High fat diet consumption results in mitochondrial dysfunction, oxidative stress, and oligodendrocyte loss in the central nervous system

High fat diet consumption results in mitochondrial dysfunction, oxidative stress, and oligodendrocyte loss in the central nervous system

Journal Pre-proof High fat diet consumption results in mitochondrial dysfunction, oxidative stress, and Oligodendrocyte loss in the central nervous sy...

3MB Sizes 0 Downloads 12 Views

Journal Pre-proof High fat diet consumption results in mitochondrial dysfunction, oxidative stress, and Oligodendrocyte loss in the central nervous system

Monica R. Langley, Hyesook Yoon, Ha-Neui Kim, Chan-Il Choi, Whitney Simon, Laurel Kleppe, Ian R. Lanza, Nathan K. LeBrasseur, Aleksey Matveyenko, Isobel A. Scarisbrick PII:

S0925-4439(19)30358-8

DOI:

https://doi.org/10.1016/j.bbadis.2019.165630

Reference:

BBADIS 165630

To appear in:

BBA - Molecular Basis of Disease

Received date:

20 September 2019

Revised date:

14 November 2019

Accepted date:

2 December 2019

Please cite this article as: M.R. Langley, H. Yoon, H.-N. Kim, et al., High fat diet consumption results in mitochondrial dysfunction, oxidative stress, and Oligodendrocyte loss in the central nervous system, BBA - Molecular Basis of Disease(2019), https://doi.org/10.1016/j.bbadis.2019.165630

This is a PDF file of an article that has undergone enhancements after acceptance, such as the addition of a cover page and metadata, and formatting for readability, but it is not yet the definitive version of record. This version will undergo additional copyediting, typesetting and review before it is published in its final form, but we are providing this version to give early visibility of the article. 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.

© 2019 Published by Elsevier.

Journal Pre-proof

1

High Fat Diet Consumption Results in Mitochondrial Dysfunction, Oxidative Stress, and Oligodendrocyte Loss in the Central Nervous System Short running title: High fat diet causes oligodendrocyte loss Authors Monica R. Langleya, Hyesook Yoona,b, Ha-Neui Kima, Chan-Il Choia, Whitney Simona, Laurel Kleppea, Ian R. Lanzab,c, Nathan K. LeBrasseura,b, Aleksey Matveyenkob,c, Isobel

ro

of

A. Scarisbricka,b,*.

Affiliations

Department of Physical Medicine & Rehabilitation, Rehabilitation Medicine Research

-p

a

Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN

lP

b

re

Center, Mayo Clinic, Rochester, MN 55905, USA

Department of Endocrinology, Mayo Clinic, Rochester, MN 55905, USA

Jo

ur

c

na

55905, USA

* Correspondence to: Isobel A. Scarisbrick, Ph.D., Neurobiology of Disease Program, Physical Medicine a& Rehabilitation, Rehabilitation Medicine Research Center, 642B Guggenheim Building, Mayo Clinic, Rochester, 200 First St SW, Rochester, MN 55905, Tel: 507-284-0124, Fax:507-266-4716, Email: [email protected]

Journal Pre-proof

2

Abstract Metabolic syndrome is a key risk factor and co-morbidity in multiple sclerosis (MS) and other neurological conditions, such that a better understanding of how a high fat diet contributes to oligodendrocyte loss and the capacity for myelin regeneration has the potential to highlight new treatment targets. Results demonstrate that modeling metabolic dysfunction in mice with chronic high fat diet (HFD) consumption promotes

of

loss of oligodendrocyte progenitors across the brain and spinal cord. A number of

ro

transcriptomic and metabolomic changes in ER stress, mitochondrial dysfunction, and

-p

oxidative stress pathways in HFD-fed mouse spinal cords were also identified.

re

Moreover, deficits in TCA cycle intermediates and mitochondrial respiration were observed in the chronic HFD spinal cord tissue. Oligodendrocytes are known to be

lP

particularly vulnerable to oxidative damage, and we observed increased markers of

na

oxidative stress in both the brain and spinal cord of HFD-fed mice. We additionally identified that increased apoptotic cell death signaling is underway in oligodendrocytes

ur

from mice chronically fed a HFD. When cultured under high saturated fat conditions,

Jo

oligodendrocytes decreased both mitochondrial function and differentiation. Overall, our findings show that HFD-related changes in metabolic regulators, decreased mitochondrial function, and oxidative stress contribute to a loss of myelinating cells. These studies identify HFD consumption as a key modifiable lifestyle factor for improved myelin integrity in the adult central nervous system and in addition new tractable metabolic targets for myelin protection and repair strategies.

Keywords high fat diet, myelin, oligodendrocyte, mitochondria, apoptosis

Journal Pre-proof Abbreviations 4-HNE= 4-hydroxynonenal aNSCs= adult neural stem cells CLAMS= Comprehensive Lab Animal Monitoring System EDSS= expanded disability status score EAE= experimental autoimmune encephalitis

of

HFD= high fat diet

ro

MetSyn = metabolic syndrome MS= multiple sclerosis

-p

MBP= myelin basic protein (MBP

re

OLC= oligodendrocyte lineage cell

lP

OPCs= Oligodendrocyte progenitor cells

Olig2= oligodendrocyte lineage transcription factor 2

na

PLP= proteolipid protein RER= respiratory exchange rate

ur

RRMS= relapsing remitting MS

Jo

SPMS= secondary progressive MS TCA= tricarboxylic acid

VWMD= vanishing white matter disease

3

Journal Pre-proof

4

1. INTRODUCTION White matter pathology including myelin sheath abnormalities, demyelination, loss of oligodendrocytes, and impaired myelin repair processes are significant pathophysiological characteristics of diverse neurological conditions, including multiple sclerosis (MS), Alzheimer’s disease, neuropsychiatric disorders, and traumatic brain and spinal cord injury [1-6]. Oligodendrocytes not only serve as insulation by

of

ensheathing axons and allowing for increased speed and temporal control of action

ro

potential propagation, but also provide key metabolic and trophic support for neurons [1,

-p

7, 8]. Development of new strategies to prevent oligodendrocyte loss and guide efficient

re

oligodendrogenesis and myelination has the potential to therapeutically benefit individuals affected by white matter-related neurological injury and disease. Since

lP

oligodendrocyte function is intimately linked to neighboring glial cells and neuronal

na

activity, in addition to microenvironmental factors, the most promising interventions will

ur

likely involve a combinatorial approach [1, 9].

Jo

Obesity during childhood or adolescence is associated with an increased risk of developing MS [10-12]. Recent studies support a causal role for elevated body mass index (BMI) in MS etiology which is further increased when interacting with other known genetic and environmental risk factors such as HLA polymorphisms or Epstein Barr virus positivity [10, 13, 14]. Interestingly, metabolic dysfunction resulting from excessive dietary fat intake may also influence the initiation, progression, or disability burden of MS [13, 15, 16]. In terms of conversion from relapsing remitting (RRMS) to secondary progressive (SPMS), elevated BMI is associated with an increased risk of SPMS in smokers, but not non-smokers [15]. There is also a significant positive association

Journal Pre-proof

5

between higher BMI or adverse lipid profile and higher Expanded Disability Status Score (EDSS) [16]. Since obesity and metabolic syndrome are common co-morbidities which impact quality of life for individuals with MS, recent studies have evaluated the utility of drugs and interventions to overcome metabolic syndrome as a strategy to improve patients’ physical and mental health or radiological outcomes [16-18].

of

Disruptions in brain lipid metabolism, serum lipid profiles, and cholesterol levels

ro

have all been reported during active disease in MS [16, 19]. How to manage dietary fat consumption for optimal CNS health is still controversial since accurate myelin

-p

assembly requires large amounts of lipids and fatty acid synthesis, yet overconsumption

re

of saturated fats is detrimental for overall brain function [19, 20]. HFD consumption

lP

exacerbates both autoreactive immune responses and neuroinflammation in experimental autoimmune encephalitis (EAE) [21, 22]. Metabolic derangement resulting

na

from obesogenic diets or excessive saturated fatty acids can lead to oxidative and ER

ur

stress responses and ultimate death of vulnerable cell types [23-25]. Elevated levels of oxidative stress and ER stress-related proteins have been observed in MS lesions [26,

Jo

27]. Oligodendrocytes are particularly sensitive and susceptible to oxidative stress having extremely high energetic and ER demands in order to support myelin membranes [28]. Moreover, our recent findings show that chronic consumption of a Western style diet, containing high fat and high sucrose, causes significant loss of oligodendrocytes and their progenitors in the intact adult murine spinal cord [29]. Therefore, to elucidate mechanisms underlying oligodendrocyte loss observed in the context of a Western diet, here we specifically address whether similar effects occur with consumption of a diet high in saturated fat alone. The brain and spinal cord of mice

Journal Pre-proof

6

consuming a regular or a high fat diet for 4 to 12 wk were analyzed using transcriptomic, metabolomic, functional, and immunohistochemical outcome measures to reveal potential mechanisms of oligodendrocyte loss. These approaches identified perturbations in mitochondrial function among the key mechanisms by which HFDconsumption is likely to impair myelinating cell function in vivo. Parallel disruptions in mitochondrial function and differentiation were also identified when HFD was modeled

of

in cultures of purified oligodendrocytes grown under high saturated fat conditions.

ro

Altogether these findings highlight new pathogenic mechanisms by which excess

-p

dietary fat contributes to myelin injury in the adult brain and spinal cord and uncover

lP

2. MATERIALS AND METHODS

re

new targets for therapy.

2.1 Experimental Method Details

na

2.1.1 Animal husbandry and diet

ur

All diets, procedures, and experiments were approved by The Mayo Clinic

Jo

Institutional Animal Care and Use Committee (IACUC) and performed in accordance with appropriate institutional and regulatory guidelines. Male C57BL6/J mice were obtained from Jackson Labs (Bar Harbor, ME) at 8 wk of age and were randomly assigned to feeding on either a regular diet (RD, 10 % kcal fat, D12450K, Research Diets, New Brunswick, NJ) or a high fat diet (HFD, 60% kcal fat, D12492, Research Diets, New Brunswick, NJ) beginning at 10 wk of age. Saturated fat comprised 22.7 and 32% of the total fat content within the diets, respectively. Carbohydrate comprised 70% kcal for RD and 20% HFD, while both diets had 20% kcal protein. Mice were

Journal Pre-proof

7

provided ad libitum access to the diets along with tap water and marked by tail tattoo for individual identification. Food intake and body weights were monitored weekly. 2.1.2 Glucose tolerance test Following a 6 h fast, baseline glucose levels were measured by needle puncture of the tail of mice consuming RD or HFD for 3 wk (Bayer Breeze2, Bayer, Mishawaka, IN). Next, an intraperitoneal injection of glucose (1g/kg) was administered and blood

of

glucose levels measured again at each time point (0, 15, 30, 60, 90, 120 min) as

ro

previously described [30].

-p

2.1.3 Behavioral Assessment

re

Behavioral analysis was performed at baseline and following 4 or 12 wk of RD or HFD consumption. To examine spontaneous movement, mice were placed in a

lP

chamber (27 x 27 x 20.3 cm, Med Associates Inc., St. Albans, VT) equipped with

na

infrared beams to record horizontal and vertical movements over a 30 min interval during the light phase. Mice were allowed to acclimate to the room conditions for 1 h

ur

prior to recording at 50 ms resolution in Activity Monitor Software. Equipment was

Jo

cleaned with 70% ethanol between mice. The same testing chamber and period was also used to evaluate anxiety-like behavior by quantifying the amount of time a mouse spent in the center zone (4.5, 4.5 to 12, 12 cm) versus the residual area. 2.1.4 Immunohistochemistry 4% Paraformaldehyde-fixed, paraffin-embedded brain and lumbosacral spinal cord tissues were cut to 6 µm and utilized for all immunostaining procedures. Immunoperoxidase and immunofluorescence was performed as previously described [29]. Briefly, sections were washed, steamed in antigen retrieval solution (sodium

Journal Pre-proof

8

citrate,10 mM, pH 6.0), blocked (20 % normal goat serum with 0.25 % Triton-x), then incubated in primary antibodies for Olig2 (Millipore, MABN50, 1:200), CC-1 (Millipore, OP80, 1:50), Nkx2.2 (Developmental Studies Hybridoma Bank, University of Iowa, Iowa city, IA), NG2 (Millipore, ab5320), 4-HNE (Abcam, ab46545, 1:500), Bax (Invitrogen, MA514003,1:200 ), Bcl-2 (Abcam, AB59348, 1:500), cleaved caspase-3 (Cell Signaling, 9664, 1:1000), GFAP (Abcam, ab4647, 1:2000) for 18 h. For

of

immunoperoxidase staining, subsequent incubation with an appropriate biotinylated

ro

secondary antibody (Jackson, 1:200) followed by incubation with an avidin peroxidase

-p

solution (Vector) was used to yield a brown reaction product after incubation with 3,3-

re

diaminobenzidine (DAB, Sigma D5637). Immunostained sections were counter stained with methyl green (Sigma, 67060), dehydrated, cover slipped using DPX Mountant

lP

(Sigma), and imaged using an Olympus BX51 microscope (Olympus, Center Valley,

na

PA).

Immunofluorescence staining was achieved by incubating with appropriate

ur

fluorochrome-conjugated secondary antibodies (Jackson, 1:200) and counterstaining

Jo

with 4’,6’-diamindino-2-phenylindole (DAPI). Cell counting and thresholding of images from the dorsal column of the spinal cord and corpus callosum of the brain was performed in ImageJ (NIH) without knowledge of treatment groups using cropped regions of interest averaged from 2-3 sections per animal with at least n= 4 per group. 2.1.5 Western blotting One half of the spinal cord (cervical and thoracic) was homogenized in radioimmunoprecipitation buffer and 35 µg protein per sample was resolved by SDS-PAGE (Bio-Rad Laboratories, Hercules, CA) and electroblotted as described previously [29].

