13
RESEARCH ARTICLE Independent effects of dietary fat and sucrose content on chondrocyte metabolism and osteoarthritis pathology in mice Elise L. Donovan 1, *, Erika Barboza Prado Lopes 1 , Albert Batushansky 1 , Mike Kinter 1,2 and Timothy M. Griffin 1,2,3, ABSTRACT Obesity is one of the most significant risk factors for knee osteoarthritis. However, therapeutic strategies to prevent or treat obesity-associated osteoarthritis are limited because of uncertainty about the etiology of disease, particularly with regard to metabolic factors. High-fat-diet-induced obese mice have become a widely used model for testing hypotheses about how obesity increases the risk of osteoarthritis, but progress has been limited by variation in disease severity, with some reports concluding that dietary treatment alone is insufficient to induce osteoarthritis in mice. We hypothesized that increased sucrose content of typical low-fat control diets contributes to osteoarthritis pathology and thus alters outcomes when evaluating the effects of a high-fat diet. We tested this hypothesis in male C57BL/6J mice by comparing the effects of purified diets that independently varied sucrose or fat content from 6 to 26 weeks of age. Outcomes included osteoarthritis pathology, serum metabolites, and cartilage gene and protein changes associated with cellular metabolism and stress-response pathways. We found that the relative content of sucrose versus cornstarch in low-fat iso-caloric purified diets caused substantial differences in serum metabolites, joint pathology, and cartilage metabolic and stress-response pathways, despite no differences in body mass or body fat. We also found that higher dietary fat increased fatty acid metabolic enzymes in cartilage. The findings indicate that the choice of control diets should be carefully considered in mouse osteoarthritis studies. Our study also indicates that altered cartilage metabolism might be a contributing factor to how diet and obesity increase the risk of osteoarthritis. KEY WORDS: Osteoarthritis, Obesity, Cartilage, Metabolism, High-fat diet, Mouse INTRODUCTION Obesity is a major risk factor for knee osteoarthritis (OA) (Deshpande et al., 2016; Felson et al., 1988), doubling the lifetime risk compared with individuals with a body mass index (BMI) below 25 (Murphy et al., 2008). Biomechanical, inflammatory and metabolic factors are all believed to contribute to obesity-associated OA pathology (Courties et al., 2015; Issa and Griffin, 2012; Messier, 2009; Rai and Sandell, 2011; Thijssen et al., 2015). Furthermore, many studies have shown that components of metabolic syndrome, including hypertension, hyperglycemia, dyslipidemia and central adiposity, are associated with OA pathology and the risk of progression (Berenbaum et al., 2017; Katz et al., 2010; Sowers et al., 2009; Zhuo et al., 2012). In particular, articular cartilage is susceptible to the deleterious effects of both elevated glucose and lipids. For example, high dietary fat consumption (Lu et al., 2016) and type 2 diabetes (Eymard et al., 2015) are each associated with more rapidly progressing joint space narrowing in individuals with knee OA, even after adjusting for BMI. However, the causal role of metabolic syndrome and its components in knee OA progression remain unclear (Appleton et al., 2017; Niu et al., 2017). Thus, experimentally isolating the effects of specific dietary macronutrients on cartilage metabolism and joint pathology might improve our understanding of how metabolic factors contribute to OA pathogenesis. The use of diet-induced obese animal models to study OA pathogenesis has expanded rapidly in the past 10 years (Berenbaum et al., 2017; Griffin and Guilak, 2008). However, variability in the timing and severity of high-fat-diet-induced OA phenotypes, particularly in mice, has raised questions about the robustness and repeatability of the model (Kozijn et al., 2017). One potential source of variation is the comparison of defined high-fat diets to undefined chow diets, which differ in many ways other than just fat content (Warden and Fisler, 2008). For example, chow diets contain high levels of isoflavones, which can influence hormone-dependent rodent phenotypes (Brown and Setchell, 2001). Another difference between chow and defined diets is fiber type and content. Defined diets often include an insoluble fiber, cellulose, which contributes to alterations in gut morphology and microbiota content (Chassaing et al., 2015; Dalby et al., 2017). Moreover, even when defined control and high-fat diets are used, the specific composition of macronutrients can vary and contribute to metabolic perturbations. Wu and colleagues elegantly showed the importance of dietary fatty acid composition in post-traumatic OA pathogenesis (Wu et al., 2015). An area that has received less attention in diet-induced obese animal studies of OA is carbohydrate type and content. We recently reported the time course of OA pathogenesis in male C57BL/6J mice fed a high-fat diet (60% kcal fat; ResearchDiets, D12492) compared with a defined control-fat diet (10% kcal fat; ResearchDiets, D12450B) (Barboza et al., 2017). High-fat feeding did not increase the overall knee OA score versus control diet until >32 weeks of high-fat feeding. This duration was longer than expected, based on our previous work showing early-stage OA changes after 12 weeks using the same 60% high-fat diet; however, in this earlier study, the control animals ate an undefined chow diet (Griffin et al., 2012). Received 5 April 2018; Accepted 9 July 2018 1 Aging and Metabolism Research Program, Oklahoma Medical Research Foundation (OMRF), Oklahoma City, OK 73104, USA. 2 Department of Geriatric Medicine, Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA. 3 Department of Biochemistry and Molecular Biology and Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA. *Present address: Oregon Institute of Technology, Klamath Falls, OR, USA. Author for correspondence ([email protected]) T.M.G., 0000-0001-8840-9940 This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/3.0), which permits unrestricted use, distribution and reproduction in any medium provided that the original work is properly attributed. 1 © 2018. Published by The Company of Biologists Ltd | Disease Models & Mechanisms (2018) 11, dmm034827. doi:10.1242/dmm.034827 Disease Models & Mechanisms

Independent effects of dietary fat and sucrose content on ...consistency.Basedonmaximumscores,thekneeOApathologywas moderate and did not differ between the three diets using either

  • Upload
    others

  • View
    2

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Independent effects of dietary fat and sucrose content on ...consistency.Basedonmaximumscores,thekneeOApathologywas moderate and did not differ between the three diets using either

RESEARCH ARTICLE

Independent effects of dietary fat and sucrose content onchondrocyte metabolism and osteoarthritis pathology in miceElise L. Donovan1,*, Erika Barboza Prado Lopes1, Albert Batushansky1, Mike Kinter1,2 andTimothy M. Griffin1,2,3,‡

ABSTRACTObesity is one of the most significant risk factors for kneeosteoarthritis. However, therapeutic strategies to prevent or treatobesity-associated osteoarthritis are limited because of uncertaintyabout the etiology of disease, particularly with regard to metabolicfactors. High-fat-diet-induced obese mice have become a widelyused model for testing hypotheses about how obesity increases therisk of osteoarthritis, but progress has been limited by variation indisease severity, with some reports concluding that dietary treatmentalone is insufficient to induce osteoarthritis in mice. We hypothesizedthat increased sucrose content of typical low-fat control diets contributesto osteoarthritis pathology and thus alters outcomes when evaluatingthe effects of a high-fat diet. We tested this hypothesis in maleC57BL/6J mice by comparing the effects of purified diets thatindependently varied sucrose or fat content from 6 to 26 weeks of age.Outcomes included osteoarthritis pathology, serum metabolites, andcartilage gene andprotein changes associatedwith cellularmetabolismand stress-response pathways. We found that the relative content ofsucrose versus cornstarch in low-fat iso-caloric purified diets causedsubstantial differences in serum metabolites, joint pathology, andcartilage metabolic and stress-response pathways, despite nodifferences in body mass or body fat. We also found that higherdietary fat increased fatty acid metabolic enzymes in cartilage. Thefindings indicate that the choice of control diets should be carefullyconsidered in mouse osteoarthritis studies. Our study also indicatesthat altered cartilage metabolism might be a contributing factor tohow diet and obesity increase the risk of osteoarthritis.

KEY WORDS: Osteoarthritis, Obesity, Cartilage, Metabolism,High-fat diet, Mouse

INTRODUCTIONObesity is a major risk factor for knee osteoarthritis (OA)(Deshpande et al., 2016; Felson et al., 1988), doubling the lifetimerisk compared with individuals with a body mass index (BMI)below 25 (Murphy et al., 2008). Biomechanical, inflammatory and

metabolic factors are all believed to contribute to obesity-associatedOA pathology (Courties et al., 2015; Issa and Griffin, 2012; Messier,2009; Rai and Sandell, 2011; Thijssen et al., 2015). Furthermore,many studies have shown that components of metabolic syndrome,including hypertension, hyperglycemia, dyslipidemia and centraladiposity, are associated with OA pathology and the risk ofprogression (Berenbaum et al., 2017; Katz et al., 2010; Sowerset al., 2009; Zhuo et al., 2012). In particular, articular cartilage issusceptible to the deleterious effects of both elevated glucose andlipids. For example, high dietary fat consumption (Lu et al., 2016)and type 2 diabetes (Eymard et al., 2015) are each associatedwith more rapidly progressing joint space narrowing in individualswith knee OA, even after adjusting for BMI. However, the causalrole of metabolic syndrome and its components in knee OAprogression remain unclear (Appleton et al., 2017; Niu et al.,2017). Thus, experimentally isolating the effects of specific dietarymacronutrients on cartilage metabolism and joint pathology mightimprove our understanding of how metabolic factors contribute toOA pathogenesis.

The use of diet-induced obese animal models to study OApathogenesis has expanded rapidly in the past 10 years (Berenbaumet al., 2017; Griffin and Guilak, 2008). However, variability inthe timing and severity of high-fat-diet-induced OA phenotypes,particularly in mice, has raised questions about the robustness andrepeatability of the model (Kozijn et al., 2017). One potential sourceof variation is the comparison of defined high-fat diets to undefinedchow diets, which differ in many ways other than just fat content(Warden and Fisler, 2008). For example, chow diets contain highlevels of isoflavones, which can influence hormone-dependentrodent phenotypes (Brown and Setchell, 2001). Another differencebetween chow and defined diets is fiber type and content. Defineddiets often include an insoluble fiber, cellulose, which contributes toalterations in gut morphology and microbiota content (Chassainget al., 2015; Dalby et al., 2017). Moreover, even when definedcontrol and high-fat diets are used, the specific composition ofmacronutrients can vary and contribute to metabolic perturbations.Wu and colleagues elegantly showed the importance of dietaryfatty acid composition in post-traumatic OA pathogenesis(Wu et al., 2015).

