Skip to main content
Journal of Bone and Mineral Research logoLink to Journal of Bone and Mineral Research
. 2010 Dec 16;26(2):229–238. doi: 10.1002/jbmr.320

The Amazing Osteocyte

Lynda F Bonewald 1
PMCID: PMC3179345  PMID: 21254230

Abstract

The last decade has provided a virtual explosion of data on the molecular biology and function of osteocytes. Far from being the “passive placeholder in bone,” this cell has been found to have numerous functions, such as acting as an orchestrator of bone remodeling through regulation of both osteoclast and osteoblast activity and also functioning as an endocrine cell. The osteocyte is a source of soluble factors not only to target cells on the bone surface but also to target distant organs, such as kidney, muscle, and other tissues. This cell plays a role in both phosphate metabolism and calcium availability and can remodel its perilacunar matrix. Osteocytes compose 90% to 95% of all bone cells in adult bone and are the longest lived bone cell, up to decades within their mineralized environment. As we age, these cells die, leaving behind empty lacunae that frequently micropetrose. In aged bone such as osteonecrotic bone, empty lacunae are associated with reduced remodeling. Inflammatory factors such as tumor necrosis factor and glucocorticoids used to treat inflammatory disease induce osteocyte cell death, but by different mechanisms with potentially different outcomes. Therefore, healthy, viable osteocytes are necessary for proper functionality of bone and other organs. © 2011 American Society for Bone and Mineral Research.

Keywords: OSTEOCYTE, OSTEOCYTIC RESORPTION, REMODELING, OSTEOCYTIC OSTEOLYSIS

Pioneers in the Study of Osteocytes

Before the introduction of PubMed and easy access to papers, some of the earliest observations regarding osteocytes, like the cells themselves, laid buried and difficult to find. My colleagues and I, like others, made assumptions that many of our observations were new and novel until these publications were made readily available. For example, over 100 years ago, it was postulated that osteocytes could remodel their extracellular matrix(1); over 40 years ago, it was thought that osteocytes were responsive to parathyroid hormone,(2) could remodel bone(3) and could express tartrate-specific acid phosphatase(4); and over 20 years ago, it was said that osteocytes were mechanosensory cells.(5) Marotti and Palumbo drew beautiful diagrams to illustrate their theories regarding osteocyte function and communication.(6) Histology was the major tool used by these early pioneers to generate their theories. Peter Nijweide was the first to isolate avian osteocytes.(7) Some of the earliest videos of bone cells including osteocytes were recorded by Kumegawa and colleagues.(8) With new technology, such as molecular and transgenic approaches, imaging, cell lines, systems biology, advanced instrumentation, and others, a dramatic increase in information on osteocyte biology has occurred in the last decade, leading to validation of old theories and the generation of new ones. These are highlighted in this review.

Osteocytes as Descendants of Osteoblasts

The osteocyte, defined as a cell located within the bone matrix, is descended from mesenchymal stem cells through osteoblast differentiation (Fig. 1). It was proposed by Manolagas(9) that the matrix-producing osteoblast either can become an osteocyte, a lining cell, or can undergo programmed cell death. His theories were based on the observations of another very early pioneer, Michael Parfitt, who proposed that osteoblasts must die by apoptosis.(10) Osteocytogenesis has been thought to be a passive process whereby a subpopulation of osteoblasts becomes passively encased in osteoid that passively mineralizes. It was theorized that an osteoblast-producing matrix/osteoid becomes trapped when its neighbor osteoblasts place osteoid on top of the embedding cell.(11) However, there are several arguments against osteocytogenesis being a passive process.

Fig. 1.

Fig. 1

Expression of markers during osteoblast-to-osteocyte ontogeny. The osteocyte appears to be the descendant of the matrix-producing osteoblast, which is a descendant of the mesenchymal stem cell known to express markers such as Stro1, CD29, CD105, CD166. Matrix-producing osteoblasts express Cbfa1 and Osterix, necessary for osteoblast differentiation, followed by alkaline phosphase and collagen, necessary for the production of osteoid. Osteocalcin is produced by the late osteoblast and continues to be expressed by the osteocyte. By some unknown mechanism, some designated cells begin to embed in osteoid and begin to extend dendritic projections, keeping connections with already embedded cells and cells on the bone surface. Molecules such as E11/gp38 and MT1-MMP appear to play a role in dendrite/canaliculi formation, whereas molecules such as destrin and CapG regulate the cytoskeleton. PHEX, MEPE, and DMP-1 regulate biomineralization and mineral metabolism, and FGF-23 regulates renal phosphate excretion. FGF-23 is elevated not only in osteocytes from hypophosphatemic animals but also in those of normal rats.(112) Sclerostin is a marker of the mature osteocyte and is a negative regulator of bone formation.(45) ORP150 may preserve viability of this cell in a hypoxic environment.(23)

One of the first changes to take place in the embedding cell is the formation of dendritic processes. The cell undergoes a dramatic transformation from a polygonal cell to a cell extending dendrites toward the mineralizing front, which is followed by dendrites extending to either the vascular space or bone surface. The cell, once embedded in bone, especially cortical bone, has a polarity, especially with regard to directionality of mineral formation. The osteoid osteocyte must do two major functions simultaneously: regulate mineralization and form connective dendritic processes. The osteoid osteocyte can control and regulate mineralization,(12) and Holmbeck and colleagues(13) have shown osteocytogenesis to be an active invasive process requiring cleavage of collagen and potentially other matrix molecules. Osteocytes in mice null for the metalloproteinase MT1-MMP have significantly reduced number and length of dendritic processes. MT1-MMP is a membrane-anchored proteinase that can cleave collagens type I, II, and III, fibrin, fibronectin, and other matrix molecules. In this mouse model, the almost complete lack of dendritic processes did not appear to affect viability or density of osteocytes in contrast to studies by Zhao and coworkers,(14) where osteocytes in a mouse model of collagenase-resistant type I collagen did show increased apoptosis. However, it was impossible to determine the effect of a lack of dendritic processes on either osteocyte function or the effect(s) on the skeleton because the MT1-MMP null mouse exhibits multiple defects such as dwarfism owing to a lack of MT1-MMP in other skeletal tissues.(15)

Osteocyte morphology may be controlled by E11/gp38/podoplanin, a marker for the embedding osteoid osteocyte (Fig. 2). E11, also called podoplanin, OTS-8, gp38, or PA2.25, was first detected on the cell surface of osteocytes in rat bone(16,17) and odontoblasts in rat tooth.(18) It is also expressed in type I cells of rat lung and other tissues of brain, kidney, the lymphatic system, and skin. Application of fluid-flow shear stress on osteocyte-like MLO-Y4 cells increased the number and length of dendrites and was blocked by small interfering RNA against E11/gp38.(19) Conditional deletion in bone cells in vivo resulted in decreased canaliculi and increased trabecular bone.(20) In addition to E11, organized expression of tubulin, vimentin, and actin in cell bodies and dendrites of osteocytes is crucial to maintain their dendritic morphology.(21) Differences in distribution of fimbrin, villin, filamin, and spectrin, all actin-binding proteins, have been described accompanying the differentiation of osteoblasts to osteocytes,(22) as well as CapG and destrin, molecules necessary for cytoskeletal rearrangement.(23) Intriguingly, two groups of investigators have shown an increase in number of dendritic processes with skeletal age, suggesting that the already embedded osteocyte can either generate new processes or, alternatively, the newly embedded cells have a greater number of dendrites.(13,24)

Fig. 2.

