published version of the manuscript.

Funding

This project was funded by the German Research Foundation (Deutsche Forschungsgemeinschaft (DFG)), Sonderforschungsbereich SFB1118 project B06.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Erkkinen, M.G.; Kim, M.-O.; Geschwind, M.D. Clinical Neurology and Epidemiology of the Major Neurodegenerative Diseases. Cold Spring Harb. Perspect. Biol. 2017, 10, a033118. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Gardner, R.C.; Yaffe, K. Epidemiology of mild traumatic brain injury and neurodegenerative disease. Mol. Cell. Neurosci. 2015, 66, 75–80. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Cicero, C.E.; Mostile, G.; Vasta, R.; Rapisarda, V.; Signorelli, S.S.; Ferrante, M.; Zappia, M.; Nicoletti, A. Metals and neurodegenerative diseases. A systematic review. Environ. Res. 2017, 159, 82–94. [Google Scholar] [CrossRef]
  4. Dugger, B.N.; Dickson, D.W. Pathology of Neurodegenerative Diseases. Cold Spring Harb. Perspect. Biol. 2017, 9, a028035. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Gan, L.; Cookson, M.R.; Petrucelli, L.; La Spada, A.R. Converging pathways in neurodegeneration, from genetics to mechanisms. Nat. Neurosci. 2018, 21, 1300–1309. [Google Scholar] [CrossRef] [PubMed]
  6. Prince, M.; Bryce, R.; Albanese, E.; Wimo, A.; Ribeiro, W.; Ferri, C.P. The global prevalence of dementia: A systematic review and metaanalysis. Alzheimers Dement. 2013, 9, 63–75.e2. [Google Scholar] [CrossRef] [PubMed]
  7. Hurd, M.D.; Martorell, P.; Delavande, A.; Mullen, K.J.; Langa, K.M. Monetary costs of dementia in the United States. N. Engl. J. Med. 2013, 368, 1326–1334. [Google Scholar] [CrossRef] [Green Version]
  8. Soto, C.; Pritzkow, S. Protein misfolding, aggregation, and conformational strains in neurodegenerative diseases. Nat. Neurosci. 2018, 21, 1332–1340. [Google Scholar] [CrossRef]
  9. Golpich, M.; Amini, E.; Mohamed, Z.; Azman Ali, R.; Mohamed Ibrahim, N.; Ahmadiani, A. Mitochondrial Dysfunction and Biogenesis in Neurodegenerative diseases: Pathogenesis and Treatment. CNS Neurosci. Ther. 2017, 23, 5–22. [Google Scholar] [CrossRef] [PubMed]
  10. Singh, E.; Devasahayam, G. Neurodegeneration by oxidative stress: A review on prospective use of small molecules for neuroprotection. Mol. Biol. Rep. 2020, 47, 3133–3140. [Google Scholar] [CrossRef] [PubMed]
  11. Uttara, B.; Singh, A.V.; Zamboni, P.; Mahajan, R.T. Oxidative stress and neurodegenerative diseases: A review of upstream and downstream antioxidant therapeutic options. Curr. Neuropharmacol. 2009, 7, 65–74. [Google Scholar] [CrossRef] [Green Version]
  12. Lewerenz, J.; Maher, P. Chronic Glutamate Toxicity in Neurodegenerative Diseases-What is the Evidence? Front. Neurosci. 2015, 9, 469. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Didonna, A.; Benetti, F. Post-translational modifications in neurodegeneration. AIMS Biophys. 2015, 3, 27–49. [Google Scholar] [CrossRef]
  14. Gupta, R.; Sahu, M.; Srivastava, D.; Tiwari, S.; Ambasta, R.K.; Kumar, P. Post-translational modifications: Regulators of neurodegenerative proteinopathies. Ageing Res. Rev. 2021, 68, 101336. [Google Scholar] [CrossRef] [PubMed]
  15. Schaffert, L.-N.; Carter, W.G. Do Post-Translational Modifications Influence Protein Aggregation in Neurodegenerative Diseases: A Systematic Review. Brain Sci. 2020, 10, 232. [Google Scholar] [CrossRef] [PubMed]
  16. Chen, L.; Kashina, A. Post-translational Modifications of the Protein Termini. Front. Cell Dev. Biol. 2021, 9, 719590. [Google Scholar] [CrossRef] [PubMed]
  17. Conibear, A.C. Deciphering protein post-translational modifications using chemical biology tools. Nat. Rev. Chem. 2020, 4, 674–695. [Google Scholar] [CrossRef]
  18. Leutert, M.; Entwisle, S.W.; Villén, J. Decoding Post-Translational Modification Crosstalk with Proteomics. Mol. Cell. Proteom. 2021, 20, 100129. [Google Scholar] [CrossRef]
  19. Meluh, P.B.; Koshland, D. Evidence that the MIF2 gene of Saccharomyces cerevisiae encodes a centromere protein with homology to the mammalian centromere protein CENP-C. Mol. Biol. Cell 1995, 6, 793–807. [Google Scholar] [CrossRef] [PubMed]
  20. Shen, Z.; Pardington-Purtymun, P.E.; Comeaux, J.C.; Moyzis, R.K.; Chen, D.J. UBL1, a human ubiquitin-like protein associating with human RAD51/RAD52 proteins. Genomics 1996, 36, 271–279. [Google Scholar] [CrossRef]
  21. van der Veen, A.G.; Ploegh, H.L. Ubiquitin-like proteins. Annu. Rev. Biochem. 2012, 81, 323–357. [Google Scholar] [CrossRef] [PubMed]
  22. Flotho, A.; Melchior, F. Sumoylation: A regulatory protein modification in health and disease. Annu. Rev. Biochem. 2013, 82, 357–385. [Google Scholar] [CrossRef] [PubMed]
  23. Tatham, M.H.; Jaffray, E.; Vaughan, O.A.; Desterro, J.M.; Botting, C.H.; Naismith, J.H.; Hay, R.T. Polymeric chains of SUMO-2 and SUMO-3 are conjugated to protein substrates by SAE1/SAE2 and Ubc9. J. Biol. Chem. 2001, 276, 35368–35374. [Google Scholar] [CrossRef] [Green Version]
  24. Saitoh, H.; Hinchey, J. Functional heterogeneity of small ubiquitin-related protein modifiers SUMO-1 versus SUMO-2/3. J. Biol. Chem. 2000, 275, 6252–6258. [Google Scholar] [CrossRef] [Green Version]
  25. Guo, D.; Li, M.; Zhang, Y.; Yang, P.; Eckenrode, S.; Hopkins, D.; Zheng, W.; Purohit, S.; Podolsky, R.H.; Muir, A.; et al. A functional variant of SUMO4, a new I kappa B alpha modifier, is associated with type 1 diabetes. Nat. Genet. 2004, 36, 837–841. [Google Scholar] [CrossRef]
  26. Owerbach, D.; McKay, E.M.; Yeh, E.T.H.; Gabbay, K.H.; Bohren, K.M. A proline-90 residue unique to SUMO-4 prevents maturation and sumoylation. Biochem. Biophys. Res. Commun. 2005, 337, 517–520. [Google Scholar] [CrossRef]
  27. Wei, W.; Yang, P.; Pang, J.; Zhang, S.; Wang, Y.; Wang, M.-H.; Dong, Z.; She, J.-X.; Wang, C.-Y. A stress-dependent SUMO4 sumoylation of its substrate proteins. Biochem. Biophys. Res. Commun. 2008, 375, 454–459. [Google Scholar] [CrossRef]
  28. Han, Z.-J.; Feng, Y.-H.; Gu, B.-H.; Li, Y.-M.; Chen, H. The post-translational modification, SUMOylation, and cancer (Review). Int. J. Oncol. 2018, 52, 1081–1094. [Google Scholar] [CrossRef] [Green Version]
  29. Baczyk, D.; Audette, M.C.; Drewlo, S.; Levytska, K.; Kingdom, J.C. SUMO-4: A novel functional candidate in the human placental protein SUMOylation machinery. PLoS ONE 2017, 12, e0178056. [Google Scholar] [CrossRef] [Green Version]
  30. Liang, Y.-C.; Lee, C.-C.; Yao, Y.-L.; Lai, C.-C.; Schmitz, M.L.; Yang, W.-M. SUMO5, a Novel Poly-SUMO Isoform, Regulates PML Nuclear Bodies. Sci. Rep. 2016, 6, 26509. [Google Scholar] [CrossRef] [Green Version]
  31. Celen, A.B.; Sahin, U. Sumoylation on its 25th anniversary: Mechanisms, pathology, and emerging concepts. FEBS J. 2020, 287, 3110–3140. [Google Scholar] [CrossRef]
  32. Johnson, E.S. Protein modification by SUMO. Annu. Rev. Biochem. 2004, 73, 355–382. [Google Scholar] [CrossRef] [Green Version]
