Fat accumulates in bone marrow (BM) of patients with diabetes. In this study, we investigated the mechanisms and consequences of this phenomenon. BM mesenchymal stromal cells (BM-MSCs) from patients with type 2 diabetes (T2D) constitutively express adipogenic markers and robustly differentiate into adipocytes (ADs) upon in vitro induction as compared with BM-MSCs from subjects without diabetes. Moreover, BM-ADs from subjects with T2D (T2D BM-ADs) paracrinally stimulate a transcriptional adipogenic program in BM-MSCs. Antagonism of MCP-1, a chemokine pivotally expressed in T2D BM-ADs, prevented the T2D BM-AD secretome from converting BM-MSCs into ADs. Mechanistic validation of human data was next performed in an obese T2D mouse model. Systemic antagonism of MCP-1 improved metabolic control, reduced BM fat, and increased osteocyte density. It also indirectly re-established the abundance of long-term versus short-term hematopoietic stem cells. We reveal a diabetic feedback loop in which 1) BM-MSCs are constitutively inclined to make ADs, and 2) mature BM-ADs, via secreted MCP-1, relentlessly fuel BM-MSC determination into new fat. Pharmacological inhibition of MCP-1 signaling can contrast this vicious cycle, restoring, at least in part, the balance between adipogenesis and hematopoiesis in BM from subjects with T2D.

Type 2 diabetes (T2D) is a major risk factor for cardiovascular disease because of its damaging impact on the micro- and macrovasculature. Patients with T2D have twice the chance of dying from ischemic events as subjects without diabetes (1), and those who survive suffer a markedly slower and more arduous recovery (2). This worse outcome has been attributed to the deleterious effect of T2D on angiogenesis and vasculogenesis, two intertwined reparative processes brought about by local and circulating cells (3). A defect in the mobilization of progenitor cells, such as CD34+ cells, from the bone marrow (BM) represents a common feature (4) and a predictor for cardiovascular mortality in patients with diabetes (5,6). Moreover, the reduced availability and functionality of circulating vasculogenic cells may be the consequence of pathogenic processes occurring within the BM microenvironment (7).

Excessive adiposity, typically associated with T2D, does not spare the BM. However, the marrow is not an ectopic fat deposit, like organs that normally contain only small amounts of fat, such as the liver, skeletal muscle, and heart. Instead, fat is a physiological component of the BM, representing ∼10% of the total fat mass in a lean individual (8). In patients with T2D, BM adipose tissue (BMAT) is remarkably increased at the expense of the hematopoietic tissue. Other pathological conditions, such as calorie restriction and radiation exposure, as well as aging and obesity have been associated with BM adiposity (912).

BMAT shares some phenotypic similarities with but constitutes a distinct category from both white and brown adipose tissue. Moreover, its function and pathogenic implications remain largely unknown (13). BM adipocytes (ADs) originate from resident mesenchymal stromal cells (MSCs), renamed recently as skeletal stem cells (14), which can also differentiate into osteoblastic and chondrogenic lineages (11). The relationship between BM-ADs and other marrow cell populations is increasingly complex. Several studies suggest that BMAT accumulation is deleterious to the integrity of bone and the hematopoietic niche. However, this association is not universal and depends on the experimental and clinical context. For example, in mice, both the expansion of endogenous BMAT, as in obesity or aging, and exogenous AD transplantation cause a decrease in LinnegSca1poscKitpos (LSK), long-term CD34neg (LT), and short-term CD34pos (ST) hematopoietic stem cells (HSCs), which also contain multipotent progenitor cells (11). In contrast, a recent study showed that BM-ADs support regenerative hematopoiesis after irradiation or chemotherapy (15). BMAT and bone are also intimately linked, as their levels are often found to be inversely correlated, with a reduced bone-to-BMAT ratio being considered a risk factor for bone fractures (16). Mature osteoblasts inhibit the differentiation of MSCs into the adipose tissue through paracrine mechanisms. Conversely, mature BM-ADs influence bone mass by inhibiting the progression of BM-MSCs to osteoblasts and enhancing osteoclastogenesis (17). In addition, BMAT functions as an endocrine organ, surpassing the more widespread white fat as a source of adiponectin (ADIPOQ) (12) and secreting an extensive number of cytokines and adipokines, such as leptin (LEP), resistin (RETN), and monocyte chemoattractant protein-1 (MCP-1/CCL2) (18,19).

MCP-1 is expressed by various cell types, either constitutively or after induction by oxidative stress, cytokines, or growth factors (20). Circulating levels of MCP-1 are elevated in obese patients and patients with T2D (21). Furthermore, gene deletion (22) and pharmacological inhibition of MCP-1 reduce visceral white fat volume in obese mice (23,24), suggesting direct participation in adipogenesis. Activation of MCP-1 receptor CCR2 leads to the induction of the transcription of MCP-1–induced protein (MCPIP), which promotes AD differentiation in a peroxisome proliferator–activated receptor γ (PPARγ)–independent manner (25,26). MCP-1 is also pivotally involved in the modulation and recruitment of macrophages and T cells, which accounts for its participation in the induction of fat tissue inflammation in T2D and obesity (27).

The current study aims to unveil the molecular mechanisms and consequences of AD accumulation in the BM of patients with T2D. Results highlight a feedback-loop mechanism by which mature BM-ADs from subjects with T2D (T2D BM-ADs) release high levels of MCP-1, which in turn stimulates the differentiation of BM-MSCs into new ADs. Importantly, in vivo antagonism of the MCP-1/CCR2 signaling in T2D mice not only decreased BMAT accumulation but also rescued LT-HSC depletion.

Patient Recruitment

Patients undergoing hip replacement surgery were recruited under informed consent at the Avon Orthopaedic Centre, Southmead Hospital. The study protocol complied with the Declaration of Helsinki, was covered by institutional ethical approval (REC14/SW/1083 and REC14/WA/1005), and was registered as an observational clinical study in the National Institute for Health Research Clinical Research Network Portfolio, UK Clinical Trials Gateway, and ClinicalTrials.gov.

T2D was diagnosed according to the American Diabetes Association guidelines. Specifically, it was defined as 1) patient/referring doctor reports a previous diagnosis of diabetes, 2) HbA1c >48 mmol/mol, and 3) not requiring insulin treatment for at least 12 months after the initial diagnosis. Exclusion criteria comprised acute disease/infection, immune diseases, current or past hematological disorders or malignancy, unstable angina, recent (within 6 months) myocardial infarction or stroke, critical limb ischemia, liver failure, dialysis due to renal failure, pregnancy, and lack of consent to participate. Available data on patients’ characteristics are provided in Supplementary Table 1.

Human BM Isolation

BM samples were obtained from scooped femur heads remaining from hip replacement surgery. During the replacement procedure, the femoral head was removed with a saw, and the proximal femoral canal was opened with reamers and rasps. The BM displaced into the wound was scooped into a sterile pot with a curette. The superfluous cancellous bone was removed from the femoral neck and proximal metaphysis and placed with the marrow. The sample was decanted into a collection tube with 0.5 mol/L EDTA, pH 8. Only material that would otherwise be discarded was collected for study.

Histology

Bone samples were fixed in 1% paraformaldehyde (Thermo Fisher Scientific, Loughborough, U.K.), decalcified for 16 h with 10% formic acid, and embedded in paraffin. Blocks were sectioned on a rotary microtome at 2 μm, and the samples were stained with hematoxylin and eosin (H&E; Sigma-Aldrich, Gillingham, U.K.).

Human BM Cell Isolation and Culture

BM samples were stratified on Ficoll Histopaque 1077 (Thermo Fisher Scientific) and centrifuged without acceleration or brake at 300g for 30 min at 25°C. A total of 1 × 107 mononuclear cells from the Ficoll separation was seeded in plastic flasks in α–minimum essential medium Eagle (αMEM) basal media (Thermo Fisher Scientific) supplemented with 20% FBS (Thermo Fisher Scientific) for 48 h at 37°C, 5% CO2. The adherent cells were considered BM-MSCs and expanded in αMEM supplemented with 20% FBS.

Mature ADs from the floating phase after stratification were collected, strained through a 100-μm filter, and washed with Hanks’ balanced salt solution (Sigma-Aldrich). The cells were then cultured using a published method (28). Briefly, 1 × 107 cells were seeded in a well of a 12-well culture plate (CELLSTAR; Greiner Bio-One, Cardiff, U.K.), which was filled to the brim with αMEM containing 20% FBS. A 22-mm borosilicate glass coverslip (VWR International, Leighton, U.K.) was added on top of the well. Mature ADs were incubated for 4 days until adhesion to the glass plate and then flipped and put inside a well of a six-well plate containing αMEM. ADs were then incubated 48 h in FBS-free αMEM. The conditioned media (CM) was then harvested and filtered through a 22-μm filter (Sigma-Aldrich). Unilocularity of ADs was evaluated by microscopy, and absence of foam-cell contamination was confirmed by negative expression of the macrophage markers CD11b and CD68 mRNA by quantitative PCR (qPCR) (Supplementary Fig. 1).

ELISA

Chemokines were quantified using ELISA kits on 100 μL of CM, following the manufacturer’s protocol (Bio-Techne, Minneapolis, MN). Glycated and total hemoglobin was measured using ELISA kits (Generon, Slough, U.K.) using serum diluted 1:500.

