Human type H vessels are a sensitive biomarker of bone mass

Liang Wang, Fei Zhou, Peng Zhang, Hongzhen Wang, Zhipeng Qu, Peng Jia, Zhe Yao, Guangsi Shen, Guangfei Li, Guoyang Zhao, Jian Li, Yongtao Mao, Zonggang Xie, Wei Xu, Youjia Xu, Ying Xu, Liang Wang, Fei Zhou, Peng Zhang, Hongzhen Wang, Zhipeng Qu, Peng Jia, Zhe Yao, Guangsi Shen, Guangfei Li, Guoyang Zhao, Jian Li, Yongtao Mao, Zonggang Xie, Wei Xu, Youjia Xu, Ying Xu

Abstract

Vascularization is fundamental for bone formation and bone tissue homeostasis. However, in human subjects, a direct molecular relationship has not been identified between angiogenesis and agents that promote bone disease or factors related to age. Osteopenia is a condition in which bone mineral density is lower than normal, and it represents a sign of normal aging. Here we tested whether the type H vessel, which was recently identified as strongly positive for CD31 and Endomucin (CD31hiEmcnhi) in mice, is an important indicator of aging and osteopenia in human subjects. We found that age-dependent losses of type H vessels in human bone sections conform to the observations in aged mice. The abundance of human type H vessels and osteoprogenitors may be relevant to changes in the skeletal microarchitecture and advanced osteopenia. Furthermore, ovariectomized mice, a widely used model for postmenopausal osteoporosis, exhibited significantly reduced type H vessels accompanied by reduced osteoprogenitors, which is consistent with impaired bone microarchitecture and osteoporosis, suggesting that this feature is an indicator of bone mass independent of aging. More importantly, administration of desferrioxamine led to significantly increased bone mass via enhanced angiogenesis and increased type H vessels in ovariectomized mice. Altogether, these data represent a novel finding that type H vessels are regulated in aged and osteopenia subjects. The abundance of human type H vessels is an early marker of bone loss and represents a potential target for improving bone quality via the induction of type H vessels.

Trial registration: ClinicalTrials.gov NCT02750046.

Conflict of interest statement

The authors declare no conflict of interest.

Figures

Figure 1
Figure 1
Age series of mouse tibia and femur immunostaining. (ac) Tibia immunostaining for CD31 (green), Endomucin (red), Osterix (gray), and DAPI (blue) for juvenile (3 weeks, a), adult (11 weeks, b), and aged (52 weeks, c) mice. (df), Femur (proximal region) immunostaining for CD31 (green), Endomucin (red), Osterix (gray), and DAPI (blue) for juvenile (5 weeks, d), adult (16 weeks, e), and aged (47 weeks, f) mice. Line in each panel indicates the boundary of metaphysis and growth plate. Scale bar: 100 μm
Figure 2
Figure 2
Decreased type H vessels in aged patients. (a), Schematic illustration of bone sample collection from patients during surgery. (b), Typical human bone sample collected from PFN surgery. (c,d), Pre- and post-surgical X-ray images of a typical patient. (eg), Bone immunostaining for CD31 (green), Endomucin (red), Osterix (gray), and DAPI (blue) for young (e), adult (f) and aged (g) patients. (h) Quantification of type H vessel percentage (HV/TV%) in young (N=7), adult (N=8), and aged patients (N=24). Graphs represent mean±S.D.; *P<0.05; ***P<0.0001; Scale bar: 100 μm
Figure 3
Figure 3
Synergistic decrease of type H vessel and bone mineral density in human bone. (ac) Micro-CT 3D reconstructed image of bone samples from normal (a), osteopenia (b), and osteoporosis (c) patients. (di) Quantification of the trabecular index of bone samples from normal (N=6), osteopenia (N=10), and osteoporosis (N=8) patients. (jl) Bone immunostaining for CD31 (green), Endomucin (red), Osterix (gray), and DAPI (blue) for normal (j), osteopenia (k), and osteoporosis (l) patients. Graphs represent mean±S.D.; *P<0.05; **P<0.01; Scale bar: 100 μm
Figure 4
Figure 4
Change of type H vessels in osteoporotic mice. (af) Tibia immunostaining for CD31 (green), Endomucin (red), Osterix (gray), and DAPI (blue) of Sham (ab), OVX (cd), and OVX+DFO (ef) group mice. Lines in b, d, and f indicate the boundary of the metaphysis and growth plate. (gi) Micro-CT 3D reconstructed images of cancellous bone from the Sham (g), OVX (h), and OVX+DFO (i) mice. (jn) Quantification of the BMD and trabecular index of bone from the Sham, OVX, and OVX+DFO mice. Graphs represent mean±S.D.; *P<0.05; **P<0.01; Scale bar: 200 μm (e), 100 μm (f)

