Open access peer-reviewed chapter

The Role of Lysine 63-Linked Ubiquitylation in Health and Disease

Written By

Paola Pontrelli, Francesca Conserva and Loreto Gesualdo

Submitted: 27 August 2018 Reviewed: 19 December 2018 Published: 01 February 2019

DOI: 10.5772/intechopen.83659

From the Edited Volume

Ubiquitin Proteasome System - Current Insights into Mechanism Cellular Regulation and Disease

Edited by Matthew Summers

Chapter metrics overview

1,377 Chapter Downloads

View Full Metrics

Abstract

A specific subfamily within the E2 protein family is involved in the synthesis of noncanonical poly-ubiquitin chains, linked through lysine 63 residues. The role of lysine 63-linked polyubiquitylation in diseases has emerged only recently. Under physiological conditions, this process does not seem to be involved in the classical protein degradation by the proteasome, but it is involved in the regulation of intracellular signaling, DNA damage response, cellular trafficking, and lysosomal targeting. The alteration of this process has been described in a number of pathological conditions, including immune disorders, diabetes, and cancer. In this chapter, we will describe the role of lysine 63-linked ubiquitylation in the regulation of diverse signaling pathways involved in cell behavior. We will also describe some pathological conditions in which altered lysine 63-linked ubiquitylation has been referred to play an important role.

Keywords

  • lysine 63-linked ubiquitylation
  • immune system
  • diabetes complications
  • autophagy
  • cancer

1. Introduction

The ubiquitin signaling system, often referred to as “ubiquitin code”, is very complex, although the ubiquitin moieties engaged in protein ubiquitylation are always identical. In order to understand the complexity of the ubiquitin code, we need to remember that the ubiquitin moiety contains seven lysine residues, all of which can be potentially engaged in the formation of polyubiquitin chains, and the protein fate depends upon the specific lysine residue involved in the polyubiquitin link as well as the length of the polyubiquitin chain [1, 2].

Several types of polyubiquitin chains exist in cells and the type of chain defines how ubiquitinated proteins are regulated. For instance, we know that ubiquitin chains generated via lysine (K)48 of ubiquitin (K48 chains) function as a signal for proteolysis, while chains generated via K63 (K63 chains) are involved in nonproteolytic functions, such as DNA repair, protein kinase activation, and membrane trafficking [3].

The ubiquitin signaling starts with the activation of the ubiquitin moiety by an ubiquitin-activating enzyme E1, and the energy to initiate this process is provided by an ATP molecule. Exploiting its active site containing a cysteine, E1s attack the ubiquitin-AMP intermediate, forming a thioester bond. The subsequent reaction involves the transfer of the activated ubiquitin from the E1 to an E2 enzyme through a transthioesterification reaction; E2 enzymes also contain an active site that includes a cysteine residue. The final step is carried out by ubiquitin protein ligase E3 enzymes, which allows the transfer of ubiquitin to the lysine of the target protein [4, 5]. Although target specificity is given by E3 enzymes, not all E3s are able to ligate the ubiquitin molecule to their target directly, as this ability is dependent on the type of active site they are furnished with. There are two main types of catalytic domains on E3 enzymes. The E3s possessing a RING (really interesting new gene) domain in their active site do not contain a cysteine residue within the active-site; thus, they work by bringing the “charged” E2 in close proximity to the target protein, and ubiquitin is transferred by the E2. E3s that possess a HECT domain [homology to E6-AP carboxyl terminus] instead, contain a cysteine residue in their active site that allows the formation of a thioester intermediate and the subsequent transfer of ubiquitin to the target protein [6, 7].

In summary, the enzymatic cascade that leads to the formation of an isopeptide bond between ubiquitin and its target molecule involves E1, E2, and E3 enzymes, and the ubiquitin chain linkage specificity is generally conferred by E2s.

Among the E2s that participate in the specific formation of K63-linked ubiquitin chains, Ubc13 (also known as Ube2n) is probably the best characterized. To finalize the reaction, Ubc13 requires the concomitant presence of specific E2-like partner proteins, among which Ube2v1 (also known as Uev1A) is involved in the formation of K63-linked ubiquitin chains in the cytosol, while the protein Mms2 participates in the nuclear K63-linked chain formation. It was also shown that this enzymatic complex exerts its activity on previously mono-ubiquitinated substrates; thus, the priming of the substrate with the first ubiquitin molecule can likely be promoted by different E2s [8].

Overall, the complexity of the cellular responses elicited by ubiquitin is actually greater than previously foreseen; it was recently discovered that heterogeneous ubiquitin chains also exist, possessing both proteolytic and nonproteolytic functions. Also, ubiquitin itself can be modified through phosphorylation and/or acetylation [9, 10]. Finally, deubiquitinating enzymes (DUBs) exist which operate through either the editing or disassembly of ubiquitin chains, allowing the fine-tuning of the entire system [11].

Importantly, both the proteolytic and nonproteolytic functions of ubiquitin are crucial to regulate different intracellular signaling pathways involved in the modulation of immunity, inflammation, and cell survival [12]. Here we report an overview on the main pathways modulated by K63-linked ubiquitylation and the role of this post-translational modification in health and disease.

Advertisement

2. Physiological roles of lysine 63-linked ubiquitylation

2.1. Lysine 63-linked ubiquitylation in NF-κB signaling

NF-κB is a dimeric transcription factor that controls cytokine production and cell growth, ultimately modulating processes such as inflammation and immune responses. This protein complex is ubiquitously present in the cell cytoplasm in an inactive state but it can rapidly be processed into its active form by different regulatory mechanisms including ubiquitylation.

The canonical ubiquitylation trough K48-linked chains controls processing of the NF-κB precursor p100 and p105, as well as degradation of the NF-κB inhibitor IκB when it becomes phosphorylated by the IκB kinase (IKK) complex. The noncanonical ubiquitylation through K63-linked chains is instead involved in the activation of the IκB kinase complex IKK [13]. These events are mediated, among others, by a class of E3 enzymes known as the TNF receptor-associated factor (TRAF). TRAF proteins mediate NF-κB activation from a number of receptors such as the TNF receptor (TNFR), IL-1 receptor (IL-1R), and Toll-like receptor (TLR). TRAF6 transduces signals from IL1-R/TLR while TRAF2 transduces signals from TNFR. Both TRAF-2 and TRAF-6 are able to form K63-linked ubiquitin chains on their specific targets [14].

