In vivo experiments
All zebrafish (Danio rerio) used for this project were located in the aquaria at the Bateson Centre, at the University of Sheffield (UK). Zebrafish were present in tanks at a density of no more than four zebrafish per liter, with 14 hours light and 10 hours dark cycle, at a temperature of 28°C. All experimental procedures were carried out in accordance with the UK Home Office Project License PPL70/8178 and personal license IO6008638. All transgenic mice (Mus Musculus) used for this project were housed under pathogen-free conditions at the Experimental Therapy Unit at the Faculty of Medicine of the University of Nantes, France (Agreement D44015 and DUO 6781). All protocols applied in the present study were first validated by the French ethical committee of the “Pays de la Loire” (CEEA-PdL-06) and authorized by the French ministry of agriculture and fisheries (authorization # 18415-201901101823350 v2).
Generation of IL34 mutant zebrafish
The zebrafish Il34 gene (ENSDARG00000091003.2 or ZDB-GENE-050419-150) contains seven exons as human and mouse genes (Figure 1). IL34 mutant zebrafish was generated using the CRISPR-Cas9 technology as previously described 53,54. Exon 3 was targeted using the sequence shown in Supplementary Figure 1A and the corresponding 20 bp spacer region was placed into a guide RNA template for in vitro transcription. The gRNA was then transcribed using the MEGAshortscript T7 kit (Life Technologies, UK) and microinjected with Cas9 protein (NEB, UK) into the yolk of zebrafish embryos the one cell stage. F0 adult fish were crossed with wild-type fish to identify founder with germline transmission. Primers used for genotyping were (Fw 5’-TCA GCC AAT AAA TAT CAG ATC CA-3’ and Rv 5’-CGT CTC CTG GTT GCA TTT-3’) which amplify a 300bp fragment of the wild type sequence of Zebrafish IL34 exon3 covering the chosen CRISPR target sequence. Obtained fragments of shorter sizes were sequenced to identify mutations induced in the different founders. Two mutations corresponding to a 23bp deletion (mutant #1 in Figure 1) and a 50bp deletion combined to a 6bp insertion (mutant #2 in Figure 1) were obtained. Phenotypes of Zebrafishes homozygous for each of these mutations (Il34-/- from F3 or following generations) were compared to ensure for link to Il34 deficiency and not from potential background mutations. Genotyping was performed on DNA extracted from the caudal fins by PCR using same primers as those used to identify founders. Fragments of 300bp, 277bp and 256bp were amplified respectively for Il34 exon3 WT, mutant #1 and mutant #2 sequences.
Van Kossa and Acian Blue staining of zebrafish skeleton
For Von Kossa staining, samples were fixed in 4% PFA for 2 hours at room temperature, rinsed in water containing 0.01% tween 20, and left to incubate in a solution of silver nitrate under a 60 W light bulb for 1 hour. After rinsing with water containing 0.01% tween 20, samples were fixed in 2.5% sodium-thiosulfate for 10 minutes, rinsed and again fixed in 4% PFA for 30 minutes at room temperature. Preservation was done in glycerol and samples were kept at room temperature in dark until images were taken.
For Alcian Blue Staining, samples were fixed overnight in 4% PFA at 4ºC. After several washes in a phosphate buffer solution containing 0.1% tween 20 (PBS-T) and dehydration using methanol, samples were transferred into Alcian blue staining solution (0.1% Alcian Blue, 70% ethanol, 1% concentrated hydrochloric acid) and left to stain overnight at room temperature. Samples where then rinsed in PBS-T and bleached in 30% hydrogen peroxide for 10 minutes at 37ºC. A 30% saturated borate solution was then used to eliminate all residues of bleaching solution before putting the samples into a trypsin digestion solution for 30 minutes at 37ºC until brains and eyes appeared translucent. A rehydration was performed and samples were put in glycerol for preservation until images were taken.
Zebrafish were imaged for both stains using the SMZ1500 stereomicroscope, with a DS-Fi1 camera (both Nikon, Japan), at 20 X magnification and Nikon Elements software.
