FoxO proteins restrain osteoclastogenesis and bone resorption by attenuating H2O2 accumulation

Shoshana M Bartell, Ha-Neui Kim, Elena Ambrogini, Li Han, Srividhya Iyer, S Serra Ucer, Peter Rabinovitch, Robert L Jilka, Robert S Weinstein, Haibo Zhao, Charles A O'Brien, Stavros C Manolagas, Maria Almeida, Shoshana M Bartell, Ha-Neui Kim, Elena Ambrogini, Li Han, Srividhya Iyer, S Serra Ucer, Peter Rabinovitch, Robert L Jilka, Robert S Weinstein, Haibo Zhao, Charles A O'Brien, Stavros C Manolagas, Maria Almeida

Abstract

Besides their cell-damaging effects in the setting of oxidative stress, reactive oxygen species (ROS) play an important role in physiological intracellular signalling by triggering proliferation and survival. FoxO transcription factors counteract ROS generation by upregulating antioxidant enzymes. Here we show that intracellular H2O2 accumulation is a critical and purposeful adaptation for the differentiation and survival of osteoclasts, the bone cells responsible for the resorption of mineralized bone matrix. Using mice with conditional loss or gain of FoxO transcription factor function, or mitochondria-targeted catalase in osteoclasts, we demonstrate this is achieved, at least in part, by downregulating the H2O2-inactivating enzyme catalase. Catalase downregulation results from the repression of the transcriptional activity of FoxO1, 3 and 4 by RANKL, the indispensable signal for the generation of osteoclasts, via an Akt-mediated mechanism. Notably, mitochondria-targeted catalase prevented the loss of bone caused by loss of oestrogens, suggesting that decreasing H2O2 production in mitochondria may represent a rational pharmacotherapeutic approach to diseases with increased bone resorption.

Figures

Figure 1. RANKL decreases FoxO levels and…
Figure 1. RANKL decreases FoxO levels and activity.
(a) FoxO mRNA (triplicates) and (b) protein levels in BMMs, pre-osteoclasts (pOC) and mature (m) osteoclast cultures. (c) Luciferase activity in RAW264.7 cells transfected with a FoxO-luc reporter construct and incubated with the indicated doses of RANKL in the presence or absence of fetal bovine serum (FBS) for 16 h (triplicates). RLU, relative luminescence units. *P<0.05 versus respective untreated cells, #P<0.05 versus untreated cells in 10% serum. (d) Luciferase activity as in c in cells co-transfected with a control plasmid (pcDNA) or with plasmids expressing FoxO1, 3 or 4 and treated with 60 ng ml−1 RANKL. *P<0.05 versus respective vehicle (veh)-treated cells, #P<0.05 versus veh-treated cells with pcDNA. (e) Protein levels of FoxOs and the nuclear protein lamin A in nuclear extract of BMMs incubated with or without RANKL in the absence or presence of 10% FBS. (f) Catalase mRNA levels in cells described in a. *P<0.05. (g) RANKL-induced Akt, Erk, FoxO1 and FoxO3 phosphorylation in BMMs. (h) Luciferase activity as in d in cells treated with veh, 50 nM PD98059 and 10 μM LY294002. *P<0.05 versus respective veh-treated cells, #P<0.05 versus similar treated cells with pcDNA. (i) Akt, Erk and FoxO1 phosphorylation in BMMs induced by RANKL for 15 min in the presence or absence of 10 μM LY294002 or 0.2 μM wortmannin. (j) Protein levels of FoxOs in cells treated similar to that in i for 2 days. (k,l) BMM incubated with or without RANKL for 6 h (k) or 3 h (l) in the presence or absence of 20 mM MG132. *P<0.05 versus respective veh-treated cells, #P<0.05 versus RANKL alone. Bars represent mean and s.d.; Student’s t-test.
Figure 2. Deletion of FoxOs in osteoclasts…
Figure 2. Deletion of FoxOs in osteoclasts increases bone resorption.