Gene Expression Analysis

RNA Extraction and Purification

RNA was purified using Tri-Reagent (Sigma-Aldrich) per the manufacturer’s protocol and repurified using acid-phenol phase separation. RNA was quantified using a NanoDrop (Thermo Fisher Scientific).

cDNA Library Generation

cDNA was generated using the High-Capacity RNA-to-cDNA Kit (Thermo Fisher Scientific) and 100 ng total RNA following the manufacturer’s protocol. A T100 Thermal Cycler (Bio-Rad, Watford, U.K.) was used.

qPCR

mRNA expression was assayed using real-time qPCR. The reaction was made with Power SYBR Green Master Mix (Thermo Fisher Scientific) and 200 nmol/L gene-specific primers (Supplementary Table 2) in a QuantStudio 6 Flex RT-PCR machine (Thermo Fisher Scientific).

Flow Cytometry

Freshly isolated BM cells or cultured BM-MSCs were labeled with primary antibodies in staining buffer (Hanks’ balanced salt solution supplemented with 1% BSA) and acquired with an LSR Fortessa ×20 (BD Biosciences, Oxford, U.K.), and quantification of the antigenic profile was performed using FlowJo v10 software (FlowJo, LLC). Antibodies used are listed in Supplementary Table 3.

Differentiation of BM-MSCs

Adipose differentiation was induced by treatment with 1 μmol/L dexamethasone, 0.5 mmol/L isobutylmethylxanthine, 10 μg/mL insulin, and 1 μmol/L indomethacin (all from Sigma-Aldrich) in DMEM supplemented with 10% FBS for 21 days, changing the media every 3 days. Cells were fixed with 10% formaldehyde, treated with 60% isopropanol, and exposed to Oil Red O (ORO) 0.3% solution (all from Sigma-Aldrich) for 30 min and then washed with H2O. Osteoblast differentiation was induced by treatment with 0.1 mmol/L dexamethasone, 50 μg/mL l-ascorbic acid 2-phosphate, and 10 mmol/L β-glycerophosphate (all from Sigma-Aldrich) in DMEM supplemented with 10% FBS for 14 days, changing the media every 3 days. Cells were fixed with ice-cold methanol (Sigma-Aldrich) and treated with a solution of 0.4 mg/mL nitro-blue tetrazolium chloride and 0.19 mg/mL 5-bromo-4-chloro-3’-indolyphosphate p-toluidine (both from Thermo Fisher Scientific) to assess alkaline phosphatase activity.

Animal Studies

Experiments were performed in accordance with the Guide for the Care and Use of Laboratory Animals (The Institute of Laboratory Animal Resources, 1996) and with the approval of the University of Bristol and the British Home Office (license 30/2811). As a model of T2D and obesity, we used 4-week-old male obese leptin-receptor homozygous mutant C57BLKS/J-Leprdb/Leprdb/Dock7+ (Db/Db) mice (Charles River Laboratories, Harlow, U.K.). Age- and sex-matched lean heterozygous C57BLKS/J-Leprdb/LeprWT Dock7+ (Wt/Db) mice served as controls. Animals were fed standard chow (Charles River Laboratories) and provided water ad libitum.

Mice were randomly allocated to receive 4 g/kg/day of the MCP-1 receptor antagonist RS504393 (Tocris Bioscience, Bristol, U.K.) diluted in DMSO in drinking water (1% of water volume) or the equivalent volume of DMSO for 8 weeks. Glucose was measured in urine using Diastix colorimetric reagent strips (Bayer, Reading, U.K.).

Mouse BM Processing Protocol

Mouse femurs were cut longitudinally. Pictures were taken by a Fast1384 Qicam camera (QImaging) using Infinity Analyze capture software. Cells were counted and analyzed using ImageJ software (National Institutes of Health).

Whole BM cells were obtained from the mouse tibia. The marrow was flushed from the bone using PBS and then subjected to red blood cell lysis. Cells were then washed and used for the appropriate experiment.

Statistical Analysis

Values are presented as mean ± SEM. Two-tailed independent-samples t test was used to compare groups with T2D and without diabetes assuming unequal variances. One- and two-way ANOVA were used to compare four group experiments, followed by multivariate analysis to compare each group individually. Nonparametric sample groups were evaluated using the Kruskal-Wallis test. P values <0.05 were considered statistically significant.

Features of Fat Accumulation in the BM of Patients With T2D

We confirm patients with T2D show a remarkable remodeling of the BM at a cellular level, as described in another cohort evaluated previously (10). H&E staining shows a large increase in the area covered by ADs as compared with control subjects without diabetes (Fig. 1A and B). This effect is due to an increase in both AD number and size (Fig. 1C and D). The average AD size was 4,487 ± 675 and 7,812 ± 666 μm2 in patients without diabetes and with T2D, respectively, corresponding to a 1.74-fold increase in the former group (P < 0.05).

Figure 1

Adipocyte accumulation in BM of patients with T2D compared with control subjects without diabetes (ND). A: H&E staining of human femoral bone samples from subjects without diabetes and those with T2D. Scatter plots show individual values, with means and SE bars, of the percentage of total BM area covered by ADs (B) and AD density (C). D: The bar graph illustrates the frequency of ADs according to different size categories. Means and SE bars for each category are shown. Mann-Whitney test for non-normally distributed variables (B) and unpaired t test for normally distributed variables (C and D). N = 8 biological replicates/group. Scale bars, 250 μm. *P < 0.05; ***P < 0.001 vs. subjects without diabetes.

Figure 1

Adipocyte accumulation in BM of patients with T2D compared with control subjects without diabetes (ND). A: H&E staining of human femoral bone samples from subjects without diabetes and those with T2D. Scatter plots show individual values, with means and SE bars, of the percentage of total BM area covered by ADs (B) and AD density (C). D: The bar graph illustrates the frequency of ADs according to different size categories. Means and SE bars for each category are shown. Mann-Whitney test for non-normally distributed variables (B) and unpaired t test for normally distributed variables (C and D). N = 8 biological replicates/group. Scale bars, 250 μm. *P < 0.05; ***P < 0.001 vs. subjects without diabetes.

Close modal

BM-MSCs From Subjects With Diabetes Have an Increased Propensity to Differentiate Into ADs

We next investigated if adipogenic predetermination of BM-MSCs was the cause and source of AD accumulation in the BM of subjects with diabetes. We first measured the effect of T2D on the expression of a spectrum of genes implicated in adipogenesis (Fig. 2A). Results show that T2D induces an increase in the mRNA levels of proadipogenic genes CEBPa, PDGFRb, and IGFR1. In addition, T2D caused the downregulation of the antiadipogenic genes SIRT1 and GLI1. STAT5a, which can be pro- and antiadipogenic, was also downregulated in T2D BM-MSCs. FACS analysis of cultured BM-MSCs showed a twofold increase in PDGFRβ protein levels in the group with T2D, whereas the preadipocyte factor PREF-1 remained unchanged (Fig. 2B).

Figure 2

BM-MSCs from patients with T2D have an increased intrinsic potential to differentiate into the adipocyte lineage. A: The bar graph shows basal mRNA expression levels of adipogenesis regulators in BM-MSCs from subjects without diabetes (ND) and subjects with T2D. Values are means with respective SE bars. B: Flow cytometry analysis of BM-MSCs consisted of gating live single cells, selecting for dual-positive CD105-CD73 MSCs fluorescence minus one (FMO; Bi), and finally examining the frequency of cell fractions positive for PDGFRβ (Bii) and PREF-1 (Biii). Scatter plots showing the quantification of CD73posCD105pos (Biv), CD73posCD105posPDGFRβpos (Bv), and CD73posCD105posPREF-1pos (Bvi) cell populations. C: BM-MSC differentiation in ADs. ADs are stained with ORO-positive cells (arrows; Ci) and quantified by morphometry of microscopic images, with values in the ND and T2D groups illustrated by the scatter plot (Cii). Bar graphs show the results from qPCR analysis of PPARγ (Ciii), ADIPOQ (Civ), and FABP4 (Cv) mRNA expression in BM-MSCs at different times of the adipogenesis induction assay. Values are means with SE bars. *P < 0.05; **P < 0.01; ***P < 0.001 vs. subjects without diabetes; +P < 0.05; +++P < 0.001 vs. time 0 of the adipogenic assay. D: BM-MSC differentiation in osteoblasts, detected by staining with alkaline phosphatase. Representative images with positive cells indicated by arrows (Di) and scatter plot showing means with SE bars (Dii). No difference was detected in this comparison. Student t test with unequal distribution was used in all of the comparisons. N = 4 biological replicates/group. Scale bars, 250 μm. APC, allophycocyanin; FITC, fluorescein isothiocyanate.

Figure 2

BM-MSCs from patients with T2D have an increased intrinsic potential to differentiate into the adipocyte lineage. A: The bar graph shows basal mRNA expression levels of adipogenesis regulators in BM-MSCs from subjects without diabetes (ND) and subjects with T2D. Values are means with respective SE bars. B: Flow cytometry analysis of BM-MSCs consisted of gating live single cells, selecting for dual-positive CD105-CD73 MSCs fluorescence minus one (FMO; Bi), and finally examining the frequency of cell fractions positive for PDGFRβ (Bii) and PREF-1 (Biii). Scatter plots showing the quantification of CD73posCD105pos (Biv), CD73posCD105posPDGFRβpos (Bv), and CD73posCD105posPREF-1pos (Bvi) cell populations. C: BM-MSC differentiation in ADs. ADs are stained with ORO-positive cells (arrows; Ci) and quantified by morphometry of microscopic images, with values in the ND and T2D groups illustrated by the scatter plot (Cii). Bar graphs show the results from qPCR analysis of PPARγ (Ciii), ADIPOQ (Civ), and FABP4 (Cv) mRNA expression in BM-MSCs at different times of the adipogenesis induction assay. Values are means with SE bars. *P < 0.05; **P < 0.01; ***P < 0.001 vs. subjects without diabetes; +P < 0.05; +++P < 0.001 vs. time 0 of the adipogenic assay. D: BM-MSC differentiation in osteoblasts, detected by staining with alkaline phosphatase. Representative images with positive cells indicated by arrows (Di) and scatter plot showing means with SE bars (Dii). No difference was detected in this comparison. Student t test with unequal distribution was used in all of the comparisons. N = 4 biological replicates/group. Scale bars, 250 μm. APC, allophycocyanin; FITC, fluorescein isothiocyanate.

