Articles

  • KSBNS 2024

Article

Original Article

Exp Neurobiol 2019; 28(3): 414-424

Published online May 24, 2019

https://doi.org/10.5607/en.2019.28.3.414

© The Korean Society for Brain and Neural Sciences

Abnormal Mitochondria in a Non-human Primate Model of MPTP-induced Parkinson's Disease: Drp1 and CDK5/p25 Signaling

Junghyung Park1,†, Jincheol Seo1,2,†, Jinyoung Won1, Hyeon-Gu Yeo1,3, Yu-Jin Ahn1,3, Keonwoo Kim1,4, Yeung Bae Jin1, Bon-Sang Koo1, Kyung Seob Lim5, Kang-Jin Jeong1, Philyong Kang5, Hwal-Yong Lee1, Seung Ho Baek1, Chang-Yeop Jeon1, Jung-Joo Hong1, Jae-Won Huh1,3, Young-Hyun Kim1,3, Sang-Je Park1, Sun-Uk Kim3,5, Dong-Seok Lee2, Sang-Rae Lee1,3*, and Youngjeon Lee1,3*

1National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea.

2School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Korea.

3Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology, Daejeon 34113, Korea.

4Department of Physical Therapy, Graduate School of Inje University, Gimhae 50834, Korea.

5Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea.

Correspondence to: *To whom correspondence should be addressed.
Sang-Rae Lee, TEL: 82-43-240-6322, FAX: 82-43-240-6309
e-mail: srlee@kribb.re.kr
Youngjeon Lee, TEL: 82-43-240-6316, FAX: 82-43-240-6309
e-mail: neurosci@kribb.re.kr
These authors contributed equally to work.

Received: February 27, 2019; Revised: April 25, 2019; Accepted: May 14, 2019

Mitochondria continuously fuse and divide to maintain homeostasis. An impairment in the balance between the fusion and fission processes can trigger mitochondrial dysfunction. Accumulating evidence suggests that mitochondrial dysfunction is related to neurodegenerative diseases such as Parkinson's disease (PD), with excessive mitochondrial fission in dopaminergic neurons being one of the pathological mechanisms of PD. Here, we investigated the balance between mitochondrial fusion and fission in the substantia nigra of a non-human primate model of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD. We found that MPTP induced shorter and abnormally distributed mitochondria. This phenomenon was accompanied by the activation of dynamin-related protein 1 (Drp1), a mitochondrial fission protein, through increased phosphorylation at S616. Thereafter, we assessed for activation of the components of the cyclin-dependent kinase 5 (CDK5) and extracellular signal-regulated kinase (ERK) signaling cascades, which are known regulators of Drp1(S616) phosphorylation. MPTP induced an increase in p25 and p35, which are required for CDK5 activation. Together, these findings suggest that the phosphorylation of Drp1(S616) by CDK5 is involved in mitochondrial fission in the substantia nigra of a non-human primate model of MPTP-induced PD.


Keywords: Cyclin-dependent kinases, Mitochondria, Mitochondrial dynamics, Non-human primate, Parkinson disease

Parkinson's disease (PD) is the most common age-related neurodegenerative disease affecting motor control. Clinically, it is characterized by four cardinal signs: rigidity, bradykinesia, resting tremor, and postural instability. The motor symptoms are accompanied by dopaminergic neuron degeneration in the substantia nigra pars compacta, leading to a dopamine deficit in the striatum, including the caudate and putamen [1,2]. The causes of PD pathogenesis are complex, with various contributors, such as genetic susceptibility and environmental factors. Recently, accumulating evidence has suggested a link between PD pathogenesis and mitochondrial dysfunction [3,4].

Mitochondria are the main subcellular organelles responsible for production of adenosine triphosphate (ATP) and regulation of metabolite synthesis, intracellular calcium homeostasis, and programmed cell death. In particular, the central nervous system (CNS) has a high demand for mitochondrial ATP as an energy source to maintain ionic gradients across the axonal membrane, a process that is essential for neurotransmission [5,6]. Mitochondria are highly dynamic; they continuously undergo fission, which is regulated by Drp1 and Fis1, and fusion, which is regulated by Mfn1, Mfn2, and Opa1 [7,8,9]. The balance between mitochondrial fission and fusion significantly affects the role of mitochondria in the maintenance of cellular process [7,8,10]. Excessive mitochondrial fission triggers mitochondrial fragmentation and dysfunction, subsequently leading to a reduction in the mitochondrial membrane potential, depletion of ATP, accumulation of reactive oxygen species (ROS), and release of apoptotic factors [11,12]. In view of this, abnormal mitochondrial dynamics is also thought to be involved in various neurodegenerative diseases, including PD [13,14]. Indeed, a change in Drp1 activity has been implicated in various neurodegenerative disorders [15,16]. Drp1-dependent mitochondrial morphology and distribution are key factors in modulating mitochondrial homeostasis in dopaminergic neurons in models of PD [17,18]. Drp1 activity is controlled by post-translational modifications, including phosphorylation [19]. Specifically, phosphorylation of a serine residue, S616, results in increased Drp1 activity, reflecting variant pathological processes [20,21]. However, more information is needed on the precise relationship between abnormal mitochondrial dynamics and the causative factors of PD.

