Submit manuscript...
MOJ
eISSN: 2374-6912

Cell Science & Report

Correspondence:

Received: January 01, 1970 | Published: ,

Citation: DOI:

Download PDF

Abstract

Neurodegenerative disorders (NDDs) are progressive and chronic disorders characterized by destruction of neurons in sensory, motor and cognitive systems. Free radical’s accumulation, oxidative stress, ER stress and a dysfunctional ubiquitin proteasome system can regulate prognosis in NDDs. Oxidative trauma in the brain can result from high rate of oxidative metabolism in contrast to the diminished functional levels of the antioxidant enzymes responsible for detoxification. Endoplasmic Reticulum (ER) advocates a degree of control on cellular parameters such as proper protein folding, posttranslational modification and subsequent protein trafficking in order to maintain normal cellular homeostasis. However, an abnormal ER functioning can lead to loss of integrity of the ER thus resulting in ER stress. In addition to this impairment in the ubiquitin proteasome system (UPS) machinery results in the accumulation of toxic proteins in the brain thus resulting in severe neuronal trauma and subsequent damage. This review explores the disease critical interactions and roles of three critical NDD determinants viz. oxidative stress, ER stress and UPS dysfunction in neurodegenerative conditions.

Keywords: ndd, free radicals, ros, oxidative stress, er stress, ups, e3-ligases, mitochondrial dysfunction

Abbreviations

NDD, neurodegenerative disorders; ER, endoplasmic reticulum; UPS, ubiquitin proteasome system; ROS, reactive oxygen species; AD, alzheimer’s disease; PD, parkinson’s disease; MS, multiple sclerosis; HD, huntington’s disease; ALS, amyotrophic lateral sclerosis; PS, presenilin; APP, amyloid beta (a4) precursor protein; MARK1, microtubule affinity-regulating kinase 1; SOD-1, superoxide dismutase 1; VAMP, vesicle-associated membrane protein; ALS2, amyotrophic lateral sclerosis 2; DCTN1, dynactin 1; FUS, fus rna binding protein; TDP-43, tar dna binding protein; PERK, phosphorylation of the er stress kinases; IRE 1, inositol-requiring enzyme 1; SCA, spinocerebellar ataxia; GSK, glycogen synthase Kinase; ASK1, apoptosis signal-regulating kinase 1; JNK, c-jun nh2-terminal kinase; Keap1, kelch-like ech-associated protein 1; MGRN1, mahogunin ring finger 1, E3 ubiquitin protein ligase; MYCBP2, myc binding protein 2, E3 ubiquitin protein ligase; UHRF2, ubiquitin-like with phd and ring finger domains 2; ZNRF1, zinc and ring finger 1, E3 ubiquitin protein ligase; NEDD4, neural precursor cell expressed, developmentally down-regulated 4, E3 ubiquitin protein ligase; NEDD4L, neural precursor cell expressed, developmentally down-regulated 4-like, E3 ubiquitin protein Ligase; HECTD2, hect domain containing e3 ubiquitin protein ligase 2; PJA2, praja ring finger 2, E3, ubiquitin protein ligase; RNF19, ring finger protein 19a, rbr e3 ubiquitin protein ligase; HECTD1, hect domain containing e3 ubiquitin protein ligase 1; MULAN, mitochondrial e3 ubiquitin protein ligase 1; HACE1, hect domain and ankyrin repeat containing e3 ubiquitin protein ligase 1; TRIM13, tripartite motif containing 13; AIMP2, aminoacyl trna synthetase complex-interacting multifunctional protein 2; NRF2, nuclear factor, erythroid 2-like 2; DVL1, dishevelled segment polarity protein 1; MYC, v-myc avian myelocytomatosis viral oncogene homolog; TSC2, tuberous sclerosis 2; FBXO45, f-box protein 45; PCNP, pest proteolytic signal containing nuclear protein; SMAD2, smad family member 2

Introduction

Neurodegenerative disorders (NDDs) are characterized by the gradual and progressive loss of neurons and neuronal death that ultimately leads to deficient nervous system functioning. It can result due to diverse factors such as oxidative stress, ER stress, mitochondrial dysfunction, impaired ubiquitin proteasomal system and several other determinants such as endocrine conditions, gender, poor education, inflammation, stroke, smoking, hypertension, diabetes, infection, head trauma, depression, tumors, vitamin deficiencies, immune and metabolic conditions, chemical exposure, accumulation of reactive oxygen species (ROS), loss of mitochondrial membrane potential, and ATP depletion. The two hit hypothesis of neurodegeneration states that neuronal cells that have been subjected to a severely stress once, becomes more vulnerable to the negative impact of a second hit and the effect of the toxicity of both the hits of severe stress may be synergistic in nature. Most common neurodegenerative diseases include Alzheimer`s disease (AD), Parkinson`s disease (PD), Huntington`s disease (HD), Schizophrenia, Amyotrophic lateral sclerosis (ALS) and Multiple Sclerosis (MS).1–5

Oxidative stress plays a critical role in the progression of several age related brain disorders. Severe oxidative trauma to the neurons can result in neuronal dysfunction and death. However, the neuronal cells are equipped with an arsenal of protective mechanism to prevent the damaging effects of oxidative stress on neuronal integrity and homeostasis. The removal of aberrantly functioning proteins by proteolysis and the synthesis of new and protective counterparts are critical during periods of continuous oxidative trauma.6 Reactive oxygen species (ROS) can result in oxidative stress and subsequently lead to mitochondrial dysfunction. Moreover, disturbed equilibrium between pro-oxidant/antioxidant homeostasis can generate ROS and free radicals which are detrimental for neurons. ROS in turn modulates the functionality of antioxidants and biomolecules thus leading to neuronal dysfunction and advances the brain towards progressive neurodegeneration.7–10

