Elsevier

Bone

Volume 116, November 2018, Pages 307-314
Bone

Full Length Article
Vhl deficiency in osteocytes produces high bone mass and hematopoietic defects

https://doi.org/10.1016/j.bone.2018.08.022Get rights and content

Highlights

  • Osteocytes exert a robust influence on bone mass through their capacity to perceive pericellular oxygen.

  • Osteocytic Hif1a is dispensible for proper skeletal development.

  • Osteocytic Vhl deletion promotes canonical Wnt signaling.

  • Absence of osteocytic Vhl reduces B cell maturation in bone marrow.

Abstract

Tissue oxygen (O2) levels vary during development and disease; adaptations to decreased O2 (hypoxia) are mediated by hypoxia-inducible factor (HIF) transcription factors. HIFs are active in the skeleton, and stabilizing HIF-α isoforms cause high bone mass (HBM) phenotypes. A fundamental limitation of previous studies examining the obligate role for HIF-α isoforms in the skeleton involves the persistence of gene deletion as osteolineage cells differentiate into osteocytes. Because osteocytes orchestrate skeletal development and homeostasis, we evaluated the influence of Vhl or Hif1a disruption in osteocytes. Osteocytic Vhl deletion caused HBM phenotype, but Hif1a was dispensable in osteocytes. Vhl cKO mice revealed enhanced canonical Wnt signaling. B cell development was reduced while myelopoiesis increased in osteocytic Vhl cKO, revealing a novel influence of Vhl/HIF-α function in osteocytes on maintenance of bone microarchitecture via canonical Wnt signaling and effects on hematopoiesis.

Introduction

Oxygen (O2) levels vary throughout the body. They change spatiotemporally during development, and can be acutely impacted during pathophysiological insults, such as stroke or bone fracture. Adaptive responses to decreased O2 (hypoxia) are rectified by transcriptional induction of genes promoting angiogenesis and anaerobic glycolysis, which are mediated, in part, by the hypoxia-inducible factor (HIF) family of transcription factors [1]. Constitutively expressed HIF-β subunits bind to one of three distinct HIF-α isoforms (HIF-1α, HIF-2α, and HIF-3α) to initiate gene expression, although HIF-3α is considered an inhibitor of HIF-1α and HIF-2α. Oxygen levels regulate HIF-responsive gene expression. Under normoxic conditions, HIF-α subunits are proline hydroxylated by proline hydroxylase-domain proteins (PHDs), targeted for ubiquitination by the E3 ligase complex von Hippel-Lindau (Vhl) and are ultimately degraded by the 26S proteasome. Under hypoxic conditions, prolyl hydroxylation of HIF-α is inhibited, enabling HIF-α to accumulate, translocate into the nucleus and complex with HIF-β/ARNT, and ultimately bind to HIF-response elements (HRE) within target genes.

The VHL/HIF system is highly active in the skeleton. Multiple strategies that use osteolineage-specific cre drivers and deletion of PHD or Vhl to stabilize HIF-1α and HIF-2α increase bone mass. Individually or combinatorially deleting Phd1, Phd2, or Phd3 in Osterix-positive osteoprogenitors improves trabecular microarchitecture dependent upon the degree of HIF-α stabilization [2]. Similarly, deletion of Vhl in mature osteoblasts using an osteocalcin (Bglap) cre driver increases trabecular and cortical microarchitecture [3].

The murine models described above clearly demonstrate that manipulating HIF-α levels in bone cells influences skeletal development and homeostasis. In these models, Vhl or Phd isoform elimination persists as osteoprogenitors differentiate into osteoblasts and then transition into osteocytes. Thus, these models cannot elucidate the influence of Vhl deletion in osteocytes compared to Vhl deletion in osteoprogenitors or osteoblasts. Osteocytes are the most abundant cells within bone, comprising >90% of total cells [4]. Osteocytes exert critical roles in mechano-sensing, skeletal remodeling, mineral homeostasis and hematopoiesis [5]. Indeed, the osteocyte and the extensive osteocytic network in bone are now recognized as a central regulator of skeletal activity [5,6]. The frequency of HIF-1α-positive osteocytes increases in mice under disuse conditions [7]; in vitro, exposing osteocyte-like cells to hypoxia augments secretion of chemotactic factors [8] and GDF15 to promote osteoclastogenesis [9], and influences the osteoblast-to-osteocyte transition [10]. Other osteocytic functions attributed to HIF-α function include directing appropriate bone formation in response to mechanical loading [11], sensing microdamage [12], and development of the lacunocanalicular network in which osteocytes reside [13].

