Javascript required
Skip to content Skip to sidebar Skip to footer

Ppargamma Expression Adipose Tissue High Fat Diet

Deficiency of PPARĪ³ in Bone Marrow Stromal Cells Does not Prevent High-Fat Diet-Induced Bone Deterioration in Mice

Jay J Cao,

Grand Forks Human Nutrition Research Center, Agricultural Research Service

, USDA, Grand Forks, ND,

USA

Search for other works by this author on:

Brian R Gregoire,

Grand Forks Human Nutrition Research Center, Agricultural Research Service

, USDA, Grand Forks, ND,

USA

Search for other works by this author on:

Kim G Michelsen,

Grand Forks Human Nutrition Research Center, Agricultural Research Service

, USDA, Grand Forks, ND,

USA

Search for other works by this author on:

Xingming Shi

Department of Neuroscience and Regenerative Medicine, Augusta University

, Augusta, GA,

USA

Department of Orthopaedic Surgery, Augusta University

, Augusta, GA,

USA

Search for other works by this author on:

Revision received:

08 April 2021

ABSTRACT

Background

Bone marrow osteoblasts and adipocytes are derived from a common mesenchymal stem cell and have a reciprocal relationship. Peroxisome proliferator-activated receptor gamma (PPARĪ³), a regulator for adipocyte differentiation, may be a potential target for reducing obesity and increasing bone mass.

Objectives

This study tested the hypothesis that bone-specific Pparg conditional knockout (cKO), via deletion of Pparg from bone marrow stromal cells (BMSC) using Osterix 1 (Osx1)-Cre, would prevent high-fat (HF) diet–induced bone deterioration in mice.

Methods

PPARĪ³ cKO (PPARĪ³fl/fl: Osx1-Cre) and floxed littermate control (PPARĪ³fl/fl Osx1-Cre ) mice that were 6 weeks old were randomly assigned to 4 groups (n = 12/group, 6 male and 6 female) and fed ad libitum with either a normal-fat (NF) purified diet (3.85 kcal/g; 10% energy as fat) or an HF diet (4.73 kcal/g; 45% energy as fat) for 6 mo. Bone structure, body composition, and serum bone-related cytokines were measured. Data were analyzed by 2-way ANOVA with Tukey post hoc comparison.

Results

The HF diet decreased the tibial and lumbar vertebrae trabecular bone volume/total volume (BV/TV) by 28% and 18%, respectively, compared to the NF diet (P < 0.01). PPARĪ³ cKO mice had 23% lower body fat mass and 9% lower lean mass than control mice. PPARĪ³ cKO mice had 41% greater tibial trabecular BV/TV compared to control mice. None of trabecular bone parameters at the second lumbar vertebra were affected by genotype. PPARĪ³ cKO mice had decreased cortical thickness compared to control mice. PPARĪ³ cKO mice had a 14% lower (P < 0.01) serum concentration of leptin and a 35% higher (P < 0.05) concentration of osteocalcin compared with control mice.

Conclusions

These data indicate that PPARĪ³ has site-specific impacts on bone structures in mice and that knockout PPARĪ³ in BMSC increased bone mass (BV/TV) in the tibia but not the lumbar vertebrae. PPARĪ³ disruption in BMSC did not prevent HF diet–induced bone deterioration in mice.

Introduction

Obesity is a major public health problem affecting 36% of the population in the United States (1), and is associated with many chronic disorders (2–4). Osteoporosis, a disease characterized by low bone mass and microarchitectural deterioration, affects about 10 million Americans over the age of 50 (5, 6). Although obesity is generally associated with increased bone mineral density and content in humans (7, 8), studies have also documented detrimental effects of obesity on bone in humans (9) and mice (10, 11).

However, the pathophysiological effects of obesity on bone are complex: the mechanisms through which obesity affects bone metabolism are not well defined, and multiple mechanisms have been proposed (9, 11, 12). One possible mechanism is that because both marrow osteoblasts (bone-forming cells) and adipocytes (energy-storing cells) are derived from a common mesenchymal stem cell (13), obesity would increase bone marrow adipocyte differentiation and fat accumulation at the expense of decreasing osteoblast differentiation and bone formation (12). In supporting this mechanism, studies have demonstrated that the decreased bone marrow osteoblastogenesis with aging or estrogen deficiency is accompanied by increased marrow adipogenesis (14, 15). Experimentally, agents that inhibit adipogenesis stimulate osteoblast differentiation, and vice versa (16–19).

Obesity affects bone metabolism through systemic or local cytokines secreted by adipocytes, especially proinflammatory cytokines. Obesity is associated with low-grade elevated chronic inflammation because adipose tissue is infiltrated with the increased amount of macrophages, which are an important source of proinflammatory cytokines (20–22). Proinflammatory cytokines, such as TNFĪ±, IL-1Ī², and IL-6, promote osteoclast activity and bone resorption (10, 23–25).

The specific mechanism controlling the fate of a mesenchymal stem cell, to either adipocytes or osteoblasts, is the area of interest for our research. Peroxisome proliferator-activated receptor gamma (PPARĪ³), as a ligand-activated nuclear receptor and a master regulator of adipocyte differentiation (26), has been considered a primary target for potential anti-osteoporosis and anti-obesity therapy (27–29).

Homozygous global PPARĪ³ deficient (PPARĪ³–/–) mice are embryonically lethal due to placental dysfunction, and heterozygous global PPARĪ³ deficient (PPARĪ³+/–) mice are resistant to high-fat (HF) diet–induced obesity and insulin resistance (30) and show a high bone mass phenotype (31). To overcome embryonic lethality and further investigate PPARĪ³ and adipocyte functions in specific tissue, mice with various tissue-specific PPARĪ³ deletions have been generated. For example, deletion of PPARĪ³ in the adipose tissue of mice protects against HF diet–induced obesity and insulin resistance (32) and increases bone formation (33).

Several mouse models with bone-specific PPARĪ³ deletion have been generated (29, 34, 35). We previously demonstrated that adult mice with bone-specific deletion of the PPARĪ³ gene from osteoprogenitor cells (PPARĪ³fl/fl: Col3.6Cre) have a higher bone mass [bone volume/total volume (BV/TV)] during development than control mice (29) and that deletion of the PPARĪ³ gene from bone marrow mesenchymal stromal cells (BMSC; PPARĪ³fl/fl: Dermo1-Cre) protects against cortical bone loss in aging mice (34).

However, the extent to which bone marrow adipocytes affect bone in diet-induced obesity has not been investigated. In this study, we hypothesized that bone marrow adipocyte deficiency would prevent bone deterioration in HF diet–induced obesity. To test this hypothesis, we generated mice in which the PPARĪ³ gene was deleted from BMSC/progenitor cells using Osterix 1 (Osx1)-Cre, which is expressed in the earliest stages of commitment to the osteoblast lineage (36).

Methods

Animals, breeding, and genotyping

Double-floxed PPARĪ³ (PPARĪ³fl/fl) mice were kindly donated by Dr. Frank J Gonzales at the National Cancer Institute (37) and rederived at The Jackson Laboratory. In addition, a breeding pair of Osx1-GFP::Cre [B6 .Cg-Tg(Sp7-tTA, tetO-EGFP/cre)1Amc/J] mice were purchased from The Jackson Laboratory. The same Osx1 Cre line has been used successfully in bone-related research (35).

Mice were housed in the Tecniplast HEPA-ventilated micro-isolator caging system in an isolated barrier room with a 12-hour light/dark cycle. The animal protocol for the study was approved by the USDA Agricultural Research Service Grand Forks Human Nutrition Research Center Institutional Animal Care and Use Committee. Animals were maintained and processed in accordance with the NIH Guide for the Care and Use of Laboratory Animals. Mice were fed Purina Rat Chow #5012 (Ralston-Purina) during the breeding period before the experiment started.

PPARĪ³ conditional knockout (cKO, PPARĪ³fl/fl: Osx1-Cre) mice were created by breeding Osx1-Cre transgenic mice with PPARĪ³fl/fl mice using standard breeding procedures as previously described (29, 34, 35, 37). Genotyping was performed around weaning and analyzed using primers and PCR conditions previously described (29, 38). In brief, ear genomic DNA was extracted and amplified with 1 forward primer (F1: 5'-CTCCAATGTTCTCAAACTTAC-3') and 2 reverse primers (R1: 5'-GATGAGTCATGTAAGTTGACC-3'; R2: 5'-GTATTCTATGGCTTCCAGTG C-3'). The expected sizes of the PCR products are ∼250 bp for a wild-type allele, ∼285 bp for a floxed allele, and ∼450 bp for a null allele. PCR analysis of Cre was performed using primers 5'-GCATTTCTGGGGATTGCTTA-3' (forward) and 5'-GTCATCCTTAGCGCCGTAAA-3' (reverse). The expected size of a PCR product for Cre is ∼350 bp.

At 6 weeks of age, floxed littermate control (PPARĪ³fl/fl Osx1-Cre) and cKO mice were randomly assigned to 4 groups (n = 12/group, 6 male and 6 female) and fed ad libitum with either a normal-fat (NF) purified diet (3.85 kcal/g; 10% energy as fat; D12450B) or an HF diet (4.73 kcal/g; 45% energy as fat with extra fat from lard; D12451), which were formulated based on AIN-93G (39) and purchased from Research Diet for 6 mo (Supplemental Table 1). The diets were formulated based on energy, with concentrations by weight of essential nutrients adjusted to be greater in the HF diet than in the NF diet. Mice had free access to diets and water throughout the study. Body mass was recorded every 2 wk and food consumption was recorded for 2 consecutive days every 2 wk.

