Hypoxia mediates osteocyte ORP150 expression and cell death in vitro

  • Authors:
    • Monica Montesi
    • Katharina Jähn
    • Lynda Bonewald
    • Susanna Stea
    • Barbara Bordini
    • Alina Beraudi
  • View Affiliations

  • Published online on: September 26, 2016     https://doi.org/10.3892/mmr.2016.5790
  • Pages: 4248-4254
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Skeletal unloading leads to hypoxia in the bone microenvironment, resulting in imbalanced bone remodeling that favors bone resorption. Osteocytes, the mechanosensors of bone, have been demonstrated to orchestrate bone homeostasis. Hypoxic osteocytes either undergo apoptosis or actively stimulate osteoclasts to remove bone matrix during hypoxia. Oxygen‑regulated protein 150 (ORP150) is an endoplasmic reticulum‑associated chaperone that has been observed to serve an important role in the cellular adaptation to hypoxia and in preventing cellular apoptosis in various tissue types. The current study hypotheses that expression of ORP150 would be increased in osteocytes under hypoxic conditions (1% O2). The MLO‑Y4 osteocyte cell line was cultured under normoxic or hypoxic conditions for up to 72 h. It was demonstrated that 1% O2 significantly induced hypoxia after 16 h and up to 72 h, significantly reduced cell number at 8 and 48 h, induced cell death at 8, 24 and 48 h and induced apoptosis at 16, 24 and 48 h. Significant differences in ORP150 mRNA were observed at 72 h, however no differences were observed in the protein expression levels. The relative increase in ORP150 mRNA observed in hypoxia, compared with normoxia, may support its cytoprotective role in oxygen‑deprived conditions.

Introduction

Bone remodeling is a fundamental mechanism for removing and replacing bone during skeleton adaptation to mechanical loads. Osteocytes are the most abundant cells in bone (13), and they are actively involved in the orchestration of both bone-forming osteoblasts and bone-resorbing osteoclasts (4,5). Osteocytes implement their role by activating the signaling pathways involved in the maintenance of bone homeostasis, specifically the Wnt/β-catenin pathway (6). This pathway has been demonstrated to be activated by mechanical loading to protect against bone loss (7) and to lead to the production of sclerostin, an inhibitor of osteoblast activity in response to unloading (8). It has been previously observed that osteocytes express receptor activator of nuclear factor κB ligand, a pre-osteoclastogenic cytokine involved in bone resorption activation (5,9,10). Osteocytes are known as the mechanosensor of bone (11,12) due to their location deep within the bone matrix and their dendritic network that allows the detection of variations in the levels of strain placed upon bone (4,13). Skeletal loading is fundamental in maintaining osteocyte viability (14), as demonstrated by increasing osteocyte apoptosis with disuse (15,16).

Skeletal unloading, as observed in patients undergoing bed rest, results in imbalanced bone remodeling that favors bone resorption (16,17). It has been suggested that the loss of bone observed during disuse is the result of osteocyte hypoxia, resulting from unloading that reduces the interstitial fluid flow and, consequentially, reduces oxygen transport (18,19). It is widely regarded that disruption of the lacuno-canalicular network, which is necessary for nutrient and gaseous exchange for osteocytes, results in localized hypoxia in bone (20,21).

It has been hypothesized that oxygen deprivation, due to reduced mechanical loading, may be the cause of the osteocyte apoptosis (22,23) and osteoclast recruitment, in order to remove bone matrix during hypoxia (2427). Hypoxia and oxidative stress increase the expression of different factors that mediate the adaptive responses to hypoxia/ischemia. The role of certain molecular endoplasmic reticulum (ER) chaperones has been extensively investigated, in particular regarding their functions in the quality control of proteins processed in the ER and in the regulation of ER signaling in response to stress (28).

Oxygen-regulated protein 150 (ORP150) protein (150 kDa), a novel endoplasmic-reticulum-associated chaperone induced by hypoxia/ischemia (29), has been identified to be highly expressed in osteocytes compared with osteoblasts (30). ORP150 has been proposed to be involved in the prevention of apoptosis induced by oxygen deprivation (31,32).

The cytoprotective role of ORP150 in different cell types has been previously demonstrated and it has been hypothesized that ORP150 is required by osteocytes to survive in their reduced oxygen state into the bone matrix (3335). The current study aimed to establish whether ORP150 levels correlated with osteocyte cell death and apoptosis under hypoxic conditions.

Materials and methods

Cell culture

The murine long bone osteocyte Y4 (MLO-Y4) cell line (supplied from University of Missouri, Kanas City, USA) was used and cultured as previously described (36,37). Briefly, the cells were cultured on collagen-coated (rat tail type I collagen; BD Biosciences, Franklin Lakes, NJ, USA) plastic ware and were grown at 37°C, 5% CO2, 95% air with α-minimal essential medium (α-MEM; 1X; Invitrogen; Thermo Fisher Scientific, Inc., Waltham, MA, USA) supplemented with 2.5% fetal bovine serum (FBS) (PAA; GE Healthcare Life Sciences, Pittsburgh, PA, USA), 2.5% bovine calf serum (BCS; GE Healthcare Life Sciences, Logan, UT, USA) and 100 U/ml penicillin/streptomycin at (Invitrogen; Thermo Fisher Scientific, Inc). For the hypoxia experiments, 5,000 cells/cm2 were seeded onto collagen-coated multi-well dishes, incubated in α-MEM without phenol red, 2.5% FBS + 2.5% BCS and 100 U/ml penicillin/streptomycin (Invitrogen; Thermo Fisher Scientific, Inc.).

