bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

Suppression of Oxidation by Superfamily

Member 2 in Skeletal Muscle Promotes Hepatic Steatosis and

Resistance

Norihiro Imai1, Hayley T. Nicholls1, Michele Alves-Bezerra1, Yingxia Li1, Anna A.

Ivanova2, Eric A. Ortlund2, and David E. Cohen1

1Division of Gastroenterology and Hepatology, Joan & Sanford I. Weill Department of

Medicine, Weill Cornell Medical College, NY 10021 USA

2Department of Biochemistry, Emory University, Atlanta, GA 30322 USA

Current addresses:

Norihiro Imai - Department of Gastroenterology and Hepatology, Nagoya University

School of Medicine, Aichi 4668560 Japan

Michele Alves-Bezerra - Department of Molecular Physiology and Biophysics, Baylor

College of Medicine, Houston, TX 77030 USA

bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

Figure number: 8 Supplemental figure number: 10 Supplemental table number: 2 References: 48

Keywords: Hepatic steatosis, , acyl-CoA thioesterase, fatty acid oxidation, insulin

resistance

Conflict of interest: The authors have declared that no conflict of interest exists.

Author contributions: N.I.: designed research studies, conducted experiments,

acquired data, analyzed data and wrote manuscript. H.T.N.: conducted experiments and

analyzed data, M.A.B.: designed research studies and conducted experiments, Y.L.:

acquired data, A.A.I.: conducted experiments and analyzed data, E.A.O.: analyzed data,

D.E.C.: designed research studies, analyzed data and wrote manuscript.

Corresponding author:

David E. Cohen, M.D., Ph.D. 413 E 69th St, New York, NY 10021 Phone: 646-962-7680 Fax: 646-962-0427 Email: [email protected]

2 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

Financial Support:

Supported by the National Institute of Diabetes and Digestive and Kidney Diseases (R37

DK048873 and R01 DK056626 to D.E.C.); the Georgia Clinical & Translational Science

Alliance of the National Institutes of Health (UL1TR002378 to E.A.O.); American Heart

Association Postdoctoral Fellowship (19POST34380692 to N.I. and 18POST33990445

to M.A-B.); the American Liver Foundation Irwin M. Arias, MD Postdoctoral Research

Fellowship Award American (to N.I.); the Liver Foundation NASH Fatty Liver Disease

Postdoctoral Research Fellowship Award (to M.A.B.).

Acknowledgements:

The authors thank Dr. Scott Rodeo, Hospital for Special Surgery for assistance with

treadmill experiments. The Weill Cornell Medicine Metabolic Phenotyping Core and the

Emory Integrated Lipidomics Core are subsidized by Weill Cornell Medicine and the

Emory University School of Medicine respectively.

3 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

ABSTRACT

Thioesterase superfamily member 2 (Them2) is highly expressed in oxidative tissues

where it hydrolyzes long chain fatty acyl-CoA esters to free fatty acids and CoA. Although

mice globally lacking Them2 (Them2-/-) are protected against diet-induced obesity, insulin

resistance and hepatic steatosis, liver-specific Them2-/- mice remain susceptible. To

explore the contribution of Them2 in extrahepatic tissues, we created mice with Them2

deleted in skeletal muscle (S-Them2-/-), cardiac muscle (C-Them2-/-) or (A-

Them2-/-). When fed a high-fat diet, S-Them2-/- but not C-Them2-/- or A-Them2-/- mice

exhibited reduced weight gain. Only S-Them2-/- mice exhibited improved glucose

homeostasis together with improved insulin sensitivity in skeletal muscle. Increased rates

of fatty acid oxidation in skeletal muscle of S-Them2-/- mice were reflected in alterations

in skeletal muscle metabolites, including short chain fatty acids, branched chain amino

acids and the pentose phosphate pathway. Protection from diet-induced hepatic steatosis

in S-Them2-/- mice was attributable to increased VLDL triglyceride secretion rates in

support of demands of increased muscle fatty acid utilization. These results reveal a key

role for skeletal muscle Them2 in the pathogenesis of diet-induced obesity, insulin

resistance and hepatic steatosis.

4 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

INTRODUCTION

Rates of obesity have increased rapidly over the past 40 years and currently affect 600

million people worldwide1. Obesity increases the risk of developing and

non-alcoholic fatty liver disease (NAFLD). These common conditions contribute

significantly to rising morbidity and mortality, highlighting the unmet need for effective new

therapeutic targets in NAFLD.

Thioesterase superfamily member 2 (Them2; also known as acyl-CoA thioesterase (Acot)

13) catalyzes the hydrolysis of long chain fatty acyl-CoA esters into free fatty acids and

CoA2. Them2 is abundant in the liver and primarily localized to mitochondria, where it

regulates trafficking of fatty acids to oxidative versus lipid biosynthetic pathways,

depending upon metabolic conditions2. Them2 is also expressed at high levels in other

oxidative tissues, including skeletal and cardiac muscle, as well as in adipose tissue.

However, its function in these tissues is not understood3. Mice globally lacking Them2

(Them2-/-) are protected against diet-induced obesity and hepatic steatosis, and exhibit

markedly improved glucose homeostasis4,5. The global absence of Them2 is also

associated with reduced hepatic ER stress in the setting of overnutrition6. Conversely,

Them2 liver-specific knockout (L-Them2-/-) mice exhibit similar body weights as controls,

and are not protected against hepatic steatosis or insulin resistance in response to high-

5 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

fat diet (HFD) feeding7. Studies in L-Them2-/- mice revealed a mechanistic role for Them2

in the hepatic trafficking of acyl-CoA molecules towards triglyceride synthesis and

incorporation into VLDL particles7.

To explore the hypothesis that Them2 activity in extrahepatic tissues may be required for

the development of insulin resistance and hepatic steatosis, we generated tissue-specific

knockout mice lacking Them2 in skeletal muscle, cardiac muscle and adipose tissue. Only

skeletal muscle-specific Them2 knockout mice recapitulated the protection from insulin

resistance and hepatic steatosis that was observed in Them2-/- mice. Our results

demonstrate that Them2 limits rates of fatty acid oxidation in skeletal muscle, and its

upregulation in the setting of overnutrition promotes skeletal muscle insulin resistance

and hepatic steatosis by complementary mechanisms. In addition to reducing the demand

for hepatic VLDL secretion, the reduction in fatty acid oxidation in skeletal muscle

decreases the production of short chain fatty acids and pentose phosphate pathway

intermediates that maintain insulin sensitivity. In addition, Them2 suppresses the

expression of myokines capable of modulating skeletal muscle physiology, as well as

crosstalk with liver and adipose tissue. When taken together, these findings suggest that

Them2 in skeletal could be targeted as therapeutic strategy in the management of NAFLD.

6 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

RESULTS

Them2 suppresses insulin sensitivity in skeletal muscle and limits exercise

capacity in HFD-fed mice

Because skeletal muscle and heart extensively utilize fatty acids to produce ATP, we

examined the potential contribution of Them2 to the regulation of insulin action in these

tissues. We first confirmed that Them2-/- mice on a C57BL6J genetic background were

protected against HFD-induced obesity (Figure 1-A,B) and glucose intolerance (Figure 1-

C), as we have observed for a mixed genetic background4. In response to exogenous

insulin administration (Figure 1-D), HFD-fed Them2-/- mice exhibited a 62 % reduction in

blood glucose concentrations compared with a non-significant 24 % decrease in Them2+/+

mice, with an associated 2.6-fold increase in Akt phosphorylation in skeletal muscle,

which together indicated improved insulin sensitivity in the absence of Them2 expression.

There was a 1.7-fold increase in Them2 expression in skeletal muscle, but not in cardiac

muscle, in HFD-fed compared to chow-fed mice (Figure 1-E).

Notwithstanding reduced body weight of Them2-/- mice, both skeletal muscle and cardiac

tissues from Them2-/- mice were proportionate in size and histologically normal (Suppl.

Figure 1-A,B). Whereas skeletal muscle of Them2-/- mice exhibited no changes in mRNA

expression levels of myosin heavy chains, there were modest decreases in the

7 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

proportions of type I muscle fiber type relative to type II fiber type (Suppl. Figure

1-C,D). Them2-/- mice also exhibited similar expression of myosin heavy chains and

hypertrophy-related in cardiac muscle as control mice (Suppl. Figure 1-E,F). In

chow-fed mice, Them2 expression did not influence in exercise capacity, as assessed by

treadmill testing (Figure 1-F) and lactate production (Suppl. Figure 1-G). However, in the

setting of HFD feeding, Them2-/- mice showed improved exercise capacity compared to

wild type controls (Figure 1-F). We next investigated the impact of Them2 on cardiac

function in response to chronic ionotropic stimulation. Whereas cardiac hypertrophy is

accompanied by decreased rates of fatty acid oxidation and increased glucose utilization8,

Them2 expression did not influence development of hypertrophy (Figure 1-G).

Metabolic monitoring revealed no differences in voluntary (wheel) running (Figure 1-H),

spontaneous physical activity or food intake (data not shown). Similar to Them2-/- mice on

a mixed genetic background4, chow-fed Them2-/- C57BL6J mice exhibited increased

energy expenditure (Figure 1-I). Although no differences were found in energy

expenditure during voluntary running (Figure 1-I), respiratory exchange ratios of Them2-

/- mice were elevated during the light cycle after acclimation to running wheel (Figure 1-

J), indicative of greater glucose utilization in skeletal muscle in the absence of Them2

8 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

expression during a period when mice were more sedentary. Taken together, these results

suggest that Them2 regulates energy substrate utilization in skeletal muscle.

Skeletal muscle expression of Them2 promotes obesity and insulin resistance in

HFD-fed mice

To systematically explore contributions of extrahepatic Them2 to obesity, hepatic

steatosis and insulin resistance, we created mice lacking Them2 in skeletal muscle,

cardiac muscle and adipose tissues, each on a C57BL/6 genetic background (Figure 2-

A). Skeletal muscle deletion of Them2 with preserved cardiac expression (S-Them2-/-

mice) was accomplished using mice expressing Cre recombinase driven by the Myf5

promoter. Because Myf5 is expressed in brown adipose tissue (BAT) in addition to skeletal

muscle, mice lacking Them2 in skeletal muscle were also deficient in BAT. The Myh6

promoter was used to generate cardiac muscle deletion of Them2 with preserved skeletal

muscle expression (C-Them2-/- mice). A-Them2-/- mice lacking Them2 in BAT and white

adipose tissue (WAT), which expresses Them2 at low levels, were generated using mice

expressing Cre recombinase driven by the adiponectin promoter. Only S-Them2-/- mice

were protected against diet-induced overweight (Figure 2-B), excess fat accumulation

(Figure 2-C), glucose intolerance (Figure 2-D), and insulin resistance (Figure 2-E), as

9 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

observed for global Them2-/- mice (Figure 1-A-D). These differences were not associated

with substantial alterations in expression of other skeletal muscle Acot or Acsl isoforms

(Suppl. Figure 2-A,B). The possibility that this phenotype was attributable to the absence

of Them2 expression in BAT was excluded by demonstrating similar weight differences

(Suppl. Figure 3-A) and body compositions (Suppl. Figure 3-B) for mice housed at 30 °C,

a thermoneutral temperature that suppresses BAT activity. This was further supported by

the phenotypes of A-Them2-/- mice (Figure 2-B-D), which also lack Them2 expression in

BAT, but were not protected against HFD-induced obesity and insulin resistance. We used

a gain of function approach to confirm the metabolic contribution of skeletal muscle

Them2: AAV8-Mck driven rescue of Them2 expression in muscle of global Them2-/- mice

(Suppl. Figure 3-C) resulted in higher body weights than AAV8- Mck-LacZ treated Them2-

/- mice (Suppl. Figure 3-D) with increases in both lean and fat mass (Suppl. Figure 3-E).

Taken together, these results indicate that skeletal muscle Them2 plays a central role in

the development of obesity and insulin resistance in response to HFD feeding.

To investigate mechanisms of improved glucose homeostasis in HFD-fed S-Them2-/- mice,

we performed hyperinsulinemic-euglycemic clamp studies. S-Them2-/- mice exhibited

higher glucose infusion rates than controls (Figure 3-A), as well as increased rates of

glucose uptake in skeletal muscle (Figure 3-B). By contrast, basal and clamped hepatic

10 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

glucose production rates were not influenced by skeletal muscle expression of Them2

(Figure 3-C). In keeping with these findings, there were marked increases in Akt

phosphorylation by 2.5-fold in skeletal muscle of HFD-fed S-Them2-/- mice compared to

controls in response to acute administration of insulin (Figure 3-D). These results reveal

that Them2 in skeletal muscle promotes whole body glucose intolerance by reducing

skeletal muscle insulin sensitivity.

Them2 controls rates of fatty acid oxidation in skeletal muscle

We next examined the role of Them2 in the regulation of fatty acid oxidation in skeletal

muscle. Rates of fatty acid oxidation in skeletal muscle homogenates were increased in

both Them2-/- (Figure 4-A) and S-Them2-/- mice (Figure 4-B). Muscle strips isolated from

Them2-/- mice exhibited increased oxidation rates of palmitate from endogenous (pulsed)

palmitate but not from exogenous (chase) sources (Figure 4-C).

To gain mechanistic insights, we performed immunoblot analyses of gastrocnemius

muscle samples to assess the expression of key proteins that control fatty acid oxidation.

In contrast to chow-fed control mice, HFD-fed controls exhibited 2-fold increases in

Them2 expression levels (Figure 4-D,E). Suggestive of its role in regulating Them2

expression in skeletal muscle, the Them2-interacting PC-TP3, was reduced in

11 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

chow-fed S-Them2-/- mice relative to controls (Figure 4-D,E). Although phosphorylation of

adenosine monophosphate-activated protein kinase (AMPK (Thr172)), which increases

fatty acid oxidation rates in skeletal muscle, showed no genotype-dependent changes,

chow-fed S-Them2-/- mice exhibited reduced phosphorylation of acetyl-CoA carboxylase

(ACC) compared to control mice (Figure 4-D,E). Phosphorylated ACC is inactive, whereas

dephosphorylated ACC enhances conversion of acetyl-CoA to malonyl-CoA, and this in

turn reduces fatty acid oxidation owing to inhibition of carnitine palmitoyltransferase I

(CPT1)9. Although CPT1 protein expression was not altered in chow-fed mice, relative

mRNA expression of CPT1 was reduced in skeletal muscle of chow-fed S-Them2-/- mice

(Suppl. Figure 4-A). Notwithstanding the absence of changes in ACC phosphorylation in

HFD-fed mice, increased CPT1 protein expression in HFD-fed S-Them2-/- mice suggests

increased mitochondrial fatty acid utilization in skeletal muscle of S-Them2-/- mice.

