Manganese exposure induces neuroinflammation by impairing mitochondrial dynamics in astrocytes

Manganese exposure induces neuroinflammation by impairing mitochondrial dynamics in astrocytes

Accepted Manuscript Title: Manganese exposure induces neuroinflammation by impairing mitochondrial dynamics in astrocytes Author: Souvarish Sarkar Emi...

882KB Sizes 0 Downloads 66 Views

Accepted Manuscript Title: Manganese exposure induces neuroinflammation by impairing mitochondrial dynamics in astrocytes Author: Souvarish Sarkar Emir Malovic Dilshan S. Harischandra Hilary A. Ngwa Anamitra Ghosh Colleen Hogan Dharmin Rokad Gary Zenitsky Huajun Jin Vellareddy Anantharam Anumantha G. Kanthasamy Arthi Kanthasamy PII: DOI: Reference:

S0161-813X(17)30094-3 http://dx.doi.org/doi:10.1016/j.neuro.2017.05.009 NEUTOX 2177

To appear in:

NEUTOX

Received date: Revised date: Accepted date:

31-3-2017 19-5-2017 19-5-2017

Please cite this article as: Sarkar S, Malovic E, Harischandra DS, Ngwa HA, Ghosh A, Hogan C, Rokad D, Zenitsky G, Jin H, Anantharam V, Kanthasamy AG, Kanthasamy A, Manganese exposure induces neuroinflammation by impairing mitochondrial dynamics in astrocytes, Neurotoxicology (2017), http://dx.doi.org/10.1016/j.neuro.2017.05.009 This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

1

Manganese exposure induces neuroinflammation by impairing mitochondrial

2

dynamics in astrocytes

3

Souvarish Sarkar#, Emir Malovic#, Dilshan S. Harischandra, Hilary A. Ngwa, Anamitra

5

Ghosh, Colleen Hogan, Dharmin Rokad, Gary Zenitsky, Huajun Jin, Vellareddy

6

Anantharam, Anumantha G. Kanthasamy, Arthi Kanthasamy*

cr

ip t

4

us

7

Parkinson Disorders Research Laboratory, Iowa Center for Advanced Neurotoxicology,

9

Department of Biomedical Sciences, 2062 Veterinary Medicine Building, Iowa State University, Ames, IA 50011.

M

10

an

8

11

14

d

13

#These authors made equal contributions

te

12

*To whom correspondence should be addressed: Arthi Kanthasamy, Ph.D. Professor,

16

Department of Biomedical Sciences, Iowa State University, Ames, IA 50011,

17

Telephone: (515) 294-2516; Fax: (515) 294-2315, Email: [email protected]

Ac ce p

15

1 Page 1 of 49

Abstract

2

Chronic manganese (Mn) exposure induces neurotoxicity, which is characterized by

3

Parkinsonian symptoms resulting from impairment in the extrapyramidal motor system

4

of the basal ganglia. Mitochondrial dysfunction and oxidative stress are considered key

5

pathophysiological features of Mn neurotoxicity. Recent evidence suggests astrocytes

6

as a major target of Mn neurotoxicity since Mn accumulates predominantly in

7

astrocytes. However, the primary mechanisms underlying Mn-induced astroglial

8

dysfunction and its role in metal neurotoxicity are not completely understood. In this

9

study, we examined the interrelationship between mitochondrial dysfunction and

10

astrocytic inflammation in Mn neurotoxicity. We first evaluated whether Mn exposure

11

alters mitochondrial bioenergetics in cultured astrocytes. Metabolic activity assessed by

12

MTS assay revealed an IC50 of 92.68 µM Mn at 24 h in primary mouse astrocytes

13

(PMAs) and 50.46 µM in the human astrocytic U373 cell line. Mn treatment reduced

14

mitochondrial mass, indicative of impaired mitochondrial function and biogenesis, which

15

was substantiated by the significant reduction in mRNA of mitofusin-2, a protein that

16

serves as a ubiquitination target for mitophagy. Furthermore, Mn increased

17

mitochondrial circularity indicating augmented mitochondrial fission. Seahorse analysis

18

of bioenergetics status in Mn-treated astrocytes revealed that Mn significantly impaired

19

the basal mitochondrial oxygen consumption rate as well as the ATP-linked respiration

20

rate. The effect of Mn on mitochondrial energy deficits was further supported by a

21

reduction in ATP production. Mn-exposed primary astrocytes also exhibited a severely

22

quiescent energy phenotype, which was substantiated by the inability of oligomycin to

23

increase the extracellular acidification rate. Since astrocytes regulate immune functions

Ac ce p

te

d

M

an

us

cr

ip t

1

2 Page 2 of 49

in the CNS, we also evaluated whether Mn modulates astrocytic inflammation. Mn

2

exposure in astrocytes not only stimulated the release of proinflammatory cytokines, but

3

also exacerbated the inflammatory response induced by aggregated α-synuclein. The

4

novel mitochondria-targeted antioxidant, mito-apocynin, significantly attenuated Mn-

5

induced inflammatory gene expression, further supporting the role of mitochondria

6

dysfunction and oxidative stress in mediating astrogliosis. Lastly, intranasal delivery of

7

Mn in vivo elevated GFAP and depressed TH levels in the olfactory bulbs, clearly

8

supporting the involvement of astrocytes in Mn-induced dopaminergic neurotoxicity.

9

Collectively, our study demonstrates that Mn drives proinflammatory events in

cr

us

an

astrocytes by impairing mitochondrial bioenergetics.

M

10

ip t

1

Ac ce p

te

d

11

3 Page 3 of 49

Introduction

2

Central nervous system (CNS) aberrations resulting from prolonged exposure to

3

transition metals such as iron (Febbraro et al., 2012; Galvin et al., 2000; Gregory et al.,

4

2009), lead (Bihaqi and Zawia, 2013), and manganese (Gorojod et al., 2015; Hesketh et

5

al., 2008), lend support to the potential involvement of environmental metal exposure in

6

etiology of chronic neurodegeneration (Rokad et al., 2016). Manganese (Mn) toxicity

7

resulting from overexposure to airborne sources was first noted in miners (Peres et al.,

8

2016; Rodier, 1955), and subsequently in industrial workers like welders (Horning et al.,

9

2015; Kwakye et al., 2015). Although Mn has a multitude of physiological functions

10

(e.g., enzyme cofactor for superoxide dismutase and arginase, immunity, bone

11

development, and reproduction) (Karki et al., 2015), the accruing toxic levels has been

12

shown to predominately localize to the globus pallidus, as well as other brain areas of

13

the extrapyramidal motor system, thereby leading to a condition termed manganism,

14

which is phenotypically similar to PD (Kwakye et al., 2015; Milatovic, D. et al., 2007). In

15

addition, diets including grains, legumes, baby formulations and nuts represent the main

16

source of Mn, of which <5% is intestinally absorbed (Karki et al., 2013; Kwakye et al.,

17

2015). Approximately, 30-40% of inhaled Mn is absorbed into the bloodstream, where it

18

is transported primarily bound to albumin and β-globulin in oxidation state 2 (Mn2+)

19

(Karki et al., 2013); however, the absorbed proportion could be smaller (~10.6%)

20

because of oxidative state-dependent differences in solubility (Ellingsen et al., 2003).

21

Availability of the Mn2+ oxidation state in the central nervous system (CNS) is facilitated

22

by transporters such as divalent metal ion transporter 1 (DMT1) and SLC13A10, while

Ac ce p

te

d

M

an

us

cr

ip t

1

4 Page 4 of 49

1

Mn3+ is facilitated by transferrin (Chen et al., 2015; Karki et al., 2015; Leyva-Illades et

2

al., 2014; Sidoryk-Wegrzynowicz and Aschner, 2013).

3

As a ubiquitous constituent and physiological glue of the CNS, astrocytes are

5

indispensable for cellular homeostatic maintenance (Volterra and Meldolesi, 2005).

6

Astrocytes regulate extracellular glutamate levels among neurons in tripartite synapses

7

(Karki et al., 2013; Popoli et al., 2011). Excessive Mn can disrupt this regulation by

8

inducing tumor necrosis factor-α (TNFα), a secreted proinflammatory cytokine. Previous

9

studies demonstrated that Mn impairs calcium (Ca2+) homeostasis in astrocytes and

10

activates key proinflammatory events such as NFκB, NOS2 and cytokine production

11

(Karki et al., 2015; Karki et al., 2014). This inflammatory response increases expression

12

of yin yang-1, a transcription repressor that inhibits production of excitatory amino acid

13

transporter-2, which is necessary for glutamate reuptake (Karki et al., 2015; Karki et al.,

14

2014). Recent studies also demonstrate that Mn interferes with neurodegenerative

15

disease-specific proteins including prions, alpha-synuclein and huntingtin (Bichell et al.,

16

2017; Choi et al., 2010; Choi et al., 2007; Harischandra et al., 2015), as well as protein

17

misfolding, which may further contribute to neuroinflammation. Thus, Mn exposure can

18

activate the inflammatory response in astrocytes, which can further contribute to Mn

19

neurotoxicity.

Ac ce p

te

d

M

an

us

cr

ip t

4

20 21

Along with pericytes, the end-feet of astrocytes envelope blood vessels to

22

regulate blood-brain barrier trafficking (Pekny and Pekna, 2014). Interestingly,

23

astrocytes have been demonstrated to have an increased affinity for Mn, with

5 Page 5 of 49

concentrations up to 50-fold greater than in neurons (Aschner et al., 1992; Gonzalez et

2

al., 2008). Once internalized, the Mn is sequestered by mitochondria via the Ca2+

3

uniporter (Gunter et al., 2006). Kinetical analyses revealed the influx of Mn through this

4

mitochondrial uniporter is slower than Ca2+ per se, but Mn efflux is far slower (Gavin et

5

al., 1990; Martinez-Finley et al., 2013). Even though the atomic charge properties and

6

size of Mn are quite analogous to Ca2+, the two Ca2+ efflux mechanisms are poor

7

exporters of Mn, thus explaining the mitochondrial sequestration of Mn (Gunter et al.,

8

1975; Gunter and Sheu, 2009; Martinez-Finley et al., 2013). On the other hand,

9

although sodium (Na+)-independent Ca2+ efflux does transport Mn, Na+-dependent Ca2+

10

efflux, the predominant mechanism in CNS mitochondria, has not been reported to

11

export Mn (Gavin et al., 1990; Gunter and Sheu, 2009; Martinez-Finley et al., 2013).

