International Journal of Molecular Sciences

Article β-Hydroxybutyrate Elicits Favorable Mitochondrial Changes in Skeletal Muscle

Brian A. Parker 1, Chase M. Walton 1, Sheryl T. Carr 1, Jacob L. Andrus 1 ID , Eric C. K. Cheung 1, Michael J. Duplisea 1, Esther K. Wilson 1, Carrie Draney 2, Daniel R. Lathen 2 ID , Kyle B. Kenner 2, David M. Thomson 1, Jeffery S. Tessem 2 ID and Benjamin T. Bikman 1,*

1 Department of Physiology and Developmental Biology, Brigham Young University, Provo, UT 84604, USA; [email protected] (B.A.P.); [email protected] (C.M.W.); [email protected] (S.T.C.); [email protected] (J.L.A.); [email protected] (E.C.K.C.); [email protected] (M.J.D.); [email protected] (E.K.W.); [email protected] (D.M.T.) 2 Department of Nutrition, Dietetics and Food Science, Brigham Young University, Provo, UT 84604, USA; [email protected] (C.D.); [email protected] (D.R.L.); [email protected] (K.B.K.); [email protected] (J.S.T.) * Correspondence: [email protected]; Tel.: +1-801-422-1798

 Received: 10 July 2018; Accepted: 27 July 2018; Published: 1 August 2018 

Abstract: The clinical benefit of has historically and almost exclusively centered on neurological conditions, lending insight into how alter mitochondrial function in neurons. However, there is a gap in our understanding of how ketones influence mitochondria within skeletal muscle cells. The purpose of this study was to elucidate the specific effects of β-hydroxybutyrate (β-HB) on muscle cell mitochondrial physiology. In addition to increased cell viability, murine myotubes displayed beneficial mitochondrial changes evident in reduced H2O2 emission and less mitochondrial fission, which may be a result of a β-HB-induced reduction in ceramides. Furthermore, muscle from rats in sustained ketosis similarly produced less H2O2 despite an increase in mitochondrial respiration and no apparent change in mitochondrial quantity. In sum, these results indicate a general improvement in muscle cell mitochondrial function when β-HB is provided as a fuel.

Keywords: ketones; β-hydroxybutyrate; mitochondria; ceramides

1. Introduction Once considered “metabolic garbage,” ketones have become the focus of significant efforts within the realm of cardiometabolic research. Recent discoveries have revealed that ketones, such as acetoacetate and its precursor β-hydroxybutyrate (β-HB), are not only viable fuel sources for all cells with mitochondria, including the brain [1] but are also legitimate signaling molecules, eliciting advantageous changes in inflammation [2], cognition [3,4], oxidative stress [5] and more. Beyond pathology, ketones may also be a relevant metabolic fuel in the context of physical activity, improving athletic performance [6] and myocardial adenosine triphosphate (ATP) generation [7]. Relevant, if not central, to each of these instances is the degree to which the ketones alter mitochondrial function. Unsurprisingly, mitochondria are responsive to ketones, as it is converted to acetyl-Coa and catabolized within the mitochondrial matrix. However, and fatty acid oxidation notably utilizes the same oxidative processes (i.e., via acetyl-Coa conversion) without eliciting the same numerous cellular benefits noted above. Thus, simple oxidation of the fails to explain the disparate effects of these nutrients on organismal function.

Int. J. Mol. Sci. 2018, 19, 2247; doi:10.3390/ijms19082247 www.mdpi.com/journal/ijms Int. J. Mol. Sci. 2018, 19, 2247 2 of 12 Int. J. Mol. Sci. 2018, 19, x 2 of 12

Skeletal muscle represents the largest -responsive tissue in the body and a defect in Skeletal muscle represents the largest insulin-responsive tissue in the body and a defect in insulin insulin signaling in muscle critically contributes to the mounting glycemic burden evident with signaling in muscle critically contributes to the mounting glycemic burden evident with prolonged prolonged insulin resistance, eventually resulting in frank type 2 diabetes. Disrupted mitochondrial insulin resistance, eventually resulting in frank type 2 diabetes. Disrupted mitochondrial function function is thought to at least contribute to muscle insulin resistance [8,9], likely due to increased is thought to at least contribute to muscle insulin resistance [8,9], likely due to increased oxidative oxidative stress and altered insulin signaling [10–12], both of which may be affected by changes in stress and altered insulin signaling [10–12], both of which may be affected by changes in mitochondrial mitochondrial morphology [8]. morphology [8]. To a degree, previous findings of beneficial mitochondrial adaptations to ketones may offer To a degree, previous findings of beneficial mitochondrial adaptations to ketones may offer insight insight into the benefits of ketosis, a state of mildly elevated blood ketones. Whether through the use into the benefits of ketosis, a state of mildly elevated blood ketones. Whether through the use of of a or the consumption of exogenous ketones, limited evidence suggests a generally a ketogenic diet or the consumption of exogenous ketones, limited evidence suggests a generally favorable metabolic milieu [13–16]. To this end, the purpose of this study was to elucidate the effects favorable metabolic milieu [13–16]. To this end, the purpose of this study was to elucidate the effects of of β-HB on myriad aspects of skeletal muscle mitochondrial physiology, including respiration, ATP β-HB on myriad aspects of skeletal muscle mitochondrial physiology, including respiration, ATP and and H2O2 emission and morphology. H2O2 emission and morphology.

2.2. Results Results

2.1.2.1. ββ--HydroxybutyrateHydroxybutyrate Elicits Elicits Favorable Favorable Changes Changes in in Mitochondrial Mitochondrial Respiration Respiration in in Muscle Muscle C Cellsells FollowingFollowing 24 24 h h of of treatment treatment with with β-hydroxybutyrateβ-hydroxybutyrate β-HB),β-HB), C2C12 C2C12 myotubes myotubes had hadslight, slight, yet significantyet significant elevations elevations in mitochondrial in mitochondrial respiration respiration raterate (Figure (Figure 1A),1A), including including after after the the additi additionon of ADPof ADP (GMD) (GMD) that continued that continued with addition with addition of succinate of succinate (GMSD) (GMSD)but not with but carbonyl not with cyanide carbonyl-4- (trifluoromethoxy)phenylhydrazonecyanide-4-(trifluoromethoxy)phenylhydrazone (FCCP). These (FCCP). changes These were changes further were evident further in aevident significant in a elevationsignificant in elevation respiratory in respiratory control ratio control (RCR), ratio a general (RCR), aindicator general indicatorof mitochondrial of mitochondrial “fitness” “fitness”(Figure 1B).(Figure Uncoupling1B). Uncoupling control controlratio (UCR; ratio Figure (UCR; 1C) Figure was1C) similar was similar across acrossthe treatments. the treatments. AnalysisAnalysis of of mitochondrial mitochondrial respiration respiration (Figure (Figure 11AA–C)–C) simply indicates the rate of oxygen use. Thus, we sought to better understand the the func functionaltional effect effect of of this altered oxygen use use by measuring ATP production and H2OO22 emission.emission. Firstly, Firstly, we we found found that that ATP ATP production production was was similar similar across across the the treatmentstreatments——β--HBHB addition addition had had no no significant significant effect, effect, despite despite an upward an upward trend (Figure trend1 D,E). (Figure However, 1D,E). However,H2O2 emission H2O2 (Figureemission1F) (Figure tended 1F) to tended change, to significantly change, significantly diminishing diminishing when compared when compared with O 2 withconsumption O2 consumption (Figure1 (FigureG). 1G).