Consistent with a lack of effect of Them2 expression on fatty acid oxidation rates of

exogenous fatty acids, we observed no changes in skeletal muscle expression of CD36.

To further ascertain mechanisms underlying the increased skeletal muscle fatty acid

oxidation, we surveyed the expression of genes that control lipid and glucose

homeostasis. The expression of these genes was largely unchanged either in chow

(Suppl. Figure 4-A,B) or HFD-fed mice (Suppl. Figure 4-A,B), suggesting that increased

12 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

fatty acid oxidation in S-Them2-/- mice was not compensated by fatty acid uptake or

glucose utilization.

In keeping with increased rates of fatty acid oxidation, total fatty acyl-CoAs in skeletal

muscle of chow-fed S-Them2-/- mice tended to decrease (Figure 4-F), but without

changes in individual molecular species (Suppl. Figure 5A). The absence of a change in

fatty acyl-CoAs following HFD-feeding is likely attributable to abundant cellular nutrients

in the setting of overnutrition. Acetyl-carnitine forms when acetyl-CoA concentrations

exceed what is required by the TCA cycle, and is essential molecules for maintaining

metabolic flexibility and insulin sensitivity10. Skeletal muscle of S-Them2-/- mice exhibited

increased acetyl-carnitine concentrations for mice fed HFD (Figure 4-G), without

discernible changes in individual acyl-carnitine molecular species (Suppl. Figure 5-B).

These results are consistent with increased fatty acid oxidation rates in skeletal muscle

lacking Them2 even after HFD feeding. Free fatty acid concentrations were higher in

skeletal muscle of S-Them2-/- mice (Figure 4-H), with increases being most pronounced

in short chain molecular species fatty acids (Figure 4-I, Suppl. Figure 5-C,D), which can

influence glucose homeostasis11.

To gain further insight into Them2 metabolic regulation in skeletal muscle, we conducted

targeted metabolomics profiling for 216 predefined polar metabolites. Unsupervised

13 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

clustering of differences identified both diets and genotypes (Figure 4-J). Metabolic

pathway analysis identified significant alterations in S-Them2-/- mice fed either chow

(Suppl. Figure 6A) or HFD (Suppl. Figure 6B). Of these, and the

pentose phosphate pathway (PPP) were affected by skeletal muscle Them2 expression

in mice fed both diets.

The absence of Them2 in skeletal muscle increased tissue concentrations of 9 amino

acids in chow-fed mice (Suppl. Figure 7-A), whereas it decreased concentrations of 4

amino acids in HFD-fed mice (Suppl. Figure 7-B). For both diets, ablation of Them2 mainly

altered branched chain amino acids. Branched-chain amino acids promote protein and

glutamine synthesis in skeletal muscle, and increased levels of branched-chain amino

acids is known to correlate with an improvement in glucose metabolism12. Therefore,

increased levels of branched-chain amino acids in muscle of chow-fed S-Them2-/- mice

may have contributed to protection against diet-induced insulin resistance.

Whereas most metabolites in the PPP pathway were unchanged in both chow diet and in

HFD (Figure 4-K), ribose 5-phosphate was significantly reduced in chow-fed mice and

both ribulose 5-phosphate and ribose 5-phosphate were increased in skeletal muscle of

HFD-fed S-Them2-/- mice, suggestive of upregulation of this pathway by overnutrition in

the absence of Them2. PPP generates reducing equivalents to prevent oxidative stress13,

14 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

and its upregulation may have contributed to improved insulin sensitivity by clearing

excess glucose-induced reactive oxygen species in skeletal muscle. Them2 expression

in skeletal muscle did not influence activity of superoxide dismutase (Suppl. Figure 8-A)

or thiobarbituric acid reactive substances (Suppl. Figure 8-B), suggesting that the ablation

of Them2 did not promote oxidative stress in skeletal muscle. Taken together, these

studies revealed that Them2 in skeletal muscle modulates tissue concentrations of short

chain fatty acids, branched chain amino acids and PPP metabolites, each of which may

play a role in the regulation of glucose homeostasis.

Them2 limits fatty acid utilization in cultured myotubes

To assess whether Them2 alters mitochondrial fatty acid utilization in skeletal muscle, we

utilized C2C12 myotubes that were transduced to overexpress mouse Them2. AAV8-

Them2 transduction increased Them2 expression by 1.5-fold (Figure 5-A). Although

increased Them2 expression did not alter myotube diameter or fusion index14, AAV8-

Them2 treated myotubes exhibited greater myotube areas, suggesting Them2 expression

enhances myotube elongation (Figure 5-B,C). Myotubes that overexpressed Them2 also

exhibited higher rates of oxygen consumption (OCR) during both basal and maximal

respiration (Figure 5-D). Of note, when palmitate was made available, there were no

15 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

differences in basal or maximal respiration (Figure 5-D), with differences between bovine

serum albumin (BSA) treatment alone and palmitate plus BSA exposure signifying

oxidation of extracellular palmitate. As evidenced by higher OCR values following

treatment with BSA alone, AAV8-Them2 transduced myotubes utilized less extracellular

(Figure 5-E).

A reduction in OCR after etomoxir treatment under substrate limited conditions provides

a measure of intracellular fatty acid oxidation (Figure 5-F), which was not influenced by

Them2 expression (Figure 5-G). Taken together, these results suggest that Them2

regulates extracellular fatty acid utilization through its regulation of the intracellular fatty

acid pool that is available for oxidation.

Skeletal muscle Them2 expression enables HFD-fed mice to exceed a body weight

threshold value for hepatic steatosis and metabolic dysfunction

In beings, hepatic steatosis is largely a function of adipose tissue insulin

resistance and serves an important barometer of metabolic dysfunction, which becomes

fully established at a threshold level of 5.5% liver fat15. Although HFD-fed mice are widely

utilized in studies of NAFLD, a threshold value for pathogenic intrahepatic triglyceride

accumulation and its relationship to body composition has not been systematically

16 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

explored. To enable accurate weight-based comparisons between S-Them2-/- and control

mice, 87 C57BL6J male mice were fed a HFD for 12 w. Body weights exhibited a normal

distribution ranging from 31.1 g to 52.2 g (mean ± SEM; 42.3 ± 0.5 g; median 42.5g)

(Figure 6-A). Both lean and fat mass increased linearly as functions of body weight (Figure

6-B), and fat mass was a linear function of lean mass (Figure 6-C). By contrast, liver

weight exhibited two very distinct linear relationships to body weight (Figure 6-D): Up to

a body weight threshold of 41.6 g (corresponding to fat mass of 15.6 g), there was a

relatively flat slope of liver weight versus body weight (0.031 g liver/g BW). In excess of

41.6 g, the slope was 4.3-fold steeper (0.134 g liver/g BW). Additionally, there was a

similar biphasic dependence of liver weight on fat (Figure 6-E) but not lean weight of mice

(Figure 6-F).

Hepatic steatosis exhibited a biphasic relationship to body weight, with threshold body

weight of 41.6 g corresponded to 31% hepatic steatosis (Figure 6-G). By contrast, both

liver weights (Figure 6-H) and hepatic triglyceride concentrations (Figure 6-I) were linearly

correlated with histologic steatosis. The body weight threshold of 41.6 g also

discriminated plasma concentrations of free fatty acids, glucose and cholesterol (Figure

6-J), as well as hepatic mRNA expression levels of specific genes that govern the

metabolism of fatty acids (Aox and CD36) and lipid droplets (Cidea) (Figure 6-K).

17 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

Indicative that the 41.6 g weight threshold for hepatic steatosis reflected changes in

skeletal muscle dysfunction, expression of genes that mediate fatty acid activation (Acsls)

and deactivation (Acots) were significantly altered in skeletal muscle of mice exceeding

this body weight (Figure 6-L). Additionally, five myokine genes were downregulated above

this weight threshold, as was Pgc1a, which controls mitochondrial biogenesis in skeletal

muscle (Figure 6-M). Taken together, these findings reveal two distinct phases in the

development of hepatic steatosis and metabolic dysfunction in HFD-fed C57BL/6J mice

with a threshold of 31% at a body weight of 41.6 g. After 12 w of HFD-feeding, body

weights of most (80%) of S-Them2-/- mice fell below the 41.6 g threshold, compared with

36 % of controls (Figure 6-N).

In keeping with reduced weight and improved glucose homeostasis, HFD-fed S-Them2-/-

but not L-Them2-/-7 or C-Them2-/- mice were also protected against hepatic steatosis, as

evidence by histopathology (Figure 7-A) and hepatic triglyceride concentrations (Figure

7-B), as was previously observed in Them2-/- mice4. Pyruvate tolerance tests revealed

reduced hepatic glucose production only in S-Them2-/- mice (Figure 7-C), as was also

observed in global Them2-/- mice4. There were no differences in plasma activities of

aspartate aminotransferase, alanine aminotransferase or creatine kinase or in insulin

concentrations (Suppl. Figure 9-A,B). Of note, for mice under the 41.6 g body weight

18 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

threshold, hepatic triglyceride concentrations were also reduced in HFD-fed S-Them2-/-

mice compared to controls (Figure 7-D). These findings suggested that metabolic

improvements in S-Them2-/- mice were in part attributable to reduced body weight gain

because most mice lacking Them2 did not achieve a body weight threshold for the

development of more severe hepatic steatosis, but importantly, were also due in part to

expression of skeletal muscle Them2 per se.

Skeletal muscle Them2 expression promotes hepatic steatosis by suppressing

VLDL secretion

We next explored the mechanisms underlying skeletal muscle Them2-mediated

development of hepatic steatosis. We first examined evidence for increased rates of

hepatic fatty acid oxidation. Fasting concentrations of ß-hydroxybutyrate were reduced in

HFD-fed control mice compared with chow-fed controls, but this was not observed for S-

Them2-/- mice, in which levels remained unchanged by HFD-feeding and were

significantly higher than in HFD-fed control mice (Suppl. Figure 9-C). This suggested that

the absence of Them2 in skeletal muscle might be associated with increase rates of fatty

acid oxidation in liver. However, direct measurement of hepatic fatty acid oxidation in

HFD-fed S-Them2-/- mice did not reveal increases (Figure 7-E), suggesting that

19 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

differences in ß-hydroxybutyrate may instead have reflected Them2-mediated changes

in rates of ketone body utilization in skeletal muscle. Chow but not HFD-fed S-Them2-/-

mice exhibited increased fasting plasma concentrations of free fatty acids (Suppl. Figure

9-D), which argues against reduced hepatic uptake of fatty acids as a mechanism by

which S-Them2-/- mice were protected against steatosis. Plasma concentrations of

triglycerides, total and free cholesterol, as well as phospholipids were unaffected by

skeletal muscle Them2 expression in either chow or HFD-fed mice (Suppl. Figure 9-D),

and despite dramatic reductions in hepatic steatosis, RNAseq analysis of liver identified

only a limited number of genes with altered expression (Suppl. Figure 10-A). Among these

was the hepatic steatosis marker Cidea, which was markedly downregulated, suggesting

reductions in triglyceride storage. This was also implicated by pathway analysis, which

identified increased degradation of hepatic triglycerides as the most upregulated pathway

(Suppl. Figure 10-B). In keeping with this possibility, S-Them2-/- mice exhibited increased

rates of VLDL triglyceride secretion (Figure 7-F). Taken together, these findings suggest

that increased demand by skeletal muscle for fatty acids in the form of secreted hepatic

triglycerides may have explained the reduced steady state concentrations in liver.

20 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

Muscle Them2 mediates crosstalk between skeletal muscle and the liver

Lastly, we examined evidence of regulatory cross-talk between muscle and liver that may

have also contributed to protection against hepatic steatosis and metabolic dysfunction

in S-Them2-/- mice. Conditioned medium from C2C12 myotubes transduced to

overexpress Them2, suppressed basal and maximal OCR values in Hepa1-6 cells (Figure

8-A). We next assessed for Them2-dependent expression of myokines in skeletal muscle.

Ablation of Them2 altered myokine expression depending on dietary conditions

(Figure 8-B). Overnight fasting induced substantial changes in myokine genes expression

in S-Them2-/- mice (Figure 8-B). Skeletal muscle of chow-fed S-Them2-/- mice induced

expression of insulin like growth factor 1 (Igf-1) and interleukin 15 (IL-15), both of which

regulate muscle mass16,17, whereas IL-15 also decreases plasma triglycerides and

adipose tissue mass18. In HFD-fed mice, S-Them2-/- mice exhibited modest increases in

chemokine ligand 1 (CXCL1), which regulates angiogenesis and fatty acid metabolism19.

Independent of diet, there was consistent down-regulation in osteoclin which is

upregulated by palmitic acid and promotes insulin resistance in muscle20–22. Because

skeletal muscle of HFD-fed S-Them2-/- mice is protected against excess body weight gain,

these alterations in myokine genes might have been secondary to changes in body weight.

However, differences in relative mRNA expression were still observed in weight matched

21 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

HFD-fed mice, as defined by body weights that fell below the threshold of 41.6 g (Figure

8-C): myostatin and osteoclin were downregulated in skeletal muscle of S-Them2-/- mice

compared to control mice. These results suggested that Them2-mediated control of

skeletal muscle myokine may contribute to crosstalk among skeletal

muscle, adipose tissue and the liver (Figure 8-D).

22 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

DISCUSSION

This study identified a key role for skeletal muscle Them2 in the regulation of skeletal

muscle metabolism, as well as in the pathogenesis of HFD-induced insulin resistance and

hepatic steatosis in mice. Them2 limits rates of fatty acid oxidation by skeletal muscle,

which in turn reduces rates of insulin-mediated glucose disposal by reducing insulin

sensitizing metabolites and myokines. This also reduced demand for exogenous fatty

acids, leading to the development of hepatic steatosis.

