Vol. 38, No. 6795 Biol. Pharm. Bull. 38, 795–808 (2015)

Review Common Mechanisms of Onset of Cancer and Neurodegenerative Diseases Hiroyoshi Ariga Faculty of Pharmaceutical Sciences, Hokkaido University; Kita 12, Nishi 6, Kita-ku, Sapporo 060–0812, Japan. Received February 9, 2015 Onset of cancer and neurodegenerative disease occurs by abnormal cell growth and neuronal cell death, respectively, and the number of patients with both diseases has been increasing in parallel with an increase in mean lifetime, especially in developed countries. Although both diseases are sporadic, about 10% of the diseases are genetically inherited, and analyses of such familial forms of gene products have contributed to an understanding of the molecular mechanisms underlying the onset and pathogenesis of these diseases. I have been working on c-myc, a protooncogene, for a long time and identified various c-Myc-binding proteins that play roles in c-Myc-derived tumorigenesis. Among these proteins, some proteins have been found to be also responsible for the onset of neurodegenerative diseases, including Parkinson’s disease, retinitis pigmentosa and cerebellar atrophy. In this review, I summarize our findings indicating the common mechanisms of onset between cancer and neurodegenerative diseases, with a focus on genes such as DJ-1 and Myc-Modulator 1 (MM-1) and signaling pathways that contribute to the onset and pathogenesis of cancer and neurodegenerative diseases. Key words

1.

c-Myc; DJ-1; Myc-Modulator 1 (MM-1); cancer; Parkinson’s disease; neurodegenerative disease

INTRODUCTION

Products of oncogenes and tumor suppressor genes play pivotal roles in regulation of cell growth, differentiation and death, and aberrant activation of oncogene products and inactivation of tumor suppressor proteins lead to abnormal cell growth, resulting in the onset of cancer. While mutation in both alleles of an oncogene is necessary for tumorigenesis, mutation in either allele of a tumor suppressor gene is sufficient. Onset and pathogenesis of neurodegenerative diseases, including Alzheimer’s disease, Parkinson’s disease (PD) and Huntington disease, are affected by environmental and genetic factors that lead to abrogation of synaptic function, culminating in neuronal cell death. Although exits of cancer and neurodegenerative diseases are completely opposite, there are some common points of view between both diseases, including growth and death signaling pathways. Diseases are categorized by two types, non-inherited (sporadic) and inherited (familial) cases, and investigation of products of causative genes for familial cases has contributed to the unraveling of molecular mechanisms underlying the onsets of diseases. Furthermore, recent findings, including ours, have shown that some genes are responsible for both cancer and neurodegenerative diseases, thereby advocating the scenario of “common mechanisms of onsets of cancer and neurodegenerative diseases.” I have been working on oncogene c-myc for a long time and identified various c-Myc-binding proteins. Of the proteins identified, some proteins have been found to directly affect the onset and pathogenesis of both cancer and neurodegenerative diseases. In this review, I show such cases, focusing on DJ-1 and Myc-Modulator 1 (MM-1), and discuss the common mechanism between cancer and neurodegenerative diseases.



2. FUNCTION OF C-MYC AND IDENTIFICATION OF C-MYC-BINDING PROTEINS The c-myc gene is a protooncogene, and its gene product c-Myc is comprised of 439 amino acids and harbors two domains, Myc-box I/II and basic-helix-loop-helix-leucine zipper (bHLHzip) at the N-terminus and C-terminus, respectively (Fig. 1). c-Myc is a transcription factor and binds to the E-box sequence, CAC GTG, through its basic-helix-loop-helix region after dimerization with another bHLHzip protein, Max, via the leucine zipper structure.1) In addition to the bHLHzip region, Myc-boxes I and II are also essential for transcription activity of c-Myc through association with the co-activator complex.2–4) c-Myc is known to regulate transcription of more than 50% of genes present in cells, including genes related to metabolism, ribosomal synthesis and progression of the cell cycle, thereby leading to increase of cell mass.3–5) We have found for the first time that c-Myc is a factor that regulates initiation of DNA replication6) and that c-Myc binds to one of the replication origins located at the region upstream of the c-myc gene, which also acts as a transcriptional enhancer, in association with origin recognition complex (ORC) and sequence-specific DNA-binding protein Myc Single-Strand binding Proteins (MSSP), one of the c-Myc-binding proteins we identified.7–11) This case is also true for the replication origin/enhancer in the heat shock protein 70 gene to which c-Myc/NF-Y binds,12) giving rise to the idea of a concerted mechanism of DNA replication and transcription.13,14) Although DNA replication function of c-Myc had been debated for more than 10 years after our proposal,15) many reports supporting DNA replication function of c-Myc have been published and it has finally been corroborated.16) Amplification and mutation in the c-myc gene have been observed in almost all cancer cells. Translocation of the c-myc gene to other genes has also been observed in Burkitt lymphoma, acute lymphoblastic leukemia and multiple myeloma.17)

e-mail: [email protected] © 2015 The Pharmaceutical Society of Japan

Biol. Pharm. Bull.

796

Fig. 1.

Vol. 38, No. 6 (2015)

Structure of c-Myc and a Role of MM-1

c-Myc comprises two functional domains at the N-terminus and C-terminus. Max binds to the leucine zipper structure to bind to the E-box sequence in the target genes. MM-1 binds to Myc-box II to repress transcriptional activity of c-Myc after recruiting the co-repressor complex containing HDAC. Repressive activity of MM-1 toward c-Myc activity is lost by the A157R mutant, and c-Myc is aberrantly activated, leading to the onset of cancer. MBI: Myc-box I; MBII: Myc-box II; b: basic region; HLH: helix-loop-helix; zip: leucine zipper.

Of the mutations in c-Myc, threonine 58 (T58) and serine 62 (S62) located in Myc-box 1 have frequently been mutated in cancer cells. c-Myc is an unstable protein and its half-life is only about 20 min.18) Although c-Myc is degraded by the ubiquitin–proteasome system after the cell cycle progresses to the S-phase, mutations of T58 and S62 render c-Myc stable, and the cell cycle-promoting activity of c-Myc therefore continues, resulting in the onset of cancer.19) Although mutation in Myc-box II in cancer cells is rare, Myc-box II is essential for the transforming activity of c-Myc. Transforming activity of c-Myc is lost after mutation in Myc-box II, and c-Myc lacking Myc-box I but containing Myc-box II still possesses transforming activity.20) Thus, both Myc-boxes I and II are necessary for transcriptional activity, but Myc-box II is sufficient for transforming activity of c-Myc. As described above, c-Myc with Max binds to the E-box sequence and positively regulates transcription of E-box-containing genes. In addition, c-Myc indirectly regulates transcription or DNA replication by binding to other DNA-binding factors. There are several cascades that lead to activation or repression of c-Myc activity. To investigate c-Myc function and c-Myc cascades,

c-Myc-binding proteins have been identified by using various strategies, including two-hybrid screening and immunoprecipitation followed by mass spectrometry, and more than 30 c-Myc-binding proteins have been identified.21) These proteins are roughly divided into two groups, proteins binding to Mycbox I/II and proteins binding to bHLHzip, and they positively or negatively modulate transcription activity and tumorigenicity of c-Myc. We have identified approximately 10 c-Myc-binding proteins. Among them, novel proteins are MM-1 and Associate of MYC-1 (AMY-1), which bind to Myc-box II, and MSSP, pTwenty-One and CDK-associated protein-1 (TOK-1) and PAP1-Associated protein-1 (PAPA-1), which bind to bHLHzip. While MM-1 and TOK-1 negatively regulate c-Myc activity, AMY-1 and MSSP positively regulate c-Myc activity.9,22–25) We have also identified novel c-Myc-modulator proteins, DJ-1 and Pim-1 Associated Protein-1 (PAP-1).26,27) Since MM-1, PAP-1 and DJ-1 were found to be related to onsets of neurodegenerative diseases, I hereafter focus on these three proteins.

Dr. Hiroyoshi Ariga was born in Okaya, Nagano prefecture, in 1951. He obtained his Ph.D. degree from the University of Tokyo in 1979 under the supervisor of Prof. Hiroto Shimojo. After spending his postdoctoral training at the Albert Einstein College of Medicine, New York, U.S.A., he joined to the Institute of Medical Sciences at The University of Tokyo as an assistant professor in 1981. After spending time as a research associate at the State University of New York at Stony Brook, Stony Brook, U.S.A., he returned to the Institute of Medical Sciences in 1983 and then moved to the Faculty of Pharmaceutical Sciences at Hokkaido University as a full professor in 1989. He was awarded the Japanese Biochemical Society award for the Young Investigator in 1987 and the Pharmaceutical Society of Japan Award in 2015. His major research fields are molecular biology and neurochemistry, with a focus on elucidation of the mechanisms of onset of cancer and neurodegenerative diseases.

Hiroyoshi Ariga

Biol. Pharm. Bull. Vol. 38, No. 6 (2015)797

Fig. 2.

Two Roles of MM-1

MM-1 acts as a prefoldin subunit, prefoldin 5, in the cytoplasm. Prefoldin inhibits oligomer formation of pathogenic Huntingtin and α-synuclein, causative gene products of Huntington’s disease and Parkinson’s disease, respectively. MM-1 acts as a tumor suppressor in the nucleus as shown in Fig. 1.

3. COMMON MECHANISMS OF ONSETS OF CANCER AND NEURODEGENERATIVE DISEASES 3.1. Tumor Suppressor MM-1 and Neurodegeneration MM-1 was isolated by two-hybrid screening using c-Myc as a bait in 1998 and found to bind to Myc-box II.22) MM-1 contains four splicing variants, MM-1α, MM-1β, MM-1γ and MM-1δ from the sequence of chromosome 12, and subcellular localization of MM-1 splicing variants differs in the cytoplasm or nucleus.28) The first isolated MM-1 is MM-1α with additional 13 amino acids from the sequence of chromosome 14 at the N-terminus, but the function of MM-1 has been found to be the same as that of MM-1α. I therefore use MM-1α as MM-1 in this review. MM-1 negatively regulates transcriptional activity of c-Myc through the TIF1β/mSin3 co-repressor complex,29) and one of the c-Myc/MM-1 target genes is protooncogene c-fms30) (Figs. 1, 2). Missense mutation of amino acid number 157 from alanine to arginine (A157R) was observed in about 50–60% of patients with leukemia and lymphoma and in more than 75% of patients with squamous cell carcinoma of tongue cancer, and A157R lost the activities to repress both the E-boxdependent transcriptional activity of c-Myc and the myc/ras cooperative transforming activity, indicating that MM-1 is a tumor suppressor.31) MM-1 also negatively regulates expression of the wnt4 gene through binding to Egr-1 on the wnt4 gene promoter. The wnt4 gene is located at the top of the Wnt signal that leads to activation of c-myc expression. MM-1 therefore indirectly represses c-myc expression via the Wnt signal.32) Furthermore, MM-1 directly binds to a subunit of the proteasome and to Rabring7, a modulator of ubiquitin ligase, thereby facilitating ubiquitination and subsequent degradation of c-Myc.33,34) Thus, MM-1 represses c-Myc activity in various ways in the nucleus. Prefoldin is a molecular chaperone comprised of 6 subunits, PFD1–6, and PFD5 has been found to be MM-1.35,36) Prefoldin

