Growth hormone (GH), a pleiotropic hormone secreted by the pituitary gland, regulates immune and inflammatory responses. In this study, we show that GH regulates the phenotypic and functional plasticity of macrophages both in vitro and in vivo. Specifically, GH treatment of GM-CSF–primed monocyte–derived macrophages promotes a significant enrichment of anti-inflammatory genes and dampens the proinflammatory cytokine profile through PI3K-mediated downregulation of activin A and upregulation of MAFB, a critical transcription factor for anti-inflammatory polarization of human macrophages. These in vitro data correlate with improved remission of inflammation and mucosal repair during recovery in the acute dextran sodium sulfate–induced colitis model in GH-overexpressing mice. In this model, in addition to the GH-mediated effects on other immune cells, we observed that macrophages from inflamed gut acquire an anti-inflammatory/reparative profile. Overall, these data indicate that GH reprograms inflammatory macrophages to an anti-inflammatory phenotype and improves resolution during pathologic inflammatory responses.

Growth hormone (GH) is produced and secreted by somatotropic cells of the anterior pituitary gland in mammals and is a main regulator of postnatal growth and development. Although initially implicated in somatic growth control, GH is a pleiotropic hormone with myriad functions, including anabolic actions on muscle and bone and catabolic effects on adipose tissue (1). It also promotes fluid retention in the kidney (2), exerts metabolic effects on the liver (3), triggers sexual maturation (4), and induces insulin resistance (5). Many of these effects are mediated through insulin-like growth factor (IGF)-1, which is produced in response to GH in liver and other tissues (6).

Studies both in vitro and in vivo have also demonstrated a role for GH in immune regulation. GH receptor (GHR) is expressed by various leukocyte subsets (7), including B and T cells (8) and macrophages (9), and GH has been shown to mediate thymic development (8), promote T cell engraftment in SCID mice (10), improve B cell responses and Ab production (11, 12), and modulate the activity of NK cells (13) and macrophages (9). In myeloid cells, GH stimulates the proliferation of RAW 264.7 macrophages (14) and functions as a human macrophage–activating factor (15). Beneficial effects of GH administration have been reported in autoimmunity, as it alters tolerization mechanisms through activation of regulatory T cells and modulation of Th17 cell plasticity, reduces type I diabetes development (16), and contributes to improve collagen-induced arthritis symptoms (17). In inflammatory diseases, GH limits mucosal inflammation in experimental colitis (18) and has protective effects in patients with active Crohn’s disease (19, 20).

Tissue-resident macrophages are a heterogeneous and highly plastic cell population whose effector functions are dependent on their ontogeny and the surrounding tissue microenvironment. Their functional versatility enables them to play critical roles in both the initiation and resolution of inflammatory and immune responses. Reflecting this functional versatility, macrophages primed by GM-CSF (GM-Mϕ) or macrophages primed by M-CSF (M-Mϕ) exert opposite functions in terms of T cell stimulation, inflammatory cytokine production, tumor cell growth, and methotrexate sensitivity (2125). Indeed, GM-Mϕ and M-Mϕ are considered as pro- and anti-inflammatory macrophages, respectively, as they display specific transcriptional profiles resembling those of macrophages from the synovia of rheumatoid arthritis patients or tumor-associated macrophages, respectively (26, 27).

We report in this study that GH downmodulates the transcriptional and cytokine profile of proinflammatory GM-Mϕ by reducing, in a PI3K-dependent manner, the secretion of activin A, a protein involved in tilting the balance of macrophages toward a proinflammatory phenotype (28), and upregulating the expression of MAFB, a transcription factor that promotes the anti-inflammatory polarization of macrophages (29, 30). In line with these in vitro results, we demonstrate that GH-overexpressing transgenic (GHTg)–mice show improved remission of inflammation and mucosal repair during recovery from dextran sodium sulfate (DSS)–induced acute colitis. Indeed, together with possible effects on other immune cells, the polarization of gut macrophages from GHTg mice was shifted toward an anti-inflammatory/reparative profile during the recovery phase. Overall, our results point to GH as a regulatory factor for macrophage polarization under physiological and pathological settings.

Mice transgenic for bovine GH under the control of the phosphoenolpyruvate carboxykinase promoter were on a C57BL/6J background (31) and were maintained by repeated backcrosses to C57BL/6J females. A total of 40 GHTg mice and 40 control littermates (10–14 wk old) were subjected to the DSS-induced experimental model of colitis, using matched sex ratios in each experiment. Mice were handled according to national and European Union guidelines, and experiments were approved by the Animal Experimentation Ethics Committee of the Madrid regional government (PROEX 316- 15).

Acute colitis was induced with 3% DSS (w/v, molecular mass 36,000–50,000 Da; MP Biomedicals, Solon, OH) in drinking water for 5 d, followed by a recovery period of an additional 9 d as indicated. Mice were monitored for body weight and signs of distress, diarrhea, and rectal bleeding and (when required) were killed. The colon was removed, washed with PBS, and opened longitudinally for analysis.

When needed, infiltrating immune cells were obtained by enzymatic digestion of the intestinal tissues. Briefly, tissue samples were incubated (30 min, 37°C) in 10 ml HBSS with 0.4 mg/ml Dispase (Life Technologies) and 10,000 U/ml penicillin/streptomycin. FBS was added to 5% (v/v), and tissue debris was removed by sequential filtering through 100, 70, and 40 μm cell strainers. Cells were collected by centrifugation (150 × g, 10 min, 4°C). Colon cells were stained with combinations of fluorescence-labeled Abs to the cell surface markers CD45, CD11b, F4/80, Gr1, CD11c and CD86 and analyzed in a Gallios Flow Cytometer (Beckman Coulter, Brea, CA). The profiles obtained were analyzed with FlowJo software (BD Biosciences); leukocytes were gated as CD45+ cells. F4/80+ cells from mouse colon samples were isolated using anti-F4/80 MicroBeads UltraPure (Miltenyi Biotech) (∼80% purity).

Full-length colon samples were longitudinally divided into two halves for histological and transcriptional analysis. Inflammation and crypt damage were scored on formalin-fixed, paraffin-embedded colon sections stained with H&E. The scoring scheme (32) included inflammation severity (0, none; 1, mild; 2, moderate; and 3, severe), inflammation extent (0, none; 1, mucosa; 2, submucosa; and 3, transmural), crypt damage (0, none; 1, basal one-third damage; 2, basal two-thirds damage; 3, crypt loss, surface epithelium present; and 4, crypt and surface epithelium loss), and percentage of colon involvement (0, 0%; 1, <25%; 2, 25–50%; 3, 50–75%; and 4, >75%). Histological evaluation was performed by two independent investigators in a double-blinded fashion.

For analysis, formalin-fixed, paraffin-embedded colon sections were stained with anti–arginase-1 (BD Biosciences, San Jose, CA) and K13-a anti–inducible NO synthase (iNOS) (Novus Biologicals, Littleton, CO) Abs and immunodetected with Alexa 488–labeled goat anti-rabbit IgG (Molecular Probes, Eugene, OR). Slides were mounted for fluorescence with ProLong Gold reagent with DAPI (Invitrogen, San Diego, CA) and visualized on a TCS SP5 Confocal Microscope (Leica Microsystems, Wetzlar, Germany).

Human PBMCs were isolated from buffy coats of healthy donors by density gradient centrifugation using endotoxin-tested (<0.12 endotoxin units/ml) Ficoll Paque Plus (GE Healthcare, Uppsala, Sweden), according to standard procedures. Monocytes were purified from PBMCs by magnetic cell sorting using CD14 MicroBeads (Miltenyi Biotec, Bergisch Gladbach, Germany). Monocytes (>94% CD14+ cells) were cultured at 0.5 × 106 cells per milliliter for 7 d (37°C, 5% CO2) in RPMI 1640 medium supplemented with 10% FCS, sodium pyruvate and l-glutamine, and containing GM-CSF (1000 U/ml) or M-CSF (10 ng/ml) (both from ImmunoTools, Friesoythe, Germany) to generate GM-Mϕ and M-Mϕ, respectively. Cytokines were added every 2 d. To evaluate the direct effect of GH on monocytes, cells were incubated with recombinant human GH (rhGH) (1 μg/ml Genotonorm; Pfizer, New York, NY; for 7 d, 37°C, 5% CO2). To determine the effect of GH on differentiated GM-Mϕ and M-Mϕ, cells were treated with rhGH (1 μg/ml) in fresh medium for 24 h. To evaluate the effect of IGF-1 on differentiated GM-Mϕ, cells were treated with rhIGF-1 (100 ng/ml) for 24 h, or it was added at the same time as GM-CSF. For signaling analysis, GM-Mϕ and M-Mϕ were stimulated with rhGH (1 μg/ml) for the indicated times. Where appropriate, GM-Mϕ and M-Mϕ were treated with LY294002 (10 μM, 60 min) prior to rhGH addition. Functional polarization of GM-Mϕ and M-Mϕ was determined by quantification of cytokine (TNF-α, IL-10, IL-6) production. Murine GM-Mϕ and M-Mϕ bone marrow–derived macrophages were generated using human M-CSF (10 ng/ml; ImmunoTools) or murine GM-CSF (20 ng/ml; PreproTech), respectively, as previously described (21, 33).

RNA sequencing (RNAseq) was performed on RNA obtained from three independent human samples of untreated or GH-treated (1 μg/ml, 24 h) GM-Mϕ, using the BGISEQ-500 platform (BGI, Hong Kong, China), with 20 million reads and 100 paired-end sequencing reads. Statistical analysis of RNAseq data were performed following previously described procedures (29). Differential gene expression analysis by DSeq2 revealed no statistically significant differences between the experimental conditions. RNAseq data have been deposited in the Sequence Read Archive (https://www.ncbi.nlm.nih.gov/sra) under accession no. PRJNA555143. Gene-set enrichment analysis (GSEA) was performed using data sets available at the Web site (http://software.broadinstitute.org/gsea/index.jsp) (34), as well as data sets containing the genes differentially expressed by either human M-Mϕ (anti-inflammatory gene set) or GM-Mϕ (proinflammatory gene set) (GSEA68061) (28, 35), and the genes specifically downregulated in MAFB-silenced human M-Mϕ (GSE84622) (29).

Mouse samples were collected in RNAlater, and total RNA was extracted with TriReagent (both from Sigma-Aldrich, St Louis, MO). Total RNA from cultured cells was extracted using the Nucleospin RNA/Protein Kit (Macherey-Nagel, Düren, Germany). RNA was retrotranscribed using SuperScript II Reverse Transcriptase (Invitrogen), and real-time quantitative PCR (RT-qPCR) analysis was performed with Power SYBR Green PCR Master Mix (Applied Biosystems, Foster City, CA). Triplicate samples were quantified using the ABI Prism HT7900 sequence detection system (Applied Biosystems). Oligonucleotides for selected genes were designed employing the Roche Universal ProbeLibrary Assay Design Center (Table I). Assays were performed in triplicates and results normalized according to the expression levels of TBP and GAPDH; for murine samples, results were normalized using Actb expression levels. Results were obtained using the 2−∆Ct comparative threshold method for quantification.

Supernatants from untreated and GH-treated GM-Mϕ or M-Mϕ were evaluated for the presence of cytokines and growth factors using commercially available ELISA kits for activin A and CCL2 (Quantikine immunoassay; R&D Systems, Minneapolis, MN) and for IL-6, IL-10, and TNF-α (Human ELISA Max Standard; BioLegend, San Diego, CA) following the protocols supplied by the manufacturers.