We previously demonstrated that Them2-/- mice exhibited reduced plasma free fatty acid

concentrations, as well as reduced hepatic steatosis and glucose production5,23. However,

these effects were not recapitulated in L-Them2-/- mice, suggesting that extrahepatic

Them2 expression is required in order to recapitulate the phenotype in the setting of

overnutrition7. In both mice and , skeletal and cardiac muscles substantially

contribute to basal calorie consumption. Skeletal plays critical roles in energy metabolism,

glucose uptake and locomotion24. The metabolic activity of the heart is a major

determinant of the body’s oxygen consumption, predominantly utilizing long chain fatty

acids as substrates for energy production8. Consequently, glucose and

in skeletal muscle and cardiac muscle contribute substantially to whole body energy

consumption.

23 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

Whereas Them2 is most highly expressed in heart, its deletion from cardiac muscle did

result in discernable differences in organ function or whole body metabolism. To preserve

function, cardiac muscle has extraordinary metabolic flexibility that enable it to adapt to

changes in substrate availability8. For this reason, we speculate that alterations in this

specific of long chain would not cause obvious functional

or metabolic consequences in C-Them2-/- mice. This notion is supported by our

observation that Them2 expression in heart of control mice was unaffected by HFD

feeding. By contrast, HFD feeding robustly upregulated Them2 expression in skeletal

muscle, suggesting a pathogenic role in skeletal muscle mechanical dysfunction and

alteration in metabolism.

As evidenced by reduced rates of glucose uptake during clamp studies and diminished

insulin-stimulated Akt phosphorylation, Them2 contributed to the development of skeletal

muscle insulin resistance, presumably as a reflection of reduced rates of fatty acid

oxidation25. Previous studies have demonstrated that the accumulation of excess free

fatty acids and their derivatives including long chain acyl-CoAs in skeletal muscle are key

determinants of insulin resistance26. Although the absence of a fatty acyl-CoA

thioesterase might have been expected to increase skeletal muscle concentrations fatty

acyl-CoAs and decrease concentrations of free fatty acids, we observed the opposite in

24 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

S-Them2-/- mice. However, the preferred substrate of Them2 is long chain fatty acyl-CoA,

and the observed increase in free fatty acid concentrations was mainly attributable to

increases in short chain fatty acids. Short chain fatty acids are predominantly generated

by colonic bacteria and are metabolized by enterocytes and hepatocytes. Short chain

fatty acids are also formed in peroxisomes during shortening of very long chain fatty acyl-

CoAs that can be hydrolyzed by peroxisomal Acots and released into the cytosol27. We

speculate, increased rates of mitochondrial ß-oxidation in skeletal muscle of S-Them2-/-

mice were also accompanied by increases in peroxisomal oxidation, which resulted in the

production of more short chain fatty acids. The reduction in skeletal muscle fatty acyl-CoA

concentrations most likely reflects their increased rates of oxidation, as evidence by the

reciprocal rise in acetyl-carnitine concentrations.

Short chain fatty acids influence muscle fatty acid oxidation, glucose uptake and insulin

sensitivity via both direct and indirect mechanisms11,28. Of note, acetate and butyrate

improve skeletal muscle glucose metabolism and insulin sensitivity11. When taken

together with observed increases in branched chain amino acids and PPP metabolites12,13,

these data suggest that elevated concentrations of short chain fatty acids in skeletal

muscle of S-Them2-/- mice contributed to the improved insulin sensitivity observed the

setting of overnutrition.

25 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

Although the reduction in lean mass of S-Them2-/- mice at the thermoneutrality was similar

as at room temperature, there was less reduction in fat mass. This may have been

because Them2 in BAT also plays a role in HFD-induced obesity via adaptive

thermogenesis5. Because fat mass is tightly correlated with lean mass in HFD-fed mice,

we further postulate that reduction in lean body mass due to loss of skeletal muscle

Them2 also exerted metabolic control through associated decreases in adipose tissue.

In human NAFLD, hepatic steatosis is strongly influenced by relatively modest changes

in total body weight: An 11% loss in body weight was associated with a 52 % reduction in

hepatic triglycerides, owing largely to the restoration of insulin sensitivity29. Interestingly,

these observations in humans are of a similar magnitude to our findings in HFD-fed mice,

whereby at 16 w of age, S-Them2-/- mice exhibited a 12 % reduction in BW (Figure 2-B),

which was associated with a 52 % reduction in hepatic triglyceride concentrations.

However, unlike observations in humans15, C57BL6J mice exhibited remarkable

accelerations in hepatic triglyceride accumulation at a discrete threshold of body weight

(41.6 g). Moreover, fat weight is tightly correlated with lean weight in HFD-fed mice. Hence,

we concluded that reduced hepatic steatosis in S-Them2-/- mice is attributable in part to

the reduced body weight gain such that mice rarely exceeded body weights of 41.6 g.

Nevertheless, hepatic triglyceride concentrations were still reduced in HFD-fed S-Them2-

26 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

/- mice compared to HFD-fed control mice weighing less than 41.6 g revealing that Them2

expression per se is sufficient to regulate hepatic lipid metabolism.

A potential mechanism whereby skeletal muscle Them2 directly controls hepatic steatosis

is organ crosstalk through the secretion of myokines, which are bioactive proteins that

regulate organ function by autocrine, paracrine and endocrine actions30. Deletion of

Them2 in skeletal muscle induced expression of several myokines. Skeletal muscle of

chow-fed S-Them2-/- mice showed significantly higher gene expressions of Igf-1 and IL-

15, which regulate muscle hypertrophy16,17. IL-15 also decreases lipid deposition in pre-

adipocytes and reduces white adipose tissue mass18. In the setting of overnutrition, there

was also a modest increase in skeletal muscle expression of CXCL1 in S-Them2-/- mice.

CXCL1 is the functional homologue to human IL-831, and it plays a role in angiogenesis

and neutrophil chemoattractant activity32. Increased expression of CXCL1 attenuates diet

induced obesity in mice through enhanced fatty acid oxidation in muscle19. Osteoclin

(synonym: musclin), which was downregulated in skeletal muscle of S-Them2-/- mice, is

a myokine that is controlled by nutritional status and promotes insulin resistance. Musclin

expression inhibits glucose uptake and glycogen synthesis20, and Musclin mRNA is

regulated by the forkhead box O1 transcription factor (FOXO1) downstream of PI3K/Akt

pathway in response to insulin action22.

27 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

Our findings demonstrate a central role for skeletal muscle Them2 in the control of nutrient

metabolism and in the pathogenesis of diet-induced insulin resistance and hepatic

steatosis. The inhibition of Them2 activity in muscle could offer an attractive strategy for

the management of obesity and nonalcoholic fatty liver disease.

28 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

MATERIALS AND METHODS

Animals and diets

Mixed background global Them2 knockout mice were backcrossed 20 generations with

C56BL/6J4,5. Wild type C56BL/6J mice were obtained from the Jackson Laboratory, Bar

Harbor, ME, USA (Stock #000664). Tissue specific knockout mice were created by a

LoxP/Cre system7. C56BL6 background mice with Them2 flanked by two LoxP sites

(Them2flox/flox) were crossed with mice expressing Cre recombinase driven by the Myf5

promoter to generate skeletal muscle specific deletion of Them2-/- (S-Them2-/-), the Myh6

promoter to generate cardiac muscle specific deletion of Them2-/- (C-Them2-/-) and the

Adipoq promoter was used to generate adipose tissue specific deletion of Them2-/- (A-

Them2-/-) respectively (The Jackson Laboratory, Myf5-cre; Stock #007893, Myh6-cre;

Stock #011038, Adipoq-cre; Stock #010803)33–35. These mice were viable and displayed

no apparent developmental abnormalities. The presence of Cre allele was determined by

PCR analysis using the primers specified by the Jackson Laboratory. Mice were housed

in a barrier facility on a 12 h light/dark cycle with free access to water and diet. Male mice

were weaned at 4 w of age and fed chow (PicoLab Rodent Diet 20; LabDiet, St. Louis,

MO, USA). Alternatively, 5 w old male mice were fed a HFD (D12492: protein 20 % kcal,

fat 60 % kcal, carbohydrate 20 % kcal, energy density 5.21 kcal/g; Research Diets Inc.,

29 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

New Brunswick, NJ, USA) for 12 w. Following 6 h fasting (9:00 AM to 3:00 PM) or 18 h

fasting (6:00 PM to 12:00 PM), 17 w old mice were euthanized and plasma was collected

by cardiac puncture. Tissues were harvested for immediate use or snap frozen in liquid

nitrogen and stored at -80 °C. Mice were monitored by daily health status observation by

technicians supported by veterinary care. Housing and husbandry were conducted in

facilities with a sentinel colony health monitoring program and strict biosecurity measures

to prevent, detect, and eradicate adventitious infections. Animal use and euthanasia

protocols were approved by the Institutional Animal Care and Use Committee of Weill

Cornell Medical College.

Adeno-associated -mediated Them2 expression in skeletal muscle

Adeno-associated virus (AAV) vectors used in this study were generated by Vigene

Biosciences, Rockville, Maryland, USA. Vectors are based on the AAV8 genome and

transgenes are driven by the muscle creatine kinase (Mck) promoter. The mouse Them2

gene (gene ID: 55856) was packaged with Flag as AAV8- Mck-Flag-Them2 and a LacZ

gene was used to generate AAV8-Mck-LacZ as a control. Mice were anesthetized with

2% isoflurane vapors prior to injection to ensure no movement. Both hindlimbs were

shaved and sterilized with isopropanol swab sticks. AAVs (1-5x1011 particles in 30μl

30 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

saline) were injected to the gastrocnemius muscle by intramuscular injection using 30-

gauge insulin syringe.36 Mice were then return to home cages for 12 w to allow for

recombinant protein expression.

Maximal exercise capacity

Mice were acclimated to treadmill running tracks incrementally for 5 d starting from 10

min at 0.3 km/h to 60 min at 1.2 km/h. To determine maximal exercise capacity, treadmill

speed was started at 0.3 km/h and increased 0.3 km/h every 3 min until the speed

reached 1.2 km/h. The speed was kept constant (1.2 km/h) until mice reached exhaustion.

Blood lactate levels were measured from the tail tip before and after 30 min treadmill

running at 1.2 km/h using lactate pro2 LT-1730 (ARKRAY, Kyoto, Japan).

Cardiac function

Cardiac function was evaluated by echocardiography as response to 2 w chronic

isoproterenol infusion as previously described37–39. In brief, 10 w old mice were implanted

with ALZET osmotic minipumps Model 1002 (DURECT Corporation, Cupertino, CA, USA)

and infused with isoproterenol at 30 mg/kg/day for 2 w. Echocardiography was carried out

at 10 w (pre) and 12 w (post) to monitor cardiac hemodynamics using a Vevo 3100

31 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

(FUJIFILM Visual sonics, Toronto, Canada). M mode was used to measure left ventricle

ejection fraction and wall thickness. Each mouse was anesthetized with inhaled isoflurane

(2%).

Metabolic monitoring

Mice were housed in individual cages for 2 w for acclimation prior to metabolic

monitoring40. Mice were then housed in temperature-controlled cabinets with a 12 h

light/dark cycle and monitored using the Promethion Metabolic Screening System (Sable

Systems International, North Las Vegas, NV). Rates of O2 consumption (VO2) and CO2

production (VCO2) were determined at 5 min intervals. Values of respiratory exchange

ratio (RER) were calculated as VCO2/VO2. After 2 d acclimation, metabolic parameters

were recorded over 24 h with or without a running wheel inside the cage. Physical

activities and voluntary running on wheel were determined according to beam breaks

within a grid of photosensors built outside the cages. Energy expenditure was calculated

by indirect calorimetry41 and adjusted by ANCOVA using VassarStats

(www.vassarstats.net) to adjust for differences in lean body mass using lean body

composition determined by magnetic resonance imaging EchoMRI (EchoMRI, Houston,

TX).

32 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

Tolerance Tests

Glucose tolerance tests, insulin tolerance tests, and pyruvate tolerance tests were

performed with minor modifications7,42. In brief, mice were fasted 6 h for glucose tolerance

tests, 4 h for insulin tolerance tests, and overnight (16 h) for pyruvate tolerance tests. A

drop of blood from the tail tip was subjected to glucose measurement at baseline and at

regular intervals using GE 100 Blood Glucose Monitor (General Electric, Ontario, CA).

Glucose solution was administered by intraperitoneal injection with 1 g/kg for global

Them2-/- mice and their controls and with 1.5 g/kg for A-Them2-/- mice and their littermate

controls, or orally using gavage needle with fixed dose at 40 mg for C-Them2-/- mice, S-

Them2-/- mice and their littermate controls. Insulin solution was administered by

intraperitoneal injection with 0.75 U/kg for A-Them2-/- mice, C-Them2-/- mice, S-Them2-/-

mice and littermate controls. Pyruvate solution was administered by orally using a gavage

needle with 2 g/kg for C-Them2-/- mice, S-Them2-/- mice and littermate controls.

Hyperinsulinemic-euglycemic clamp studies

Hyperinsulinemic-euglycemic clamp studies were performed at Animal Physiology Core

(Albert Einstein College of Medicine, New York, NY, USA) as described privously43. Briefly,

33 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

vascular catheters were inserted, 8 d prior to the clamps. Hyperglycemic-euglycemic

clamp studies were performed in unrestrained mice. Mice were fasted for 5 h prior to the

study. The clamp protocol consisted of a 90 min tracer (5-uCi bolus of [3-3H]-glucose

followed by a 0.05 uCi/min infusion) equilibration period followed by a 120 min

experimental period. A blood sample (10 ul) was taken for the assessment of basal

glucose and insulin levels and glucose turnover. The insulin clamp was begun with a

primed-continuous infusion of human insulin (300 mU/kg bolus followed by 5 mU/kg/min

1; Novolin; Novo Nordisk, Princeton, NJ). The [3-3H]-glucose infusion was increased to

0.1 uCi/min for the remainder of the experiment. Euglycemia (150 mg/dl) was maintained

during clamps by measuring blood glucose every 10 min and infusing 45% dextrose as

necessary. Blood samples (60–200 µl) were taken and processed to determine glucose

specific activity. All blood samples were resuspended in heparinized saline and infused

back to mice to maintain hematocrit. Plasma samples were collected to determine

glucose levels and specific activities of [3-3H]-glucose. Tissue samples were stored at

−80°C for glucose uptake analyses43.