Generation of Il34 mutant mouse
The Il34 mutant mouse was generated at the Mouse Clinical Institute (IGBMC, Illkirch, France; Project IR00004258 / K4258) by classical embryonic stem cells (ES) injection in blastocyst stage embryo. Three JM8.N4 ES cell clones carrying the targeted Il34tm1a(EUCOMM)Wtsi allele were purchased at the European Conditional Mouse Mutagenesis Consortium (EUCOMM) and the clone EPD0146_4_F02 (embryonic stem line JM8.N4; C57BL/6) that was confirmed by PCR and Sanger sequencing (Supplementary Figure 2) as being correctly targeted was used to generate the Il34 conditional mutant mouse line. Breeding with ERT2-Cre mice (B6.Cg‐Tg(UBC‐cre/ERT2)1Ejb/J, JR#8085, Jackson Laboratory, Bar Harbor, Maine, USA) enabled to (Tamoxifen dependently) delete exons 3–5 of Il34 and the neomycin-resistance cassette generating the Il34+/LacZ mice (Supplementary Figure 2). Breeding with CAG-FLPe mice (C57BL/6-Tg(CAG-flpe)16Ito, RBRC10707, RIKEN BRC, Tsukuba, Ibaraki 305-0074, Japan) allowed to delete the whole LacZ–NeoR cassette and generate mice carrying a loxP-flanked Il34 allele (Il34+/f). Homozygous Il34LacZ/LacZ mice (called Il34-/- in the manuscript) were used for analysis. Mice were genotyped by PCR (Supplementary Figure 2) with the primers Il34-S2: 5’-GTC AGT ATC GGC GGA ATT-3’, Il34-S3: 5’-GTT TGG CCG ATG CTG GCA AAG G-3’ and Il34-AS2: 5’-CTG TCT TAT GAA GAT GGC ATG CC-3’. Il34-S2 and Il34-AS2 primers enable to amplify a 440bp fragment in presence of Il34LacZ allele, and Il34-S3 and Il34-AS2 primers fragments of 240bp and 290bp respectively in presence of wild type (WT) and Il34f alleles (Supplementary Figure 2).
Alizarin Red and Alcian Blue double staining of mouse skeleton
The whole-mount skeletal staining protocol used is derived from the protocol of Rigueur and Lyons 55. Briefly, after euthanasia, all skin, internal organs, adipose tissue and as much as possible muscle were removed before fixation in a PBS 1X pH 7.4 solution containing 2% of paraformaldehyde and 0.2% glutaraldehyde. Skeletons were then dehydrated in ethanol and placed in acetone for permeabilization. Cartilage staining was then realized by submerging the skeletons in the Alcian blue stain (Alcian blue 8GX 0.03 % (w/v), 80 % EtOH, 20 % glacial acetic acid). After washes in 70 % and 95 % ethanol, a pre-clear of the tissue was realized in a 1 % KOH solution. Bone staining was then carried out in Alizarin red stain (Alizarin red 0.005 % (w/v) in 1 % (w/v) KOH). The Alizarin red solution was then replaced with a v/v mix of glycerol and 1 % KOH to remove the excess red color. Skeleton were transferred to 100 % glycerol for long-term storage and imaging.
New-born mice treatment with blocking antibodies
The protocol used to treat newborn mice with blocking antibodies was previously described 38. Briefly, newborn C57BL/6 mice from naïve and transgenic IL34+/LacZ mothers received four subcutaneous injections (25 mg/kg of body weight) of respectively Sheff-5 (rat anti-mouse IL34 blocking IgG1 antibody, Diaclone, Besançon, France) and IK22-5 rat anti- mouse RANKL blocking IgG2a antibody 56 or isotopic corresponding control every 2 days beginning at day 1 after birth (Supplementary Figure 4b). The mice were finally sacrificed at postnatal day 15 for phenotyping.
Micro-CT analysis
A Skyscan 1076 micro-CT scanner (Skyscan, Kontich, Belgium) was used to analyze and compare between the different groups of mice the bone morphometric, structural and mineral parameters at different anatomical sites namely the tibia, the mandible, the vertebra and the cranium. All samples were scanned using the same parameters (pixel size 9 μm, 50 kV, 0.5 mm Aluminum filter, 20 minutes of scanning). The scanner reconstruction was carried out using the NRecon software and the analyses were performed using CTAn, CTVox, and DataViewer software (Skyscan). In order to obtain the different measurements, the IMAGE-J software (National Institutes of Health, Bethesda, MD, USA) was used. In this way, the acquisition of the image in CTVox was systematically calibrated with a phantom of 5 mm (known size) and all measurements were finally sized using the analysis scale in the IMAGE-J software.