GHR expression was determined in human samples using an anti-GHR mAb (36). The incubations were done in the presence of 20 μg/ml human IgG to prevent biding through the Fc portion of the Abs. Infiltrating immune cells from mouse intestinal tissues were phenotypically characterized using anti-CD45, anti-CD19, anti-CD11b, anti-Gr1, anti-F4/80, anti-CD11c and anti-CD86 mAb (BioLegend), as described previously (37). Analysis was performed on a Gallios Flow Cytometer (Beckman Coulter).

PBMCs were isolated from healthy donors as described and depleted of CD14+ cells using specific MicroBeads (Miltenyi Biotec) and magnetic cell sorting. Allogenic cells were then labeled with 5 μM Cell Trace Violet (CTV; Invitrogen) for 20 min at 37°C and washed twice with RPMI 1640 medium containing 10% FCS. CTV-labeled cells were seeded onto 96-well plates containing GM-Mϕ (1 or 2 × 105 cells per well) or M-Mϕ (1 × 104 cells per well) in the presence or absence of rhGH (1 μg/ml, 24 h, 37°C). Cells were cocultured in RPMI 1640 medium with 10% FCS (10 d) and supplemented with rhGH (1 μg/ml) 24 h before cell proliferation evaluation. As negative controls, we used isolated CTV-labeled cells in the presence or absence of rhGH. CTV-labeled cells exposed to PMA (100 ng/ml, 24 h) were used as positive control. Cell viability and proliferation (dye dilution) were determined on CD4+ cells using a Gallios Flow Cytometer (Beckman Coulter). The percentage of dividing cells was calculated using FlowJo (Tree Star, Ashland, OR).

Listeria monocytogenes coupled to GFP (Listeria-GFP), kindly provided by Dr. E. Veiga (Centro Nacional de Biotecnología/Consejo Superior de Investigaciones Científicas, Madrid, Spain), was grown in brain-heart infusion medium (overnight, 37°C), diluted, recovered at midlogarithmic growth phase (OD600nm = 0.5), and washed in PBS before use.

Cells were infected with Listeria-GFP as described (38). Briefly, 30 min (37°C, 5% CO2) after inoculation, gentamicin (100 μg/ml) was added (60 min; 37°C) to cultures to eliminate extracellular bacteria. Cells were then extensively washed, fixed with paraformaldehyde (4% w/v, 20 min, room temperature) and seeded (30 min, 37°C) onto poly-L-lysine–coated plates (20 μg/ml, 10 min, room temperature; Sigma-Aldrich) prior to their incubation with wheat germ agglutinin (Thermo Fisher Scientific, Waltham, MA) and DAPI (Sigma-Aldrich) for 5 min at 4°C. Cell staining was evaluated by confocal microscopy (Zeiss Axiovert LSM 510-META) and images were analyzed and quantified using ImageJ software (National Institutes of Health).

Cell lysates were prepared in RIPA buffer (50 mM HEPES, 150 mM NaCl, 0.5% sodium deoxycholate, 0.1% SDS, 1% Triton X-100, 10 mM NaF, 5 mM EDTA, 0.1 mM NaPO4, 5% ethylene glycol) supplemented with 10 μg/ml of aprotinin and leupeptin, 10 μM sodium orthovanadate, and 1 mM PMSF. Lysates (50 μg) were subjected to electrophoresis and transferred to nitrocellulose membranes (GE Healthcare, Freiburg, Germany). After blocking with 5% BSA in TBS buffer (100 mM Tris 1 HCl [pH 8] and 1.5 M NaCl), membranes were incubated with anti-MAFB (BioLegend), anti-pGSK3α/β, -AKT, -pAKT, (Cell Signaling Technology, Danvers, MA), anti-ERK1/2 or anti-pERK1/2 (Santa Cruz Biotechnology, Santa Cruz, CA), or anti-vinculin (Sigma-Aldrich) Abs, followed by the corresponding HRP-conjugated secondary Ab.

For comparison of means, and unless otherwise indicated, statistical significance of the data were evaluated using paired Student t test and two-way ANOVA (Sidak multiple comparations test). The p value < 0.05 was considered significant (*p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001). Statistical parameters used in the GSEA analysis were as previously described (34).

Previous reports have demonstrated that GH modulates immune (16, 17) and inflammatory (1720) responses. Because GH has also been shown to modulate adipocyte differentiation in a macrophage-dependent manner (9), we sought to determine whether GH affects macrophage polarization during inflammatory responses. To this end, we initially made use of a murine model of DSS-induced acute colitis in which GH limits pathological conditions (18). We observed that both GHTg and wild-type (WT) mice survived 5 d of treatment with 3% DSS. We did not conduct survival analysis during the recovery phase as euthanasia was performed at the designed time points to cover the kinetics of immune cell infiltration, but we did not observe differences in mortality in any of the groups of mice maintained until the end of the experiment (9 d). Along the 5-d DSS treatment, both GHTg and WT mice were positive for occult blood (Hemoccult test) with no significant differences between the two groups of mice (Fig. 1A). Likewise, weight loss (Fig. 1B) and histological scores for severity of inflammation were similar in both groups (Fig. 1C, upper panel). Lastly, we also detected comparable levels of destruction of the crypt structure, a disturbed epithelial layer and massive infiltration of inflammatory cells in mucosal and submucosal layers in both mouse strains (Fig. 1C, middle [WT] and bottom [GHTg]; for higher resolution images of these micrographs, see Supplemental Fig. 1A [WT] and Supplemental Fig. 1B [GHTg]), confirming the lack of significant differences between the two groups of mice during disease onset.

Table I.
Human and mouse oligonucleotides used for RT-qPCR analysis
GeneForwardReverse
hCCR2 5′-CCCATCATCTATGCCTTCGT-3′ 5′-GGCAATCCTACAGCCAAGAG-3′ 
hEGLN3 5′-ATCGACAGGCTGGTCCTCTA-3′ 5′-GATAGCAAGCCACCATTGC-3′ 
hFOLR2 5′-GAGAGAGGCCAACTCAGACAC-3′ 5′-CCAGACCATGTCTTTCTGTCC-3′ 
hIGF1 5′-TGTGGAGACAGGGGCTTTTA-3′ 5′-ATCCACGATGCCTGTCTGA-3′ 
hINHBA 5′-CTCGGAGATCATCACGTTTG-3′ 5′-CCTTGGAAATCTCGAAGTGC-3′ 
hMMP12 5′-TGTCACTACCGTGGGAAATAAG-3′ 5′-AACACTGGTCTTTGGTCTCTCAG-3′ 
hCCL2 5′-AGTCTCTGCCGCCCTTCT-3′ 5′-GTGACTGGGGCATTGATTG-3′ 
hHMOX1 5′-GCCATGAACTTTGTCCGGTG-3′ 5′-TTTCGTTGGGGAAGATGCCA-3′ 
hSTAB1 5′-CGCCTGAAGCCCCACC-3′ 5′-TCTTGCCACGGTGAGAAGTC-3′ 
HIL-10 5′-TCACTCATGGCTTTGTAGATGC-3′ 5′-GTGGAGCAGGTGAAGAATGC-3′ 
hCLEC5A 5′-ACGGCTTCATTACCACAAGG-3′ 5′-CTTGCTTGATAAAATTCCCAGTC-3′ 
hTBP 5′-CGGCTGTTTAACTTCGCTTC-3′ 5′-CACACGCCAAGAAACAGTGA-3′ 
hGAPDH 5′-GAAGATGGTGATGGGATTTC-3′ 5′-GAAGGTGAAGGTCGGAGTC-3′ 
mArg1 5′-AGCCGCTGGAACCCAGAGAGA-3′ 5′-AACAAAGGCCAGGTCCCCGTG-3′ 
mNos2 5′-GGCAGCCTGTGAGACCTTTG-3′ 5′-GCATTGGAAGTGAAGCGTTTC-3′ 
mCd206 5′-CCACAGCATTGAGGAGTTTG-3′ 5′-ACAGCTCATCATTTGGCTCA-3′ 
mYm1 5′-TCTGGGTACAAGATCCCTGAA-3′ 5′-TCATATGGAGATTTATAGAGGGGACT-3′ 
mIl10 5′-CAGAGCCACATGCTCCTA-3′ 5′-GTCCAGCTGGTCCTTTGT-3′ 
mInhba 5′-ATCATCACCTTTGCCGAGTC-3′ 5′-TCACTGCCTTCCTTGGAAAT-3′ 
mTnf 5′-TCTTCTCATTCCTGCTTGTGG-3′ 5′-GGTCTGGGCCATAGAACTGA-3′ 
mActb 5′-TACCCAGGCATTGCTGACAG-3′ 5′-ACTTGCGGTGCACGATGGA-3′ 
GeneForwardReverse
hCCR2 5′-CCCATCATCTATGCCTTCGT-3′ 5′-GGCAATCCTACAGCCAAGAG-3′ 
hEGLN3 5′-ATCGACAGGCTGGTCCTCTA-3′ 5′-GATAGCAAGCCACCATTGC-3′ 
hFOLR2 5′-GAGAGAGGCCAACTCAGACAC-3′ 5′-CCAGACCATGTCTTTCTGTCC-3′ 
hIGF1 5′-TGTGGAGACAGGGGCTTTTA-3′ 5′-ATCCACGATGCCTGTCTGA-3′ 
hINHBA 5′-CTCGGAGATCATCACGTTTG-3′ 5′-CCTTGGAAATCTCGAAGTGC-3′ 
hMMP12 5′-TGTCACTACCGTGGGAAATAAG-3′ 5′-AACACTGGTCTTTGGTCTCTCAG-3′ 
hCCL2 5′-AGTCTCTGCCGCCCTTCT-3′ 5′-GTGACTGGGGCATTGATTG-3′ 
hHMOX1 5′-GCCATGAACTTTGTCCGGTG-3′ 5′-TTTCGTTGGGGAAGATGCCA-3′ 
hSTAB1 5′-CGCCTGAAGCCCCACC-3′ 5′-TCTTGCCACGGTGAGAAGTC-3′ 
HIL-10 5′-TCACTCATGGCTTTGTAGATGC-3′ 5′-GTGGAGCAGGTGAAGAATGC-3′ 
hCLEC5A 5′-ACGGCTTCATTACCACAAGG-3′ 5′-CTTGCTTGATAAAATTCCCAGTC-3′ 
hTBP 5′-CGGCTGTTTAACTTCGCTTC-3′ 5′-CACACGCCAAGAAACAGTGA-3′ 
hGAPDH 5′-GAAGATGGTGATGGGATTTC-3′ 5′-GAAGGTGAAGGTCGGAGTC-3′ 
mArg1 5′-AGCCGCTGGAACCCAGAGAGA-3′ 5′-AACAAAGGCCAGGTCCCCGTG-3′ 
mNos2 5′-GGCAGCCTGTGAGACCTTTG-3′ 5′-GCATTGGAAGTGAAGCGTTTC-3′ 
mCd206 5′-CCACAGCATTGAGGAGTTTG-3′ 5′-ACAGCTCATCATTTGGCTCA-3′ 
mYm1 5′-TCTGGGTACAAGATCCCTGAA-3′ 5′-TCATATGGAGATTTATAGAGGGGACT-3′ 
mIl10 5′-CAGAGCCACATGCTCCTA-3′ 5′-GTCCAGCTGGTCCTTTGT-3′ 
mInhba 5′-ATCATCACCTTTGCCGAGTC-3′ 5′-TCACTGCCTTCCTTGGAAAT-3′ 
mTnf 5′-TCTTCTCATTCCTGCTTGTGG-3′ 5′-GGTCTGGGCCATAGAACTGA-3′ 
mActb 5′-TACCCAGGCATTGCTGACAG-3′ 5′-ACTTGCGGTGCACGATGGA-3′ 
FIGURE 1.