250 A CON bHB 200

) ** g

150

m

/

x c

u *

l

e

F s

/ l

2 100

o

O

m

p ( 50

0 GM D S F GML GMD GMSD GMSF 2.0 1.5 B C 1.5 *

1.0

R

R C

1.0 C

R U

0.5 0.5

0.0 0.0 CON bHB CON bHB 0.7 0.007 D 0.6 FigureE 1. Cont.0.006

) 0.5 0.005

g

o

m i /

0.4 t 0.004

c

a

P

e

R

T s

/ 0.003 l

A 0.3

O

:

o P

m 0.2 0.002

p ( 0.1 0.001

0 0 CON bHB CON bHB 140 1.5

F 120 G **

) 100

g 1

o

i

m t

/ 80

a

c 2

R

e

O

2 s

2 60

/

l

H O

: o

2 0.5

40

m

O

2

p ( 20 H

0 0 CON bHB CON bHB 250 A CON bHB 220500 A CON

) **

bHB g

125000

m

/

x c

u *

l e

) **

F s

/

g

l 2

110500

o

m

O

/

x

m c

u *

l

e

p

(

F s

/ l

2 15000

o

O

m

p ( 500 GM D S F GML GMD GMSD GMSF 2.0 1.5 0 GM D S F B GML GMD GMCSD GMSF 12.5.0 * 1.5 1.0

BR C R C *

11.0.5 C R

U 1.0

R 0.5 R C 0.5

1.0 C R Int. J. Mol. Sci. 2018, 19, 2247 U 3 of 12 Int. J. Mol. Sci. 2018, 19, x 0.5 3 of 12 00.0.5 0.0 CON bHB CON bHB 0.7 0.007 Int. J. Mol. Sci. 2018, 19, x 3 of 12 0.0 0.0 D 0.6 E 0.006 CON bHB CON bHB 0.7 ) 0.007

0.5 0.005

g

o

m i

D/ 0.6 E 0.006

0.4 t 0.004

c

a

P

e

R

)

T

s 0.5 0.005 /

g 0.003 l

A 0.3

O

o

:

o

m i

/ t

0.4 P 0.004

m c

0.2 a 0.002

P

e

p

R

(

T s

/ 0.003 l A 0.3 O 0.001

0.1 :

o P m 0.2 0.002

p 0 0 ( 0.1 CON bHB 0.001 CON bHB 140 1.5 0 0 F 120 G CON bHB CON bHB 140 1.5 **

) 100

g 1 o

F i t m 120 G

/ 80

a

c 2 R

e ** O

) 100

2 s

2 60 /

g 1

l

o

H O

i

:

o

m t

2 0.5 / 80

40 a

m

c 2

O

R

2

e

p

O

(

2

s 2

60 H /

l 20

H O

: o

2 0.5

40 m

0 O 0

2

p ( CON bHB H CON bHB 20 0 0 FigureFigure 1. β -Hydroxybutyrate1. β-HydroxybutyrateCON favorably favorablyb altersaltersHB mitochondrial function. function.CON C2C12 C2C12 myotubes myotubesbH Bwere weretreated treated

withwith PBS PBS (CON) (CON) or withor withβ-Hydroxybutyrate β-Hydroxybutyrate (β--HB;HB; 5 5 mM) mM) for for 24 24 h. h. To To measure measure mitochondrial mitochondrial respirationFigure 1. β -(HydroxybutyrateA), cells were treated favorably with GM altersL: glutamate mitochondrial (10 mM) function. + malate C2C12 (2 mM);myotubes GMD were: + ADP treated (2.5 respiration (A), cells were treated with GML: glutamate (10 mM) + malate (2 mM); GMD: + ADP mM);with PBS GMS (CON)D: + succinate or with (10 mM); β-Hydroxybutyrate GMSF: + FCCP (β-HB (0.05; 5 mM)μM). forRespiratory 24 h. To control measure ratio mitochondrial ((B); RCR; (2.5 mM); GMSD: + succinate (10 mM); GMSF: + FCCP (0.05 µM). Respiratory control ratio ((B); GMrespirationD/GML) ( andA), cells uncoupling were treated control with ratio GM L(: (glutamateC); UCR; (10 GMS mM)F/GMS + malateD) were (2 mM); determined GMD: + fromADP the(2.5 RCR; GM /GM ) and uncoupling control ratio ((C); UCR; GMS /GMS ) were determined from the respirationmM);D GMS assay.DL: + succinate ATP production (10 mM); (D GMS), ATP:OF: + FCCP2 ratio (P:O; (0.05 (μEM).), H 2 RespiratoryOF2 productionD control (F) and ratio H2 O((2B:O);2 ratio RCR; respiration(GMG) wereD/GM assay. determinedL) and ATP uncoupling production by measuring control (D ),fluorescence. ATP:O ratio 2((ratioC) ;* p UCR; (P:O;< 0.05 GMS; ( E**), pFH

12

y a

s 1104

As

A

e 182

u

y

l

a

B s

160

r

s

a

A

m

e 48

a

l

u

l

A B

26

r a

m 04

a l

A CON bHB 2 B 2.5 0 CON bHB

2 e

c 2.5

Bn a

b 1.5 r

o 2

e

s

c b

n 1

A

a T

b 1.5

r

T o

M 0.5 s

b 1

A

T 0 T M 0.5 CON bHB Figure 2. β-Hydroxybutyrate increases0 myotube viability. Following treatment with PBS (CON) or β- CON bHB Hydroxybutyrate (β-HB; 5 mM) for 24 h, myotubes were measured for viability via Alamar Blue (A) FigureorFigure with 2. β -Hydroxybutyrate2.MTT β-Hydroxybutyrate (B) to determine increases increasesmetabolic myotube activity. viability.p viability.< 0.05; n Following = Following6. treatment treatment with PBS with (CON) PBS (CON)or β- or β-HydroxybutyrateHydroxybutyrate (β (β-HB;-HB;5 5 mM)mM) for 24 24 h, h, myotubes myotubes were were measured measured for forviability viability via Alamar via Alamar Blue Blue(A) (A) or with MTT (B) to determine metabolic activity. p < 0.05; n = 6. or with MTT (B) to determine metabolic activity. p < 0.05; n = 6.