(a) FoxOs mRNA levels in BMM cultures (triplicates). (b) FoxOs mRNA (right) and protein (left) in mature osteoclast cultures. (c) Representative μCT images of distal femur of 6-week-old male mice. Scale bar, 1 mm. (d) Cancellous BMD, bone volume relative to the total volume (BV/TV) and trabecular (Tb) number, thickness and spacing in distal femur of 6-week-old male FoxO1,3,4f/f (n=10) and FoxO1,3,4f/f;LysM-Cre (n=8) littermates. (e) Cortical bone measurements determined using micro-CT in femoral diaphysis of the samples described in d. Pm, perimeter. (f) Osteoclasts in cancellous bone of vertebra from FoxO1,3,4f/f (n=8) and FoxO1,3,4f/f;LysM-Cre (n=11) mice described in d. (g) Histological sections of bone, described in f, stained for TRAP activity (osteoclasts stain red). Scale bar, 20 μm. (h) Serum concentration of a collagen degradation product (CTx) in mice described in d (n=8 mice per group). (i) Cathepsin K (CatK), TRAP, calcitonin receptor (calcr) and osteocalcin (OCN) mRNA levels in vertebrae of mice described in d (n=6 mice per group). Bars represent mean and s.d. *P<0.05 versus FoxO1,3,4f/f by Student’s t-test.
Figure 3. Overexpression of FoxO3 in osteoclasts…
Figure 3. Overexpression of FoxO3 in osteoclasts decreases bone resorption.
(a) FoxO3 transgene mRNA and (b) protein in mature osteoclast cultures (triplicates); ND, not detected. (c) Representative μCT images of distal femur of 12-week-old male mice. Scale bar, 1 mm. (d) Cancellous BMD and bone volume relative to the total volume (BV/TV) of 12-week-old male LysM-Cre (n=12) and FoxO3C;LysM-Cre (n=13) littermates. (e) Osteoclasts in cancellous bone of vertebra from mice described in d (n=6 mice per group). (f) Histological sections of bone, described in e, stained for TRAP activity (osteoclasts stain red). Scale bar, 20 μm. Bars represent mean and s.d. *P<0.05 by Student’s t-test.
Figure 4. FoxOs attenuate osteoclast progenitor proliferation…
Figure 4. FoxOs attenuate osteoclast progenitor proliferation and decrease mature osteoclast lifespan.
(a) Representative pictures (left) and number of TRAP-positive multinucleated cells (MNCs) generated from BMMs (right, triplicates). Scale bar, 500 μm. (b) Cathepsin K (CatK), TRAP and calcitonin receptor (calcr) mRNA levels in osteoclast cultures (triplicates). (c) Representative pictures of BrdU (green) and propidium iodide (red) staining (left) and percentage of BrdU-positive cells in BMMs cultures (right; triplicates). Scale bar, 20 μm. (df) Same as ad using cells from FoxO3C;LysM-Cre and LysM-Cre littermates. (g) Caspase-3 activity in mature osteoclast cultures (triplicates). AFU, arbitrary fluorescent units. (h) Cyclin D1 mRNA and (i) protein levels in BMM cell cultures (triplicate). Bars represent mean and s.d.; *P<0.05 versus respective FoxO1,3,4f/f or LysM-Cre cells by Student’s t-test.
Figure 5. FoxOs attenuate bone resorption by…
Figure 5. FoxOs attenuate bone resorption by decreasing H2O2.
(a) Catalase mRNA levels in BMM and in mature osteoclast (mOC) cultures (triplicates). (b) Catalase protein levels in mature osteoclast. (c) MnSOD mRNA (triplicates). (d) H2O2 levels using Amplex Red in osteoclast progenitors (triplicates). (e) Proliferation by BrdU labelling; and (f) cyclin D1 mRNA levels in BMM cultures treated with vehicle (veh) or 75 Units ml−1 of pegylated catalase (triplicates). (g) Cyclin D1 mRNA levels in BMM silenced for catalase (triplicates). Sh, short hairpin. (h) Four-week-old male mice were administered veh or pegylated catalase (CAT) for 6 weeks. Bone volume relative to the total volume (BV/TV), and trabecular (Tb) number and spacing in vertebrae; and (i) cortical thickness at the midshaft of femur (n=10–11 per group). Bars represent mean and s.d. *P<0.05 by Student’s t-test. #P<0.05 by two-way analysis of variance (ANOVA).