Journal Pre-proof

9

Membranes were probed for AMPK (Cell Signaling, 2532S), pAMPK (Cell Signaling, 2531S), SIRT1 (Abcam, Ab121193), PGC1α (Abcam, Ab54481), and β-actin (Novus Biological, NB600-501). Detection was accomplished using appropriate HRPconjugated secondary antibodies (GE Healthcare), Pierce chemiluminescent developer (ThermoFisher), and ImageLab 2.0 Software (Bio-Rad). Relative optical density (ROD) for each protein was normalized to the actin of the same membrane.

of

2.1.6 RNA isolation, qPCR, and sequencing

ro

Total RNA was isolated from half of the spinal cords or primary murine cells

-p

using RNA Stat60 (Tel-Test, Friendswood, TX) per manufacturer’s instructions and

re

used for qPCR using BioRad iTaq Universal Probes (ThermoFisher) or primers (Integrated DNA Technologies) and SYBR Green One-step kits. RNA expression of

lP

each gene was normalized to Rn18s.

na

RNA libraries were prepared using 200 ng of total RNA according to the manufacturer’s instructions for the TruSeq RNA Sample Prep Kit v2 (Illumina, San

ur

Diego, CA). The concentration and size distribution of the completed libraries was

Jo

determined using an Agilent Bioanalyzer DNA 1000 chip (Santa Clara, CA) and Qubit fluorometry (Invitrogen, Carlsbad, CA). Libraries were sequenced at 100 million reads per sample following Illumina’s standard protocol using the Illumina cBot and HiSeq 3000/4000 PE Cluster Kit. The flow cells were sequenced as 100 X 2 paired end reads on an Illumina HiSeq 4000 using HiSeq 3000/4000 sequencing kit and HCS v3.3.20 collection software. Base-calling was performed using Illumina’s RTA version 2.5.2. One regular diet-fed mouse was excluded because of poor sequencing.

Journal Pre-proof

10

mRNA-seq data was processed by the Mayo Bioinformatics Core Facility to identify genes with differential expression among groups. MAP-RSeq [31] workflow (v1.2.1.3) was used to process mRNA-seq data, including read alignment, quality control, gene expression quantification and finally summarizing the data across samples. Paired-end reads were aligned by TopHat (v2.0.12) against the mouse genome build (mm10) using the bowtie1aligner [32], and the gene counts were

of

generated using Subread package [33] (v1.4.4). The workflow also provides detailed

ro

quality control metrics across genes using the RSeQC [34] (v2.3.2).

-p

Differential expression analysis was performed with edgeR [35] package (v2.6.2)

re

to identify genes with altered expression. A cutoff for false discovery rate-adjusted pvalue was set at < 0.05 to determine the genes with significant expression changes

lP

between conditions. The differential genes were further submitted to Ingenuity Pathway

na

Analysis (IPA®) for pathway enrichment analysis and causal network analysis. For causal network analysis (also described as “master regulator analysis”), only directed

ur

and experimentally observed interactions with a false discovery rate of less than 0.1

Jo

were used. The overall heatmap represents RPKM values representing the entire gene profile differences between groups irrespective of FDR organized by hierarchical clustering with the row distance of one minus Pearson correlation, while the scatterplot depicts only significantly differentially expressed genes based on a FDR of less than 0.15. Heatmaps derived from select PantherGo Pathways as well as the scatterplots from 500 gene sets for each cell type [36] were represented regardless of FDR. 2.1.7 Qualitative large-scale metabolite profiling and data analysis

Journal Pre-proof

11

Untargeted metabolomics analysis was performed by the Metabolomics Resource Core Facility at Mayo Clinic in spinal cord tissues of RD or HFD-fed animals by adapting previously described methods [37]. At study endpoints, spinal cords were quickly removed by flushing PBS through the vertebral column, split in half vertically and immediately snap frozen on dry ice for subsequent metabolic and transcriptomic analyses. Briefly, spinal cord samples that were stored at -80 C were pulverized in liquid

of

nitrogen using a custom mortar and pestle into a fine powder. Accurate weight was

ro

obtained using an analytical balance making sure the pulverized sample remained

-p

frozen. Pulverized samples were kept at -80 0C. Before metabolite extraction, samples

re

were thawed on ice for 15 min and 1X PBS was added to make a 100 mg/ml homogenate. An aliquot of 50 µl (~ 5 mg tissue) was used for metabolite extraction. 13

lP

C6-phenylalanine (2 µl at 250 ng/µl) was added as internal standard (IS) to all samples

na

and tubes were sonicated in the ice bath for 2 min (30 sec x 4 pulses). Samples were then deproteinated with cold acetonitrile: methanol (1:6 ratio), kept on ice with

ur

intermittent vortexing and centrifuged at 18000xg for 30 min at 4 C. The supernatant

Jo

was divided into 2 aliquots and dried down using a stream of nitrogen gas for analysis on a Quadruple Time-of-Flight Mass Spectrometer (Agilent Technologies 6550 Q-TOF) coupled with an Ultra High Pressure Liquid Chromatograph (1290 Infinity UHPLC Agilent Technologies). Profiling data was acquired under both positive and negative electrospray ionization conditions over a mass range m/z of 100 - 1700 at a resolution of 10,000 (separate runs) in scan mode. Metabolite separation was achieved using two columns of differing polarity, a hydrophilic interaction column (HILIC, ethylene-bridged hybrid 2.1

Journal Pre-proof

12

x 150 mm, 1.7 mm; Waters) and a reversed-phase C18 column (high-strength silica 2.1 x 150 mm, 1.8 mm; Waters) with gradient described previously [37]. Run time was 18 min for the HILIC and 27 min for the C18 column using a flow rate of 400 µl/min. A total of four runs per sample were performed to give maximum coverage of metabolites. Samples were injected in duplicate, wherever necessary, and a pooled quality control (QC) sample, made up of all of the samples from each study was injected several times

of

during a run. A separate plasma quality control (QC) sample was analyzed with pooled

ro

QC to account for analytical and instrumental variability. Dried samples were stored at -

-p

20 C until analysis. Samples were reconstituted in running buffer and analyzed within

re

48 h of reconstitution. Auto-MS/MS data was also acquired with pooled QC sample to aid in unknown metabolite identification using fragmentation pattern. The tissue pellet

lP

from pulverized samples remaining after the extraction was stored at -80 C.

na

Total protein content from the tissue pellet was determined using Pierce BCA protein assay kit using a Spectramax Plus microplate reader. Briefly, the tissue pellet,

ur

after metabolite extraction, was solubilized in 0.3 M sodium hydroxide and 10 µl lysate

Jo

was used to determine total protein concentration. Protein determination was performed in triplicate and bovine serum albumin (BSA) was used to generate the standard curve. Data alignment, filtering, univariate, multivariate statistical and differential analysis was performed using Mass Profiler Professional (Agilent Inc, USA). Metabolites detected in at least ≥ 80% of one of two groups were selected for differential expression analyses. Metabolite peak intensities and differential regulation of metabolites between groups were determined as described previously [37]. Each

Journal Pre-proof

13

sample was normalized to the protein content and log2 transformed. Unpaired t-test with multiple testing correction p < 0.05 was used to find the differentially expressed metabolites between two groups. Missing values are excluded from the analysis for the calculation of fold change and p-values. Default settings were used with the exception of signal-to-noise ratio threshold (3), mass limit (0.0025 units), and time limit (9 s). Putative identification of each metabolite was done based on accurate mass (m/z)

of

against METLIN database using a detection window of ≤ 7 ppm. The putatively

ro

identified metabolites were annotated as Chemical Abstracts Service (CAS), Kyoto

-p

Encyclopedia of Genes and Genomes (KEGG), Human Metabolome Project (HMP) database, and LIPID MAPS identifiers. The differentially expressed metabolites are

re

analyzed for pathway enrichment using MetaCore (Genego, St. Joseph, MI) – this

lP

identifies pathways using 3 metabolite identifiers – that is, KEGG, CAS and HMDB.

na

2.1.8 Targeted TCA cycle metabolomics

Spinal cord levels of tricarboxylic acid (TCA) cycle metabolites were measured

ur

using the same samples prepared for untargeted metabolomics after the derivatization

Jo

with N-methyl-N-(t-butyldimethylsilyl)-trifluoroacetamide + 1% tbutyldimethylchlorosilane by gas chromatography/mass spectrometry under electron impact and selected ion monitoring conditions as previously described [37]. All measurements were performed against 12-point calibration curves that underwent the same derivatization with internal standard. 2.1.9 Mitochondrial functional and structural analyses Oroboros oxygraph O2k was used for high resolution respirometry readings from isolated spinal cord mitochondria and Seahorse extracellular flux assay (Agilent) was

Journal Pre-proof

14

used to assess mitochondrial function in cell cultures as described in more detail in our previous publications [38]. Oxygen consumption rates (OCR) were normalized to BCA protein estimation. In brief, fresh whole spinal cord tissue was placed in mitochondria isolation buffer (225 mM Mannitol, 75 mM sucrose, 20 mM MOPS, 1 mM EGTA, 0.5 mM DTT) containing 0.05% Nargase (w/v)/1g tissue and homogenized for 10 min. Next, differential centrifugation was used to isolate mitochondria from other fractions, the

of

mitochondria pellet was resuspended in mitochondria isolation buffer with 0.02%

ro

Digitonin for 10 min, then centrifuged and resuspended again in fresh buffer (125 µl/100

-p

mg tissue) for running the assay. 50 µl of isolated mitochondria suspension was run for

re

each of two technical duplicates. The oxygraph chamber was pre-washed and filled with respiration buffer (120 mM mannitol, 40 mM MOPS, 5mM KH 2PO4, 0.1mM EGTA,

lP

60mM KCl, 5 mM MgCl2) to equilibrate and oxygenate for at least 30 minutes. Oxygen

na

calibrations for ambient air and zero were acquired for each chamber during each run. Oxygen consumption rates were measured in DatLab (Oroboros) after adding the

ur

mitochondria suspension (state I), glutamate (10 mM)/Malate (2 mM)/succinate (10 mM)

Jo

mixture (state II), ADP (14.25 mM), oligomycin (2 µg/ml), FCCP (0.005 mM) titrations, and antimycin A (2.5 mM) using Hamilton syringes. State 3 mitochondrial respiration was assessed following the addition of glutamate, malate, and succinate and ADP to the chamber, giving the basal respiration rate for complexes I and II. Oligomycin injection then inhibited ATP synthase to induce state 4 respiration, attributable to proton leak across the inner mitochondrial membrane. FCCP addition results in an uncoupled state, allowing for determination of maximum capacity, while non-mitochondrial respiration was assessed after antimycin inhibits complex III. State III respiration measurements

Journal Pre-proof

15

were taken from a stable segment following the addition of ADP, while State IV measurements followed oligomycin injection. Maximum respiration was recorded during the highest segment of uncoupled respiration following the FCCP titrations, and nonmitochondrial respiration was measured after anitmycin A addition. All measurements were normalized to protein content of the mitochondria fraction. The Seahorse XF24 (Agilent) extracellular flux assay was used to monitor

of

oxygen consumption rates at various stages of respiration in primary murine

ro

oligodendrocytes as previously described [39]. Following cartridge equilibration with

-p

injection ports filled with oligomycin (1 μg/ml), FCCP (1 μM), and antimycin A (10 μM), a

re

plate pre-treated in serum-free media was placed into the Seahorse analyzer. OCR readouts at each stage were measured in pmol/min and normalized to protein.

lP

For structural analysis of mitochondria, 0.09 x 106 cells primary oligodendrocytes

na

were grown per well on poly-L-lysine (PLL, 10 μg/ml) coated glass coverslips in 24-well plates, treated with 0 or 100 μM palmitate (PA) for 24 h, and then washed twice with

0

ur

fresh media. CMXRos MitoTracker Red (ThermoFisher, 100 nM) was incubated at 37

Jo

C for 30 min. Immunocytochemistry procedures and DAPI counterstaining were then

performed. ImageJ was used to quantify mitochondrial parameters as described previously [39, 40]. Averaged values are reported from analyzed cells within five separate images per coverslip. 2.1.10 Primary glia isolation, OLC culture Mixed glial preparations from postnatal day 0-3 murine cerebral cortex were cultured in DMEM (Fisher 11960-044) with 1 mM sodium pyruvate (Corning 11360070), 20 mM HEPES (Gibco 15630-080), 100 U/ml PenStrep (Life Technologies 15140122),

Journal Pre-proof

16

and 10% fetal bovine serum (FBS, Atlanta Biologicals S11150) for 10-12 d. Oligodendrocytes, astrocytes, and microglia cultures were separated by differential adhesion/shaking overnight at 225 rpm on an Innova 2000 orbital shaker as previously described [41]. For oligodendrocyte treatments and differentiation, 0.09 x 10 6 cells were plated on each well of PLL-coated coverslips or 24 well plates and grown in Neurobasal A

of

media (Life Technologies, 10888022) with N2 (17502048), B27 (17504044), PenStrep,

ro

sodium pyruvate, 2 mM glutamax (35050-061), and 5% BSA (Sigma A4503) for two d

-p

before treating with palmitate (PA, 100 µM, 24 h). This dose was lower than toxic

re

amounts previously reported in other cells types at the same time point [42, 43] and did not result in significant lipotoxicity to oligodendrocytes or aNSCs. RNA levels for MBP,

lP

PLP, and Olig2 were determined in technical duplicates using and an iCycler iQ5

na

system (BioRad) with primers identified within the Key Resources Table. Relative starting quantities were normalized to Rn18S and reported as percent of control. PLP

ur

protein levels were determined based on threshold of immunoreactivity from five fields

Jo

per coverslip quantified in ImageJ. 2.1.11 Adult neural stem cell culture Adult neural stem cells were isolated from the subventricular zone (SVZ) of 8 wk old C57Bl6/J mice as previously described [44]. Briefly, mice were anesthetized with sodium pentobarbital and decapitated to collect coronal brain sections and further dissect stem cells from within the SVZ. Collected cells were dissociated with Accutase and grown in DMEM/F12 with B27, antibiotic-antimycotic, insulin (20 µg/mL), epidermal

Journal Pre-proof

17

growth factor (EGF, 20 ng/mL) and basic fibroblast growth factor (bFGF, 20 ng/mL, PeproTech, Rocky Hill, NJ) in T75 tissue culture treated flasks as neurospheres. For myelin RNA and protein expression experiments, cells were dissociated and plated onto poly-L-ornithine (20 µg/ml) and laminin (10 µg/ml) coated 6 well plates and glass coverslips, respectively. To induce differentiation toward oligodendrocyte lineage, cells were switched to media containing Neurobasal A, antibiotic-antimycotic, B27,

of

insulin (20 µg/mL), T3 (60 ng/ml), glutamax (2 mM) and cultured for an additional 72 h.

ro

To identify effects of PA (100µM) on differentiation to oligodendrocyte lineage, we

-p

compared protein and RNA marker expression to controls by immunofluorescence

re

staining and qRT-PCR. 2.1.12 Statistical analysis

lP

Data were tested for normality using the Shapiro-Wilk test, graphed, and analyzed by

na

Student’s t-test unless noted otherwise in GraphPad Prism 7.0 software. Mann-Whitney nonparametric test was used for histological analysis of 4-HNE immunoreactivity in

ur

brain and spinal cord tissue, while body weight, food intake, and glucose tolerance test

Jo

in mice were analyzed by two-way ANOVA with Bonferroni post-test. p-values ≤ 0.05 were considered significant.