Int. J. Mol. Sci. 2018, 19, 2247 4 of 12 Int. J. Mol. Sci. 2018, 19, x 4 of 12

2.3. β-Hydroxybutyrate Induces Mitochondrial Fusion 2.3. β-Hydroxybutyrate Induces Mitochondrial Fusion We previously found that mitochondrial morphology alters mitochondrial physiology, We previously found that mitochondrial morphology alters mitochondrial physiology, including including respiration and reactive oxygen species production [8]. In an effort to better understand a respiration and reactive oxygen species production [8]. In an effort to better understand a potential non-energetic mechanism whereby β-HB alters mitochondrial respiration, we scrutinized potential non-energetic mechanism whereby β-HB alters mitochondrial respiration, we scrutinized mitochondrial morphology and amount in C2C12 myotubes following β-HB treatment. There were mitochondrial morphology and amount in C2C12 myotubes following β-HB treatment. There were no no detectable differences in mitochondrial amount between treatments (Figure 3D). Whereas the detectable differences in mitochondrial amount between treatments (Figure3D). Whereas the control control cells had a generally even degree of mitochondria that exhibited contact or no contact with cells had a generally even degree of mitochondria that exhibited contact or no contact with adjacent adjacent mitochondria (Figure 3A), which we defined as fusion or fission, respectively, the β-HB- mitochondria (Figure3A), which we defined as fusion or fission, respectively, the β-HB-treated cells treated cells exhibited a shift in increased mitochondrial fusion (Figure 3B,C). These morphological exhibited a shift in increased mitochondrial fusion (Figure3B,C). These morphological differences were differences were supported by disparate levels of mitochondrial proteins that mediate fusion and supported by disparate levels of mitochondrial proteins that mediate fusion and fission. While β-HB fission. While β-HB treatment elicited no significant effect on MFN2, which promotes fusion, it did treatment elicited no significant effect on MFN2, which promotes fusion, it did significantly reduce significantly reduce DRP1, a key protein known to mediate mitochondrial fission (Figure 3D,E). DRP1, a key protein known to mediate mitochondrial fission (Figure3D,E). A B

10 µm CON bHB

C 75% D 1,000,000

Fusion Fission ** e 900,000

c n

e 800,000

c

s d

e 700,000

e r

r 50%

u e

o 600,000

l

t l

# F

a

d 500,000 t

*** e

n R

400,000

e r e

c 25% k

r 300,000

c

e

a

r P

t 200,000

o t

i 100,000 M 0% 0 CON bHB CON bHB E F 3500

) 3000

MFN2 U A

( 2500

n i

t 2000

DRP1 c A

/ 1500 ***

n i

e 1000

t o

Actin r 500 P 0 CON bHB CON bHB CON bHB MFN2 DRP1 Figure 3. β-Hydroxybutyrate increases mitochondrial content and function. C2C12 myotubes were Figure 3. β-Hydroxybutyrate increases mitochondrial content and function. C2C12 myotubes were treated with vehicle (PBS; CON) or with β-Hydroxybutyrate (β-HB; 5 mM) for 24 h then incubated treated with vehicle (PBS; CON) or with β-Hydroxybutyrate (β-HB; 5 mM) for 24 h then incubated with MitoTracker and florescence was imaged to determine morphology (A–C) and quantified by with MitoTracker and florescence was imaged to determine morphology (A–C) and quantified by plate plate reader assay ((D); n = 6). In parallel, cells were used to detect mitofusin-2 (MFN2) and dynamin- reader assay ((D); n = 6). In parallel, cells were used to detect mitofusin-2 (MFN2) and dynamin-related relatedpeptide-1 peptide (DRP1)-1 (DRP1) protein protein expression expression ((E,F); n((E=,F 3).); n ** = p3).< ** 0.01; p < ***0.01;p < *** 0.001 p < for0.001β-HB for β versus-HB versus CON; CON;# p < 0.05# p < for 0.05 fission for fission vs. fusion. vs. fusion.

2.4. Myotube Ceramides Are Reduced with β-HB Treatment Despite being necessary to cellular health, ceramides mediate multiple pathological cellular processes [17–19], including forcing sustained mitochondrial fission [8]. Due to our findings of reduced

Int. J. Mol. Sci. 2018, 19, x 5 of 12

2.4. Myotube Ceramides Are Reduced with β-HB Treatment Despite being necessary to cellular health, ceramides mediate multiple pathological cellular Int. J. Mol. Sci. 2018, 19, 2247 5 of 12 processes [17–19], including forcing sustained mitochondrial fission [8]. Due to our findings of reduced mitochondrial fission in myotubes following β-HB treatment, we probed ceramide levels mitochondrialand found a small fission but in significant myotubes reduction following inβ -HBmultiple treatment, ceramide we probedspecies, ceramideincluding levels C16, C22 and and found C24 a small(Figure but 4). significant reduction in multiple ceramide species, including C16, C22 and C24 (Figure4).

0.020

0.018 CON

0.016 bHB

0.014

) s **

e 0.012

g

d

u

i /

l 0.010

m

o

a

r

m e

p 0.008

( C 0.006 0.004 * 0.002 * *

0.000 C16 C18 C20 C22 C24 C24:1 Total FigureFigure 4.4. β--HydroxybutyrateHydroxybutyrate reduces select ceramide species. Following treatment with vehicle (PBS; CON)CON) oror withwith ββ-Hydroxybutyrate-Hydroxybutyrate (ββ-HB;-HB; 5 mM) for 24 h,h, lipidslipids werewere isolatedisolated fromfrom C2C12C2C12 myotubesmyotubes thenthen analyzedanalyzed viavia lipidomicslipidomics ((nn == 6). * p < 0.05; ** p < 0.01 0.01 for for ββ--HBHB versus versus CON. CON.

