Mobilization of hematopoietic stem/progenitor cells (HSPC) from the bone marrow (BM) is impaired in diabetes. Excess oncostatin M (OSM) produced by M1 macrophages in the diabetic BM signals through p66Shc to induce Cxcl12 in stromal cells and retain HSPC. BM adipocytes are another source of CXCL12 that blunts mobilization. We tested a strategy of pharmacologic macrophage reprogramming to rescue HSPC mobilization. In vitro, PPAR-γ activation with pioglitazone switched macrophages from M1 to M2, reduced Osm expression, and prevented transcellular induction of Cxcl12. In diabetic mice, pioglitazone treatment downregulated Osm, p66Shc, and Cxcl12 in the hematopoietic BM, restored the effects of granulocyte-colony stimulation factor (G-CSF), and partially rescued HSPC mobilization, but it increased BM adipocytes. Osm deletion recapitulated the effects of pioglitazone on adipogenesis, which was p66Shc independent, and double knockout of Osm and p66Shc completely rescued HSPC mobilization. In the absence of OSM, BM adipocytes produced less CXCL12, being arguably devoid of HSPC-retaining activity, whereas pioglitazone failed to downregulate Cxcl12 in BM adipocytes. In patients with diabetes on pioglitazone therapy, HSPC mobilization after G-CSF was partially rescued. In summary, pioglitazone reprogrammed BM macrophages and suppressed OSM signaling, but sustained Cxcl12 expression by BM adipocytes could limit full recovery of HSPC mobilization.

Studies performed in experimental and human diabetes have consistently shown an impaired mobilization of hematopoietic stem/progenitor cells (HSPC) from the bone marrow (BM) to peripheral blood (PB) after stimulation with granulocyte-colony stimulation factor (G-CSF) (1). Such mobilization defect precedes the reduction of circulating HSPC (2,3), which, in turn, is a driver of micro- and macrovascular complications (4,5). On this basis, it has been hypothesized that restoring normal HSPC levels could exert protective effects against the future risk of complications (6,7). It is therefore of interest to identify therapeutic strategies to counter the so-called “diabetic stem cell mobilopathy” (8).

We have previously described a molecular pathway contributing to mobilopathy. Hyperglycemia stimulates myelopoiesis by BM progenitors (9), thereby expanding proinflammatory (M1) macrophages, which secrete excess amounts of oncostatin M (OSM) (10). In turn, OSM signals in BM mesenchymal stem/stromal cells (MSC) and induces the expression and release of CXCL12 (10). This effect requires cytosolic p66Shc, serving as an adaptor protein for OSM receptor (OSMR) and leading to Cxcl12 expression (11). CXCL12 is the major regulator of HSPC traffic in and out the BM: high levels of CXCL12 keep HSPC in their niches (12), whereas mobilizing stimuli act by reducing intramarrow CXCL12 concentrations (like G-CSF [13]) by desensitizing cells to CXCL12 (like plerixafor [14]) or by inverting the gradient of CXCL12 from the BM to PB (dipeptidyl peptidase 4 [DPP4] inhibitors [15]). The observation that DPP-4 inhibitors and plerixafor mobilize HSPC in diabetes (7) supports the concept that an ineffective CXCL12 switch causes diabetes-associated G-CSF unresponsiveness (1). In addition, the BM of diabetic rodents and patients with diabetes undergoes extensive remodeling, including adipocyte accumulation (8,16,17). BM adipocytes contribute to forming the HSPC niche and their expansion after BM injury favors hematopoietic reconstitution (18). Adipocytes are a source of CXCL12 and other niche gene products capable of retaining HSPC in the niche and preventing their mobilization (1820).

Since OSM inhibition or deletion reduced BM Cxcl12 expression and rescued HSPC mobilization in diabetic mice (10,11), and given that proinflammatory M1 macrophages selectively produce OSM (10), we herein tested a pharmacologic strategy of macrophage reprogramming to counter mobilopathy. To this end, we took advantage of prior observations that PPAR-γ promotes M2 macrophage polarization (21). As PPAR-γ prominently regulates adipose tissue biology (22), we also explored the interplay among the OSM-p66Shc pathway, BM adipogenesis, and HSPC mobilization.

Mice

C57BL/6J wild-type (Wt) mice were purchased from The Jackson Laboratory and established as a colony since 2001. p66Shc−/− mice were obtained from Pier Giuseppe Pelicci’s laboratory (European Institute of Oncology, Milan, Italy); a colony was established in 2010 and backcrossed on the C57BL/6J background for >10 generations. Osm−/− mice on the C57BL/6J background were obtained from GSK (Stevenage, U.K.), and a colony was established since 2015. p66Shc−/−Osm−/− double-knockout (DKO) mice were generated by crossing p66Shc−/− mice and Osm−/−. For all the experiments, 2- to 4-month-old male mice were used; whenever female mice were included, the male:female ratio was 1:1 in all groups of each experiment. Diabetes was induced in 2-month-old animals by a single intraperitoneal injection of streptozotocin (STZ) at 175 mg/kg in 100 mmol/L Na-Citrate buffer, pH 4.5. Blood glucose was measured using a glucometer (FreeStyle; Abbott, Abbott Park, IL). HSPC mobilization was induced by injecting animals subcutaneously with 200 μg/kg/die human recombinant G-CSF daily for four consecutive days. Pioglitazone (DOC Generici, Milano, Italy) was given to animals by daily gavage at 20 mg/kg for 4 weeks. Diabetic mice were treated with pioglitazone since the onset of diabetes. For evaluation of in vivo OSM effects, 3-month-old male mice were injected subcutaneously with PBS or carrier-free recombinant mouse OSM (495-MO/CF; R&D Systems, Minneapolis, MN) at 0.5 mg per injection every 6 h for 48 h before analysis. Total WBC count was performed using the CELL-DYN Emerald hematology analyzer (Abbott) on fresh EDTA-treated mouse blood.

Assignment of mice to treatments or experimental groups was always determined randomly, based on a computer-generated sequence. All animal studies were approved by the Animal Care and Use Committee of Venetian Institute of Molecular Medicine and by the Italian Health Ministry.

Patients

Patients with diabetes and individuals without diabetes were recruited at the University Hospital of Padova. The study for G-CSF–induced mobilization was approved by the local ethics committee, was conducted in accordance with the Declaration of Helsinki as revised in 2000, and is registered at ClinicalTrials.gov (NCT01102699). This was a prospective, parallel-group study of direct BM stimulation with human recombinant G-CSF in subjects with and subjects without diabetes. Specific methods for quantifying blood cells and HSPC have previously been published (2). Informed consent was obtained from all human participants.

Flow Cytometry

Flow cytometry was performed on murine BM or EDTA-treated PB. BM cells were isolated by flushing femurs and tibias with ice-cold MACS Separation Buffer (Miltenyi Biotec, Gladbach, Germany) through a 40 μmol/L cell strainer. PB or BM cells (100 μL) were incubated with antibodies for 15 min in the dark at room temperature. After red blood cell lysis, samples were resuspended in 200 μL PBS, and data were acquired with a FACSCanto (BD Biosciences) cytometer followed by analysis using FlowJo software (Tree Star, Inc.). Antibodies for Linc-kit+Sca-1+ (LKS) cells were anti–Lineage Cocktail, Pacific Blue (clone, B255716; BioLegend); anti–CD117 (c-Kit)-FITC (clone, 2B8; Thermo Fisher Scientific, Waltham, MA); and anti–Ly-6A/E (Sca-1)-PE (clone, D7; Thermo Fisher Scientific). BM macrophages were stained with the following antibodies: anti–Ly-6G (Gr-1)-PE (clone, RB6-8C5; eBioscience), anti-CD115 (c-fms)–Alexa Fluor 488 (clone, AFS98; Thermo Fisher Scientific), and F4/80 APC-eFluor 780 (clone, BM8; Thermo Fisher Scientific).