Endoplasmic reticulum (ER) mediated stress on the other side results from disturbances in the structural integrity and function of the ER, thus leading to the accumulation of misfolded proteins and deviations in the calcium homeostasis. The endoplasmic reticulum (ER) acts as protein quality control in the secretory pathway to prevent protein misfolding and aggregation. Under conditions of stress, the ER mediated machinery can reestablish homeostasis by sophistically regulating various transcriptionally and translationally mediated signaling networks and proteins.11 The normal ER response is depicted by reduction in damaged proteins levels, caused by translational attenuation, induction of ER chaperones and misfolded proteins proteolysis. However, under prolonged or provoked ER stress ambience, can lead to the activation of apoptotic pathways resulting in neuronal death.. Therefore, ER stress situation and remains a subject of curious debate involving the pathogenesis of common NDDs such as Parkinson’s disease (PD) and Alzheimer’s disease (AD).12

Improper functioning of the Ubiquitin proteasome system (UPS) under conditions of severe ER and oxidative trauma leads to the derogatory accumulation of damaged and misfiring stress consistent proteins. The UPS in collaboration with chaperones and co-chaperones constitute the regulatory mechanism responsible for neuronal quality control and survival. In addition, UPS can also operate as machinery for protein quality control and degradation in conjunction with autophagy, Dysfunctional UPS has been held capable in various NDDs and recent reports on Drosophila suggested that the role of UPS in protein turnover is essential for maintaining axon guidance, synaptic function and growth, axon pruning, and neuronal maintenance.13 Under the ambit of this review we have made an attempt to explore the role of oxidative stress, ER stress and UPS dysfunction respectively in neurodegenerative conditions. This interaction shall than be crucial in embellishing the development of potential neurotherapeutics.

Role of oxidative stress in neurodegeneration

Oxidative stress (OS) condition in the brain results from imbalance between ROS and the body’s detoxification mechanism, which results in accumulation of ROS and subsequent neuronal damage. Hence, the outcome of oxidative stress on neuronal cells depends upon the ability of the cell to maintain oxidative homeostasis. High stress levels can cause ATP depletion, necrosis and prevent apoptotic cell death.14

Any disproportion in the usual redox state can result in toxicity via the activation of peroxides and free radicals which in turn damages lipids, proteins and cellular DNA. A mammalian cell as a consequence of mitochondrial aerobic respiration generates superoxide radical. Superoxide is sequentially reduced to hydroxyl radicals and hydrogen peroxide that cause severe traumatic injury to the DNA thus leading to mutations, which might be causative factors leading to severe neurodegeneration.15

Reactive oxygen species (ROS) also plays a discrete role in cell signaling, by a mechanism known as redox signaling. In order to sustain proper cellular homeostasis, a balance must be reached between ROS production and consumption. Therefore, it is obvious that free radicals need to either be reduced and detoxified by converting them into metabolically nondestructive molecules or be neutralized right after their generation. Any aberration in the cellular antioxidant defense system, which protects the neurons from free radical assaults, therefore can lead to neurodegenerative conditions and aging.16

Brain is the most metabolically active organ of the body that including the spinal cord comprises the central nervous system (CNS), which even in resting condition consumes an estimated 20-22% of the total oxygen uptake. In addition, during active state the brain oxygen demand considerably rises in order to establish normal physiological homeostasis. Blockage or oxygen deprivation can lead to severe and irreversible injuries to the neurons. Oxygen consumption in the brain of oxygen results in production of free radicals and higher oxygen levels in brain leads to even higher concentration of reactive oxygen/nitrogen species. However, in spite of the fact that brain has higher necessity for oxygen, it is relatively deficient in the enzymes capable of metabolizing a number of these toxic oxygen-based reactants to harmless residues. In contrast, CNS is highly enriched with polyunsaturated fatty acids and toxic oxygen derivatives oxidizes these polyunsaturated fatty acids.17,18 This then makes the neurons more vulnerable to oxidation related damages and the role of the cellular detoxification machinery in these conditions is vital.

Oxidative stress has major impact on several NDDs such as Alzheimer’s disease (AD), Parkinson’s disease (PD), Amyotrophic lateral sclerosis (ALS) and Huntington’s disease (HD) In AD, oxidative stress is associated with a number of critical events which ultimately influence amyloid precursor protein (APP) processing. Tau modification leads to increased brain toxicity and as a result of oxidative stress, APP and Tau processing is altered via activation of different signaling pathways. In general most of the AD cases are late onset and sporadic although there are approximately 10-15% familial AD (FAD) cases. Mutation in three genes namely, APP, Presenilin 1 and 2 (PS1, PS2) can potentially lead to FAD. During normal physiological state proteolytic cleavage of APP is commenced by α-secretase followed by γ-secretase mediated second cleavage to yield non amyloidogenic fragments.19 However, mutations in APP results in an altered proteolytic processing where α- secretase is replaced by β-secretase (BACE1), followed by γ-secretase mediated cleavage in order to yield amyloidogenic Aβ42 which aggregates as insoluble plaques. Widespread cell culture studies have revealed Aβ42 to have toxic effect on brain and can emanate cell death via apoptosis.20 The hyperphosphorylation of tau protein, by various kinases such as MARK, MAPK and GSK-3αβ, results in the formation of paired helical filaments (PHFs), which further combine to form insoluble NFTs.21 Abnormal hyperphosphorylation of tau is indicative of both an abnormal activation of kinases and decreased phosphatase activity.22 Experiments on Pin1 knockout mice illustrates a rise in amyloidogenic APP processing thus increasing the levels of Aβ42 and additionally also display tau hyperphosphorylation thus leading to behavioral deficits, motor and neuronal degeneration.23

