BBC Russian
Svoboda | Graniru | BBC Russia | Golosameriki | Facebook

This website requires cookies, and the limited processing of your personal data in order to function. By using the site you are agreeing to this as outlined in our privacy notice and cookie policy.

Abstract 


We recently described myonectin (also known as erythroferrone) as a novel skeletal muscle-derived myokine with metabolic functions. Here, we use a genetic mouse model to determine myonectin's requirement for metabolic homeostasis. Female myonectin-deficient mice had larger gonadal fat pads and developed mild insulin resistance when fed a high-fat diet (HFD) and had reduced food intake during refeeding after an unfed period but were otherwise indistinguishable from wild-type littermates. Male mice lacking myonectin, however, had reduced physical activity when fed ad libitum and in the postprandial state but not during the unfed period. When stressed with an HFD, myonectin-knockout male mice had significantly elevated VLDL-triglyceride (TG) and strikingly impaired lipid clearance from circulation following an oral lipid load. Fat distribution between adipose and liver was also altered in myonectin-deficient male mice fed an HFD. Greater fat storage resulted in significantly enlarged adipocytes and was associated with increased postprandial lipoprotein lipase activity in adipose tissue. Parallel to this was a striking reduction in liver steatosis due to significantly reduced TG accumulation. Liver metabolite profiling revealed additional significant changes in bile acids and 1-carbon metabolism pathways. Combined, our data affirm the physiologic importance of myonectin in regulating local and systemic lipid metabolism.-Little, H. C., Rodriguez, S., Lei, X., Tan, S. Y., Stewart, A. N., Sahagun, A., Sarver, D. C., Wong, G. W. Myonectin deletion promotes adipose fat storage and reduces liver steatosis.

Free full text 


Logo of fasebjClick here to go to the full-featured web site.Click here to learn how to submit an article to The FASEB Journal.Never miss an issue. Click here to subscribe to The FASEB Journal s full content.Click here for the latest news from The FASEB Journal.Find out what s being published every day. Click here to subscribe to RSS feeds.Click here to visit The FASEB Journal's full-featured site.
FASEB J. 2019 Jul; 33(7): 8666–8687.
Published online 2019 Apr 19. https://doi.org/10.1096/fj.201900520R
PMCID: PMC6593887
PMID: 31002535

Myonectin deletion promotes adipose fat storage and reduces liver steatosis

Abstract

We recently described myonectin (also known as erythroferrone) as a novel skeletal muscle–derived myokine with metabolic functions. Here, we use a genetic mouse model to determine myonectin’s requirement for metabolic homeostasis. Female myonectin-deficient mice had larger gonadal fat pads and developed mild insulin resistance when fed a high-fat diet (HFD) and had reduced food intake during refeeding after an unfed period but were otherwise indistinguishable from wild-type littermates. Male mice lacking myonectin, however, had reduced physical activity when fed ad libitum and in the postprandial state but not during the unfed period. When stressed with an HFD, myonectin-knockout male mice had significantly elevated VLDL–triglyceride (TG) and strikingly impaired lipid clearance from circulation following an oral lipid load. Fat distribution between adipose and liver was also altered in myonectin-deficient male mice fed an HFD. Greater fat storage resulted in significantly enlarged adipocytes and was associated with increased postprandial lipoprotein lipase activity in adipose tissue. Parallel to this was a striking reduction in liver steatosis due to significantly reduced TG accumulation. Liver metabolite profiling revealed additional significant changes in bile acids and 1-carbon metabolism pathways. Combined, our data affirm the physiologic importance of myonectin in regulating local and systemic lipid metabolism.—Little, H. C., Rodriguez, S., Lei, X., Tan, S. Y., Stewart, A. N., Sahagun, A., Sarver, D. C., Wong, G. W. Myonectin deletion promotes adipose fat storage and reduces liver steatosis.

Keywords: myokine, lipid metabolism, carbohydrate metabolism, obesity, diabetes

Skeletal muscle takes up the majority of circulating postprandial glucose and plays a vital role in maintaining whole-body energy balance (1, 2). Impaired insulin responsiveness in skeletal muscle is mechanistically linked to type 2 diabetes (3, 4). As a major metabolic organ, skeletal muscle is believed to communicate with other organs and tissues via secreted hormones to influence whole-body metabolism. The discovery that skeletal muscle dynamically secretes a variety of myokines (proteins that stimulate muscle and nonmuscle tissues to regulate various biologic processes) has provided a novel and critical conceptual framework to understand skeletal muscle’s role in coordinating integrated physiology (5, 6).

Of the hundreds of proteins secreted by skeletal muscle (79), only a few have been functionally characterized, including myostatin (1012), IL-6 (13), fibroblast growth factor 21 (14, 15), insulin-like 6 (16), follistatin-like 1 (17), leukemia inhibitory factor (18), IL-7 (19), IL-15 (20), musclin (21, 22), and irisin (23). These myokines either act locally within skeletal muscle as autocrine and paracrine factors or circulate in blood as endocrine factors, linking skeletal muscle to the regulation of physiologic processes in nonmuscle tissues (6, 11, 13, 14, 2327).

We recently described myonectin [complement component 1q/TNF-related protein (CTRP) 15] as a novel myokine expressed predominantly by skeletal muscle (28) and determined that exercise can up-regulate its expression and circulating level (28, 29). Myonectin is a member of the CTRP family with a signature C-terminal globular complement component 1q domain (28, 3035). Since our initial identification and characterizations of myonectin (28, 36), it has also been referred to as erythroferrone, a secreted protein induced in erythroblasts that links stress erythropoiesis to iron mobilization in the liver in response to blood loss (37, 38).

Using genetic gain- or loss-of-function mouse models, we and others provided in vivo evidence for the important metabolic and cardiovascular functions of several CTRP family members (29, 33, 3959). Unlike other family members, myonectin is the only CTRP whose basal expression is primarily restricted to skeletal muscle. Several lines of evidence suggest that myonectin is a physiologically relevant metabolic regulator (28, 36). First, overnight food removal reduces and refeeding increases myonectin mRNA and serum levels. Second, and similar to refeeding, a bolus of glucose or emulsified lipid administered to overnight-unfed mice increases circulating myonectin (28). The addition of glucose, amino acids, or free fatty acids to cultured myotubes also up-regulates myonectin expression, suggesting that nutrient uptake and metabolism by muscle cells is sufficient to induce production of this protein. Third, the expression and circulating levels of myonectin are significantly reduced in diet-induced obese and insulin-resistant mice (28). Fourth, infusion of recombinant myonectin into mice reduces circulating free fatty acid levels by promoting free fatty acid uptake (28). Fifth, myonectin suppresses liver autophagy (36), an intracellular recycling pathway activated in the unfed state and inhibited in the fed state. Finally, the circulating level of myonectin is associated with insulin resistance and type 2 diabetes in humans (60, 61). The caveat of our previous functional studies, however, is the reliance on recombinant protein infusion in mice and the use of established muscle and adipocyte cell lines (28, 36); it is unclear whether myonectin (erythroferrone) is required for metabolic homeostasis in a physiologic context (62). Therefore, we aimed to determine the physiologic function of myonectin using a genetic loss-of-function mouse model. Our data provide evidence that myonectin does indeed have a role in regulating lipid metabolism in the context of metabolic stress induced by high-fat feeding, and that this effect is sex-dependent.

MATERIALS AND METHODS

Animals

Myonectin (Erfe/Fam132b+/−) heterozygous mice were obtained from the Mutant Mouse Regional Resource Center at University of California–Davis (Davis, CA, USA) (strain B6; 129S5-Fam132btm1Lex/Mmucd, identifier: MMRRC:032289-UCD). Mice were generated by Lexicon Pharmaceuticals (The Woodlands, TX, USA) (63). We maintained the mice on the C57BL/6N genetic background for >6 generations. PCR genotyping strategy was provided by Mutant Mouse Regional Resource Center. Genotyping primers for the myonectin wild-type (WT) allele were: forward, 5′-GTCAGCCTTACCTGCCCAG-3′ and reverse, 5′-GACGTGAATCTC AGTCTGGC-3′, yielding a WT PCR product of 216 bp. Primers for the knockout (KO) allele were: forward, 5′-GCAGCGCATCGCCTTCTATC-3′ and reverse, 5′-GACCGTCACTGAGGTTCCAC-3′, yielding a KO PCR product of 390 bp. To confirm loss of myonectin mRNA from KO mice, quantitative PCR was performed on muscle samples using 2 independent sets of myonectin-specific primers: Primer set 1: forward, 5′-TGCTTGGATGCTGTTCGTCAA-3′ and reverse, 5′-CAGATGGGATAAAGGGGCCTG-3′; primer set 2: forward, 5′-GCGAGAGAGCCATCTGGAGCACTG-3′ and reverse, 5′-GGTCCCTTTCTTGGCTGCTCGGTG-3′. All mice were housed in polycarbonate cages with ad libitum access to water and food in a temperature-controlled room with 12-h light/dark cycles. Mice were fed either a high-fat diet (HFD) (60% calories from fat, D12492; Research Diets, New Brunswick, NJ, USA) or a matched control low-fat diet (LFD) (10% calories from fat, D12450B; Research Diets) throughout the study beginning at 5–6 wk of age. Body weights of WT and myonectin-KO mice were measured weekly. At the end of the study, mice were euthanized 2 h after food removal in the morning or after an overnight unfed period followed by 2 h of ad libitum refeeding. Tissues were dissected, weighed, and snap-frozen in liquid nitrogen for RNA and protein extraction or prepared for histology. Additional tissues were stored at −80°C. All animal experiments were approved by the Animal Care and Use Committee of the Johns Hopkins University School of Medicine.

Mouse body composition analysis

Body composition of myonectin-KO mice and WT littermates was determined using a quantitative NMR instrument (Echo-MRI-100; Echo Medical Systems, Houston, TX, USA) at the Johns Hopkins University School of Medicine mouse phenotyping core facility. Echo-MRI analyses measured total fat mass, lean mass, and water content.

Indirect calorimetry

Indirect calorimetry was performed as previously described by Petersen et al. (54). In brief, WT and myonectin-KO mice were used for simultaneous assessments of daily body weight change, food intake (corrected for spillage), physical activity, and whole-body metabolic profile in an open-flow indirect calorimeter (Comprehensive Lab Animal Monitoring System; Columbus Instruments, Columbus, OH, USA). Data were collected for 3–4 consecutive d to confirm that mice were acclimated to calorimetry chambers (indicated by stable body weights, food intakes, and diurnal metabolic patterns), and data from the following days were analyzed. Rates of Vo2 and Vco2 in each chamber were measured throughout the studies. Respiratory exchange ratio (RER; Vco2/Vo2) was calculated by Comprehensive Lab Animal Monitoring System software (v.4.02) to estimate relative oxidation of carbohydrates (RER = 1.0) vs. fats (RER = 0.7), not accounting for protein oxidation. Energy expenditure (EE) was calculated as EE = Vo2 × [3.815 + (1.232 × RER)]. Vo2, Vco2, and EE data were normalized to lean mass. Physical activity was measured by infrared beam breaks in the metabolic chamber. Mean metabolic values were calculated per subject and averaged across subjects for statistical analysis.

Mouse serum and blood chemistry analysis

Blood samples were collected by tail bleeds using capillary blood collection tubes (Microvette CB 300; Sarstedt, Nümbrecht, Germany). Blood samples were allowed to clot on ice and then centrifuged for 10 min at 10,000 g to collect serum. Serum triglyceride (TG) and cholesterol levels were measured using an Infinity kit (Thermo Fisher Scientific, Waltham, MA, USA). Nonesterified free fatty acids (NEFAs) were measured using a NEFA-HR (2) kit (Fujifilm, Tokyo, Japan). β-Hydroxybutyrate was measured using a LiquiColor assay (EKF Diagnostics, Cardiff, United Kingdom). Assays were performed according to the manufacturer’s protocol.

ELISA

Insulin (MilliporeSigma, Burlington, MA, USA), angiopoietin-like protein (ANGPTL) 3 (Thermo Fisher Scientific), and ANGPTL4 (Aviva Systems Biology, San Diego, CA, USA) levels were measured in serum according to the manufacturer’s protocol.

Glucose and insulin tolerance tests

For glucose tolerance tests, mice were unfed overnight (~15 h) and subsequently intraperitoneally injected or orally gavaged with a glucose solution in saline at a dose of 1 g glucose/kg body weight. Blood was collected from the tail vein into capillary blood collection tubes before injection and 15 min postinjection for assessment of insulin levels. Serum was isolated from blood and stored at −80°C until analysis. Tail vein blood glucose levels at the indicated time points were monitored with a glucometer (Nova Max Plus, Nova Biomedical, Waltham, MA, USA). For insulin tolerance tests (ITTs), mice were intraperitoneally injected with recombinant human insulin (Thermo Fisher Scientific) diluted in saline after a 2-h unfed period at a dose of 1–1.5 U insulin/kg body weight. Food was removed between 9:00 am and 10:00 am. Tail vein blood glucose was monitored with a glucometer at the indicated time points.

Lipid tolerance test

After an overnight unfed period (~15 h), mice were orally gavaged with 20% emulsified intralipid (soybean oil; MilliporeSigma) at a dose of 10 μl/g body weight. Blood was collected from the tail vein into capillary blood collection tubes before gavage and 1, 2, 3, and 4 h postgavage. Serum was isolated from blood samples and assayed for TG and NEFAs using commercially available kits according to the manufacturer’s protocol.

Hepatic VLDL–TG secretion

Mice were unfed for 4 h, beginning 2 h into the light cycle. Poloxamer 407 (Merck, Darmstadt, Germany) diluted in saline was intraperitoneally injected at a dose of 1 mg/g body weight to inhibit lipoprotein lipase (LPL) activity. Blood was collected from the tail vein into capillary blood collection tubes before injection and at 1, 2, 4, and 8 h postinjection. Serum was isolated from blood and assayed for TG using a colorimetric kit.

Intestinal lipid absorption and secretion

Overnight-unfed mice (~15 h) were intraperitoneally injected with poloxamer 407 diluted in saline at a dose of 1 mg/g body weight to inhibit LPL activity. After 1 h, mice were orally gavaged with 20% emulsified intralipid (10 μl/g body weight). Blood was collected from the tail vein into capillary blood collection tubes before gavage and at 1, 2, 3, and 4 h postgavage. Serum was isolated from blood samples and assayed for TG using a colorimetric kit.

Blood and tissue collection

Mice were anesthetized with isoflurane, and blood was collected from the retro-orbital plexus through heparin-coated capillary tubes (Thermo Fisher Scientific). Blood samples were allowed to clot on ice and were then centrifuged for 10 min at 10,000 g. Separated serum was divided into aliquots and stored at −80°C. Organs were excised, weighed, and flash-frozen in liquid nitrogen before being stored at −80°C.