12

This extremely sluggish efflux of Mn potentially explains the prolonged CNS clearance

13

and the buildup of toxicity (Martinez-Finley et al., 2013). Mn can also competitively

14

inhibit both efflux pathways, thereby raising the mitochondrial Ca2+ concentration, which

15

can impair aerobic respiration and elicit reactive oxygen species (ROS) generation

16

(Gavin et al., 1990; Martinez-Finley et al., 2013; Streifel et al., 2013; Tjalkens et al.,

17

2006).

cr

us

an

M

d

te

Ac ce p

18

ip t

1

19

Despite the known effects of Mn on astrocytes, the extent of Mn-induced structural and

20

functional impairment of mitochondrial dynamics and its consequent neuroinflammatory

21

processes as it relates to Mn neurotoxicity have not been systematically examined. In

22

the present study, we characterized the impact of Mn on mitochondrial bioenergetic flux

23

in astrocytes using the Seahorse bioenergetic analyzer and its functional consequence

6 Page 6 of 49

on neuroinflammation. Our results demonstrate that overexposure to Mn i) lowers basal

2

respiration in astrocytes, ii) induces astrocytic inflammation that is not exclusive to

3

TNFα, and iii) potentiates aggregated α-Synuclein (αSynagg)-induced inflammation in

4

astrocytes. Thus, our study establishes a functional interaction between mitochondrial

5

dysfunction and astrocytic inflammation in Mn-induced neurotoxicity. In addition, we

6

report that the mitochondria-targeted anti-oxidant mito-apocynin attenuates Mn-induced

7

astrocyte inflammation.

8

Methods

9

Chemicals and Reagents

an

us

cr

ip t

1

Dulbecco’s modified eagle medium (DMEM), fetal bovine serum (FBS), L-glutamine (Q),

11

and penicillin/streptomycin (P/S) were obtained from Invitrogen (Carlsbad, CA).

12

MitoTracker Green FM and MitoTracker Red CMXRos, were purchased from Molecular

13

Probes (Eugene, OR). Manganese Chloride (MnCl2) was obtained from Sigma-Aldrich

14

(St. Louis, MO). The CellTiter® 961 AQueous Non-Radioactive Cell Proliferation Assay

15

kit and CellTiter® Glo Luminescent Cell Viability Assay kit were obtained from Promega

16

(Madison, WI). The CD11b magnetic separation kit was purchased from Stem Cell

17

Technologies (Vancouver, Canada). Tyrosine hydroxylase (TH) (AB_2201528) and glial

18

fibrillary acidic protein (GFAP) (AB_2109815) antibodies were purchased from EMD

19

Millipore. Inducible nitric oxide synthase (iNOS) (AB_631831) antibody was purchased

20

from Santa Cruz Biotechnologies (Dallas, TX). Mitofusin-2 (Mfn2) (#11925) was

21

purchased from Cell Signaling Technologies. All the standards used for the Luminex

22

multiplex cytokine assay were purchased from PeproTech Inc (Rocky Hill, NJ).

23

Streptavidin-Biotin and biotinylated antibodies used for Luminex were purchased from

Ac ce p

te

d

M

10

7 Page 7 of 49

eBioSciences (San Diego, CA). MA was obtained from Dr. Kalyanaraman (Medical

2

College of Wisconsin, Milwaukee).

3

Animal Study

ip t

1

Eight-week-old male C57BL/6Ncrl mice, obtained from Charles River, were

5

housed under standard conditions of constant temperature (22 ± 1°C), humidity

6

(relative, 30%), and a 12 h light/dark cycle. After acclimating for 3 days, mice were

7

exposed to 50 µL of 20 mM of Mn (200 µg). This dose is consistent with previously

8

published literature (Moberly et al., 2012). According to reports from Centers for

9

Disease Control and Prevention, the ambient level of Mn near industries is 0.2-0.33

an

us

cr

4

10

mg/m3.

11

(https://www.atsdr.cdc.gov/toxprofiles/tp151-c2.pdf).

12

procedures were approved by the Institutional Animal Care and Use Committee

13

(IACUC) at Iowa State University (Ames, IA, USA). Intranasal delivery was preferred

14

because it takes advantage of an incomplete blood-brain barrier in the olfactory

15

epithelium. The olfactory nerves can completely bypass the blood-brain barrier, thus

16

chemicals can be taken up by these neurons and transported directly into the brain

17

(Graff and Pollack, 2005). Also, environmental exposure is usually occupation-related

18

inhalation. Following the treatments, we carried out behavioral, biochemical and

19

neurochemical studies.

20

Cell culture and Treatments

21

One-day-old C57BL/6Ncrl pups were sacrificed, their brains dissected out, and a single

22

cell suspension was prepared. After growing in culture for 16 days, the astrocytes were

the

dose

used

is

environmentally

relevant

Use of the animals and protocol

Ac ce p

te

d

M

Hence

8 Page 8 of 49

isolated after microglia were removed using our CD11b magnetic-bead separation

2

technique (Gordon et al., 2011; Sarkar et al., 2017). The human astrocytic U373 cell

3

line was obtained from ATCC and grown per ATCC protocol. Primary mouse astrocytes

4

(PMAs) were cultured in DMEM, 10% FBS, 1% Q, 1% P/S. Treatments for PMAs were

5

performed in 2% FBS-containing DMEM, with 1% Q and P/S. U373s were cultured in

6

MEM, 10% FBS, 1% Q, and 1% P/S. Treatments for U373s were performed in 2%-FBS-

7

containing MEM, with 1% Q and P/S. All treatments were for 24 h using Mn at 100 µM,

8

αSynagg at 1 µM, and mito-apocynin at 10 µM.

9

Recombinant human α-synuclein purification and aggregation

an

us

cr

ip t

1

BL21(DE3) strain E. coli cells were transformed using a plasmid encoding for wild-type

11

human α-synuclein and grown on agar plate with Ampicillin (Amp) resistance. Pre-

12

culture was prepared by inoculating a single colony from the agar plate into a tube

13

containing 10 mL of LB broth with Amp and incubated overnight at 37 °C. The following

14

day, the pre-culture was inoculated into 1 L of LB medium containing Amp, and OD600

15

was taken every hour till the culture reached an OD of 0.5. Recombinant α-synuclein

16

expression was induced by adding 1 mM isopropyl β-D-1-thiogalactopyranoside (IPTG)

17

(Invitrogen), and the cells were further incubated at 37 °C for 8 h before harvesting.

18

Cells were lysed and recombinant α-synuclein was purified as previously described

19

(Giasson et al., 1999; Narhi et al., 1999). Finally, protein was lyophilized and stored at -

20

80 °C. For α-synuclein aggregation, a 70-µM recombinant protein solution was prepared

21

by dissolving 1 mg in 1 mL of ultrapure water and shaken at a speed of 1000 rpm at 37

22

°C for 7 days (Luk et al., 2012b).

23

qRT-PCR

Ac ce p

te

d

M

10

9 Page 9 of 49

RNA extraction and qRT-PCR were performed as described previously(Gordon, R. et

2

al., 2016a; Seo et al., 2014). Total RNA was extracted using TRIZOL reagent as per the

3

manufacturers’ protocol and concentration was measured using NanoDrop. First strand

4

cDNA synthesis was performed using an Affinity Script qPCR cDNA synthesis system

5

(Agilent Technologies). Real-time PCR was performed with the RT2 SYBR Green

6

master mix (Thermo-Fisher #K0172). The following genes from QuantiTect Primer

7

Assay (Qiagen) were used for qRT-PCR: pro-IL-1β, pro-IL-18, CSF-2, IL-12b, and IL-6.

8

The house keeping gene 18S rRNA (Qiagen #PPM57735E) was used as the reference

9

for all qRT-PCR experiments. The dissociation curves were run to ensure a single

10

amplicon peak was obtained. The results are reported as fold change in gene

11

expression, which was determined via the ∆∆Ct method using the threshold cycle (Ct)

12

value for the housekeeping gene and for the respective gene of interest in each sample

13

(Gordon, Richard et al., 2016; Lawana et al., 2017).

14

Western Blotting

15

Western blot analysis was performed according to previous published protocols

16

(Harischandra et al., 2017; Kanthasamy et al., 2012). Briefly, tissue/cell samples were

17

homogenized using a tissue homogenizer in RIPA buffer. Following protein isolation, the

18

Bradford assay was performed for protein estimation. Next, 25-40 µg of protein was

19

loaded in each well of 10-15% SDS-acrylamide gels and ran for 2 h at 110 V. After

20

running, proteins were transferred to a nitrocellulose membrane at 27 V for 18 h at 4°C.

21

After transfer, the membranes were blocked using LI-COR blocking buffer for 1 h at RT.

22

Following blocking, the membranes were incubated in primary antibodies for 3 to 18 h,

23

then washed with PBS-Tween (0.01%), incubated in infrared LI-COR secondary

Ac ce p

te

d

M

an

us

cr

ip t

1

10 Page 10 of 49

antibodies for 1 h, then washed again with PBS-Tween, and scanned using LI-COR

2

scanner. The following antibodies were used: GFAP (1:1000), TH (1:1000), and iNOS

3

(1:500). Secondary antibodies were used according to manufacturer’s instructions.

4

Luminex Multiplex Cytokine Analysis

5

Cytokine levels were assessed via Luminex cytokine assay according to our previous

6

publications (Gordon, R. et al., 2016b; Panicker et al., 2015). Briefly, PMAs were treated

7

in 96-well plates (20,000 cells/well) with 100 µL of treatment medium. Post-treatment,

8

40 µL of the treatment medium was collected and added to 40 µL of primary antibody

9

conjugated to magnetic microspheres and incubated overnight at 4°C in a clear bottom,

10

black 96-well plate. For serum cytokine levels, serum was diluted 5 times with

11

block/store buffer containing BSA (Sigma-Aldrich), and 40 µL of diluted serum was used

12

as the sample. After incubation, each well was triple-washed using a magnetic washer

13

and then incubated for 1 h with secondary antibodies followed by three more washes.

14

Lastly, samples were incubated for 30 min with streptavidin/phycoerythrin followed by

15

another two washes. A Bio-Plex reader was used to read the 96-well plates.

16

MTS Assays

17

MTS assay was performed according to our previous publications (Brenza et al., 2016;

18

Charli et al., 2016). Briefly, 20,000 cells/well were plated in a 96-well tissue culture plate

19

and exposed to 100 µL of treatment medium after attachment. Post-treatment, 10 µL of

20

MTS dye was added to each well and incubated for 45 min at 37°C. After incubation,

21

absorbance readings were taken using a plate reader at 490 nm, and a 640-nm readout

22

was used for background subtraction.

Ac ce p

te

d

M

an

us

cr

ip t

1

11 Page 11 of 49

Seahorse Mito-Stress Analysis

2

A Seahorse XFe24 Analyzer was used to measure mitochondrial extracellular

3

acidification rates (ECAR) and oxygen consumption rates (OCR) using the Mito-Stress

4

test following a previously published protocol (Charli et al., 2016). PMAs were plated at

5

20,000 cells/well of a Seahorse plate. The calibration plate was hydrated overnight in a

6

non-CO2 incubator. For the Mito-Stress test, 0.75 µM oligomycin, 1 µM FCCP, and 0.5

7

µM rotenone/antimycin were used. The wave report generator (Agilent) and cell-

8

phenotype report generator were used for analysis.