An area that has received less attention in diet-induced obeseanimal studies of OA is carbohydrate type and content. We recentlyreported the time course of OA pathogenesis in male C57BL/6J micefed a high-fat diet (60% kcal fat; ResearchDiets, D12492) comparedwith a defined control-fat diet (10% kcal fat; ResearchDiets,D12450B) (Barboza et al., 2017). High-fat feeding did not increasethe overall knee OA score versus control diet until >32 weeks ofhigh-fat feeding. This duration was longer than expected, based onour previous work showing early-stage OA changes after 12 weeksusing the same 60% high-fat diet; however, in this earlier study, thecontrol animals ate an undefined chow diet (Griffin et al., 2012).Received 5 April 2018; Accepted 9 July 2018

1Aging and Metabolism Research Program, Oklahoma Medical ResearchFoundation (OMRF), Oklahoma City, OK 73104, USA. 2Department of GeriatricMedicine, Reynolds Oklahoma Center on Aging, University of Oklahoma HealthSciences Center, Oklahoma City, OK 73104, USA. 3Department of Biochemistryand Molecular Biology and Department of Physiology, University of OklahomaHealth Sciences Center, Oklahoma City, OK 73104, USA.*Present address: Oregon Institute of Technology, Klamath Falls, OR, USA.

‡Author for correspondence ([email protected])

T.M.G., 0000-0001-8840-9940

This is an Open Access article distributed under the terms of the Creative Commons AttributionLicense (http://creativecommons.org/licenses/by/3.0), which permits unrestricted use,distribution and reproduction in any medium provided that the original work is properly attributed.

1

© 2018. Published by The Company of Biologists Ltd | Disease Models & Mechanisms (2018) 11, dmm034827. doi:10.1242/dmm.034827

Disea

seModels&Mechan

isms

Page 2: Independent effects of dietary fat and sucrose content on ...consistency.Basedonmaximumscores,thekneeOApathologywas moderate and did not differ between the three diets using either

Thus, differences in the dietary compositions of the control dietsmight contribute to the variation in outcomes. In particular, it iscommon for many defined low-fat control diets, which often containnearly 70% kcal from carbohydrates, to include a substantial amountof sucrose. Sucrose is a disaccharide composed of glucose andfructose. In people, fructose consumption independently causesdyslipidemia and abdominal adiposity (Warden and Fisler, 2008).Thus, diet manufacturers began producing diets that matched thesucrose content of control low-fat diets to that in the high-fat diet tocontrol for potential effects of sucrose. We incorporated a recentlydeveloped low-fat diet with lower sucrose content to furtherunderstand how dietary sucrose independent of fat contributesto OA pathophysiology in the C57BL/6J mouse model ofdiet-induced OA.We compared the independent effects of dietary sucrose and

fat on knee OA pathology and cartilage metabolism by feedingC57BL/6J mice one of three defined diets: (1) 10% kcal fat diet withsucrose content matched to that contained in the high-fat diet (7%),herein defined as low fat low sucrose (LFLS); (2) the previouslyused 10% kcal fat diet with higher sucrose content (35%), referredto as low fat high sucrose (LFHS); and (3) the previously used high-fat diet containing 60% kcal fat and 7% kcal sucrose, referred to ashigh fat low sucrose (HFLS) (Table S1). The definitions of the dietgroups as LFLS, LFHS and HFLS are intended to distinguish therelative sucrose and fat content between the groups, although theLFLS group is similar to chow diets in terms of sucrose and fatcontent. We hypothesized that increased dietary sucrose and fat(i.e. LFHS and HFLS diets, respectively) would induce systemicmetabolic pathology, cartilage stress and early-stage markers of OApathology compared with the LFLS diet group. We focused on20 weeks of diet treatment, which causes early-stage OA pathologyin the HFLS group (Barboza et al., 2017).

RESULTSDietary sucrose content in low-fat diets changes serummetabolic biomarkers without altering body weight or fatHigh dietary sucrose content did not alter body weight or percentagebody fat content in mice fed low-fat diets (i.e. 10% kcal fat, 70%kcal carbohydrate) (Fig. 1A,B, LFLS versus LFHS). In contrast,mice fed a HFLS diet gained substantially more body weight

(∼40% increase) and body fat (∼2.5-fold increase) compared withlow-fat fed mice, regardless of sucrose content (Fig. 1A,B).To further characterize the metabolic effects of dietary sucroseand fat content, we also analyzed a panel of diagnostic serummetabolic markers in blood collected at the time of death. Uponnecropsy, we observed tumor-like nodules in the liver of two LFLSanimals. The presence of nodules was associated with altered serummetabolites and synovial thickening (Fig. S1); therefore, thesesamples were excluded from the final serum and synovial analyses,as well as cartilage gene and protein analyses. We did not, however,observe differences in cartilage or osteophyte histopathology scoresassociated with liver nodules so these samples were included inthose outcome analyses.

Overall, metabolic markers were more likely to be significantlyelevated in mice fed a HFLS diet when compared with mice fed alow-fat diet that containedmore, not less, sucrose (i.e. LFHS) (Fig. 2).This is because metabolic markers were generally lower in micefed the LFHS diet compared with those fed the LFLS diet (Fig. 2).For example, serum albumin, bilirubin, gamma-glutamyltransferase,globulins, protein and urea nitrogen were all significantly reduced inLFHS versus LFLS mice. Moreover, serum calcium, glucose andtriglycerides also trended lower in LFHS mice. In fact, mice fed aLFLS diet had elevated levels of total serum protein and urea nitrogencompared with mice fed any other diet, suggesting that this diet mightstimulate protein catabolism.

Notably, only two metabolites were elevated in the HFLS versusLFLS diet group: alanine aminotransferase (ALT) and inorganicphosphate (Fig. 2). In addition, serum cholesterol was greater inmice fed a HFLS diet regardless of sucrose content in the low-fatdiets. High dietary sucrose did not significantly elevate anymetabolitelevels compared with the other diets. Overall, these data show thatdietary sucrose significantly alters serum markers of metabolicfunction independent of changes in body weight or fat. Contrary toour expectation, more serum metabolites were altered in animalsfed a high-fat diet when compared with those fed a control dietwith more, not less, sucrose. Consequently, interpreting the effectof high-fat-diet-induced obesity on serum metabolites depends onthe carbohydrate composition of the control low-fat diet.

Low dietary sucrose and high dietary fat induce differenthistological changes in knee cartilage and bone that areassociated with early-stage OAWe evaluated OA histopathology scoring using two complementaryapproaches, the maximum joint score and the overall location-averaged joint score (Fig. 3A). The maximum score comparison ismore consistent with clinical definitions of disease burden, whereasthe average score is less sensitive to small focal changes andthus better represents more widespread pathological changes.We previously reported overall site-average scoring for the LFHSandHFLS joints (Barboza et al., 2017), but these histological sectionswere blinded and re-graded together with the LFLS sections forconsistency. Based on maximum scores, the knee OA pathology wasmoderate and did not differ between the three diets using either theOsteoarthritis Research Society International (OARSI) or modifiedMankin scoring systems (Fig. 3A; Table S2). The averagewhole-jointOA pathology, however, showed minor significant or trendingincreases for the LFLS group (Fig. 3A). The primary factor drivingthe slight increase in early signs of OA in the LFLS group was agreater loss of Safranin-O staining, indicating reduced proteoglycancontent. These changes were greatest in the lateral tibial plateau,where superficial cartilage damage was also increased (Fig. 3B,C).In contrast, other histopathological changes were greatest in the

Fig. 1. Effect of diet on body mass and adiposity. (A,B) Increased dietaryfat content, but not sucrose, increased body mass (A) and body fat (B).Data were collected at 25 weeks of age after 19 weeks of diet treatment.Body fat, expressed as a percent of total body mass, was measured by dual-energy X-ray absorptiometry (DEXA). Data shown are for one cohort ofanimals, although values are consistent with data from additional cohorts.Values are mean±s.e.m. Lines over bars indicate statistically significantdifferences (****P<0.0001 by one-way ANOVA and Tukey’s multiplecomparisons test; LFLS, n=10; LFHS, n=8; HFLS, n=10).

2

RESEARCH ARTICLE Disease Models & Mechanisms (2018) 11, dmm034827. doi:10.1242/dmm.034827

Disea

seModels&Mechan

isms

Page 3: Independent effects of dietary fat and sucrose content on ...consistency.Basedonmaximumscores,thekneeOApathologywas moderate and did not differ between the three diets using either

LFHS and HFLS diet groups. Notably, a high-fat diet increasedosteophyte severity and cartilage tidemark duplication (Fig. 3A,B).Moreover, when scores were averaged throughout the joint, theLFHS and HFLS groups showed increased numbers of hypertrophicchondrocytes in the calcified cartilage (Fig. 3A,B). The LFHS andHFLS groups also showed increases in the maximal synovialcellularity and thickness in focal areas adjacent to the meniscus(Fig. 3F,G; Fig. S2). Thus, animals on each diet showed differentcharacteristics of early-stage OA pathology.