CDK5 is a proline-directed serine-threonine kinase that is mainly expressed in post-mitotic neurons [22,23]. CDK5 activity is mainly controlled by neuron-specific activators, p35 and p39, which are activated after being cleaved into p25 and p29, resulting in CDK5 hyperactivity [24,25]. CDK5 plays an important role in the regulation of CNS development and synaptic plasticity [26,27]. However, inappropriate activation of CDK5 plays an early role in the cell death cascade, even before the initiation of mitochondrial dysfunction, and CDK5 inhibition prevents mitochondrial damage and cell death in a model of PD [28,29,30]. Interestingly, CDK5 modulates mitochondrial morphology during neuronal apoptosis as an upstream signaling kinase [31,32]. Furthermore, CDK5-mediated phosphorylation of Drp1 is related to mitochondrial morphology control during neuronal injury [33]. However, the mechanisms via which CDK5 regulates mitochondrial fission by phosphorylation of Drp1 at S616 during dopaminergic neuronal loss are still not completely understood.

The neurotoxin, 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), can trigger parkinsonism in non-human primates, and has been used extensively in experimental models of PD [34,35,36]. However, it is difficult to develop macaque models of MPTP-induced chronic parkinsonism owing to symptomatic variation. To induce a stable non-human primate PD model, adjustments of MPTP administration at an individual-level are required according to the severity of behavioral symptoms [37]. Recently, we established and verified a primate model of chronic stable PD by repeated low-dose MPTP administration based on automatic quantification of individual global activity in cynomolgus monkeys (Macaca fascicularis) [38]. In our MPTP-treated monkeys, parkinsonian symptoms and decreased dopamine transporter activity persisted until 1 year. Dopaminergic neuronal cell death was confirmed by immunohistochemistry and western blotting [38]. Although the clinical features in human chronic PD patients can be observed in this model, further investigation is needed to support its use for chronic PD research and drug discovery. In the present study, we investigated pathological alterations and molecular mechanisms of mitochondrial dynamics in the substantia nigra of MPTP-treated cynomolgus monkeys at 1 year after the first MPTP administration.

Animals

All experimental animals were derived from our previous study [38]. Briefly, four female adult cynomolgus monkeys were obtained from the Zhaoqing Laboratory Animal Research Centre (Guangdong Province, China). They were maintained in individual indoor cages (60×80×80 cm) at the National Primate Research Center of the Korea Research Institute of Bioscience and Biotechnology (KRIBB) at a temperature of 24±2℃, a relative humidity of 50±5%, and under a 12-h light/12-h dark cycle. The monkeys were able to have visual contact and voice interaction with neighbors but no physical contact (to avoid aggression), as described previously [39,40]. The dimensions of the cages met that provided by the guidelines of the USA National Institutes of Health. The monkeys were fed commercial monkey chow (Harlan Teklad, Indianapolis, IN, USA) supplemented with various fruits and were given water ad libitum. They were also given various rubber and plastic toys and fruits as environmental enrichment. The attending veterinarian monitored the monkeys' health in accordance with the recommendations of the Weatherall report on the use of non-human primates in research [41]. They were also monitored through a once yearly administration of microbiological tests for B virus, simian retrovirus, simian immunodeficiency virus, simian virus 40, and simian T-cell lymphotropic virus. All procedures were approved by the KRIBB Institutional Animal Care and Use Committee (Approval No. KRIBB-AEC-16068). All animal experiments complied with the ARRIVE guidelines [42].

MPTP administration

MPTP (0.2 mg/kg; Sigma-Aldrich, St. Louis, MO, USA) was dissolved in saline to a final concentration of 2 mg/mL and intramuscularly injected into the left femoral region of the cynomolgus monkeys daily, from Monday to Friday each week, as described previously [38]. The total number of MPTP injections were commensurate with each individual animal's global activity intensity. The stop point thresholds for MPTP administration were indicated by a global activity intensity lower than 8% (arbitrary) of baseline intensity.

Tissue preparation

Four monkeys were transcardially perfused with 400 mL of 100 mM phosphate-buffered solution (PBS) under deep anesthesia induced by an intramuscular injection of ketamine (1 mg/kg) at 48 weeks following the first MPTP administration. Whole brains were removed from the skull, washed in cold PBS, and bilaterally separated. For immunohistochemical staining, the left hemispheres were post-fixed with 4% paraformaldehyde and incubated in 30% sucrose solution at 4℃.

Western blot analysis

The tissues were harvested from the substantia nigra of the monkey brains using punches on 4-mm-thick slices, snap-frozen, and stored at −80℃. Whole protein lysates of the substantia nigra were prepared using the PRO-PREP protein extraction solution (Intron Biotechnology, Seongnam, Korea). Equal amounts of proteins were separated by electrophoresis on 10~15% sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) gels and transferred onto nitrocellulose membranes (BD Biosciences, Franklin Lakes, NJ, USA). The membranes were blocked using incubation in blocking buffer (BD Biosciences) and primarily blotted with primary antibodies against anti-TH (MAB318; Merck Millipore, Darmstadt, Germany), anti-GFAP (AB5804), anti-β-actin (A5316; Sigma-Aldrich, St. Louis, MO, USA), anti-Iba-1 (ab108539) anti-Mfn1 (ab57602; Abcam, Cambridge, MA, USA), anti-Drp1 (#8570), anti-phospho(p)-Drp1 (#3455), anti-Mfn2 (#9482), anti-Opa1 (#67589), anti-CDK5 (#2506), anti-ERK (#9102), anti-p-ERK (#9101; Cell Signaling, Danvers, MA, USA), anti-Fis1 (PA1-41082), and anti-p35 (MA5-14834; Thermo Scientific, Waltham, MA, USA) antibodies at 4℃ overnight. The membranes were washed with 10 mM Tris-HCl (pH 7.5) containing 150 mM NaCl and 0.1% Tween-20 (TBST) and incubated with horseradish peroxidase-conjugated secondary antibodies (Cell Signaling) for 1 h at room temperature. After the removal of excess antibodies by washing with TBST, specific binding was detected using a chemiluminescence detection system (Thermo Scientific) according to the manufacturer's instructions.