  33. Yang, Y.; He, Y.; Wang, X.; Liang, Z.; He, G.; Zhang, P.; Zhu, H.; Xu, N.; Liang, S. Protein SUMOylation modification and its associations with disease. Open Biol. 2017, 7, 170167. [Google Scholar] [CrossRef]
  34. Wilkinson, K.A.; Henley, J.M. Mechanisms, regulation and consequences of protein SUMOylation. Biochem. J. 2010, 428, 133–145. [Google Scholar] [CrossRef] [Green Version]
  35. Cremona, C.A.; Sarangi, P.; Yang, Y.; Hang, L.E.; Rahman, S.; Zhao, X. Extensive DNA damage-induced sumoylation contributes to replication and repair and acts in addition to the mec1 checkpoint. Mol. Cell 2012, 45, 422–432. [Google Scholar] [CrossRef] [Green Version]
  36. Agarwal, N.; Taberner, F.J.; Rangel Rojas, D.; Moroni, M.; Omberbasic, D.; Njoo, C.; Andrieux, A.; Gupta, P.; Bali, K.K.; Herpel, E.; et al. SUMOylation of Enzymes and Ion Channels in Sensory Neurons Protects against Metabolic Dysfunction, Neuropathy, and Sensory Loss in Diabetes. Neuron 2020, 107, 1141–1159.e7. [Google Scholar] [CrossRef]
  37. Pichler, A.; Fatouros, C.; Lee, H.; Eisenhardt, N. SUMO conjugation—A mechanistic view. Biomol. Concepts 2017, 8, 13–36. [Google Scholar] [CrossRef]
  38. Yasugi, T.; Howley, P.M. Identification of the structural and functional human homolog of the yeast ubiquitin conjugating enzyme UBC9. Nucleic Acids Res. 1996, 24, 2005–2010. [Google Scholar] [CrossRef] [Green Version]
  39. Su, Y.-F.; Yang, T.; Huang, H.; Liu, L.F.; Hwang, J. Phosphorylation of Ubc9 by Cdk1 enhances SUMOylation activity. PLoS ONE 2012, 7, e34250. [Google Scholar] [CrossRef] [Green Version]
  40. Nacerddine, K.; Lehembre, F.; Bhaumik, M.; Artus, J.; Cohen-Tannoudji, M.; Babinet, C.; Pandolfi, P.P.; Dejean, A. The SUMO pathway is essential for nuclear integrity and chromosome segregation in mice. Dev. Cell 2005, 9, 769–779. [Google Scholar] [CrossRef]
  41. Chu, Y.; Yang, X. SUMO E3 ligase activity of TRIM proteins. Oncogene 2011, 30, 1108–1116. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Hochstrasser, M. SP-RING for SUMO. Cell 2001, 107, 5–8. [Google Scholar] [CrossRef] [Green Version]
  43. Song, J.; Durrin, L.K.; Wilkinson, T.A.; Krontiris, T.G.; Chen, Y. Identification of a SUMO-binding motif that recognizes SUMO-modified proteins. Proc. Natl. Acad. Sci. USA 2004, 101, 14373–14378. [Google Scholar] [CrossRef] [Green Version]
  44. Seu, C.S.; Chen, Y. Identification of SUMO-binding motifs by NMR. Methods Mol. Biol. 2009, 497, 121–138. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Kahyo, T.; Nishida, T.; Yasuda, H. Involvement of PIAS1 in the sumoylation of tumor suppressor p53. Mol. Cell 2001, 8, 713–718. [Google Scholar] [CrossRef]
  46. Kagey, M.H.; Melhuish, T.A.; Wotton, D. The Polycomb Protein Pc2 Is a SUMO E3. Cell 2003, 113, 127–137. [Google Scholar] [CrossRef] [Green Version]
  47. Pichler, A.; Gast, A.; Seeler, J.S.; Dejean, A.; Melchior, F. The Nucleoporin RanBP2 Has SUMO1 E3 Ligase Activity. Cell 2002, 108, 109–120. [Google Scholar] [CrossRef] [Green Version]
  48. Eisenhardt, N.; Chaugule, V.K.; Koidl, S.; Droescher, M.; Dogan, E.; Rettich, J.; Sutinen, P.; Imanishi, S.Y.; Hofmann, K.; Palvimo, J.J.; et al. A new vertebrate SUMO enzyme family reveals insights into SUMO-chain assembly. Nat. Struct. Mol. Biol. 2015, 22, 959–967. [Google Scholar] [CrossRef]
  49. Guervilly, J.-H.; Takedachi, A.; Naim, V.; Scaglione, S.; Chawhan, C.; Lovera, Y.; Despras, E.; Kuraoka, I.; Kannouche, P.; Rosselli, F.; et al. The SLX4 complex is a SUMO E3 ligase that impacts on replication stress outcome and genome stability. Mol. Cell 2015, 57, 123–137. [Google Scholar] [CrossRef] [Green Version]
  50. Sentis, S.; Le Romancer, M.; Bianchin, C.; Rostan, M.-C.; Corbo, L. Sumoylation of the estrogen receptor alpha hinge region regulates its transcriptional activity. Mol. Endocrinol. 2005, 19, 2671–2684. [Google Scholar] [CrossRef]
  51. Kotaja, N.; Karvonen, U.; Jänne, O.A.; Palvimo, J.J. PIAS proteins modulate transcription factors by functioning as SUMO-1 ligases. Mol. Cell. Biol. 2002, 22, 5222–5234. [Google Scholar] [CrossRef] [Green Version]
  52. Rosas-Acosta, G.; Langereis, M.A.; Deyrieux, A.; van Wilson, G. Proteins of the PIAS family enhance the sumoylation of the papillomavirus E1 protein. Virology 2005, 331, 190–203. [Google Scholar] [CrossRef] [Green Version]
  53. Wong, K.A.; Kim, R.; Christofk, H.; Gao, J.; Lawson, G.; Wu, H. Protein inhibitor of activated STAT Y (PIASy) and a splice variant lacking exon 6 enhance sumoylation but are not essential for embryogenesis and adult life. Mol. Cell. Biol. 2004, 24, 5577–5586. [Google Scholar] [CrossRef] [Green Version]
  54. Potts, P.R.; Yu, H. Human MMS21/NSE2 is a SUMO ligase required for DNA repair. Mol. Cell. Biol. 2005, 25, 7021–7032. [Google Scholar] [CrossRef] [Green Version]
  55. Moreno-Ayala, R.; Schnabel, D.; Salas-Vidal, E.; Lomelí, H. PIAS-like protein Zimp7 is required for the restriction of the zebrafish organizer and mesoderm development. Dev. Biol. 2015, 403, 89–100. [Google Scholar] [CrossRef] [Green Version]
  56. Tago, K.; Chiocca, S.; Sherr, C.J. Sumoylation induced by the Arf tumor suppressor: A p53-independent function. Proc. Natl. Acad. Sci. USA 2005, 102, 7689–7694. [Google Scholar] [CrossRef] [Green Version]
  57. Colomer-Lluch, M.; Serra-Moreno, R. BCA2/Rabring7 Interferes with HIV-1 Proviral Transcription by Enhancing the SUMOylation of IκBα. J. Virol. 2017, 91, e02098-16. [Google Scholar] [CrossRef] [Green Version]
  58. Zhu, G.; Harischandra, D.S.; Ghaisas, S.; Zhang, P.; Prall, W.; Huang, L.; Maghames, C.; Guo, L.; Luna, E.; Mack, K.L.; et al. TRIM11 Prevents and Reverses Protein Aggregation and Rescues a Mouse Model of Parkinson’s Disease. Cell Rep. 2020, 33, 108418. [Google Scholar] [CrossRef]
  59. Yamashita, D.; Moriuchi, T.; Osumi, T.; Hirose, F. Transcription Factor hDREF Is a Novel SUMO E3 Ligase of Mi2α. J. Biol. Chem. 2016, 291, 11619–11634. [Google Scholar] [CrossRef] [Green Version]
  60. Zhao, X.; Sternsdorf, T.; Bolger, T.A.; Evans, R.M.; Yao, T.-P. Regulation of MEF2 by histone deacetylase 4- and SIRT1 deacetylase-mediated lysine modifications. Mol. Cell. Biol. 2005, 25, 8456–8464. [Google Scholar] [CrossRef]
  61. Grégoire, S.; Yang, X.-J. Association with class IIa histone deacetylases upregulates the sumoylation of MEF2 transcription factors. Mol. Cell. Biol. 2005, 25, 2273–2287. [Google Scholar] [CrossRef] [Green Version]
  62. García-Gutiérrez, P.; Juárez-Vicente, F.; Gallardo-Chamizo, F.; Charnay, P.; García-Domínguez, M. The transcription factor Krox20 is an E3 ligase that sumoylates its Nab coregulators. EMBO Rep. 2011, 12, 1018–1023. [Google Scholar] [CrossRef] [Green Version]
  63. Braschi, E.; Zunino, R.; McBride, H.M. MAPL is a new mitochondrial SUMO E3 ligase that regulates mitochondrial fission. EMBO Rep. 2009, 10, 748–754. [Google Scholar] [CrossRef]