9

MitoTracker Green FM and MitoTracker Red CMXRos

an

us

cr

ip t

1

MitoTracker Green FM and MitoTracker Red CMXRos were obtained from Molecular

11

Probes and used according to our previous publications (Charli et al., 2016; Gordon,

12

Richard et al., 2016). After treatment, cells were washed and incubated in MitoTracker

13

green FM according to the concentration suggested by the manufacturer for 10-20 min

14

in HBSS, followed by washing with HBSS. Hoechst nuclear stain was used to normalize

15

the results. Excitation and emission readings were taken using a plate reader with 490

16

and 516 nm, respectfully, for MitoTracker green FM and 350 and 461 nm for Hoechst.

17

For the MitoTracker Red CMXRos assay, post-treatment cells were washed in fresh

18

media and incubated in MitoTracker red dye (200 nM) for 15 min. Following incubation,

19

cells were washed twice, fixed in 4% paraformaldehyde, mounted on slides, dried

20

overnight, and imaged using a Nikon Eclipse C1 microscope equipped with a SPOT

21

RT3 digital camera (Diagnostic Instruments, Sterling Heights, MI).

Ac ce p

te

d

M

10

22

12 Page 12 of 49

ATP Assay

2

ATP assay was performed following Charli et al. (2016) with minor modifications. Briefly,

3

20,000 cells/well were plated in an opaque 96-well plate and exposed to 100 µL of

4

treatment medium after attachment. After treatment, 100 µL of CellTiter-Glo

5

Luminescent reagent from Promega was added to each well containing 100 µL of

6

media. The samples were placed on an orbital shaker for 3 min after which they were

7

incubated for 10 min at RT. Readings were taken using a luminometer (BioTek).

8

Background luminescence was determined using the corresponding cell-free treatment

9

media and the average was subtracted from the sample values.

an

us

cr

ip t

1

HPLC analysis

11

HPLC analysis of dopamine was performed according to our previous publication

12

(Gordon, Richard et al., 2016). In brief, olfactory bulb tissues were extracted using an

13

antioxidant extraction solution (0.1 M perchloric acid containing 0.05% Na2EDTA and

14

0.1% Na2S2O5) and isoproterenol (internal standard). A reversed-phase C-18 column

15

was used to isocratically separate dopamine, 3,4-dihydroxyphenylacetic acid (DOPAC),

16

and homovanillic acid (HVA) at a flow rate of 0.6 ml/min using a Dionex Ultimate 3000

17

HPLC system (pump ISO-3100SD, Thermo Scientific, Bannockburn, IL) equipped with a

18

refrigerated automatic sampler (model WPS-3000TSL). The electrochemical detection

19

system included a CoulArray model 5600A coupled with an analytical cell (microdialysis

20

cell 5014B) and a guard cell (model 5020) with potentials set at 350, 0, -150, and 220

21

mV. Data acquisition and analysis were performed using Chromeleon 7 and ESA

22

CoulArray 3.10 HPLC Software.

Ac ce p

te

d

M

10

13 Page 13 of 49

Behavior Analysis

2

The automated VersaMax Monitor (model RXYZCM-16, AccuScan, Columbus, OH) was

3

used to measure the spontaneous open-field locomotor activity of mice in an activity

4

chamber made of clear Plexiglas and covered with a ventilated Plexiglas lid, as

5

described previously (Ghosh et al., 2013; Ngwa et al., 2014). Horizontal and vertical

6

activity data were collected and analyzed by a VersaMax Analyzer (model CDA-8,

7

AccuScan). Locomotor activities were monitored during a 10-min test session following

8

a 2-min acclimation period.

9

Data Analysis

an

us

cr

ip t

1

GraphPad 5.0 was used for statistical analysis with p≤0.05 considered statistically

11

significant. One-way ANOVA was used for comparison among multiple groups. In most

12

cases, Tukey post analysis was applied. For comparing 2 groups, Student’s t-test was

13

used.

d

te Ac ce p

14

M

10

15

Results

16

Mn induced a dose-dependent decrease in mitochondrial metabolic activity in

17

astrocytes

18

Mn has been shown to accumulate in mitochondria, block the electron transport chain

19

(ETC), and alter mitochondrial permeability in astrocytes (Hazell, 2002; Rao and

20

Norenberg, 2004). In this experiment, we examined the effect of Mn on metabolic

21

activity in PMA and U373 cells. Both sets of astrocytes were exposed to increasing

14 Page 14 of 49

concentrations of Mn (0.001-1 mM) for 24 h. We observed a dose-dependent decrease

2

in metabolic activity as determined by MTS assay. The IC50 for inhibiting metabolic

3

activity was determined to be 92.7 µM for PMAs (Fig. 1A) and 50.5 µM for U373s

4

(Fig.1B). Thus, we used 100 µM Mn for all subsequent studies. To further validate the

5

effect of Mn on astrocytic mitochondria, both PMAs and U373s were treated with 100

6

µM Mn for 24 h, and then subjected to MitoTracker green, a fluorescent dye that binds

7

to intact mitochondria in live cells irrespective of mitochondrial potential and its intensity

8

indicates mitochondrial health. In this assay, we observed a statistically significant loss

9

of mitochondrial mass, suggesting a loss of mitochondrial function in PMAs (Fig. 2A)

10

and U373s (Fig. 2B) compared to untreated controls. Additionally, we used MitoTracker

11

red dye to verify mitochondrial morphology in Mn treated PMAs (Fig. 2C). Mn exposure

12

was found to increase mitochondrial circularity, indicating augmentation of mitochondrial

13

fission (Scott and Youle, 2010), and it positively correlated with diminished ATP

14

production in PMAs as determined by CellTiter-Glo Luminescent assay (Fig. 2D).

15

Furthermore, qRT-PCR analysis revealed that transcript levels of Mfn2, a gene involved

16

in mitochondrial fusion and linked to PD pathology (Tang et al., 2015), was down-

17

regulated in response to Mn treatment (Fig. 2E). Immunoblot analysis further showed

18

that Mn exposure downregulated Mfn2 protein levels (Fig. 2F). Together, these data

19

suggest that Mn treatment reduces metabolic activity, mitochondrial mass, and

20

dysregulates mitochondrial fission/fusion processes in both mouse and human

21

astrocytes.

22

Altered mitochondrial dynamics and bioenergetics in Mn-treated astrocytes

Ac ce p

te

d

M

an

us

cr

ip t

1

15 Page 15 of 49

Mn is known to alter brain mitochondrial influx/efflux kinetics, which affects energy

2

metabolism (Gavin et al., 1990). Thus, we examined the effects Mn treatment exerts on

3

mitochondrial bioenergetics using the Seahorse XFe24 analyzer as described in our

4

recent publication (Charli et al., 2016). PMAs were treated with 100 µM Mn for 24 h,

5

after which the Mito Stress assay was performed (Fig. 3 and 4). The Seahorse directly

6

measures oxygen consumption rate (OCR) (Fig. 3A), while also monitoring the

7

extracellular acidification rate (ECAR), an indicator of glycolysis (Fig. 4A). Mn exposure

8

led to a statistically significant decrease in basal mitochondrial respiration and in oxygen

9

consumption (Fig. 3A-B) when compared to untreated PMAs. Similarly, Mn exposure

10

also reduced mitochondrial ATP production (Fig. 3A,C) as well as the non-mitochondrial

11

respiration rate (Fig. 4A-B). Thus, Mn not only diminishes astrocytic mitochondrial

12

function, but also markedly reduces glycolytic function. Additionally, cell phenotype

13

analysis revealed that Mn exposure shifts the astrocytes’ bioenergetic phenotype from

14

aerobic to a quiescent state (Fig. 4C). These data collectively suggest that Mn exposure

15

not only lowers the basal respiration rate and ATP production, but also alters the cellular

16

phenotype by lowering glycolytic flux.

17

Upregulation of inflammatory genes in Mn-treated astrocytes

18

Although the role of mitochondrial bioenergetics has been well established in PD

19

models, most of that research focused on neurons. More recent studies have suggested

20

a potential link between neuroinflammation and mitochondrial defects in neuroimmune

21

cells that may contribute to the amplification of inflammatory and neurodegenerative

22

processes (Alfonso-Loeches et al., 2014). To test for a potential link between

23

mitochondrial dysfunction and inflammation in astrocytes, we treated PMAs with 100 µM

Ac ce p

te

d

M

an

us

cr

ip t

1

16 Page 16 of 49

Mn for 24 h. The cells were then subjected to qRT-PCR analyses and the treatment

2

media were analyzed for the release of pro-inflammatory cytokines. As revealed by

3

qRT-PCR, Mn treatment increased mRNA levels of the pro-inflammatory factors IL-12b

4

(Fig. 5A), IL-6 (Fig. 5B), TNFα (Fig. 5C), NOS2 (Fig. 5D), pro-IL-1β (Fig. 5E) and CSF-2

5

(Fig. 5F). Luminex cytokine assay revealed that Mn induced the release of the pro-

6

inflammatory cytokines IL-6 (Fig. 5G), IL-12 (Fig. 5H), and TNFα (Fig. 5I) in the

7

extracellular milieu. These data suggest that Mn can activate astrocytes, leading to the

8

induction and maturation of pro-inflammatory factors.

9

Mn potentiates αSynagg-induced inflammation in astrocytes

an

us

cr

ip t

1

Misfolded αsyn (PARK1,4) has been linked to PD pathogenesis and is one of the major

11

components of Lewy bodies (Kim et al., 2013). In terms of gene-environment interaction

12

between αsyn and Mn, our study showed that prolonged exposure to Mn induces αsyn

13

aggregation (Harischandra et al., 2015). Furthermore, we and others have shown that

14

αSynagg induces inflammation in microglia and astrocytes (Codolo et al., 2013; Gordon,

15

R. et al., 2016b; Kim et al., 2013; Su et al., 2008). Hence, we hypothesized that

16

exposure to an environmental toxicant like Mn in tandem with αSynagg will either have

17

an additive or synergistic effect on inflammation. PMAs were treated with either 100 µM

18

Mn plus 1 µM αSynagg, 100 µM Mn alone or 1 µM αSynagg for 24 h, and then the cells

19

were harvested and treatment media were subjected to qRT-PCR and Luminex assays.

