Nrf2—a therapeutic target for the treatment of neurodegenerative diseases

https://doi.org/10.1016/j.freeradbiomed.2015.07.147Get rights and content

Highlights

  • Oxidative stress in neurodegenerative diseases

  • The Keap1-Nrf2-ARE pathway in brain

  • Changes in Nrf2 and Nrf2-dependent genes in diseased brain tissue

  • Transgenic, viral, and chemical-mediated activation of Nrf2 in animal models

  • Effect of Nrf2 activation on progression of neurodegeneration in animal models

Abstract

The brain is very sensitive to changes in redox status; thus maintaining redox homeostasis in the brain is critical for the prevention of accumulating oxidative damage. Aging is the primary risk factor for developing neurodegenerative diseases. In addition to age, genetic and environmental risk factors have also been associated with disease development. The primary reactive insults associated with the aging process are a result of oxidative stress (OS) and nitrosative stress (NS). Markers of increased oxidative stress, protein and DNA modification, inflammation, and dysfunctional proteostasis have all been implicated in contributing to the progression of neurodegeneration. The ability of the cell to combat OS/NS and maintain a clearance mechanism for misfolded aggregating proteins determines whether or not it will survive. A critical pathway in this regard is the Nrf2 (nuclear factor erythroid 2-related factor 2)- antioxidant response element (ARE) pathway. Nrf2 activation has been shown to mitigate a number of pathologic mechanisms associated with Alzheimer’s disease, Parkinson’s disease, amyotrophic lateral sclerosis, Huntington’s disease, and multiple sclerosis. This review will focus on the role of Nrf2 in these diseases and the potential for Nrf2 activation to attenuate disease progression.

Introduction

The brain is very sensitive to changes in redox status; thus maintaining redox homeostasis in the brain is critical for the prevention of accumulating oxidative damage. Aging is the primary risk factor for developing neurodegenerative diseases. In addition to age, genetic and environmental risk factors have also been associated with disease development. The primary reactive insults associated with the aging process are a result of oxidative stress (OS) and nitrosative stress (NS). OS/NS are produced endogenously via enzymatic and spontaneous reactions through a variety of sources and normal physiological functions [1], [2].

Downstream markers of OS/NS have been identified clearly in all neurodegenerative diseases. Signs of lipid peroxidation, such as aldehydes including 4-hydroxynonenol (4-HNE), are increased in Parkinson’s disease (PD) [3], [4], [5], Alzheimer’s disease (AD) [6], [7], [8], and amyotrophic lateral sclerosis (ALS) [9], [10]. There is also a correlation between the affected brain regions and 4-HNE adducted proteins [11], [12], [13]. Another major marker of OS production is protein carbonyls, representing protein oxidation. Carbonyls are present in the substantia nigra (SN) in PD [14], [15], AD [16], and ALS [17] and in affected brain regions in other diseases [18], [19], [20], [21]. Oxidative damage to DNA/RNA has been evaluated by measuring 8-hydroxy-2-deoxyguanosine (8-OHDG) and is increased in central and peripheral nervous systems of patients with neurodegenerative diseases [22], [23], [24], [25], [26], [27], [28], [29], [30]. Post-translational modification proteins modify protein structure and function. Protein structure and function can be altered by phosphorylation, nitration, ubiquitination, and glycosylation. Such modifications have been observed in alpha-synuclein (SYN) in patients with synucleinopathies including PD and dementia with Lewy bodies (DLB) [31], [32], [33]. Nitration and phosphorylation of tau protein has been found in the hippocampus and neocortex of patients with AD and other tau pathologies [34], [35]. Nitrotyrosine-modified proteins are elevated eight-fold in the hippocampus and neocortex of AD brains [36], [37]. These region-specific protein modifications correlate with areas of increased OS/NS in the brain. Furthermore, such modifications are thought to contribute to protein misfolding and subsequent aggregate/inclusion formation. Because most neurodegenerative diseases have characteristic misfolded protein aggregates, such as SYN in PD, beta-amyloid (Aβ) plaques and hyperphosphorylated tau neurofibrillary tangles (NFTs) in AD, huntingtin (Htt) in Huntington’s disease (HD), and superoxide dismutase 1 (SOD1) in ALS, protein aggregation and regulation of misfolded protein clearance by the proteasome and autophagy appear to be vital to pathogenesis.