2.1.1. IL1-R/TLR-induced NF-κB signaling

In the NF-κB signaling triggered by activation of the IL1-R/TLR, the E2 protein complex consisting of Ubc13-Uev1A, in concert with the E3 enzyme TRAF6, leads to the formation of K63-linked ubiquitin chains on several target proteins including Interleukin 1 Receptor Associated Kinase 1 (IRAK1), NF-kappa-B essential modulator (NEMO), and TRAF6 itself [15]. Once ubiquitinated, TRAF6 can be recognized by a specific ubiquitin binding domain (UBD) within the protein TGF-β Activated Kinase 1 (MAP3K7) Binding Protein 2 (TAB2) and this interaction activates the TAB2-associated TAK1 kinase, which in turn phosphorylates and activates IKK. Active IKK promotes degradation of IκB and ultimately releases inhibition on NF-κB (Figure 1). It was shown that TAK1 can activate IKK only in the presence of the NF-κB essential modulator NEMO. Importantly, this protein contains a C-terminal domain that binds preferentially to K63 ubiquitin chains. Thus, this type of ubiquitylation might be useful to provide a scaffold that facilitates protein interactions [14, 16, 17].

Figure 1.

IL1-R/TLR-induced NF-κB signaling.

2.1.2. TNFR-induced NF-κB signaling

NF-κB activation can also be triggered by molecules that bind TNF-R1. As for IL-1/TLR, TNF-R1 signal transduction involves a cascade of reactions that are regulated at different levels by protein ubiquitylation.

The model proposed for NF-κB activation suggests that upon exposure to TNF-α, TNF-R1 undergoes a conformational change that allows recruitment of the adaptor tumor necrosis factor receptor type 1-associated death domain protein (TRADD) within the cytosol. This interaction elicits the enrollment of two additional proteins: TRAF2 and kinase receptor-interacting serine/threonine-protein kinase 1 (RIPK1). Unlike TRAF6, TRAF2 is unable to attach ubiquitin moieties to RIPK1 independently, but acts as a scaffold for the recruitment of two different E3 enzymes: cellular inhibitor of apoptosis protein-1 (c-IAP1) and c-IAP2. These proteins add, among others, K63-linked chains on RIPK1; thus, they are the actual effectors of RIPK1 ubiquitylation. The following events that lead to NF-κB activation involve the formation of a second E3 protein complex at the initial site of ubiquitylation known as linear ubiquitin chain assembly complex (LUBAC). In linear ubiquitin chains, the C-ter Gly76 of one ubiquitin is linked to the α-NH2 group of Met1 of another ubiquitin moiety. Thus, the LUBAC complex catalyzes the formation of linear (M1)-linked ubiquitin chains on RIPK1 proteins, c-IAP1 and c-IAP2. Once these linear chains are added, RIPK1 is structurally able to attract the kinase complexes TAK1 and IKK through their ubiquitin binding domain-containing subunits (TAB2/TAB3 and NEMO). This ultimately triggers IKK phosphorylation by TAK1 and NF-κB activation [18] (Figure 2). In summary, a variety of ubiquitin chain modifications seem to be required for NF-κB activation, and further studies will shed light on the many roles of each specific type of polyubiquitin chains.

Figure 2.

TNFR1-induced NF-κB signaling.

2.2. Lysine 63-linked ubiquitylation in Wnt/β-catenin signaling

The Wnt/β-catenin signaling pathway is essential in the regulation of events such as cell proliferation, organized migration, self-renewal, and tissue polarity. Disruption of the Wnt/β-catenin signaling pathway has been linked with oncogenesis and other pathological conditions. Regulation of Wnt signaling is controlled by protein ubiquitylation at many levels, and K48- and K63-linked ubiquitin chains in particular have been shown to regulate this pathway through both proteolytic and nonproteolytic functions.

Within the canonical Wnt/β-catenin signaling, the absence of a Wnt ligand at the transmembrane receptor Frizzled (Fz) determines the rapid phosphorylation of the free β-catenin in the cytosol. This reaction is catalyzed by a protein complex that includes the proteins axin, adenomatous polyposis coli (APC), casein kinase I (CK1), and glycogen synthase kinase 3 beta (GSK3β). When phosphorylated, β-catenin is recognized and ubiquitinated by a specific E3 ubiquitin ligase complex (SCF βTrCP), and degradation through the proteasome occurs (Figure 3A). Hence, one of the roles of the axin-APC complex is to maintain low cytosolic levels of β-catenin [19].

Figure 3.

Wnt/β-catenin signaling in the absence (A) or presence (B) of a Wnt ligand.

When a Wnt ligand binds a Fz receptor, in the presence of specific adapters known as low-density lipoprotein receptor-related proteins 5/6 (Lrp5/6), recruitment of the proteins Axin and Disheveled (DvI) occurs, and the formation of the protein complex that drives β-catenin degradation is inhibited. As a consequence, the levels of cytoplasmic β-catenin rise and the protein is transported into the nucleus where it forms a complex with the T cell factor (TCF) family of transcription factors, and activates transcription of its target genes [20] (Figure 3B).

A number of evidence demonstrate that K63-linked polyubiquitylation plays a role in the regulation of Wnt signaling although the precise molecular mechanisms that regulate these complex interactions have not been fully elucidated yet. We know that the formation of K63-linked polyubiquitin chains is promoted by the E2 protein complex Ubc13-Uev1a and recent evidence suggests that deletion of Ubc13 is associated with accumulation of β-catenin and increased transcription of Wnt target genes; however, the precise molecular mechanisms that drive these signaling events remain to be fully elucidated [21]. As largely known, deubiquitinating enzymes (DUBs) act in concert with ubiquitinating enzymes to precisely adjust the extent and duration of ubiquitin signals. Cellular experiments show that Trabid is a DUB protein able to reverse K63-linked hyperubiquitylation of the APC complex, thus acting as a positive regulator of the Wnt/β-catenin signaling [22].

Abnormal Wnt signaling underlies a wide range of pathological conditions in humans, including cancer. Hyperactivation of the Wnt pathway, for instance, is a characteristic of tumor cells from patients with cylindromatosis. These patients present mutations in the CYLD gene; this gene encodes for a DUB enzyme whose loss in human cells causes K63-linked hyperubiquitylation of the upstream Fz-binding effector protein Disheveled (Dvl) resulting in enhanced responsiveness to Wnt [23].

2.3. Lysine 63-linked ubiquitylation in membrane protein trafficking

Membrane proteins serve different functions: they allow cells to sense and/or interact with molecules in the extracellular space, confer a proper shape to the cell, regulate the osmotic pressure, and channel the passage of ions, endogenous compounds, xenobiotics, etc. Both the sorting and degradation of membrane-bound proteins are regulated at least in part by K63-linked protein ubiquitylation.

The synthesis of membrane protein takes place specifically on those ribosomes attached to the endoplasmic reticulum (ER). Once newly synthesized, proteins migrate from the inner lumen of the ER to the cis face of the Golgi apparatus (more proximal to the nucleus) where they undergo refinement and quality control to ensure proper folding. Within the Golgi apparatus, proteins are also sorted according to their predetermined cellular destination and finally secreted through the trans face (more distal to the nucleus). The migration of proteins through these cellular compartments is guided by lipid vesicles known as endosomes. Importantly, endosomes also coordinate the downregulation of cell-surface receptors through internalization of these proteins and subsequent degradation in the lysosomes.