Immunohistochemistry and mitochondrial imaging

The left hemispheres of the brains were sectioned in the coronal plane at 30 µm of thickness using a cryostat (Leica Biosystems, Wetzlar, Germany). For blocking, 30-µm free-floating tissue sections were incubated with 4% normal horse serum (S-2000; Vector Laboratories, Burlingame, CA, USA) in 0.3% Triton X-100 for 2 h at room temperature. For immunohistochemistry and immunofluorescent staining, the tissue sections were incubated with anti-TH (AB152; Merck Millipore), anti-GFAP (AB5804; Sigma-Aldrich), anti-Iba-1 (ab108539; Abcam), and anti-TOM20 (#42406; Cell Signaling) antibodies at 4℃ overnight. The appropriate secondary antibodies (Vector Laboratories and Thermo Scientific) were incubated for 2 h at room temperature to allow binding to the primary antibody. Immunohistochemistry staining was visualized using the ABC method (Vector Laboratories) with 3, 3′-diaminobenzidine as the peroxidase substrate. The tissue sections were observed using the Precipoint M8 digital microscope (PreciPoint, Freising, Germany). Fluorescent images were acquired using the LSM-710 confocal microscope (Carl Zeiss, Jena, Germany). Measurement of mitochondrial length was performed as described previously [43].

Statistical analysis

The data represent the mean and standard deviation (SD) from three independent experiments (n=3). Experimental differences were tested for statistical significance using two-way analysis of variance (ANOVA) using GraphPad Prism 5 software (San Diego, CA, USA). A p-value <0.05 was deemed to be statistically significant and is indicated on graphs by an asterisk; p-values <0.01 and <0.001 are indicated by two and three asterisks, respectively.

Loss of dopaminergic neurons in the basal ganglia region of monkeys with MPTP-induced PD

We previously developed a model of chronic PD in non-human primates using a novel strategy of MPTP administration that was based on global activity evaluation in individual cynomolgus monkeys [38]. In this model, we first confirmed damage of dopaminergic neurons in the basal ganglia region of the monkey brain by determining the protein level of tyrosine hydroxylase (TH), a marker of dopaminergic neurons, using immunoblotting. Our results showed that the protein level of TH was dramatically reduced in the substantia nigra than in the saline group (Fig. 1A, 1B, and 1C). We also investigated neuroinflammation, an important physiological alteration in PD, by determining the protein level of GFAP (a marker of astrocytes) and Iba-1 (a marker of microglia), as described in our earlier study [39]. Our results indicated that the protein level of GFAP in the substantia nigra was higher in the MPTP group than in the saline group, whereas there was no significant difference in Iba-1 between the two groups (Fig. 1D and 1E). Altogether, we demonstrated that our MPTP-induced PD model successfully reflected dopaminergic neuronal loss and neuroinflammation in the substantia nigra.

MPTP-induced increase of abnormal mitochondria in the substantia nigra

Abnormal mitochondrial dynamics significantly affect dopaminergic neuronal loss in patients with PD [3]. Therefore, we first observed dopaminergic mitochondrial morphology by immunohistochemistry for TOM20, a mitochondria outer membrane protein and a marker of mitochondria, co-stained with TH. Our observation indicated that the mitochondria of the dopaminergic neurons in the substantia nigra contained a high number of interconnected structures and were widely distributed throughout the whole cell, including the perinuclear and synaptic regions in the saline group. On the other hand, the number of mitochondria in the MPTP group was markedly reduced; moreover, mitochondria were distributed around the nuclear region in a punctate manner (Fig. 2A). The average length of mitochondria in the MPTP group was significantly shorter than that in the saline group (Fig. 2B). Moreover, the neurite structure in the saline-injected group was more developed than that in the MPTP-injected group. Therefore, we determined the protein level of synaptophysin, a pre-synapse marker, using immunoblotting to verify its possible decrease induced by MPTP in the substantia nigra. We noted that synaptophysin levels was decreased after MPTP than after saline injection (Fig. 2C). Our findings indicated that MPTP induced abnormal mitochondrial morphology and distribution in the substantia nigra of the monkey brain.

MPTP-induced phosphorylation-mediated Drp1 activation in the substantia nigra

Drp1-mediated control of mitochondrial morphology and distribution is crucial for modulating dopaminergic neurons in models of PD [17]. Thus, we assessed the mitochondrial fission and fusion proteins, including the phosphorylation level of Drp1(S616), using immunoblotting. Our results showed that phosphorylation of Drp1(S616) was markedly increased by MPTP injection, with no change in the expression level of the mitochondrial fission proteins, Drp1 and Fis1 (Fig. 3A). The expression of mitochondrial fusion proteins, Mfn1, Mfn2, and Opa1 were not significantly changed by MPTP (Fig. 3B). Although the expression of mitochondrial fusion proteins was independent of MPTP, there were differences among individuals. Taken together, our data suggested that abnormal mitochondrial phenotype in the substantia nigra of MPTP-injected monkeys was accompanied by an increase in Drp1(S616) phosphorylation.