Journal Pre-proof 2.2 KEY RESOURCES TABLE

Abcam

ab28486; AB_776593

AB12193; AB_298923 74.5A5; AB_531794

ab5320; AB_11213678

lP

na

Jo

rabbit anti-4hydroxynonenol rabbit anti-cleaved caspase-3 rabbit anti-Bcl-2 rabbit anti-Neurofilament biotinylated goat anti-rat secondary biotinylated donkey antirabbit secondary biotinylated donkey antimouse secondary goat-anti-rabbit AF488 secondary goat anti-rat Cy3

2531S; AB_330330

Millipore

MAB386; AB_94975

Abcam Novus Biological Abcam

Ab54481; AB_881987 NB600-501; AB_10077656

Cell Signaling Technology Abcam Sigma Aldrich Jackson

9664; AB_10694088

Jackson

711-065-152; AB_2340593

Jackson

715-066-151; AB_2340788

Jackson

111-545-047; AB_2338051

Jackson

112-166-072; AB_2338257

ur

anti-Neural glial antigen2 (NG2) rabbit anti-Proteolipid protein (PLP) rat anti-Myelin basic protein (MBP) rabbit anti-PGC1-α mouse anti-ᵝ-actin

MABN50; AB_10807410 MA514003; AB_10979735 Ab9610; AB_10141047 Ab16794; AB_443473 2532S; AB_330331

re

mouse anti-SIRT1 anti-Nkx2.2

Millipore Invitrogen Millipore Abcam Cell Signaling Technology Cell Signaling Technology Abcam Development al Studies Hybridoma Bank, University of Iowa, Iowa city, IA Millipore

of

rabbit anti-pAMPK

Identifier

ro

Antibodies mouse anti-Olig2 rabbit anti-BAX rabbit anti-Olig2 mouse anti-CC-1 rabbit anti-AMPK

Source

-p

Reagent or Resource

ab46545; AB_722490

AB59348; AB_2064155 N4142; AB_477272 112-066-072; AB_2338185

18

Journal Pre-proof

FIS1

PINK1

SOD2 GFAP

SOD2 Rn18s

Primary isolation (Yoon 2015) Primary isolation (Choi 2017)

N/A

The Jackson Laboratory

N/A

of

N/A

NM_016967; Mm.PT.58.42319010

lP

Applied Biosystems Integrated DNA Technologies Integrated DNA Technologies Applied Biosystems Integrated DNA Technologies Integrated DNA Technologies

ur Jo

PGC1-α

711-166-152; AB_2313568

na

MBP

PLP

Jackson

re

oligonucleotides, (primers/probes) OLIG2

111-545-041; AB_2341130

ro

Experimental models: cell lines Mouse:C57BL/6 oligodendrocyte lineage cells (OLCs) Mouse:C57BL/6 adult neural stem cells (aNSCs) Experimental models: organisms/strains Mouse: C57BL/6J

Jackson

-p

secondary goat anti-rabbit AF488 secondary donkey anti-rabbit Cy3 secondary

19

Integrated DNA Technologies Applied Biosystems Applied Biosystems

NM_001025251; CCAGTAGTCCATTTCTTCAAGAACAT/GC CGATTTATAGTCGGAAGCTC NM_011123.2; TCTTTGGCGACTACAAGACCAC/CACAA ACTTGTCGGGATGTCCTA Mm01208835_m1 Mm.PT.56a.21878911

Mm.PT.58.23711353

Mm01313000_m1 NM_010277.2; GCAGATGAAGCCACCCTGG/ GAGGTCTGGCTTGGCCAC NM_013671.3; Mm01313000_m1 NR_003278.3; Mm03928990_g1

Journal Pre-proof

Chemicals, Peptides, and recombinant proteins palmitic acid dapi Mitotracker Red

20

Sigma Aldrich Cat#P0500 ThermoFisher Cat#D1306; AB_2629482 ThermoFisher Cat#m7512

Commercial kits Seahorse XF Cell MitoStress Test Pierce BCA protein estimation

Agilent

Cat#103015-100

Other Regular diet (10 kcal % fat) High fat diet (60 kcal % fat)

Research Diets Research Diets

https://www.graphpad.com/scientificsoftwar e/prism https://imagej.net/Fiji

D12450K D12492

Jo

ur

na

lP

GraphPad Prism 7

https://www.med-associates.com/

-p

Fuji ImageJ

Med Associates Inc GraphPad Software NIH

re

Software and Algorithms Activity Monitor Software

ro

of

ThermoFisher Cat#23225

2.3 CONTACT FOR REAGENT AND RESOURCE SHARING Additional information and requests for resources can be directed to the corresponding author/Lead Contact, Isobel Scarisbrick ([email protected]).

Journal Pre-proof

21

3. RESULTS 3.1 Chronic high fat diet consumption impairs metabolic parameters and diminishes the number of oligodendrocytes and oligodendrocyte progenitors in the central nervous system Since our prior studies revealed that a diet high in fat and sucrose results in a loss of myelinating cells and their progenitors in the spinal cord [29], we chose to

of

investigate the differential impact of high fat alone and any temporal effects by feeding

ro

10 wk old male C57Bl/6 mice a regular diet (RD, 10% of kcal from fat) or high fat diet

-p

(HFD, 60% kcal from fat) for either 4 or 12 wk (Figure 1A). As expected, the HFD mice appeared noticeably larger (Figure 1B) compared to the RD-fed controls and consumed

re

more kcal of food per day (Figure 1C). Beginning at 3 wk through 12 wk, mice also

lP

gained a significant amount of weight (Figure 1D). After 3 wk, glucose sensitivity was

na

assessed using a glucose tolerance test. As expected, HFD-fed mice demonstrated significantly higher fasting blood glucose levels and impaired glucose tolerance (Figure

ur

1E, F) [30]. A set of RD and HFD mice were monitored in a Comprehensive Lab Animal

Jo

Monitoring System (CLAMS) to reveal significant changes in several metabolic parameters including percent fat/lean mass, VO2, VCO2, respiratory exchange rate (RER), and metabolic rate (Supplemental Figure 1). In a separate cohort, a number of movement measurements were also affected by HFD consumption including anxietylike behavior and decreased vertical motion at 4 wk, and also involving horizontal activity by 12 wk (Supplemental Figure 2). After establishing the systemic and behavioral effects of HFD consumption in mice, we next tested the direct consequences on oligodendrocyte lineage cells (OLCs).

Journal Pre-proof

22

At study endpoints, spinal cord tissues were separated into cervical-thoracic and lumbosacral segments for analysis as depicted in (Figure 1G). The impact of consuming a HFD on myelin producing cells was determined by counting OLC markers in the postfixed lumbosacral spinal cord of each group. Oligodendrocyte progenitor cells (OPCs) were identified using antibodies recognizing NG2, Nkx2.2, and Olig2 (oligodendrocyte lineage transcription factor 2, marks progenitors and early mature cells), while mature

of

oligodendrocytes were considered CC-1+. After 4 wk of HFD, no change in the number

ro

of Olig2+ cells was observed in the spinal cord dorsal column (Figure 1H). RNA

-p

expression of oligodendrocyte markers Olig2 (Figure 1I), myelin basic protein (MBP,

re

Figure 1J), and proteolipid protein (PLP, Figure 1K) remained unchanged after 12 wk of HFD-consumption. By contrast counts of oligodendrocyte progenitors immunoreactive

lP

for Nkx2.2 (Figure 1L, 40x, and Supplemental Figure 3A , 20x), NG2 (Figure 1 M, 40x,

na

and Supplemental Figure 3B, 20x) and Olig2 (Figure 1N, 40x, and Supplemental Figure 3C, 20x) were all reduced at 12 wk. Furthermore, counts of CC-1+ mature

ur

oligodendrocytes (Figure 1O, 40x, and Supplemental Figure 3D, 20x) demonstrated a

Jo

significant depletion. We did not observe a significant change in Olig2 (Supplemental Figure 4A, B), in MBP isoforms (Supplemental Figure 1A, C and Supplemental Figure 5 E-H) or PLP (Supplemental Figure 5 A,D) protein expression by Western blot. Considering we observed a loss of oligodendrocytes and their progenitors in the spinal cord at 12 wk of HFD consumption, we next examined any regional specificity by parallel quantification in the brain. The corpus callosum (Figure 1P) from mice fed a HFD had fewer Olig2+ cells (Figure 1Q) at 12 wk, but staining for MBP (Supplemental Figure 3E), a marker of myelin structural integrity, and mature oligodendrocytes (CC-1+,

Journal Pre-proof

23

Supplemental Figure 3F) were unaffected. Taken together, these findings suggest that oligodendrocyte progenitors in both the brain and spinal cord are vulnerable to death in response to chronic HFD consumption; however, only spinal cord mature oligodendrocytes appeared susceptible to HFD-induced toxicity at the timepoint examined. Interestingly, remaining oligodendrocytes within both central nervous system

Jo

ur

na

lP

re

-p

ro

of

(CNS) regions maintained normal PLP and MBP levels at all time points examined.

Jo

ur

na

lP

re

-p

ro

of

Journal Pre-proof

24

Journal Pre-proof

25

Fig 1. Chronic high fat diet consumption impairs metabolic parameters and diminishes the number of oligodendrocytes and progenitors in the central nervous system. A, Schematic depicting experimental design in which 10 wk old male mice were fed a regular diet (RD) or a high fat diet (HFD) for 4 or 12 wk. Representative images show mice consuming a HFD gained weight (B) after consuming more kcal food per day (C, n=3 cages of 5 mice per group). HFD mice gained weight (D, n=10 per

of

group. RD 25.84 g and HFD 24.97 g at study start; RD 27.53 g and HFD 43.02 g at

ro

study end) and had impaired glucose tolerance after 3 wk (E, F; AUC=area under the

-p

curve, n=8 mice per group). Graphical results represented as the mean ± SEM. G, Schematic shows spinal cord regions used in analyses. H, Representative images from

re

immunostained sections of spinal cord dorsal column and counts of the number of

lP

Olig2+ cells at 4 wk (n=5 mice per group). RNA expression of Olig2 (I), MBP (J), and

na

PLP (K) at 12 wk (n=4 mice per group). Representative images of dorsal column and corresponding counts of Nkx2.2+ (L), NG2+ (M), Olig2+ (N), and CC-1+ (O) cells after

ur

12 wk (n=7 mice per group). P, Schematic depicting region of corpus callosum

Jo

examined for immunostaining. Representative images from immunostained sections of the corpus callosum (Q) and counts of Olig2+ cells at 12 wk (n=5 mice per group). Bar graphs shown represent the mean number of cells per area of spinal cord ± SEM and are expressed as percent of RD-fed control mice. (*, p<0.05; **, p<0.01; and ***, p<0.001). Scale bar = 20 μm (L-O) and 50 μm (Q).

Journal Pre-proof

26

3.2 Chronic high fat diet consumption differentially regulates the transcriptional profile of the adult mouse spinal cord To begin to address the mechanisms driving oligodendrocyte depletion in the face of HFD-consumption, we performed RNA sequencing from half of each spinal cord as demonstrated in Figure 2A. An overall heatmap (Figure 2B and Supplemental Figure 4) depicts differential gene expression in the spinal cords of mice consuming a RD (n=3)

of

and HFD (n=4) for 12 wk, where blue is the relative row minimum and red is the relative

ro

row maximum. Using a false discovery rate (FDR) of P < 0.15, a scatter plot of

-p

differentially expressed genes (DEGs, Figure 2C) showed 75 to be transcriptionally

re

distinct between diet groups (32 up and 43 down). Among these, we observed an upregulation of Hba-a2, Sdf2L1, Hmgcs2, Creld2, Cryab, Hspa5/Grp78, Grn, Kdr,

lP

Ddit4l, Fos, Xbp1, Paqr6, Fn14, Gstp1, Ccdc42ep2, and Litaf. Significantly

na

downregulated genes included Lcn2, Ch25h, Sgk1, Slc17a7, Pdyn, Socs3, Plaur, Dbp, Acer2, Pla2g3, Kirrel2, Cp, Fn1, and Klf9.

ur

A number of ER stress induced genes including Creld2, Xbp1, and Hspa5 were

Jo

upregulated in HFD spinal cord. Not surprisingly, ingenuity canonical pathway analysis revealed ER Stress and the Unfolded Protein Response pathways as top pathways affected in spinal cord tissue by HFD consumption (Table 1). Next, to identify transcription factors which may act as master regulators for the observed transcriptional changes, we performed a causal network analysis with only directed and experimentally observed interactions with a FDR less than 0.1 (Table 2). PTK6 (Figure 2D), predicted to be activated, and NFKBIA (Figure 2E), predicted as an inhibited upstream transcriptional regulator, influence expression of Xbp1 and HSPA5, respectively.

Journal Pre-proof

27

Moreover, PDGFRA is a crucial mediator of oligodendrogenesis [45] and was predicted to be inactivated. Loss of Mecp2 in oligodendrocytes down regulates myelin gene expression and was predicted to have lower activation in the HFD mice [46]. We next focused attention on expression of myelin and cholesterol related genes using PantherGO Pathways (Figure 3A), where there was surprisingly a trend for increases in a number of genes, which may reflect a compensatory response in

of

remaining oligodendrocytes and explain why frank loss of myelin was not observed at

ro

the 12 wk endpoint despite the loss of mature myelinating cells and progenitors. Having

-p

characterized overall transcriptomic differences between the spinal cord of mice

re

consuming a RD or HFD, we next used the gene sets described by Zhang et al [36] to profile transcriptional changes enriched at distinct stages of oligodendrocyte lineage

lP

progression (Figure 3B-E), including oligodendrocyte precursors (OPCs, Figure 3C),

na

new OLCs (Figure 3D), and myelinating OLCs (Figure 3E)[36]. Of the OPC-enriched gene set, Slc6a3 (p=0.27), Lox (p=0.18) , Cdc25c (p=0.16), Snora17 (p=0.015), Bglap2

ur

(p=0.79), Gsx1 (p=0.07), Ccnb1 (p=0.18), Alas2 (p=0.10) , Raet1d, and Troap (p=0.19)

Jo

were all increased above 1.5-fold, while Ddx43 (p=0.002) and Cpz were below 0.5-fold in the spinal cord of mice consuming HFD relative to RD (Figure 3C, all genes included regardless of FDR). New OL gene changes included increased Krt28 (p=0.10), S100a14 (p=0.19), Snora26 (p=0.25), 1700001P01Rik (p=0.10), and Sis (p=0.08) and decreased Erp27 (p=0.18), Corin (p=0.09), and Chrng (Figure 3D). In myelinating OLs, Hist1h2bg (p=0.40), 1700001P01Rik (p=0.10), A930003A15Rik (p=0.09), Hist1h2ae (p=0.43), Hist1h4i (p=0.04), and Hist1h2ac (p=0.34) were increased, while Hist1h2bn (p=0.006), Erp27 (p=0.18), and Mpl (p=0.04) were decreased (Figure 3E).