2.5.2.5. Ketogenic DietDiet in Rats Alters MuscleMuscle MitochondrialMitochondrial PhysiologyPhysiology RatsRats on on a a ketogenic ketogenic diet diet (KETO) (KETO) diet diet were were pair pair-fed-fed based based on the on consumption the consumption of standard of standard (STD)- (STD)-fedfed rats, such rats, that such caloric that caloric intake intake was not was different not different between between diets. diets.Within Within 1 week 1 weekon the on KETO the KETO diet, diet,body bodyweight weight was significantly was significantly reduced reduced by 6–8% by compared 6–8% compared to the STD to the-fed STD-fed group (Figure group 5A) (Figure and5 thisA) andpersisted this persistedthrough the through 4-week the experimental 4-week experimental period. After period. 3.5 weeks After of 3.5feeding, weeks plasma of feeding, ketones plasma were ketones140% higher were in 140% the KETO higher- inversus the KETO- STD-fed versus rats ( STD-fedFigure 5B; rats 0.49 (Figure vs. 0.25B; mM 0.49, respectively vs. 0.2 mM,). respectively). Muscle mass Musclewas not mass affected was bynot diet affected (Figure by diet5C). (Figure Mitochondrial5C). Mitochondrial respiration respiration was slightly was elevated slightly in elevated the red 2 2 2 inquadriceps the red quadriceps (Figure 5D), (Figure while5 D),H O while to O H ratio2O2 wasto O lower2 ratio in was muscle lower from in muscle KETO- fromfed rats KETO-fed (Figure rats5E). (FigureComponents5E). Components of the oxidative of the phosphorylation oxidative phosphorylation complexes complexes were not significantly were not significantly different differentin KETO inversus KETO STD versus-fed STD-fedgroups, though groups, they though tended they to tended be lower to be in lowerthe KETO in the-fed KETO-fed rats (Figure rats 5F (Figure,H). Citrate5F,H). Citratesynthase synthase activity, activity, on the other on the hand, other was hand, significantly was significantly lower in lower the red in quadriceps the red quadriceps muscles muscles(but not (butwhite not quadriceps) white quadriceps) of KETO of versus KETO versusSTD-fed STD-fed rats (Figure rats (Figure 5G). MFN25G). MFN2 and andDRP1 DRP1 levels levels were were also also not notsignificantly significantly affected affected by diet by diet(Figure (Figure 5I,J),5 I,J),although although a very a very strong strong trend trend (p = 0.051) ( p = 0.051) for decreased for decreased DRP1 DRP1levels levelsin the inKETO the KETO-fed-fed muscle muscle was observed. was observed.

Int.Int. J. Mol. J. Mol. Sci. Sci.2018 2018, 19, ,19 2247, x 6 of6 12 of 12

STD KETO A400 B C *

350

) g

( 300

t

h 250 *

g i

e 200

W

y

d 150 o

B 100 50 0 Pre Wk1 Wk2 Wk3 Wk4 D E p = 0.08

* * p = 0.05

F Diet: STD KETO STD KETO Ponceau G C. V- C. III - * C. IV - C. II -

C. I -

H Diet: STD KETO STD KETO I Diet: STD KETO STD KETO J Diet: STD KETO STD KETO

p = 0.051

FigureFigure 5. Ketogenic 5. Ketogenic diet dietinduces induces weight weight loss and loss enhances and enhances mitochondrial mitochondrial respiration respiration without without increased mitochondrialincreased mitochondrial content in skeletalcontent musclein skeletal from muscle rats. from Fisher rats. 344 Fisher rats were344 rats pair-fed were pair either-fed a either standard a (STD)standard or ketogenic (STD) or (KETO) ketogenic diet (KETO) for 4 weeks. diet for Body 4 weeks. weight Body (A weight) was measured (A) was measured weekly. Atweekly. the end At ofthe the treatmentend of period, the treatment blood and period, skeletal blood muscle and wereskeletal removed muscle from were the removed rats. Plasma fromβ the-hydroxybutyrate rats. Plasma β- (B) andhydroxybutyrate gastrocnemius (Gastroc) (B) and weight gastrocnemius (C) were (Gastroc) measured. weight Mitochondrial (C) were respiration measured. (D Mitochondrial) and H2O2:O 2 Figure 5. Ketogenic diet induces ratiowrespiratione (Ei)g wereht ( determinedDlo) andss Ha2On2 as:Od2 in ratioe Figuren (hE) a1were.n Componentsc determines medi oft aso the inc hFigure oxidativeon 1.d Componentsr phosphorylationial res ofp theira systemoxidativetion ( F ),without increased citratephosphorylation synthase activity system (G) and(F), cytochrome citrate synthase C content activity (H ) ( wereG) and measured cytochrome as markers C content of mitochondrial (H) were content.measured Protein as markers expression of mitochondrial for mitofusin-2 content. ((I); MFN2)Protein expression and dynamin-related for mitofusin peptide-1-2 ((I); MFN2) ((J); DRP1)and proteindynamin expression-related werepeptide assessed-1 ((J); DRP1) as markers protein of expression mitochondrial were fusion assessed vs. as fission. markers * p of< mitochondrial 0.05; n = 6. fusion vs. fission. * p < 0.05; n = 6. 3. Discussion 3. Discussion The purpose of this study was to establish an underlying understanding of the actions of ketones The purpose of this study was to establish an underlying understanding of the actions of ketones on skeletal muscle cell physiology and mitochondrial function. We found that β-hydroxybutyrate on skeletal muscle cell physiology and mitochondrial function. We found that β-hydroxybutyrate (β- (β-HB) increased mitochondrial respiration, with no loss of ATP production, yet a significant reduction HB) increased mitochondrial respiration, with no loss of ATP production, yet a significant reduction in H2O2 emission. Furthermore, β-HB enhanced muscle viability, promoting survival in the wake of a in H2O2 emission. Furthermore, β-HB enhanced muscle viability, promoting survival in the wake of noxious stimulus. Lastly, β-HB promoted a general shift towards mitochondrial fusion, which may

