Craniometaphyseal Dysplasia Mutations in ANKH Negatively Affect Human Induced Pluripotent Stem Cell Differentiation into Osteoclasts

I-Ping Chen, Raj Luxmi, Jitendra Kanaujiya, Zhifang Hao, Ernst J Reichenberger, I-Ping Chen, Raj Luxmi, Jitendra Kanaujiya, Zhifang Hao, Ernst J Reichenberger

Abstract

We identified osteoclast defects in craniometaphyseal dysplasia (CMD) using an easy-to-use protocol for differentiating osteoclasts from human induced pluripotent stem cells (hiPSCs). CMD is a rare genetic bone disorder, characterized by life-long progressive thickening of craniofacial bones and abnormal shape of long bones. hiPSCs from CMD patients with an in-frame deletion of Phe377 or Ser375 in ANKH are more refractory to in vitro osteoclast differentiation than control hiPSCs. To exclude differentiation effects due to genetic variability, we generated isogenic hiPSCs, which have identical genetic background except for the ANKH mutation. Isogenic hiPSCs with ANKH mutations formed fewer osteoclasts, resorbed less bone, expressed lower levels of osteoclast marker genes, and showed decreased protein levels of ANKH and vacuolar proton pump v-ATP6v0d2. This proof-of-concept study demonstrates that efficient and reproducible differentiation of isogenic hiPSCs into osteoclasts is possible and a promising tool for investigating mechanisms of CMD or other osteoclast-related disorders.

Keywords: craniometaphyseal dysplasia; hiPSC differentiated osteoclasts; osteoclastogenesis; rare genetic bone disorder.

Copyright © 2017 The Authors. Published by Elsevier Inc. All rights reserved.