Journal Pre-proof Together, these results suggest that HFD consumption promotes complex transcriptional changes in the spinal cord and in oligodendrocyte lineage cells that include pathways essential for maintaining homoeostasis and highlight several

Jo

ur

na

lP

re

-p

ro

of

important new targets for further investigation.

28

Jo

ur

na

lP

re

-p

ro

of

Journal Pre-proof

29

Journal Pre-proof

30

Fig 2. Transcriptional changes in the spinal cord of adult high fat-fed mice. A, One half of the spinal cord from regular diet (RD, n=3) and high fat diet (HFD, n=4) fed mice were processed whole genome RNA sequencing. B, Heat map of transcriptional changes observed in the spinal cord of male mice fed RD or HFD for 12 wk representing the entire gene profile differences between groups irrespective of FDR, organized by hierarchical clustering with the row distance of one minus Pearson

of

correlation where blue is relative row minimum and red is the row maximum. C, Scatter

ro

plot of differentially expressed genes (DEGs, red dots; 32 up, 43 down with a FDR <

-p

0.15) where the x-axis is false discovery rate (FDR) and y-axis is log fold change (log FC), with annotated gene names for select up (red) and down (blue) regulated genes.

Jo

ur

na

lP

upstream transcriptional regulator.

re

PTK6 (D) was predicted to be activated while NFKBIA (E) was predicted as an inhibited

2.67

2.55E-06

MYOCD

Transcription regulator

3

2.12

31

Target molecules in dataset

Network biascorrected p-value

Predicting activation Z-score

3

0.013

CDC42EP2, CRYAB, GNA12, GSTP1,HBA1/HBA2,H MGCS2,KDR, PDIA6,XBP1 ARL4D, COL8A2,CP, FBN1,FN1, GRN, LCN2,LFNG,

-p

ro

of

Molecule Type Kinase

Exp Log Ratio

PTK6

Master Regulator

Depth

P-value of overlap

Journal Pre-proof

0.012

2

5.69E-04

0.015

HSPA5,PDIA6,SDF2L1, FN1

3

2

1.76E-04

0.022

HSPA5,KDR,SGK1, FN1

2

1.89

3.92E-05

0.026

ARC, GSTP1,KDR,LCN2, COL8A2,FBN1,FN1

transcription regulator

2

1.73

1.06E-03

0.042

KDR,SGK1, FN1

0

transcription regulator

3

1.5

7.52E-05

0.033

ARC, CRYAB, HBA1/HBA2,HMGCS2, HSPA5,KDR, SERPINB1,TNFRSF12 A,XBP1, CH25H,COL8A2, DBP,FBN1,GRN, LAMA4,LRG1

0.04

transcription

1

1.41

6.47E-05

0.0032

HSPA5,XBP1

0.38

Transcription regulator

TAF12

0.04

transcription regulator

SRF

0.11

transcription regulator

NFYC

-0.03

CCNE1

CREB3

1

Jo

ur

na

XBP1

lP

re

8.00E-06

ARC,GSTP1,KDR, XBP1, COL8A2,FBN1,FN1, LCN2,

Journal Pre-proof

32

regulator -0.07

transcription regulator

3

1.41

6.39E-06

0.0094

ARC,CDC42EP2,GSTP 1,KDR,LCN2, COL8A2,FBN1,FN1

ATF6B

0.10

transcription regulator

1

1

6.30E-03

0.0318

XBP1

NFYC

-0.03

transcription regulator

1

1

1.67E-02

0.0449

SGK1

PIK3CA

-0.06

kinase

3

1

1.32E-05

0.049

ARC, CDC42EP2, CRYAB, GNA12,GSTP1,HMGCS 2,HSPA5, PDIA6, XBP1, ARL4D, CIART,CP, DBP, LCN2,LFNG, SGK1

DDIT3

0.13

transcription regulator

1

0.58

PRKAB1

0.00 5

kinase

PRKAG1

0.00 2

RPS6KA 4

-0.04

PRKACA

0.07

2

0.45

1.11E-04

0.0435

HMGCS2, DBP,FN1, LCN2,SGK1

kinase

2

0.45

1.11E-04

0.0435

HMGCS2, DBP,FN1, LCN2,SGK1

kinase

2

0.33

6.49E-06

0.0083

ARC,GSTP1,HSPA5, ,CIART,COL8A2,FBN1, FN1, LCN2,SGK1

chemical endogenous mammalian

3

0.33

3.15E-05

0.0162

ARC,GSTP1,HSPA5, PDIA6, XBP1, CIART,FN1, LCN2, SGK1

kinase

3

0.23

7.34E-06

0.032

ARC, CRYAB,GSTP1,HMGC S2,HSPA5,KDR, SERPINB1, ARL4D,CH25H,CIART, COL8A2,CP,

ur

na

lP

re

heme

0.008

Jo

-p

ro

of

MKL1

1.60E-04

HSPA5, XBP1, LCN2

Journal Pre-proof

33 DBP,FBN1,FN1,GRN, LCN2,LFNG,Podxl

CLOCK

0.02

ARNTL

0.28

PPRC1

-0.05

SMARCA 2

-0.02

kinase

3

0

3.62E-07

0.0008

1

0

3.33E-04

0.0049

HMGCS2, DBP

1

0

0.0184

HMGCS2, DBP

4.05E-03

0.0319

GNG11,LCN2

2.20E-03

0.0344

KDR, CP

0.0153

CDC42EP2, CRYAB, GNA12, HBA1/HBA2,HSPA5,K DR, PDIA6,SERPINB1, XBP1, CH25H,COL8A2,CP, FBN1,FN1, GRN, LCN2,LFNG, SGK1

0.0126

ARC, CDC42EP2, CRYAB,GNA12, GSTP1,HBA1/HBA2,H MGCS2,HSPA5,KDR, PDIA6, SDF2L1,SERPINB1, XBP1, ARL4D, CH25H,CIART,CP, DBP,FN1, GRN, LAMA4,LFNG, PLA2G3, SGK1

re 0

lP 1

na

-0.12

1

kinase

3

2.07E-03

0

-0.23

1.19E-06

Jo

ur

PDGFRA

transcription regulator transcription regulator transcription regulator transcription regulator

ro

of

-0.07

-p

MAP3K3

ARC,CDC42EP2, ,CRYAB,GNA12,HSPA 5,SDF2L1,SERPINB1,T NFRSF12A, ARL4D, ,CH25H,CIART,COL8A 2,CP, ,DBP,FBN1,FN1,GRN, LCN2,LFNG,LRG1,PLA 2G3, SGK1

HTT

-0.04

transcription regulator

3

-0.41

9.99E-07

Journal Pre-proof

MECP2

-0.11

transcription regulator

1

-0.58

1.25E-04

0.0021

HBA1/HBA2,SGK1,SL C17A7

CREB1

0.05

transcription regulator

1

-0.82

2.90E-04

0.0314

ARC,HSPA5, CIART, FN1, LCN2,SGK1

1

-1

8.39E-03

0.0162

SLC17A7

-0.19

transcription regulator transcription regulator transcription regulator

1

-1

1.46E-02

0.0341

DBP

1

-1

1.67E-02

0.0408

LFNG

of

34

ARC, HSPA5, CIART, FN1, GRN, LCN2, SGK1

FEZF2 PER2 MSGN1

0.05

kinase

2

-1.13

1.43E-04

NFKBIA

-0.37

transcription regulator

1

-1.41

liganddependent nuclear receptor

1

-1.73

-p

transcription regulator

re

0.07

CRYAB ,HSPA5, CH25H, CP, FN1,GRN,LCN2, SGK1

2.27E-03

0.0453

GSTP1,HMGCS2,LCN2

3

-2

1.76E-04

0.0222

HSPA5,KDR, FN1, SGK1

6.19E-07

na

TAF11

0.0025

lP

NR1I2

0.0373

ro

PASK

ur

Table 1. Summary of ingenuity canonical pathways significantly affected in spinal cord

Jo

tissue by HFD consumption. Bold indicates increased.

Journal Pre-proof

35

Ingenuity Canonical Pathways

-log(pvalue)

log(B -H pvalue )

Endoplasmic Reticulum Stress Pathway

3.45

1.56

0.0004

0.028

2

XBP1,HSPA5

Aldosterone Signaling in Epithelial Cells

2.86

1.29

0.0014

0.051

3

CRYAB,HSPA5,SGK1

Unfolded protein response

2.63

1.29

0.002

0.051

Glutamate Receptor Signaling

2.58

1.29

0.003

Huntington's Disease Signaling

2.4

1.21

0.004

Ceramide Degradation

2.1

Sphingosine and Sphingosine-1phosphate Metabolism

1.97

#D EG s

Molecules

2

GNG11,SLC17A7

0.062

3

GNG11,HSPA5,SGK1

0.008

0.104

1

ACER2

0.94

0.011

0.115

1

ACER2

1.89

0.94

0.013

0.115

2

GNG11,PLA2G3

0.051

-p

re

lP

ur

0.99

ro

XBP1,HSPA5

na

of

FDR

2

Jo

CCR3 Signaling in Eosinophils

P-value

Ketogenesis

1.88

0.94

0.013

0.115

1

HMGCS2

Mevalonate Pathway I

1.76

0.87

0.017

0.134

1

HMGCS2

Table 2. Master regulator analysis shows significantly affected transcriptional regulators in HFD-fed mouse spinal cord. Bold indicates increased.

Jo

ur

na

lP

re

-p

ro

of

Journal Pre-proof

36

Journal Pre-proof

37

Fig 3. Myelin-related transcriptional changes in the spinal cord of adult high fatfed mice. A, Heat map of myelin-related genes in the spinal cord of male mice fed RD or HFD for 12 wk where blue indicates relative minimum value and red is the row maximum value. Genes within groups are sorted by highest to lowest average HFD relative values compared to RD, with red asterisk indicating genes with p<0.05 Schematic depicting differentiation of oligodendrocyte lineage cells (B) and scatterplots

of

of RNA expression of genes enriched in oligodendrocyte progenitors (OPCs, C), new

ro

oligodendrocytes (OL, D) or myelinating OLs (E) in HFD mice normalized relative to RD

Jo

ur

na

lP

re

-p

controls.

Journal Pre-proof

38

3.3 Functional and metabolic changes in spinal cord mitochondria following HFD consumption are time-dependent Prior to initiation of cell death, ER stress pathway activation can have a number of effects on metabolic and bioenergetic processes [28, 47, 48]. Therefore, since the ER Stress Pathway was the top ingenuity canonical pathway identified from transcriptomic profiling of spinal cord tissue, we further explored potential mechanisms for

of

oligodendrocyte loss in this model by comparing metabolite profiles from the spinal

ro

cords of mice fed a RD or HFD using untargeted UPLC-ToF-MS-based metabolomics

-p

as described previously in detail [37].

HFD consumption significantly altered 238 metabolites analyzed using an

re

unpaired t-test with Benjamini-Hochberg multiple testing correction and identified using

lP

the METLIN metabolite database. To determine which pathways were most affected by

na

the metabolites altered due to chronic HFD-consumption, we performed metabolite sets enrichment analysis (MSEA) with MetabaCore depicted with most significant p-values in

ur

red and least significant in white (Figure 4A). These results suggest that protein

Jo

biosynthesis, TCA cycle, glutathione metabolism, and the mitochondrial electron transport chain were among the significantly enriched pathways. Accordingly, we sought to validate these findings by quantifying TCA cycle metabolites at 4 and 12 wk using a targeted GC-MS-based approach [37]. No significant differences were observed at 4 wk (Figure 4B), but intermediates from the TCA cycle including oxaloacetate (p=0.04) , citric acid (p=0.003), and α-ketoglutarate (p=0.03) were strongly depleted in the 12 wk HFD spinal cord when compared to RD controls (Figure 4B, C and Supplemental Figure 7).

Journal Pre-proof

39

A separate cohort of mice was fed a RD or HFD for 4 or 12 wk to assess the functional impact on spinal cord mitochondria using Oroboros oxygraph [38]. Unexpectedly, we observed a significant increase in State 3 Respiration at 4 wk on HFD (Figure 4D), while 12 wk HFD spinal cord mitochondria had diminished State 3 oxygen consumption compared to RD (Figure 4D). Even State 4 respiration was increased at 4 wk, but declined at 12 wk of HFD consumption (Figure 4E). Maximum (Figure 4F) and

of

non-mitochondrial respiration (Figure 4G) were not altered by HFD consumption at 4 or

ro

12 wk. Taken together, these data provide evidence for temporally distinct changes in

-p

TCA cycle metabolites and mitochondrial function, where at 12 wk TCA cycle

Jo

ur

na

lP

re

metabolites are depleted and mitochondrial function is impaired.