Bone morphometric parameters including tibia total length and width were sized using specific reference marks (Figure 1c and Supplementary Figure 4a), and for the cranium measurements were made using the method previously described 57. Briefly, seven measurements regarding the sagittal, vertical and transversal planes of craniofacial growth were made (Figure 1c and Supplementary Figure 4a).
Bone mineral and structural parameters including the bone mineral density (BMD), the percentage of bone volume (BV/TV), the trabecula thickness (Tb.Th), the trabecula separation (Tb.Sp) and the trabecula number (Tb.N) were analyzed for each bone at different anatomical sites using a volume of interest (VOI) measuring 2.0 mm x 1.1mm × 1.1 mm. The VOI was sectioned using the Data Viewer software, and analyzed using the CTAn software. The different points chosen for the analysis are presented in Supplementary Figure 4. To facilitate the identification of changes in the different structures, a “color density range” was used in the CTAn software that made it possible to adjust the correspondence of color and brightness values using image gray scales. For tibia and head images, a brightness level of -32 and a contrast level of 6 from the color density range of the CTAn software were systematically used.
Histology, histo-enzymology and immunohistochemistry
Histology, histo-enzymology and immunohistochemistry were performed on 3 µm thickness paraffin embedded sections of the different samples prepared as previously described 58. Masson's trichrome and Safranin-O stains were performed following classical protocols and tartrate-resistant acid phosphatase (TRAP) histo-enzymology was carried out as previously described 59. Immunohistochemistry was performed by using the protocol as previously described 60 and the following antibodies: rabbit monoclonal anti-RUNX2 (Abcam, ref#ab192256, 1/1000), rabbit polyclonal anti- osterix (OSX) (Abcam ref#ab22552, 1/1000), anti-CD207 (eBioscience, ref# 14-2073-80, 1/100).
LacZ staining
Sections (12μm) of IL34+/LacZ mice epidermis embedded in OCT were cut using Cryostat Leica CM3050S. Slices were fixed with PFA 1% 5min, rinsed with PBS 1x and incubated in Xgal (5-bromo-4-chloro-3-indolyl-beta-D-galactopyranoside) solution overnight at 37°C. Sections were rinsed with PBS 1X, left to dry and mounted with EUKITT® medium.
In vitro experiments
Reagents
Recombinant human Macrophage-Colony Stimulating Factor (M-CSF), human interleukin-34 (IL-34), human M-CSF receptor (M-CSFR/CD115), human TGF-β1, human bone morphogenetic protein 2 (BMP-2), human bone morphogenetic protein 4 (BMP-4), human bone morphogenetic protein 7 (BMP-7), human Noggin, Activin RIIA receptor (ActRIIA), human Activin RIIB receptor (ActRIIB), human TRANCE (RANKL) and antibody anti-human M-CSFR, Anti-Phosho-M-CSFR (Y723) were obtained from R&D Systems (Abingdon, UK). Anti-human IL34 (BT-34) mouse IgG1 monoclonal antibody was produced by Diaclone (Besançon, France) under patent (Heymann D, Ségaliny A, Brion R. University of Nantes /Nantes Hospital/INSERM, “Anti-IL-34 antibodies”. WO/2016/097420 A1, 2016). Antibodies directed against human Smad1 (D59D7), human Smad2 (D43B4), anti Phospho-Smad1/5 (ser463/465) (41D10), anti-phospho-Smad2 (Ser465/467) (138D), β-Actin (8H10D10) and HRP-conjugated secondary antibodies were purchased from Cell Signalling (Ozyme, Saint Quentin Yvelines, France). AlphaLISA® SureFire® Ultra Total SMAD1 and p-SMAD1 (Ser463/465) Assay kits were purchase from PerkinElmer (Villebon-sur-Yvette, France).
Cell cultures
The cell lines used in the present study were purchased from the American Tissue Cell Collection (ATCC, Molsheim, France). HEK293 (HEK) transfected with the pCDNA3 empty plasmid or the pCDNA3 plasmid containing the M-CSFR gene as described by Segaliny et al., 6. Human Mesenchymal Stem Cells-Bone Marrow (HMSC-BM) (CLS catalog number 300665) were cultured in Dulbecco's Modified Eagle's Medium (DMEM, Lonza, Levallois-Perret, France) supplemented with 10% fetal bovine serum (FBS; Hyclone Perbio, Bezons, France) and 2 mmol/L of L-glutamine.