Human Monocyte–Derived Macrophages

Human peripheral blood mononuclear cells (PBMCs) were isolated from buffy coat from healthy donors by Histopaque-1077 (Sigma-Aldrich) density gradient centrifugation followed by a high-density hyperosmotic Percoll (GE Healthcare) gradient. PBMCs were seeded at 5 × 104 cells/cm2 in RPMI 1640 medium (Corning) containing 10% FBS, 2 mmol/L glutamine, penicillin-streptomycin, and 20 nmol/L recombinant human macrophage–colony stimulating factor (M-CSF; ImmunoTools) for 7 days to obtain resting (M0) macrophages. Medium was changed every 3 days. Polarization stimuli were added for 48 h as follows: for M1 macrophages with lipopolysaccharide (1 μg/mL) (Sigma-Aldrich) and rh interferon-γ (rh IFN-γ [10 ng/mL]) (ImmunoTools) and for M2 macrophages with rh interleukin-4 (rh IL-4 [20 ng/mL]) (ImmunoTools) and rh IL-13 (5 ng/mL) (ImmunoTools). Pioglitazone (10 μmol/L) (Sigma-Aldrich) was added 16 h before polarization and maintained throughout it. Conditioned medium was obtained by adding serum-free medium without stimuli for 48 h.

Mouse BM–Derived Macrophages

BM cells were obtained by flushing with sterile ice-cold PBS both femurs and tibia of 3-month-old mice. Red blood cells were lysed with ammonium-chloride-potassium lysing buffer. To obtain resting (M0) macrophages, we plated 150,000 cells/cm2 on 6-well plates (BD Falcon, New York, NY) with RPMI 1640 medium (Corning) supplemented with glutamine 2 mmol/L, penicillin-streptomycin, and 10% FBS + 10 nmol/L murine M-CSF for 7 days without medium change. For M1 macrophages, cells were incubated 48 h with lipopolysaccharide (1 μg/mL) and mouse recombinant IFN-γ (10 ng/mL). For M2 polarization, resting macrophages were incubated with IL-4 (20 ng/mL) and IL-13 (5 ng/mL). Pioglitazone (10 μmol/L) (Sigma-Aldrich) was added 16 h before polarization and maintained throughout it. Conditioned medium was obtained by adding serum-free medium without stimuli for 48 h.

BM-Derived MSC

Murine BM-Derived MSC (BM-MSC) were isolated by flushing the BM of 3 month-old-mice and cultivating adherent cells with minimum essential medium (MEM)-α medium containing 10% FBS, glutamine 2 mmol/L, and penicillin-streptomycin. Passage 3–6 was used in all experiments. For gene expression analysis, cells were treated with murine recombinant oncostatin-M (R&D Systems) for 48 h in serum-free media.

Human BM-MSC were obtain from the BM of patients undergoing orthopedic surgery at the University Hospital of Padova (ethics committee protocol no. 2868P). Aspirates were washed twice with ice-cold sterile PBS, and the cell pellets were plated on tissue culture petri dishes (BD Falcon) with complete mesenchymal medium (MesenCult Mesenchymal Stem Cell Stimulatory Supplement (human); STEMCELL Technologies Inc.). The medium was changed when fibroblast-like cells began to appear and then every other day. Experiments were performed with MCS up to passage 5.

Tissue Analysis

Femurs were fixed in 4% paraformaldehyde, decalcified, and cut in longitudinal sections. Sections were either stained with hematoxylin-eosin (H-E) or processed for immunofluorescence. Hoechst was used to visualize cellularity, whereas an anti-perilipin antibody identified adipocytes. In separate experiments, adipocytes were isolated as previously described (23). For further detail, see Supplementary Material.

Adipogenic Differentiation

Murine BM-MSC were seeded into 12-well plates (BD Falcon) and grown with MEM-α medium containing 10% FBS, glutamine 2 mmol/L, and penicillin-streptomycin until a confluent layer was reached. Adipogenic differentiation was induced by incubating the cells for 8 days with DMEM/F12 (1:1) medium (Sigma-Aldrich) containing 10% FBS, glutamine 2 mmol/L, and penicillin-streptomycin; 170 μmol/L pantothenic acid (MP Biomedicals); 33 μmol/L biotin (MP Biomedicals); 66 nmol/L insulin (Sigma-Aldrich); 0.25 mmol/L 3-isobutyl-1-methylxanthine (Sigma-Aldrich); 100 nmol/L dexamethasone (Sigma-Aldrich); 1 nmol/L 3,3′,5-triiodo-l-thyronine; Sigma-Aldrich); and 10 μmol/L rosiglitazone. Medium was changed every 3 days. For the oil red O (ORO) staining, cells were fixed with 4% paraformaldehyde for 10 min and after a rinse with PBS were stained for 15 min at room temperature with a 0.5% solution (w/v) of ORO in isopropanol. ORO was eluted by adding 100% isopropanol, and absorbance was read at 540 nm. Alternatively, after fixation cells were stained with 10 μmol/L BODIPY 493/503 (Invitrogen, Milano, Italy) for 30 min and counterstained with H33342 (Sigma-Aldrich) for 5 min.

Molecular Biology

RNA was isolated from flushed BM or cells by use of QIAzol or with a Total RNA Purification Micro Kit (Norgen Biotek) and quantified with a NanoDrop 2000 Spectrophotometer (Thermo Fisher Scientific). cDNA was synthesized from 500 ng RNA using a SensiFAST cDNA Synthesis Kit (Bioline, London, U.K.). Quantitive PCR was performed using SensiFAST SYBR Lo-ROX Kit (Bioline) via a QuantStudio 5 Real-Time PCR System (Thermo Fisher Scientific). A list of primers can be found in Supplementary Table 1.

GEO Data Set Gene Expression Analysis

GSE27017 data sets (24) were browsed using the open-source GEO2R software. After defining two groups that contained BM adipocyte sample data and epididymal adipocyte sample data, respectively, we extracted and analyzed expression values of selected genes.

Statistical Analysis

Continuous data are expressed as mean ± SE unless otherwise specified, whereas categorical data are presented as percentage or as fold of basal. Normality was checked using the Kolmogorov-Smirnov test, and nonnormal data were log transformed before analysis. Comparison between two or more groups was performed using the Student t test and ANOVA for normal variables or the Mann-Whitney U test and Kruskal-Wallis test for nonnormal variables that could not be log transformed (e.g., because of frequent zero values). All tests were two tailed. Bonferroni adjustment was used to account for multiple testing. Biological replicates are shown for every experiment. Whenever there were <7 of 10 replicates, individual data points are shown. Significance was conventionally accepted at P < 0.05. Number of biological replicates and significance are reported in the figure legends.

Data and Resource Availability

The data sets generated during or analyzed during the current study are available from the corresponding author upon reasonable request.

Rescue of HSPC Mobilization by Pharmacologic OSM Suppression in Diabetic Mice

We first tested whether PPAR-γ activation modified BM macrophage phenotype and reduced OSM signaling. We found that, in vitro, the antidiabetes PPAR-γ agonist pioglitazone reduced expression of the M1 gene Tnfa by ∼50% and increased that of the M2 gene Mrc ∼4× in murine BM-derived macrophages, concomitantly reducing Osm expression in M1-polarized macrophages (Fig. 1A). In contrast to the conditioned medium of control M1 macrophages, the condition medium of pioglitazone-treated M1 macrophages failed to upregulate Cxcl12 in murine BM-MSC (Fig. 1B and C). The surge in OSM protein concentration found in the conditioned medium of M1 macrophages was significantly blunted by pioglitazone (Fig. 1D), suggesting that reduction of OSM in the medium was responsible for blocking Cxcl12 induction. Indeed, the conditioned medium from Osm−/− BM macrophages, treated with either vehicle or pioglitazone, failed to upregulate Cxcl12 in BM-MSC (Fig. 1E).