Body composition

Body composition (fat compared with lean mass) was measured 1 d before starting the experimental diets and at the end of the study by an EchoMRI-100 whole-body composition analyzer (Echo Medical Systems, LLC) according to the manufacturer's instruction and as previously described in detail (25).

Bone structure evaluation with micro–computed tomography

The tibia and the second lumbar vertebra (LV2) of each mouse were scanned at an isotopic voxel size of 12 Ī¼m and were analyzed for bone structure using a Scanco micro–computed tomography (Ī¼CT) scanner 40 (Scanco Medical AG) with the setting as previously described (10, 40). For the tibial trabecular bone, 100 slices starting from about 0.1 mm distal to the growth plate were chosen for analyses. For the LV2 analysis, the entire secondary spongiosa between the cranial and the caudal area was scanned and analyzed. For assessment of cortical indices, a scan of 100 slices at the tibial mid-diaphysis was performed. The recommended guidelines for Ī¼CT scanning (41) and bone histomorphometry nomenclature were followed (42).

Measurements of serum biochemical markers

Serum concentrations of bone biochemical markers were determined using commercial anti-mouse ELISA kits according to the manufacturers' instructions, with kits for adiponectin and IL-6 (R&D Systems), bone-specific alkaline phosphatase (BALP, Cusabio Technology), osteocalcin (Biomedical Technologies Inc.), leptin (ALPCO Diagnostics), and tartrate-resistant acid phosphatase 5b (TRAP, Immunodiagnostic System). The same cytokine was measured for all samples with the same batch kits on the same day to minimize variation. Quality control of each bone biomarker was performed using 2 control samples in duplicate for each plate. The control samples with known concentrations (low and high values) were supplied by the respective kit manufacturers. The concentrations of the low- and high-value control samples must fall within the recommended range for the assay to be valid. The calculated CVs for biomarkers were 2.3% for adiponectin, 4.2% for IL-6, 3.2% for BALP, 3.7% for leptin, 0.6% for osteocalcin, and 1.5% for TRAP.

Measurement of mRNA in bone

Total RNA was obtained from pulverized bone samples that included bone marrow using Trizol (Invitrogen), reverse transcribed, and amplified and quantified using a Sequence Detection System (SDS 7300) as previously described (10). Relative mRNA expression was normalized to the expression of Gapdh mRNA in the same sample. The sequence information of oligonucleotide primers is presented in Supplemental Table 2. All oligonucleotide primers for PCR amplification were designed using PrimerQuest software, synthesized, and purified by HPLC (Integrated DNA Technologies).

Statistical analyses

Data are expressed as means ± SDs. The main effects of dietary fat, genotype, sex, and their interactions were analyzed using 3-factor ANOVA with JMP (version 15.0.0, SAS Institute, Inc.). Data from both sexes were then pooled and analyzed using 2-factor ANOVA (or repeated 2-factor ANOVA for body weight changes in Supplemental Figure 1) because no interactions were detected between sex and fat level or genotype. Tukey contrasts were performed to compare group means if the fat-by-genotype interaction was significant. In all analyses, P values ≤ 0.05 and > 0.05 were considered statistically significant and nonsignificant, respectively. A power analysis indicated that detecting a significant difference in the primary outcome variable of tibial BV/TV (3 percentage points) among the treatments with 0.90 power and an alpha of 0.05 (10) would require 12 mice/group.

Results

There were no differences in initial body weight between the NF and HF groups, but PPARĪ³ cKO mice had 11% lower body weight than control mice (Figure 1A). All animals gained weight (the body weight changes are presented in Supplemental Figure 1). Body masses of mice fed the HF diet were significantly greater than the masses of those fed the NF diet (P < 0.01). Compared to those in the floxed littermate control group, PPARĪ³ cKO mice had a 15% lower body mass at the end of study (P < 0.05). We further examined the extent to which the HF diet or genotype affected body composition. The HF diet increased the fat mass for both floxed littermate control and cKO mice (P < 0.05) but did not affect the lean body mass (P > 0.05; Figure 1B andC). Floxed littermate control mice had more fat mass than PPARĪ³ cKO mice before the study (P < 0.01) and at the end of the study (P = 0.05). Floxed littermate control mice also had more lean mass at the beginning (P < 0.05) and the end of the study (P < 0.05) than PPARĪ³ cKO mice.

FIGURE 1

(A) Body mass, (B) fat mass, (C) lean mass, and (D) food and energy intake of floxed littermate control (PPARĪ³fl/fl Osx1-Cre–) or PPARĪ³ cKO (PPARĪ³fl/fl: Osx1-Cre) mice fed either a purified normal-fat (10% energy as fat) or a high-fat (45% energy as fat) diet for 6 mo. Values are means ± SDs (n = 12). The main effects of dietary fat and genotype and their interactions were analyzed using 2-factor ANOVA. Abbreviations: C-HF, control mice fed a high-fat diet; C-NF, control mice fed a normal-fat diet; cKO, conditional knockout; cKO-NF, conditional knockout mice fed a NF diet; cKO-HF, conditional knockout mice fed an HF diet.

(A) Body mass, (B) fat mass, (C) lean mass, and (D) food and energy intake of floxed littermate control (PPARĪ³fl/fl Osx1-Cre) or PPARĪ³ cKO (PPARĪ³fl/fl: Osx1-Cre) mice fed either a purified normal-fat (10% energy as fat) or a high-fat (45% energy as fat) diet for 6 mo. Values are means ± SDs (n = 12). The main effects of dietary fat and genotype and their interactions were analyzed using 2-factor ANOVA. Abbreviations: C-HF, control mice fed a high-fat diet; C-NF, control mice fed a normal-fat diet; cKO, conditional knockout; cKO-NF, conditional knockout mice fed a NF diet; cKO-HF, conditional knockout mice fed an HF diet.

FIGURE 1

(A) Body mass, (B) fat mass, (C) lean mass, and (D) food and energy intake of floxed littermate control (PPARĪ³fl/fl Osx1-Cre–) or PPARĪ³ cKO (PPARĪ³fl/fl: Osx1-Cre) mice fed either a purified normal-fat (10% energy as fat) or a high-fat (45% energy as fat) diet for 6 mo. Values are means ± SDs (n = 12). The main effects of dietary fat and genotype and their interactions were analyzed using 2-factor ANOVA. Abbreviations: C-HF, control mice fed a high-fat diet; C-NF, control mice fed a normal-fat diet; cKO, conditional knockout; cKO-NF, conditional knockout mice fed a NF diet; cKO-HF, conditional knockout mice fed an HF diet.

(A) Body mass, (B) fat mass, (C) lean mass, and (D) food and energy intake of floxed littermate control (PPARĪ³fl/fl Osx1-Cre) or PPARĪ³ cKO (PPARĪ³fl/fl: Osx1-Cre) mice fed either a purified normal-fat (10% energy as fat) or a high-fat (45% energy as fat) diet for 6 mo. Values are means ± SDs (n = 12). The main effects of dietary fat and genotype and their interactions were analyzed using 2-factor ANOVA. Abbreviations: C-HF, control mice fed a high-fat diet; C-NF, control mice fed a normal-fat diet; cKO, conditional knockout; cKO-NF, conditional knockout mice fed a NF diet; cKO-HF, conditional knockout mice fed an HF diet.

The mean food intake (Figure 1D) by animals fed the HF diets was lower than the intake by those fed the NF diet (P < 0.01) but the total energy intake was similar between groups (P = 0.80) due to the higher energy content of the HF diet (3.85 kcal/g compared with 4.73 kcal/g for the NF and HF diets, respectively). Since the diet was formulated based on energy, with the concentrations by weight of essential nutrients adjusted to be greater in the HF diet than that in the NF diet, while food intake was lower for the mice fed the HF diet compared to those fed the NF diet (2.85 g compared with 3.53 g for the HF and NF diets, respectively), the intake of micronutrients, including Ca, was the same for both groups (Supplemental Table 3). PPARĪ³ cKO mice had reduced food intake and energy intake compared with floxed littermate control mice (P < 0.05).

The effects of the HF diet and genotype on bone structures in the proximal tibia, mid-diaphysis of the tibia, and LV2 were evaluated with Ī¼CT (Table 1;Figure 2). As expected, the HF diet adversely affected the 3-dimensional bone microstructure. Compared with the NF diet, the HF diet decreased the tibial and lumbar vertebrae trabecular BV/TV (P < 0.01) by 28% and 18%, respectively. Mice fed the HF diet also had lower bone mineral density (BMD) at the tibia (P < 0.01) and LV2 (P  = 0.01). The HF diet did not affect any tibial mid-diaphysis parameters. PPARĪ³ cKO mice had greater tibial BV/TV, a higher trabecular number (Tb.N), greater connectivity density (Conn.Dn), a higher BMD, and a lower structure model index (SMI) compared to control mice. None of the trabecular bone parameters at LV2 were affected by genotype. PPARĪ³ cKO mice had a lower bone area, total area, medullary area (Me.Ar), bone perimeter, cortical thickness, and BMD compared to control mice. There were no significant interactions between diet and genotype on other bone parameters (P > 0.05), except the tibial structure mode index (SMI; P < 0.05). PPARĪ³ cKO mice fed the HF diet had a higher SMI than those fed the NF diet, indicating the adverse effect of the HF diet on the bones of PPARĪ³ cKO mice.