assists folding of newly synthesized proteins such as actin and tubulin after proteins have been transferred to chaperonin in the cytoplasm. We found that prefoldin inhibited oligomer formation of pathogenic Huntingtin and α-synuclein, causative gene products for Huntington disease and PD, respectively, thereby inhibiting Huntingtin- and α-synuclein-induced neuronal cell death (Fig. 2). Knockdown of MM-1/PFD5 resulted in disruption of the prefoldin complex and loss of protective activity against oligomer formation of pathogenic Huntingtin and α-synuclein.37–39) Prefoldin also modulates aggregation of Amyloid-β (Aβ).40,41) Furthermore, mice possessing a missense mutation in MM-1α at amino acid number 110 from leucine to arginine (L110R) exhibit various symptoms, including degeneration of the cerebellum and retina and male infertility.42) The levels of ubiquitinated proteins and cytotoxicity were higher in L110R MM-1 cells than in wild-type cells under normal conditions and were increased by various stresses. The level of polyubiquitinated protein aggregation was increased in the brains of L110R MM-1α mice.43) These results suggest that MM-1-containing prefoldin plays a role in quality control against protein aggregation and that dysfunction of prefoldin is one of the causes of neurodegenerative diseases. 3.2. Splicing Factor PAP-1 and Retinitis Pigmentosa PAP-1 has been isolated by two-hybrid screening using Pim-1 as a bait.27) The Pim-1 gene is an oncogene that cooperates with c-myc in tumorigenesis. Since Pim-1 and c-Myc are usually located in the cytoplasm and nucleus, respectively, and since both proteins are not directly associated, we have screened for Pim-1- and c-Myc-binding proteins and identified two novel proteins, PAP-1 and PAPA-1. Pim-1 directly binds to PAP-1, and PAPA-1 directly binds to PAP-1 and c-Myc, indicating a Pim-1 cascade from Pim-1, PAP-1, PAPA-1 and c-Myc. Missense mutations in the RP9 gene, a causative gene for autonomously dominant inherited retinitis pigmentosa, have been identified, and it has been shown that the PAP-1 gene is identical to the RP9 gene.44) Retinitis pigmentosa is a

Biol. Pharm. Bull.

798

neurodegenerative disease that occurs in the retina in which retinal apoptosis is elicited. We found that PAP-1 is a transcriptional splicing factor located in the spliceosome and that mutations of PAP-1 observed in RP9 patients disturb correct splicing.45,46) Although the molecular mechanism underlying the onset of retinitis pigmentosa by PAP-1 mutations is still not known, it is thought that PAP-1 regulates splicing of the gene(s) that is expressed in the retina and is responsible for formation or function of the retina and that mutation of PAP-1 compromises proper splicing of the gene, leading to retinal apoptosis. 3.3. Oncoprotein DJ-1 and Parkinson’s Disease DJ-1 was isolated in the course of screening for c-Myc-binding proteins in 1997.26) Although DJ-1 does not directly bind to c-Myc, the DJ-1 gene was found to transform NIH3T3 cells in cooperation with the activated ras gene, indicating that the DJ-1 gene is a novel oncogene.26) DJ-1 also accelerates transformation by c-myc through activating the Erk pathway.47) DJ-1 is localized in the cytoplasm, nucleus and mitochondria26,48) and is secreted into culture medium49–51) or serum,52–59) cerebrospinal fluid,53,54,60) saliva61,62) and nipple

Fig. 3.

Vol. 38, No. 6 (2015)

fluid.63) DJ-1 is translocated from the cytoplasm to nucleus upon addition of a mitogen to the culture medium26) and is translocated to mitochondria after oxidative stress.64) Increased levels of DJ-1 expression have been observed in various cancer cells and tissues, including breast,51,52) prostate,65,66) lung,67,68) and endometrial,69,70) thyroid,71,72) pancreas,73–76) esophageal,77) ovary,78,79) cervical,80) liver,81) gastric,82,83) supraglottic84) cancers, cholangiocarcinoma,85) glioma/glioblastomas,86,87) laryngeal carcinoma,88) bladder carcinoma89) and melanoma57,90) and leukemia.91,92) Secretion of DJ-1 into the serum or nipple fluid has been found in patients with breast cancer.52,63) Increased levels of DJ-1 expression in cancer cells are parallel to severity of cancer with poor prognosis, including metastasis and invasion.82,83,87,90,93–98) PD is a progressive neuronal degenerative disease. Since dopaminergic neuron death occurs in the substantia nigra pars compacta, the level of dopamine in the stratus is reduced, leading to movement dysfunction, a phenomenon of PD. Although the molecular mechanism of the onset of PD is still not fully understood, it is thought that oxidative stress and mitochondrial dysfunction are the main causes of PD.99–102) Oxida-

Model of the Onset of Parkinson’s Disease

When neuronal cells receive oxidative stress, mitochondria, especially complex I of the respiration complexes, are compromised, facilitating production of large amounts of reactive oxygen species (ROS). ROS enhance oxidative stress to cells, leading to a vicious cycle of oxidative stress. ROS injure and oxidize proteins, making the proteins insoluble. When activity of protein degradation systems such as the ubiquitin–proteasome system is reduced, insoluble proteins are aggregated. Aggregated proteins are degraded by chaperone-mediated autophagy. Damaged mitochondria are degraded by mitophagy. Proteins identified as causative gene products in a familial form of PD play crucial roles in each step during the onset of PD. Reduction and loss of functions of these proteins induce synaptic dysfunction followed by neuronal cell death.

Biol. Pharm. Bull. Vol. 38, No. 6 (2015)799

Fig. 4.

Oxidation of Cysteine Residues in DJ-1

DJ-1 has three cysteine residues located at amino acid numbers 46, 54 and 106 (C46, C53 and C106). Of the three cysteine residues, C106 is the most sensitive to oxidative stress and is oxidized from SH to SOH, SO2H and SO3H, and then C46 and C53 are oxidized. This figure was a modified version of Fig. 2 in our previous manuscript.120)

tive stress compromises mitochondrial function, which results in the production of a large amount of reactive oxygen species (ROS), thus triggering activation of cell death pathways and inducing oxidation of proteins. Proteins oxidized by ROS become insoluble. When the activity of protein degradation systems such as the ubiquitin–proteasome system is reduced, insoluble proteins are aggregated, and the aggregations are degraded by chaperone-mediated autophagy. Damaged mitochondria, on the other hand, are degraded by mitochondriaspecific autophagy (mitophagy) as described below. When protective systems against oxidative stress and against harmful aggregated proteins are hampered, synaptic dysfunction followed by neuronal cell death is induced. Recent studies have shown that causative gene products for familial PD play critical roles in specific steps during the onset of PD99,103,104) (Fig. 3). In 2003, mutations, including a large deletion and homozygous missense mutation, were observed in the park7 locus in patients with a familial form of PD, and the DJ-1 gene was identified as park7.105) This is the first case of an oncogene also being a causative gene for a neurodegenerative disease. More than 10 homozygous and heterozygous mutations have been identified in the DJ-1 gene (see the database, http:// www.molgen.ua.ac.be/PDmutDB/).106,107) Gene product DJ-1 is comprised of 189 amino acids without a specific domain and it works as a homodimer.108–111) DJ-1 has three cysteine residues at amino acid numbers 46, 53 and 106 (C46, C53 and C106, respectively). DJ-1 is a sensor protein against various stresses, including oxidative stress. When cells receive oxidative stress, C106 of DJ-1 is first oxidized from SH (reduced form) to SOH, SO2H and SO3H forms, and then the other cysteine residues C46 and C53 are oxidized112–114) (Fig. 4). DJ-1 at C106 with SO3H is believed to be an inactive form of DJ-1, and accumulation of excessively oxidized DJ-1 has been observed in brains of patients with sporadic PD, Alzheimer’s disease and Huntington’s disease,115–117) suggesting that DJ-1 participates in the onsets of at least both familial PD and sporadic PD.

Fig. 5.

Function of DJ-1

DJ-1 is a multifunctional protein, and excess activation and loss of function of DJ-1 activity are related to onset of cancer and various oxidative stress-related diseases, respectively. FAP: familial amyloid polyneuropathy; COPD: chronic obstructive pulmonary disease. This figure was a modified version of Fig. 1 in our previous manuscript.120)

DJ-1 is a multi-functional protein,118–120) having functions in transcriptional regulation,121–135) anti-oxidative stress reaction,64,112,113,136,137) mitochondrial regulation,48,138–146) regulation of signal transduction,96,123,132,147–152) and functions as a protease,58,153–155) a chaperone156,157) and a deglycase158) (Fig. 5). Aberrant activation of and loss of DJ-1 function are thought to lead to onsets of cancer and oxidative stress-related diseases, including PD,105) stroke,159,160) male infertility,161–163) chronic obstructive pulmonary disease (COPD)164) and type 2 diabetes165) (Fig. 5). 3.3.1. Anti-oxidative Stress Function The main function of DJ-1 is thought to be an anti-oxidative stress function.

Biol. Pharm. Bull.

800

There are several ways for DJ-1 to exert an anti-oxidative stress function: 1) DJ-1 quenches ROS through self-oxidation of its cysteine residues,113,114) 2) DJ-1 activates and stabilizes nuclear factor erythroid-2 related factor 2 (Nrf2), a master transcription factor for redox and detoxification-related genes, through sequestering Keap1, an inhibitor of Nrf2, leading to degradation of Nrf2 by the ubiquitin–proteasome system,126) and 3) DJ-1 upregulates the expression of superoxide dismutase 3 (SOD3) and glutathione ligase, anti-oxidant enzymes, by an unknown mechanism.166,167) Furthermore, DJ-1 directly binds to SOD1 to regulate its activity.168–170) Although DJ-1 is an abundant protein in cells, the contribution level of reduction of oxidative stress by quenching ROS through self-oxidation of DJ-1 is thought to be less than 10% of the total contribution, and aforementioned reactions 2) and 3) contribute to the rest of the anti-oxidative stress function. DJ-1 mutants observed in PD patients have loss of or weaken anti-oxidative stress activity,171) and DJ-1-knockout mice are more vulnerable to neuronal toxins such as 1-methyl-4-phenyl1,2,3,6-tetrahydr (MPTP) than are wild-type mice.172) DJ-1 is expressed in both neuron and glia,115,173,174) and the secretion level of DJ-1 from glia is higher than that from neuron.50) Although it is possible that DJ-1 secreted from glia protects neuron from oxidative stress-induced cell death,160,175,176) there is no direct evidence so far. 3.3.2. Transcriptional Activity of DJ-1 and the Effect of DJ-1 on Synthesis and Secretion of Dopamine DJ-1 binds to various transcription factors to act as a co-activator or co-repressor for regulating their target genes without direct binding to DNA. DJ-1-binding transcription factors or modified proteins identified so far include p53,123,128,132) androgen receptor and its regulatory proteins,121,122,127) poly-

Fig. 6.