Figures

Figure 1
Figure 1
Differentiating Healthy Control hiPSCs into Osteoclasts (A) Schematic protocol of differentiating hiPSCs into osteoclasts. (B) Embryoid body (EB) formation and mesoderm gene expression. EBs cultured for 4 days (left panel). Scale bar, 200 μm. Expression of mesoderm marker genes in EBs cultured for 1, 2, 3, and 4 days by qPCR. ∗p < 0.05 by one-way ANOVA. Data presented are means ± SD. (C) Myelomonocytic cell expansion. Single cells released from EBs into suspension (top panel). Scale bar, 100 μm. Percentage of cells positive for hematopoietic cell surface markers CD14, CD43, and CD45 in cells released from 10, 13, 17, and 21 day adherent EBs by flow cytometry. Data presented are means ± SD. (D) TRAP+ osteoclasts differentiated from hiPSCs (left panel), resorption pits on bone chips (middle panel), and expression of OC marker genes, TRAP and CTSK by RT-PCR (right panel). HPRT served as internal control. Ctl1, control1; Ctl2, control2; 1w, 2w, 1 or 2 weeks in stage 3. Scale bar, 100 μm (left panel) and 200 μm (middle panel). Three independent experiments (three technical replicates per experiment) for each hiPSC line. Data were pooled from four wild-type hiPSC lines (hiPSCs from two healthy subjects, two hiPSC clones of each individual donor).
Figure 2
Figure 2
Osteoclasts Differentiated from Healthy Control and CMD hiPSCs (A) Comparison of control and CMD osteoclasts derived from hiPSCs. Numbers of TRAP+ multinucleated cells (nuclei ≥3) are shown (mean ± SD) in the left corners of the images. Different letters (a, b) indicate statistically significant difference (p TRAP, CATHEPSIN K, CALCITONIN RECEPTOR mRNA message in osteoclasts differentiated from CMD hiPSCs shown by qPCR. ∗p < 0.05 by two-way ANOVA. Data presented are means ± SD. Three independent experiments (technical replicates) were performed for each hiPSC line.
Figure 3
Figure 3
Osteoclasts Differentiated from Isogenic hiPSC with or without CMD ANKH Mutations (A) Electropherograms showing partial sequences of ANKH exon 9 with insertion of Phe377del into wild-type hiPSCs and correction of Ser375del ANKH in CMD hiPSCs. Blue arrows indicate silent mutations introduced to minimize recutting of a repaired site. (B) Decreased numbers of TRAP+ multinucleated osteoclasts formed from isogenic CMD mutant hiPSCs compared with wild-type controls. Numbers of TRAP+ multinucleated cells (nuclei ≥3) are shown (mean ± SD) in the left corners of the images. ∗ indicates statistical significance compared with the parental isogenic hiPSCs by Student’s t test. Scale bar, 100 μm. (C) Decreased resorbed area on bone chips by osteoclasts derived from isogenic CMD mutant hiPSCs compared with wild-type controls. ∗ on bone images (left panel) indicate resorption pits. Bar figures show the percentage of resorbed area (mean ± SD). Scale bar, 200 μm. ∗ indicates statistical significance compared with the parental isogenic hiPSCs by Student’s t test. (D) Decreased nuclei numbers in CMD hiPSC-derived osteoclasts (n = 70–156 cells per group). Yellow bar indicates the mean value of each group. Different letters (a, b, c) indicate statistically significant difference (p TRAP, CATHEPSIN K, CALCITONIN RECEPTOR, NFATc1 in osteoclasts differentiated from isogenic hiPSCs with a S375del mutation in ANKH by qPCR. ∗p < 0.05 by Student's t test. Data presented are means ± SD. (F) A representative Atp6v0d2 (40 kDa) immunoblot of hiPSCs (left panel) and hiPSC-differentiated osteoclasts (right panel) with or without S375del or F377del ANKH mutations. GAPDH served as loading control. Samples with wild-type and a S375del mutation in ANKH are isogenic hiPSCs. The F377del cell lysate is from patient CMD1 hiPSCs and hiPSC-derived osteoclasts. Numbers below indicate ratio of Atp6v0d2 to GAPDH. (G) A representative ANKH (52 kDa) immunoblot of undifferentiated hiPSCs (left panel) and hiPSC-derived osteoclasts (right panel) with or without S375del or F377del ANKH mutations. GAPDH served as loading control. Samples with wild-type and a S375del mutation are isogenic hiPSCs. The F377del cell lysate is from patient CMD1 hiPSCs and hiPSC-derived osteoclasts. Numbers below indicate the ratio of ANKH to GAPDH. Three independent experiments (technical replicates) were performed for each isogenic hiPSC line.