FIGURE 2

Representative images of (A) proximal tibia trabecular bone, (B) tibial mid-diaphysis cortical bone, and (C) the second lumbar vertebra trabecular bone (data are presented in Table 1) of floxed littermate control (PPARĪ³fl/fl Osx1-Cre–) or PPARĪ³ cKO (PPARĪ³fl/fl: Osx1-Cre) mice fed either a purified normal-fat (10% energy as fat) or a high-fat (45% energy as fat) diet for 6 mo. For the tibial trabecular bone, 100 slices starting from about 0.1 mm distal to the growth plate were chosen for analyses. For the second lumbar vertebra analysis, the entire secondary spongiosa between the cranial and the caudal area was scanned and analyzed. For assessment of cortical indices, a scan of 100 slices at the tibial mid-diaphysis was performed. Abbreviations: C-HF, control mice fed a high-fat diet; C-NF, control mice fed a normal-fat diet; cKO, conditional knockout; cKO-NF, conditional knockout mice fed a NF diet; cKO-HF, conditional knockout mice fed an HF diet.

Representative images of (A) proximal tibia trabecular bone, (B) tibial mid-diaphysis cortical bone, and (C) the second lumbar vertebra trabecular bone (data are presented in Table 1) of floxed littermate control (PPARĪ³fl/fl Osx1-Cre) or PPARĪ³ cKO (PPARĪ³fl/fl: Osx1-Cre) mice fed either a purified normal-fat (10% energy as fat) or a high-fat (45% energy as fat) diet for 6 mo. For the tibial trabecular bone, 100 slices starting from about 0.1 mm distal to the growth plate were chosen for analyses. For the second lumbar vertebra analysis, the entire secondary spongiosa between the cranial and the caudal area was scanned and analyzed. For assessment of cortical indices, a scan of 100 slices at the tibial mid-diaphysis was performed. Abbreviations: C-HF, control mice fed a high-fat diet; C-NF, control mice fed a normal-fat diet; cKO, conditional knockout; cKO-NF, conditional knockout mice fed a NF diet; cKO-HF, conditional knockout mice fed an HF diet.

FIGURE 2

Representative images of (A) proximal tibia trabecular bone, (B) tibial mid-diaphysis cortical bone, and (C) the second lumbar vertebra trabecular bone (data are presented in Table 1) of floxed littermate control (PPARĪ³fl/fl Osx1-Cre–) or PPARĪ³ cKO (PPARĪ³fl/fl: Osx1-Cre) mice fed either a purified normal-fat (10% energy as fat) or a high-fat (45% energy as fat) diet for 6 mo. For the tibial trabecular bone, 100 slices starting from about 0.1 mm distal to the growth plate were chosen for analyses. For the second lumbar vertebra analysis, the entire secondary spongiosa between the cranial and the caudal area was scanned and analyzed. For assessment of cortical indices, a scan of 100 slices at the tibial mid-diaphysis was performed. Abbreviations: C-HF, control mice fed a high-fat diet; C-NF, control mice fed a normal-fat diet; cKO, conditional knockout; cKO-NF, conditional knockout mice fed a NF diet; cKO-HF, conditional knockout mice fed an HF diet.

Representative images of (A) proximal tibia trabecular bone, (B) tibial mid-diaphysis cortical bone, and (C) the second lumbar vertebra trabecular bone (data are presented in Table 1) of floxed littermate control (PPARĪ³fl/fl Osx1-Cre) or PPARĪ³ cKO (PPARĪ³fl/fl: Osx1-Cre) mice fed either a purified normal-fat (10% energy as fat) or a high-fat (45% energy as fat) diet for 6 mo. For the tibial trabecular bone, 100 slices starting from about 0.1 mm distal to the growth plate were chosen for analyses. For the second lumbar vertebra analysis, the entire secondary spongiosa between the cranial and the caudal area was scanned and analyzed. For assessment of cortical indices, a scan of 100 slices at the tibial mid-diaphysis was performed. Abbreviations: C-HF, control mice fed a high-fat diet; C-NF, control mice fed a normal-fat diet; cKO, conditional knockout; cKO-NF, conditional knockout mice fed a NF diet; cKO-HF, conditional knockout mice fed an HF diet.

TABLE 1

Bone structural properties of proximal tibia and lumbar vertebrae from floxed littermate control or PPARĪ³ cKO mice fed either a purified normal-fat or a high-fat diet for 6 mo 1

ANOVA (P value)
Indices C-NF cKO-NF C-HF cKO-HF Fat Genotype Fat × Genotype
Proximal tibia (trabecular bone)
 BV/TV, % 7.47 ± 1.88 9.07 ± 3.75 4.34 ± 1.38 7.60 ± 3.26 <0.01 <0.01 0.30
 Tb.N, mm–1 2.72 ± 0.56 3.52 ± 1.20 2.44 ± 0.72 2.99 ± 1.51 0.19 0.03 0.70
 Tb.Th, mm 0.06 ± 0.01 0.05 ± 0.01 0.06 ± 0.01 0.05 ± 0.01 0.99 0.47 0.65
 Tb.Sp, mm 0.38 ± 0.08 0.32 ± 0.13 0.44 ± 0.12 0.41 ± 0.17 0.05 0.16 0.69
 Conn.Dn, mm–3 18.3 ± 17.9 44.0 ± 35.9 5.3 ± 4.2 40.7 ± 53.4 0.40 <0.01 0.61
 SMI 2.64 ± 0.48ab 2.46 ± 0.31b 3.06 ± 0.43a 2.32 ± 0.39b 0.24 <0.01 <0.05
 BMD, mg hydroxyapatite/ccm 115 ± 23 140 ± 44 74 ± 32 115 ± 45 <0.01 <0.01 0.43
Tibial mid-diaphysis (cortical bone)
 B.Ar, mm2 0.73 ± 0.06 0.65 ± 0.05 0.71 ± 0.06 0.63 ± 0.06 0.24 <0.01 0.90
 T.Ar, mm2 1.08 ± 0.10 0.97 ± 0.08 1.05 ± 0.08 0.94 ± 0.09 0.29 <0.01 0.97
 Me.Ar, mm2 0.35 ± 0.07 0.31 ± 0.04 0.34 ± 0.04 0.30 ± 0.04 0.51 0.01 0.95
 B.Pm, mm 6.3 ± 0.4 6.0 ± 0.4 6.2 ± 0.3 5.8 ± 0.4 0.43 <0.01 0.71
 Ct.Th, mm 0.26 ± 0.03 0.23 ± 0.01 0.24 ± 0.01 0.23 ± 0.01 0.19 <0.01 0.30
 BMD, mg hydroxyapatite/ccm 787 ± 70 748 ± 38 772 ± 41 750 ± 33 0.64 0.03 0.51
Second lumbar vertebra (trabecular bone)
 BV/TV, % 22.3 ± 6.6 21.3 ± 3.0 18.0 ± 5.5 17.5 ± 3.3 <0.01 0.62 0.85
 Tb.N, mm–1 4.3 ± 0.7 4.5 ± 0.7 4.3 ± 0.7 4.1 ± 0.9 0.33 0.87 0.36
 Tb.Th, mm 0.05 ± 0.01 0.05 ± 0.01 0.05 ± 0.01 0.05 ± 0.01 0.45 0.50 0.22
 Tb.Sp, mm 0.23 ± 0.04 0.22 ± 0.04 0.23 ± 0.04 0.25 ± 0.05 0.24 0.79 0.38
 Conn.Dn, mm–3 116 ± 36 152 ± 44 120 ± 46 119 ± 72 0.32 0.25 0.24
 SMI 0.63 ± 0.77 0.70 ± 0.45 1.16 ± 0.75 1.16 ± 0.42 <0.01 0.84 0.86
 BMD, mg hydroxyapatite/ccm 240 ± 59 232 ± 28 203 ± 50 200 ± 32 0.01 0.68 0.87
ANOVA (P value)
Indices C-NF cKO-NF C-HF cKO-HF Fat Genotype Fat × Genotype
Proximal tibia (trabecular bone)
 BV/TV, % 7.47 ± 1.88 9.07 ± 3.75 4.34 ± 1.38 7.60 ± 3.26 <0.01 <0.01 0.30
 Tb.N, mm–1 2.72 ± 0.56 3.52 ± 1.20 2.44 ± 0.72 2.99 ± 1.51 0.19 0.03 0.70
 Tb.Th, mm 0.06 ± 0.01 0.05 ± 0.01 0.06 ± 0.01 0.05 ± 0.01 0.99 0.47 0.65
 Tb.Sp, mm 0.38 ± 0.08 0.32 ± 0.13 0.44 ± 0.12 0.41 ± 0.17 0.05 0.16 0.69
 Conn.Dn, mm–3 18.3 ± 17.9 44.0 ± 35.9 5.3 ± 4.2 40.7 ± 53.4 0.40 <0.01 0.61
 SMI 2.64 ± 0.48ab 2.46 ± 0.31b 3.06 ± 0.43a 2.32 ± 0.39b 0.24 <0.01 <0.05
 BMD, mg hydroxyapatite/ccm 115 ± 23 140 ± 44 74 ± 32 115 ± 45 <0.01 <0.01 0.43
Tibial mid-diaphysis (cortical bone)
 B.Ar, mm2 0.73 ± 0.06 0.65 ± 0.05 0.71 ± 0.06 0.63 ± 0.06 0.24 <0.01 0.90
 T.Ar, mm2 1.08 ± 0.10 0.97 ± 0.08 1.05 ± 0.08 0.94 ± 0.09 0.29 <0.01 0.97
 Me.Ar, mm2 0.35 ± 0.07 0.31 ± 0.04 0.34 ± 0.04 0.30 ± 0.04 0.51 0.01 0.95
 B.Pm, mm 6.3 ± 0.4 6.0 ± 0.4 6.2 ± 0.3 5.8 ± 0.4 0.43 <0.01 0.71
 Ct.Th, mm 0.26 ± 0.03 0.23 ± 0.01 0.24 ± 0.01 0.23 ± 0.01 0.19 <0.01 0.30
 BMD, mg hydroxyapatite/ccm 787 ± 70 748 ± 38 772 ± 41 750 ± 33 0.64 0.03 0.51
Second lumbar vertebra (trabecular bone)
 BV/TV, % 22.3 ± 6.6 21.3 ± 3.0 18.0 ± 5.5 17.5 ± 3.3 <0.01 0.62 0.85
 Tb.N, mm–1 4.3 ± 0.7 4.5 ± 0.7 4.3 ± 0.7 4.1 ± 0.9 0.33 0.87 0.36
 Tb.Th, mm 0.05 ± 0.01 0.05 ± 0.01 0.05 ± 0.01 0.05 ± 0.01 0.45 0.50 0.22
 Tb.Sp, mm 0.23 ± 0.04 0.22 ± 0.04 0.23 ± 0.04 0.25 ± 0.05 0.24 0.79 0.38
 Conn.Dn, mm–3 116 ± 36 152 ± 44 120 ± 46 119 ± 72 0.32 0.25 0.24
 SMI 0.63 ± 0.77 0.70 ± 0.45 1.16 ± 0.75 1.16 ± 0.42 <0.01 0.84 0.86
 BMD, mg hydroxyapatite/ccm 240 ± 59 232 ± 28 203 ± 50 200 ± 32 0.01 0.68 0.87