Fig. 2

Visualization of early, embedding osteocytes. Using anti-E11 immunostaining and visualization of the actin cytoskeleton by alexa488 staining for phalloidin, one can visualize the embedding osteocyte and the early osteocyte in 12-day murine calvaria. The merged image shows that the majority of the E11 is on the cell surface and along the dendritic processes. Also, if one looks closely, the dendrites that end on the cell surface have a bulbous tip of unknown function. This structure must interface with the cells on the bone surface. The image is provided by Dr Sarah Dallas, University of Missouri at Kansas City. The second image is of an acid-etched resin-embedded murine sample showing an osteocyte lacuna sending canaliculi to the bone surface. Note the rough surfaces of canaliculi toward the bone surface and the smooth surface of canaliculi that project away from the bone surface, suggesting a difference between forming and formed canaliculi. Both sets of images demonstrate the complexity of this network and the interface of osteocytes with the bone surface.

As the osteoblast transitions to an osteocyte, alkaline phosphatase is reduced, and casein kinase II is elevated, as is osteocalcin.(25) Additional markers are expressed, including phosphate-regulating gene with homologies to endopeptidases on the X chromosome (PHEX), matrix extracellular phosphoglycoprotein (MEPE), dentin matrix protein 1 (DMP-1), fibroblast growth factor 23, (FGF-23), sclerostin, and ORP150, a factor thought to protect against hypoxia. The function of these molecules will be discussed below. Surprisingly, osteocytes also can express markers of osteoclasts, such as acid phosphatase and cathepsin K, under certain conditions such as lactation to remodel their perilacunar matrix.(26) These markers, as discussed in this review, show the versatility of this bone cell.

Osteocytes as Inducers of Osteoclast Activation

The earliest data supporting the concept that osteocytes can send signals of bone resorption were gathered using isolated avian osteocytes showing support of osteoclast formation and activation in the absence of any osteotropic factors.(27) These observations were later duplicated using the osteocyte-like cell line MLO-Y4.(28) RANK ligand (RANKL) expression on dendritic processes appeared to be responsible. Conditioned medium from the MLO-Y4 cells also supports osteoblast differentiation(29) and, surprisingly, mesenchymal stem cell differentiation,(30) supporting the theory that osteocytes could be orchestrators of bone remodeling.

Both healthy and dying osteocytes can recruit osteoclasts to sites of remodeling. Osteocyte apoptosis can occur at sites of microdamage. Proapoptotic molecules are elevated in osteocytes immediately at the microcrack locus, whereas antiapoptotic molecules are expressed 1 to 2 mm from the microcrack,(31) showing that some osteocytes have protective mechanisms against apoptosis. Apoptotic osteocytes release apoptotic bodies expressing RANKL to recruit osteoclasts.(32) There are different forms of “dying,” such as apoptosis and necrosis, and osteocytes in these states may send different signals. Targeted ablation of osteocytes through necrosis was performed using the 10-kb Dmp1 promoter to drive the diptheria toxin receptor expression in osteocytes.(33) Injection of a single dose of diphtheria toxin eliminated approximately 70% of osteocytes in cortical bone and generated osteoclast activation in these mice. Another model of defective osteocytes was the deletion of β-catenin in osteocytes(34) (Fig. 3). Clearly β-catenin is necessary for normal osteocyte function, and deletion results is an increase in osteoclast activity and the generation of a porous bone phenotype. Osteocyte cell death and cell survival are reviewed below.

Fig. 3.

Fig. 3

The Wnt/β-catenin pathway plays an important role in osteocyte function and viability and the maintenance of normal bone. Deletion of β-catenin in osteocytes results in bone with a “moth-eaten appearance.” The calvaria shown are from 14-week-old female control and conditional knockout (cKO) mice in which β-catenin is deleted using a Dmp1-Cre. The long bones from these animals show even greater porosity and fragility, thought to be responsible for death of these transgenic animals at an early age.(34) This bone porosity was due to increased osteoclast number and activity most likely owing to reduced expression of osteoprotegerin and an increase in RANKL, both found to be expressed in osteocytes. These observations support the role of β-catenin in osteocyte viability. The diagram integrates a number of observations regarding how mechanical loading in the form of fluid-flow shear stress regulates osteocyte viability,(40) function.(34) and communication(59) through the Wnt/β-catenin pathway. The unique triggering or crosstalk between prostaglandin and this pathway has been detailed previously.(58)

The Importance of Osteocyte Death

Osteocyte cell death can occur in association with such pathologic conditions as osteoporosis and osteoarthritis, leading to increased skeletal fragility.(35) Such fragility is considered to be due to loss of the ability to sense microdamage and/or signal repair. Oxygen deprivation, such as occurs during immobilization, has been shown to promote osteocyte apoptosis,(36) as does glucocorticoid treatment(35) and withdrawal of estrogen.(37,38) Tumor necrosis factor α (TNF-α) and interleukin 1 (IL-1) have been reported to increase with estrogen deficiency and also induce osteocyte apoptosis (for review, see ref. (39)).

Inhibitors of osteocyte cell death include estrogen and selective estrogen receptor modulators, bisphosphonates, calcitonin, CD40 ligand, calbindin-D28k, and monocyte chemotactic proteins 1 and 3 (MCP1 and MCP3; for review, see ref. (39)). Recently, mechanical loading in the form of fluid-flow shear stress to mimic bone fluid flow in the osteocyte lacunocanalicular network has been shown to block glucocorticoid-induced apoptosis.(40) This was shown to be mediated through the release of prostaglandin, which activated the Wnt/β-catenin pathway. Alternatively, it also has been postulated that the reason for the opposite effects of mechanical loading and glucocorticoid on apoptosis of osteocytes is their opposing actions on the family of focal adhesion kinases (FAKs) and the proline-rich tyrosine kinase 2 (Pyk2). These investigators used substrate stretching as a form of mechanical loading to prevent apoptosis and showed activation of FAKs and extracellular signals–regulated kinases (ERKs).(41) It also was proposed that glucocorticoids oppose FAK/ERK signaling through Pyk2 and c-Jun N-terminal kinase (JNK)(42) and that extracellular matrix (ECM)–integrin/FAK signaling is linked with the the Wnt/β-catenin pathway.(43) Therefore, whether mechanical loading is applied through fluid-flow shear stress or through substrate stretching, the Wnt/β-catenin pathway appears to be involved.

In addition to undergoing programmed cell death, osteocytes can undergo a process of self-preservation called autophagy, especially in response to glucocorticoid.(44) Autophagy is a lysosomal degradation process necessary for recycling of cellular products. During autophagy, parts of the cytoplasm and intracellular organelles are localized within autophagic vacuoles for delivery to lysosomes for degradation. Autophagy has been proposed as a “double-edged sword” that can protect cells from apoptosis but, on the other hand, can destroy cellular components. Autophagy can preserve viability or, alternatively, can be a self-destructive process that leads to cell death. Therefore, in addition to responding to external agents or stimuli with necrosis and apoptosis, osteocytes can undergo autophagy to preserve self until favorable conditions return.

Osteocyte viability clearly plays a significant role in the maintenance of bone homeostasis and integrity. However, whereas blocking osteocyte apoptosis may improve diseases such as bone loss owing to aging or to glucocorticoid therapy, osteocyte apoptosis may be essential for damage repair and normal skeletal replacement. Any agents that block this process may exacerbate conditions in which repair is required.