Oxidative can modulate pathogenesis in Parkinson’s. PD is the most common neurodegenerative disorder and is clinically demarcated by bradykinesia, progressive rigidity and tremor. Like all other neurodegenerative disorders determinants such as environmental factors, mitochondrial dysfunction, oxidative damage, and genetic predisposition together play a crucial role in both sporadic as well as familial PD.24 Neurotoxic compounds, such as N-methyl-4-phenyl-1,2,3,6-tetrahydropyridine or its active derivative, MPPT and 6-hydroxydopamine (6-OHDA) can provoke oxidative stress, impair mitochondrial respiration and energy metabolism which in turn leads to neurodegeneration. Postmortem tissues from PD patients have revealed a significant insight into the failure of complex I in the substantia nigra. Complex I is involved in the mitochondrial electron-transport chain, and 30-40% decrease in activity may be the central prognosis of sporadic PD.25 The decreased activity of complex I could be the result of self-inflected oxidative damage, under production of certain, complex I subunits, and may be due to complex I disassembly.26 Immunocytochemical confirmation of protein glycation and nitration in substantia nigra region of human PD brain revealed oxidative damage to DNA and protein resulting from persistent oxidative trauma.27

Huntington’s disease (HD) is autosomaly inherited and is characterized by progressive cognitive impairment, choreiform movements, psychiatric disturbances and loss of long projection neurons, resulting in atrophy of the caudate nucleus, globus pallidus, and putamen.28 HD mutation is an extension of the CAG trinucleotide repeat inside exon 1 of the huntingtin (HTT) gene, the exact role of which is unknown.29 CAG triplet codes for glutamine expansion and upon mutation, presents a polyglutamine tract at the N-terminus, results in a conformational change of the protein, which eventually results in abnormal protein-protein interaction. Mutant HTT presents a dominant “gain of function” to the protein, due to the stretched polyglutamine segment, which finally leads to neurodegeneration. Empirical evidences suggest that the mitochondrial metabolic defect resulting in impaired energy metabolism may be the consequences of HTT gene expansion.30 Altered mitochondrial energy metabolism raises the production of free radicals thus resulting in severe neuronal trauma. Mitochondrial dysfunction is a critical hallmark in the pathogenesis of PD. The activity of mitochondrial complexes I, II, III and IV is significantly altered during HD pathogenesis. Biochemical studies of HD brain tissue have reported defects in the caudate and decreased activities of complex II and III activity. However, no such deviation was observed with complex I or IV.31

Amyotrophic lateral sclerosis (ALS) is clinically identified by progressive atrophy, weakness, and spasticity of muscle tissue. ALS is characterized as an adult-onset neurodegenerative disease reflecting the degeneration of upper and lower motor neurons in the spinal cord, cortex, and brainstem.32 Mutations in the ubiquitous enzyme; Cu/Zn-superoxide dismutase (SOD-1), accounts for upto 5-20% all of major genetic defect in ALS. In addition to SOD-1 gene few other genes such as VAMP-associate protein B (VAPB), Alsin (ALS2), Dynactin (DCTN1), fused in sarcoma protein (FUS), TAR DNA-binding protein-43 (TDP-43), and lipid phosphatase FIG4 (FIG4) can also contribute to ALS pathogenesis. Postmortem tissues from ALS patients have clearly revealed that oxidative stress is the main causative factor that contributes to accumulation of oxidative damage to lipids, proteins, and DNA thus suggesting a direct role in ALS progression.33,34

Role of ER stress in neurodegenerative conditions

Endoplasmic reticulum (ER) is an imperative organelle responsible for the post-translation modification, proper folding, and transport of nascent proteins to target destinations. Loss of ER integrity results in ER stress and may be established due to changes in the calcium homeostasis within the ER and due to the accumulation of unfolded proteins. ER stress plays a crucial role in several signaling cascades including the unfolded protein response (UPR) which counteracts the effects of the original stress.35,36 The activity of the ubiquitin proteasome system (UPS) is significantly misregulated in these stress conditions and leads to protein aggregates and other toxic product accumulation thus leading to brain damaging conditions.37,38 Furthermore, intracellular ER calcium concentration and its release from the ER play a significant role in controlling neuronal death.39

As discussed earlier AD is a neurodegenerative disease characterized by the progressive loss of cognitive functions and memory loss. ER stress and an altered calcium homeostasis have major impact on severity of AD pathogenesis.40,41 Brain tissue from AD patients reports an alteration in calcium metabolism and subsequent neurodegeneration. Neurons containing NFTs shows an increase in the levels of free and protein bound calcium as compared to tangle free neurons. In addition to change in the level of calcium ion due to ER stress, an alteration in APP or PS proteins activity also can define AD prognosis.42 PS1 and PS2 proteins are the major catalytic components of the γ-secretase complex that facilitates the intramembranous cleavage of APP. ER stress mutations can cause a change in the pattern of APP processing in the affected neurons and as a result increase the amount of the toxic Aβ1-42 peptide. PS1 and PS2 are ER transmembrane proteins that are richly expressed by the brain neurons and which facilitates a linkage between AD and ER stress. ER stress related alteration in PS1 activity demonstrates altered calcium homeostasis, increased production of Aβ peptides, and enhanced apoptotic sensitivity. Mutant PS1 attaches to and restrains the ER kinase, IRE1 which senses the gathering of misfolded proteins in the ER lumen. IRE also triggers the downstream signals to mediate the transcription of the ER chaperone, BiP.43

Cultured neuronal cells, including dopaminergic neurons, reveals that neurotoxic compounds such as N-methyl-4-phenyl-1,2,3,6-tetrahydroyridine or its active derivative, MPPT and 6-hydroxydopamine (6-OHDA) can elicit ER stress and activate a number of genes such as the ER chaperones and other machinery of the UPR for instance the transcription factor, CHOP/Gadd153. In addition it can also lead to the phosphorylation of the ER stress kinases, PERK and IRE.44,45 Thus, ER stress in combination with abnormal protein degradation can contribute to the pathophysiology of NDDs.