Abbreviations: B.Ar, cross-sectional bone area; BMD, bone mineral density; B.Pm, bone perimeter; BV, bone volume; C-HF, control mice fed a high-fat diet; C-NF, control mice fed a normal-fat diet; cKO, conditional knockout; cKO-HF, conditional knockout mice fed a high-fat diet; cKO-NF, conditional knockout mice fed a normal-fat diet; Conn.Dn, connectivity density; Ct.Th, cortical thickness; Me.Ar, cross-sectional medullary area; SMI, structure model index; T.Ar, cross-sectional total area; Tb.N, trabecular number; Tb.Sp, trabecular separation; Tb.Th, trabecular thickness; TV, total volume.

1

Values are means ± SDs (n = 12). Labeled means in a row without a common letter differ at a P value ≤ 0.05. The main effects of dietary fat and genotype and their interactions were analyzed using a 2-factor ANOVA. Control mice were PPARĪ³fl/fl Osx1-Cre mice and cKO mice were PPARĪ³fl/fl: Osx1-Cre mice.

TABLE 1

Bone structural properties of proximal tibia and lumbar vertebrae from floxed littermate control or PPARĪ³ cKO mice fed either a purified normal-fat or a high-fat diet for 6 mo 1

ANOVA (P value)
Indices C-NF cKO-NF C-HF cKO-HF Fat Genotype Fat × Genotype
Proximal tibia (trabecular bone)
 BV/TV, % 7.47 ± 1.88 9.07 ± 3.75 4.34 ± 1.38 7.60 ± 3.26 <0.01 <0.01 0.30
 Tb.N, mm–1 2.72 ± 0.56 3.52 ± 1.20 2.44 ± 0.72 2.99 ± 1.51 0.19 0.03 0.70
 Tb.Th, mm 0.06 ± 0.01 0.05 ± 0.01 0.06 ± 0.01 0.05 ± 0.01 0.99 0.47 0.65
 Tb.Sp, mm 0.38 ± 0.08 0.32 ± 0.13 0.44 ± 0.12 0.41 ± 0.17 0.05 0.16 0.69
 Conn.Dn, mm–3 18.3 ± 17.9 44.0 ± 35.9 5.3 ± 4.2 40.7 ± 53.4 0.40 <0.01 0.61
 SMI 2.64 ± 0.48ab 2.46 ± 0.31b 3.06 ± 0.43a 2.32 ± 0.39b 0.24 <0.01 <0.05
 BMD, mg hydroxyapatite/ccm 115 ± 23 140 ± 44 74 ± 32 115 ± 45 <0.01 <0.01 0.43
Tibial mid-diaphysis (cortical bone)
 B.Ar, mm2 0.73 ± 0.06 0.65 ± 0.05 0.71 ± 0.06 0.63 ± 0.06 0.24 <0.01 0.90
 T.Ar, mm2 1.08 ± 0.10 0.97 ± 0.08 1.05 ± 0.08 0.94 ± 0.09 0.29 <0.01 0.97
 Me.Ar, mm2 0.35 ± 0.07 0.31 ± 0.04 0.34 ± 0.04 0.30 ± 0.04 0.51 0.01 0.95
 B.Pm, mm 6.3 ± 0.4 6.0 ± 0.4 6.2 ± 0.3 5.8 ± 0.4 0.43 <0.01 0.71
 Ct.Th, mm 0.26 ± 0.03 0.23 ± 0.01 0.24 ± 0.01 0.23 ± 0.01 0.19 <0.01 0.30
 BMD, mg hydroxyapatite/ccm 787 ± 70 748 ± 38 772 ± 41 750 ± 33 0.64 0.03 0.51
Second lumbar vertebra (trabecular bone)
 BV/TV, % 22.3 ± 6.6 21.3 ± 3.0 18.0 ± 5.5 17.5 ± 3.3 <0.01 0.62 0.85
 Tb.N, mm–1 4.3 ± 0.7 4.5 ± 0.7 4.3 ± 0.7 4.1 ± 0.9 0.33 0.87 0.36
 Tb.Th, mm 0.05 ± 0.01 0.05 ± 0.01 0.05 ± 0.01 0.05 ± 0.01 0.45 0.50 0.22
 Tb.Sp, mm 0.23 ± 0.04 0.22 ± 0.04 0.23 ± 0.04 0.25 ± 0.05 0.24 0.79 0.38
 Conn.Dn, mm–3 116 ± 36 152 ± 44 120 ± 46 119 ± 72 0.32 0.25 0.24
 SMI 0.63 ± 0.77 0.70 ± 0.45 1.16 ± 0.75 1.16 ± 0.42 <0.01 0.84 0.86
 BMD, mg hydroxyapatite/ccm 240 ± 59 232 ± 28 203 ± 50 200 ± 32 0.01 0.68 0.87
ANOVA (P value)
Indices C-NF cKO-NF C-HF cKO-HF Fat Genotype Fat × Genotype
Proximal tibia (trabecular bone)
 BV/TV, % 7.47 ± 1.88 9.07 ± 3.75 4.34 ± 1.38 7.60 ± 3.26 <0.01 <0.01 0.30
 Tb.N, mm–1 2.72 ± 0.56 3.52 ± 1.20 2.44 ± 0.72 2.99 ± 1.51 0.19 0.03 0.70
 Tb.Th, mm 0.06 ± 0.01 0.05 ± 0.01 0.06 ± 0.01 0.05 ± 0.01 0.99 0.47 0.65
 Tb.Sp, mm 0.38 ± 0.08 0.32 ± 0.13 0.44 ± 0.12 0.41 ± 0.17 0.05 0.16 0.69
 Conn.Dn, mm–3 18.3 ± 17.9 44.0 ± 35.9 5.3 ± 4.2 40.7 ± 53.4 0.40 <0.01 0.61
 SMI 2.64 ± 0.48ab 2.46 ± 0.31b 3.06 ± 0.43a 2.32 ± 0.39b 0.24 <0.01 <0.05
 BMD, mg hydroxyapatite/ccm 115 ± 23 140 ± 44 74 ± 32 115 ± 45 <0.01 <0.01 0.43
Tibial mid-diaphysis (cortical bone)
 B.Ar, mm2 0.73 ± 0.06 0.65 ± 0.05 0.71 ± 0.06 0.63 ± 0.06 0.24 <0.01 0.90
 T.Ar, mm2 1.08 ± 0.10 0.97 ± 0.08 1.05 ± 0.08 0.94 ± 0.09 0.29 <0.01 0.97
 Me.Ar, mm2 0.35 ± 0.07 0.31 ± 0.04 0.34 ± 0.04 0.30 ± 0.04 0.51 0.01 0.95
 B.Pm, mm 6.3 ± 0.4 6.0 ± 0.4 6.2 ± 0.3 5.8 ± 0.4 0.43 <0.01 0.71
 Ct.Th, mm 0.26 ± 0.03 0.23 ± 0.01 0.24 ± 0.01 0.23 ± 0.01 0.19 <0.01 0.30
 BMD, mg hydroxyapatite/ccm 787 ± 70 748 ± 38 772 ± 41 750 ± 33 0.64 0.03 0.51
Second lumbar vertebra (trabecular bone)
 BV/TV, % 22.3 ± 6.6 21.3 ± 3.0 18.0 ± 5.5 17.5 ± 3.3 <0.01 0.62 0.85
 Tb.N, mm–1 4.3 ± 0.7 4.5 ± 0.7 4.3 ± 0.7 4.1 ± 0.9 0.33 0.87 0.36
 Tb.Th, mm 0.05 ± 0.01 0.05 ± 0.01 0.05 ± 0.01 0.05 ± 0.01 0.45 0.50 0.22
 Tb.Sp, mm 0.23 ± 0.04 0.22 ± 0.04 0.23 ± 0.04 0.25 ± 0.05 0.24 0.79 0.38
 Conn.Dn, mm–3 116 ± 36 152 ± 44 120 ± 46 119 ± 72 0.32 0.25 0.24
 SMI 0.63 ± 0.77 0.70 ± 0.45 1.16 ± 0.75 1.16 ± 0.42 <0.01 0.84 0.86
 BMD, mg hydroxyapatite/ccm 240 ± 59 232 ± 28 203 ± 50 200 ± 32 0.01 0.68 0.87