References

    1. Bhatia M., Bonnet D., Kapp U., Wang J.C., Murdoch B., Dick J.E. Quantitative analysis reveals expansion of human hematopoietic repopulating cells after short-term ex vivo culture. J. Exp. Med. 1997;186:619–624.
    1. Chen I.P., Fukuda K., Fusaki N., Iida A., Hasegawa M., Lichtler A., Reichenberger E.J. Induced pluripotent stem cell reprogramming by integration-free Sendai virus vectors from peripheral blood of patients with craniometaphyseal dysplasia. Cell Reprogram. 2013;15:503–513.
    1. Chen I.P., Wang C.J., Strecker S., Koczon-Jaremko B., Boskey A., Reichenberger E.J. Introduction of a Phe377del mutation in ANK creates a mouse model for craniometaphyseal dysplasia. J. Bone Miner Res. 2009;24:1206–1215.
    1. Chen I.P., Wang L., Jiang X., Aguila H.L., Reichenberger E.J. A Phe377del mutation in ANK leads to impaired osteoblastogenesis and osteoclastogenesis in a mouse model for craniometaphyseal dysplasia (CMD) Hum. Mol. Genet. 2011;20:948–961.
    1. Choi K.D., Vodyanik M.A., Slukvin I.I. Generation of mature human myelomonocytic cells through expansion and differentiation of pluripotent stem cell-derived lin-CD34+CD43+CD45+ progenitors. J. Clin. Invest. 2009;119:2818–2829.
    1. Cong L., Ran F.A., Cox D., Lin S., Barretto R., Habib N., Hsu P.D., Wu X., Jiang W., Marraffini L.A. Multiplex genome engineering using CRISPR/Cas systems. Science. 2013;339:819–823.
    1. Costa-Rodrigues J., Fernandes A., Fernandes M.H. Spontaneous and induced osteoclastogenic behaviour of human peripheral blood mononuclear cells and their CD14(+) and CD14(-) cell fractions. Cell Prolif. 2011;44:410–419.
    1. Dutra E.H., Chen I.P., Reichenberger E.J. Dental abnormalities in a mouse model for craniometaphyseal dysplasia. J. Dent. Res. 2013;92:173–179.
    1. Grigoriadis A.E., Kennedy M., Bozec A., Brunton F., Stenbeck G., Park I.H., Wagner E.F., Keller G.M. Directed differentiation of hematopoietic precursors and functional osteoclasts from human ES and iPS cells. Blood. 2010;115:2769–2776.
    1. Ho A.M., Johnson M.D., Kingsley D.M. Role of the mouse ank gene in control of tissue calcification and arthritis. Science. 2000;289:265–270.
    1. Hoffman R., Tong J., Brandt J., Traycoff C., Bruno E., McGuire B.W., Gordon M.S., McNiece I., Srour E.F. The in vitro and in vivo effects of stem cell factor on human hematopoiesis. Stem Cells. 1993;11(Suppl 2):76–82.
    1. Jacquin C., Gran D.E., Lee S.K., Lorenzo J.A., Aguila H.L. Identification of multiple osteoclast precursor populations in murine bone marrow. J. Bone Miner Res. 2006;21:67–77.
    1. Jeon O.H., Panicker L.M., Lu Q., Chae J.J., Feldman R.A., Elisseeff J.H. Human iPSC-derived osteoblasts and osteoclasts together promote bone regeneration in 3D biomaterials. Sci. Rep. 2016;6:26761.
    1. Kang H., Shih Y.R., Nakasaki M., Kabra H., Varghese S. Small molecule-driven direct conversion of human pluripotent stem cells into functional osteoblasts. Sci. Adv. 2016;2:e1600691.
    1. Kanke K., Masaki H., Saito T., Komiyama Y., Hojo H., Nakauchi H., Lichtler A.C., Takato T., Chung U.I., Ohba S. Stepwise differentiation of pluripotent stem cells into osteoblasts using four small molecules under serum-free and feeder-free conditions. Stem Cell Rep. 2014;2:751–760.
    1. Kuhn L.T., Liu Y., Boyd N.L., Dennis J.E., Jiang X., Xin X., Charles L.F., Wang L., Aguila H.L., Rowe D.W. Developmental-like bone regeneration by human embryonic stem cell-derived mesenchymal cells. Tissue Eng. Part A. 2014;20:365–377.
    1. Lee S.H., Rho J., Jeong D., Sul J.Y., Kim T., Kim N., Kang J.S., Miyamoto T., Suda T., Lee S.K. v-ATPase V0 subunit d2-deficient mice exhibit impaired osteoclast fusion and increased bone formation. Nat. Med. 2006;12:1403–1409.