Close modal

Next, we examined the effect of T2D on BM-MSC differentiation into ADs or osteoblasts. Quantification of ORO-positive cells shows an increase in the adipogenic activity of T2D BM-MSCs (Fig. 2Ci and ii). Real-time qPCR analysis also shows that T2D BM-MSCs express higher transcript levels of the AD differentiation markers PPARγ, ADIPOQ, and fatty acid–binding protein 4 (FABP4) (Fig. 2Ciii–v). These results indicate that T2D programs BM-MSCs with an enhanced propensity into adipogenesis. In contrast, T2D did not influence the osteogenic differentiation of BM-MSCs, as assessed by the alkaline phosphatase activity assay (Fig. 2Di and ii).

Mature BM-ADs Modulate BM-MSC Differentiation Through Paracrine Mechanisms

We next investigated the possibility that T2D BM-ADs directly interfere with BM-MSC differentiation, creating an imbalance in the typical lineage specification. BM-MSCs from subjects without diabetes were exposed to AD or osteoblast differentiation media, which were supplemented with the CM from cultured BM-ADs from subjects without diabetes (ND-CM) or BM-ADs from subjects with T2D (T2D-CM) or no CM as a control group (No CM). We observed that the T2D-CM increases the differentiation of BM-MSCs into ORO-positive ADs over the levels found in groups treated with ND-CM or No CM (Fig. 3Ai and ii). The enhanced activation of BM-MSC adipogenesis by the T2D-CM was associated with a greater induction of the ADIPOQ and PPARγ expression (Fig. 3Aiii and iv), whereas FABP4 was similarly upregulated among all groups (Fig. 3Av). These data suggest T2D causes an incremental feedback loop by which BM-ADs will increase MSC fate toward adipogenesis, which, in turn, will stimulate more BM-MSCs to do the same.

Figure 3

Secreted factors from human mature BM adipocytes paracrinally regulate the differentiation of BM-MSCs into adipocyte and osteoblasts. A: Adipocyte differentiation of BM-MSCs from subjects without diabetes following treatment with regular induction medium alone (No CM), the same medium supplemented with BM-AD CM from subjects without diabetes (ND-CM), or T2D-CM. ADs stained with ORO indicated by arrows (Ai) and related quantification, with single values, means, and SE bars illustrated by the scatter plot (Aii). Bar graphs illustrating the results of the qPCR analysis of ADIPOQ (Aiii), PPARγ (Aiv), and FABP4 (Av) mRNA expression at different times of the adipogenesis induction assay. Values are means with SE bars. B: Osteoblast differentiation of BM-MSCs treated with No CM, ND-CM, or T2D-CM. Alkaline phosphatase–positive osteoblasts indicated by arrows (Bi) and related quantification with single values, means, and SE bars illustrated by the scatter plot (Bii). A similar illustration of the qPCR analysis of RUNX2 mRNA expression after 2 weeks of induction (Biii). One-way ANOVA Kruskal-Wallis test, followed by Dunn multiple-comparisons test. N = 4–7 biological replicates/group. Scale bars, 250 μm. *P < 0.05; **P < 0.01; ***P < 0.001 vs. No CM; +P < 0.05; ++P < 0.01; +++P < 0.001 vs. ND-CM.

Figure 3

Secreted factors from human mature BM adipocytes paracrinally regulate the differentiation of BM-MSCs into adipocyte and osteoblasts. A: Adipocyte differentiation of BM-MSCs from subjects without diabetes following treatment with regular induction medium alone (No CM), the same medium supplemented with BM-AD CM from subjects without diabetes (ND-CM), or T2D-CM. ADs stained with ORO indicated by arrows (Ai) and related quantification, with single values, means, and SE bars illustrated by the scatter plot (Aii). Bar graphs illustrating the results of the qPCR analysis of ADIPOQ (Aiii), PPARγ (Aiv), and FABP4 (Av) mRNA expression at different times of the adipogenesis induction assay. Values are means with SE bars. B: Osteoblast differentiation of BM-MSCs treated with No CM, ND-CM, or T2D-CM. Alkaline phosphatase–positive osteoblasts indicated by arrows (Bi) and related quantification with single values, means, and SE bars illustrated by the scatter plot (Bii). A similar illustration of the qPCR analysis of RUNX2 mRNA expression after 2 weeks of induction (Biii). One-way ANOVA Kruskal-Wallis test, followed by Dunn multiple-comparisons test. N = 4–7 biological replicates/group. Scale bars, 250 μm. *P < 0.05; **P < 0.01; ***P < 0.001 vs. No CM; +P < 0.05; ++P < 0.01; +++P < 0.001 vs. ND-CM.

Close modal

We next examined the effect of T2D on osteoblast differentiation. Osteoblastogenesis assays indicate that the ND-CM increases BM-MSC differentiation into alkaline phosphatase–positive osteoblasts. In contrast, this stimulatory effect was negated to the T2D-CM (Fig. 3Bi and ii). The same pattern was confirmed when assessing the mRNA expression of the osteoblast marker RUNX2 (Fig. 3Biii). Altogether, these results indicate that BM-ADs of patients with T2D promote a paracrine induction of BM-MSC adipogenesis at the expense of osteoblastogenesis.

T2D Alters the BM-AD Secretome

To identify which factor secreted by BM-ADs is responsible for the regulation of BM-MSC adipogenic determination, we investigated by qPCR the mRNA expression of multiple adipokines and cytokines in freshly harvested human BM-ADs. We found that the adipokines LEP and RETN were upregulated in T2D as compared with BM-ADs from subjects without diabetes, whereas IGF1, PEDF, and ADIPOQ were downregulated (Fig. 4Ai). Cytokine expression analysis revealed a large increase in TNFa and MCP-1 levels in T2D BM-ADs as well as a modest increase in matrix metalloproteinase 2 (MMP2). We also observed that T2D reduced the levels of IL18, MIF, and angiopoietin 1 (ANGPT1) (Fig. 4Aii). We next verified if the observed transcriptional changes were associated with similar modifications of secreted proteins. ELISA of the AD CM confirmed that the levels of LEP and RETN were increased in T2D (Fig. 4Bi and ii), and ADIPOQ was decreased (Fig. 4Biii), whereas PEDF protein levels were comparable between the two groups (Fig. 4Biv). We observed a striking increase in the secretion of MCP-1 protein by T2D BM-ADs as well as a milder increase in MMP2 and interferon-γ (Fig. 4Bv–vii). Tumor necrosis factor-α was similar between the two groups (Fig. 4Bviii), and ANGPT1 was increased in the T2D BM-AD CM (Fig. 4Bix). Altogether, these data indicate T2D has a profound effect on the BM-AD secretome, with MCP-1 being the most prominently affected factor. This latter phenomenon is well conserved among species, as we have confirmed the elevated MCP-1 levels in the CM of BM-ADs from obese, diabetic Db/Db mice as compared with lean nondiabetic (ND) Wt/Db controls (Supplementary Fig. 2).

Figure 4

T2D modulates the expression and secretion of adipokines and cytokines in BM adipocytes. A: Bar graphs show the results of the qPCR analysis of adipokine (Ai) and cytokine (Aii) mRNA levels in human mature BM-ADs from subjects without diabetes (ND) and with T2D. Values are means with SE bars. B: Scatter plots showing values, means, and SE bars of secreted factors in CM from BM-ADs of the two groups, as assessed by ELISA kits. Secreted factors measured are LEP (Bi), RETN (Bii), ADIPOQ (Biii), PEDF (Biv), MCP-1 (Bv), MMP2 (Bvi), interferon-γ (IFNγ; Bvii), tumor necrosis factor-α (TNFα; Bviii), and ANGPT1 (Bix). Student t test with unequal distribution for all comparisons. N = 6 biological replicates/group. *P < 0.05; ** P < 0.01; ***P < 0.001 vs. subjects without diabetes.

Figure 4

T2D modulates the expression and secretion of adipokines and cytokines in BM adipocytes. A: Bar graphs show the results of the qPCR analysis of adipokine (Ai) and cytokine (Aii) mRNA levels in human mature BM-ADs from subjects without diabetes (ND) and with T2D. Values are means with SE bars. B: Scatter plots showing values, means, and SE bars of secreted factors in CM from BM-ADs of the two groups, as assessed by ELISA kits. Secreted factors measured are LEP (Bi), RETN (Bii), ADIPOQ (Biii), PEDF (Biv), MCP-1 (Bv), MMP2 (Bvi), interferon-γ (IFNγ; Bvii), tumor necrosis factor-α (TNFα; Bviii), and ANGPT1 (Bix). Student t test with unequal distribution for all comparisons. N = 6 biological replicates/group. *P < 0.05; ** P < 0.01; ***P < 0.001 vs. subjects without diabetes.