Improved mucosal repair after DSS-induced colitis in GHTg mice correlates with altered expression of macrophage polarization–specific markers. (A) Percentage of GHTg and WT mice that were Hemoccult positive as determined by daily evaluation during DSS treatment and recovery. (B) Percentage of body weight change during DSS treatment and recovery in GHTg mice and WT littermates. Mean ± SD of 44 mice are shown. Two-way ANOVA. **p < 0.01. (C) Histologic inflammation score and the representative photomicrographs of H&E-stained sections from longitudinally opened and spirally arranged colon obtained from GHTg mice and WT littermates at day DSS + 1. Original magnification ×10; scale bar, 1000 μm. (D) Representative photomicrographs of H&E-stained colon sections from GHTg mice and WT littermates at day DSS + 7. Scale bar, 1000 μm (black arrows: area with crypt damage; white arrows: area showing infiltrating foci). Zoom images (original magnification ×40): regions of the colon with immune infiltration (insets) that were obtained from consecutive sections stained with anti-arginase and anti-iNOS mAbs. Nuclei were counterstained with DAPI. Scale bar, 50 μm. Black arrow depict area with crypt damage; white arrow depict area showing infiltrating foci. (E) Histologic scores for severity, extent of inflammation, crypt damage, and percentage of colon involvement. Mean ± SD of four mice at DSS + 7 are shown. Paired Student t test. ***p < 0.001, ****p < 0.0001.

FIGURE 1.

Improved mucosal repair after DSS-induced colitis in GHTg mice correlates with altered expression of macrophage polarization–specific markers. (A) Percentage of GHTg and WT mice that were Hemoccult positive as determined by daily evaluation during DSS treatment and recovery. (B) Percentage of body weight change during DSS treatment and recovery in GHTg mice and WT littermates. Mean ± SD of 44 mice are shown. Two-way ANOVA. **p < 0.01. (C) Histologic inflammation score and the representative photomicrographs of H&E-stained sections from longitudinally opened and spirally arranged colon obtained from GHTg mice and WT littermates at day DSS + 1. Original magnification ×10; scale bar, 1000 μm. (D) Representative photomicrographs of H&E-stained colon sections from GHTg mice and WT littermates at day DSS + 7. Scale bar, 1000 μm (black arrows: area with crypt damage; white arrows: area showing infiltrating foci). Zoom images (original magnification ×40): regions of the colon with immune infiltration (insets) that were obtained from consecutive sections stained with anti-arginase and anti-iNOS mAbs. Nuclei were counterstained with DAPI. Scale bar, 50 μm. Black arrow depict area with crypt damage; white arrow depict area showing infiltrating foci. (E) Histologic scores for severity, extent of inflammation, crypt damage, and percentage of colon involvement. Mean ± SD of four mice at DSS + 7 are shown. Paired Student t test. ***p < 0.001, ****p < 0.0001.

Close modal

However, although both groups of mice showed similar relative weight loss during disease onset, the recovery phase was significantly quicker in GHTg mice in terms of changes in body weight (Fig. 1B). Histological analysis of samples harvested 7 d after the DSS treatment (DSS + 7) (Fig. 1D, upper, Fig. 1E; Supplemental Fig. 1C [WT] and Supplemental Fig. 1D [GHTg] for higher resolution images), further supported the role of GH in enhancing tissue repair after DSS-induced colitis based on the extent and severity of inflammation, crypt damage, and percentage of affected colon. To assess the state of macrophage polarization at the recovery phase, we determined the expression of conventional proinflammatory (iNOS) and anti-inflammatory/reparative (arginase 1) macrophage markers in the recovering tissue. Immunohistological analysis of DSS + 7 mice revealed that the proportion of arginase-1+ cells infiltrating the colon was higher in GHTg mice than in WT littermates, with the latter group showing an increase in the number of iNOS+ cells compared with GHTg mice (Fig. 1D, bottom). Of note, histological scores were also significantly different between GHTg and WT littermates at this stage, with a lower grade of mucosal destruction and inflammation in the colon of GHTg mice (Fig. 1E). Indeed, flow cytometry analysis of the infiltrating macrophages of the intestinal tissues revealed a higher percentage of anti-inflammatory macrophages (CD45+F4/80+Gr1+CD11b+ CD86high) in GHTg mice (0.72 ± 0.13) than in controls (0.28 ± 0.09) (Table II). We also found a higher CD4+/CD8+ ratio in GHTg than in control animals (0.74 versus 0.38) in the recovery phase (DSS + 7), whereas the opposite occurred during inflammation (DSS + 1 d, GHTg 0.24 versus controls 0.68) (Table II). Notwithstanding the possibility of a GH-mediated effect on other immune cells, which might also contribute to improved recovery from DSS-induced colitis in GHTg mice, our results indicate a GH-triggered change in the polarization state of macrophages within the inflamed tissue.

Table II.
Immune cell infiltration (gated for CD45+) of affected intestinal tissue at DSS + 1 and DSS + 7 in WT and GHTg mice subjected to the DSS-induced model of colitis


DSS + 1
DSS + 7
WTGHTgWTGHTg
CD3+ CD4+ CD8 8.20 ± 2.46 4.52 ± 0.84 7.14 ± 0.92 9.32 ± 1.60 
CD4 CD8+ 12.04 ± 5.57 18.41 ± 4.63 18.49 ± 7.90 12.56 ± 3.07 
CD3+ CD69+ CD4+ CD8 2.01 ± 0.45 1.61 ± 0.33 2.40 ± 0.26 2.39 ± 0.61 
CD4 CD8+ 8.14 ± 3.20 16.38 ± 4.81 14.71 ± 6.41 8.41 ± 2.65 
F4/80+ Gr1+ CD11chighCD86low 3.86 ± 1.52 5.42 ± 2.27 0.44 ± 0.01 0.19 ± 0.09 
CD11clowCD86high 1.82 ± 0.36 2.05 ± 0.39 0.28 ± 0.09 0.72 ± 0.13 


DSS + 1
DSS + 7
WTGHTgWTGHTg
CD3+ CD4+ CD8 8.20 ± 2.46 4.52 ± 0.84 7.14 ± 0.92 9.32 ± 1.60 
CD4 CD8+ 12.04 ± 5.57 18.41 ± 4.63 18.49 ± 7.90 12.56 ± 3.07 
CD3+ CD69+ CD4+ CD8 2.01 ± 0.45 1.61 ± 0.33 2.40 ± 0.26 2.39 ± 0.61 
CD4 CD8+ 8.14 ± 3.20 16.38 ± 4.81 14.71 ± 6.41 8.41 ± 2.65 
F4/80+ Gr1+ CD11chighCD86low 3.86 ± 1.52 5.42 ± 2.27 0.44 ± 0.01 0.19 ± 0.09 
CD11clowCD86high 1.82 ± 0.36 2.05 ± 0.39 0.28 ± 0.09 0.72 ± 0.13 

The above results led us to hypothesize that GH might directly modulate macrophage polarization. To address this issue, we switched to the analysis of human monocyte–derived proinflammatory (GM-Mϕ) and anti-inflammatory (M-Mϕ) macrophages. Human GHR was found on monocytes, proinflammatory GM-Mϕ, and anti-inflammatory M-Mϕ, as demonstrated by flow cytometry using a GHR-specific mAb (36) (Supplemental Fig. 2A). Because the expression of the major GH effector molecule IGF-1 (39) is restricted to anti-inflammatory M-Mϕ (28), we evaluated the influence of GH on macrophage polarization by comparing the transcriptome of GM-Mϕ untreated or exposed to 1 μg/ml of rhGH for 24 h (Fig. 2A–C). GSEA of the RNAseq data revealed a significant positive enrichment of M-Mϕ-specific genes, but not GM-Mϕ-specific genes (28, 35), following GH exposure (Fig. 2D). We validated these findings by RT-qPCR analysis, which showed that GH treatment of GM-Mϕ significantly upregulated the expression of genes associated with the M-Mϕ anti-inflammatory profile (HMOX1, STAB1, IGF1, and FOLR2), whereas the GM-Mϕ–specific genes CCR2, MMP12, and EGLN3 were significantly downregulated (Fig. 3A). The effect of GH was specific for GM-Mϕ, as it failed to modify the transcriptome of M-Mϕ (Fig. 3B) or monocytes (Supplemental Fig. 2B). Overall, these data revealed that GH modulates the human macrophage gene signature, favoring the expression of genes specifically linked to anti-inflammatory M-Mϕ. To correlate these observations with data from GHTg mice, we derived murine GM-Mϕ and M-Mϕ from bone marrow progenitors using GM-CSF or M-CSF, respectively, followed by GH exposure. RT-qPCR analysis showed that GH treatment of murine GM-Mϕ significantly upregulated the expression of genes associated with the M-Mϕ anti-inflammatory profile (Arg1, Cd206, and Ym1), whereas it downregulated the expression of GM-Mϕ–associated inflammatory markers (Nos2, Inhba, and Tnf) (Fig. 3C). Thus, these data confirmed that GH also modulates the mouse macrophage gene signature, favoring the acquisition of anti-inflammatory markers.

FIGURE 2.

GH modulates the global transcriptomal signature of GM-Mϕ. (A) Relative mRNA expression of M-Mϕ–specific genes (HMOX1, IGF1, FOLR2) in untreated or rhGH-treated (1 ng/ml–2 μg/ml) GM-Mϕ. Results are shown as 2−∆Ct relative to the mean of internal GAPDH and TBP expression. Determinations were done in triplicates; box plots represent the mean and the minimum to maximum values (n = 3). Paired Student t test. *p < 0.05. (B) Experimental design of the gene-profiling experiment. (C) MA plot representing the comparative analysis of the transcriptome from untreated versus rhGH-treated GM-Mϕ. (D) GSEA on the “log fold change–ranked” list of genes obtained from untreated versus rhGH-treated GM-Mϕ according to limma analysis, using the previously defined M-Mϕ (left panel) and GM-Mϕ (right panel) gene sets (GSE68061). Vertical black lines indicate the position of each of the genes comprising the M-Mϕ and GM-Mϕ gene sets.

FIGURE 2.

GH modulates the global transcriptomal signature of GM-Mϕ. (A) Relative mRNA expression of M-Mϕ–specific genes (HMOX1, IGF1, FOLR2) in untreated or rhGH-treated (1 ng/ml–2 μg/ml) GM-Mϕ. Results are shown as 2−∆Ct relative to the mean of internal GAPDH and TBP expression. Determinations were done in triplicates; box plots represent the mean and the minimum to maximum values (n = 3). Paired Student t test. *p < 0.05. (B) Experimental design of the gene-profiling experiment. (C) MA plot representing the comparative analysis of the transcriptome from untreated versus rhGH-treated GM-Mϕ. (D) GSEA on the “log fold change–ranked” list of genes obtained from untreated versus rhGH-treated GM-Mϕ according to limma analysis, using the previously defined M-Mϕ (left panel) and GM-Mϕ (right panel) gene sets (GSE68061). Vertical black lines indicate the position of each of the genes comprising the M-Mϕ and GM-Mϕ gene sets.

Close modal
FIGURE 3.

GH drives an M-Mϕ–specific gene signature in human and murine GM-Mϕ, but does not alter the expression profile of M-Mϕ. (A) Relative mRNA expression of marker genes corresponding to GM-Mϕ (CCR2, MMP12, EGLN3) and M-Mϕ (HMOX1, STAB1, IGF1, FOLR2) polarization in untreated (white bars) or rhGH-treated (gray bars) GM-Mϕ. (B) Relative mRNA expression of marker genes as in (A) in untreated (white bars) or rhGH-treated (gray bars) M-Mϕ. In both cases, results are shown as 2−∆Ct relative to the mean of internal GAPDH and TBP expression. Determinations were done in triplicates; box and whisker plots represent the mean, lower and upper deciles, and the minimum to maximum values (n = 8). Paired Student t test. (C) Relative mRNA expression of mouse GM-Mϕ–specific (Nos2, Inhba, Tnf) and M-Mϕ–specific (Arg1, Cd206, Ym1) polarization genes in untreated or rhGH-treated GM-Mϕ and M-Mϕ. Results are shown as mean ± SD values of 2−ΔCt relative to internal Actb expression. Determinations were done in triplicates (n = 4). Paired Student t test. *p < 0.05, **p < 0.01, ***p < 0.001.