Provided the recognition that osteocytes orchestrate many aspects of skeletal development and homeostasis, we sought to evaluate the influence of Vhl or Hif1a disruption in osteocytes. We used a cre-loxP system under control of the non-inducible 10 kb-Dmp1 promoter [14] to address the function of Vhl in osteocytes. Similar to results using Bglap-cre to delete Vhl in mature osteoblasts [3], osteocytic deletion of Vhl produced mice with a high bone mass (HBM) phenotype that dramatically increased trabecular and cortical microarchitecture. Femoral osteocytes in Vhl cKO mice revealed qualitative and quantitative reductions in sclerostin immunostaining, and concomitant increases in activated β-catenin levels, suggesting that Vhl/HIF-α signaling influences canonical Wnt signaling in osteocytes. Vhl cKO mice mated to mice lacking the Wnt co-receptor Lrp5 or to transgenic mice over-expressing the human SOST gene revealed that Vhl functions epistatically to canonical Wnt signaling: cortical microarchitecture resolved to wild-type phenotype in compound mice, but not trabecular microarchitecture. Osteocytic HIF-α contributes to hematopoiesis, as B cell development is blocked in osteocytic Vhl cKO mice. These data reveal a novel influence of Vhl/HIF-α function in osteocytes on maintenance of bone microarchitecture via canonical Wnt signaling and effects on hematopoiesis.

Section snippets

Mice

Vhlhf/f [15] (#004081) and Hif1af/f [16] (#007561) were purchased from Jackson Laboratories and bred to control C57Bl/6 or with mice expressing cre recombinase from the 10 kb-Dmp1 promoter (Dmp1-cre) [B6N.FVB-Tg(Dmp1-cre)1Jqfe/BwdJ] [14] (kindly provided by Dr. Jian Q. Feng, Department of Biomedical Sciences, Texas A&M University Baylor College of Dentistry) to generate Dmp1-cre; Vhlhf/f and Dmp1-cre; Hif1af/f mice with osteocyte conditional deletion of Vhlh (hereafter, Vhl cKO) or Hif1a (Hif1a

Deletion of Vhl in osteocytes has robust effects on trabecular and cortical microarchitecture

Deletion of Vhl and subsequent stabilization of HIF-1α and HIF-2α in mature osteoblasts promotes bone modeling and accrual, whereas deletion of Hif1a produces an inverse skeletal phenotype [3]. Because Vhl deletion in mature osteoblasts persists as osteoblasts differentiate into osteocytes, previous data do not identify the impact of osteocytic Vhl deletion on skeletal mass and architecture. In order to examine the impact of Vhl and Hif1a expression in osteocytes, we used a cre-loxP system

Osteocytic Vhl contributes to skeletal development

Stabilizing HIF-α isoforms, via deletion of Phd1, Phd2, or Phd3 [2], or Vhl [3], produced HBM mice, which affected both trabecular and cortical bone. Osteoblastic Vhl deletion also increased bone formation in response to distraction osteogenesis [31], and pharmacologic activation of HIF-αs increased bone volume in distraction gaps [31,32] and fracture callus [33]. Thus, using genetic and pharmacologic means, it is clear that stabilizing HIF-α isoforms influences skeletal microarchitecture,

Acknowledgements

Research reported in this publication was supported by the University of California Merced faculty research awards (JOM), Veterans Administration award number BX001478 (to A.G.R) and the National Institute of Arthritis and Musculoskeletal and Skin Diseases of the National Institutes of Health under award numbers R01AR053237 (AGR), R03AR057547 (DCG), and R01AR064255 (DCG). The content is solely the responsibility of the authors and does not necessarily represent the official views of the

References (52)

  • Y. Gong et al.

    LDL receptor-related protein 5 (LRP5) affects bone accrual and eye development

    Cell

    (2001)
  • H.Y. Cho et al.