The Role of the Wnt/β-Catenin Pathway in Osteocyte Function and Viability

Negative regulators of the Wnt/β-catenin pathway such as Dkk1 and sclerostin are highly expressed in osteocytes. Dkk1 is expressed throughout the body, but sclerostin is expressed mainly in osteocytes. Mature osteocytes have been shown to produce sclerostin, coded by the gene SOST, that can inhibit osteoblast activity.(45) Downregulation of Dkk1 and SOST may create a permissive environment in which Wnt proteins already present can activate the Wnt pathway. Mutations of SOST causes high bone mass in humans,(46) as does deletion in mice.(47) Sclerostin was thought to be a bone morphogenetic protein (BMP) antagonist but later was discovered to be involved in the Wnt pathway as an antagonist against lipoprotein receptor 5 (LRP5), a positive regulator of bone mass.(48,49) Mechanical loading also has been reported to reduce sclerostin expression,(50) as does parathyroid hormone (PTH), which may account for some of the anabolic effect of PTH on bone formation.(51,52) PTH also appears to reduce sclerostin levels in patients.(53) Bone response to unloading was proposed to be due to elevated sclerostin.(50,54) Antibody to sclerostin is being considered as a new drug against postmenopausal osteoporosis(55,56) because of its specificity and its anabolic effect on bone formation. Other potential uses include disuse-induced bone loss, bone repair, fracture healing, and accelerated implant fixation. This molecule, more than any other, has served to raise interest in the osteocyte.

Mechanical loading of MLO-Y4 cells by fluid-flow shear stress protects against dexamethasone-induced apoptosis, and the mechanism for this protective effect of mechanical loading appears to be partially mediated through prostaglandin E2 (PGE2) crosstalk with β-catenin signaling.(40) Both PGE2 and fluid-flow shear-stress treatment result in increased phosphorylation of GSK-3β and β-catenin nuclear translocation.(57) Shear stress, through PGE2 release, activates both PI3K/Akt and cAMP-PKA signaling, which converge to inactivate GSK-3, leading to the increase in nuclear accumulation of β-catenin. Therefore, the Wnt/β-catenin signaling pathway plays a role not only in bone response to loading but also in osteocyte apoptosis.(58) β-Catenin also has been shown to bind to the Cx43 promoter, stimulating Cx43 expression and functional gap junctions between osteocytes.(59) Therefore, in addition to playing a role in osteocyte viability in response to shear stress, the β-catenin pathway is important in osteocyte communication (Fig. 3).

Other signaling pathways are activated in response to mechanical loading and also may cross talk with the Wnt/β-catenin signaling pathway. The estrogen receptor α isoform (ERα) may play a role in shuttling β-catenin into the nucleus in response to mechanical strain in osteoblasts.(60) This may explain in part how estrogen regulates bone mass through crosstalk between ERα with Wnt/β-catenin signaling. Therefore, multiple signaling pathways such as the estrogen and prostaglandin signaling pathways may converge through β-catenin.

As stated earlier, targeted deletion of β-catenin in osteocytes results in a bone phenotype with a “moth-eaten appearance,” clearly showing the importance of this pathway in the maintenance of normal bone(34) (Fig. 3). These published studies showed that osteocyte β-catenin is required for the expression of the antiosteoclastogenic factor osteoprotegerin (OPG) in osteocytes. Surprisingly, it was found that osteocytes express RANKL and OPG at levels comparable with or exceeding those of osteoblasts. These data support previous findings that osteocytes recruit osteoclasts and that osteocytes are key regulators of bone homeostasis.

Osteocytes as a Source of Factors and Regulators of Mineral Metabolism

Osteocyte-specific or highly selective proteins have been shown to function in mineral metabolism. One of these is sclerostin, which was described earlier. Osteocytes appear to also regulate phosphate and biomineralization through molecules such as PHEX, DMP-1, MEPE, and FGF-23.(61) All of these are highly expressed in osteocytes.(6264) Autosomal recessive hypophosphatemic rickets in patients is due to mutations in DMP1.(65) Dmp1 null mice have a similar phenotype to Hyp mice carrying a Phex mutation, that of osteomalacia and rickets owing to elevated FGF-23 levels in osteocytes.(64,65) Both Dmp1 and Phex appear to downregulate FGF-23 expression, which, in turn, allows reabsorption of phosphate by the kidney, thereby maintaining sufficient circulating phosphate to maintain normal bone mineral content. In the absence of either Dmp1 or Phex, FGF-23 is elevated in the osteocyte and in the circulation, leading to phosphate excretion by the kidney, thereby reducing circulating phosphate responsible for osteomalacia and rickets. Based on these observations, my colleagues and I proposed that the osteocyte lacunocanalicular network can function as an endocrine system, targeting distant organs such as kidney.(65)

FGF-23 may be more than just a regulator of renal phosphate handling.(66) In addition to hypophosphatemic rickets, FGF-23 is elevated in osteocytes in patients with chronic kidney disease (CKD).(67) Progression in CKD is associated with elevated FGF-23.(68) Also, increased death with cardiovascular disease is associated with elevated FGF-23, as is calcification.(69) It will be important to determine if this osteocyte-derived factor has a direct effect on tissues other than the kidney. The unraveling of these interactions should lead to insight into diseases of both hyper- and hypophosphatemia, in addition to other diseases such as CKD and cardiovascular disease.

Osteocytic Remodeling of the Perilacunar Matrix

Since the surface area of the osteocyte lacunocanalicular system within bone is several orders of magnitude greater than the area of the bone surface,(70) removal of only a few angstroms of mineral per osteocyte would have significant effects on circulating, systemic ion levels. Belanger coined the term osteocytic osteolysis and suggested that either PTH or low-calcium diet can induce this function in osteocytes.(2,3) Enlarged lacunae were found in bone from renal osteodystrophy patients,(71) in rats sent into space,(72) and in alveolar bone of hibernating ground squirrels.(73) Periosteocytic lesions in patients with X-linked hypophosphatemic rickets have been described,(74) as has a sphere of hypomineralized matrix around lacunae in prednisolone-treated rats.(75) Changes in properties of perilacunar bone matrix and lacunar size would influence fracture risk, and any mechanism that changes the material properties of the perilacunar matrix will have consequences on mechanosensation by osteocytes.(76,77)

Conversely, in 1971, Baylink showed tetracycline binding to the osteocyte perilacunar matrix, which led him to suggest that osteocytes have the ability to form bone.(4) Later, Zambonin-Zallone and coworkers used the egg-laying hen and several methods, including autoradiography and tetracycline labeling, to show that at least 20% of the osteocytes are active in bone formation.(78) Both these early observations support the hypothesis that osteocytes can form new perilacunar matrix.

During lactation, osteocyte lacunae are enlarged significantly in both cortical and trabecular bone compared with virgin and postweaned animals,(79) showing that healthy osteocytes can remove and replace their perilacunar matrix and potentially play a role in mineral homeostasis during a calcium-demanding condition such as lactation. Additionally, the widths of canaliculi were found to be significantly increased. Gene array analysis of osteocytes from lactating animals revealed an elevation of genes products known to be used by osteoclasts to remove bone, including tartrate-resistant acid phosphatase, cathepsin K, and others, and these returned to levels similar to virgin gene expression levels on weaning. These observations suggest that the healthy osteocyte can both add and remove mineral from its lacunae and canaliculi. Therefore, the osteocyte is a unique cell expressing genes of both the mesenchymal and hemapoietic lineage. This remodeling was mediated through the PTH type 1 receptor because lactating animals lacking this gene in osteocytes failed to remodel their perilacunar matrix.(26) Targeted expression of constitutively active PTH type 1 receptor in osteocytes results in elevated bone remodeling and elevated bone mass.(80) Therefore, the PTH type 1 receptor most likely plays an important role is osteocyte viability and function.

In contrast to the young, healthy osteocyte within a hypomineralized matrix, the aging osteocyte may be subjected to hypermineralization of its perilacunar matrix,(81) and micropetrosis has been described in aging bone, where the lacunae fill in with mineral. This hypermineralization may lead to cell death with age, which, in turn, leads to the filling in of the lacunae with mineral. This would change the dynamics of bone fluid flow through the osteocyte lacunocanalicular network dramatically, potentially affecting osteocyte function and viability.