Complete blood count analysis

A complete blood count on blood samples was performed at the Pathology Phenotyping Core at the Johns Hopkins University School of Medicine. Blood samples were collected from the femoral vein using EDTA-coated blood collection tubes (Sarstedt) and analyzed using Procyte Dx analyzer (Idexx Laboratories, Westbrook, ME, USA).

Serum lipoprotein analysis

Lipoprotein analyses were performed at the Metabolism core at the Baylor College of Medicine (Houston, TX, USA). Serum (300 μl) was pooled from 10 mice of each genotype (30 μl/mouse). Pooled serum samples from WT and KO mice were subjected to fractionation using FPLC followed by quantification of TG and cholesterol levels in each fraction.

Heparin releasable LPL activity in serum

Mice were unfed overnight (~13 h) and then given free access to food for 2 h. The experiment was conducted as previously described by Qiao et al. (64). Mice were briefly anesthetized with isoflurane and retro-orbitally injected with heparin sodium salt (MilliporeSigma) diluted in saline at a dose of 300 U/kg body weight. Blood was collected from the tail vein into capillary blood collection tubes before injection and 5 min postinjection. Serum was isolated from blood samples. Preheparin serum was used to measure serum TG levels. Total lipase activity was measured using a commercial kit (BioVision, Milpitas, CA, USA) in postheparin serum.

Mouse histology

Adipose and liver tissues from littermate-matched WT and myonectin-KO mice were fixed in 10% formalin at 4°C overnight. Fixed tissues were embedded in paraffin, sectioned, and stained with hematoxylin and eosin (H&E) at the Histology Reference Laboratory at the Johns Hopkins University School of Medicine.

Adipocyte cell size analysis

Adipocyte size was measured using the MRI Adipocyte Tools macro in the ImageJ software [National Institutes of Health (NIH), Bethesda, MD, USA]. All cells in 1 field of view at ×100 magnification per tissue section per mouse were analyzed from a total of 5 WT and 5 KO littermate-matched mice for visceral (gonadal) white adipose tissue (WAT) and 4 WT and 5 KO littermate-matched mice for subcutaneous inguinal WAT (iWAT). On average, 230 cells were measured for each sample.

Extraction and quantification of mouse hepatic lipid contents

A total of 50–80 mg of frozen liver tissue was homogenized in 500 μl ultra-pure water. A total of 200 μl lysate was transferred to a new tube, and 1 ml of 2:1 chloroform:methanol was added. After thorough mixing, samples were spun at 1700 rpm for 5 min at 4°C. The lower chloroform phase was transferred to a new tube and dried in a speed vacuum. Extracted lipids were resuspended in 5% Triton X-100 in saline. Samples were assayed for TG and cholesterol using commercially available colorimetric kits (Thermo Fisher Scientific). Protein content was measured in the original water lysate using a bicinchoninic acid assay (Thermo Fisher Scientific), and lipid levels were normalized to total protein.

Western blotting

Frozen tissue samples were homogenized in RIPA buffer (50 mM Tris-HCl, pH 7.4; 150 mM NaCl; 1 mM EDTA; 1% Triton X-100; 0.25% deoxycholate) supplemented with a protease inhibitor cocktail (MilliporeSigma) and a phosphatase inhibitor cocktail (Roche, Basel, Switzerland). Protein content was measured using a bicinchoninic acid assay. Samples for Western blotting were diluted in SDS loading dye (final concentration: 50 mM Tris-HCl, pH 7.4, 2% SDS, 6% glycerol, 1% 2-ME, and 0.01% bromphenol blue) and denatured at 95°C for 10 min. A total of 10–20 μg total protein was loaded into each lane of a 10% polyacrylamide gel (Bio-Rad, Hercules, CA, USA) and separated by electrophoresis. The Bio-Rad Trans-Blot Turbo semidry system was used to transfer protein onto PVDF membranes. Blots were blocked in 5% nonfat milk in PBS with Tween 20 (PBST) and exposed to primary antibodies overnight at 4°C. After washing, blots were exposed to secondary antibodies conjugated to horseradish peroxidase (HRP) for 1 h and developed in ECL (Amersham ECL Select; GE Healthcare, Chicago, IL, USA). Bands were visualized with MultiImage III FluorChem Q (Alpha Innotech, San Leandro, CA, USA) and quantified using Alphaview Software (Alpha Innotech). All primary antibodies were diluted 1:1000 in PBST + 0.02% sodium azide, and all secondary antibodies were diluted 1:5000 in 5% nonfat milk in PBST. Antibodies used: LPL (GTX101125; GeneTex, Irvine, CA, USA), heat shock cognate 71 kDa protein (Hsc70) (sc-7298; Santa Cruz Biotechnology, Dallas, TX, USA), perilipin 2 (PA1-16972; Thermo Fisher Scientific), mouse IgG-HRP (7076; Cell Signaling Technology, Danvers, MA, USA), and rabbit IgG-HRP (7074; Cell Signaling Technology).

Liver metabolomics analysis

Metabolite quantifications of WT (n = 10) and myonectin-KO (n = 10) liver samples were performed by Metabolon (Morrisville, NC, USA) using the metabolomics platform. Sample processing, compound identification by tandem mass spectrometry, metabolite quantification and data normalization, and data extraction and curation were all performed according to the manufacturer’s protocol.

Stimulated adipose tissue lipolysis

Mice were unfed for 5 h beginning at 9:30 am. The β3-adrenergic receptor agonist, CL 316,243 (Santa Cruz Biotechnology), was diluted in saline and intraperitoneally injected at a dose of 1 mg/kg body weight. Blood was collected from the tail vein into capillary blood collection tubes before injection and at 15 min postinjection. Blood glucose was monitored with a glucometer before and 15 min after CL 316,243 injection. Serum was separated from blood and assayed for NEFAs and glycerol using calorimetric kits (Fujifilm and MilliporeSigma, respectively).

Tissue LPL activity

Tissue LPL activity was measured as previously described by Mizunoya et al. (65). Briefly, frozen tissue samples were pulverized in liquid nitrogen using a mortar and pestle. A total of ~50 mg pulverized tissue was incubated in Krebs-Ringer solution containing bovine serum albumin and heparin. After clarifying, the supernatant was used to measure LPL activity using a commercial kit (Roar Biomedical, New York, NY, USA). Activity was normalized to tissue weight.

Real-time quantitative PCR analysis

RNA was isolated from tissues using Trizol reagent (Thermo Fisher Scientific) according to the manufacturer’s protocol and treated with DNAse I (New England Biolabs, Ipswich, MA, USA) to remove genomic DNA. For each sample, 1–2 μg total RNA was reverse transcribed using random primers and the GoScript reverse transcription system from Promega (Madison, WI, USA). Real-time quantitative PCR was performed on a CFX connect system using iTaq Universal SYBR Green PCR master mix (Bio-Rad). Gene expression was first normalized to 36b4 (also known as acidic ribosomal phosphoprotein P0) to generate a ΔCt value, and ΔΔCt was obtained by normalizing data to mean ΔCt of the control group (66). Information on primers used are available upon request.

Maximal exercise capacity

Maximal sprint and endurance exercise tests were performed as we previously described (67). Briefly, 12-wk-old mice fed a standard chow diet were first subjected to the endurance exercise test. Mice were acclimated to the treadmill for 3 d before the experiment, and the exercise test was carried out on the fourth day. Blood glucose and lactate values were measured before and after exercise using glucose and lactate meters (Nova Biomedical). Blood was collected from the tail vein into capillary blood collection tubes before and after exercise, and silver nitrate sticks were used to cauterize the wound. Mice were run in multiple groups, beginning at 9:30 am until exhaustion. After 2.5 wk of recovery, mice were subjected to the sprint exercise test. At 1 d before the experiment, mice were reacclimated to the treadmill using the same protocol as the original d-3 acclimation. For the sprint exercise experiment, mice were run in multiple groups, beginning at 8:30 am until exhaustion. As with the endurance test, blood, glucose, and lactate were collected before and after exercise. The experimenter was blinded with respect to genotype during all exercise tests.

Recovery after endurance exercise

Mice used in this experiment were fed a standard chow diet. Mice were acclimated to the treadmill as previously described, but with the addition of a fourth acclimation day in which mice ran on the treadmill at 14 m/min for 5 min at a 10° incline (67). The endurance exercise run was performed on the fifth day, when the mice were 11 wk of age. Mice were unfed for 2 h in the morning before exercise. The run was conducted as previously described by Leick et al. (68): 60 min at a speed of 14 m/min at a 10° incline. A shock grid at the back of the treadmill encouraged mice to continue running by administering a mild shock each time they stepped off the belt. If any mice met the predetermined criteria for exhaustion before the end of the run, they were removed from the treadmill and their data were omitted from analysis (67). The experimenter was blinded with respect to genotype during the treadmill run. Blood glucose and lactate values were measured at the indicated time points using glucose and lactate meters (Nova Biomedical). A single cohort of mice received a glucose gavage within 5 min of the end of the run (0.5 mg glucose/g body weight dissolved in saline). All mice were allowed to recover in their cages with free access to water but no food. After 2 h of recovery following the end of the run, mice were euthanized. Blood and tissues were collected as described above.

Tissue glycogen measurement

Tissue glycogen was measured using a protocol adapted from the NIH-funded Mouse Metabolic Phenotyping Centers (https://mmpc.org/shared/protocols.aspx). Frozen whole liver and gastrocnemius muscle were pulverized in liquid nitrogen. A total of ~25 mg liver tissue or 50 mg muscle were homogenized in 400 μl 0.9 N perchloric acid. Samples were clarified by centrifuging at 10,000 g for 10 min at 4°C and transferring the supernatant to a new tube. Supernatant (50 μl) was transferred to each of 2 reaction tubes, along with 25 μl 1 M potassium bicarbonate and 125 μl 0.4 M sodium acetate buffer (pH 4.8) with or without 5 mg/ml amyloglucosidase (MilliporeSigma). Reactions were incubated for 2 h at 40°C on a heat block. After 2 h, reactions were neutralized with NaOH and then vortexed and centrifuged for 3 min at 10,000 g at room temperature. A total of 20 μl of supernatant from each reaction was used to measure glucose using an Amplex Red glucose assay kit (Thermo Fisher Scientific) according to the manufacturer’s protocol. Amyloglucosidase hydrolyzes glycogen into free glucose monomers. For each tissue sample, the amount of glucose in the condition without amyloglucosidase (measuring free glucose) was subtracted from the amount of glucose in the condition with amyloglucosidase (measuring free glucose + glucose from glycogen) to yield the amount of glucose from glycogen only. This value was then normalized to tissue weight.

Statistical analysis

Comparisons between 2 groups of data were performed using 2-tailed Student’s t tests with Welch’s correction and 95% confidence intervals. For experiments with 2 independent variables, a 2-way ANOVA was conducted, and the Bonferroni method was used for multiple comparisons. Prism 7 software (GraphPad Software, La Jolla, CA, USA) was used to conduct analyses. Values were considered significant at P < 0.05. All data are presented as means ± sem.

RESULTS

Myonectin-deficient mouse model

We used a whole-body KO mouse model to assess the metabolic function of myonectin in vivo. To create a myonectin-null allele, exons 2 through 7 of the myonectin gene were replaced with a neomycin cassette (Fig. 1A). Using 1 set of primers that targeted the WT myonectin gene and another set that targeted the neomycin cassette, we could distinguish between WT, heterozygous, and homozygous-null mice (Fig. 1B). To verify that myonectin was not expressed in the KO mice, we measured myonectin mRNA levels in the skeletal muscle [the tissue with the highest expression in WT mice (28)] by quantitative PCR. Using 2 independent primer sets, we confirmed the absence of myonectin transcript (Fig. 1C). In the absence of a reliable antibody that can recognize endogenous myonectin without cross-reactivity, we did not quantify myonectin protein levels in WT and KO mice. Myonectin-KO mice were viable, fertile, born at the expected Mendelian ratio, and exhibited no gross developmental abnormalities.

An external file that holds a picture, illustration, etc.
Object name is fj.201900520Rf1.jpg

Myonectin-deficient mouse model. A) Schematic of the gene targeting strategy used to generate myonectin-KO mice. The majority of the myonectin gene (exons 2–7) was replaced with a neomycin resistance gene and lacZ reporter cassette. SA, splice acceptor; pA, polyadenylation signal. B) PCR genotyping showing successful production of WT (+/+), heterozygous (+/−), and homozygous KO (−/−) mice. C) The absence of myonectin transcript in the mouse gastrocnemius muscle was confirmed by quantitative PCR using 2 independent primer pairs. Primer set 1 targets exons 2 and 3, yielding a PCR product size of 143 bp. Primer set 2 targets exons 4 and 6, yielding a PCR product size of 381 bp. WT, n = 6; KO, n = 6 female mice. lacZ, β-galactosidase; loxP, locus of X-over in P1; Neg. Ctrl., negative control; neo, neomycin resistant.

Myonectin is not required for the physiologic response to exercise

Under basal conditions, myonectin is primarily produced in skeletal muscle, and its expression can be further induced by exercise (28, 29). We therefore first tested whether it is required for the physiologic response to exercise. During both maximal sprint and endurance exercise on a treadmill, loss of myonectin had no impact on total running distance and the postexercise serum levels of glucose, lactate, NEFA, TG, and ketone (β-hydroxybutyrate) in female WT and KO mice (Table 1). However, we did observe significantly higher pre-exercise serum ketones (β-hydroxybutyrate) in KO female mice relative to WT littermate controls. For male mice, with the exception of postsprint exercise serum TG levels that were significantly lower in the KO animals, no significant genotypic differences were noted in exercise parameters (Table 1). In the postexercise recovery phase (with or without glucose gavage postexercise), myonectin deficiency did not significantly affect the ability of KO animals to replenish their skeletal muscle or liver glycogen stores (Table 2). These results indicate that myonectin is not required for physiologic response and adaptation to an acute bout of exercise.