MPTP-induced activation of the CDK5/p35 signaling pathway in the substantia nigra

Drp1-mediated excessive mitochondrial fission was mainly induced by increased Drp1 phosphorylation. Drp1 can be phosphorylated by various kinases, such as CDK5 and ERK [44,45]. Therefore, we confirmed the activity of kinases upstream of Drp1(S616) phosphorylation using immunoblotting. Our data showed that the protein level of CDK5 was unchanged, but those of p35 and p25, the neuron-specific activators of CDK5, were increased in the substantia nigra of the MPTP group (Fig. 4A). In contrast, other upstream kinases of Drp1(S616) phosphorylation, ERK, were not different between the two groups (Fig. 4B). ERK phosphorylation level was lower in the MPTP group than in the saline group. These results suggested that Drp1-mediated abnormal mitochondrial morphology involved CDK5 activation via elevated p35 and p25 levels.

Mitochondria are important organelles in PD, and dopaminergic neurons appear to be particularly sensitive to mitochondrial dysfunction. One of the possible reasons for such vulnerability is the lower basal level of mitochondria in dopaminergic neurons than in other midbrain neurons [46,47]. Therefore, emphasis has been placed on maintaining mitochondrial function in dopaminergic neurons. The homeostasis of mitochondrial dynamics is not only associated with the maintenance of mitochondrial function, but also with an imbalance between mitochondrial fission and fusion, which can trigger dopaminergic neuronal loss [14,15,48]. However, little is known regarding the molecular mechanisms underlying the mitochondrial dynamics in PD.

MPTP has been commonly used to induce stable PD in non-human primates, with bilateral clinical features closely resembling idiopathic PD [35]. Therefore, we investigated the mechanisms of mitochondria dynamics in a non-human primate model of MPTP-induced PD. First, we confirmed the loss of dopaminergic neurons and an increase of neuroinflammation in the basal ganglia region of cynomolgus monkeys injected with MPTP using our own strategy based on global activity evaluation [38]. In this model of PD, mitochondrial fission as well as unusual mitochondrial distribution were observed in the MPTP group. In MPTP-injected monkeys, mitochondria were located closer to the nucleus than was observed in the saline group. Mitochondrial distribution within the regions of high energy demand is critical for various functions, and impaired mitochondrial transport and distribution have been linked to abnormal neuronal synaptic functions as in PD [6,49,50,51,52,53]. In addition, we found a decrease in the protein level of synaptophysin, a marker of synaptic number and function. Accordingly, we showed that mitochondrial distribution and synaptic function were disrupted in our experimental model. These findings were consistent with those of earlier studies, which showed loss of dopaminergic synapses followed by substantia nigra cell bodies in mice treated with MPTP [54,55].

Recent evidence has suggested that the balance between mitochondrial fission and fusion is correlated with axonal mitochondrial transport and distribution [8,14,48,56]. Although the mitochondrial fission process is essential for axonal mitochondrial transport and the degradation of damaged mitochondria [57,58], excessive mitochondrial fission is an early event of synaptic degradation [59]. Furthermore, Drp1 activity has been closely associated with the fate of dopaminergic neurons [17,18], and inhibition of Drp1 activation attenuates disrupted synaptic function in diverse neurodegenerative models, including PD [16,60,61]. Our results indicated that excessive mitochondrial fission in MPTP-induced PD in monkeys was accompanied by phosphorylation of Drp1(S616), which triggers Drp1 activation. These results are consistent with other published results that an increase in Drp1(S616) phosphorylation is associated with various neurodegenerative diseases involving dopaminergic neurons [18,62,63]. However, the precise molecular mechanisms of excessive mitochondria fission mediated by Drp1 phosphorylation in experimental PD models are still unclear.

CDK5 has been identified as a regulator of mitochondrial fragmentation during neuronal apoptosis by modulating Drp1 phosphorylation, and its suppression attenuates excessive mitochondrial fission leading to apoptosis [31,32,45]. However, the precise mechanism underlying the relationship between mitochondrial morphology and activated CDK5 in PD is not fully understood. Our findings indicated that Drp1(S616) phosphorylation was induced by CDK5 activation, which was accompanied by an increased level of p35 and p25 in the substantia nigra of MPTP-injected monkeys. On the other hand, another kinase of Drp1, ERK, remained unchanged after MPTP injection. Our results indicated that MPTP-induced CDK5 activation regulates mitochondrial fragmentation by modulating the phosphorylation of Drp1(S616). In PD, CDK5 hyperactivation is a classical pathology that is associated with loss of dopaminergic neurons in the substantia nigra [64]. Inhibition of CDK5 hyperactivation provides a neuroprotective effect in experimental PD models [65,66] . Furthermore, hyperactivation of CDK5 is involved in pre-synaptic loss, and ultimately neurodegeneration, by regulating neuronal actin cytoskeleton remodeling [67]. Therefore, our model of MPTP-induced PD indicated that CDK5-mediated increase of Drp1 phosphorylation at the S616 residue may trigger mitochondrial fission, ultimately inducing dopaminergic neuronal loss in the substantia nigra.

Human PD symptoms were observed in our non-human primate model of MPTP-induced PD. However, the degree of physical response to MPTP varies according to each individual monkey. Therefore, we developed a new strategy for MPTP-induced chronic PD, with consistent symptoms [38]. In this chronic PD model, we evaluated the molecular pathology more precisely, focusing on altered mitochondrial morphology, which is a marker of various genetic and pharmacological mechanisms of PD [68]. Thus, our model showed that CDK5-mediated increase of Drp1(S616) phosphorylation triggers mitochondrial fission, and ultimately induces dopaminergic neuronal loss in the substantia nigra. Therefore, inhibition of CDK5-relative signaling and excessive mitochondrial fission may provide therapeutic strategies. Altogether, our MPTP-mediated non-human primate PD model reflects PD pathology with both behavioral symptoms and molecular mechanisms. Therefore, our findings could contribute to the development of therapeutic strategies.