20

As revealed by qRT-PCR analysis, Mn synergistically increased the αSynagg-induced

21

expression of pro-IL-1β (Fig. 6A) and the release of IL-1β (Fig. 6B). Mn had an additive

22

effect on the αSynagg-induced pro-inflammatory factor IL-6 (Fig. 6C) and it potentiated

23

the αSynagg-induced increase in CSF-2 (Fig. 6E). Furthermore, Mn potentiated the

Ac ce p

te

d

M

10

17 Page 17 of 49

αSynagg-induced release of the pro-inflammatory cytokines IL-12 (Fig. 6D) and TNFα

2

(Fig. 6F). Collectively, these data indicate that Mn exposure exacerbates inflammation

3

elicited by αSynagg in astrocytes.

4

Mito-apocynin,

5

attenuated Mn-induced inflammatory genes in human astrocytic culture

6

Our group has recently shown that mito-apocynin, a novel mitochondrially targeted

7

derivative of apocynin, can reduce inflammation and neurodegeneration in an MPTP

8

mouse model of PD by reducing mitochondria-mediated oxidative stress (Ghosh et al.,

9

2016a). If Mn-induced mitochondrial ROS contributes to astrocytic neuroinflammatory

novel

mitochondrially targeted

derivative

of

apocynin,

10

processes,

the

anti-oxidant

mito-apocynin

should

dampen

Mn-induced

11

inflammation in human astrocytic culture. U373s were co-treated with 100 µM Mn and

12

10 µM mito-apocynin for 24 h. Following treatment, cells were collected for mRNA

13

analysis. As revealed by qRT-PCR, mito-apocynin significantly reduced the expression

14

level of the pro-inflammatory factors, pro-IL-1β (Fig. 7B), and pro-IL-18 (Fig. 7C)

15

induced by Mn alone. Mito-apocynin further reduced the expression of CSF-2 (Fig. 7A).

16

Lastly, the MitoTracker green assay revealed that mito-apocynin prevented the Mn-

17

induced reduction of astrocytic mitochondrial mass (Fig. 7D). These data collectively

18

suggest that the mitochondrially targeted anti-oxidant mito-apocynin protects against

19

Mn-induced inflammation in astrocytes by possibly dampening mitochondrial oxidative

20

stress.

21

Intranasal delivery of Mn induces inflammation and TH loss in the olfactory bulbs

22

of mice

Ac ce p

te

d

M

then

an

us

cr

a

ip t

1

18 Page 18 of 49

To validate our findings from primary astrocyte cultures in animal models, we subjected

2

C57BL/6 mice to intranasal doses of 200 µg Mn in 50 µL three times a week for four

3

weeks. At the end of the treatment period, behavioral tests were performed and then

4

OB tissues were collected for neurochemical and biochemical analyses. Mn reduced

5

motor activity, as indicated by the significant reduction in total distance traveled (Fig.

6

8A), horizontal activity (Fig. 8B), and total movement time (Fig. 8C). Furthermore,

7

neurochemical analysis of the olfactory bulb revealed that intranasal delivery of Mn

8

significantly

9

downregulation of TH expression (Fig. 8E). Immunoblot analysis of OB tissues showed

10

that Mn induced the expression of GFAP, an activation marker of astrocytes (Fig. 8F).

11

Additionally, iNOS expression (Fig. 8G) and serum IL-12 (Fig. 8H) were elevated,

12

indicating that Mn elicited a pro-inflammatory response. Although Mn has been shown

13

to accumulate in the striatal region, we did not observe any changes in striatal pro-

14

inflammatory factors or TH levels (data not shown) even after 30 days. Together these

15

data corroborate our in vitro findings, whereby Mn-induced astroglial activation is

16

accompanied by an enhanced pro-inflammatory response.

cr

levels

(Fig.

us

dopamine

8D),

which

coincided

with

a

te

d

M

an

decreased

Ac ce p

17

ip t

1

18

Discussion

19

Neuroinflammation plays a key role in neurodegenerative disorders, including

20

Parkinsonian syndrome (Glass et al., 2010; Golde, 2002; Herrero et al., 2015; Tansey

21

and Goldberg, 2010; Whitton, 2007). Even though the etiology of these diseases is not

22

well established, the consensus is that environment plays an important role in these

23

neurodegenerative disorders. Astrocytes, the most abundant glial cells of the brain, are 19 Page 19 of 49

a recognized source of sustained inflammation upon activation (Phillips et al., 2014).

2

Besides showing how the environmental neurotoxin Mn affects astrocytes, we also

3

demonstrate its role in triggering a synergistic interaction between mitochondrial

4

dysfunction and inflammation. Here, we show that Mn inhibits astrocytic metabolic

5

activity and further contributes to mitochondrial dysfunction at a lower micromolar

6

concentration than reported elsewhere for a 24-h IC50 (Gonzalez et al., 2008; Park and

7

Park, 2010; Yin et al., 2008). For the first time, we demonstrate via the Seahorse

8

bioanalyzer that Mn exposure can change the metabolic phenotype of astrocytes. We

9

also show that Mn exposure increased the release of pro-inflammatory factors from

10

astrocytes and potentiated the release of IL-1β initially induced by αSynagg. Finally, we

11

demonstrate in vivo that intranasal Mn exposure increased astrocytic activation, motor

12

deficits and the loss of TH.

13

Although several studies have suggested that Mn induces mitochondrial dysfunction

14

and impairs glutamate uptake in astrocytes (Milatovic, D. et al., 2007; Rao and

15

Norenberg, 2004), none have linked mitochondrial dysfunction to inflammation. The

16

accumulation of Mn in mitochondria induces mitochondrial dysfunction by increasing

17

oxidative stress and impairing membrane potential (Hazell, 2002; Milatovic, Dejan et al.,

18

2007; Rao and Norenberg, 2004). Our findings further support the role of Mn toxicity in

19

astrocytic and mitochondrial dysfunction. We show Mn-induced mitochondrial

20

dysfunction is associated with decreased mitochondrial mass and reduced expression

21

of the mitochondrial fusion protein Mfn2 (Fig. 2). Mfn2 reduction has been linked to

22

progressive loss of dopaminergic neurons in the nigrostriatal tract (Pham et al., 2012).

23

Furthermore, recent studies have shown that VPS35 (PARK17) modulates Mfn2

Ac ce p

te

d

M

an

us

cr

ip t

1

20 Page 20 of 49

degradation leading to mitochondrial dysfunction in dopaminergic neurons (Tang et al.,

2

2015). On the other hand, Mfn2 overexpression can reduce astrogliosis in vitro (Liu et

3

al., 2014). From the bioenergetic profile, we found that basal oxygen consumption and

4

maximal respiratory capacity were significantly attenuated by Mn, indicating that Mn

5

damages mitochondrial function, as further evidenced by a parallel reduction in ATP

6

levels. Interestingly ECAR, which serves as an index of glycolysis, was reduced in cells

7

exposed to Mn as compared to control cells. A previous study involving U87

8

astrocytoma cells reported a concentration-dependent decrease in the activity of several

9

glycolytic enzymes such as hexokinase, lactate dehydrogenase, and pyruvate kinase

10

(Malthankar et al., 2004). Based on this study, the inhibition we observed may not be a

11

direct effect of Mn, but rather a secondary impairment following the disruption of the

12

electron transport chain and tricarboxylic acid cycle. Our studies raise the possibility that

13

Mn can also inhibit glycolysis, thereby accounting for the pronounced depletion of ATP

14

in Mn-treated cells (Fig. 2), but further studies will be essential to understand the role

15

glycolytic flux under different Mn exposure times. Astrocytes provide neurotrophic and

16

nutrient support to neighboring neurons by producing growth factors and helping to

17

maintain neuronal function, tissue repair, and homeostasis. In contrast, reactive

18

astroglia produce proinflammatory factors, which become toxic to neurons when

19

persistently activated under chronic stress, thus leading to degeneration. The

20

phenotypic switch observed in our study upon exposure to Mn may limit nutrient

21

availability, thereby contributing to neuronal stress.

22

Although microglia are the major inflammatory cells in the brain, recent studies have

23

identified astrocytes to also be a key player driving neuroinflammation and

Ac ce p

te

d

M

an

us

cr

ip t

1

21 Page 21 of 49

neurodegeneration (Colombo and Farina, 2016). A few studies have suggested a

2

possible link between Mn and inflammation in astrocytes (Hazell, 2002; Moreno et al.,

3

2009; Moreno et al., 2008), however, the signaling mechanisms associated with this

4

inflammation remain elusive. In this study, we demonstrate that exposing astrocytes to

5

Mn induced the expression and release of pro-inflammatory factors such as IL-1β,

6

TNFα and IL-6 (Fig. 5). Recently, IL-1β has been linked to inflammasome signaling,

7

which has been implicated in various neurodegenerative disorders (Freeman and Ting,

8

2016). In particular, the multi-protein NLRC4 inflammasome complex has been

9

implicated in the inflammatory signaling process in astrocytes (Liu and Chan, 2014). We

10

are currently investigating the role of these inflammasomes in the Mn-induced

11

inflammatory cascade in astrocytes.

12

Aggregated α-synuclein is the key constituent of Lewy bodies, and has been shown to

13

be secreted from stressed or dying neurons. Microglia cells can be activated by

14

secreted αSynagg, thereby inducing inflammation in PD (Alvarez-Erviti et al., 2011; Kim

15

et al., 2013; Lee et al., 2010; Lotharius and Brundin, 2002; Luk et al., 2012a; Su et al.,

16

2008). Our group has previously demonstrated that long-term Mn exposure induces

17

αSyn aggregation leading to neuronal death (Harischandra et al., 2015). We recently

18

demonstrated

19

promote aggregation (Yen et al., 2016). In this study, we demonstrate for the first time

20

that Mn can also potentiate an αSynagg-induced inflammatory response in astrocytes,

21

further fueling the possibility of a gene-environment interaction driving inflammation and

22

neurodegeneration (Fig. 6).

Ac ce p

te

d

M

an

us

cr

ip t

1

that other divalent metals like copper interact with prion proteins to

22 Page 22 of 49

The role of ROS generation in inflammation and neurodegenerative disorders has been

2

well documented (Amor et al., 2010; Mittal et al., 2014). Furthermore, oxidative stress

3

has been linked to Mn-induced neurotoxicity (Chen and Liao, 2002). In fact, in a recent

4

report using antioxidants, resistance was conferred against neurotoxicity (Ghosh et al.,

5

2016b) by reducing inflammation and oxidative stress in a neurotoxin model of PD. We

6

also recently demonstrated that mito-apocynin, a mitochondrially targeted derivative of

7

apocynin, can reduce inflammation in the MPTP mouse model of PD (Ghosh et al.,

8

2016a). We have since demonstrated that mito-apocynin reduces mitochondrial

9

superoxide generation in neuronal cells as well as oxidative damage in MitoPark mice, a

10

mitochondrially defective animal model of PD (Langley et al., 2017). In this study, we

11

demonstrate that mito-apocynin attenuated the Mn-induced inflammatory response in

12

astrocytes by mitigating Mn’s effect on mitochondrial mass (Fig. 7). This further confirms

13

that mitochondrial damage can directly augment inflammation in astroglial cells.