Osteocytes as Mechanosensory Cells

Another early postulated function for osteocytes is as a mechanosensory cell because of their location in bone and their complex dendritic network. The lining cell also has been hypothesized to be a major mechanosensory cell in the adult skeleton, but little is known about this type of bone cell. In vivo, the ideal frequency, intensity, and timing of loading are known that will increase bone mass.(8284) Whereas parameters of in vivo loading are well characterized, a major challenge has been to identify in vitro experiments that replicate in vivo results. The fact that mechanical loading and unloading change osteocyte gene expression in vivo shows that load affects osteocyte function.(50,8587)

Little is known about the bone fluid that flows through the osteocyte lacunocanalicular system, except that a molecular weight/mass cutoff of 70 kDa, the size of bovine serum albumin (BSA), exists.(88) Injection of dye into the tail vein of a mouse results in complete diffusion through the lacunocanalicular system within minutes. It has been proposed that bone fluid flow is driven by extravascular pressure as well as applied cyclic mechanical loading of osteocytes and that the peak physiologic loads are in the range of 8 to 30 dyn/cm2.(89) Whereas bone loss owing to hind limb unloading is restored with restored blood flow,(90) blood pressure does not play a role in regulating bone mass.(91) Recently, real-time measurement of load-induced solute transport has been shown, suggesting a peak shear stress on osteocyte processes of 5 Pa.(92) However, there are still many unanswered questions with regard to magnitude, frequency, and type of flow, such as pulsatile or oscillatory, to which the osteocyte may be subjected.

Early in vitro experiments used hydrostatic pressure and substrate stretching, whereas now fluid-flow shear stress is used because primary osteocytes are more sensitive than osteoblasts and more sensitive to shear stress than to substrate stretching.(93,94) MLO-Y4 osteocyte-like cells are several orders of magnitude more sensitive to fluid-flow shear stress with regard to release of prostaglandin than 2T3 osteoblast-like cells.(57) Shear stress has many effects on MLO-Y4 cells, including the release of nitric oxide, adenosine triphosphate (ATP), and prostaglandins; opening of hemichannels and gap junctions; promotion of dendrite elongation; bending of cilia; prevention of apoptosis; initiation of signaling pathways such as the Wnt/β-catenin, protein kinase A (PKA), and other signaling pathways; induction of β-catenin translocation to the nucleus; activation of gene transcription and translation; etc.—and the list continues to grow. To validate observations using cell lines, primary osteocytes can be prepared by sequential alternating collagenase digestions with EDTA,(7,95) but the yields continue to be low, especially with increasing age. Mice in which the 8-kb Dmp1 promoter, driving green fluorescent protein (GFP) expression in osteocytes, is active have been used to obtain a highly purified GFP-positive population.(96) New transgenics are being made with labeled osteocytes such as the Sost promoter driving GFP, which will allow tracking of isolated mature osteocytes.

The osteocyte may have several means to sense load, such as through the cell body, the dendritic processes, or bending of cilia.(97) It has been proposed that the osteocyte senses load only through its processes(98) or through both the cell body and the processes.(76) Recently, it has been shown that glycocalyces on the surfaces of dendritic processes, but not on the cell body, play an essential role in mechanotransduction, whereas another, different mechanosensing mechanism is active on the cell body.(99) It also has been proposed that the osteocyte senses load through cilia, single flagellar-like structures found on every cell.(100,101) Unlike kidney cells, cilia in bone cells do not mediate calcium flux in response to fluid flow but do induce the release of prostaglandin.(101) Mice with impaired polycystin 1 (PC-1) function develop osteopenia owing to impaired osteoblast-mediated bone formation.(100) Mice with conditional deletion of Pkd1 were developed using DMP-1-Cre, and such mice are smaller than controls at 1 month, but only minor differences were observed in 16-week-old mice, showing a recovery of bone mass. However, a dramatic decrease in response to anabolic loading was observed.(102) This shows that PC-1 in osteocytes is essential for the bone anabolic response to load.

Rapid osteocyte signals in response to shear stress include release of nitric oxide (NO), ATP, and prostaglandin. Just deleting only one of the three rapidly released small molecules will inhibit the bone anabolic response to loading. NO is a mechanical mediator that appears to be released around the same time as PGE2 from osteocytes,(93) and endothelial NO synthase is found in osteocytes.(103) In bone, NO inhibits resorption and promotes bone formation. Both osteoblasts and osteocytes release NO in response to mechanical strain or fluid-flow shear stress.(104) ATP and intracellular calcium also can be released from osteocytes in response to extracellular calcium or mechanical stimulation.(105,106) The P2X7 nucleotide receptor, an ATP-gated ion channel expressed in many cell types, may play a role in mechanosensation because deletion resulted in a 70% reduction in bone anabolic response in mice.(107) Blockers of P2X7 receptors suppressed prostaglandin release, whereas agonists enhanced release in bone cells, suggesting that the P2X7 receptor is necessary for release of prostaglandin in response to mechanical load.

In vivo prostaglandin induces new bone formation, and indomethacin blocks the effects of anabolic loading.(108) Prostaglandin appears to be released through hemichannels, unopposed halves of gap-junction channels, in response to shear stress.(109) Hemichannels are opened through association with perturbed integrin in response to shear stress and the glycocalyces of the osteocyte dendritic processes are necessary for integrin attachment to hemichannels.(99) Hemichannels expressed in MLO-Y4 cells also function as transducers of the antiapoptotic effects of bisphosphonates.(110) Therefore, hemichannels in osteocytes have multiple functions, including the release of signaling factors and protection of cell viability (for review, see refs. (39) and (111)).

What's Next?

Where will the next decade or two take us with regard to osteocyte biology and function? Will other osteocyte factors be discovered that can be used for drug targeting and potential therapeutics? Will additional factors be discovered that have targeted effects on distant organs? Bone frequently has been referred to as a storehouse of factors, and this usually refers to factors within the bone matrix. But what if the osteocyte lacunar network is a storehouse for regulatory factors rapidly released—unlike the bone matrix, which requires some degradation. Preserving the health and function of this network becomes imperative.

Acknowledgments

The author's work is supported by the National Institutes of Health NIAMS AR-46798 and RC2-AR058962.

Disclosures

The author states that she has no conflicts of interest.