Close modal

MCP-1 Plays a Key Role in T2D BM-AD–Induced Promotion of BM-MSC Adipogenesis

MCP-1 and its receptor CCR2 have been implicated in obesity through the promotion of AD differentiation and consequent increase in adipose tissue mass (26,29). However, it remains unknown if MCP-1 plays a role in fat accumulation within the BM of patients with T2D. To verify this possibility, we performed adipogenesis assays on human BM-MSCs stimulated with No CM, ND-CM, or T2D-CM as in the previous experiment, but with the addition of the MCP-1 receptor antagonist RS504393 or its vehicle. Inhibition of MCP-1 signaling moderately decreased BM-MSC adipogenesis in the No-CM and ND-CM groups (Fig. 5A and B). In the T2D-CM group, in which adipogenesis was increased, MCP-1 antagonism resulted in a striking inhibition to levels even lower than the No-CM group (Fig. 5A and B). These phenomena were associated with consensual changes in the expression of AD markers. The antagonist reduced the ADIPOQ mRNA expression in BM-MSCs exposed to No CM or ND-CM and completely abrogated the inductive action exerted by the T2D-CM (Fig. 5C). Second, RS504393 selectively inhibited the T2D-CM–induced upregulation of PPARγ (Fig. 5D). Third, the antagonist downregulated FABP4 mRNA levels in all experimental groups (Fig. 5E). Fourth, the antagonist inhibited MCPIP, which was selectively upregulated in BM-MSCs following exposure to T2D-CM (Fig. 5F). This latter result confirms the effective inhibition of molecular signaling downstream to the MCP-1/CCR2 duo.

Figure 5

Antagonism of MCP-1 signaling by a CCR2 antagonist inhibits the promotion of adipogenesis in BM-MSCs from subjects without diabetes exposed to the CM from BM adipocytes of subjects with T2D. BM-MSCs from subjects without diabetes underwent the adipogenic assay in the presence or absence of the specific MCP-1 receptor antagonist RS504393. Three groups were studied in which BM-MSCs were incubated with regular induction medium alone (No CM), the same medium supplemented with BM-AD CM from subjects without diabetes (ND-CM), or BM-AD CM from subjects with T2D (T2D-CM). ADs stained with ORO indicated by arrows (A) and related quantification illustrated by bar graph (B). Using the same protocol, we performed qPCR analyses of ADIPOQ (C), PPARγ (D), FABP4 (E), and MCPIP (F) mRNA at the end of the adipogenesis induction. Values are means with SE bars. Two-way ANOVA with Tukey correction. N = 5 biological replicates/group. Scale bars, 250 μm. *P < 0.05; **P < 0.01; ***P < 0.001 vs. No CM; #P < 0.05; ##P < 0.01 vs. ND-CM; &P < 0.05; &&P < 0.01; &&&P < 0.001 vs. No inhibitor.

Figure 5

Antagonism of MCP-1 signaling by a CCR2 antagonist inhibits the promotion of adipogenesis in BM-MSCs from subjects without diabetes exposed to the CM from BM adipocytes of subjects with T2D. BM-MSCs from subjects without diabetes underwent the adipogenic assay in the presence or absence of the specific MCP-1 receptor antagonist RS504393. Three groups were studied in which BM-MSCs were incubated with regular induction medium alone (No CM), the same medium supplemented with BM-AD CM from subjects without diabetes (ND-CM), or BM-AD CM from subjects with T2D (T2D-CM). ADs stained with ORO indicated by arrows (A) and related quantification illustrated by bar graph (B). Using the same protocol, we performed qPCR analyses of ADIPOQ (C), PPARγ (D), FABP4 (E), and MCPIP (F) mRNA at the end of the adipogenesis induction. Values are means with SE bars. Two-way ANOVA with Tukey correction. N = 5 biological replicates/group. Scale bars, 250 μm. *P < 0.05; **P < 0.01; ***P < 0.001 vs. No CM; #P < 0.05; ##P < 0.01 vs. ND-CM; &P < 0.05; &&P < 0.01; &&&P < 0.001 vs. No inhibitor.

Close modal

In Vivo MCP-1 Antagonism Ameliorates the Metabolic Status of Obese, Diabetic Db/Db Mice

We posited that in vivo administration of the same antagonist would result in a decrease in BM-AD accumulation. To validate this hypothesis, we exposed T2D Db/Db mice to either 4 mg/kg/day of RS504393 (DR group) or DMSO (DD group) in drinking water for 8 weeks. We established a third group of ND Wt/Db (wild-type [WT]) to receive DMSO and be used as a reference group. During the 8-week experiment, no significant differences in weight were observed between the DD and DR groups (Supplementary Fig. 3). In contrast, we observed that RS504393 treatment had an inhibitory effect on body weight gain (Fig. 6Ai) without altering food intake (Supplementary Fig. 3). We found this lowered growth rate associated with selective changes in specific fat deposits. Although the weight of either pericardial or inguinal fat was similar in DD and DR groups, RS504393 treatment resulted in a reduction of the epididymal fat weight (Fig. 6Aii). Histology of the epididymal fat shows that the DR-treated mice exhibited smaller ADs compared with the DD-treated mice (Fig. 6Aiii and iv). This indicates that the antagonist selectively reduces epididymal fat accumulation by acting on AD hypertrophy. Interestingly, we also observed RS504393 treatment ameliorated the metabolic control in T2D mice as indicated by mitigation of glycosuria (Fig. 6Bi) and reduction of blood glycated hemoglobin levels (Fig. 6Bii).

Figure 6

In vivo inhibition of MCP-1 signaling improves metabolic control in T2D mice. An in vivo study was performed in mice to assess the effect of MCP-1 antagonism on systemic and BM end points. Treatment was started in 4-week-old mice for 8 weeks. This figure illustrates the systemic outcomes. A: Body weight (BW) progression and fat deposit profiles of ND vehicle (DMSO)–treated Wt/Db mice [ND WT (DMSO) group], DMSO-treated obese T2D Db/Db mice [T2D DD (DMSO) group], and MCP-1 inhibitor (RS504393)–treated T2D Db/Db mice [T2D DR (RS504393) group]. The line graph shows the weight increase of mice during the 8 weeks of treatment (Ai). The bar graph illustrates the pericardial, inguinal, and epididymal fat pad weight at the end of the study (at week 8 of treatment) (Aii). Histology of epididymal fat pad (Aiii) and size quantification of ADs of the epididymal fat pad illustrated by scatter plot (Aiv). B: Glucosuria and HbA1c profiles of mice in the different treatment groups. Line graph shows the urinary glucose levels measured weekly for 8 weeks with urinalysis sticks (Bi), and scatter plot shows plasma HbA1c levels at week 8 of treatment (Bii). Values are means with SE bars. Student t test with unequal distribution (Ai and Bi), two-way ANOVA with Tukey correction (Aii), and one-way ANOVA with Tukey correction (Aiv and Bii). N = 6 biological replicates/group. Scale bar, 100 μm. **P < 0.01; ***P < 0.001 vs. ND WT; #P < 0.05; ##P < 0.01; ###P < 0.001 vs. T2D DD group.

Figure 6

In vivo inhibition of MCP-1 signaling improves metabolic control in T2D mice. An in vivo study was performed in mice to assess the effect of MCP-1 antagonism on systemic and BM end points. Treatment was started in 4-week-old mice for 8 weeks. This figure illustrates the systemic outcomes. A: Body weight (BW) progression and fat deposit profiles of ND vehicle (DMSO)–treated Wt/Db mice [ND WT (DMSO) group], DMSO-treated obese T2D Db/Db mice [T2D DD (DMSO) group], and MCP-1 inhibitor (RS504393)–treated T2D Db/Db mice [T2D DR (RS504393) group]. The line graph shows the weight increase of mice during the 8 weeks of treatment (Ai). The bar graph illustrates the pericardial, inguinal, and epididymal fat pad weight at the end of the study (at week 8 of treatment) (Aii). Histology of epididymal fat pad (Aiii) and size quantification of ADs of the epididymal fat pad illustrated by scatter plot (Aiv). B: Glucosuria and HbA1c profiles of mice in the different treatment groups. Line graph shows the urinary glucose levels measured weekly for 8 weeks with urinalysis sticks (Bi), and scatter plot shows plasma HbA1c levels at week 8 of treatment (Bii). Values are means with SE bars. Student t test with unequal distribution (Ai and Bi), two-way ANOVA with Tukey correction (Aii), and one-way ANOVA with Tukey correction (Aiv and Bii). N = 6 biological replicates/group. Scale bar, 100 μm. **P < 0.01; ***P < 0.001 vs. ND WT; #P < 0.05; ##P < 0.01; ###P < 0.001 vs. T2D DD group.

Close modal

In Vivo Inhibition of MCP-1 Reduces BM-AD Coverage and Number and Increases Bone Osteocytes in Obese, T2D Db/Db Mice

After 8 weeks of treatment, when mice were 12 weeks of age, AD coverage at the metaphysis of the femur averaged 4% of the BM space in the ND WT group and 85% in the T2D DD group (Fig. 7Ai and ii). RS504393 treatment blunted the effect of T2D on fat encroachment, lowering the total area covered by ADs from 85 to 73% (Fig. 7Ai and ii). The decreased BMAT coverage was not associated with differences in the average AD size (Fig. 7Aiii), although the DR group contained more small-size ADs (1–10 μm2), which, in white adipose tissue, are considered metabolically healthy compared with large ADs (Fig. 7Aiv). We also observed that the antagonist caused a decrease in the number of ADs (Fig. 7Av). This decrease in number in the absence of changes in average size suggests that MCP-1 most likely affects the differentiation of BM cells into ADs in situ.