Vol. 38, No. 6 (2015)

pyrimidine tract-binding protein-associated splicing factor (PSF),125) Keap1, an inhibitor for nuclear factor erythroid-2 related factor 2 (Nrf2),126) sterol regulatory element-binding protein (SREBP),131) Ras-responsive element-binding protein (RREB1)133) and signal transducer and activator of transcription 1 (STAT1)135) to modulate their transcriptional activity, thereby affecting various cell functions. Positive regulation of SREBP1 and RREB1 functions by DJ-1 may participate in the onset and pathogenesis of metabolic syndrome.131,133) Indeed, recent studies have shown relationships of DJ-1 with glucose, hypertension and obesity markers.165,177–183) As for Parkinson’s disease, dopamine is synthesized from tyrosine by two enzymes, tyrosine hydroxylase (TH) and dopamine decarboxylase (DDC), and packed into vesicles by vesicular monoamine transporter 2 (VMAT2). Expression of the TH gene is negatively regulated by PSF, a repressor of transcription.125) We found that DJ-1 positively regulates TH gene expression by sequestering PSF from the TH gene promoter in human cells but not in mouse cells due to the presence of the PSF recognition sequence in the human TH gene but not in the mouse TH gene.130) Furthermore, we found that DJ-1 directly binds to TH and DDC proteins to activate these enzymatic activities in an oxidative status of C106 of DJ-1-dependent manner184) and that DJ-1 positively regulates VMAT2 by upregulating VMAT2 gene expression and by direct binding to VMAT2 protein.185) These results suggest that DJ-1 directly regulates biosynthesis and secretion of dopamine. 3.3.3. Activation and Repression of Cell Growth and Cell Death Signaling Pathways, Respectively, by DJ-1 DJ-1 binds to various proteins that play roles in signaling pathways (Fig. 6). DJ-1 binds to p53, a tumor suppressor protein and transcription factor, to modulate p53 activity. p53

DJ-1-Mediated Cell Growth and Cell Death Pathways

DJ-1 binds to various proteins located in cell growth and cell death pathways and affects positively and negatively cell growth and cell death pathways, respectively.

Biol. Pharm. Bull. Vol. 38, No. 6 (2015)801

is well known to respond to various stresses. When cells are exposed to ultraviolet light (UV), for instance, the expression level of p53 is increased, and p53 is activated by its phosphorylation. Activated p53 attenuates cell growth (G1 arrest) to check the injured level of DNA and facilitates repair of injured DNA. When the injured level of DNA is too severe, p53 up-regulates pro-apoptotic genes such as Bax gene to kill cells containing damaged DNA. DJ-1 modulates p53 activity in two ways: DJ-1 binds to Topors/p53-binding protein3, which is small ubiquitin-related modifier-1 (SUMO-1) ligase. When p53 is sumoylated by Topors, p53 activity is compromised, and Topors-inhibited p53 activity is restored by DJ-1 through the canceling binding of Topors to p53.123) SUMO-1 conjugation modifies the activities of many proteins, including PSF described above,125) and DJ-1 regulates sumoylation levels of proteins by binding to SUMO-1 ligases such as Topors and protein inhibitor of activated STAT (PIAS) family proteins.121,123) DJ-1 itself is sumoylated at lysine 106. Sumoylation of DJ-1 is necessary for full activities of DJ-1, and L166P DJ-1, a pathogenic mutant DJ-1 observed in a PD patient, is excessively sumoylated, resulting in aggregation.186) The other way for DJ-1 to modulate p53 is by directly binding to the DNA-binding region of p53 and inhibiting p53 activity in both oxidative status of C106 of DJ-1- and DNA-binding affinity of p53-dependent manners.132) In this case, DJ-1 inhibits expression of the p53-target gene DUSP1 encoding protein phosphatase, resulting in activation of Erk1/2 and thereby stimulating cell growth.132) When cells receive signals from growth factors, their receptors with tyrosine kinase activity are activated by self-phosphorylation. Activated receptors then phosphorylate adaptor proteins to transduce growth signals to Ras or phosphoinositide 3-kinase (PI3K), and Ras or PI3K activates Erk and Akt pathways, respectively, to stimulate cell growth and inhibit apoptosis (Fig. 6). Phosphatase and tensin homolog deleted on chromosome10 (PTEN) is a lipid phosphatase and is a negative regulator of the PI3K/Akt pathway. Under an oxidative stress condition, weakly oxidized DJ-1 inhibits PTEN phosphatase activity by direct interaction with PTEN to activate Akt.147,148) When cells receive a signal from a growth factor such as epidermal growth factor (EGF), DJ-1 activates the Erk pathway by an unknown mechanism.96,149) Thus, DJ-1 facilitates cell growth by activating both Akt and Erk pathways. DJ-1 has been identified as an oncogene in cooperation with activated ras.26) Since Ras activates the Erk pathway, it is thought that the mechanism underlying ras-dependent transforming activity of DJ-1 is activation of the Erk pathway. Death associated protein (Daxx) and homeodomain interacting protein kinase 1 (HIPK1) are activators of apoptosis-activating kinase 1 (ASK1) and stimulate ASK1 in the cytoplasm upon the apoptosis signal.187,188) DJ-1 binds to Daxx in the nucleus to inhibit translocation of Daxx into the cytoplasm.150) DJ-1 also binds to ASK1 and HIPK1.151,152) All of these associations of DJ-1 with Daxx, ASK1 and HIPK1 inhibit the apoptosisinducing activity of ASK1, leading to cell growth (Fig. 6). These cell growth and apoptosis signals have been known to be used both in cancer and neurodegenerative diseases. In animal models of and patients with Parkinson’s disease, for instance, highly oxidized DJ-1 is accumulated concomitantly with activation of PTEN, thus reducing Akt and Erk activities and thereby stimulating the apoptosis signaling pathway. In

cancer cells, on the other hand, an elevated expression level of DJ-1 is observed with inactivation of PTEN, thus increasing Akt and Erk activities, culminating in the stimulation of cell growth. Also, opposite phenomena of p53 pathways have been reported in cancer and neurodegenerative diseases. 3.3.4. Regulation of Mitochondrial Activity by DJ-1 A small amount of DJ-1 is localized in the outer and inner membranes of mitochondria under normal conditions, and DJ-1 is translocated to mitochondria upon oxidative stress.48,64,145) Mitochondria with abnormal morphology such as fragmented mitochondria and with reduced mitochondrial membrane potential are observed in DJ-1-knockout mice.142,143) We have found that DJ-1 binds to subunits of mitochondrial complex I in respiration complexes, enhances its activity, and is co-localized with mitochondrial complex I in the mitochondrial inner membrane.48) Furthermore, the N-terminal 12 amino acids are necessary for DJ-1 to localize to mitochondria, and pathogenic mutants of DJ-1 such as L166P and M26I DJ-1 are localized in mitochondria as monomers.145) Since dimer formation is necessary for DJ-1 to exert its activity, it is reasonable that monomers of L166P and M26I DJ-1 are inactive. DJ-1 also binds to pyrroline-5-carboxylate reductase 1 (PYCR1), a mitochondrial enzyme that catalyzes the last step in proline biosynthesis and responds to oxidative stress, to protect cells from oxidative stress-induced cell death.146) DJ-1 is associated with PGC-1α, a master regulator of mitochondrial gene expression, and enhances the expression of oxidative stress-related genes.189) All of the findings described above suggest a role of DJ-1 in quality control of mitochondria. Damaged mitochondria are degraded by the mitochondriaspecific autophagy system called mitophagy. When mitochondria are injured, mitochondrial membrane potential is reduced. Parkin, a park2 gene product and ubiquitin ligase, is recruited to the outer mitochondrial membrane in damaged mitochondria and is activated through its phosphorylation by Pink1, a park6 gene product and protein kinase. Activated Parkin then ubiquitinates outer mitochondrial membrane-resident proteins, including mitofusion 2 and voltage-dependent anion-selective channel protein 1 (VDAC1), and recruits the macroautophagy system in which damaged mitochondria are engulfed.190) The role of DJ-1 in mitophagy is not clear and is still in debate: some studies have shown that DJ-1 is essential for the basal level of autophagy140) and facilitates Parkin/Pink1-mediated mitophagy.141–143) Further studies are needed to clarify the role of DJ-1 in mitophagy. 3.4. Identification of DJ-1-Binding Compounds and Their Application to Treatment of Neurodegenerative Diseases Symptomatic treatments have been used for neurodegenerative diseases. Since dopamine level is decreased in the striatum of PD brains and since dopamine does not penetrate the blood brain barrier (BBB), a precursor and releaser of dopamine or an inhibitor of the enzyme that breaks down dopamine have been used for PD therapy. However, neuronal cell death still occurs during these therapies. A drug(s) that inhibits oxidative stress-induced neuronal cell death is therefore needed. We first examined the effect of recombinant DJ-1 protein on protective activity against toxin 6-hydroxydopamine (6-OHDA)-induced PD in model rats. When 6-OHDA is injected into the substantia nigra, neuronal cell death in both

Biol. Pharm. Bull.

802

the substantia nigra and striatum occurs, and subsequent locomotion defect is observed. Co-injection of 6-OHDA with recombinant wild-type DJ-1, but not with L166P DJ-1, into the substantia nigra ameliorated such PD symptoms.191) Administration of recombinant DJ-1 also protected against neurodegeneration caused by focal cerebral ischemia and reperfusion in rats, a stroke model of rats.160) These findings suggest that DJ-1 itself is a target for PD and stroke therapies. As described above, all of the activities of DJ-1 depend on the oxidative level of C106, and excessively oxidized DJ-1, an inactive form of DJ-1, has been observed in brains of patients with PD, Alzheimer’s disease and Huntington’s disease.115–117) Therefore, it is thought that a drug(s) inhibiting excess oxidation of C106 of DJ-1 may serve as a useful drug for PD therapy. We first clarified the X-ray crystal structure of DJ-1 and found that there is a pocket around C106 of DJ-1.108) After in silico screening for DJ-1-binding compounds using a university compound library and Zinc library, various compounds, including compound B/UCP0054278, compound A/

Fig. 7.

Vol. 38, No. 6 (2015)

UCP0045037 and compound-23, were obtained192–195) (Fig. 7). These compounds inhibit excess oxidation of C106 of DJ-1 and maintain the reduced form DJ-1, thereby inhibiting oxidative stress-induced neuronal cell death and mitigating locomotion defect in mouse and rat PD models concomitantly with increased TH and mitochondrial complex I activities. These compounds are able to penetrate the BBB192,195,196) and are also effective in stroke model rats.197) Protective activities of DJ-1-binding compounds are lost in DJ-1-knockdown or -knockout cells and in DJ-1-knockout mice, indicating the DJ-1-dependency of compounds.192,195,198) These DJ-1-binding compounds are promising for wide ranges of neurodegenerative diseases and need further studies such as a study on their structure–activity relationship to improve their activities.

4.

SUMMARY

A low level of ROS acts as a second messenger to promote cell growth. A high level of ROS, however, damages DNA,

Isolation of DJ-1-Binding Compounds

A. Crystal structure of DJ-1. DJ-1 works as a dimer, and there is a pocket around the C106 region. Compounds that bind to the C106 region have been identified by in silico screening. B. Chemical structures of two DJ-1-binding compounds, compound B and compound-23.

Table 1.

Genes Responsible for the Onset of Both Cancer and Neurodegenerative Diseases

Gene

Gene in cancer

Neurodegenerative disease

Function of protein

DJ-1 MM-1 PAP-1 Pink1 Parkin APP

Oncogene Tumor suppressor gene Modulator Modulator Tumor suppressor gene Oncogene?

Parkinson’s disease Cerebellar atrophy Retinitis pigmentosa Parkinson’s disease Parkinson’s disease Alzheimer’s disease

Anti-oxidative stress, mitochondrial regulation Transcriptional repression, inhibition of protein aggregation Splicing reaction Mitophagy Mitophagy, ubiquitination Precursor of Aβ

Biol. Pharm. Bull. Vol. 38, No. 6 (2015)803

proteins and lipids and compromises mitochondrial function, leading to the onset of various diseases, including cancer and neurodegenerative diseases. Human cells possess defense systems against ROS and ROS-induced cell death, and genes/ proteins that have been identified as causes for either cancer or neurodegenerative diseases play crucial roles in these defense systems. In this review, I introduced these players with focus on our findings. In addition, common players in cancer or neurodegenerative diseases that have so far been identified are listed in Table 1. Although the precise mechanisms of commonality and difference between these diseases have just started to be investigated, investigation of these mechanisms is expected to lead to the development of a drug(s) for therapies of mostly inevitable diseases such as cancer and neurodegenerative diseases. Acknowledgment I would like to thank all of the members who have participated in our projects. Conflict of Interest interest.