Int. J. Mol. Sci. 2018, 19, 2247 7 of 12 be a result of reduced ceramides. Altogether, these results suggest a favorable influence of β-HB on muscle cell function. Current dietary patterns, including macronutrient composition and meal frequency, result in very few instances of ketosis due to consistent elevations in insulin, which inhibits ketogenesis. However, ketosis, a stated of mildly elevated ketones, can be achieved through either dietary intervention (i.e., fasting or low- diet) or the consumption of exogenous ketones. While neither intervention has been studied in the context of understanding ketone-induced muscle cell mitochondrial physiology to the extent outlined in our study, there are nevertheless clues that suggests enhanced mitochondrial function in muscle. The bulk of research exploring the mitochondrial effects of ketones use neurons as the cell of interest, which is not surprising given the pronounced neurological benefits of ketones, including reduced seizures [20], migraines [21] and improved cognition in both states of compromised [22] and healthy [23] neurological function. Some of these reported mitochondrial benefits of ketones in neurons are a reduction in oxidative stress and enhanced survival [24], similar to what we found in muscle cells in the studies herein. Despite this neuron-heavy focus, some previous evidence exists that suggests a mitochondrial benefit in muscle, albeit indirectly. Ahola-Erkkila et al. [25], in studying a model of mitochondrial myopathy, found that β-HB helped maintain mitochondrial respiration and morphology within muscle tissue, potentially slowing muscle loss with myopathies. This, combined with our observation of enhanced β-HB-induced myotube viability, presents a provocative paradigm with states of ketosis—that ketones promote muscle cell survival. Thus, perhaps ketones, in addition to serving other roles, act to preserve muscle mass in insulin-reduced or -deficient states, such as untreated type 1 diabetes, fasting, or a ketogenic diet. Beyond skeletal muscle, it is tempting to speculate that ketones may also alter cardiac muscle viability. Aubert et al. [26] recently found that heart failure due to hypertrophic cardiomyopathy is typified by a fuel shift, wherein cardiomyocytes rely more heavily on ketone catabolism. Moreover, Sato et al. [7] observed an increased ATP efficiency in myocardium fueled with ketones compared with other fuel sources. Our findings of increased muscle cell viability when incubated with ketones may explain observations of maintained, or even increased, muscle mass in humans when fed an isocaloric, low-carbohydrate ketogenic diet. Specifically, Volek et al. [27] found that lean body mass increased roughly 1 kg in six weeks compared with control-fed subjects, which may stem from a ketone-induced muscle protein sparing effect [6,28,29]. Regardless of whether ketones induce a muscle-sparing effect via enhanced muscle cell viability, ketosis does not appear to hinder muscle function. Our findings of maintained ATP production, reduced H2O2 emission and increased muscle cell viability may provide molecular insight into how muscle performance is enhanced, or at least not compromised, when relying more heavily on ketones as ketone availability is increased. Ketone-induced enhanced muscle cell viability may not only reveal a novel mechanism whereby muscle is able to resist damage following intense physical activity but also perhaps in explicit muscle pathologies, such as muscle dystrophy [30]. Much of the mitochondrial and viability improvements in muscle cells could be explained by the β-HB-induced reduction in ceramides. Ceramides mediate deleterious changes in both mitochondrial function [8] and nutrient transport [31]. Moreover, the reduction in ketones may elicit improved viability due to a reduction in ceramide-mediated apoptotic signaling [17]. To determine the effect of nutritional ketosis on skeletal muscle in vivo, rats were fed a very low carbohydrate ketogenic diet for 4 weeks. Despite equal caloric intake, the KETO-fed rats weighed significantly less after 1 week on KETO versus STD-fed controls. A loss of muscle mass did not account for this weight loss; diet did not affect gastrocnemius weight. It may be due, at least in part, to a loss of glycogen and associated water from the liver and muscle, though this was not measured in the current study and evidence in humans suggests muscle glycogen is comparable in low-carbohydrate-fed to high-carbohydrate-fed individuals [32]. We observed decreased maximal citrate synthase activity in red but not white, muscle from KETO versus STD-fed rats, which may indicate reduced total mitochondrial number. This is consistent with Int. J. Mol. Sci. 2018, 19, 2247 8 of 12 previous work [33] in which similar findings were found in the mixed gastrocnemius of rats fed a ketogenic diet for 8 weeks. Despite the reduction in citrate synthase activity, red quadriceps muscles from KETO-fed rats had increased respiration, perhaps evidence of a compensatory mechanism. Moreover, consistent with our findings in βHB-treated myotubes, KETO feeding also resulted in a very nearly significant (p = 0.051) reduction in DRP1 protein content, suggesting that nutritional ketosis protects muscle against mitochondrial fragmentation. This effect is likely specific to an isocaloric ketogenic diet versus ad libitum high fat (60%) diet, which was reported to increase DRP1 in young rats [34] and may explain the improved H2O2 emission, something we’ve reported previously [8]. Our observed changes in DRP1 tempt the conclusion that KETO alters ceramides in whole muscle tissue [8]. However, the lack of ceramide analysis from the in vivo studies prevents such a conclusion and is an obvious limitation. We expect to report on the specific ketone-ceramide relationship in whole tissue in the near future. In sum, the results of this study lend support for the growing scientific reevaluation of ketones as viable, even beneficial, metabolic signals. Moreover, our findings elucidate a potential mechanism whereby ketones elicit a protective effect on muscle cells and help maintain an overall favorable metabolic milieu. Future studies from human muscle tissue in states of ketosis are necessary to establish the relevance of these findings.

4. Materials and Methods

4.1. Cell Culture C2C12 murine myoblast cells were maintained in DMEM (Dulbecco’s modified Eagle’s medium; D6546, Sigma-Aldrich, Saint Louis, MO, USA) plus 10% FBS (Invitrogen, Carlsbad, CA, USA). For differentiation into myotubes, C2C12 myoblasts were grown to confluency and the medium was replaced with DMEM plus 10% horse serum (Invitrogen). Myotubes were used for experiments on day 4 of differentiation. For β-hydroxybutyrate treatments (β-HB), cells were incubated with β-HB (54965, Sigma-Adrich) for 24 h at 5 mM.

4.2. Animals All animal procedures were approved (18-0101, 9 February 2018) by the institutional animal care and use committee at Brigham Young University. 5-month-old male Fisher 344 rats (n = 6/group) were acclimatized for 1 week after arrival at the animal facility and were then pair-fed with either standard diet (STD; Envigo Teklad Rodent Diet, 8604; 32% protein, 14% fat, 54% carbohydrate) or ketogenic diet (KETO; Envigo Teklad custom diet, TD.10911; 22.4% protein, 77.1% fat, 0.5% carbohydrate) for 4 weeks. Each day the chow remaining in the cage was weighed and replaced with fresh chow. Body weight for the rats was recorded weekly. At 4 weeks, the rats were euthanized and blood, gastrocnemius and quadriceps muscles were removed. Plasma ketone levels were measured using the Abbott Precision Xtra Ketone Monitoring System. White and red quadriceps muscle was separated and a small sample of the red quadriceps (~10 mg) was prepared for assessment of mitochondrial respiration as described above. The remaining muscle was frozen at the temperature of liquid nitrogen and stored at −90 ◦C for later analysis.

4.3. Cell Viability Assays Cells were plated at a concentration of 2 × 105 cells/mL in 24-well plates (at 1 mL/well) or in 96-well plates (at 100 µL/well), then cultured for 48 h total with vehicle (water; CON) or 5 mM β-HB (β-HB). Cellular proliferation was determined by MTT assays (Sigma-Aldrich) and Alamar Blue assays (Sigma-Aldrich). Absorbance for the MTT and Alamar Blue assays were determined on a BioTek Synergy 2 plate reader. Int. J. Mol. Sci. 2018, 19, 2247 9 of 12

4.4. Mitochondrial Morphology C2C12 myoblasts were grown to confluence in chamber slides (NUNC Lab-Tek II Chambered Coverglass System; 155382) and differentiated at day 4. The mitochondrial dye MitoTracker Red CMXRos (Molecular Probes, Eugene, OR, USA; M7512), dissolved in anhydrous DMSO, was added to cultured myotubes at a concentration of 250 nM. The cells were incubated for 30 min at 37 ◦C in the dark and then visualized using a confocal microscope (Olympus IX81, Center Valley, PA, USA). Following image capture, a blind assessment of mitochondrial morphology was performed and quantified.