Figure 6. Mitochondria-derived H 2 O 2…
Figure 6. Mitochondria-derived H2O2 promotes osteoclast generation and bone resorption.
(a) Catalase activity in soft tissues and osteoclast (Oc) cultures from LysM-Cre (n=4) and MitoCat-FS;LysM-Cre (n=4) littermates. RLU, relative light units. (b) Representative μCT images of distal femur of 12-week-old male mice. Scale bar, 1 mm. (c) Cancellous BMD and bone volume relative to the total volume (BV/TV) of 12-week-old male LysM-Cre (n=12) and MitoCat-FS;LysM-Cre (n=10) littermates. (d) Cortical thickness determined by μCT in femoral diaphysis of the samples described in c. (e) Osteoclasts in cancellous bone of vertebra from mice described in c (n=6 mice per group). (f) Histological sections of bones, described in e, stained for TRAP activity (osteoclasts stain red). Scale bar, 20 μm. (g) Number of TRAP-positive MNCs generated from BMMs (triplicates). (h) Cathepsin K (CatK), TRAP and calcitonin receptor (calcr) mRNA levels in osteoclast cultures (triplicates). (i) Proliferation by BrdU labelling in BMM cultures (triplicates). (j) Caspase-3 activity in mature osteoclast cultures (triplicates). AFU, arbitrary fluorescent units. (k) Twelve-week-old mice were sham-operated or OVX and killed 6 weeks later. BMD by DEXA as % change from the initial BMD in LysM-Cre sham (n=10), LysM-Cre OVX (n=9), MitoCat-FS;LysM-Cre sham (n=7) and MitoCat-FS;LysM-Cre OVX (n=7) mice. (l) Serum concentration of a collagen degradation product (CTx), in mice described in k, at the time of killing. Bars represent mean and s.d. *P<0.05 by Student’s t-test. #P<0.05 by two-way ANOVA.
Figure 7. Effects of FoxOs on the…
Figure 7. Effects of FoxOs on the birth and death of osteoblasts and osteoclasts.
Aberrant cell number is the culprit of most metabolic bone diseases, including osteoporosis. Earlier work, with targeted loss or gain of FoxO function at different stages of osteoblast differentiation in mice, revealed a restraining effect of FoxOs on Wnt signalling in mesenchymal progenitors. This effect leads to a decrease in the number of osteoblasts and is independent of the antioxidant properties of FoxOs. On the other hand, FoxOs promote the survival of mature osteoblasts. Previous work has also elucidated that FoxOs reduce ROS and promote stemness in haematopoietic stem cells. The present report shows that FoxOs prevent the accumulation of H2O2 in osteoclasts and their progenitors by increasing the expression of catalase and, thereby, promote cell cycle arrest and apoptosis.

References

    1. Teitelbaum S. L. & Ross F. P. Genetic regulation of osteoclast development and function. Nat. Rev. Genet. 4, 638–649 (2003).
    1. Parfitt A. M. Targeted and nontargeted bone remodeling: relationship to basic multicellular unit origination and progression. Bone 30, 5–7 (2002).
    1. Manolagas S. C. Birth and death of bone cells: basic regulatory mechanisms and implications for the pathogenesis and treatment of osteoporosis. Endocr. Rev. 21, 115–137 (2000).
    1. Boyde A., Ali N. N. & Jones S. J. Resorption of dentine by isolated osteoclasts in vitro. Br Dent. J 156, 216–220 (1984).
    1. Chambers T. J., Revell P. A., Fuller K. & Athanasou N. A. Resorption of bone by isolated rabbit osteoclasts. J. Cell Sci. 66, 383–399 (1984).