Figure 1

Pharmacologic rescue of mobilopathy by PPAR-γ activation. A: Gene expression of mouse BM macrophages cultured in M1 or M2 polarizing conditions with (w) and without (wo, control) treatment with the PPAR-γ agonist pioglitazone (*P < 0.05 vs. control). B: Schematic representation of the experiment performed to evaluate whether macrophage conditioned medium (CM) induces Cxcl12 expression in BM-MSC. C: Mouse BM-MSC were incubated with conditioned medium from Wt mouse BM macrophages polarized toward M0, M1, or M2 in the presence or in the absence (control) of pioglitazone (*P < 0.05 vs. M0; #P < 0.05 vs. untreated control). D: Concentrations of OSM, measured by ELISA, in conditioned medium from M0 macrophages and M1 macrophages without (control) or with pioglitazone treatment (*P < 0.05 for the indicated comparison). E: Mouse BM-MSC were incubated with conditioned medium from Osm−/− BM macrophages polarized toward M0, M1, or M2 in the presence or in the absence (control) of pioglitazone. Histograms indicate mean ± SEM with superimposed individual data points. For isolation of macrophages and MSC, we used an equal number of male (50%) and female (50%) mice.

Figure 1

Pharmacologic rescue of mobilopathy by PPAR-γ activation. A: Gene expression of mouse BM macrophages cultured in M1 or M2 polarizing conditions with (w) and without (wo, control) treatment with the PPAR-γ agonist pioglitazone (*P < 0.05 vs. control). B: Schematic representation of the experiment performed to evaluate whether macrophage conditioned medium (CM) induces Cxcl12 expression in BM-MSC. C: Mouse BM-MSC were incubated with conditioned medium from Wt mouse BM macrophages polarized toward M0, M1, or M2 in the presence or in the absence (control) of pioglitazone (*P < 0.05 vs. M0; #P < 0.05 vs. untreated control). D: Concentrations of OSM, measured by ELISA, in conditioned medium from M0 macrophages and M1 macrophages without (control) or with pioglitazone treatment (*P < 0.05 for the indicated comparison). E: Mouse BM-MSC were incubated with conditioned medium from Osm−/− BM macrophages polarized toward M0, M1, or M2 in the presence or in the absence (control) of pioglitazone. Histograms indicate mean ± SEM with superimposed individual data points. For isolation of macrophages and MSC, we used an equal number of male (50%) and female (50%) mice.

Close modal

To verify the relevance of this effect in vivo, we used mice with type 1–like diabetes induced by STZ. This model was chosen for two reasons: 1) mobilopathy is complete and severe in STZ-induced diabetes, while it is milder in mice with high-fat diet–induced type 2–like diabetes (Supplementary Fig. 1), and 2) pioglitazone is expected to induce less confounding metabolic effects in STZ than in high-fat diet mice. We treated diabetic and nondiabetic mice with pioglitazone for 4 weeks. In diabetic mice, PPAR-γ activation nonsignificantly reduced glucose levels (361 ± 21 vs. 412 ± 25 mg/dL; P = 0.44) and did not affect body weight (Fig. 2A and B), indicating that pioglitazone exerted no substantial systemic metabolic effects in this model. Successful PPAR-γ stimulation in the BM was confirmed by transactivation of typical target genes (Plin1, 2×, and Abca1, 3×), transrepression of the M1 macrophage gene Il1b (0.4×), and induction of M2 macrophage genes Cd36 (>40×) and Mertk (5×) (Fig. 2C). Treatment of diabetic mice with pioglitazone did not improve features of enhanced myelopoiesis associated with diabetes, including the raise in granulocyte-to-lymphocyte ratio in PB and the increased number of macrophages in the BM (Fig. 2D and E). However, pioglitazone restored normal suppression of BM macrophages (Fig. 2F) and rescued the Cxcl12 switch (70% suppression; P = 0.015) (Fig. 2G) after G-CSF in diabetic mice. Suppression of OSM and CXCL12 levels in the BM plasma by pioglitazone was confirmed at the protein level (Fig. 2H). While nondiabetic mice promptly mobilized HSPC approximately eightfold after G-CSF, diabetic mice treated with vehicle were fully unresponsive to G-CSF (Fig. 2I). On the contrary, diabetic mice treated with pioglitazone showed a partial rescue of HSPC mobilization (4.1×).

Figure 2

In vivo effects of pioglitazone. A and B: Average glucose levels (A) and body weight (B) of nondiabetic control mice and mice with STZ-induced diabetes treated with pioglitazone for 4 weeks (n = 8–10/group). C: Expression of PPAR-γ target genes in the BM of diabetic mice treated with pioglitazone vs. untreated controls (*P < 0.05 vs. control). DF: White blood cell (WBC) count (D), the granulocyte-to-lymphocyte ratio (G/L) (E), and the percentage of BM macrophages over total BM cellularity (F) (*P < 0.05 vs. basal; #P < 0.05 vs. nondiabetic) in nondiabetic and diabetic mice with or without pioglitazone treatment. GI: BM Cxcl12 gene expression (G), OSM and CXCL12 protein level (H), and HSPC mobilization (I) after G-CSF stimulation in nondiabetic control mice and in diabetic mice with or without pioglitazone treatment (*P < 0.05 vs. basal; #P < 0.05 vs. nondiabetic). Histograms indicate mean ± SEM. For all experiments, we used an equal number of male (50%) and female (50%) mice. Pioglit., pioglitazone.

Figure 2

In vivo effects of pioglitazone. A and B: Average glucose levels (A) and body weight (B) of nondiabetic control mice and mice with STZ-induced diabetes treated with pioglitazone for 4 weeks (n = 8–10/group). C: Expression of PPAR-γ target genes in the BM of diabetic mice treated with pioglitazone vs. untreated controls (*P < 0.05 vs. control). DF: White blood cell (WBC) count (D), the granulocyte-to-lymphocyte ratio (G/L) (E), and the percentage of BM macrophages over total BM cellularity (F) (*P < 0.05 vs. basal; #P < 0.05 vs. nondiabetic) in nondiabetic and diabetic mice with or without pioglitazone treatment. GI: BM Cxcl12 gene expression (G), OSM and CXCL12 protein level (H), and HSPC mobilization (I) after G-CSF stimulation in nondiabetic control mice and in diabetic mice with or without pioglitazone treatment (*P < 0.05 vs. basal; #P < 0.05 vs. nondiabetic). Histograms indicate mean ± SEM. For all experiments, we used an equal number of male (50%) and female (50%) mice. Pioglit., pioglitazone.