A growing number of studies seem to suggest that K63-linked ubiquitin chains act as a signal for the internalization and intracellular sorting of integral membrane proteins [24]; more specifically, K63-linked ubiquitin chains have been shown to direct proteins to a specialized subclass of endosomes known as multivesicular bodies (MVBs). Once in the MVBs, proteins are either sent to the lysosomes for degradation, or secreted as exosomes via fusion with the plasma membrane; this process also serves to position membrane bound receptors to their specific location.

The first evidence that K63-linked ubiquitin chains could function as a signal to stimulate internalization of plasma membrane proteins through endocytosis and targeting into the lysosomal degradation pathway was acquired in Saccharomyces cerevisiae [25]. These preliminary observations led to the discovery of several mammalian proteins undergoing a similar regulatory mechanism, for example, the epidermal growth factor receptor (EGFR) [26], the human dopamine transporter (DAT) [27], the nerve growth factor receptor tyrosine receptor kinase A (TrkA) [28], major histocompatibility complex class I molecules [29], and the prolactin receptor [30] and possibly the low-density lipoprotein receptor (LDLR) [31].

Advertisement

3. Lysine 63-linked ubiquitylation in diseases

3.1. Lysine 63 ubiquitylation in immune disorders

Protein ubiquitylation has emerged as a key mechanism in the modulation of the immune system development and intensity of the immune responses [32, 33]. K63-linked ubiquitylation is involved in immune cell development since germline ablation of Ubc13 induces embryonic lethality [33]. Moreover, through the regulation of different intracellular signaling pathways, K63 ubiquitylation has emerged as critical for T cell differentiation [34]. K63 ubiquitylation chains have been described as fundamental for both the innate and adaptive immune systems given their involvement in master pathways controlling immune responses, such as NF-κB signaling [35] and MAPK activation [36], as previously described.

3.1.1. Lysine 63 ubiquitylation and adaptive immune response

The adaptive immune response, also called acquired immunity, refers to antigen-specific immune response; thus, antigen presentation induces the development of effective T- and B-cell responses. B-cells can be divided into memory B cells that express membrane-bound antibodies, or plasma B cells that can secrete antibodies to identify free pathogens circulating into the body. All B cells express a B cell receptor involved in antigen binding, internalization, and processing of antigens, other than activation of intracellular signaling pathways. T cells instead mature into the thymus where they start to express T cell receptors (TCRs) and CD4 and CD8 receptors. T cell receptors, assisted by CD4 or CD8 receptors, recognize antigens bound to certain major histocompatibility complex class 1 (MHCI) and class 2 (MHCII), expressed by antigen presenting cells such as macrophages and dendritic cells. Mature T cells can be mainly divided into Helper T cells, CD4+ cells involved in the activation of other immune cells, cytotoxic T cells, CD8+ that removes pathogens and infected cells, and T regulatory cells. T regulatory cells (Treg) play a central role in the regulation of the adaptive immune response and represent a T lymphocyte subpopulation that maintains tolerance to self-antigens and prevents autoimmune disease [37]. Tregs are produced in the thymus as a subpopulation of T cells and express a transcription factor (Forkhead box protein 3) involved in Treg development and function [38]. Tregs can also be induced from naive T cells in the periphery in the presence of transforming growth factor b (TGF-β).

K63 ubiquitylation is specifically involved in the suppressive function of Treg cells, and it has been described as fundamental for the immunosuppressive function of Tregs in murine models in vivo [39]. It is also well known that NF-κB signaling can modulate Treg cell differentiation [40]. Chang et al. demonstrated that Ubc13 deficiency in Treg, with the subsequent reduction in K63 ubiquitylation, impaired the in vivo suppressive function of these cells. Ubc13 deficiency in Ubc13Treg−KO mice, in fact, was able to influence the IKK signaling axis normally required for the expression of specific Treg functional factors, such as IL-10 and SOCS1 [39]. Both IL-10 and SOCS1 can specifically regulate Treg stability and inhibition activity.

Defects in Treg cells have been described in several human immune disorders including systemic lupus erythematosus (SLE). Treg disorders in SLE patients are characterized by abnormal peripheral tolerance that has been linked to a deficiency in the E3 ubiquitin ligase Cbl-b [41], involved in the regulation of T cell receptor signaling, during the induction of peripheral tolerance. Interestingly SLE patients were also characterized by an altered pattern of K63 ubiquitinated proteins in Tregs, with a decreased expression of K63 ubiquitinated proteins, related to increased pSTAT-3 expression [42]. These processes could be responsible for the loss of Treg suppressive capacity in SLE patients.

K63 ubiquitylation can also influence B cell receptor, T cell receptor, and IL-1 receptor (IL-1R)-mediated immune responses. Murine Ubc13-deficient T cells showed altered proliferation in response to diverse stimuli and impaired intracellular signaling altering the activation of both NF-κB and MAP kinases into T cells [43]. Also, murine Ubc13-deficient (Ubc13−/−) B cells showed impaired activation of the B cell receptor and CD40-induced activation, as well as Toll-like receptor mediated activation [44]. All this evidence underlies the meaning of this process in the mammalian immune response, thus indicating the importance of investigating K63 ubiquitylation in immune disorders.

3.1.2. Lysine 63 ubiquitylation and innate immune response

Innate immune response is triggered upon infections with pathogens such as bacteria, parasites, and viruses. It includes several mechanisms consisting in the physical and chemical barrier to infectious agents, in the activation of the complement cascade, and in the recruitment to the sites of infection of immune cells [such as macrophages and neutrophils] able to produce cytokines, thus inducing the inflammatory response. Innate immune response can also influence the adaptive immune system through antigen presentation.

It has been demonstrated that K63 ubiquitylation facilitates the innate immune signaling activated by diverse receptors such as Toll-like receptors, able to recognize pathogen components (lipopolysaccharide—LPS—from Gram-negative bacteria or lipoteichoic acid from Gram-positive bacteria), or cytokine receptors [45]. Also, in the regulation of the innate immune response, such as the adaptive immune response, the IKK-NF-κB pathway and MAPK activation plays a central role, since they are activated by the engagement of Toll-like receptors (TLRs).

An important component of the innate immune system is represented by natural killer (NK) cells, involved in the direct elimination of infected or transformed cells and able to secrete diverse cytokines, including IFN-γ, thus increasing the inflammatory response and the recruitment of immune cells. Also, IFN-γ production in NK cells is regulated by K63 ubiquitylation through its involvement in the NF-κB pathway [46], underlying once again the importance of this mechanism in the modulation of the immune response.

Ubiquitylation signaling has been described as specifically involved in the anti-viral innate immune response. In fact, the importance of the ubiquitylation pathway in the innate immune response has been validated by the discovery of some viruses encoding deubiquitinating proteases. Deubiquitinating enzymes (DUBs) catalyze the removal of ubiquitin from different cellular substrates, thus influencing several intracellular processes [47]. These deubiquitinating proteases produced by viruses, can lead to the suppression of the anti-viral immune response in order to promote viral replication [48].