Fig. 1. Effect of MPTP on dopaminergic neuronal loss and neuroinflammation in the substantia nigra of cynomolgus monkeys. (A) TH protein expression level in the substantia nigra of saline- or MPTP-injected cynomolgus monkeys was determined by using western blotting. (B) Immunohistochemistry staining of TH-positive neurons in the cynomolgus monkey brain injected with saline or MPTP was performed using anti-TH antibody. The bottom panels show magnified images of the substantia nigra pars compacta (SNpc) region indicated by the black squares in the top panels; scale bars=10 mm. (C) Fluorescent imaging results of TH proteins were validated using anti-TH antibody in the substantia nigra of saline- or MPTP-injected cynomolgus monkeys; scale bars=100 µm. (D) GFAP and Iba-1 protein expression in the substantia nigra of saline- or MPTP-injected cynomolgus monkeys was confirmed using western blotting. (E) Expression of GFPA and Iba-1 proteins were identified in the SNpc region of saline- or MPTP-injected cynomolgus monkeys using immunohistochemistry; scale bars=2 µm. C1 and C2 indicate the saline-injected group, and C3 and C4 indicate the MPTP-injected group. The data are presented as mean values±SD (n=2). ***denotes p<0.001.
Fig. 2. Effect of MPTP on mitochondrial morphology and synaptic function in the substantia nigra. (A) Mitochondrial morphology in the substantia nigra of saline- or MPTP-injected cynomolgus monkeys was observed using immunofluorescent staining with anti-TOM20 and anti-TH antibodies. The right end panels show magnified images of the regions indicated by white squares in the left end panels; scale bars=5 µm. (B) The graph shows the average mitochondrial length in the substantia nigra of saline- and MPTP-injected cynomolgus monkeys. (C) The expression of synaptophysin, a pre-synaptic marker, was determined using western blotting with anti-synaptophysin antibody. The data are presented as mean values±SD (n=2). *denotes p<0.05.
Fig. 3. Expression level of proteins involved in mitochondrial dynamics. (A) The levels of the mitochondrial fission proteins, p-Drp1(S616), Drp1, and Fis1, and (B) the mitochondrial fusion proteins, Mfn1, Mfn2, and Opa1, in the substantia nigra of saline- or MPTP-injected cynomolgus monkeys were identified using western blot analysis. Drp1 was used as the loading control for p-Drp1(S616). The data are presented as mean values±SD (n=2). **denotes p<0.01.
Fig. 4. The CDK5/p25 signaling pathway in the substantia nigra of MPTP-injected cynomolgus monkeys. (A) The protein levels of CDK5, p35, and p25, and (B) p-ERK in the substantia nigra of saline- and MPTP-injected cynomolgus monkeys were determined using western blotting. ERK was used as the loading control for p-ERK. The data are presented as mean values±SD (n=2). *denotes p<0.05, **denotes p<0.01, and ***denotes p<0.001.
  1. Kalia LV, Lang AE. Parkinson's disease. Lancet 2015;386:896-912.
    Pubmed
  2. Poewe W, Seppi K, Tanner CM, Halliday GM, Brundin P, Volkmann J, Schrag AE, Lang AE. Parkinson disease. Nat Rev Dis Primers 2017;3:17013.
    Pubmed
  3. Cardoso SM. The mitochondrial cascade hypothesis for Parkinson's disease. Curr Pharm Des 2011;17:3390-3397.
    Pubmed
  4. Exner N, Lutz AK, Haass C, Winklhofer KF. Mitochondrial dysfunction in Parkinson's disease: molecular mechanisms and pathophysiological consequences. EMBO J 2012;31:3038-3062.
    Pubmed
  5. Du H, Guo L, Yan SS. Synaptic mitochondrial pathology in Alzheimer's disease. Antioxid Redox Signal 2012;16:1467-1475.
    Pubmed
  6. Yang Y, Lu B. Mitochondrial morphogenesis, distribution, and Parkinson disease: insights from PINK1. J Neuropathol Exp Neurol 2009;68:953-963.
    Pubmed
  7. Knott AB, Perkins G, Schwarzenbacher R, Bossy-Wetzel E. Mitochondrial fragmentation in neurodegeneration. Nat Rev Neurosci 2008;9:505-518.
    Pubmed
  8. Westermann B. Mitochondrial fusion and fission in cell life and death. Nat Rev Mol Cell Biol 2010;11:872-884.
    Pubmed
  9. Cho DH, Nakamura T, Lipton SA. Mitochondrial dynamics in cell death and neurodegeneration. Cell Mol Life Sci 2010;67:3435-3447.
    Pubmed
  10. Detmer SA, Chan DC. Functions and dysfunctions of mitochondrial dynamics. Nat Rev Mol Cell Biol 2007;8:870-879.
    Pubmed
  11. Park J, Choi H, Min JS, Kim B, Lee SR, Yun JW, Choi MS, Chang KT, Lee DS. Loss of mitofusin 2 links beta-amyloid-mediated mitochondrial fragmentation and Cdk5-induced oxidative stress in neuron cells. J Neurochem 2015;132:687-702.
    Pubmed
  12. Ruiz A, Alberdi E, Matute C. Mitochondrial Division Inhibitor 1 (mdivi-1) protects neurons against excitotoxicity through the modulation of mitochondrial function and intracellular Ca2+ signaling. Front Mol Neurosci 2018;11:3.
    Pubmed
  13. Poole AC, Thomas RE, Andrews LA, McBride HM, Whitworth AJ, Pallanck LJ. The PINK1/Parkin pathway regulates mitochondrial morphology. Proc Natl Acad Sci U S A 2008;105:1638-1643.
    Pubmed
  14. Berthet A, Margolis EB, Zhang J, Hsieh I, Zhang J, Hnasko TS, Ahmad J, Edwards RH, Sesaki H, Huang EJ, Nakamura K. Loss of mitochondrial fission depletes axonal mitochondria in midbrain dopamine neurons. J Neurosci 2014;34:14304-14317.
    Pubmed
  15. Filichia E, Hoffer B, Qi X, Luo Y. Inhibition of Drp1 mitochondrial translocation provides neural protection in dopaminergic system in a Parkinson's disease model induced by MPTP. Sci Rep 2016;6:32656.
    Pubmed
  16. Rappold PM, Cui M, Grima JC, Fan RZ, de Mesy-Bentley KL, Chen L, Zhuang X, Bowers WJ, Tieu K. Drp1 inhibition attenuates neurotoxicity and dopamine release deficits in vivo. Nat Commun 2014;5:5244.
    Pubmed
  17. Galindo MF, Solesio ME, Atienzar-Aroca S, Zamora MJ, Jordán Bueso J. Mitochondrial dynamics and mitophagy in the 6-hydroxydopamine preclinical model of Parkinson's disease. Parkinsons Dis 2012;2012:131058.
    Pubmed
  18. Zhang Z, Liu L, Jiang X, Zhai S, Xing D. The essential role of Drp1 and Its regulation by S-nitrosylation of parkin in dopaminergic neurodegeneration: implications for Parkinson's disease. Antioxid Redox Signal 2016;25:609-622.
    Pubmed
  19. Elgass K, Pakay J, Ryan MT, Palmer CS. Recent advances into the understanding of mitochondrial fission. Biochim Biophys Acta 2013;1833:150-161.
    Pubmed
  20. Prieto J, León M, Ponsoda X, Sendra R, Bort R, Ferrer-Lorente R, Raya A, López-García C, Torres J. Early ERK1/2 activation promotes DRP1-dependent mitochondrial fission necessary for cell reprogramming. Nat Commun 2016;7:11124.
    Pubmed
  21. Bradshaw TY, Romano LE, Duncan EJ, Nethisinghe S, Abeti R, Michael GJ, Giunti P, Vermeer S, Chapple JP. A reduction in Drp1-mediated fission compromises mitochondrial health in autosomal recessive spastic ataxia of Charlevoix Saguenay. Hum Mol Genet 2016;25:3232-3244.
    Pubmed
  22. Dhavan R, Tsai LH. A decade of CDK5. Nat Rev Mol Cell Biol 2001;2:749-759.
    Pubmed
  23. Cheung ZH, Ip NY. The roles of cyclin-dependent kinase 5 in dendrite and synapse development. Biotechnol J 2007;2:949-957.
    Pubmed
  24. Tsai LH, Delalle I, Caviness VS, Chae T, Harlow E. p35 is a neural-specific regulatory subunit of cyclin-dependent kinase 5. Nature 1994;371:419-423.
    Pubmed
  25. Tang D, Yeung J, Lee KY, Matsushita M, Matsui H, Tomizawa K, Hatase O, Wang JH. An isoform of the neuronal cyclin-dependent kinase 5 (Cdk5) activator. J Biol Chem 1995;270:26897-26903.
    Pubmed
  26. Lew J, Huang QQ, Qi Z, Winkfein RJ, Aebersold R, Hunt T, Wang JH. A brain-specific activator of cyclin-dependent kinase 5. Nature 1994;371:423-426.
    Pubmed
  27. van den Heuvel S, Harlow E. Distinct roles for cyclin-dependent kinases in cell cycle control. Science 1993;262:2050-2054.
    Pubmed
  28. Sun KH, de Pablo Y, Vincent F, Shah K. Deregulated Cdk5 promotes oxidative stress and mitochondrial dysfunction. J Neurochem 2008;107:265-278.
    Pubmed
  29. Czapski GA, Gąssowska M, Wilkaniec A, Cieślik M, Adamczyk A. Extracellular alpha-synuclein induces calpain-dependent overactivation of cyclin-dependent kinase 5 in vitro. FEBS Lett 2013;587:3135-3141.
    Pubmed
  30. Binukumar BK, Shukla V, Amin ND, Grant P, Bhaskar M, Skuntz S, Steiner J, Pant HC. Peptide TFP5/TP5 derived from Cdk5 activator P35 provides neuroprotection in the MPTP model of Parkinson's disease. Mol Biol Cell 2015;26:4478-4491.
    Pubmed
  31. Meuer K, Suppanz IE, Lingor P, Planchamp V, Göricke B, Fichtner L, Braus GH, Dietz GP, Jakobs S, Bähr M, Weishaupt JH. Cyclin-dependent kinase 5 is an upstream regulator of mitochondrial fission during neuronal apoptosis. Cell Death Differ 2007;14:651-661.
    Pubmed
  32. Cherubini M, Puigdellívol M, Alberch J, Ginés S. Cdk5-mediated mitochondrial fission: a key player in dopaminergic toxicity in Huntington's disease. Biochim Biophys Acta 2015;1852:2145-2160.
    Pubmed
  33. Jahani-Asl A, Huang E, Irrcher I, Rashidian J, Ishihara N, Lagace DC, Slack RS, Park DS. CDK5 phosphorylates DRP1 and drives mitochondrial defects in NMDA-induced neuronal death. Hum Mol Genet 2015;24:4573-4583.
    Pubmed
  34. Gubellini P, Kachidian P. Animal models of Parkinson's disease: an updated overview. Rev Neurol (Paris) 2015;171:750-761.
    Pubmed
  35. Blesa J, Trigo-Damas I, Del Rey NL, Obeso JA. The use of nonhuman primate models to understand processes in Parkinson's disease. J Neural Transm (Vienna) 2018;125:325-333.
    Pubmed
  36. Jeong HS, Lee SR, Kim JE, Lyoo IK, Yoon S, Namgung E, Chang KT, Kim BS, Yang S, Im JJ, Jeon S, Kang I, Ma J, Chung YA, Lim SM. Brain structural changes in cynomolgus monkeys administered with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine: a longitudinal voxel-based morphometry and diffusion tensor imaging study. PLoS One 2018;13:e0189804.
    Pubmed
  37. Potts LF, Wu H, Singh A, Marcilla I, Luquin MR, Papa SM. Modeling Parkinson's disease in monkeys for translational studies, a critical analysis. Exp Neurol 2014;256:133-143.
    Pubmed
  38. Seo J, Lee Y, Kim BS, Park J, Yang S, Yoon HJ, Yoo J, Park HS, Hong JJ, Koo BS, Baek SH, Jeon CY, Huh JW, Kim YH, Park SJ, Won J, Ahn YJ, Kim K, Jeong KJ, Kang P, Lee DS, Lim SM, Jin YB, Lee SR. A non-human primate model for stable chronic Parkinson's disease induced by MPTP administration based on individual behavioral quantification. J Neurosci Methods 2019;311:277-287.
    Pubmed
  39. Yeo HG, Lee Y, Jeon CY, Jeong KJ, Jin YB, Kang P, Kim SU, Kim JS, Huh JW, Kim YH, Sim BW, Song BS, Park YH, Hong Y, Lee SR, Chang KT. Characterization of cerebral damage in a monkey model of Alzheimer's disease induced by intracerebroventricular injection of streptozotocin. J Alzheimers Dis 2015;46:989-1005.
    Pubmed
  40. Lee Y, Kim YH, Park SJ, Huh JW, Kim SH, Kim SU, Kim JS, Jeong KJ, Lee KM, Hong Y, Lee SR, Chang KT. Insulin/IGF signaling-related gene expression in the brain of a sporadic Alzheimer's disease monkey model induced by intracerebroventricular injection of streptozotocin. J Alzheimers Dis 2014;38:251-267.
    Pubmed
  41. Weatherall D. The use of non-human primates in research. London: Academy of Medical Sciences, 2006.
  42. Kilkenny C, Browne WJ, Cuthill IC, Emerson M, Altman DG. Improving bioscience research reporting: the ARRIVE guidelines for reporting animal research. PLoS Biol 2010;8:e1000412.
    Pubmed
  43. Park J, Choi H, Min JS, Park SJ, Kim JH, Park HJ, Kim B, Chae JI, Yim M, Lee DS. Mitochondrial dynamics modulate the expression of pro-inflammatory mediators in microglial cells. J Neurochem 2013;127:221-232.
    Pubmed
  44. Yu T, Jhun BS, Yoon Y. High-glucose stimulation increases reactive oxygen species production through the calcium and mitogen-activated protein kinase-mediated activation of mitochondrial fission. Antioxid Redox Signal 2011;14:425-437.
    Pubmed
  45. Guo MY, Shang L, Hu YY, Jiang LP, Wan YY, Zhou QQ, Zhang K, Liao HF, Yi JL, Han XJ. The role of Cdk5-mediated Drp1 phosphorylation in Aβ1-42 induced mitochondrial fission and neuronal apoptosis. J Cell Biochem 2018;119:4815-4825.
    Pubmed
  46. Liang CL, Wang TT, Luby-Phelps K, German DC. Mitochondria mass is low in mouse substantia nigra dopamine neurons: implications for Parkinson's disease. Exp Neurol 2007;203:370-380.
    Pubmed
  47. Pickrell AM, Pinto M, Hida A, Moraes CT. Striatal dysfunctions associated with mitochondrial DNA damage in dopaminergic neurons in a mouse model of Parkinson's disease. J Neurosci 2011;31:17649-17658.
    Pubmed
  48. Pham AH, Meng S, Chu QN, Chan DC. Loss of Mfn2 results in progressive, retrograde degeneration of dopaminergic neurons in the nigrostriatal circuit. Hum Mol Genet 2012;21:4817-4826.
    Pubmed
  49. Obashi K, Okabe S. Regulation of mitochondrial dynamics and distribution by synapse position and neuronal activity in the axon. Eur J Neurosci 2013;38:2350-2363.
    Pubmed
  50. Chen H, Chan DC. Mitochondrial dynamics--fusion, fission, movement, and mitophagy--in neurodegenerative diseases. Hum Mol Genet 2009;18:R169-R176.
    Pubmed
  51. López-Doménech G, Higgs NF, Vaccaro V, Roš H, Arancibia-Cárcamo IL, MacAskill AF, Kittler JT. Loss of dendritic complexity precedes neurodegeneration in a mouse model with disrupted mitochondrial distribution in mature dendrites. Cell Reports 2016;17:317-327.
    Pubmed
  52. Hsieh CH, Shaltouki A, Gonzalez AE, Bettencourt da Cruz A, Burbulla LF, St Lawrence E, Schüle B, Krainc D, Palmer TD, Wang X. Functional impairment in miro degradation and mitophagy is a shared feature in familial and sporadic Parkinson's disease. Cell Stem Cell 2016;19:709-724.
    Pubmed
  53. Prots I, Grosch J, Brazdis RM, Simmnacher K, Veber V, Havlicek S, Hannappel C, Krach F, Krumbiegel M, Schütz O, Reis A, Wrasidlo W, Galasko DR, Groemer TW, Masliah E, Schlötzer-Schrehardt U, Xiang W, Winkler J, Winner B. α-Synuclein oligomers induce early axonal dysfunction in human iPSC-based models of synucleinopathies. Proc Natl Acad Sci U S A 2018;115:7813-7818.
    Pubmed
  54. Li Y, Liu W, Li L, Hölscher C. Neuroprotective effects of a GIP analogue in the MPTP Parkinson's disease mouse model. Neuropharmacology 2016;101:255-263.
    Pubmed
  55. Reeve AK, Grady JP, Cosgrave EM, Bennison E, Chen C, Hepplewhite PD, Morris CM. Mitochondrial dysfunction within the synapses of substantia nigra neurons in Parkinson's disease. NPJ Parkinsons Dis 2018;4:9.
    Pubmed
  56. Misko A, Jiang S, Wegorzewska I, Milbrandt J, Baloh RH. Mitofusin 2 is necessary for transport of axonal mitochondria and interacts with the Miro/Milton complex. J Neurosci 2010;30:4232-4240.
    Pubmed
  57. Trevisan T, Pendin D, Montagna A, Bova S, Ghelli AM, Daga A. Manipulation of mitochondria dynamics reveals separate roles for form and function in mitochondria distribution. Cell Reports 2018;23:1742-1753.
    Pubmed
  58. Pozo Devoto VM, Dimopoulos N, Alloatti M, Pardi MB, Saez TM, Otero MG, Cromberg LE, Marín-Burgin A, Scassa ME, Stokin GB, Schinder AF, Sevlever G, Falzone TL. αSynuclein control of mitochondrial homeostasis in human-derived neurons is disrupted by mutations associated with Parkinson's disease. Sci Rep 2017;7:5042.
    Pubmed
  59. Verstreken P, Ly CV, Venken KJ, Koh TW, Zhou Y, Bellen HJ. Synaptic mitochondria are critical for mobilization of reserve pool vesicles at Drosophila neuromuscular junctions. Neuron 2005;47:365-378.
    Pubmed
  60. Shirendeb UP, Calkins MJ, Manczak M, Anekonda V, Dufour B, McBride JL, Mao P, Reddy PH. Mutant huntingtin's interaction with mitochondrial protein Drp1 impairs mitochondrial biogenesis and causes defective axonal transport and synaptic degeneration in Huntington's disease. Hum Mol Genet 2012;21:406-420.
    Pubmed
  61. Calkins MJ, Manczak M, Mao P, Shirendeb U, Reddy PH. Impaired mitochondrial biogenesis, defective axonal transport of mitochondria, abnormal mitochondrial dynamics and synaptic degeneration in a mouse model of Alzheimer's disease. Hum Mol Genet 2011;20:4515-4529.
    Pubmed
  62. Roe AJ, Qi X. Drp1 phosphorylation by MAPK1 causes mitochondrial dysfunction in cell culture model of Huntington's disease. Biochem Biophys Res Commun 2018;496:706-711.
    Pubmed
  63. Bossy B, Petrilli A, Klinglmayr E, Chen J, Lütz-Meindl U, Knott AB, Masliah E, Schwarzenbacher R, Bossy-Wetzel E. S-Nitrosylation of DRP1 does not affect enzymatic activity and is not specific to Alzheimer's disease. J Alzheimers Dis 2010;20:S513-S526.
    Pubmed
  64. Alvira D, Ferrer I, Gutierrez-Cuesta J, Garcia-Castro B, Pallàs M, Camins A. Activation of the calpain/cdk5/p25 pathway in the girus cinguli in Parkinson's disease. Parkinsonism Relat Disord 2008;14:309-313.
    Pubmed
  65. Qu D, Rashidian J, Mount MP, Aleyasin H, Parsanejad M, Lira A, Haque E, Zhang Y, Callaghan S, Daigle M, Rousseaux MW, Slack RS, Albert PR, Vincent I, Woulfe JM, Park DS. Role of Cdk5-mediated phosphorylation of Prx2 in MPTP toxicity and Parkinson's disease. Neuron 2007;55:37-52.
    Pubmed
  66. Binukumar BK, Pant HC. TFP5/TP5 peptide provides neuroprotection in the MPTP model of Parkinson's disease. Neural Regen Res 2016;11:698-701.
    Pubmed
  67. Shah K, Rossie S. Tale of the good and the bad Cdk5: remodeling of the actin cytoskeleton in the brain. Mol Neurobiol 2018;55:3426-3438.
    Pubmed
  68. Haddad D, Nakamura K. Understanding the susceptibility of dopamine neurons to mitochondrial stressors in Parkinson's disease. FEBS Lett 2015;589:3702-3713.
    Pubmed