  64. Stindt, M.H.; Carter, S.; Vigneron, A.M.; Ryan, K.M.; Vousden, K.H. MDM2 promotes SUMO-2/3 modification of p53 to modulate transcriptional activity. Cell Cycle 2011, 10, 3176–3188. [Google Scholar] [CrossRef] [Green Version]
  65. Ikeuchi, Y.; Dadakhujaev, S.; Chandhoke, A.S.; Huynh, M.A.; Oldenborg, A.; Ikeuchi, M.; Deng, L.; Bennett, E.J.; Harper, J.W.; Bonni, A.; et al. TIF1γ protein regulates epithelial-mesenchymal transition by operating as a small ubiquitin-like modifier (SUMO) E3 ligase for the transcriptional regulator SnoN1. J. Biol. Chem. 2014, 289, 25067–25078. [Google Scholar] [CrossRef] [Green Version]
  66. Subramaniam, S.; Sixt, K.M.; Barrow, R.; Snyder, S.H. Rhes, a striatal specific protein, mediates mutant-huntingtin cytotoxicity. Science 2009, 324, 1327–1330. [Google Scholar] [CrossRef] [Green Version]
  67. Qiao, H.; Prasada Rao, H.B.D.; Yang, Y.; Fong, J.H.; Cloutier, J.M.; Deacon, D.C.; Nagel, K.E.; Swartz, R.K.; Strong, E.; Holloway, J.K.; et al. Antagonistic roles of ubiquitin ligase HEI10 and SUMO ligase RNF212 regulate meiotic recombination. Nat. Genet. 2014, 46, 194–199. [Google Scholar] [CrossRef]
  68. Hu, M.-M.; Liao, C.-Y.; Yang, Q.; Xie, X.-Q.; Shu, H.-B. Innate immunity to RNA virus is regulated by temporal and reversible sumoylation of RIG-I and MDA5. J. Exp. Med. 2017, 214, 973–989. [Google Scholar] [CrossRef] [Green Version]
  69. Carbia-Nagashima, A.; Gerez, J.; Perez-Castro, C.; Paez-Pereda, M.; Silberstein, S.; Stalla, G.K.; Holsboer, F.; Arzt, E. RSUME, a small RWD-containing protein, enhances SUMO conjugation and stabilizes HIF-1alpha during hypoxia. Cell 2007, 131, 309–323. [Google Scholar] [CrossRef] [Green Version]
  70. Pelisch, F.; Gerez, J.; Druker, J.; Schor, I.E.; Muñoz, M.J.; Risso, G.; Petrillo, E.; Westman, B.J.; Lamond, A.I.; Arzt, E.; et al. The serine/arginine-rich protein SF2/ASF regulates protein sumoylation. Proc. Natl. Acad. Sci. USA 2010, 107, 16119–16124. [Google Scholar] [CrossRef]
  71. Kung, C.-P.; Khaku, S.; Jennis, M.; Zhou, Y.; Murphy, M.E. Identification of TRIML2, a novel p53 target, that enhances p53 SUMOylation and regulates the transactivation of proapoptotic genes. Mol. Cancer Res. 2015, 13, 250–262. [Google Scholar] [CrossRef] [Green Version]
  72. Weger, S.; Hammer, E.; Engstler, M. The DNA topoisomerase I binding protein topors as a novel cellular target for SUMO-1 modification: Characterization of domains necessary for subcellular localization and sumolation. Exp. Cell Res. 2003, 290, 13–27. [Google Scholar] [CrossRef]
  73. Morita, Y.; Kanei-Ishii, C.; Nomura, T.; Ishii, S. TRAF7 sequesters c-Myb to the cytoplasm by stimulating its sumoylation. Mol. Biol. Cell 2005, 16, 5433–5444. [Google Scholar] [CrossRef] [Green Version]
  74. Oh, Y.; Chung, K.C. UHRF2, a ubiquitin E3 ligase, acts as a small ubiquitin-like modifier E3 ligase for zinc finger protein 131. J. Biol. Chem. 2013, 288, 9102–9111. [Google Scholar] [CrossRef] [Green Version]
  75. Rodriguez, M.S.; Dargemont, C.; Hay, R.T. SUMO-1 conjugation in vivo requires both a consensus modification motif and nuclear targeting. J. Biol. Chem. 2001, 276, 12654–12659. [Google Scholar] [CrossRef] [Green Version]
  76. Matic, I.; Schimmel, J.; Hendriks, I.A.; van Santen, M.A.; van de Rijke, F.; van Dam, H.; Gnad, F.; Mann, M.; Vertegaal, A.C.O. Site-specific identification of SUMO-2 targets in cells reveals an inverted SUMOylation motif and a hydrophobic cluster SUMOylation motif. Mol. Cell 2010, 39, 641–652. [Google Scholar] [CrossRef]
  77. Hietakangas, V.; Anckar, J.; Blomster, H.A.; Fujimoto, M.; Palvimo, J.J.; Nakai, A.; Sistonen, L. PDSM, a motif for phosphorylation-dependent SUMO modification. Proc. Natl. Acad. Sci. USA 2006, 103, 45–50. [Google Scholar] [CrossRef] [Green Version]
  78. Yang, S.-H.; Galanis, A.; Witty, J.; Sharrocks, A.D. An extended consensus motif enhances the specificity of substrate modification by SUMO. EMBO J. 2006, 25, 5083–5093. [Google Scholar] [CrossRef] [Green Version]
  79. Knopman, D.S.; Amieva, H.; Petersen, R.C.; Chételat, G.; Holtzman, D.M.; Hyman, B.T.; Nixon, R.A.; Jones, D.T. Alzheimer disease. Nat. Rev. Dis. Prim. 2021, 7, 33. [Google Scholar] [CrossRef]
  80. Breijyeh, Z.; Karaman, R. Comprehensive Review on Alzheimer’s Disease: Causes and Treatment. Molecules 2020, 25, 5789. [Google Scholar] [CrossRef]
  81. Abubakar, M.B.; Sanusi, K.O.; Ugusman, A.; Mohamed, W.; Kamal, H.; Ibrahim, N.H.; Khoo, C.S.; Kumar, J. Alzheimer’s Disease: An Update and Insights into Pathophysiology. Front. Aging Neurosci. 2022, 14, 742408. [Google Scholar] [CrossRef]
  82. Long, J.M.; Holtzman, D.M. Alzheimer Disease: An Update on Pathobiology and Treatment Strategies. Cell 2019, 179, 312–339. [Google Scholar] [CrossRef]
  83. He, Z.; Guo, J.L.; McBride, J.D.; Narasimhan, S.; Kim, H.; Changolkar, L.; Zhang, B.; Gathagan, R.J.; Yue, C.; Dengler, C.; et al. Amyloid-β plaques enhance Alzheimer’s brain tau-seeded pathologies by facilitating neuritic plaque tau aggregation. Nat. Med. 2018, 24, 29–38. [Google Scholar] [CrossRef]
  84. Nortley, R.; Korte, N.; Izquierdo, P.; Hirunpattarasilp, C.; Mishra, A.; Jaunmuktane, Z.; Kyrargyri, V.; Pfeiffer, T.; Khennouf, L.; Madry, C.; et al. Amyloid β oligomers constrict human capillaries in Alzheimer’s disease via signaling to pericytes. Science 2019, 365, 357059. [Google Scholar] [CrossRef]
  85. Krumova, P.; Meulmeester, E.; Garrido, M.; Tirard, M.; Hsiao, H.-H.; Bossis, G.; Urlaub, H.; Zweckstetter, M.; Kügler, S.; Melchior, F.; et al. Sumoylation inhibits alpha-synuclein aggregation and toxicity. J. Cell Biol. 2011, 194, 49–60. [Google Scholar] [CrossRef] [Green Version]
  86. Lee, L.; Dale, E.; Staniszewski, A.; Zhang, H.; Saeed, F.; Sakurai, M.; Fa’, M.; Orozco, I.; Michelassi, F.; Akpan, N.; et al. Regulation of synaptic plasticity and cognition by SUMO in normal physiology and Alzheimer’s disease. Sci. Rep. 2014, 4, 7190. [Google Scholar] [CrossRef] [Green Version]
  87. Lai, Y.-J.; Liu, L.; Hu, X.-T.; He, L.; Chen, G.-J. Estrogen Modulates ubc9 Expression and Synaptic Redistribution in the Brain of APP/PS1 Mice and Cortical Neurons. J. Mol. Neurosci. 2017, 61, 436–448. [Google Scholar] [CrossRef]
  88. Long, Y.; Cheng, Y.; Yang, J.; Yang, T.; Lai, Y. Abeta-induced presynaptic release of UBC9 through extracellular vesicles involves SNAP23. Neurosci. Lett. 2022, 785, 136771. [Google Scholar] [CrossRef]
  89. Zhang, Y.-Q.; Sarge, K.D. Sumoylation of amyloid precursor protein negatively regulates Abeta aggregate levels. Biochem. Biophys. Res. Commun. 2008, 374, 673–678. [Google Scholar] [CrossRef] [Green Version]
  90. Maruyama, T.; Abe, Y.; Niikura, T. SENP1 and SENP2 regulate SUMOylation of amyloid precursor protein. Heliyon 2018, 4, e00601. [Google Scholar] [CrossRef]
  91. Bao, J.; Qin, M.; Mahaman, Y.A.R.; Zhang, B.; Huang, F.; Zeng, K.; Xia, Y.; Ke, D.; Wang, Q.; Liu, R.; et al. BACE1 SUMOylation increases its stability and escalates the protease activity in Alzheimer’s disease. Proc. Natl. Acad. Sci. USA 2018, 115, 3954–3959. [Google Scholar] [CrossRef] [Green Version]