Close modal

Role of BM Adipogenesis

By staining BM sections for perilipin, we observed that, in both diabetic and nondiabetic mice, treatment with pioglitazone increased BM adipocytes (Fig. 3A and B). This effect is known to occur also in humans (25,26) and is consistent with the ability of PPAR-γ to promote adipogenic differentiation of MSC (27). Importantly, STZ-induced diabetes per se resulted in accumulation of BM adipocytes (Fig. 3C and D). Average adipocyte area was not affected, ruling out that increased perilipin staining was due to hypertrophy (Fig. 3E). Excess adipocytes are believed to contribute to BM remodeling and HSPC mobilopathy in diabetes, owing to the expression and release of CXCL12 (19) and other paracrine factors (e.g., MCP-1) (28). In fact, by immunofluorescence staining of the BM, we found an intact CXCL12 signal close to adipocytes (Fig. 3F). Also, Cxcl12 gene expression in freshly isolated BM adipocytes (BMAds) was markedly higher than in the whole BM (Fig. 3G). Since Cxcl12 expression in BM-MSC is controlled by OSM through p66Shc (11), we verified whether the same was in adipocytes. Using an ex vivo organoid culture system (Fig. 3H), we found that stimulation of visceral adipose tissue (VAT) with OSM induced Cxcl12 expression in Wt (approximately threefold; P = 0.03) but not in p66Shc−/− cells (Fig. 3I). As origin and properties of BM adipose tissue differ from those of VAT (24), we measured Cxcl12 expression after treating mice with recombinant OSM or vehicle: while OSM induced Cxcl12 in the unfractioned BM, the very high Cxcl12 expression in freshly isolated BMAds could not be further increased by OSM (Fig. 3J). To explain this site-specific effect of OSM, we mined publicly available gene expression profiles comparing BMAds with peripheral adipose tissue: this analysis confirmed higher expression of Cxcl12 in BMAds and showed that BMAds had significantly lower gene expression for OSMR and its cognate gp130 and LIF receptors than VAT (Supplementary Fig. 2). This finding can explain why BMAd Cxcl12 expression could not be stimulated by OSM. However, consistent with the notion that OSM drives osteogenic at the expense of adipogenic differentiation of MSC (29), we found that Osm−/− mice displayed strikingly increased amounts of BM adipocytes compared with Wt mice either in the diabetic or nondiabetic condition (Fig. 3K and L). Immunostaining signal for intact CXCL12 around BM adipocytes was diminished by ∼40% in Osm−/− versus Wt mice (P = 0.013) (Fig. 3M and N), and Cxcl12 gene expression was lower in freshly isolated BMAds of Osm−/− mice compared with Wt mice (Fig. 3O). Thus, despite the fact that Cxcl12 expression was very high in BMAds and could not be further stimulated by OSM, these data still support that OSM can control Cxcl12 expression in BMAds as in MSC.

Figure 3

BM adipogenesis. A: Representative serial BM sections stained with H-E (left panels) or immunostained (right panels) for Hoechst (blue [total cellularity]) and perilipin (red [adipocytes]) from nondiabetic or diabetic mice with or without pioglitazone treatment. B: Quantification of adipocyte number (Ad.N) per marrow area (Ma.Ar) in nondiabetic or diabetic mice with or without pioglitazone treatment (two-way ANOVA showed significant effects of diabetes and treatment; *P < 0.05 vs. control; #P < 0.05 vs. nondiabetic). C: Representative serial BM sections stained with H-E or immunostained with Hoechst and perilipin to quantify BMAds in nondiabetic control mice and in mice with STZ-induced diabetes. D: Quantification of BMAds in nondiabetic control mice and in mice with STZ-induced diabetes (*P < 0.05 vs. nondiabetic). E: Average adipocyte area in nondiabetic control mice and in mice with STZ-induced diabetes. F: Representative BM sections stained for Hoechst (blue) and intact (full-length) CXCL12 (red). For diabetic and nondiabetic mice, a longitudinal femur section stained with H-E or immunostained is presented with the respective magnification (arrows indicate adipocytes, identified by morphology). G: Comparative gene expression analysis of Cxcl12 in the unfractioned whole BM vs. the BMAds (*P < 0.05). H: Schematic representation of the culture and stimulation of VAT organoids. I: Induction of Cxcl12 gene expression in VAT organoids from Wt or p66Shc−/− mice by OSM treatment (*P < 0.05 vs. control). J: Analysis of Cxcl12 gene expression in the whole unfractioned BM and in freshly isolated BMAds in mice treated in vivo with control vehicle (Ctrl) (white) or with recombinant OSM (red) (*P < 0.05 vs. control). K: Representative BM sections stained with Hoechst and perilipin to quantify BMAds in nondiabetic and diabetic Wt or Osm−/− mice (representative low-magnification images are shown in Supplementary Fig. 3). L: Quantification of BMAds in nondiabetic and diabetic Wt or Osm−/− mice (*P < 0.05 vs. Wt; #P < 0.05 vs. control). M: Serial BM sections of Wt and Osm−/− nondiabetic mice stained with H-E or immunostained (IF) with Hoechst (blue) and for perilipin (green) (adipocytes) and intact (full-length) CXCL12 (red). Magnifications of the areas inside white squares are shown to illustrate signal colocalization. N: CXCL12 signal relative to the background staining (excluding bone areas) was quantified around adipocytes in an area between two cell diameters (*P < 0.05 vs. Wt). O: Gene expression analysis of Cxcl12 in freshly isolated BMAds of Wt and Osm−/− mice (*P < 0.05 vs. Wt). Histograms indicate mean ± SEM with superimposed individual data points. Sex of mice: 100% males in panels AG and KO and 50% males and 50% females, respectively, in panels HJ.

Figure 3

BM adipogenesis. A: Representative serial BM sections stained with H-E (left panels) or immunostained (right panels) for Hoechst (blue [total cellularity]) and perilipin (red [adipocytes]) from nondiabetic or diabetic mice with or without pioglitazone treatment. B: Quantification of adipocyte number (Ad.N) per marrow area (Ma.Ar) in nondiabetic or diabetic mice with or without pioglitazone treatment (two-way ANOVA showed significant effects of diabetes and treatment; *P < 0.05 vs. control; #P < 0.05 vs. nondiabetic). C: Representative serial BM sections stained with H-E or immunostained with Hoechst and perilipin to quantify BMAds in nondiabetic control mice and in mice with STZ-induced diabetes. D: Quantification of BMAds in nondiabetic control mice and in mice with STZ-induced diabetes (*P < 0.05 vs. nondiabetic). E: Average adipocyte area in nondiabetic control mice and in mice with STZ-induced diabetes. F: Representative BM sections stained for Hoechst (blue) and intact (full-length) CXCL12 (red). For diabetic and nondiabetic mice, a longitudinal femur section stained with H-E or immunostained is presented with the respective magnification (arrows indicate adipocytes, identified by morphology). G: Comparative gene expression analysis of Cxcl12 in the unfractioned whole BM vs. the BMAds (*P < 0.05). H: Schematic representation of the culture and stimulation of VAT organoids. I: Induction of Cxcl12 gene expression in VAT organoids from Wt or p66Shc−/− mice by OSM treatment (*P < 0.05 vs. control). J: Analysis of Cxcl12 gene expression in the whole unfractioned BM and in freshly isolated BMAds in mice treated in vivo with control vehicle (Ctrl) (white) or with recombinant OSM (red) (*P < 0.05 vs. control). K: Representative BM sections stained with Hoechst and perilipin to quantify BMAds in nondiabetic and diabetic Wt or Osm−/− mice (representative low-magnification images are shown in Supplementary Fig. 3). L: Quantification of BMAds in nondiabetic and diabetic Wt or Osm−/− mice (*P < 0.05 vs. Wt; #P < 0.05 vs. control). M: Serial BM sections of Wt and Osm−/− nondiabetic mice stained with H-E or immunostained (IF) with Hoechst (blue) and for perilipin (green) (adipocytes) and intact (full-length) CXCL12 (red). Magnifications of the areas inside white squares are shown to illustrate signal colocalization. N: CXCL12 signal relative to the background staining (excluding bone areas) was quantified around adipocytes in an area between two cell diameters (*P < 0.05 vs. Wt). O: Gene expression analysis of Cxcl12 in freshly isolated BMAds of Wt and Osm−/− mice (*P < 0.05 vs. Wt). Histograms indicate mean ± SEM with superimposed individual data points. Sex of mice: 100% males in panels AG and KO and 50% males and 50% females, respectively, in panels HJ.

Close modal

We reasoned that suppression of Cxcl12 in BMAds could contribute to the rescued HSPC mobilization that we have previously shown in diabetic Osm−/− mice (11), and thus we checked whether the same was true in pioglitazone-treated mice. While the expression of Osm and Cxcl12 was reduced by pioglitazone in the unfractioned diabetic BM, expression of Cxcl12 specifically in BMAds was unaffected and remained elevated (Fig. 4A and B). Therefore, contrary to what was observed in Osm−/− mice (Fig. 3M–O), sustained Cxcl12 expression by the expanded BM adipose tissue in pioglitazone-treated mice could limit HSPC mobilization.