Jo

ur

na

lP

re

-p

ro

of

Journal Pre-proof

40

Journal Pre-proof

41

Fig 4. Functional and metabolic changes in spinal cord mitochondria following HFD consumption are time-dependent. Metabolite Sets Enrichment Overview (A) summarizing significantly affected pathways in HFD-fed mice as revealed by untargeted LC-MS-MS profiling of spinal cord tissue. Heat map (B) summarizes targeted LC-MSMS profiling of tricarboxylic acid (TCA) cycle metabolites at 4 and 12 wk of RD or HFD consumption with metabolites depleted at 12 wk identified within the schematic by blue

of

arrows (C). Gray indicates metabolite was not detected in samples, red indicated

ro

highest fold expression and blue lowest fold abundance. High resolution respirometry by

-p

Oroboros oxygraph of isolated spinal cord mitochondria indicates oxygen consumption is increased in 4 wk HFD mice (D, E), but decreased by 12 wk (D, E). Injections of

re

glutamate/malate/succinate (GMS), ADP, oligomycin (O), and trifluoromethoxy

lP

carbonylcyanide phenylhydrazon (FCCP) allow for oxygen consumption rates (OCRs) to

na

be determined during various stages of mitochondrial respiration. State 3 mitochondrial respiration was assessed following the addition of glutamate, malate, and succinate to

ur

the chamber, giving the basal respiration rate for complexes I and II. Oligomycin

Jo

injection then inhibited ATP synthase to induce state 4 respiration, attributable to proton leak across the inner mitochondrial membrane. FCCP addition results in an uncoupled state, allowing for determination of maximum capacity, while non-mitochondrial respiration was assessed after antimycin inhibits complex III. OCRs during state 3 (D), state 4 (E), Maximum (F), and non-mitochondrial respiration (G). Graphical results represented as the mean ± SEM (*, p<0.05; **, p<0.01; and ***, p<0.001; n=4-6/group).

Journal Pre-proof

42

3.4 Altered mitochondrial quality control processes, oxidative stress, and apoptosis in the spinal cord following 12 wk of high fat diet consumption. HFD consumption was previously shown to alter the metabolic regulators AMPK and SIRT1 in peripheral tissues and to affect mitochondrial function and quality control processes [49, 50]. Therefore, we next explored this signaling pathway (Figure 5A) in the spinal cord of RD and HFD-fed mice. With 12 wk of HFD, immunoblotting

of

demonstrated reductions in AMPK phosphorylation (Figure 5B, C) and in total AMPK

ro

(Figure 5B, C). SIRT1 protein levels (Figure 5B, C) were also significantly reduced in

-p

the spinal cord of HFD mice compared to controls. PGC1α, a direct target of AMPK and

re

SIRT1 and crucial regulator of mitochondrial biogenesis , showed diminished protein levels (Figure 5B, C) and RNA expression (Figure 5D) in the HFD-fed mice compared to

lP

RD-fed controls. There was also an upregulation of the fission gene, Fis1 (Figure 5D),

na

and a reduction in the mitophagy-related gene PINK1 (Figure 5D), which could indicate an overall accumulation of fragmented mitochondria in the spinal cord of HFD-fed mice.

ur

Oligodendrocytes are particularly vulnerable to cell death mediated by oxidative

Jo

damage due to their relatively low antioxidant levels and high amounts of iron storage [51, 52]. Notably, gene expression of the oxidative defense enzyme SOD2 (Figure 5D) was significantly lower in HFD spinal cords compared to RD. Next, in a heatmap of mitochondrial-related genes (Figure 5E), we noticed a trend for increased Bax gene expression (p=0.03, FDR=0.63), while anti-apoptosis Bcl-2 appeared less (p=0.69, FDR=0.99) in the spinal cords of HFD mice versus RD controls. Free radical production results in oxidative modification of lipids which can damage biological membranes including the myelin sheath. 4-hydroxynonenal (4-HNE) lipid peroxidation products are

Journal Pre-proof

43

found in demyelinating MS lesions and are highly toxic to OLCs [47, 53, 54]. White matter tracts within both the spinal cord dorsal column (Figure 5F) and corpus callosum (Figure 5G) of mice consuming HFD had significantly greater immunoreactivity for 4HNE compared to RD controls. Furthermore, immunofluorescence staining demonstrated increased levels of pro-apoptotic protein Bax (Figure 5H) and decreased levels of anti-apoptotic Bcl-2 (Figure 5I) in the dorsal column of HFD-fed mice.

of

Quantification of the Bax/Bcl-2 ratio (Figure 5H) and double positive Olig2/cleaved-

ro

caspase-3 cells (Figure 5I) suggest apoptotic cell death signaling is underway in

-p

oligodendrocytes chronically fed a HFD. These results point to HFD-related changes in

re

key metabolic regulators such as AMPK and SIRT1, decreased mitochondrial biogenesis and an impaired potential for antioxidant responses as contributing factors to

lP

the increases in oxidative stress markers such as 4-HNE and loss of myelinating cells

Jo

ur

na

observed in mice consuming HFD.

Jo

ur

na

lP

re

-p

ro

of

Journal Pre-proof

44

Journal Pre-proof

45

Fig 5. Altered Mitochondrial Quality Control Processes, Oxidative Stress, and Apoptosis in the spinal cord following 12 wk of high fat diet consumption. Schematic (A) and Western blots (B) demonstrate significant reductions in AMPK phosphorylation, SIRT1, and PGC1α. C, Bar graphs show quantification of relative optical density (ROD) highlighting reductions in AMPK phosphorylation and total AMPK, SIRT1, and PGC1α protein expression in the spinal cord of high fat diet (HFD)-

of

consuming mice compared to those fed a regular diet (RD). The first HFD mouse

ro

samples protein level changes were less affected than others due to animal variability

-p

despite consistent conditions between mice. D, Quantitative PCR demonstrated differences in RNA expression of PGC1α, Fis1, PINK1, and SOD2. E, Heat map

re

summarizes RNAseq data for mitochondria-related transcriptional changes.

lP

Representative 4-hydroxynonenol (4-HNE) immunostained sections and corresponding

na

quantification of immunoreactivity normalized to RD control revealing increased lipid peroxidation in the spinal cord (SC) (F) and corpus callosum (G) of HFD-fed mice by 12

ur

wk. Immunofluorescence images and associated bar graphs show increased Bax/Olig2

Jo

double positive cells (H) and an increased Bax/Bcl2 ratio as well as more cleaved caspase-3/Olig2 double positive cells (I) in the white matter of spinal cord tissue from HFD-fed mice versus RD controls, indicating oligodendrocyte apoptosis. Graphical results represented as the mean ± SEM (*, p<0.05, **, p<0.01, and ***, p<0.001 (n=5-7 mice per group)). Scale bar= 50 µm (F), 100 µm (G), and 20 µm (H, I).

Journal Pre-proof

46

3.5 Mitochondrial changes and differentiation in oligodendrocytes treated with saturated fat. To model HFD consumption in vitro and study potential direct effects towards oligodendrocytes, we cultured primary murine oligodendrocytes in media containing the saturated fatty acid palmitate (PA, 100 µM) for 24 h. Seahorse extracellular flux analysis (Figure 6A) was performed to evaluate mitochondrial bioenergetics. PA significantly

of

decreased basal, ATP-linked, and maximal oxygen consumption rates in

ro

oligodendrocytes, but had no effect on proton leak. Since we observed transcriptional

-p

changes in fission and mitophagy genes in the spinal cord of mice consuming a HFD, we also visualized the integrity and structure of oligodendrocyte mitochondria using

re

MitoTracker. Healthy control oligodendrocytes have a long, filamentous network of

lP

mitochondria (Figure 6B), while PA-treated oligodendroglia have mitochondria that are

na

smaller, more compact, and circular. Overall, mitochondrial function and structure are impaired in oligodendrocytes cultured with high levels of saturated fat, in a manner

ur

recapitulating the changes observed in the spinal cord of HFD-fed mice (Figure 4).

Jo

To determine the influence of PA on oligodendrocyte differentiation, PLPimmunoreactivity and RNA transcripts for PLP, MBP and Olig2 were quantified. Addition of PA (100 µM, 24 h) to differentiating primary oligodendrocyte cultures reduced PLP protein (Figure 6C) to half that of controls. Supporting this, PLP, MBP and Olig2 RNA expression were also depleted (Figure 6D). In addition to oligodendrocyte progenitors, neural stem cells also contribute to replacement of oligodendrocytes following injury [44, 55]. Therefore, we next determined if the oligotoxic effects of PA extend to monolayer cultures of neural stem cells (aNSCs) derived from the adult subventricular zone (SVZ).

Journal Pre-proof

47

Supporting deleterious effects of PA across the oligodendrocytes and neural stem cells, the abundance of PLP protein (Figure 6E) and MBP RNA expression (Figure 6F) were significantly less in aNSCs cultured in the presence of PA. Taken together, these findings suggest excess PA impairs the ability of OLCs or aNSCs to differentiate into

Jo

ur

na

lP

re

-p

ro

of

myelinating oligodendrocytes.

Jo

ur

na

lP

re

-p

ro

of

Journal Pre-proof

48

Journal Pre-proof

49

Fig 6. Differentiation and mitochondrial changes in oligodendrocyte progenitors cultured in the presence of high saturated fatty acid. High fat consumption was modeled in primary murine oligodendrocyte lineage cell (OLC) cultures by including palmitate in the media (PA, 100µM, 24 h). Bioenergetics characterization (A) of PA in OLCs was measured using a Seahorse XF24 analyzer. Basal, ATP-linked, and maximum capacity oxygen consumption rates were decreased in OLCs treated with PA.

of

Significant changes in proton leak respiration were not observed. MitoTracker staining

ro

(B) showed increased mitochondrial fragmentation in PA-treated OLCs as determined

-p

by mitochondrial parameters including significantly increased solidity and circularity and a smaller average size. Proteolipid (PLP) immunoreactivity of OLCs (C) and qPCR

re

quantification of RNA (D) for PLP, myelin basic protein (MBP) and Olig2 illustrate

lP

impaired differentiation toward a mature myelinating oligodendrocyte when treated with

na

PA. Similarly PA-treated adult SVZ-derived neural stem cell (aNSC) cultures (M) also express less PLP protein (E) and MBP RNA (F). Graphical results represented as the

Jo

ur

mean ± SEM (n=3-6/group; *, p<0.05, **, p<0.01, and ***, p<0.001). Scale bar= 50 µm.

Journal Pre-proof

50

4. Discussion Diet-induced obesity increases the risk for a number of diseases including diabetes, cardiovascular disease, cancer, and cognitive dysfunction and has been steadily growing in prevalence in recent decades [20, 56-59]. The role of oligodendrocytes and myelin plasticity is likewise implicated in a variety of neuropsychiatric and cognitive conditions [1, 2, 6]. Notably, white matter integrity is

of

negatively correlated with BMI and is decreased in the corpus callosum of obese

ro

individuals [4, 14, 60]. Elevated BMI is also linked to an increased incidence and

-p

disease burden in MS, especially in combination with other risk factors [10, 13, 61, 62].

re

Here we explored the potential link between a HFD, myelin integrity and metabolic dysfunction in the CNS. Our findings demonstrate for the first time that a HFD alone

lP

depletes oligodendrocyte progenitors in the brain and spinal cord and high saturated fat

na

impedes oligodendrocyte differentiation in vitro. Overall, we observed changes in genes and pathways involving oligodendrogenesis and differentiation, iron homeostasis,

ur

oxidative stress, ER stress, insulin signaling, and inflammation all of which may

Jo

contribute mechanistically alone or collectively to the loss of myelinating cells in the CNS of mice chronically consuming HFD. In the spinal cords of HFD fed mice, RNA sequencing and untargeted metabolomics point towards ER stress and mitochondrial dysfunction as key mechanisms contributing to oligodendrogliopathy. These findings were validated by results showing depletion of TCA cycle intermediates and reduced oxygen consumption rates in the spinal cords of HFD-consuming mice and in isolated oligodendrocytes. Moreover, deficits in AMPK-SIRT1-PGC1α signaling were present in HFD spinal cords, whereas the oxidative stress marker 4HNE was increased. Overall,

Journal Pre-proof

51

the vast influence of metabolic dysfunction on oligodendrocytes observed here is of high relevance to demyelinating, neuropsychiatric and cognitive disorders and to considerations for strategies to manage myelin protection and repair. The results of the current study suggest that consumption of excess high fat alone is sufficient to trigger oligodendrogliopathy and impairments in oligodendrocyte differentiation across the brain and spinal cord and links these to several disturbances

of

in metabolic status. Oligodendrocytes play critical roles in modulating motor [63, 64] and

ro

psychiatric [5, 65] function by improving the speed and integrity of impulse conduction,

-p

providing neuronal metabolic support, and regulating synapses [8]. In a recent study, we

re

discovered that a diet high in saturated fat (41%) and sucrose (34%), the so-called Western diet, depletes oligodendrocytes and their precursors after 7 wk of consumption

lP

in the adult (4 month) spinal cord [29]. A later study, reported neuroinflammation,

na

cerebrovascular decline, and white matter damage with an increase in Olig2+ progenitors, but a decrease in MBP in the frontal parietal cortex/corpus callosum

ur

following long term (10 month) consumption of a “Western-style” diet (16.4% calories

Jo

from fat with added high-fructose corn syrup) [4]. Importantly, in both prior studies examining the effects of a high fat high sugar diet on myelin, exercise prevented the deleterious effects. The findings of the current study suggest that consumption of high fat alone is sufficient to trigger demyelinating effects in the brain and spinal cord of adult mice. Additional study will be needed to determine if the demyelinating effects of HFD can likewise be prevented by exercise interventions. Many of the transcriptional changes we observed are increased in the spinal cord of mice consuming a HFD are already established to be differentially expressed in

Journal Pre-proof

52

individuals with MS and its experimental models. Fos, Grn, Sdf2l1, Fn14, and Paqr6 RNA accumulation are each separately found in glia of white matter tissue of MS patients, and Grn polymorphisms may influence MS disease course and relapse recovery [66-72]. Down regulated genes in the spinal cord of HFD mice were also affected in MS animal models, include Slc17a7 and Pdyn [73]. Although Cryab was shown to inhibit inflammation and disease in the EAE model, recent studies in the

of

Cuprizone model suggest that Cryab activates astrocytes and exacerbates

ro

demyelination [74]. Moreover, activation of the Sgk1 pathway following oxidative stress

-p

exacerbates EAE [75]. Among many genes identified by Moyon et. al in a study of

re

transcriptional profiling of Cuprizone-“activated” oligodendrocytes, higher expression of Socs3, Ddit4, and lower expression of Kdr and Cdc42ep2 genes may be required to

lP

activate OPCs enabling remyelination [76]. Kruppel like factor 9 (Klf9), down regulated

na

by HFD, is a transcription factor downstream of T3 that regulates oligodendrocyte differentiation and myelin regeneration in the Cuprizone model [77, 78]. Ddit3 is

ur

expressed by activated astrocytes following Cuprizone exposure, while knockout mice

Jo

had less oligodendrocyte apoptosis, demyelination, and microglial and astrocyte activation [79]. HFD also exacerbates disease symptoms and neuroinflammation in EAE models [21, 22]. Notably, the genes identified as having higher expression in HFD-EAE versus RD-EAE did not overlap with those we observed in the spinal cord of adult mice consuming HFD, indicating unique mechanisms contribute to HFD-triggered pathology depending on the disease context [22]. ER stress and inflammation closely follow lipid dysregulation in diabetic neuropathy and obesity-associated allodynia models and cooperate in a feed forward

Journal Pre-proof

53

manner [80-82]. In line with studies showing ER stress in liver, heart, muscle, hypothalamus, and hippocampus in diet-induced obesity models [7, 48, 83, 84], RNA sequencing results in the current study uncovered ER Stress and the Unfolded Protein Response as prominently altered pathways in the adult spinal cord after 12 wk of HFD consumption. Since myelinating cells produce large amounts of plasma membrane, they are particularly susceptible to disruptions in secretory pathways including ER stress and

of

eventual apoptosis if left unchecked [3, 26, 85]. Thus, controlling ER overload may be

ro

beneficial to restoring oligodendrocytes and myelin in various white matter diseases [3,

-p

26, 85, 86]. Xbp1, involved in ER stress responses, is reported to increase in MS

re

lesions as well as in vanishing white matter disease (VWMD) [3, 26]. ER chaperones help control cellular responses to ER stress and some were shown to be required for

lP

oligodendrocyte survival during EAE [85]. ER and mitochondria interact both physically

MS pathology [27].

na

and functionally, and alterations in these connections have recently been described in

ur

Alterations in ER stress are connected to mitochondrial deficits in MS [27].