Figure 7

In vivo inhibition of MCP-1 signaling decreases fat accumulation and increases osteocyte content in BM of T2D mice. An in vivo study was performed in mice to assess the effect of MCP-1 antagonism on systemic and BM end points. This figure illustrates the BM outcomes for fat and bone at 12 weeks of age after 8 weeks of treatment. Abbreviations are the same as in Fig. 6: ND vehicle (DMSO)–treated Wt/Db mice [ND WT (DMSO) group], DMSO-treated obese T2D Db/Db mice [T2D DD (DMSO) group], and MCP-1 inhibitor (RS504393)–treated T2D Db/Db mice [T2D DR (RS504393) group]. A: Histology of mouse femoral metaphysis BM. H&E staining image at original magnification ×4 and ×20 (Ai). Scatter plots illustrating the quantification of AD coverage over total BM area (Aii) and average AD size (Aiii) and bar graph showing the AD distribution according to size categories (Aiv). Scatter plot showing the quantification of the number of ADs per microscopic field (Av). B: Scatter plots illustrating the results of qPCR analysis of FABP4 (Bi), ADIPOQ (Bii), and PLIN1 (Biii) mRNA expression in the whole BM cell population. C: Analyses performed on the whole BM cell population from tibia at week 8 of treatment with mRNA quantification of murine Mcp-1 (Ci) and Mcpip (Cii) expression. MCP-1 levels measured by ELISA at week 8 of treatment in plasma (Ciii) and cell-free BM lavage (Civ). Values are means with SE bars. One-way ANOVA (A and B) and nonparametric one-way ANOVA with Kruskal-Wallis correction (C). N = 6 biological replicates/group. Scale bars, 500 μm (×4) and 100 μm (×20). *P < 0.05; ***P < 0.001 vs. ND WT; #P < 0.05; ##P > 0.01; ###P < 0.001 vs. T2D DD group.

Figure 7

In vivo inhibition of MCP-1 signaling decreases fat accumulation and increases osteocyte content in BM of T2D mice. An in vivo study was performed in mice to assess the effect of MCP-1 antagonism on systemic and BM end points. This figure illustrates the BM outcomes for fat and bone at 12 weeks of age after 8 weeks of treatment. Abbreviations are the same as in Fig. 6: ND vehicle (DMSO)–treated Wt/Db mice [ND WT (DMSO) group], DMSO-treated obese T2D Db/Db mice [T2D DD (DMSO) group], and MCP-1 inhibitor (RS504393)–treated T2D Db/Db mice [T2D DR (RS504393) group]. A: Histology of mouse femoral metaphysis BM. H&E staining image at original magnification ×4 and ×20 (Ai). Scatter plots illustrating the quantification of AD coverage over total BM area (Aii) and average AD size (Aiii) and bar graph showing the AD distribution according to size categories (Aiv). Scatter plot showing the quantification of the number of ADs per microscopic field (Av). B: Scatter plots illustrating the results of qPCR analysis of FABP4 (Bi), ADIPOQ (Bii), and PLIN1 (Biii) mRNA expression in the whole BM cell population. C: Analyses performed on the whole BM cell population from tibia at week 8 of treatment with mRNA quantification of murine Mcp-1 (Ci) and Mcpip (Cii) expression. MCP-1 levels measured by ELISA at week 8 of treatment in plasma (Ciii) and cell-free BM lavage (Civ). Values are means with SE bars. One-way ANOVA (A and B) and nonparametric one-way ANOVA with Kruskal-Wallis correction (C). N = 6 biological replicates/group. Scale bars, 500 μm (×4) and 100 μm (×20). *P < 0.05; ***P < 0.001 vs. ND WT; #P < 0.05; ##P > 0.01; ###P < 0.001 vs. T2D DD group.

Close modal

To assess if the observed changes in BM-AD accumulation correspond to consensual modifications in adipogenesis markers in BM, we quantified the mRNA expression of FABP4, ADIPOQ, and PLIN1 (Fig. 7Bi–iii). We observed an increase in the mRNA levels of the three markers in the untreated T2D DD group, which correlated with the increased AD presence in the BM of these mice. In contrast, antagonism of MCP-1 signaling decreased the levels of these markers, in line with the observed AD decrease. Analysis of murine Mcp-1 mRNA levels in whole BM did not show any difference among the three groups (Fig. 7Ci). In contrast, Mcpip mRNA levels were increased in the DD group as compared with the WT group, with this increase being inhibited by RS504393 in the DR group (Fig. 7Cii). Serum and cell-free BM lavage concentrations of MCP-1 were elevated in T2D DD mice compared with WT controls, with no differences observed between the DD and DR groups (Fig. 7Ciii and iv).

The cortical bone thickness of the tibia was reduced by 50% in T2D mice regardless of treatment (Supplementary Fig. 4Ai and ii). The density of osteocytes embedded inside the bone was also reduced in the DD group compared with the WT group, with this reduction being blunted by RS504393 in the DR group (Supplementary Fig. 4Aiii). We also observed a twofold increase in osteoclast density in DD and DR mice with a corresponding decrease in osteoblast as compared with WT (Supplementary Fig. 4B and C).

Circulating levels of COOH-terminal telopeptide, a marker of bone resorption, did not change among all groups, whereas RANKL and OPG levels were decreased in both groups with diabetes regardless of treatment compared with those without diabetes, resulting in an unchanged RANKL to OPG ratio (Supplementary Fig. 4Di–iv). Sclerostin, a circulating marker of osteocyte activity, was reduced in the DD group compared with ND, with this reduction being attenuated by the MCP-1 antagonist (Supplementary Fig. 4Dv).

Inhibition of MCP-1 Alleviates the Consequences of T2D on HSC Abundance and Fate

HSCs are significantly affected both regarding amount and functionality by T2D (10,30), a result that was confirmed in the present investigation. We further analyzed by flow cytometry whether RS504393 could influence HSC availability in the BM of T2D mice. We defined BM-HSCs as lineage-negative (Linneg), cKitpos, and Sca-1pos cells. LT-HSCs were characterized as LinnegcKitposSca-1posCD34neg, whereas ST-HSCs as LinnegcKitposSca-1posCD34pos (Fig. 8Ai). The percentage of HSCs in the BM of the T2D mice was lower than that of the ND WT group (Fig. 8Aii), with no difference between the DD and DR groups. When considering HSC subpopulations according to their maturity, we found that T2D reduced the relative abundance of LT-HSCs (Fig. 8Aiii) while increasing the abundance of ST-HSCs (Fig. 8Aiv). This unbalance was corrected by treatment with the MCP-1 antagonist in the T2D DR group (Fig. 8Aiii and iv), suggesting that T2D impinges upon the HSC fate determination and that MCP-1 inhibition blocks this effect. Stem cell factor (SCF) has been shown to be secreted by BM-ADs and able to support hematopoiesis (15). However, the levels of SCF in the cell-free BM lavage were similar among the studied groups (Fig. 8Av). To verify if MCP-1 acts directly on HSCs, we exposed BM mononuclear cells to the CM of BM-ADs from subjects without diabetes or T2D BM-ADs for 96 h and analyzed the antigenic phenotypes of the HSC population by flow cytometry (Fig. 8Bi). Compared with the No-CM group, both the ND-CM and T2D-CM groups showed a slight but significant decrease in total HSCs (Fig. 8Bii) and LT-HSCs (Fig. 8Biii), which was associated with a corresponding increase in ST-HSC (Fig. 8Biv). Cotreatment with RS504393 did not affect LT- and SC-HSC levels compared with the vehicle while still inhibiting MCPIP expression (Fig. 8Bii–v).

Figure 8

In vivo inhibition of MCP-1 signaling rescues the content of primitive HSCs in BM of T2D mice. An in vivo study was performed in mice to assess the effect of MCP-1 antagonism on systemic and BM end points. This figure illustrates the BM outcomes about HSCs. Abbreviations are the same as in Fig. 6: ND vehicle (DMSO)–treated Wt/Db mice [ND WT (DMSO) group], DMSO-treated obese T2D Db/Db mice [T2D DD (DMSO) group], and MCP-1 inhibitor (RS504393)–treated T2D Db/Db mice [T2D DR (RS504393) group]. Flow cytometry analysis of tibia BM. A: Analysis of the BM HSC population. Total BM HSCs were defined as LinnegcKitposSca-1pos, LT-HSCs as LinnegcKitposSca-1posCD34neg, and ST-HSCs as LinnegcKitposSca-1posCD34pos (Ai). Quantification of HSCs (Aii), LT-HSCs (Aiii), and ST-HSCs (Aiv) in total BM cells. Scatter plot with means and SE bars. *P < 0.05; **P < 0.01 vs. ND WT; #P < 0.05 vs. T2D DD group. N = 6 biological replicates/group. B: In a separate in vitro experiment, murine BM mononuclear cells were exposed to BM-AD CM from subjects without diabetes (ND-CM) or with T2D (T2D-CM) (using the unconditioned medium [No CM] as control) for 96 h in the presence or absence of the MCP-1 antagonist, and the HSC population was analyzed by flow cytometry. (Bi) Representative flow cytometry gating. Quantification of total HSCs (Bii), LT-HSCs (Biii), and ST-HSCs (Biv). Relative expression of MCPIP mRNA (Bv). Values are means with SE bars. One-way ANOVA (A) and two-way ANOVA with Tukey correction (B). N = 4 biological replicates/group. *P < 0.05; **P < 0.01; ***P < 0.001 vs. corresponding No CM group; ##P < 0.01 vs. No CM group; &P < 0.05; &&P < 0.01 vs. corresponding No Inhibitor group. FITC, fluorescein isothiocyanate; FSC-a, forward light scatter area; SSC-a, side scatter area.