Diet composition alters the expression of cellular stressresponse genes in cartilageWe first evaluated the expression of pro-anabolic and pro-catabolicgenes in cartilage that was isolated from the femoral condyles andtibial plateau (Fig. 4A,B, respectively). The minor changes in kneejoint histopathology were not associated with significant changes inthe expression of primary cartilage anabolic genes (e.g. Acan orCol2a1) or catabolic genes (e.g. Adamts5 andMmp13) (Fig. 4A,B).Thus, we next evaluated the expression of genes associated with theprotection against, and induction of, cellular stress. The expressionof the AMP-activated protein kinase subunit Prkaa1 and thenuclear NAD-dependent deacetylase sirtuin-1 (Sirt1) showed

near-significant trends for differences across the diet groups(P=0.058 and P=0.063, respectively; one-way ANOVA) (Fig. 4A).These trends were primarily caused by increased expression in theLFLS versus LFHS diet groups [P=0.058 and P=0.067, respectively;false discovery rate (FDR)-adjusted post-hoc comparison].Moreover,the expression of the transcription factor Foxo3, which positivelyregulates chondrocyte autophagy and antioxidant expression,was significantly altered by diet (P=0.0002; one-way ANOVA).Like Prkaa1 and Sirt1, Foxo3 was also most highly expressed inthe LFLS group (Fig. 4A). Diet did not alter the expression ofgenes transcribing the cellular stress mediators PGE2 and HIF-2α(Ptgs2 and Epas1, respectively; Fig. 4B). However, diet did alterthe expression of the endoplasmic reticulum (ER) stress mediatorDdit3, also known as Chop, with expression greatest in the LFLSgroup (P<0.0001, one-way ANOVA; Fig. 4B). CHOP is atranscription factor for which expression is upregulated in responseto ER stress and the mitochondrial unfolded protein response(mtUPR) (Shpilka and Haynes, 2017).

To further characterize these diet-dependent cellular stressresponses, we next analyzed the expression of genes involved incellular protein homeostasis (i.e. ‘proteostasis’) and metabolism.These findings suggest that dietary sucrose and fat content led to

Fig. 2. Dietary sucrose and fat independently alter serum metabolic biomarkers. Metabolic markers were generally lower in mice fed the LFHS dietcompared with those fed the LFLS diet. Consequently, markers were more likely to be significantly elevated in mice fed a HFLS diet when compared with micefed a low-fat diet that contained more, not less, sucrose. Blood was obtained by terminal cardiac puncture between 09:00 and 11:00 following a 1-2 h fast.Data points are values for individual animals, and horizontal bars are mean±s.e.m. Results that include two LFLS samples excluded due to liver nodules areshown in Fig. S1. Lines over bars indicate statistically significant differences (*P<0.05, **P<0.01, ***P<0.001 by one-way ANOVA and Tukey’s multiplecomparisons test or Kruskal–Wallis test with Dunn’s multiple comparisons test; LFLS, n=6-8; LFHS, n=5-6; HFLS, n=4-9). Variations in sample size were due tomarker detectability. ALKP, alkaline phosphatase; ALT, alanine aminotransferase; GGT, gamma-glutamyltransferase.

3

RESEARCH ARTICLE Disease Models & Mechanisms (2018) 11, dmm034827. doi:10.1242/dmm.034827

Disea

seModels&Mechan

isms

Page 4: Independent effects of dietary fat and sucrose content on ...consistency.Basedonmaximumscores,thekneeOApathologywas moderate and did not differ between the three diets using either

differences in cartilage mitochondrial proteostasis and lipidmetabolism. Diet did not significantly alter the expression of thetranscription factorX box-binding protein 1 (Xbp1), a key regulator ofthe ER stress response (Fig. 4C); however, it did alter the expressionof the ubiquitous adenosine triphosphate (ATP)-dependent molecular

chaperone Hsp90 (also known as Hsp90aa1) and the mitochondrial-localized chaperone Hsp10 (also known as Hspe1). In both cases,expression was greatest in cartilage from the LFLS diet group(Fig. 4C). Furthermore, diet also altered the expression of themitochondrial protease subunits Clpp and Lonp1, which were also

Fig. 3. See next page for legend.

4

RESEARCH ARTICLE Disease Models & Mechanisms (2018) 11, dmm034827. doi:10.1242/dmm.034827

Disea

seModels&Mechan

isms

Page 5: Independent effects of dietary fat and sucrose content on ...consistency.Basedonmaximumscores,thekneeOApathologywas moderate and did not differ between the three diets using either

most highly expressed in the LFLS diet group (P=0.0012 andP=0.09, respectively; one-way ANOVA). There were no differencesin the expression of Hif1a or the autophagy mediator Bcl2 acrossdiets, although a key regulator ofmitochondrial translation,Mtif2, wasmore highly expressed in the LFLS versus LFHS group (Fig. 4D).Based on the potential role of diet-induced changes in cartilage

lipid metabolism, we also evaluated the expression of two importantlipid metabolism regulatory enzymes: acetyl-CoA carboxylase (Acc)and carnitine palmitoyltransferase 1 (Cpt1a). Acc activity inhibits thebeta-oxidation of fatty acids and promotes lipid synthesis; whereas,Cpt1a shuttles fatty acids into the mitochondria and is the rate-limiting step of beta-oxidation. Diet did not alter Acc expression,although variation was much greater in the LFLS group comparedwith the LFHS or HFLS groups. In contrast, a high-fat dietsignificantly increased Cpt1a expression regardless of the sucrosecontent of the low-fat diets, suggesting a potential increase incartilage beta-oxidation (Fig. 4D). Detailed cellular stress andmetabolism gene expression data are provided in Table S3.

High dietary sucrose is associated with lower levels ofcartilagemetabolic and antioxidant proteins comparedwithhigh dietary fatWe next utilized selected-reaction-monitoring (SRM) massspectrometry to quantify protein abundance in knee cartilagehomogenates. The analysis targeted 120 proteins spanning theglycolysis, beta-oxidation, tricarboxylic acid (TCA) cycle andelectron transport chain pathways, as well as enzymes involved incellular antioxidant defense and proteostasis. Of these, 100 proteinswere sufficiently abundant for quantification. A list of all detectedproteins and their diet-specific differences in abundance is providedin Table S4. Twenty-seven proteins were significantly altered bydiet. These proteins were distributed throughout all the metabolicand antioxidant pathways that were evaluated (Fig. 5).Overall, the greatest number of diet-induced differences occurred

with elevated dietary sucrose (20 proteins; LFHS versus LFLS)compared with dietary fat (7 proteins; HFLS versus LFLS).These differences were generally due to protein levels being lowerin the LFHS versus LFLS group, with HFLS group levels beingintermediate. For example, the proteins that were less abundant withincreased dietary sucrose included nearly all of the antioxidantproteins and many involved in glycolysis [e.g. glycogenphosphorylase (Pygb), phosphofructokinase (Pfkl), lactatedehydrogenase (Ldhb)] and the TCA cycle [e.g. aconitase (Aco2),fumarate hydratase (Fh1), pyruvate carboxylase (Pc)] (Fig. 5). Therewere some notable exceptions, however, where protein abundancewas increased with high dietary sucrose. These proteins include the

TCA cycle enzyme isocitrate dehydrogenase (Idh3b), complex V ofthe electron transport chain [i.e. mitochondrial ATP synthase(Atp5b, Atp5a1)], and aldo-keto reductase family 1 member B3(Akr1b3), which is the first and rate-limiting enzyme in the polyolpathway (Fig. 5).

As previously mentioned, there were fewer differences inprotein abundance associated with high dietary fat (i.e. HFLSversus LFLS). Proteins that were uniquely elevated in theHFLS group involve those that regulate the beta-oxidation offatty acids, including acetyl-coenzyme A acyltransferase 2 (Acaa2)and enoyl-coenzyme A delta isomerase (Eci2). More proteins,however, were lower in the HFLS group, including Ldhb,Aco2, Pc, superoxide dismutase 2 (Sod2), and dihydrolipoamidedehydrogenase (DLD), an E3 component of pyruvate andα-ketoglutarate dehydrogenases. When comparing elevated dietaryfat versus sucrose (i.e. HFLS versus LFHS), high dietary fat wasassociated with greater levels of proteins involved in glycolysis(Pfkl, Eno1), beta-oxidation (Ech1, Hadha, Acaa2), and antioxidantdefense (Gpx1) (Fig. 5; Table S4).

High dietary fat greatly reduces the network connectivity ofcartilage metabolic and cellular stress proteinsThe analysis of metabolic and antioxidant protein levels in cartilagefrom LFLS, LFHS and HFLS diet groups showed differences acrossmultiple pathways, suggesting substantial effects of dietary sucroseand fat content on cartilage metabolic regulation. Therefore, wesought an integrative analytical approach to evaluate these effectsusing a correlation-based network analysis. Correlation networkanalyses, also referred to as co-expression analyses, can provideinference into biological function when quantitative relationshipsbetween pairs of values, in this case protein levels, are consideredin aggregate. For our analysis, we evaluated each diet separately todefine a diet-specific protein network (Fig. 6A). These networkswere based on protein pairs with significant correlations amongindependent within-diet sample replicates (i.e. correlation coefficient|r|≥0.8 and P≤0.05). We focused on differences in overall networksize and density (Fig. 6A-C), metabolic pathway-specific intrinsicnetwork density (Fig. 6D) and ‘hub’ proteins with the greatestnumber of correlations (Fig. 6E).

We observed substantial differences in network topology amongthe three diets (Fig. 6A). The diet effects were almost exclusivelycaused by differences in the number of correlations among proteinpairs (i.e. the number of ‘links’) rather than differences in the numberof proteins with ≥1 significant correlation (i.e. ‘number of nodes’),which was nearly identical among diets (Fig. 6B). Consequently, thenetwork density varied considerably among diets, being greatest inthe LFHS diet (0.12) and least in the HFLS diet (0.04) (Fig. 6B).Notably, the LFLS diet network was intermediate to both diets andcontained a similar number of positive and negative correlations(Fig. 6A,B). Of >750 protein pair correlations among all threediets, only nine were common to all three diets (Fig. 6C). Ingeneral, the majority of protein pair correlations were unique toeach diet. We next evaluated the network densities that wereintrinsic to specific metabolic pathways. These results showed thatnetwork densities of enzymes involved in carbohydratemetabolism and the TCA cycle were relatively greatest in theLFLS and LFHS diets, whereas fatty acid metabolism enzymedensities were relatively greatest in the HFLS diet (Fig. 6D).Finally, we compared the proteins with the greatest number ofsignificant correlations with other proteins within each dietnetwork (i.e. ‘hub’ proteins) (Fig. 6E). This comparison showedthat stress-related proteins (i.e. antioxidant and proteostasis

Fig. 3. Dietary sucrose and fat independently alter knee OA pathology.(A) Cartilage OA histopathology scoring of medial and lateral kneecompartments. Osteophyte scoring from medial tibial compartment.Data points represent values for individual animals, and horizontal bars aremean±s.e.m. (B) Location-specific OA histopathology (LF, lateral femur; LT,lateral tibia; MF, medial femur; MT, medial tibia). Data are mean±s.e.m.(C-E) Representative sagittal section histology images from the lateral kneecompartment. Scale bars: 100 µm. Dashed line rectangular area from thelateral tibial plateau is shown at higher magnification in the lower image toillustrate the greater prevalence of cartilage fibrillation (arrowheads) andSafranin-O staining loss (arrow) in the LFLS group than in the LFHS and HFLSgroups. (F) Representative and maximal synovial lining cellularity andthickness. (G) Maximal focal synovial cellularity and thickness by jointcompartment. Lines over bars indicate statistically significant differences(*P<0.05, **P<0.01) by Kruskal–Wallis (A,F) or repeated-measures two-wayANOVA (B,G) with FDR-corrected post hoc tests (0.05). n=10 for each dietgroup, except for the LFLS group in F and G (n=8).