Abbreviations: B.Ar, cross-sectional bone area; BMD, bone mineral density; B.Pm, bone perimeter; BV, bone volume; C-HF, control mice fed a high-fat diet; C-NF, control mice fed a normal-fat diet; cKO, conditional knockout; cKO-HF, conditional knockout mice fed a high-fat diet; cKO-NF, conditional knockout mice fed a normal-fat diet; Conn.Dn, connectivity density; Ct.Th, cortical thickness; Me.Ar, cross-sectional medullary area; SMI, structure model index; T.Ar, cross-sectional total area; Tb.N, trabecular number; Tb.Sp, trabecular separation; Tb.Th, trabecular thickness; TV, total volume.

1

Values are means ± SDs (n = 12). Labeled means in a row without a common letter differ at a P value ≤ 0.05. The main effects of dietary fat and genotype and their interactions were analyzed using a 2-factor ANOVA. Control mice were PPARĪ³fl/fl Osx1-Cre mice and cKO mice were PPARĪ³fl/fl: Osx1-Cre mice.

Mice fed the HF diet had higher (P < 0.01) serum concentrations of leptin and the bone resorption marker TRAP and a lower (P < 0.05) concentration of osteocalcin than those fed the NF diet (Figure 3). Compared with control mice, PPARĪ³ cKO mice had a 14% lower (P < 0.01) serum concentration of leptin and a 35% higher (P < 0.05) concentration of osteocalcin. There were no differences in serum concentrations of adiponectin, BALP, and IL-6 between mice fed the HF and NF diets or between control and PPARĪ³ cKO mice.

FIGURE 3

Serum concentrations of (A) leptin, (B) adiponectin, (C) BALP, (D) IL-6, (E) osteocalcin, and (F) TRAP of floxed littermate control (PPARĪ³fl/fl Osx1-Cre–) or PPARĪ³ cKO (PPARĪ³fl/fl: Osx1-Cre) mice fed either a purified normal-fat (10% energy as fat) or a high-fat (45% energy as fat) diet for 6 mo. Values are means ± SDs (n = 12). The main effects of dietary fat and genotype and their interactions were analyzed using 2-factor ANOVA. Abbreviations: BALP, bone-specific alkaline phosphatase; C-HF, control mice fed a high-fat diet; C-NF, control mice fed a normal-fat diet; cKO, conditional knockout; cKO-NF, conditional knockout mice fed a normal-fat diet; cKO-HF, conditional knockout mice fed a high-fat diet; TRAP, tartrate-resistant acid phosphatase.

Serum concentrations of (A) leptin, (B) adiponectin, (C) BALP, (D) IL-6, (E) osteocalcin, and (F) TRAP of floxed littermate control (PPARĪ³fl/fl Osx1-Cre) or PPARĪ³ cKO (PPARĪ³fl/fl: Osx1-Cre) mice fed either a purified normal-fat (10% energy as fat) or a high-fat (45% energy as fat) diet for 6 mo. Values are means ± SDs (n = 12). The main effects of dietary fat and genotype and their interactions were analyzed using 2-factor ANOVA. Abbreviations: BALP, bone-specific alkaline phosphatase; C-HF, control mice fed a high-fat diet; C-NF, control mice fed a normal-fat diet; cKO, conditional knockout; cKO-NF, conditional knockout mice fed a normal-fat diet; cKO-HF, conditional knockout mice fed a high-fat diet; TRAP, tartrate-resistant acid phosphatase.

FIGURE 3

Serum concentrations of (A) leptin, (B) adiponectin, (C) BALP, (D) IL-6, (E) osteocalcin, and (F) TRAP of floxed littermate control (PPARĪ³fl/fl Osx1-Cre–) or PPARĪ³ cKO (PPARĪ³fl/fl: Osx1-Cre) mice fed either a purified normal-fat (10% energy as fat) or a high-fat (45% energy as fat) diet for 6 mo. Values are means ± SDs (n = 12). The main effects of dietary fat and genotype and their interactions were analyzed using 2-factor ANOVA. Abbreviations: BALP, bone-specific alkaline phosphatase; C-HF, control mice fed a high-fat diet; C-NF, control mice fed a normal-fat diet; cKO, conditional knockout; cKO-NF, conditional knockout mice fed a normal-fat diet; cKO-HF, conditional knockout mice fed a high-fat diet; TRAP, tartrate-resistant acid phosphatase.

Serum concentrations of (A) leptin, (B) adiponectin, (C) BALP, (D) IL-6, (E) osteocalcin, and (F) TRAP of floxed littermate control (PPARĪ³fl/fl Osx1-Cre) or PPARĪ³ cKO (PPARĪ³fl/fl: Osx1-Cre) mice fed either a purified normal-fat (10% energy as fat) or a high-fat (45% energy as fat) diet for 6 mo. Values are means ± SDs (n = 12). The main effects of dietary fat and genotype and their interactions were analyzed using 2-factor ANOVA. Abbreviations: BALP, bone-specific alkaline phosphatase; C-HF, control mice fed a high-fat diet; C-NF, control mice fed a normal-fat diet; cKO, conditional knockout; cKO-NF, conditional knockout mice fed a normal-fat diet; cKO-HF, conditional knockout mice fed a high-fat diet; TRAP, tartrate-resistant acid phosphatase.

The HF diet increased Pparg (P < 0.05) and leptin (P < 0.01) expression in whole bone with marrow (P < 0.01;Figure 4), and there were no differences in their expressions between control and PPARĪ³ cKO mice (P > 0.05).

FIGURE 4

Expression of Pparg and Lep in femurs of floxed littermate control (PPARĪ³fl/fl Osx1-Cre–) or PPARĪ³ cKO (PPARĪ³fl/fl: Osx1-Cre) mice fed either a purified normal-fat (10% energy as fat) or a high-fat (45% energy as fat) diet for 6 mo. Values are means ± SDs (n = 12). The main effects of dietary fat and genotype and their interactions were analyzed using 2-factor ANOVA. Abbreviations: C-HF, control mice fed a high-fat diet; C-NF, control mice fed a normal-fat diet; cKO, conditional knockout; cKO-NF, conditional knockout mice fed a normal-fat diet; cKO-HF, conditional knockout mice fed a high-fat diet; Lep, leptin.

Expression of Pparg and Lep in femurs of floxed littermate control (PPARĪ³fl/fl Osx1-Cre) or PPARĪ³ cKO (PPARĪ³fl/fl: Osx1-Cre) mice fed either a purified normal-fat (10% energy as fat) or a high-fat (45% energy as fat) diet for 6 mo. Values are means ± SDs (n = 12). The main effects of dietary fat and genotype and their interactions were analyzed using 2-factor ANOVA. Abbreviations: C-HF, control mice fed a high-fat diet; C-NF, control mice fed a normal-fat diet; cKO, conditional knockout; cKO-NF, conditional knockout mice fed a normal-fat diet; cKO-HF, conditional knockout mice fed a high-fat diet; Lep, leptin.

FIGURE 4

Expression of Pparg and Lep in femurs of floxed littermate control (PPARĪ³fl/fl Osx1-Cre–) or PPARĪ³ cKO (PPARĪ³fl/fl: Osx1-Cre) mice fed either a purified normal-fat (10% energy as fat) or a high-fat (45% energy as fat) diet for 6 mo. Values are means ± SDs (n = 12). The main effects of dietary fat and genotype and their interactions were analyzed using 2-factor ANOVA. Abbreviations: C-HF, control mice fed a high-fat diet; C-NF, control mice fed a normal-fat diet; cKO, conditional knockout; cKO-NF, conditional knockout mice fed a normal-fat diet; cKO-HF, conditional knockout mice fed a high-fat diet; Lep, leptin.