TABLE 1

Time to exhaustion, total distance run, and blood parameters during an endurance exercise capacity test or a sprint exercise capacity test in chow-fed male and female WT and myonectin-KO mice

ParameterFemale mice
Male mice
WTKOPWTKOP
Endurance exercise capacity
 N1415914
 Time to exhaustion (min)116.1 ± 5.253125.0 ± 4.3170.205299.1 ± 7.172101.5 ± 3.6940.7714
 Running distance (m)1770 ± 117.51968 ± 101.60.21351406 ± 141.21447 ± 75.80.8031
 Pre-exercise glucose (mg/dl)161.9 ± 4.655157.9 ± 3.9330.5253190.2 ± 6.357183.3 ± 3.7580.3640
 Postexercise glucose (mg/dl)148.5 ± 11.27128.9 ± 6.840.1508175.1 ± 17.39161.4 ± 8.850.4953
 Pre-exercise lactate (mM)2.000 ± 0.09891.873 ± 0.11970.42191.433 ± 0.14241.550 ± 0.17630.6120
 Postexercise lactate (mM)5.232 ± 0.31795.013 ± 0.35700.65085.300 ± 0.63884.779 ± 0.49600.5278
 Pre-exercise TG (mg/dl)56.75 ± 5.87250.17 ± 4.2730.374053.01 ± 7.72657.32 ± 6.1430.6675
 Postexercise TG (mg/dl)52.00 ± 2.62953.31 ± 2.4850.720237.13 ± 2.47243.26 ± 2.3630.0905
 Pre-exercise NEFA (mM)1.225 ± 0.09401.035 ± 0.06520.11131.196 ± 0.05431.172 ± 0.12840.8662
 Postexercise NEFA (mM)2.387 ± 0.17642.756 ± 0.14660.11961.633 ± 0.19792.020 ± 0.21820.2049
 Pre-exercise β-hydroxybutyrate (μM)122.4 ± 8.09*163.8 ± 17.07* 0.0405*139.2 ± 10.93133.0 ± 15.280.7445
 Postexercise β-hydroxybutyrate (μM)545.7 ± 47.10623.3 ± 52.920.2830377.0 ± 58.62370.2 ± 44.520.9278
Sprint exercise capacity
 N1515912
 Time to exhaustion (min)14.52 ± 0.473414.71 ± 0.58620.799912.90 ± 0.659012.97 ± 0.40130.9373
 Running distance (m)244.4 ± 13.20250.6 ± 17.110.7764200.8 ± 17.31201.2 ± 10.790.9851
 Pre-exercise glucose (mg/dl)169.6 ± 5.266176.1 ± 4.3920.3491180.8 ± 4.481186.1 ± 5.1400.4461
 Postexercise glucose (mg/dl)180.8 ± 3.558176.3 ± 5.0920.4788221.9 ± 8.093209.8 ± 5.0420.2219
 Pre-exercise lactate (mM)2.270 ± 0.27502.027 ± 0.15840.45121.711 ± 0.11841.917 ± 0.17530.3440
 Postexercise lactate (mM)11.02 ± 0.441910.29 ± 0.58220.32938.872 ± 1.1169.700 ± 0.8030.5559
 Pre-exercise TG (mg/dl)48.40 ± 4.33249.07 ± 4.9850.920359.03 ± 4.00754.92 ± 3.5920.4559
 Postexercise TG (mg/dl)46.93 ± 2.45848.55 ± 2.0330.615956.55 ± 3.604*45.31 ± 2.351* 0.0202*
 Pre-exercise NEFA (mM)1.083 ± 0.11900.856 ± 0.14530.23681.199 ± 0.09831.307 ± 0.13140.5187
 Postexercise NEFA (mM)1.012 ± 0.05260.919 ± 0.12310.49640.982 ± 0.10550.814 ± 0.08940.2403
 Pre-exercise β-hydroxybutyrate (μM)199.6 ± 13.41*244.9 ± 15.74* 0.0371*161.9 ± 21.62203.9 ± 33.470.3055
 Postexercise β-hydroxybutyrate (μM)224.0 ± 17.87239.6 ± 14.980.5103204.8 ± 24.29224.4 ± 20.160.5423

Mice were 12 wk of age for the endurance test and 14.5 wk of age for the sprint test. *P < 0.05.

TABLE 2

Chow-fed male and female WT and myonectin-KO mice (at 11 wk of age) were subjected to an endurance run on the treadmill, and blood glucose and lactate values were measured at the indicated time points

ParameterFemale mice
Male mice
WTKOPWTKOP
No glucose gavage postexercise
 N6556
 Body weight (g)18.23 ± 0.383618.10 ± 0.87010.893425.50 ± 0.766824.38 ± 0.76310.3293
 Liver weight (g)0.7950 ± 0.01930.7340 ± 0.03700.19281.076 ± 0.03671.040 ± 0.03570.4999
 Pre-exercise glucose (mg/dl)181.5 ± 5.30187.0 ± 10.700.6614211.8 ± 6.63201.0 ± 8.670.3487
 0 min postexercise glucose (mg/dl)184.5 ± 6.35191.6 ± 13.310.6478207.0 ± 13.02211.3 ± 11.130.8064
 30 min postexercise glucose (mg/dl)158.2 ± 7.85150.4 ± 11.600.5959159.0 ± 6.46158.0 ± 6.810.9175
 60 min postexercise glucose (mg/dl)144.7 ± 13.11154.8 ± 6.840.5139140.8 ± 10.96163.3 ± 5.900.1183
 120 min postexercise glucose (mg/dl)127.8 ± 5.88135.2 ± 5.080.3682145.4 ± 12.84154.2 ± 4.740.5496
 Pre-exercise lactate (mM)1.750 ± 0.10571.560 ± 0.09800.22002.400 ± 0.45722.033 ± 0.13330.4783
 0 min postexercise lactate (mM)3.950 ± 0.28144.300 ± 0.79870.69654.460 ± 0.17784.400 ± 0.39070.8928
 Muscle glycogen (nmol glucose from glycogen/mg tissue)9.75 ± 0.582410.53 ± 1.31980.611511.16 ± 1.08512.37 ± 1.9810.6097
 Liver glycogen (nmol glucose from glycogen/mg tissue)30.81 ± 7.692712.10 ± 0.88490.059167.37 ± 19.7078.24 ± 11.160.6470
With glucose gavage postexercise
 N5655
 Body weight (g)19.94 ± 0.344420.33 ± 0.88870.693224.12 ± 0.541724.86 ± 0.68230.4216
 Liver weight (g)0.8300 ± 0.03190.8417 ± 0.04640.84091.006 ± 0.02791.008 ± 0.03230.9638
 Pre-exercise glucose (mg/dl)162.2 ± 7.51160.7 ± 4.820.8684198.2 ± 7.37192.2 ± 5.210.5270
 0 min postexercise glucose (mg/dl)150.8 ± 17.89159.5 ± 8.310.6754197.4 ± 12.48192.0 ± 10.220.7467
 30 min postexercise glucose (mg/dl)137.6 ± 9.48153.2 ± 4.520.1906179.0 ± 2.19172.6 ± 8.350.4948
 60 min postexercise glucose (mg/dl)127.7 ± 6.74129.0 ± 2.000.8649156.0 ± 13.00154.7 ± 0.670.9350
 120 min postexercise glucose (mg/dl)103.8 ± 12.01105.5 ± 7.980.9094139.4 ± 7.90137.8 ± 4.420.8652
 Pre-exercise lactate (mM)1.580 ± 0.10681.517 ± 0.06010.62251.540 ± 0.08721.740 ± 0.15680.3060
 0 min postexercise lactate (mM)6.520 ± 1.37824.433 ± 0.76970.23162.800 ± 0.11404.600 ± 1.14060.1901
 Muscle glycogen (nmol glucose from glycogen/mg tissue)9.63 ± 0.576210.56 ± 0.27630.197612.15 ± 0.83512.91 ± 0.8360.5347
 Liver glycogen (nmol glucose from glycogen/mg tissue)27.91 ± 4.612144.47 ± 9.48560.159642.61 ± 17.0447.36 ± 11.020.8216

Half of the mice received a glucose gavage immediately after completing the run, and mice were allowed to recover for 2 h before euthanization.

Myonectin-KO mice fed a control LFD show no overt metabolic phenotypes

Given its potential role as a postprandial metabolic regulator (28, 36), we tested whether myonectin is necessary for metabolic homeostasis under basal conditions. Male and female WT and KO mice were fed a control LFD for 12 wk, beginning at 6 wk of age. Neither sex displayed genotypic differences in body weight over time or in body composition (Supplemental Fig. S1A, B). Organ weights (heart, kidney, spleen, liver, and adipose tissue) were also not different between genotypes of either sex (Supplemental Table S1). Indirect calorimetry revealed no differences in ad libitum food intake, metabolic rate, or EE between WT and KO mice of either sex (Table 3). However, ad libitum fed female KO mice had a higher RER in the dark cycle and lower physical activity in the light cycle. Similar to the female KO mice, ad libitum fed male KO mice also had significantly lower physical activity in the light cycle and over a 24-h period. Myonectin is robustly induced upon refeeding following an unfed period (36). We therefore challenged the WT and KO mice with a 24-h unfed period followed by 24 h of free access to food while they were in the metabolic cages. No genotypic differences in either sex were observed during the unfed period (Table 3). However, in the refeeding phase, female KO mice had lower RER in the light cycle, and male KO mice had significantly reduced physical activity in the dark cycle and over a 24-h period (Table 3). Because myonectin is also up-regulated after an acute bout of exercise (28), we provided the WT and KO mice with a voluntary running wheel in the metabolic cages in a separate experiment. No major genotypic differences in either sex were observed (Supplemental Table S2).

TABLE 3.

Indirect calorimetry, physical activity, and food intake analysis of male and female mice fed a control LFD

ParameterDark cycle
Light cycle
24 h
WTKOPWTKOPWTKOP
LFD (female)
Ad libitum
 N666666
 Body weight (g)23.69 ± 0.7123.67 ± 0.780.9816
 Food intake (kcal)11.34 ± 0.552513.30 ± 0.86160.09794.00 ± 0.30273.52 ± 0.48330.429315.34 ± 0.675716.98 ± 0.71560.1262
 Vo2 (ml/kg lean mass/h)6226 ± 360.156211 ± 266.790.97515078 ± 221.795153 ± 47.770.75625650 ± 239.435680 ± 131.950.9144
 Vco2 (ml/kg lean mass/h)5980 ± 288.226222 ± 229.130.52644337 ± 213.504453 ± 68.200.62195153 ± 193.945333 ± 91.150.4282
 RER (Vco2/Vo2)0.9632 ± 0.0100*1.0035 ± 0.0139* 0.0424*0.8533 ± 0.01190.8644 ± 0.01200.52420.9133 ± 0.00870.9398 ± 0.01010.0750
 EE (kcal/kg lean mass/h)31.12 ± 1.728531.36 ± 1.28950.912524.72 ± 1.101725.14 ± 0.23140.719427.90 ± 1.148528.24 ± 0.60700.8019
 Total activity (beam breaks)89,169 ± 22,95576,618 ± 13,1120.647727,651 ± 1674*20,527 ± 1654* 0.0128*116,819 ± 24,43897,145 ± 13,9920.5047
 Ambulatory activity (counts)62,096 ± 18,03551,144 ± 89780.602817,165 ± 1826*11,441 ± 1462* 0.0355*79,261 ± 19,21262,585 ± 94590.4607
Unfed
 N565656
 Body weight (g)20.68 ± 0.8420.50 ± 0.900.8808
 Food intake (kcal)000000
 Vo2 (ml/kg lean mass/h)5658 ± 165.675958 ± 312.730.42394020 ± 259.174275 ± 335.130.56104837 ± 208.505108 ± 296.490.4736
 Vco2 (ml/kg lean mass/h)4219 ± 141.644365 ± 201.450.56833000 ± 211.403130 ± 221.650.67983607 ± 172.873742 ± 196.150.6204
 RER (Vco2/Vo2)0.7453 ± 0.00740.7341 ± 0.00630.27910.7451 ± 0.00970.7342 ± 0.01180.49230.7453 ± 0.00780.7336 ± 0.00670.2872
 EE (kcal/kg lean mass/h)26.78 ± 0.800628.11 ± 1.44060.446719.03 ± 1.246120.17 ± 1.54780.581222.90 ± 1.004824.10 ± 1.37100.4981
 Total activity (beam breaks)96,004 ± 13,58092,982 ± 17,3400.893946,965 ± 11,93248,531 ± 22,6730.9529142,969 ± 22,646141,513 ± 35,9270.9735
 Ambulatory activity (counts)68,721 ± 947567,799 ± 12,9920.955632,873 ± 861034,541 ± 17,3170.9336101,594 ± 15,363102,340 ± 26,9040.9814
Refeed
 N565656
 Body weight (g)23.17 ± 1.0622.45 ± 0.790.6028
 Food intake (kcal)14.65 ± 0.773515.42 ± 2.06330.74325.50 ± 0.17795.13 ± 0.48250.504020.15 ± 0.910120.19 ± 1.69920.9856
 Vo2 (ml/kg lean mass/h)5347 ± 222.175673 ± 110.570.23764839 ± 148.654978 ± 104.460.46995102 ± 182.805340 ± 51.570.2694
 Vco2 (ml/kg lean mass/h)5408 ± 213.095666 ± 131.890.33735000 ± 148.674944 ± 140.850.79225211 ± 179.395321 ± 53.040.5836
 RER (Vco2/Vo2)1.0119 ± 0.00890.9986 ± 0.00870.31151.0338 ± 0.0145*0.9925 ± 0.0087* 0.0463*1.0219 ± 0.01100.9965 ± 0.00430.0825
 EE (kcal/kg lean mass/h)27.06 ± 1.105628.62 ± 0.57680.256224.62 ± 0.734825.08 ± 0.57100.635325.89 ± 0.910526.93 ± 0.25720.3237
 Total activity (beam breaks)38,270 ± 307843,979 ± 47780.343816,763 ± 146314,686 ± 15360.353255,032 ± 421858,665 ± 58620.6276
 Ambulatory activity (counts)23,748 ± 424027,082 ± 26750.52768759 ± 19787406 ± 8310.553832,507 ± 612434,487 ± 32670.7848
LFD (male)
Ad libitum
 N666666
 Body weight (g)33.78 ± 0.9332.27 ± 1.540.4267
 Food intake (kcal)12.10 ± 0.448312.19 ± 1.06830.94474.68 ± 0.52104.32 ± 0.28480.569216.78 ± 0.939316.51 ± 1.25160.8670
 Vo2 (ml/kg lean mass/h)5140 ± 336.814764 ± 122.470.33274507 ± 317.604266 ± 87.280.49254821 ± 327.534512 ± 93.370.3996
 Vco2 (ml/kg lean mass/h)4979 ± 325.834702 ± 169.580.47374037 ± 276.073855 ± 106.020.55944506 ± 300.884274 ± 130.210.5038
 RER (Vco2/Vo2)0.9691 ± 0.00990.9857 ± 0.01410.36070.8976 ± 0.01570.9032 ± 0.00930.76300.9355 ± 0.01200.9463 ± 0.01110.5245
 EE (kcal/kg lean mass/h)25.74 ± 1.683323.97 ± 0.67100.362022.17 ± 1.544721.02 ± 0.45940.504623.94 ± 1.616022.48 ± 0.51440.4205
 Total activity (beam breaks)31,086 ± 257325,107 ± 29270.156514,819 ± 1041*10,042 ± 1007* 0.0080*45,905 ± 2828*35,148 ± 3672* 0.0443*
 Ambulatory activity (counts)15,473 ± 190112,719 ± 18000.31766390 ± 9104123 ± 5280.063121,862 ± 230516,841 ± 21670.1436
Unfed
 N666666
 Body weight (g)30.40 ± 1.0029.26 ± 1.540.5519
 Food intake (kcal)000000
 Vo2 (ml/kg lean mass/h)4821 ± 351.504499 ± 137.610.42403773 ± 252.603497 ± 141.250.36834292 ± 299.083996 ± 125.830.3935
 Vco2 (ml/kg lean mass/h)3560 ± 273.933319 ± 101.560.44042715 ± 185.862508 ± 110.250.36503133 ± 227.982912 ± 97.030.4025
 RER (Vco2/Vo2)0.7376 ± 0.00520.7377 ± 0.00170.98390.7192 ± 0.00370.7165 ± 0.00580.71100.7294 ± 0.00370.7284 ± 0.00290.8459
 EE (kcal/kg lean mass/h)22.78 ± 1.677221.25 ± 0.64990.426817.74 ± 1.192316.43 ± 0.67300.367320.23 ± 1.421418.83 ± 0.59890.3950
 Total activity (beam breaks)46,032 ± 345939,156 ± 40270.225014,328 ± 152312,284 ± 12980.331760,360 ± 360751,439 ± 44030.1493
 Ambulatory activity (counts)26,838 ± 249222,431 ± 28430.27136571 ± 10265685 ± 8030.512733,409 ± 262528,116 ± 30970.2223
Refeed
 N666666
 Body weight (g)32.65 ± 1.0031.51 ± 1.230.4913
 Food intake (kcal)16.11 ± 0.783915.39 ± 0.75090.52294.01 ± 0.22763.91 ± 0.19320.738320.12 ± 0.905219.29 ± 0.82380.5175
 Vo2 (ml/kg lean mass/h)5289 ± 306.054745 ± 95.610.14134366 ± 278.174085 ± 89.060.37164848 ± 289.284430 ± 90.210.2167
 Vco2 (ml/kg lean mass/h)5092 ± 280.394618 ± 120.980.16574179 ± 242.683952 ± 104.510.42054655 ± 257.944300 ± 105.020.2446
 RER (Vco2/Vo2)0.9637 ± 0.00670.9725 ± 0.00880.44100.9595 ± 0.01550.9675 ± 0.01370.70860.9618 ± 0.00990.9704 ± 0.00890.5320
 EE (kcal/kg lean mass/h)26.45 ± 1.511723.79 ± 0.51010.145821.81 ± 1.353520.45 ± 0.45410.378724.23 ± 1.418522.20 ± 0.46740.2214
 Total activity (beam breaks)33,740 ± 2923*23,819 ± 1962* 0.0207*10,661 ± 9868470 ± 8910.130744,401 ± 3607*32,289 ± 2359* 0.0212*
 Ambulatory activity (counts)15,749 ± 188011,041 ± 10460.06084111 ± 4523088 ± 4490.139319,860 ± 2129*14,129 ± 1240* 0.0483*