    1. Destaing O., Saltel F., Geminard J. C., Jurdic P. & Bard F. Podosomes display actin turnover and dynamic self-organization in osteoclasts expressing actin-green fluorescent protein. Mol. Biol. Cell 14, 407–416 (2003).
    1. Downing J. R., Rettenmier C. W. & Sherr C. J. Ligand-induced tyrosine kinase activity of the colony-stimulating factor 1 receptor in a murine macrophage cell line. Mol. Cell Biol. 8, 1795–1799 (1988).
    1. Sengupta A. et al. Identification and subcellular localization of proteins that are rapidly phosphorylated in tyrosine in response to colony-stimulating factor 1. Proc. Natl Acad. Sci. USA 85, 8062–8066 (1988).
    1. Hamilton J. A. CSF-1 signal transduction. J. Leukoc. Biol. 62, 145–155 (1997).
    1. Boyle W. J., Simonet W. S. & Lacey D. L. Osteoclast differentiation and activation. Nature 423, 337–342 (2003).
    1. Garrett I. R. et al. Oxygen-derived free radicals stimulate osteoclastic bone resorption in rodent bone in vitro and in vivo. J. Clin. Invest. 85, 632–639 (1990).
    1. Ha H. et al. Reactive oxygen species mediate RANK signaling in osteoclasts. Exp. Cell Res. 301, 119–127 (2004).
    1. Lee N. K. et al. A crucial role for reactive oxygen species in RANKL-induced osteoclast differentiation. Blood 106, 852–859 (2005).
    1. Bhatt N. Y. et al. Macrophage-colony-stimulating factor-induced activation of extracellular-regulated kinase involves phosphatidylinositol 3-kinase and reactive oxygen species in human monocytes. J. Immunol. 169, 6427–6434 (2002).
    1. Ishii K. A. et al. Coordination of PGC-1beta and iron uptake in mitochondrial biogenesis and osteoclast activation. Nat. Med. 15, 259–266 (2009).
    1. Manolagas S. C. From estrogen-centric to aging and oxidative stress: a revised perspective of the pathogenesis of osteoporosis. Endocr. Rev. 31, 266–300 (2010).
    1. Lean J. M. et al. A crucial role for thiol antioxidants in estrogen-deficiency bone loss. J. Clin. Invest. 112, 915–923 (2003).
    1. Yamasaki N. et al. High oxygen tension prolongs the survival of osteoclast precursors via macrophage colony-stimulating factor. Bone 44, 71–79 (2009).
    1. Balaban R. S., Nemoto S. & Finkel T. Mitochondria, oxidants, and aging. Cell 120, 483–495 (2005).
    1. Tothova Z. et al. FoxOs are critical mediators of hematopoietic stem cell resistance to physiologic oxidative stress. Cell 128, 325–339 (2007).
    1. de Keizer P., Burgering B. & Dansen T. B. FOXO as a sensor, mediator and regulator of redox signaling. Antioxid. Redox Signal. 14, 1093–1105 (2011).
    1. Calnan D. R. & Brunet A. The FoxO code. Oncogene 27, 2276–2288 (2008).
    1. Ambrogini E. et al. FoxO-mediated defense against oxidative stress in osteoblasts is indispensable for skeletal homoeostasis in mice. Cell Metab. 11, 136–146 (2010).
    1. Rached M. T. et al. FoxO1 is a positive regulator of bone formation by favoring protein synthesis and resistance to oxidative stress in osteoblasts. Cell Metab. 11, 147–160 (2010).
    1. Iyer S. et al. FoxOs attenuate bone formation by suppressing Wnt signaling. J. Clin. Invest. 123, 3404–3419 (2013).
    1. Tsuchiya K. et al. Expanded granulocyte/monocyte compartment in myeloid-specific triple FoxO knockout increases oxidative stress and accelerates atherosclerosis in mice. Circ. Res. 112, 992–1003 (2013).
    1. Brunet A. et al. Akt promotes cell survival by phosphorylating and inhibiting a Forkhead transcription factor. Cell 96, 857–868 (1999).