Expression of Pparg and Lep in femurs of floxed littermate control (PPARĪ³fl/fl Osx1-Cre) or PPARĪ³ cKO (PPARĪ³fl/fl: Osx1-Cre) mice fed either a purified normal-fat (10% energy as fat) or a high-fat (45% energy as fat) diet for 6 mo. Values are means ± SDs (n = 12). The main effects of dietary fat and genotype and their interactions were analyzed using 2-factor ANOVA. Abbreviations: C-HF, control mice fed a high-fat diet; C-NF, control mice fed a normal-fat diet; cKO, conditional knockout; cKO-NF, conditional knockout mice fed a normal-fat diet; cKO-HF, conditional knockout mice fed a high-fat diet; Lep, leptin.

Discussion

In this study, we examined the role of marrow adipocytes in bone-related changes in an HF diet–induced obese mouse model using animals carrying a deletion of the PPARĪ³ gene in Osx1-expressing bone marrow stromal cells and using their floxed littermate as controls. Our data showed that Pparg cKO mice (PPARĪ³fl/fl: Osx1-Cre) have an increased trabecular bone mass (BV/TV) in the proximal tibia and an increased serum concentration of osteocalcin, a bone formation marker, but with decreased fat mass and a lower serum leptin concentration, compared to their littermate controls. We found that Pparg cKO mice were not protected from the detrimental impact of an HF diet on bone structure, a finding different from our original hypothesis.

As expected, we found that the HF diet increased the body mass but negatively affected the bone structure at the tibia and LV2, which has been repeatedly demonstrated (43–46). Similarly, leptin-deficient (ob/ob) mice are obese and weigh twice as much as wild-type animals but have a lower bone mass at the weight-bearing bone femur (47). Apparently, the mechanical loading and stimulation of bone formation conferred by body weight does not counter the detrimental effects of excessive adiposity on bone (47). The mechanisms through which obesity or adiposity negatively affect bone health have been extensively discussed and reviewed (9, 11, 12). Obesity in humans is multifaceted and the micronutrient intake is not always at the recommended level; the use of rodent models of obesity allows us to control the intake of nutrients and to delineate the mechanisms underlying the negative impact of adiposity upon bone health.

The noteworthy finding of this study is that, contrary to our hypothesis, our data show that the knockout of Pparg in BMSC improved bone outcome measures but did not protect against bone deterioration in HF diet–induced obesity. Bone is a dynamic organ that undergoes significant turnover via a process involving bone resorption by osteoclasts and bone formation by osteoblasts (48). HF diet–induced obesity is known to increase the production of proinflammatory cytokines and increase osteoclast-mediated bone resorption (24, 25, 49), despite the increase in body weight or osteoblast number or activity (10, 24). Considering osteoclast differentiation and function are not affected by a global Pparg deficiency (31) or bone-specific Pparg deficiency (29), it is possible that the HF diet could induce osteoclastogenesis that is not regulated by PPARĪ³ in the mesenchymal lineage. Osteoclasts are derived from hematopoietic precursors of the monocyte-macrophage lineage (48), and mice lacking Pparg in osteoclast lineage cells (Tie2-Cre: PPARĪ³f/f) developed osteopetrosis, characterized by an increased bone mass (50). Therefore, it is possible that PPARĪ³ also plays an important role in obesity-induced osteoclastogenesis and bone resorption, in addition to osteoblastogenesis and bone formation. In addition, the possibility of systemic effects of PPARĪ³ needs to be explored.

One of the proposed mechanisms relating to the detrimental effects of obesity on bone is that both adipocytes and osteoblasts are derived from a common mesenchymal stem cell (13), and the increase in 1 population in the bone marrow would decrease the other. Existing evidence does support a reciprocal relationship between bone marrow adipocytes and osteoblasts (14–19). Our data in part support this mechanism, in that mice carrying deletion of Pparg in Osx1-expressing cells (PPARĪ³fl/fl: Osx1-Cre) had increased tibial Tb.N, BMD, and Conn.Dn and decreased cortical thickness. Decreased cortical thickness and/or Me.Ar in Pparg cKO mice could indicate modeling for growth was limited when compared to the control mice. In addition, the serum concentration of osteocalcin was increased compared to the concentration in floxed littermate control mice, indicating an increase in the osteoblast number and/or activity and bone formation. Similar to our findings, Sun et al. (35), using the same Pparg cKO model, also reported that the femoral Tb.N was significantly increased and trabecular separation was decreased in Pparg cKO mice. However, it is possible that this is due to a limitation of bone modeling for growth rather than to an increase in bone mass (BV/TV) in the proximal tibia per se, since Pparg cKO mice were smaller than the controls.

Interestingly, we found that deletion of the Pparg gene did not affect the bone structural parameters at lumbar vertebrae. Contrary to the findings of this study, we found in an earlier study that mice deficient in Pparg from cells expressing a 3.6-kb type I collagen promoter fragment (PPARĪ³fl/fl: Col3.6-Cre) exhibited moderate reductions in BMD and bone mineral content (BMC) in the fourth LV but not in the femur (29). Mice with Pparg deletion of Dermo1-expressing mesenchymal lineage cells (PPARĪ³fl/fl: Dermo1-Cre) had increased BMD and BMC in both the femur and LV at 6 mo but not at 21 mo of age (34). We do not have a good explanation for the discrepancies in these site-specific bone structural changes among the different studies. Contrasting phenotypes in the femur and spine have also been reported in leptin-deficient ob/ob mice (47). Different cKO models in these studies could contribute to these different results. Other mechanisms or pathways might be involved. For example, mechanical loading conferred by body weight may have differential effects on long bones, such as the tibia in our study, compared to the lumbar spine. David et al. (17) found that mechanical loading downregulates PPARĪ³ in BMSC and adipogenesis while increasing osteoblastogenesis.

The finding that Pparg cKO mice had a lower fat mass than floxed littermate controls was unexpected, since adipocyte deletion should have been restricted to bone marrow. Similar to this observation, we also found that mice with Pparg deletion in Dermol-expressing cells had a reduction in whole-body adiposity (34). While Osx1 was identified as being expressed in developing bones, which is required for osteoblast differentiation and bone formation (51), whether or how much it is expressed or its function in other tissues besides bone has not been investigated. It is also possible that bone-specific ablation of adipocytes would affect peripheral adipogenesis and/or adipocyte functions through certain feedback loops, such as bone-secreted cytokines. For example, osteocalcin, an osteoblast-secreted protein used as a bone formation marker, has been demonstrated to behave as a hormone that regulates glucose and energy metabolism (52). Osteocalcin-deficient (osteocalcin –/–) mice have abnormal visceral fat (53), and long-term treatment of wild-type mice with nanomolar amounts of osteocalcin can significantly decrease the body weight and fat mass of mice fed an HF diet (52). Whether the decrease in fat mass in Pparg cKO mice is the consequence of the increase in the serum concentration of osteocalcin, as found in this study, or of other bone-secreted cytokines remains to be investigated.

Obviously, the relationship between obesity and bone is multifaceted, and PPARĪ³ is 1 of the important factors regulating both obesity and bone development. Bone-secreted cytokines can affect energy, the glucose metabolism, and adiposity (as discussed above), and bone itself is also modulated by obesity or paracrine effects of adipokines, such as leptin and adiponectin (11, 54, 55).

In summary, this study demonstrated that mice deficient of Pparg in BMSC had a decreased fat mass and an increased bone mass (BV/TV) in the proximal tibia but not in lumbar vertebrae when compared with the PPARĪ³-floxed littermate controls. Deletion of PPARĪ³ did not prevent HF diet–induced bone deterioration in mice. Further studies are needed to understand the complex relationship between adipocytes or adiposity and bone metabolism, which may help develop therapeutic agents to prevent or treat both obesity and osteoporosis.

Acknowledgments

We thank Dr. Frank J. Gonzalez, National Cancer Institute, National Institute of Health, for providing floxed PPARĪ³ mice. We thank James Lindlauf for help in animal breeding and care and food intake measurements, and Matthew J. Picklo for critical review of the manuscript.

The authors' responsibilities were as follows—JJC: contributed to the study design, implementation, data analysis and interpretation, and manuscript preparation; XS: contributed to the study design, data interpretation, and manuscript preparation: BRG and KGM: contributed to the study design, the experiment execution, data collection, and manuscript revision; and all authors: read and approved the final manuscript.

Notes

This work was supported by the Agricultural Research Service of the USDA (#3062-51000-053-00D). Funding support for XS was from the National Institute on Aging of the National Institutes of Health under Award Number R01AG046248.

Mention of trade names or commercial products in this publication is solely for the purpose of providing specific information and does not imply recommendation or endorsement by the USDA. The USDA is an equal opportunity provider and employer. The findings and conclusions in this manuscript are those of the authors and should not be construed to represent any official USDA or US Government determination or policy.

Author disclosures: Funding support for XS was from the National Institute on Aging of the National Institutes of Health under Award Number R01AG046248. JJC is a member of the Journal's Editorial Board. All other authors report no conflicts of interest.

Supplemental Tables 1–3 and Supplemental Figure 1 are available from the "Supplementary data" link in the online posting of the article and from the same link in the online table of contents at https://academic.oup.com/jn.