At the time of the study, LFD-fed mice were 27 wk of age. LFD-fed mice were given 2 d to acclimate to the metabolic cages. Ad libitum data were collected on the third day. Mice were denied access to food but had free access to water on d 4 (unfed data), and refeed data were collected on d 5 when mice regained free access to food. *P < 0.05.

To assess whether myonectin deficiency alters carbohydrate metabolism, glucose tolerance tests (GTTs) were performed. The rate of glucose uptake in peripheral tissues in response to glucose injection was not different between genotypes of either sex (Supplemental Fig. S1C, D). The magnitude of insulin secretion in response to a rise in blood glucose during GTT did not differ between genotypes in males (Supplemental Fig. S1E). We also performed ITTs to assess insulin action in peripheral tissue. The rate of glucose disposal in response to a bolus of insulin did not differ between genotypes of either sex (Supplemental Fig. S1F, G). We previously showed that recombinant myonectin infusion promotes lipid uptake in mice (28). To test whether myonectin deficiency affects the ability of mice to handle an acute lipid load, we performed lipid tolerance tests. Overnight-unfed mice were orally gavaged with a bolus of emulsified intralipid, and serum lipid levels were measured over time to assess the rate of lipid uptake and clearance by peripheral tissues. No genotypic differences were observed in the rate of clearance of serum TG or NEFA (Supplemental Fig. S1H, I). Aside from physical activity in the ad libitum fed state or during refeeding, our data indicate that myonectin is dispensable for whole-body carbohydrate and lipid metabolism under basal conditions.

Minimal impact of myonectin deficiency on carbohydrate metabolism in the context of obesity

We next determined whether myonectin is required for a proper response to metabolic stress. A separate cohort of male and female WT and KO mice was challenged with an HFD, beginning at 5 wk of age, to induce obesity and insulin resistance. We performed the same sets of metabolic tests as in the control LFD-fed cohorts to determine whether myonectin deficiency exacerbates or improves the pathophysiological phenotypes associated with diet-induced obesity and insulin resistance. Body weight and body composition did not differ between genotypes of either sex (Fig. 2A, B). Despite no difference in overall fat mass, the gonadal fat pad was larger in myonectin-KO female mice relative to WT littermates (Supplemental Table S1). Indirect calorimetry analyses indicated no differences in metabolic rate, physical activity, or EE between genotypes of either sex under ad libitum fed conditions or during a 24-h unfed period and 24-h refeed period (Table 4). However, we noted that ad libitum fed female KO mice had significantly reduced food intake in the light cycle. In the refeeding phase following an unfed period, female KO mice had reduced food intake and lower RER in the dark cycle; a reduction in food intake was also observed over the 24-h refeed period. In contrast to female mice, the only difference seen in KO male mice was reduced RER in the light cycle in the ad libitum fed state (Table 4). Myonectin-deficient female mice also showed a modest impairment in the rate of glucose uptake in peripheral tissues in response to glucose or insulin injection (Fig. 2C, F). Myonectin-deficient male mice, however, were indistinguishable from WT littermates in GTTs and ITTs (Fig. 2D, G). The magnitude of insulin secretion in response to glucose injection during the GTT did not differ between genotypes in males (Fig. 2E). Together, these results indicate that myonectin deficiency has minimal impact on whole-body carbohydrate metabolism in the pathophysiological context of diet-induced obesity.

An external file that holds a picture, illustration, etc.
Object name is fj.201900520Rf2.jpg

There are no overt carbohydrate metabolism phenotypes in the myonectin-KO mice fed an HFD. A) Body weights of female (WT, n = 8; KO, n = 8) and male (WT, n = 12–13; KO, n = 16) mice over time. Mice were weaned at 3.5 wk of age onto a standard chow diet. At 5 wk of age, the diet was switched to an HFD. B) Body composition analysis of female (WT, n = 7; KO, n = 8) and male (WT, n = 12; KO, n = 16) mice at 28 wk of age. C) Blood glucose levels during intraperitoneal GTTs in 18-wk-old female mice (WT, n = 8; KO, n = 8). D) Blood glucose levels during oral GTT in 19-wk-old male mice (WT, n = 12; KO, n = 12). E) Serum insulin levels in male mice at 0 min before glucose gavage (WT, n = 11; KO, n = 9) and at 15 min after glucose gavage (WT, n = 12; KO, n = 11); for some samples, insulin levels were below the threshold of detection. F, G) Blood glucose levels during ITTs in female mice (WT, n = 7; KO, n = 8) and male mice (WT, n = 11; KO, n = 15) at 23 wk of age. Insulin was injected at a dose of 1.2 U/kg body weight for female mice and 1.5 U/kg body weight for male mice. *P < 0.05.

TABLE 4.

Indirect calorimetry, physical activity, and food intake analysis of male and female mice fed an HFD

ParameterDark cycle
Light cycle
24 h
WTKOPWTKOPWTKOP
HFD (female)
Ad libitum
 N666666
 Body weight (g)39.26 ± 2.1639.25 ± 2.030.9978
 Food intake (kcal)7.78 ± 0.9366.02 ± 1.1330.26292.77 ± 0.278*1.70 ± 0.346*0.0419*10.55 ± 1.1187.71 ± 1.4040.1522
 Vo2 (ml/kg lean mass/h)5829 ± 241.715891 ± 32.680.80865275 ± 214.595244 ± 153.420.90725550 ± 225.095567 ± 74.910.9464
 Vco2 (ml/kg lean mass/h)4417 ± 174.414366 ± 30.950.78213981 ± 126.933886 ± 103.030.57564198 ± 147.354126 ± 52.170.6601
 RER (Vco2/Vo2)0.7584 ± 0.01050.7412 ± 0.00700.20640.7561 ± 0.00820.7415 ± 0.00460.15910.7573 ± 0.00880.7412 ± 0.00550.1582
 EE (kcal/kg lean mass/h)27.68 ± 1.128527.85 ± 0.12660.884025.03 ± 0.973524.79 ± 0.70970.848226.35 ± 1.035526.32 ± 0.34090.9822
 Total activity (beam breaks)26,652 ± 504230,357 ± 45710.598313,739 ± 284914,363 ± 25500.873640,391 ± 766544,720 ± 62270.6709
 Ambulatory activity (counts)12,799 ± 246615,179 ± 22890.49565513 ± 12555936 ± 10650.802918,313 ± 354921,115 ± 24850.5340
Unfed
 N666666
 Body weight (g)36.85 ± 2.1237.01 ± 1.990.9586
 Food intake (kcal)000000
 Vo2 (ml/kg lean mass/h)5643 ± 221.605644 ± 101.860.99704585 ± 230.514547 ± 157.970.89535115 ± 212.535091 ± 126.550.9234
 Vco2 (ml/kg lean mass/h)4027 ± 144.583996 ± 70.730.85083286 ± 154.033245 ± 106.040.83123657 ± 140.153617 ± 85.750.8125
 RER (Vco2/Vo2)0.7142 ± 0.00330.7080 ± 0.00190.14120.7174 ± 0.00330.7139 ± 0.00230.41370.7155 ± 0.00290.7107 ± 0.00180.1953
 EE (kcal/kg lean mass/h)26.49 ± 1.023126.46 ± 0.47500.976121.54 ± 1.069021.35 ± 0.73290.883824.02 ± 0.983223.88 ± 0.58800.9035
 Total activity (beam breaks)35,897 ± 604540,484 ± 53370.582212,202 ± 274612,706 ± 24950.894748,099 ± 847353,190 ± 74250.6612
 Ambulatory activity (counts)19,678 ± 337622,210 ± 29240.58345290 ± 12975383 ± 11430.958324,968 ± 442627,593 ± 36700.6580
Refeed
 N666666
 Body weight (g)37.79 ± 2.0637.57 ± 1.980.9405
 Food intake (kcal)11.11 ± 0.135*9.48 ± 0.561*0.0423*3.44 ± 0.3862.90 ± 0.3120.307014.55 ± 0.4454*12.39 ± 0.5515*0.0155*
 Vo2 (ml/kg lean mass/h)5914 ± 239.536071 ± 125.660.57725201 ± 254.855187 ± 174.100.96575552 ± 239.425624 ± 145.890.8049
 Vco2 (ml/kg lean mass/h)4506 ± 157.984526 ± 81.210.91223984 ± 165.083904 ± 109.040.69574241 ± 155.714211 ± 92.230.8723
 RER (Vco2/Vo2)0.7629 ± 0.0054*0.7458 ± 0.0040*0.0311*0.7675 ± 0.00740.7534 ± 0.00530.15360.7650 ± 0.00600.7493 ± 0.00440.0629
 EE (kcal/kg lean mass/h)28.11 ± 1.107428.74 ± 0.57720.630524.75 ± 1.174624.60 ± 0.79740.917926.41 ± 1.104526.64 ± 0.66870.8595
 Total activity (beam breaks)31,069 ± 806035,028 ± 58330.699912,508 ± 268011,956 ± 20660.873943,577 ± 10,66146,984 ± 78390.8025
 Ambulatory activity (counts)15,373 ± 464417,250 ± 27220.73635174 ± 14184881 ± 8520.863620,547 ± 603622,131 ± 35470.8267
HFD (male)
Ad libitum
 N666666
 Body weight (g)44.71 ± 1.2146.41 ± 2.740.5898
 Food intake (kcal)11.05 ± 0.48510.92 ± 0.6740.88203.69 ± 0.4152.89 ± 0.3260.168014.73 ± 0.506513.81 ± 0.79650.3621
 Vo2 (ml/kg lean mass/h)4896 ± 178.315268 ± 157.820.14944340 ± 179.534720 ± 163.000.14854615 ± 175.504992 ± 157.970.1426
 Vco2 (ml/kg lean mass/h)3803 ± 136.514071 ± 120.760.17293339 ± 131.943561 ± 122.980.24693569 ± 129.953814 ± 120.270.1971
 RER (Vco2/Vo2)0.7769 ± 0.00440.7728 ± 0.00310.46420.7697 ± 0.0034*0.7545 ± 0.0041*0.0181*0.7735 ± 0.00310.7641 ± 0.00320.0600
 EE (kcal/kg lean mass/h)23.36 ± 0.846825.11 ± 0.74970.153220.67 ± 0.846822.39 ± 0.77190.163822.00 ± 0.829023.74 ± 0.74980.1516
 Total activity (beam breaks)19,968 ± 223318,985 ± 18790.743410,056 ± 4469179 ± 10610.472130,024 ± 239228,164 ± 26930.6170
 Ambulatory activity (counts)8471 ± 15497870 ± 12800.77133449 ± 3262999 ± 6140.536411,920 ± 176810,869 ± 18100.6866
Unfed
 N656565
 Body weight (g)41.54 ± 1.2144.41 ± 2.820.3888
 Food intake (kcal)000000
 Vo2 (ml/kg lean mass/h)4666 ± 216.334867 ± 194.460.50553693 ± 133.103930 ± 159.850.28694177 ± 170.754396 ± 162.820.3770
 Vco2 (ml/kg lean mass/h)3340 ± 147.613470 ± 137.690.53482659 ± 83.472806 ± 113.430.33082998 ± 112.113136 ± 113.670.4094
 RER (Vco2/Vo2)0.7162 ± 0.00270.7130 ± 0.00250.40040.7207 ± 0.00410.7139 ± 0.00210.18420.7182 ± 0.00330.7134 ± 0.00200.2482
 EE (kcal/kg lean mass/h)21.91 ± 1.006822.84 ± 0.91090.510717.37 ± 0.610218.45 ± 0.74930.294119.63 ± 0.789220.63 ± 0.76090.3825
 Total activity (beam breaks)17,821 ± 173122,326 ± 31950.25956245 ± 4597229 ± 8940.365424,066 ± 204629,555 ± 38260.2512
 Ambulatory activity (counts)7579 ± 89010,425 ± 23600.30901672 ± 2152318 ± 5860.34669250 ± 101912,743 ± 28670.3028
Refeed
 N656565
 Body weight (g)42.57 ± 1.1545.43 ± 2.830.3890
 Food intake (kcal)11.14 ± 0.97710.66 ± 0.8980.72284.42 ± 0.3183.28 ± 0.4350.069015.56 ± 1.10813.94 ± 0.85410.2759
 Vo2 (ml/kg lean mass/h)4992 ± 136.645113 ± 150.940.56824295 ± 129.024374 ± 122.970.66564639 ± 128.564738 ± 134.790.6089
 Vco2 (ml/kg lean mass/h)3764 ± 83.213849 ± 107.150.54543329 ± 75.073332 ± 89.300.97953544 ± 76.373587 ± 95.730.7344
 RER (Vco2/Vo2)0.7546 ± 0.00660.7531 ± 0.00310.84060.7761 ± 0.00790.7618 ± 0.00320.14060.7646 ± 0.00690.7572 ± 0.00270.3527
 EE (kcal/kg lean mass/h)23.68 ± 0.620424.25 ± 0.70670.562920.48 ± 0.581920.79 ± 0.57820.716622.07 ± 0.581722.50 ± 0.63140.6292
 Total activity (beam breaks)26,584 ± 507119,906 ± 11660.25068855 ± 13948000 ± 7020.600035,440 ± 637227,905 ± 17260.2993
 Ambulatory activity (counts)12,476 ± 30908073 ± 8460.22063173 ± 8262498 ± 4610.496515,649 ± 388810,571 ± 12630.2603

At the time of the study, HFD-fed mice were 30 wk of age. HFD-fed mice were given 3 d to acclimate to the metabolic cages. Ad libitum data were collected on the fourth day. Mice were denied access to food but had free access to water on d 5 (unfed data), and refeed data were collected on d 6 when mice regained free access to food. *P < 0.05.