14

Intranasal delivery of Mn can cause deficits in both spatial memory and motor function

15

(Blecharz-Klin et al., 2012). Likewise, in our study, intranasal delivery of Mn to C57BL/6

16

mice induced significant deficits in motor behavior. Our Mn treatment paradigm also

17

depleted TH in the olfactory bulb (OB) region of the brain and increased expression of

18

the astrocyte activation marker GFAP. In contrast, Dorman et al. (2004) did not observe

19

upregulation of GFAP in the rat OB after 90 days of Mn exposure. This discrepancy may

20

be attributed to species differences (mice vs. rats) and application of different Mn

21

compounds (MnCl2 vs. MnSO4). Our studies also reveal that Mn increased the levels of

22

pro-inflammatory cytokines (Fig. 8). Mn is known to accumulate in the striatum, but with

23

this acute treatment paradigm, we did not observe any changes in striatal dopamine or

Ac ce p

te

d

M

an

us

cr

ip t

1

23 Page 23 of 49

1

GFAP. Longer chronic studies are required to determine the relationship between

2

dopamine and GFAP levels in the striatum of Mn-treated mice. Our study collectively

3

demonstrates that Mn can 1) induce mitochondrial damage by altering mitochondrial

4

dynamics

5

proinflammatory factors in astrocytes, 3) potentiate αSynagg-induced inflammation in

6

astrocytes, further validating the gene-environment relationship, and 4) induce astrocyte

7

activation and TH loss in the OB, leading to OB inflammation and decreased motor

8

activity when administered intranasally in vivo. Our novel findings provide an important

9

insight into how the environmental toxicant Mn modulates inflammation and metabolic

10

activity in astrocytes. Future studies should explore the variable mechanistic basis of

11

astrocyte-mediated neuroinflammation in Mn neurotoxicity.

12

Conflict of interest

13

A. G.K. and V.A. are shareholders of PK Biosciences Corporation (Ames, IA), which is

14

interested in identifying novel biomarkers and potential therapeutic targets for PD.

15

A.G.K and V.A Do not have any direct interest in the work presented in present work.

16

Acknowledgements

17

This work was supported by National Institutes of Health (NIH) Grants: ES026892 and

18

NS088206. The authors would like to thank Dr. Balaraman Kalyanaraman for providing

19

access to Mito-apocynin.

20

acknowledged.

states

in

astrocytes,

2)

increase

production

ip t

bioenergetic

of

Ac ce p

te

d

M

an

us

cr

and

Lloyds chair to AGK and Salsbury Chair to AK are also

21

24 Page 24 of 49

References

2

Alfonso-Loeches, S., Urena-Peralta, J.R., Morillo-Bargues, M.J., Oliver-De La Cruz, J.,

3

Guerri, C., 2014. Role of mitochondria ROS generation in ethanol-induced NLRP3

4

inflammasome activation and cell death in astroglial cells. Front Cell Neurosci 8, 216.

5

Alvarez-Erviti, L., Couch, Y., Richardson, J., Cooper, J.M., Wood, M.J., 2011. Alpha-

6

synuclein release by neurons activates the inflammatory response in a microglial cell

7

line. Neurosci Res 69(4), 337-342.

8

Amor, S., Puentes, F., Baker, D., van der Valk, P., 2010. Inflammation in

9

neurodegenerative diseases. Immunology 129(2), 154-169.

an

us

cr

ip t

1

Aschner, M., Gannon, M., Kimelberg, H.K., 1992. Manganese uptake and efflux in

11

cultured rat astrocytes. J Neurochem 58(2), 730-735.

12

Bichell, T.J., Wegrzynowicz, M., Grace Tipps, K., Bradley, E.M., Uhouse, M.A., Bryan,

13

M., Horning, K., Fisher, N., Dudek, K., Halbesma, T., Umashanker, P., Stubbs, A.D.,

14

Holt, H.K., Kwakye, G.F., Tidball, A.M., Colbran, R.J., Aschner, M., Diana Neely, M., Di

15

Pardo, A., Maglione, V., Osmand, A., Bowman, A.B., 2017. Reduced bioavailable

16

manganese causes striatal urea cycle pathology in Huntington's disease mouse model.

17

Biochim Biophys Acta.

18

Bihaqi, S.W., Zawia, N.H., 2013. Enhanced taupathy and AD-like pathology in aged

19

primate brains decades after infantile exposure to lead (Pb). Neurotoxicology 39, 95-

20

101.

21

Blecharz-Klin, K., Piechal, A., Joniec-Maciejak, I., Pyrzanowska, J., Widy-Tyszkiewicz,

22

E., 2012. Effect of intranasal manganese administration on neurotransmission and

23

spatial learning in rats. Toxicol Appl Pharmacol 265(1), 1-9.

Ac ce p

te

d

M

10

25 Page 25 of 49

Brenza, T.M., Ghaisas, S., Ramirez, J.E., Harischandra, D., Anantharam, V.,

2

Kalyanaraman, B., Kanthasamy, A.G., Narasimhan, B., 2016. Neuronal protection

3

against oxidative insult by polyanhydride nanoparticle-based mitochondria-targeted

4

antioxidant therapy. Nanomedicine.

5

Charli, A., Jin, H., Anantharam, V., Kanthasamy, A., Kanthasamy, A.G., 2016.

6

Alterations in mitochondrial dynamics induced by tebufenpyrad and pyridaben in a

7

dopaminergic neuronal cell culture model. Neurotoxicology 53, 302-313.

8

Chen, C.J., Liao, S.L., 2002. Oxidative stress involves in astrocytic alterations induced

9

by manganese. Exp Neurol 175(1), 216-225.

an

us

cr

ip t

1

Chen, P., Bowman, A.B., Mukhopadhyay, S., Aschner, M., 2015. SLC30A10: A novel

11

manganese transporter. Worm 4(3), e1042648.

12

Choi, C.J., Anantharam, V., Martin, D.P., Nicholson, E.M., Richt, J.A., Kanthasamy, A.,

13

Kanthasamy, A.G., 2010. Manganese upregulates cellular prion protein and contributes

14

to altered stabilization and proteolysis: relevance to role of metals in pathogenesis of

15

prion disease. Toxicol Sci 115(2), 535-546.

16

Choi, C.J., Anantharam, V., Saetveit, N.J., Houk, R.S., Kanthasamy, A., Kanthasamy,

17

A.G., 2007. Normal cellular prion protein protects against manganese-induced oxidative

18

stress and apoptotic cell death. Toxicol Sci 98(2), 495-509.

19

Codolo, G., Plotegher, N., Pozzobon, T., Brucale, M., Tessari, I., Bubacco, L., de

20

Bernard, M., 2013. Triggering of inflammasome by aggregated alpha-synuclein, an

21

inflammatory response in synucleinopathies. PLoS One 8(1), e55375.

22

Colombo, E., Farina, C., 2016. Astrocytes: Key Regulators of Neuroinflammation.

23

Trends Immunol 37(9), 608-620.

Ac ce p

te

d

M

10

26 Page 26 of 49

Dorman, D.C., McManus, B.E., Marshall, M.W., James, R.A., Struve, M.F., 2004. Old

2

age and gender influence the pharmacokinetics of inhaled manganese sulfate and

3

manganese phosphate in rats. Toxicol Appl Pharmacol 197(2), 113-124.

4

Ellingsen, D.G., Hetland, S.M., Thomassen, Y., 2003. Manganese air exposure

5

assessment and biological monitoring in the manganese alloy production industry. J

6

Environ Monit 5(1), 84-90.

7

Febbraro, F., Giorgi, M., Caldarola, S., Loreni, F., Romero-Ramos, M., 2012. alpha-

8

Synuclein expression is modulated at the translational level by iron. Neuroreport 23(9),

9

576-580.

an

us

cr

ip t

1

Freeman, L.C., Ting, J.P., 2016. The pathogenic role of the inflammasome in

11

neurodegenerative diseases. J Neurochem 136 Suppl 1, 29-38.

12

Galvin, J.E., Giasson, B., Hurtig, H.I., Lee, V.M., Trojanowski, J.Q., 2000.

13

Neurodegeneration with brain iron accumulation, type 1 is characterized by alpha-,

14

beta-, and gamma-synuclein neuropathology. Am J Pathol 157(2), 361-368.

15

Gavin, C.E., Gunter, K.K., Gunter, T.E., 1990. Manganese and calcium efflux kinetics in

16

brain mitochondria. Relevance to manganese toxicity. Biochem J 266(2), 329-334.

17

Ghosh, A., Langley, M.R., Harischandra, D.S., Neal, M.L., Jin, H., Anantharam, V.,

18

Joseph, J., Brenza, T., Narasimhan, B., Kanthasamy, A., Kalyanaraman, B.,

19

Kanthasamy, A.G., 2016a. Mitoapocynin Treatment Protects Against Neuroinflammation

20

and Dopaminergic Neurodegeneration in a Preclinical Animal Model of Parkinson's

21

Disease. J Neuroimmune Pharmacol 11(2), 259-278.

22

Ghosh, A., Saminathan, H., Kanthasamy, A., Anantharam, V., Jin, H., Sondarva, G.,

23

Harischandra, D.S., Qian, Z., Rana, A., Kanthasamy, A.G., 2013. The peptidyl-prolyl

Ac ce p

te

d

M

10

27 Page 27 of 49

isomerase Pin1 up-regulation and proapoptotic function in dopaminergic neurons:

2

relevance to the pathogenesis of Parkinson disease. J Biol Chem 288(30), 21955-

3

21971.

4

Ghosh, A., Tyson, T., George, S., Hildebrandt, E.N., Steiner, J.A., Madaj, Z., Schulz, E.,

5

Machiela, E., McDonald, W.G., Escobar Galvis, M.L., Kordower, J.H., Van Raamsdonk,

6

J.M., Colca, J.R., Brundin, P., 2016b. Mitochondrial pyruvate carrier regulates

7

autophagy, inflammation, and neurodegeneration in experimental models of Parkinson's

8

disease. Sci Transl Med 8(368), 368ra174.

9

Giasson, B.I., Uryu, K., Trojanowski, J.Q., Lee, V.M., 1999. Mutant and wild type human

10

alpha-synucleins assemble into elongated filaments with distinct morphologies in vitro. J

11

Biol Chem 274(12), 7619-7622.

12

Glass, C.K., Saijo, K., Winner, B., Marchetto, M.C., Gage, F.H., 2010. Mechanisms

13

underlying inflammation in neurodegeneration. Cell 140(6), 918-934.