References

  • 1.Recklinghausen FV. Untersuchungen uber Rachitis und Osteomalacia. Jena: Gustav Fischer; 1920. [Google Scholar]
  • 2.Belanger LF, Robichon J. Parathormone-induced osteolysis in dogs: a microradiographic and alpharadiographic survey. J Bone Joint Surg Am. 1964;46:1008–1012. [PubMed] [Google Scholar]
  • 3.Belanger LF. Osteocytic osteolysis. Calcif Tissue Res. 1969;4:1–12. doi: 10.1007/BF02279101. [DOI] [PubMed] [Google Scholar]
  • 4.Baylink DJ, Wergedal JE. Bone formation by osteocytes. Am J Physiol. 1971;221:669–678. doi: 10.1152/ajplegacy.1971.221.3.669. [DOI] [PubMed] [Google Scholar]
  • 5.Pead MJ, Suswillo R, Skerry TM, Vedi S. Lanyon LE. Increased 3H-uridine levels in osteocytes following a. single short period of dynamic bone loading in vivo. Calcif Tissue Int. 1988;43:92–96. doi: 10.1007/BF02555153. [DOI] [PubMed] [Google Scholar]
  • 6.Marotti G, Cane V, Palazzini S, Palumbo C. Structure-function relationships in the osteocyte. Ital. J. Min. Electrolyte Metab. 1990;4:93–106. [Google Scholar]
  • 7.van der Plas A, Nijweide PJ. Isolation and purification of osteocytes. J Bone Miner Res. 1992;7:389–396. doi: 10.1002/jbmr.5650070406. [DOI] [PubMed] [Google Scholar]
  • 8.Tanaka K, Matsuo T, Ohta M, et al. Time-lapse microcinematography of osteocytes. Miner Electrolyte Metab. 1995;1995:189–192. [PubMed] [Google Scholar]
  • 9.Manolagas SC. Birth and death of bone cells: basic regulatory mechanisms and implications for the pathogenesis and treatment of osteoporosis. Endocr Rev. 2000;21:115–137. doi: 10.1210/edrv.21.2.0395. [DOI] [PubMed] [Google Scholar]
  • 10.Parfitt AM. Bone-forming cells in Clinical Conditions. In: BK H, editor. Bone. The Osteoblast and Osteocyte. Vol. 1. Boca Raton, Fl: Telford Press and CRC Press; 1990. pp. 351–429. [Google Scholar]
  • 11.Franz-Odendaal TA, Hall BK, Witten PE. Buried alive: how osteoblasts become osteocytes. Dev Dyn. 2006;235:176–190. doi: 10.1002/dvdy.20603. [DOI] [PubMed] [Google Scholar]
  • 12.Barragan-Adjemian C, Nicolella D, Dusevich V, Dallas MR, Eick JD, Bonewald LF. Mechanism by which MLO-A5 late osteoblasts/early osteocytes mineralize in culture: similarities with mineralization of lamellar bone. Calcif Tissue Int. 2006;79:340–353. doi: 10.1007/s00223-006-0107-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Holmbeck K, Bianco P, Pidoux I, et al. The metalloproteinase MT1-MMP is required for normal development and maintenance of osteocyte processes in bone. J Cell Sci. 2005;118:147–156. doi: 10.1242/jcs.01581. [DOI] [PubMed] [Google Scholar]
  • 14.Zhao W, Byrne MH, Wang Y, Krane SM. Osteocyte and osteoblast apoptosis and excessive bone deposition accompany failure of collagenase cleavage of collagen. J Clin Invest. 2000;106:941–949. doi: 10.1172/JCI10158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Holmbeck K, Bianco P, Caterina J, et al. MT1-MMP-deficient mice develop dwarfism, osteopenia, arthritis, and connective tissue disease due to inadequate collagen turnover. Cell. 1999;99:81–92. doi: 10.1016/s0092-8674(00)80064-1. [DOI] [PubMed] [Google Scholar]
  • 16.Wetterwald A, Hoffstetter W, Cecchini MG, et al. Characterization and cloning of the E11 antigen, a marker expressed by rat osteoblasts and osteocytes. Bone. 1996;18:125–132. doi: 10.1016/8756-3282(95)00457-2. [DOI] [PubMed] [Google Scholar]
  • 17.Schulze E, Witt M, Kasper M, Lowik CW, Funk RH. Immunohistochemical investigations on the differentiation marker protein E11 in rat calvaria, calvaria cell culture and the osteoblastic cell line ROS 17/2.8. Histochem Cell Biol. 1999;111:61–69. doi: 10.1007/s004180050334. [DOI] [PubMed] [Google Scholar]
  • 18.Schwab W, Schulze E, Witt M, Funk RH, Kasper M. Immunohistochemical localization of the differentiation marker E11 in dental development of rats. Acta Histochem. 1999;101:431–436. doi: 10.1016/S0065-1281(99)80043-9. [DOI] [PubMed] [Google Scholar]
  • 19.Zhang K, Barragan-Adjemian C, Ye L, et al. E11/gp38 selective expression in osteocytes: regulation by mechanical strain and role in dendrite elongation. Mol Cell Biol. 2006;26:4539–4552. doi: 10.1128/MCB.02120-05. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Guo D, Zhao H, Mishina Y, Feng J, Harris S, Bonewald L. Mice with Targeted Deletion of E11/gp38 in Late Osteoblasts Have Reduced Canaliculi per Osteocyte Which May Be Responsible for The Enhanced Trabecular Bone Volume. FR0268. J Bone Min Res. 2009:S126. [Google Scholar]
  • 21.Tanaka-Kamioka K, Kamioka H, Ris H, Lim SS. Osteocyte shape is dependent on actin filaments and osteocyte processes are unique actin-rich projections. J Bone Miner Res. 1998;13:1555–1568. doi: 10.1359/jbmr.1998.13.10.1555. [DOI] [PubMed] [Google Scholar]
  • 22.Kamioka H, Sugawara Y, Honjo T, Yamashiro T, Takano-Yamamoto T. Terminal differentiation of osteoblasts to osteocytes is accompanied by dramatic changes in the distribution of actin-binding proteins. J Bone Miner Res. 2004;19:471–478. doi: 10.1359/JBMR.040128. [DOI] [PubMed] [Google Scholar]
  • 23.Guo D, Keightley A, Guthrie J, Veno PA, Harris SE, Bonewald LF. Identification of osteocyte-selective proteins. Proteomics. 2010;10 doi: 10.1002/pmic.201000306. in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Okada S, Yoshida S, Ashrafi SH, Schraufnagel DE. The canalicular structure of compact bone in the rat at different ages. Microsc Microanal. 2002;8:104–115. doi: 10.1017/s1431927601020037. [DOI] [PubMed] [Google Scholar]
  • 25.Mikuni-Takagaki Y, Kakai Y, Satoyoshi M, et al. Matrix mineralization and the differentiation of osteocyte-like cells in culture. J Bone Miner Res. 1995;10:231–242. doi: 10.1002/jbmr.5650100209. [DOI] [PubMed] [Google Scholar]
  • 26.Qing H, Divieti Pajevic P, Barry K, Dusevich V, Wysolmerski J, Bonewald L. PTHR1 in Osteocytes Plays a Major role in Perilacunar Remodeling through the Activation of “Osteoclastic” Genes in Osteocytes. #1082J Bone Min Res. 2010;(Suppl):S25. [Google Scholar]
  • 27.Tanaka K, Yamaguchi Y, Hakeda Y. Isolated chick osteocytes stimulate formation and bone-resorbing activity of osteoclast-like cells. Journal of Bone and Mineral Metabolism. 1995;13:61–70. [Google Scholar]
  • 28.Zhao S, Zhang YK, Harris S, Ahuja SS, Bonewald LF. MLO-Y4 osteocyte-like cells support osteoclast formation and activation. J Bone Miner Res. 2002;17:2068–2079. doi: 10.1359/jbmr.2002.17.11.2068. [DOI] [PubMed] [Google Scholar]
  • 29.Heino TJ, Hentunen TA, Vaananen HK. Osteocytes inhibit osteoclastic bone resorption through transforming growth factor-beta: enhancement by estrogen. J Cell Biochem. 2002;85:185–197. doi: 10.1002/jcb.10109. [DOI] [PubMed] [Google Scholar]