One of the primary endogenous sources of OS is the mitochondrial electron transport chain. Increased mitochondrial dysfunction associated with neurodegenerative diseases leads to increased OS generation and reduction in the production of ATP. In addition and associated with neuroinflammation, the enzymes of the NADPH oxidase system generate superoxide anions. The combination of superoxide anion with nitric oxide, produced by nitric oxide synthase, generates the highly reactive NS peroxynitrite. Combating OS/NS is dependent upon the cell’s ability to maintain cellular redox homeostasis. A critical pathway in this regard is the Nrf2 (nuclear factor erythroid 2-related factor 2)- antioxidant response element (ARE) pathway. The ARE is an enhancer element having the consensus sequence RTGACnnnGC, which is located in the 5’ flanking region of many phase II detoxifying and antioxidant genes [38], [39]. Nrf2 is a cytoplasmic protein sequestered by the actin-bound protein Keap1 (Kelch ECH associating protein) [40], [41]. Keap1, a Cul3-based E3 ligase, polyubiquitinates Nrf2, targeting it for subsequent proteasomal degradation [42], [43]. Oxidative stress or exposure to electrophilic agents that react with Keap1 stabilize Nrf2, leading to increased Nrf2 protein levels and nuclear accumulation of Nrf2. Once in the nucleus, Nrf2 dimerizes with small Maf proteins and binds to the ARE, transcriptionally driving expression of several detoxifying and antioxidant genes [44], [45].

Nrf2 contains six well-conserved Nrf2-ECH homologous (Neh) domains that support molecular functions. The CNC (cap’n’ collar) and DNA binding regions are located in the Neh1 domain, as is the Maf dimerization site. Neh4 and Neh5 are necessary for recruitment of transcription factors and other canonical proteins required for gene expression [46]. Molecular studies have determined that the Neh2 domain is required for the cytoplasmic localization of Nrf2, because deletion of Neh2 leads to continuous nuclear translocation of Nrf2 to the nucleus. Yeast two-hydridization screening using the Neh2 domain from Nrf2 as bait identified Keap1 as an Nrf2 binding protein. Eighty percent of the independently isolated clones screened were Keap1 positive, suggesting specificity of the Keap1–Nrf2 interaction [40]. Keap1 has two canonical domains, the Kelch domain and the bric-a-brac, tramtrack, broad-complex (BTB) domain. The Kelch domain binds actin and thus tethers the Keap1–Nrf2 complex to the cytoskeleton. The BTB domain is important for protein dimerization of Keap1 molecules. There are many cysteine residues in the Keap1 protein that potentially function as sensors of oxidants and electrophiles; humans have 27 and rat and mouse have 25 [44], [47]. The “hinge and latch” model proposes that two Keap1 molecules bind Nrf2 at high- and low-affinity sites located in the Neh2 domain [48]. The hinge domain, EGTE, supports high affinity and the latch domain, DLG, low affinity. When Keap1 senses oxidative or electrophilic stress, the low-affinity domain binding Nrf2 is abolished and proteosomal degradation of Nrf2 is disrupted [49]. In addition to Keap1, Nrf2 turnover can be regulated by GSK3β/β-TrCP- and Hrd1-dependent mechanisms in different pathological states [50], [51]. GSK3β phosphorylates the Neh6 domain of Nrf2. This facilitates binding of the β-TrCP/Cul1 E3 ligase complex to Nrf2. Nrf2 is then ubiquitinated and degraded through β-TrCP-mediated proteasomal degradation. Hrd1 is another E3 ubiquitin ligase that resides in the endoplasmic reticulum membrane. Hrd1 directly interacts with Nrf2 at the Neh4-5 domains by binding to the cytoplasmic C-terminal region of Hrd1, leading to Nrf2 ubiquitination and degradation.