    1. Nemoto S. & Finkel T. Redox regulation of forkhead proteins through a p66shc-dependent signaling pathway. Science 295, 2450–2452 (2002).
    1. Yang J. Y. et al. ERK promotes tumorigenesis by inhibiting FOXO3a via MDM2-mediated degradation. Nat. Cell Biol. 10, 138–148 (2008).
    1. Huang H. & Tindall D. J. Regulation of FOXO protein stability via ubiquitination and proteasome degradation. Biochim. Biophys. Acta 1813, 1961–1964 (2011).
    1. Essaghir A., Dif N., Marbehant C. Y., Coffer P. J. & Demoulin J. B. The transcription of FOXO genes is stimulated by FOXO3 and repressed by growth factors. J. Biol. Chem. 284, 10334–10342 (2009).
    1. Clausen B. E., Burkhardt C., Reith W., Renkawitz R. & Forster I. Conditional gene targeting in macrophages and granulocytes using LysMcre mice. Transgenic Res. 8, 265–277 (1999).
    1. Marinkovic D. et al. Foxo3 is required for the regulation of oxidative stress in erythropoiesis. J Clin. Invest. 117, 2133–2144 (2007).
    1. Almeida M. et al. Skeletal involution by age-associated oxidative stress and its acceleration by loss of sex steroids. J. Biol. Chem. 282, 27285–27297 (2007).
    1. Dai D. F. et al. Mitochondrial oxidative stress mediates angiotensin ii-induced cardiac hypertrophy and G{alpha}q overexpression-induced heart failure. Circ. Res. 108, 837–846 (2011).
    1. Giorgio M., Trinei M., Migliaccio E. & Pelicci P. G. Hydrogen peroxide: a metabolic by-product or a common mediator of ageing signals? Nat. Rev. Mol. Cell Biol. 8, 722–728 (2007).
    1. Newmeyer D. D. & Ferguson-Miller S. Mitochondria: releasing power for life and unleashing the machineries of death. Cell 112, 481–490 (2003).
    1. Chance B., Sies H. & Boveris A. Hydroperoxide metabolism in mammalian organs. Physiol. Rev. 59, 527–605 (1979).
    1. D'Autreaux B. & Toledano M. B. ROS as signalling molecules: mechanisms that generate specificity in ROS homoeostasis. Nat. Rev. Mol. Cell Biol. 8, 813–824 (2007).
    1. Harman D. Aging: a theory based on free radical and radiation chemistry. J. Gerontol. 11, 298–300 (1956).
    1. Janssen-Heininger Y. M. et al. Redox-based regulation of signal transduction: principles, pitfalls, and promises. Free Radic. Biol. Med. 45, 1–17 (2008).
    1. Sena L. A. & Chandel N. S. Physiological roles of mitochondrial reactive oxygen species. Mol. Cell 48, 158–167 (2012).
    1. Finkel T. Signal transduction by mitochondrial oxidants. J. Biol. Chem. 287, 4434–4440 (2012).
    1. Pinton P. et al. Protein kinase C beta and prolyl isomerase 1 regulate mitochondrial effects of the life-span determinant p66Shc. Science 315, 659–663 (2007).
    1. Tormos K. V. et al. Mitochondrial complex III ROS regulate adipocyte differentiation. Cell Metab. 14, 537–544 (2011).
    1. Leloup C. et al. Mitochondrial reactive oxygen species are obligatory signals for glucose-induced insulin secretion. Diabetes 58, 673–681 (2009).
    1. Pan Y., Schroeder E. A., Ocampo A., Barrientos A. & Shadel G. S. Regulation of yeast chronological life span by TORC1 via adaptive mitochondrial ROS signaling. Cell Metab. 13, 668–678 (2011).
    1. Lee S. J., Hwang A. B. & Kenyon C. Inhibition of respiration extends C. elegans life span via reactive oxygen species that increase HIF-1 activity. Curr. Biol. 20, 2131–2136 (2010).