The author declares no conflict of

REFERENCES 1) Blackwood EM, Eisenman RN. Max: a helix-loop-helix zipper protein that forms a sequence-specific DNA-binding complex with Myc. Science, 251, 1211–1217 (1991). 2) Diolaiti D, McFerrin L, Carroll PA, Eisenman RN. Functional interactions among members of the MAX and MLX transcriptional network during oncogenesis. Biochim. Biophys. Acta, 1849, 484–500 (2015). 3) Sabò A, Amati B. Genome recognition by MYC. Cold Spring Harb. Perspect. Med., 4, a014191 (2014). 4) Rahl PB, Young RA. MYC and transcription elongation. Cold Spring Harb. Perspect. Med., 4, a020990 (2014). 5) Kusnadi EP, Hannan KM, Hicks RJ, Hannan RD, Pearson RB, Kang J. Regulation of rDNA transcription in response to growth factors, nutrients and energy. Gene, 556, 27–34 (2015). 6) Iguchi-Ariga SMM, Itani T, Kiji Y, Ariga H. Possible function of c-myc products: promotion of cellular DNA replication. EMBO J., 6, 2365–2371 (1987). 7) Iguchi-Ariga SMM, Okazaki T, Itani T, Ogata M, Sato Y, Ariga H. An initiation site of DNA replication with transcriptional enhancer activity present upstream of c-myc gene. EMBO J., 7, 3135–3142 (1988). 8) Ariga H, Imamura Y, Iguchi-Ariga SMM. DNA replication origin and transcriptional enhancer in c-myc gene share the c-myc protein binding sequences. EMBO J., 8, 4273–4279 (1989). 9) Negishi Y, Iguchi-Ariga SMM, Ariga H. Protein complexes bearing myc-like antigenicity recognize two distinct DNA sequences. Oncogene, 7, 543–548 (1992). 10) Negishi Y, Nishita Y, Saëgusa Y, Kakizaki I, Galli I, Kihara F, Tamai K, Miyajima N, Iguchi-Ariga SMM, Ariga H. Identification and cDNA cloning of single-stranded DNA binding proteins that interact with the region upstream of the human c-myc gene. Oncogene, 9, 1133–1143 (1994). 11) Takayama M, Taira T, Tamai K, Iguchi-Ariga SMM, Ariga H. ORC1 interacts with c-Myc to inhibit E-box-dependent transcription by abrogating c-Myc-SNF5/INI1 interaction. Genes Cells, 5, 481–490 (2000). 12) Taira T, Sawai M, Ikeda M, Tamai K, Iguchi-Ariga SMM, Ariga H. Cell cycle-dependent switch of up- and down-regulation of human hsp70 gene expression by interaction between c-Myc and CBF/ NF-Y. J. Biol. Chem., 274, 24270–24279 (1999).

13) Iguchi-Ariga SMM, Ariga H. Concerted mechanism of DNA replication and transcription. Cell Struct. Funct., 14, 649–651 (1989). 14) DePamphilis ML. Transcriptional elements as components of eukaryotic origins of DNA replication. Cell, 52, 635–638 (1988). 15) Gutierrez C, Guo ZS, Burhans W, DePamphilis ML, Farrell-Towt J, Ju G. Is c-myc protein directly involved in DNA replication? Science, 240, 1202–1203 (1988). 16) Dominguez-Sola D, Ying CY, Grandori C, Ruggiero L, Chen B, Li M, Galloway DA, Gu W, Gautier J, Dalla-Favera R. Non-transcriptional control of DNA replication by c-Myc. Nature, 448, 445–451 (2007). 17) Said J, Lones M, Yea S. Burkitt lymphoma and MYC: what else is new? Adv. Anat. Pathol., 21, 160–165 (2014). 18) Hann SR, Eisenman RN. Proteins encoded by the human c-myc oncogene: differential expression in neoplastic cells. Mol. Cell. Biol., 4, 2486–2497 (1984). 19) Farrell AS, Sears RC. MYC degradation. Cold Spring Harb. Perspect. Med., 4, a014365 (2014). 20) Hirst SK, Grandori C. Differential activity of conditional MYC and its variant MYC-S in human mortal fibroblasts. Oncogene, 19, 5189–5197 (2000). 21) Tu WB, Helander S, Pilstål R, Hickman KA, Lourenco C, Jurisica I, Raught B, Wallner B, Sunnerhagen M, Penn LZ. Myc and its interactors take shape. Biochim. Biophys. Acta, 1849, 469–483 (2015). 22) Mori K, Maeda Y, Kitaura H, Taira T, Iguchi-Ariga SMM, Ariga H. MM-1, a novel C-MYC associating protein which represses transcriptional activity of C-MYC. J. Biol. Chem., 273, 29794–29800 (1998). 23) Taira T, Maëda J, Onishi T, Kitaura H, Yoshida S, Kato H, Ikeda M, Tamai K, Iguchi-Ariga SMM, Ariga H. AMY-1, a novel C-MYC binding protein that stimulates transcriptional activity of C-MYC. Genes Cells, 3, 549–565 (1998). 24) Ono T, Kitaura H, Ugai H, Murata T, Yokoyama KK, Iguchi-Ariga SMM, Ariga H. TOK-1, a novel p21Cip1-binding protein that cooperatively enhances p21- dependent inhibitory activity toward CDK2 kinase. J. Biol. Chem., 275, 31145–31154 (2000). 25) Kuroda TS, Maita H, Tabata T, Taira T, Kitaura H, Ariga H, IguchiAriga SMM. A novel nucleolar protein, PAPA-1, induces growth arrest as a result of cell cycle arrest at the G1 phase. Gene, 340, 83–98 (2004). 26) Nagakubo D, Taira T, Kitaura H, Ikeda M, Tamai K, Iguchi-Ariga SMM, Ariga H. DJ-1, a novel oncogene which transforms mouse NIH3T3 cells in cooperation with ras. Biochem. Biophys. Res. Commun., 231, 509–513 (1997). 27) Maita H, Harada Y, Nagakubo D, Kitaura H, Ikeda M, Tamai T, Takahashi K, Ariga H, Iguchi-Ariga SMM. PAP-1, a novel target protein of phosphorylation by Pim-1 kinase. Eur. J. Biochem., 267, 5168–5178 (2000). 28) Hagio Y, Kimura Y, Taira T, Fujioka Y, Iguchi-Ariga SMM, Ariga H. Distinct localizations and repression activities of MM-1 isoforms toward c-Myc. J. Cell. Biochem., 97, 145–155 (2006). 29) Satou A, Taira T, Iguchi-Ariga SMM, Ariga H. A novel transrepression pathway of c-Myc: Recruitment of a transcriptional corepressor complex to c-Myc by MM-1, a c-Myc-binding protein. J. Biol. Chem., 276, 46562–46567 (2001). 30) Satou A, Hagio Y, Taira T, Iguchi-Ariga SMM, Ariga H. Repression of the c-fms gene in fibroblast cells by c-Myc-MM-1-TIF1β complex. FEBS Lett., 572, 211–215 (2004). 31) Fujioka Y, Taira T, Maeda Y, Tanaka S, Nishihara H, Iguchi-Ariga SMM, Nagashima K, Ariga H. MM-1, a c-Myc-binding protein, is a candidate for a tumor suppressor in leukemia/lymphoma and tongue cancer. J. Biol. Chem., 276, 45137–45144 (2001). 32) Yoshida T, Kitaura H, Hagio Y, Sato T, Iguchi-Ariga SMM, Ariga H. Negative regulation of the Wnt signal by MM-1 through inhibiting expression of the wnt4 gene. Exp. Cell Res., 314, 1217–1228 (2008).

804

Biol. Pharm. Bull.

33) Kimura Y, Nagao A, Fujioka Y, Satou A, Taira T, Iguchi-Ariga SMM, Ariga H. MM-1 facilitates degradation of c-Myc by recruiting proteasome and a novel ubiquitin E3 ligase. Int. J. Oncol., 31, 829–836 (2007). 34) Narita R, Kitaura H, Torii A, Tashiro E, Miyazawa M, Ariga H, Iguchi-Ariga SMM. Rabring7 degrades c-Myc through complex formation with MM-1. PLoS ONE, 7, e41891 (2012). 35) Geissler S, Siegers K, Schiebel E. A novel protein complex promoting formation of functional alpha- and gamma-tubulin. EMBO J., 17, 952–966 (1998). 36) Vainberg IE, Lewis SA, Rommelaere H, Ampe C, Vandekerckhove J, Klein HL, Cowan NJ. Prefoldin, a chaperone that delivers unfolded proteins to cytosolic chaperonin. Cell, 93, 863–873 (1998). 37) Miyazawa M, Tashiro E, Kitaura H, Maita H, Suto H, Iguchi-Ariga SMM, Ariga H. Prefoldin subunits are protected from ubiquitinproteasome system-mediated degradation by forming complex with other constituent subunits. J. Biol. Chem., 286, 19191–19203 (2011). 38) Tashiro E, Zako T, Muto H, Itoo Y, Sörgjerd K, Terada N, Abe A, Miyazawa M, Kitamura A, Kitaura H, Kubota H, Maeda M, Momoi T, Iguchi-Ariga SM, Kinjo M, Ariga H. Prefoldin protects neuronal cells from polyglutamine toxicity by preventing aggregation formation. J. Biol. Chem., 288, 19958–19972 (2013). 39) Takano M, Tashiro E, Kitamura A, Maita H, Iguchi-Ariga SMM, Kinjo M, Ariga H. Prefoldin prevents aggregation of α-synuclein. Brain Res., 1542, 186–194 (2014). 40) Sakono M, Zako T, Ueda H, Yohda M, Maeda M. Formation of highly toxic soluble amyloid beta oligomers by the molecular chaperone prefoldin. FEBS J., 275, 5982–5993 (2008). 41) Sörgjerd KM, Zako T, Sakono M, Stirling PC, Leroux MR, Saito T, Nilsson P, Sekimoto M, Saido TC, Maeda M. Human prefoldin inhibits Amyloid-β (Aβ) fibrillation and contributes to formation of nontoxic Aβ aggregates. Biochemistry, 52, 3532–3542 (2013). 42) Lee Y, Smith RS, Jordan W, King BL, Won J, Valpuesta JM, Naggert JK, Nishina PM. Prefoldin 5 is required for normal sensory and neuronal development in a murine model. J. Biol. Chem., 286, 726–736 (2011). 43) Abe A, Takahashi-Niki K, Takekoshi Y, Shimizu T, Kitaura H, Maita H, Iguchi-Ariga SMM, Ariga H. Prefoldin plays a role as a clearance factor in preventing proteasome inhibitor-induced protein aggregation. J. Biol. Chem., 288, 27764–27776 (2013). 44) Keen TJ, Hims MM, McKie AB, Moore AT, Doran RM, Mackey DA, Mansfield DC, Mueller RF, Bhattacharya SS, Bird AC, Markham AF, Inglehearn CF. Mutations in a protein target of the Pim-1 kinase associated with the RP9 form of autosomal dominant retinitis pigmentosa. Eur. J. Hum. Genet., 10, 245–249 (2002). 45) Maita H, Kitaura H, Keen TJ, Inglehearn CF, Ariga H, Iguchi-Ariga SMM. PAP-1, the mutated gene underlying the RP9 form of dominant retinitis pigmentosa, is a splicing factor. Exp. Cell Res., 300, 283–296 (2004). 46) Maita H, Kitaura H, Ariga H, Iguchi-Ariga SMM. Association of PAP-1 and Prp3p, the products of causative genes of dominant retinitis pigmentosa, in the tri-snRNP complex. Exp. Cell Res., 302, 61–68 (2005). 47) Kim YC, Kitaura H, Iguchi-Ariga SMM, Ariga H. DJ-1, an oncogene and causative gene for familial Parkinson’s disease, is essential for SV40 transformation in mouse fibroblasts through up-regulation of c-Myc. FEBS Lett., 584, 3891–3895 (2010). 48) Hayashi T, Ishimori C, Takahashi-Niki K, Taira T, Kim Y-C, Maita H, Maita C, Ariga H, Iguchi-Ariga SMM. DJ-1 binds to mitochondrial complex I and maintains its activity. Biochem. Biophys. Res. Commun., 390, 667–672 (2009). 49) Tsuboi Y, Munemoto H, Ishikawa S, Matsumoto K, Iguchi-Ariga SMM, Ariga H. DJ-1, a causative gene product of a familial form of Parkinson’s disease, is secreted through microdomains. FEBS Lett., 582, 2643–2649 (2008). 50) Yanagida T, Tsushima J, Kitamura Y, Yanagisawa D, Takata K,