Human inherited neurodegenerative disorders such as Huntington’s disease (HD), dentatorubral-pallidoluysian atrophy, spinobulbar muscular atrophy, and six spinocerebellar ataxias (SCA 1, 2, 3, 6, 7 and 17) are caused due to expanded polyglutamine (polyQ) repeats in the brain. In cultured cells, transgenic animals and in human post-mortem brain tissue these disorders are significantly characterized by aggregation of intracellular protein aggregates and selective neuronal death.46 Additionally, in HD the mutant Huntingtin gene can also have an effect on the calcium metabolism in the cell and sensitize the IP3 receptors in the ER.47

Evidence about the role of ER stress in polyQ diseases approaches from studies showing the colocalization of polyQ fragments with various molecular chaperones viz. Hsp70 and Hsp40 that are induced during ER stress. Drosophila overexpression of Hsp70 restrains polyQ toxicity. This Hsp mediated effect has also been observed in a few, but not all mouse models of polyQ diseases.46 SCA3 polyQ fragments also triggers ER stress mediated neuronal cell death, as shown by the activation of PERK, IRE1 and the stimulation of CHOP/Gadd153 and BiP/Grp78. However, this effect is mainly due to impairment in the interaction between the ER and the UPS. Another study reveals that deficient mouse embryonic fibroblasts show activation in the apoptosis signal-regulating kinase 1 (ASK1) which is indispensable for polyQ induced ER-mediated cell death.48 Moreover, ASK1 forms a complex with TRAF2 and IRE proteins at the ER and consequently trigger downstream signals, such as the c-Jun NH2-terminal Kinase (JNK).49

Transgenic ALS mice and human samples reports intracellular cytoplasmic inclusions in motor neurons. In ALS mice these contain deposits of SOD1 and ubiquitin. The aggregates appear prior to the first appearance of disease symptoms. Although the significance of inclusion bodies in ALS is not clear yet it has been attributed to act as a neurotoxin and inhibits critical cellular functionalities.50 Dorfin is one such ubiquitin E3 Ligase which play a crucial role in ALS mediated neurodegeneration. In ALS infected neurons, mutant SOD1is degraded by such ubiquitin E3-ligase through the UPS. Mutant a-synuclein and aggregated SOD1 in combination with other proteins can alter the function of the UPS and also affects the motor neurons. Although, motor neurons are not the only cell type targeted in ALS. The role of glial neuron and glial cells interactions at some stage in development of the disease suggests that the increase in ROS production combined with ER and oxidative damage to crucial proteins and other cell machinery may play a role in ALS pathogenesis.51

Glutamate metabolism is associated with prolonged stimulation of excitatory amino-acid receptors and results in increased intracellular calcium levels, which can easily damage the integrity and functional aspects of mitochondria and the ER. This then result in cleavage of caspase-12 in the spinal cord of transgenic ALS mice. Caspase-12 activity and cleavage in the ALS mice may be due to the activity of the calcium dependent enzyme, Calpain. Caspase-12 acts as substrate for calpain in some cells including neurons. Other biomarkers for ER stress, Bip/ Grp78 function is also altered in the ALS mice.52 Furthermore, evidence for ER stress in ALS comes from studies showing an increase in Bip/Grp78 level in spinal motor neurons of transgenic ALS mice proceeding to onset of motor symptoms. It has been also accounted that mutant SOD1is associated with ER stress, but not wild type SOD1.53 These findings provide credence to the fact that ER stress is part of the mechanism by which mutant SOD1 contributes to ALS related motor neuron degeneration.

UPS and E3 ligases in neurodegeneration

Dysfunction of the ubiquitin proteasome system is one of the major events that lead to the progression of neuronal loss. An in vivo report suggests that oxidative stress is caused directly by neuronal proteasome dysfunction in the mammalian brain.13,54,55 The UPS plays a vital role in regulated degradation of cellular proteins under diverse physiological conditions. Aggregation of misfolded proteins has been attributed in the progression of various neurodegenerative diseases, such as Alzheimer's disease (AD), Parkinson's disease (PD) and Huntington's disease (HD). Ubiquitin E3 ligases are key regulators involved in mediating the proteasomal degradation of misfolded proteins in the endoplasmic reticulum (ER), as a result protecting neurons against oxidative stress, mitochondrial dysfunction and ER stress.56 Furthermore, Ubiquitin proteasome system can crtically modulate the level of proteins in cells, and robustly control cellular mechanisms. Aberration in UPS function in susceptible neurons results in protein aggregation, increased, oxidative stress, ER stress, and ultimately neuronal death. Conversely, neurons depend on the proper functioning of E3 ligases and UPS to maintain neuronal homeostasis.57 Table 1 highlights the prospective role E3 ligases in neurodegeneration.58–80

E3 ligase

Substrates

Functional significance

References

Keap1

Nrf2

Involved in degradation of Nrf2. disruption of protein degradation systems and sustained activation of the Keap1-Nrf2 system occur in the AD brain.