References

    1. Long F. Building strong bones: molecular regulation of the osteoblast lineage. Nat Rev Mol Cell Biol 2012; 13: 27–38.
    1. Aliprantis AO, Ueki Y, Sulyanto R, Park A, Sigrist KS, Sharma SM et al. NFATc1 in mice represses osteoprotegerin during osteoclastogenesis and dissociates systemic osteopenia from inflammation in cherubism. J Clin Invest 2008; 118: 3775–3789.
    1. Chang J, Wang Z, Tang E, Fan Z, McCauley L, Franceschi R et al. Inhibition of osteoblastic bone formation by nuclear factor-kappaB. Nat Med 2009; 15: 682–689.
    1. Cao H, Yu S, Yao Z, Galson DL, Jiang Y, Zhang X et al. Activating transcription factor 4 regulates osteoclast differentiation in mice. J Clin Invest 2010; 120: 2755–2766.
    1. Wang X, Guo B, Li Q, Peng J, Yang Z, Wang A et al. miR-214 targets ATF4 to inhibit bone formation. Nat Med 2013; 19: 93–100.
    1. Yu B, Chang J, Liu Y, Li J, Kevork K, Al-Hezaimi K et al. Wnt4 signaling prevents skeletal aging and inflammation by inhibiting nuclear factor-kappaB. Nat Med 2014; 20: 1009–1017.
    1. Scholtysek C, Katzenbeisser J, Fu H, Uderhardt S, Ipseiz N, Stoll C et al. PPARbeta/delta governs Wnt signaling and bone turnover. Nat Med 2013; 19: 608–613.
    1. Moverare-Skrtic S, Henning P, Liu X, Nagano K, Saito H, Borjesson AE et al. Osteoblast-derived WNT16 represses osteoclastogenesis and prevents cortical bone fragility fractures. Nat Med 2014; 20: 1279–1288.
    1. Maeda K, Kobayashi Y, Udagawa N, Uehara S, Ishihara A, Mizoguchi T et al. Wnt5a-Ror2 signaling between osteoblast-lineage cells and osteoclast precursors enhances osteoclastogenesis. Nat Med 2012; 18: 405–412.
    1. Baron R, Kneissel M. WNT signaling in bone homeostasis and disease: from human mutations to treatments. Nat Med 2013; 19: 179–192.
    1. Cui Y, Niziolek PJ, MacDonald BT, Zylstra CR, Alenina N, Robinson DR et al. Lrp5 functions in bone to regulate bone mass. Nat Med 2011; 17: 684–691.
    1. Brown JP, Prince RL, Deal C, Recker RR, Kiel DP, de Gregorio LH et al. Comparison of the effect of denosumab and alendronate on BMD and biochemical markers of bone turnover in postmenopausal women with low bone mass: a randomized, blinded, phase 3 trial. J Bone Miner Res 2009; 24: 153–161.
    1. Tomlinson RE, Silva MJ. Skeletal blood flow in bone repair and maintenance. Bone Res 2013; 1: 311–322.
    1. Kanczler JM, Oreffo RO. Osteogenesis and angiogenesis: the potential for engineering bone. Eur Cell Mater 2008; 15: 100–114.
    1. Saran U, Gemini Piperni S, Chatterjee S. Role of angiogenesis in bone repair. Arch Biochem Biophys 2014; 561: 109–117.
    1. Maes C, Kobayashi T, Selig MK, Torrekens S, Roth SI, Mackem S et al. Osteoblast precursors, but not mature osteoblasts, move into developing and fractured bones along with invading blood vessels. Dev Cell 2010; 19: 329–344.
    1. Schipani E, Maes C, Carmeliet G, Semenza GL. Regulation of osteogenesis-angiogenesis coupling by HIFs and VEGF. J Bone Miner Res 2009; 24: 1347–1353.
    1. Kusumbe AP, Ramasamy SK, Adams RH. Coupling of angiogenesis and osteogenesis by a specific vessel subtype in bone. Nature 2014; 507: 323–328.
    1. Xie H, Cui Z, Wang L, Xia Z, Hu Y, Xian L et al. PDGF-BB secreted by preosteoclasts induces angiogenesis during coupling with osteogenesis. Nat Med 2014; 20: 1270–1278.
    1. Nakashima K, Zhou X, Kunkel G, Zhang Z, Deng JM, Behringer RR et al. The novel zinc finger-containing transcription factor osterix is required for osteoblast differentiation and bone formation. Cell 2002; 108: 17–29.
    1. Kusumbe AP, Adams RH. Osteoclast progenitors promote bone vascularization and osteogenesis. Nat Med 2014; 20: 1238–1240.
    1. Bouxsein ML, Boyd SK, Christiansen BA, Guldberg RE, Jepsen KJ, Muller R. Guidelines for assessment of bone microstructure in rodents using micro-computed tomography. J Bone Miner Res 2010; 25: 1468–1486.