In addition, there are endogenous proteins that have been shown to interact with Keap1 and activate the Nrf2 pathway. An initial screen using Nrf2-dependent ARE-luciferase activity identified seven activating proteins: sequestosome 1 (SQSTM1 or p62), d-site of albumin promoter binding protein (DBP), dipeptidylpeptidase 3 (DPP3), BCL2-like 1 (BCL2L1 or Bcl-xL), the kinesin family member 26B (KIF26B), cAMP-responsive element binding protein-regulated transcription coactivator 1 (TORC1), myeloid cell leukemia sequence 1 (Mcl1), and the splicing factor arginine/serine-rich 10 (SFRS10) [52]. Subsequently, both SQSTM1 and DPP3 were shown to interact with Keap1 [53], [54]. The Wilms tumor gene on the X chromosome (WTX) and PALB2, a major BRCA2 binding partner, also bind to Keap1, whereas p21 directly binds to Nrf2, leading to inhibition of Nrf2 ubiquitination and increased Nrf2-dependent gene expression [55], [56], [57]. This review will focus on the role of Nrf2 in neurodegeneration and the potential for Nrf2 activation to mitigate progression of neurological diseases.

Section snippets

Nrf2 in Alzheimer’s disease

AD is an age-associated progressive neurodegenerative disorder characterized by memory loss and cognitive dysfunction. It is the most common form of dementia. There are currently over 5 million AD patients in the United States and it is projected that, without the development of disease-modifying therapies, the number will increase to approximately 13.8 million by 2050 [58]. Pathological hallmarks of AD include brain atrophy due to neuronal and synapse loss, senile plaques predominantly

Nrf2 in Parkinson’s disease

PD is the most common neurodegenerative movement disorder, characterized by motor symptoms such as tremor, bradykinesia, posture instability, and rigidity, as well as a number of nonmotor neuropsychiatric problems [95]. One pathological hallmark of PD is the loss of dopaminergic neurons in the substantia nigra pars compacta (SN), resulting in reduced levels of dopamine in nerve terminals projecting to the striatum. A second hallmark is the formation of intracellular inclusions made up primarily

Nrf2 in amyotrophic lateral sclerosis

Amyotrophic lateral sclerosis (ALS) is the most common adult-onset motor neuron disease, caused by the progressive degeneration of motor neurons in the spinal cord, brain stem, and motor cortex [[124]. Motor neurons contain ubiquitin-positive hyaline and skein-like inclusions [125]. These inclusions contain SOD1 that co-localizes with p62 [126]. TDP-43 (TAR DNA-binding protein)- and FUS-positive inclusions have also been identified in ALS [127], [128]. As with AD and PD, the majority of ALS

Nrf2 in Huntington’s disease

HD is an autosomal dominant neurodegenerative disease that is associated with expansion of a CAG repeat in the gene encoding the Htt protein [149]. This results in a stretch of N-terminal glutamine residues, and the severity and onset of disease correlates with the length of this polyglutamine repeat. Furthermore, a conformational change in the Htt increases self–aggregation associated with intracellular inclusion formation [150]. HD is pathologically characterized by degeneration in

Nrf2 in multiple sclerosis

It is estimated that as many as 400,000 people in the United States and more than 2.3 million people worldwide are affected by multiple sclerosis (MS). MS is a chronic neuroinflammatory disease onset by activation of peripheral CD4+ T cells that traverse the blood–brain barrier and mount a damaging autoimmune attack on myelin and oligodendrocytes in the central nervous system (CNS) [167]. Neurological deficits manifest in heterogenic conditions that include but are not limited to vision loss,

Nrf2 and neuroinflammation

A pathological hallmark feature of neurodegenerative disease is neuroinflammation, mediated primarily by activated resident innate immune cells, the microglia. When activated, microglia secrete cytotoxic reactive oxygen and nitrogen species, contributing to the deleterious affects on neurons, especially in cases of microglial overactivation and dysregulation. Furthermore, components of dead or damaged neurons also activate microglia (reactive microgliosis) via pattern recognition receptors

NFE2L2 and Keap1 associated haplotypes and neurodegenerative diseases

Based on substantial evidence (as discussed in this review) that neurodegenerative effects of OS/NS in diseases such as PD, AD, and ALS can be modulated and/or prevented by up-regulation of Nrf2, genomic studies have been undertaken to ascertain if common genetic variations exist in the genes encoding Nrf2 and its repressor protein Keap1, NFE2L2 and Keap1, thus contributing to the onset or risk of these diseases. Interestingly, based on compilation of publicly available SNPs and other genetic