Tanji et al.58

PARK2

AIMP2

Parkin is an E3 ubiquitin ligase that has been shown to be a key regulator of the autophagy pathway. Although, mutations in Parkin results into Parkinson’s Disease.

Segura-Aguilar et al. & Imam et al.59,60

HECW1

DVL1, p53, and mutant SOD1

NEDL1 is another name of HECW1. It mainly interacts with p53 and the Wnt signaling protein DVL1, and may play a critical role in p53-mediated cell death in neurons.

Li et al.61

HUWE1

TopBP1, N-Myc, C-Myc, p53, Mcl-1

HUWE1 regulates neuronal differentiation by destabilizing N-Myc, and also modulates p53-dependent and independent tumor suppression via ARF. It is also known as Mule. HUWE1 is a HECT domain E3 ubiquitin ligase which involves in degradation of Mcl-1 and thus regulates DNA damage-induced apoptosis.

Zhong et al.62

MGRN1

Involved in melanocortin signaling. Loss of mahogunin function leads to neurodegeneration and loss of pigmentation, and also has mechanism of action in prion disease.

Perez-Oliva et al.63

MYCBP2

TSC2, Fbxo45

MYCBP2 associates with Fbxo45 to play a crucial role in neuronal development. MycBP2 is an E3 ubiquitin ligase also known as PAM. MycBP2 also modulates the mTOR pathway through ubiquitination of TSC2.

Han et al.64

UHRF2

PCNP

UHRF2 ubiquitinates PCNP and has been shown to play a role in degradation of nuclear aggregates containing polyglutamine repeats mediated Neurodegeneration. UHRF2 is also known as NIRF. UHRF2 is a nuclear protein that may regulate cell cycle progression through association with Chk2.

Mori et al.65

ZNRF1

Highly expressed in neuronal cells. ZNRF1 is found in synaptic vesicle helpful in neuronal transmissions and plasticity. It also contains a RING finger motif, which expression is up regulated in the Schwann cells mediated nerve injury.

Araki and Milbrandt66 & Saitoh and Araki67

NEDD4

Highly expressed in the early mouse embryonic central nervous system. It down regulates both neuronal voltage-gated Na+ channels and epithelial Na+ channels in response to increased intracellular Na+ concentrations.

Goulet et al.68

NEDD4L

Smad2

It also highly expressed in the early mouse embryonic central nervous system. NEDD4L negatively regulates TGF-β signaling by targeting Smad2 for degradation.

Gao et al.69

HECTD2

HECTD2 is a likely E3 ubiquitin ligase and may act as a vulnerable gene for neurodegeneration especially in prion disease.

Lloyd et al.70

PJA2

Expressed in neuronal synapses. The exact role and substrates of PJA2 are unclear.

Yu et al.71

RNF19

SOD1

RNF19 is also known as Dorfin. Accumulation of mutant SOD1 results into ALS disease. RNF19 ubiquitinates mutant SOD1 protein, causing less neurotoxicity in brain.

Sone et al.72

HECTD1

HECTD1 is required for normal development of the mesenchyme and neural tube closure.

Zohn et al.73

MULAN

mnd2

Involved in degradation of mnd2. mnd2 causes neuromuscular disorder due to loss of Omi/HtrA2's protease activity.

Cilenti et al.74

HACE1

NRF2

HACE1 plays a crucial in the NRF2 mediated antioxidative stress response pathway and also involved in HD pathogenesis.

Rotblat et al.75

CUL4

TSC2

It promotes proteasomal degradation of TSC2. As a result, Tnfaip8 l1/Oxi-β competes with TSC2 to bind FBXW5, increasing TSC2 stability through preventing its ubiquitination in PD progression.

Ha et al.76

TRIM13

Involved in regulation of ER stress induced cell death. However, the expression of TRIM13 sensitizes cells to ER stress induced neuronal cell death.

Tomar et al.77

MGRN1

Over expression of MGRN1 protects against cell death mediated by ER and oxidative stress and also interacts with Cytosolic Hsp70. Lack of MGRN1 functionalities are the hall mark of age dependent spongiform disease in the brain.

Chhangani and Mishra78

NEDD4-1

FOXM1B

Up regulated in cultured neurons in response to various neurotoxins, including, hydrogen superoxide, and zinc via transcriptional activation likely mediated by the reactive oxygen species. A level of the insulin-like growth factor receptor (IGF-1Rβ) is also maintained due to up regulation of NEDD4-1.

Kwak et al.79

APC/C

Involved in cell cycle progression in proliferating cells, plays a significant role in post-mitotic neurons. APC/C-activating cofactor, Cdh1, is also helpful for the function of APC/C in neuronal survival.

Almeida80

Table 1 E3 ligases in the brain and their functional prospect in neuro degeneration

Conclusion

The future of neurodegenerative disorders depends on the researchers’ ability to adjust actions to circumstances and have a clear projection relating to the aberrant mechanisms that ultimately decides the fate of the neurons and henceforth degeneration. Despite tremendous advancement in the field of neurobiology, still the future of such therapies hangs on torrid balance and deceptive hopes Neuronal damage is caused due to free radical’s accumulation, oxidative stress and ER stress. In addition, dysfunctional of ubiquitin proteasome systems can also regulate prognosis in NDDs. These factors can cause an imbalance between cellular-antioxidant defence and reactive oxygen species concentration. Further research is therefore needed in order to make bio molecule based neurotherapeutics a blatant reality in conditions of neurodegeneration.