Abbreviations used: B.Ar, bone area; BALP, bone-specific alkaline phosphatase; BMC, bone mineral content; BMD, bone mineral density; BMSC, bone marrow stromal cells; B.Pm, bone perimeter; BV, bone volume; cKO, conditional knockout; Conn.D, connectivity density; Ct.Th, cortical thickness; HF, high-fat; LV2, second lumbar vertebra; Me.Ar, medullary area; NF, normal-fat; Osx1, Osterix 1; PPARĪ³, peroxisome proliferator-activated receptor gamma; SMI, structure model index; T.Ar, total area; Tb.N, trabecular number; Tb.Sp, trabecular separation; Tb.Th, trabecular thickness; TRAP, tartrate-resistant acid phosphatase; TV, total volume; Ī¼CT, micro–computed tomography.

References

1.

Ogden

CL

,

Carroll

MD

,

Fryar

CD

,

Flegal

KM

.

Prevalence of obesity among adults and youth: United States, 2011–2014

.

NCHS Data Brief

.

2015

;(

219

):

1

8

.

2.

Messerli

FH

,

Christie

B

,

DeCarvalho

JG

,

Aristimuno

GG

,

Suarez

DH

,

Dreslinski

GR

,

Frohlich

E

D.

Obesity and essential hypertension. Hemodynamics, intravascular volume, sodium excretion, and plasma renin activity

.

Arch Intern Med

.

1981

;

141

(

1

):

81

5

.

3.

Kopelman

PG

.

Obesity as a medical problem

.

Nature

.

2000

;

404

(

6778

):

635

43

.

4.

Mokdad

AH

,

Bowman

BA

,

Ford

ES

,

Vinicor

F

,

Marks

JS

,

Koplan

JP

.

The continuing epidemics of obesity and diabetes in the United States

.

JAMA

.

2001

;

286

(

10

):

1195

200

.

5.

Melton

LJ

3rd.

The prevalence of osteoporosis: gender and racial comparison

.

Calcif Tissue Int

.

2001

;

69

(

4

):

179

81

.

6.

Wright

NC

,

Looker

AC

,

Saag

KG

,

Curtis

JR

,

Delzell

ES

,

Randall

S

,

Dawson-Hughes

B

.

The recent prevalence of osteoporosis and low bone mass in the United States based on bone mineral density at the femoral neck or lumbar spine

.

J Bone Miner Res

.

2014

;

29

(

11

):

2520

6

.

7.

Jensen

VFH

,

Molck

AM

,

Dalgaard

M

,

McGuigan

FE

,

Akesson

KE

.

Changes in bone mass associated with obesity and weight loss in humans: applicability of animal models

.

Bone

.

2021

;

145

:

115781

.

8.

Qiao

D

,

Li

Y

,

Liu

X

,

Zhang

X

,

Qian

X

,

Zhang

H

,

Zhang

G

,

Wang

C

.

Association of obesity with bone mineral density and osteoporosis in adults: a systematic review and meta-analysis

.

Public Health

.

2020

;

180

:

22

28

.

9.

Zhao

LJ

,

Liu

YJ

,

Liu

PY

,

Hamilton

J

,

Recker

RR

,

Deng

HW

.

Relationship of obesity with osteoporosis

.

J Clin Endocrinol Metab

.

2007

;

92

(

5

):

1640

6

.

10.

Cao

JJ

,

Gregoire

BR

,

Gao

H

.

High-fat diet decreases cancellous bone mass but has no effect on cortical bone mass in the tibia in mice

.

Bone

.

2009

;

44

(

6

):

1097

104

.

11.

Cao

JJ

.

Effects of obesity on bone metabolism

.

J Orthop Surg Res

.

2011

;

6

:

30

.

12.

Rosen

CJ

,

Bouxsein

ML

.

Mechanisms of disease: is osteoporosis the obesity of bone?

.

Nat Clin Pract Rheumatol

.

2006

;

2

(

1

):

35

43

.

13.

Gregoire

FM

,

Smas

CM

,

Sul

HS

.

Understanding adipocyte differentiation

.

Physiol Rev

.

1998

;

78

(

3

):

783

809

.

14.

Moerman

EJ

,

Teng

K

,

Lipschitz

DA

,

Lecka-Czernik

B

.

Aging activates adipogenic and suppresses osteogenic programs in mesenchymal marrow stroma/stem cells: the role of PPAR-gamma2 transcription factor and TGF-beta/BMP signaling pathways

.

Aging Cell

.

2004

;

3

(

6

):

379

89

.

15.

Lei

Z

,

Xiaoying

Z

,

Xingguo

L

.

Ovariectomy-associated changes in bone mineral density and bone marrow haematopoiesis in rats

.

Int J Exp Pathol

.

2009

;

90

(

5

):

512

9

.

16.

Gimble

JM

,

Robinson

CE

,

Wu

X

,

Kelly

KA

,

Rodriguez

BR

,

Kliewer

SA

,

Lehmann

JM

,

Morris

DC

.

Peroxisome proliferator-activated receptor-gamma activation by thiazolidinediones induces adipogenesis in bone marrow stromal cells

.

Mol Pharmacol

.

1996

;

50

(

5

):

1087

94

.

17.

David

V

,

Martin

A

,

Lafage-Proust

MH

,

Malaval

L

,

Peyroche

S

,

Jones

DB

,

Vico

L

,

Guignandon

A

.

Mechanical loading down-regulates peroxisome proliferator-activated receptor gamma in bone marrow stromal cells and favors osteoblastogenesis at the expense of adipogenesis

.

Endocrinology

.

2007

;

148

(

5

):

2553

62

.

18.

Sen

B

,

Xie

Z

,

Case

N

,

Ma

M

,

Rubin

C

,

Rubin

J

.

Mechanical strain inhibits adipogenesis in mesenchymal stem cells by stimulating a durable beta-catenin signal

.

Endocrinology

.

2008

;

149

(

12

):

6065

75

.

19.

Beresford

JN

,

Bennett

JH

,

Devlin

C

,

Leboy

PS

,

Owen

ME

.

Evidence for an inverse relationship between the differentiation of adipocytic and osteogenic cells in rat marrow stromal cell cultures

.

J Cell Sci

.

1992

;

102

(

2

):

341

51

.

20.

Hotamisligil

GS

,

Shargill

NS

,

Spiegelman

BM

.

Adipose expression of tumor necrosis factor-alpha: direct role in obesity-linked insulin resistance

.

Science

.

1993

;

259

(

5091

):

87

91

.

21.

Xu

H

,

Barnes

GT

,

Yang

Q

,

Tan

G

,

Yang

D

,

Chou

CJ

,

Sole

J

,

Nichols

A

,

Ross

JS

,

Tartaglia

LA

et al.

Chronic inflammation in fat plays a crucial role in the development of obesity-related insulin resistance

.

J Clin Invest

.

2003

;

112

(

12

):

1821

30

.

22.

Weisberg

SP

,

McCann

D

,

Desai

M

,

Rosenbaum

M

,

Leibel

RL

,

Ferrante

AW

Jr
.

Obesity is associated with macrophage accumulation in adipose tissue

.

J Clin Invest

.

2003

;

112

(

12

):

1796

808

.

23.

Kyung

TW

,

Lee

JE

,

Phan

TV

,

Yu

R

,

Choi

HS

.

Osteoclastogenesis by bone marrow-derived macrophages is enhanced in obese mice

.

J Nutr

.

2009

;

139

(

3

):

502

6

.

24.

Shu

L

,

Beier

E

,

Sheu

T

,

Zhang

H

,

Zuscik

MJ

,

Puzas

EJ

,

Boyce

BF

,

Mooney

RA

,

Xing

L

.

High-fat diet causes bone loss in young mice by promoting osteoclastogenesis through alteration of the bone marrow environment

.

Calcif Tissue Int

.

2015

;

96

(

4

):

313

23

.

25.

Cao

JJ

,

Gregoire

BR

,

Shen

CL

.

A high-fat diet decreases bone mass in growing mice with systemic chronic inflammation induced by low-dose, slow-release lipopolysaccharide pellets

.

J Nutr

.

2017

;

147

(

10

):

1909

16

.

26.

Rosen

ED

,

Sarraf

P

,

Troy

AE

,

Bradwin

G

,

Moore

K

,

Milstone

DS

,

Spiegelman

BM

,

Mortensen

RM

.

PPAR gamma is required for the differentiation of adipose tissue in vivo and in vitro

.

Mol Cell

.

1999

;

4

(

4

):

611

7

.

27.

Justesen

J

,

Stenderup

K

,

Ebbesen

EN

,

Mosekilde

L

,

Steiniche

T

,

Kassem

M

.

Adipocyte tissue volume in bone marrow is increased with aging and in patients with osteoporosis

.

Biogerontology

.

2001

;

2

(

3

):

165

71

.

28.

Verma

S

,

Rajaratnam

JH

,

Denton

J

,

Hoyland

JA

,

Byers

RJ

.

Adipocytic proportion of bone marrow is inversely related to bone formation in osteoporosis

.

J Clin Pathol

.

2002

;

55

(

9

):

693

8

.

29.

Cao

J

,

Ou

G

,

Yang

N

,

Ding

K

,

Kream

BE

,

Hamrick

MW

,

Isales

CM

,

Shi

XM

.

Impact of targeted PPAR gamma disruption on bone remodeling

.

Mol Cell Endocrinol

.

2015

;

410

:

27

34

.