Herpes Simplex Virus 1 (HSV1), a dsDNA virus belonging to the alpha-herpesvirus subfamily, can cause humans gingivostomatitis, cold sores, and herpetic keratitis. HSV1 dsDNA induces NF-κB signaling activation that promotes the anti-viral immune-response. However, to evade the innate immune system, these viruses encode for a DUB domain, called UL36USP, which is also similar to an open reading frame encoded by other viruses such as the human cytomegalovirus (HCMV) [49]. HCMV is a member of the beta-herpes virus subfamily, whose infection, normally asymptomatic, once reactivated can cause severe disease in immune-compromised and immune-suppressed individuals. Interestingly the UL36USP deubiquitinase activity inhibits NF-κB activation, by deubiquitinating IκBα, thus blocking its degradation and, consequently, finally quenching IFN production [50], a cytokine important for the anti-viral immune response.

Also, retroviral infections, including HIV-1, are mediated by modulation of the ubiquitylation system. In particular, the retrovirus factor TRIM5 (tripartite motif-containing protein 5) promotes innate immune signaling by activating, through K63 ubiquitin, MAP3K7 kinase complex with the subsequent stimulation of AP-1 and NF-κB signaling [51].

K63 ubiquitylation is also involved in bacterial cytoplasmic infections. These infections induce the cytosolic exposure of peptidoglycans and are characterized by the activation of the nuclear oligomerization domain 2 (NOD2) intracellular signaling. Polymorphisms in NOD2 have been associated with 15–30% of genetic Crohn’s disease [52], an inflammatory bowel disease that may affect any part of the gastrointestinal tract, from mouth to anus, characterized by a dysfunctional immune response to normal microbiota [53]. It has been demonstrated that the activation by NOD2 of the K63-specific E3 ubiquitin ligase TRAF6, leading to NF-κB stimulation, is seriously compromised in Crohn’s disease-patients with the NOD2 allele L1007insC [54]. In addition, NOD2 (nucleotide oligomerization domain 2) regulates the formation of K63-linked polyubiquitin chains on the I kappa kinase (IKK) scaffolding protein, NEMO.

Thus, ubiquitin-mediated regulation of the innate immune response could represent an important node in the management of pathogen infection and could symbolize a novel target for future therapies.

3.2. Lysine 63 ubiquitylation in diabetes and diabetic nephropathy

Diabetes mellitus is a metabolic disorder characterized by the reduction and altered function of pancreatic insulin-producing β-cells, and by organ damage [55]. Type 1 diabetes is induced by autoimmune destruction of β-cells responsible for insulin insufficiency, while type 2 diabetes is due to peripheral insulin resistance and subsequent β-cell expansion and hyperinsulinemia [56]. The number of diabetic patients is increasing decade by decade, and this high prevalence is registered worldwide with a projection of more than 438 million of diabetic patients with 7.8% prevalence by 2030 all over the world.

Chronic hyperglycemia and oxidative stress have been described as pro-apoptotic signals for pancreatic β-cell, thus influencing the metabolic state of these cells and the cell fate decisions [56]. Several intracellular signaling pathways can contribute to modulate β-cell function. Among these, also post-translational modification has been recognized to play a role. SUMOylation, a post-translational modification consisting in covalent attachment to target proteins of the small ubiquitin-like modifier (SUMO) peptides, has been recently described as a key event regulating β-cell survival and function [57]. Different cytokines can induce autoimmune destruction of pancreatic β-cell through the modulation of several intracellular signaling pathways characterized by the activation of phosphorylation and ubiquitylation events, including K63 ubiquitylation, in the cells. In fact, one of the factors involved into cytokine-mediated apoptosis of β-cell is represented by the mixed lineage kinase MLK3 [58], a pro-apoptotic factor involved in a cascade of events ultimately leading to mitochondrial outer membrane permeabilization, thus compromising mitochondrial integrity. Humphrey et al. demonstrated that IL-1β, one of the cytokines involved in the autoimmune destruction of pancreatic β-cells, stimulates K63-linked ubiquitylation of MLK3, thus promoting its activity and finally influencing the progression toward β-cell death [59]. Thus, K63 ubiquitylation could represent a potential target for therapeutic intervention in promoting β cell survival in diabetic patients.

Diabetes is also responsible for the insurgence of different complications such as retinopathy, cardiovascular diseases, and renal diseases. In the last years, the importance of the ubiquitylation pathway in diabetes and diabetic complications, such as cardiac diseases [60] and diabetic nephropathy [61, 62, 63, 64], has emerged significantly [65].

Hyperglycemia, hypertension, and other hemodynamic changes intensify the filtration and reabsorption processes and this can lead to kidney failure progressing toward end-stage renal disease (ESRD). The incidence of ESRD due to diabetes varies among countries between 15 and 45%, with a mean value of 33%, which means that 33% of patients are starting renal replacement therapy.

Renal damage in type 2 diabetic patients can be characterized by different patterns including diabetic glomerulosclerosis, vascular and ischemic glomerular changes, and other glomerulonephritis in the presence or absence of diabetic lesions [66]. Pure diabetic nephropathy is characterized by mesangial proliferation, podocyte loss, glomerular basal membrane thickening, and nodular extracellular matrix accumulation with the classical Kimmelstiel-Wilson lesions.

The specific role of K63 ubiquitylation in diabetic nephropathy has been recently described [67]. Hyperglycemic conditions induce in tubular cells an increased expression of specifically K63-ubiquitinated proteins. Also, kidney biopsies from diabetic nephropathy patients are characterized by increased K63 ubiquitylation at tubular cells when compared to diabetic patients without renal damage or patients with other nephritides such as membranous nephropathy. Interestingly, increased K63 ubiquitylation in glucose-stimulated tubular cells was able to promote epithelial to mesenchymal transition, a process already described as involved in diabetic nephropathy dysfunction [68]. Epithelial to mesenchymal transition represents a potential source of myofibroblasts involved in the progression of kidney fibrosis. Also, in in vivo kidney biopsies of diabetic nephropathy patients, tubular cells characterized by increased accumulation of K63 ubiquitinated proteins were also characterized by expression of mesenchymal markers [67], thus underlying the importance of K63 ubiquitylation in the progression of renal fibrosis in diabetic patients.

Other than in epithelial-to-mesenchymal transition, hyperglycemia-induced K63 ubiquitylation is also involved in the apoptotic death of tubular cells through the deregulation of autophagy. Autophagy is an intracellular process involved in degradation of damaged proteins/organelle or in the intracellular response to nutrient deprivation, stress, and extracellular environmental changes. In human glomerulopathies, changes in the ubiquitin-proteasome system have been correlated with autophagy [69]. Impaired autophagy has also been described as a characteristic feature of diabetics [70], and recently, the molecular mechanisms responsible for this alteration have been correlated to hyperglycemia-induced K63 ubiquitylation [71]. In diabetic nephropathy patients in vivo, those tubules characterized by increased expression of the autophagic factor LC3 were also characterized by increase in K63 ubiquitinated protein accumulation. Interestingly, accumulation of autophagic particles into tubular cells, due to K63 ubiquitinated protein accumulation, could be responsible for increased apoptosis of these cells, as observed both in vitro and in vivo in kidney biopsies from diabetic nephropathy patients [70]. Taken together, all this evidence support the role of K63 ubiquitylation in the progression of tubular damage in diabetic nephropathy patients, which could be responsible for the progression of kidney fibrosis and for the induction of apoptosis of tubular cells with the consequent reduction of renal function.