Insulin signaling

Mice were fasted for 6 h during the light cycle (9:00 AM to 3:00 PM). Tissues were

34 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

collected 10 min following intraperitoneal insulin (10 U/kg) injection or saline injection as

a control. Insulin signaling was evaluated by immunoblot analysis for phosphorylation of

Akt44.

Quantitative PCR

Relative mRNA expression was determined by quantitative PCR (qPCR) using SYBR

Green Real-Time PCR Master Mix (Applied Biosystems, Foster City, CA, USA). Total RNA

was isolated from soleus muscle (10 µg) and apex of ventricle (20 µg) using RNeasy

fibrous tissue mini kit (QIAGEN, Hilden, Germany) following manufacturer’s instructions.

cDNA was synthesized with a High-Capacity cDNA Reverse transcription Kit (Applied

Biosystems, Foster City, CA, USA). Equal amount of mRNA samples were subjected to

qPCR using QuantStudio 6 Flex Real-Time PCR System (Applied Biosystems, Foster

City, CA, USA) in a 384 plate. Genes were analyzed relative to cyclophilin A. The

nucleotide sequences of oligonucleotides used for qPCR are presented in Suppl. Table 2.

Immunoblot analysis

Immunoblot analyses were performed by standard techniques7. Briefly, tissues or cells

were homogenized in a RIPA buffer containing cOmplete Protease Inhibitor Cocktail and

35 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

PhosphoSTOP Phosphate Inhibitor Cocktail Tablets (Roche, Indianapolis, IN, USA) using

a Bead Ruptor 24 Elite bead mill homogenizer (Omni International, Kennesaw, GA, USA).

Protein concentrations were determined by using a BCA reagent (Thermo Fisher

Scientific, Springfield Township, NJ). Equal amounts of protein samples were separated

by SDS-PAGE and transferred to nitrocellulose membranes for immunoblot analysis.

Membranes were incubated with primary (Suppl. Table 1) overnight and

probed with respective secondary antibodies (Dako-Agilent, Santa Clara, CA, USA) for 1

h. Bands were then developed using enhanced chemiluminescence (SuperSignal West

DURA, Thermo Fisher Scientific, Springfield Township, NJ, USA) and imaged by

ChemiDoc XRS+ (BioRad, Hercules, CA, USA).

Electrophoresis for myosin heavy chains

Frozen soleus muscle strips were homogenized in a lysis buffer containing 0.5M NaCl,

20mM NaPPi, 50mM Tris, 1mM EDTA and 1mM DTT, adjusted pH 6.8 using a Bead

Ruptor 24 Elite bead mill homogenizer (Omni International, Kennesaw, GA, USA).

Supernatants from 10 min centrifugation at 2500 x g were diluted 1:1 with .

Samples are then suspended in 1:1 in a Laemmli sample buffer and incubate at room

temperature for 10 min and then heated (70°C) for 10 min. Myosin heavy chains were

36 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

separated by SDS-PAGE in a 6% acrylamide gel containing 30% glycerol over 24 h.

Bands were visualized by Coomassie Brilliant Blue staining45.

Histopathology

Freshly harvested tissues from 17 w-old mice were immersed in 10% neutralized

formaldehyde for 2 d. Following to a paraffin embedding, sectioned tissues were stained

with Hematoxylin and Eosin by the Laboratory of Comparative Pathology, Memorial Sloan

Kettering Cancer Center, NY, USA. Slides were visualized using an Eclipse Ti microscope

(Nikon, Tokyo, Japan).

Tissue triglyceride concentrations

Lipids were extracted from frozen specimens with a mixture of chloroform/methanol (2:1)

using Folch’s method as previously described7. Concentrations of triglycerides were

assayed enzymatically (FUJIFILM Wako Diagnostics, Mountain View, CA).

Plasma assays

Enzymatic assay kits were used to measure plasma concentrations of triglycerides, free

fatty acids, total cholesterol, free cholesterol, phospholipid (FUJIFILM Wako Diagnostics,

37 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

Mountain View, CA), ß-hydroxybutyrate (Stanbio Laboratories, Boerne, TX), aspartate

aminotransferase, alanine aminotransferase (Infinity AST/ALT, Thermo Scientific,

Waltham, MA, USA) and creatine kinase (Stanbio CK Liqui-UV Test, EKF Diagnostics,

Cardiff, UK). Plasma concentrations of insulin were measured by using an ELISA kit

(Crystal Chem, Downers Grove, IL), according to the manufacturer’s protocol.

Rates of fatty acid oxidation

Rates of fatty acid oxidation in muscle tissues were measured by degradation of 14C-

palmitate (American Radiolabeled Chemicals; St. Louis, MO, USA) into 14C acid soluble

14 7,46 metabolites (ASM) and C-labeled CO2 . Briefly, gastrocnemius muscle strips were

collected from 6 h fasted mice. Tissues were kept on ice no longer than 30 min. Muscle

tissues were minced and homogenized in a Dounce homogenizer followed by

centrifugation for 10 min at 420 x g. Supernatants were transferred to microtubes

containing 0.4 μCi 14C-palmitate / 500 μM palmitate conjugated with 0.7 % fatty acid-free

14 BSA and incubated at 37 °C for 30 min. C-labeled CO2 produced by TCA cycle was

captured onto filter paper soaked with 1 M NaOH and 14C-labeled ASM were separated

with 1 M perchloric acid. 14C-labeled CO2 in the filter paper and 14C-labeled ASM in the

supernatant were dissolved in Ecoscint H (National Diagnostics; Atlanta, GA, USA) and

38 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

were counted using a liquid scintillation counter (Beckman Coulter; Danvers, MA).

Fatty acid oxidation in skeletal muscle was also evaluated by applying pulse chase

design47. Soleus muscle strips were freshly dissected and incubated in preoxygenated

HBSS with 10 mM HEPES, Ca and Mg supplemented 2 mM pyruvate, 4 % fatty acid free

BSA and 1 mM palmitic acid adjusted pH 7.4. Isolated soleus muscles were then

transferred into pulse buffer (HBSS with 10 mM HEPES, Ca and Mg supplemented 2 mM

pyruvate, 4% fatty acid free BSA plus 2 uCi of 9,10 3H-palmitate). After washing following

the pulse incubation, the muscle strips with preoxygenated with incubation buffer in the

absence of 3H-palmitate. 3H-palmitate pulsed muscles were then transferred to the tubes

containing chase buffer (buffer: HBSS with 10 mM HEPES, Ca and Mg supplemented 2

14 14 mM pyruvate, 4 % fatty acid free BSA plus 0.5 uCi/ml of C-palmitate) and a CO2 trap.

3 3 H2O produced from the endogenous oxidation of [9,10- H] palmitate was separated from

14 chase buffer with a mixture of chloroform/methanol (2:1). Gaseous CO2 produced from

the exogenous oxidation of [1-14C] palmitate was measured by transferring the chase

14 buffer to 1 M H2SO4 and a paper disc with 1 M NaOH. Liberated CO2 was trapped in the

paper disc. Samples were dissolved in Ecoscint H (National Diagnostics; Atlanta, GA,

USA) and counted using a liquid scintillation counter (Beckman Coulter; Danvers, MA).

39 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

Hepatic VLDL triglyceride secretion rates

Following a 12 h fast, the lipoprotein lipase inhibitor Tyloxapol (500 mg/kg of body weight)

(Sigma-Aldrich) was administered through retro-orbital injection.7 Tail tip blood samples

(25 μL) were collected into microtubes before Tyloxapol injection and at regular intervals

for up to 2 hours. Serum triglyceride concentrations were determined using the enzymatic

assay described above. Rates of hepatic triglyceride secretion were calculated from the

time-dependent linear increases in serum triglyceride concentrations.

Lipidomics

Fatty acids, fatty acyl-CoAs and acyl-carnitines were analyzed by LC MS/MS via Multiple

Reaction Monitoring on an ExionLC AC system coupled to triple quadrupole mass

spectrometer QTRAP5500 (ABSciex, Framingham, MA, USA) at the Emory Integrated

Lipidomics Core (Emory University, Atlanta, GA, USA)7. Briefly, quadriceps muscle

samples were homogenized using Bead Ruptor Elite (Omni International, Kennesaw, GA,

USA). Extracted samples were derivatized with 3-Nitrophenylhydrazine for fatty acids and

n-butanol containing 5 % acetyl chloride for carnitines. The resuspended samples were

subjected to LC MS/MS analysis. Data processing, peak determination, and peak area

integration were performed using the MultiQuant 3.0.2 software (AB Sciex, USA).

40 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

Polar metabolite profiling

Predefined 216 targeted polar metabolites were analyzed at Proteomics & Metabolomics

Core Facility (Weill Cornell Medicine, New York, NY, USA). Briefly, gastrocnemius muscle

tissues were homogenized in 80 % methanol and lyophilized. Measurements were

normalized by tissue weight as described priviously48. Metabolomics pathway analysis

and clustering analysis were performed using Metabo Analyst ver.4.0

(https://www.metaboanalyst.ca/home.xhtml).

RNA sequencing

RNA sequencing was performed at Genomics Resources Core Facility (Weill Cornell

Medicine, New York, NY, USA). Briefly, RNA samples from liver tissues were extracted by

RNeasy Fibrous Tissue Mini Kit and RNeasy Mini Kit following to the manufacturer’s

instruction (QIAGEN, Venlo, Netherlands). After quality check by electrophoresis, RNA

sequencing was performed at 50 million reads per sample using Nova Seq 6000 (illumina,

San Diego, CA, USA).

Cell culture

C2C12 myoblasts (ATCC #CRL-1772) were maintained in high glucose DMEM with 10 %

41 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

bovine serum and 1 % penicillin-streptomycin. To induce myotubes differentiation, C2C12

myoblasts were plated at 25,000 cells/cm2. Once reaching 100 % confluency, media was

changed to DMEM based differentiation medium with 2 % HS and 1 % penicillin-

streptomycin. Differentiation medium was changed 24 h later and every 48 h thereafter.

Myotubes were transduced with AAV8-Mck-Flag-Them2 or AAV8-Mck-LacZ at 1x105

genome copies after 4 d in differentiation medium. Experiments were conducted on d 6

of differentiation.

Mitochondrial respiration

Cellular metabolic flux was determined by measuring oxygen consumption rate (OCR) in

Hepa1-6 cells (ATCC #CRL-1830) with conditioned medium obtained from C2C12

myotubes reconstituted with AAVs. OCR values were measured using a Seahorse XFe

96 extracellular flux analyzer (Seahorse Bioscience; North Billerica, MA, USA). OCR

values were measured under basal conditions and following exposure to oligomycin,

carbonyl cyanide-p-trifluoromethoxyphenylhydrazone (FCCP), rotenone and antimycin A

respectively. After retrieving the plate from Seahorse instrument, live nuclei were

stained with NucRed™ Live 647 ReadyProbes™ Reagent (Thermo Fisher Scientific,

Waltham, MA, USA) following to the manufacturer’s instruction. Live cell nuclei were

42 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

counted in each whole well images which obtained by Spectramax i3x (Molecular Devices,

San Jose, CA, USA). Normalized OCR values were calculated according to the numbers

of live cell nuclei in each well.

Statistical Analyses

Data are presented as mean values with error bars representing SEM. Statistical

significance was determined by using two-tailed unpaired Student’s t-tests when two

groups were compared. Correlations were evaluated by Pearson’s correlation coefficient

analysis. Threshold values were determined by segmental linear regression. Multiple

group comparisons were performed using two-way ANOVA. Differences were considered

significant for P < 0.05 (GraphPad Prism 8, GraphPad Software, La Jolla, CA, USA).

43 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

REFERENCES

1. GBD 2015 Obesity Collaborators, G. 2015 O. et al. Health Effects of Overweight

and Obesity in 195 Countries over 25 Years. N. Engl. J. Med. 377, 13–27 (2017).

2. Cao, J., Xu, H., Zhao, H., Gong, W. & Dunaway-Mariano, D. The mechanisms of

human hotdog-fold thioesterase 2 (h0054HEM2) substrate recognition and

catalysis illuminated by a structure and function based analysis. Biochemistry 48,

1293–1304 (2009).

3. Kanno, K. et al. Interacting proteins dictate function of the minimal START domain

phosphatidylcholine transfer protein/StarD2. J. Biol. Chem. 282, 30728–30736

(2007).

4. Kang, H. W., Niepel, M. W., Han, S., Kawano, Y. & Cohen, D. E. Thioesterase

superfamily member 2/acyl-CoA thioesterase 13 (Them2/Acot13) regulates hepatic

lipid and glucose metabolism. FASEB J. 26, 2209–2221 (2012).

5. Kang, H. W. et al. Thioesterase superfamily member 2/Acyl-CoA thioesterase 13

(Them2/Acot13) regulates adaptive thermogenesis in mice. J. Biol. Chem. 288,

33376–33386 (2013).

6. Ersoy, B. A., Maner-Smith, K. M., Li, Y., Alpertunga, I. & Cohen, D. E. Thioesterase-

mediated control of cellular calcium homeostasis enables hepatic ER stress. J. Clin.

44 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

Invest. 128, 141–156 (2018).

7. Alves-Bezerra, M. et al. Thioesterase Superfamily Member 2 Promotes Hepatic

VLDL Secretion by Channeling Fatty Acids Into Triglyceride Biosynthesis.

Hepatology 70, 496–510 (2019).

8. Pascual, F. & Coleman, R. A. Fuel availability and fate in cardiac metabolism: A

tale of two substrates. Biochimica et Biophysica Acta - Molecular and Cell Biology

of Lipids (2016) doi:10.1016/j.bbalip.2016.03.014.

9. German, N. J. et al. PHD3 Loss in Cancer Enables Metabolic Reliance on Fatty

Acid Oxidation via Deactivation of ACC2. Mol. Cell 63, 1006–1020 (2016).

10. Lindeboom, L. et al. Long-echo time MR spectroscopy for skeletal muscle

acetylcarnitine detection. J. Clin. Invest. 124, 4915–4925 (2014).

11. Canfora, E. E., Jocken, J. W. & Blaak, E. E. Short-chain fatty acids in control of

body weight and insulin sensitivity. Nat. Rev. Endocrinol. 11, 577–591 (2015).

12. Yoon, M. S. The emerging role of branched-chain amino acids in insulin resistance

and metabolism. Nutrients (2016) doi:10.3390/nu8070405.