  92. Soares, E.S.; de Souza, A.C.G.; Zanella, C.A.; Carmichael, R.E.; Henley, J.M.; Wilkinson, K.A.; Cimarosti, H.I. Effects of amyloid-β on protein SUMOylation and levels of mitochondrial proteins in primary cortical neurons. IBRO Neurosci. Rep. 2022, 12, 142–148. [Google Scholar] [CrossRef]
  93. Dorval, V.; Fraser, P.E. Small ubiquitin-like modifier (SUMO) modification of natively unfolded proteins tau and alpha-synuclein. J. Biol. Chem. 2006, 281, 9919–9924. [Google Scholar] [CrossRef] [Green Version]
  94. Luo, H.-B.; Xia, Y.-Y.; Shu, X.-J.; Liu, Z.-C.; Feng, Y.; Liu, X.-H.; Yu, G.; Yin, G.; Xiong, Y.-S.; Zeng, K.; et al. SUMOylation at K340 inhibits tau degradation through deregulating its phosphorylation and ubiquitination. Proc. Natl. Acad. Sci. USA 2014, 111, 16586–16591. [Google Scholar] [CrossRef] [Green Version]
  95. Hoppe, J.B.; Salbego, C.G.; Cimarosti, H. SUMOylation: Novel Neuroprotective Approach for Alzheimer’s Disease? Aging Dis. 2015, 6, 322–330. [Google Scholar] [CrossRef] [Green Version]
  96. Poewe, W.; Seppi, K.; Tanner, C.M.; Halliday, G.M.; Brundin, P.; Volkmann, J.; Schrag, A.-E.; Lang, A.E. Parkinson disease. Nat. Rev. Dis. Prim. 2017, 3, 17013. [Google Scholar] [CrossRef]
  97. Radhakrishnan, D.M.; Goyal, V. Parkinson’s disease: A review. Neurol. India 2018, 66, S26–S35. [Google Scholar] [CrossRef]
  98. Del Rey, N.L.-G.; Quiroga-Varela, A.; Garbayo, E.; Carballo-Carbajal, I.; Fernández-Santiago, R.; Monje, M.H.G.; Trigo-Damas, I.; Blanco-Prieto, M.J.; Blesa, J. Advances in Parkinson’s Disease: 200 Years Later. Front. Neuroanat. 2018, 12, 113. [Google Scholar] [CrossRef] [Green Version]
  99. Simon, D.K.; Tanner, C.M.; Brundin, P. Parkinson Disease Epidemiology, Pathology, Genetics, and Pathophysiology. Clin. Geriatr. Med. 2020, 36, 1–12. [Google Scholar] [CrossRef]
  100. Cerri, S.; Mus, L.; Blandini, F. Parkinson’s Disease in Women and Men: What’s the Difference? J. Park. Dis. 2019, 9, 501–515. [Google Scholar] [CrossRef]
  101. Capouch, S.D.; Farlow, M.R.; Brosch, J.R. A Review of Dementia with Lewy Bodies’ Impact, Diagnostic Criteria and Treatment. Neurol. Ther. 2018, 7, 249–263. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Leitão, A.D.G.; Rudolffi-Soto, P.; Chappard, A.; Bhumkar, A.; Lau, D.; Hunter, D.J.B.; Gambin, Y.; Sierecki, E. Selectivity of Lewy body protein interactions along the aggregation pathway of α-synuclein. Commun. Biol. 2021, 4, 1124. [Google Scholar] [CrossRef] [PubMed]
  103. Lashuel, H.A.; Overk, C.R.; Oueslati, A.; Masliah, E. The many faces of α-synuclein: From structure and toxicity to therapeutic target. Nat. Rev. Neurosci. 2013, 14, 38–48. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Di Maio, R.; Barrett, P.J.; Hoffman, E.K.; Barrett, C.W.; Zharikov, A.; Borah, A.; Hu, X.; McCoy, J.; Chu, C.T.; Burton, E.A.; et al. α-Synuclein binds to TOM20 and inhibits mitochondrial protein import in Parkinson’s disease. Sci. Transl. Med. 2016, 8, 342ra78. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. De Miranda, B.R.; Rocha, E.M.; Castro, S.L.; Greenamyre, J.T. Protection from α-Synuclein induced dopaminergic neurodegeneration by overexpression of the mitochondrial import receptor TOM20. NPJ Park. Dis. 2020, 6, 38. [Google Scholar] [CrossRef]
  106. Lindersson, E.; Beedholm, R.; Højrup, P.; Moos, T.; Gai, W.; Hendil, K.B.; Jensen, P.H. Proteasomal inhibition by alpha-synuclein filaments and oligomers. J. Biol. Chem. 2004, 279, 12924–12934. [Google Scholar] [CrossRef] [Green Version]
  107. Yang, Y.; Nishimura, I.; Imai, Y.; Takahashi, R.; Lu, B. Parkin Suppresses Dopaminergic Neuron-Selective Neurotoxicity Induced by Pael-R in Drosophila. Neuron 2003, 37, 911–924. [Google Scholar] [CrossRef] [Green Version]
  108. Tsai, Y.C.; Fishman, P.S.; Thakor, N.V.; Oyler, G.A. Parkin facilitates the elimination of expanded polyglutamine proteins and leads to preservation of proteasome function. J. Biol. Chem. 2003, 278, 22044–22055. [Google Scholar] [CrossRef] [Green Version]
  109. Hawk, B.J.D.; Khounlo, R.; Shin, Y.-K. Alpha-Synuclein Continues to Enhance SNARE-Dependent Vesicle Docking at Exorbitant Concentrations. Front. Neurosci. 2019, 13, 216. [Google Scholar] [CrossRef] [Green Version]
  110. Burré, J.; Sharma, M.; Tsetsenis, T.; Buchman, V.; Etherton, M.R.; Südhof, T.C. Alpha-synuclein promotes SNARE-complex assembly in vivo and in vitro. Science 2010, 329, 1663–1667. [Google Scholar] [CrossRef]
  111. Cartelli, D.; Aliverti, A.; Barbiroli, A.; Santambrogio, C.; Ragg, E.M.; Casagrande, F.V.; Cantele, F.; Beltramone, S.; Marangon, J.; De Gregorio, C.; et al. α-Synuclein is a Novel Microtubule Dynamase. Sci. Rep. 2016, 6, 33289. [Google Scholar] [CrossRef] [Green Version]
  112. Alim, M.A.; Ma, Q.-L.; Takeda, K.; Aizawa, T.; Matsubara, M.; Nakamura, M.; Asada, A.; Saito, T.; Kaji, H.; Yoshii, M.; et al. Demonstration of a role for alpha-synuclein as a functional microtubule-associated protein. J. Alzheimers Dis. 2004, 6, 435–442; discussion 443–9. [Google Scholar] [CrossRef] [PubMed]
  113. Nemani, V.M.; Lu, W.; Berge, V.; Nakamura, K.; Onoa, B.; Lee, M.K.; Chaudhry, F.A.; Nicoll, R.A.; Edwards, R.H. Increased expression of alpha-synuclein reduces neurotransmitter release by inhibiting synaptic vesicle reclustering after endocytosis. Neuron 2010, 65, 66–79. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Scott, D.; Roy, S. α-Synuclein inhibits intersynaptic vesicle mobility and maintains recycling-pool homeostasis. J. Neurosci. 2012, 32, 10129–10135. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Rott, R.; Szargel, R.; Shani, V.; Hamza, H.; Savyon, M.; Abd Elghani, F.; Bandopadhyay, R.; Engelender, S. SUMOylation and ubiquitination reciprocally regulate α-synuclein degradation and pathological aggregation. Proc. Natl. Acad. Sci. USA 2017, 114, 13176–13181. [Google Scholar] [CrossRef] [Green Version]
  116. Hasegawa, M.; Fujiwara, H.; Nonaka, T.; Wakabayashi, K.; Takahashi, H.; Lee, V.M.-Y.; Trojanowski, J.Q.; Mann, D.; Iwatsubo, T. Phosphorylated alpha-synuclein is ubiquitinated in alpha-synucleinopathy lesions. J. Biol. Chem. 2002, 277, 49071–49076. [Google Scholar] [CrossRef] [Green Version]
  117. Giasson, B.I.; Duda, J.E.; Murray, I.V.; Chen, Q.; Souza, J.M.; Hurtig, H.I.; Ischiropoulos, H.; Trojanowski, J.Q.; Lee, V.M. Oxidative damage linked to neurodegeneration by selective alpha-synuclein nitration in synucleinopathy lesions. Science 2000, 290, 985–989. [Google Scholar] [CrossRef] [PubMed]
  118. Hart, G.W.; Housley, M.P.; Slawson, C. Cycling of O-linked beta-N-acetylglucosamine on nucleocytoplasmic proteins. Nature 2007, 446, 1017–1022. [Google Scholar] [CrossRef]
  119. Paleologou, K.E.; Oueslati, A.; Shakked, G.; Rospigliosi, C.C.; Kim, H.-Y.; Lamberto, G.R.; Fernandez, C.O.; Schmid, A.; Chegini, F.; Gai, W.P.; et al. Phosphorylation at S87 is enhanced in synucleinopathies, inhibits alpha-synuclein oligomerization, and influences synuclein-membrane interactions. J. Neurosci. 2010, 30, 3184–3198. [Google Scholar] [CrossRef] [Green Version]