    1. Schulz T. J. et al. Glucose restriction extends Caenorhabditis elegans life span by inducing mitochondrial respiration and increasing oxidative stress. Cell Metab. 6, 280–293 (2007).
    1. Yang W. & Hekimi S. A mitochondrial superoxide signal triggers increased longevity in Caenorhabditis elegans. PLoS Biol. 8, e1000556 (2010).
    1. West A. P. et al. TLR signalling augments macrophage bactericidal activity through mitochondrial ROS. Nature 472, 476–480 (2011).
    1. Zhou R., Yazdi A. S., Menu P. & Tschopp J. A role for mitochondria in NLRP3 inflammasome activation. Nature 469, 221–225 (2011).
    1. Lambeth J. D. NOX enzymes and the biology of reactive oxygen. Nat. Rev. Immunol. 4, 181–189 (2004).
    1. Paik J. H. et al. FoxOs cooperatively regulate diverse pathways governing neural stem cell homoeostasis. Cell Stem Cell 5, 540–553 (2009).
    1. Renault V. M. et al. FoxO3 regulates neural stem cell homoeostasis. Cell Stem Cell 5, 527–539 (2009).
    1. Lopez-Otin C., Blasco M. A., Partridge L., Serrano M. & Kroemer G. The hallmarks of aging. Cell 153, 1194–1217 (2013).
    1. Manolagas S. C., Kousteni S. & Jilka R. L. Sex steroids and bone. Recent Prog. Horm. Res. 57, 385–409 (2002).
    1. Nakamura T. et al. Estrogen prevents bone loss via estrogen receptor alpha and induction of Fas ligand in osteoclasts. Cell 130, 811–823 (2007).
    1. Martin-Millan M. et al. The estrogen receptor alpha in osteoclasts mediates the protective effects of estrogens on cancellous but not cortical bone. Mol. Endocrinol. 24, 323–334 (2010).
    1. Almeida M. et al. Estrogen receptor-alpha signaling in osteoblast progenitors stimulates cortical bone accrual. J. Clin. Invest. 123, 394–404 (2013).
    1. Onal M. et al. Receptor activator of nuclear factor kappaB ligand (RANKL) protein expression by B lymphocytes contributes to ovariectomy-induced bone loss. J. Biol. Chem. 287, 29851–29860 (2012).
    1. Haidar R., Musallam K. M. & Taher A. T. Bone disease and skeletal complications in patients with beta thalassemia major. Bone 48, 425–432 (2011).
    1. Tsay J. et al. Bone loss caused by iron overload in a murine model: importance of oxidative stress. Blood 116, 2582–2589 (2010).
    1. Lee H. Y. et al. Targeted expression of catalase to mitochondria prevents age-associated reductions in mitochondrial function and insulin resistance. Cell Metab. 12, 668–674 (2010).
    1. Mao P. et al. Mitochondria-targeted catalase reduces abnormal APP processing, amyloid beta production and BACE1 in a mouse model of Alzheimer's disease: implications for neuroprotection and lifespan extension. Hum. Mol. Genet. 21, 2973–2990 (2012).
    1. Someya S. et al. Age-related hearing loss in C57BL/6J mice is mediated by Bak-dependent mitochondrial apoptosis. Proc. Natl Acad. Sci. USA 106, 19432–19437 (2009).
    1. Paik J. H. et al. FoxOs are lineage-restricted redundant tumor suppressors and regulate endothelial cell homoeostasis. Cell 128, 309–323 (2007).
    1. Parfitt A. M. et al. Bone histomorphometry: standardization of nomenclature, symbols, and units. Report of the ASBMR Histomorphometry Nomenclature Committee. J. Bone Miner. Res. 2, 595–610 (1987).
    1. Huang X., Frenkel K., Klein C. B. & Costa M. Nickel induces increased oxidants in intact cultured mammalian cells as detected by dichlorofluorescein fluorescence. Toxicol. Appl. Pharmacol. 120, 29–36 (1993).
    1. Livak K. J. & Schmittgen T. D. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods 25, 402–408 (2001).

Source: PubMed

3
Prenumerera