3.3. Lysine 63 ubiquitylation and neurodegenerative disorders

Neurodegenerative disorders like Parkinson’s disease or dementia or Alzheimer’s disease are, in some cases, characterized by the presence of insoluble deposits in neurons containing components of the ubiquitin-proteasome system. It has been reported, in fact, that the ubiquitylation pathways play an important role in the pathogenesis of these diseases.

The most common cause of familial Parkinson’s disease is characterized by mutations in Parkin [72], a ubiquitin ligase whose loss of function leads to both toxic accumulation of its substrates [73], and impaired formation of Lewy bodies, fibrillary masses of molecules implicated in the formation and degradation of alpha-synuclein aggregates [74]. Alpha-synuclein is a protein expressed in neurons involved in the formation of synaptic vesicles in presynaptic terminals and in the release of the dopamine, one of the brain’s neurotransmitters. It has been demonstrated that K63 ubiquitylation plays an important role in the generation of these aggregates. Lim et al. observed that parkin-mediated ubiquitylation of proteins within Lewy-body-like inclusions was augmented by K63 ubiquitylation and occurs mainly through K63 linkages [75]. Interestingly, it has been demonstrated that the ubiquitin hydrolase UCHL1 is able to promote also K63-linked ubiquitylation of alpha-synuclein [76]. Thus, K63 ubiquitylation could represent a mechanism by which protein inclusion can occur and by which proteins are stabilized [77] forming aggregates observed in neurodegenerative disorders including Parkinson disease and dementia.

Interestingly, K63 ubiquitylation can contribute not only to inclusion biogenesis, but also to the clearance of inclusions. Intracellular alpha-synuclein can be ubiquitinated in K63 residues also by the E3 ubiquitin ligase Nedd4-1 [neural precursor cell expressed developmentally down-regulated protein 4-1], thus inducing its degradation by the endo-lysosomal pathway [78]. The first evidence of the role of K63 ubiquitylation in modulating autophagy clearance of aggregates in neurodegenerative disorders was described by Tan et al. in 2008 [79]. These authors demonstrated that K63 polyubiquitin chains linked to protein inclusions represent a target, driving aggregates to the clearance by autophagy.

Poly-ubiquitinated proteins can also be accumulated into the mitochondria during proteolytic stress and can be responsible for mitochondria-mediated cell death during proteasomal dysfunction, as demonstrated in an in vitro model of dopaminergic degeneration [80]. Interestingly, also monoamine oxidases (MAOs), located on the outer mitochondrial membrane and involved in the control of the neurotransmitters levels in the brain, can induce K63 ubiquitylation of mitochondrial proteins and promote autophagy of damaged organelles in neuroblastoma cells [81].

Taken together, these results evidenced the importance of K63 ubiquitylation in neurodegenerative disorders and open novel scenarios for the treatment of these diseases.

3.4. Lysine 63 ubiquitylation in cancer

The involvement of ubiquitin proteasome system dysregulation in the degradations of apoptotic proteins and subsequent induction of tumor formation has been well established, whereas, the finding that nonproteolytic ubiquitylation has a role in cancer and metastasis is recent.

With respect to K63-linked ubiquitylation, it was recently shown that both Ube2v1 and its partner Ubc13 are overexpressed in breast cancer, ovarian cancer, prostate cancer, and colorectal cancer, as well as in lymphoma [82, 83]. In breast cancer in particular, Ubc13 was identified as a key protein for metastasis spreading to the lung. This action appears to be mediated through TGF-β-induced activation of the TAK1-p38 MAP Kinase cascade. Notably, it was also demonstrated that in vivo inhibition of UBE2v1 through RNA interference can prevent breast tumor growth and metastasis formation [84, 85].

The role of Ubc13-Uev1A in tumorigenesis was also suggested by Pulvino et al., in accordance with the finding that small-molecule inhibitor of Ubc13-Uev1A interaction, known as NSC697923, can inhibit proliferation and survival of diffuse large B-cell lymphoma cells via inhibition of the Nf-KB signaling in these cells [86].

Additional proof that K63-linked ubiquitylation is involved in the regulation of tumorigenesis comes from the observation that several E3s and DUBs involved in this particular type of ubiquitylation/deubiquitylation are important regulators of proteins that guide cell cycle, DNA damage, and cell death [87, 88, 89, 90]. For a complete review on the role of ubiquitylation on tumor formation and metastasis, refer to the work by Gallo et al. [83].

Advertisement

4. Conclusions

The role of post-translational modification in the regulation of cell behavior in response to diverse stimuli is emerging more and more and provides additional information regarding layers of regulation in cells. In this scenario, K63 ubiquitylation is starting to play an increasingly important role, since several crucial mechanisms involved in cell-signaling are regulated by this type of post-translational modification.

A vast group of human disorders such as Alzheimer’s disease, Parkinson’s disease, Type II Diabetes, and cancer have been investigated. In this chapter, we focused on the description of the main intracellular signaling pathways regulated by post-translational modification with respect to K63-linked ubiquitylation. The goal of this overview was to summarize the main findings regarding the main regulatory mechanisms that contribute to the disease pathogenesis or progression.

A better understanding of the different layers of regulation within molecular pathways, including the ubiquitin code, will indeed clear the path to a more precise manipulation of those aberrant post-translational signaling events that cause disease, marking a new era in therapeutic management and personalized medicine.

The specific targets of these modifications, influencing intracellular signaling pathways and cellular behavior, represent the future of target therapy; thus, the investigation of these mechanisms should be further analyzed in depth.

Advertisement

Acknowledgments

The personnel working on this project have received funding from the Innovative Medicines Initiative 2 Joint Undertaking under grant agreement No 115974. This Joint Undertaking receives support from the European Union’s Horizon 2020 research and innovation program and EFPIA with JDRF.

Advertisement

Conflict of interest

None.