51)

52)

53)

54)

55)

56)

57)

58)

59)

60)

61)

62)

63)

64)

65) 66)

Vol. 38, No. 6 (2015)

Shibaike T, Yamamoto A, Taniguchi T, Yasui H, Taira T, Morikawa S, Inubushi T, Tooyama I, Ariga H. Oxidative stress induction of DJ-1 protein in reactive astrocytes scavenges free radicals and reduces cell injury. Oxid. Med. Cell. Longev., 2, 36–42 (2009). Tsuchiya B, Iwaya K, Kohno N, Kawate T, Akahoshi T, Matsubara O, Mukai K. Clinical significance of DJ-1 as a secretory molecule: retrospective study of DJ-1 expression at mRNA and protein levels in ductal carcinoma of the breast. Histopathology, 61, 69–77 (2012). Le Naour F, Misek DE, Krause MC, Deneux L, Giordano TJ, Scholl S, Hanash SM. Proteomics-based identification of RS/DJ-1 as a novel circulating tumor antigen in breast cancer. Clin. Cancer Res., 7, 3328–3335 (2001). Maita C, Tsuji S, Yabe I, Hamada S, Ogata A, Maita H, IguchiAriga SMM, Sasaki H, Ariga H. Secretion of DJ-1 into the serum of patients with Parkinson’s disease. Neurosci. Lett., 431, 86–89 (2008). Waragai M, Wei J, Fujita M, Nakai M, Ho GJ, Masliah E, Akatsu H, Yamada T, Hashimoto M. Increased level of DJ-1 in the cerebrospinal fluids of sporadic Parkinson’s disease. Biochem. Biophys. Res. Commun., 345, 967–972 (2006). Waragai M, Nakai M, Wei J, Fujita M, Mizuno H, Ho G, Masliah E, Akatsu H, Yokochi F, Hashimoto M. Plasma levels of DJ-1 as a possible marker for progression of sporadic Parkinson’s disease. Neurosci. Lett., 425, 18–22 (2007). Akazawa YO, Saito Y, Hamakubo T, Masuo Y, Yoshida Y, Nishio K, Shichiri M, Miyasaka T, Iwanari H, Mochizuki Y, Kodama T, Noguchi N, Niki E. Elevation of oxidized DJ-1 in the brain and erythrocytes of Parkinson’s disease model animals. Neurosci. Lett., 483, 201–205 (2010). Bande MF, Santiago M, Blanco MJ, Mera P, Capeans C, RodríguezAlvarez MX, Pardo M, Piñeiro A. Serum DJ-1/PARK 7 is a potential biomarker of choroidal nevi transformation. Invest. Ophthalmol. Vis. Sci., 53, 62–67 (2012). Koide-Yoshida S, Niki T, Ueda M, Himeno S, Taira T, Iguchi-Ariga SMM, Ando Y, Ariga H. DJ-1 degrades transthyretin and an inactive form of DJ-1 is secreted in familial amyloidotic polyneuropathy. Int. J. Mol. Med., 19, 885–893 (2007). Allard L, Burkhard PR, Lescuyer P, Burgess JA, Walter N, Hochstrasser DF, Sanchez JC. PARK7 and nucleoside diphosphate kinase A as plasma markers for the early diagnosis of stroke. Clin. Chem., 51, 2043–2051 (2005). Hirotani M, Maita C, Niino M, Iguchi-Ariga SMM, Hamada S, Ariga H, Sasaki H. Correlation between DJ-1 levels in the cerebrospinal fluid and the progression of disabilities in multiple sclerosis patients. Mult. Scler., 14, 1056–1060 (2008). Devic I, Hwang H, Edgar JS, Izutsu K, Presland R, Pan C, Goodlett DR, Wang Y, Armaly J, Tumas V, Zabetian CP, Leverenz JB, Shi M, Zhang J. Salivary α-synuclein and DJ-1: potential biomarkers for Parkinson’s disease. Brain, 134, e178–e178 (2011). Kang WY, Yang Q, Jiang XF, Chen W, Zhang LY, Wang XY, Zhang LN, Quinn TJ, Liu J, Chen SD. Salivary DJ-1 could be an indicator of Parkinson’s disease progression. Front. Aging Neurosci, 6, 102 (2014). Oda M, Makita M, Iwaya K, Akiyama F, Kohno N, Tsuchiya B, Iwase T, Matsubara O. High levels of DJ-1 protein in nipple fluid of patients with breast cancer. Cancer Sci., 103, 1172–1176 (2012). Canet-Avilés RM, Wilson MA, Miller DW, Ahmad R, McLendon C, Bandyopadhyay S, Baptista MJ, Ringe D, Petsko GA, Cookson MR. The Parkinson’s disease protein DJ-1 is neuroprotective due to cysteine-sulfinic acid-driven mitochondrial localization. Proc. Natl. Acad. Sci. U.S.A., 101, 9103–9108 (2004). Hod Y. Differential control of apoptosis by DJ-1 in prostate benign and cancer cells. J. Cell. Biochem., 92, 1221–1233 (2004). Osman WM, Abd El Atti RM, Abou Gabal HH. DJ-1 and androgen receptor immunohistochemical expression in prostatic carcinoma: a possible role in carcinogenesis. J. Egypt. Natl. Canc. Inst., 25,

Biol. Pharm. Bull. Vol. 38, No. 6 (2015)805 223–230 (2013). 67) MacKeigan JP, Clements CM, Lich JD, Pope RM, Hod Y, Ting JP. Proteomic profiling drug-induced apoptosis in non-small cell lung carcinoma: identification of RS/DJ-1 and RhoGDIalpha. Cancer Res., 63, 6928–6934 (2003). 68) Kim RH, Peters M, Jang Y, Shi W, Pintilie M, Fletcher GC, DeLuca C, Liepa J, Zhou L, Snow B, Binari RC, Manoukian AS, Bray MR, Liu FF, Tsao MS, Mak TW. DJ-1, a novel regulator of the tumor suppressor PTEN. Cancer Cell, 7, 263–273 (2005). 69) Shu K, Xiao Z, Long S, Yan J, Yu X, Zhu Q, Mei T. Expression of DJ-1 in endometrial cancer: Close correlation with clinicopathological features and apoptosis. Int. J. Gynecol. Cancer, 23, 1029–1035 (2013). 70) Morelli M, Scumaci D, Di Cello A, Venturella R, Donato G, Faniello MC, Quaresima B, Cuda G, Zullo F, Costanzo F. DJ-1 in endometrial cancer: a possible biomarker to improve differential diagnosis between subtypes. Int. J. Gynecol. Cancer, 24, 649–658 (2014). 71) Srisomsap C, Subhasitanont P, Otto A, Mueller EC, Punyarit P, Wittmann-Liebold B, Svasti J. Detection of cathepsin B up-regulation in neoplastic thyroid tissues by proteomic analysis. Proteomics, 2, 706–712 (2002). 72) Giusti L, Iacconi P, Ciregia F, Giannaccini G, Donatini GL, Basolo F, Miccoli P, Pinchera A, Lucacchini A. Fine-needle aspiration of thyroid nodules: proteomic analysis to identify cancer biomarkers. J. Proteome Res., 7, 4079–4088 (2008). 73) Melle C, Ernst G, Escher N, Hartmann D, Schimmel B, Bleul A, Thieme H, Kaufmann R, Felix K, Friess HM, Settmacher U, Hommann M, Richter KK, Daffner W, Täubig H, Manger T, Claussen U, von Eggeling F. Protein profiling of microdissected pancreas carcinoma and identification of HSP27 as a potential serum marker. Clin. Chem., 53, 629–635 (2007). 74) Tian M, Cui YZ, Song GH, Zong MJ, Zhou XY, Chen Y, Han JX. Proteomic analysis identifies MMP-9, DJ-1 and A1BG as overexpressed proteins in pancreatic juice from pancreatic ductal adenocarcinoma patients. BMC Cancer, 8, 241 (2008). 75) He XY, Liu BY, Yao WY, Zhao XJ, Zheng Z, Li JF, Yu BQ, Yuan YZ. Serum DJ-1 as a diagnostic marker and prognostic factor for pancreatic cancer. J. Dig. Dis., 12, 131–137 (2011). 76) Tsiaousidou A, Lambropoulou M, Chatzitheoklitos E, Tripsianis G, Tsompanidou C, Simopoulos C, Tsaroucha AK. B7H4, HSP27 and DJ-1 molecular markers as prognostic factors in pancreatic cancer. Pancreatology, 13, 564–569 (2013). 77) Yuen HF, Chan YP, Law S, Srivastava G, El-Tanani M, Mak TW, Chan KW. DJ-1 could predict worse prognosis in esophageal squamous cell carcinoma. Cancer Epidemiol. Biomarkers Prev., 17, 3593–3602 (2008). 78) Barua A, Edassery SL, Bitterman P, Abramowicz JS, Dirks AL, Bahr JM, Hales DB, Bradaric MJ, Luborsky JL. Prevalence of antitumor antibodies in laying hen model of human ovarian cancer. Int. J. Gynecol. Cancer, 19, 500–507 (2009). 79) Chow SN, Chen RJ, Chen CH, Chang TC, Chen LC, Lee WJ, Shen J, Chow LP. Analysis of protein profiles in human epithelial ovarian cancer tissues by proteomic technology. Eur. J. Gynaecol. Oncol., 31, 55–62 (2010). 80) Arnouk H, Merkley MA, Podolsky RH, Stöppler H, Santos C, Alvarez M, Mariategui J, Ferris D, Lee JR, Dynan WS. Characterization of molecular markers indicative of cervical cancer progression. Proteomics Clin., 3, 516–527 (2009). 81) Liu S, Yang Z, Wei H, Shen W, Liu J, Yin Q, Li X, Yi J. Increased DJ-1 and its prognostic significance in hepatocellular carcinoma. Hepatogastroenterology, 57, 1247–1256 (2010). 82) Shimwell NJ, Ward DG, Mohri Y, Mohri T, Pallan L, Teng M, Miki YC, Kusunoki M, Tucker O, Wei W, Morse J, Johnson PJ. Macrophage migration inhibitory factor and DJ-1 in gastric cancer: differences between high-incidence and low-incidence areas. Br. J.