Impact of myonectin deletion on hematologic parameters

Myonectin (also referred to as erythroferrone) plays important roles in linking stress erythropoiesis and hepatic iron mobilization in response to blood loss or anemia by suppressing hepcidin expression in the liver (37, 38). Here, under basal nonstressed conditions, myonectin deficiency did not alter any blood parameters, including hemoglobin level, hematocrit, and population counts of lymphoid and myeloid cell types in male mice (Supplemental Table S3), consistent with previous findings (38). Interestingly, under basal conditions when mice were fed a control LFD, female mice lacking myonectin had significantly elevated total white blood cell counts as well as lymphocyte counts. Although diet-induced obesity can adversely affect multiple cell types in the immune system (69), metabolic stress from an HFD did not alter any of the blood parameters in myonectin-deficient mice of either sex relative to control WT littermates (Supplemental Table S3). Liver hepcidin expression was also indistinguishable between WT and KO mice (unpublished results). Thus, myonectin is not required for iron homeostasis under conditions in which stress erythropoiesis is not induced.

Myonectin deletion impairs lipid handling in diet-induced obese male mice

Although HFD-fed female WT and myonectin-KO mice had similar rates of lipid clearance following an oral lipid gavage (unpublished results), male KO mice exhibited significant impairment in uptake of TGs and free fatty acids from the circulation compared with WT littermates (Fig. 3). The impaired lipid clearance phenotype following lipid loading was confirmed in 2 additional independent cohorts of HFD-fed mice. Mice develop insulin resistance after exposure to an HFD for a short period (1–4 wk), whereas more pronounced differences in body weight gain occur after longer exposure to high-fat feeding (70). Because lipid tolerance was different between WT and KO male mice fed an HFD, we sought to determine whether insulin resistance due to HFD is sufficient to impair lipid disposal in response to oral lipid gavage. In contrast to chronic high-fat feeding, relatively short exposure to an HFD for 4 wk did not alter lipid tolerance in myonectin-deficient mice compared with WT littermates (unpublished results). This result suggests that excess adiposity from chronic high-fat feeding is necessary to alter lipid handling capacity in myonectin-null animals.

An external file that holds a picture, illustration, etc.
Object name is fj.201900520Rf3.jpg

Impaired lipid tolerance in myonectin-KO male mice fed an HFD. Serum TG (A) and NEFA (B) during lipid tolerance test in male mice (WT, n = 12; KO, n = 16) at 17 wk of age. *P < 0.05, **P < 0.01, ***P < 0.001.

Impact of myonectin deficiency on metabolic response to high-fat feeding later in life

Diet-induced obese mice in our studies were exposed to HFD beginning at 5 wk of age, which corresponds to adolescence. To determine whether myonectin is required for metabolic flexibility in adult animals, we exposed another cohort of mice to HFD after they reached 21 wk of age. After switching from standard chow to HFD for 12 wk, mice were assessed by several tolerance tests. Body weight and glucose and insulin tolerance did not differ between genotypes of either sex (Supplemental Fig. S2AG). No differences in lipid tolerance were noted between WT and KO female mice (Supplemental Fig. S2H, I). Male myonectin-KO mice, however, exhibited reduced capacity to clear serum TG from the circulation following an oral lipid gavage (Supplemental Fig. S2J, K). The magnitude of difference between genotypes in lipid tolerance was smaller in this older cohort of myonectin-deficient male mice as compared with younger cohorts (Fig. 3). These results indicate that the severity of the lipid tolerance phenotype is contingent upon the age at which mice are exposed to the metabolic stress of high-fat feeding.

Elevated postprandial VLDL–TG levels in myonectin-deficient male mice

Although myonectin deficiency impairs the capacity to acutely handle orally delivered emulsified intralipid, it is unclear whether myonectin is required for the proper handling of dietary lipids in a postprandial state following physiologically normal ad libitum feeding. To test this, HFD-fed male mice were unfed overnight and then given free access to food for 2 h. Blood samples were taken before and after refeeding, and serum lipid levels were measured. Although fasting serum TG levels were similar between WT and KO mice, postprandial serum TG levels were significantly higher in myonectin-KO mice upon refeeding following an unfed period (Fig. 4A). Fasting and postprandial serum NEFA and cholesterol levels were not different between genotypes (Fig. 4B, C). Food intake did not differ between WT and KO mice (Table 4); thus, differences in calorie intake cannot explain differences in postprandial serum TG levels. Fractionation of pooled postprandial serum revealed that myonectin-KO male mice had significantly elevated and larger very LDL (VLDL)–TG particle levels compared with WT males (Fig. 4D). Lipoprotein cholesterol profiles showed that the size, but not the amount, of HDL cholesterol was larger in the myonectin-KO animals (Fig. 4E). These data indicate that myonectin is required for proper postprandial handling of dietary TG derived from normal ad libitum feeding of an HFD.

An external file that holds a picture, illustration, etc.
Object name is fj.201900520Rf4.jpg

Higher postprandial VLDL-TG levels in myonectin-KO male mice fed an HFD. A–C) Serum TG (A), NEFA (B), and cholesterol (C) levels in male mice after an overnight 14-h unfed period ("fasted") or after an unfed period overnight followed by ad libitum refeeding for 2 h (refed). Mice (36 wk of age) were euthanized after refeeding for tissue collection. A total of 2 WT mice did not eat during the refeed period as assessed by no increase in serum TG levels over fasting levels and an empty stomach. Therefore, refed data from these mice were not included in the analysis. Unfed: WT, n = 12; KO, n = 17. Refed: WT, n = 10; KO, n = 17. D, E) Refed serum samples from 10 mice of each genotype were pooled and subjected to FPLC fractionation to separate lipoprotein species. TG (D) and cholesterol (E) levels were measured in each fraction. IDL, intermediate-density lipoprotein. *P < 0.05.

Loss of myonectin does not affect intestinal lipid absorption and secretion

Several mechanisms could affect lipid clearance from the circulation. For example, elevated serum TG levels in myonectin-KO mice after oral lipid gavage or ad libitum feeding could be due to enhanced intestinal lipid absorption, greater secretion of TG-rich chylomicrons by the enterocytes, or both. To test this, overnight-unfed mice were injected with poloxamer 407, a detergent that inhibits LPL activity, preventing lipid uptake in peripheral tissue. After 1 h, mice were orally gavaged with intralipid. Accumulation of serum TG over time reflects intestinal lipid absorption and subsequent secretion of TG-rich chylomicrons. No differences in the kinetics of serum TG rise were noted between HFD-fed WT and KO mice (Supplemental Fig. S3A). Alternatively, impaired lipid uptake into peripheral tissues could result in the elevated TG levels seen in KO animals. LPL plays a major role in the cellular uptake of lipids derived from circulating lipoprotein particles; the enzyme is located on the surface of endothelial cells in most tissues and hydrolyzes TG to liberate free fatty acids, which can then be taken up by peripheral tissues (71). We therefore measured whole-body LPL activity in postheparin sera collected from mice in the postprandial state. As we previously observed, a 2-h ad libitum feeding following an overnight unfed period was sufficient to induce a difference in serum TG between genotypes; the heparin-displaceable total LPL activity, however, was not different between WT and myonectin-KO mice (Supplemental Fig. S3B, C).

Myonectin deletion alters adipose fat storage and lipid accumulation in the liver

We next sought to determine whether myonectin deficiency alters lipid storage in peripheral tissues. Although HFD-fed WT and KO mice had similar body weights and body compositions (Fig. 2A, B), the distribution of fat among tissues was altered. We noted that the KO mice (39 wk old) had significantly increased fat mass (both visceral and subcutaneous fat depots) and a modest reduction in liver weight (Fig. 5A–C and Supplemental Table S1). Liver histology revealed a marked reduction in steatosis in KO mice relative to WT littermates (Fig. 5D). Quantification of lipids also revealed significantly reduced hepatic TG content and lower cholesterol levels (Fig. 5E, F). Consistent with the histologic data and hepatic TG content, the lipid droplet–associated protein perilipin 2 was also correspondingly reduced in myonectin-KO livers (Fig. 5G). Histologic analysis of visceral epididymal WAT (eWAT) and subcutaneous iWAT indicated significantly larger adipocytes in myonectin-KO animals (Fig. 5H, I); this was further confirmed by cell size quantification (Fig. 5J, K). Differences in adipocyte cell size were more pronounced in the visceral fat depot (eWAT). These data suggest that myonectin plays a role in lipid partitioning between tissues and its deficiency promotes adipose tissue lipid storage and reduces lipid accumulation in the liver.

An external file that holds a picture, illustration, etc.
Object name is fj.201900520Rf5.jpg

Myonectin deficiency alters lipid distribution in male mice fed an HFD. A–D) WT and myonectin-KO mice were euthanized at 39 wk of age after a 2–4-h unfed period in the morning. Unless otherwise specified, data in this figure are from the analyses of WT (n = 12) and KO (n = 16) male mice. The wet weight of liver (A), visceral (gonadal) fat pad (B), and subcutaneous (inguinal) fat pad (C) are presented along with representative images of H&E-stained liver sections from WT and KO mice (D). E, F) Quantification of TG (E) and cholesterol (F) levels in livers. G) Western blot analysis of Perilipin 2. A total of 2 separate gels were run, each with WT (n = 6) and KO (n = 8) samples. Perilipin 2 (PLIN2) levels were first normalized to HSC70 levels from the same gel and then normalized to the mean WT value. Data were then combined from both gels for analysis. H, I) Representative images of H&E-stained eWAT (H) and iWAT (I) sections from WT and KO male mice. J) Quantification of adipocyte size in eWAT sections (WT, n = 5; KO, n = 5). K) Quantification of adipocyte size in iWAT sections (WT, n = 4; KO, n = 5). *P < 0.05, **P < 0.01.

Myonectin deletion alters multiple liver metabolite levels

Given the striking differences in lipid partitioning between adipose and liver of HFD-fed WT and KO animals, we measured circulating lipids from blood collected at the same time as tissue samples as shown in Fig. 5. Serum TG, NEFA, and β-hydroxybutyrate levels were similar between HFD-fed WT and KO male mice; serum cholesterol levels, however, were lower in myonectin-KO animals (Fig. 6A–D). To explore the genotypic difference in hepatic fat accumulation, we also performed unbiased metabolomics analysis to assess global changes in liver metabolites. Of the 642 metabolites quantified, only a small subset (32 in total) of metabolites significantly differed between WT and KO mice (Fig. 6E). Our data indicate that HFD-fed myonectin-KO male mice had elevated secondary bile acids, as well as increased levels of metabolites involved in 1-carbon metabolism. Interestingly, the hepatic level of imidazole propionate, a molecule reported to induce insulin resistance (72), was also significantly elevated in myonectin-KO male mice; however, this did not impair insulin sensitivity in male mice as measured by GTTs and ITTs (Fig. 2D, G). In addition to metabolite profiling, we performed extensive quantitative PCR to assess the expression of genes involved in lipid synthesis and catabolism in the liver and adipose tissue. Of the major genes involved in lipid synthesis and oxidation and lipoprotein and iron metabolism, there were no significant differences in the expression levels between genotypes in the liver or 2 major fat depots (Supplemental Fig. S4). However, for genes involved in bile acid metabolism, the expression of cholesterol 7 α-hydroxylase, the rate-limiting enzyme in bile acid synthesis, was significantly up-regulated in the liver of myonectin-KO male mice (Supplemental Fig. S4E).

An external file that holds a picture, illustration, etc.
Object name is fj.201900520Rf6.jpg

Steady-state serum and liver metabolite levels in WT and myonectin-KO male mice fed an HFD. Sera and livers were harvested from mice (39 wk old) 2–4 h after food removal. AD) Quantification of serum TG (A), NEFA (B), cholesterol (C), and ketones (β-hydroxybutyrate) (D) (WT, n = 12; KO, n = 16). E) All liver metabolites that were significantly different (P < 0.05) between WT (n = 10) and myonectin-KO (n = 10) male mice. *P < 0.05.

Myonectin deficiency does not alter hepatic VLDL-TG secretion or fat oxidation

Because steady-state levels of metabolites (after 2 h of food removal) and gene expression could not fully explain the differential lipid accumulation in adipose and liver, we tested specific pathways of lipid flux in vivo. Reduced hepatic steatosis in HFD-fed KO mice could be due to increased hepatic VLDL-TG secretion. To test this, mice were first unfed for 4 h, beginning at 2 h into the light cycle, to ensure the absence of food-derived circulating chylomicrons secreted by enterocytes; in this physiologic state, most of the circulating TG is likely derived from VLDL-TG particles secreted from liver. Mice were next injected with poloxamer 407 to inhibit lipid uptake in peripheral tissue. Thus, the accumulation of serum TG over time reflects hepatic VLDL-TG secretion. This experiment revealed no differences between WT and KO mice, indicating that hepatic VLDL-TG secretion cannot account for the reduced hepatic TG accumulation in myonectin-deficient male mice (Supplemental Fig. S5A). Alternatively, enhanced hepatic fat oxidation can result in lower hepatic TG content. Although not a direct measure of fat oxidation in the liver, serum ketones are good proxies because ketogenesis in the liver is biochemically linked to fat oxidation (73); hence, circulating ketone levels correspond to the extent of hepatic fat oxidation. Circulating ketones (β-hydroxybutyrate) did not differ between WT and KO mice in either the unfed or fed state, indicating that there were no differences in hepatic fat oxidation between genotypes (Supplemental Fig. S5B).