FIGURE 3.

GH drives an M-Mϕ–specific gene signature in human and murine GM-Mϕ, but does not alter the expression profile of M-Mϕ. (A) Relative mRNA expression of marker genes corresponding to GM-Mϕ (CCR2, MMP12, EGLN3) and M-Mϕ (HMOX1, STAB1, IGF1, FOLR2) polarization in untreated (white bars) or rhGH-treated (gray bars) GM-Mϕ. (B) Relative mRNA expression of marker genes as in (A) in untreated (white bars) or rhGH-treated (gray bars) M-Mϕ. In both cases, results are shown as 2−∆Ct relative to the mean of internal GAPDH and TBP expression. Determinations were done in triplicates; box and whisker plots represent the mean, lower and upper deciles, and the minimum to maximum values (n = 8). Paired Student t test. (C) Relative mRNA expression of mouse GM-Mϕ–specific (Nos2, Inhba, Tnf) and M-Mϕ–specific (Arg1, Cd206, Ym1) polarization genes in untreated or rhGH-treated GM-Mϕ and M-Mϕ. Results are shown as mean ± SD values of 2−ΔCt relative to internal Actb expression. Determinations were done in triplicates (n = 4). Paired Student t test. *p < 0.05, **p < 0.01, ***p < 0.001.

Close modal

As macrophage polarization is essentially defined by the prevailing cytokine secretion profile, we assessed the ability of GH to influence the cytokine signature of GM-Mϕ and M-Mϕ. To do this, we cultured both macrophage populations in the absence or presence of rhGH for 24 h and then measured the levels of TNF-α, IL-6, and IL-10 in the corresponding culture supernatants. Results showed that, compared with control cultures, GH exposure for 24 h triggered a significant decrease of TNF-α and a significant increase of IL-10 in GM-Mϕ supernatants, whereas the levels of IL-6 were unaffected (Fig. 4A). By contrast, and in accordance with the gene profile of M-Mϕ, GH treatment failed to modify the cytokine signature of anti-inflammatory macrophages (Fig. 4A). Because M-Mϕ secrete CCL2 (28), we also evaluated CCL2 levels in the culture supernatant of untreated or rhGH-treated GM-Mϕ or M-Mϕ and found that GH also induced a significant increase in the levels of CCL2 in GM-Mϕ supernatants (Fig. 4A). Altogether, these results indicate that GH treatment reprograms human macrophages, as it impairs the phenotypic and functional polarization of GM-Mϕ and causes a shift toward an anti-inflammatory profile.

FIGURE 4.

GH treatment modifies the secreted cytokine profile of GM-Mϕ and decreases the secretion of activin A. (A). TNF-α, IL-6, IL-10, and CCL2 abundance in culture supernatants of untreated (white bars) or rhGH-treated (gray bars) GM-Mϕ and M-Mϕ by ELISA. Shown are the mean ± SD of triplicate determinations (n = 8). Paired Student t test. (B) Abundance of activin A in culture supernatants of untreated (white bars) or rhGH-treated (gray bars) GM-Mϕ and M-Mϕ by ELISA. (C) INHBA expression determined by RT-qPCR in untreated (white bars) or rhGH-treated (gray bars) GM-Mϕ and M-Mϕ. Results are shown as 2−∆Ct relative to the mean of internal GAPDH and TBP expression. Results in (B) and (C) correspond to triplicate determinations (n = 8); box and whisker plots represent the mean, lower and upper deciles, and the minimum to maximum values. Paired Student t test. *p < 0.05, **p < 0.01.

FIGURE 4.

GH treatment modifies the secreted cytokine profile of GM-Mϕ and decreases the secretion of activin A. (A). TNF-α, IL-6, IL-10, and CCL2 abundance in culture supernatants of untreated (white bars) or rhGH-treated (gray bars) GM-Mϕ and M-Mϕ by ELISA. Shown are the mean ± SD of triplicate determinations (n = 8). Paired Student t test. (B) Abundance of activin A in culture supernatants of untreated (white bars) or rhGH-treated (gray bars) GM-Mϕ and M-Mϕ by ELISA. (C) INHBA expression determined by RT-qPCR in untreated (white bars) or rhGH-treated (gray bars) GM-Mϕ and M-Mϕ. Results are shown as 2−∆Ct relative to the mean of internal GAPDH and TBP expression. Results in (B) and (C) correspond to triplicate determinations (n = 8); box and whisker plots represent the mean, lower and upper deciles, and the minimum to maximum values. Paired Student t test. *p < 0.05, **p < 0.01.

Close modal

Activins are pluripotent growth and differentiation factors of the TGF-β superfamily (40). Like TGF-β, activins exert immunostimulatory and immunosuppressive functions (41). Activin A is considered a crucial modulator of inflammatory responses (42) and, in fact, activin A skews macrophage polarization by promoting the acquisition of a proinflammatory phenotype (28). We thus measured the levels of activin A in the culture supernatants of untreated or rhGH-treated GM-Mϕ and M-Mϕ and observed that GH treatment led to a significant decrease in activin A in GM-Mϕ cultures (Fig. 4B), whereas it did not modify activin A secretion from M-Mϕ (Fig. 4B). These data agree with the decrease in the mRNA levels of the activin A-encoding gene (INHBA) (43) detected in rhGH-treated GM-Mϕ (Fig. 4C). Thus, our results suggest an active role of GH in downregulating activin A expression and secretion by GM-Mϕ, which would ultimately contribute to shape the transcriptional and functional profile of these macrophages.

The physiological effects of GH are partially mediated by IGF-1 produced in the liver (44). IGF-1 is nonetheless produced by several cell types (45), including M-Mϕ (46). We thus evaluated whether GH-induced GM-Mϕ repolarization might be mediated by IGF-1. We found that treatment of fully differentiated proinflammatory GM-Mϕ with IGF-1 had no effect on their characteristic gene expression profile (Supplemental Fig. 3A, 3B), or on activin A secretion (Supplemental Fig. 3C), suggesting that the polarizing action of GH is independent of IGF-1 production.

Recent reports indicate that MAFB plays an essential role in human macrophage anti-inflammatory polarization (29, 30). MAFB is known to constrain the ability of M-CSF to instruct myeloid cell proliferation, promotes macrophage differentiation (47), and controls the acquisition of an anti-inflammatory transcriptional profile of M-Mϕ (29). We thus performed GSEA on the GH-treated GM-Mϕ RNAseq data and observed a significant positive enrichment of MAFB-regulated genes (Fig. 5A). Indeed, Western blot analysis showed that GH treatment increased the level of MAFB protein in GM-Mϕ, whereas no changes were observed in the already elevated expression of MAFB in M-Mϕ (29) (Fig. 5B, left). In line with these results, and with the previous demonstration that MAFB protein stability is controlled by GSK3β-mediated phosphorylation and subsequent proteasomal degradation (48), we found a significant increase in the inactivating serine 9 phosphorylation of GSK3β (49) in rhGH-treated GM-Mϕ but not in rhGH-treated M-Mϕ (Fig. 5B, right). The relevance of these results was supported by the increase in the expression of MAFB-dependent genes (IL-10, CCL2) and the decreased levels of the proinflammatory gene CLEC5A, whose expression is inhibited by MAFB (29), in GH-treated GM-Mϕ (Fig. 5C). These observations indicate that GH modifies the polarization state of GM-Mϕ likely through the inhibition of GSK3β, which would block MAFB degradation and result in the upregulation of MAFB-dependent genes and the subsequent acquisition of an anti-inflammatory profile.

FIGURE 5.

GH-mediated upregulation of MAFB in GM-Mϕ correlates with M-Mϕ marker expression. (A) GSEA analysis on the “log fold change–ranked” list of genes obtained from untreated versus rhGH-treated GM-Mϕ according to limma analysis, using the previously defined list of genes downregulated in MAFB-silenced cells (GSE84622). (B) Representative Western blot of MAFB (left panel) and pGSK3β (right panel) expression in lysates of untreated or rhGH-treated GM-Mϕ and M-Mϕ. Vinculin was used as a loading control. Densitometric analysis of MAFB and pGSK3β levels normalized to that of vinculin. Mean ± SD (n = 8). Paired Student t test. **p < 0.01, ****p < 0.0001. (C) CLEC5A, CCL2, and IL-10 mRNA expression levels determined by RT-qPCR in untreated or rhGH-treated GM-Mϕ and M-Mϕ. Results are shown as 2−∆Ct relative to the mean of internal GAPDH and TBP expression and correspond to triplicate determinations (n = 8); box and whisker plots represent the mean, lower and upper deciles, and the minimum to maximum values. Paired Student t test. *p < 0.05.

FIGURE 5.

GH-mediated upregulation of MAFB in GM-Mϕ correlates with M-Mϕ marker expression. (A) GSEA analysis on the “log fold change–ranked” list of genes obtained from untreated versus rhGH-treated GM-Mϕ according to limma analysis, using the previously defined list of genes downregulated in MAFB-silenced cells (GSE84622). (B) Representative Western blot of MAFB (left panel) and pGSK3β (right panel) expression in lysates of untreated or rhGH-treated GM-Mϕ and M-Mϕ. Vinculin was used as a loading control. Densitometric analysis of MAFB and pGSK3β levels normalized to that of vinculin. Mean ± SD (n = 8). Paired Student t test. **p < 0.01, ****p < 0.0001. (C) CLEC5A, CCL2, and IL-10 mRNA expression levels determined by RT-qPCR in untreated or rhGH-treated GM-Mϕ and M-Mϕ. Results are shown as 2−∆Ct relative to the mean of internal GAPDH and TBP expression and correspond to triplicate determinations (n = 8); box and whisker plots represent the mean, lower and upper deciles, and the minimum to maximum values. Paired Student t test. *p < 0.05.

Close modal

Kinetic analysis indicated that MAFB expression in GM-Mϕ was evident ∼1 h after exposure to GH and that GH-induced serine 9 phosphorylation of GSK3β remained elevated 4 and 24 h after treatment (48, 49) (Fig. 6A, Supplemental Fig. 4). To dissect the molecular mechanisms underlying these events, and based on previous data on the regulation of GSK3β activity (50, 51), we tested the initial signaling events triggered by GH in GM-Mϕ. We detected GH-mediated phosphorylation of ERK1/2 and AKT after 30 min and 1 h, respectively (Fig. 6A, Supplemental Fig. 4). Of note, blocking PI3K activity in GM-Mϕ with LY294002 (52) inhibited the GH-mediated phosphorylation of GSK3β and the increase in MAFB expression (Fig. 6A) and activin A secretion (Fig. 6B). These data are in accord with a previous report describing a role for AKT in GSK3β inactivation (49). Correspondingly, LY294002 pretreatment of GM-Mϕ abrogated the GH-mediated upregulation of CCL2, FOLR2, IGF1, and IL-10 and the downregulation of CLEC5A and IHNBA (Fig. 6C). Overall, these results demonstrate the involvement of a PI3K-GSK3β-MAFB axis in the GH-mediated polarization of GM-Mϕ.

FIGURE 6.