References

  1. 1. Ikeda F, Dikic I. Atypical ubiquitin chains: New molecular signals. ‘Protein Modifications: Beyond the Usual Suspects’ review series. EMBO Reports. 2008;9(6):536-542
  2. 2. Akutsu M, Dikic I, Bremm A. Ubiquitin chain diversity at a glance. Journal of Cell Science. 2016;129(5):875-880
  3. 3. Chen ZJ, Sun LJ. Nonproteolytic functions of ubiquitin in cell signaling. Molecular Cell. 2009;33(3):275-286
  4. 4. Ye Y, Rape M. Building ubiquitin chains: E2 enzymes at work. Nature Reviews. Molecular Cell Biology. 2009;10(11):755-764
  5. 5. Schulman BA, Harper JW. Ubiquitin-like protein activation by E1 enzymes: The apex for downstream signalling pathways. Nature Reviews. Molecular Cell Biology. 2009;10(5):319-331
  6. 6. Komander D, Rape M. The ubiquitin code. Annual Review of Biochemistry. 2012;81:203-229
  7. 7. Deshaies RJ, Joazeiro CA. RING domain E3 ubiquitin ligases. Annual Review of Biochemistry. 2009;78:399-434
  8. 8. Hodge CD, Spyracopoulos L, Glover JN. Ubc13: The Lys63 ubiquitin chain building machine. Oncotarget. 2016;7(39):64471-64504
  9. 9. Swatek KN, Komander D. Ubiquitin modifications. Cell Research. 2016;26(4):399-422
  10. 10. Swaney DL, Rodriguez-Mias RA, Villen J. Phosphorylation of ubiquitin at Ser65 affects its polymerization, targets, and proteome-wide turnover. EMBO Reports. 2015;16(9):1131-1144
  11. 11. Amerik AY, Hochstrasser M. Mechanism and function of deubiquitinating enzymes. Biochimica et Biophysica Acta. 2004;1695(1-3):189-207
  12. 12. Hayden MS, Ghosh S. Shared principles in NF-kappaB signaling. Cell. 2008;132(3):344-362
  13. 13. Oeckinghaus A, Ghosh S. The NF-kappaB family of transcription factors and its regulation. Cold Spring Harbor Perspectives in Biology. 2009;1(4):a000034
  14. 14. Chen ZJ. Ubiquitin signalling in the NF-kappaB pathway. Nature Cell Biology. 2005;7(8):758-765
  15. 15. Keating SE, Bowie AG. Role of non-degradative ubiquitination in interleukin-1 and toll-like receptor signaling. The Journal of Biological Chemistry. 2009;284(13):8211-8215
  16. 16. Deng L, Wang C, Spencer E, Yang L, Braun A, You J, et al. Activation of the IkappaB kinase complex by TRAF6 requires a dimeric ubiquitin-conjugating enzyme complex and a unique polyubiquitin chain. Cell. 2000;103(2):351-361
  17. 17. Wang C, Deng L, Hong M, Akkaraju GR, Inoue J, Chen ZJ. TAK1 is a ubiquitin-dependent kinase of MKK and IKK. Nature. 2001;412(6844):346-351
  18. 18. Wajant H, Scheurich P. TNFR1-induced activation of the classical NF-kappaB pathway. The FEBS Journal. 2011;278(6):862-876
  19. 19. Tauriello DV, Maurice MM. The various roles of ubiquitin in Wnt pathway regulation. Cell Cycle. 2010;9(18):3700-3709
  20. 20. Clevers H. Wnt/beta-catenin signaling in development and disease. Cell. 2006;127(3):469-480
  21. 21. Wu X, Yamamoto M, Akira S, Sun SC. Regulation of hematopoiesis by the K63-specific ubiquitin-conjugating enzyme Ubc13. Proceedings of the National Academy of Sciences of the United States of America. 2009;106(49):20836-20841
  22. 22. Tran H, Hamada F, Schwarz-Romond T, Bienz M. Trabid, a new positive regulator of Wnt-induced transcription with preference for binding and cleaving K63-linked ubiquitin chains. Genes & Development. 2008;22(4):528-542
  23. 23. Tauriello DV, Haegebarth A, Kuper I, Edelmann MJ, Henraat M, Canninga-van Dijk MR, et al. Loss of the tumor suppressor CYLD enhances Wnt/beta-catenin signaling through K63-linked ubiquitination of Dvl. Molecular Cell. 2010;37(5):607-619
  24. 24. Mukhopadhyay D, Riezman H. Proteasome-independent functions of ubiquitin in endocytosis and signaling. Science. 2007;315(5809):201-205
  25. 25. Galan JM, Haguenauer-Tsapis R. Ubiquitin lys63 is involved in ubiquitination of a yeast plasma membrane protein. The EMBO Journal. 1997;16(19):5847-5854
  26. 26. Huang F, Zeng X, Kim W, Balasubramani M, Fortian A, Gygi SP, et al. Lysine 63-linked polyubiquitination is required for EGF receptor degradation. Proceedings of the National Academy of Sciences of the United States of America. 2013;110(39):15722-15727
  27. 27. Vina-Vilaseca A, Sorkin A. Lysine 63-linked polyubiquitination of the dopamine transporter requires WW3 and WW4 domains of Nedd4-2 and UBE2D ubiquitin-conjugating enzymes. The Journal of Biological Chemistry. 2010;285(10):7645-7656
  28. 28. Geetha T, Jiang J, Wooten MW. Lysine 63 polyubiquitination of the nerve growth factor receptor TrkA directs internalization and signaling. Molecular Cell. 2005;20(2):301-312
  29. 29. Duncan LM, Piper S, Dodd RB, Saville MK, Sanderson CM, Luzio JP, et al. Lysine-63-linked ubiquitination is required for endolysosomal degradation of class I molecules. The EMBO Journal. 2006;25(8):1635-1645
  30. 30. Varghese B, Barriere H, Carbone CJ, Banerjee A, Swaminathan G, Plotnikov A, et al. Polyubiquitination of prolactin receptor stimulates its internalization, postinternalization sorting, and degradation via the lysosomal pathway. Molecular and Cellular Biology. 2008;28(17):5275-5287
  31. 31. Zhang L, Xu M, Scotti E, Chen ZJ, Tontonoz P. Both K63 and K48 ubiquitin linkages signal lysosomal degradation of the LDL receptor. Journal of Lipid Research. 2013;54(5):1410-1420
  32. 32. Bhoj VG, Chen ZJ. Ubiquitylation in innate and adaptive immunity. Nature. 2009;458(7237):430-437
  33. 33. Wu X, Karin M. Emerging roles of Lys63-linked polyubiquitylation in immune responses. Immunological Reviews. 2015;266(1):161-174
  34. 34. Chen Z, Luo X, Lu Y, Zhu T, Wang J, Tsun A, et al. Ubiquitination signals critical to regulatory T cell development and function. International Immunopharmacology. 2013;16(3):348-352
  35. 35. Ajibade AA, Wang HY, Wang RF. Cell type-specific function of TAK1 in innate immune signaling. Trends in Immunology. 2013;34(7):307-316