  120. Abeywardana, T.; Pratt, M.R. Extent of inhibition of α-synuclein aggregation in vitro by SUMOylation is conjugation site- and SUMO isoform-selective. Biochemistry 2015, 54, 959–961. [Google Scholar] [CrossRef] [PubMed]
  121. Liang, Z.; Chan, H.Y.E.; Lee, M.M.; Chan, M.K. A SUMO1-Derived Peptide Targeting SUMO-Interacting Motif Inhibits α-Synuclein Aggregation. Cell Chem. Biol. 2021, 28, 180–190.e6. [Google Scholar] [CrossRef]
  122. Ruffmann, C.; Zini, M.; Goldwurm, S.; Bramerio, M.; Spinello, S.; Rusconi, D.; Gambacorta, M.; Tagliavini, F.; Pezzoli, G.; Giaccone, G. Lewy body pathology and typical Parkinson disease in a patient with a heterozygous (R275W) mutation in the Parkin gene (PARK2). Acta Neuropathol. 2012, 123, 901–903. [Google Scholar] [CrossRef]
  123. Kitada, T.; Asakawa, S.; Hattori, N.; Matsumine, H.; Yamamura, Y.; Minoshima, S.; Yokochi, M.; Mizuno, Y.; Shimizu, N. Mutations in the parkin gene cause autosomal recessive juvenile parkinsonism. Nature 1998, 392, 605–608. [Google Scholar] [CrossRef] [PubMed]
  124. Dawson, T.M.; Dawson, V.L. The role of parkin in familial and sporadic Parkinson’s disease. Mov. Disord. 2010, 25, S32–S39. [Google Scholar] [CrossRef] [Green Version]
  125. Um, J.W.; Chung, K.C. Functional modulation of parkin through physical interaction with SUMO-1. J. Neurosci. Res. 2006, 84, 1543–1554. [Google Scholar] [CrossRef] [PubMed]
  126. Okarmus, J.; Havelund, J.F.; Ryding, M.; Schmidt, S.I.; Bogetofte, H.; Heon-Roberts, R.; Wade-Martins, R.; Cowley, S.A.; Ryan, B.J.; Færgeman, N.J.; et al. Identification of bioactive metabolites in human iPSC-derived dopaminergic neurons with PARK2 mutation: Altered mitochondrial and energy metabolism. Stem Cell Rep. 2021, 16, 1510–1526. [Google Scholar] [CrossRef] [PubMed]
  127. Bonifati, V.; Rizzu, P.; van Baren, M.J.; Schaap, O.; Breedveld, G.J.; Krieger, E.; Dekker, M.C.J.; Squitieri, F.; Ibanez, P.; Joosse, M.; et al. Mutations in the DJ-1 gene associated with autosomal recessive early-onset parkinsonism. Science 2003, 299, 256–259. [Google Scholar] [CrossRef] [Green Version]
  128. Repici, M.; Giorgini, F. DJ-1 in Parkinson’s Disease: Clinical Insights and Therapeutic Perspectives. J. Clin. Med. 2019, 8, 1377. [Google Scholar] [CrossRef] [Green Version]
  129. Shinbo, Y.; Niki, T.; Taira, T.; Ooe, H.; Takahashi-Niki, K.; Maita, C.; Seino, C.; Iguchi-Ariga, S.M.M.; Ariga, H. Proper SUMO-1 conjugation is essential to DJ-1 to exert its full activities. Cell Death Differ. 2006, 13, 96–108. [Google Scholar] [CrossRef] [PubMed]
  130. Strobbe, D.; Robinson, A.A.; Harvey, K.; Rossi, L.; Ferraina, C.; De Biase, V.; Rodolfo, C.; Harvey, R.; Campanella, M. Distinct Mechanisms of Pathogenic DJ-1 Mutations in Mitochondrial Quality Control. Front. Mol. Neurosci. 2018, 11, 68. [Google Scholar] [CrossRef]
  131. Ross, C.A. Polyglutamine Pathogenesis. Neuron 2002, 35, 819–822. [Google Scholar] [CrossRef] [Green Version]
  132. Zoghbi, H.Y.; Orr, H.T. Glutamine repeats and neurodegeneration. Annu. Rev. Neurosci. 2000, 23, 217–247. [Google Scholar] [CrossRef] [PubMed]
  133. MacDonald, M.E.; Ambrose, C.M.; Duyao, M.P.; Myers, R.H.; Lin, C.; Srinidhi, L.; Barnes, G.; Taylor, S.A.; James, M.; Groot, N.; et al. A novel gene containing a trinucleotide repeat that is expanded and unstable on Huntington’s disease chromosomes. Cell 1993, 72, 971–983. [Google Scholar] [CrossRef]
  134. Wexler, N.S.; Lorimer, J.; Porter, J.; Gomez, F.; Moskowitz, C.; Shackell, E.; Marder, K.; Penchaszadeh, G.; Roberts, S.A.; Gayán, J.; et al. Venezuelan kindreds reveal that genetic and environmental factors modulate Huntington’s disease age of onset. Proc. Natl. Acad. Sci. USA 2004, 101, 3498–3503. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Langbehn, D.R.; Brinkman, R.R.; Falush, D.; Paulsen, J.S.; Hayden, M.R. A new model for prediction of the age of onset and penetrance for Huntington’s disease based on CAG length. Clin. Genet. 2004, 65, 267–277. [Google Scholar] [CrossRef] [PubMed]
  136. Langbehn, D.R.; Hayden, M.R.; Paulsen, J.S. CAG-repeat length and the age of onset in Huntington disease (HD): A review and validation study of statistical approaches. Am. J. Med. Genet. B Neuropsychiatr. Genet. 2010, 153, 397–408. [Google Scholar] [CrossRef] [Green Version]
  137. DiFiglia, M.; Sapp, E.; Chase, K.; Schwarz, C.; Meloni, A.; Young, C.; Martin, E.; Vonsattel, J.-P.; Carraway, R.; Reeves, S.A.; et al. Huntingtin is a cytoplasmic protein associated with vesicles in human and rat brain neurons. Neuron 1995, 14, 1075–1081. [Google Scholar] [CrossRef] [Green Version]
  138. Block-Galarza, J.; Chase, K.O.; Sapp, E.; Vaughn, K.T.; Vallee, R.B.; DiFiglia, M.; Aronin, N. Fast transport and retrograde movement of huntingtin and HAP 1 in axons. Neuroreport 1997, 8, 2247–2251. [Google Scholar] [CrossRef]
  139. Humbert, S.; Bryson, E.A.; Cordelières, F.P.; Connors, N.C.; Datta, S.R.; Finkbeiner, S.; Greenberg, M.E.; Saudou, F. The IGF-1/Akt Pathway Is Neuroprotective in Huntington’s Disease and Involves Huntingtin Phosphorylation by Akt. Dev. Cell 2002, 2, 831–837. [Google Scholar] [CrossRef] [Green Version]
  140. Rangone, H.; Poizat, G.; Troncoso, J.; Ross, C.A.; MacDonald, M.E.; Saudou, F.; Humbert, S. The serum- and glucocorticoid-induced kinase SGK inhibits mutant huntingtin-induced toxicity by phosphorylating serine 421 of huntingtin. Eur. J. Neurosci. 2004, 19, 273–279. [Google Scholar] [CrossRef]
  141. Zeitlin, S.; Liu, J.P.; Chapman, D.L.; Papaioannou, V.E.; Efstratiadis, A. Increased apoptosis and early embryonic lethality in mice nullizygous for the Huntington’s disease gene homologue. Nat. Genet. 1995, 11, 155–163. [Google Scholar] [CrossRef]
  142. Nasir, J.; Floresco, S.B.; O’Kusky, J.R.; Diewert, V.M.; Richman, J.M.; Zeisler, J.; Borowski, A.; Marth, J.D.; Phillips, A.G.; Hayden, M.R. Targeted disruption of the Huntington’s disease gene results in embryonic lethality and behavioral and morphological changes in heterozygotes. Cell 1995, 81, 811–823. [Google Scholar] [CrossRef] [Green Version]
  143. Dragatsis, I.; Levine, M.S.; Zeitlin, S. Inactivation of Hdh in the brain and testis results in progressive neurodegeneration and sterility in mice. Nat. Genet. 2000, 26, 300–306. [Google Scholar] [CrossRef]
  144. Miller, J.; Arrasate, M.; Shaby, B.A.; Mitra, S.; Masliah, E.; Finkbeiner, S. Quantitative relationships between huntingtin levels, polyglutamine length, inclusion body formation, and neuronal death provide novel insight into huntington’s disease molecular pathogenesis. J. Neurosci. 2010, 30, 10541–10550. [Google Scholar] [CrossRef] [Green Version]
  145. Arrasate, M.; Mitra, S.; Schweitzer, E.S.; Segal, M.R.; Finkbeiner, S. Inclusion body formation reduces levels of mutant huntingtin and the risk of neuronal death. Nature 2004, 431, 805–810. [Google Scholar] [CrossRef] [Green Version]