GH-mediated MAFB upregulation in GM-Mϕ is dependent on PI3K activity. (A) Western blot analysis of MAFB, pGSK3α/β, pAKT, and pERK1/2 expression in lysates of untreated or rhGH-treated GM-Mϕ pretreated or not with the PI3K inhibitor LY294002. Anti-AKT and anti-ERK1/2 mAbs were used as loading controls. One representative experiment from five is shown. (B). ELISA determination of activin A in culture supernatants of untreated (solid bars) or rhGH-treated (1 μg/ml, 24 h, striped bars) cells. GM-Mϕ were pretreated with LY294002 (gray background bars) or untreated (white background bars). Mean ± SD from three independent experiments are shown. Paired Student t test. *p < 0.05, ****p < 0.0001. (C) FOLR2, IL-10, CCL2, IGF1, CLEC5A, and INHBA mRNA expression levels were determined by RT-qPCR in untreated (white background bars) and pretreated with LY294002 (gray background bars) GM-Mϕ treated with GH (striped bars) or untreated (solid bars). Data show mean ± SD of triplicate determinations, relative to the mean of GAPDH and TBP expression (n = 3). Paired Student t test. *p < 0.05, **p < 0.01.

FIGURE 6.

GH-mediated MAFB upregulation in GM-Mϕ is dependent on PI3K activity. (A) Western blot analysis of MAFB, pGSK3α/β, pAKT, and pERK1/2 expression in lysates of untreated or rhGH-treated GM-Mϕ pretreated or not with the PI3K inhibitor LY294002. Anti-AKT and anti-ERK1/2 mAbs were used as loading controls. One representative experiment from five is shown. (B). ELISA determination of activin A in culture supernatants of untreated (solid bars) or rhGH-treated (1 μg/ml, 24 h, striped bars) cells. GM-Mϕ were pretreated with LY294002 (gray background bars) or untreated (white background bars). Mean ± SD from three independent experiments are shown. Paired Student t test. *p < 0.05, ****p < 0.0001. (C) FOLR2, IL-10, CCL2, IGF1, CLEC5A, and INHBA mRNA expression levels were determined by RT-qPCR in untreated (white background bars) and pretreated with LY294002 (gray background bars) GM-Mϕ treated with GH (striped bars) or untreated (solid bars). Data show mean ± SD of triplicate determinations, relative to the mean of GAPDH and TBP expression (n = 3). Paired Student t test. *p < 0.05, **p < 0.01.

Close modal

We next examined whether the phenotypic, transcriptional, and signaling effects of GH on GM-Mϕ influenced the effector functions normally mediated by this macrophage subtype. As innate immune cells, macrophages efficiently phagocytose pathogens and stress-inducing agents (53), with M-Mϕ displaying a higher phagocytic activity than GM- Mϕ (22). We thus tested whether GH treatment altered the phagocytic properties of GM-Mϕ by coculturing untreated and rhGH-treated GM-Mϕ or M-Mϕ with Listeria-GFP for 30 min, and determining bacterial uptake by confocal microscopy. Results showed that bacterial uptake was significantly higher in rhGH-treated GM-Mϕ than in untreated cells, whereas this effect was absent in M-Mϕ (Fig. 7A).

FIGURE 7.

GH treatment increases bacterial uptake by GM-Mϕ and diminishes the ability of macrophages to trigger allogenic T cell proliferation. (A) GM-Mϕ treated or not with rhGH and M-Mϕ were infected with Listeria-GFP, fixed, and added to poly-l-lysine–coated plates. Left panel shows representative confocal images of GFP fluorescence (green), membrane staining (wheat germ agglutinin, red) and nuclei (DAPI, cyan). Scale bar, 10 μm. Right panel shows GFP fluorescence quantitation using ImageJ. Equatorial-plane images from 200 to 250 cells were collected in random fields. Results are mean ± SD of background-subtracted GFP intensities (arbitrary units [a.u.]) (n = 3) Paired Student t test. (B) Representative flow cytometry panels to illustrate the procedure for CD4+ proliferation determination. In the coculture, viable CD4+ cells were identified using anti–CD4-PerCpCy5.5 mAb and then CTV dilution (gated) was evaluated. (C) Percentage of CD4+ cell proliferation, measured by CTV dilution in cells cocultured with GM-Mϕ or M-Mϕ in the absence (−) or presence (+) of rhGH. As controls proliferation of CD4+ cells cultured in the absence or presence of rhGH without macrophages and of CD4+ cells exposed to PMA for 24 h are shown. Results are expressed as a percentage of the maximum CD4+ cell proliferation (CD4+ cell proliferation in the presence of GM-Mϕ without GH stimulation, 100%). Mean ± SD (n = 5). Paired Student t test. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.

FIGURE 7.

GH treatment increases bacterial uptake by GM-Mϕ and diminishes the ability of macrophages to trigger allogenic T cell proliferation. (A) GM-Mϕ treated or not with rhGH and M-Mϕ were infected with Listeria-GFP, fixed, and added to poly-l-lysine–coated plates. Left panel shows representative confocal images of GFP fluorescence (green), membrane staining (wheat germ agglutinin, red) and nuclei (DAPI, cyan). Scale bar, 10 μm. Right panel shows GFP fluorescence quantitation using ImageJ. Equatorial-plane images from 200 to 250 cells were collected in random fields. Results are mean ± SD of background-subtracted GFP intensities (arbitrary units [a.u.]) (n = 3) Paired Student t test. (B) Representative flow cytometry panels to illustrate the procedure for CD4+ proliferation determination. In the coculture, viable CD4+ cells were identified using anti–CD4-PerCpCy5.5 mAb and then CTV dilution (gated) was evaluated. (C) Percentage of CD4+ cell proliferation, measured by CTV dilution in cells cocultured with GM-Mϕ or M-Mϕ in the absence (−) or presence (+) of rhGH. As controls proliferation of CD4+ cells cultured in the absence or presence of rhGH without macrophages and of CD4+ cells exposed to PMA for 24 h are shown. Results are expressed as a percentage of the maximum CD4+ cell proliferation (CD4+ cell proliferation in the presence of GM-Mϕ without GH stimulation, 100%). Mean ± SD (n = 5). Paired Student t test. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001.

Close modal

Macrophages are also Ag-presenting and T cell–activating cells (54), and GM-Mϕ display a considerably higher T cell–activating capacity than M-Mϕ (55). To determine whether GH modifies the T cell–activating ability of GM-Mϕ, we cocultured untreated and rhGH-treated GM-Mϕ with allogeneic T cells for 10 d and examined T cell proliferation by evaluation of dye dilution using flow cytometry (Fig. 7B). Results showed that rhGH-treated GM-Mϕ promoted a significantly lower proliferation of allogenic T cells than untreated GM-Mϕ (Fig. 7C). These results indicate that GH-mediated repolarization of GM-Mϕ not only affects phenotypic and transcriptional parameters, but also negatively affects effector functions that characterize proinflammatory GM-Mϕ (T cell activation) while favoring the acquisition of functions preferentially associated with anti-inflammatory M-Mϕ (bacterial capture and uptake).

To investigate the pathophysiological relevance of the GH-induced MAFB-mediated switch in macrophage polarization, we evaluated the polarization state of colon-infiltrating recovery-phase macrophages from mice subjected to DSS-induced colitis. In agreement with immunohistochemistry results, analysis of colon samples from DSS + 7 mice revealed a higher expression of Arg1 and significantly diminished levels of Nos2 in GHTg mice (Fig. 8A). We then isolated the infiltrated F4/80+ cells at DSS + 7 d (Fig. 8B) and observed a significant increase of Arg1, Cd206, and Ym1, three specific markers of anti-inflammatory macrophages (56, 57), in cells from GHTg mice compared with control littermates, and a reduced expression of Nos2, Tnf, and Inhba (Fig. 8C). These data corroborate the switch in macrophage polarization toward an anti-inflammatory phenotype observed in GHTg mice. Accordingly, although a direct effect of GH on other immune cells cannot be discarded, the presence of macrophages with a more reparative and less proinflammatory phenotype correlates with the improved recovery from DSS-induced colitis in GHTg mice. Overall, these results support the in vivo relevance of the macrophage-repolarizing ability of GH, and further illustrate the capacity of GH to drive macrophage polarization to a more anti-inflammatory/reparative phenotype.

FIGURE 8.

Colon-infiltrating recovery-phase macrophages from DSS-induced colitis exhibit an enhanced anti-inflammatory and MAFB-dependent signature in GHTg mice. (A) Expression levels of Arg1 and Nos2 were determined by RT-qPCR using full-length colon samples from GHTg mice and WT littermates at day DSS + 7 (n = 4). Mean ± SD values of 2−∆Ct, relative to Actb expression, from triplicate determinations are shown. Student t test. *p < 0.05, **p < 0.01, ****p < 0.0001. (B) After enzymatic digestion of the intestinal tissue of both GHTg and control littermates at DSS + 7 d (left panel, Before), F4/80+ cells were isolated using magnetic beads, and the sample purity analyzed by flow cytometry using an anti-F4/80 mAb (gated for CD45+) (right panel, After). (C) Expression levels of Arg1, Nos2, and MAFB-dependent genes were determined by RT-qPCR using isolated F4/80+ cells from GHTg mice and WT littermates’ colon at day DSS + 7 (n = 3). Mean ± SD values of 2−∆Ct, relative to Actb expression, from triplicate determinations are shown. Student t test. **p < 0.01.

FIGURE 8.

Colon-infiltrating recovery-phase macrophages from DSS-induced colitis exhibit an enhanced anti-inflammatory and MAFB-dependent signature in GHTg mice. (A) Expression levels of Arg1 and Nos2 were determined by RT-qPCR using full-length colon samples from GHTg mice and WT littermates at day DSS + 7 (n = 4). Mean ± SD values of 2−∆Ct, relative to Actb expression, from triplicate determinations are shown. Student t test. *p < 0.05, **p < 0.01, ****p < 0.0001. (B) After enzymatic digestion of the intestinal tissue of both GHTg and control littermates at DSS + 7 d (left panel, Before), F4/80+ cells were isolated using magnetic beads, and the sample purity analyzed by flow cytometry using an anti-F4/80 mAb (gated for CD45+) (right panel, After). (C) Expression levels of Arg1, Nos2, and MAFB-dependent genes were determined by RT-qPCR using isolated F4/80+ cells from GHTg mice and WT littermates’ colon at day DSS + 7 (n = 3). Mean ± SD values of 2−∆Ct, relative to Actb expression, from triplicate determinations are shown. Student t test. **p < 0.01.

Close modal

GH modulates immune function through direct effects on immune cells that express the GHR. For example, in macrophage-specific GHR-knockout mice, the loss of GH action exacerbates inflammation in adipose tissue (58). We previously showed that transgenic expression of GH ameliorates inflammation in diabetic NOD mice (16) and reduces the incidence of arthritis in a collagen-induced murine model (17). Similarly, GH attenuates the inflammatory burden in inflammatory bowel disease (59) and reduces NF-κB activation in colitis (18). A preliminary study in humans revealed that GH might be a beneficial treatment for Crohn’s disease (20). Although some of these observations have been associated with GH resistance processes, they might also be related to a potential role of GH modulating the immune system and favoring anti-inflammatory responses.