Jo

Mitochondrial morphology and impaired mitochondrial function and antioxidant responses are also reported in experimental models of demyelinating disease [87-89]. Here we document fragmented mitochondria in cultures of oligodendrocytes grown under high fat conditions. Fis1 is involved in mitochondrial fission and in early events in cell-death signaling, and expression was also upregulated in the spinal cord of mice consuming high fat. Similar changes are likewise engaged in OLCs in the context of oxidative stress or inflammation [40, 89-91]. Moreover, inhibition of fission is protective against demyelination and neuroinflammation, although has little impact on OLC

Journal Pre-proof

54

differentiation in EAE and Cuprizone MS models [89, 92]. Impairments in mitochondria also lead to increased reactive oxygen species and downstream effects on proteins, lipids, and nucleic acids [92-94]. Notably, in both the corpus callosum and spinal cord white matter regions where OLC death occurred following chronic HFD consumption, we observed increased lipid peroxidation. Increased lipid peroxidation was also observed in the spinal cord with consumption of high fat and sucrose [29]. In MS

of

pathogenesis and its animal models, oxidative stress mediates apoptosis of

ro

oligodendrocytes and is commonly documented by an elevated Bax/Bcl2 ratio and

-p

caspase-3 cleavage [95, 96]. Our in vivo data suggest that mitochondrial-dependent

re

apoptosis also occurs in oligodendrocytes in relation to HFD-induced mitochondrial dysfunction and increased oxidative damage.

lP

In the current study, we discovered substantial deficits in TCA cycle

na

intermediates and oxygen consumption rates in the spinal cords of HFD mice. Key substrates at all stages of the TCA cycle, from citric acid to oxaloacetate were reduced

ur

in the spinal cord of mice consuming HF. In addition, these effects were paralleled by

Jo

diminished mitochondrial oxidative respiration. Oligodendrocytes have extremely high energy demands and die hours faster than neurons following nutrient deprivation by arterial occlusion [28, 97]. Impaired energy metabolism has already been proposed as a contributing factor to oligodendrogliopathy in pattern III MS lesions and is described in animal models of the disease [87, 89, 93]. Normally, metabolism of pyruvate and other fuel sources through the mitochondrial TCA cycle leads to ATP synthesis via the electron transport chain. Our findings that the dysmyelinating effects of HFD were associated with significant impairments in TCA cycle function, are of particular interest

Journal Pre-proof

55

given the efficacy of dimethyl fumarate, itself a TCA cycle metabolite, as a first line treatment for relapsing remitting MS [98]. AMPK, SIRT1 and PGC1α were all substantially reduced in the spinal cord of mice consuming HF. AMPK and SIRT1 are crucial metabolic sensors that impact PCG1α and thus mitochondria biogenesis and function. In addition to promoting cell survival, AMPK-SIRT1 activation and mitochondrial function regulate oligodendrocyte

of

differentiation [50, 99-102]. Recently, folate was determined to regulate oligodendrocyte

ro

survival and differentiation through AMPK activity [99]. SIRT1 and SIRT2 activation

-p

redundantly mediate differentiation of neural stem or progenitor cells towards the oligodendrocyte lineage, likely by upregulation of PDGFRα, Sox10, Nkx2.2 and

re

downregulation of p21 [9, 103, 104]. Moreover, SIRT1 activation through

lP

overexpression or agonists is neuroprotective in MS models, while expansion of

na

oligodendrocyte progenitors occurs following SIRT1 inactivation [101, 104]. Exercise and other activators of the AMPK-SIRT1-PGC1α system may be beneficial in reducing

ur

deleterious effects of obesogenic diets on oligodendrocyte lineage cells and myelin

Jo

integrity by promoting mitochondrial homeostasis [4, 29, 49, 101]. Interestingly, we observed a predicted decrease in PDGFRα signaling in the transcriptomic data along with decreased AMPK-SIRT1 signaling, leading to a loss of both progenitors and mature oligodendrocytes in the spinal cord following long term HFD. Whether or not the disruptions we observe in TCA cycle metabolite depletion, impaired mitochondrial function and oligodendrocyte loss observed in HFD-fed spinal cords are a direct consequence of the altered AMPK-SIRT signaling, as specified in other tissues and cell types, should be more directly addressed in future studies.

Journal Pre-proof

56

Mature CC-1+ oligodendrocytes along with Olig2+ progenitors are lost in the spinal cord white matter with HFD consumption, but only the later are impacted in the corpus callosum at the same 12 wk time point. Given their increased energy demands for proliferation and myelin membrane production it is possible that oligodendrocyte progenitors and young oligodendrocytes as labeled by Olig2 are more vulnerable to the stresses associated with HF consumption compared to mature oligodendrocytes. We

of

cannot exclude that mature oligodendroglia in the corpus callosum would be lost with

ro

even more chronic HFD feeding. Region specific and possible temporal differences in

-p

HF-elicited oligotoxicity may also relate to heterogeneity in the local oligodendrocyte

re

populations, region specific effects on neurons, or differential effects across the astrocyte and microglial compartments that so significantly contribute to the

lP

microenvironment [41]. Our studies also demonstrate an anxiety-like phenotype and

na

overall activity level changes in the HFD-fed mice that mirror a number of previous reports [20, 58], however future studies will be required to determine if HFD-induced

ur

behavioral changes are in fact dependent on the oligodendrocyte loss documented.

Jo

Although we show that impaired differentiation and mitochondrial fragmentation occurring with HFD are recapitulated across regions and in cultures of oligodendrocytes, a Ribotag approach or single cell sequencing will be beneficial for the identification of cell-specific transcriptional changes in each tissue, and this is another goal for future experiments.

Journal Pre-proof

57

5. Conclusions The findings of the current study demonstrate significant perturbations in CNS function occur in response to long term consumption of a diet laden with excessive fat, including direct impairment of oligodendrocyte mitochondrial function that ultimately impairs oligodendrocyte survival and differentiation. Previous reports in Western diet-fed mice suggest exercise may mitigate deleterious effects on myelin integrity and

of

behavioral outcomes, and our new data in mice fed a high fat diet suggest that

ro

modulation of mitochondrial function, ER and oxidative stress pathways, in addition to

-p

the TCA cycle may also be important areas to explore as targets to improve myelin

re

protection and regeneration. Accordingly, this work should inspire new research to evaluate ways to mitigate the negative consequences of a chronic high fat diet on

6. Author Contributions

na

lP

oligodendrocytes alone and in the context of white matter injury.

ur

IAS, MRL and AM conceived and designed the study. MRL, HY and AM performed all

Jo

in vivo experiments, necropsies, histological staining and analysis. WS and HNK assisted with necropsies and LK performed some of the histological staining. CC generated heat maps of RNA sequencing data and contributed to neural stem cell cultures. MRL performed and analyzed oxygraph and in vitro experiments. MRL and IAS interpreted the data and wrote the manuscript. All authors reviewed and edited the manuscript.

Journal Pre-proof

58

7. Acknowledgements The work was supported by Mayo Clinic Center for Multiple Sclerosis and Autoimmune Neurology (CMSAN), the Eugene and Marcia Applebaum Foundation, the Mayo Clinic Center for Biomedical Discovery (CBD) and the Mayo Clinic Metabolomics Core (U24DK100469 and UL1TR000135). Portions of this work were also supported by R01NS052741, RG4958 from the National Multiple Sclerosis Society, a grant from the

of

Craig H. Neilsen Foundation, and the Minnesota State Spinal Cord Injury and Traumatic

ro

Brain Injury Research Program.

-p

We would like to thank Thomas White for his assistance in analyzing the glucose

re

tolerance data and Katherine Klaus for her help the Oroboros oxygraph system. We also thank Xuewei Wang and Tumpa Dutta for their initial assistance with Bioinformatics

lP

for RNAseq and LC-MS-MS measurements, respectively. The diagrams within figures

ur

Declaration of Interests

na

were created using elements from the Biomedical-PPT-Toolkit-Suite (Motifolio).

Jo

The authors have no declarations of interests to disclose.

Journal Pre-proof

59

References

Jo

ur

na

lP

re

-p

ro

of

[1] R. Ohtomo, A. Iwata, K. Arai, Molecular Mechanisms of Oligodendrocyte Regeneration in White Matter-Related Diseases, Int J Mol Sci 19(6) (2018). [2] R.C. Armstrong, A.J. Mierzwa, G.M. Sullivan, M.A. Sanchez, Myelin and oligodendrocyte lineage cells in white matter pathology and plasticity after traumatic brain injury, Neuropharmacology 110(Pt B) (2016) 654-659. [3] B. van Kollenburg, J. van Dijk, J. Garbern, A.A. Thomas, G.C. Scheper, J.M. Powers, M.S. van der Knaap, Glia-specific activation of all pathways of the unfolded protein response in vanishing white matter disease, J Neuropathol Exp Neurol 65(7) (2006) 707-15. [4] L.C. Graham, W.A. Grabowska, Y. Chun, S.L. Risacher, V.M. Philip, A.J. Saykin, I. Alzheimer's Disease Neuroimaging, S.J. Sukoff Rizzo, G.R. Howell, Exercise prevents obesity-induced cognitive decline and white matter damage in mice, Neurobiol Aging 80 (2019) 154-172. [5] K. Roy, J.C. Murtie, B.F. El-Khodor, N. Edgar, S.P. Sardi, B.M. Hooks, M. BenoitMarand, C. Chen, H. Moore, P. O'Donnell, D. Brunner, G. Corfas, Loss of erbB signaling in oligodendrocytes alters myelin and dopaminergic function, a potential mechanism for neuropsychiatric disorders, Proc Natl Acad Sci U S A 104(19) (2007) 8131-6. [6] V. Haroutunian, P. Katsel, P. Roussos, K.L. Davis, L.L. Altshuler, G. Bartzokis, Myelination, oligodendrocytes, and serious mental illness, Glia 62(11) (2014) 1856-77. [7] M. Cai, H. Wang, J.J. Li, Y.L. Zhang, L. Xin, F. Li, S.J. Lou, The signaling mechanisms of hippocampal endoplasmic reticulum stress affecting neuronal plasticity-related protein levels in high fat diet-induced obese rats and the regulation of aerobic exercise, Brain Behav Immun 57 (2016) 347-359. [8] R.E. Pepper, K.A. Pitman, C.L. Cullen, K.M. Young, How Do Cells of the Oligodendrocyte Lineage Affect Neuronal Circuits to Influence Motor Function, Memory and Mood?, Front Cell Neurosci 12 (2018) 399. [9] A. Alizadeh, S. Karimi-Abdolrezaee, Microenvironmental regulation of oligodendrocyte replacement and remyelination in spinal cord injury, J Physiol 594(13) (2016) 3539-52. [10] M.A. Gianfrancesco, B. Acuna, L. Shen, F.B. Briggs, H. Quach, K.H. Bellesis, A. Bernstein, A.K. Hedstrom, I. Kockum, L. Alfredsson, T. Olsson, C. Schaefer, L.F. Barcellos, Obesity during childhood and adolescence increases susceptibility to multiple sclerosis after accounting for established genetic and environmental risk factors, Obes Res Clin Pract 8(5) (2014) e435-47. [11] A. Langer-Gould, S.M. Brara, B.E. Beaber, C. Koebnick, Childhood obesity and risk of pediatric multiple sclerosis and clinically isolated syndrome, Neurology 80(6) (2013) 548-52. [12] K.L. Munger, T. Chitnis, A. Ascherio, Body size and risk of MS in two cohorts of US women, Neurology 73(19) (2009) 1543-50. [13] T. Olsson, L.F. Barcellos, L. Alfredsson, Interactions between genetic, lifestyle and environmental risk factors for multiple sclerosis, Nat Rev Neurol 13(1) (2017) 2536.