Conclusions

Based on the information provided herein, the Nrf2-ARE pathway is a high-value therapeutic target for neurodegenerative diseases. The existing data are strongest for PD, ALS, and MS, but ongoing experiments in AD and HD should provide more insight into how important Nrf2 is in these diseases as well. Numerous cell-based and in silico high-throughput screens have identified novel Nrf2-activating compounds [199], [200], [201], [202], [203], [204], [205]. The most promising approach may be to find

Acknowledgments

This work was supported by Grants R01 ES08089 and R01 ES10042 from the National Institute of Environmental Health Sciences, Grants R01 AG033493 and P50 AG033514 from the National Institute on Aging, and the CHDI Foundation.

References (229)

  • T. Horiguchi et al.

    Nitration of tau protein is linked to neurodegeneration in tauopathies

    Am J Pathol

    (2003)
  • T.H. Rushmore et al.

    Transcriptional regulation of the rat glutathione S-transferase Ya subunit gene. Characterization of a xenobiotic-responsive element controlling inducible expression by phenolic antioxidants

    J Biol Chem

    (1990)
  • T.H. Rushmore et al.

    The antioxidant responsive element. Activation by oxidative stress and identification of the DNA consensus sequence required for functional activity

    J Biol Chem

    (1991)
  • L.M. Zipper et al.

    The Keap1 BTB/POZ dimerization function is required to sequester Nrf2 in cytoplasm’

    J Biol Chem

    (2002)
  • M. McMahon et al.

    Keap1-dependent proteasomal degradation of transcription factor Nrf2 contributes to the negative regulation of antioxidant response element-driven gene expression

    J Biol Chem

    (2003)
  • W. Chen et al.

    Direct interaction between Nrf2 and p21(Cip1/WAF1) upregulates the Nrf2-mediated antioxidant response

    Mol Cell

    (2009)
  • N.D. Camp et al.

    Wilms tumor gene on X chromosome (WTX) inhibits degradation of Nrf2 protein through competitive binding to Keap1 protein

    J Biol Chem

    (2012)
  • W.L. Klein et al.

    Targeting small Abeta oligomers: the solution to an Alzheimer’s disease conundrum?

    Trends Neurosci

    (2001)
  • Y. Wang et al.

    NAD(P)H:quinone oxidoreductase activity is increased in hippocampal pyramidal neurons of patients with Aalzheimer’s disease

    Neurobiol Aging

    (2000)
  • K.S. SantaCruz et al.

    Regional NAD(P)H:quinone oxidoreductase activity in Alzheimer’s disease

    Neurobiol Aging

    (2004)
  • M.Y. Aksenov et al.

    Changes in thiol content and expression of glutathione redox system genes in the hippocampus and cerebellum in Alzheimer’s disease

    Neurosci Lett

    (2001)
  • G. Joshi et al.

    Increased Alzheimer’s disease-like pathology in the APP/ PS1DeltaE9 mouse model lacking Nrf2 through modulation of autophagy

    Neurobiol Aging

    (2015)
  • K. Kanninen et al.

    Nuclear factor erythroid 2-related factor 2 protects against beta amyloid

    Mol Cell Neurosci

    (2008)
  • V. Torres-Lista et al.

    Neophobia, NQO1 and SIRT1 as premorbid and prodromal indicators of AD in 3xTg-AD mice

    Behav Brain Res

    (2014)
  • A. Jain et al.

    .’p62/SQSTM1 is a target gene for transcription factor Nrf2 and creates a positive feedback loop by inducing antioxidant response element-driven gene transcription

    J Biol Chem

    (2010)
  • S.A. Farr et al.

    Antisense oligonucleotide against GSK-3beta in brain of SAMP8 mice improves learning and memory and decreases oxidative stress: Involvement of transcription factor Nrf2 and implications for Alzheimer disease

    Free Radic Biol Med

    (2014)
  • M. Leiros et al.