  146. Yahr, M.D. Garrison’s History of Neurology. Arch. Neurol. 1969, 21, 342. [Google Scholar] [CrossRef]
  147. Rubinsztein, D.C.; Leggo, J.; Coles, R.; Almqvist, E.; Biancalana, V.; Cassiman, J.J.; Chotai, K.; Connarty, M.; Crauford, D.; Curtis, A.; et al. Phenotypic characterization of individuals with 30–40 CAG repeats in the Huntington disease (HD) gene reveals HD cases with 36 repeats and apparently normal elderly individuals with 36–39 repeats. Am. J. Hum. Genet. 1996, 59, 16–22. [Google Scholar]
  148. Duyao, M.; Ambrose, C.; Myers, R.; Novelletto, A.; Persichetti, F.; Frontali, M.; Folstein, S.; Ross, C.; Franz, M.; Abbott, M. Trinucleotide repeat length instability and age of onset in Huntington’s disease. Nat. Genet. 1993, 4, 387–392. [Google Scholar] [CrossRef]
  149. Harper, P.S. Major Problems in Neurology: Huntington’s Disease, 2nd ed.; W.B. Saunders Company Ltd.: London, UK, 1996. [Google Scholar]
  150. Kalchman, M.A.; Graham, R.K.; Xia, G.; Koide, H.B.; Hodgson, J.G.; Graham, K.C.; Goldberg, Y.P.; Gietz, R.D.; Pickart, C.M.; Hayden, M.R. Huntingtin is ubiquitinated and interacts with a specific ubiquitin-conjugating enzyme. J. Biol. Chem. 1996, 271, 19385–19394. [Google Scholar] [CrossRef] [Green Version]
  151. Davies, S.W.; Turmaine, M.; Cozens, B.A.; DiFiglia, M.; Sharp, A.H.; Ross, C.A.; Scherzinger, E.; Wanker, E.E.; Mangiarini, L.; Bates, G.P. Formation of Neuronal Intranuclear Inclusions Underlies the Neurological Dysfunction in Mice Transgenic for the HD Mutation. Cell 1997, 90, 537–548. [Google Scholar] [CrossRef] [Green Version]
  152. Saudou, F.; Finkbeiner, S.; Devys, D.; Greenberg, M.E. Huntingtin Acts in the Nucleus to Induce Apoptosis but Death Does Not Correlate with the Formation of Intranuclear Inclusions. Cell 1998, 95, 55–66. [Google Scholar] [CrossRef]
  153. Steffan, J.S.; Agrawal, N.; Pallos, J.; Rockabrand, E.; Trotman, L.C.; Slepko, N.; Illes, K.; Lukacsovich, T.; Zhu, Y.-Z.; Cattaneo, E.; et al. SUMO modification of Huntingtin and Huntington’s disease pathology. Science 2004, 304, 100–104. [Google Scholar] [CrossRef] [Green Version]
  154. O’Rourke, J.G.; Gareau, J.R.; Ochaba, J.; Song, W.; Raskó, T.; Reverter, D.; Lee, J.; Monteys, A.M.; Pallos, J.; Mee, L.; et al. SUMO-2 and PIAS1 modulate insoluble mutant huntingtin protein accumulation. Cell Rep. 2013, 4, 362–375. [Google Scholar] [CrossRef] [Green Version]
  155. Sedighi, F.; Adegbuyiro, A.; Legleiter, J. SUMOylation Prevents Huntingtin Fibrillization and Localization onto Lipid Membranes. ACS Chem. Neurosci. 2020, 11, 328–343. [Google Scholar] [CrossRef]
  156. Morozko, E.L.; Smith-Geater, C.; Monteys, A.M.; Pradhan, S.; Lim, R.G.; Langfelder, P.; Kachemov, M.; Kulkarni, J.A.; Zaifman, J.; Hill, A.; et al. PIAS1 modulates striatal transcription, DNA damage repair, and SUMOylation with relevance to Huntington’s disease. Proc. Natl. Acad. Sci. USA 2021, 118, e2021836118. [Google Scholar] [CrossRef]
  157. Ochaba, J.; Monteys, A.M.; O’Rourke, J.G.; Reidling, J.C.; Steffan, J.S.; Davidson, B.L.; Thompson, L.M. PIAS1 Regulates Mutant Huntingtin Accumulation and Huntington’s Disease-Associated Phenotypes In Vivo. Neuron 2016, 90, 507–520. [Google Scholar] [CrossRef] [Green Version]
  158. Ramírez-Jarquín, U.N.; Sharma, M.; Shahani, N.; Li, Y.; Boregowda, S.; Subramaniam, S. Rhes protein transits from neuron to neuron and facilitates mutant huntingtin spreading in the brain. Sci. Adv. 2022, 8, eabm3877. [Google Scholar] [CrossRef]
  159. Heikkinen, T.; Bragge, T.; Kuosmanen, J.; Parkkari, T.; Gustafsson, S.; Kwan, M.; Beltran, J.; Ghavami, A.; Subramaniam, S.; Shahani, N.; et al. Global Rhes knockout in the Q175 Huntington’s disease mouse model. PLoS ONE 2021, 16, e0258486. [Google Scholar] [CrossRef]
  160. Brugnoli, A.; Napolitano, F.; Usiello, A.; Morari, M. Genetic deletion of Rhes or pharmacological blockade of mTORC1 prevent striato-nigral neurons activation in levodopa-induced dyskinesia. Neurobiol. Dis. 2016, 85, 155–163. [Google Scholar] [CrossRef]
  161. Mealer, R.G.; Subramaniam, S.; Snyder, S.H. Rhes deletion is neuroprotective in the 3-nitropropionic acid model of Huntington’s disease. J. Neurosci. 2013, 33, 4206–4210. [Google Scholar] [CrossRef] [Green Version]
  162. Pinna, A.; Napolitano, F.; Pelosi, B.; Di Maio, A.; Wardas, J.; Casu, M.A.; Costa, G.; Migliarini, S.; Calabresi, P.; Pasqualetti, M.; et al. The Small GTP-Binding Protein Rhes Influences Nigrostriatal-Dependent Motor Behavior during Aging. Mov. Disord. 2016, 31, 583–589. [Google Scholar] [CrossRef]
  163. Gu, M.; Gash, M.T.; Mann, V.M.; Javoy-Agid, F.; Cooper, J.M.; Schapira, A.H. Mitochondrial defect in Huntington’s disease caudate nucleus. Ann. Neurol. 1996, 39, 385–389. [Google Scholar] [CrossRef]
  164. Milakovic, T.; Johnson, G.V.W. Mitochondrial respiration and ATP production are significantly impaired in striatal cells expressing mutant huntingtin. J. Biol. Chem. 2005, 280, 30773–30782. [Google Scholar] [CrossRef] [Green Version]
  165. Yablonska, S.; Ganesan, V.; Ferrando, L.M.; Kim, J.; Pyzel, A.; Baranova, O.V.; Khattar, N.K.; Larkin, T.M.; Baranov, S.V.; Chen, N.; et al. Mutant huntingtin disrupts mitochondrial proteostasis by interacting with TIM23. Proc. Natl. Acad. Sci. USA 2019, 116, 16593–16602. [Google Scholar] [CrossRef] [Green Version]
  166. Reddy, P.H. Increased mitochondrial fission and neuronal dysfunction in Huntington’s disease: Implications for molecular inhibitors of excessive mitochondrial fission. Drug Discov. Today 2014, 19, 951–955. [Google Scholar] [CrossRef] [Green Version]
  167. Shirendeb, U.P.; Calkins, M.J.; Manczak, M.; Anekonda, V.; Dufour, B.; McBride, J.L.; Mao, P.; Reddy, P.H. Mutant huntingtin’s interaction with mitochondrial protein Drp1 impairs mitochondrial biogenesis and causes defective axonal transport and synaptic degeneration in Huntington’s disease. Hum. Mol. Genet. 2012, 21, 406–420. [Google Scholar] [CrossRef] [Green Version]
  168. Yano, H.; Baranov, S.V.; Baranova, O.V.; Kim, J.; Pan, Y.; Yablonska, S.; Carlisle, D.L.; Ferrante, R.J.; Kim, A.H.; Friedlander, R.M. Inhibition of mitochondrial protein import by mutant huntingtin. Nat. Neurosci. 2014, 17, 822–831. [Google Scholar] [CrossRef] [Green Version]
  169. Song, W.; Chen, J.; Petrilli, A.; Liot, G.; Klinglmayr, E.; Zhou, Y.; Poquiz, P.; Tjong, J.; Pouladi, M.A.; Hayden, M.R.; et al. Mutant huntingtin binds the mitochondrial fission GTPase dynamin-related protein-1 and increases its enzymatic activity. Nat. Med. 2011, 17, 377–382. [Google Scholar] [CrossRef] [Green Version]
  170. Zheng, J.; Winderickx, J.; Franssens, V.; Liu, B. A Mitochondria-Associated Oxidative Stress Perspective on Huntington’s Disease. Front. Mol. Neurosci. 2018, 11, 329. [Google Scholar] [CrossRef]