The phenotypic and functional heterogeneity of macrophages is essential to create the appropriate extracellular environment for immune responses to occur. Macrophages are highly plastic and can adopt proinflammatory or anti-inflammatory/resolving properties depending on microenvironmental signals, and the deregulation of this balance leads to chronic inflammatory diseases (60). Modulation of macrophage polarization is thus a potential target for therapeutic intervention in these chronic pathologies (61). The results of the current study reveal that GH does not promote monocyte polarization, but repolarizes the GM-Mϕ phenotype to an anti-inflammatory phenotype. Consistent with this, GSEA of RNAseq data demonstrated a significant enrichment of M-Mϕ–specific genes in rhGH-treated GM-Mϕ, some of them also confirmed by RT-qPCR analysis (HMOX1, IL-10, and IGF1), whereas the expression of others associated with GM-Mϕ were reduced (INHBA, MMP12, and EGLN3). The reprogramming effect of GH on human macrophages was also seen in murine GM-Mϕ, whose treatment with GH resulted in upregulation of M-Mϕ–associated anti-inflammatory genes (Arg1, Cd206, and Ym1) and reduction of GM-Mϕ-specific inflammatory genes (Nos2, Inhba, and Tnf). Functional in vitro analysis showed that GH reduces TNF-α release by GM-Mϕ and concomitantly increases their ability to secrete IL-10. Likewise, we also detected CCL2 secretion by rhGH-treated GM-Mϕ. By contrast, GH failed to alter TNF-α, IL-10, or CCL2 production in M-Mϕ. Further studies are needed to determine whether these findings are a consequence of a partial effect of GH on M-Mϕ, a dosage-mediated effect, or other unknown cofactors required to complete the repolarization process.

Initially characterized as inducers of follicle-stimulating hormone production, activins are now recognized to regulate the growth of numerous cell types, for example, by contributing to the maintenance of pluripotency in embryonic stem cells, and exerting antitumorigenic effects (41). Moreover, activin A is a crucial modulator of inflammatory responses (42), and its expression is upregulated in response to inflammatory mediators (62, 63). Activin A, which is encoded by the INHBA gene, induces arginase A expression in murine peritoneal macrophages and is important for the differential gene expression profiles and effector functions exhibited by proinflammatory macrophages (28). We observed that, in vitro, rhGH treatment of GM-Mϕ reduces the expression of INHBA and activin A, pointing to a possible role for GH in triggering GM-Mϕ repolarization.

We also found that GH triggers MAFB upregulation via a PI3K-dependent pathway. MAFB is a member of the MAF family of transcription factors and is involved in many cellular functions, such as control of lymphangiogenesis (64), or differentiation of pancreatic α and β cells (65, 66). Within the murine myeloid lineage, MAFB restricts the ability of M-CSF to trigger myeloid cell proliferation (67), and also critically determines the acquisition of the anti-inflammatory transcriptional and functional profiles in human macrophages (29, 30). Simultaneously, it negatively regulates genes associated with proinflammatory polarization (29, 30). Our GSEA of RNAseq data indicated a significant enrichment of MAFB-specific genes in rhGH-treated GM-Mϕ. In the current study, GH reduced activin A secretion and promoted MAFB upregulation, and thus supporting the role of GH in GM-Mϕ reprogramming. In accord with these results, activin A was shown to suppress MAFB expression in a pancreatic islet cell line (68). MAFB stabilization is controlled by GSK3β (48), an inhibitory serine/threonine kinase that is inactivated by phosphorylation at serine-9 (49). We found that these GH-mediated processes were blocked in cells pretreated with LY294002, a classical PI3K inhibitor, suggesting a role for p-AKT in this signaling event. Activated AKT has been associated with phosphorylation of GSK3β and inhibition of its activity (69), indicating that GSK3β is a substrate for AKT, as was described several years ago (70). Nonetheless, the effect triggered by GH was not completely abolished by LY294002 treatment. Although several mechanisms might be behind this effect, our results indicate that GH also triggered ERK1/2 activation, a pathway that has also been implicated in GSK3β inactivation (71, 72).

The polarization state of macrophages not only conditions the secreted cytokine profile, but also affects other cellular processes such as phagocytosis, production of reactive oxygen species (22), and capacity to trigger allogenic T cell proliferation. As expected, bacterial capture ability was higher in M-Mϕ than in GM-Mϕ (22), and we found that GH treatment increased the bacterial capture and microbial killing properties of GM-Mϕ. In addition, GH treatment significantly limited the ability of GM-Mϕ to promote allogeneic T cell proliferation. All of these data confirm the role of this hormone in repolarizing GM-Mϕ.

Available evidence supports the use of GH to treat inflammatory bowel diseases, including Crohn disease and ulcerative colitis (73). Although in most of the cases the observed effects have been associated with metabolic changes triggered by GH (3), a role for GH in enhancing survival, remission of inflammation, and mucosal repair during recovery has been linked to a local decrease of IL-1β production in a murine model of DSS-induced colitis in GHTg mice (74). This in vivo data suggest that GH reduces inflammation by altering the cytokine profile secreted by immune cells. We thus analyzed the in vivo relevance of GH-mediated GM-Mϕ reprogramming by comparing the phenotype of macrophages infiltrating the intestinal tissue of GHTg and WT littermates after DSS-induced colitis. As reported (74), GH did not influence the susceptibility of mice to acute DSS-induced colitis, but instead improved the remission of inflammation and mucosal repair during recovery. Importantly, GHTg mice presented a high proportion of infiltrating M-Mϕ (arginase 1+) when compared with WT littermates, which showed GM-Mϕ (iNOS+) infiltration. Flow cyometry analysis of these isolated macrophages confirmed the data. In GHTg mice, we found higher numbers of anti-inflammatory macrophages (CD45+F4/80+Gr1+CD11b+CD86high) than in controls. In addition, the infiltrating F4/80+ cells in GHTg mice showed higher expression of Arg1, Cd206, and Ym1, three genes whose expression marks murine anti-inflammatory macrophages (56, 57), whereas the expression of inflammatory genes (Nos2, Inhba, and Tnf) was reduced.

It has been previously described that IGF-1 secreted by satellite cells, myofibers, fibroblasts, endothelial cells, and inflammatory cells, plays an important role in muscle regeneration (75), with the authors of this study proposing an autocrine role for IGF-1 in modulating murine macrophage polarization to an anti-inflammatory phenotype. This observation correlates with in vitro analyses showing that both murine and human macrophages express significant quantities of IGF-1 propeptides, which are upregulated by stimulation with so-called M2-polarizing stimuli, including IL-4, IL-13, or M-CSF (76, 77). Our in vitro studies clearly showed an increase of IGF-1 production by GM-Mϕ treated with GH, but we excluded an effect of IGF-1 on GM-Mϕ reprogramming, per se. It is nonetheless known that, in vivo, IGF-1 influences the magnitude of tissue inflammation by redirecting epithelial cells to a phagocytic phenotype (78).

In conclusion, our findings provide strong support for a role of GH in reprograming inflammatory macrophages to an anti-inflammatory phenotype, which contributes to dampening the inflammatory microenvironment where the immune response takes place.

This work was supported in part by grants from the Spanish Ministry of Science, Innovation and Universities (SAF2017-82940-R Agencia Estatal de Investigación/Fondo Europeo de Desarrollo Regional (AEI/FEDER), Unión Europea [UE] [to M.M.], SAF2017-83785-R AEI/FEDER, UE [to Á.L.C.] and FJCI-2016-29990 AEI/FEDER, UE [to B.S.P.]), from the Redes Temáticas de Investigación Cooperativa en Salud Program of Instituto de Salud Carlos III (RD12/0012/0006 and RD12/0012/0007, Red de Investigación en Inflamación y Enfermedades Reumáticas), and the Regional Government of Madrid (B2017/BMD-3804 [to C.M.-A.]).

The sequence presented in this article has been submitted to the Sequence Read Archive (https://www.ncbi.nlm.nih.gov/sra) under accession number PRJNA555143.

The online version of this article contains supplemental material.

Abbreviations used in this article:

Ct

cycle threshold

CTV

Cell Trace Violet

DSS

dextran sodium sulfate

GH

growth hormone

GHR

GH receptor

GHTg

GH-overexpressing transgenic

GM-Mϕ

macrophage primed by GM-CSF

GSEA

gene-set enrichment analysis

IGF

insulin-like growth factor

iNOS

inducible NO synthase

Listeria-GFP

Listeria monocytogenes coupled to GFP

M-Mϕ

macrophage primed by M-CSF

rhGH

recombinant human GH

RNAseq

RNA sequencing

RT-qPCR

real-time quantitative PCR

WT

wild-type.