Figure 8

In vivo inhibition of MCP-1 signaling rescues the content of primitive HSCs in BM of T2D mice. An in vivo study was performed in mice to assess the effect of MCP-1 antagonism on systemic and BM end points. This figure illustrates the BM outcomes about HSCs. Abbreviations are the same as in Fig. 6: ND vehicle (DMSO)–treated Wt/Db mice [ND WT (DMSO) group], DMSO-treated obese T2D Db/Db mice [T2D DD (DMSO) group], and MCP-1 inhibitor (RS504393)–treated T2D Db/Db mice [T2D DR (RS504393) group]. Flow cytometry analysis of tibia BM. A: Analysis of the BM HSC population. Total BM HSCs were defined as LinnegcKitposSca-1pos, LT-HSCs as LinnegcKitposSca-1posCD34neg, and ST-HSCs as LinnegcKitposSca-1posCD34pos (Ai). Quantification of HSCs (Aii), LT-HSCs (Aiii), and ST-HSCs (Aiv) in total BM cells. Scatter plot with means and SE bars. *P < 0.05; **P < 0.01 vs. ND WT; #P < 0.05 vs. T2D DD group. N = 6 biological replicates/group. B: In a separate in vitro experiment, murine BM mononuclear cells were exposed to BM-AD CM from subjects without diabetes (ND-CM) or with T2D (T2D-CM) (using the unconditioned medium [No CM] as control) for 96 h in the presence or absence of the MCP-1 antagonist, and the HSC population was analyzed by flow cytometry. (Bi) Representative flow cytometry gating. Quantification of total HSCs (Bii), LT-HSCs (Biii), and ST-HSCs (Biv). Relative expression of MCPIP mRNA (Bv). Values are means with SE bars. One-way ANOVA (A) and two-way ANOVA with Tukey correction (B). N = 4 biological replicates/group. *P < 0.05; **P < 0.01; ***P < 0.001 vs. corresponding No CM group; ##P < 0.01 vs. No CM group; &P < 0.05; &&P < 0.01 vs. corresponding No Inhibitor group. FITC, fluorescein isothiocyanate; FSC-a, forward light scatter area; SSC-a, side scatter area.

Close modal

In this study, we provide novel extensive evidence about the cellular and molecular mechanisms underpinning fat accumulation in the BM of patients with T2D and also show BMAT could adversely influence the BM microenvironment by paracrine signaling to neighbor cells.

T2D BM-MSCs Are Primed to an Adipogenic Fate

In animal models of obesity, AD hypertrophy precedes hyperplasia in response to the increase in calories requiring storage (31). These dynamic changes are hard to monitor in real time in patients. Our study captured an advanced stage of BM remodeling in subjects with T2D, in whom a twofold accumulation of BMAT was associated with increased AD size and density. Paracrine factors released by enlarged ADs may induce neighboring pre-ADs to grow and differentiate, strengthening the adipose tissue energy reservoir. In line with this, we observed BM-ADs paracrinally stimulate the intrinsic capacity of the BM-MSC pool to differentiate into new ADs. In theory, a reduced clearance may also contribute to expanding the AD pool. However, evidence indicates the canonical apoptotic molecular machinery is upregulated in the visceral adipose tissue of patients with T2D (32,33). Therefore, apoptosis may not be a good explanation for enhanced adiposity in our setting. More likely, hyperglycemia and oxidative stress, which we showed to be activated in BM from subjects with diabetes (10,30), could be responsible for a dysregulated differentiation of precursor cells into ADs. A recent report has attributed a pathogenic role to reactive oxygen species in peripheral obesity (34). Moreover, reactive oxygen species are acknowledged to facilitate differentiation of pre-ADs into mature ADs by accelerating mitotic clonal expansion (35,36). In line with this, we show that BM-MSCs of patients with T2D have an increased intrinsic adipogenic potential, without changing their osteogenic potential. Undifferentiated T2D BM-MSCs show upregulated expression of proadipogenic factors and downregulation of antiadipogenic factors at steady state. C/EBPα is central for AD differentiation and lipid accumulation (37). Hence, high levels of C/EBPα suppose a priming of BM-MSCs toward adipogenesis. PDGFRβ is pivotally implicated in the control of adipogenesis, as supported by lineage-tracing experiments in mice showing PDGFRβ-positive mural stem cells are the originators of adipose tissue hyperplasia under high-fat diet conditions (38). Our finding of a twofold increase in PDGFRβ-positive BM-MSCs of patients with T2D further supports a readiness toward fat accumulation.

Adipogenesis Feedback Loop Between BMAT and BM-MSCs in the BM From Subjects With Diabetes

Using an in vitro approach, we showed that the secretome of T2D BM-ADs could bolster the fate commitment of naive BM-MSCs into ADs. This phenomenon creates a vicious cycle of fat accumulation in the BM from subjects with diabetes, as the appearance of mature ADs will send more adipogenic signals to the BM-MSCs, which will differentiate into more ADs. Furthermore, our findings suggest that it is not the BMAT volume that negatively modulates osteogenesis, but the pathophysiological status of the adipose tissue itself. For example, secretion of adiponectin by BM-ADs (39), which acts positively on osteoblast differentiation, is downregulated in patients with T2D. This decrease could explain why AD CM from subjects without diabetes promotes osteoblastogenesis, whereas T2D AD CM does not.

T2D Alters Adipokine Expression and Secretion in BM-ADs

In line with our results, previous studies have shown that high glucose and hypoxia can induce MCP-1 expression in ADs and other cellular systems via reactive oxygen species (36,40). Analysis of AD transcripts and secretome showed the upregulation of LEP and RETN and downregulation of ADIPOQ in human BM-ADs, which follows the patterns observed in other white adipose deposits of patients with T2D (41). LEP reportedly induces adipogenesis and inhibits osteoblastogenesis in the BM (42). RETN is also relevant to bone remodeling, as it activates osteoclast differentiation (43). Moreover, BMAT is a major source of ADIPOQ (12). Hence, the decrease in ADIPOQ production may contribute to the decrease in circulating levels of ADIPOQ previously reported in patients with T2D (44). MMP2 could also participate in the positive-feedback loop of adipogenesis in the BM of individuals with T2D, possibly through a previously reported in vitro action on pre-AD differentiation (45).

T2D BM-ADs Stimulate Adipogenesis in BM-MSCs via MCP-1

We decided to conduct a more in-depth investigation on MCP-1 for several reasons. First, we know from the literature that MCP-1 can induce AD differentiation, osteoclastogenesis, and bone reabsorption via the transcription factor MCPIP (25,26,46) and that high levels of circulating MCP-1 are associated with insulin resistance through induction of an inflammatory response in adipose tissue (21). Second, we found that, within a spectrum of chemokines and growth factors differentially modulated in T2D BM-ADs, MCP-1 is upregulated by 10-fold as compared with counterparts without diabetes. Third, we demonstrated that this phenomenon is conserved among species, as MCP-1 levels are augmented in BM-ADs of T2D Db/Db mice. Nevertheless, no previous study has examined the possibility that BM-AD–produced MCP-1 drives the conversion of MSCs into new ADs in diabetes. We verified this possibility by performing an adipogenic induction assay on naive BM-MSCs stimulated with BM-AD CM in the presence of RS504393, an MCP-1 receptor antagonist, or its vehicle. Results indicate RS504393 inhibits the transcription of the adipogenic markers ADIPOQ, PPARγ, and FABP4. Importantly, in line with our hypothesis, this molecular response was associated with the complete abolition of the T2D BM-AD secretome’s ability to induce BM-MSC differentiation into mature ADs.

Systemic Inhibition of MCP-1 Rescues BM Adipogenic Remodeling and Increases Osteocyte Density in T2D Mice

We next performed an in vivo validation study in T2D Db/Db mice, with the primary objective of demonstrating MCP-1 antagonism can halt the accumulation of BM-ADs. Systemic RS504393 for 8 weeks resulted in the inhibition of MCP-1 downstream effector MCPIP, downregulation of adipogenic factors FABP4, ADIPOQ, and PLIN1, and, last but not least, amelioration of several pathological features, including a significant effect on the designed primary end point. BMAT reduction by RS504393 was mainly due to a lowered number of ADs, which was associated with a shift toward smaller ADs. This suggests that MCP-1 antagonism not only limits adipogenesis but also promotes the formation of healthier and metabolically active ADs (47). Investigation of BM-AD content by osmium tetroxide and micro computed tomography imaging may allow a more direct approach to visualize global changes of adiposity throughout the bone (48).

Similar to the study published by Kang et al. (23), we observed a slower weight gain in treated Db/Db mice, which was reflected by a lowered epididymal fat pad weight through a reduction in AD size. In addition, RS504393 treatment reduced urinary glucose and plasma HbA1c levels, thus suggesting MCP-1 receptor antagonism may benefit metabolic control. Further investigation is necessary to determine if reduced BM adiposity has contributed in improving insulin resistance in our experimental model.