  171. Kumar, S.; Ashe, H.A.; Parnell, L.N.; Fernando, D.J.; Tsigos, C.; Young, R.J.; Ward, J.D.; Boulton, A.J. The prevalence of foot ulceration and its correlates in type 2 diabetic patients: A population-based study. Diabet. Med. 1994, 11, 480–484. [Google Scholar] [CrossRef]
  172. Yovera-Aldana, M.; Velásquez-Rimachi, V.; Huerta-Rosario, A.; More-Yupanqui, M.D.; Osores-Flores, M.; Espinoza, R.; Gil-Olivares, F.; Quispe-Nolazco, C.; Quea-Vélez, F.; Morán-Mariños, C.; et al. Prevalence and incidence of diabetic peripheral neuropathy in Latin America and the Caribbean: A systematic review and meta-analysis. PLoS ONE 2021, 16, e0251642. [Google Scholar] [CrossRef]
  173. Feldman, E.L.; Callaghan, B.C.; Pop-Busui, R.; Zochodne, D.W.; Wright, D.E.; Bennett, D.L.; Bril, V.; Russell, J.W.; Viswanathan, V. Diabetic neuropathy. Nat. Rev. Dis. Prim. 2019, 5, 41. [Google Scholar] [CrossRef]
  174. Rojas, D.R.; Kuner, R.; Agarwal, N. Metabolomic signature of type 1 diabetes-induced sensory loss and nerve damage in diabetic neuropathy. J. Mol. Med. 2019, 97, 845–854. [Google Scholar] [CrossRef] [Green Version]
  175. Tehrani, K.H.N. A Study of Nerve Conduction Velocity in Diabetic Patients and its Relationship with Tendon Reflexes (T-Reflex). Open Access Maced. J. Med. Sci. 2018, 6, 1072–1076. [Google Scholar] [CrossRef] [Green Version]
  176. Charles, M.; Soedamah-Muthu, S.S.; Tesfaye, S.; Fuller, J.H.; Arezzo, J.C.; Chaturvedi, N.; Witte, D.R. Low peripheral nerve conduction velocities and amplitudes are strongly related to diabetic microvascular complications in type 1 diabetes: The EURODIAB Prospective Complications Study. Diabetes Care 2010, 33, 2648–2653. [Google Scholar] [CrossRef] [Green Version]
  177. Dahlin, L.B.; Rix, K.R.; Dahl, V.A.; Dahl, A.B.; Jensen, J.N.; Cloetens, P.; Pacureanu, A.; Mohseni, S.; Thomsen, N.O.B.; Bech, M. Three-dimensional architecture of human diabetic peripheral nerves revealed by X-ray phase contrast holographic nanotomography. Sci. Rep. 2020, 10, 7592. [Google Scholar] [CrossRef]
  178. Kowalski, G.M.; Bruce, C.R. The regulation of glucose metabolism: Implications and considerations for the assessment of glucose homeostasis in rodents. Am. J. Physiol. Endocrinol. Metab. 2014, 307, E859–E871. [Google Scholar] [CrossRef] [Green Version]
  179. Brownlee, M. Biochemistry and molecular cell biology of diabetic complications. Nature 2001, 414, 813–820. [Google Scholar] [CrossRef]
  180. Yan, L.-J. Redox imbalance stress in diabetes mellitus: Role of the polyol pathway. Anim. Model. Exp. Med. 2018, 1, 7–13. [Google Scholar] [CrossRef]
  181. Yan, S.F.; Ramasamy, R.; Schmidt, A.M. Mechanisms of disease: Advanced glycation end-products and their receptor in inflammation and diabetes complications. Nat. Clin. Pr. Endocrinol. Metab. 2008, 4, 285–293. [Google Scholar] [CrossRef]
  182. Diskin, C.; Ryan, T.A.J.; O’Neill, L.A.J. Modification of Proteins by Metabolites in Immunity. Immunity 2021, 54, 19–31. [Google Scholar] [CrossRef]
  183. Zhu, S.; Gu, H.; Peng, C.; Xia, F.; Cao, H.; Cui, H. Regulation of Glucose, Fatty Acid and Amino Acid Metabolism by Ubiquitination and SUMOylation for Cancer Progression. Front. Cell Dev. Biol. 2022, 10, 849625. [Google Scholar] [CrossRef] [PubMed]
  184. Xiong, Y.; Guan, K.-L. Mechanistic insights into the regulation of metabolic enzymes by acetylation. J. Cell Biol. 2012, 198, 155–164. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Yu, Y.; Chen, Y.; Liu, K.; Cheng, J.; Tu, J. SUMOylation enhances the activity of IDH2 under oxidative stress. Biochem. Biophys. Res. Commun. 2020, 532, 591–597. [Google Scholar] [CrossRef] [PubMed]
  186. Hou, Z.; Chen, J.; Yang, H.; Hu, X.; Yang, F. PIAS1 alleviates diabetic peripheral neuropathy through SUMOlation of PPAR-γ and miR-124-induced downregulation of EZH2/STAT3. Cell Death Discov. 2021, 7, 372. [Google Scholar] [CrossRef] [PubMed]
  187. Schorova, L.; Martin, S. Sumoylation in Synaptic Function and Dysfunction. Front. Synaptic Neurosci. 2016, 8, 9. [Google Scholar] [CrossRef] [Green Version]
  188. Benson, M.; Iñiguez-Lluhí, J.A.; Martens, J. Sumo Modification of Ion Channels. Adv. Exp. Med. Biol. 2017, 963, 127–141. [Google Scholar] [CrossRef]
  189. Plant, L.D.; Marks, J.D.; Goldstein, S.A. SUMOylation of NaV1.2 channels mediates the early response to acute hypoxia in central neurons. Elife 2016, 5, e20054. [Google Scholar] [CrossRef] [PubMed]
  190. Rajan, S.; Plant, L.D.; Rabin, M.L.; Butler, M.H.; Goldstein, S.A.N. Sumoylation silences the plasma membrane leak K+ channel K2P1. Cell 2005, 121, 37–47. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  191. Benson, M.D.; Li, Q.-J.; Kieckhafer, K.; Dudek, D.; Whorton, M.R.; Sunahara, R.K.; Iñiguez-Lluhí, J.A.; Martens, J.R. SUMO modification regulates inactivation of the voltage-gated potassium channel Kv1.5. Proc. Natl. Acad. Sci. USA 2007, 104, 1805–1810. [Google Scholar] [CrossRef] [Green Version]
  192. Julius, D. TRP channels and pain. Annu. Rev. Cell Dev. Biol. 2013, 29, 355–384. [Google Scholar] [CrossRef] [PubMed]
  193. Facer, P.; Casula, M.A.; Smith, G.D.; Benham, C.D.; Chessell, I.P.; Bountra, C.; Sinisi, M.; Birch, R.; Anand, P. Differential expression of the capsaicin receptor TRPV1 and related novel receptors TRPV3, TRPV4 and TRPM8 in normal human tissues and changes in traumatic and diabetic neuropathy. BMC Neurol. 2007, 7, 11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. Hosseini, A.; Abdollahi, M. Diabetic neuropathy and oxidative stress: Therapeutic perspectives. Oxid. Med. Cell. Longev. 2013, 2013, 168039. [Google Scholar] [CrossRef] [PubMed]
  195. Vincent, A.M.; Russell, J.W.; Low, P.; Feldman, E.L. Oxidative stress in the pathogenesis of diabetic neuropathy. Endocr. Rev. 2004, 25, 612–628. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  196. Liu, Y.; Bridges, R.; Wortham, A.; Kulesz-Martin, M. NF-κB repression by PIAS3 mediated RelA SUMOylation. PLoS ONE 2012, 7, e37636. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  197. Stankovic-Valentin, N.; Drzewicka, K.; König, C.; Schiebel, E.; Melchior, F. Redox regulation of SUMO enzymes is required for ATM activity and survival in oxidative stress. EMBO J. 2016, 35, 1312–1329. [Google Scholar] [CrossRef]
Figure 1. The catalytic cycle of SUMOylation and deSUMOylation. Maturation: SENPs process the SUMO precursor into mature SUMO. Activation: SUMO is activated in an ATP-dependent 2-step reaction by the heterodimeric E1 enzyme and bound via a thioester bond on cysteine residues to the SAE2 subunit. Conjugation: SUMO is actively transferred to the cysteine residue of the E2 enzyme UBC9. Ligation: SUMO is attached to a specific lysine residue in the substrate; this step can be directly done by E2-SUMO but usually requires an E3 ligase that enhances the conjugation. DeSUMOylation: SUMO proteins are cleaved off from substrates by SENPs, free SUMO is available for another catalytic cycle. Figure was created with BioRender.com.