1
Velloso
,
C. P.
2008
.
Regulation of muscle mass by growth hormone and IGF-I.
Br. J. Pharmacol.
154
:
557
568
.
2
Ogle
,
G. D.
,
A. R.
Rosenberg
,
G.
Kainer
.
1992
.
Renal effects of growth hormone. II. Electrolyte homeostasis and body composition.
Pediatr. Nephrol.
6
:
483
489
.
3
Vijayakumar
,
A.
,
S.
Yakar
,
D.
Leroith
.
2011
.
The intricate role of growth hormone in metabolism.
Front. Endocrinol. (Lausanne)
2
:
32
.
4
Veldhuis
,
J. D.
,
J. N.
Roemmich
,
A. D.
Rogol
.
2000
.
Gender and sexual maturation-dependent contrasts in the neuroregulation of growth hormone secretion in prepubertal and late adolescent males and females--a general clinical research center-based study.
J. Clin. Endocrinol. Metab.
85
:
2385
2394
.
5
Takano
,
A.
,
T.
Haruta
,
M.
Iwata
,
I.
Usui
,
T.
Uno
,
J.
Kawahara
,
E.
Ueno
,
T.
Sasaoka
,
M.
Kobayashi
.
2001
.
Growth hormone induces cellular insulin resistance by uncoupling phosphatidylinositol 3-kinase and its downstream signals in 3T3-L1 adipocytes.
Diabetes
50
:
1891
1900
.
6
Lu
,
M.
,
J. U.
Flanagan
,
R. J.
Langley
,
M. P.
Hay
,
J. K.
Perry
.
2019
.
Targeting growth hormone function: strategies and therapeutic applications.
Signal Transduct. Target. Ther.
4
:
3
.
7
Hattori
,
N.
,
T.
Saito
,
T.
Yagyu
,
B. H.
Jiang
,
K.
Kitagawa
,
C.
Inagaki
.
2001
.
GH, GH receptor, GH secretagogue receptor, and ghrelin expression in human T cells, B cells, and neutrophils.
J. Clin. Endocrinol. Metab.
86
:
4284
4291
.
8
Welniak
,
L. A.
,
R.
Sun
,
W. J.
Murphy
.
2002
.
The role of growth hormone in T-cell development and reconstitution.
J. Leukoc. Biol.
71
:
381
387
.
9
Lu
,
C.
,
P. A.
Kumar
,
Y.
Fan
,
M. A.
Sperling
,
R. K.
Menon
.
2010
.
A novel effect of growth hormone on macrophage modulates macrophage-dependent adipocyte differentiation.
Endocrinology
151
:
2189
2199
.
10
Murphy
,
W. J.
,
S. K.
Durum
,
D. L.
Longo
.
1992
.
Human growth hormone promotes engraftment of murine or human T cells in severe combined immunodeficient mice.
Proc. Natl. Acad. Sci. USA
89
:
4481
4485
.
11
Kimata
,
H.
,
M.
Fujimoto
.
1994
.
Growth hormone and insulin-like growth factor I induce immunoglobulin (Ig)E and IgG4 production by human B cells.
J. Exp. Med.
180
:
727
732
.
12
Kimata
,
H.
,
A.
Yoshida
.
1994
.
Differential effect of growth hormone and insulin-like growth factor-I, insulin-like growth factor-II, and insulin on Ig production and growth in human plasma cells.
Blood
83
:
1569
1574
.
13
Bidlingmaier
,
M.
,
C. J.
Auernhammer
,
H.
Feldmeier
,
C. J.
Strasburger
.
1997
.
Effects of growth hormone and insulin-like growth factor I binding to natural killer cells.
Acta Paediatr. Suppl.
423
:
80
81
.
14
Smith
,
J. R.
,
H.
Benghuzzi
,
M.
Tucci
,
A.
Puckett
,
J. L.
Hughes
.
2000
.
The effects of growth hormone and insulin-like growth factor on the proliferation rate and morphology of RAW 264.7 macrophages.
Biomed. Sci. Instrum.
36
:
111
116
.
15
Warwick-Davies
,
J.
,
D. B.
Lowrie
,
P. J.
Cole
.
1995
.
Growth hormone is a human macrophage activating factor. Priming of human monocytes for enhanced release of H2O2.
J. Immunol.
154
:
1909
1918
.
16
Villares
,
R.
,
D.
Kakabadse
,
Y.
Juarranz
,
R. P.
Gomariz
,
C.
Martínez-A
,
M.
Mellado
.
2013
.
Growth hormone prevents the development of autoimmune diabetes.
Proc. Natl. Acad. Sci. USA
110
:
E4619
E4627
.
17
Villares
,
R.
,
G.
Criado
,
Y.
Juarranz
,
M.
Lopez-Santalla
,
E. M.
García-Cuesta
,
J. M.
Rodríguez-Frade
,
J.
Leceta
,
P.
Lucas
,
J. L.
Pablos
,
C.
Martínez-A
, et al
.
2018
.
Inhibitory role of growth hormone in the induction and progression phases of collagen-induced arthritis.
Front. Immunol.
9
:
1165
.
18
Han
,
X.
,
N.
Benight
,
B.
Osuntokun
,
K.
Loesch
,
S. J.
Frank
,
L. A.
Denson
.
2007
.
Tumour necrosis factor alpha blockade induces an anti-inflammatory growth hormone signalling pathway in experimental colitis.
Gut
56
:
73
81
.
19
Scolapio
,
J. S.
,
M.
Camilleri
,
C. R.
Fleming
,
L. V.
Oenning
,
D. D.
Burton
,
T. J.
Sebo
,
K. P.
Batts
,
D. G.
Kelly
.
1997
.
Effect of growth hormone, glutamine, and diet on adaptation in short-bowel syndrome: a randomized, controlled study.
Gastroenterology
113
:
1074
1081
.
20
Slonim
,
A. E.
,
L.
Bulone
,
M. B.
Damore
,
T.
Goldberg
,
M. A.
Wingertzahn
,
M. J.
McKinley
.
2000
.
A preliminary study of growth hormone therapy for Crohn’s disease.
N. Engl. J. Med.
342
:
1633
1637
.
21
Fleetwood
,
A. J.
,
T.
Lawrence
,
J. A.
Hamilton
,
A. D.
Cook
.
2007
.
Granulocyte-macrophage colony-stimulating factor (CSF) and macrophage CSF-dependent macrophage phenotypes display differences in cytokine profiles and transcription factor activities: implications for CSF blockade in inflammation.
J. Immunol.
178
:
5245
5252
.
22
Mendoza-Coronel
,
E.
,
E.
Ortega
.
2017
.
Macrophage polarization modulates FcγR- and CD13-mediated phagocytosis and reactive oxygen species production, independently of receptor membrane expression.
Front. Immunol.
8
:
303
.
23
Municio
,
C.
,
B.
Soler Palacios
,
L.
Estrada-Capetillo
,
A.
Benguria
,
A.
Dopazo
,
E.
García-Lorenzo
,
S.
Fernández-Arroyo
,
J.
Joven
,
M. E.
Miranda-Carús
,
I.
González-Álvaro
,
A.
Puig-Kröger
.
2016
.
Methotrexate selectively targets human proinflammatory macrophages through a thymidylate synthase/p53 axis.
Ann. Rheum. Dis.
75
:
2157
2165
.
24
Puig-Kröger
,
A.
,
E.
Sierra-Filardi
,
A.
Domínguez-Soto
,
R.
Samaniego
,
M. T.
Corcuera
,
F.
Gómez-Aguado
,
M.
Ratnam
,
P.
Sánchez-Mateos
,
A. L.
Corbí
.
2009
.
Folate receptor beta is expressed by tumor-associated macrophages and constitutes a marker for M2 anti-inflammatory/regulatory macrophages.
Cancer Res.
69
:
9395
9403
.
25
Verreck
,
F. A. W.
,
T.
de Boer
,
D. M. L.
Langenberg
,
M. A.
Hoeve
,
M.
Kramer
,
E.
Vaisberg
,
R.
Kastelein
,
A.
Kolk
,
R.
de Waal-Malefyt
,
T. H. M.
Ottenhoff
.
2004
.
Human IL-23-producing type 1 macrophages promote but IL-10-producing type 2 macrophages subvert immunity to (myco)bacteria.
Proc. Natl. Acad. Sci. USA
101
:
4560
4565
.
26
Soler Palacios
,
B.
,
L.
Estrada-Capetillo
,
E.
Izquierdo
,
G.
Criado
,
C.
Nieto
,
C.
Municio
,
I.
González-Alvaro
,
P.
Sánchez-Mateos
,
J. L.
Pablos
,
A. L.
Corbí
,
A.
Puig-Kröger
.
2015
.
Macrophages from the synovium of active rheumatoid arthritis exhibit an activin A-dependent pro-inflammatory profile.
J. Pathol.
235
:
515
526
.
27
Zheng
,
X.
,
K.
Turkowski
,
J.
Mora
,
B.
Brüne
,
W.
Seeger
,
A.
Weigert
,
R.
Savai
.
2017
.
Redirecting tumor-associated macrophages to become tumoricidal effectors as a novel strategy for cancer therapy.
Oncotarget
8
:
48436
48452
.
28
Sierra-Filardi
,
E.
,
A.
Puig-Kröger
,
F. J.
Blanco
,
C.
Nieto
,
R.
Bragado
,
M. I.
Palomero
,
C.
Bernabéu
,
M. A.
Vega
,
A. L.
Corbí
.
2011
.
Activin A skews macrophage polarization by promoting a proinflammatory phenotype and inhibiting the acquisition of anti-inflammatory macrophage markers.
Blood
117
:
5092
5101
.
29
Cuevas
,
V. D.
,
L.
Anta
,
R.
Samaniego
,
E.
Orta-Zavalza
,
J.
Vladimir de la Rosa
,
G.
Baujat
,
Á.
Domínguez-Soto
,
P.
Sánchez-Mateos
,
M. M.
Escribese
,
A.
Castrillo
, et al
.
2017
.
MAFB determines human macrophage anti-inflammatory polarization: relevance for the pathogenic mechanisms operating in multicentric carpotarsal osteolysis.
J. Immunol.
198
:
2070
2081
.
30
Kim
,
H.
2017
.
The transcription factor MafB promotes anti-inflammatory M2 polarization and cholesterol efflux in macrophages.
Sci. Rep.
7
:
7591
.
31
Cecim
,
M.
,
J.
Kerr
,
A.
Bartke
.
1995
.
Effects of bovine growth hormone (bGH) transgene expression or bGH treatment on reproductive functions in female mice.
Biol. Reprod.
52
:
1144
1148
.
32
Dieleman
,
L. A.
,
M. J.
Palmen
,
H.
Akol
,
E.
Bloemena
,
A. S.
Peña
,
S. G.
Meuwissen
,
E. P.
Van Rees
.
1998
.
Chronic experimental colitis induced by dextran sulphate sodium (DSS) is characterized by Th1 and Th2 cytokines.
Clin. Exp. Immunol.
114
:
385
391
.
33
Fleetwood
,
A. J.
,
H.
Dinh
,
A. D.
Cook
,
P. J.
Hertzog
,
J. A.
Hamilton
.
2009
.
GM-CSF- and M-CSF-dependent macrophage phenotypes display differential dependence on type I interferon signaling.
J. Leukoc. Biol.
86
:
411
421
.
34
Subramanian
,
A.
,
P.
Tamayo
,
V. K.
Mootha
,
S.
Mukherjee
,
B. L.
Ebert
,
M. A.
Gillette
,
A.
Paulovich
,
S. L.
Pomeroy
,
T. R.
Golub
,
E. S.
Lander
,
J. P.
Mesirov
.
2005
.
Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles.
Proc. Natl. Acad. Sci. USA
102
:
15545
15550
.
35
González-Domínguez
,
É.
,
Á.
Domínguez-Soto
,
C.
Nieto
,
J. L.
Flores-Sevilla
,
M.
Pacheco-Blanco
,
V.
Campos-Peña
,
M. A.
Meraz-Ríos
,
M. A.
Vega
,
Á. L.
Corbí
,
C.
Sánchez-Torres
.
2016
.
Atypical activin A and IL-10 production impairs human CD16+ monocyte differentiation into anti-inflammatory macrophages.
J. Immunol.
196
:
1327
1337
.
36
Mellado
,
M.
,
J. M.
Rodríguez-Frade
,
L.
Kremer
,
C.
von Kobbe
,
A. M.
de Ana
,
I.
Mérida
,
C.
Martinez-A
.
1997
.
Conformational changes required in the human growth hormone receptor for growth hormone signaling.
J. Biol. Chem.
272
:
9189
9196
.
37
Wensveen
,
F. M.
,
V.
Jelenčić
,
S.
Valentić
,
M.
Šestan
,
T. T.
Wensveen
,
S.
Theurich
,
A.
Glasner
,
D.
Mendrila
,
D.
Štimac
,
F. T.
Wunderlich
, et al
.
2015
.
NK cells link obesity-induced adipose stress to inflammation and insulin resistance.
Nat. Immunol.
16
:
376
385
.
38
Neu
,
C.
,
A.
Sedlag
,
C.
Bayer
,
S.
Förster
,
P.
Crauwels
,
J. H.
Niess
,
G.
van Zandbergen
,
G.
Frascaroli
,
C. U.
Riedel
.
2013
.