Cancer, 107, 1595–1601 (2012). 83) Li Y, Cui J, Zhang CH, Yang DJ, Chen JH, Zan WH, Li B, Li Z, He YL. High-expression of DJ-1 and loss of PTEN associated with tumor metastasis and correlated with poor prognosis of gastric carcinoma. Int. J. Med. Sci., 10, 1689–1697 (2013). 84) Zhu XL, Wang ZF, Lei WB, Zhuang HW, Hou WJ, Wen YH, Wen WP. Tumorigenesis role and clinical significance of DJ-1, a negative regulator of PTEN, in supraglottic squamous cell carcinoma. J. Exp. Clin. Cancer Res., 31, 94 (2012). 85) Kawase H, Fujii K, Miyamoto M, Kubota KC, Hirano S, Kondo S, Inagaki F. Differential LC-MS-based proteomics of surgical human cholangiocarcinoma tissues. J. Proteome Res., 8, 4092–4103 (2009). 86) Hinkle DA, Mullett SJ, Gabris BE, Hamilton RL. DJ-1 expression in glioblastomas shows positive correlation with p53 expression and negative correlation with epidermal growth factor receptor amplification. Neuropathology, 31, 29–37 (2011). 87) Wang C, Fang M, Zhang M, Li W, Guan H, Sun Y, Xie S, Zhong X. The positive correlation between DJ-1 and β-catenin expression shows prognostic value for patients with glioma. Neuropathology, 33, 628–636 (2013). 88) Shen Z, Ren Y, Ye D, Guo J, Kang C, Ding H. Significance and relationship between DJ-1 gene and surviving gene expression in laryngeal carcinoma. Eur. J. Histochem., 55, e9 (2011). 89) Lee H, Choi SK, Ro JY. Overexpression of DJ-1 and HSP90α, and loss of PTEN associated with invasive urothelial carcinoma of urinary bladder: Possible prognostic markers. Oncol. Lett., 3, 507–512 (2012). 90) Pardo M, Garcia A, Thomas B, Pineiro A, Akoulitchev A, Dwek RA, Zitzmann N. The characterization of the invasion phenotype of uveal melanoma tumour cells shows the presence of MUC18 and HMG-1 metastasis markers and leads to the identification of DJ-1 as a potential serum biomarker. Int. J. Cancer, 119, 1014–1022 (2006). 91) Zhou X, Xu N, Li R, Xiao Y, Gao G, Lu Q, Ding L, Li L, Li Y, Du Q, Liu X. A comparative proteomic study of homoharringtonineinduced apoptosis in leukemia K562 cells. Leuk. Lymphoma, 14, 1–8 (2015). 92) Liu H, Wang M, Li M, Wang D, Rao Q, Wang Y, Xu Z, Wang J. Expression and role of DJ-1 in leukemia. Biochem. Biophys. Res. Commun., 375, 477–483 (2008). 93) Bindukumar B, Schwartz S, Aalinkeel R, Mahajan S, Lieberman A, Chadha K. Proteomic profiling of the effect of prostate-specific antigen on prostate cancer cells. Prostate, 68, 1531–1545 (2008). 94) Zhu XL, Wang ZF, Lei WB, Zhuang HW, Jiang HY, Wen WP. DJ-1: a novel independent prognostic marker for survival in glottic squamous cell carcinoma. Cancer Sci., 101, 1320–1325 (2010). 95) Bai J, Guo C, Sun W, Li M, Meng X, Yu Y, Jin Y, Tong D, Geng J, Huang Q, Qi J, Fu S. DJ-1 may contribute to metastasis of nonsmall cell lung cancer. Mol. Biol. Rep., 39, 2697–2703 (2012). 96) He X, Zheng Z, Li J, Ben Q, Liu J, Zhang J, Ji J, Yu B, Chen X, Su L, Zhou L, Liu B, Yuan Y. DJ-1 promotes invasion and metastasis of pancreatic cancer cells by activating SRC/ERK/uPA. Carcinogenesis, 33, 555–562 (2012). 97) Kawate T, Iwaya K, Kikuchi R, Kaise H, Oda M, Sato E, Hiroi S, Matsubara O, Kohno N. DJ-1 protein expression as a predictor of pathological complete remission after neoadjuvant chemotherapy in breast cancer patients. Breast Cancer Res. Treat., 139, 51–59 (2013). 98) Abd El Atti RM, Abou Gabal HH, Osman WM, Saad AS. Insights into the prognostic value of DJ-1 and MIB-1 in astrocytic tumors. Diagn. Pathol., 8, 126 (2013). 99) Han JY, Kim JS, Son JH. Mitochondrial homeostasis molecules: regulation by a trio of recessive Parkinson’s disease genes. Exp. Neurobiol, 23, 345–351 (2014). 100) Camilleri A, Vassallo N. The centrality of mitochondria in the pathogenesis and treatment of Parkinson’s disease. CNS Neurosci. Ther., 20, 591–602 (2014). 101) Segura-Aguilar J, Paris I, Muñoz P, Ferrari E, Zecca L, Zucca FA.

806

102) 103)

104) 105)

106)

107)

108)

109)

110)

111)

112)

113)

114)

115)

116)

117)

118)

119)

Biol. Pharm. Bull. Protective and toxic roles of dopamine in Parkinson’s disease. J. Neurochem., 129, 898–915 (2014). Dias V, Junn E, Mouradian MM. The role of oxidative stress in Parkinson’s disease. J. Parkinson’s Dis., 3, 461–491 (2013). Abdel-Salam OM. The paths to neurodegeneration in genetic Parkinson’s disease. CNS Neurol. Disord. Drug Targets, 13, 1485–1512 (2014). Bonifati V. Genetics of Parkinson’s disease–state of the art, 2013. Parkinsonism Relat. Disord., 20 (Suppl. 1), S23–S28 (2014). Bonifati V, Rizzu P, van Baren MJ, Schaap O, Breedveld GJ, Krieger E, Dekker MC, Squitieri F, Ibanez P, Joosse M, van Dongen JW, Vanacore N, van Swieten JC, Brice A, Meco G, van Duijn CM, Oostra BA, Heutink P. Mutations in the DJ-1 gene associated with autosomal recessive early-onset parkinsonism. Science, 299, 256–259 (2003). Bonifati V, Oostra BA, Heutink P. Linking DJ-1 to neurodegeneration offers novel insights for understanding the pathogenesis of Parkinson’s disease. J. Mol. Med. (Berl.), 82, 163–174 (2004). Nuytemans K, Theuns J, Cruts M, Van Broeckhoven C. Genetic etiology of Parkinson disease associated with mutations in the SNCA, PARK2, PINK1, PARK7, and LRRK2 genes: a mutation update. Hum. Mutat., 31, 763–780 (2010). Honbou K, Suzuki NN, Horiuchi M, Niki T, Taira T, Ariga H, Inagaki F. The crystal structure of DJ-1, a protein related to male fertility and Parkinson’s disease. J. Biol. Chem., 278, 31380–31384 (2003). Tao X, Tong L. Crystal structure of human DJ-1, a protein associated with early onset Parkinson’s disease. J. Biol. Chem., 278, 31372–31379 (2003). Lee SJ, Kim SJ, Kim IK, Ko J, Jeong CS, Kim GH, Park C, Kang SO, Suh PG, Lee HS, Cha SS. Crystal structures of human DJ-1 and Escherichia coli Hsp31, which share an evolutionarily conserved domain. J. Biol. Chem., 278, 44552–44559 (2003). Wilson MA, Collins JL, Hod Y, Ringe D, Petsko GA. The 1.1-A resolution crystal structure of DJ-1, the protein mutated in autosomal recessive early onset Parkinson’s disease. Proc. Natl. Acad. Sci. U.S.A., 100, 9256–9261 (2003). Mitsumoto A, Nakagawa Y, Takeuchi A, Okawa K, Iwamatsu A, Takanezawa Y. Oxidized forms of peroxiredoxins and DJ-1 on two-dimensional gels increased in response to sublethal levels of paraquat. Free Radic. Res., 35, 301–310 (2001). Taira T, Saito Y, Niki T, Iguchi-Ariga SMM, Takahashi K, Ariga H. DJ-1 has a role in antioxidative stress to prevent cell death. EMBO Rep., 5, 213–218 (2004). Kinumi T, Kimata J, Taira T, Ariga H, Niki E. Cysteine-106 of DJ-1 is the most sensitive cysteine residue to hydrogen peroxidemediated oxidation in vivo in human umbilical vein endothelial cells. Biochem. Biophys. Res. Commun., 317, 722–728 (2004). Bandopadhyay R, Kingsbury AE, Cookson MR, Reid AR, Evans IM, Hope AD, Pittman AM, Lashley T, Canet-Aviles R, Miller DW, McLendon C, Strand C, Leonard AJ, Ariga H, Wood NW, de Silva R, Hardy JA, Lees AJ, Revesz T. The expression of DJ-1 (PARK7) in normal human CNS and idiopathic Parkinson’s disease. Brain, 127, 420–430 (2004). Choi J, Sullards MC, Olzmann JA, Rees HD, Weintraub ST, Bostwick DE, Gearing M, Levey AI, Chin LS, Li L. Oxidative damage of DJ-1 is linked to sporadic Parkinson’s and Alzheimer’s diseases. J. Biol. Chem., 281, 10816–10824 (2006). Sajjad MU, Green EW, Miller-Fleming L, Hands S, Herrera F, Campesan S, Khoshnan A, Outeiro TF, Giorgini F, Wyttenbach A. DJ-1 modulates aggregation and pathogenesis in models of Huntington’s disease. Hum. Mol. Genet., 23, 755–766 (2014). Kahle PJ, Waak J, Gasser T. DJ-1 and prevention of oxidative stress in Parkinson’s disease and other age-related disorders. Free Radic. Biol. Med., 47, 1354–1361 (2009). Wilson MA. The role of cysteine oxidation in DJ-1 function and

Vol. 38, No. 6 (2015)