About the status of the bone, a secondary end point of our in vivo study, we found that the cortical bone thickness of Db/Db mice was half as thin as that of control ND Wt/Db mice, and RS504393 could not rescue this phenotype. However, we did observe changes in bone-regulating osteocytes. In untreated Db/Db mice, the density of osteocytes was greatly reduced as compared with the ND Wt/Db mice. Osteocytes are responsible for bone mechano-sensing, homeostasis, and remodeling, as they coordinate the activity of osteoclasts and osteoblasts (49). It has been shown that a reduction of osteocyte lacunae in older patients is associated with defects in bone remodeling and fracture healing (50,51). MCP-1 antagonist treatment of Db/Db mice increased osteocyte density in the femoral bone to an intermediate level between ND Wt/Db and untreated Db/Db, possibly suggesting this may be a viable strategy to improve bone health in T2D. Sclerostin is produced by the osteocytes, and its levels in the circulation followed the changes in osteocyte density, decreasing in T2D and being partially rescued by RS504393. However, these changes could be counterintuitive, as sclerostin has antianabolic effects on bone formation through inhibition of the Wnt signaling (52). Also, we could not observe any benefit on various cellular and molecular end points of bone formation and degradation, including osteoblast and osteoclast density, RANKL to OPG ratio, and circulating levels of COOH-terminal telopeptide, a specific marker for the degradation of mature type I collagen, which is elevated in patients with increased bone resorption.

MCP-1 Inhibition Rescues the Deficit of LT-HSCs in T2D BM

We showed a decrease in total HSC levels in the two groups with T2D treated with the MCP-1 antagonist or its vehicle as compared with those without diabetes. These data confirm the depletive action of diabetes and also suggest that MCP-1 does not directly contribute to HSC rarefaction. However, when looking at the abundance of specific subpopulations, we found that MCP-1 antagonism rescued the T2D-associated primitive LT-HSC deficit, restoring the levels observed in ND mice. This was contrasted by the lack of effect of RS504393 in an in vitro assay, in which the antagonist failed to inhibit the effect AD CM on the relative abundance of CD34pos and CD34neg HSCs. Altogether, these findings suggest that secreted factors other than MCP-1 are responsible for the paracrine influence of T2D BM-ADs on HSC lineage progression and that treatment with RS504393 corrects the proportion of LT- and SC-HSCs indirectly through amelioration of general and local conditions, such as improved metabolic control and inhibition of BM adipogenesis. Reduced AD presence in the BM signifies a decrease in secretion of factors that negatively affect HSC maintenance, thus permitting the recovery of proper primitive LT-HSC levels. In line with this, BM reconstitution studies showed that inhibition of adipogenesis is beneficial for hematopoietic tissue recovery (53). In contrast, other studies suggest that BMAT is instrumental to HSC maintenance, mainly via the secretion of SCF (15). In this regard, we could not find any significant change in SCF levels in the cell-free murine BM lavage among studied groups.

In conclusion, this study highlights a key paracrine mechanism by which adipose tissue accumulates in the BM of subjects with diabetes and perpetuates BM-MSC adipogenic differentiation. This study demonstrates for the first time that MCP-1 inhibition provides a means for protection of the BM microenvironment in diabetes, reducing BMAT volume and AD size and supporting hematopoiesis. These results have important clinical and therapeutic implications for the maintenance of the BM integrity. We suggest that clinical MCP-1 receptor antagonism could pave the way to novel treatments of T2D and associated comorbidities.

Acknowledgments. The authors thank Dr. Andrew Herman, Lorena Sueiro Ballesteros, and the University of Bristol, Faculty of Biomedical Sciences Flow Cytometry Facility for assistance. The authors also thank the Italian Leukemia-Lymphomas Association, a section of Treviso, for the valid contribution to the project.

Funding. This study was supported by British Heart Foundation grant RG/13/17/30545, “Unravelling mechanisms of stem cell depletion for the preservation of regenerative fitness in patients with diabetes,” and Italian Ministry of Health, Istituto di Ricovero e Cura a Carattere Scientifico MultiMedica Ricerca Corrente and grant RF-2011-02346867.

Duality of Interest. No potential conflicts of interest relevant to this article were reported.

Author Contributions. D.F.-M. contributed to the article by establishing the hypothesis and research protocol of this study, generating the data of Figs. 28, as well as drafting and correcting the manuscript. D.M. contributed to the article by generating the data of Fig. 1, correcting the manuscript, and participating in the scientific discussion on the structure and data of the paper. G.S. and M.S. contributed to the article by providing human BM histology samples, correcting the manuscript, and participating in the scientific discussion on the structure and data of the paper. N.S. and A.B. contributed to the article through surgical expertise and collection of human BM. P.M. is the principal investigator of the research project and contributed to the article by obtaining British Heart Foundation funding, generating ideas, participating in the scientific discussion on the structure and data of the paper, participating in the drafting and correction of the article, as well as providing invaluable mentoring to D.F.-M. D.F.-M. and P.M. are the guarantors of this work and, as such, had full access to all of the data in the study and take responsibility for the integrity of the data and the accuracy of the data analysis.

Prior Presentation. Parts of this study were presented in poster form at the Keystone Symposia on Molecular and Cellular Biology: Bioenergetics and Metabolic Disease (J4), Keystone, CO, 21–25 January 2018.