CD14-dependent monocyte isolation enhances phagocytosis of listeria monocytogenes by proinflammatory, GM-CSF-derived macrophages.
PLoS One
8
: e66898.
39
Chia
,
D. J.
2014
.
Minireview: mechanisms of growth hormone-mediated gene regulation.
Mol. Endocrinol.
28
:
1012
1025
.
40
Chen
,
Y. G.
,
Q.
Wang
,
S. L.
Lin
,
C. D.
Chang
,
J.
Chuang
,
S. Y.
Ying
.
2006
.
Activin signaling and its role in regulation of cell proliferation, apoptosis, and carcinogenesis. [Published erratum appears in 2006 Exp. Biol. Med. (Maywood) 231: 1448.]
Exp. Biol. Med. (Maywood)
231
:
534
544
.
41
Xia
,
Y.
,
A. L.
Schneyer
.
2009
.
The biology of activin: recent advances in structure, regulation and function.
J. Endocrinol.
202
:
1
12
.
42
Phillips
,
D. J.
,
D. M.
de Kretser
,
M. P.
Hedger
.
2009
.
Activin and related proteins in inflammation: not just interested bystanders.
Cytokine Growth Factor Rev.
20
:
153
164
.
43
Brown
,
C. W.
,
D. E.
Houston-Hawkins
,
T. K.
Woodruff
,
M. M.
Matzuk
.
2000
.
Insertion of Inhbb into the Inhba locus rescues the Inhba-null phenotype and reveals new activin functions.
Nat. Genet.
25
:
453
457
.
44
Ohlsson
,
C.
,
S.
Mohan
,
K.
Sjögren
,
A.
Tivesten
,
J.
Isgaard
,
O.
Isaksson
,
J. O.
Jansson
,
J.
Svensson
.
2009
.
The role of liver-derived insulin-like growth factor-I.
Endocr. Rev.
30
:
494
535
.
45
Perrini
,
S.
,
L.
Laviola
,
M. C.
Carreira
,
A.
Cignarelli
,
A.
Natalicchio
,
F.
Giorgino
.
2010
.
The GH/IGF1 axis and signaling pathways in the muscle and bone: mechanisms underlying age-related skeletal muscle wasting and osteoporosis.
J. Endocrinol.
205
:
201
210
.
46
Spadaro
,
O.
,
C. D.
Camell
,
L.
Bosurgi
,
K. Y.
Nguyen
,
Y. H.
Youm
,
C. V.
Rothlin
,
V. D.
Dixit
.
2017
.
IGF1 shapes macrophage activation in response to immunometabolic challenge.
Cell Rep.
19
:
225
234
.
47
Sarrazin
,
S.
,
N.
Mossadegh-Keller
,
T.
Fukao
,
A.
Aziz
,
F.
Mourcin
,
L.
Vanhille
,
L.
Kelly Modis
,
P.
Kastner
,
S.
Chan
,
E.
Duprez
, et al
.
2009
.
MafB restricts M-CSF-dependent myeloid commitment divisions of hematopoietic stem cells.
Cell
138
:
300
313
.
48
Herath
,
N. I.
,
N.
Rocques
,
A.
Garancher
,
A.
Eychène
,
C.
Pouponnot
.
2014
.
GSK3-mediated MAF phosphorylation in multiple myeloma as a potential therapeutic target.
Blood Cancer J.
4
: e175.
49
Jope
,
R. S.
,
G. V.
Johnson
.
2004
.
The glamour and gloom of glycogen synthase kinase-3.
Trends Biochem. Sci.
29
:
95
102
.
50
Beurel
,
E.
,
S. F.
Grieco
,
R. S.
Jope
.
2015
.
Glycogen synthase kinase-3 (GSK3): regulation, actions, and diseases.
Pharmacol. Ther.
148
:
114
131
.
51
Hermida
,
M. A.
,
J.
Dinesh Kumar
,
N. R.
Leslie
.
2017
.
GSK3 and its interactions with the PI3K/AKT/mTOR signalling network.
Adv. Biol. Regul.
65
:
5
15
.
52
Gharbi
,
S. I.
,
M. J.
Zvelebil
,
S. J.
Shuttleworth
,
T.
Hancox
,
N.
Saghir
,
J. F.
Timms
,
M. D.
Waterfield
.
2007
.
Exploring the specificity of the PI3K family inhibitor LY294002.
Biochem. J.
404
:
15
21
.
53
Baroja-Mazo
,
A.
,
F.
Martín-Sánchez
,
A. I.
Gomez
,
C. M.
Martínez
,
J.
Amores-Iniesta
,
V.
Compan
,
M.
Barberà-Cremades
,
J.
Yagüe
,
E.
Ruiz-Ortiz
,
J.
Antón
, et al
.
2014
.
The NLRP3 inflammasome is released as a particulate danger signal that amplifies the inflammatory response.
Nat. Immunol.
15
:
738
748
.
54
Murray
,
P. J.
2017
.
Macrophage polarization.
Annu. Rev. Physiol.
79
:
541
566
.
55
Morrissey
,
P. J.
,
L.
Bressler
,
L. S.
Park
,
A.
Alpert
,
S.
Gillis
.
1987
.
Granulocyte-macrophage colony-stimulating factor augments the primary antibody response by enhancing the function of antigen-presenting cells.
J. Immunol.
139
:
1113
1119
.
56
Murray
,
P. J.
,
J. E.
Allen
,
S. K.
Biswas
,
E. A.
Fisher
,
D. W.
Gilroy
,
S.
Goerdt
,
S.
Gordon
,
J. A.
Hamilton
,
L. B.
Ivashkiv
,
T.
Lawrence
, et al
.
2014
.
Macrophage activation and polarization: nomenclature and experimental guidelines.
Immunity
41
:
14
20
.
57
Raes
,
G.
,
R.
Van den Bergh
,
P.
De Baetselier
,
G. H.
Ghassabeh
,
C.
Scotton
,
M.
Locati
,
A.
Mantovani
,
S.
Sozzani
.
2005
.
Arginase-1 and Ym1 are markers for murine, but not human, alternatively activated myeloid cells.
J. Immunol.
174
: 6561; author reply 6561–6562.
58
Lu
,
C.
,
P. A.
Kumar
,
J.
Sun
,
A.
Aggarwal
,
Y.
Fan
,
M. A.
Sperling
,
C. N.
Lumeng
,
R. K.
Menon
.
2013
.
Targeted deletion of growth hormone (GH) receptor in macrophage reveals novel osteopontin-mediated effects of GH on glucose homeostasis and insulin sensitivity in diet-induced obesity.
J. Biol. Chem.
288
:
15725
15735
.
59
Andreassen
,
M.
,
J.
Frystyk
,
J.
Faber
,
L. O.
Kristensen
.
2012
.
GH activity and markers of inflammation: a crossover study in healthy volunteers treated with GH and a GH receptor antagonist.
Eur. J. Endocrinol.
166
:
811
819
.
60
Ruffell
,
D.
,
F.
Mourkioti
,
A.
Gambardella
,
P.
Kirstetter
,
R. G.
Lopez
,
N.
Rosenthal
,
C.
Nerlov
.
2009
.
A CREB-C/EBPbeta cascade induces M2 macrophage-specific gene expression and promotes muscle injury repair.
Proc. Natl. Acad. Sci. USA
106
:
17475
17480
.
61
Mantovani
,
A.
,
S. K.
Biswas
,
M. R.
Galdiero
,
A.
Sica
,
M.
Locati
.
2013
.
Macrophage plasticity and polarization in tissue repair and remodelling.
J. Pathol.
229
:
176
185
.
62
Jones
,
K. L.
,
A.
Mansell
,
S.
Patella
,
B. J.
Scott
,
M. P.
Hedger
,
D. M.
de Kretser
,
D. J.
Phillips
.
2007
.
Activin A is a critical component of the inflammatory response, and its binding protein, follistatin, reduces mortality in endotoxemia.
Proc. Natl. Acad. Sci. USA
104
:
16239
16244
.
63
Robson
,
N. C.
,
D. J.
Phillips
,
T.
McAlpine
,
A.
Shin
,
S.
Svobodova
,
T.
Toy
,
V.
Pillay
,
N.
Kirkpatrick
,
D.
Zanker
,
K.
Wilson
, et al
.
2008
.
Activin-A: a novel dendritic cell-derived cytokine that potently attenuates CD40 ligand-specific cytokine and chemokine production.
Blood
111
:
2733
2743
.
64
Dieterich
,
L. C.
,
S.
Klein
,
A.
Mathelier
,
A.
Sliwa-Primorac
,
Q.
Ma
,
Y. K.
Hong
,
J. W.
Shin
,
M.
Hamada
,
M.
Lizio
,
M.
Itoh
, et al
.
2015
.
DeepCAGE transcriptomics reveal an important role of the transcription factor MAFB in the lymphatic endothelium.
Cell Rep.
13
:
1493
1504
.
65
Conrad
,
E.
,
C.
Dai
,
J.
Spaeth
,
M.
Guo
,
H. A.
Cyphert
,
D.
Scoville
,
J.
Carroll
,
W. M.
Yu
,
L. V.
Goodrich
,
D. M.
Harlan
, et al
.
2016
.
The MAFB transcription factor impacts islet α-cell function in rodents and represents a unique signature of primate islet β-cells.
Am. J. Physiol. Endocrinol. Metab.
310
:
E91
E102
.
66
Hang
,
Y.
,
R.
Stein
.
2011
.
MafA and MafB activity in pancreatic β cells.
Trends Endocrinol. Metab.
22
:
364
373
.
67
Lavin
,
Y.
,
D.
Winter
,
R.
Blecher-Gonen
,
E.
David
,
H.
Keren-Shaul
,
M.
Merad
,
S.
Jung
,
I.
Amit
.
2014
.
Tissue-resident macrophage enhancer landscapes are shaped by the local microenvironment.
Cell
159
:
1312
1326
.
68
Andrzejewski
,
D.
,
M. L.
Brown
,
N.
Ungerleider
,
A.
Burnside
,
A. L.
Schneyer
.
2015
.
Activins A and B regulate fate-determining gene expression in islet cell lines and islet cells from male mice.
Endocrinology
156
:
2440
2450
.
69
Bijur
,
G. N.
,
R. S.
Jope
.
2003
.
Rapid accumulation of Akt in mitochondria following phosphatidylinositol 3-kinase activation.
J. Neurochem.
87
:
1427
1435
.
70
Cross
,
D. A.
,
D. R.
Alessi
,
P.
Cohen
,
M.
Andjelkovich
,
B. A.
Hemmings
.
1995
.
Inhibition of glycogen synthase kinase-3 by insulin mediated by protein kinase B.
Nature
378
:
785
789
.
71
Ding
,
Q.
,
W.
Xia
,
J. C.
Liu
,
J. Y.
Yang
,
D. F.
Lee
,
J.
Xia
,
G.
Bartholomeusz
,
Y.
Li
,
Y.
Pan
,
Z.
Li
, et al
.
2005
.
Erk associates with and primes GSK-3beta for its inactivation resulting in upregulation of beta-catenin.
Mol. Cell
19
:
159
170
.
72
Wen
,
A. Y.
,
K. M.
Sakamoto
,
L. S.
Miller
.
2010
.
The role of the transcription factor CREB in immune function.
J. Immunol.
185
:
6413
6419
.
73
Soendergaard
,
C.
,
P. H.
Kvist
,
P.
Thygesen
,
M.
Reslow
,
O. H.
Nielsen
,
J. J.
Kopchick
,
T. L.
Holm
.
2017
.
Characterization of growth hormone resistance in experimental and ulcerative colitis.
Int. J. Mol. Sci.
18
:
2046
.
74
Williams
,
K. L.
,
C. R.
Fuller
,
L. A.
Dieleman
,
C. M.
DaCosta
,
K. M.
Haldeman
,
R. B.
Sartor
,
P. K.
Lund
.
2001
.
Enhanced survival and mucosal repair after dextran sodium sulfate-induced colitis in transgenic mice that overexpress growth hormone.
Gastroenterology
120
:
925
937
.
75
Tonkin
,
J.
,
L.
Temmerman
,
R. D.
Sampson
,
E.
Gallego-Colon
,
L.
Barberi
,
D.
Bilbao
,
M. D.
Schneider
,
A.
Musarò
,
N.
Rosenthal
.
2015
.
Monocyte/macrophage-derived IGF-1 orchestrates murine skeletal muscle regeneration and modulates autocrine polarization.
Mol. Ther.
23
:
1189
1200
.
76
Arkins
,
S.
,
N.
Rebeiz
,
A.
Biragyn
,
D. L.
Reese
,
K. W.
Kelley
.
1993
.
Murine macrophages express abundant insulin-like growth factor-I class I Ea and Eb transcripts.
Endocrinology
133
:
2334
2343
.
77
Martinez
,
F. O.
,
S.
Gordon
,
M.
Locati
,
A.
Mantovani
.
2006
.
Transcriptional profiling of the human monocyte-to-macrophage differentiation and polarization: new molecules and patterns of gene expression.
J. Immunol.
177
:
7303
7311
.
78
Han
,
C. Z.
,
I. J.
Juncadella
,
J. M.
Kinchen
,
M. W.
Buckley
,
A. L.
Klibanov
,
K.
Dryden
,
S.
Onengut-Gumuscu
,
U.
Erdbrügger
,
S. D.
Turner
,
Y. M.
Shim
, et al
.
2016
.
Macrophages redirect phagocytosis by non-professional phagocytes and influence inflammation.
Nature
539
:
570
574
.

The authors have no financial conflicts of interest.

Supplementary data