14

Golde, T.E., 2002. Inflammation takes on Alzheimer disease. Nat Med 8(9), 936-938.

15

Gonzalez, L.E., Juknat, A.A., Venosa, A.J., Verrengia, N., Kotler, M.L., 2008.

16

Manganese activates the mitochondrial apoptotic pathway in rat astrocytes by

17

modulating the expression of proteins of the Bcl-2 family. Neurochem Int 53(6-8), 408-

18

415.

19

Gordon, R., Hogan, C.E., Neal, M.L., Anantharam, V., Kanthasamy, A.G., Kanthasamy,

20

A., 2011. A simple magnetic separation method for high-yield isolation of pure primary

21

microglia. J Neurosci Methods 194(2), 287-296.

22

Gordon, R., Neal, M.L., Luo, J., Langley, M.R., Harischandra, D.S., Panicker, N., Charli,

23

A., Jin, H., Anantharam, V., Woodruff, T.M., Zhou, Q.-Y., Kanthasamy, A.G.,

Ac ce p

te

d

M

an

us

cr

ip t

1

28 Page 28 of 49

Kanthasamy, A., 2016. Prokineticin-2 upregulation during neuronal injury mediates a

2

compensatory protective response against dopaminergic neuronal degeneration. Nature

3

communications 7, 12932.

4

Gordon, R., Neal, M.L., Luo, J., Langley, M.R., Harischandra, D.S., Panicker, N., Charli,

5

A., Jin, H., Anantharam, V., Woodruff, T.M., Zhou, Q.Y., Kanthasamy, A.G.,

6

Kanthasamy, A., 2016a. Prokineticin-2 upregulation during neuronal injury mediates a

7

compensatory protective response against dopaminergic neuronal degeneration. Nature

8

communications 7, 12932.

9

Gordon, R., Singh, N., Lawana, V., Ghosh, A., Harischandra, D.S., Jin, H., Hogan, C.,

10

Sarkar, S., Rokad, D., Panicker, N., Anantharam, V., Kanthasamy, A.G., Kanthasamy,

11

A., 2016b. Protein kinase Cdelta upregulation in microglia drives neuroinflammatory

12

responses and dopaminergic neurodegeneration in experimental models of Parkinson's

13

disease. Neurobiology of disease 93, 96-114.

14

Gorojod, R.M., Alaimo, A., Porte Alcon, S., Pomilio, C., Saravia, F., Kotler, M.L., 2015.

15

The autophagic- lysosomal pathway determines the fate of glial cells under manganese-

16

induced oxidative stress conditions. Free radical biology & medicine 87, 237-251.

17

Graff, C.L., Pollack, G.M., 2005. Nasal drug administration: potential for targeted central

18

nervous system delivery. J Pharm Sci 94(6), 1187-1195.

19

Gregory, A., Polster, B.J., Hayflick, S.J., 2009. Clinical and genetic delineation of

20

neurodegeneration with brain iron accumulation. J Med Genet 46(2), 73-80.

21

Gunter, R.E., Puskin, J.S., Russell, P.R., 1975. Quantitative magnetic resonance

22

studies of manganese uptake by mitochondria. Biophys J 15(4), 319-333.

Ac ce p

te

d

M

an

us

cr

ip t

1

29 Page 29 of 49

Gunter, T.E., Gavin, C.E., Aschner, M., Gunter, K.K., 2006. Speciation of manganese in

2

cells and mitochondria: a search for the proximal cause of manganese neurotoxicity.

3

Neurotoxicology 27(5), 765-776.

4

Gunter, T.E., Sheu, S.S., 2009. Characteristics and possible functions of mitochondrial

5

Ca(2+) transport mechanisms. Biochim Biophys Acta 1787(11), 1291-1308.

6

Harischandra, D.S., Ghaisas, S., Rokad, D., Zamanian, M., Jin, H., Anantharam, V.,

7

Kimber, M., Kanthasamy, A., Kanthasamy, A., 2017. Environmental Neurotoxicant

8

Manganese Regulates Exosome-mediated Extracellular miRNAs in Cell Culture Model

9

of Parkinson's Disease: Relevance to alpha-Synuclein Misfolding in Metal Neurotoxicity.

an

us

cr

ip t

1

Neurotoxicology.

11

Harischandra, D.S., Jin, H., Anantharam, V., Kanthasamy, A., Kanthasamy, A.G., 2015.

12

alpha-Synuclein protects against manganese neurotoxic insult during the early stages of

13

exposure in a dopaminergic cell model of Parkinson's disease. Toxicol Sci 143(2), 454-

14

468.

15

Hazell, A.S., 2002. Astrocytes and manganese neurotoxicity. Neurochem Int 41(4), 271-

16

277.

17

Herrero, M.T., Estrada, C., Maatouk, L., Vyas, S., 2015. Inflammation in Parkinson's

18

disease: role of glucocorticoids. Front Neuroanat 9, 32.

19

Hesketh, S., Sassoon, J., Knight, R., Brown, D.R., 2008. Elevated manganese levels in

20

blood and CNS in human prion disease. Mol Cell Neurosci 37(3), 590-598.

21

Horning, K.J., Caito, S.W., Tipps, K.G., Bowman, A.B., Aschner, M., 2015. Manganese

22

Is Essential for Neuronal Health. Annu Rev Nutr 35, 71-108.

Ac ce p

te

d

M

10

30 Page 30 of 49

Kanthasamy, A.G., Choi, C., Jin, H., Harischandra, D.S., Anantharam, V., Kanthasamy,

2

A., 2012. Effect of divalent metals on the neuronal proteasomal system, prion protein

3

ubiquitination and aggregation. Toxicol Lett 214(3), 288-295.

4

Karki, P., Lee, E., Aschner, M., 2013. Manganese neurotoxicity: a focus on glutamate

5

transporters. Ann Occup Environ Med 25(1), 4.

6

Karki, P., Smith, K., Johnson, J., Jr., Aschner, M., Lee, E., 2015. Role of transcription

7

factor yin yang 1 in manganese-induced reduction of astrocytic glutamate transporters:

8

Putative mechanism for manganese-induced neurotoxicity. Neurochem Int 88, 53-59.

9

Karki, P., Webb, A., Smith, K., Johnson, J., Jr., Lee, K., Son, D.S., Aschner, M., Lee, E.,

10

2014. Yin Yang 1 is a repressor of glutamate transporter EAAT2, and it mediates

11

manganese-induced decrease of EAAT2 expression in astrocytes. Mol Cell Biol 34(7),

12

1280-1289.

13

Kim, C., Ho, D.H., Suk, J.E., You, S., Michael, S., Kang, J., Joong Lee, S., Masliah, E.,

14

Hwang, D., Lee, H.J., Lee, S.J., 2013. Neuron-released oligomeric alpha-synuclein is an

15

endogenous

16

communications 4, 1562.

17

Kwakye, G.F., Paoliello, M.M., Mukhopadhyay, S., Bowman, A.B., Aschner, M., 2015.

18

Manganese-Induced

19

Distinguishable Features. Int J Environ Res Public Health 12(7), 7519-7540.

20

Langley, M., Ghosh, A., Charli, A., Sarkar, S., Ay, M., Luo, J., Zielonka, J., Brenza, T.,

21

Bennett, B., Jin, H., Ghaisas, S., Schlichtmann, B., Kim, D., Anantharam, V.,

22

Kanthasamy, A., Narasimhan, B., Kalyanaraman, B., Kanthasamy, A., 2017. Mito-

23

apocynin Prevents Mitochondrial Dysfunction, Microglial Activation, Oxidative Damage

of

TLR2

Ac ce p

agonist

te

d

M

an

us

cr

ip t

1

for

Parkinsonism

paracrine

and

activation

Parkinson's

of

microglia.

Disease:

Shared

Nature

and

31 Page 31 of 49

and Progressive Neurodegeneration in MitoPark Transgenic Mice. Antioxidants &

2

Redox Signaling.

3

Lawana, V., Singh, N., Sarkar, S., Charli, A., Jin, H., Anantharam, V., Kanthasamy,

4

A.G., Kanthasamy, A., 2017. Involvement of c-Abl Kinase in Microglial Activation of

5

NLRP3 Inflammasome and Impairment in Autolysosomal System. J Neuroimmune

6

Pharmacol.

7

Lee, E.J., Woo, M.S., Moon, P.G., Baek, M.C., Choi, I.Y., Kim, W.K., Junn, E., Kim,

8

H.S., 2010. Alpha-synuclein activates microglia by inducing the expressions of matrix

9

metalloproteinases and the subsequent activation of protease-activated receptor-1.

an

us

cr

ip t

1

Journal of immunology 185(1), 615-623.

11

Leyva-Illades, D., Chen, P., Zogzas, C.E., Hutchens, S., Mercado, J.M., Swaim, C.D.,

12

Morrisett, R.A., Bowman, A.B., Aschner, M., Mukhopadhyay, S., 2014. SLC30A10 is a

13

cell surface-localized manganese efflux transporter, and parkinsonism-causing

14

mutations block its intracellular trafficking and efflux activity. The Journal of

15

neuroscience : the official journal of the Society for Neuroscience 34(42), 14079-14095.

16

Liu, L., Chan, C., 2014. IPAF inflammasome is involved in interleukin-1beta production

17

from astrocytes, induced by palmitate; implications for Alzheimer's Disease.

18

Neurobiology of aging 35(2), 309-321.

19

Liu, T., Xue, C.C., Shi, Y.L., Bai, X.J., Li, Z.F., Yi, C.L., 2014. Overexpression of

20

mitofusin 2 inhibits reactive astrogliosis proliferation in vitro. Neurosci Lett 579, 24-29.

21

Lotharius, J., Brundin, P., 2002. Pathogenesis of Parkinson's disease: dopamine,

22

vesicles and alpha-synuclein. Nat Rev Neurosci 3(12), 932-942.

Ac ce p

te

d

M

10

32 Page 32 of 49

1

Luk, K.C., Kehm, V., Carroll, J., Zhang, B., O'Brien, P., Trojanowski, J.Q., Lee, V.M.,

2

2012a.

3

neurodegeneration in nontransgenic mice. Science 338(6109), 949-953.

4

Luk, K.C., Kehm, V.M., Zhang, B., O'Brien, P., Trojanowski, J.Q., Lee, V.M., 2012b.

5

Intracerebral inoculation of pathological alpha-synuclein initiates a rapidly progressive

6

neurodegenerative alpha-synucleinopathy in mice. J Exp Med 209(5), 975-986.

7

Malthankar, G.V., White, B.K., Bhushan, A., Daniels, C.K., Rodnick, K.J., Lai, J.C.,

8

2004. Differential lowering by manganese treatment of activities of glycolytic and

9

tricarboxylic acid (TCA) cycle enzymes investigated in neuroblastoma and astrocytoma

10

cells is associated with manganese-induced cell death. Neurochem Res 29(4), 709-717.