13. Krüger, A. et al. The pentose phosphate pathway is a metabolic redox sensor and

regulates transcription during the antioxidant response. Antioxidants Redox Signal.

(2011) doi:10.1089/ars.2010.3797.

45 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

14. Sun, Y. et al. Mammalian target of rapamycin regulates miRNA-1 and follistatin in

skeletal myogenesis. J. Cell Biol. 189, 1157–1169 (2010).

15. Bril, F. et al. Metabolic and histological implications of intrahepatic triglyceride

content in nonalcoholic fatty liver disease. Hepatology 65, 1132–1144 (2017).

16. Adams, G. R. Exercise effects on muscle insulin signaling and action invited review:

Autocrine/paracrine IGF-I and skeletal muscle adaptation. J. Appl. Physiol. 93,

1159–1167 (2002).

17. Busquets, S., Figueras, M., Almendro, V., López-Soriano, F. J. & Argilés, J. M.

Interleukin-15 increases glucose uptake in skeletal muscle An antidiabetogenic

effect of the cytokine. Biochim. Biophys. Acta - Gen. Subj. 1760, 1613–1617 (2006).

18. Carbó, N. et al. Interleukin-15 mediates reciprocal regulation of adipose and muscle

mass: A potential role in body weight control. Biochim. Biophys. Acta - Gen. Subj.

1526, 17–24 (2001).

19. Pedersen, L., Olsen, C. H., Pedersen, B. K. & Hojman, P. Muscle-derived

expression of the chemokine CXCL1 attenuates diet-induced obesity and improves

fatty acid oxidation in the muscle. Am. J. Physiol. - Endocrinol. Metab. 302, 831–

840 (2012).

20. Nishizawa, H. et al. Musclin, a Novel Skeletal Muscle-derived Secretory Factor. J.

46 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

Biol. Chem. 279, 19391–19395 (2004).

21. Gu, N. et al. Palmitate increases musclin gene expression through activation of

PERK signaling pathway in C2C12 myotubes. Biochem. Biophys. Res. Commun.

467, 521–526 (2015).

22. Yasui, A. et al. Foxo1 represses expression of musclin, a skeletal muscle-derived

secretory factor. Biochem. Biophys. Res. Commun. 364, 358–365 (2007).

23. Ersoy, B. A. et al. Phosphatidylcholine transfer protein interacts with thioesterase

superfamily member 2 to attenuate insulin signaling. Sci. Signal. 6, 1–12 (2013).

24. Merz, K. E. & Thurmond, D. C. Role of skeletal muscle in insulin resistance and

glucose uptake. Compr. Physiol. 10, 785–809 (2020).

25. Stinkens, R., Goossens, G. H., Jocken, J. W. E. & Blaak, E. E. Targeting fatty acid

metabolism to improve glucose metabolism. Obes. Rev. 16, 715–757 (2015).

26. Rachek, L. I. Free fatty acids and skeletal muscle insulin resistance. Progress in

Molecular Biology and Translational Science vol. 121 (Elsevier Inc., 2014).

27. Schönfeld, P. & Wojtczak, L. Short- and medium-chain fatty acids in energy

metabolism: The cellular perspective. J. Lipid Res. 57, 943–954 (2016).

28. Den Besten, G. et al. The role of short-chain fatty acids in the interplay between

diet, gut microbiota, and host energy metabolism. J. Lipid Res. 54, 2325–2340

47 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

(2013).

29. Magkos, F. et al. Effects of Moderate and Subsequent Progressive Weight Loss on

Metabolic Function and Adipose Tissue Biology in Humans with Obesity. Cell

Metab. 23, 591–601 (2016).

30. Trovato, E., Di Felice, V. & Barone, R. Extracellular vesicles: Delivery vehicles of

myokines. Front. Physiol. 10, 1–13 (2019).

31. Rubio, N. & Sanz-Rodriguez, F. Induction of the CXCL1 (KC) chemokine in mouse

astrocytes by infection with the murine encephalomyelitis virus of Theiler. Virology

358, 98–108 (2007).

32. Lira, S. A. et al. Expression of the chemokine N51/KC in the thymus and epidermis

of transgenic mice results in marked infiltration of a single class of inflammatory

cells. J. Exp. Med. 180, 2039–48 (1994).

33. Tallquist, M. D., Weismann, K. E., Hellstrom, M. & Soriano, P. Early myotome

specification regulates PDGFA expression and axial skeleton development.

Development 127, 5059–5070 (2000).

34. Agah, R. et al. Gene recombination in postmitotic cells: Targeted expression of Cre

recombinase provokes cardiac-restricted, site-specific rearrangement in adult

ventricular muscle in vivo. J. Clin. Invest. 100, 169–179 (1997).

48 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

35. Eguchi, J. et al. Transcriptional control of adipose lipid handling by IRF4. Cell Metab.

13, 249–259 (2011).

36. Rodino-Klapac, L. R. et al. Persistent expression of FLAG-tagged micro dystrophin

in nonhuman primates following intramuscular and vascular delivery. Mol. Ther. 18,

109–117 (2010).

37. Wang, J. J. C. et al. Genetic Dissection of Cardiac Remodeling in an Isoproterenol-

Induced Heart Failure Mouse Model. PLoS Genet. 12, 1–30 (2016).

38. Galindo, C. L. et al. Transcriptional profile of isoproterenol-induced cardiomyopathy

and comparison to exercise-induced cardiac hypertrophy and human cardiac failure.

BMC Physiol. 9, 1–22 (2009).

39. Yoshida, T., Yamashita, M., Horimai, C. & Hayashi, M. Kruppel-like Factor 4 Protein

Regulates Isoproterenol-induced Cardiac Hypertrophy by Modulating Myocardin

Expression and Activity *. 289, 26107–26118 (2014).

40. Krisko, T. I. et al. Dissociation of Adaptive Thermogenesis from Glucose

Homeostasis in Microbiome-Deficient Mice. Cell Metab. 31, 592-604.e9 (2020).

41. De, J. B. & Weir, V. (x949) I09, I-9. J. Physiol.

42. Desai, A. et al. Regulation of fatty acid trafficking in liver by thioesterase superfamily

member 1. J. Lipid Res. 59, 368–379 (2018).

49 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

43. Li, X., Wu, X., Camacho, R., Schwartz, G. J. & LeRoith, D. Intracerebroventricular

leptin infusion improves glucose homeostasis in lean type 2 diabetic MKR mice via

hepatic vagal and non-vagal mechanisms. PLoS One 6, 1–7 (2011).

44. Agouni, A., Owen, C., Czopek, A., Mody, N. & Delibegovic Mirela, M. In vivo

differential effects of fasting, re-feeding, insulin and insulin stimulation time course

on insulin signaling pathway components in peripheral tissues. Biochem. Biophys.

Res. Commun. 401, 104–111 (2010).

45. Picard, B., Barboiron, C., Chadeyron, D. & Jurie, C. Protocol for high-resolution

electrophoresis separation of myosin heavy chain isoforms in bovine skeletal

muscle. Electrophoresis 32, 1804–1806 (2011).

46. Huynh, F. K., Green, M. F., Koves, T. R. & Hirschey, M. D. Measurement of fatty

acid oxidation rates in animal tissues and cell lines. Methods in Enzymology vol.

542 (Elsevier Inc., 2014).

47. Dzamko, N. et al. AMPK-independent pathways regulate skeletal muscle fatty acid

oxidation. J. Physiol. 586, 5819–5831 (2008).

48. Goncalves, M. D. et al. Fenofibrate prevents skeletal muscle loss in mice with lung

cancer. Proc. Natl. Acad. Sci. U. S. A. 115, E743–E752 (2018).

50 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

FIGURE LEGENDS

Figure 1. Them2 is upregulated in skeletal muscle by HFD feeding and its ablation

improves skeletal muscle insulin sensitivity and exercise capacity in HFD-fed mice

Mice were fed a HFD (H) from 5-16 w. (A) Body weights and (B) Lean and fat mass

(determined at 16 w of age). Tolerance tests to glucose (C) were conducted between 16

and 17 w of age. Mice were injected IP with saline or insulin (10mU/g) prior to (D)

measurements of blood glucose concentrations, as well as immunoblot analysis of Akt

phosphorylation in cardiac and gastrocnemius muscle tissues. (E) Expression of Them2

in gastrocnemius muscle (skeletal muscle, SM) and heart (cardiac muscle, CM). (F)

Maximal exercise capacity was analyzed by graded treadmill test (n = 7-15/group). (G)

Cardiac ejection fraction and left ventricle wall thickness were evaluated by

echocardiography before and after isoproterenol osmotic mini pump implantation (†P <

0.05 before vs. after treatment). Chow-fed mice were individually housed in metabolic

cages for the measurement of (H) voluntary running on an exercise wheel, (I) energy

expenditure and (J) respiratory exchange ratio (n = 14/group), *P < 0.05 Them2-/- vs

Them2+/+, †P < 0.05 with running wheel vs without running wheel. Data are mean ± SEM.

Statistical analyses were conducted using Student’s t test or two-way ANOVA with

repeated-measures where appropriate.

51 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

Figure 2. Selective deletion of Them2 in skeletal muscle reduces weight gain and

improves glucose homeostasis and insulin sensitivity in HFD-fed mice

Male C57BL/6 mice with (L to R) skeletal muscle deletion of Them2 (S-Them2-/-,

Them2Flox/Flox x Myf5-CreTg/0); cardiac muscle deletion of Them2 (C-Them2-/-,

Them2Flox/Flox x Myh6-CreTg/0); and adipose tissue deletion of Them2 (A-Them2-/-,

Them2Flox/Flox x Adipoq-CreTg/0). (A) Immunoblot analysis of Them2 with HSP90 as a

loading control. Mice were fed a HFD (H) from 5-17 w. (B) Body weights and (C) lean and

fat mass (determined at 16 w). Tolerance tests to (D) glucose and (E) insulin were

conducted between 16 and 17 w of age. (B-E) Data are mean ± SEM; n=6-18/group.

Statistical analyses were conducted using 2-way ANOVA with repeated measures (B, D,

E) or Students t-test (C); *P < 0.05 compared to control.

Figure 3. Increased glucose uptake and improved insulin sensitivity in skeletal

muscle of HFD-fed S-Them2-/- mice

Hyperinsulinemic-euglycemic clamp studies were performed in mice fed a HFD for 12 w.

(A) Time course of glucose infusion rates, with bar plot presenting glucose infusion rates

during the clamp. (B) Glucose uptake rates were measured for whole body,

52 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

gastrocnemius muscle (skeletal muscle, SM), inguinal white adipose tissue (iWAT) and

brown adipose tissue (BAT). (C) Hepatic glucose production rates measured under basal

and clamp conditions. (S-Them2-/-, n = 6; Control, n = 4), (D) Gastrocnemius muscle

samples were collected from mice fed a HFD for 5-17 w, 10 min after i.p. injection with

saline or insulin (10mU/g), and subjected to immunoblot analysis (n = 4 mice/group). **P

< 0.01, S-Them2-/- vs Control. Statistical analyses were conducted using Student’s t test.

Data are mean ± SEM.

Figure 4. Increased oxidation of endogenous fatty acids and associated metabolic

changes in skeletal muscle of S-Them2-/- mice

Fatty acid oxidation rates were assessed according to degradation rates of [14C]-palmitate

14 into acid-soluble metabolites [ C]-labeled (ASM) and CO2 in homogenized

gastrocnemius (skeletal) muscle from chow-fed (A) global Them2-/- and (B) S-Them2-/-

mice (n = 8/group). (C) Fatty acid oxidation rates were measured in isolated soleus

muscle strips from chow-fed mice using a using [3H]-palmitate in a pulse-chase design

that differentiated between endogenous and exogenous fatty acids (n = 10/group). (D-E)

Gastrocnemius samples were collected from chow-fed mice (C) or HFD (H) for 12 w. (D)

Immunoblot analyses and (E) corresponding densitometry of proteins that regulate fatty

53 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

acid metabolism. Skeletal muscle concentrations of (F) fatty acyl-CoAs, (G) acetyl-

carnitine, (H) free fatty acids and (I) short chain fatty acids (n = 6-8/group). (J)

Unsupervised clustering of data obtained by targeted profiling of 216 polar metabolites in

gastrocnemius muscle. (K) Intermediates of the pentose phosphate pathway (n =

8/group). Statistical analyses were conducted using Student’s t test; *P < 0.05, **P <

0.01, ***P < 0.001, S-Them2-/- vs Control. †P < 0.05, ††P < 0.01, †††P < 0.001, chow vs

HFD. Data are mean ± SEM.

Figure 5. Them2 regulated myocellular fatty acid utilization

Cultured C2C12 myotubes were transduced with 1 x 105 GC AAV8- Mck-LacZ or AAV8-

Mck-Them2. (A) Immunoblot analysis of Them2, with GAPDH used to control for unequal

loading. (B) Representative images of transduced C2C12 myotubes. Cells were fixed and

immunostained with anti-myosin (red) to reveal sarcomeres. Cell nuclei were

stained with DAPI (blue). (C) Myotube area, diameter and fusion index were evaluated

using Image J software. Oxygen consumption rates (OCR) of AAV8 transduced C2C12

myotubes were measured by Seahorse XF96e following to the sequential injections of (i)

oligomycin, (ii) FCCP and (iii) rotenone/antimycin A. (E) OCR values were in the presence

of bovine serum albumin (BSA) treated and palmitic acid (PA) plus BSA. (F) Utilization of

54 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

exogenous fatty acids (FA) was determined by subtracting OCR values in the presence

of BSA alone from PA/BSA. (F) OCR values were measured in the presence of the CPT-

1 inhibitor etomoxir or vehicle (DMSO). (G) Utilization of endogenous FA was determined

by subtracting OCR in the presence of etomoxir from vehicle. Data represent combined

results from 3 independent experiments. Bar plots present FA utilization during the

baseline (basal) and the 3 time points following the addition of FCCP (maximal). Statistical

analyses were conducted using Student’s t test; *P < 0.05, **P < 0.01. Data are mean ±

SEM.

Figure 6. Skeletal muscle Them2 expression enables HFD-fed mice to exceed a

threshold value for accelerated hepatic steatosis and metabolic dysfunction

C57BL/6J mice were fed a HFD for 12 w. (A) Body weight distribution (n = 84) and (B) fat

and lean weights. (C) Linear relationship between fat and lean weights. (D-E) Biphasic

relationships between liver weight and (D) body weight and (E) fat weight (n=64).