Acknowledgements

Authors are thankful to DTU senior management for constant support and encouragement.

Conflict of interest

The author declares no conflict of interest.

References

  1. Soto C. Unfolding the role of protein misfolding in neurodegenerative diseases. Nat Rev Neurosci. 2003;4(1):49–60.
  2. Bueler H. Impaired mitochondrial dynamics and function in the pathogenesis of Parkinson’s disease. Exp Neurol. 2009;218(2):235–246.
  3. Kieper N, Holmstrom KM, Ciceri D, et al. Modulation of mitochondrial function and morphology by interaction of Omi/ HtrA2 with the mitochondrial fusion factor OPA1. Exp Cell Res. 2010;316(7):1213–1224.
  4. Brown RC, Lockwood AH, Sonawane BR. Neurodegenerative diseases: an overview of environmental risk factors. Environ Health Perspect. 2005;113(9):1250–1256.
  5. Unnithan AS, Jiang Y, Rumble JL, et al. N–acetyl cysteine prevents synergistic, severe toxicity from two hits of oxidative stress. Neurosci Lett. 2014;560:71–76.
  6. Dasuri K, Zhang L, Keller JN. Oxidative stress, neurodegeneration, and the balance of protein degradation and protein synthesis. Free Radic Biol Med. 2013;62:170–185.
  7. Mesman S, von Oerthel L, Smidt MP. Mesodiencephalic Dopaminergic Neuronal Differentiation Does Not Involve GLI2A–Mediated SHH–Signaling and Is under the Direct Influence of Canonical WNT Signaling. PLoS One. 2014;9(5):e97926.
  8. Alvarez–Buylla A, Ihrie RA. Sonic hedgehog signaling in the postnatal brain. Semin Cell Dev Biol. 2014;33:105–111.
  9. Hardy J, Orr H. The genetics of neurodegenerative diseases. J Neurochem. 2006;97(6):1690–1699.
  10. Prokai L, Rivera–Portalatin NM, Prokai–Tatrai K. Quantitative structure–activity relationships predicting the antioxidant potency of 17β–estradiol–related polycyclic phenols to inhibit lipid peroxidation. Int J Mol Sci. 2013;14(1):1443–1454.
  11. Pereira CMF. Crosstalk between Endoplasmic Reticulum Stress and Protein Misfolding in Neurodegenerative Diseases. ISRN Cell Biology. 2013;2013:22.
  12. Lindholm D, Wootz H, Korhonen L. ER stress and Neurodegenerative diseases. Cell Death Differ. 2006;13(3):385–392.
  13. Jaiswal M, Sandoval H, Zhang K, et al. Probing mechanisms that underlie human neurodegenerative diseases in Drosophila. Annu Rev Genet. 2012;46:371–396.
  14. Beal MF. Mitochondria take center stage in aging and neurodegeneration. Ann of Neurol. 2005;58(4):495–505.
  15. Halliwell B. Free radicals, antioxidants, and human disease: curiosity, cause, or consequence? Lancet. 1994;344(8924):721–724.
  16. Yu BP. Cellular defenses against damage from reactive oxygen species. Physiol Rev. 1994;74 (1):139–162.
  17. Jaiswal M, Sandoval H, Zhang K, et al. Oxidative damage to RNA in neurodegenerative diseases. Journal of Biomedicine and Biotechnology. 2006;2006(3):82323.
  18. Perry G, Nunomura A, Hirai K, et al. Is oxidative damage the fundamental pathogenic mechanism of Alzheimer’s and other neurodegenerative diseases? Free Radic Biol Med. 2002;33(11):1475–1479.
  19. O’Brien RJ, Wong PC. Amyloid precursor protein processing and Alzheimer’s disease. Annu Rev Neurosci. 2011;34:185–204.
  20. Zheng YL, Li BS, Amin ND, et al. A peptide derived from cyclin–dependent kinase activator (p35) specifically inhibits Cdk5 activity and phosphorylation of tau protein in transfected cells. Eur J Biochem. 2002;269(18):4427–4434.
  21. Hanger DP, Betts JC, Loviny TL, et al. New phosphorylation sites identified in hyperphosphorylated tau (paired helical filament–tau) from Alzheimer’s disease brain using nanoelectrospray mass spectrometry. J Neurochem. 1998;71(6):2465–2476.
  22. Stoothoff WH, Johnson GV. Tau phosphorylation: physiological and pathological consequences. Biochim Biophy Acta. 2005;1739 (2–3):280–297.
  23. Liou YC, Sun A, Ryo A, et al. Role of the prolyl isomerase Pin1 in protecting against age–dependent neurodegeneration. Nature. 2003;424(6948):556–561.
  24. Jenner P. Oxidative stress in Parkinson’s disease. Ann Neurol. 2003;53(3):S26–S38.
  25. Dawson TM, Dawson VL. Molecular pathways of neurodegeneration in Parkinson’s disease. Science. 2003;302(5646):819–822.
  26. Keeney PM, Xie J, Capaldi RA, et al. Parkinson’s disease brain mitochondrial complex I has oxidatively damaged subunits and is functionally impaired and misassembled. J Neurosci. 2006;26(19):5256–5264.
  27. Floor E, Wetzel MG. Increased protein oxidation in human substantia nigra pars compacta in comparison with basal ganglia and prefrontal cortex measured with an improved dinitrophenylhydrazine assay. J Neurochem. 1998;70(1):268–275.