4.5. Mitochondrial Respiration Cells and tissue were prepared for mitochondrial respiration as described previously [35–38] before being transferred to respirometer chambers using the Oroboros O2K oxygraph (Oroboros, Innsbruck, Austria). Electron flow through complex I was supported by glutamate + malate (10 mM and 2 mM, respectively) to determine leak oxygen consumption (GML). Following stabilization, adenosine diphosphate (ADP) (2.5 mM) was added to determine oxidative phosphorylation capacity (GMD). Succinate was added (GMSD) for complex I + II electron flow into the Q-junction. To determine full electron transport system capacity in cells over oxidative phosphorylation, the chemical uncoupler carbonyl cyanide 4-(trifluoromethoxy) phenylhydrazone (FCCP) was added (0.05 µM, followed by 0.025 µM steps until maximal O2 flux was reached). Lastly, residual oxygen consumption was measured by adding antimycin A (2.5 µM) to block complex III action, effectively stopping any electron flow, which provides a baseline rate of respiration. Following respiration protocol, samples were removed from the chambers and used for further analysis, including protein quantification.

4.6. ATP Analysis Following the culture period, cells were transferred to 2.5 mM glucose in SAB buffer for 2 h, followed by transfer to either 2.5 mM glucose SAB buffer or 16.7 mM glucose SAB buffer for 1 h. Cells were washed with PBS, harvested by trypsinisation and pelleted by centrifugation. The cells were lysed in 150 µL 1M perchloric acid on ice to precipitate cellular proteins. Lysate was centrifuged at 20,000× g for 10 min, after which 150 µL supernatant was transferred to a new tube with 150 µL 1M KOH. ATP was quantified using the ATP assay kit (Life Technologies, Carlsbad, CA, USA).

4.7. H2O2 Emission

H2O2 emission was measured using an Amplex Red Hydrogen Peroxide/Peroxidase Assay kit (Molecular Probes; A22188) as described previously [8]. A reaction mixture containing 50 µM Amplex Red and 0.1 unit/mL HRP in KRPG (Krebs-Ringer phosphate glucose) buffer was prepared (145 mM NaCl, 5.7 mM sodium phosphate, 4.86 mM KCl, 0.54 mM CaCl2, 1.22 mM MgSO4 and 5.5 mM glucose). The reaction mixture was pre-warmed in a 96-well plate with 100 µL of mixture per well. A 20 µL aliquot of cells suspended in KRPG buffer (~1.5 × 104 cells) were added to each well. Samples were incubated for 1 h. Fluorescence was measured with a microplate reader (Molecular Devices; San Jose, CA, USA).

4.8. Lipids Lipids were quantified by shotgun lipidomics using an ABI 5600+ (AB Sciex, Framingham, MA, USA), as described previously [39,40]. Briefly, we simultaneously identified changes in hundreds of distinct lipid species via a nonbiased approach following direct infusion of extracted lipids containing 18 mM ammonium fluoride to aid in ionization of neutral lipids and to reduce salt adducts. Data from the AB Sciex 5600+ was collected and calibrated with Analyst and PeakView Software (AB Sciex). The in-house-developed Lipid Explorer software assists with simplifying the data by identifying lipid species based on exact mass and fragmentation patterns. Int. J. Mol. Sci. 2018, 19, 2247 10 of 12

4.9. Tissue Homogenization Muscles were ground-glass homogenized in lysis buffer (50 mM Tris-HCl, pH 7.4; 250 mM mannitol; 50 mM NaF; 5 mM sodium pyrophosphate; 1 mM EDTA; 1 mM EGTA; 1% Triton X-100; 50 mM β-glycerophosphate; 1 mM sodium orthovanadate; 1 mM DTT; 1 mM benzamidine; 0.1 mM phenylmethane sulfonyl fluoride; 5 µg/mL soybean trypsin inhibitor). An aliquot of the homogenate was separated for the citrate synthase assay, while the rest was centrifuged at 10,000× g for 10 min. The supernatant was retained for western blotting analysis.

4.10. Citrate Synthase Activity Whole, uncentrifuged homogenates were assayed for citrate synthase activity as described previously [41], with the exception that reaction volumes were scaled down to a final volume of 200 µL to be run in 96-well plates.

4.11. Western Blotting Muscle homogenates were analyzed for protein concentration using a modified Lowry assay (DC Protein Assay; Bio-Rad Laboratories, Hercules, CA, USA) according to manufacturer’s protocols. Equal amounts of protein were then separated by electrophoresis and transferred to polyvinylidene fluoride (PVDF) membranes. Proper transfer and equal protein loading were verified by Ponceau-S staining of the PVDF membranes after transfer. Primary antibodies [OXPHOS (Invitrogen #457999), cytochrome C (Santa Cruz #sc-13156), MFN2 (EMD-Millipore #ABC42), DRP1 (Cell Signaling, #8570)] were applied overnight at 4 ◦C. Membranes were exposed to autoradiographic film and resulting band intensities were determined using Gel-Pro software (Media Cybernetics, Rockville, MD, USA).

4.12. Statistical Methods Data are presented as means ± SEM. Data were compared with Student’s t-test (Graphpad Prism; Microsoft Excel). Significance was set at p < 0.05.

Author Contributions: B.A.P., C.M.W. and B.T.B. conceived the hypothesis and study design. B.A.P., C.M.W., S.T.C., J.L.A., C.D., E.C.K.C., M.J.D., E.K.W., D.R.L., K.B.K., D.M.T., J.S.T. and B.T.B. conducted the experiments, compiled and analyzed data. J.S.T., D.M.T. and B.T.B. wrote the manuscript. Funding: This research was supported by an endowment from Kevin and Mimi Sayer. Conflicts of Interest: The authors declare no conflict of interest.

References

1. Owen, O.E.; Morgan, A.P.; Kemp, H.G.; Sullivan, J.M.; Herrera, M.G.; Cahill, G.F., Jr. Brain metabolism during fasting. J. Clin. Investig. 1967, 46, 1589–1595. [CrossRef][PubMed] 2. Youm, Y.H.; Nguyen, K.Y.; Grant, R.W.; Goldberg, E.L.; Bodogai, M.; Kim, D.; D’Agostino, D.; Planavsky, N.; Lupfer, C.; Kanneganti, T.D.; et al. The ketone metabolite beta-hydroxybutyrate blocks NLRP3 inflammasome-mediated inflammatory disease. Nat. Med. 2015, 21, 263–269. [CrossRef][PubMed] 3. Ijff, D.M.; Postulart, D.; Lambrechts, D.; Majoie, M.; de Kinderen, R.J.A.; Hendriksen, J.G.M.; Evers, S.; Aldenkamp, A.P. Cognitive and behavioral impact of the ketogenic diet in children and adolescents with refractory epilepsy: A randomized controlled trial. Epilepsy Behav 2016, 60, 153–157. [CrossRef][PubMed] 4. Kashiwaya, Y.; Bergman, C.; Lee, J.H.; Wan, R.; King, M.T.; Mughal, M.R.; Okun, E.; Clarke, K.; Mattson, M.P.; Veech, R.L. A ketone ester diet exhibits anxiolytic and cognition-sparing properties, and lessens amyloid and tau pathologies in a mouse model of Alzheimer’s disease. Neurobiol. Aging 2013, 34, 1530–1539. [CrossRef] [PubMed] 5. Kim, D.Y.; Davis, L.M.; Sullivan, P.G.; Maalouf, M.; Simeone, T.A.; van Brederode, J.; Rho, J.M. are protective against oxidative stress in neocortical neurons. J. Neurochem. 2007, 101, 1316–1326. [CrossRef] [PubMed] Int. J. Mol. Sci. 2018, 19, 2247 11 of 12