Threshold value of 41.6 g body weight was determined by segmental linear regression.

(F) Linear relationship between liver weight and lean weight. (G) Biphasic relationship

between hepatic steatosis determines histologically and body weight. (H) Linear

relationship of liver weight to hepatic steatosis. (I) Linear relationship of hepatic

55 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

triglycerides concentrations to hepatic steatosis. (n=64). (J) Plasma concentrations of free

fatty acids, glucose and total cholesterol. (K) Hepatic mRNA expression levels of selected

genes that govern the metabolism of fatty acids and lipid droplets (n = 8/group). Relative

mRNA expression in skeletal muscle (L) of genes that regulate fatty acid utilization and

(M) myokines. (N) Body and liver weights of HFD-fed S-Them2-/- and control mice (n =

10-14/group) relative to the biphasic linear regression derived for HFD-fed C57BL/6J mice

(panel D). Statistical analyses were conducted using Student’s t test; *P < 0.05, **P <

0.01, ***P < 0.001, body weight < 41.6 g vs body weight > 41.6 g. Data are mean ± SEM.

Abbreviations: Pctp: phosphatidylcholine transfer protein, Aox: alternative oxidase, CPT-

1: carnitine palmitoyltransferase I, Fatp: fatty acid transport protein, Fabp: fatty acid

binding protein, Cidea: cell death activator, Acot: acyl-CoA thioesterase, Acsl: acyl-CoA

synthetase long chain family member, Adipoq: adiponectin, Angptl4: angiopoietin-like 4,

Bdnf: brain derived neurotrophic factor, CXCL1: chemokine ligand 1, Epo: erythropoietin,

Fgf21: fibroblast growth factor 21, Fstl1: follistatin-like 1, Igf1: insulin like growth factor 1,

IL-6: interleukin-6, IL-15: interleukin-15, Lif: leukemia inhibitory factor, Mstn: myostatin,

Osm: oncostatin M, Ostn: osteocrin, Pgc1a: Pparg coactivator 1 alpha, Sparc: secreted

protein acidic and rich in cysteine, Vegfa: vascular endothelial growth factor A.

56 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

Figure 7. Them2 promotes hepatic steatosis by suppressing VLDL secretion

(L to R) Mice with skeletal muscle (S-Them2-/-) and cardiac muscle (C-Them2-/-) Them2

deletion were fed a HFD for 12 w and then livers were analyzed for (A) hepatic steatosis

by H&E staining (bars represent 100µm) and (B) hepatic triglyceride concentrations. (C)

Pyruvate tolerance tests conducted in HFD-fed mice at 16 w of age. Data are mean ±

SEM (n=6-12/group). Statistical analyses were conducted by Students t-test (B) or 2-way

ANOVA with repeated measures (C); *P < 0.05 vs control. Data are presented as mean

± SEM (D) Hepatic triglyceride concentrations of HFD-fed S-Them2-/- and control mice

with body weights of less than 41.6 g (n = 3-7/group). (E) Fatty acid oxidation rates in

livers of HFD-fed S-Them2-/- mice (n = 8/group). (F) Rates of triglyceride hepatic secretion

in HFD-fed mice (n = 8-10/group). Statistical analyses were conducted using Student’s t

test or two-way ANOVA with repeated-measures where appropriate. *P < 0.05, S-Them2-

/- vs Control. Data are mean ± SEM.

Figure 8. Skeletal muscle Them2 mediates crosstalk between muscle and the liver

(A) Rates of oxygen consumption (OCR) of Hepa1-6 cells cultured with conditioned

medium. Sequential additions of oligomycin (i), FCCP (ii) and rotenone/antimycin A (iii)

57 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

are indicated. Conditioned medium was collected from C2C12 myotubes reconstituted

with AAV-LacZ or AAV-Them2. Data represent combined results from 3 independent

experiments. (B) Relative mRNA expression of myokine genes in soleus (skeletal) muscle

dissected from mice fed chow diet or a HFD for 12 w. Myokine genes were also analyzed

in chow-fed mice after 18 h fasting. Heat map represents relative values to controls for

the same diet. (C) Relative mRNA expression of myokine genes in body weight matched

(<41.6g) mice fed HFD for 12 w (n = 5-7/ group). *P < 0.05, S-Them2-/- vs body weight

matched (<41.6 g) control. (D) Schematic summary of metabolic role of Them2 in skeletal

muscle and its influence on the development of hepatic steatosis. Them2 in skeletal

muscle decreases dependence on fatty acid oxidation to produce ATP, therefore utilizing

less circulating free fatty acids. Them2 limits fatty acid oxidation in skeletal muscle

consequently altering the production of short chain fatty acids, branched chain amino

acids and pentose phosphate pathway. These alterations are most evident in the setting

of over nutrition and limit insulin sensitivity in skeletal muscle. Them2 also regulates

muscle-to-organ crosstalk by multiple myokine genes depending on dietary condition,

therefore Them2 may coordinate metabolism of fatty acids in skeletal muscle, adipose

tissue and the liver. Statistical analyses were conducted using Student’s t test (B,C) or

two-way ANOVA with repeated-measures (A). Data are mean ± SEM.

58 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

Supplemental Figure 1. Ablation of Them2 does not promote muscle atrophy or

hypertrophy

(A) Weights of cardiac and gastrocnemius (skeletal) muscle from mice fed chow (C) or

HFD (H) for 12 w (n = 15-17/group, C57BL6J control mice; Them2+/+, global Them2

knockout mice; Them2-/-). (B) Histology of skeletal (gastrocnemius) muscle (SM) and

cardiac muscle (CM). (C) Relative mRNA expression of myosin heavy chain genes in

soleus muscle. (D) Relative mRNA expression of myosin heavy chain and cardiac

hypertrophy genes in ventricle (n = 8/group). (E) Myosin heavy chain isoforms in soleus

muscle determined by electrophoresis separation (n = 8/group, *P < 0.05, Them2-/- vs

Them2+/+). (F) Blood lactate concentrations were measured before (sedentary) and 30

min after treadmill running (exercised mice) (n =8/group). Statistical analyses were

conducted using Student’s t test. Data are mean ± SEM. Abbreviations: Myh, myosin

heavy chain; Anf, atrial natriuretic factor; aSka, α-skeletal actin; Bnp, brain natriuretic

peptide.

59 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

Supplemental Figure 2. Effect of skeletal muscle Them2 on the gene expression

of Acot and Acsl genes

(A) Relative mRNA expression levels of Acot and Acsl genes in skeletal muscle of chow-

fed mice (n = 8/group). (B) Relative mRNA expression of Acot and Acsl genes in skeletal

muscle of HFD-fed mice (n = 8/group). Heat maps represent relative gene expression

compared to C57BL6 for Them2-/- and floxed control for S-Them2-/-.

Supplemental Figure 3. Skeletal muscle expression of Them2 mediates excess

weight gain in mice fed HFD

(A) Body weights of HFD-fed mice housed at 30 °C (n=6-7/group). *P < 0.05 vs control.

(B) Body composition of mice fed a HFD for 12 w while housed at 30 °C (n=6-7/group).

Statistical comparisons were conducted using Student’s t-test; *P < 0.05 vs control. (C)

Immunoblot analysis of Them2 in gastrocnemius muscle with GAPDH as a control for

unequal loading. (D) Body weights of HFD-fed Them2-/- mice following treatment with AAV

carrying Them2 or LacZ as a control (n=7/group). P < 0.05; AAV-LacZ vs AAV-Them2. (E)

Body composition of HFD-fed mice. Mice were maintained at 30 ℃ (n=7/group). *P <

0.05; AAV-LacZ vs AAV-Them2. Statistical analyses were conducted using Student’s t test

or two-way ANOVA with repeated-measures where appropriate. Data are mean ± SEM.

60 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

Supplemental Figure 4. Effect of skeletal muscle Them2 expression on the

expression of genes that control lipid and glucose homeostasis

Mice were (A,B) fed chow or (C,D) HFD for 12 w. Relative mRNA expression of genes in

skeletal muscle that (A,C) regulate fatty acid uptake and oxidation and (B,D) lipid and

glucose metabolism (n = 8/group). Statistical analyses were conducted using Student’s t

test; *P < 0.05, **P < 0.01 vs control. Data are mean ± SEM.

Supplemental Figure 5. Influence of Them2 expression in skeletal muscle on

molecular species of acyl-CoAs, acyl-carnitines and free fatty acids.

Mice were fed chow (C) or a HFD (H) for 12 w. Molecular species of (A) fatty acyl-CoAs,

(B) fatty acyl-carnitines, and (C) saturated and unsaturated long chain and short chain

free fatty acids. (n = 8/ group). Statistical analyses were conducted using Student’s t test;

*P < 0.05, **P < 0.01, ***P < 0.001 vs control. Data are mean ± SEM.

Supplemental Figure 6. Pathway analysis of metabolomic data illustrating the

influence of Them2 expression in skeletal muscle

Mice were fed (A) chow or (B) a HFD for 12 w after which gastrocnemius muscles were

subjected to targeted profiling of 216 polar metabolites. Data were analyzed by

61 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

MetaboAnalyst. Each node contains all the matched pathways arranged by p values (from

pathway enrichment analysis) on vertical-axis, and pathway impact values (from pathway

topology analysis) on horizontal-axis.

The node color is based on its p value and the node radius is determined based on their

pathway impact values.

Supplemental Figure 7. Effect of skeletal muscle Them2 expression on amino acids

contents

(A,B) Relative concentrations of amino acids in gastrocnemius muscle. (n = 8/group)

Statistical analyses were conducted using Student’s t test; *P < 0.05, **P < 0.01, ***P <

0.001 vs Control. Data are mean ± SEM.

Supplemental Figure 8. Effect of skeletal muscle Them2 expression on measures

of oxidative stress

Mice were fed chow or a HFD for 12 w. (A) Superoxide dismutase (SOD) activities were

assessed in skeletal muscle (n = 6/group). (B) Tissue-specific lipid peroxidation level was

analyzed using a thiobarbituric acid reactive substances (TBARS) assay in skeletal

62 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

muscle (n = 7/group). Statistical analyses were conducted using Student’s t test. Data are

mean ± SEM

Supplemental Figure 9. Plasma characteristics of S-Them2-/- mice

Mice were fed chow or a HFD for 12 w and the plasma was collected for (A) Activities of

aspartate aminotransferase (AST), alanine aminotransferase (ALT) and creatine kinase

(CK) (n = 9 – 14/group). (B) Fasting concentrations plasma insulin (n = 8/group). (C)

Fasting concentrations of ß-hydroxybutyrate (b-HB/group) (n = 16). (D) Plasma

concentrations of triglycerides (TG), non-esterified fatty acid (NEFA), total cholesterol (T-

Cho), free cholesterol (F-Cho) and phospholipids (PL) (n = 8 – 10/group). Statistical

analyses were conducted using Student’s t test; *P < 0.05, **P < 0.01 vs control. †P<0.05,

†† P<0.01, ††† P<0.001 chow vs HFD. Data are mean ± SEM.

Supplemental Figure 10. Influence of skeletal muscle Them2 expression on hepatic

mRNA expression

(A) Volcano plot of RNAseq data comparing liver of control and S-Them2-/- mice fed a

HFD for 12 w. Red font represents significant changes in gene expression after

adjustment for false discovery rate. The top ten genes are annotated. (B) Identification of

63 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

top 10 pathways influenced by Them2 expression as assessed by Ingenuity Pathway

Analysis (Qiagen) of RNAseq data. Bar color indicates predicted directionally. Z-scores,

which represent directionality were calculated based on the data set’s correlation with the

activated state. White indicates z-score = 0 (no activation) and gray indicates no activity

pattern.

64 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

Supplemental Table 1

Protein Source Reference number ACC Cell Signaling Technology (Danvers, MA, USA) 3661 Akt Cell Signaling Technology 9272 alphaGAPDH Novus Biologicals (Centennial, CL, USA) NB100-56875 AMPK Cell Signaling Technology 2532 CD36 Santa Cruz Biotechnology (Dallas, TX, USA) 7309 CPT-1 Proteintech (Rosemont, IL, USA) 15184-1-AP HSP-90 Santa Cruz Biotechnology sc-25840 Phospho-ACC (Ser79) Cell Signaling Technology 3662 Phospho-Akt (Ser473) Cell Signaling Technology 9271 Phospho-Akt (Thr308) Cell Signaling Technology 4056 Phospho-AMPK (Thr172) Cell Signaling Technology 2535 PCTP Home Shoda et al., 2001, Hepatol 33,1194-1205 Them2 Home Kanno et al., 2007, J Biol Chem 282, 30728-30736

65 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

Supplemental Table 2 Gene Forward primer (5' - 3') Rverse primer (5' - 3') Acot1 CTGGCGCATGCAGGATC GGCACTTTTCTTGGATAGCTCC Acot2 CTGAAGTCAACGACGCAAAA CGGCGGAGGTACAAACAG Acot3 GCTCAGTCACCCTCAGGTAA AAGTTTCCGCCGATGTTGGA Acot4 TTGAAGAAGCAGTGCGGTACA AAAGACCCAGAAGCCCAATGT Acot5 TCAGGATGGTGCCAACAGTA GATGCTCAGCGGTTCGTC Acot6 ACGCCATCCTCAGGTGAAAG TGTCTCCAAAGGTGCTGATCTGTGCC Acot7 TGACCAATAAAGCCACCTTGTG CCTGCTCCTGCCGTAAATACAC Acot8 GATGCTGTACGAGTGTGAGAGC ACAGCAAGGACCCCATCC Acot9/10 TGCTACATGCACAACCACAA AAGGTTGCATCCAAAACAGG Acot11 (Them1) AGATCATGGCTTGGATGGAG AAAGGCGTTATTCACGATGG Acot12 GGAGATTACCACCACCTTGG TTCAACCTTAACAGATATGGCATC Acot13 (Them2) TCGACCATGGCTCTAATGTG TCCTGTGGTCTTGTTGGTCA Acsl1 TGGGGTGGAAATCATCAGCC CACAGCATTACACACTGTACAACGG Acsl3 CAATTACAGAAGTGTGGGACT CACCTTCCTCCCAGTTCTTT Acsl4 TGCCAAAAGTGACCAGTCCTATG TGTTACCAAACCAGTCTCGGGG Acsl5 TCCCAGCCCACCTCTGATGATGTG ACAAACTGTCCCGCCGAATG Acsl6 TGAATGCACAGCTGGGTGTA ATGTGGTTGCAGGGCAGAG