    1. Moon S.H., Ju J., Park S.J., Bae D., Chung H.M., Lee S.H. Optimizing human embryonic stem cells differentiation efficiency by screening size-tunable homogenous embryoid bodies. Biomaterials. 2014;35:5987–5997.
    1. Nakano T., Kodama H., Honjo T. Generation of lymphohematopoietic cells from embryonic stem cells in culture. Science. 1994;265:1098–1101.
    1. Ng E.S., Davis R.P., Azzola L., Stanley E.G., Elefanty A.G. Forced aggregation of defined numbers of human embryonic stem cells into embryoid bodies fosters robust, reproducible hematopoietic differentiation. Blood. 2005;106:1601–1603.
    1. Nurnberg P., Thiele H., Chandler D., Hohne W., Cunningham M.L., Ritter H., Leschik G., Uhlmann K., Mischung C., Harrop K. Heterozygous mutations in ANKH, the human ortholog of the mouse progressive ankylosis gene, result in craniometaphyseal dysplasia. Nat. Genet. 2001;28:37–41.
    1. Ochiai-Shino H., Kato H., Sawada T., Onodera S., Saito A., Takato T., Shibahara T., Muramatsu T., Azuma T. A novel strategy for enrichment and isolation of osteoprogenitor cells from induced pluripotent stem cells based on surface marker combination. PLoS One. 2014;9:e99534.
    1. Panicker L.M., Miller D., Park T.S., Patel B., Azevedo J.L., Awad O., Masood M.A., Veenstra T.D., Goldin E., Stubblefield B.K. Induced pluripotent stem cell model recapitulates pathologic hallmarks of Gaucher disease. Proc. Natl. Acad. Sci. USA. 2012;109:18054–18059.
    1. Pick M., Azzola L., Mossman A., Stanley E.G., Elefanty A.G. Differentiation of human embryonic stem cells in serum-free medium reveals distinct roles for bone morphogenetic protein 4, vascular endothelial growth factor, stem cell factor, and fibroblast growth factor 2 in hematopoiesis. Stem Cells. 2007;25:2206–2214.
    1. Ran F.A., Hsu P.D., Wright J., Agarwala V., Scott D.A., Zhang F. Genome engineering using the CRISPR-Cas9 system. Nat. Protoc. 2013;8:2281–2308.
    1. Reichenberger E., Tiziani V., Watanabe S., Park L., Ueki Y., Santanna C., Baur S.T., Shiang R., Grange D.K., Beighton P. Autosomal dominant craniometaphyseal dysplasia is caused by mutations in the transmembrane protein ANK. Am. J. Hum. Genet. 2001;68:1321–1326.
    1. Taylor R.M., Kashima T.G., Hemingway F.K., Dongre A., Knowles H.J., Athanasou N.A. CD14- mononuclear stromal cells support (CD14+) monocyte-osteoclast differentiation in aneurysmal bone cyst. Lab Invest. 2012;92:600–605.
    1. Watanabe K., Ueno M., Kamiya D., Nishiyama A., Matsumura M., Wataya T., Takahashi J.B., Nishikawa S., Nishikawa S., Muguruma K. A ROCK inhibitor permits survival of dissociated human embryonic stem cells. Nat. Biotechnol. 2007;25:681–686.
    1. Wolins N.E., Quaynor B.K., Skinner J.R., Tzekov A., Park C., Choi K., Bickel P.E. OP9 mouse stromal cells rapidly differentiate into adipocytes: characterization of a useful new model of adipogenesis. J. Lipid Res. 2006;47:450–460.
    1. Yamamoto T., Kurihara N., Yamaoka K., Ozono K., Okada M., Yamamoto K., Matsumoto S., Michigami T., Ono J., Okada S. Bone marrow-derived osteoclast-like cells from a patient with craniometaphyseal dysplasia lack expression of osteoclast-reactive vacuolar proton pump. J. Clin. Invest. 1993;91:362–367.
    1. Yoshimi K., Kunihiro Y., Kaneko T., Nagahora H., Voigt B., Mashimo T. ssODN-mediated knock-in with CRISPR-Cas for large genomic regions in zygotes. Nat. Commun. 2016;7:10431.
    1. Zandstra P.W., Conneally E., Piret J.M., Eaves C.J. Ontogeny-associated changes in the cytokine responses of primitive human haemopoietic cells. Br. J. Haematol. 1998;101:770–778.

Source: PubMed

3
Sottoscrivi