Human osteoclast differentiation
CD14+ monocytes were isolated from peripheral blood of 3 healthy donors CD14+ cells were initially isolated from human peripheral blood donors provided by the French blood bank institute (Etablissement Français du Sang, Nantes, France, authorization number: NTS 2000-24), by using MACS microbeads (MiltenyiBiotec, Bergisch Gladbach, Germany) as previously described 61. For osteoclast differentiation, CD14+ cells were cultured in alpha-MEM (Lonza) supplemented with 10% human serum (Invitrogen, France) and in the presence of human M-CSF (25 ng/mL) or human IL34 (100 ng/mL) +/- human BMP2 (40 or 100 ng/mL) for 3 days. Then cells were treated with same molecules in the presence of human RANKL (100 ng/mL) for 11 days. Medium was renewed every 3 days. After 11 days of treatment, osteoclasts were analyzed by Acid Phosphatase (TRAP) staining kits (Sigma Aldrich, Saint-Quentin Fallavier, France). TRAP+ multinucleated cells with 3 nuclei and more were considered as osteoclasts and were manually enumerated.
Human osteoblastic differentiation
Human Mesenchymal Stem Cells-Bone Marrow (HMSC-BM) (CLS catalog number 300665) were purchased from CLS (Germany). Osteoblast differentiation assays were performed as previously described 60,62. Briefly, HMSC-BM were cultured in DMEM was supplemented 10% of FBS, vitamin D3 (10−8 M; Sigma) and dexamethasone (10−7 M; Sigma). After 3 days, ascorbic acid (50 ng/mL; Sigma) and b-glycerophosphate (10 mM; Sigma) were added to allow mineralization detected by alizarin red-S staining for three weeks. Images were captured using a stereomicroscope (Nikon), and mineralized surfaces were quantified using Image J software. Mineralization process was carried out in the presence or absence of human cytokine IL-34 (25 ng/mL), BMP2 (10 ng/mL) or combination of both molecules for 3 weeks. RNA samples were collected at days 3, 4, 14 and 21 after the induction of differentiation.
Flow cytometry
FACS analysis of CD11b monocytic bone marrow and spleen cells were performed as previously described 63. Briefly, after red blood cell lysis (Sigma-Aldrich), bone marrow and spleen cells were labelled with anti- CD11b (clone M1/70; BD Bioscience, Le Pont de Claix, France). Data were acquired using a FACS Canto-II (BD Biosciences).
Western blot
The cells were collected in a RIPA buffer (10 mM Tris pH 8, 1 mM EDTA, 150 mM NaCl, 1% NP40, 0.1% SDS containing a cocktail of protease and phosphatase inhibitors Halt™ (Thermo Fisher, Waltham, MA, USA). The protein concentration was determined using a BCA (bicinchoninic acid) method by BC Assay Protein Quantitation Kit (Interchim, Montluçon, France). 50 μg of protein extracts were prepared in a Laëmmli buffer (62.5 mM Tris–HCl, pH 6.8, 2% SDS, 10% glycerol, 5% 2-mercaptoethanol, 0.001% bromophenol blue) and then separated by SDS-polyacrylamide gel electrophoresis. After electrophoretic transfer, the immobilon-P membranes (Millipore, Molsheim, France) were blotted with the antibodies referenced in the “Reagents” section. The membranes were then probed with secondary antibodies coupled with horseradish peroxidase. Antibody binding was visualized with an enhanced chemiluminescence (ECL) kit Clarity™ Western ECL Substrate (Bio-Rad, Marnes-la-Coquette, France). The luminescence was detected with a ChemiDoc MP Imaging System (Bio-Rad). Blots images and semi-quantitative analysis were done using ImageJ software (USA).
SMAD1/5 signaling measured by Alpha SureFire® Technology
Direct quantification analysis of cell signaling was performed by using Alpha SureFire® Technology from PerkinElmer in a Victor® Nivo™ multimode microplate reader (ALSU-PSM1; PerkinElmer, Villebon-sur-Yvette, France).