Myh1 (Type IIx) TCTGCAGACGGAGTCAGGT TTGAGTGAATGCCTGTTTGC Myh2 (Type IIa) GTCAAAGGAGGAGGAACAGCAGC GTTGAGTGAATGCTTGCTTCCCC Myh4 (Type IIb) TGGCCGAGCAAGAGCTAC TTGATGAGGCTGGTGTTCTG Myh6 (MHC-α) GCCCAGTACCTCCGAAAGTC GCCTTAACATACTCCTCCTTGTC Myh7 (Type I, MHC-β) CCAAGGGCCTGAATGAGGAG GCAAAGGCTCCAGGTCTGAG Anf CCATATTGGAGCAAATCCTGTG CGGCATCTTCTCCTCCAGGT aSKa TTGACGTGTACATAGATTGACTCGTTT TGGCTGGCTTTAATGCTTCA Bnp GGGAGAACACGGCATCATTG ACAGCACCTTCAGGAGATCCA

Adipoq TGTTCCTCTTAATCCTGCCCA CCAACCTGCACAAGTTCCCTT Angptl4 CATCCTGGGACGAGATGAACT TGACAAGCGTTACCACAGGC Bdnf TCATACTTCGGTTGCATGAAGG AGACCTCTCGAACCTGCCC Cxcl1 ACGAAATGCGAAATCATGTGC CTGTGTCGTCTCCAGGACAA Epo ACTCTCCTTGCTACTGATTCCT ATCGTGACATTTTCTGCCTCC Fgf21 CTGCTGGGGGTCTACCAAG CTGCGCCTACCACTGTTCC Fstl1 CACGGCGAGGAGGAACCTA TCTTGCCATTACTGCCACACA Igf-1 GTGAGCCAAAGACACACCCA ACCTCTGATTTTCCGAGTTGC IL-6 TAGTCCTTCCTACCCCAATTTCC TTGGTCCTTAGCCACTCCTTC IL-15 ACATCCATCTCGTGCTACTTGT GCCTCTGTTTTAGGGAGACCT Irisin TTGCCATCTCTCAGCAGAAGA GGCCTGCACATGGACGATA Lif ATTGTGCCCTTACTGCTGCTG GCCAGTTGATTCTTGATCTGGT Mstn AGTGGATCTAAATGAGGGCAGT GTTTCCAGGCGCAGCTTAC Osm CCCGGCACAATATCCTCGG TCTGGTGTTGTAGTGGACCGT Ostn CGTCTTGATGATCTGGTGTCC TGGGAATACCAAACCGCTTTT Sparc GTGGAAATGGGAGAATTTGAGGA CTCACACACCTTGCCATGTTT Vegfa GCACATAGAGAGAATGAGCTTCC CTCCGCTCTGAACAAGGCT

CD36 CAACCACTGTTTCTGCACTG CAGGCTTTCCTTCTTTGCAC Fatp1 CGCTTTCTGCGTATCGTCTGCAAG AAGATGCACGGGATCGTGTCT Fabp3 CACAGAGATCAACTTTCAGCTGGG CCATGAGTGAGAGTCAGGATGAG Aox TCAACAGCCCACTGTGACTTCCATTA TCAGGTAGCATTATCCATCTCTCA Cpt1 TTTGACTTTGAGAAATACCCTGACTA TGGATGAAATTCTCTCCCACAATAA Cpt2 CAGTGCACAGAAGCCTCTCTTG CTTCCCAATGCCGTTCTCAA Acc GAGAGGGCTCAAGTCCTTCC ACATCCACTTCCACACACGA Acadl CTCCGCCCGATGTTCTCATTC CTCTACTCACTTCTCCAGCTTTC

Pctp CCAGAGTATCTCGGCACCTC ACGCTTTCACCATGTCCTTC Pgc1a CAACATGCTCAAGCCAAACCAACA CGCTCAATAGTCTTGTTCTCAAATGGG Ppara GTGGCTGCTATAATTTGCTGTG GAAGGTGTCATCTGGATGGTT Pparg CACAATGCCATCAGGTTTGG GCTGGTCGATATCACTGGAGATC Ppard TCCATCGTCAACAAAGACGGG ACTTGGGCTCAATGATGTCAC Ucp2 TGCCTTCCTTTCTCCGCTTGG GAAAGGTGCCTCCCGAGATTGGTAG Ucp3 CCGATACATGAACGCTCCCCTA ATCATCACGTTCCAAGCTCCC Pdk4 AGGGAGGTCGAGCTGTTCTC GGAGTGTTCACTAAGCGGTCA Glut4 TCGTCATTGGCATTCTGGTTG AGCTCGTTCTACTAAGAGCAC Dgat1 GGCCCAAGGTAGAAGAGGAC GATCAGCATCACCACACACC Dgat2 AGGATCTGCCCTGTCACG GCCAGCCAGGTGAAGTAGAG

CyclophilinA GGCCGATGACGAGCCC TGTCTTTGGAACTTTGTCTGCAA

66

bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

A B C

50 30 8 +/+ L 700

Them2 d P = 0.08 / P = 0.07

* 25 g 600 g -/-

* ) 

 Them2 m

40 0 t

6 ,

500 0

h ]

g 20

0

g

e

i

0

s s

e 400

1 s 15 o

w 30

x

a c

 4

(

u y

M 300

l

d C

10 g

o U

+/+ d 200

B 20 A

Them2 5 o 2 o

Them2-/- l 100 B 0 [ 10 0 0 6 8 10 12 14 16 Lean Fat 0 30 60 90 120 Agew Time, min D p-Akt/Akt

L 400

+

d

/

/

+ g

2 *

m

300 m

, 4

]

e

e

h

s

T

o o

c 200

t

u

l * e

g 2

v

i

t d

100 a

o

l

o

e

l

R B [ 0 0 Saline Insulin SM CM

E n

o i

s C s 2.0

e *

r H

p x

e 1.5

2 m

e 1.0

h

T

e

v 0.5

i

t

a l e 0.0 R SM CM

F G H

200 † † † ,

m

% g

e 2.0

, s

90 n

m

i

i

n

n

i

, c

n 4

o

r

3

s

i

n m

150 t

e

s

0

1.5 u

c

,

x

r

e

1

y

a

e

n

t

r y

60

i

x

l

f

r

k

c

a a

100 c

n

i t

a 1.0

m

m

o

h n

p 2

i

i

t

t

u

a

x

l

l

c

l c

a 30

o

e a

50 j 0.5

* V

M

E W 0 0 0.0 0 C H - + - + Total Light Dark Isoproterenol Isoproterenol

I J

1.0

Wheel

o

i

t

a r 15

† e ,

† g

e * n

r 0.9

a u

t *

i

h c

d * * x n 10 †

l †

e e

*

a

p

y

c

x

r

k

e *

o

t y

a 0.8 r

g 5

i

r

p

e s

n +/+

e Them2

E R 0 Wheel acclimation Them2-/- Wheel - + - + - + - + - + - + 0.7 0 4 8 12 16 20 0 4 8 12 16 20 24 Total Light Dark Time, h Fig 1 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

A

S-Them2-/- C-Them2-/- A-Them2-/-

Them2

HSP90

Control Control Control

Them2

HSP90

B 50 50 50

* g

 40 40 40

t

h

g i

e 30 30 30

w

y d

o 20 Control 20 Control 20 Control B S-Them2-/- C-Them2-/- A-Them2-/- 10 10 10 6 8 10 12 14 16 6 8 10 12 14 16 6 8 10 12 14 16 Age, w Agew Agew C 30 30 30 25 * 25 25

20 * 20 20

g

 s

s 15 15 15 a

M 10 10 10 5 5 5 0 0 0 Lean Fat Lean Fat Lean Fat D

L 700 700 700

d /

g 600 600 600

m *

,

] 500 500 500 e

s 400 400 400

o

c u

l 300 300 300

g

d 200 200 200

o o

l 100 100 100 B [ 0 0 0 0 30 60 90 120 0 30 60 90 120 0 30 60 90 120

400

E L 400 400

d /

g * m

300 300 300

,

]

e s

o 200

c 200 200

u

l

g

d 100 100 100

o

o

l B [ 0 0 0 0 15 30 45 60 0 15 30 45 60 0 15 30 45 60 Time, min Time, min Time, min

Fig 2 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

A B

Whole body SM iWAT BAT 2.0

, 0.08 0.4 0.5 0.8 ,

e Control e

t Control

e

t

t a

a 0.010 r

-/- a * r

-/- r

0.4 ,

S-Them2 n

S-Them2 i n 1.5

n 0.06 0.3 0.6

p

n

n

i

e

i

o

o

i

m

k

i

m

/

m

s

m

/

a

s

a

/ g

t 0.3

l

u

g

u

k

f

g

p

c

f

/

k

l

k 1.0 n / 0.04 0.2

u 0.4

n

i

/

l

i

g

o

0.005

g

o

e n

e 0.2

i

e

m

s

r

s

m

m

s

o

u

m

o

o

c

m c

0.5 d 0.02 0.1 0.2

c

u u

l 0.1

u

l

l

G

G G 0.0 0.000 0.00 0.0 0.0 0.0 0 30 60 90 120 Time, min

C D

Basal Clamp Akt phosphorylation

, n

o 0.15 0.08

i

t l

c 4

o u

r ** **

t

d

n

n o

i 0.06 o

r 3

c p

m 0.10

/

o

e

g

t

k

s

/ l

0.04 e

o 2

v

o

c

i

t

u

l

m a

0.05 l

g

m 0.02 e

c 1

i

R

t

a p e 0.00 0.00 0 H S473 T308

Fig 3 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

A B C D

, ,

3 2.0 , e

e 0.5

t t

+/+ e

Control t a

Them2 a

l

l

r

r

a

o o * r

r *

r

n -/- n

t * * -/- t 0.4

S-Them2 n o

* o 1.5 n

i Them2

n

i

t

o

t

g

o

i o

2 /

a

t

a

c

c

n

d

a

d

i

i i

o 0.3

o

d

x

t

x

i

t

m

1.0

/

o

x

o

l

e

e

o

o

v

d

v

d

i

i

i

i t

t 0.2 e

c 1

c

m

t

a

a

l

a

l

a

n

a

0.5

t

e

e

i

y

y

r

r

t

t t

t 0.1

m

l

a

a

a F 0 F 0.0 P 0.0 CO2 ASM CO2 ASM s s u u o o n n e e g g E o o n d x

o n E i 4 E

s †††

s

e r

p 3

x ††† e

n i

e 2

t †† ** o

r * *

p

e 1

v

i

t

a l

e 0 R C H C H C H C H C H C H

2 P K 1 6 m T P C 3 e - T h C M C P D T P /A /A C C K C P C M A A p p

F G H I ,

2.5 600 ] d

i 100

, **

P = 0.08 ,

** c

]

]

e

,

e

] a

* s

e

u

u

2.0 e

e d

A *

n

y

s

s

i

u i

* t

u o

t 4

s

t

s

c

s

i

i i

s 400

t

t

a

a

C

s

n

s

f

i

-

t

i

t

l

g

r

t y

g 1.5

t

y

n

a

i

t

g

m

g

c

m

c

/

a a

/ 50

-

a

f

m

l

d

m

h

d

/

i

/

i

y y

1.0 c

e

t

g

t

p

g

p 2 200

i

t

t

i

e

l

e

n

l

r

n

r

a

c o

g **

F

g

F

[

A

[

h n

0.5 [

n * *** ***

S [ 0.0 0 0 0 C H C H C H C H C H C H C H C H C H te te te te te te a a a a a a t n r r o rm e o ty le n o c i u a a F A p B v x ro yl e P th H e -M 3 J K

Pentose phosphate pathway

2.5 l

o * * r

t 2.0

n *

o c

1.5

o

t

e

v 1.0

i

t

a l

e 0.5 R 0.0 C H C H C H C H C H C H e te te te te id Class s a a a a c o h h h h a c p p p p lu s s s s ic C Control G o o o o v - h h h h ru C S-Them2-/- D -p -p -p -p y 6 6 5 5 P e e e e H Control s s s s o o o o -/- c t l b H S-Them2 u c u i l u ib R G r - F -R D D

Fig 4 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

A B

Them2

n o

i 2.0 s

LacZ mThem2 LacZ

s e

r *

p 1.5 Them2

x e

Them2

n i

e 1.0

t

o

r p

GAPDH

e 0.5

v

i

t

a l

e 0.0 R

C l 20 l

a 0.6 50

/

m

e

µ

b **

,

%

r u

40

t ,

e 15

t

o

a

y e

0.4 e

r m

m 30

a

,

a

i x

10 e

d

e

b

d u

e 20

t

n

b i

0.2 o

u

y t

5 n

o M

o 10

i

y

s

M u

0 F 0.0 0

D E

i LacZ Them2 e

l ii iii Exogenous FA utilization c

400 BSA i

u

e

l

n c

0 150

PA/BSA u 0

i n

0

,

0

0

0

1

0 /

300 , *

n 0

i 100

1

/

m

/

n

l

i

o

m

/

m

l p 200 o

50 *

,

m

R

p

C

, R O Baseline Maximal C 0

100 O 0 20 40 60 80 Basal Maximal Time, min F G

LacZ Them2

i iii Endogenous FA utilization i

e ii l

Vehicle e

c

l c

u 80 u

n 300 Etomoxir

n

0

0

0

0 0

, i

0 60

0

,

1

0

/

1

n

/

i

n i

m 40

/

l m

200 /

o

l

o

m

p m

20

,

p

,

R

R

C C O Baseline Maximal 0 100 O Basal Maximal 0 20 40 60 80 Time, min

Fig 5 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

A B C D E

40 25 Median 42.5g Fat < 41.6 g 10 41.6 Lean 3 > 41.6 g 3

30 20

e

g

g

c

i

,

,

t

t

g

m

h

h

,

f

g

t g g 2

2 i

i

o

h

,

e

e

t

r

g i

20 w

a 15

w

e

e

r F

b 5

r

e

e

W

m

v

v

i

i

u

L L N 1 1 10 10

0 0 5 0 0 25 30 35 40 45 50 55 30 35 40 45 50 55 20 25 30 30 35 40 45 50 55 10 15 20 Body weight, g Body weight, g Lean, g Fat weight, g Body weight, g F G H I