5

RESEARCH ARTICLE Disease Models & Mechanisms (2018) 11, dmm034827. doi:10.1242/dmm.034827

Disea

seModels&Mechan

isms

Page 6: Independent effects of dietary fat and sucrose content on ...consistency.Basedonmaximumscores,thekneeOApathologywas moderate and did not differ between the three diets using either

proteins) were the most common hub proteins in each network.However, the specific proteins mostly differed between the diets,and the number of correlations associated with the top-rankedproteins reflected the diet-specific network density differences (i.e.greatest in LFHS and least in HFLS) (Fig. 6E).

DISCUSSIONWe hypothesized that increased dietary sucrose and fat would inducesystemic metabolic pathology, cartilage stress, and early-stagemarkers of OA pathology compared with the LFLS diet group.Our findings showed broad effects of dietary sucrose and fat in all of

Fig. 4. Effects of dietary sucrose and fat on the expression of metabolic and stress-response genes in knee cartilage. (A-D) Data are presentedas expression relative to the geometric mean of three reference genes rather than as fold-change relative to the LFLS group, owing to the absence oflongitudinal data for assessing diet-induced up- versus downregulation. Diet did not substantially alter the expression of cartilage anabolic genes (A) (e.g. Acan orCol2a1) or cartilage catabolic genes (B) (e.g. Adamts5 and Mmp13). However, diet did alter the expression of cellular stress mediators Foxo3, a transcriptionfactor that positively regulates chondrocyte autophagy and antioxidant expression, and Ddit3 (i.e. Chop), a mediator of endoplasmic reticulum stress andthe mitochondrial unfolded protein response. Further evaluation of genes involved in cellular proteostasis (C) and metabolism (D) showed significant effectsof diet on mediators of cartilage mitochondrial proteostasis and lipid metabolism. Detailed cellular stress and metabolism gene expression data, includinggeneNCBI RefSeq numbers, are provided in Table S3. Lines over bars indicate statistically significant differences. *P<0.05, **P<0.01 and ***P<0.001 by one-wayANOVA and Tukey’s multiple comparisons test or Kruskal–Wallis test with Dunn’s multiple comparisons test; n=8 per diet group.

6

RESEARCH ARTICLE Disease Models & Mechanisms (2018) 11, dmm034827. doi:10.1242/dmm.034827

Disea

seModels&Mechan

isms

Page 7: Independent effects of dietary fat and sucrose content on ...consistency.Basedonmaximumscores,thekneeOApathologywas moderate and did not differ between the three diets using either

these outcomes, as summarized in Table 1. Notably, the greatestnumber of changes occurred between the low- and high-sucrosediets despite no differences in body mass or body fat. For example,there were more than twice as many differences in cartilage geneexpression and protein abundance between the low- and high-sucrose diets compared with the low- and high-fat diets (Table 1).However, contrary to our hypothesis, more pathologic changeswere associated with a low-sucrose diet. For example, cartilagepathology and serum metabolic disease biomarkers were reducedin the high-sucrose diet group (Table 1). Consequently, for studiesusing high-fat diets to study obesity-induced OA, the carbohydratecomposition of the low-fat control diet has the potential tosignificantly alter the interpretation of obesity-related outcomes

(e.g. compare the ‘Increased fat’ and ‘Mixed comparison’ columnsummaries in Table 1).

The importance of the composition of control diets inhigh-fat-diet-induced obesity studies of OA pathophysiologyhas not been previously recognized. In fact, we recently reviewedhow the variation in high-fat diet content and composition betweenstudies is a potential reason for differences in OA outcomes(Berenbaum et al., 2017). However, based on our current findings,variation in control diets could also be a contributing factor.We compiled a follow-up to Table 2 of Berenbaum et al. (2017),which includes the control diets used in studies of diet-inducedobesity and OA (Table S5). This table shows that like the variationin high-fat diets, there is also substantial variation among controldiets, including the use of nonpurified ‘chow’ diets versus purifiedlow-fat diets. This added variation in control diet composition couldfurther contribute to differences reported in the literature (Kozijnet al., 2017).

What is the ideal control diet for diet-induced obese mousestudies of OA? From a macro-nutrient perspective (i.e. % kcal fromprotein, carbohydrates and fat), both the LFLS and LFHS diets aresimilar to many chow diets. However, the lower sucrose content ofthe LFLS diet is more similar to chow, which typically contains littleto no sucrose. The LFLS diet is also the most appropriate control dietfor comparison to the HFLS diet because the only differences arethe relative content of cornstarch versus lard. However, the use ofcellulose as an insoluble fiber supplement in many purified defineddiets, such as those used in the current study, causes substantialchanges in gut morphology and microbiota content comparedwith chow-fed mice (Chassaing et al., 2015; Dalby et al., 2017).These changes could underlie our findings indicating that the LFLSdiet was moderately stressful to the mice. Numerous serummetabolicbiomarkers were elevated in the LFLS diet group, including serumalbumin, bilirubin, gamma-glutamyltransferase, globulins, proteinand urea nitrogen. These findings suggest that liver function wasimpaired with the LFLS diet and that protein catabolism might havebeen increased. In addition, two animals developed tumor-like livernodules. A direct comparison to chow-fed mice would be needed totest this idea, although the comparison to LFHS-fed mice suggests apotential benefit of sucrose.

It is not known how increased sucrose content improved metabolichealth, although oneway could be by increasing palatability. A recentdietary preference study in rats using the same diets as the currentstudy showed a strong preference for the HFLS diet and a secondarypreference for the LFHS diet, with the relative consumption being4%, 10% and 86% of total calories for the LFLS, LFHS, andHFLS diets, respectively (Lee et al., 2017). Thus, palatability couldcontribute to differences among diets, perhaps by reducing stress(Finger et al., 2011). An additional factor that could be modified toimprove the health of mice fed purified LFLS diets is by changingthe composition of the fiber supplement. Swapping insolublecellulose for a soluble fiber, inulin, has been shown to protectagainst the loss of gut mass and recover fecal short-chain fatty acidlevels close to those observed in chow-fed mice (Chassaing et al.,2015). Recent studies have linked altered gut microbiota to OApathology (Collins et al., 2015; Schott et al., 2018), suggesting thatthe fiber composition of purified, defined diets is an importantconsideration in diet selection. Thus, although the current study doesnot clearly identify the appropriate ‘control’ diet for diet-inducedobese mouse studies of OA, it shows that controlling for body weightor body fat alone is insufficient. Future studies are needed that includelongitudinal testing and the modification of purified, defined low-fatdiets to improve palatability and fiber composition.

Fig. 5. Effects of dietary sucrose and fat on changes in metabolic andantioxidant enzyme abundance in knee cartilage. Heatmap of proteinabundance levels as measured by selected-reaction-monitoring (SRM) massspectrometry. Enzymes with a significant diet effect (one-way ANOVA;P<0.05) are shown, with relative abundance indicated by color scale.More diet-induced differences occurred with elevated dietary sucrose(20 proteins; LFHS versus LFLS) compared with dietary fat (7 proteins; HFLSversus LFLS). These differences were generally caused by protein levelsbeing lower in the LFHS versus LFLS group, with HFLS group levelsbeing intermediate. When comparing elevated dietary fat versus sucrose(i.e. HFLS versus LFHS), high dietary fat was associated with greater levelsof proteins involved in glycolysis (Pfkl, Eno1), β-oxidation (Ech1, Hadha,Acaa2), and antioxidant defense (Gpx1). For β-oxidation enzymes, ‘p’indicates localization to the peroxisome and ‘m’ indicates localization tomitochondria. Protein names and diet-specific differences based on Tukeypost hoc statistical analysis are provided in Table S4. TCA, tricarboxylic acid.