Journal Pre-proof

60

Jo

ur

na

lP

re

-p

ro

of

[14] T.M. Wassenaar, K. Yaffe, Y.D. van der Werf, C.E. Sexton, Associations between modifiable risk factors and white matter of the aging brain: insights from diffusion tensor imaging studies, Neurobiol Aging 80 (2019) 56-70. [15] A. Manouchehrinia, A.K. Hedstrom, L. Alfredsson, T. Olsson, J. Hillert, R. Ramanujam, Association of Pre-Disease Body Mass Index With Multiple Sclerosis Prognosis, Front Neurol 9 (2018) 232. [16] P. Tettey, S. Simpson, Jr., B. Taylor, L. Blizzard, A.L. Ponsonby, T. Dwyer, K. Kostner, I. van der Mei, An adverse lipid profile is associated with disability and progression in disability, in people with MS, Mult Scler 20(13) (2014) 1737-44. [17] L. Negrotto, M.F. Farez, J. Correale, Immunologic Effects of Metformin and Pioglitazone Treatment on Metabolic Syndrome and Multiple Sclerosis, JAMA Neurol 73(5) (2016) 520-8. [18] A. Sicras-Mainar, E. Ruiz-Beato, R. Navarro-Artieda, J. Maurino, Comorbidity and metabolic syndrome in patients with multiple sclerosis from Asturias and Catalonia, Spain, BMC Neurol 17(1) (2017) 134. [19] P. Dimas, L. Montani, J.A. Pereira, D. Moreno, M. Trotzmuller, J. Gerber, C.F. Semenkovich, H.C. Kofeler, U. Suter, CNS myelination and remyelination depend on fatty acid synthesis by oligodendrocytes, Elife 8 (2019). [20] M. Ogrodnik, Y. Zhu, L.G.P. Langhi, T. Tchkonia, P. Kruger, E. Fielder, S. Victorelli, R.A. Ruswhandi, N. Giorgadze, T. Pirtskhalava, O. Podgorni, G. Enikolopov, K.O. Johnson, M. Xu, C. Inman, A.K. Palmer, M. Schafer, M. Weigl, Y. Ikeno, T.C. Burns, J.F. Passos, T. von Zglinicki, J.L. Kirkland, D. Jurk, Obesity-Induced Cellular Senescence Drives Anxiety and Impairs Neurogenesis, Cell Metab (2019). [21] S. Timmermans, J.F. Bogie, T. Vanmierlo, D. Lutjohann, P. Stinissen, N. Hellings, J.J. Hendriks, High fat diet exacerbates neuroinflammation in an animal model of multiple sclerosis by activation of the Renin Angiotensin system, J Neuroimmune Pharmacol 9(2) (2014) 209-17. [22] M. Hasan, J.E. Seo, K.A. Rahaman, H. Min, K.H. Kim, J.H. Park, C. Sung, J. Son, M.J. Kang, B.H. Jung, W.S. Park, O.S. Kwon, Novel genes in brain tissues of EAE-induced normal and obese mice: Upregulation of metal ion-binding protein genes in obese-EAE mice, Neuroscience 343 (2017) 322-336. [23] R. Ghemrawi, S.F. Battaglia-Hsu, C. Arnold, Endoplasmic Reticulum Stress in Metabolic Disorders, Cells 7(6) (2018). [24] C.M. Mayer, D.D. Belsham, Palmitate attenuates insulin signaling and induces endoplasmic reticulum stress and apoptosis in hypothalamic neurons: rescue of resistance and apoptosis through adenosine 5' monophosphate-activated protein kinase activation, Endocrinology 151(2) (2010) 576-85. [25] K.S. Gwiazda, T.L. Yang, Y. Lin, J.D. Johnson, Effects of palmitate on ER and cytosolic Ca2+ homeostasis in beta-cells, Am J Physiol Endocrinol Metab 296(4) (2009) E690-701. [26] A.N. Mhaille, S. McQuaid, A. Windebank, P. Cunnea, J. McMahon, A. Samali, U. FitzGerald, Increased expression of endoplasmic reticulum stress-related signaling pathway molecules in multiple sclerosis lesions, J Neuropathol Exp Neurol 67(3) (2008) 200-11.

Journal Pre-proof

61

Jo

ur

na

lP

re

-p

ro

of

[27] Y. Haile, X. Deng, C. Ortiz-Sandoval, N. Tahbaz, A. Janowicz, J.Q. Lu, B.J. Kerr, N.J. Gutowski, J.E. Holley, P. Eggleton, F. Giuliani, T. Simmen, Rab32 connects ER stress to mitochondrial defects in multiple sclerosis, J Neuroinflammation 14(1) (2017) 19. [28] L. Rosko, V.N. Smith, R. Yamazaki, J.K. Huang, Oligodendrocyte Bioenergetics in Health and Disease, Neuroscientist (2018) 1073858418793077. [29] H. Yoon, A. Kleven, A. Paulsen, L. Kleppe, J. Wu, Z. Ying, F. Gomez-Pinilla, I.A. Scarisbrick, Interplay between exercise and dietary fat modulates myelinogenesis in the central nervous system, Biochim Biophys Acta 1862(4) (2016) 545-55. [30] M.E. McGee-Lawrence, T.A. White, N.K. LeBrasseur, J.J. Westendorf, Conditional deletion of Hdac3 in osteoprogenitor cells attenuates diet-induced systemic metabolic dysfunction, Mol Cell Endocrinol 410 (2015) 42-51. [31] K.R. Kalari, A.A. Nair, J.D. Bhavsar, D.R. O'Brien, J.I. Davila, M.A. Bockol, J. Nie, X. Tang, S. Baheti, J.B. Doughty, S. Middha, H. Sicotte, A.E. Thompson, Y.W. Asmann, J.P. Kocher, MAP-RSeq: Mayo Analysis Pipeline for RNA sequencing, BMC Bioinformatics 15 (2014) 224. [32] B. Langmead, C. Trapnell, M. Pop, S.L. Salzberg, Ultrafast and memory-efficient alignment of short DNA sequences to the human genome, Genome Biol 10(3) (2009) R25. [33] S. Anders, P.T. Pyl, W. Huber, HTSeq--a Python framework to work with highthroughput sequencing data, Bioinformatics 31(2) (2015) 166-9. [34] L. Wang, S. Wang, W. Li, RSeQC: quality control of RNA-seq experiments, Bioinformatics 28(16) (2012) 2184-5. [35] M.D. Robinson, D.J. McCarthy, G.K. Smyth, edgeR: a Bioconductor package for differential expression analysis of digital gene expression data, Bioinformatics 26(1) (2010) 139-40. [36] Y. Zhang, K. Chen, S.A. Sloan, M.L. Bennett, A.R. Scholze, S. O'Keeffe, H.P. Phatnani, P. Guarnieri, C. Caneda, N. Ruderisch, S. Deng, S.A. Liddelow, C. Zhang, R. Daneman, T. Maniatis, B.A. Barres, J.Q. Wu, An RNA-sequencing transcriptome and splicing database of glia, neurons, and vascular cells of the cerebral cortex, J Neurosci 34(36) (2014) 11929-47. [37] T. Dutta, Y.C. Kudva, X.M. Persson, L.A. Schenck, G.C. Ford, R.J. Singh, R. Carter, K.S. Nair, Impact of Long-Term Poor and Good Glycemic Control on Metabolomics Alterations in Type 1 Diabetic People, J Clin Endocrinol Metab 101(3) (2016) 1023-33. [38] G.N. Ruegsegger, S. Manjunatha, P. Summer, S. Gopala, P. Zabeilski, S. Dasari, P.M. Vanderboom, I.R. Lanza, K.A. Klaus, K.S. Nair, Insulin deficiency and intranasal insulin alter brain mitochondrial function: a potential factor for dementia in diabetes, FASEB J 33(3) (2019) 4458-4472. [39] M. Langley, A. Ghosh, A. Charli, S. Sarkar, M. Ay, J. Luo, J. Zielonka, T. Brenza, B. Bennett, H. Jin, S. Ghaisas, B. Schlichtmann, D. Kim, V. Anantharam, A. Kanthasamy, B. Narasimhan, B. Kalyanaraman, A.G. Kanthasamy, MitoApocynin Prevents Mitochondrial Dysfunction, Microglial Activation, Oxidative Damage, and Progressive Neurodegeneration in MitoPark Transgenic Mice, Antioxid Redox Signal 27(14) (2017) 1048-1066.

Journal Pre-proof

62

Jo

ur

na

lP

re

-p

ro

of

[40] R.K. Dagda, S.J. Cherra, 3rd, S.M. Kulich, A. Tandon, D. Park, C.T. Chu, Loss of PINK1 function promotes mitophagy through effects on oxidative stress and mitochondrial fission, J Biol Chem 284(20) (2009) 13843-55. [41] H. Yoon, G. Walters, A.R. Paulsen, I.A. Scarisbrick, Astrocyte heterogeneity across the brain and spinal cord occurs developmentally, in adulthood and in response to demyelination, PLoS One 12(7) (2017) e0180697. [42] Y.W. Ng, Y.H. Say, Palmitic acid induces neurotoxicity and gliatoxicity in SH-SY5Y human neuroblastoma and T98G human glioblastoma cells, PeerJ 6 (2018) e4696. [43] S. Kim, C. Kim, S. Park, Mdivi-1 Protects Adult Rat Hippocampal Neural Stem Cells against Palmitate-Induced Oxidative Stress and Apoptosis, Int J Mol Sci 18(9) (2017). [44] C.I. Choi, H. Yoon, K.L. Drucker, M.R. Langley, L. Kleppe, I.A. Scarisbrick, The Thrombin Receptor Restricts Subventricular Zone Neural Stem Cell Expansion and Differentiation, Sci Rep 8(1) (2018) 9360. [45] A. Hall, N.A. Giese, W.D. Richardson, Spinal cord oligodendrocytes develop from ventrally derived progenitor cells that express PDGF alpha-receptors, Development 122(12) (1996) 4085-94. [46] X.R. Jin, X.S. Chen, L. Xiao, MeCP2 Deficiency in Neuroglia: New Progress in the Pathogenesis of Rett Syndrome, Front Mol Neurosci 10 (2017) 316. [47] A.D. Roth, M.T. Nunez, Oligodendrocytes: Functioning in a Delicate Balance Between High Metabolic Requirements and Oxidative Damage, Adv Exp Med Biol 949 (2016) 167-181. [48] K.R. Bohnert, J.D. McMillan, A. Kumar, Emerging roles of ER stress and unfolded protein response pathways in skeletal muscle health and disease, J Cell Physiol 233(1) (2018) 67-78. [49] N.L. Price, A.P. Gomes, A.J. Ling, F.V. Duarte, A. Martin-Montalvo, B.J. North, B. Agarwal, L. Ye, G. Ramadori, J.S. Teodoro, B.P. Hubbard, A.T. Varela, J.G. Davis, B. Varamini, A. Hafner, R. Moaddel, A.P. Rolo, R. Coppari, C.M. Palmeira, R. de Cabo, J.A. Baur, D.A. Sinclair, SIRT1 is required for AMPK activation and the beneficial effects of resveratrol on mitochondrial function, Cell Metab 15(5) (2012) 675-90. [50] M. Bonora, E. De Marchi, S. Patergnani, J.M. Suski, F. Celsi, A. Bononi, C. Giorgi, S. Marchi, A. Rimessi, J. Duszynski, T. Pozzan, M.R. Wieckowski, P. Pinton, Tumor necrosis factor-alpha impairs oligodendroglial differentiation through a mitochondria-dependent process, Cell Death Differ 21(8) (2014) 1198-208. [51] O. Baud, R.F. Haynes, H. Wang, R.D. Folkerth, J. Li, J.J. Volpe, P.A. Rosenberg, Developmental up-regulation of MnSOD in rat oligodendrocytes confers protection against oxidative injury, Eur J Neurosci 20(1) (2004) 29-40. [52] H. Lassmann, J. van Horssen, Oxidative stress and its impact on neurons and glia in multiple sclerosis lesions, Biochim Biophys Acta 1862(3) (2016) 506-10. [53] E. McCracken, V. Valeriani, C. Simpson, T. Jover, J. McCulloch, D. Dewar, The lipid peroxidation by-product 4-hydroxynonenal is toxic to axons and oligodendrocytes, J Cereb Blood Flow Metab 20(11) (2000) 1529-36. [54] G.G. Ortiz, F.P. Pacheco-Moises, O.K. Bitzer-Quintero, A.C. Ramirez-Anguiano, L.J. Flores-Alvarado, V. Ramirez-Ramirez, M.A. Macias-Islas, E.D. Torres-

Journal Pre-proof

63

Jo

ur

na

lP

re

-p

ro

of

Sanchez, Immunology and oxidative stress in multiple sclerosis: clinical and basic approach, Clin Dev Immunol 2013 (2013) 708659. [55] M.E. Silva, S. Lange, B. Hinrichsen, A.R. Philp, C.R. Reyes, D. Halabi, J.B. Mansilla, P. Rotheneichner, A. Guzman de la Fuente, S. Couillard-Despres, L.F. Batiz, R.J.M. Franklin, L. Aigner, F.J. Rivera, Pericytes Favor Oligodendrocyte Fate Choice in Adult Neural Stem Cells, Front Cell Neurosci 13 (2019) 85. [56] C.S. Fox, S.H. Golden, C. Anderson, G.A. Bray, L.E. Burke, I.H. de Boer, P. Deedwania, R.H. Eckel, A.G. Ershow, J. Fradkin, S.E. Inzucchi, M. Kosiborod, R.G. Nelson, M.J. Patel, M. Pignone, L. Quinn, P.R. Schauer, E. Selvin, D.K. Vafiadis, L. American Heart Association Diabetes Committee of the Council on, H. Cardiometabolic, C.o.C. Council on Clinical Cardiology, C.o.C.S. Stroke Nursing, C.o.Q.o.C. Anesthesia, R. Outcomes, A. American Diabetes, Update on Prevention of Cardiovascular Disease in Adults With Type 2 Diabetes Mellitus in Light of Recent Evidence: A Scientific Statement From the American Heart Association and the American Diabetes Association, Diabetes Care 38(9) (2015) 1777-803. [57] R.J. Koene, A.E. Prizment, A. Blaes, S.H. Konety, Shared Risk Factors in Cardiovascular Disease and Cancer, Circulation 133(11) (2016) 1104-14. [58] W. Mizunoya, K. Ohnuki, K. Baba, H. Miyahara, N. Shimizu, K. Tabata, T. Kino, Y. Sato, R. Tatsumi, Y. Ikeuchi, Effect of dietary fat type on anxiety-like and depression-like behavior in mice, Springerplus 2(1) (2013) 165. [59] D. Frasca, B.B. Blomberg, R. Paganelli, Aging, Obesity, and Inflammatory AgeRelated Diseases, Front Immunol 8 (2017) 1745. [60] J. Xu, Y. Li, H. Lin, R. Sinha, M.N. Potenza, Body mass index correlates negatively with white matter integrity in the fornix and corpus callosum: a diffusion tensor imaging study, Hum Brain Mapp 34(5) (2013) 1044-52. [61] L.E. Mokry, S. Ross, N.J. Timpson, S. Sawcer, G. Davey Smith, J.B. Richards, Obesity and Multiple Sclerosis: A Mendelian Randomization Study, PLoS Med 13(6) (2016) e1002053. [62] J.J. Guerrero-Garcia, L. Carrera-Quintanar, R.I. Lopez-Roa, A.L. Marquez-Aguirre, A.E. Rojas-Mayorquin, D. Ortuno-Sahagun, Multiple Sclerosis and Obesity: Possible Roles of Adipokines, Mediators Inflamm 2016 (2016) 4036232. [63] I.A. McKenzie, D. Ohayon, H. Li, J.P. de Faria, B. Emery, K. Tohyama, W.D. Richardson, Motor skill learning requires active central myelination, Science 346(6207) (2014) 318-22. [64] S. Schneider, A. Gruart, S. Grade, Y. Zhang, S. Kroger, F. Kirchhoff, G. Eichele, J.M. Delgado Garcia, L. Dimou, Decrease in newly generated oligodendrocytes leads to motor dysfunctions and changed myelin structures that can be rescued by transplanted cells, Glia 64(12) (2016) 2201-2218. [65] X. Chen, W. Zhang, T. Li, Y. Guo, Y. Tian, F. Wang, S. Liu, H.Y. Shen, Y. Feng, L. Xiao, Impairment of Oligodendroglia Maturation Leads to Aberrantly Increased Cortical Glutamate and Anxiety-Like Behaviors in Juvenile Mice, Front Cell Neurosci 9 (2015) 467. [66] M. Luan, Z. Shang, Y. Teng, X. Chen, M. Zhang, H. Lv, R. Zhang, The shared and specific mechanism of four autoimmune diseases, Oncotarget 8(65) (2017) 108355-108374.