6. Cox, P.J.; Kirk, T.; Ashmore, T.; Willerton, K.; Evans, R.; Smith, A.; Murray, A.J.; Stubbs, B.; West, J.; McLure, S.W.; et al. Nutritional Ketosis Alters Fuel Preference and Thereby Endurance Performance in Athletes. Cell Metab. 2016, 24, 256–268. [CrossRef][PubMed] 7. Sato, K.; Kashiwaya, Y.; Keon, C.A.; Tsuchiya, N.; King, M.T.; Radda, G.K.; Chance, B.; Clarke, K.; Veech, R.L. Insulin, ketone bodies, and mitochondrial energy transduction. FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol. 1995, 9, 651–658. [CrossRef] 8. Smith, M.E.; Tippetts, T.S.; Brassfield, E.S.; Tucker, B.J.; Ockey, A.; Swensen, A.C.; Anthonymuthu, T.S.; Washburn, T.D.; Kane, D.A.; Prince, J.T.; et al. Mitochondrial fission mediates ceramide-induced metabolic disruption in skeletal muscle. Biochem. J. 2013, 456, 427–439. [CrossRef][PubMed] 9. Petersen, K.F.; Befroy, D.; Dufour, S.; Dziura, J.; Ariyan, C.; Rothman, D.L.; DiPietro, L.; Cline, G.W.; Shulman, G.I. Mitochondrial dysfunction in the elderly: Possible role in insulin resistance. Science 2003, 300, 1140–1142. [CrossRef][PubMed] 10. Shikuma, C.M.; Day, L.J.; Gerschenson, M. Insulin resistance in the HIV-infected population: The potential role of mitochondrial dysfunction. Curr. Drug Targets Infect. Disord. 2005, 5, 255–262. [CrossRef][PubMed] 11. Anderson, E.J.; Lustig, M.E.; Boyle, K.E.; Woodlief, T.L.; Kane, D.A.; Lin, C.T.; Price, J.W., 3rd; Kang, L.;

Rabinovitch, P.S.; Szeto, H.H.; et al. Mitochondrial H2O2 emission and cellular redox state link excess fat intake to insulin resistance in both rodents and humans. J. Clin. Investig. 2009, 119, 573–581. [CrossRef] [PubMed] 12. Fisher-Wellman, K.H.; Neufer, P.D. Linking mitochondrial bioenergetics to insulin resistance via redox biology. Trends Endocrinol. Metab. 2012, 23, 142–153. [CrossRef][PubMed] 13. Myette-Cote, E.; Neudorf, H.; Rafiei, H.; Clarke, K.; Little, J.P. Prior ingestion of exogenous ketone monoester attenuates the glycaemic response to an oral glucose tolerance test in healthy young individuals. J. Physiol. 2018, 596, 1385–1395. [CrossRef][PubMed] 14. McClernon, F.J.; Yancy, W.S.; Eberstein, J.A.; Atkins, R.C.; Westman, E.C. The effects of a low-carbohydrate ketogenic diet and a low-fat diet on mood, hunger, and other self-reported symptoms. Obesity 2007, 15, 182–187. [CrossRef][PubMed] 15. Yancy, W.S., Jr.; Olsen, M.K.; Guyton, J.R.; Bakst, R.P.; Westman, E.C. A low-carbohydrate, ketogenic diet versus a low-fat diet to treat obesity and hyperlipidemia: A randomized, controlled trial. Ann. Intern. Med. 2004, 140, 769–777. [CrossRef][PubMed] 16. Yancy, W.S.; Foy, M.E.; Westman, E.C. A low-carbohydrate, ketogenic diet for type 2 diabetes mellitus. J. Gen. Intern. Med. 2004, 19, 110. 17. Summers, S.A. Ceramides in insulin resistance and lipotoxicity. Prog. Lipid Res. 2006, 45, 42–72. [CrossRef] [PubMed] 18. Park, M.; Kaddai, V.; Ching, J.; Fridianto, K.T.; Sieli, R.J.; Sugii, S.; Summers, S.A. A Role for Ceramides, but Not Sphingomyelins, as Antagonists of Insulin Signaling and Mitochondrial Metabolism in C2C12 Myotubes. J. Biol. Chem. 2016, 291, 23978–23988. [CrossRef][PubMed] 19. Chaurasia, B.; Summers, S.A. Ceramides—Lipotoxic Inducers of Metabolic Disorders. Trends Endocrinol. Metab. 2015, 26, 538–550. [CrossRef][PubMed] 20. Buchhalter, J.R.; D’Alfonso, S.; Connolly, M.; Fung, E.; Michoulas, A.; Sinasac, D.; Singer, R.; Smith, J.; Singh, N.; Rho, J.M. The relationship between d-beta-hydroxybutyrate blood concentrations and seizure control in children treated with the ketogenic diet for medically intractable epilepsy. Epilepsia Open 2017, 2, 317–321. [CrossRef][PubMed] 21. Barbanti, P.; Fofi, L.; Aurilia, C.; Egeo, G.; Caprio, M. Ketogenic diet in migraine: Rationale, findings and perspectives. Neurol. Sci. 2017, 38 (Suppl. 1), 111–115. [CrossRef][PubMed] 22. Ciavardelli, D.; Piras, F.; Consalvo, A.; Rossi, C.; Zucchelli, M.; Di Ilio, C.; Frazzini, V.; Caltagirone, C.; Spalletta, G.; Sensi, S.L. Medium-chain plasma acylcarnitines, ketone levels, cognition, and gray matter volumes in healthy elderly, mildly cognitively impaired, or Alzheimer’s disease subjects. Neurobiol. Aging 2016, 43, 1–12. [CrossRef][PubMed] 23. Murray, A.J.; Knight, N.S.; Cole, M.A.; Cochlin, L.E.; Carter, E.; Tchabanenko, K.; Pichulik, T.; Gulston, M.K.; Atherton, H.J.; Schroeder, M.A.; et al. Novel ketone diet enhances physical and cognitive performance. FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol. 2016, 30, 4021–4032. [CrossRef][PubMed] 24. Milder, J.; Patel, M. Modulation of oxidative stress and mitochondrial function by the ketogenic diet. Epilepsy Res. 2012, 100, 295–303. [CrossRef][PubMed] Int. J. Mol. Sci. 2018, 19, 2247 12 of 12