    1. Parfitt AM, Drezner MK, Glorieux FH, Kanis JA, Malluche H, Meunier PJ et al. Bone histomorphometry: standardization of nomenclature, symbols, and units. Report of the ASBMR Histomorphometry Nomenclature Committee. J Bone Miner Res 1987; 2: 595–610.
    1. Li ZC, Dai LY, Jiang LS, Qiu S. Difference in subchondral cancellous bone between postmenopausal women with hip osteoarthritis and osteoporotic fracture: implication for fatigue microdamage, bone microarchitecture, and biomechanical properties. Arthritis Rheum 2012; 64: 3955–3962.
    1. Wan C, Gilbert SR, Wang Y, Cao X, Shen X, Ramaswamy G et al. Activation of the hypoxia-inducible factor-1alpha pathway accelerates bone regeneration. Proc Natl Acad Sci USA 2008; 105: 686–691.
    1. Ramasamy SK, Kusumbe AP, Wang L, Adams RH. Endothelial Notch activity promotes angiogenesis and osteogenesis in bone. Nature 2014; 507: 376–380.
    1. Jilka RL. The relevance of mouse models for investigating age-related bone loss in humans. J Gerontol A Biol Sci Med Sci 2013; 68: 1209–1217.
    1. Schneider P, Krucker T, Meyer E, Ulmann-Schuler A, Weber B, Stampanoni M et al. Simultaneous 3D visualization and quantification of murine bone and bone vasculature using micro-computed tomography and vascular replica. Microsc Res Tech 2009; 72: 690–701.
    1. Kusumbe AP, Ramasamy SK, Itkin T, Mae MA, Langen UH, Betsholtz C et al. Age-dependent modulation of vascular niches for haematopoietic stem cells. Nature 2016; 532: 380–384.
    1. Brandi ML, Collin-Osdoby P. Vascular biology and the skeleton. J Bone Miner Res 2006; 21: 183–192.
    1. He X, Dziak R, Yuan X, Mao K, Genco R, Swihart M et al. BMP2 genetically engineered MSCs and EPCs promote vascularized bone regeneration in rat critical-sized calvarial bone defects. PLoS ONE 2013; 8: e60473.
    1. Schipani E, Wu C, Rankin EB, Giaccia AJ. Regulation of bone marrow angiogenesis by osteoblasts during bone development and homeostasis. Front Endocrinol (Lausanne) 2013; 4: 85.
    1. Wang H, Li X, Tomin E, Doty SB, Lane JM, Carney DH et al. Thrombin peptide (TP508) promotes fracture repair by up-regulating inflammatory mediators, early growth factors, and increasing angiogenesis. J Orthop Res 2005; 23: 671–679.
    1. Glowacki J. Angiogenesis in fracture repair. Clin Orthop Relat Res 1998; 355(Suppl): S82–S89.
    1. Peng J, Lai ZG, Fang ZL, Xing S, Hui K, Hao C et al. Dimethyloxalylglycine prevents bone loss in ovariectomized C57BL/6J mice through enhanced angiogenesis and osteogenesis. PLoS ONE 2014; 9: e112744.
    1. Kyriakides ZS, Kremastinos DT, Karayannakos P. Estrogen stimulates angiogenesis in normoperfused skeletal muscle in rabbits. Circulation 2001; 103: E107–E108.
    1. Losordo DW, Isner JM. Estrogen and angiogenesis: a review. Arterioscler Thromb Vasc Biol 2001; 21: 6–12.
    1. Mekraldi S, Lafage-Proust MH, Bloomfield S, Alexandre C, Vico L. Changes in vasoactive factors associated with altered vessel morphology in the tibial metaphysis during ovariectomy-induced bone loss in rats. Bone 2003; 32: 630–641.
    1. Prestwood KM, Kenny AM, Unson C, Kulldorff M. The effect of low dose micronized 17ss-estradiol on bone turnover, sex hormone levels, and side effects in older women: a randomized, double blind, placebo-controlled study. J Clin Endocrinol Metab 2000; 85: 4462–4469.
    1. Kanis JA, Adachi JD, Cooper C, Clark P, Cummings SR, Diaz-Curiel M et al. Standardising the descriptive epidemiology of osteoporosis: recommendations from the Epidemiology and Quality of Life Working Group of IOF. Osteoporos Int 2013; 24: 2763–2764.
    1. Dempster DW, Compston JE, Drezner MK, Glorieux FH, Kanis JA, Malluche H et al. Standardized nomenclature, symbols, and units for bone histomorphometry: a 2012 update of the report of the ASBMR Histomorphometry Nomenclature Committee. J Bone Miner Res 2013; 28: 2–17.

Source: PubMed

3
Abonnere