Journal Pre-proof

64

Jo

ur

na

lP

re

-p

ro

of

[67] J.S. Yu, T. Hayashi, E. Seboun, R.M. Sklar, T.H. Doolittle, S.L. Hauser, Fos RNA accumulation in multiple sclerosis white matter tissue, J Neurol Sci 103(2) (1991) 209-15. [68] M. Vercellino, C. Fenoglio, D. Galimberti, A. Mattioda, C. Chiavazza, E. Binello, L. Pinessi, D. Giobbe, E. Scarpini, P. Cavalla, Progranulin genetic polymorphisms influence progression of disability and relapse recovery in multiple sclerosis, Mult Scler 22(8) (2016) 1007-12. [69] A. Groves, Y. Kihara, D. Jonnalagadda, R. Rivera, G. Kennedy, M. Mayford, J. Chun, A Functionally Defined In Vivo Astrocyte Population Identified by c-Fos Activation in a Mouse Model of Multiple Sclerosis Modulated by S1P Signaling: Immediate-Early Astrocytes (ieAstrocytes), eNeuro 5(5) (2018). [70] R. Waller, M.N. Woodroofe, S.B. Wharton, P.G. Ince, S. Francese, P.R. Heath, A. Cudzich-Madry, R.H. Thomas, N. Rounding, B. Sharrack, J.E. Simpson, Gene expression profiling of the astrocyte transcriptome in multiple sclerosis normal appearing white matter reveals a neuroprotective role, J Neuroimmunol 299 (2016) 139-146. [71] B. Serafini, R. Magliozzi, B. Rosicarelli, R. Reynolds, T.S. Zheng, F. Aloisi, Expression of TWEAK and its receptor Fn14 in the multiple sclerosis brain: implications for inflammatory tissue injury, J Neuropathol Exp Neurol 67(12) (2008) 1137-48. [72] S. Desplat-Jego, L. Feuillet, R. Creidy, I. Malikova, R. Rance, M. Khrestchatisky, K. Hahm, L.C. Burkly, J. Pelletier, J. Boucraut, TWEAK is expressed at the cell surface of monocytes during multiple sclerosis, J Leukoc Biol 85(1) (2009) 132-5. [73] I. Sevastou, G. Pryce, D. Baker, D.L. Selwood, Characterisation of Transcriptional Changes in the Spinal Cord of the Progressive Experimental Autoimmune Encephalomyelitis Biozzi ABH Mouse Model by RNA Sequencing, PLoS One 11(6) (2016) e0157754. [74] H.F. Kuipers, J. Yoon, J. van Horssen, M.H. Han, P.L. Bollyky, T.D. Palmer, L. Steinman, Phosphorylation of alphaB-crystallin supports reactive astrogliosis in demyelination, Proc Natl Acad Sci U S A 114(9) (2017) E1745-E1754. [75] L. Wang, B. Li, M.Y. Quan, L. Li, Y. Chen, G.J. Tan, J. Zhang, X.P. Liu, L. Guo, Mechanism of oxidative stress p38MAPK-SGK1 signaling axis in experimental autoimmune encephalomyelitis (EAE), Oncotarget 8(26) (2017) 42808-42816. [76] S. Moyon, A.L. Dubessy, M.S. Aigrot, M. Trotter, J.K. Huang, L. Dauphinot, M.C. Potier, C. Kerninon, S. Melik Parsadaniantz, R.J. Franklin, C. Lubetzki, Demyelination causes adult CNS progenitors to revert to an immature state and express immune cues that support their migration, J Neurosci 35(1) (2015) 4-20. [77] M.M. Herrmann, S. Barth, B. Greve, K.M. Schumann, A. Bartels, R. Weissert, Identification of gene expression patterns crucially involved in experimental autoimmune encephalomyelitis and multiple sclerosis, Dis Model Mech 9(10) (2016) 1211-1220. [78] J.C. Dugas, A. Ibrahim, B.A. Barres, The T3-induced gene KLF9 regulates oligodendrocyte differentiation and myelin regeneration, Mol Cell Neurosci 50(1) (2012) 45-57. [79] F. Fischbach, J. Nedelcu, P. Leopold, J. Zhan, T. Clarner, L. Nellessen, C. Beissel, Y. van Heuvel, A. Goswami, J. Weis, B. Denecke, C. Schmitz, T. Hochstrasser,

Journal Pre-proof

65

Jo

ur

na

lP

re

-p

ro

of

S. Nyamoya, M. Victor, C. Beyer, M. Kipp, Cuprizone-induced graded oligodendrocyte vulnerability is regulated by the transcription factor DNA damage-inducible transcript 3, Glia 67(2) (2019) 263-276. [80] K.K. Ryan, S.C. Woods, R.J. Seeley, Central nervous system mechanisms linking the consumption of palatable high-fat diets to the defense of greater adiposity, Cell Metab 15(2) (2012) 137-49. [81] S. Lupachyk, P. Watcho, A.A. Obrosov, R. Stavniichuk, I.G. Obrosova, Endoplasmic reticulum stress contributes to prediabetic peripheral neuropathy, Exp Neurol 247 (2013) 342-8. [82] C.K. Gavini, A.L. Bookout, R. Bonomo, L. Gautron, S. Lee, V. Mansuy-Aubert, Liver X Receptors Protect Dorsal Root Ganglia from Obesity-Induced Endoplasmic Reticulum Stress and Mechanical Allodynia, Cell Rep 25(2) (2018) 271-277 e4. [83] H.C. Hsu, C.Y. Chen, B.C. Lee, M.F. Chen, High-fat diet induces cardiomyocyte apoptosis via the inhibition of autophagy, Eur J Nutr 55(7) (2016) 2245-54. [84] X. Wang, Z.F. Zhang, G.H. Zheng, A.M. Wang, C.H. Sun, S.P. Qin, J. Zhuang, J. Lu, D.F. Ma, Y.L. Zheng, The Inhibitory Effects of Purple Sweet Potato Color on Hepatic Inflammation Is Associated with Restoration of NAD(+) Levels and Attenuation of NLRP3 Inflammasome Activation in High-Fat-Diet-Treated Mice, Molecules 22(8) (2017). [85] Y. Hussien, J.R. Podojil, A.P. Robinson, A.S. Lee, S.D. Miller, B. Popko, ER Chaperone BiP/GRP78 Is Required for Myelinating Cell Survival and Provides Protection during Experimental Autoimmune Encephalomyelitis, J Neurosci 35(48) (2015) 15921-33. [86] M.S. Elitt, H.E. Shick, M. Madhavan, K.C. Allan, B.L.L. Clayton, C. Weng, T.E. Miller, D.C. Factor, L. Barbar, B.S. Nawash, Z.S. Nevin, A.M. Lager, Y. Li, F. Jin, D.J. Adams, P.J. Tesar, Chemical Screening Identifies Enhancers of Mutant Oligodendrocyte Survival and Unmasks a Distinct Pathological Phase in Pelizaeus-Merzbacher Disease, Stem Cell Reports 11(3) (2018) 711-726. [87] P. Acs, S. Komoly, Selective ultrastructural vulnerability in the cuprizone-induced experimental demyelination, Ideggyogy Sz 65(7-8) (2012) 266-70. [88] M. Faizi, A. Salimi, E. Seydi, P. Naserzadeh, M. Kouhnavard, A. Rahimi, J. Pourahmad, Toxicity of cuprizone a Cu(2+) chelating agent on isolated mouse brain mitochondria: a justification for demyelination and subsequent behavioral dysfunction, Toxicol Mech Methods 26(4) (2016) 276-83. [89] F. Luo, K. Herrup, X. Qi, Y. Yang, Inhibition of Drp1 hyper-activation is protective in animal models of experimental multiple sclerosis, Exp Neurol 292 (2017) 21-34. [90] R.J. Youle, M. Karbowski, Mitochondrial fission in apoptosis, Nat Rev Mol Cell Biol 6(8) (2005) 657-63. [91] H.Y. Lin, S.W. Weng, Y.H. Chang, Y.J. Su, C.M. Chang, C.J. Tsai, F.C. Shen, J.H. Chuang, T.K. Lin, C.W. Liou, C.Y. Lin, P.W. Wang, The Causal Role of Mitochondrial Dynamics in Regulating Insulin Resistance in Diabetes: Link through Mitochondrial Reactive Oxygen Species, Oxid Med Cell Longev 2018 (2018) 7514383. [92] L. Haider, M.T. Fischer, J.M. Frischer, J. Bauer, R. Hoftberger, G. Botond, H. Esterbauer, C.J. Binder, J.L. Witztum, H. Lassmann, Oxidative damage in multiple sclerosis lesions, Brain 134(Pt 7) (2011) 1914-24.

Journal Pre-proof

66

Jo

ur

na

lP

re

-p

ro

of

[93] M.E. Witte, D.J. Mahad, H. Lassmann, J. van Horssen, Mitochondrial dysfunction contributes to neurodegeneration in multiple sclerosis, Trends Mol Med 20(3) (2014) 179-87. [94] C.M. Rice, M. Sun, K. Kemp, E. Gray, A. Wilkins, N.J. Scolding, Mitochondrial sirtuins--a new therapeutic target for repair and protection in multiple sclerosis, Eur J Neurosci 35(12) (2012) 1887-93. [95] G. Vakilzadeh, F. Khodagholi, T. Ghadiri, A. Ghaemi, F. Noorbakhsh, M. Sharifzadeh, A. Gorji, The Effect of Melatonin on Behavioral, Molecular, and Histopathological Changes in Cuprizone Model of Demyelination, Mol Neurobiol 53(7) (2016) 4675-84. [96] N. Sanadgol, F. Golab, H. Askari, F. Moradi, M. Ajdary, M. Mehdizadeh, Alphalipoic acid mitigates toxic-induced demyelination in the corpus callosum by lessening of oxidative stress and stimulation of polydendrocytes proliferation, Metab Brain Dis 33(1) (2018) 27-37. [97] U. Funfschilling, L.M. Supplie, D. Mahad, S. Boretius, A.S. Saab, J. Edgar, B.G. Brinkmann, C.M. Kassmann, I.D. Tzvetanova, W. Mobius, F. Diaz, D. Meijer, U. Suter, B. Hamprecht, M.W. Sereda, C.T. Moraes, J. Frahm, S. Goebbels, K.A. Nave, Glycolytic oligodendrocytes maintain myelin and long-term axonal integrity, Nature 485(7399) (2012) 517-21. [98] D. Mehta, C. Miller, D.L. Arnold, E. Bame, A. Bar-Or, R. Gold, J. Hanna, L. Kappos, S. Liu, A. Matta, J.T. Phillips, D. Robertson, C.A. von Hehn, J. Campbell, K. Spach, L. Yang, R.J. Fox, Effect of dimethyl fumarate on lymphocytes in RRMS: Implications for clinical practice, Neurology 92(15) (2019) e1724-e1738. [99] Q. Weng, J. Wang, J. Wang, B. Tan, J. Wang, H. Wang, T. Zheng, Q.R. Lu, B. Yang, Q. He, Folate Metabolism Regulates Oligodendrocyte Survival and Differentiation by Modulating AMPKalpha Activity, Sci Rep 7(1) (2017) 1705. [100] R. Schoenfeld, A. Wong, J. Silva, M. Li, A. Itoh, M. Horiuchi, T. Itoh, D. Pleasure, G. Cortopassi, Oligodendroglial differentiation induces mitochondrial genes and inhibition of mitochondrial function represses oligodendroglial differentiation, Mitochondrion 10(2) (2010) 143-50. [101] J. Wang, C. Zhao, P. Kong, H. Sun, Z. Sun, G. Bian, Y. Sun, L. Guo, Treatment with NAD(+) inhibited experimental autoimmune encephalomyelitis by activating AMPK/SIRT1 signaling pathway and modulating Th1/Th17 immune responses in mice, Int Immunopharmacol 39 (2016) 287-94. [102] I. Ziabreva, G. Campbell, J. Rist, J. Zambonin, J. Rorbach, M.M. Wydro, H. Lassmann, R.J. Franklin, D. Mahad, Injury and differentiation following inhibition of mitochondrial respiratory chain complex IV in rat oligodendrocytes, Glia 58(15) (2010) 1827-37. [103] L.R. Stein, S. Imai, Specific ablation of Nampt in adult neural stem cells recapitulates their functional defects during aging, EMBO J 33(12) (2014) 132140. [104] B. Jablonska, M. Gierdalski, L.J. Chew, T. Hawley, M. Catron, A. Lichauco, J. Cabrera-Luque, T. Yuen, D. Rowitch, V. Gallo, Sirt1 regulates glial progenitor proliferation and regeneration in white matter after neonatal brain injury, Nat Commun 7 (2016) 13866.

Jo

ur

na

lP

re

-p

ro

of

Journal Pre-proof

67

Journal Pre-proof

ro

of

Graphical abstract

-p

Highlights

Chronic high fat diet impairs TCA cycle function in the mouse spinal cord



Mitochondria dysfunction occurs in the spinal cord after high fat consumption



Excess fat promotes mitochondrial dysfunction in purified oligodendrocytes



Chronic high fat diet causes oxidative stress in the brain and spinal cord



Chronic high fat diet results in oligodendrocyte loss in the brain and spinal cord

Jo

ur

na

lP

re



68