30.

Kubota

N

,

Terauchi

Y

,

Miki

H

,

Tamemoto

H

,

Yamauchi

T

,

Komeda

K

,

Satoh

S

,

Nakano

R

,

Ishii

C

,

Sugiyama

T

et al.

PPAR gamma mediates high-fat diet-induced adipocyte hypertrophy and insulin resistance

.

Mol Cell

.

1999

;

4

(

4

):

597

609

.

31.

Akune

T

,

Ohba

S

,

Kamekura

S

,

Yamaguchi

M

,

Chung

UI

,

Kubota

N

,

Terauchi

Y

,

Harada

Y

,

Azuma

Y

,

Nakamura

K

et al.

PPAR gamma insufficiency enhances osteogenesis through osteoblast formation from bone marrow progenitors

.

J Clin Invest

.

2004

;

113

(

6

):

846

55

.

32.

Jones

JR

,

Barrick

C

,

Kim

KA

,

Lindner

J

,

Blondeau

B

,

Fujimoto

Y

,

Shiota

M

,

Kesterson

RA

,

Kahn

BB

,

Magnuson

MA

.

Deletion of PPAR gamma in adipose tissues of mice protects against high fat diet-induced obesity and insulin resistance

.

Proc Natl Acad Sci

.

2005

;

102

(

17

):

6207

12

.

33.

Cock

TA

,

Houten

SM

,

Auwerx

J

.

Peroxisome proliferator-activated receptor-gamma: too much of a good thing causes harm

.

EMBO Rep

.

2004

;

5

(

2

):

142

7

.

34.

Cao

J

,

Ding

K

,

Pan

G

,

Rosario

R

,

Su

Y

,

Bao

Y

,

Zhou

H

,

Xu

J

,

McGee Lawrence

ME

,

Hamrick

MW

et al.

Deletion of PPAR gamma in mesenchymal lineage cells protects against aging-induced cortical bone loss in mice

.

J Gerontol A Biol Sci Med Sci

.

2020

;

75

(

5

):

826

34

.

35.

Sun

H

,

Kim

JK

,

Mortensen

R

,

Mutyaba

LP

,

Hankenson

KD

,

Krebsbach

PH

.

Osteoblast-targeted suppression of PPAR gamma increases osteogenesis through activation of mTOR signaling

.

Stem Cells

.

2013

;

31

(

10

):

2183

92

.

36.

Rodda

SJ

,

McMahon

AP

.

Distinct roles for Hedgehog and canonical Wnt signaling in specification, differentiation and maintenance of osteoblast progenitors

.

Development

.

2006

;

133

(

16

):

3231

44

.

37.

Akiyama

TE

,

Sakai

S

,

Lambert

G

,

Nicol

CJ

,

Matsusue

K

,

Pimprale

S

,

Lee

YH

,

Ricote

M

,

Glass

CK

,

Brewer

HB

Jr.
et al.

Conditional disruption of the peroxisome proliferator-activated receptor gamma gene in mice results in lowered expression of ABCA1, ABCG1, and apoE in macrophages and reduced cholesterol efflux

.

Mol Cell Biol

.

2002

;

22

(

8

):

2607

19

.

38.

Liu

F

,

Woitge

HW

,

Braut

A

,

Kronenberg

MS

,

Lichtler

AC

,

Mina

M

,

Kream

BE

.

Expression and activity of osteoblast-targeted Cre recombinase transgenes in murine skeletal tissues

.

Int J Dev Biol

.

2004

;

48

(

7

):

645

53

.

39.

Reeves

PG

,

Nielsen

FH

,

Fahey

GC

Jr
.

AIN-93 purified diets for laboratory rodents: final report of the American Institute of Nutrition ad hoc writing committee on the reformulation of the AIN-76A rodent diet

.

J Nutr

.

1993

;

123

(

11

):

1939

51

.

40.

Cao

JJ

,

Picklo

MJ

Sr.

Involuntary wheel running improves but does not fully reverse the deterioration of bone structure of obese rats despite decreasing adiposity

.

Calcif Tissue Int

.

2015

;

97

(

2

):

145

55

.

41.

Bouxsein

ML

,

Boyd

SK

,

Christiansen

BA

,

Guldberg

RE

,

Jepsen

KJ

,

Muller

R

.

Guidelines for assessment of bone microstructure in rodents using micro-computed tomography

.

J Bone Miner Res

.

2010

;

25

(

7

):

1468

86

.

42.

Parfitt

AM

,

Drezner

MK

,

Glorieux

FH

,

Kanis

JA

,

Malluche

H

,

Meunier

PJ

,

Ott

SM

,

Recker

RR

.

Bone histomorphometry: standardization of nomenclature, symbols, and units. Report of the ASBMR Histomorphometry Nomenclature Committee

.

J Bone Miner Res

.

1987

;

2

(

6

):

595

610

.

43.

Blum

M

,

Harris

SS

,

Must

A

,

Naumova

EN

,

Phillips

SM

,

Rand

WM

,

Dawson-Hughes

B

.

Leptin, body composition and bone mineral density in premenopausal women

.

Calcif Tissue Int

.

2003

;

73

(

1

):

27

32

.

44.

Zhao

LJ

,

Jiang

H

,

Papasian

CJ

,

Maulik

D

,

Drees

B

,

Hamilton

J

,

Deng

HW

.

Correlation of obesity and osteoporosis: effect of fat mass on the determination of osteoporosis

.

J Bone Miner Res

.

2008

;

23

(

1

):

17

29

.

45.

Goulding

A

,

Jones

IE

,

Taylor

RW

,

Williams

SM

,

Manning

PJ

.

Bone mineral density and body composition in boys with distal forearm fractures: a dual-energy x-ray absorptiometry study

.

J Pediatr

.

2001

;

139

(

4

):

509

15

.

46.

Hsu

YH

,

Venners

SA

,

Terwedow

HA

,

Feng

Y

,

Niu

T

,

Li

Z

,

Laird

N

,

Brain

JD

,

Cummings

SR

,

Bouxsein

ML

et al.

Relation of body composition, fat mass, and serum lipids to osteoporotic fractures and bone mineral density in Chinese men and women

.

Am J Clin Nutr

.

2006

;

83

(

1

):

146

54

.

47.

Hamrick

MW

,

Pennington

C

,

Newton

D

,

Xie

D

,

Isales

C

.

Leptin deficiency produces contrasting phenotypes in bones of the limb and spine

.

Bone

.

2004

;

34

(

3

):

376

83

.

48.

Novack

DV

,

Teitelbaum

SL

.

The osteoclast: friend or foe?

.

Annu Rev Pathol

.

2008

;

3

:

457

84

.

49.

Cao

JJ

,

Gregoire

BR

,

Michelsen

KG

,

Picklo

MJ

.

Increasing dietary fish oil reduces adiposity and mitigates bone deterioration in growing C57BL/6 mice fed a high-fat diet

.

J Nutr

.

2020

;

150

(

1

):

99

107

.

50.

Wan

Y

,

Chong

LW

,

Evans

RM

.

PPAR-gamma regulates osteoclastogenesis in mice

.

Nat Med

.

2007

;

13

(

12

):

1496

503

.

51.

Nakashima

K

,

Zhou

X

,

Kunkel

G

,

Zhang

Z

,

Deng

JM

,

Behringer

RR

,

de Crombrugghe

B

.

The novel zinc finger-containing transcription factor Osterix is required for osteoblast differentiation and bone formation

.

Cell

.

2002

;

108

(

1

):

17

29

.

52.

Ferron

M

,

Hinoi

E

,

Karsenty

G

,

Ducy

P

.

Osteocalcin differentially regulates beta cell and adipocyte gene expression and affects the development of metabolic diseases in wild-type mice

.

Proc Natl Acad Sci

.

2008

;

105

(

13

):

5266

70

.

53.

Lee

NK

,

Sowa

H

,

Hinoi

E

,

Ferron

M

,

Ahn

JD

,

Confavreux

C

,

Dacquin

R

,

Mee

PJ

,

McKee

MD

,

Jung

DY

et al.

Endocrine regulation of energy metabolism by the skeleton

.

Cell

.

2007

;

130

(

3

):

456

69

.

54.

Ducy

P

,

Amling

M

,

Takeda

S

,

Priemel

M

,

Schilling

AF

,

Beil

FT

,

Shen

J

,

Vinson

C

,

Rueger

JM

,

Karsenty

G

.

Leptin inhibits bone formation through a hypothalamic relay: a central control of bone mass

.

Cell

.

2000

;

100

(

2

):

197

207

.

55.

Shinoda

Y

,

Yamaguchi

M

,

Ogata

N

,

Akune

T

,

Kubota

N

,

Yamauchi

T

,

Terauchi

Y

,

Kadowaki

T

,

Takeuchi

Y

,

Fukumoto

S

et al.

Regulation of bone formation by adiponectin through autocrine/paracrine and endocrine pathways

.

J Cell Biochem

.

2006

;

99

(

1

):

196

208

.

Published by Oxford University Press on behalf of the American Society for Nutrition 2021.

This work is written by (a) US Government employee(s) and is in the public domain in the US.

  • Supplementary data

    Ppargamma Expression Adipose Tissue High Fat Diet

    Source: https://academic.oup.com/jn/article/151/9/2697/6296112