11

Martinez-Finley, E.J., Gavin, C.E., Aschner, M., Gunter, T.E., 2013. Manganese

12

neurotoxicity and the role of reactive oxygen species. Free radical biology & medicine

13

62, 65-75.

14

Milatovic, D., Yin, Z., Gupta, R.C., Sidoryk, M., Albrecht, J., Aschner, J.L., Aschner, M.,

15

2007. Manganese induces oxidative impairment in cultured rat astrocytes. Toxicol Sci

16

98(1), 198-205.

17

Milatovic, D., Yin, Z., Gupta, R.C., Sidoryk, M., Albrecht, J., Aschner, J.L., Aschner, M.,

18

2007. Manganese Induces Oxidative Impairment in Cultured Rat Astrocytes.

19

Toxicological Sciences 98(1), 198-205.

20

Mittal, M., Siddiqui, M.R., Tran, K., Reddy, S.P., Malik, A.B., 2014. Reactive oxygen

21

species in inflammation and tissue injury. Antioxid Redox Signal 20(7), 1126-1167.

22

Moberly, A.H., Czarnecki, L.A., Pottackal, J., Rubinstein, T., Turkel, D.J., Kass, M.D.,

23

McGann, J.P., 2012. Intranasal exposure to manganese disrupts neurotransmitter

alpha-synuclein

transmission

initiates

Parkinson-like

Ac ce p

te

d

M

an

us

cr

ip t

Pathological

33 Page 33 of 49

release from glutamatergic synapses in the central nervous system in vivo.

2

Neurotoxicology 33(5), 996-1004.

3

Moreno, J.A., Streifel, K.M., Sullivan, K.A., Legare, M.E., Tjalkens, R.B., 2009.

4

Developmental exposure to manganese increases adult susceptibility to inflammatory

5

activation of glia and neuronal protein nitration. Toxicol Sci 112(2), 405-415.

6

Moreno, J.A., Sullivan, K.A., Carbone, D.L., Hanneman, W.H., Tjalkens, R.B., 2008.

7

Manganese potentiates nuclear factor-kappaB-dependent expression of nitric oxide

8

synthase 2 in astrocytes by activating soluble guanylate cyclase and extracellular

9

responsive kinase signaling pathways. J Neurosci Res 86(9), 2028-2038.

an

us

cr

ip t

1

Narhi, L., Wood, S.J., Steavenson, S., Jiang, Y., Wu, G.M., Anafi, D., Kaufman, S.A.,

11

Martin, F., Sitney, K., Denis, P., Louis, J.C., Wypych, J., Biere, A.L., Citron, M., 1999.

12

Both familial Parkinson's disease mutations accelerate alpha-synuclein aggregation. J

13

Biol Chem 274(14), 9843-9846.

14

Ngwa, H.A., Kanthasamy, A., Jin, H., Anantharam, V., Kanthasamy, A.G., 2014.

15

Vanadium exposure induces olfactory dysfunction in an animal model of metal

16

neurotoxicity. Neurotoxicology 43, 73-81.

17

Panicker, N., Saminathan, H., Jin, H., Neal, M., Harischandra, D.S., Gordon, R.,

18

Kanthasamy, K., Lawana, V., Sarkar, S., Luo, J., Anantharam, V., Kanthasamy, A.G.,

19

Kanthasamy, A., 2015. Fyn Kinase Regulates Microglial Neuroinflammatory Responses

20

in Cell Culture and Animal Models of Parkinson's Disease. The Journal of neuroscience

21

: the official journal of the Society for Neuroscience 35(27), 10058-10077.

Ac ce p

te

d

M

10

34 Page 34 of 49

Park, E.J., Park, K., 2010. Induction of oxidative stress and inflammatory cytokines by

2

manganese chloride in cultured T98G cells, human brain glioblastoma cell line. Toxicol

3

In Vitro 24(2), 472-479.

4

Pekny, M., Pekna, M., 2014. Astrocyte reactivity and reactive astrogliosis: costs and

5

benefits. Physiol Rev 94(4), 1077-1098.

6

Peres, T.V., Schettinger, M.R., Chen, P., Carvalho, F., Avila, D.S., Bowman, A.B.,

7

Aschner, M., 2016. "Manganese-induced neurotoxicity: a review of its behavioral

8

consequences and neuroprotective strategies". BMC Pharmacol Toxicol 17(1), 57.

9

Pham, A.H., Meng, S., Chu, Q.N., Chan, D.C., 2012. Loss of Mfn2 results in

10

progressive, retrograde degeneration of dopaminergic neurons in the nigrostriatal

11

circuit. Hum Mol Genet 21(22), 4817-4826.

12

Phillips, E.C., Croft, C.L., Kurbatskaya, K., O'Neill, M.J., Hutton, M.L., Hanger, D.P.,

13

Garwood, C.J., Noble, W., 2014. Astrocytes and neuroinflammation in Alzheimer's

14

disease. Biochem Soc Trans 42(5), 1321-1325.

15

Popoli, M., Yan, Z., McEwen, B.S., Sanacora, G., 2011. The stressed synapse: the

16

impact of stress and glucocorticoids on glutamate transmission. Nat Rev Neurosci

17

13(1), 22-37.

18

Rao, K.V., Norenberg, M.D., 2004. Manganese induces the mitochondrial permeability

19

transition in cultured astrocytes. J Biol Chem 279(31), 32333-32338.

20

Rodier, J., 1955. Manganese poisoning in Moroccan miners. Br J Ind Med 12(1), 21-35.

21

Rokad, D., Ghaisas, S., Harischandra, D.S., Jin, H., Anantharam, V., Kanthasamy, A.,

22

Kanthasamy, A.G., 2016. Role of neurotoxicants and traumatic brain injury in alpha-

23

synuclein protein misfolding and aggregation. Brain Res Bull.

Ac ce p

te

d

M

an

us

cr

ip t

1

35 Page 35 of 49

Sarkar, S., Malovic, E., Plante, B., Zenitsky, G., Jin, H., Anantharam, V., Kanthasamy,

2

A., Kanthasamy, A.G., 2017. Rapid and Refined CD11b Magnetic Isolation of Primary

3

Microglia with Enhanced Purity and Versatility. J Vis Exp(122).

4

Scott, I., Youle, R.J., 2010. Mitochondrial fission and fusion. Essays in biochemistry 47,

5

85-98.

6

Seo, J., Ottesen, E.W., Singh, R.N., 2014. Antisense methods to modulate pre-mRNA

7

splicing. Methods Mol Biol 1126, 271-283.

8

Sidoryk-Wegrzynowicz, M., Aschner, M., 2013. Role of astrocytes in manganese

9

mediated neurotoxicity. BMC Pharmacol Toxicol 14, 23.

an

us

cr

ip t

1

Streifel, K.M., Miller, J., Mouneimne, R., Tjalkens, R.B., 2013. Manganese inhibits ATP-

11

induced calcium entry through the transient receptor potential channel TRPC3 in

12

astrocytes. Neurotoxicology 34, 160-166.

13

Su, X., Maguire-Zeiss, K.A., Giuliano, R., Prifti, L., Venkatesh, K., Federoff, H.J., 2008.

14

Synuclein activates microglia in a model of Parkinson's disease. Neurobiology of aging

15

29(11), 1690-1701.

16

Tang, F.L., Liu, W., Hu, J.X., Erion, J.R., Ye, J., Mei, L., Xiong, W.C., 2015. VPS35

17

Deficiency or Mutation Causes Dopaminergic Neuronal Loss by Impairing Mitochondrial

18

Fusion and Function. Cell Rep 12(10), 1631-1643.

19

Tansey, M.G., Goldberg, M.S., 2010. Neuroinflammation in Parkinson's disease: its role

20

in neuronal death and implications for therapeutic intervention. Neurobiology of disease

21

37(3), 510-518.

22

Tjalkens, R.B., Zoran, M.J., Mohl, B., Barhoumi, R., 2006. Manganese suppresses ATP-

23

dependent intercellular calcium waves in astrocyte networks through alteration of

Ac ce p

te

d

M

10

36 Page 36 of 49

mitochondrial and endoplasmic reticulum calcium dynamics. Brain Res 1113(1), 210-

2

219.

3

Volterra, A., Meldolesi, J., 2005. Astrocytes, from brain glue to communication

4

elements: the revolution continues. Nat Rev Neurosci 6(8), 626-640.

5

Whitton, P.S., 2007. Inflammation as a causative factor in the aetiology of Parkinson's

6

disease. Br J Pharmacol 150(8), 963-976.

7

Yen, C.F., Harischandra, D.S., Kanthasamy, A., Sivasankar, S., 2016. Copper-induced

8

structural conversion templates prion protein oligomerization and neurotoxicity. Sci Adv

9

2(7), e1600014.

an

us

cr

ip t

1

Yin, Z., Aschner, J.L., dos Santos, A.P., Aschner, M., 2008. Mitochondrial-dependent

11

manganese neurotoxicity in rat primary astrocyte cultures. Brain Res 1203, 1-11.

M

10

Ac ce p

te

d

12

37 Page 37 of 49

Figure Legends

2

Fig. 1: Determination of IC50 for MnCl2 based on metabolic response of astrocytic

3

cell culture. (A) MTS assay performed on primary mouse astrocytes after treatment

4

with 1, 3, 10, 30, 100, 300, and 1000 µM MnCl2 for 24 h. (B) MTS assay performed on

5

U373 human astrocyte cell line after treatment with 1, 3, 10, 30, 100, 300, and 1000 µM

6

MnCl2 for 24 h. Data represented as mean±SEM with 8 biological replicates from 1-2

7

independent experiments.

8

Fig. 2: Mn exposure reduced mitochondrial mass and induced mitochondrial

9

dysfunction in astrocyte cell cultures. 100 µM Mn for 24 h induced loss in

10

mitochondrial mass in primary mouse astrocytes (A) and in U373 cell line (B), as shown

11

by MitoTracker green assay. (C) 100 µM Mn exposure shifted mitochondrial morphology

12

in primary astrocytes, as shown by MitoTracker Red. (D) CellTiter Glo assay reveals Mn

13

reduced ATP production in primary astrocytes. (E) q-RT-PCR and (F) Western blot

14

analyses show 100 µM Mn exposure for 24 h reduced Mitofusin2 (Mfn2) in primary

15

mouse astrocytes. Data analyzed via Student’s t test, *p<0.05, **p<0.01, ***p<0.001.

16

Data represented as mean±SEM with 3-8 biological replicates from 2-3 independent

17

experiments.