dysfunction. Antioxid. Redox Signal., 15, 111–122 (2011). 120) Ariga H, Takahashi-Niki K, Kato I, Maita H, Niki T, Iguchi-Ariga SMM. Neuroprotective function of DJ-1 in Parkinson’s disease. Oxid. Med. Cell. Longev., 2013, 683920 (2013), review. 121) Takahashi K, Taira T, Niki T, Seino C, Iguchi-Ariga SMM, Ariga H. DJ-1 positively regulates the androgen receptor by impairing the binding of PIASx alpha to the receptor. J. Biol. Chem., 276, 37556–37563 (2001). 122) Niki T, Takahashi-Niki K, Taira T, Iguchi-Ariga SMM, Ariga H. DJBP: a novel DJ-1-binding protein, negatively regulates the androgen receptor by recruiting histone deacetylase complex, and DJ-1 antagonizes this inhibition by abrogation of this complex. Mol. Cancer Res., 1, 247–261 (2003). 123) Shinbo Y, Taira T, Niki T, Iguchi-Ariga SMM, Ariga H. DJ-1 restores p53 transcription activity inhibited by Topors/p53BP3. Int. J. Oncol., 26, 641–648 (2005). 124) Xu J, Zhong N, Wang H, Elias JE, Kim CY, Woldman I, Pifl C, Gygi SP, Geula C, Yankner BA. The Parkinson’s disease-associated DJ-1 protein is a transcriptional co-activator that protects against neuronal apoptosis. Hum. Mol. Genet., 14, 1231–1241 (2005). 125) Zhong N, Kim CY, Rizzu P, Geula C, Porter DR, Pothos EN, Squitieri F, Heutink P, Xu J. DJ-1 transcriptionally up-regulates the human tyrosine hydroxylase by inhibiting the sumoylation of pyrimidine tract-binding protein-associated splicing factor. J. Biol. Chem., 281, 20940–20948 (2006). 126) Clements CM, McNally RS, Conti BJ, Mak TW, Ting JP. DJ-1, a cancer- and Parkinson’s disease-associated protein, stabilizes the antioxidant transcriptional master regulator Nrf2. Proc. Natl. Acad. Sci. U.S.A., 103, 15091–15096 (2006). 127) Tillman JE, Yuan J, Gu G, Fazli L, Ghosh R, Flynt AS, Gleave M, Rennie PS, Kasper S. DJ-1 binds androgen receptor directly and mediates its activity in hormonally treated prostate cancer cells. Cancer Res., 67, 4630–4637 (2007). 128) Fan J, Ren H, Fei E, Jia N, Ying Z, Jiang P, Wu M, Wang G. Sumoylation is critical for DJ-1 to repress p53 transcriptional activity. FEBS Lett., 582, 1151–1156 (2008). 129) Fan J, Ren H, Jia N, Fei E, Zhou T, Jiang P, Wu M, Wang G. DJ-1 decreases Bax expression through repressing p53 transcriptional activity. J. Biol. Chem., 283, 4022–4030 (2008). 130) Ishikawa S, Taira T, Takahashi-Niki K, Niki T, Ariga H, IguchiAriga SMM. Human DJ-1-specific transcriptional activation of tyrosine hydroxylase gene. J. Biol. Chem., 285, 39718–39731 (2010). 131) Yamaguchi S, Yamane T, Takahashi-Niki K, Kato I, Niki T, Goldberg MS, Shen J, Ishimoto K, Doi T, Iguchi-Ariga SMM, Ariga H. Transcriptional activation of low-density lipoprotein receptor gene by DJ-1 and effect of DJ-1 on cholesterol homeostasis. PLoS ONE, 7, e38144 (2012). 132) Kato I, Maita H, Takahashi-Niki K, Saito Y, Noguchi N, IguchiAriga SMM, Ariga H. Oxidized DJ-1 inhibits p53 by sequestering p53 from promoters in a DNA-binding affinity-dependent manner. Mol. Cell. Biol., 33, 340–359 (2013). 133) Yamane T, Suzui S, Kitaura H, Takahashi-Niki K, Iguchi-Ariga SMM, Ariga H. Transcriptional activation of the cholecystokinin gene by DJ-1 through interaction of DJ-1 with RREB1 and the effect of DJ-1 on the cholecystokinin level in mice. PLoS ONE, 8, e78374 (2013). 134) Yamane T, Sugimoto N, Maita H, Watanabe K, Takahashi-Niki K, Maita C, Kato-Ose I, Ishikawa S, Gao J, Kitaura H, Niki T, IguchiAriga SMM, Ariga H. Identification of DJ-1-associated regions on human genes from SH-SY5Y cells using chromatin immunoprecipitation sequence technique. Mol. Biol., 3, 15 (2013). 135) Kim JH, Choi DJ, Jeong HK, Kim J, Kim DW, Choi SY, Park SM, Suh YH, Jou I, Joe EH. DJ-1 facilitates the interaction between STAT1 and its phosphatase, SHP-1, in brain microglia and astrocytes: A novel anti-inflammatory function of DJ-1. Neurobiol. Dis., 60, 1–10 (2013).

Biol. Pharm. Bull. Vol. 38, No. 6 (2015)807 136) Yokota T, Sugawara K, Ito K, Takahashi R, Ariga H, Mizusawa H. Down regulation of DJ-1 enhances cell death by oxidative stress, ER stress, and proteasome inhibition. Biochem. Biophys. Res. Commun., 312, 1342–1348 (2003). 137) Martinat C, Shendelman S, Jonason A, Leete T, Beal MF, Yang L, Floss T, Abeliovich A. Sensitivity to oxidative stress in DJ1-deficient dopamine neurons: an ES-derived cell model of primary Parkinsonism. PLoS Biol., 2, e327 (2004). 138) Li HM, Niki T, Taira T, Iguchi-Ariga SMM, Ariga H. Association of DJ-1 with chaperones and enhanced association and colocalization with mitochondrial Hsp70 by oxidative stress. Free Radic. Res., 39, 1091–1099 (2005). 139) Zhang L, Shimoji M, Thomas B, Moore DJ, Yu SW, Marupudi NI, Torp R, Torgner IA, Ottersen OP, Dawson TM, Dawson VL. Mitochondrial localization of the Parkinson’s disease related protein DJ-1: implications for pathogenesis. Hum. Mol. Genet., 14, 2063– 2073 (2005). 140) Krebiehl G, Ruckerbauer S, Burbulla LF, Kieper N, Maurer B, Waak J, Wolburg H, Gizatullina Z, Gellerich FN, Woitalla D, Riess O, Kahle PJ, Proikas-Cezanne T, Krüger R. Reduced basal autophagy and impaired mitochondrial dynamics due to loss of Parkinson’s disease-associated protein DJ-1. PLoS ONE, 5, e9367 (2010). 141) Thomas KJ, McCoy MK, Blackinton J, Beilina A, van der Brug M, Sandebring A, Miller D, Maric D, Cedazo-Minguez A, Cookson MR. DJ-1 acts in parallel to the PINK1/parkin pathway to control mitochondrial function and autophagy. Hum. Mol. Genet., 20, 40–50 (2011). 142) Wang X, Petrie TG, Liu Y, Liu J, Fujioka H, Zhu X. Parkinson’s disease-associated DJ-1 mutations impair mitochondrial dynamics and cause mitochondrial dysfunction. J. Neurochem., 121, 830–839 (2012). 143) Heo JY, Park JH, Kim SJ, Seo KS, Han JS, Lee SH, Kim JM, Park JI, Park SK, Lim K, Hwang BD, Shong M, Kweon GR. DJ-1 null dopaminergic neuronal cells exhibit defects in mitochondrial function and structure: involvement of mitochondrial complex I assembly. PLoS ONE, 7, e32629 (2012). 144) Giaime E, Yamaguchi H, Gautier CA, Kitada T, Shen J. Loss of DJ-1 does not affect mitochondrial respiration but increases ROS production and mitochondrial permeability transition pore opening. PLoS ONE, 7, e40501 (2012). 145) Maita C, Maita H, Iguchi-Ariga SMM, Ariga H. Monomer DJ-1 and its N-terminal sequence are necessary for mitochondrial localization of DJ-1 mutants. PLoS ONE, 8, e54087 (2013). 146) Yasuda T, Kaji Y, Agatsuma T, Niki T, Arisawa M, Shuto S, Ariga H, Iguchi-Ariga SMM. DJ-1 cooperates with PYCR1 in cell protection against oxidative stress. Biochem. Biophys. Res. Commun., 436, 289–294 (2013). 147) Kim RH, Peters M, Jang Y, Shi W, Pintilie M, Fletcher GC, DeLuca C, Liepa J, Zhou L, Snow B, Binari RC, Manoukian AS, Bray MR, Liu FF, Tsao MS, Mak TW. DJ-1, a novel regulator of the tumor suppressor PTEN. Cancer Cell, 7, 263–273 (2005). 148) Kim YC, Kitaura H, Taira T, Iguchi-Ariga SMM, Ariga H. Oxidation of DJ-1-dependent cell transformation through direct binding of DJ-1 to PTEN. Int. J. Oncol., 35, 1331–1341 (2009). 149) Gao H, Yang W, Qi Z, Lu L, Duan C, Zhao C, Yang H. DJ-1 protects dopaminergic neurons against rotenone-induced apoptosis by enhancing ERK-dependent mitophagy. J. Mol. Biol., 423, 232–248 (2012). 150) Junn E, Taniguchi H, Jeong BS, Zhao X, Ichijo H, Mouradian MM. Interaction of DJ-1 with Daxx inhibits apoptosis signal-regulating kinase 1 activity and cell death. Proc. Natl. Acad. Sci. U.S.A., 102, 9691–9696 (2005). 151) Waak J, Weber SS, Görner K, Schall C, Ichijo H, Stehle T, Kahle PJ. Oxidizable residues mediating protein stability and cytoprotective interaction of DJ-1 with apoptosis signal-regulating kinase 1. J. Biol. Chem., 284, 14245–14257 (2009).

152) Sekito A, Koide-Yoshida S, Niki T, Taira T, Iguchi-Ariga SMM, Ariga H. DJ-1 interacts with HIPK1 and affects H2O2-induced cell death. Free Radic. Res., 40, 155–165 (2006). 153) Olzmann JA, Brown K, Wilkinson KD, Rees HD, Huai Q, Ke H, Levey AI, Li L, Chin LS. Familial Parkinson’s disease-associated L166P mutation disrupts DJ-1 protein folding and function. J. Biol. Chem., 279, 8506–8515 (2004). 154) Chen J, Li L, Chin LS. Parkinson’s disease protein DJ-1 converts from a zymogen to a protease by carboxyl-terminal cleavage. Hum. Mol. Genet., 19, 2395–2408 (2010). 155) Mitsugi H, Niki T, Takahashi-Niki K, Tanimura K, Yoshizawa-Kumagaye K, Tsunemi M, Iguchi-Ariga SMM, Ariga H. Identification of the recognition sequence and target proteins for DJ-1 protease. FEBS Lett., 587, 2493–2499 (2013). 156) Shendelman S, Jonason A, Martinat C, Leete T, Abeliovich A. DJ-1 is a redox-dependent molecular chaperone that inhibits alphasynuclein aggregate formation. PLoS Biol., 2, e362 (2004). 157) Zhou W, Zhu M, Wilson MA, Petsko GA, Fink AL. The oxidation state of DJ-1 regulates its chaperone activity toward alpha-synuclein. J. Mol. Biol., 356, 1036–1048 (2006). 158) Richarme G, Mihoub M, Dairou J, Bui LC, Leger T, Lamouri A. Parkinsonism-associated protein DJ-1/Park7 is a major protein deglycase that repairs methylglyoxal- and glyoxal-glycated cysteine, arginine, and lysine residues. J. Biol. Chem., 290, 1885–1897 (2015). 159) Aleyasin H, Rousseaux MW, Phillips M, Kim RH, Bland RJ, Callaghan S, Slack RS, During MJ, Mak TW, Park DS. The Parkinson’s disease gene DJ-1 is also a key regulator of stroke-induced damage. Proc. Natl. Acad. Sci. U.S.A., 104, 18748–18753 (2007). 160) Yanagisawa D, Kitamura Y, Inden M, Takata K, Taniguchi T, Morikawa S, Morita M, Inubushi T, Tooyama I, Taira T, Iguchi-Ariga SMM, Akaike A, Ariga H. DJ-1 protects against neurodegeneration caused by focal cerebral ischemia and reperfusion in rats. J. Cereb. Blood Flow Metab., 28, 563–578 (2008). 161) Wagenfeld A, Gromoll J, Cooper TG. Molecular cloning and expression of rat contraception associated protein 1 (CAP1), a protein putatively involved in fertilization. Biochem. Biophys. Res. Commun., 251, 545–549 (1998). 162) Okada M, Matsumoto K, Niki T, Taira T, Iguchi-Ariga SMM, Ariga H. DJ-1, a target protein for an endocrine disrupter, participates in the fertilization in mice. Biol. Pharm. Bull., 25, 853–856 (2002). 163) An CN, Jiang H, Wang Q, Yuan RP, Liu JM, Shi WL, Zhang ZY, Pu XP. Down-regulation of DJ-1 protein in the ejaculated spermatozoa from Chinese asthenozoospermia patients. Fertil. Steril., 96, 19–23.e2 (2011). 164) Malhotra D, Thimmulappa R, Navas-Acien A, Sandford A, Elliott M, Singh A, Chen L, Zhuang X, Hogg J, Pare P, Tuder RM, Biswal S. Decline in NRF2-regulated antioxidants in chronic obstructive pulmonary disease lungs due to loss of its positive regulator, DJ-1. Am. J. Respir. Crit. Care Med., 178, 592–604 (2008). 165) Jain D, Jain R, Eberhard D, Eglinger J, Bugliani M, Piemonti L, Marchetti P, Lammert E. Age- and diet-dependent requirement of DJ-1 for glucose homeostasis in mice with implications for human type 2 diabetes. J. Mol. Cell Biol., 4, 221–230 (2012). 166) Nishinaga H, Takahashi-Niki K, Taira T, Andreadis A, Iguchi-Ariga SMM, Ariga H. Expression profiles of genes in DJ-1-knockdown and L166P DJ-1 mutant cells. Neurosci. Lett., 390, 54–59 (2005). 167) Zhou W, Freed CR. DJ-1 up-regulates glutathione synthesis during oxidative stress and inhibits A53T alpha-synuclein toxicity. J. Biol. Chem., 280, 43150–43158 (2005). 168) Yamashita S, Mori A, Kimura E, Mita S, Maeda Y, Hirano T, Uchino M. DJ-1 forms complexes with mutant SOD1 and ameliorates its toxicity. J. Neurochem., 113, 860–870 (2010). 169) Wang Z, Liu J, Chen S, Wang Y, Cao L, Zhang Y, Kang W, Li H, Gui Y, Chen S, Ding J. DJ-1 modulates the expression of Cu/Znsuperoxide dismutase-1 through the Erk1/2-Elk1 pathway in neuroprotection. Ann. Neurol., 70, 591–599 (2011).