7

RESEARCH ARTICLE Disease Models & Mechanisms (2018) 11, dmm034827. doi:10.1242/dmm.034827

Disea

seModels&Mechan

isms

Page 8: Independent effects of dietary fat and sucrose content on ...consistency.Basedonmaximumscores,thekneeOApathologywas moderate and did not differ between the three diets using either

Our findings shed light on an important question within the field,which is to what extent do changes in systemic metabolism alterjoint tissue metabolism? In terms of systemic factors, we observedthat elevated levels of total serum protein and urea nitrogen, factorspotentially linked to increased protein catabolism or kidney or liverdisease, were associated with reduced cartilage Safranin-O staining inLFLS-fed mice. Low-grade systemic inflammation and hypertensionare both linked to OA progression clinically (Attur et al., 2015;Niu et al., 2017), although they were not tested in the current study.In HFLS-fedmice, elevated cholesterol was associated with increasedcartilage tidemark duplication and osteophyte formation. Highcholesterol has previously been shown to increase ectopic boneformation in experimental mouse studies (de Munter et al., 2013),and dysregulated cholesterol levels and elevated dietary fat havebeen associated with OA in humans (Berenbaum et al., 2017;Clockaerts et al., 2011; Lu et al., 2016).In cartilage, we observed changes in gene expression and

protein abundance, consistent with distinct diet effects on substratemetabolism. For example, our findings suggest that a high-fat dietincreases cartilage lipid metabolism. The HFLS diet increased

Cpt1a expression, which encodes the enzyme mediating therate-limiting step of beta-oxidation and has been implicated in arole for the nuclear receptor peroxisome proliferator-activatedreceptor δ as a promoter of cartilage degeneration (Ratneswaranet al., 2015, 2017). The HFLS diet also increased the terminalenzyme of beta-oxidation, acetyl-coenzyme A acyltransferase 2.Furthermore, a pathway-specific network analysis of carbohydrateversus fatty acid metabolic pathways showed that the fatty acidnetwork density was elevated with the HFLS diet. Conversely, anincrease in dietary sucrose modestly increased the carbohydratenetwork density relative to the LFLS group. In addition, the increasein Akr1b3 in the LFHS group suggests that sucrose increased thepolyol metabolic pathway, which was also observed in primarychondrocytes cultured in high glucose (Laiguillon et al., 2015).The consequences of increased polyol pathway flux are not wellunderstood in chondrocytes, but they could involve altered cellularredox homeostasis and enhanced chondrocyte inflammation(Laiguillon et al., 2015).

We previously reported that high-fat-diet-induced obesity did notincrease overt OA cartilage pathology until >32 weeks of diet

Fig. 6. Correlation-based networkanalysis of cartilage metabolic andstress-response antioxidant enzymesfollowing high-sucrose and high-fatdiets. Networks were developed based onsignificantly correlated protein pairs (i.e.correlation coefficient |r|≥0.8, P≤0.05)among independent diet-specific cartilagesamples analyzed by SRM massspectrometry. (A) Graphical networkrepresentation for each diet. Squaresrepresent one protein (i.e. ‘node’) with atleast one significant correlation (i.e. ‘link’) toanother protein. Enzyme functionalcategories and direction of correlation areindicated by the key. (B) Network densitycalculations show substantial diet effects,being greatest with a high-sucrose diet andleast with a high-fat diet. (C) Venn diagramof the correlated enzyme pairs showsthat most correlations are diet specific.(D) Network densities intrinsic to specificmetabolic pathways. Carbohydratemetabolism and TCA cycle pathwaydensities were elevated for LFLS and LFHSdiets, whereas fatty acid metabolismdensity was relatively greatest with a HFLSdiet. (E) Diet-specific comparison ofproteins with the greatest number of links toother proteins (i.e. ‘hub’ proteins). Fullprotein names are available in Table S4.

8

RESEARCH ARTICLE Disease Models & Mechanisms (2018) 11, dmm034827. doi:10.1242/dmm.034827

Disea

seModels&Mechan

isms

Page 9: Independent effects of dietary fat and sucrose content on ...consistency.Basedonmaximumscores,thekneeOApathologywas moderate and did not differ between the three diets using either

Table 1. Summary of outcomes for all diet-specific comparisons

Category Specific outcomeOveralldiet effect

Increased fat(HFLS:LFLS)

Increased sucrose(LFHS:LFLS)

Mixed comparison(HFLS:LFHS)

Knee OA scores (whole joint averages) Modified Mankin OA * – – –

Safranin-O stain loss ** – – – –

Tidemark duplication ** ++ ++Hypertrophic chondrocytes * ++ ++Osteophyte severity *** ++ ++Maximum synovial thickness ** ++Maximum synovial cellularity * ++ ++

Body habitus Body mass (g) **** ++ ++Body fat (%) **** ++ ++

Serum metabolic panel Albumin ** – –

ALT *** ++Amylase *** ++Bilirubin (total) * – – ++Calcium ** – ++Cholesterol **** ++ ++GGT ** – – ++Globins (total) * – –

Glucose * – ++Inorganic phosphate ** ++Protein (total) *** – – – –

Triglycerides * – +Urea nitrogen ** – – – –

Cartilage gene expression Ddit3 **** – – – –

Clpp ** – – – –

Clpx * – –

Cpt1a ** ++ ++Foxo3 *** – – – –

Hsp10 ** – – – –

Hsp90 * – –

Lonp1 * – –

Mtif2 * – –

Sod1 *** – – – –

Sod2 * – – – –

Cartilage protein content ABCD3 * – –

ACAA2 ** ++ ++ACO2 * – – – –

AKR1B3 * ++ALB_M ** – –

ALDH2 * – –

ATP5A1 ** ++ATP5B ** ++CAT * – –

DLD * – –

ECH1 ** ++ECI1 ** ++ECI2 * ++ENO1 * ++FH1 * – –

GPX1 ** – – ++HADHA ** – – ++IDH3B * ++LDHB ** – – – –

PC * – – – –

PFKL * – – ++PRDX1 ** – –

PRDX2 ** – –

PYGB * – –

SOD1 * – –

SOD2 ** – – – –

TXNRD1 * – –

The relative increase in sucrose versus cornstarch in low-fat iso-caloric purified diets (i.e., LFHS:LFLS) caused substantial differences in serummetabolites, jointpathology, and cartilage metabolic and stress-response pathways, despite no differences in body mass or body fat. These differences were greater than thosecaused by elevated dietary fat (i.e., HFLS:LFLS). Consequently, the carbohydrate composition of low-fat control diets has the potential to substantially alter theinterpretation of the effect of elevated dietary fat on OA-related pathology. Comparing the ‘Increased fat’ and ‘Mixed comparisons’ columns shows this effect.Asterisks refer to ANOVA P-values: *P<0.05, **P<0.01, ***P<0.001, ****P<0.0001. Plus/minus symbols refer to post hoc statistical comparisons as follows:++P<0.05 upregulation, +0.05<P<0.10 upregulation, – –P<0.05 downregulation, –0.05<P<0.10 downregulation.

9

RESEARCH ARTICLE Disease Models & Mechanisms (2018) 11, dmm034827. doi:10.1242/dmm.034827

Disea

seModels&Mechan

isms

Page 10: Independent effects of dietary fat and sucrose content on ...consistency.Basedonmaximumscores,thekneeOApathologywas moderate and did not differ between the three diets using either

treatment using the HFLS versus the LFHS diets (Barboza et al.,2017). Therefore, an additional goal of this study was to identifyearly molecular changes in cartilage that might contribute to thefuture increase in cartilage pathology. There were few differences incartilage gene expression or protein abundance in the HFLS versusLFHS groups. The most notable differences were in genes andproteins associated with elevated lipid metabolism. However, acorrelation network analysis of metabolic and stress-responseproteins revealed stark differences between these two diets interms of the density of the networks and the specific pairs ofcorrelated proteins (i.e. ‘links’). The HFLS network density wasonly one-third that of the LFHS diet, despite a similar-sizednetwork. Changes in network density have previously beenassociated with different types and durations of cellular stressresponses. In yeast, oxidative stress induces denser and moretightly co-regulated gene and protein co-expression networks(Lehtinen et al., 2013). In contrast, chronic or pathologic stressconditions can lead to a reduction in network density (Szalay et al.,2007). In both cases, these changes in network density might beadaptive or reflect different temporal phases of stress responses.Therefore, it is not possible to determine from our data whether thedifferences in network densities between HFLS and LFHS groupsreflect different adaptive or temporal readjustments to cellularstress. However, the low density of the HFLS network leaves itmore susceptible to collapse. Furthermore, the stress-responsive70 kDa heat shock proteins HSPA1A and HSPA9 were the mosthighly linked ‘hub’ proteins in the LFLS and LFHS diets,respectively, whereas the HFLS network did not contain anyhighly linked heat shock proteins.This study has several limitations. We found that numerous

stress-related genes and proteins were elevated in the cartilage fromthe LFLS group. However, with our study design, it is not possibleto interpret whether gene expression is elevated in the LFLS group orsuppressed in the LFHS and HFLS groups without data from earliertime points to establish a basal level of expression. Interpretation isalso hindered from a functional perspective because stress-responsivegenes are often initially upregulated in a homeostatic response. Theselimitations also apply to the interpretation of the network analyses.Future longitudinal studies are needed to establish the causalrelationship of these diet treatments.Our results do not directly indicate the molecular etiology of OA

pathologies observed among the different diet groups. For example,there were no differences in gene expression of matrix components(Acan, Col2a1) or matrix degrading proteases (Adamts5, Mmp9,Mmp13), although gene expression might not reflect differences inprotein abundance or activity. It is also possible that diet treatments,which were started prior to skeletal maturation, altered joint tissuestructure by impairing matrix synthesis anabolic pathways. Forexample, the reduced Safranin-O staining observed in the LFLSdiet group was associated with a significant increase in glycogenphosphorylase (Pygb), the enzyme that mediates glycogenolysis.This might indicate that glucose was limiting, which could affectmonosaccharide supply for proteoglycan synthesis. Although wedid not independently measure proteoglycan content of cartilageto verify the Safranin-O staining results, our proteomic resultsare consistent with them.We found that the cartilage albumin contentwas significantly increased in the LFLS group (Table S4, ALB_M),and albumin content in cartilage has been shown to be inverselyproportional to proteoglycan content owing to changes in the cartilagesolute diffusion partition coefficient (Snowden andMaroudas, 1976).Other limitations are that we did not evaluate other OA-related jointtissue changes, such as muscle weakness, subchondral bone sclerosis,

intra-articular fat inflammation, and meniscus or ligamentdegradation.

An additional limitation is that the diet treatments differed in therelative period of time that animals were housed at The JacksonLaboratory versus the Oklahoma Medical Research Foundation(OMRF). LFHS and HFLS animals began diet treatments at 6 weeksof age in The Jackson Laboratory’s Diet-Induced Obese Mouseservice before being purchased at 23-24 weeks of age. These animalswere therefore only housed at the OMRF for up to 3 weeks prior totissue collection. In contrast, the LFLS diet is not available throughThe Jackson Laboratory’s Diet-Induced Obese Mouse service.Therefore, animals were purchased from The Jackson Laboratoryat 5 weeks of age and fed the LFLS diet at the OMRF from6 to 26 weeks of age. We did, however, attempt to minimize otherpotential sources of variation by evaluating joint histopathology in ablinded fashion for all groups at the same time. We also conductedall serum and gene expression analyses for all groups at the sametime, and the proteomic analyses were completed in two batches thatinvolved mixed diet group samples.