    Gracilins: Spongionella-derived promising compounds for Alzheimer disease

    Neuropharmacology

    (2015)
  • H.V. Kim et al.

    Amelioration of Alzheimer’s disease by neuroprotective effect of sulforaphane in animal model

    Amyloid

    (2013)
  • L. Yu et al.

    Orientin alleviates cognitive deficits and oxidative stress in Abeta1-42-induced mouse model of Alzheimer’s disease

    Life Sci

    (2015)
  • M.F. Beal

    Therapeutic approaches to mitochondrial dysfunction in Parkinson’s disease

    Parkinsonism Relat Disord

    (2009)
  • A. Samii et al.

    Parkinson’s disease

    Lancet

    (2004)
  • K. Dasuri et al.

    Oxidative stress, neurodegeneration, and the balance of protein degradation and protein synthesis

    Free Radic Biol Med

    (2012)
  • D.T. Dexter et al.

    Basal lipid peroxidation in substantia nigra is increased in Parkinson’s disease

    J Neurochem

    (1989)
  • D.T. Dexter et al.

    Increased levels of lipid hydroperoxides in the parkinsonian substantia nigra: an HPLC and ESR study

    Mov Disord

    (1994)
  • A. Yoritaka et al.

    Immunohistochemical detection of 4-hydroxynonenal protein adducts in Parkinson disease

    Proc Natl Acad Sci U S A

    (1996)
  • R.G. Smith et al.

    Presence of 4-hydroxynonenal in cerebrospinal fluid of patients with sporadic amyotrophic lateral sclerosis

    Ann Neurol

    (1998)
  • E.P. Simpson et al.

    Increased lipid peroxidation in sera of ALS patients: a potential biomarker of disease burden

    Neurology

    (2004)
  • K.S. Montine et al.

    4-hydroxy-2-nonenal pyrrole adducts in human neurodegenerative disease

    J Neuropathol Exp Neurol

    (1997)
  • L.M. Sayre et al.

    4-Hydroxynonenal-derived advanced lipid peroxidation end products are increased in Alzheimer’s disease

    J Neurochem

    (1997)
  • W.A. Pedersen et al.

    Protein modification by the lipid peroxidation product 4-hydroxynonenal in the spinal cords of amyotrophic lateral sclerosis patients

    Ann Neurol

    (1998)
  • Z.I. Alam et al.

    A generalised increase in protein carbonyls in the brain in Parkinson’s but not incidental Lewy body disease

    J Neurochem

    (1997)
  • E. Floor et al.

    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)
  • A.C. Bowling et al.

    Superoxide dismutase activity, oxidative damage, and mitochondrial energy metabolism in familial and sporadic amyotrophic lateral sclerosis

    J Neurochem

    (1993)
  • K. Hensley et al.

    Brain regional correspondence between Alzheimer’s disease histopathology and biomarkers of protein oxidation

    J Neurochem

    (1995)
  • L. Lyras et al.

    An assessment of oxidative damage to proteins, lipids, and DNA in brain from patients with Alzheimer’s disease

    J Neurochem

    (1997)
  • Z.I. Alam et al.

    Oxidative DNA damage in the parkinsonian brain: an apparent selective increase in 8-hydroxyguanine levels in substantia nigra

    J Neurochem

    (1997)
  • N. Shibata et al.

    Nonoxidative protein glycation is implicated in familial amyotrophic lateral sclerosis with superoxide dismutase-1 mutation

    Acta Neuropathol

    (2000)
  • M.A. Lovell et al.

    Ratio of 8-hydroxyguanine in intact DNA to free 8-hydroxyguanine is increased in Alzheimer disease ventricular cerebrospinal fluid

    Arch Neurol

    (2001)
  • Y. Ihara et al.

    Oxidative stress and metal content in blood and cerebrospinal fluid of amyotrophic lateral sclerosis patients with and without a Cu, Zn-superoxide dismutase mutation

    Neurol Res

    (2005)
  • T. Murata et al.

    Increased mitochondrial oxidative damage and oxidative DNA damage contributes to the neurodegenerative process in sporadic amyotrophic lateral sclerosis

    Free Radic Res

    (2008)
  • Cited by (256)

    View all citing articles on Scopus
    View full text