1.
Beckman
JA
,
Paneni
F
,
Cosentino
F
,
Creager
MA
.
Diabetes and vascular disease: pathophysiology, clinical consequences, and medical therapy: part II
.
Eur Heart J
2013
;
34
:
2444
2452
[PubMed]
2.
Ding
D
,
Qiu
J
,
Li
X
, et al
.
Hyperglycemia and mortality among patients with coronary artery disease
.
Diabetes Care
2014
;
37
:
546
554
[PubMed]
3.
Giacco
F
,
Brownlee
M
.
Oxidative stress and diabetic complications
.
Circ Res
2010
;
107
:
1058
1070
[PubMed]
4.
Fadini
GP
,
Ciciliot
S
,
Albiero
M
.
Concise review: perspectives and clinical implications of bone marrow and circulating stem cell defects in diabetes
.
Stem Cells
2017
;
35
:
106
116
[PubMed]
5.
Patel
RS
,
Li
Q
,
Ghasemzadeh
N
, et al
.
Circulating CD34+ progenitor cells and risk of mortality in a population with coronary artery disease
.
Circ Res
2015
;
116
:
289
297
[PubMed]
6.
Rigato
M
,
Bittante
C
,
Albiero
M
,
Avogaro
A
,
Fadini
GP
.
Circulating progenitor cell count predicts microvascular outcomes in type 2 diabetic patients
.
J Clin Endocrinol Metab
2015
;
100
:
2666
2672
[PubMed]
7.
Ferraro
F
,
Lymperi
S
,
Méndez-Ferrer
S
, et al
.
Diabetes impairs hematopoietic stem cell mobilization by altering niche function
.
Sci Transl Med
2011
;
3
:
104ra101
[PubMed]
8.
Hardouin
P
,
Rharass
T
,
Lucas
S
.
Bone marrow adipose tissue: to be or not to be a typical adipose tissue
?
Front Endocrinol (Lausanne)
2016
;
7
:
85
[PubMed]
9.
Green
DE
,
Rubin
CT
.
Consequences of irradiation on bone and marrow phenotypes, and its relation to disruption of hematopoietic precursors
.
Bone
2014
;
63
:
87
94
[PubMed]
10.
Spinetti
G
,
Cordella
D
,
Fortunato
O
, et al
.
Global remodeling of the vascular stem cell niche in bone marrow of diabetic patients: implication of the microRNA-155/FOXO3a signaling pathway
.
Circ Res
2013
;
112
:
510
522
[PubMed]
11.
Ambrosi
TH
,
Scialdone
A
,
Graja
A
, et al
.
Adipocyte accumulation in the bone marrow during obesity and aging impairs stem cell-based hematopoietic and bone regeneration
.
Cell Stem Cell
2017
;
20
:
771
784
.e776
12.
Cawthorn
WP
,
Scheller
EL
,
Learman
BS
, et al
.
Bone marrow adipose tissue is an endocrine organ that contributes to increased circulating adiponectin during caloric restriction
.
Cell Metab
2014
;
20
:
368
375
[PubMed]
13.
Krings
A
,
Rahman
S
,
Huang
S
,
Lu
Y
,
Czernik
PJ
,
Lecka-Czernik
B
.
Bone marrow fat has brown adipose tissue characteristics, which are attenuated with aging and diabetes
.
Bone
2012
;
50
:
546
552
[PubMed]
14.
Bianco
P
,
Robey
PG
.
Skeletal stem cells
.
Development
2015
;
142
:
1023
1027
[PubMed]
15.
Zhou
BO
,
Yu
H
,
Yue
R
, et al
.
Bone marrow adipocytes promote the regeneration of stem cells and haematopoiesis by secreting SCF
.
Nat Cell Biol
2017
;
19
:
891
903
[PubMed]
16.
Schwartz
AV
,
Sigurdsson
S
,
Hue
TF
, et al
.
Vertebral bone marrow fat associated with lower trabecular BMD and prevalent vertebral fracture in older adults
.
J Clin Endocrinol Metab
2013
;
98
:
2294
2300
[PubMed]
17.
Holt
V
,
Caplan
AI
,
Haynesworth
SE
.
Identification of a subpopulation of marrow MSC-derived medullary adipocytes that express osteoclast-regulating molecules: marrow adipocytes express osteoclast mediators
.
PLoS One
2014
;
9
:
e108920
[PubMed]
18.
Falank
C
,
Fairfield
H
,
Reagan
MR
.
Signaling interplay between bone marrow adipose tissue and multiple myeloma cells
.
Front Endocrinol (Lausanne)
2016
;
7
:
67
[PubMed]
19.
Hardaway
AL
,
Herroon
MK
,
Rajagurubandara
E
,
Podgorski
I
.
Bone marrow fat: linking adipocyte-induced inflammation with skeletal metastases
.
Cancer Metastasis Rev
2014
;
33
:
527
543
[PubMed]
20.
Deshmane
SL
,
Kremlev
S
,
Amini
S
,
Sawaya
BE
.
Monocyte chemoattractant protein-1 (MCP-1): an overview
.
J Interferon Cytokine Res
2009
;
29
:
313
326
[PubMed]
21.
Panee
J
.
Monocyte chemoattractant protein 1 (MCP-1) in obesity and diabetes
.
Cytokine
2012
;
60
:
1
12
[PubMed]
22.
Weisberg
SP
,
Hunter
D
,
Huber
R
, et al
.
CCR2 modulates inflammatory and metabolic effects of high-fat feeding
.
J Clin Invest
2006
;
116
:
115
124
[PubMed]
23.
Kang
YS
,
Lee
MH
,
Song
HK
, et al
.
CCR2 antagonism improves insulin resistance, lipid metabolism, and diabetic nephropathy in type 2 diabetic mice
.
Kidney Int
2010
;
78
:
883
894
[PubMed]
24.
Yang
SJ
,
IglayReger
HB
,
Kadouh
HC
,
Bodary
PF
.
Inhibition of the chemokine (C-C motif) ligand 2/chemokine (C-C motif) receptor 2 pathway attenuates hyperglycaemia and inflammation in a mouse model of hepatic steatosis and lipoatrophy
.
Diabetologia
2009
;
52
:
972
981
[PubMed]
25.
Younce
CW
,
Azfer
A
,
Kolattukudy
PE
.
MCP-1 (monocyte chemotactic protein-1)-induced protein, a recently identified zinc finger protein, induces adipogenesis in 3T3-L1 pre-adipocytes without peroxisome proliferator-activated receptor gamma
.
J Biol Chem
2009
;
284
:
27620
27628
[PubMed]
26.
Younce
C
,
Kolattukudy
P
.
MCP-1 induced protein promotes adipogenesis via oxidative stress, endoplasmic reticulum stress and autophagy
.
Cell Physiol Biochem
2012
;
30
:
307
320
[PubMed]
27.
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
:
1796
1808
[PubMed]
28.
Zhang
HH
,
Kumar
S
,
Barnett
AH
,
Eggo
MC
.
Ceiling culture of mature human adipocytes: use in studies of adipocyte functions
.
J Endocrinol
2000
;
164
:
119
128
[PubMed]
29.
Hemmrich
K
,
Thomas
GP
,
Abberton
KM
, et al
.
Monocyte chemoattractant protein-1 and nitric oxide promote adipogenesis in a model that mimics obesity
.
Obesity (Silver Spring)
2007
;
15
:
2951
2957
[PubMed]
30.
Oikawa
A
,
Siragusa
M
,
Quaini
F
, et al
.
Diabetes mellitus induces bone marrow microangiopathy
.
Arterioscler Thromb Vasc Biol
2010
;
30
:
498
508
[PubMed]
31.
Jo
J
,
Gavrilova
O
,
Pack
S
, et al
.
Hypertrophy and/or hyperplasia: dynamics of adipose tissue growth
.
PLoS Comput Biol
2009
;
5
:
e1000324
[PubMed]
32.
Herold
C
,
Rennekampff
HO
,
Engeli
S
.
Apoptotic pathways in adipose tissue
.
Apoptosis
2013
;
18
:
911
916
[PubMed]
33.
Alkhouri
N
,
Gornicka
A
,
Berk
MP
, et al
.
Adipocyte apoptosis, a link between obesity, insulin resistance, and hepatic steatosis
.
J Biol Chem
2010
;
285
:
3428
3438
[PubMed]
34.
Youn
JY
,
Siu
KL
,
Lob
HE
,
Itani
H
,
Harrison
DG
,
Cai
H
.
Role of vascular oxidative stress in obesity and metabolic syndrome
.
Diabetes
2014
;
63
:
2344
2355
[PubMed]
35.
Lee
H
,
Lee
YJ
,
Choi
H
,
Ko
EH
,
Kim
JW
.
Reactive oxygen species facilitate adipocyte differentiation by accelerating mitotic clonal expansion
.
J Biol Chem
2009
;
284
:
10601
10609
[PubMed]
36.
Yang
X
,
Wang
Y
,
Gao
G
.
High glucose induces rat mesangial cells proliferation and MCP-1 expression via ROS-mediated activation of NF-κB pathway, which is inhibited by eleutheroside E
.
J Recept Signal Transduct Res
2016
;
36
:
152
157
[PubMed]
37.
Lin
FT
,
Lane
MD
.
Antisense CCAAT/enhancer-binding protein RNA suppresses coordinate gene expression and triglyceride accumulation during differentiation of 3T3-L1 preadipocytes
.
Genes Dev
1992
;
6
:
533
544
[PubMed]
38.
Vishvanath
L
,
MacPherson
KA
,
Hepler
C
, et al
.
Pdgfrβ+ mural preadipocytes contribute to adipocyte hyperplasia induced by high-fat-diet feeding and prolonged cold exposure in adult mice
.
Cell Metab
2016
;
23
:
350
359
[PubMed]
39.
Liu
X
,
Chen
T
,
Wu
Y
,
Tang
Z
.
Role and mechanism of PTEN in adiponectin-induced osteogenesis in human bone marrow mesenchymal stem cells
.
Biochem Biophys Res Commun
2017
;
483
:
712
717
[PubMed]
40.
Famulla
S
,
Horrighs
A
,
Cramer
A
,
Sell
H
,
Eckel
J
.
Hypoxia reduces the response of human adipocytes towards TNFα resulting in reduced NF-κB signaling and MCP-1 secretion
.
Int J Obes
2012
;
36
:
986
992
[PubMed]
41.
Jung
UJ
,
Choi
MS
.
Obesity and its metabolic complications: the role of adipokines and the relationship between obesity, inflammation, insulin resistance, dyslipidemia and nonalcoholic fatty liver disease
.
Int J Mol Sci
2014
;
15
:
6184
6223
[PubMed]
42.
Yue
R
,
Zhou
BO
,
Shimada
IS
,
Zhao
Z
,
Morrison
SJ
.
Leptin receptor promotes adipogenesis and reduces osteogenesis by regulating mesenchymal stromal cells in adult bone marrow
.
Cell Stem Cell
2016
;
18
:
782
796
[PubMed]
43.
Thommesen
L
,
Stunes
AK
,
Monjo
M
, et al
.
Expression and regulation of resistin in osteoblasts and osteoclasts indicate a role in bone metabolism
.
J Cell Biochem
2006
;
99
:
824
834
[PubMed]
44.
Aleidi
S
,
Issa
A
,
Bustanji
H
,
Khalil
M
,
Bustanji
Y
.
Adiponectin serum levels correlate with insulin resistance in type 2 diabetic patients
.
Saudi Pharm J
2015
;
23
:
250
256
[PubMed]
45.
Bouloumié
A
,
Sengenès
C
,
Portolan
G
,
Galitzky
J
,
Lafontan
M
.
Adipocyte produces matrix metalloproteinases 2 and 9: involvement in adipose differentiation
.
Diabetes
2001
;
50
:
2080
2086
[PubMed]
46.
Miyamoto
K
,
Ninomiya
K
,
Sonoda
KH
, et al
.
MCP-1 expressed by osteoclasts stimulates osteoclastogenesis in an autocrine/paracrine manner
.
Biochem Biophys Res Commun
2009
;
383
:
373
377
[PubMed]
47.
Weyer
C
,
Foley
JE
,
Bogardus
C
,
Tataranni
PA
,
Pratley
RE
.
Enlarged subcutaneous abdominal adipocyte size, but not obesity itself, predicts type II diabetes independent of insulin resistance
.
Diabetologia
2000
;
43
:
1498
1506
[PubMed]
48.
Scheller
EL
,
Doucette
CR
,
Learman
BS
, et al
.
Region-specific variation in the properties of skeletal adipocytes reveals regulated and constitutive marrow adipose tissues
.
Nat Commun
2015
;
6
:
7808
[PubMed]
49.
Jilka
RL
,
O’Brien
CA
.
The role of osteocytes in age-related bone loss
.
Curr Osteoporos Rep
2016
;
14
:
16
25
[PubMed]
50.
Torres-Lagares
D
,
Tulasne
JF
,
Pouget
C
,
Llorens
A
,
Saffar
JL
,
Lesclous
P
.
Structure and remodelling of the human parietal bone: an age and gender histomorphometric study
.
J Craniomaxillofac Surg
2010
;
38
:
325
330
[PubMed]
51.
Bach-Gansmo
FL
,
Brüel
A
,
Jensen
MV
,
Ebbesen
EN
,
Birkedal
H
,
Thomsen
JS
.
Osteocyte lacunar properties and cortical microstructure in human iliac crest as a function of age and sex
.
Bone
2016
;
91
:
11
19
[PubMed]
52.
Ellies
DL
,
Viviano
B
,
McCarthy
J
, et al
.
Bone density ligand, Sclerostin, directly interacts with LRP5 but not LRP5G171V to modulate Wnt activity
.
J Bone Miner Res
2006
;
21
:
1738
1749
[PubMed]
53.
Naveiras
O
,
Nardi
V
,
Wenzel
PL
,
Hauschka
PV
,
Fahey
F
,
Daley
GQ
.
Bone-marrow adipocytes as negative regulators of the haematopoietic microenvironment
.
Nature
2009
;
460
:
259
263
[PubMed]
Readers may use this article as long as the work is properly cited, the use is educational and not for profit, and the work is not altered. More information is available at http://www.diabetesjournals.org/content/license.

Supplementary data