808

Biol. Pharm. Bull.

170) Girotto S, Cendron L, Bisaglia M, Tessari I, Mammi S, Zanotti G, Bubacco L. DJ-1 is a copper chaperone acting on SOD1 activation. J. Biol. Chem., 289, 10887–10899 (2014). 171) Takahashi-Niki K, Niki T, Taira T, Iguchi-Ariga SMM, Ariga H. Reduced anti-oxidative stress activities of DJ-1 mutants found in Parkinson’s disease patients. Biochem. Biophys. Res. Commun., 320, 389–397 (2004). 172) Kim RH, Smith PD, Aleyasin H, Hayley S, Mount MP, Pownall S, Wakeham A, You-Ten AJ, Kalia SK, Horne P, Westaway D, Lozano AM, Anisman H, Park DS, Mak TW. Hypersensitivity of DJ1-deficient mice to 1-methyl-4-phenyl-1,2,3,6-tetrahydropyrindine (MPTP) and oxidative stress. Proc. Natl. Acad. Sci. U.S.A., 102, 5215–5220 (2005). 173) Rizzu P, Hinkle DA, Zhukareva V, Bonifati V, Severijnen LA, Martinez D, Ravid R, Kamphorst W, Eberwine JH, Lee VM, Trojanowski JQ, Heutink P. DJ-1 colocalizes with tau inclusions: a link between Parkinsonism and dementia. Ann. Neurol., 55, 113–118 (2004). 174) Yanagida T, Takata K, Inden M, Kitamura Y, Taniguchi T, Yoshimoto K, Taira T, Ariga H. Distribution of DJ-1, Parkinson’s disease-related protein PARK7, and its alteration in 6-hydroxydopamine-treated hemiparkinsonian rat brain. J. Pharmacol. Sci., 102, 243–247 (2006). 175) Mullett SJ, Hamilton RL, Hinkle DA. DJ-1 immunoreactivity in human brain astrocytes is dependent on infarct presence and infarct age. Neuropathology, 29, 125–131 (2009). 176) Mullett SJ, Hinkle DA. DJ-1 knock-down in astrocytes impairs astrocyte-mediated neuroprotection against rotenone. Neurobiol. Dis., 33, 28–36 (2009). 177) Collins AR, Lyon CJ, Xia X, Liu JZ, Tangirala RK, Yin F, Boyadjian R, Bikineyeva A, Praticò D, Harrison DG, Hsueh WA. Ageaccelerated atherosclerosis correlates with failure to upregulate antioxidant genes. Circ. Res., 104, e42–e54 (2009). 178) Malaud E, Merle D, Piquer D, Molina L, Salvetat N, Rubrecht L, Dupaty E, Galea P, Cobo S, Blanc A, Saussine M, Marty-Ané C, Albat B, Meilhac O, Rieunier F, Pouzet A, Molina F, Laune D, Fareh J. Local carotid atherosclerotic plaque proteins for the identification of circulating biomarkers in coronary patients. Atherosclerosis, 233, 551–558 (2014). 179) Cuevas S, Zhang Y, Yang Y, Escano C, Asico L, Jones JE, Armando I, Jose PA. Role of renal DJ-1 in the pathogenesis of hypertension associated with increased reactive oxygen species production. Hypertension, 59, 446–452 (2012). 180) Billia F, Hauck L, Grothe D, Konecny F, Rao V, Kim RH, Mak TW. Parkinson-susceptibility gene DJ-1/PARK7 protects the murine heart from oxidative damage in vivo. Proc. Natl. Acad. Sci. U.S.A., 110, 6085–6090 (2013). 181) Klawitter J, Klawitter J, Agardi E, Corby K, Leibfritz D, Lowes BD, Christians U, Seres T. Association of DJ-1/PTEN/AKT- and ASK1/p38-mediated cell signalling with ischaemic cardiomyopathy. Cardiovasc. Res., 97, 66–76 (2013). 182) Liu M, Zhou B, Xia ZY, Zhao B, Lei SQ, Yang QJ, Xue R, Leng Y, Xu JJ, Xia Z. Hyperglycemia-induced inhibition of DJ-1 expression compromised the effectiveness of ischemic postconditioning cardioprotection in rats. Oxid. Med. Cell. Longev., 2013, 564902 (2013). 183) Yamane T, Murao S, Kozuka M, Shimizu M, Suzuki J, Kubo C, Yamaguchi A, Musashi M, Minegishi Y, Momose I, Matsushita M, Shirahata A, Furukawa N, Kobayashi R, Umezawa A, Sakamoto M, Moriya K, Saito M, Makita A, Ohkubo I, Ariga H. Serum DJ-1 level is positively associated with improvement of metabolic syndrome in Japanese women through lifestyle intervention. Nutr. Res., 34, 851–855 (2014). 184) Ishikawa S, Taira T, Niki T, Takahashi-Niki K, Maita C, Maita H,

185)

186)

187)

188)

189)

190) 191)

192)

193)

194)

195)

196)

197)

198)

Vol. 38, No. 6 (2015)

Ariga H, Iguchi-Ariga SMM. Oxidative status of DJ-1-dependent activation of dopamine synthesis through interaction of tyrosine hydroxylase and 4-dihydroxy-l-phenylalanine (l-DOPA) decarboxylase with DJ-1. J. Biol. Chem., 284, 28832–28844 (2009). Ishikawa S, Tanaka Y, Takahashi-Niki K, Niki T, Ariga H, IguchiAriga SMM. Stimulation of vesicular monoamine transporter 2 activity by DJ-1 in SH-SY5Y cells. Biochem. Biophys. Res. Commun., 421, 813–818 (2012). Shinbo Y, Niki T, Taira T, Ooe H, Takahashi-Niki K, Maita C, Seino C, Iguchi-Ariga SMM, Ariga H. Proper SUMO-1 conjugation is essential to DJ-1 to exert its full activities. Cell Death Differ., 13, 96–108 (2006). Hattori K, Naguro I, Runchel C, Ichijo H. The roles of ASK family proteins in stress responses and diseases. Cell Commun. Signal., 7, 9 (2009). Shiizaki S, Naguro I, Ichijo H. Activation mechanisms of ASK1 in response to various stresses and its significance in intracellular signaling. Adv. Biol. Regul., 53, 135–144 (2013). Zhong N, Xu J. Synergistic activation of the human MnSOD promoter by DJ-1 and PGC-1alpha: regulation by SUMOylation and oxidation. Hum. Mol. Genet., 17, 3357–3367 (2008). Pickrell AM, Youle RJ. The Roles of PINK1, Parkin, and mitochondrial fidelity in Parkinson’s disease. Neuron, 85, 257–273 (2015). Inden M, Taira T, Kitamura Y, Yanagida T, Tsuchiya D, Takata K, Yanagisawa D, Nishimura K, Taniguchi T, Kiso Y, Yoshimoto K, Agatsuma T, Koide-Yoshida S, Iguchi-Ariga SMM, Shimohama S, Ariga H. PARK7 DJ-1 protects against degeneration of nigral dopaminergic neurons in Parkinson’s disease rat model. Neurobiol. Dis., 24, 144–158 (2006). Miyazaki S, Yanagida T, Nunome K, Ishikawa S, Inden M, Kitamura Y, Nakagawa S, Taira T, Hirota K, Niwa M, Iguchi-Ariga SMM, Ariga H. DJ-1-binding compounds prevent oxidative stress-induced cell death and movement defect in Parkinson’s disease model rats. J. Neurochem., 105, 2418–2434 (2008). Qu W, Fan L, Kim Y-C, Ishikawa S, Iguchi-Ariga SMM, Pu X-P, Ariga H. Kaempferol derivatives prevent oxidative stress-induced cell death in a DJ-1-dependent manner. J. Pharmacol. Sci., 110, 191–200 (2009). Gao J-W, Yamane T, Maita H, Ishikawa S, Iguchi-Ariga SMM, Pu X-P, Ariga H. DJ-1-mediated protective effect of protocatechuic aldehyde against oxidative stress in SH-SY5Y cells. J. Pharmacol. Sci., 115, 36–44 (2011). Kitamura Y, Watanabe S, Taguchi M, Takagi K, Kawata T, Takahashi-Niki K, Yasui H, Maita H, Iguchi-Ariga SMM, Ariga H. Neuroprotective effect of a new DJ-1-binding compound against neurodegeneration in Parkinson’s disease and stroke model rats. Mol. Neurodegener., 6, 48 (2011). Inden M, Kitamura M, Takahashi K, Takata K, Ito N, Niwa R, Funayama R, Nishimura K, Taniguchi T, Honda T, Taira T, Ariga H. Protection against dopaminergic neurodegeneration in Parkinson’s disease-model animals by a modulator of the oxidized form of DJ-1, a wild-type of familial Parkinson’s disease-linked PARK7. J. Pharmacol. Sci., 117, 189–203 (2011). Yanagida T, Kitamura Y, Yamane K, Takahashi K, Takata K, Yanagisawa D, Yasui H, Taniguchi T, Taira T, Honda T, Ariga H. Protection against oxidative stress-induced neurodegeneration by a modulator for DJ-1, a wild-type of familial Parkinson’s diseaselinked PARK7. J. Pharmacol. Sci., 109, 463–468 (2009). Takahashi-Niki K, Inafune A, Michitani N, Hatakeyama Y, Suzuki K, Sasaki M, Kitamura Y, Niki T, Iguchi-Ariga SMM, Ariga H. DJ-1-dependent protective activity of DJ-1-binding compound No. 23 against neuronal cell death in MPTP-treated mouse model of Parkinson’s disease. J. Pharmacol. Sci., 127, 305–310 (2015).

Common mechanisms of onset of cancer and neurodegenerative diseases.

Onset of cancer and neurodegenerative disease occurs by abnormal cell growth and neuronal cell death, respectively, and the number of patients with bo...
1MB Sizes 2 Downloads 12 Views