  36. 36. Matsuzawa A, Tseng PH, Vallabhapurapu S, Luo JL, Zhang W, Wang H, et al. Essential cytoplasmic translocation of a cytokine receptor-assembled signaling complex. Science. 2008;321(5889):663-668
  37. 37. Schmidt A, Oberle N, Krammer PH. Molecular mechanisms of treg-mediated T cell suppression. Frontiers in Immunology. 2012;3:51
  38. 38. Sakaguchi S, Yamaguchi T, Nomura T, Ono M. Regulatory T cells and immune tolerance. Cell. 2008;133(5):775-787
  39. 39. Chang JH, Xiao Y, Hu H, Jin J, Yu J, Zhou X, et al. Ubc13 maintains the suppressive function of regulatory T cells and prevents their conversion into effector-like T cells. Nature Immunology. 2012;13(5):481-490
  40. 40. Long M, Park SG, Strickland I, Hayden MS, Ghosh S. Nuclear factor-kappaB modulates regulatory T cell development by directly regulating expression of Foxp3 transcription factor. Immunity. 2009;31(6):921-931
  41. 41. Gomez-Martin D, Ibarra-Sanchez M, Romo-Tena J, Cruz-Ruiz J, Esparza-Lopez J, Galindo-Campos M, et al. Casitas B lineage lymphoma b is a key regulator of peripheral tolerance in systemic lupus erythematosus. Arthritis and Rheumatism. 2013;65(4):1032-1042
  42. 42. Romo-Tena J, Rajme-Lopez S, Aparicio-Vera L, Alcocer-Varela J, Gomez-Martin D. Lys63-polyubiquitination by the E3 ligase casitas B-lineage lymphoma-b (Cbl-b) modulates peripheral regulatory T cell tolerance in patients with systemic lupus erythematosus. Clinical and Experimental Immunology. 2018;191(1):42-49
  43. 43. Yamamoto M, Sato S, Saitoh T, Sakurai H, Uematsu S, Kawai T, et al. Cutting edge: Pivotal function of Ubc13 in thymocyte TCR signaling. Journal of Immunology. 2006;177(11):7520-7524
  44. 44. Yamamoto M, Okamoto T, Takeda K, Sato S, Sanjo H, Uematsu S, et al. Key function for the Ubc13 E2 ubiquitin-conjugating enzyme in immune receptor signaling. Nature Immunology. 2006;7(9):962-970
  45. 45. Fiil BK, Gyrd-Hansen M. Met1-linked ubiquitination in immune signalling. The FEBS Journal. 2014;281(19):4337-4350
  46. 46. Chen J, Hao L, Li C, Ye B, Du Y, Zhang H, et al. The endoplasmic reticulum adaptor protein ERAdP initiates NK cell activation via the Ubc13-mediated NF-kappaB pathway. Journal of Immunology. 2015;194(3):1292-1303
  47. 47. Komander D, Clague MJ, Urbe S. Breaking the chains: Structure and function of the deubiquitinases. Nature Reviews. Molecular Cell Biology. 2009;10(8):550-563
  48. 48. Bailey-Elkin BA, Knaap RCM, Kikkert M, Mark BL. Structure and function of viral deubiquitinating enzymes. Journal of Molecular Biology. 2017;429(22):3441-3470
  49. 49. Kattenhorn LM, Korbel GA, Kessler BM, Spooner E, Ploegh HL. A deubiquitinating enzyme encoded by HSV-1 belongs to a family of cysteine proteases that is conserved across the family Herpesviridae. Molecular Cell. 2005;19(4):547-557
  50. 50. Ye R, Su C, Xu H, Zheng C. Herpes simplex virus 1 ubiquitin-specific protease UL36 abrogates NF-kappaB activation in DNA sensing signal pathway. Journal of Virology. 2017;91(5)
  51. 51. Pertel T, Hausmann S, Morger D, Zuger S, Guerra J, Lascano J, et al. TRIM5 is an innate immune sensor for the retrovirus capsid lattice. Nature. 2011;472(7343):361-365
  52. 52. Fritz JH, Ferrero RL, Philpott DJ, Girardin SE. Nod-like proteins in immunity, inflammation and disease. Nature Immunology. 2006;7(12):1250-1257
  53. 53. Podolsky DK. Inflammatory bowel disease. The New England Journal of Medicine. 2002;347(6):417-429
  54. 54. Abbott DW, Yang Y, Hutti JE, Madhavarapu S, Kelliher MA, Cantley LC. Coordinated regulation of Toll-like receptor and NOD2 signaling by K63-linked polyubiquitin chains. Molecular and Cellular Biology. 2007;27(17):6012-6025
  55. 55. Weir GC, Bonner-Weir S. Islet beta cell mass in diabetes and how it relates to function, birth, and death. Annals of the New York Academy of Sciences. 2013;1281:92-105
  56. 56. Remedi MS, Emfinger C. Pancreatic beta-cell identity in diabetes. Diabetes, Obesity & Metabolism. 2016;18 Suppl 1:110-116
  57. 57. MacDonald PE. A post-translational balancing act: The good and the bad of SUMOylation in pancreatic islets. Diabetologia. 2018;61(4):775-779
  58. 58. Humphrey RK, Newcomb CJ, Yu SM, Hao E, Yu D, Krajewski S, et al. Mixed lineage kinase-3 stabilizes and functionally cooperates with TRIBBLES-3 to compromise mitochondrial integrity in cytokine-induced death of pancreatic beta cells. The Journal of Biological Chemistry. 2010;285(29):22426-22436
  59. 59. Humphrey RK, Yu SM, Bellary A, Gonuguntla S, Yebra M, Jhala US. Lysine 63-linked ubiquitination modulates mixed lineage kinase-3 interaction with JIP1 scaffold protein in cytokine-induced pancreatic beta cell death. The Journal of Biological Chemistry. 2013;288(4):2428-2440
  60. 60. Portbury AL, Ronnebaum SM, Zungu M, Patterson C, Willis MS. Back to your heart: Ubiquitin proteasome system-regulated signal transduction. Journal of Molecular and Cellular Cardiology. 2012;52(3):526-537
  61. 61. Pontrelli P, Oranger A, Barozzino M, Conserva F, Papale M, Gesualdo L. The pathological role of the ubiquitination pathway in diabetic nephropathy. Minerva Medica. 2018;109(1):53-67
  62. 62. Goru SK, Kadakol A, Gaikwad AB. Hidden targets of ubiquitin proteasome system: To prevent diabetic nephropathy. Pharmacological Research. 2017;120:170-179
  63. 63. Yang XD, Xiang DX, Yang YY. Role of E3 ubiquitin ligases in insulin resistance. Diabetes, Obesity & Metabolism. 2016;18(8):747-754
  64. 64. Gao C, Huang W, Kanasaki K, Xu Y. The role of ubiquitination and sumoylation in diabetic nephropathy. BioMed Research International. 2014;2014:160692
  65. 65. Aghdam SY, Gurel Z, Ghaffarieh A, Sorenson CM, Sheibani N. High glucose and diabetes modulate cellular proteasome function: Implications in the pathogenesis of diabetes complications. Biochemical and Biophysical Research Communications. 2013;432(2):339-344