ConclusionsObesity is a primary risk factor for knee OA, but how pro-obesogenicdiets and their various macronutrient components affect chondrocytepathophysiology is not well understood. The results from this studyshow that dietary sucrose and fat content have independent effectson serum metabolic biomarkers and knee joint pathophysiology,including significant changes in cartilage metabolic and stress-responsive genes and proteins. The relative content of sucrose versuscornstarch in low-fat iso-caloric purified diets caused substantialdifferences in serum metabolites, joint pathology, and cartilagemetabolic and stress-response pathways, despite no differences inbodymass or body fat. These results illustrate the impact of metabolicfactors on OA pathophysiology independent of changes in adiposity.The findings also indicate that the choice of control diets in mousediet-induced obesity studies of OA should be carefully considered instudy design and the interpretation of results. Finally, our findingssuggest that increased dietary fat induces a metabolic shift in cartilagemetabolism towards increased utilization of fatty acids. Thus, alteredcartilage metabolism might be a contributing factor to how obesityincreases the risk of OA.

MATERIALS AND METHODSAnimalsAll experiments were conducted in accordance with protocolsapproved by the Association for Assessment and Accreditation ofLaboratory Animal Care-accredited Institutional Animal Care andUse Committee at the OMRF.Male C57BL/6J mice were purchasedfrom The Jackson Laboratory (USA). Beginning at 6 weeks of age,animals were fed one of three irradiated, purified open source diets(Research Diets Inc., USA; Table S1): (1) low-fat low-sucrose(LFLS) diet containing 10% kcal fat and 7% sucrose (productD12450Ji); (2) low-fat high-sucrose (LFHS) diet containing 10%kcal fat and 35% sucrose (product D12450Bi); or (3) high-fatlow-sucrose (HFLS) diet containing 60% kcal fat and 7% sucrose(product D12492i). Diets were provided ad libitum for 20 weeks,and animals were euthanized by either CO2 asphyxiation orexsanguination under isoflurane anesthesia at 26 weeks of age.LFHS and HFLS animals were purchased at 23-24 weeks of ageas part of The Jackson Laboratory’s Diet-Induced Obese Mouseservice, which randomizes animal to diet groups. Thus, theseanimals were acclimated to the OMRF vivarium for up to 3 weeksprior to serum and tissue collection. Animals receiving the LFLS

10

RESEARCH ARTICLE Disease Models & Mechanisms (2018) 11, dmm034827. doi:10.1242/dmm.034827

Disea

seModels&Mechan

isms

Page 11: Independent effects of dietary fat and sucrose content on ...consistency.Basedonmaximumscores,thekneeOApathologywas moderate and did not differ between the three diets using either

diet were shipped at 5 weeks of age and underwent diet treatmentfrom 6 to 26 weeks of age in the OMRF vivarium. Mice were grouphoused (≤5 animals/cage) in ventilated cages in a temperature-controlled room maintained at 22±3°C on 14/10 h light/dark cycleswith ad libitum access to food and water. Animals were weighedweekly and received daily inspection and routine veterinaryassessment. Body composition, excluding the head, was measuredunder isoflurane anesthesia using a DEXA system at 25 weeks of age(Lunar PIXImus2, GE LUNAR Corp., USA).

Serum metabolite analysisAnimals were placed in transport cages and moved to the laboratoryfor a period of 1-2 h prior to blood collection between 09:00 and11:00. We collected blood as a terminal procedure by cardiacpuncture under isoflurane anesthesia. Blood was allowed to clot inmicrovette tubes (CB 300 Z, SARSTEDT, Germany) at roomtemperature for 20 min and then centrifuged at 10,000 g for 5 min.Serum was aliquoted and frozen at −80°C until analysis. Serummetabolites were measured in samples with ≥200 µl serumusing an IDEXX Catalyst Dx Chemistry Analyzer (USA) withChem 17, triglycerides and fructosamine panels following themanufacturer’s instructions.

Histopathology analysisFollowing death, the left limb was skinned and the kneewas isolatedby manual dissection mid-femur to mid-tibia. Periarticular skeletalmuscle was retained to facilitate fixation at a physiologic joint angle,and a small incision was made near the origin of the patellarligament to expose the joint cavity. Joints were then placed in 4%paraformaldehyde for 24 h at 4°C for tissue fixation. Followingfixation, periarticular muscles were removed by gross dissection,knees were rinsed in phosphate buffered saline, and joints weredecalcified using CalEx HCl-based decalcifying solution (ThermoFisher Scientific, USA) for 3 days at 4°C. Knees were thendehydrated in an ethanol gradient prior to paraffin embedding andsagittal sectioning. Slides were stained with Hematoxylin, Fast Greenand Safranin-O for histological grading as described previously(Cai et al., 2014; Griffin et al., 2010).Two experienced graders (E.B.P.L. and T.M.G.) evaluatedmultiple

stained sections from the medial and lateral joint compartments.Slides were organized by knee joint sample, randomized by diettreatment, and assigned a temporary identification code to blindgraders to diet treatment and minimize any order effect. ModifiedMankin grading and OARSI mouse OA grading scores wereassigned separately for the medial tibia, medial femur, lateral tibiaand lateral femur independently by each grader (Table S2).Graders also evaluated osteophyte severity semiquantitatively aspreviously described (Barboza et al., 2017). Subcomponent scoresthat differed by >2 (modified Mankin cartilage damage andSafranin-O loss) or >1 (all other scores) between graders werere-evaluated for consensus scoring. Scores were then averaged forboth graders to obtain a final score per section and location.OA severity was evaluated using two complementary approaches:the maximum joint score and the overall location-averaged jointscore. The maximum score is the highest score per joint for anysection or joint location; this approach is more consistent withclinical definitions of disease. In contrast, the average score is lesssensitive to small focal changes and thus better represents morewidespread pathological changes. We previously reported overallsite-average OA scoring for the LFHS and HFLS joints (Barbozaet al., 2017); however, these joint sections were re-blinded andintegrated with the LFLS sections to be graded again to prevent

any grader-dependent discrepancies. Importantly, the previouslyreported findings were replicated here, indicating reproducibilityof the evaluation process. Synovial cellularity and thickness wasevaluated in the medial and lateral joint compartments superiorand inferior to the anterior horn of the meniscus. Quantitative andsemiquantitative data were collected in a blinded fashion usingmethods modified from Lewis and colleagues (Lewis et al., 2011)and described in detail in Fig. S2 and Table S2.

RNA and protein extractionArticular cartilage from the right knee was carefully collected withthe aid of a stereomicroscope immediately following animal death.Femur and tibia were gently disarticulated, and meniscus andconnective tissues were carefully removed from the articular surfacesusing McPherson-Vannus scissors and fine forceps. Then, with afresh size-11 scalpel blade held initially at a 45-60° angle to thecartilage surface, the blade was inserted into the cartilage and rotatedalmost parallel to the surface to remove cartilage without disturbingthe subchondral bone. Cartilage pieces were placed in 250 µlTRIzol™ Reagent (Invitrogen, USA) on ice, flash-frozen in liquidnitrogen, and stored at −80°C until further processing. Histologicalevaluation of joints after cartilage removal indicates that the processcaptures uncalcified and calcified cartilage (data not shown). Thawedsamples were mechanically homogenized using a microcentrifugetube homogenizing pestle and rotary motor (RAECorporation, USA)system. Samples were homogenized 3×30 s, placing samples onice in between to minimize heating. Total TRIzol™ volume was thenincreased to 500 µl, and 100 µl chloroform was added beforecentrifuging samples for 15 min at 12,000 g at 4°C. The aqueouslayer containing the chondrocyte RNA was then collected intoequal volume ethanol and further processed using RNA cleanand Concentrator Columns per manufacturer protocol (ZymoResearch, USA). RNA was eluted in 10 μl DNase/RNase-FreeWater. RNA concentration and purity were determined using aNanoDrop spectrophotometer (Thermo Fisher Scientific). TotalRNA yields are typically ∼1 µg with 260/280 ratios between1.87 and 2.05. Protein was collected from the lower organic phasefraction remaining from the TRIzol™ extraction according to themanufacturer’s protocol and processed as previously described formass spectrometry analysis (Fu et al., 2016).

Gene expression analysis by quantitative reversetranscription PCR arrayWe designed custom quantitative PCR arrays (Qiagen, USA)containing 42 target genes related to OA pathology, cellularmetabolism and cellular stress (Table S3). The array included threereference genes (B2m, Gapdh and Actb), a positive PCR control, apositive reverse transcription control, and a negative genomicDNA control. Then, 200 ng cDNAwas synthesized using the RT2

First-Strand kit (Qiagen) and loaded onto plates followingthe manufacturer’s instructions. Plates were run on a CFX96thermocycler (Bio-Rad, USA). Data for each target gene werenormalized to the geometric mean of the three reference genes,and gene expression was evaluated using the 2-ΔCt method(Schmittgen and Livak, 2008).

Protein abundance analysis by SRM mass spectrometryProtein abundance was quantified using SRM mass spectrometry, aspreviously reported (Rindler et al., 2013). Cartilage protein (20 μg)was suspended in 1% sodium dodecyl sulfate (SDS), with 8 pmolbovine serum albumin (BSA) added as an internal standard. Sampleswere heated to equilibrate and proteins were precipitated with acetone.