Myonectin deficiency does not alter adipose tissue lipolysis

Given that myonectin-KO mice had greater adiposity, decreased adipose tissue lipolysis could account for this phenotype. Although recombinant myonectin treatment does not affect ex vivo lipolysis in WAT explants (28), the impact of its deficiency on adipose lipolysis in vivo is unknown. To test this, mice were injected with a β3-adrenergic receptor agonist (CL 316,243) to stimulate lipolysis following a 5-h unfed period. CL-stimulated lipolysis was assessed by measuring serum glucose, glycerol, and NEFA before injection and 15 min postinjection; no differences were observed between WT and KO mice (Supplemental Fig. S6). Our data thus indicate that both basal (no changes in steady-state serum NEFA levels; Fig. 6B) and stimulated lipolysis were not altered in mice lacking myonectin.

Myonectin-deficient male mice have higher postprandial LPL activity in the subcutaneous fat depot

Increased lipid uptake into adipocytes could result in greater adiposity. The capacity for peripheral tissues to take up circulating lipid is largely dependent on LPL whose activity is regulated in a tissue-specific and metabolic state-dependent manner (71). In WAT, LPL expression is down-regulated while unfed to divert lipid away from adipose to other oxidative tissues. In contrast, LPL activity is activated in WAT postprandially to promote lipid uptake and storage. We therefore measured LPL activity in subcutaneous (iWAT) and visceral (eWAT) fat depots obtained from HFD-fed mice euthanized after 2 h of food removal or in a postprandial state (overnight unfed period followed by 2 h ad libitum feeding). LPL activity was significantly increased in iWAT from KO mice relative to WT controls, but only in the postprandial state (Fig. 7A, B). Increased LPL enzymatic activity was not due to increased expression of LPL mRNA or protein in the iWAT from KO animals (Fig. 7C–E). ANGPTLs and apolipoproteins (Apos) are known to regulate LPL activity (74). We therefore assessed the expression of known LPL inhibitors (Angptl3, Angptl4, Angptl8, and ApoC3) and activators (ApoC2, ApoA4, and ApoA5) in the same fat tissue samples as well as in the liver. We also examined the level of farnesoid X receptor mRNA, the transcriptional regulator of ANGPTLs and Apos, as well as glycosylphosphatidylinositol-anchored HDL-binding protein 1 and VLDL receptor, which are important for translocation of LPL to the cell surface. At the mRNA level, no significant genotypic differences were observed for these transcripts in WAT and liver (Supplemental Fig. S7AC). We also measured protein levels of ANGPTL3 and ANGPTL4, 2 of the major LPL inhibitors, in fasting serum as well as the postprandial serum samples; their protein levels were also not significantly different between genotypes (Supplemental Fig. S7D, E).

An external file that holds a picture, illustration, etc.
Object name is fj.201900520Rf7.jpg

Postprandial LPL activity in the s.c. (inguinal) fat depot is increased in myonectin-KO male mice fed an HFD. A) LPL activity in eWAT (WT, n = 12; KO, n = 16) and iWAT (WT, n = 12; KO, n = 15) from mice euthanized at 39 wk of age following a 2–4-h unfed period in the morning. B) LPL activity in eWAT and iWAT from mice (WT, n = 10; KO, n = 17) euthanized at 36 wk of age after 2-h refeeding following food removal overnight. C) LPL mRNA expression in the same eWAT (WT, n = 10; KO, n = 16) and iWAT (WT, n = 10; KO, n = 15) samples as in B. D, E) Western blot analysis of LPL in the same iWAT samples as in B. A total of 2 separate gels were run: 1 with 6 WT and 8 KO samples, and the other with 4 WT and 8 KO samples. LPL levels were first normalized to HSC70 levels from the same gel, then normalized to the mean WT value. Data were then combined from both gels for analysis (WT, n = 10; KO, n = 16) (E). Images from 1 gel are shown in D. ***P < 0.001.

DISCUSSION

We used a genetic loss-of-function mouse model to determine whether myonectin is required physiologically to maintain metabolic homeostasis. Under basal conditions in which mice were fed a control LFD comparable to standard chow, myonectin was largely dispensable for regulating whole-body carbohydrate and fat metabolism, consistent with a recent report (62). However, there is one notable phenotype in male mice fed a control LFD: myonectin deficiency significantly reduced physical activity in the ad libitum fed state, as well as in the refed (postprandial) state following a 24 h unfed period. In the unfed state, in which mice typically have heightened foraging behavior, physical activity level was not significantly different between genotypes; thus, there appears to be a physiologic state-dependent change in ambulatory and total physical activity in the KO animals. We speculate that myonectin may modulate, directly or indirectly, a central pathway controlling physical activity in male mice between 2 opposing (fed and unfed) metabolic states. Of note, the reduced physical activity seen in myonectin-KO male mice is not the result of a functional deficit in the skeletal muscle because the capacity for an exhaustive sprint or endurance run was not significantly different between genotypes; nor were the pre- and postexercise blood glucose, lactate, NEFA, or ketone levels. The ability of skeletal muscle and liver to replenish their glycogen stores following exercise (with or without glucose gavage postexercise) was also not significantly different, indicating that myonectin is not required for the metabolic response or adaptation to a single bout of exercise. It appears that myonectin is also dispensable for physiologic adaptation to chronic voluntary exercise. When indirect calorimetry analyses were conducted in WT and KO mice with access to a running wheel, we observed no significant differences in metabolic rate, food intake, or the amount of voluntary exercise (wheel running) between genotypes of either sex. Similarly, a recent report indicates that myonectin is not required for normal heart function under normal, nonstressed conditions (29).

In the pathophysiological context of obesity and insulin resistance induced by an HFD, we observed 2 significant and notable phenotypes related to lipid metabolism in the myonectin-deficient animals. First, myonectin-KO mice displayed an impaired ability to handle an acute oral lipid load. Second, lipids appear to be partitioned differentially between adipose and liver; specifically, myonectin-KO male mice had strikingly reduced TG accumulation in the liver and, in parallel, a significant increase in adiposity due to greater lipid storage in hypertrophied adipocytes. These 2 lipid phenotypes in KO animals are sex-dependent. Apart from developing mild insulin resistance and a bigger gonadal fat pad, myonectin-KO female mice fed an HFD were largely indistinguishable from WT littermates with respect to whole-body lipid metabolism. It is known that female mice on a C57BL/6 genetic background are much less susceptible to diet-induced obesity (75, 76). Many studies (77), including our previous studies (5456) and the present study, reinforce the importance of sex as a biologic variable that can influence the impact and phenotypic outcomes of a given genotype. Intriguingly, although our current study and previous findings (62) do not support a major role for myonectin in regulating glucose metabolism, recent reports suggest that circulating myonectin in human blood is correlated with insulin resistance, impaired glucose metabolism, and type 2 diabetes (60, 61). Whether myonectin plays a causal role in human carbohydrate metabolism and insulin sensitivity distinct from rodents remains to be determined.

We previously showed that myonectin infusion lowers serum lipid levels in mice and, when given to cultured hepatocytes and adipocytes, also promotes lipid uptake into cells (28). Additionally, ingestion of lipid via oral gavage can up-regulate myonectin expression (28). Based on these previous findings, we hypothesized that myonectin deficiency would result in impaired lipid clearance following an oral lipid load. Indeed, myonectin-KO male mice exhibited a significantly greater rise in both serum TG and NEFA levels following an oral lipid challenge relative to WT littermates, a functional deficit we observed in multiple independent cohorts of HFD-fed male KO mice. Several mechanisms could account for the observed phenotype, including impaired lipid clearance and uptake by peripheral tissues as hypothesized and increased intestinal lipid absorption and chylomicron secretion. We ruled out the latter as the cause of the lipid intolerance seen in myonectin-deficient animals. To determine lipid uptake by peripheral tissues, we focused on LPL activity because this enzyme is a major determinant of lipid clearance in extrahepatic tissues (71). The biggest difference in serum lipid levels between genotypes was seen at 2 h after oral lipid gavage or 2 h postfeed following an unfed period; we therefore quantified the total heparin-displaceable LPL activity in the serum 2 h postfeeding. Although there were no significant genotypic differences in total lipase activity, we could not rule out potential differences in LPL-dependent lipid uptake between WT and KO mice. Of note, both WT and KO mice converged on the same baseline lipid levels 4 h after oral lipid gavage. Thus, the observed significant differences at earlier time points likely reflect differences in the kinetics of lipid uptake rather than the overall capacity for lipid disposal in peripheral tissues. For example, relative to WT littermates, the overall LPL activity may be lower in myonectin-KO mice shortly after oral lipid gavage, reflecting slower activation of the enzyme in response to altered nutritional state, which would result in a much greater rise in serum TG at 0–2 h postgavage that we observed. At later time points, total LPL activity in KO mice may reach the same maximum level attained in WT mice, reflecting the marked decline in serum TG observed in KO mice at 2–4 h postgavage, leading to the same baseline lipid levels between genotypes 4 h after the oral lipid challenge. Hence, it is possible that when we measured total LPL activity at 2 h postfeeding, LPL activity in WT mice was in the process of decreasing back to baseline, and the activity in the KO animals was increasing to its maximal level. Future studies will help to determine the total heparin-displaceable LPL activity at different time points after oral lipid challenge.

The second major phenotype observed in myonectin-KO male mice is a significant difference in lipid distribution between liver and adipose tissue. We observed significantly greater adipocyte hypertrophy due to greater fat storage in lipid droplets in both the visceral and subcutaneous fat depots of HFD-fed KO mice. Parallel to increased adiposity, we observed a striking reduction in fat accumulation in the liver of myonectin-deficient animals. Several mechanisms could underlie this phenotype, including liver-specific processes, adipose-specific processes, or a combination of both. Reduced hepatic steatosis could be a consequence of increased fat oxidation, increased VLDL secretion, decreased lipid uptake from circulation, or decreased de novo lipogenesis. Serum ketones (β-hydroxybutyrate), a good proxy for liver fat oxidation (73), were not significantly different between WT and KO mice in either fed or unfed states, thus ruling out hepatic fat oxidation as the cause for reduced steatosis. We also ruled out enhanced hepatic VLDL-TG secretion as a contributing factor to reduced fatty liver. Although lipid uptake and fatty acid synthesis have not been directly measured, our liver metabolite profiling, in addition to extensive PCR quantification of genes involved in hepatic lipid metabolism, does not suggest any differences in these 2 processes.

Interestingly, despite a striking reduction in ectopic lipid accumulation in the liver, myonectin-KO male mice did not exhibit better hepatic glucose metabolism profiles relative to WT littermates, and fasting blood glucose levels (dominated by hepatic gluconeogenesis) and GTTs did not reveal any significant differences between genotypes. Although fatty liver resulting from excessive ectopic fat deposition in the obese state is frequently associated with impaired hepatic insulin action (as reflected in greater hepatic glucose output) (4), these 2 phenomena appear to be uncoupled in myonectin-KO animals. Intriguingly, one of the liver metabolites that was significantly up-regulated in myonectin-KO male mice is imidazole propionate. This molecule, thought to be produced from histidine by gut microbiota, was recently reported to be elevated in obese humans with type 2 diabetes, and when given to mice, it can induce insulin resistance (72). Although it seems that diabetic humans have a relatively wide range of imidazole propionate concentrations relative to nondiabetic controls, the level of up-regulation (2–3 fold) we observed in the myonectin-KO mice is not sufficient to disrupt normal insulin signaling as indicated by a lack of significant differences in fasting blood glucose, as well as glucose and insulin tolerance.

An alternative explanation for the reduced fat accumulation seen in the myonectin-KO liver could be secondary to changes in fat storage in adipose tissue, leading to reduced ectopic lipid deposition in other organs and tissues. Enlarged adipocytes seen in our myonectin-KO mice could result from decreased lipolysis, increased lipid uptake and storage, decreased fat oxidation, or increased de novo lipogenesis. We ruled out reduced adipose lipolysis as a cause of increased adiposity because neither basal nor β3-adrenergic agonist–stimulated lipolysis was significantly different between WT and KO mice. LPL activity was measured in adipose tissue as a proxy for the capacity for lipid uptake. Intriguingly, LPL activity was significantly greater in the subcutaneous (inguinal) fat depots of KO mice relative to WT littermates in the postprandial state. Increased fat uptake into adipocytes via LPL may, in part, account for the increase in adiposity and adipocyte cell size. However, this appears to be fat depot–specific; we did not observe a significant increase in LPL activity in the visceral (gonadal) fat of KO animals. Although fat oxidation and synthesis in adipose tissue were not directly measured in this study, none of the lipid synthesis or catabolism genes we considered were significantly different in their expression levels between WT and KO mice. What accounts for the increased LPL activity seen in the iWAT of myonectin-KO mice is presently unclear because none of the known regulators (angiopoetin-like proteins and Apos) of LPL activity and transcytosis were different in their expression or serum protein levels between WT and KO animals.

In summary, we provide here a detailed analysis of the metabolic consequences of myonectin deletion in a genetic mouse model. Although myonectin is largely dispensable for whole-body glucose metabolism, it plays an important role in regulating local and systemic lipid metabolism. Loss of myonectin causes lipid intolerance in response to oral lipid loading, and it alters lipid distribution between adipose and liver. Thus, in addition to regulating stress erythropoiesis in the context of blood loss (38), myonectin (erythroferrone) is also a secreted metabolic regulator of fat metabolism in the physiologic context of diet-induced obesity. Future studies will help pinpoint the mechanisms by which myonectin controls fat metabolism in a tissue-specific manner and the receptor (currently unknown) through which it exerts its biologic functions.

ACKNOWLEDGMENTS

The authors thank Susan Aja (Johns Hopkins University) for help with indirect calorimetry. This work was supported in part by a Grant from the U.S. National Institutes of Health (NIH), National Institute of Diabtes and Digestive and Kidney Diseases (DK084171 to G.W.W.). H.C.L. was supported by a National Research Service Award pre-doctoral fellowship (F31DK116537) and a NIH, National Institute of General Medical Sciences training grant (T32 GM007445). S.R. was supported by a postdoctoral fellowship from the American Diabetes Association (1-18-PMF-022). The authors declare no conflicts of interest.