    Transgenic mice overexpressing secreted frizzled-related proteins (sFRP)4 under the control of serum amyloid P promoter exhibit low bone mass but did not result in disturbed phosphate homeostasis

    Bone

    (2010)
  • J. Zhu et al.

    Osteoblasts support B-lymphocyte commitment and differentiation from hematopoietic stem cells

    Blood

    (2007)
  • K. Fulzele et al.

    Myelopoiesis is regulated by osteocytes through Gsα-dependent signaling

    Blood

    (2013)
  • L.M. Calvi et al.

    Osteoblastic expansion induced by parathyroid hormone receptor signaling in murine osteocytes is not sufficient to increase hematopoietic stem cells

    Blood

    (2012)
  • Y. Fujiwara et al.

    RANKL (receptor activator of NFκB ligand) produced by osteocytes is required for the increase in B cells and bone loss caused by estrogen deficiency in mice

    J. Biol. Chem.

    (2016)
  • M. Sato et al.

    Osteocytes regulate primary lymphoid organs and fat metabolism

    Cell Metab.

    (2013)
  • G.L. Semenza

    Hydroxylation of HIF-1: oxygen sensing at the molecular level

    Physiology (Bethesda)

    (2004)
  • C. Wu et al.

    Oxygen-sensing PHDs regulate bone homeostasis through the modulation of osteoprotegerin

    Genes Dev.

    (2015)
  • Y. Wang et al.

    The hypoxia-inducible factor alpha pathway couples angiogenesis to osteogenesis during skeletal development

    J. Clin. Invest.

    (2007)
  • M.B. Schaffler et al.

    Osteocyte signaling in bone

    Curr. Osteoporos. Rep.

    (2012)
  • S.L. Dallas et al.

    The osteocyte: an endocrine cell and more

    Endocr. Rev.

    (2013)
  • J. Delgado-Calle et al.

    Osteocytes and skeletal pathophysiology

    Curr. Mol. Biol. Rep.

    (2015)
  • T.S. Gross et al.

    Upregulation of osteopontin by osteocytes deprived of mechanical loading or oxygen

    J. Bone Miner. Res.

    (2005)
  • E. Hinoi et al.

    Positive regulation of osteoclastic differentiation by growth differentiation factor 15 upregulated in osteocytic cells under hypoxia

    J. Bone Miner. Res.

    (2012)
  • G.-L. Zuo et al.

    Activation of HIFa pathway in mature osteoblasts disrupts the integrity of the osteocyte/canalicular network

    PLoS ONE

    (2015)
  • Cited by (29)

    • Energy homeostasis in the bone

      2024, Trends in Endocrinology and Metabolism
    • Hypoxia signaling in bone physiology and energy metabolism

      2023, Current Opinion in Endocrine and Metabolic Research
    • TRIM21 drives intervertebral disc degeneration induced by oxidative stress via mediating HIF-1α degradation

      2021, Biochemical and Biophysical Research Communications
      Citation Excerpt :

      HIFs are one of the most important factors that directly mediate cellular responses to hypoxia [4]. The HIF family consists of three α subunits: HIF-1α, HIF-2α, HIF-3α, and one β subunit, HIF-1β [22]. Under normoxic conditions, HIF-α subunits are proline hydroxylated through proline hydroxylase-domain proteins (PHDs), targeted by the E3 ligase complex (Vhl) for proteasomal degradation.

    • HIF-1α induces hypoxic apoptosis of MLO-Y4 osteocytes via JNK/caspase-3 pathway and the apoptotic-osteocyte-mediated osteoclastogenesis in vitro

      2020, Tissue and Cell
      Citation Excerpt :

      As for osteocyte, some recent studies showed that in vivo oxygen tension of osteocyte lacunae was well below 10 % (Spencer et al., 2014), a key extracorporeal threshold for PHD enzyme activity, suggesting the importance of strict regulation of HIF signaling in normal osteocyte function (Stegen et al., 2018). The VHL-deficient osteocytes, activating more HIF-1α protein expression, were significantly thinner than WT cells in both the volume and surface area and showed clear signs of apoptosis such as cytoplasmic shrinkage, chromatin condensation, and nuclear disintegration (Shi et al., 2015; Loots et al., 2018). In light of the findings summmarized above, we hypothesized that HIF-1α positively regulated the apoptosis of osteocytes.

    View all citing articles on Scopus
    View full text