Figure 4

Effects of pioglitazone on gene expression. Gene expression of Osm (A), Cxcl12 (B), p66Shc (C), and Dpp4 (D) was analyzed in the whole unfractioned BM and in freshly isolated BMAds of diabetic mice treated with vehicle (Ctrl) or pioglitazone. *P < 0.05 vs. control; #P < 0.05 vs. BM. E: DPP4 activity was measured in the BM fluid in diabetic mice treated with vehicle or pioglitazone. Histograms indicate mean ± SEM with superimposed individual data points. Only male mice were used.

Figure 4

Effects of pioglitazone on gene expression. Gene expression of Osm (A), Cxcl12 (B), p66Shc (C), and Dpp4 (D) was analyzed in the whole unfractioned BM and in freshly isolated BMAds of diabetic mice treated with vehicle (Ctrl) or pioglitazone. *P < 0.05 vs. control; #P < 0.05 vs. BM. E: DPP4 activity was measured in the BM fluid in diabetic mice treated with vehicle or pioglitazone. Histograms indicate mean ± SEM with superimposed individual data points. Only male mice were used.

Close modal

As an additional pathway, we explored DPP4, which is released by VAT (30) and degrades CXCL12 (31). We found that gene expression of Dpp4 was not significantly different between BMAds and the rest of BM (Fig. 4D) or VAT (Supplementary Fig. 2); neither Dpp4 expression nor DPP4 activity was affected by pioglitazone treatment (Fig. 4D and E).

Intriguingly, pioglitazone significantly reduced p66Shc expression in the whole BM and in BMAds (Fig. 4C). Since p66Shc regulates HSPC mobilization in different ways (11,32), we explored the interplay among Osm and p66Shc. Consistent with prior studies showing that p66Shc-derived H2O2 sustains adipogenesis (33), we found less adipocyte accumulation in the BM of diabetic p66Shc−/− versus Wt mice (Fig. 5A–C). We then evaluated whether and how p66Shc affected the adipogenic differentiation of BM-derived MSC. In vitro, MSC isolated from the BM of p66Shc−/− had a 60% lower adipogenic differentiation capacity compared with that of Wt MSC (Fig. 5D and E). Adipogenesis by p66Shc−/− BM-MSC could be partially rescued by transfection with a vector encoding for Wt but not with that encoding for 36Ser→Ala p66Shc mutant (Fig. 5F and G), which does not translocate to mitochondria (34). Since OSMR recruits cytosolic p66Shc to activate the cascade culminating in Cxcl12 expression (11,35), it is possible that Osm deletion increased the pool of p66Shc available for mitochondrial translocation, thereby stimulating adipogenesis. To verify this hypothesis, we generated Osm−/−p66Shc−/− DKO mice. DKO mice were viable and fertile, with a normal reproductive fitness and no overt pathologic phenotype (data not shown). The extent of BM adipocyte accumulation in DKO mice was even greater than that observed in Osm−/− mice, ruling out that adipogenesis induced by Osm deficiency depended on p66Shc (Fig. 5H and I). As expected, BMAds from DKO expressed markedly lower levels of Cxcl12 compared with Wt mice (Supplementary Fig. 4). DKO mice showed high levels of phenotypically (LKS) and functionally (colonies) defined HSPC in the unstimulated PB (Fig. 5J and K). In addition, despite massively increased BM adipocytes, HSPC mobilization after G-CSF in diabetic DKO mice was restored to normal (Fig. 5L).

Figure 5

OSM-p66Shc interplay in BM adipogenesis. A: Representative serial BM sections stained with H-E or immunostained with Hoechst (blue) and for perilipin (red) to visualize BMAds in nondiabetic and diabetic Wt and p66Shc−/− mice (note that images for Wt mice are reproduced from Fig. 3C for consistency). B: Quantification of BM adipocytes (Ad.N) per marrow area (Ma.Ar) in nondiabetic and diabetic Wt and p66Shc−/− mice (*P < 0.05 vs. nondiabetic; #P < 0.05 vs. Wt). C: Average adipocyte area in nondiabetic and diabetic Wt and p66Shc−/− mice. D: BM-derived MSC from Wt or p66Shc−/− mice were incubated with control or adipogenic medium for 8 days. Undifferentiated cells had a classical spindle-shaped morphology and virtually no lipid accumulation, while differentiated cells were round shaped and accumulated single or multiple lipid droplets. Representative images display a merge of phase contrast, Hoechst staining (blue [nuclei]), and BODIPY staining (green [lipids]). E: Adipogenesis was quantified from ORO optical density as fold change vs. undifferentiated cells (*P < 0.05 vs. control; #P < 0.05 vs. wild type). F: BM-MSC from p66Shc−/− mice transfected with vectors encoding Wt p66Shc (p66WT) or 36Ser→Ala mutant (p66S36A) were incubated with control or adipogenic medium, and lipid accumulation was evaluated with BODIPY qualitative staining. G: Quantification of lipid accumulation was performed with ORO quantitative staining in p66Shc−/− MSC transfected with p66WT or p66S36A under unstimulated (white [control]) or adipogenic (red) condition (*P < 0.05 vs. control). H: Representative serial BM sections from Wt or p66Shc−/−Osm−/− DKO mice were stained with H-E or with Hoechst (blue) and anti-perilipin (red). I: Quantification of BMAds in Wt and DKO mice (*P < 0.05 vs. Wt). J: Levels of HSPC, quantified as LKS cells, in unstimulated PB of Wt and Osm−/−p66Shc−/− DKO mice (*P < 0.05 vs. Wt). K: Representative macroscopic photographs of the colony-forming unit (CFU) assay from unstimulated PB of Wt and DKO mice, with quantification (*P < 0.05 vs. Wt). L: Mobilization of phenotypically defined HSPC (LKS cells) in diabetic and nondiabetic DKO mice (*P < 0.05 vs. basal). Histograms indicate mean ± SEM with superimposed individual data points when there were <7–10 biological replicates/group. Sex of mice: 100% males in panels AC and JL, and 50% and 50% males and females, respectively, in panels DI. Ctrl, control.

Figure 5

OSM-p66Shc interplay in BM adipogenesis. A: Representative serial BM sections stained with H-E or immunostained with Hoechst (blue) and for perilipin (red) to visualize BMAds in nondiabetic and diabetic Wt and p66Shc−/− mice (note that images for Wt mice are reproduced from Fig. 3C for consistency). B: Quantification of BM adipocytes (Ad.N) per marrow area (Ma.Ar) in nondiabetic and diabetic Wt and p66Shc−/− mice (*P < 0.05 vs. nondiabetic; #P < 0.05 vs. Wt). C: Average adipocyte area in nondiabetic and diabetic Wt and p66Shc−/− mice. D: BM-derived MSC from Wt or p66Shc−/− mice were incubated with control or adipogenic medium for 8 days. Undifferentiated cells had a classical spindle-shaped morphology and virtually no lipid accumulation, while differentiated cells were round shaped and accumulated single or multiple lipid droplets. Representative images display a merge of phase contrast, Hoechst staining (blue [nuclei]), and BODIPY staining (green [lipids]). E: Adipogenesis was quantified from ORO optical density as fold change vs. undifferentiated cells (*P < 0.05 vs. control; #P < 0.05 vs. wild type). F: BM-MSC from p66Shc−/− mice transfected with vectors encoding Wt p66Shc (p66WT) or 36Ser→Ala mutant (p66S36A) were incubated with control or adipogenic medium, and lipid accumulation was evaluated with BODIPY qualitative staining. G: Quantification of lipid accumulation was performed with ORO quantitative staining in p66Shc−/− MSC transfected with p66WT or p66S36A under unstimulated (white [control]) or adipogenic (red) condition (*P < 0.05 vs. control). H: Representative serial BM sections from Wt or p66Shc−/−Osm−/− DKO mice were stained with H-E or with Hoechst (blue) and anti-perilipin (red). I: Quantification of BMAds in Wt and DKO mice (*P < 0.05 vs. Wt). J: Levels of HSPC, quantified as LKS cells, in unstimulated PB of Wt and Osm−/−p66Shc−/− DKO mice (*P < 0.05 vs. Wt). K: Representative macroscopic photographs of the colony-forming unit (CFU) assay from unstimulated PB of Wt and DKO mice, with quantification (*P < 0.05 vs. Wt). L: Mobilization of phenotypically defined HSPC (LKS cells) in diabetic and nondiabetic DKO mice (*P < 0.05 vs. basal). Histograms indicate mean ± SEM with superimposed individual data points when there were <7–10 biological replicates/group. Sex of mice: 100% males in panels AC and JL, and 50% and 50% males and females, respectively, in panels DI. Ctrl, control.