18

Fig. 3: Mn exposure altered mitochondrial respiration in primary mouse

19

astrocytes. (A) Seahorse Mito Stress Test measuring oxygen consumption rate (OCR).

20

Mn exposure for 24 h reduced basal mitochondrial respiration (B) and mitochondrial

21

ATP generation (C) in primary mouse astrocytes. Data analyzed via Student’s t test,

22

*p<0.05, **p <0.01, ***p<0.001. Data represented as mean±SEM with 3-5 biological

23

replicates from 2-3 independent experiments.

Ac ce p

te

d

M

an

us

cr

ip t

1

38 Page 38 of 49

Fig. 4: Mn exposure alters non-mitochondrial respiration and cellular energy

2

phenotype in primary mouse astrocytes. (A) Seahorse Mito-Stress Test measuring

3

extracellular acidification rate (ECAR). Mn exposure for 24 h reduced non-mitochondrial

4

respiration (B). 100 µM Mn altered cellular energy phenotype in primary mouse

5

astrocytes (C). Data analyzed via Student’s t-test, *p<0.05, **p <0.01, ***p<0.001. Data

6

represented as mean±SEM with 3-5 biological replicates from 2-3 independent

7

experiments.

8

Fig. 5: Mn-induced production of pro-inflammatory factors in astrocytes. (A-F) q-

9

RT-PCR analysis of primary mouse astrocytes treated with 100 µM Mn for 24 h induced

10

expression of the pro-inflammatory factors IL-12b (A), IL-6 (B), TNFα (C), Nos2 (D), pro-

11

IL-1β (E) and GM-CSF (F). (G-I) Luminex analysis of treatment medium from primary

12

astrocytes exposed to 100 µM Mn for 24 h, revealing induction of the released pro-

13

inflammatory factors IL-6 (G), IL-12 (H), TNFα (I). Data analyzed via Student’s t-test,

14

*p<0.05, **p <0.01, ***p<0.001. Data represented as mean±SEM with 2-8 biological

15

replicates from 2-3 independent experiments.

16

Fig. 6: Mn potentiated production and release of pro-inflammatory factors

17

induced by αSynagg. (A) Co-treatment of 100 µM Mn with 1 µM αSynagg (Agg-αsyn)

18

potentiated pro-IL-1β mRNA levels as shown by q-RT-PCR. (B) Luminex assay reveals

19

Mn potentiated release of IL-1β levels from astrocytes induced by αSynagg. (C, E) Mn

20

aggravated αSynagg-induced mRNA expression of IL-6 (C) and GM-CSF (E) as revealed

21

by q-RT-PCR. (D, F) Luminex assay showing Mn has an additive effect on the release

22

of αSynagg-induced IL-12 (D) and TNFα (F). Data analyzed via ANOVA with Tukey post

Ac ce p

te

d

M

an

us

cr

ip t

1

39 Page 39 of 49

analysis, *p<0.05, **p <0.01, ***p<0.001. Data represented as mean±SEM with 3-8

2

biological replicates from 2-3 independent experiments.

3

Fig. 7: Mito-apocynin attenuated Mn-induced pro-inflammatory factors in human

4

astrocytic culture. Co-treatment with 10 µM mito-apocynin (MA) and 100 µM MnCl2

5

attenuated the mRNA upregulation of GM-CSF (A), pro-IL-1β (B) and pro-IL-18 (C). (D)

6

Co-treatment with 10 µM mito-apocynin and 100 µM MnCl2 modulated the change in

7

mitochondrial mass induced by Mn, as shown by MitoTracker Green assay. Data

8

analyzed via ANOVA with Tukey post analysis, *p<0.05, **p<0.01, ***p<0.001. Data

9

represented as mean±SEM with 3-8 biological replicates from 2-3 independent

an

us

cr

ip t

1

experiments.

11

Fig. 8: Intranasal exposure to Mn caused motor deficits and inflammation in vivo.

12

(A-C) Intranasal exposure of 50 µl of 20 mM MnCl2 caused motor deficits in C57BL/6

13

mice as indicated by total distance traveled (A), horizontal activity, (B) and total

14

movement time (C). Neurochemical analysis of OB revealing Mn reduced dopamine

15

levels (D). Western blot analysis shows Mn reduced TH (E) and increased GFAP (F)

16

and iNOS (G). (H) Luminex analysis shows Mn exposure increased serum levels of IL-

17

12. Data analyzed via Student’s t-test, *p<0.05, **p<0.01, ***p<0.001. Data represented

18

as mean±SEM with n=6-16.

Ac ce p

te

d

M

10

19

40 Page 40 of 49

Figure 1 A IC50= 92.68 μM

ip t

80

cr

60

us

40

an

20 0 -6.5

-6.0

-5.5

-5.0

-4.5

M

Metabolic Activity (% Control)

100

-4.0

-3.5

-3.0

Log [Mn] (M)

ed

Metabolic Activity (% Control)

B

pt

100

60 40 20

Ac ce

80

IC50= 50.46 μM

0 -6.5

-6.0

-5.5

-5.0

-4.5

-4.0

-3.5

-3.0

Log [Mn] (M)

Page 41 of 49

M

n

ro l

on t

1.2

C D M

0

n

0 80 20

n

ip t

B

M

cr

120

us

C on tr ol

Mitochondrial Mass (%Control)

80 20

on tr ol

n

***

C

an

M

M

C on tr ol

Mitochondrial Mass (%Control) 100

ATP (% Control)

ed

pt

Ac ce

E

C

Mitofusin 2 mRNA Fold Change

A Figure 2

120

100

***

120

100

***

80 20

0

F

1.0

*

0.8 0.2

0.0

Page 42 of 49

Figure 3

A Oligomycin

Rotenone/Antimycin

an

us

cr

ip t

FCCP

***

Ac ce

pt

ed

M

B

C

***

Page 43 of 49

Figure 4

A Rotenone/Antimycin Oligomycin

us

cr

ip t

FCCP

***

Ac ce

pt

ed

M

an

B

C

Page 44 of 49

2000

0

***

0.0988

1

0

H

800

I

600

***

400

200

0 0.00

cr 1.75

20

n

0.25

ip t

0.50

M

TNF mRNA Fold change

0.75

n

0.0

1.00

M

3 Co nt ro l

us

2.5

CSF-2 mRNA Fold Change

5.0

Co nt ro l

E

an

n

***

n

***

7.5

M

0

2 M

B

Co nt ro l

10

TNF- (pg/mL)

30

M

0.0

n

D ro l

0.5

ed Co nt

1.0

IL-6 mRNA Fold Change

*

M

20

Pro-IL-1 mRNA Fold Change

n

M

ro l

1.5

Co nt ro l

pt

n

Co nt

IL-12b mRNA Fold Change

2.0

Mn

4000 M

Co nt ro l

NOS2 mRNA Fold Change

A

Co nt ro l

6000

IL-12 (pg/mL)

8000

Ac ce

G

Mn

Co nt ro l

IL-6 (pg/mL)

Figure 5

C 1.75

*

1.50

1.25

F 0.0909

1.50

1.25

1.00

0.75

0.50

0.25

0.00

25

***

15

10

5

0

Page 45 of 49

5000 M

n

n

17500

n

M

ag g+

S yn

ag g+

M

n

n

M

cr S yn

ag g

S yn

ip t

10.0

M

n

M

0

ag g+

20 2.5

S yn

F ag g

D

us

0.0

S yn

0

ag g

40 Co nt ro l

B

S yn

15

ro l

*** 15000

an

2 IL-1 (pg/mL)

4

Co nt

10

M

n

6

IL-12 (pg/mL)

M

n

M

ag g+

S yn

ag g

10

Co nt ro l

60

TNF- (pg/mL)

ed

0

n

5

M

pt

20

n

n

M

ag g+

S yn

ag g

S yn

Co nt ro l

Pro-IL-1 mRNA Fold Change

8

M

n

M

ag g+

S yn

80

*

Ac ce

100

ag g

E S yn

C

S yn

Co nt ro l

IL-6 mRNA Fold Change

A

Co nt ro l

CSF-2 mRNA Fold Change

Figure 6 ***

*** 7.5

5.0

**

12500

10000

7500

5000

2500

0

***

4000

3000

2000

1000

0

Page 46 of 49

oc yn in

***

to -ap

110

Mi

120 ito

M

-a p

cy ni n

n

oc yn in

-a po

M

us

cr

ip t

6

cy nin

D

n+ Mi to

0

an Co nt ro l

Pro-IL-1 mRNA Fold Change

2

M

0

4

-ap o

0

M

oc yn in

yn in

n

1

Mitochondrial Mass (% Control)

-A p

-a po c

M

2

Mn

*

ed

pt ito

M

ito

M

B

Co nt ro l

oc yn in

n+

M

3

Mn +M ito

-a p

3

ito

cy ni n

Ac ce

4

M

-a po

n

1

M

2

n+ M ito

C

M

Co nt ro l

CSF-2 mRNA Fold Change

A

Co nt ro l

Pro-IL-18 mRNA Fold Change

Figure 7

8

***

***

100

90

80 20

0

Page 47 of 49

B

C *** 50

0

ol

ip t

C

on tr

M

ol C

on tr

M

ol on tr C

D Dopamine level (% Control)

Control

3

1

2

3

us

2

Manganese

TH

62kDa

β-Actin

an

42kDa

Control

1

2

G

M

F

Manganese

3

1

2

GFAP

130kDa

β-Actin

42kDa

Control

1

2

Control

Mn

Mn

Manganese

3

1

2

3 iNOS β-Actin

0.0855

0.75 iNOS (Relative Expression)

pt

1.2 1.1 1.0 0.9 0.8 0.7 0.6 0.5 0.4 0.3 0.2 0.1 0.0

**

Ac ce

GFAP (Relative Expression)

42kDa

**

Control

ed

50kDa

3

1.2 1.1 1.0 0.9 0.8 0.7 0.6 0.5 0.4 0.3 0.2 0.1 0.0

cr

E 1

50

n

0

n

0

100

n

*** 50

100

M

100

150

Movement time (% control)

150

Horizontal activity (% control)

150

Total distance travelled (% control)

Figure 8

TH (Relative Expression)

A

0.50

0.25

0.00 Control

Mn

H

Page 48 of 49

Highlights 1. Mn induces mitochondrial dysfunction in astrocytes

ip t

2. Mn exposure induces inflammatory response in astrocytes and exacerbates αSynagg-induced inflammatory response in astrocytes

cr

3. Mito-apocynin attenuates Mn-induced inflammatory response in astrocytes by reducing

us

mitochondrial damage

4. Intranasal Mn exposure induces neuroinflammation and behavioral deficits in mouse

an

model

M

5. Thus, Mn-induced mitochondrial defects contributes to astroglial neuroinflammation

Ac ce p

te

d

 

Page 49 of 49