RNA isolation and real-time PCR
Total RNA was extracted using NucleoSpin® RNA Plus (Macherey-Nagel, Duren, Germany). 1μg of total RNA was used for first strand cDNA synthesis using the OneScript® RT Mix (Ozyme). Real-time PCR was performed on 20 ng of reverse transcribed total RNA (cDNA), 300 nM of primers (QuantiTect Primer® Assays, Qiagen) and PowerUp™ SYBR™ Master Mix from Applied Biosystems™ (Thermo Fisher) in a CFX96 Touch Deep Well Real-Time PCR Detection system from Bio-Rad. Thermal cycle conditions were perform by following manufacture protocol. The analysis was performed with CFX Manager Software (Bio-Rad) using human glyceraldehyde 3-phosphate dehydrogenase (GAPDH), Hypoxanthine Phosphoribosyl transferase 1 (HPRT1) and TATA box binding protein (TBP) as invariant controls (QuantiTect Primer® Assays, Qiagen). Oligonucleotides were designed with Primer-Blast software (NCBI) and purchased from Eurogentec (Eurogentec, Angers, France). The 2−ΔΔCt (cycle threshold) method was used to calculate expression levels. List of primers and gene name symbols with corresponding full names are indicated in Tables S1 and S2 below.
Surface plasmon resonance (SPR) assays
All SPR experiments were performed on a T200 apparatus (Cytiva) at 25 °C in PBS pH7.4 containing 0.05% of surfactant P20. Human recombinant BMP2, BMP4 and BMP7 proteins were immobilized (1500- 2300 RU) at pH 4.5 on CM5-S sensor chip by amine coupling following the manufacturer’s instructions (Cytiva, Velizy-Villacoublay, France). IL34 kinetics were measured using one cycle titration, for these five increasing concentrations of recombinant human IL-34 (12.5, 25, 50, 100, 200 nM) were injected during 60s at 100 µL/min on coated BMPs. The last injection was followed by a 600s dissociation time in running buffer. The KD values were evaluated using a bivalent fitting model (T200 Evaluation software 3.2.1, Cytiva). All sensorgrams were corrected by subtracting the low signal from the control reference surface (without any immobilized protein) and blank buffer injections before fitting. For KD evaluation of IL34 on human recombinant receptors BMPRIIA, Act RIIA and Act RIIB, these receptors were captured on immobilized anti-human Fc (Cytiva), four increasing concentrations of IL34 (18.75, 37.5, 75, 150, 300 nM) were injected. The KD values were evaluated by using a steady-state fitting model. The binding responses of IL-34 (50 nM) alone, Nogging (50 nM) alone and a mixing of IL-34 and Noggin were measured by 180s injection on different coated BMP proteins (BMP-2, BMP-4, BMP-7) at a flow rate of 30 µL/min followed by a dissociation time of 400s in running buffer.
Protein-Protein docking and analysis
Structures of M-CSFR, BMPR1 and IL-34 were extracted from their bound crystallographic forms (1REW for BMPR1A + BMP2 64, 4WRL for M-CSF:M-CSFR1 65 and 4DKD for IL-34:M-CSFR1 66). Docking experiments were performed using either BMP-2 fixed and the partner protein mobile, or the reverse, as previously published 64. ClusPro analysis 67 was performed in balanced mode, only the first 10 binding modes clusters were considered for analysis, the best modes were selected by visual inspection. Interface analysis was performed using the PISA web server 68. Visualization and superimposition of docking poses and crystallographic structures were done using PyMOL (The PyMOL Molecular Graphics System, Version 2.5 Schrödinger, LLC; Schrödinger, LLC 2015).
Statistical analysis
All experiments were repeated at least three times in independent experiments. The differences between the experimental conditions were assessed with Student’s t test or a one-way ANOVA followed by the Mann–Whitney test or Kruskal-Wallis test (in the case of more than two independent samples of equal or different sample size). The results are given as a mean ± SD. Results were considered significant at p-values of ≤0.05, p-values of ≤0.01 and p-values of ≤0.001. GraphPad Prism 6 software (GraphPad Software, San Diego, CA, USA) and Real Statistics Resource Pack Software (Release 8.91), copyright (2013-2023) Charles Zaiontz (www.real-statistics.com) were used for statistical analyses.