Close modal

PPAR-γ Activation Reprograms Macrophages and Rescues HSPC Mobilization in Humans

To establish the clinical impact of pioglitazone therapy on HSPC mobilization, we performed in vitro experiments with human macrophages. Pioglitazone reduced OSM expression in M1-polarized macrophages by ∼50% (Fig. 6A). Consequently, the ability of pioglitazone-treated M1 macrophage–conditioned medium to induce CXCL12 expression in human BM-MSC was abolished (Fig. 6B), which was consistent with what was observed with murine cells. We then analyzed data from a study wherein patients with and patients without diabetes received low-dose G-CSF to test HSPC mobilization (2) and divided them in pioglitazone users and nonusers (Table 1). While control subjects without diabetes had doubled PB-HSPC levels 24 h after G-CSF, this was not observed in patients with type 1 diabetes (T1D) (1.0×; P = 0.96) or patients with type 2 diabetes (T2D) not taking pioglitazone (1.2×; P = 0.50). On the contrary, patients with T2D patients on pioglitazone treatment displayed a significant 1.7× mobilization of CD34+ HSPC (Fig. 6C and D). This beneficial effect occurred despite the fact that T2D patients on pioglitazone were older, more obese, and had a higher prevalence of hypertension and atherosclerotic disease (Table 1)—factors that are expected to negatively impact HSPC availability (17). The fold change in CD34+ HSPC levels in pioglitazone-treated patients with diabetes was higher than in T1D patients and T2D patients not taking pioglitazone combined but still lower than that observed in individuals without diabetes.

Figure 6

Effects of pioglitazone on HSPC mobilization in humans. A: Expression of Osm in human PB monocyte-derived macrophages cultured in M0 or M1 polarizing conditions (*P < 0.05 vs. control; #P < 0.05 vs. M0). B: Human BM-MSC were incubated with conditioned medium from human macrophages polarized toward M0 or M1 in the presence or in the absence of pioglitazone (*P < 0.05 vs. control; #P < 0.05 vs. M0). C: Levels of human circulating HSPC, defined as CD34+ cells, before and after G-CSF stimulation in patients without diabetes (control [Ctrl]), in patients with T1D, and in patients with T2D with or without pioglitazone treatment (*P < 0.05 vs. baseline; #P < 0.05 vs. control). D: Fold change increase in human HSPC from panel C (*P < 0.05 vs. control; #P < 0.05 for the comparison between pioglitazone-treated T2D patients and other T1D and T2D patients combined). Histograms indicate mean ± SEM with superimposed individual data points. Piogl, pioglitazone.

Figure 6

Effects of pioglitazone on HSPC mobilization in humans. A: Expression of Osm in human PB monocyte-derived macrophages cultured in M0 or M1 polarizing conditions (*P < 0.05 vs. control; #P < 0.05 vs. M0). B: Human BM-MSC were incubated with conditioned medium from human macrophages polarized toward M0 or M1 in the presence or in the absence of pioglitazone (*P < 0.05 vs. control; #P < 0.05 vs. M0). C: Levels of human circulating HSPC, defined as CD34+ cells, before and after G-CSF stimulation in patients without diabetes (control [Ctrl]), in patients with T1D, and in patients with T2D with or without pioglitazone treatment (*P < 0.05 vs. baseline; #P < 0.05 vs. control). D: Fold change increase in human HSPC from panel C (*P < 0.05 vs. control; #P < 0.05 for the comparison between pioglitazone-treated T2D patients and other T1D and T2D patients combined). Histograms indicate mean ± SEM with superimposed individual data points. Piogl, pioglitazone.

Close modal
Table 1

Characteristics of patients in the HSPC mobilization substudy

No diabetesT1DT2D without pioglitazoneT2D with pioglitazoneP
n 14 13 11  
Age, years 40.1 ± 14.3 38.5 ± 9.0 55.4 ± 9.4* 59.6 ± 5.2* <0.001 
Male sex 78.5 84.6 81.8 80.0 0.937 
BMI, kg/m2 25.8 ± 4.6 25.1 ± 3.0 29.6 ± 8.2 34.0 ± 6.7 0.017 
Diabetes duration, years 0.0 18.5 ± 9.9* 12.8 ± 12.4* 13.6 ± 6.1* <0.001 
HbA1c, % (mmol/mol) 5.0 ± 0.2 (31.0 ± 2.0) 8.0 ± 1.2 (64.0 ± 9.6)* 7.9 ± 1.1 (63.0 ± 8.8)* 8.1 ± 1.8 (65.0 ± 14.4)* <0.001 
Hypertension 14.3 30.7 90.9* 80.0* <0.001 
Retinopathy 0.0 46.2* 18.2 0.0 0.009 
Nephropathy 0.0 7.7 9.0 20.0 0.511 
Neuropathy 0.0 30.7 18.2 40.0 0.116 
Atherosclerosis 0.0 15.4 36.4 60.0* 0.014 
Insulin 0.0 100.0* 72.7* 60.0* <0.001 
Oral agents 0.0 7.6 72.7* 100.0* <0.001 
 Metformin 0.0 7.6 81.8* 80.0*  
 Sulfonylurea 0.0 0.0 36.3* 40.0*  
 DPP4i inhibitors 0.0 0.0 27.2* 20.0  
Statin 0.0 15.4 81.8* 20.0# 0.014 
ACEi/ARBs 7.1 15.4 81.8* 20.0# <0.001 
No diabetesT1DT2D without pioglitazoneT2D with pioglitazoneP
n 14 13 11  
Age, years 40.1 ± 14.3 38.5 ± 9.0 55.4 ± 9.4* 59.6 ± 5.2* <0.001 
Male sex 78.5 84.6 81.8 80.0 0.937 
BMI, kg/m2 25.8 ± 4.6 25.1 ± 3.0 29.6 ± 8.2 34.0 ± 6.7 0.017 
Diabetes duration, years 0.0 18.5 ± 9.9* 12.8 ± 12.4* 13.6 ± 6.1* <0.001 
HbA1c, % (mmol/mol) 5.0 ± 0.2 (31.0 ± 2.0) 8.0 ± 1.2 (64.0 ± 9.6)* 7.9 ± 1.1 (63.0 ± 8.8)* 8.1 ± 1.8 (65.0 ± 14.4)* <0.001 
Hypertension 14.3 30.7 90.9* 80.0* <0.001 
Retinopathy 0.0 46.2* 18.2 0.0 0.009 
Nephropathy 0.0 7.7 9.0 20.0 0.511 
Neuropathy 0.0 30.7 18.2 40.0 0.116 
Atherosclerosis 0.0 15.4 36.4 60.0* 0.014 
Insulin 0.0 100.0* 72.7* 60.0* <0.001 
Oral agents 0.0 7.6 72.7* 100.0* <0.001 
 Metformin 0.0 7.6 81.8* 80.0*  
 Sulfonylurea 0.0 0.0 36.3* 40.0*  
 DPP4i inhibitors 0.0 0.0 27.2* 20.0  
Statin 0.0 15.4 81.8* 20.0# 0.014 
ACEi/ARBs 7.1 15.4 81.8* 20.0# <0.001 

Continuous variables are presented as mean ± SD, whereas categorical variables are presented as percentages. P values are from ANOVA or χ2 tests. ACEi, ACE inhibitors; ARBs, angiotensin receptor blockers; DPP4i, DPP4 inhibitors.