Figure 1. The catalytic cycle of SUMOylation and deSUMOylation. Maturation: SENPs process the SUMO precursor into mature SUMO. Activation: SUMO is activated in an ATP-dependent 2-step reaction by the heterodimeric E1 enzyme and bound via a thioester bond on cysteine residues to the SAE2 subunit. Conjugation: SUMO is actively transferred to the cysteine residue of the E2 enzyme UBC9. Ligation: SUMO is attached to a specific lysine residue in the substrate; this step can be directly done by E2-SUMO but usually requires an E3 ligase that enhances the conjugation. DeSUMOylation: SUMO proteins are cleaved off from substrates by SENPs, free SUMO is available for another catalytic cycle. Figure was created with BioRender.com.
Cells 11 03395 g001
Figure 2. SUMOylation in Alzheimer’s disease: SUMOylation of APP leads to lower levels of Aβ aggregate levels. In contrast, the SUMOylation of β-secretase leads to the increased formation of Aβ aggregates. The SUMOylation of Tau prevents ubiquitination and leads to the formation of Tau protein tangles. UBC9 overexpression was able to rescue Aβ-mediated deficits. Figure was created with BioRender.com.
Figure 2. SUMOylation in Alzheimer’s disease: SUMOylation of APP leads to lower levels of Aβ aggregate levels. In contrast, the SUMOylation of β-secretase leads to the increased formation of Aβ aggregates. The SUMOylation of Tau prevents ubiquitination and leads to the formation of Tau protein tangles. UBC9 overexpression was able to rescue Aβ-mediated deficits. Figure was created with BioRender.com.
Cells 11 03395 g002
Figure 3. SUMOylation in Parkinson’s disease: SUMO-modified α-synuclein remains soluble and less amyloid fibers are formed. SUMOylation of DJ-1 promotes its activity and results in reduced mitochondrial stress. The SUMOylation of parkin promotes its nuclear transport. Figure was created with BioRender.com.
Figure 3. SUMOylation in Parkinson’s disease: SUMO-modified α-synuclein remains soluble and less amyloid fibers are formed. SUMOylation of DJ-1 promotes its activity and results in reduced mitochondrial stress. The SUMOylation of parkin promotes its nuclear transport. Figure was created with BioRender.com.
Cells 11 03395 g003
Figure 4. SUMOylation in Huntington’s disease: SUMOylation of Httex1p has an ambivalent effect and can either decrease or increase the solubility, depending on if it is SUMO1- or SUMO2-attached. The downregulation of PIAS1 in the striatum leads to the reduced SUMOylation of HTT and decreased accumulation. Rhes knockout in mice has been shown to have beneficial and neuroprotective effects in Huntington’s disease. Figure was created with BioRender.com.
Figure 4. SUMOylation in Huntington’s disease: SUMOylation of Httex1p has an ambivalent effect and can either decrease or increase the solubility, depending on if it is SUMO1- or SUMO2-attached. The downregulation of PIAS1 in the striatum leads to the reduced SUMOylation of HTT and decreased accumulation. Rhes knockout in mice has been shown to have beneficial and neuroprotective effects in Huntington’s disease. Figure was created with BioRender.com.
Cells 11 03395 g004
Figure 5. SUMOylation in diabetic peripheral neuropathy: Prolonged hyperglycemia leads to reduced levels of SUMO-conjugated proteins in neuron and Schwann cells. The tissue-specific knockout of SUMOylation in peripheral sensory neurons of DRGs leads to a functional impairment of TRPV1 channels, altered the function of metabolic enzymes and increased the onset of DPN. The SUMOylation of PPAR- γ results in higher miR-124 levels, which inhibit Schwann cell apoptosis. Figure was created with Biorender.com.
Figure 5. SUMOylation in diabetic peripheral neuropathy: Prolonged hyperglycemia leads to reduced levels of SUMO-conjugated proteins in neuron and Schwann cells. The tissue-specific knockout of SUMOylation in peripheral sensory neurons of DRGs leads to a functional impairment of TRPV1 channels, altered the function of metabolic enzymes and increased the onset of DPN. The SUMOylation of PPAR- γ results in higher miR-124 levels, which inhibit Schwann cell apoptosis. Figure was created with Biorender.com.
Cells 11 03395 g005
Table 1. Categorization of SUMO E3 ligases.
Table 1. Categorization of SUMO E3 ligases.
SP-RING FamilyDescribed bySIM FamilyDescribed by
PIAS1Kahyo et al. 2001 [45]CBX4Kagey et al. 2003 [46]
PIAS3Sentis et al. 2005 [50]KIAA1586Eisenhardt et al. 2015 [48]
PIAS3-βSentis et al. 2005 [50]RanBP2Pichler et al. 2002 [47]
PIASx-αKotaja et al. 2002 [51]SLX4Guervilly et al. 2015 [49]
PIASx-βAcosta et al. 2005 [52]ZNF451Eisenhardt et al. 2015 [48]
PIASyWong et al. 2004 [53]
PIASy-E6Wong et al. 2004 [53]
MMS21Potts and Yu 2005 [54]
ZMIZ1Moreno-Ayala et al. 2015 [55]
ZMIZ2Moreno-Ayala et al. 2015 [55]
TRIM SuperfamilyDescribed byOthersDescribed by
TRIM1Chu and Yang 2011 [41]ARFTago et al. 2005 [56]
TRIM9Chu and Yang 2011 [41]BCA2Lluch and Moreno 2017 [57]
TRIM11Zhu et al. 2020 [58]DREFYamashita et al. 2016 [59]
TRIM19Chu and Yang 2011 [41]HDAC4Zhao et al. 2005 [60]
TRIM22Chu and Yang 2011 [41]HDAC-5, -7, -9 Grégoire and Yang 2005 [61]
TRIM27Chu and Yang 2011 [41]KROX2Gutiérrez et al. 2011 [62]
TRIM28Chu and Yang 2011 [41]MAPLBraschi et al. 2009 [63]
TRIM32Chu and Yang 2011 [41]MDM2Stindt et al. 2011s [64]
TRIM33Ikeuchi et al. 2014 [65]RHESSubramaniam et al. 2009 [66]
TRIM36Chu and Yang 2011 [41]RNFQiao et al. 2014 [67]
TRIM38Hu et al. 2017 [68]RSUMENagashima et al. 2007 [69]
TRIM39Chu and Yang 2011 [41]SF2Pelisch et al. 2010 [70]
TRIML2Kung et al. 2015 [71]TOPORSWeger et al. 2003 [72]
TRAF7Morita et al. 2005 [73]
UHRF2Oh and Chung 2013 [74]
Table 2. The different SUMOylation motifs.
Table 2. The different SUMOylation motifs.
SUMOylation MotifsAmino AcidsDescribed by
Classical Consensus motifψKXE/DRodriguez et al. 2001 [75]
Inverted Consensus motifE/DXKψMatic et al. 2010 [76]
Hydrophobic cluster motif(ψ)nKXEMatic et al. 2010 [76]
PDSMψKXEXXS-℗Hietakangas et al. 2005 [77]
NDSMψKXEXX(E)nYang et al. 2006 [78]
Ψ = bulky hydrophobic residues; K = the lysine modified; E/D = acidic residues; X = any residue; n = two or more; S-℗ = phosphorylated serine.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Mandel, N.; Agarwal, N. Role of SUMOylation in Neurodegenerative Diseases. Cells 2022, 11, 3395. https://doi.org/10.3390/cells11213395

AMA Style

Mandel N, Agarwal N. Role of SUMOylation in Neurodegenerative Diseases. Cells. 2022; 11(21):3395. https://doi.org/10.3390/cells11213395

Chicago/Turabian Style

Mandel, Nicolas, and Nitin Agarwal. 2022. "Role of SUMOylation in Neurodegenerative Diseases" Cells 11, no. 21: 3395. https://doi.org/10.3390/cells11213395

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop