+ All Categories
Home > Documents > Post-treatment With an Ultra-low Dose Of

Post-treatment With an Ultra-low Dose Of

Date post: 06-Nov-2015
Category:
Upload: lauriejerie
View: 6 times
Download: 1 times
Share this document with a friend
Description:
Post-treatment with an ultra-low dose ofNADPH oxidase inhibitor diphenyleneiodoniumattenuates disease progression in multipleParkinson’s disease models
Popular Tags:
16
Post-treatment with an ultra-low dose of NADPH oxidase inhibitor diphenyleneiodonium attenuates disease progression in multiple Parkinson’s disease models Qingshan Wang, 1 Li Qian, 1 Shih-Heng Chen, 1 Chun-Hsien Chu, 1 Belinda Wilson, 1 Esteban Oyarzabal, 1 Syed Ali, 2 Bonnie Robinson, 2 Deepa Rao 3 and Jau-Shyong Hong 1 Nicotinamide adenine dinucleotide phosphate oxidase, a key superoxide-producing enzyme, plays a critical role in microglia- mediated chronic neuroinflammation and subsequent progressive dopaminergic neurodegeneration in Parkinson’s disease. Although nicotinamide adenine dinucleotide phosphate oxidase-targeting anti-inflammatory therapy for Parkinson’s disease has been proposed, its application in translational research remains limited. The aim of this study was to obtain preclinical evidence supporting this therapeutic strategy by testing the efficacy of an ultra-low dose of the nicotinamide adenine dinucleotide phosphate oxidase inhibitor diphenyleneiodonium in both endotoxin (lipopolysaccharide)- and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine- treated mice using post-treatment regimens. Our data revealed that post-treatment with diphenyleneiodonium significantly attenuated progressive dopaminergic degeneration and improved rotarod activity. Remarkably, post-treatment with diphenyleneio- donium 10 months after lipopolysaccharide injection when mice had 30% loss of nigral dopaminergic neurons, showed high efficacy in protecting the remaining neuronal population and restoring motor function. Diphenyleneiodonium-elicited neuroprotec- tion was associated with the inhibition of microglial activation, a reduction in the expression of proinflammatory factors and an attenuation of a-synuclein aggregation. A pathophysiological evaluation of diphenyleneiodonium-treated mice, including assess- ment of body weight, organs health, and neuronal counts, revealed no overt signs of toxicity. In summary, infusion of ultra-low dose diphenyleneiodonium potently reduced microglia-mediated chronic neuroinflammation by selectively inhibiting nicotinamide adenine dinucleotide phosphate oxidase and halted the progression of neurodegeneration in mouse models of Parkinson’s disease. The robust neuroprotective effects and lack of apparent toxic side effects suggest that diphenyleneiodonium at ultra-low dose may be a promising candidate for future clinical trials in Parkinson’s disease patients. 1 Neuropharmacology Section, Laboratory of Neurobiology, National Institute of Environmental Health Sciences, Research Triangle Park, NC 27709, USA 2 Neurochemistry Laboratory, Division of Neurotoxicology, National Centre for Toxicological Research/USFDA, Jefferson, AR 72079, USA 3 National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27709, USA Correspondence to: Qingshan Wang, National Institute of Environmental Health Sciences, 111 T.W. Alexander Dr., Research Triangle Park, North Carolina, 27709, USA E-mail: [email protected] Correspondence may also be addressed to: Jau-Shyong Hong. E-mail: [email protected]. doi:10.1093/brain/awv034 BRAIN 2015: 138; 1247–1262 | 1247 Received September 12, 2014. Revised November 25, 2014. Accepted December 16, 2014. Advance Access publication February 25, 2015 Published by Oxford University Press on behalf of the Guarantors of Brain 2015. This work is written by US Government employees and is in the public domain in the US by guest on June 16, 2015 Downloaded from
Transcript
  • Post-treatment with an ultra-low dose ofNADPH oxidase inhibitor diphenyleneiodoniumattenuates disease progression in multipleParkinsons disease models

    Qingshan Wang,1 Li Qian,1 Shih-Heng Chen,1 Chun-Hsien Chu,1 Belinda Wilson,1

    Esteban Oyarzabal,1 Syed Ali,2 Bonnie Robinson,2 Deepa Rao3 and Jau-Shyong Hong1

    Nicotinamide adenine dinucleotide phosphate oxidase, a key superoxide-producing enzyme, plays a critical role in microglia-

    mediated chronic neuroinammation and subsequent progressive dopaminergic neurodegeneration in Parkinsons disease.

    Although nicotinamide adenine dinucleotide phosphate oxidase-targeting anti-inammatory therapy for Parkinsons disease has

    been proposed, its application in translational research remains limited. The aim of this study was to obtain preclinical evidence

    supporting this therapeutic strategy by testing the efcacy of an ultra-low dose of the nicotinamide adenine dinucleotide phosphate

    oxidase inhibitor diphenyleneiodonium in both endotoxin (lipopolysaccharide)- and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine-

    treated mice using post-treatment regimens. Our data revealed that post-treatment with diphenyleneiodonium signicantly

    attenuated progressive dopaminergic degeneration and improved rotarod activity. Remarkably, post-treatment with diphenyleneio-

    donium 10 months after lipopolysaccharide injection when mice had 30% loss of nigral dopaminergic neurons, showed high

    efcacy in protecting the remaining neuronal population and restoring motor function. Diphenyleneiodonium-elicited neuroprotec-

    tion was associated with the inhibition of microglial activation, a reduction in the expression of proinammatory factors and an

    attenuation of a-synuclein aggregation. A pathophysiological evaluation of diphenyleneiodonium-treated mice, including assess-

    ment of body weight, organs health, and neuronal counts, revealed no overt signs of toxicity. In summary, infusion of ultra-low

    dose diphenyleneiodonium potently reduced microglia-mediated chronic neuroinammation by selectively inhibiting nicotinamide

    adenine dinucleotide phosphate oxidase and halted the progression of neurodegeneration in mouse models of Parkinsons disease.

    The robust neuroprotective effects and lack of apparent toxic side effects suggest that diphenyleneiodonium at ultra-low dose may

    be a promising candidate for future clinical trials in Parkinsons disease patients.

    1 Neuropharmacology Section, Laboratory of Neurobiology, National Institute of Environmental Health Sciences, ResearchTriangle Park, NC 27709, USA

    2 Neurochemistry Laboratory, Division of Neurotoxicology, National Centre for Toxicological Research/USFDA, Jefferson, AR72079, USA

    3 National Toxicology Program, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina27709, USA

    Correspondence to: Qingshan Wang,

    National Institute of Environmental Health Sciences,

    111 T.W. Alexander Dr.,

    Research Triangle Park,

    North Carolina, 27709, USA

    E-mail: [email protected]

    Correspondence may also be addressed to: Jau-Shyong Hong. E-mail: [email protected].

    doi:10.1093/brain/awv034 BRAIN 2015: 138; 12471262 | 1247

    Received September 12, 2014. Revised November 25, 2014. Accepted December 16, 2014. Advance Access publication February 25, 2015

    Published by Oxford University Press on behalf of the Guarantors of Brain 2015. This work is written by US Government employees and is in the public domain in the US

    by guest on June 16, 2015D

    ownloaded from

  • Keywords: Parkinsons disease; microglia; NADPH oxidase; neuroinammation; superoxide

    Abbreviations: DOPAC = dihydroxyphenylacetic acid; DPI = diphenyleneiodonium; LPS = lipopolysaccharide;MPTP = 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine; NADPH = nicotinamide adenine dinucleotide phosphate

    IntroductionParkinsons disease is an age-associated movement disorder

    that progresses over decades in aficted individuals.

    The pathological hallmark of Parkinsons disease is the

    progressive nigrostriatal dopaminergic neurodegeneration

    coupled with intracellular inclusions known as Lewy

    bodies (Olanow and Tatton, 1999). Current clinical dopa-

    mine replacement interventions for patients with

    Parkinsons disease provide temporary symptomatic relief

    but fail to halt disease progression (Salawu et al., 2010).

    Thus, alternative strategies must be developed to target

    Parkinsons disease progression to modify the course of

    the disease.

    Microglia-mediated neuroinammation has been linked

    to multiple neurodegenerative diseases, including

    Parkinsons disease (Gao et al., 2002, 2003; Gao and

    Hong, 2008; Perry et al., 2010; Czirr and Wyss-Coray,

    2012; Phani et al., 2012). These ndings prompted

    pharmaceutical companies to investigate the use of anti-

    inammatory drugs as potential treatments for

    Parkinsons disease. Early epidemiological and animal stu-

    dies supported that non-steroidal anti-inammatory drugs

    have been shown to reduce the risk of acquiring

    Parkinsons disease (Teismann and Ferger, 2001; Chen

    et al., 2005). However, recent meta-analyses and

    case-control studies failed to support these ndings (Samii

    et al., 2009; Becker et al., 2011). The development of novel

    anti-inammatory strategies to treat neurodegenerative dis-

    eases has been further hampered by the failure of several

    clinical trials (McGeer and McGeer, 2007). The inability of

    translating successful strategies from animal studies to

    human therapy highlighted the need for better therapeutic

    strategies and more suitable animal models in Parkinsons

    disease therapy development.

    One recent strategy for Parkinsons disease therapy has

    been to deviate from conventional anti-inammatory tar-

    gets and inhibit upstream mediators, such as microglial

    nicotinamide adenine dinucleotide phosphate (NADPH)

    oxidase (Gao et al., 2012), a key superoxide-producing

    enzyme. Once activated, NADPH oxidase produces extra-

    cellular and intracellular reactive oxygen species (Lambeth,

    2004), which are critical in initiating and maintaining

    chronic neuroinammatory responses, leading to progres-

    sive dopaminergic neurodegeneration (Block et al., 2007;

    Gao and Hong, 2008; Lambeth et al., 2008). As a proof

    of concept, we used the NADPH oxidase inhibitor diphe-

    nyleneiodonium (DPI) as a therapy for Parkinsons disease.

    Although DPI lacks clinical use at its recommended dose

    (mg/kg) because of non-specicity and high toxicity (Aldieri

    et al., 2008), we recently reported that DPI at

    sub-picomolar concentrations (1014 to 1013M) specic-ally inhibits NADPH oxidase activation and protects dopa-

    minergic neurons in vitro (Wang et al., 2014a). Beyond

    using a new class of anti-inammatory drugs,

    we recognized that the choice of suitable animal models

    was essential for the successful development of

    Parkinsons disease therapies. Widely used parkinsonian ani-

    mal models, including those generated by

    1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) or

    6-hydroxydopamine, acutely lesion dopaminergic neurons

    within days but fail to recapitulate the progressive feature

    of Parkinsons disease (Cannon and Greenamyre, 2010).

    Moreover, the use of pretreatment regimens for candidate

    therapies in these acute parkinsonian animal models to-

    gether with their toxicities after long-term usage have ham-

    pered the progress of successful translational therapies.

    Here, we aimed to overcome these obstacles by

    post-administering an ultra-low dose of DPI in chronic in-

    ammation-based Parkinsons disease models, which recap-

    itulate the delayed, progressive features of dopaminergic

    degeneration. We found that DPI at an extreme low dose

    (10 ng/kg/day, subcutaneously for 2-week infusion) showed

    no apparent toxicity and successfully protected dopamin-

    ergic neurons in two inammation-driven mouse models

    after the loss of 30% dopaminergic neurons. Thetherapeutic potential of DPI was further veried in a sub-

    chronic MPTP Parkinsons disease mouse model. Parallel

    experiments in NADPH oxidase-decient mice validated

    that NADPH oxidase is the molecular target of DPI-elicited

    neuroprotection. Our ndings suggest a promising strat-

    egy for arresting Parkinsons disease progression by miti-

    gating neuroinammation through the inhibition of

    NADPH oxidase.

    Materials and methods

    Animal treatments

    A repeated MPTP regimen (15mg/kg, subcutaneously for sixconsecutive days) or a single systemic lipopolysaccharide (LPS)injection (Escherichia coli 0111:B4, Sigma) were administeredto C57BL/6J and/or transgenic mice over-expressing humanA53T mutant -synuclein (B6.C3-Tg [Prnp-SNCA*A53T]83Vle/J, The Jackson Laboratory) mice. The dosage ofMPTP (Zhang et al., 2004; Wang et al., 2014b) or LPS (Qinet al., 2007, 2013) was selected based on our previous studies.Mice used as vehicle controls received an equal volume of0.9% saline. In both MPTP and LPS regimens, mice weretreated subcutaneously with DPI at 10 ng/kg/day for 2 weeksvia an Alzet osmotic pump. In LPS-treated C57BL/6J mice,DPI was administered after 3 months (pre-motor group,

    1248 | BRAIN 2015: 138; 12471262 Q. Wang et al.

    by guest on June 16, 2015D

    ownloaded from

  • n = 8 to 11 each group with total 37 mice) or 10 months(motor group, n = 6 each group with total 24 mice) afterLPS injection. LPS-treated transgenic mice over-expressinghuman A53T mutant -synuclein (n = 5 to 6 each groupwith total 16 mice) received DPI infusion after 1 month ofLPS challenge. In MPTP-injected C57BL/6J mice (n = 8 to 10each group with total 27 mice), DPI infusion started 3 daysafter the rst injection of MPTP. All the mice were euthanizedat the desired time points. Housing, breeding and experimentaluse of the animals were performed in strict accordance withthe National Institutes of Health guidelines. All procedureswere approved by the National Institute of EnvironmentalHealth Sciences/National Institutes of Health animal care anduse committee.

    Immunohistochemistry and double-labelling immunofluorescence

    Immunostaining and double-labelling immunouorescencewere performed as described previously (Gao et al., 2011;Wang et al., 2012, 2014b). For details of immunostainingassays and quantitative analysis see the online Supplementarymaterial.

    Cell counts

    The number of tyrosine hydroxylase-immunoreactive neuronsin the substantia nigra pars compacta was estimated usingstereological methodology with the optical fractionatorsmethod (MBF Science) as described previously (Wang et al.,2014b). For details see Supplementary material.

    Rotarod test

    The rotarod behaviour test was measured using a Rotamexdevice (Columbus Instruments). The parameters of therotarod system include start speed, acceleration and highestspeed (1 rpm, accelerate 12 rpm/2 s, 50 rpm). The miceunderwent three consecutive trials. The rest period betweeneach trial was 30min. The mean latency time to fall offthe rotating rod for the last two trials was used for theanalysis.

    In situ visualization of superoxide andsuperoxide-derived oxidantproduction

    In situ visualization of oxidative stress was assessed bydihydroethidium histochemistry according to previous reportswith minor modications (Quick and Dugan, 2001; Wu et al.,2003). Briey, LPS-injected mice were administered singleinjections [intraperitoneally (i.p.)] of dihydroethidium ata dose of 20mg/kg. Eighteen hours later, mice were perfusedtranscardially with PBS, and coronal substantia nigra sectionswere examined for the dihydroethidium productusing uorescence microscopy (excitation 534 nm; emission580 nm).

    Real-time PCR analysis

    Total RNA was extracted with the RNeasy Mini kit andreverse-transcribed with an oligo dT primer. Real-timePCR amplication was performed using SYBR Green PCRMaster Mix (Applied Biosystems) and Applied Biosystems7900HT Fast Real-Time PCR System according to the manu-facturers protocols. The primers were designed by Vector NTIVersion: Advance 11 software (Invitrogen, SupplementaryTable 1). The PCR conditions were 95C for 10 s, 55C for30 s, and 72C for 30 s for 40 cycles. All of the data werenormalized to Gadph.

    Catecholamine content analysis

    The levels of dopamine and its metabolite dihydroxyphenyla-cetic acid (DOPAC) were measured using high-performanceliquid chromatography and coupled with electrochemical de-tection as described previously (Zhang et al., 2004). For detailssee Supplementary material.

    Statistical analysis

    Data are expressed as the mean standard error of the mean(SEM) and were analysed statistically with Graph-Pad Prism(GraphPad Software Inc.). Differences with two groups wereanalysed using unpaired two-tailed Students t-test. Formore than two groups, one- or two-way ANOVA was applied.When ANOVA showed signicant differences, pair-wise com-parisons between means were tested by Tukeys post hoctesting. In all analyses, P50.05 was considered statisticallysignicant.

    Results

    Post-treatment with an ultra-lowdose of DPI prevents dopaminergicneurodegeneration and motordeficits in LPS-treated mice

    A neuroinammation-driven progressive dopaminergic neu-

    rodegenerative model generated by a single systemic injec-

    tion of the endotoxin LPS (Qin et al., 2007) was used to

    determine whether post-treatment of DPI can halt disease

    progression. The advantages of using this inammation-

    based model are twofold: (i) progressive nigral dopamin-

    ergic neuron loss over a period of 10 months after LPS

    injection coincides with motor decits that can be reversed

    by L-DOPA (Qin et al., 2007, Liu et al., 2008); and (ii)

    mimicking the therapeutic window for Parkinsons disease

    patients, treatment can be performed at different stages of

    disease progression. In the present study, LPS-treated mice

    were separated into two treatment groups (Fig. 1A). The

    rst group was treated with DPI 3 months after LPS injec-

    tion, a time point prior to nigral dopaminergic neuron loss

    and behavioral decits (premotor stage). The second group

    was treated with DPI after 10 months of LPS injection,

    DPI attenuates PD progression BRAIN 2015: 138; 12471262 | 1249

    by guest on June 16, 2015D

    ownloaded from

  • when signicant dopaminergic neurodegeneration (3035%

    loss of neurons) and motor decits were observed (motor

    stage). An ultra-low dose of DPI (10 ng/kg/day), which is

    approximately one-millionth of the standard doses used in

    previous reports (Vlessis et al., 1995; Miesel et al., 1996),

    was delivered systemically via a subcutaneously implanted

    mini-pump for 2 weeks. This ultra-low dose was chosen

    based on exploratory dose-response studies evaluating the

    effectiveness of doses from 0.1 to 10 ng/kg. We found that

    10 ng/kg was the lowest dose that still showed potent neu-

    roprotection. Although DPI is known to readily cross the

    bloodbrain barrier (Gatley and Martin, 1979), the meas-

    urement of the DPI concentration in the brain at this dose

    was beyond the detection sensitivity of the analysis.

    By extrapolating the pharmacokinetic data from a previous

    report (Gatley and Martin, 1979), we estimated the brain

    concentration of DPI to be in the subpicomolar range,

    which is comparable to the in vitro dose we used previously

    (Wang et al., 2014a). The efcacies of this ultra-low DPI

    on LPS-induced dopaminergic neuron loss and reduction in

    rotarod activity were determined 7 months after infusion

    for both the premotor (i.e. total 10 months after LPS injec-

    tion) and motor groups (i.e. total 17 months after LPS

    injection) (Fig. 1A). This time point was selected because

    dopaminergic neurodegeneration in premotor-group mice is

    much more evident after 10 months of LPS injection (Qin

    et al., 2007, 2013). For consistency, we also selected the

    same time point (7 months after DPI infusion) in motor-

    group mice.

    Consistent with our previous report (Qin et al., 2007),

    LPS injection reduced the number of nigral tyrosine

    hydroxylase-immunoreactive neurons in premotor stage

    mice by 32%. An almost 50% loss of tyrosine

    hydroxylase-immunoreactive neuron was detected in

    motor stage mice (Fig. 1B and C). These results replicated

    our previous nding showing progressive dopaminergic

    neurodegeneration in LPS-injected mice (Qin et al., 2007,

    2013). It is important to note that the loss of nigral dopa-

    minergic neurons was previously conrmed to reect the

    death of the neurons rather than the loss of tyrosine hydro-

    xylase immunoreactivity (Gao et al., 2011). DPI

    post-treatment exhibited signicant protection against

    LPS-induced dopaminergic neuron loss in both premotor

    (P = 0.0002, compared with LPS alone group) and motor

    (P = 0.0339, compared with LPS alone group) stage groups.

    These results indicate that the LPS-elicited progressive

    Figure 1 Post-treatment with an ultra-low dose of DPI attenuates dopaminergic neurodegeneration and motor deficits in

    LPS-treated mice. (A) Experimental designs. C57BL/6J mice received a single injection of LPS [15 106 EU/kg, intraperitoneally (i.p.)]. Three(pre-motor stage) or 10 (motor stage) months after LPS injection, the mice were infused with either vehicle or DPI (10 ng/kg/day; subcutaneously)

    via osmotic mini-pump for 2 weeks. Measurements of neuron loss and motor deficits were performed 7 months after DPI infusion. (B) Seven

    months after DPI treatment, dopaminergic neurons in the substantia nigra pars compacta were immunostained with anti-tyrosine hydroxylase

    antibody and representative images are shown. (C) The number of tyrosine hydroxylase-immunoreactive neurons in the substantia nigra pars

    compacta was counted stereologically. (D) The effects of ultra-low-dose DPI on LPS-induced motor deficits were measured using the rotarod

    test. Data are expressed as the mean SEM and were analysed by two-way ANOVA followed by Tukeys post hoc testing. *P5 0.05, **P5 0.01;#P5 0.05, ##P5 0.01; n = 611; Scale bar = 200 mm. Con = control; TH = tyrosine hydroxylase; THir = tyrosine hydroxylase-immunoreactive.

    1250 | BRAIN 2015: 138; 12471262 Q. Wang et al.

    by guest on June 16, 2015D

    ownloaded from

  • neurodegenerative process was attenuated by DPI. In con-

    currence with protecting neuronal cell bodies, DPI post-

    treatment also maintained the integrity of the neurite

    network of dopaminergic neurons in the substantia nigra

    pars reticulata, as demonstrated by greater tyrosine hydro-

    xylase-immunoreactive bre density and a reduction of

    dendritic beading (fragmented dendrites) in DPI/LPS-treated

    mice compared with the LPS alone group (Supplementary

    Fig. 1A). Consistent with these morphological observations,

    the LPS-induced decrease of striatal dopamine levels was

    signicantly attenuated by DPI post-treatment, which

    showed no differences compared with the vehicle controls

    (Supplementary Fig. 1B).

    Post-treatment with an ultra-low dose of DPI not only

    showed signicant neuroprotection but, even more impres-

    sively, displayed potent efcacy in attenuating LPS-elicited

    motor decits in both the pre-motor and motor groups.

    DPI post-treatment markedly attenuated the LPS-induced

    reduction of rotarod activity in both the premotor

    (P = 0.0132 compared with LPS alone group) and motor

    group (P = 0.0072, compared with LPS alone group)

    (Fig. 1D). It is interesting to note that DPI post-treatment

    was able to restore the rotarod activity of LPS-treated mice

    to the same level as control mice, despite its inability to

    fully prevent the loss of nigral dopaminergic neurons.

    These results suggest that ultra-low dose DPI is capable

    of halting progressive dopaminergic neurodegeneration

    and motor decits.

    Post-treatment with an ultra-lowdose of DPI is neuroprotective inLPS-treated human A53T a-synucleinover-expressing mice

    -Synuclein is a major constituent of Lewy bodies in patients

    with Parkinsons disease. The A53T mutation in the SNCA

    gene is known to increase the amount of aggregated -synu-

    clein and is closely associated with dopaminergic neurode-

    generation (Lee and Trojanowski, 2006). Although

    transgenic mice over-expressing human A53T mutant -

    synuclein have been previously used as rodent models of

    Parkinsons disease, reports have shown that this transgenic

    mouse fails to develop overt nigral dopaminergic neurode-

    generation (Giasson et al., 2002; Gao et al., 2011). We re-

    cently developed a two-hit model by injecting LPS into

    transgenic mice over-expressing human A53T mutant -

    synuclein, which showed earlier onset and a much more

    robust progressive loss of nigral dopaminergic neurons than

    C57BL/6J mice (Gao et al., 2011). Thus, the LPS-injected

    transgenic mouse can serve as an ideal model with which to

    investigate the neuroprotective effects of DPI. Because of the

    early onset (as early as 2.5 months after LPS injection) of

    dopaminergic neurodegeneration in LPS-treated transgenic

    mice over-expressing human A53T mutant -synuclein

    (Gao et al., 2011), the protective effects of DPI were deter-

    mined after 4 months of LPS injection, a time point that

    showed 4050% loss of dopaminergic neurons. Consistent

    with previous report (Gao et al., 2011), 4 months after LPS

    injection, a 43% loss of nigral dopaminergic neurons was

    noted in transgenic mice over-expressing human A53T

    mutant -synuclein compared to untreated littermate con-

    trols. Post-treatment with DPI 1 month after LPS injection

    led to a signicant protection (P = 0.0029, compared with

    LPS alone group) of the nigral tyrosine hydroxylase-immu-

    noreactive neurons (Fig. 2AC). A similar degree of DPI-eli-

    cited protection of dopaminergic terminals in the striatum

    was also observed, as shown by the high striatal tyrosine

    hydroxylase density in the DPI-treated group (87 4% ofcontrol) compared to the non-DPI-treated group (56 7%,P = 0.0006; Fig. 2B and D).

    The expression of human -synuclein in transgenic mice

    over-expressing human A53T mutant -synuclein is not

    homogeneous in the brain with high levels in brainstem

    and cortex and very low levels in the substantia nigra

    (Giasson et al., 2002). We recently conrmed this nding

    showing minimal protein expression in the substantia nigra

    by western blot and immunohistochemistry using an

    antibody again human -synuclein in untreated transgenic

    mice over-expressing human A53T mutant -synuclein

    (Gao et al., 2011). Interestingly, after LPS injection,

    increased expression and insolubility of human -synuclein

    were observed in the substantia nigra (Gao et al., 2011).

    In addition to protecting dopaminergic neurons, DPI

    post-treatment mitigated LPS-induced human -synuclein

    accumulation in the substantia nigra, as detected by immu-

    nostaining using SYN211 antibody (specic for human

    -synuclein, Fig. 3A). Double-label immunouores-

    cence analysis revealed an accumulation of -synuclein

    in the cytosol and perinuclear locations of nigral tyrosine

    hydroxylase-immunoreactive neurons after LPS injection. In

    contrast, nigral -synuclein immunoreactivity in DPI/LPS-

    treated transgenic mice over-expressing human A53T

    mutant -synuclein was diffuse and barely visible

    (Fig. 3B). Quantitative analysis of the nigral SYN211

    density indicated a 223% increase in the LPS alone

    group, which was reduced to 127% in DPI/LPS-treated

    mice (P5 0.0001, compared with LPS alone group;Fig. 3C).

    Post-treatment with an ultra-lowdose of DPI attenuates LPS-elicitedmicroglia-mediatedneuroinflammation

    To determine whether the neuroprotective effects of DPI

    were related to its anti-inammatory properties, we exam-

    ined the inhibitory effects of DPI on microglial activation

    using post-treatment regimens at different stages of the

    neurodegenerative process. Activation of microglia in

    the subsantia nigra was morphologically observed by

    immunostaining with two microglial markers: ionized cal-

    cium binding adaptor molecule 1 (AIF1, also known as

    DPI attenuates PD progression BRAIN 2015: 138; 12471262 | 1251

    by guest on June 16, 2015D

    ownloaded from

  • Iba-1) and ITGAM (formerly known as CD11b, alpha

    chain of the b-2 integrin receptor). In both the premotorand motor stage, activated microglia characterized by a

    hypertrophied morphology and intensied ITGAM and

    AIF1 staining were observed throughout the nigral reticu-

    lata area (Fig. 4A). Analysis of ITGAM and AIF1 density

    and cell body size supported these morphological observa-

    tions. Compared with the LPS alone group, post-treatment

    with DPI markedly attenuated microglial activation, as

    shown by a reduced density of ITGAM (P = 0.0020 and

    0.0001 in premotor and motor stage, respectively) and

    AIF1 (P = 0.0198 and 0.0001 in premotor and motor

    stage, respectively) staining and microglial cell body sizes

    (P50.0001 and 0.0001 in premotor and motor stage, re-spectively; Fig. 4BD).

    Activated microglia secrete a variety of toxic factors, such

    as tumor necrosis factor alpha, interleukin-1 beta and other

    proinammatory cytokines, which work in concert to cause

    neuronal damage (Block and Hong, 2005). Exposure to

    LPS produced a long-lasting 2-fold increase in the

    expression of the proinammatory genes tumor necrosis

    factor alpha, interleukin-1 beta and major histocompatibil-

    ity complex II, in both the premotor and motor groups.

    Interestingly, ultra-low dose DPI post-treatment prevented

    the LPS-induced increase in the gene expression of these

    immune factors (Fig. 4EG).

    Post-treatment with an ultra-lowdose of DPI inhibits LPS-inducedoxidative stress

    NADPH oxidase is essential for maintaining chronic micro-

    glia-mediated neuroinammation and subsequent progres-

    sive dopaminergic neurodegeneration (Qin et al., 2013).

    Although DPI exhibits potent inhibitory effects on

    NADPH oxidase at mg/kg doses, it is not clear whether

    DPI administered at ultra-low dose is still capable

    of inhibiting NADPH oxidase activation in vivo. To

    address this question, we determined the effects of

    Figure 2 Post-treatment with an ultra-low dose of DPI protects dopaminergic neurons against LPS-induced damage in

    transgenic mice over-expressing human A53T mutant a-synuclein. (A) Experimental designs. Seven-month-old transgenic mice over-expressing human A53T mutant -synuclein received a single injection of LPS [6 106 EU/kg, intraperitoneally (i.p.)]. One month after LPSinjection, the mice were infused with either vehicle or DPI (10 ng/kg/day; subcutaneously) via osmotic mini-pump for 2 weeks. (B) Three months

    after DPI post-treatment, nigral dopaminergic neurons and striatal axon fibres were immunostained with anti-tyrosine hydroxylase antibody, and

    representative images are shown. (C) The number of tyrosine hydroxylase-immunoreactive cells was counted stereologically, and the results are

    expressed as the mean SEM. (D) The density of tyrosine hydroxylase immunostaining in the striatum was quantified using densitometricanalysis. Data were analysed by one-way ANOVA followed by Tukeys post hoc testing. **P5 0.01; Scale bar = 200 mm; n = 56. TH = tyrosinehydroxylase.

    1252 | BRAIN 2015: 138; 12471262 Q. Wang et al.

    by guest on June 16, 2015D

    ownloaded from

  • ultra-low dose DPI on reactive oxygen species production

    in the brain after 10 months of LPS injection (Fig. 5A). Our

    previous report showed that a single systemic LPS injection

    causes persistent NADPH oxidase activation and related

    reactive oxygen species production in the mouse brain

    (Qin et al., 2013). Electron spin resonance analysis further

    conrmed that NADPH oxidase, but not other oxidases,

    such as xanthine oxidase, accounts for the major

    (4 90%) source of LPS-induced oxidative stress in vivo(Sato et al., 2002). In situ visualization of reactive

    oxygen species production was performed using dihy-

    droethidium, a reactive oxygen species-sensitive dye.

    Dihydroethidium can readily cross the bloodbrain barrier

    and exhibits red uorescence through interactions with

    superoxide and other free radicals in the brain (Wu et al.,

    2003). As shown in Fig. 5B, reactive oxygen species pro-

    duction in the substantia nigra was minimal in vehicle

    controls, as evidenced by the low levels of red uorescence.

    In contrast, exposure to LPS resulted in increased levels of

    red uorescence in the substantia nigra, indicating elevated

    reactive oxygen species production. Co-staining with tyro-

    sine hydroxylase antibody revealed a high degree of oxida-

    tive stress in the dopaminergic neurons of LPS-injected mice

    (Fig. 5C). Post-treatment with ultra-low dose DPI markedly

    reduced LPS-induced oxidative stress in the nigra

    and nigral dopaminergic neurons (Fig. 5B and C).

    Quantitative analysis revealed 2.3-fold increase in uores-

    cence density in LPS-injected mice compared with vehicle

    controls, which was reduced to 1.1-fold in DPI/LPS-treated

    mice (P5 0.0001; Fig. 5D). The inhibitory effects of DPIon LPS-induced reactive oxygen species production suggest

    that ultra-low dose DPI can inhibit NADPH oxidase

    activation.

    The source of reactive oxygen species production has

    recently become a subject of debate. Mitochondria have

    been traditionally considered a major source of intracellular

    reactive oxygen species production; however, our previous

    nding indicates that NADPH oxidase is a key reactive

    oxygen species-generating enzyme in microglia in response

    to LPS stimulation (Qin et al., 2013). In addition to

    NADPH oxidase, DPI at regularly used doses (between 1

    to 5mg/kg) inhibits a variety of electron-transferring avo-

    protein enzymes, including mitochondrial complex

    I (Aldieri et al., 2008). To investigate whether ultra-low

    dose DPI has specicity toward NADPH oxidase in vivo,

    we evaluated the effects of DPI on complex I activity.

    Interestingly, DPI at 10 ng/kg failed to suppress the activ-

    ities of mitochondrial complex I in the brain (Fig. 5E). This

    result indicated a high specicity of ultra-low dose DPI in

    inhibiting NADPH oxidase and revealed a critical role

    of this superoxide-producing enzyme in the generation of

    reactive oxygen species in the brain.

    Figure 3 Post-treatment with an ultra-low dose of DPI attenuates nigral a-synuclein aggregation in LPS-treated transgenicmice over-expressing human A53T mutant a-synuclein. (A) One month after LPS injection, transgenic mice over-expressing human A53Tmutant -synuclein were infused with either vehicle or DPI (10 ng/kg/day; subcutaneously) via osmotic mini-pump for 2 weeks. Three months

    after DPI post-treatment, human -synuclein was immunostained in the substantia nigra with SYN211 (specific for human -synuclein) antibody,

    and representative images are shown. (B) Magnifications of dopaminergic neuron (tyrosine hydroxylase-immunoreactive) and -synuclein double

    staining are indicated in the different groups. (C) The SYN211 density in the substantia nigra was quantified. The results are expressed as a

    percentage of the vehicle controls (mean SEM) and were analysed by one-way ANOVA followed by Tukeys post hoc testing. **P5 0.01; Scalebar = 200 mm in (A) and 50mm in (B); n = 56. -Syn = -synuclein.

    DPI attenuates PD progression BRAIN 2015: 138; 12471262 | 1253

    by guest on June 16, 2015D

    ownloaded from

  • Ultra-low dose DPI affords long-termdopaminergic neuroprotectionagainst MPTP lesions

    In addition to using LPS models, we tested the protective

    effects of ultra-low dose DPI in a MPTP Parkinsons disease

    mouse model, which has been widely used to screen for

    therapeutic agents. Unlike LPS, MPTP directly damages

    dopaminergic neurons in an acute fashion. Therefore, we

    initially evaluated the neuroprotective potential of DPI

    using a pre-treatment regimen. DPI infusion (10 ng/kg/

    day, subcutaneously) started 2 days before MPTP adminis-

    tration and lasted for 2 weeks (Supplementary Fig. 2A).

    The neuroprotective effects of DPI were evaluated at differ-

    ent time points (27, 60 and 120 days after the initial

    injection of MPTP). The chosen 27-day point was based

    on our previous report (Hu et al., 2008) showing nearly

    50% loss of dopaminergic neurons in the substantia nigra

    pars compacta. Consistently, MPTP elicited a 45% loss of

    tyrosine hydroxylase-immunoreactive neurons in the sub-

    stantia nigra pars compacta compared with vehicle controls

    at the 27-day point. The loss of nigral tyrosine hydroxy-

    lase-immunoreactive neurons remained close to the same

    degree at the 60- and 120-day time points. MPTP-elicited

    tyrosine hydroxylase-immunoreactive neuronal loss was

    signicantly attenuated by DPI (P = 0.0032, compared

    with the MPTP alone group) at the 27-day time point.

    The long-lasting protective effect of DPI was evident

    based on the results obtained at the 60- and 120-day

    time points (Supplementary Fig. 2B). To exclude the

    Figure 4 Post-treatment with an ultra-low dose of DPI attenuates chronic microglial activation. (A) Three (pre-motor) or 10

    (motor) months after LPS injection, mice (C57BL/6J) were infused with either vehicle or DPI (10 ng/kg/day; subcutaneously) via osmotic mini-

    pump for 2 weeks. Two microglial markers, ITGAM (CD11b) or AIF1 (Iba-1), were immunostained in the substantia nigra region 7 months after

    DPI treatment. Representative pictures of staining are shown. Activated microglia characterized by an enlarged cell body size and high staining

    density. (BD) The activation of microglia was quantified by measuring the density of ITGAM (CD11b; B) and AIF1 (Iba-1) (C) and the cell body

    size (D). The gene expressions of tumor necrosis factor alpha (TNF; E), interleukin-1 beta (Il-1b; F) and major histocompatibility complex II (G)in brain were determined in the rostral half of the brains using RT-PCR. Data are expressed as a percentage of time-matched vehicle controls

    (mean SEM) and were analysed by two-way ANOVA followed by Tukeys post hoc testing. *P5 0.05, **P5 0.01; n = 56; Scale bar = 50 mm.MHCII = major histocompatibility complex II.

    1254 | BRAIN 2015: 138; 12471262 Q. Wang et al.

    by guest on June 16, 2015D

    ownloaded from

  • possibility that DPI-afforded protection was due to either

    alterations of MPTP metabolism or entry into the brain, we

    measured MPP+ levels in the brain after MPTP injection

    and found no signicant difference between MPTP and

    DPI/MPTP (data not shown).

    In addition to evaluating the DPI-afforded protection of

    nigral tyrosine hydroxylase-immunoreactive neurons, stri-

    atal levels of dopamine and its metabolite, DOPAC as

    well as their turnover rate (DOPAC/dopamine) were

    measured as markers for the functional recovery of dopa-

    minergic neurons (Zigmond et al., 1990; Zigmond, 1997).

    Twenty-seven days after the MPTP injections, marked de-

    creases in striatal levels of dopamine (68%) compared with

    the vehicle controls were observed (Supplementary Fig.

    2C); in contrast, the decrease in DOPAC level was attenu-

    ated (Supplementary Fig. 2D). The higher ratio of DOPAC/

    dopamine in the MPTP-treated group further indicated

    a higher turnover of dopamine, likely due to a compensa-

    tory effect of dopaminergic neuron loss (Supplementary

    Fig. 2E). Interestingly, whereas DPI did not prevent the

    MPTP-induced loss of striatal dopamine levels, signicantly

    high levels of DOPAC (P = 0.0207, compared with the

    MPTP alone group) and DOPAC/dopamine ratios

    (P = 0.0284 compared with the MPTP alone group) were

    observed in the DPI-treated group (Supplementary Fig. 2D

    and E), suggesting enhanced functional activities of the

    surviving dopaminergic neurons (Zigmond et al., 1990;

    Zigmond, 1997). A time-dependent recovery of dopamine

    levels was found at the 60-day time point for both the

    MPTP/vehicle and the MPTP/DPI groups compared with

    the levels at the 27-day time point. Although the striatal

    levels of dopamine were the same between these two

    groups, the DOPAC level and DOPAC/dopamine ratio re-

    mained high in the DPI-treated group, suggesting that DPI

    preserved some degree of functional activity in remaining

    dopaminergic neurons. Continuing recovery of striatal

    dopamine levels was shown at the 120-day time point in

    the MPTP-treated group. It is interesting to note that the

    levels of dopamine in DPI/MPTP-treated mice returned to

    the same level as non-MPTP-injected mice (P = 0.9960;

    Figure 5 Post-treatment with an ultra-low dose of DPI inhibits the LPS-induced increase in dihydroxyethidium oxidation but

    not the activity of mitochondrial complex I. (A) Experimental designs. Ten months after LPS injection, mice (C57BL/6J) were infused with

    DPI (10 ng/kg/day). After 2 weeks of DPI infusion, the brains were perfused, and the production of superoxide was determined by measuring the

    oxidation products of dihydroethidium by fluorescence microscopy. (B) Representative images of dihydroethidium staining (red) are shown. The

    position of the substantia nigra was identified by tyrosine hydroxylase staining (green). (C) Magnification of dopaminergic neurons (tyrosine

    hydroxylase-immunoreactive) and dihydroethidium double staining are indicated in the different groups. (D) Quantified data showing the fluor-

    escence density of the dihydroethidium oxidation in the substantia nigra of different treatment groups. Fluorescence density of NADPH oxidase-

    deficient (gp91phox /) mice served as a control background. (E) The effects of ultra-low-dose DPI on the activities of complex I in the brain weredetermined using commercial assay kits. Data are expressed as a percentage of time-matched vehicle controls (mean SEM) and were analysedby Students t-test (E) or one-way ANOVA followed by Tukeys post hoc testing (D). **P5 0.01; n = 5; Scale bar = 100 mm.DHE = dihydroethidium; TH = tyrosine hydroxylase; THir = tyrosine hydroxylase-immunoreactive.

    DPI attenuates PD progression BRAIN 2015: 138; 12471262 | 1255

    by guest on June 16, 2015D

    ownloaded from

  • Supplementary Fig. 2C). The rapid recovery of striatal

    dopamine levels supported the promising neuroprotective

    effects of DPI.

    Ultra-low dose DPI attenuatesMPTP-induced oxidative stress andmicroglial activation

    Additional studies were performed to elucidate the mech-

    anism by which DPI protected against MPTP-elicited

    neurotoxicity. Although MPTP is directly toxic to dopa-

    minergic neurons, microglia-mediated neuroinammation

    contributes to its overall neurotoxicity (Wu et al., 2003;

    Hu et al., 2008; Levesque et al., 2010). Microglia are

    indirectly activated to produce cytotoxic factors (reactive

    microgliosis) in response to MPTP-induced neuronal

    damage, causing additional dopaminergic degeneration

    (Levesque et al., 2010). To provide evidence that microglia

    play a role in DPI-elicited neuroprotection, immunostaining

    of microglial markers was performed. DPI treatment signi-

    cantly attenuated MPTP-induced microglial activation,

    as shown by the reduced cell body size and densities of

    ITGAM and AIF1 staining in the substantia nigra

    (Supplementary Fig. 3A). Quantitative analysis revealed

    decreased immunoreactivities of ITGAM (P = 0.0466) and

    AIF1 (P = 0.0385) in the substantia nigra compared with

    the MPTP alone group (Supplementary Fig. 3B). Moreover,

    DPI signicantly reduced MPTP-elicited oxidative stress

    in the substantia nigra based on the low nigral density

    of dihydroethidium oxidation (red uorescence) in the

    DPI/MPTP group compared with the MPTP alone group

    (P = 0.0039; Supplementary Fig. 4A and C). Consistent

    with the LPS model, co-staining with tyrosine hydroxylase

    antibody revealed that DPI treatment reduced oxidative

    stress in dopaminergic neurons (Supplementary Fig. 4B).

    Overall, our results show that ultra-low dose DPI

    inhibits MPTP-induced reactive microgliosis and oxidative

    stress.

    NADPH oxidase deficiency abolishesultra-low dose DPI-affordedneuroprotection

    To investigate whether NADPH oxidase mediates ultra-

    low dose DPI-afforded neuroprotection, NADPH oxi-

    dase-decient (gp91phox /) and wild-type control(gp91phox+ / + ) mice were treated with DPI followed by le-

    sioning with MPTP. Consistent with the data in

    Supplementary Fig. 2B, in wild-type mice, MPTP injection

    caused a 46% loss of nigral tyrosine hydroxylase-

    immunoreactive neurons and had only 23% loss in DPI/

    MPTP-treated group (P = 0.0001; Supplementary Fig. 5A

    and C), compared to the vehicle control. In contrast, mice

    lacking gp91phox were more resistant to MPTP-induced

    lesions than wild-type controls (27 3% versus 46 3%loss of nigral tyrosine hydroxylase-immunoreactive

    neurons, P = 0.0027), supporting the potential involvement

    of NADPH oxidase in MPTP-induced dopaminergic neuron

    damage. Under this condition, ultra-low dose DPI failed to

    protect nigral dopaminergic neurons in gp91phox / micewith MPTP lesions (P = 0.9997, compared with MPTP

    alone group). Consistently, reduced degeneration of ter-

    minals of dopaminergic neurons in the striatum were

    observed in DPI/MPTP-treated wild type (gp91phox\ + )

    mice, but no difference was detected in NADPH oxidase-

    decient (gp91phox /) mice (Supplementary Fig. 5Band D).

    Post-treatment with DPI attenuatesMPTP-induced dopaminergicdegeneration and motor deficits

    The positive results from the pretreatment studies

    (Supplementary Fig. 2 to 5) led us to explore the possibility

    of a post-treatment regimen to further assess the thera-

    peutic efcacy of ultra-low dose DPI in the MPTP model.

    DPI (10 ng/kg/day) was post-administered at Day 3 after

    the rst injection of MPTP for 2 weeks (Fig. 6A). Pilot

    studies showed a 2030% loss of nigral tyrosine hydro-xylase-immunoreactive neurons when DPI infusion

    was initiated. Twenty-seven days after MPTP lesion, a

    signicantly greater number of nigral tyrosine

    hydroxylase-immunoreactive neurons was found in DPI/

    MPTP-treated mice (P = 0.0239, compared with the

    MPTP alone group; Fig. 6B). Consistent with the pretreat-

    ment regimens, DPI post-treatment enhanced the activity of

    nigral dopaminergic neurons, as shown by an increased

    dopamine turnover rate compared to the MPTP alone

    group (P = 0.0067; Fig. 6C). The neuroprotective effect of

    DPI post-treatment was correlated with the attenuation of

    MPTP-elicited motor decits (P = 0.0470), as measured by

    the rotarod test (Fig. 6D). Longer-term studies similar to

    Supplementary Fig. 2 were not conducted in this post-treat-

    ment regimen. However, based on the high turnover rate of

    dopamine and improved motor behaviour in DPI-treated

    mice, it is likely that mice post-treated with DPI would

    display an accelerated recovery from MPTP treatment at

    later time points.

    Ultra-low dose DPI displays no overttoxicity in mice

    The severe toxicity of DPI at the recommended dose ham-

    pers its potential for clinical usage in patients. Thus, we

    conducted a toxicological screen of DPI when administered

    at ultra-low dose. All parameters studied in this project,

    including the quantitative and morphological analysis

    (neurons and microglia), neurochemical measurements

    (proinammatory factors, complex I activity and dihy-

    droethidium oxidation) and behavioural observations

    (rotarod activities, Fig. 7A), showed no difference in DPI

    alone group compared to vehicle controls. General

    1256 | BRAIN 2015: 138; 12471262 Q. Wang et al.

    by guest on June 16, 2015D

    ownloaded from

  • evaluation of major organ systems and organ weights

    concluded that ultra-low dose DPI treatment had no signi-

    cant effects on body and organ weights (including liver,

    kidney and spleen; Fig. 7BE) nor any observable histo-

    pathological changes in haematoxylin and eosin-staining

    of sections from liver, spleen, kidney, testes, heart and

    lung (Fig. 7F) when compared to saline treated controls.

    Additionally, haematoxylin and eosin staining of brain sec-

    tions based on the modied National Toxicology Program

    protocol (Rao et al., 2013) also revealed no signicant

    changes in DPI-treated mice compared to vehicle controls

    (Fig. 7F). We further determined whether low doses of DPI

    affect the peripheral immune function by examining both

    high and low doses of DPI on superoxide production in

    mice neutrophils. Results indicated that high dose DPI

    (105M) completely inhibited superoxide production, butDPI in low concentrations (1013M and 1014M) failed toaffect the oxidative burst of neutrophils (Supplementary

    Fig. 6). Our preliminary data also showed no effect of

    low-dose DPI on the superoxide production of human

    neutrophils (data not shown). Overall, these ndings sug-

    gest that ultra-low dose DPI displayed no overt organ tox-

    icity and does not inuence peripheral immune cell

    functions that are critical for hosting normal immune

    responses.

    DiscussionThe present study demonstrated that DPI at an ultra-low

    dose provides potent benecial effects in three models of

    dopaminergic degeneration. One salient feature was the

    high efcacy of DPI in neuroprotection, even when admin-

    istered in a post-treatment regimen after the onset of dopa-

    minergic neuron damage (see summary in Supplementary

    Table 2). Our results strongly support that the neuropro-

    tective effects of ultra-low dose DPI occur by specically

    inhibiting NADPH oxidase and subsequently reducing

    microglia-mediated chronic neuroinammation (Fig. 8).

    Additionally, mice treated with ultra-low dose DPI

    showed no overt signs of toxicity. The efcacy of a post-

    treatment regimen in chronic progressive dopaminergic

    neuron degenerative models together with its low toxicity

    suggest that ultra-low dose DPI may be a promising drug

    candidate for future human studies.

    Over the past decade, the development of drugs capable

    of modifying disease progression in Parkinsons disease has

    been unsuccessful. Despite the encouraging results reported

    in numerous animal studies, a small percentage of these

    compounds have been tested in clinical trials with even

    fewer reaching the clinic (Hart et al., 2009; Brichta et al.,

    2013). One particular failure in translating these drugs has

    Figure 6 Post-treatment with an ultra-low dose of DPI attenuates MPTP-induced dopaminergic neuron damage and motor

    deficits. (A) Experimental designs. Repeated MPTP regimens (15 mg/kg, subcutaneously for 6 consecutive days) were administered to C57BL/6J

    mice. After 3 days of MPTP injection, the mice were infused with either vehicle or DPI (10 ng/kg/day; subcutaneously) via osmotic mini-pump for

    2 weeks. (B) Twenty-seven days after the first injection of MPTP, dopaminergic neurons in the substantia nigra pars compacta were immunos-

    tained with anti-tyrosine hydroxylase antibody, and the numbers of tyrosine hydroxylase-immunoreactive cells were counted. (C) The turnover

    rate of dopamine in the striatum was calculated by the ratio of dopamine metabolite (DOPAC) and dopamine. (D) The protective effects of DPI

    against MPTP-induced motor deficits were measured by the rotarod test. Data are expressed as the mean SEM and were analyzed by one-wayANOVA followed by Tukeys post hoc testing. *P5 0.05, **P5 0.01; n = 810. TH = tyrosine hydroxylase; THir = tyrosine hydroxylase-immunoreactive.

    DPI attenuates PD progression BRAIN 2015: 138; 12471262 | 1257

    by guest on June 16, 2015D

    ownloaded from

  • been the lack of suitable Parkinsons disease models for

    drug development, which prompted us to create two

    inammation-based progressive neurodegenerative models

    by using a systemic injection of LPS to either C57BL/6J

    or transgenic mice over-expressing human A53T mutant

    -synuclein (Qin et al., 2007; Gao et al., 2011). These

    new models improve upon existing models by recapitulat-

    ing the delayed and progressive degeneration of nigrostria-

    tal dopaminergic neurons (Qin et al., 2007; Gao et al.,

    2011), showing temporary recovery of motor decits by

    L-DOPA (Qin et al., 2007; Liu et al., 2008) and generating

    -synuclein-positive inclusion bodies in the substantia nigra

    (Liu et al., 2008; Gao et al., 2011). Administering an ultra-

    low dose of DPI to these mouse models not only halted the

    progression of neurodegeneration in mice that already

    exhibited more than a 30% loss of nigral dopaminergic

    neurons but also attenuated -synuclein accumulation in

    dopaminergic neurons in the transgenic mice over-express-

    ing human A53T mutant -synuclein (Fig. 3). To our

    knowledge, this is the rst report demonstrating a therapy

    capable of halting progressive dopaminergic neurodegen-

    eration, reducing -synuclein accumulation, attenuating

    the depletion of striatal dopamine, and improving motor

    behaviours even when administered as a post-degenerative

    intervention.

    Mechanistically, the most critical question to address

    is why such a low dose of DPI post-administered at

    the motor stage of disease progression still displays neuro-

    protection and reverses motor decits. The results of

    the present study, together with our previous reports

    (Block et al., 2007; Gao and Hong, 2008; Qin et al.,

    2013), suggest that DPI achieves these extraordinary pro-

    tective effects through the inhibition of NADPH oxidase

    and subsequent interruption of microglia-mediated chronic

    neuroinammation. Although microglial activation is essen-

    tial to restore brain homeostasis after an injury or infection

    (Streit, 2000), it may become pathological if the initial in-

    ammation is not properly resolved. Release of noxious

    endogenous ligands generated by injured neurons, such as

    m-calpain, -synuclein and high mobility group box 1, arethought to continually reactivate microglia (reactive micro-

    gliosis) resulting in additional neurodegeneration (Block

    et al., 2007; Levesque et al., 2010; Gao et al., 2011).

    Consequently, a self-propelling vicious cycle is created

    through interactions between injured neurons and dysregu-

    lated microglia, inevitably resulting in the delayed and pro-

    gressive collateral neurodegeneration of dopaminergic

    neurons in Parkinsons disease (Gao and Hong, 2008).

    We previously recognized NADPH oxidase as a key medi-

    ator in bridging chronic neuroinammation and progressive

    dopaminergic neurodegeneration (Qin et al., 2004; Zhang

    et al., 2004; Block et al., 2007; Gao and Hong, 2008). This

    nding led us to theorize that inhibiting NADPH oxidase

    could effectively disrupt this self-propelling vicious cycle,

    potentially resulting in a new disease-modifying strategy

    for Parkinsons disease. As predicted, ultra-low dose DPI

    effectively reduced LPS-induced oxidative stress, which was

    mainly derived from NADPH oxidase activation and sub-

    sequent superoxide production (Fig. 5 and Supplementary

    Fig. 4). The inhibition of NADPH oxidase not only reduced

    Figure 7 An ultra-low dose of DPI displays no toxicity. Mice were treated with either vehicle or DPI (10 ng/kg/day; subcutaneously) using

    an Alzet mini-pump for 2 weeks. The effects of ultra-low-dose DPI on rotarod (A), and body (B), liver (C), spleen (D) and kidney (E) weights

    were assessed. (F) Organs including liver, spleen, kidney, testis, heart, lung and brain were dissected and stained with haematoxylin and eosin.

    Representative images are shown. Data were analysed using a Students t-test. n = 3; Scale bar = 100 mm.

    1258 | BRAIN 2015: 138; 12471262 Q. Wang et al.

    by guest on June 16, 2015D

    ownloaded from

  • extracellular superoxide but also decreased intracellular

    reactive oxygen species levels thought to be important sec-

    ondary messengers that regulate the expression of many

    proinammatory factors by activating several downstream

    signalling pathways including protein kinase C, mitogen-

    activated protein kinase and NF-B (Block and Hong,

    2005). Consistent with this mode-of-action, post-treatment

    with DPI produced long-term inhibition of LPS-elicited

    microglial activation and suppressed gene expression of

    proinammatory factors tumor necrosis factor alpha and

    interleukin-1 beta (Fig. 4). Unlike conventional anti-inam-

    matory therapies that directly suppress certain pro-inam-

    matory factors, selective inhibition of NADPH oxidase by

    DPI can inactivate microglia and prevent the generation of

    a spectrum of pro-inammatory factors. Although this

    study focused primarily on microglial NADPH oxidase,

    neurons are also express this superoxide-producing

    enzyme at much lower quantities and thus, we cannot ex-

    clude the possibility that the inhibition of neuronal

    NADPH oxidase might also contribute to the DPI-elicited

    neuroprotection.

    In addition to using LPS models, the efcacy of

    DPI was demonstrated in a more conventional MPTP

    model. Although previous studies have demonstrated pro-

    tection by post-treatment with rottlerin (protein kinase C

    inhibitor) (Zhang et al., 2007) or caffeine (Xu et al., 2010)

    in a MPTP mouse model, their protective effects were only

    observed when the drugs were administered within a couple

    of hours after MPTP lesion. By using a subchronic MPTP

    model, we showed that DPI (10 ng/kg/day) post-treatment

    after 3 days of initial MPTP injection attenuated dopamin-

    ergic neurodegeneration and improved rotarod activity.

    It is interesting to compare the LPS and MPTP models in

    this study. LPS is known to directly activate microglia and

    trigger neuroinammation to produce delayed and progres-

    sive nigral neurodegeneration (Qin et al., 2007). In con-

    trast, MPTP causes acute dopaminergic neurotoxicity by

    inhibiting mitochondrial complex I. The different modes-

    of-action of these models begs the question how DPI can

    partially prevent dopaminergic neuronal loss, improve

    dopamine turnover and restore motor function in a

    model of direct neuronal lesion (Fig. 6). Although MPTP

    cannot directly activate microglia, stress signals released by

    dying neurons derive reactive microgliosis to generate

    superoxide via NADPH oxidase (Block et al., 2007;

    Levesque et al., 2010). We and others have previously

    reported that reactive microgliosis generated in response

    to the acute lesioning of dopaminergic neurons by MPTP

    trigger a delayed collateral neurotoxicity from neuroinam-

    mation after the acute phase of MPTP (Gao et al., 2003;

    Wu et al., 2003; Hu et al., 2008; Levesque et al., 2010).

    Thus, we believe that DPI protected against this delayed

    toxicity by attenuating reactive microgliosis through the

    inhibition of NADPH oxidase activity.

    It is important to note that although DPI potently inhibits

    microglial activation in both LPS and MPTP models, DPI

    alone did not produce microglial cytotoxicity nor did it sup-

    press the basal microglial activity as illustrated by the

    similar densities of AIF1 and ITGAM-positive microglia

    and the gene expressions of proinammatory cytokines

    Figure 8 Proposed model showing how ultra-low-dose DPI attenuates progressive dopaminergic neurodegeneration. NADPH

    oxidase is a key mediator for initiating and maintaining the self-propagating vicious cycle formed between damaged neurons and dysregulated

    microglia. The self-propelling vicious cycle is critical in driving the progressive dopaminergic degeneration in Parkinsons disease. Ultra-low dose

    DPI is capable of inhibiting the activation of NADPH oxidase and subsequent production of superoxide and other neurotoxic factors to mitigate

    chronic neuroinflammation. Once the self-propelling vicious cycle is interrupted by inhibiting NADPH oxidase on microglia, neurons or both, the

    progression of dopaminergic neuron degeneration can be halted. These results provide a novel and promising avenue for developing drug therapy

    for Parkinsons disease and other neurodegenerative diseases.

    DPI attenuates PD progression BRAIN 2015: 138; 12471262 | 1259

    by guest on June 16, 2015D

    ownloaded from

  • in comparison with the vehicle controls (Fig. 4). This

    observation is clinically relevant because DPI attenuates

    only the induced microglial activation without interfering

    with normal microglial immune surveillance function.

    Taken together, our ndings suggest that the reduction

    of chronic microglia-mediated neuroinammation through

    the inhibition of NADPH oxidase is an effective strategy

    to halt disease progression in both inammation (LPS)- and

    neurotoxin (MPTP)-based rodent Parkinsons disease

    models.

    This study also addressed some critical issues relevant to

    the potential clinical use of ultra-low dose DPI in future

    human clinical trials. The rst issue was resolving the dur-

    ation of treatment necessary to achieve an observable

    effect. To our surprise, we found that the subcutaneous

    infusion of DPI for two weeks was sufcient to provide

    potent neuroprotection. Although it is not clear why

    long-term administration was not necessary, one possible

    explanation for this effect is that once the self-propelling

    vicious cycle is interrupted by DPI, chronic neuroinamma-

    tion ceases unless additional inammation is induced in the

    brain. The second issue was whether DPI could specically

    inhibit NADPH oxidase in vivo. In micromolar concentra-

    tions, DPI inhibits several essential cytochrome-containing

    enzymes beyond just NADPH oxidase, attributing to its

    high cytotoxicity at these concentrations (Gatley and

    Martin, 1979; Aldieri et al., 2008). We recently reportedthat subpicomolar concentrations of DPI have great

    specicity to potently inhibit NADPH oxidase-generated

    superoxide without affecting the activities of other cyto-

    chrome-containing enzymes, such as inducible nitric oxide

    synthase, xanthine oxidase, cytochrome P450 reductase,

    thioredoxin reductase, and complex I, in cultured microglia

    (Wang et al., 2014a). Although the exact brain concentra-

    tion of DPI in our in vivo studies was too low to beaccurately measured, data extrapolation from a previous

    pharmacokinetic study (Gatley and Martin, 1979) estimate

    that the brain DPI levels in treated mice were similar to the

    range (1014 to 1013M) used in the aforementionedin vitro studies (Wang et al., 2014a). The specicity of

    this ultra-low dose DPI in vivo was further supported by

    showing no changes of brain mitochondrial complex I ac-

    tivity in the DPI-treated mice (Fig. 5).

    Another critical issue was whether ultra-low dose DPI

    could generate toxicity in vivo. To examine this, we con-

    ducted a standard pathological evaluation based on the

    modied National Toxicology Program/National Institutes

    of Health protocol. This method measures the weight of the

    body and organs, behavioural activity, and performs histo-

    logical assessment of organs tissues to verify the safety

    prole of putative toxicants. Although these methods

    cannot rule out subtle toxicities, no gross toxicological ef-

    fects were noted. Furthermore, as we know that patients

    with chronic granulomatous disease (CGD), a rare muta-

    tion on NADPH oxidase that render the subunit functional

    inactive, and NADPH oxidase decient mice display im-

    munodeciency, we conrmed that ultra-low dose DPI

    did not affect the peripheral immune cell functions in

    both mice (Supplementary Fig. 6) and human (data not

    shown) that are critical for hosting normal immune

    responses. Finally, it is important to point out that al-

    though we provided strong evidence for the potential clin-

    ical usage of ultra-low dose DPI in Parkinsons disease,

    there still have several issues to consider before translating

    our ndings. First, in our study, the DPI infusions are given

    when about 3035% of dopaminergic neurons are lost. In

    humans, the degree of nigral dopaminergic neuron loss is

    much greater (about 5060% loss) (Hirsch, 2007) than this

    when the patients rst show motor symptoms. It remains to

    be determined whether DPI treatment is still effective in

    Parkinsons disease patients, who already display motor

    symptoms. Second, our study showed no overt toxicity in

    low-dose DPI-treated mice; however, a more detailed evalu-

    ation for the toxicity of low-dose DPI in monkeys or even

    human is needed. Third, although neuroinammation has

    been recognized as one of the critical factors that contribute

    to the progression of Parkinsons disease, the complexity of

    disease aetiology makes it difcult to predict whether DPI

    could work as effectively in human Parkinsons disease

    patients.

    In summary, this study provides convincing evidence that

    subchronic infusion of an ultra-low dose of DPI potently

    reduced microglia-mediated chronic neuroinammation by

    selectively inhibiting NADPH oxidase and halted progres-

    sive neurodegeneration in both LPS and MPTP models.

    Our ndings may provide a novel and efcient therapeutic

    strategy for future Parkinsons disease therapy. The ability

    to halt progressive neurodegeneration, selective specicity

    in inhibiting NADPH oxidase and initial safety proles sug-

    gest that ultra-low dose DPI could be a promising candi-

    date for future clinical trials in patients with Parkinsons

    disease.

    FundingThis research was supported by the Intramural Research

    Program of the NIH, National Institute of Environmental

    Health Sciences

    Supplementary materialSupplementary material is available at Brain online.

    ReferencesAldieri E, Riganti C, Polimeni M, Gazzano E, Lussiana C, Campia I,

    et al. Classical inhibitors of NOX NAD(P)H oxidases are not

    specic. Curr Drug Metab 2008; 9: 68696.

    Becker C, Jick SS, Meier CR. NSAID use and risk of Parkinson dis-

    ease: a population-based case-control study. Eur J Neurol 2011; 18:

    133642.

    1260 | BRAIN 2015: 138; 12471262 Q. Wang et al.

    by guest on June 16, 2015D

    ownloaded from

  • Block ML, Hong JS. Microglia and inammation-mediated neurode-

    generation: multiple triggers with a common mechanism. Prog

    Neurobiol 2005; 76: 7798.

    Block ML, Zecca L, Hong JS. Microglia-mediated neurotoxicity: un-

    covering the molecular mechanisms. Nat Rev Neurosci 2007; 8:

    5769.

    Brichta L, Greengard P, Flajolet M. Advances in the pharmacological

    treatment of Parkinsons disease: targeting neurotransmitter systems.

    Trends Neurosci 2013; 36: 54354.Cannon JR, Greenamyre JT. Neurotoxic in vivo models of Parkinsons

    disease recent advances. Prog Brain Res 2010; 184: 1733.Chen H, Jacobs E, Schwarzschild MA, McCullough ML, Calle EE,

    Thun MJ, et al. Nonsteroidal antiinammatory drug use and the

    risk for Parkinsons disease. Ann Neurol 2005; 58: 9637.

    Czirr E, Wyss-Coray T. The immunology of neurodegeneration. J Clin

    Invest 2012; 122: 115663.

    Gao HM, Hong JS. Why neurodegenerative diseases are progressive:

    uncontrolled inammation drives disease progression. Trends

    Immunol 2008; 29: 35765.

    Gao HM, Jiang J, Wilson B, Zhang W, Hong JS, Liu B. Microglial

    activation-mediated delayed and progressive degeneration of rat

    nigral dopaminergic neurons: relevance to Parkinsons disease. J

    Neurochem 2002; 81: 128597.Gao HM, Liu B, Zhang W, Hong JS. Critical role of microglial

    NADPH oxidase-derived free radicals in the in vitro MPTP modelof Parkinsons disease. FASEB J 2003; 17: 19546.

    Gao HM, Zhang F, Zhou H, Kam W, Wilson B, Hong JS.

    Neuroinammation and alpha-synuclein dysfunction potentiate

    each other, driving chronic progression of neurodegeneration in a

    mouse model of Parkinsons disease. Environ Health Perspect 2011;

    119: 80714.Gao HM, Zhou H, Hong JS. NADPH oxidases: novel therapeutic

    targets for neurodegenerative diseases. Trends Pharmacol Sci 2012;

    33: 295303.

    Gao HM, Zhou H, Zhang F, Wilson BC, Kam W, Hong JS. HMGB1

    acts on microglia Mac1 to mediate chronic neuroinammation that

    drives progressive neurodegeneration. J Neurosci 2011; 31:

    108192.

    Gatley SJ, Martin JL. Some aspects of the pharmacology of dipheny-

    leneiodonium, a bivalent iodine compound. Xenobiotica 1979; 9:

    53946.Giasson BI, Duda JE, Quinn SM, Zhang B, Trojanowski JQ, Lee VM.

    Neuronal alpha-synucleinopathy with severe movement disorder in

    mice expressing A53T human alpha-synuclein. Neuron 2002; 34:

    52133.

    Hart RG, Pearce LA, Ravina BM, Yaltho TC, Marler JR.

    Neuroprotection trials in Parkinsons disease: systematic review.

    Mov Disord 2009; 24: 64754.

    Hirsch EC. How to improve neuroprotection in Parkinsons disease?

    Parkinsonism Relat Disord 2007; 13 (Suppl 3): S3325.

    Hu X, Zhang D, Pang H, Caudle WM, Li Y, Gao H, et al.

    Macrophage antigen complex-1 mediates reactive microgliosis and

    progressive dopaminergic neurodegeneration in the MPTP model of

    Parkinsons disease. J Immunol 2008; 181: 7194204.

    Lambeth JD. NOX enzymes and the biology of reactive oxygen. Nat

    Rev Immunol 2004; 4: 1819.

    Lambeth JD, Krause KH, Clark RA. NOX enzymes as novel

    targets for drug development. Semin Immunopathol 2008; 30:

    33963.

    Lee VM, Trojanowski JQ. Mechanisms of Parkinsons disease linked

    to pathological alpha-synuclein: new targets for drug discovery.

    Neuron 2006; 52: 338.

    Levesque S, Wilson B, Gregoria V, Thorpe LB, Dallas S, Polikov VS,

    et al. Reactive microgliosis: extracellular micro-calpain and micro-

    glia-mediated dopaminergic neurotoxicity. Brain 2010; 133(Pt 3):

    80821.

    Liu Y, Qin L, Wilson B, Wu X, Qian L, Granholm AC, et al.

    Endotoxin induces a delayed loss of TH-IR neurons in substantia

    nigra and motor behavioral decits. Neurotoxicology 2008; 29:

    86470.

    McGeer PL, McGeer EG. NSAIDs and Alzheimer disease: epidemiolo-

    gical, animal model and clinical studies. Neurobiol Aging 2007; 28:

    63947.

    Miesel R, Kurpisz M, Kroger H. Suppression of inammatory arthritis

    by simultaneous inhibition of nitric oxide synthase and NADPH

    oxidase. Free Radic Biol Med 1996; 20: 7581.

    Olanow CW, Tatton WG. Etiology and pathogenesis of Parkinsons

    disease. Annu Rev Neurosci 1999; 22: 12344.

    Perry VH, Nicoll JA, Holmes C. Microglia in neurodegenerative dis-

    ease. Nat Rev Neurol 2010; 6: 193201.

    Phani S, Loike JD, Przedborski S. Neurodegeneration and inamma-

    tion in Parkinsons disease. Parkinsonism Relat Disord 2012; 18

    (Suppl 1): S2079.

    Qin L, Liu Y, Hong JS, Crews FT. NADPH oxidase and aging drive

    microglial activation, oxidative stress, and dopaminergic neurode-

    generation following systemic LPS administration. Glia 2013; 61:

    85568.

    Qin L, Liu Y, Wang T, Wei SJ, Block ML, Wilson B, et al. NADPH

    oxidase mediates lipopolysaccharide-induced neurotoxicity and

    proinammatory gene expression in activated microglia. J Biol

    Chem 2004; 279: 141521.

    Qin L, Wu X, Block ML, Liu Y, Breese GR, Hong JS, et al. Systemic

    LPS causes chronic neuroinammation and progressive neurodegen-

    eration. Glia 2007; 55: 45362.

    Quick KL, Dugan LL. Superoxide stress identies neurons at

    risk in a model of ataxia-telangiectasia. Ann Neurol 2001; 49:

    62735.Rao DB, Little PB, Sills RC. Subsite awareness in neuropathology

    evaluation of national toxicology program (ntp) studies: a review

    of select neuroanatomical structures with their functional signi-

    cance in rodents. Toxicol Pathol 2013; 42: 487509.

    Salawu F, Olokoba A, Danburam A. Current management of

    Parkinsons disease. Ann Afr Med 2010; 9: 5561.

    Samii A, Etminan M, Wiens MO, Jafari S. NSAID use and the risk of

    Parkinsons disease: systematic review and meta-analysis of observa-

    tional studies. Drugs Aging 2009; 26: 76979.

    Sato K, Kadiiska MB, Ghio AJ, Corbett J, Fann YC, Holland SM,

    et al. In vivo lipid-derived free radical formation by NADPH oxi-dase in acute lung injury induced by lipopolysaccharide: a model for

    ARDS. FASEB J 2002; 16: 171320.Streit WJ. Microglial response to brain injury: a brief synopsis. Toxicol

    Pathol 2000; 28: 2830.

    Teismann P, Ferger B. Inhibition of the cyclooxygenase isoenzymes

    COX-1 and COX-2 provide neuroprotection in the MPTP-mouse

    model of Parkinsons disease. Synapse 2001; 39: 16774.Vlessis AA, Bartos D, Muller P, Trunkey DD. Role of reactive O2 in

    phagocyte-induced hypermetabolism and pulmonary injury. J Appl

    Physiol (1985) 1995; 78: 1126.

    Wang Q, Chu CH, Oyarzabal E, Jiang L, Chen SH, Wilson B, et al.

    Subpicomolar diphenyleneiodonium inhibits microglial NADPH

    oxidase with high specicity and shows great potential as a thera-

    peutic agent for neurodegenerative diseases. Glia 2014a; 62:

    203443.

    Wang Q, Chu CH, Qian L, Chen SH, Wilson B, Oyarzabal E, et al.

    Substance P exacerbates dopaminergic neurodegeneration through

    neurokinin-1 receptor-independent activation of microglial

    NADPH oxidase. J Neurosci 2014b; 34: 12490503.

    Wang Q, Shin EJ, Nguyen XK, Li Q, Bach JH, Bing G, et al.

    Endogenous dynorphin protects against neurotoxin-elicited nigros-

    triatal dopaminergic neuron damage and motor decits in mice.

    J Neuroinammation 2012; 9: 124.

    Wu DC, Teismann P, Tieu K, Vila M, Jackson-Lewis V,

    Ischiropoulos H, et al. NADPH oxidase mediates oxidative

    stress in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine model

    of Parkinsons disease. Proc Natl Acad Sci USA 2003; 100:

    614550.

    DPI attenuates PD progression BRAIN 2015: 138; 12471262 | 1261

    by guest on June 16, 2015D

    ownloaded from

  • Xu K, Xu YH, Chen JF, Schwarzschild MA. Neuroprotection by caf-feine: time course and role of its metabolites in the MPTP model of

    Parkinsons disease. Neurosci 2010; 167: 47581.

    Zhang D, Anantharam V, Kanthasamy A, Kanthasamy AG.

    Neuroprotective effect of protein kinase C delta inhibitor rottlerinin cell culture and animal models of Parkinsons disease.

    J Pharmacol Exp Ther 2007; 322: 91322.

    Zhang W, Wang T, Qin L, Gao HM, Wilson B, Ali SF, et al.

    Neuroprotective effect of dextromethorphan in the MPTP

    Parkinsons disease model: role of NADPH oxidase. FASEB J2004; 18: 58991.

    Zigmond MJ. Do compensatory processes underlie the preclinical

    phase of neurodegenerative disease? Insights from an animal

    model of parkinsonism. Neurobiol Dis 1997; 4: 24753.Zigmond MJ, Abercrombie ED, Berger TW, Grace AA, Stricker EM.

    Compensations after lesions of central dopaminergic neurons:

    some clinical and basic implications. Trends Neurosci 1990; 13:

    2906.

    1262 | BRAIN 2015: 138; 12471262 Q. Wang et al.

    by guest on June 16, 2015D

    ownloaded from


Recommended