Glossary

ANGPTLangiopoietin-like protein
Apoapolipoprotein
CTRPcomplement component 1q/TNF-related protein
EEenergy expenditure
eWATepididymal WAT
GTTglucose tolerance test
H&Ehematoxylin and eosin
HFDhigh-fat diet
HRPhorseradish peroxidase
Hsc70heat shock cognate 71 kDa protein
ITTinsulin tolerance test
iWATinguinal WAT
KOknockout
LFDlow-fat diet
LPLlipoprotein lipase
NEFAnonesterified free fatty acid
PBSTPBS with Tween 20
RERrespiratory exchange ratio
TGtriglyceride
VLDLvery LDL
WATwhite adipose tissue
WTwild type

Footnotes

This article includes supplemental data. Please visit http://www.fasebj.org to obtain this information.

AUTHOR CONTRIBUTIONS

H. C. Little and G. W. Wong contributed to the experimental design; H. C. Little, S. Rodriguez, X. Lei, S. Y. Tan, A. N. Stewart, A. Sahagun, and D. C. Sarver performed the experiments; H. C. Little and G. W. Wong analyzed and interpreted the data; and H. C. Little and G. W. Wong wrote the manuscript.

REFERENCES

1. Wasserman D. H., , Kang L., , Ayala J. E., , Fueger P. T., , Lee-Young R. S. (2011) The physiological regulation of glucose flux into muscle in vivo. J. Exp. Biol. 214, 254–262 [Europe PMC free article] [Abstract] [Google Scholar]
2. Roach P. J., , Depaoli-Roach A. A., , Hurley T. D., , Tagliabracci V. S. (2012) Glycogen and its metabolism: some new developments and old themes. Biochem. J. 441, 763–787 [Europe PMC free article] [Abstract] [Google Scholar]
3. Petersen K. F., , Dufour S., , Savage D. B., , Bilz S., , Solomon G., , Yonemitsu S., , Cline G. W., , Befroy D., , Zemany L., , Kahn B. B., , Papademetris X., , Rothman D. L., , Shulman G. I. (2007) The role of skeletal muscle insulin resistance in the pathogenesis of the metabolic syndrome. Proc. Natl. Acad. Sci. USA 104, 12587–12594 [Europe PMC free article] [Abstract] [Google Scholar]
4. Samuel V. T., , Shulman G. I. (2012) Mechanisms for insulin resistance: common threads and missing links. Cell 148, 852–871 [Europe PMC free article] [Abstract] [Google Scholar]
5. Pedersen B. K. (2013) Muscle as a secretory organ. Compr. Physiol. 3, 1337–1362 [Abstract] [Google Scholar]
6. Pedersen B. K., , Febbraio M. A. (2012) Muscles, exercise and obesity: skeletal muscle as a secretory organ. Nat. Rev. Endocrinol. 8, 457–465 [Abstract] [Google Scholar]
7. Henningsen J., , Rigbolt K. T., , Blagoev B., , Pedersen B. K., , Kratchmarova I. (2010) Dynamics of the skeletal muscle secretome during myoblast differentiation. Mol. Cell. Proteomics 9, 2482–2496 [Europe PMC free article] [Abstract] [Google Scholar]
8. Chan X. C., , McDermott J. C., , Siu K. W. (2007) Identification of secreted proteins during skeletal muscle development. J. Proteome Res. 6, 698–710 [Abstract] [Google Scholar]
9. Hartwig S., , Raschke S., , Knebel B., , Scheler M., , Irmler M., , Passlack W., , Muller S., , Hanisch F. G., , Franz T., , Li X., , Dicken H. D., , Eckardt K., , Beckers J., , de Angelis M. H., , Weigert C., , Häring H. U., , Al-Hasani H., , Ouwens D. M., , Eckel J., , Kotzka J., , Lehr S. (2014) Secretome profiling of primary human skeletal muscle cells. Biochim. Biophys. Acta 1844, 1011–1017 [Abstract] [Google Scholar]
10. McPherron A. C., , Lee S. J. (2002) Suppression of body fat accumulation in myostatin-deficient mice. J. Clin. Invest. 109, 595–601 [Europe PMC free article] [Abstract] [Google Scholar]
11. Shan T., , Liang X., , Bi P., , Kuang S. (2013) Myostatin knockout drives browning of white adipose tissue through activating the AMPK-PGC1α-Fndc5 pathway in muscle. FASEB J. 27, 1981–1989 [Europe PMC free article] [Abstract] [Google Scholar]
12. Guo T., , Bond N. D., , Jou W., , Gavrilova O., , Portas J., , McPherron A. C. (2012) Myostatin inhibition prevents diabetes and hyperphagia in a mouse model of lipodystrophy. Diabetes 61, 2414–2423 [Europe PMC free article] [Abstract] [Google Scholar]
13. Pedersen B. K. (2009) Edward F. Adolph distinguished lecture: muscle as an endocrine organ: IL-6 and other myokines. J. Appl. Physiol. 107, 1006–1014 [Abstract] [Google Scholar]
14. Hojman P., , Pedersen M., , Nielsen A. R., , Krogh-Madsen R., , Yfanti C., , Akerstrom T., , Nielsen S., , Pedersen B. K. (2009) Fibroblast growth factor-21 is induced in human skeletal muscles by hyperinsulinemia. Diabetes 58, 2797–2801 [Europe PMC free article] [Abstract] [Google Scholar]
15. Izumiya Y., , Bina H. A., , Ouchi N., , Akasaki Y., , Kharitonenkov A., , Walsh K. (2008) FGF21 is an Akt-regulated myokine. FEBS Lett. 582, 3805–3810 [Europe PMC free article] [Abstract] [Google Scholar]
16. Zeng L., , Akasaki Y., , Sato K., , Ouchi N., , Izumiya Y., , Walsh K. (2010) Insulin-like 6 is induced by muscle injury and functions as a regenerative factor. J. Biol. Chem. 285, 36060–36069 [Europe PMC free article] [Abstract] [Google Scholar]
17. Ouchi N., , Oshima Y., , Ohashi K., , Higuchi A., , Ikegami C., , Izumiya Y., , Walsh K. (2008) Follistatin-like 1, a secreted muscle protein, promotes endothelial cell function and revascularization in ischemic tissue through a nitric-oxide synthase-dependent mechanism. J. Biol. Chem. 283, 32802–32811 [Europe PMC free article] [Abstract] [Google Scholar]
18. Broholm C., , Laye M. J., , Brandt C., , Vadalasetty R., , Pilegaard H., , Pedersen B. K., , Scheele C. (2011) LIF is a contraction-induced myokine stimulating human myocyte proliferation. J. Appl. Physiol. 111, 251–259 [Abstract] [Google Scholar]
19. Haugen F., , Norheim F., , Lian H., , Wensaas A. J., , Dueland S., , Berg O., , Funderud A., , Skålhegg B. S., , Raastad T., , Drevon C. A. (2010) IL-7 is expressed and secreted by human skeletal muscle cells. Am. J. Physiol. Cell Physiol. 298, C807–C816 [Abstract] [Google Scholar]
20. Quinn L. S., , Anderson B. G., , Strait-Bodey L., , Stroud A. M., , Argilés J. M. (2009) Oversecretion of interleukin-15 from skeletal muscle reduces adiposity. Am. J. Physiol. Endocrinol. Metab. 296, E191–E202 [Europe PMC free article] [Abstract] [Google Scholar]
21. Nishizawa H., , Matsuda M., , Yamada Y., , Kawai K., , Suzuki E., , Makishima M., , Kitamura T., , Shimomura I. (2004) Musclin, a novel skeletal muscle-derived secretory factor. J. Biol. Chem. 279, 19391–19395 [Abstract] [Google Scholar]
22. Subbotina E., , Sierra A., , Zhu Z., , Gao Z., , Koganti S. R., , Reyes S., , Stepniak E., , Walsh S. A., , Acevedo M. R., , Perez-Terzic C. M., , Hodgson-Zingman D. M., , Zingman L. V. (2015) Musclin is an activity-stimulated myokine that enhances physical endurance. Proc. Natl. Acad. Sci. USA 112, 16042–16047 [Europe PMC free article] [Abstract] [Google Scholar]
23. Boström P., , Wu J., , Jedrychowski M. P., , Korde A., , Ye L., , Lo J. C., , Rasbach K. A., , Boström E. A., , Choi J. H., , Long J. Z., , Kajimura S., , Zingaretti M. C., , Vind B. F., , Tu H., , Cinti S., , Højlund K., , Gygi S. P., , Spiegelman B. M. (2012) A PGC1-α-dependent myokine that drives brown-fat-like development of white fat and thermogenesis. Nature 481, 463–468 [Europe PMC free article] [Abstract] [Google Scholar]
24. Welc S. S., , Clanton T. L. (2013) The regulation of interleukin-6 implicates skeletal muscle as an integrative stress sensor and endocrine organ. Exp. Physiol. 98, 359–371 [Europe PMC free article] [Abstract] [Google Scholar]
25. Staiger H., , Böhm A., , Scheler M., , Berti L., , Machann J., , Schick F., , Machicao F., , Fritsche A., , Stefan N., , Weigert C., , Krook A., , Häring H. U., , de Angelis M. H. (2013) Common genetic variation in the human FNDC5 locus, encoding the novel muscle-derived ‘browning’ factor irisin, determines insulin sensitivity. PLoS One 8, e61903 [Europe PMC free article] [Abstract] [Google Scholar]
26. Egan B., , Zierath J. R. (2013) Exercise metabolism and the molecular regulation of skeletal muscle adaptation. Cell Metab. 17, 162–184 [Abstract] [Google Scholar]
27. Trayhurn P., , Drevon C. A., , Eckel J. (2011) Secreted proteins from adipose tissue and skeletal muscle - adipokines, myokines and adipose/muscle cross-talk. Arch. Physiol. Biochem. 117, 47–56 [Abstract] [Google Scholar]
28. Seldin M. M., , Peterson J. M., , Byerly M. S., , Wei Z., , Wong G. W. (2012) Myonectin (CTRP15), a novel myokine that links skeletal muscle to systemic lipid homeostasis. J. Biol. Chem. 287, 11968–11980 [Europe PMC free article] [Abstract] [Google Scholar]
29. Otaka N., , Shibata R., , Ohashi K., , Uemura Y., , Kambara T., , Enomoto T., , Ogawa H., , Ito M., , Kawanishi H., , Maruyama S., , Joki Y., , Fujikawa Y., , Narita S., , Unno K., , Kawamoto Y., , Murate T., , Murohara T., , Ouchi N. (2018) Myonectin is an exercise-induced myokine that protects the heart from ischemia-reperfusion injury. Circ. Res. 123, 1326–1338 [Abstract] [Google Scholar]
30. Wong G. W., , Krawczyk S. A., , Kitidis-Mitrokostas C., , Ge G., , Spooner E., , Hug C., , Gimeno R., , Lodish H. F. (2009) Identification and characterization of CTRP9, a novel secreted glycoprotein, from adipose tissue that reduces serum glucose in mice and forms heterotrimers with adiponectin. FASEB J. 23, 241–258 [Europe PMC free article] [Abstract] [Google Scholar]
31. Wong G. W., , Krawczyk S. A., , Kitidis-Mitrokostas C., , Revett T., , Gimeno R., , Lodish H. F. (2008) Molecular, biochemical and functional characterizations of C1q/TNF family members: adipose-tissue-selective expression patterns, regulation by PPAR-gamma agonist, cysteine-mediated oligomerizations, combinatorial associations and metabolic functions. Biochem. J. 416, 161–177 [Europe PMC free article] [Abstract] [Google Scholar]
32. Wong G. W., , Wang J., , Hug C., , Tsao T. S., , Lodish H. F. (2004) A family of Acrp30/adiponectin structural and functional paralogs. Proc. Natl. Acad. Sci. USA 101, 10302–10307 [Europe PMC free article] [Abstract] [Google Scholar]
33. Wei Z., , Peterson J. M., , Lei X., , Cebotaru L., , Wolfgang M. J., , Baldeviano G. C., , Wong G. W. (2012) C1q/TNF-related protein-12 (CTRP12), a novel adipokine that improves insulin sensitivity and glycemic control in mouse models of obesity and diabetes. J. Biol. Chem. 287, 10301–10315 [Europe PMC free article] [Abstract] [Google Scholar]
34. Wei Z., , Peterson J. M., , Wong G. W. (2011) Metabolic regulation by C1q/TNF-related protein-13 (CTRP13): activation OF AMP-activated protein kinase and suppression of fatty acid-induced JNK signaling. J. Biol. Chem. 286, 15652–15665 [Europe PMC free article] [Abstract] [Google Scholar]
35. Wei Z., , Seldin M. M., , Natarajan N., , Djemal D. C., , Peterson J. M., , Wong G. W. (2013) C1q/tumor necrosis factor-related protein 11 (CTRP11), a novel adipose stroma-derived regulator of adipogenesis. J. Biol. Chem. 288, 10214–10229 [Europe PMC free article] [Abstract] [Google Scholar]
36. Seldin M. M., , Lei X., , Tan S. Y., , Stanson K. P., , Wei Z., , Wong G. W. (2013) Skeletal muscle-derived myonectin activates the mammalian target of rapamycin (mTOR) pathway to suppress autophagy in liver. J. Biol. Chem. 288, 36073–36082 [Europe PMC free article] [Abstract] [Google Scholar]
37. Kautz L., , Jung G., , Nemeth E., , Ganz T. (2014) Erythroferrone contributes to recovery from anemia of inflammation. Blood 124, 2569–2574 [Europe PMC free article] [Abstract] [Google Scholar]
38. Kautz L., , Jung G., , Valore E. V., , Rivella S., , Nemeth E., , Ganz T. (2014) Identification of erythroferrone as an erythroid regulator of iron metabolism. Nat. Genet. 46, 678–684 [Europe PMC free article] [Abstract] [Google Scholar]
39. Peterson J. M., , Aja S., , Wei Z., , Wong G. W. (2012) CTRP1 protein enhances fatty acid oxidation via AMP-activated protein kinase (AMPK) activation and acetyl-CoA carboxylase (ACC) inhibition. J. Biol. Chem. 287, 1576–1587 [Europe PMC free article] [Abstract] [Google Scholar]
40. Peterson J. M., , Seldin M. M., , Wei Z., , Aja S., , Wong G. W. (2013) CTRP3 attenuates diet-induced hepatic steatosis by regulating triglyceride metabolism. Am. J. Physiol. Gastrointest. Liver Physiol. 305, G214–G224 [Europe PMC free article] [Abstract] [Google Scholar]
41. Peterson J. M., , Wei Z., , Seldin M. M., , Byerly M. S., , Aja S., , Wong G. W. (2013) CTRP9 transgenic mice are protected from diet-induced obesity and metabolic dysfunction. Am. J. Physiol. Regul. Integr. Comp. Physiol. 305, R522–R533 [Europe PMC free article] [Abstract] [Google Scholar]