11

RESEARCH ARTICLE Disease Models & Mechanisms (2018) 11, dmm034827. doi:10.1242/dmm.034827

Disea

seModels&Mechan

isms

Page 12: Independent effects of dietary fat and sucrose content on ...consistency.Basedonmaximumscores,thekneeOApathologywas moderate and did not differ between the three diets using either

Dried protein pellets were reconstituted in 23 µl Laemmli samplebuffer, and the entire sample was run into a short (1.5 cm) SDS-polyacrylamide gel electrophoresis gel. Each sample was cut from thegel as the entire lane and divided into smaller pieces. Gel pieces werewashed to remove Coomassie Blue staining and then reduced,alkylated and digested overnight with trypsin. Peptide mixtures wereextracted from the gel, evaporated to dryness in a SpeedVac, andreconstituted in 150 µl 1% acetic acid for analysis on a TSQ Vantagetriple quadrupole mass spectrometry system (Thermo FisherScientific). The high-performance liquid chromatography systemused was an Eksigent splitless nanoflow system with a 10 cm×75 µminner diameter C18 reversed-phase capillary column. Aliquots of 7 µlwere injected and the peptides eluted with a 60 min gradient ofacetonitrile in 0.1% formic acid. The mass spectrometer was operatedin the SRM mode. For each protein, a method was developed tomeasure two ideal peptides. Assays for multiple proteins werebundled together in larger panels. Data were analyzed using theprogram SkyLine to determine the integrated peak area of theappropriate chromatographic peaks. The response for each proteinwas calculated as the geometric mean of the area of the two protein-specific peptides. Values were then normalized to the BSA internalstandard and a stable cellular reference protein,MDH1. Samples wereanalyzed in two separate runs [Run 1: LFHS (n=3) and HFLS (n=5);Run 2: LFLS (n=8), LFHS (n=5) and HFLS (n=3)], and data werenormalized to the median response of all LFHS and HFLS samplesfor each run to minimize batch effects. The integrated data were usedfor statistical comparison among the three diet groups and forconstructing the protein network analysis. Of 138 proteins included inthe SRM analysis panels, 101 were identified in mouse articular kneecartilage (Table S4).

Protein correlation network analysisProtein correlation network construction and analysis wasperformed as previously described (Batushansky et al., 2016).Briefly, diet-specific Pearson’s correlation matrices werecalculated for all samples from each diet (n=8 per diet) usingthe psych package for R-software (https://CRAN.R-project.org/package=psych). A high threshold for correlation coefficient|r|≥0.8, P≤0.05 was chosen based on sample size effects onstability of the main graph theory properties of the network, suchas transitivity, density and diameter (Pavlopoulos et al., 2011).The significant correlation matrices were transformed to adjacentlists and visualized in Cytoscape, version 3.5.1 (Shannon et al.,2003). The calculation of network properties was performed usingigraph package for R-software (Csardi and Nepusz, 2006).

Statistical analysisSample size calculations were based on our primary outcome,which was the effect of diet treatment on OA histopathology.Based on prior studies, n=10 per diet group is estimated to provide>80% power to detect a 30% difference in mean modified Mankinscores with a significance level of P=0.05. Secondary outcomesincluded diet effects on body habitus, serum metabolic biomarkers,and cartilage gene and protein expression. Sample sizes of n<10for serum metabolic biomarkers were due to insufficient sampleavailability. Cartilage gene and protein samples were collected fromten animals per group. However, samples from two LFLS animalswere excluded owing to the presence of tumor-like liver nodules;additional samples from LFHS and HFLS groups were excludedowing to poor sample quality for a final sample size of n=8 per group.Histology and body composition data for LFHS and HFLS groups(n=10 each) were analyzed from animals included in a prior study

(Barboza et al., 2017). An additional 30 animals (n=10 per group)were purchased for all additional outcomes. Diet treatment effectswere evaluated by one-way ANOVA. Data that did not meet testassumptions for homoscedasticity, even after transformation, wereanalyzed by Kruskal–Wallis test. Tests showing a significant effect ofdiet (P<0.05) were followed up with multiple-comparison post hoctests to identify individual group differences as specified in figurelegends. Statistical tests were conducted using the software Prism7.0b for Mac OS X. Data are expressed as mean±s.e.m. unlessotherwise stated. n indicates animal numbers per group, and samplesizes are provided in figure legends. P<0.05 was consideredsignificant. The heatmap in Fig. 5 was created using the ggplot2package for R.

AcknowledgementsWe thank the following investigators for their assistance with data collection andanimal care: Dr Yao Fu, Caroline Kinter, Zack Young, Mike McDaniel, EvanMcKinnis,MelindaWest, Joanna Hudson and Erin Hutchison.We also thank the OMRF ImagingCore Facility for assistance with tissue processing for histological evaluation andJapneet Kuar for assistance with synovial pathology evaluation. Finally, weacknowledge input fromDrs Luke Szweda, Ken Humphries, Sai Tummala, Mary BethHumphrey and Matlock Jeffries in the preparation of this manuscript.

Competing interestsThe authors declare no competing or financial interests.

Author contributionsConceptualization: E.L.D., T.M.G.; Methodology: E.L.D., M.K., T.M.G.; Software:A.B.; Validation: E.L.D., M.K., T.M.G.; Formal analysis: E.L.D., A.B., T.M.G.;Investigation: E.L.D., E.B.P.L., M.K.; Resources: M.K., T.M.G.; Writing - originaldraft: E.L.D., A.B., T.M.G.; Writing - review & editing: E.L.D., E.B.P.L., A.B., M.K.,T.M.G.; Visualization: E.L.D., A.B., T.M.G.; Supervision: T.M.G.; Projectadministration: E.L.D.; Funding acquisition: M.K., T.M.G.

FundingThis work was supported by the National Institutes of Health (NIH) (P20RR018758,P20GM103441, P30GM114731, P30AG050911 and R01AG049058) and theArthritis Foundation (Arthritis Investigator Award to T.M.G.). The content is solely theresponsibility of the authors and does not necessarily represent the official views ofthe NIH or the Arthritis Foundation.

Supplementary informationSupplementary information available online athttp://dmm.biologists.org/lookup/doi/10.1242/dmm.034827.supplemental

ReferencesAppleton, C. T., Hawker, G. A., Hill, C. L. and Pope, J. E. (2017). Editorial:

“Weighing in” on the framingham osteoarthritis study: measuring biomechanicaland metabolic contributions to osteoarthritis. Arthritis Rheumatol 69, 1127-1130.

Attur, M., Krasnokutsky, S., Statnikov, A., Samuels, J., Li, Z., Friese, O.,Hellio Le Graverand-Gastineau, M.-P., Rybak, L., Kraus, V. B., Jordan, J. M.et al. (2015). Low-grade inflammation in symptomatic knee osteoarthritis:prognostic value of inflammatory plasma lipids and peripheral blood leukocytebiomarkers. Arthritis Rheumatol 67, 2905-2915.

Barboza, E., Hudson, J., Chang,W.-P., Kovats, S., Towner,R.A., Silasi-Mansat,R.,Lupu, F., Kent, C. and Griffin, T. M. (2017). Profibrotic infrapatellar fat padremodeling without M1 macrophage polarization precedes knee osteoarthritis inmice with diet-induced obesity. Arthritis Rheumatol 69, 1221-1232.

Batushansky, A., Toubiana, D. and Fait, A. (2016). Correlation-based networkgeneration, visualization, and analysis as a powerful tool in biological studies: acase study in cancer cell metabolism. BioMed Res. Int. 2016, 8313272.

Berenbaum, F., Griffin, T. M. and Liu-Bryan, R. (2017). Review: metabolicregulation of inflammation in osteoarthritis. Arthritis Rheumatol 69, 9-21.

Brown, N. M. and Setchell, K. D. R. (2001). Animal models impacted byphytoestrogens in commercial chow: implications for pathways influenced byhormones. Lab. Invest. 81, 735-747.

Cai, A., Hutchison, E., Hudson, J., Kawashima, Y., Komori, N., Singh, A.,Brush, R. S., Anderson, R. E., Sonntag, W. E., Matsumoto, H. et al. (2014).Metabolic enrichment of omega-3 polyunsaturated fatty acids does not reducethe onset of idiopathic knee osteoarthritis in mice. Osteoarthritis Cartilage22, 1301-1309.

Chassaing, B., Miles-Brown, J., Pellizzon, M., Ulman, E., Ricci, M., Zhang, L.,Patterson, A. D., Vijay-Kumar, M. and Gewirtz, A. T. (2015). Lack of soluble

12

RESEARCH ARTICLE Disease Models & Mechanisms (2018) 11, dmm034827. doi:10.1242/dmm.034827

Disea

seModels&Mechan

isms

Page 13: Independent effects of dietary fat and sucrose content on ...consistency.Basedonmaximumscores,thekneeOApathologywas moderate and did not differ between the three diets using either

fiber drives diet-induced adiposity in mice. Am. J. Physiol. Gastrointest. LiverPhysiol. 309, G528-G541.

Clockaerts, S., van Osch, G. J., Bastiaansen-Jenniskens, Y. M., Verhaar, J. A.,Van Glabbeek, F., Van Meurs, J. B., Kerkhof, H. J., Hofman, A., Stricker, B. H.and Bierma-Zeinstra, S. M. (2011). Statin use is associated with reducedincidence and progression of knee osteoarthritis in the Rotterdam study. Ann.Rheum. Dis. 71, 642-647.

Collins, K. H., Paul, H. A., Reimer, R. A., Seerattan, R. A., Hart, D. A. andHerzog, W. (2015). Relationship between inflammation, the gut microbiota, andmetabolic osteoarthritis development: studies in a rat model. OsteoarthritisCartilage 23, 1989-1998.

Courties, A., Gualillo, O., Berenbaum, F. and Sellam, J. (2015). Metabolicstress-induced joint inflammation and osteoarthritis. Osteoarthritis Cartilage23, 1955-1965.

Csardi, G. and Nepusz, T. (2006). The igraph software package for complexnetwork research. InterJournal Complex Systems 1695.

Dalby, M. J., Ross, A. W., Walker, A. W. and Morgan, P. J. (2017).Dietary uncoupling of gut microbiota and energy harvesting from obesity andglucose tolerance in mice. CellReports 21, 1521-1533.

de Munter, W., Blom, A. B., Helsen, M. M., Walgreen, B., van der Kraan, P. M.,Joosten, L. A. B., van den Berg, W. B. and van Lent, P. L. E. M. (2013).Cholesterol accumulation caused by low density lipoprotein receptor deficiency ora cholesterol-rich diet results in ectopic bone formation during experimentalosteoarthritis. Arthritis Res. Ther. 15, R178.