300 e

41.6 u

s s

3 100 3 i

t

g

/

%

g

,

g

m

g

s

i

, 200

,

,

t

s

]

t

h

s

o

h

t

g

e g

2 i 2

a

i

d

e

i

e

e

r t

50 w

w

s

e

r

c

r

c

e

i

y

e

l

t

v

v

i

g

i

a i

31 L

r L

p 100

t

e 1

1 c

i

H

t

a p

0 e

H [ 0 0 0 20 25 30 30 40 50 0 20 40 60 80 100 0 20 40 60 80 100 Lean weight, g Body weight, g Hepatic steatosis, % Hepatic steatosis, % J K

< 41.6 g n o

i 80

> 41.6 g s

s 50 ***

e

r

L L

/ 1.0 500 300

p

d

/ q

x 20

g E

** L **

*** e 4

m

d ***

m

0.8 / 400

,

,

A

]

g

]

l

s

o N

m 200 r

3

d ,

i 0.6 300

e

R

]

t

c

e

s

a

m

s

e

l 2 y o * *

t 0.4 200

e

o

c

t h

100 v

u

a

i

l

f

c

t

l 1

G e a *

[ 100

0.2 a

l

t

e

r

e

o

F T

[ 0 R 0.0 0 [ 0 p x a 2 6 1 2 5 1 a -t o 1 t 3 p p p p e c A t p t t t b d P p C D a a a a i

L C C F F F F C

n o

i 2.0

s

s

e r

p 1.5 *** * **

x * ** * ** **

e

A

N 1.0

R

m

e 0.5

v

i

t

a l

e 0.0 R 1 2 3 4 5 6 7 8 0 ) 2 ) 1 3 4 5 6 t t t t t t t t 1 1 1 2 l l l l l o o o o o o o o / m t m cs cs cs cs cs c c c c c c c c t9 e o e A A A A A A A A o h c h A A A A A c T A T A ( ( 1 3 t1 1 o t c co M A A N

n 2.0

o

i s

s * e **

r 2 * *** ** *

p

x

e

g

1.5

,

t

A

h

g

N

i

e R

1 w

r

m

e

v

e

i

v L

i 1.0

t

a l

e 0 R q 4 f 1 o 1 1 1 6 5 n if n n a c a o tl n l p 2 tl f - 1 i L t m t 1 r f p p d c f s Ig L - is s s s c a g 0.5 i g B x E g F I L Ir M O O g p e d C I 6

F . A n P S V 35 40 45

A 1 4 Body weight, g Fig 6 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

A

+/+

-/-

B

g /

g 150 250

m Control Control

,

s -/- 200 -/-

e S-Them2 C-Them2

d i

r 100

e 150

c y

l * g

i 100

r t

50

c i

t 50

a

p e

H 0 0 C

L 500 500 d

/ Control Control g

400 -/- 400 -/- m

S-Them2 C-Them2

,

] e

s 300 300 o

c * u

l 200 200

g

d

o 100 100

o

l B [ 0 0 0 60 120 180 0 60 120 180 Time, min

D < 41.6 g E F

150 , 2.5

e

L

t

n i

, Control

d

a

]

l /

r *

m

s

o -/- /

g 1000 r

e 10

n 2.0 S-Them2 t

g *

e

d

m

o

i

n

i

t

m

u

r

,

o

100 ]

s

,

a

e

c

s

s

c e

d 1.5

i

i

t

e

t

o

y

l

x

t

a

d

i

r

g

o

g

r

i

/

e *

r n

e 500

v

d t

g 1.0 5

i

i

c

o

t

r i

50 * c

m

y

t

a

l

e l

a *

e

v

g

e

r

i

i y

r 0.5

r

t

c

L

t

[

e

T

a

[ S 0 F 0.0 0 0 CO2 ASM 0 30 60 90 120 Time, min

Fig 7 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

A 450 B i ii iii

) Adipoq Adipoq Adipoq

s l

l * Angptl4 Angptl4 Angptl4

e c 400 Bdnf Bdnf Bdnf 0 1.5

0 Cxcl1 Cxcl1 Cxcl1 0

, * * 5

/ Epo Epo Epo n

i 350 Fgf21 Fgf21 Fgf21 m

/ *

l Fstl1 Fstl1 Fstl1 o

m Igf1 Igf1 Igf1 p

( * * IL-6 IL-6 IL-6 R 200 *

C IL-15 IL-15 IL-15 1.0 O Irisin Ir*isin Irisin Baseline Maximal * 100 Lif Lif Lif 0 20 40 60 80 Mstn Mstn Mstn Time (minutes) Osm Osm Osm Ostn Ostn O*stn 2%HS DMEM Sparc Sp*arc Sp*arc * LacZ conditioned Vegfa Vegfa Vegfa 0.5 Them2 conditioned Chow Fasting High-fat

C Control < 41.6 g

S-Them2-/- < 41.6 g

n

o

i

s

s e

r 2

p * *

x

e

A N

R 1

m

e

v

i

t

a l

e 0 R q 4 f o 1 1 5 if n c a tl n l1 1 tl f -6 in L t m tn r f o p d c p f2 s g L -1 is s s s a g ip g B x E g F I I L Ir M O O p e d n C F I S V A A D

Fig 8 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

A B C D

%

,

n s -/- o

250 +/+ i 3

+/+ Them2 Them2

s m

Them2 r s

g 80 o

e *

f

s m

200 r

o

u -/-

,

p

i t

Them2 s

x

i

h m

e 2 60

h

g e

i 150

y

A

n

e

c

M

N w

SM

o

f

r 40

R e t *

100 o

l

s

m

c

1 n

a

s

e

o

i

u

G v t 20

50 i

t

u

m

a

b

l

i

r e 0 t

0 s 0 R C H i ) ) ) ) D I I 7 1 2 4 I h h h h e e y y y y yp p M M M M T y ( ( ( ( T I Ix a b t I I e I I I h p +/+ -/- y e e e g p p p i 0.8 Them2 Them2 T y y y

e T T T

w

y

d 0.6

o

b

%

0.4 ,

t CM

h

g i

e 0.2

w

t

r a

e 0.0 H C H

E F G

n

n o

o 15

i L

i 1.5 2.0

/

s

l

s

s

s

o

e

e

m

r

r

p p

1.5 m

x

x

, 10

] e

1.0 e

e

t

A

A

a

t N

N 1.0

c

R

R

a

l

m

m 5

0.5

d

e e

0.5 o

v

v

i

i

o

t

l

t

a

a

B

l

l

[ e

0.0 e 0.0 0

R R f y d 6 7 n a p r e h h k n ta s y y A S B n i M M a e rc d e e x S E

Fig S1 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

A B Chow High-fat 2.0 2.0 Acot1 Acot1 Acot2 Acot2 Acot3 Acot3 Acot4 Acot4

Acot5 1.5 Acot5 1.5

t t

Acot6 o Acot6

o

c

c A

A Acot7 Acot7 Acot8 Acot8 Acot9/10 1.0 Acot9/10 1.0 Acot11 Acot11 Acot12 Acot12 Acot13 Acot13

Acsl1 0.5 Acsl1 0.5 l

l Acsl3 Acsl3

s

s c

c Acsl4 Acsl4 A A Acsl5 Acsl5 Acsl6 Acsl6 0 0 Them2-/- S-Them2-/- Them2-/- S-Them2-/-

Fig S2 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

A B 50 30 * Control

* 25 -/- g

40 S-Them2

,

t

g

h 20

,

g

s

i s

e 30 15

a

w

M y

d 10 o 20 Control B 5 S-Them2-/- 10 0 6 8 10 12 14 16 Lean Fat Age, w

C D E

50

* 30 * AAV-LacZ g

25 AAV-Them2 ,

t 40

h g g *

i 20

,

e

s w 30 s

15

a

y

d M

o 10

B 20 AAV-LacZ AAV-Them2 5 10 0 6 8 10 12 14 16 Lean Fat Age, w

Fig S3 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

A B

Fatty acid uptake and oxidation Lipid and glucose homeostasis

n

n

o

o i

i 2.5 2.5 s

s Control

s

s

e

e r

2.0 -/- r 2.0 p

S-Them2 p

x

x e

** e

1.5 1.5 *

A

A

N

N R

1.0 R 1.0

m

m

e

e

v

v i

0.5 i 0.5

t

t

a

a

l

l e

0.0 e 0.0

R R 6 1 3 x 1 2 c l p a a g d 2 3 4 4 1 2 3 p p o t t c d -t 1 r r r p p k t t t t b A p p A a c c a a a c c d lu a a D a a C C c g p p p P g g C F F A P P P P P U U G D D

C D

Fatty acid uptake and oxidation Lipid and glucose homeostasis

n

n

o

o i

i 2.0 2.5

s

s

s

s

e

e r

r 2.0 * p

p 1.5 *

x

x

e

e

1.5

A

A N

N 1.0 R

R 1.0

m

m

e

e 0.5

v

v i

i 0.5

t

t

a

a

l

l e

e 0.0 0.0

R R 6 1 3 x 1 2 c l p a a g d 2 3 4 4 1 2 3 p p o t t c d -t 1 r r r p p k t t t t b A p p A a c c a a a c c d lu a a D a a C C c g p p p P g g C F F A P P P P P U U G D D

Fig S4 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

A B

2.0 2.0

Control

e

e

u u

-/- ,

s s 1.5 S-Them2 e

, 1.5

s

s

n

i

i

i

t

t

A

t

i

o

g

g

n

r

C

m

m

-

a

/ /

l 1.0 1.0

c

y

d

d

-

i

i

l

c

p

p

y

i

i

A

l

l

c

A g

g 0.5 0.5

n

n

0.0 0.0 C H C H C H C H C H C H C H C H C H l l l l l y y y y y c c c c n r o o o i i ti i o ty n n n le le i n i a a a o m o p u x c c O n l id o B e e e i a h r d L P c P H D a a x r e A H C D Saturated Unsaturated

80 , , 150 ] ***

* ]

s

s

e

d

e d

i 60

i

u

u

c

c

s

s

a

a

s

s

i

i t

y 100

t

y

t

t

t t

g 40

g

a

a

f

f

m

m

*

/

/

e

e

g

g

e

e n

r 50 n

20 r

F

F [ ** ** [ 0 0 C H C H C H C H C H C H C H C H c c c c ic ic ic ic i ti i i le le in n r s it r o l o u ri a O n o id a y lm te i in h L a S L L c M P ra A

Fig S5 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

A B

Fig S6 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

A B

Chow High-fat

L-Histidine * Control L-Histidine -/- * L-Arginine * S-Them2 L-Arginine L-Lysine * L-Lysine

c L-Glutamic acid i

l L-Glutamic acid

c

i

i

l

i

h p

L-Aspartic acid h L-Aspartic acid

o

p

r

o d

L-Glutamine r y

d L-Glutamine

y H L-Asparagine ** H L-Asparagine L-Tyrosine L-Tyrosine L-Threonine L-Threonine L-Serine L-Serine L-Proline ** L-Proline L-Tryptophan L-Tryptophan L-Phenylalanine

** L-Phenylalanine c

i L-Methionine

b

c

n

i i

o L-Methionine

a

b

n

h

i h

L-Isoleucine o

p

a

c

h

o

** L-Isoleucine h

r

p

d

c

d o

L-Leucine e *

r

y d

** h d

L-Leucine e

c

H

y h

L-Valine n **

c

H a

** r

L-Valine n B

L-Alanine a r

L-Alanine B Glycine * Glycine 0.0 0.5 1.0 1.5 2.0 Relative to Control 0.0 0.5 1.0 1.5 2.0 Relative ratio to Control

Fig S7 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

A B

100 4 n

Control i

e

t

o ,

80 -/- r y

p 3

n S-Them2

t

i

o

i

g

v

t

i

i t

60 μ

/

b

c

i

a M

h 2

μ

n

i

D ,

40

O

S

% S R 1

20 A B 0 T 0 C H C H

Fig S8 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

A B C

150

L 1.0 L

Control /

l

/ l

o †

-/- 3 o

L

m /

† S-Them2 ††† m 0.8

m

U

m

100 †††

,

,

,

] ]

] * n

a † i

B 0.6 †

l 2

m

u

H

s

-

s

a

β

n l

50 i 0.4

P

a

[ a

1 m

m s

s 0.2

a

l

a

l

P P 0 [ [ 0 0.0 C H C H C H C H C H

AST ALT CK

L L

D L

/

d

d

L

/

/

q

d

g

g / E 250

80 0.6 60 g 400

m

m

m ††† †††

, ††† ††

L

††† ,

m

]

,

]

l

l

]

d

,

/

]

o s ** o

r 200

g

r

d

d

i i

60 e 300

e

t

m

p

t

c

i

s

l s

, 0.4 40

a

]

e

o e

l 150

l

y

h

G

t

o

o

t

p T

40 h 200

h

a

s

c

f

c

a

o

l 100

e

h

e a

m 0.2 20

t

e

p

e

s

r

r

o

f

a t

20 f 100

l

a

50

a

a

a

P

m

[

m

s

m

m

s

a

s s 0 l

0 a 0.0 0 0

a

a

l

l

l

P

[

P

P

P

[ [ C H C H C H [ C H C H

Fig S9 bioRxiv preprint doi: https://doi.org/10.1101/2021.04.21.440732; this version posted April 21, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission.

A B

10 Canonical pathways 8 Cidea Triacylglycerol Degradation ) Hsp90aa1

e α-tocopherol Degradation

u l

a PXR/RXR Activation v

4 Aqp8 Gtpbp4-ps1 Heparan Sulfate Biosynthesis P

( B430212C06RikLipe Adh6-ps1 RP23-306P12.3 Basal Cell Carcinoma Signaling

0 Fam83f Avpr1a

1 Heparan Sulfate Biosynthesis

g Osteoblasts, Osteoclasts and Chondrocytes

o 2 l

Apelin Adipocyte Signaling Pathway - LPS/IL-1 Mediated Inhibition of RXR Function 0 Prostate Cancer Signaling

-2 -1 0 1 2 0.0 0.5 1.0 ld 1.5 2.0 2.5 0 5 o . . h 4 3 h - - s -log(p-value) re h log2 (fold change) T

Fig S10