  66. 66. Mazzucco G, Bertani T, Fortunato M, Bernardi M, Leutner M, Boldorini R, et al. Different patterns of renal damage in type 2 diabetes mellitus: A multicentric study on 393 biopsies. American Journal of Kidney Diseases. 2002;39(4):713-720
  67. 67. Pontrelli P, Conserva F, Papale M, Oranger A, Barozzino M, Vocino G, et al. Lysine 63 ubiquitination is involved in the progression of tubular damage in diabetic nephropathy. The FASEB Journal. 2017;31(1):308-319
  68. 68. Zhao L, Chi L, Zhao J, Wang X, Chen Z, Meng L, et al. Serum response factor provokes epithelial-mesenchymal transition in renal tubular epithelial cells of diabetic nephropathy. Physiological Genomics. 2016;48(8):580-588
  69. 69. Cybulsky AV. The intersecting roles of endoplasmic reticulum stress, ubiquitin-proteasome system, and autophagy in the pathogenesis of proteinuric kidney disease. Kidney International. 2013;84(1):25-33
  70. 70. Yamahara K, Kume S, Koya D, Tanaka Y, Morita Y, Chin-Kanasaki M, et al. Obesity-mediated autophagy insufficiency exacerbates proteinuria-induced tubulointerstitial lesions. Journal of the American Society of Nephrology. 2013;24(11):1769-1781
  71. 71. Pontrelli P, Oranger A, Barozzino M, Divella C, Conserva F, Fiore MG, et al. Deregulation of autophagy under hyperglycemic conditions is dependent on increased lysine 63 ubiquitination: A candidate mechanism in the progression of diabetic nephropathy. Journal of Molecular Medicine. 2018;96(7):645-659
  72. 72. Lucking CB, Durr A, Bonifati V, Vaughan J, De Michele G, Gasser T, et al. Association between early-onset Parkinson's disease and mutations in the parkin gene. The New England Journal of Medicine. 2000;342(21):1560-1567
  73. 73. Dawson TM. New animal models for Parkinson's disease. Cell. 2000;101(2):115-118
  74. 74. Wakabayashi K, Tanji K, Mori F, Takahashi H. The Lewy body in Parkinson's disease: Molecules implicated in the formation and degradation of alpha-synuclein aggregates. Neuropathology. 2007;27(5):494-506
  75. 75. Lim KL, Chew KC, Tan JM, Wang C, Chung KK, Zhang Y, et al. Parkin mediates nonclassical, proteasomal-independent ubiquitination of synphilin-1: Implications for Lewy body formation. The Journal of Neuroscience. 2005;25(8):2002-2009
  76. 76. Liu Y, Fallon L, Lashuel HA, Liu Z, Lansbury PT Jr. The UCH-L1 gene encodes two opposing enzymatic activities that affect alpha-synuclein degradation and Parkinson’s disease susceptibility. Cell. 2002;111(2):209-218
  77. 77. Lim KL, Dawson VL, Dawson TM. Parkin-mediated lysine 63-linked polyubiquitination: A link to protein inclusions formation in Parkinson's and other conformational diseases? Neurobiology of Aging. 2006;27(4):524-529
  78. 78. Sugeno N, Hasegawa T, Tanaka N, Fukuda M, Wakabayashi K, Oshima R, et al. Lys-63-linked ubiquitination by E3 ubiquitin ligase Nedd4-1 facilitates endosomal sequestration of internalized alpha-synuclein. The Journal of Biological Chemistry. 2014;289(26):18137-18151
  79. 79. Tan JM, Wong ES, Kirkpatrick DS, Pletnikova O, Ko HS, Tay SP, et al. Lysine 63-linked ubiquitination promotes the formation and autophagic clearance of protein inclusions associated with neurodegenerative diseases. Human Molecular Genetics. 2008;17(3):431-439
  80. 80. Sun F, Kanthasamy A, Anantharam V, Kanthasamy AG. Mitochondrial accumulation of polyubiquitinated proteins and differential regulation of apoptosis by polyubiquitination sites Lys-48 and -63. Journal of Cellular and Molecular Medicine. 2009;13(8B):1632-1643
  81. 81. Ugun-Klusek A, Theodosi TS, Fitzgerald JC, Burte F, Ufer C, Boocock DJ, et al. Monoamine oxidase-A promotes protective autophagy in human SH-SY5Y neuroblastoma cells through Bcl-2 phosphorylation. Redox Biology. 2018;20:167-181
  82. 82. Ma L, Broomfield S, Lavery C, Lin SL, Xiao W, Bacchetti S. Up-regulation of CIR1/CROC1 expression upon cell immortalization and in tumor-derived human cell lines. Oncogene. 1998;17(10):1321-1326
  83. 83. Gallo LH, Ko J, Donoghue DJ. The importance of regulatory ubiquitination in cancer and metastasis. Cell Cycle. 2017;16(7):634-648
  84. 84. Wu X, Zhang W, Font-Burgada J, Palmer T, Hamil AS, Biswas SK, et al. Ubiquitin-conjugating enzyme Ubc13 controls breast cancer metastasis through a TAK1-p38 MAP kinase cascade. Proceedings of the National Academy of Sciences of the United States of America. 2014;111(38):13870-13875
  85. 85. Wu Z, Shen S, Zhang Z, Zhang W, Xiao W. Ubiquitin-conjugating enzyme complex Uev1A-Ubc13 promotes breast cancer metastasis through nuclear factor-small ka, CyrillicB mediated matrix metalloproteinase-1 gene regulation. Breast Cancer Research. 2014;16(4):R75
  86. 86. Pulvino M, Liang Y, Oleksyn D, DeRan M, Van Pelt E, Shapiro J, et al. Inhibition of proliferation and survival of diffuse large B-cell lymphoma cells by a small-molecule inhibitor of the ubiquitin-conjugating enzyme Ubc13-Uev1A. Blood. 2012;120(8):1668-1677
  87. 87. Wu HT, Kuo YC, Hung JJ, Huang CH, Chen WY, Chou TY, et al. K63-polyubiquitinated HAUSP deubiquitinates HIF-1alpha and dictates H3K56 acetylation promoting hypoxia-induced tumour progression. Nature Communications. 2016;7:13644
  88. 88. Ng HM, Wei L, Lan L, Huen MS. The Lys63-deubiquitylating enzyme BRCC36 limits DNA break processing and repair. The Journal of Biological Chemistry. 2016;291(31):16197-16207
  89. 89. Wang B, Jie Z, Joo D, Ordureau A, Liu P, Gan W, et al. TRAF2 and OTUD7B govern a ubiquitin-dependent switch that regulates mTORC2 signalling. Nature. 2017;545(7654):365-369
  90. 90. Yao F, Zhou Z, Kim J, Hang Q , Xiao Z, Ton BN, et al. SKP2- and OTUD1-regulated non-proteolytic ubiquitination of YAP promotes YAP nuclear localization and activity. Nature Communications. 2018;9(1):2269

Written By

Paola Pontrelli, Francesca Conserva and Loreto Gesualdo

Submitted: 27 August 2018 Reviewed: 19 December 2018 Published: 01 February 2019