  • 30.Heino TJ, Hentunen TA, Vaananen HK. Conditioned medium from osteocytes stimulates the proliferation of bone marrow mesenchymal stem cells and their differentiation into osteoblasts. Exp Cell Res. 2004;294:458–468. doi: 10.1016/j.yexcr.2003.11.016. [DOI] [PubMed] [Google Scholar]
  • 31.Verborgt O, Tatton NA, Majeska RJ, Schaffler MB. Spatial distribution of Bax and Bcl-2 in osteocytes after bone fatigue: complementary roles in bone remodeling regulation? J Bone Miner Res. 2002;17:907–914. doi: 10.1359/jbmr.2002.17.5.907. [DOI] [PubMed] [Google Scholar]
  • 32.Kogianni G, Mann V, Noble BS. Apoptotic bodies convey activity capable of initiating osteoclastogenesis and localized bone destruction. J Bone Miner Res. 2008;23:915–927. doi: 10.1359/jbmr.080207. [DOI] [PubMed] [Google Scholar]
  • 33.Tatsumi S, Ishii K, Amizuka N, et al. Targeted ablation of osteocytes induces osteoporosis with defective mechanotransduction. Cell Metab. 2007;5:464–475. doi: 10.1016/j.cmet.2007.05.001. [DOI] [PubMed] [Google Scholar]
  • 34.Kramer I, Halleux C, Keller H, et al. Osteocyte Wnt/beta-catenin signaling is required for normal bone homeostasis. Mol Cell Biol. 2010;30:3071–3085. doi: 10.1128/MCB.01428-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Weinstein RS, Nicholas RW, Manolagas SC. Apoptosis of osteocytes in glucocorticoid-induced osteonecrosis of the hip. J Clin Endocrinol Metab. 2000;85:2907–2912. doi: 10.1210/jcem.85.8.6714. [DOI] [PubMed] [Google Scholar]
  • 36.Dodd JS, Raleigh JA, Gross TS. Osteocyte hypoxia: a novel mechanotransduction pathway. Am J Physiol. 1999;277:C598–602. doi: 10.1152/ajpcell.1999.277.3.C598. [DOI] [PubMed] [Google Scholar]
  • 37.Tomkinson A, Reeve J, Shaw RW, Noble BS. The death of osteocytes via apoptosis accompanies estrogen withdrawal in human bone. J Clin Endocrinol Metab. 1997;82:3128–3135. doi: 10.1210/jcem.82.9.4200. [DOI] [PubMed] [Google Scholar]
  • 38.Emerton KB, Hu B, Woo AA, et al. Osteocyte apoptosis and control of bone resorption following ovariectomy in mice. Bone. 2010;46:577–583. doi: 10.1016/j.bone.2009.11.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Bonewald L. Osteocytes. In: Marcus DF R, Nelson D, Rosen C, editors. Osteoporosis. 3rd ed. Vol. 1. Elsevier; 2007. pp. 169–190. [Google Scholar]
  • 40.Kitase Y, Barragan L, Jiang JX, Johnson ML, Bonewald LF. Mechanical induction of PGE2 in osteocytes blocks glucocorticoid-induced apoptosis through both the β-catenin and PKA pathways. J Bone Miner Res. 2010;25 doi: 10.1002/jbmr.168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Plotkin LI, Mathov I, Aguirre JI, Parfitt AM, Manolagas SC, Bellido T. Mechanical stimulation prevents osteocyte apoptosis: requirement of integrins, Src kinases, and ERKs. Am J Physiol Cell Physiol. 2005;289:C633–643. doi: 10.1152/ajpcell.00278.2004. [DOI] [PubMed] [Google Scholar]
  • 42.Plotkin LI, Manolagas SC, Bellido T. Glucocorticoids induce osteocyte apoptosis by blocking focal adhesion kinase-mediated survival. Evidence for inside-out signaling leading to anoikis. J Biol Chem. 2007;282:24120–24130. doi: 10.1074/jbc.M611435200. [DOI] [PubMed] [Google Scholar]
  • 43.Bellido T. Antagonistic interplay between mechanical forces and glucocorticoids in bone: a tale of kinases. J Cell Biochem. 111:1–6. doi: 10.1002/jcb.22660. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Xia X, Kar R, Gluhak-Heinrich J, et al. Glucocorticoid-induced autophagy in osteocytes. J Bone Miner Res. 2010;25:2479–2488. doi: 10.1002/jbmr.160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Poole KE, van Bezooijen RL, Loveridge N, et al. Sclerostin is a delayed secreted product of osteocytes that inhibits bone formation. Faseb J. 2005;19:1842–1844. doi: 10.1096/fj.05-4221fje. [DOI] [PubMed] [Google Scholar]
  • 46.Balemans W, Ebeling M, Patel N, et al. Increased bone density in sclerosteosis is due to the deficiency of a novel secreted protein (SOST) Hum Mol Genet. 2001;10:537–543. doi: 10.1093/hmg/10.5.537. [DOI] [PubMed] [Google Scholar]
  • 47.Li X, Ominsky MS, Niu QT, et al. Targeted deletion of the sclerostin gene in mice restuls in increased bone formation and bone strength. J Bone Min Res. 2008;23:860–869. doi: 10.1359/jbmr.080216. [DOI] [PubMed] [Google Scholar]
  • 48.van Bezooijen RL, Roelen BA, Visser A, et al. Sclerostin is an osteocyte-expressed negative regulator of bone formation, but not a classical BMP antagonist. J Exp Med. 2004;199:805–814. doi: 10.1084/jem.20031454. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Li X, Zhang Y, Kang H, et al. Sclerostin binds to LRP5/6 and antagonizes canonical Wnt signaling. J Biol Chem. 2005;280:19883–19887. doi: 10.1074/jbc.M413274200. [DOI] [PubMed] [Google Scholar]
  • 50.Robling AG, Niziolek PJ, Baldridge LA, et al. Mechanical stimulation of bone in vivo reduces osteocyte expression of Sost/sclerostin. J Biol Chem. 2008;283:5866–5875. doi: 10.1074/jbc.M705092200. [DOI] [PubMed] [Google Scholar]
  • 51.Bellido T, Ali AA, Gubrij I, et al. Chronic elevation of parathyroid hormone in mice reduces expression of sclerostin by osteocytes: a novel mechanism for hormonal control of osteoblastogenesis. Endocrinology. 2005;146:4577–4583. doi: 10.1210/en.2005-0239. [DOI] [PubMed] [Google Scholar]
  • 52.Keller H, Kneissel M. SOST is a target gene for PTH in bone. Bone. 2005;37:148–158. doi: 10.1016/j.bone.2005.03.018. [DOI] [PubMed] [Google Scholar]
  • 53.Drake MT, Srinivasan B, Modder UI, et al. Effects of parathyroid hormone treatment on circulating sclerostin levels in postmenopausal women. J Clin Endocrinol Metab. 2010;95:5056–5062. doi: 10.1210/jc.2010-0720. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Lin C, Jiang X, Dai Z, et al. Sclerostin mediates bone response to mechanical unloading through antagonizing Wnt/beta-catenin signaling. J Bone Miner Res. 2009;24:1651–1661. doi: 10.1359/jbmr.090411. [DOI] [PubMed] [Google Scholar]
  • 55.Lewiecki EM. Emerging drugs for postmenopausal osteoporosis. Expert Opin Emerg Drugs. 2009;14:129–144. doi: 10.1517/14728210902766813. [DOI] [PubMed] [Google Scholar]
  • 56.Padhi D, Jang G, Stouch B, Fang L, Posvar E. Single-dose, placebo-controlled, randomized study of AMG 785, a sclerostin monoclonal antibody. J Bone Miner Res. 2011;26:19–26. doi: 10.1002/jbmr.173. [DOI] [PubMed] [Google Scholar]