*

P < 0.05 vs. nondiabetic control subjects.

#

P < 0.05 vs. patients with T2D without pioglitazone (all adjusted for multiple testing using Bonferroni).

Countering the mechanisms driving chronic shortage of circulating HSPC is expected to improve micro- and macrovascular outcomes (7). Enhancing HSPC mobilization in patients with diabetes could also improve the outcome of autologous transplantation for the treatment of blood cancer (1). We show that PPAR-γ is a new candidate target to rescue the impaired HSPC mobilization consistently observed in human and experimental diabetes. PPAR-γ has previously been identified as a regulator of HSPC homeostasis (36), mainly by driving an expansion of BMAds. Indeed, PPAR-γ activation switches MSC differentiation from osteoblasts to adipocytes (37). Adipocytes contribute to forming the HSPC niche in the BM by providing soluble mediators that are critically important in regulating cellular quiescence, maintenance, and traffic (18). Under stress conditions, such as after irradiation or chemotherapy, BMAds expand and accelerate recovery of the BM niche and of hematopoietic function (18,38). By producing the HSPC-retaining chemokine CXCL12, BMAds exert antimobilizing activities (19,39). Indeed, adipocyte accumulation is a feature of the diabetic BM and associated with HSPC mobilopathy (16,17). Yet, we found that PPAR-γ activation partially rescued HSPC mobilization, despite increasing BMAds. In vitro, pioglitazone reprogrammed macrophages toward M2 and inhibited OSM signaling and production of CXCL12 by BM stromal cells. Suppression of OSM and CXCL12 was confirmed in vivo, supporting this mechanistic link. Many effects of pioglitazone were recapitulated by Osm deletion. According to our previous findings (11), Cxcl12 expression is controlled by OSM in BM-MSC via p66Shc. While the same appears to be true in VAT, BMAds expressed much higher levels of Cxcl12 that could not be stimulated further, possibly because of lower expression of OSMRs. Nonetheless, in the absence of OSM, BMAds expressed less Cxcl12, being arguably devoid of HSPC-retaining activity. Unexpectedly, despite pioglitazone-suppressed OSM gene and protein expression in the BM, pioglitazone failed to suppress Cxcl12 expression by BMAds, which remained high. This reflects that factors other than OSM concomitantly regulate Cxcl12 and can explain why pioglitazone only partially rescued HSPC mobilization.

Interestingly, we found that, in addition to suppressing OSM, pioglitazone also reduced expression of p66Shc in the whole BM and in BMAds. Cxcl12 induction by OSM requires p66Shc also in adipocytes, but Osm deletion and p66Shc deletion exerted opposite effects on adipogenesis. As cytosolic p66Shc is recruited to allow OSMR signaling (35), in the absence of OSM, more p66Shc might translocate to mitochondria, where it potentiates adipogenesis (33), Using DKO mice, we ruled out that adipogenesis induced by Osm deletion could be prevented by deleting p66Shc. Thus, OSM and p66Shc cooperate to induce Cxcl12, but they regulate the adipogenesis independently. DKO mice mimicked pioglitazone-treated mice with regard to adipogenesis and HSPC mobilization. However, pioglitazone did not completely suppress Osm, did not affect BMAd-derived Cxcl12, and only partially rescued mobilization. In addition, we acknowledge that other effects of pioglitazone could contribute to the improved mobilization and are not necessarily limited to modulation of the OSM-p66Shc pathway.

In this study, we purportedly used a model of T1D to avoid the confounding metabolic effects exerted by pioglitazone in T2D models. Importantly, while diabetic patients were normally unresponsive to the HSPC-mobilizing effect of G-CSF (40), patients with diabetes on pioglitazone showed a partial recovery of HSPC release. This lends support to the clinical transferability of our findings. Notably, pioglitazone is known to increase BM adiposity also in humans (25), which is supposed to make long bones prone to fractures (41). Although we have no information on BM adipocyte content in our patients, the emerging scenario makes a striking parallel between mice and humans, with pioglitazone stimulating BM adipogenesis and being partially able to rescue HSPC mobilization. Interestingly, this occurred despite the fact that pioglitazone did not affect myelopoiesis. Thus, even in the absence of systemic metabolic and anti-inflammatory effects, pioglitazone disconnected HSPC mobilopathy from diabetes-associated myelopoiesis. Pioglitazone has the potential to improve hard outcomes in patients with diabetes (42), and, in view of the strong association between HSPC levels and adverse diabetes outcomes (4,5), we argue that part of this protection could be mediated by rescuing HSPC traffic. Strategies to limit BM adipogenesis might potentiate such effect and improve clinical benefits of PPAR-γ activation.

S.T. and S.C. contributed equally.

This article contains supplementary material online at https://doi.org/10.2337/figshare.12179910.

Funding. The study was supported by the following grants: European Foundation for the Study of Diabetes (EFSD)/Novartis 2013 grant to G.P.F., EFSD/Lilly 2016 grant to G.P.F., Ministry of University and Education PRIN grant 2015 to G.P.F., and Italian Diabetes Society/Lilly grant 2017 to G.P.F.

Duality of Interest. S.C., M.A., and G.P.F. are the inventors of a patent, held by the University of Padova, on the use of pharmacologic oncostatin M inhibition to allow stem cell mobilization.

Author Contributions. S.T., S.C., A.C., A.A., M.A., and G.P.F. designed the research. S.T., S.C., L.M., M.D., V.S., R.C., A.A., and M.A. performed the research. S.T., S.C., L.M., M.D., V.S., A.C., A.A., M.A., and G.P.F. analyzed the data. S.C., A.A., M.A., and G.P.F. wrote the manuscript. All authors reviewed and edited the manuscript. G.P.F. is the guarantor of this work and, as such, had full access to all the data in the study and takes responsibility for the integrity of the data and the accuracy of the data analysis.