25. Ahola-Erkkila, S.; Carroll, C.J.; Peltola-Mjosund, K.; Tulkki, V.; Mattila, I.; Seppanen-Laakso, T.; Oresic, M.; Tyynismaa, H.; Suomalainen, A. Ketogenic diet slows down mitochondrial myopathy progression in mice. Hum. Mol. Genet. 2010, 19, 1974–1984. [CrossRef][PubMed] 26. Aubert, G.; Martin, O.J.; Horton, J.L.; Lai, L.; Vega, R.B.; Leone, T.C.; Koves, T.; Gardell, S.J.; Kruger, M.; Hoppel, C.L.; et al. The Failing Heart Relies on Ketone Bodies as a Fuel. Circulation 2016, 133, 698–705. [CrossRef][PubMed] 27. Volek, J.S.; Sharman, M.J.; Love, D.M.; Avery, N.G.; Gomez, A.L.; Scheett, T.P.; Kraemer, W.J. Body composition and hormonal responses to a carbohydrate-restricted diet. Metab. Clin. Exp. 2002, 51, 864–870. [CrossRef][PubMed] 28. Vandoorne, T.; De Smet, S.; Ramaekers, M.; Van Thienen, R.; De Bock, K.; Clarke, K.; Hespel, P. Intake of a Ketone Ester Drink during Recovery from Exercise Promotes mTORC1 Signaling but Not Glycogen Resynthesis in Human Muscle. Front. Physiol. 2017, 8, 310. [CrossRef][PubMed] 29. Thompson, J.R.; Wu, G. The effect of ketone bodies on nitrogen metabolism in skeletal muscle. Comp. Biochem. Physiol. B 1991, 100, 209–216. [CrossRef] 30. Zou, X.; Meng, J.; Li, L.; Han, W.; Li, C.; Zhong, R.; Miao, X.; Cai, J.; Zhang, Y.; Zhu, D. Acetoacetate Accelerates Muscle Regeneration and Ameliorates Muscular Dystrophy in Mice. J. Biol. Chem. 2016, 291, 2181–2195. [CrossRef][PubMed] 31. Guenther, G.G.; Peralta, E.R.; Rosales, K.R.; Wong, S.Y.; Siskind, L.J.; Edinger, A.L. Ceramide starves cells to death by downregulating nutrient transporter proteins. Proc. Natl. Acad. Sci. USA 2008, 105, 17402–17407. [CrossRef][PubMed] 32. Volek, J.S.; Freidenreich, D.J.; Saenz, C.; Kunces, L.J.; Creighton, B.C.; Bartley, J.M.; Davitt, P.M.; Munoz, C.X.; Anderson, J.M.; Maresh, C.M.; et al. Metabolic characteristics of keto-adapted ultra-endurance runners. Metab. Clin. Exp. 2016, 65, 100–110. [CrossRef][PubMed] 33. Kephart, W.C.; Mumford, P.W.; Mao, X.; Romero, M.A.; Hyatt, H.W.; Zhang, Y.; Mobley, C.B.; Quindry, J.C.; Young, K.C.; Beck, D.T.; et al. The 1-Week and 8-Month Effects of a Ketogenic Diet or Ketone Salt Supplementation on Multi-Organ Markers of Oxidative Stress and Mitochondrial Function in Rats. Nutrients 2017, 9, 1019. [CrossRef][PubMed] 34. Leduc-Gaudet, J.P.; Reynaud, O.; Chabot, F.; Mercier, J.; Andrich, D.E.; St-Pierre, D.H.; Gouspillou, G. The impact of a short-term high-fat diet on mitochondrial respiration, reactive oxygen species production, and dynamics in oxidative and glycolytic skeletal muscles of young rats. Physiol. Rep. 2018, 6, 4. [CrossRef] [PubMed] 35. Sampson, M.; Lathen, D.R.; Dallon, B.W.; Tessem, J.S.; Bikman, B.T. β-Hydroxybutyrate improves β-cell mitochondrial function and survival. J. Insul. Resist. 2017, 2, 1. 36. Dallon, B.W.; Parker, B.A.; Hodson, A.E.; Tippetts, T.S.; Harrison, M.E.; Appiah, M.M.A.; Witt, J.E.; Gibbs, J.L.; Gray, H.M.; Sant, T.M.; et al. Insulin selectively reduces mitochondrial uncoupling in brown adipose tissue in mice. Biochem. J. 2018, 475, 561–569. [CrossRef][PubMed] 37. Tippetts, T.S.; Winden, D.R.; Swensen, A.C.; Nelson, M.B.; Thatcher, M.O.; Saito, R.R.; Condie, T.B.; Simmons, K.J.; Judd, A.M.; Reynolds, P.R.; et al. Cigarette smoke increases cardiomyocyte ceramide accumulation and inhibits mitochondrial respiration. BMC Cardiovasc. Disord. 2014, 14, 165. [CrossRef] [PubMed] 38. Smith, M.; Tippetts, T.; Anderson, M.; Holub, Z.; Saito, R.; Swensen, A.; Prince, J.; Bikman, B.T. Insulin Increases Skeletal Muscle Ceramide Biosynthesis. Diabetes 2014, 63, A443. 39. Xia, J.Y.; Holland, W.L.; Kusminski, C.M.; Sun, K.; Sharma, A.X.; Pearson, M.J.; Sifuentes, A.J.; McDonald, J.G.; Gordillo, R.; Scherer, P.E. Targeted Induction of Ceramide Degradation Leads to Improved Systemic Metabolism and Reduced Hepatic Steatosis. Cell Metab. 2015, 22, 266–278. [CrossRef][PubMed] 40. Taylor, O.J.; Thatcher, M.O.; Carr, S.T.; Gibbs, J.L.; Trumbull, A.M.; Harrison, M.E.; Winden, D.R.; Pearson, M.J.; Tippetts, T.S.; Holland, W.L.; et al. High-Mobility Group Box 1 Disrupts Metabolic Function with Cigarette Smoke Exposure in a Ceramide-Dependent Manner. Int. J. Mol. Sci. 2017, 18, 1099. [CrossRef] [PubMed] 41. Tanner, C.B.; Madsen, S.R.; Hallowell, D.M.; Goring, D.M.; Moore, T.M.; Hardman, S.E.; Heninger, M.R.; Atwood, D.R.; Thomson, D.M. Mitochondrial and performance adaptations to exercise training in mice lacking skeletal muscle LKB1. Am. J. Physiol. Endocrinol. Metab. 2013, 305, E1018–E1029. [CrossRef] [PubMed]

© 2018 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).