Deshpande, B. R., Katz, J. N., Solomon, D. H., Yelin, E. H., Hunter, D. J.,Messier, S. P., Suter, L. G. and Losina, E. (2016). Number of persons withsymptomatic knee osteoarthritis in the US: impact of race and ethnicity, age, sex,and obesity. Arthritis Care Res (Hoboken) 68, 1743-1750.

Eymard, F., Parsons, C., Edwards, M. H., Petit-Dop, F., Reginster, J.-Y.,Bruyere, O., Richette, P., Cooper, C. and Chevalier, X. (2015). Diabetes is arisk factor for knee osteoarthritis progression. Osteoarthritis Cartilage 23, 851-859.

Felson, D. T., Anderson, J. J., Naimark, A., Walker, A. M. and Meenan, R. F.(1988). Obesity and knee osteoarthritis. The Framingham study.Ann. Intern. Med.109, 18-24.

Finger, B. C., Dinan, T. G. and Cryan, J. F. (2011). High-fat diet selectivelyprotects against the effects of chronic social stress in the mouse. Neuroscience192, 351-360.

Fu, Y., Kinter, M., Hudson, J., Humphries, K. M., Lane, R. S., White, J. R.,Hakim, M., Pan, Y., Verdin, E. and Griffin, T. M. (2016). Aging promotes sirtuin3–dependent cartilage superoxide dismutase 2 acetylation and osteoarthritis.Arthritis Rheumatol 68, 1887-1898.

Griffin, T. M. and Guilak, F. (2008). Why is obesity associated with osteoarthritis?Insights from mouse models of obesity. Biorheology 45, 387-398.

Griffin, T. M., Fermor, B., Huebner, J. L., Kraus, V. B., Rodriguiz, R. M.,Wetsel, W. C., Cao, L., Setton, L. A. and Guilak, F. (2010). Diet-induced obesitydifferentially regulates behavioral, biomechanical, and molecular risk factors forosteoarthritis in mice. Arthritis Res. Ther. 12, R130.

Griffin, T. M., Huebner, J. L., Kraus, V. B., Yan, Z. and Guilak, F. (2012). Inductionof osteoarthritis and metabolic inflammation by a very high-fat diet in mice: effectsof short-term exercise. Arthritis. Rheum. 64, 443-453.

Issa, R. I. and Griffin, T. M. (2012). Pathobiology of obesity and osteoarthritis:integrating biomechanics and inflammation. Pathobiol. Aging Age Relat. Dis. 2.

Katz, J. D., Agrawal, S. and Velasquez, M. (2010). Getting to the heart of thematter: osteoarthritis takes its place as part of themetabolic syndrome.Curr. Opin.Rheumatol. 22, 512-519.

Kozijn, A. E., Gierman, L. M., van der Ham, F., Mulder, P., Morrison, M. C.,Kuhnast, S., van der Heijden, R. A., Stavro, P. M., van Koppen, A.,Pieterman, E. J. et al. (2017). Variable cartilage degradation in mice with diet-induced metabolic dysfunction: food for thought. Osteoarthritis Cartilage 1-13.

Laiguillon, M.-C., Courties, A., Houard, X., Auclair, M., Sautet, A., Capeau, J.,Feve, B., Berenbaum, F. and Sellam, J. (2015). Characterization of diabeticosteoarthritic cartilage and role of high glucose environment on chondrocyteactivation: toward pathophysiological delineation of diabetes mellitus-relatedosteoarthritis. Osteoarthritis Cartilage 23, 1513-1522.

Lee, J. R., Muckerman, J. E., Wright, A. M., Davis, D. J., Childs, T. E.,Gillespie, C. E., Vieira-Potter, V. J., Booth, F.W., Ericsson, A. C. andWill, M. J.(2017). Sex determines effect of physical activity on diet preference: association ofstriatal opioids and gut microbiota composition. Behav. Brain Res. 334, 16-25.

Lehtinen, S., Marsellach, F. X., Codlin, S., Schmidt, A., Clement-Ziza, M.,Beyer, A., Bahler, J., Orengo, C. and Pancaldi, V. (2013). Stress inducesremodelling of yeast interaction and co-expression networks. Mol. BioSyst.9, 1697-1707.

Lewis, J. S., Hembree, W. C., Furman, B. D., Tippets, L., Cattel, D.,Huebner, J. L., Little, D., DeFrate, L. E., Kraus, V. B., Guilak, F. et al. (2011).Acute joint pathology and synovial inflammation is associated with increased intra-articular fracture severity in the mouse knee.Osteoarthritis Cartilage 19, 864-873.

Lu, B., Driban, J. B., Xu, C., Lapane, K. L., McAlindon, T. E. and Eaton, C. B.(2016). Dietary fat and progression of knee osteoarthritis dietary fat intake andradiographic progression of knee osteoarthritis: data from the osteoarthritisinitiative. Arthritis Care Res (Hoboken) 69, 368-375.

Messier, S. P. (2009). Obesity and osteoarthritis: disease genesis andnonpharmacologic weight management.Med. Clin. North Am. 93, 145-159, xi-xii.

Murphy, L., Schwartz, T. A., Helmick, C. G., Renner, J. B., Tudor, G., Koch, G.,Dragomir, A., Kalsbeek, W. D., Luta, G. and Jordan, J. M. (2008). Lifetime riskof symptomatic knee osteoarthritis. Arthritis. Rheum. 59, 1207-1213.

Niu, J., Clancy, M., Aliabadi, P., Vasan, R. and Felson, D. T. (2017). Metabolicsyndrome, its components, and knee osteoarthritis: the Framinghamosteoarthritis study. Arthritis Rheumatol 69, 1194-1203.

Pavlopoulos, G. A., Secrier, M., Moschopoulos, C. N., Soldatos, T. G.,Kossida, S., Aerts, J., Schneider, R. and Bagos, P. G. (2011). Using graphtheory to analyze biological networks. BioData Min 4, 10.

Rai, M. F. and Sandell, L. J. (2011). Inflammatory mediators: tracing links betweenobesity and osteoarthritis. Crit. Rev. Eukaryot. Gene Expr. 21, 131-142.

Ratneswaran, A., LeBlanc, E. A., Walser, E., Welch, I., Mort, J. S., Borradaile, N.and Beier, F. (2015). Peroxisome proliferator-activated receptor δ promotes theprogression of posttraumatic osteoarthritis in a mouse model. Arthritis Rheumatol67, 454-464.

Ratneswaran, A., Sun, M. M.-G., Dupuis, H., Sawyez, C., Borradaile, N. andBeier, F. (2017). Nuclear receptors regulate lipid metabolism and oxidative stressmarkers in chondrocytes. J. Mol. Med. 95, 431-444.

Rindler, P. M., Plafker, S. M., Szweda, L. I. and Kinter, M. (2013). High dietary fatselectively increases catalase expression within cardiac mitochondria. J. Biol.Chem. 288, 1979-1990.

Schmittgen, T. D. and Livak, K. J. (2008). Analyzing real-time PCR data by thecomparative CT method. Nat. Protoc. 3, 1101-1108.

Schott, E. M., Farnsworth, C. W., Grier, A., Lillis, J. A., Soniwala, S.,Dadourian, G. H., Bell, R. D., Doolittle, M. L., Villani, D. A., Awad, H. et al.(2018). Targeting the gut microbiome to treat the osteoarthritis of obesity. JCIInsight 3, 95997.

Shannon, P., Markiel, A., Ozier, O., Baliga, N. S., Wang, J. T., Ramage, D.,Amin, N., Schwikowski, B. and Ideker, T. (2003). Cytoscape: a softwareenvironment for integrated models of biomolecular interaction networks. GenomeRes. 13, 2498-2504.

Shpilka, T. and Haynes, C. M. (2017). The mitochondrial UPR: mechanisms,physiological functions and implications in ageing. Nat. Rev. Mol. Cell Biol.19, 109-120.

Snowden, J. M. and Maroudas, A. (1976). The distribution of serum albumin inhuman normal and degenerate articular cartilage. Biochim. Biophys. Acta428, 726-740.

Sowers, M., Karvonen-Gutierrez, C. A., Palmieri-Smith, R., Jacobson, J. A.,Jiang, Y. and Ashton-Miller, J. A. (2009). Knee osteoarthritis in obese womenwith cardiometabolic clustering. Arthritis. Rheum. 61, 1328-1336.

Szalay, M. S., Kovacs, I. A., Korcsmaros, T., Bode, C. and Csermely, P. (2007).Stress-induced rearrangements of cellular networks: consequences for protectionand drug design. FEBS Lett. 581, 3675-3680.

Thijssen, E., van Caam, A. and van der Kraan, P. M. (2015). Obesity andosteoarthritis, more than just wear and tear: pivotal roles for inflamed adiposetissue and dyslipidaemia in obesity-induced osteoarthritis. Rheumatology (Oxf.)54, 588-600.

Warden, C. H. and Fisler, J. S. (2008). Comparisons of diets used in animal modelsof high-fat feeding. Cell Metab. 7, 277.

Wu, C.-L., Jain, D., McNeill, J. N., Little, D., Anderson, J. A., Huebner, J. L.,Kraus, V. B., Rodriguiz, R. M., Wetsel, W. C. and Guilak, F. (2015). Dietary fattyacid content regulates wound repair and the pathogenesis of osteoarthritisfollowing joint injury. Ann. Rheum. Dis. 74, 2076-2083.

Zhuo, Q., Yang, W., Chen, J. and Wang, Y. (2012). Metabolic syndrome meetsosteoarthritis. Nat. Rev. Rheumatol. 8, 729-737.

13

RESEARCH ARTICLE Disease Models & Mechanisms (2018) 11, dmm034827. doi:10.1242/dmm.034827

Disea

seModels&Mechan

isms