1.
Fadini
GP
,
DiPersio
JF
.
Diabetes mellitus as a poor mobilizer condition
.
Blood Rev
2018
;
32
:
184
191
2.
Fadini
GP
,
Albiero
M
,
Vigili de Kreutzenberg
S
, et al
.
Diabetes impairs stem cell and proangiogenic cell mobilization in humans
.
Diabetes Care
2013
;
36
:
943
949
3.
Fadini
GP
,
Boscaro
E
,
de Kreutzenberg
S
, et al
.
Time course and mechanisms of circulating progenitor cell reduction in the natural history of type 2 diabetes
.
Diabetes Care
2010
;
33
:
1097
1102
4.
Fadini
GP
,
Rigato
M
,
Cappellari
R
,
Bonora
BM
,
Avogaro
A
.
Long-term prediction of cardiovascular outcomes by circulating CD34+ and CD34+CD133+ stem cells in patients with type 2 diabetes
.
Diabetes Care
2017
;
40
:
125
131
5.
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
6.
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
7.
Fadini
GP
,
Ferraro
F
,
Quaini
F
,
Asahara
T
,
Madeddu
P
.
Concise review: diabetes, the bone marrow niche, and impaired vascular regeneration
.
Stem Cells Transl Med
2014
;
3
:
949
957
8.
Albiero
M
,
Fadini
GP
.
Pharmacologic targeting of the diabetic stem cell mobilopathy
.
Pharmacol Res
2018
;
135
:
18
24
9.
Brownlee
M
.
Hyperglycemia-stimulated myelopoiesis causes impaired regression of atherosclerosis in type 1 diabetes
.
Cell Metab
2013
;
17
:
631
633
10.
Albiero
M
,
Poncina
N
,
Ciciliot
S
, et al
.
Bone marrow macrophages contribute to diabetic stem cell mobilopathy by producing oncostatin M
.
Diabetes
2015
;
64
:
2957
2968
11.
Albiero
M
,
Ciciliot
S
,
Tedesco
S
, et al
.
Diabetes-associated myelopoiesis drives stem cell mobilopathy through an OSM-p66Shc signaling pathway
.
Diabetes
2019
;
68
:
1303
1314
12.
Asada
N
,
Kunisaki
Y
,
Pierce
H
, et al
.
Differential cytokine contributions of perivascular haematopoietic stem cell niches
.
Nat Cell Biol
2017
;
19
:
214
223
13.
Petit
I
,
Szyper-Kravitz
M
,
Nagler
A
, et al
.
G-CSF induces stem cell mobilization by decreasing bone marrow SDF-1 and up-regulating CXCR4
.
Nat Immunol
2002
;
3
:
687
694
14.
Broxmeyer
HE
,
Orschell
CM
,
Clapp
DW
, et al
.
Rapid mobilization of murine and human hematopoietic stem and progenitor cells with AMD3100, a CXCR4 antagonist
.
J Exp Med
2005
;
201
:
1307
1318
15.
Fadini
GP
,
Albiero
M
,
Seeger
F
, et al
.
Stem cell compartmentalization in diabetes and high cardiovascular risk reveals the role of DPP-4 in diabetic stem cell mobilopathy
.
Basic Res Cardiol
2013
;
108
:
313
16.
Dang
Z
,
Maselli
D
,
Spinetti
G
, et al
.
Sensory neuropathy hampers nociception-mediated bone marrow stem cell release in mice and patients with diabetes
.
Diabetologia
2015
;
58
:
2653
2662
17.
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
18.
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
19.
Mattiucci
D
,
Maurizi
G
,
Izzi
V
, et al
.
Bone marrow adipocytes support hematopoietic stem cell survival
.
J Cell Physiol
2018
;
233
:
1500
1511
20.
Trotter
TN
,
Gibson
JT
,
Sherpa
TL
,
Gowda
PS
,
Peker
D
,
Yang
Y
.
Adipocyte-lineage cells support growth and dissemination of multiple myeloma in bone
.
Am J Pathol
2016
;
186
:
3054
3063
21.
Bouhlel
MA
,
Derudas
B
,
Rigamonti
E
, et al
.
PPARgamma activation primes human monocytes into alternative M2 macrophages with anti-inflammatory properties
.
Cell Metab
2007
;
6
:
137
143
22.
Rosen
ED
,
Sarraf
P
,
Troy
AE
, et al
.
PPAR gamma is required for the differentiation of adipose tissue in vivo and in vitro
.
Mol Cell
1999
;
4
:
611
617
23.
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
24.
Liu
LF
,
Shen
WJ
,
Ueno
M
,
Patel
S
,
Azhar
S
,
Kraemer
FB
.
Age-related modulation of the effects of obesity on gene expression profiles of mouse bone marrow and epididymal adipocytes
.
PLoS One
2013
;
8
:
e72367
25.
Beck
GR
 Jr
.,
Khazai
NB
,
Bouloux
GF
, et al
.
The effects of thiazolidinediones on human bone marrow stromal cell differentiation in vitro and in thiazolidinedione-treated patients with type 2 diabetes
.
Transl Res
2013
;
161
:
145
155
26.
Grey
A
,
Beckley
V
,
Doyle
A
, et al
.
Pioglitazone increases bone marrow fat in type 2 diabetes: results from a randomized controlled trial
.
Eur J Endocrinol
2012
;
166
:
1087
1091
27.
Tontonoz
P
,
Hu
E
,
Spiegelman
BM
.
Stimulation of adipogenesis in fibroblasts by PPAR gamma 2, a lipid-activated transcription factor
.
Cell
1994
;
79
:
1147
1156
28.
Ferland-McCollough
D
,
Maselli
D
,
Spinetti
G
, et al
.
MCP-1 feedback loop between adipocytes and mesenchymal stromal cells causes fat accumulation and contributes to hematopoietic stem cell rarefaction in the bone marrow of patients with diabetes
.
Diabetes
2018
;
67
:
1380
1394
29.
Sato
F
,
Miyaoka
Y
,
Miyajima
A
,
Tanaka
M
.
Oncostatin M maintains the hematopoietic microenvironment in the bone marrow by modulating adipogenesis and osteogenesis
.
PLoS One
2014
;
9
:
e116209
30.
Lamers
D
,
Famulla
S
,
Wronkowitz
N
, et al
.
Dipeptidyl peptidase 4 is a novel adipokine potentially linking obesity to the metabolic syndrome
.
Diabetes
2011
;
60
:
1917
1925
31.
Christopherson
KW
 II
,
Cooper
S
,
Broxmeyer
HE
.
Cell surface peptidase CD26/DPPIV mediates G-CSF mobilization of mouse progenitor cells
.
Blood
2003
;
101
:
4680
4686
32.
Albiero
M
,
Poncina
N
,
Tjwa
M
, et al
.
Diabetes causes bone marrow autonomic neuropathy and impairs stem cell mobilization via dysregulated p66Shc and Sirt1
.
Diabetes
2014
;
63
:
1353
1365
33.
Berniakovich
I
,
Trinei
M
,
Stendardo
M
, et al
.
p66Shc-generated oxidative signal promotes fat accumulation
.
J Biol Chem
2008
;
283
:
34283
34293
34.
Migliaccio
E
,
Giorgio
M
,
Mele
S
, et al
.
The p66shc adaptor protein controls oxidative stress response and life span in mammals
.
Nature
1999
;
402
:
309
313
35.
Hermanns
HM
,
Radtke
S
,
Schaper
F
,
Heinrich
PC
,
Behrmann
I
.
Non-redundant signal transduction of interleukin-6-type cytokines. The adapter protein Shc is specifically recruited to rhe oncostatin M receptor
.
J Biol Chem
2000
;
275
:
40742
40748
36.
Sertorio
M
,
Du
W
,
Amarachintha
S
,
Wilson
A
,
Pang
Q
.
In vivo RNAi screen unveils PPARγ as a regulator of hematopoietic stem cell homeostasis
.
Stem Cell Reports
2017
;
8
:
1242
1255
37.
Takada
I
,
Suzawa
M
,
Matsumoto
K
,
Kato
S
.
Suppression of PPAR transactivation switches cell fate of bone marrow stem cells from adipocytes into osteoblasts
.
Ann N Y Acad Sci
2007
;
1116
:
182
195
38.
Djazayeri
K
,
Szilvássy
Z
,
Peitl
B
, et al
.
Accelerated recovery of 5-fluorouracil-damaged bone marrow after rosiglitazone treatment
.
Eur J Pharmacol
2005
;
522
:
122
129
39.
Wilson
A
,
Fu
H
,
Schiffrin
M
, et al
.
Lack of adipocytes alters hematopoiesis in lipodystrophic mice
.
Front Immunol
2018
;
9
:
2573
40.
Fadini
GP
,
Fiala
M
,
Cappellari
R
, et al
.
Diabetes limits stem cell mobilization following G-CSF but not plerixafor
.
Diabetes
2015
;
64
:
2969
2977
41.
Meier
C
,
Kraenzlin
ME
,
Bodmer
M
,
Jick
SS
,
Jick
H
,
Meier
CR
.
Use of thiazolidinediones and fracture risk
.
Arch Intern Med
2008
;
168
:
820
825
42.
Lincoff
AM
,
Wolski
K
,
Nicholls
SJ
,
Nissen
SE
.
Pioglitazone and risk of cardiovascular events in patients with type 2 diabetes mellitus: a meta-analysis of randomized trials
.
JAMA
2007
;
298
:
1180
1188
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 https://www.diabetesjournals.org/content/license.