  28. Davies S, Ramsden DB. Huntington’s disease. Mol Pathol. 2001;54(6):409–413.
  29. Gusella JF, Wexler NS, Conneally PM, et al. A polymorphic DNA marker genetically linked to Huntington’s disease. Nature. 1983;306(5940):234–238.
  30. Smith DS, Tsai LH. Cdk5 behind the wheel: a role in trafficking and transport? Trends Cell Biol. 2002;12(1):28–36.
  31. Mann VM, Cooper J M, Javoy–Agid F, et al. Mitochondrial function and parental sex effect in Huntington’s disease. Lancet. 1990;336(8717):749.
  32. Rowland LP, Shneider NA. Amyotrophic lateral sclerosis. N Engl J Med. 2001;344(22):1688–1700.
  33. Strong MJ. The evidence for altered RNA metabolism in amyotrophic lateral sclerosis (ALS). J Neurol Sci. 2010;288(1–2):1–12.
  34. Kikuchi S, Shinpo K, Ogata A, et al. Detection of N epsilon–(carboxymethyl)lysine (CML) and non–CML advanced glycation end–products in the anterior horn of amyotrophic lateral sclerosis spinal cord. Amyotroph Lateral Scler Other Motor Neuron Disord. 2002;3(2):63–68.
  35. Breckenridge DG, Germain M, Mathai JP, et al. Regulation of apoptosis by endoplasmic reticulum pathways. Oncogene. 2003;22(53):8608–8618.
  36. Rao RV, Ellerby HM, Bredesen DE. Coupling endoplasmic reticulum stress to the cell death program. Cell Death Differ. 2004;11(4):372–380.
  37. Soto C. Unfolding the role of protein misfolding in neurodegenerative diseases. Nat Rev Neurosci. 2003;4(1):49–60.
  38. Ciechanover A, Brundin P. The ubiquitin proteasome system in neurodegenerative diseases: sometimes the chicken, sometimes the egg. Neuron. 2003;40(2):427–446.
  39. Verkhratsky A. Physiology and pathophysiology of the calcium store in the endoplasmic reticulum of neurons. Physiol Rev. 2005;85(1):201–279.
  40. Katayama T, Imaizumi K, Manabe T, et al. Induction of neuronal death by ER stress in Alzheimer’s disease. J Chem Neuroanat. 2004;28(1–2):67–78.
  41. LaFerla FM. Calcium dyshomeostasis and intracellular signalling in Alzheimer’s disease. Nat Rev Neurosci. 2002;3(11):862–872.
  42. Mattson MP. Apoptosis in neurodegenerative disorders. Nat Rev Mol Cell Biol. 2000;1(2):120–129.
  43. Wilquet V, De Strooper B. Amyloid–beta precursor protein processing in neurodegeneration. Curr Opin Neurobiol. 2004;14(5):582–584.
  44. Holtz WA, O’Malley KL. Parkinsonian mimetics induce aspects of unfolded protein response in death of dopaminergic neurons. J Biol Chem. 2003;278(21):19367–19377.
  45. Ryu EJ, Harding HP, Angelastro JM, et al. Endoplasmic reticulum stress and the unfolded protein response in cellular models of Parkinson’s disease. J Neurosci. 2002;22(24):10690–10698.
  46. Lipinski MM, Yuan J. Mechanisms of cell death in polyglutamine expansion diseases. Curr Opin Pharmacol. 2004;4(1):85–90.
  47. Tang TS, Tu H, Chan EY, et al. Huntingtin and huntingtin–associated protein 1 influence neuronal calcium signaling mediated by inositol–(1,4,5) triphosphate receptor type 1. Neuron. 2003;39(2):227–239.
  48. Nishitoh H, Matsuzawa A, Tobiume K, et al. ASK1 is essential for endoplasmic reticulum stress–induced neuronal cell death triggered by expanded polyglutamine repeats. Genes Dev. 2002;16(11):1345–1355.
  49. Thomas M, Yu Z, Dadgar N, et al. The unfolded protein response modulates toxicity of the expanded glutamine androgen receptor. J Biol Chem. 2005;280(22):21264–21271.
  50. Bruijn LI, Miller TM, Cleveland DW. Unraveling the mechanisms involved in motor neuron degeneration in ALS. Annu Rev Neurosci. 2004;27:723–749.
  51. Niwa J, Ishigaki S, Hishikawa N, et al. Dorfin ubiquitylates mutant SOD1 and prevents mutant SOD1–mediated neurotoxicity. J Biol Chem. 2002;277(39):36793–36798.
  52. Wootz H, Hansson I, Korhonen L, et al. Caspase–12 cleavage and increased oxidative stress during motoneuron degeneration in transgenic mouse model of ALS. Biochem Biophys Res Commun. 2004;322(1):281–286.
  53. Sundaramoorthy V, Walker AK, Yerbury J, et al. Extracellular wildtype and mutant SOD1 induces ER–Golgi pathway characteristic of amyotrophic lateral sclerosis in neuronal cells. Cell mol Life Sci. 2013;70(21):4181–4195.
  54. Dasuri K, Zhang L, Keller JN. Oxidative stress, neurodegeneration, and the balance of protein degradation and protein synthesis. Free Radic Biol Med. 2013;62:170–185.
  55. Elkharaz J, Ugun–Klusek A, Constantin–Teodosiu D, et al. Implications for oxidative stress and astrocytes following 26S proteasomal depletion in mouse forebrain neurones. Biochim Biophys Acta. 2013;1832(12):1930–1938.