  • 57.Kamel MA, Picconi JL, Lara-Castillo N, Johnson ML. Activation of beta-catenin signaling in MLO-Y4 osteocytic cells versus 2T3 osteoblastic cells by fluid flow shear stress and PGE(2): Implications for the study of mechanosensation in bone. Bone. 2010;47:872–881. doi: 10.1016/j.bone.2010.08.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Bonewald LF, Johnson ML. Osteocytes, Mechanosensing, and Wnt Signaling. Bone. 2008;42:606–615. doi: 10.1016/j.bone.2007.12.224. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Xia X, Batra N, Shi Q, Bonewald LF, Sprague E, Jiang JX. Prostaglandin promotion of osteocyte gap junction function through transcriptional regulation of connexin 43 by glycogen synthase kinase 3/beta-catenin signaling. Mol Cell Biol. 2010;30:206–219. doi: 10.1128/MCB.01844-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Zaman G, Jessop HL, Muzylak M, et al. Osteocytes use estrogen receptor alpha to respond to strain but their ERalpha content is regulated by estrogen. J Bone Miner Res. 2006;21:1297–1306. doi: 10.1359/jbmr.060504. [DOI] [PubMed] [Google Scholar]
  • 61.Bonewald LF. Osteocytes as Dynamic, Multifunctional Cells. Ann N Y Acad Sci. 2007;1116:281–290. doi: 10.1196/annals.1402.018. [DOI] [PubMed] [Google Scholar]
  • 62.Thompson DL, Sabbagh Y, Tenenhouse HS, et al. Ontogeny of Phex/PHEX protein expression in mouse embryo and subcellular localization in osteoblasts. J Bone Miner Res. 2002;17:311–320. doi: 10.1359/jbmr.2002.17.2.311. [DOI] [PubMed] [Google Scholar]
  • 63.Nampei A, Hashimoto J, Hayashida K, et al. Matrix extracellular phosphoglycoprotein (MEPE) is highly expressed in osteocytes in human bone. J Bone Miner Metab. 2004;22:176–184. doi: 10.1007/s00774-003-0468-9. [DOI] [PubMed] [Google Scholar]
  • 64.Liu S, Zhou J, Tang W, Jiang X, Rowe DW, Quarles LD. Pathogenic role of Fgf23 in Hyp mice. Am J Physiol Endocrinol Metab. 2006;291:E38–49. doi: 10.1152/ajpendo.00008.2006. [DOI] [PubMed] [Google Scholar]
  • 65.Feng JQ, Ward LM, Liu S, et al. Loss of DMP1 causes rickets and osteomalacia and identifies a role for osteocytes in mineral metabolism. Nat Genet. 2006;38:1310–1315. doi: 10.1038/ng1905. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Juppner H, Wolf M, Salusky IB. FGF23: more than a regulator of renal phosphate handling? J Bone Min Res. 2010;25:2091–2097. doi: 10.1002/jbmr.170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Pereira RC, Juppner H, Azucena-Serrano CE, Yadin O, Salusky IB, Wesseling-Perry K. Patterns of FGF-23, DMP1, and MEPE expression in patients with chronic kidney disease. Bone. 2009;45:1161–1168. doi: 10.1016/j.bone.2009.08.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Filser D, Kollerits B, Neyer U, et al. Fibroblast growth factor 23 (FGF23) predicts progression of chronic kidney disease: the Mild to Moderate Kidney Disease Study. J Am Soc Nephrol. 2007;18:2600–2608. doi: 10.1681/ASN.2006080936. [DOI] [PubMed] [Google Scholar]
  • 69.Gutierrez OM. Fibroblast Growth Factor 23 and Disordered Vitamin D Metabolism in Chronic Kidney Disease: Updating the “Trade-off” Hypothesis. Clin J Am Soc Nephrol. 2010;5:1710–1716. doi: 10.2215/CJN.02640310. [DOI] [PubMed] [Google Scholar]
  • 70.Marotti G, Ferretti M, Remaggi F, Palumbo C. Quantitative evaluation on osteocyte canalicular density in human secondary osteons. Bone. 1995;16:125–128. doi: 10.1016/s8756-3282(94)00019-0. [DOI] [PubMed] [Google Scholar]
  • 71.Bonucci E, Gherardi G, Mioni G, et al. [Clinico-morphological correlations in uremic osteodystrophy of patients with conservative and hemodialytic treatment with special regard to the ultrastructure] Minerva Nefrol. 1975;22:99–108. [PubMed] [Google Scholar]
  • 72.Iagodovskii VS, Triftanidi LA, Gorokhova GP. [Effect of space flight on rat skeletal bones (an optical light and electron microscopic study)] Kosm Biol Aviakosm Med. 1977;11:14–20. [PubMed] [Google Scholar]
  • 73.Haller AC, Zimny ML. Effects of hibernation on interradicular alveolar bone. J Dent Res. 1977;56:1552–1557. doi: 10.1177/00220345770560122601. [DOI] [PubMed] [Google Scholar]
  • 74.Marie PJ, Glorieux FH. Relation between hypomineralized periosteocytic lesions and bone mineralization in vitamin D-resistant rickets. Calcif Tissue Int. 1983;35:443–448. doi: 10.1007/BF02405074. [DOI] [PubMed] [Google Scholar]
  • 75.Lane NE, Yao W, Balooch M, et al. Glucocorticoid-treated mice have localized changes in trabecular bone material properties and osteocyte lacunar size that are not observed in placebo-treated or estrogen-deficient mice. J Bone Miner Res. 2006;21:466–476. doi: 10.1359/JBMR.051103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Nicolella DP, Feng JQ, Moravits DE, et al. Effects of nanomechanical bone tissue properties on bone tissue straimplications for osteocyte mechanotransduction. J Musculoskelet Neuronal Interact. 2008;8:330–331. [PMC free article] [PubMed] [Google Scholar]
  • 77.Nicolella DP, Moravits DE, Gale AM, Bonewald LF, Lankford J. Osteocyte lacunae tissue strain in cortical bone. J Biomech. 2006;39:1735–1743. doi: 10.1016/j.jbiomech.2005.04.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Zambonin Zallone A, Teti A, Primavera MV, Pace G. Mature osteocytes behaviour in a repletion period: the occurrence of osteoplastic activity. Basic Appl Histochem. 1983;27:191–204. [PubMed] [Google Scholar]
  • 79.Qing H, Ardeshirpour L, Dusevich V, Wysolmerski J, Bonewald LF. Osteocyte Perilacunar Remodeling Is Regulated Hormonally, but Not by Mechanical Unloading. Journal of Bone & Mineral Research. 2009;(supple 1):Mo0255. [Google Scholar]
  • 80.O'Brien CA, Plotkin LI, Galli C, et al. Control of bone mass and remodeling by PTH receptor signaling in osteocytes. PLoS One. 2008;3:e2942. doi: 10.1371/journal.pone.0002942. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Potter R, Miller M, Moravits D, et al. Raman spectroscopic characterization of bone tissue material properties around the osteocyte lacuna: effect of aging. J Bone Miner Res. 2009;(suppl 1):Su0266. [Google Scholar]
  • 82.Rubin C. Skeletal strain and the functional significance of bone architecture. Calcif Tissue Int. 1984;36:S11–S18. doi: 10.1007/BF02406128. [DOI] [PubMed] [Google Scholar]
  • 83.Turner CH, Forwood MR, Otter MW. Mechanotransduction in bone: do bone cells act as sensors of fluid flow? Faseb J. 1994;8:875–878. doi: 10.1096/fasebj.8.11.8070637. [DOI] [PubMed] [Google Scholar]
  • 84.Robling AG, Hinant FM, Burr DB, Turner CH. Shorter, more frequent mechanical loading sessions enhance bone mass. Med Sci Sports Exerc. 2002;34:196–202. doi: 10.1097/00005768-200202000-00003. [DOI] [PubMed] [Google Scholar]
  • 85.Skerry TM, Bitensky L, Chayen J, Lanyon LE. Early strain-related changes in enzyme activity in osteocytes following bone loading in vivo. J Bone Miner Res. 1989;4:783–788. doi: 10.1002/jbmr.5650040519. [DOI] [PubMed] [Google Scholar]