42. Peterson J. M., , Wei Z., , Wong G. W. (2010) C1q/TNF-related protein-3 (CTRP3), a novel adipokine that regulates hepatic glucose output. J. Biol. Chem. 285, 39691–39701 [Europe PMC free article] [Abstract] [Google Scholar]
43. Byerly M. S., , Swanson R., , Wei Z., , Seldin M. M., , McCulloh P. S., , Wong G. W. (2013) A central role for C1q/TNF-related protein 13 (CTRP13) in modulating food intake and body weight. PLoS One 8, e62862 [Europe PMC free article] [Abstract] [Google Scholar]
44. Enomoto T., , Ohashi K., , Shibata R., , Higuchi A., , Maruyama S., , Izumiya Y., , Walsh K., , Murohara T., , Ouchi N. (2011) Adipolin/C1qdc2/CTRP12 protein functions as an adipokine that improves glucose metabolism. J. Biol. Chem. 286, 34552–34558 [Europe PMC free article] [Abstract] [Google Scholar]
45. Su H., , Yuan Y., , Wang X. M., , Lau W. B., , Wang Y., , Wang X., , Gao E., , Koch W. J., , Ma X. L. (2013) Inhibition of CTRP9, a novel and cardiac-abundantly expressed cell survival molecule, by TNFα-initiated oxidative signaling contributes to exacerbated cardiac injury in diabetic mice. Basic Res. Cardiol. 108, 315–326 [Europe PMC free article] [Abstract] [Google Scholar]
46. Uemura Y., , Shibata R., , Ohashi K., , Enomoto T., , Kambara T., , Yamamoto T., , Ogura Y., , Yuasa D., , Joki Y., , Matsuo K., , Miyabe M., , Kataoka Y., , Murohara T., , Ouchi N. (2013) Adipose-derived factor CTRP9 attenuates vascular smooth muscle cell proliferation and neointimal formation. FASEB J. 27, 25–33 [Abstract] [Google Scholar]
47. Kambara T., , Ohashi K., , Shibata R., , Ogura Y., , Maruyama S., , Enomoto T., , Uemura Y., , Shimizu Y., , Yuasa D., , Matsuo K., , Miyabe M., , Kataoka Y., , Murohara T., , Ouchi N. (2012) CTRP9 protein protects against myocardial injury following ischemia-reperfusion through AMP-activated protein kinase (AMPK)-dependent mechanism. J. Biol. Chem. 287, 18965–18973 [Europe PMC free article] [Abstract] [Google Scholar]
48. Sun Y., , Yi W., , Yuan Y., , Lau W. B., , Yi D., , Wang X., , Wang Y., , Su H., , Wang X., , Gao E., , Koch W. J., , Ma X. L. (2013) C1q/tumor necrosis factor-related protein-9, a novel adipocyte-derived cytokine, attenuates adverse remodeling in the ischemic mouse heart via protein kinase A activation. Circulation 128 (11 Suppl 1), S113–S120 [Europe PMC free article] [Abstract] [Google Scholar]
49. Appari M., , Breitbart A., , Brandes F., , Szaroszyk M., , Froese N., , Korf-Klingebiel M., , Mohammadi M. M., , Grund A., , Scharf G. M., , Wang H., , Zwadlo C., , Fraccarollo D., , Schrameck U., , Nemer M., , Wong G. W., , Katus H. A., , Wollert K. C., , Müller O. J., , Bauersachs J., , Heineke J. (2017) C1q-TNF-related protein-9 promotes cardiac hypertrophy and failure. Circ. Res. 120, 66–77 [Abstract] [Google Scholar]
50. Yan W., , Guo Y., , Tao L., , Lau W. B., , Gan L., , Yan Z., , Guo R., , Gao E., , Wong G. W., , Koch W. L., , Wang Y., , Ma X. L. (2017) C1q/tumor necrosis factor-related protein-9 regulates the fate of implanted mesenchymal stem cells and mobilizes their protective effects against ischemic heart injury via multiple novel signaling pathways. Circulation 136, 2162–2177 [Europe PMC free article] [Abstract] [Google Scholar]
51. Kambara T., , Shibata R., , Ohashi K., , Matsuo K., , Hiramatsu-Ito M., , Enomoto T., , Yuasa D., , Ito M., , Hayakawa S., , Ogawa H., , Aprahamian T., , Walsh K., , Murohara T., , Ouchi N. (2015) C1q/tumor necrosis factor-related protein 9 protects against acute myocardial injury through an adiponectin receptor I-AMPK-dependent mechanism. Mol. Cell. Biol. 35, 2173–2185 [Europe PMC free article] [Abstract] [Google Scholar]
52. Yuasa D., , Ohashi K., , Shibata R., , Mizutani N., , Kataoka Y., , Kambara T., , Uemura Y., , Matsuo K., , Kanemura N., , Hayakawa S., , Hiramatsu-Ito M., , Ito M., , Ogawa H., , Murate T., , Murohara T., , Ouchi N. (2016) C1q/TNF-related protein-1 functions to protect against acute ischemic injury in the heart. FASEB J. 30, 1065–1075 [Abstract] [Google Scholar]
53. Lei X., , Seldin M. M., , Little H. C., , Choy N., , Klonisch T., , Wong G. W. (2017) C1q/TNF-related protein 6 (CTRP6) links obesity to adipose tissue inflammation and insulin resistance. J. Biol. Chem. 292, 14836–14850 [Europe PMC free article] [Abstract] [Google Scholar]
54. Petersen P. S., , Lei X., , Wolf R. M., , Rodriguez S., , Tan S. Y., , Little H. C., , Schweitzer M. A., , Magnuson T. H., , Steele K. E., , Wong G. W. (2017) CTRP7 deletion attenuates obesity-linked glucose intolerance, adipose tissue inflammation, and hepatic stress. Am. J. Physiol. Endocrinol. Metab. 312, E309–E325 [Europe PMC free article] [Abstract] [Google Scholar]
55. Rodriguez S., , Lei X., , Petersen P. S., , Tan S. Y., , Little H. C., , Wong G. W. (2016) Loss of CTRP1 disrupts glucose and lipid homeostasis. Am. J. Physiol. Endocrinol. Metab. 311, E678–E697 [Europe PMC free article] [Abstract] [Google Scholar]
56. Tan S. Y., , Little H. C., , Lei X., , Li S., , Rodriguez S., , Wong G. W. (2016) Partial deficiency of CTRP12 alters hepatic lipid metabolism. Physiol. Genomics 48, 936–949 [Europe PMC free article] [Abstract] [Google Scholar]
57. Wei Z., , Lei X., , Petersen P. S., , Aja S., , Wong G. W. (2014) Targeted deletion of C1q/TNF-related protein 9 increases food intake, decreases insulin sensitivity, and promotes hepatic steatosis in mice. Am. J. Physiol. Endocrinol. Metab. 306, E779–E790 [Europe PMC free article] [Abstract] [Google Scholar]
58. Wolf R. M., , Lei X., , Yang Z. C., , Nyandjo M., , Tan S. Y., , Wong G. W. (2016) CTRP3 deficiency reduces liver size and alters IL-6 and TGFβ levels in obese mice. Am. J. Physiol. Endocrinol. Metab. 310, E332–E345 [Europe PMC free article] [Abstract] [Google Scholar]
59. Lu L., , Zhang R. Y., , Wang X. Q., , Liu Z. H., , Shen Y., , Ding F. H., , Meng H., , Wang L. J., , Yan X. X., , Yang K., , Wang H. B., , Pu L. J., , Zhang Q., , Chen Q. J., , De Caterina R., , Shen W. F. (2016) C1q/TNF-related protein-1: an adipokine marking and promoting atherosclerosis. Eur. Heart J. 37, 1762–1771 [Abstract] [Google Scholar]
60. Li K., , Liao X., , Wang K., , Mi Q., , Zhang T., , Jia Y., , Xu X., , Luo X., , Zhang C., , Liu H., , Zhen H., , Li L., , Yang G. (2018) Myonectin predicts the development of type 2 diabetes. J. Clin. Endocrinol. Metab. 103, 139–147 [Abstract] [Google Scholar]
61. Toloza F. J. K., , Mantilla-Rivas J. O., , Pérez-Matos M. C., , Ricardo-Silgado M. L., , Morales-Alvarez M. C., , Pinzón-Cortés J. A., , Pérez-Mayorga M., , Arévalo-Garcia M. L., , Tolosa-González G., , Mendivil C. O. (2018) Plasma levels of myonectin but not myostatin or fibroblast-derived growth factor 21 are associated with insulin resistance in adult humans without diabetes mellitus. Front. Endocrinol. (Lausanne) 9, 5 [Europe PMC free article] [Abstract] [Google Scholar]
62. Coffey R., , Sardo U., , Kautz L., , Gabayan V., , Nemeth E., , Ganz T. (2018) Erythroferrone is not required for the glucoregulatory and hematologic effects of chronic erythropoietin treatment in mice. Physiol. Rep. 6, e13890 [Europe PMC free article] [Abstract] [Google Scholar]
63. Tang T., , Li L., , Tang J., , Li Y., , Lin W. Y., , Martin F., , Grant D., , Solloway M., , Parker L., , Ye W., , Forrest W., , Ghilardi N., , Oravecz T., , Platt K. A., , Rice D. S., , Hansen G. M., , Abuin A., , Eberhart D. E., , Godowski P., , Holt K. H., , Peterson A., , Zambrowicz B. P., , de Sauvage F. J. (2010) A mouse knockout library for secreted and transmembrane proteins. Nat. Biotechnol. 28, 749–755 [Abstract] [Google Scholar]
64. Qiao L., , Zou C., , van der Westhuyzen D. R., , Shao J. (2008) Adiponectin reduces plasma triglyceride by increasing VLDL triglyceride catabolism. Diabetes 57, 1824–1833 [Europe PMC free article] [Abstract] [Google Scholar]
65. Mizunoya W., , Haramizu S., , Shibakusa T., , Okabe Y., , Fushiki T. (2005) Dietary conjugated linoleic acid increases endurance capacity and fat oxidation in mice during exercise. Lipids 40, 265–271 [Abstract] [Google Scholar]
66. Schmittgen T. D., , Livak K. J. (2008) Analyzing real-time PCR data by the comparative C(T) method. Nat. Protoc. 3, 1101–1108 [Abstract] [Google Scholar]
67. Little H. C., , Tan S. Y., , Cali F. M., , Rodriguez S., , Lei X., , Wolfe A., , Hug C., , Wong G. W. (2018) Multiplex quantification identifies novel exercise-regulated myokines/cytokines in plasma and in glycolytic and oxidative skeletal muscle. Mol. Cell. Proteomics 17, 1546–1563 [Europe PMC free article] [Abstract] [Google Scholar]
68. Leick L., , Wojtaszewski J. F., , Johansen S. T., , Kiilerich K., , Comes G., , Hellsten Y., , Hidalgo J., , Pilegaard H. (2008) PGC-1alpha is not mandatory for exercise- and training-induced adaptive gene responses in mouse skeletal muscle. Am. J. Physiol. Endocrinol. Metab. 294, E463–E474 [Abstract] [Google Scholar]
69. Mathis D., , Shoelson S. E. (2011) Immunometabolism: an emerging frontier. Nat. Rev. Immunol. 11, 81–83 [Europe PMC free article] [Abstract] [Google Scholar]
70. Winzell M. S., , Ahrén B. (2004) The high-fat diet-fed mouse: a model for studying mechanisms and treatment of impaired glucose tolerance and type 2 diabetes. Diabetes 53 (Suppl 3), S215–S219 [Abstract] [Google Scholar]
71. Wang H., , Eckel R. H. (2009) Lipoprotein lipase: from gene to obesity. Am. J. Physiol. Endocrinol. Metab. 297, E271–E288 [Abstract] [Google Scholar]
72. Koh A., , Molinaro A., , Ståhlman M., , Khan M. T., , Schmidt C., , Mannerås-Holm L., , Wu H., , Carreras A., , Jeong H., , Olofsson L. E., , Bergh P. O., , Gerdes V., , Hartstra A., , de Brauw M., , Perkins R., , Nieuwdorp M., , Bergström G., , Bäckhed F. (2018) Microbially produced imidazole propionate impairs insulin signaling through mTORC1. Cell 175, 947.e17–961.e17 [Abstract] [Google Scholar]
73. McGarry J. D., , Foster D. W. (1980) Regulation of hepatic fatty acid oxidation and ketone body production. Annu. Rev. Biochem. 49, 395–420 [Abstract] [Google Scholar]
74. Zhang R. (2016) The ANGPTL3-4-8 model, a molecular mechanism for triglyceride trafficking. Open Biol. 6, 150272 [Europe PMC free article] [Abstract] [Google Scholar]
75. Yang Y., , Smith D. L., Jr, ., Keating K. D., , Allison D. B., , Nagy T. R. (2014) Variations in body weight, food intake and body composition after long-term high-fat diet feeding in C57BL/6J mice. Obesity (Silver Spring) 22, 2147–2155 [Europe PMC free article] [Abstract] [Google Scholar]
76. Hong J., , Stubbins R. E., , Smith R. R., , Harvey A. E., , Núñez N. P. (2009) Differential susceptibility to obesity between male, female and ovariectomized female mice. Nutr. J. 8, 11 [Europe PMC free article] [Abstract] [Google Scholar]
77. Varlamov O., , Bethea C. L., , Roberts C. T., Jr (2015) Sex-specific differences in lipid and glucose metabolism. Front. Endocrinol. (Lausanne) 5, 241 [Europe PMC free article] [Abstract] [Google Scholar]

Articles from The FASEB Journal are provided here courtesy of The Federation of American Societies for Experimental Biology

Citations & impact 


Impact metrics

Jump to Citations
Jump to Data

Citations of article over time

Alternative metrics

Altmetric item for https://www.altmetric.com/details/59270662
Altmetric
Discover the attention surrounding your research
https://www.altmetric.com/details/59270662

Smart citations by scite.ai
Smart citations by scite.ai include citation statements extracted from the full text of the citing article. The number of the statements may be higher than the number of citations provided by EuropePMC if one paper cites another multiple times or lower if scite has not yet processed some of the citing articles.
Explore citation contexts and check if this article has been supported or disputed.
https://scite.ai/reports/10.1096/fj.201900520r

Supporting
Mentioning
Contrasting
4
45
0

Article citations


Go to all (38) article citations

Data 


Data behind the article

This data has been text mined from the article, or deposited into data resources.

Funding 


Funders who supported this work.

American Diabetes Association (1)

NIDDK NIH HHS (2)

NIGMS NIH HHS (1)

National Institute of Diabetes and Digestive and Kidney Diseases (1)

National Institute of General Medical Sciences (1)

National Institutes of Health