  56. Mei J, Niu C. Alterations of Hrd1 expression in various encephalic regional neurons in 6–OHDA model of Parkinson's disease. Neurosci Lett. 2010;474(2):63–68.
  57. Jara JH, Frank DD, Ozdinler PH. Could dysregulation of UPS be a common underlying mechanism for cancer and neurodegeneration? Lessons from UCHL1. Cell Biochem Biophys. 2013;67(1):45–53.
  58. Tanji K, Miki Y, Ozaki T, et al. Phosphorylation of serine 349 of p62 in Alzheimer's disease brain. Acta Neuropathol Commun. 2014;2(1):50.
  59. Segura–Aguilar J, Paris I, Munoz P, et al. Protective and toxic roles of dopamine in Parkinson's disease. J Neurochem. 2014;129(6):898–915.
  60. Imam SZ, Trickler W, Kimura S, et al. Neuroprotective efficacy of a new brain–penetrating C–Abl inhibitor in a murine Parkinson's disease model. PLoS One. 2013;8(5):e65129.
  61. Li Y, Ozaki T, Kikuchi H, et al. A novel HECT–type E3 ubiquitin protein ligase NEDL1 enhances the p53–mediated apoptotic cell death in its catalytic activity–independent manner. Oncogene. 2008;27(26):3700–3709.
  62. Zhong Q, Gao W, Du F, et al. Mule/ARF–BP1, a BH3–only E3 ubiquitin ligase, catalyzes the polyubiquitination of Mcl–1 and regulates apoptosis. Cell. 2005;121(7):1085–1095.
  63. Perez–Oliva AB, Olivares C, Jimenez–Cervantes C, et al. Mahogunin ring finger–1 (MGRN1) E3 ubiquitin ligase inhibits signaling from melanocortin receptor by competition with Galphas. J Biol Chem. 2009;284(46):31714–31725.
  64. Han S, Witt RM, Santos TM, et al. Pam (Protein associated with Myc) functions as an E3 ubiquitin ligase and regulates TSC/mTOR signaling. Cell Signal. 2008;20(6):1084–1091.
  65. Mori T, Li Y, Hata H, et al. NIRF is a ubiquitin ligase that is capable of ubiquitinating PCNP, a PEST–containing nuclear protein. FEBS Lett. 2004;557(1–3):209–214.
  66. Araki T, Milbrandt J. ZNRF proteins constitute a family of presynaptic E3 ubiquitin ligases. J Neurosci. 2003;23(28):9385–9394.
  67. Saitoh F, Araki T. Proteasomal degradation of glutamine synthetase regulates schwann cell differentiation. J Neurosci. 2010;30(4):1204–1212.
  68. Goulet CC, Volk KA, Adams CM, et al. Inhibition of the epithelial Na+ channel by interaction of Nedd4 with a PY motif deleted in Liddle's syndrome. J Biol Chem. 1998;273(45):30012–300127.
  69. Gao S, Alarcon C, Sapkota G, et al. Ubiquitin ligase Nedd4L targets activated Smad2/3 to limit TGF–beta signaling. Mol Cell. 2009;36(3):457–468.
  70. Lloyd SE, Maytham EG, Pota H, et al. HECTD2 is associated with susceptibility to mouse and human prion disease. PLoS Genet. 2009;5(2):e1000383.
  71. Yu P, Chen Y, Tagle DA, et al. PJA1, encoding a RING–H2 finger ubiquitin ligase, is a novel human X chromosome gene abundantly expressed in brain. Genomics. 2002;79(6):869–874.
  72. Sone J, Niwa J, Kawai K, et al. Dorfin ameliorates phenotypes in a transgenic mouse model of amyotrophic lateral sclerosis. J Neurosci Res. 2010;88(1):123–135.
  73. Zohn IE, Anderson KV, Niswander L. The Hectd1 ubiquitin ligase is required for development of the head mesenchyme and neural tube closure. Dev Biol. 2007;306(1):208–221.
  74. Cilenti L, Ambivero CT, Ward N, et al. Inactivation of Omi/HtrA2 protease leads to the deregulation of mitochondrial Mulan E3 ubiquitin ligase and increased mitophagy. Biochim Biophys Acta. 2014;1843(7):1295–1307.
  75. Rotblat B, Southwell AL, Ehrnhoefer DE, et al. HACE1 reduces oxidative stress and mutant Huntingtin toxicity by promoting the NRF2 response. Proc Natl Acad Sci USA. 2014;111(8):3032–3037.
  76. Ha JY, Kim JS, Kang YH, et al. Tnfaip8 l1/Oxi–β binds to FBXW5, increasing autophagy through activation of TSC2 in a Parkinson's disease model. J Neurochem. 2014;129(3):527–538.
  77. Tomar D, Prajapati P, Sripada L, et al. TRIM13 regulates caspase–8 ubiquitination, translocation to autophagosomes and activation during ER stress induced cell death. Biochim Biophys Acta. 2013;1833(12):3134–3144.
  78. Chhangani D, Mishra A. Mahogunin ring finger–1 (MGRN1) suppresses chaperone–associated misfolded protein aggregation and toxicity. Sci Rep. 2013;3:1972.
  79. Kwak YD, Wang B, Li JJ, et al. Upregulation of the E3 ligase NEDD4–1 by oxidative stress degrades IGF–1 receptor protein in neurodegeneration. J Neurosci. 2012;32(32):10971–10981.
  80. Almeida A. Regulation of APC/C–Cdh1 and its function in neuronal survival. Mol Neurobiol. 2012;46(3):547–554.
Creative Commons Attribution License

© . This is an open access article distributed under the terms of the, which permits unrestricted use, distribution, and build upon your work non-commercially.