  • 86.Gluhak-Heinrich J, Pavlin D, Yang W, MacDougall M, Harris SE. MEPE expression in osteocytes during orthodontic tooth movement. Arch Oral Biol. 2007;52:684–690. doi: 10.1016/j.archoralbio.2006.12.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Gluhak-Heinrich J, Ye L, Bonewald LF, et al. Mechanical loading stimulates dentin matrix protein 1 (DMP1) expression in osteocytes in vivo. J Bone Miner Res. 2003;18:807–817. doi: 10.1359/jbmr.2003.18.5.807. [DOI] [PubMed] [Google Scholar]
  • 88.Wang L, Ciani C, Doty SB, Fritton SP. Delineating bone's interstitial fluid pathway in vivo. Bone. 2004;34:499–509. doi: 10.1016/j.bone.2003.11.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Weinbaum S, Cowin SC, Zeng Y. A model for the excitation of osteocytes by mechanical loading-induced bone fluid shear stresses. J Biomech. 1994;27:339–360. doi: 10.1016/0021-9290(94)90010-8. [DOI] [PubMed] [Google Scholar]
  • 90.Bergula AP, Huang W, Frangos JA. Femoral vein ligation increases bone mass in the hindlimb suspended rat. Bone. 1999;24:171–177. doi: 10.1016/s8756-3282(98)00165-3. [DOI] [PubMed] [Google Scholar]
  • 91.Li W, Gardinier JD, Price C, Wang L. Does blood pressure enhance solute transport in the bone lacunar-canalicular system? Bone. 2010;47:353–359. doi: 10.1016/j.bone.2010.05.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Price C, Zhou X, Li W, Wang L. Real-time measurement of solute transport within the lacunar-canalicular system of mechanically loaded bone: Direct evidence for load-induced fluid flow. J Bone Miner Res. 2011;26:277–285. doi: 10.1002/jbmr.211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Klein-Nulend J, van der Plas A, Semeins CM, et al. Sensitivity of osteocytes to biomechanical stress in vitro. Faseb J. 1995;9:441–445. doi: 10.1096/fasebj.9.5.7896017. [DOI] [PubMed] [Google Scholar]
  • 94.Klein-Nulend J, Semeins CM, Ajubi NE, Nijweide PJ, Burger EH. Pulsating fluid flow increases nitric oxide (NO) synthesis by osteocytes but not periosteal fibroblasts--correlation with prostaglandin upregulation. Biochem Biophys Res Commun. 1995;217:640–648. doi: 10.1006/bbrc.1995.2822. [DOI] [PubMed] [Google Scholar]
  • 95.Mikuni-Takagaki Y, Suzuki Y, Kawase T, Saito S. Distinct responses of different populations of bone cells to mechanical stress. Endocrinology. 1996;137:2028–2035. doi: 10.1210/endo.137.5.8612544. [DOI] [PubMed] [Google Scholar]
  • 96.Kalajzic I, Braut A, Guo D, et al. Dentin matrix protein 1 expression during osteoblastic differentiation, generation of an osteocyte GFP-transgene. Bone. 2004;35:74–82. doi: 10.1016/j.bone.2004.03.006. [DOI] [PubMed] [Google Scholar]
  • 97.Bonewald LF. Mechanosensation and Transduction in Osteocytes. Bonekey Osteovision. 2006;3:7–15. doi: 10.1138/20060233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Han Y, Cowin SC, Schaffler MB, Weinbaum S. Mechanotransduction and strain amplification in osteocyte cell processes. Proc Natl Acad Sci U S A. 2004;101:16689–16694. doi: 10.1073/pnas.0407429101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Burra S, Nicolella DP, Francis WL, et al. Dendritic processes of osteocytes are mechanotransducers that induce the opening of hemichannels. Proc Natl Acad Sci U S A. 2010;107:13648–13653. doi: 10.1073/pnas.1009382107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Xiao Z, Zhang S, Mahlios J, et al. Cilia-like structures and polycystin-1 in osteoblasts/osteocytes and associated abnormalities in skeletogenesis and Runx2 expression. J Biol Chem. 2006;281:30884–30895. doi: 10.1074/jbc.M604772200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Malone AM, Anderson CT, Tummala P, et al. Primary cilia mediate mechanosensing in bone cells by a calcium-independent mechanism. Proc Natl Acad Sci U S A. 2007;104:13325–13330. doi: 10.1073/pnas.0700636104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Xiao Z, Dallas M, Zhang S, et al. Conditional deletion and/or disruption of Pkd1 in osteocytes results in a significant reduction in anabolic response to mechanical loading. J Bone Miner Res. 2009;(suppl1):#1042. [Google Scholar]
  • 103.Zaman G, Pitsillides AA, Rawlinson SC, et al. Mechanical strain stimulates nitric oxide production by rapid activation of endothelial nitric oxide synthase in osteocytes. J Bone Miner Res. 1999;14:1123–1131. doi: 10.1359/jbmr.1999.14.7.1123. [DOI] [PubMed] [Google Scholar]
  • 104.Bakker AD, Soejima K, Klein-Nulend J, Burger EH. The production of nitric oxide and prostaglandin E(2) by primary bone cells is shear stress dependent. J Biomech. 2001;34:671–677. doi: 10.1016/s0021-9290(00)00231-1. [DOI] [PubMed] [Google Scholar]
  • 105.Genetos DC, Geist DJ, Liu D, Donahue HJ, Duncan RL. Fluid shear-induced ATP secretion mediates prostaglandin release in MC3T3-E1 osteoblasts. J Bone Miner Res. 2005;20:41–49. doi: 10.1359/JBMR.041009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Kamioka H, Miki Y, Sumitani K, et al. Extracellular calcium causes the release of calcium from intracellular stores in chick osteocytes. Biochem Biophys Res Commun. 1995;212:692–696. doi: 10.1006/bbrc.1995.2024. [DOI] [PubMed] [Google Scholar]
  • 107.Li J, Liu D, Ke HZ, Duncan RL, Turner CH. The P2X7 nucleotide receptor mediates skeletal mechanotransduction. J Biol Chem. 2005;280:42952–42959. doi: 10.1074/jbc.M506415200. [DOI] [PubMed] [Google Scholar]
  • 108.Forwood MR. Inducible cyclo-oxygenase (COX-2) mediates the induction of bone formation by mechanical loading in vivo. J Bone Miner Res. 1996;11:1688–1693. doi: 10.1002/jbmr.5650111112. [DOI] [PubMed] [Google Scholar]
  • 109.Cherian PP, Siller-Jackson AJ, Gu S, et al. Mechanical strain opens connexin 43 hemichannels in osteocytes: a novel mechanism for the release of prostaglandin. Mol Biol Cell. 2005;16:3100–3106. doi: 10.1091/mbc.E04-10-0912. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Plotkin LI, Bellido T. Bisphosphonate-induced, hemichannel-mediated, anti-apoptosis through the Src/ERK pathway: a gap junction-independent action of connexin43. Cell Commun Adhes. 2001;8:377–382. doi: 10.3109/15419060109080757. [DOI] [PubMed] [Google Scholar]
  • 111.Jiang JX, Siller-Jackson AJ, Burra S. Roles of gap junctions and hemichannels in bone cell functions and in signal transmission of mechanical stress. Front Biosci. 2007;12:1450–1462. doi: 10.2741/2159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Ubaidus S, Li M, Sultana S, et al. FGF23 is mainly synthesized by osteocytes in the regularly distributed osteocytic lacunar canalicular system established after physiological bone remodeling. J Electron Microsc (Tokyo). 2009;58:381–392. doi: 10.1093/jmicro/dfp032. [DOI] [PubMed] [Google Scholar]

Articles from Journal of Bone and Mineral Research are provided here courtesy of Oxford University Press

RESOURCES