To confirm the data obtained with the MLO-Y4 cells, the pre-osteoblast MC3T3-E1 cell line was additionally used. MC3T3-E1 subclone 14 was obtained from the American Type Culture Collection cell bank (Manassas, VA, USA) and was cultured as previously described (30).

Hypoxia

Time 0 was designated as 24 h post-seeding, which was when cell culture under normal conditions (normoxia 20% O2), or hypoxic conditions (hypoxia 1% O2) was initiated. Cells were cultured under these conditions for 8, 16, 24, 48 and 72 h. For the MC3T3-E1 cells, the 16 h time point was not repeated, as this set of experiments was performed to confirm the result already obtained with the MLO-Y4 cell line. For hypoxic conditions, the cells were placed inside a chamber from Billups-Rothenberg, Inc. (San Diego, CA, USA), where a mixture of gas (95% N2 and 5% CO2) was injected resulting in 1% O2. The oxygen percentage was controlled by an Oxygen Analyzer/Monitor (Vascular Technology, Inc., Nashua, NH, USA).

To evaluate hypoxia of osteocytes and pre-osteoblasts, pimonidazole hydrochloride (Hypoxyprobe™-1; Chemicon; EMD Millipore, Billerica, MA, USA) (38) was used. Pimonidazole hydrochloride is a substance with low-molecular weight that binds only to cells that have an oxygen tension of 10 mm Hg (pO2~1,2%) or lower. The cultures were stained for hypoxic cells with Hypoxyprobe™-1 according to the manufacturer's protocol.

One experiment in quadruplicate was performed for each cell culture and each time point.

Cell viability and apoptosis

Osteocyte viability and total cell number were determined by trypan blue staining (39,40). Three experiments in quadruplicate were performed for each cell culture and time point. The results were expressed as the number of trypan blue-positive cells, over the total number of cells. Apoptosis was assessed subsequent to 4′,6-diamidino-2-phenylindole nuclear staining, counting cells exhibiting chromatin condensation or nuclear blebbing over the total number of cells (41).

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

RNA isolation was performed using TRIzol reagent (Applied Biosystems; Thermo Fisher Scientific, Inc.). Total RNA (500 ng) was reverse transcribed using the High-Capacity cDNA Reverse Transcription kit (Applied Biosystems; Thermo Fisher Scientific, Inc.). Gene expression was analyzed using TaqMan Assay probes and primers for ORP150 (cat. no. Mm00491279) and β2 macroglobulin (cat. no. Mm00437762) with Master Mix TaqMan Gene Expression reagents (Applied Biosystems; Thermo Fisher Scientific, Inc.). The cycling condition used were an initial stage of 2 min incubation at 50°C then 10 min at 95°C, this was followed by 40 cycles of 95 °C for 15 min and 60°C for 1 min. All equipment and reagents used for the RT-qPCR were purchased from Applied Biosystems (Thermo Fisher Scientific, Inc.). The four experiments performed were conducted in triplicate; the quantification cycle (Cq) value was determined by the default settings, using StepOne software, version 2.1 (Applied Biosystems; Thermo Fisher Scientific, Inc.). The relative expression of ORP150 in hypoxic MLO-Y4 cells was compared with the expression in MLO-Y4 cells grown for 8 h under normoxic conditions using the 2-∆∆Cq method (42).

Western blotting

Cells cultured under hypoxia or normoxia were lysed in radioimmunoprecipitation assay buffer complete with a proteinase inhibitor cocktail (Enzo Life Sciences, Farmingdale, NY, USA). A total of four experiments for MLO-Y4 and one for MC3T3-E1 were performed to compare the ORP150 expression in hypoxic and normoxic conditions at all different time points. A total of three different experiments were conducted in order to compare ORP150 expression in normoxic osteocytes and pre-osteoblasts. Due to the fact that the experiments made with MC3T3-E1 cells were performed to confirm the results obtained with the MLO-Y4 cells, the 16 h time point for this cell line was not repeated. Protein concentration was determined in each cell lysate supernatant using a DC Protein Assay kit I colorimetric assay (Bio-Rad Laborartories, Inc., Hercules, CA, USA). Equal amounts of protein (15 µg) were applied and separated using 10% sodium dodecyl sulfate-polyacrylimide gel electrophoresis and transferred by electrophoresis to a nitrocellulose membrane. The membranes were blocked with 2.5% non-fat dried milk (Bio-Rad Laboratories, Inc.) in 1X phosphate-buffered saline at room temperature for 30 min and incubated with rabbit anti-ORP150 (dilution 1:1,000; cat. no. 3905-1; Epitomics, Burlingame, CA, USA) and rabbit anti-β-actin (dilution 1:1,000; cat. no. LF-PA0207; AbFrontier, Seoul, Korea) primary antibodies for 2 h at room temperature, with β-actin as the internal control. Horseradish peroxidase-conjugated anti-rabbit secondary antibody (dilution 1:1,000; cat. no. HAF008; R&D Systems, Inc., Minneapolis, MN, USA) was subsequently incubated with the membranes for 1.5 h at room temperature. Enhanced chemiluminescent reagents (GE Healthcare Life Sciences) and films were used to visualize the protein bands and were quantified using Adobe Photoshop (Adobe Systems, Inc., San Jose, CA, USA).

Statistical analysis

Data were analyzed for normal distribution using the Kolmogorov-Smirnov test. Differences between the two oxygen conditions were assessed using Mann-Whitney U test, and differences between the experimental times were assessed using the Kruskal-Wallis test for nonparametric data. P<0.05 was considered to indicate a statistically significant difference. Statistical analyses were performed using SPSS software, version 14.0 (SPSS, Inc., Chicago, IL, USA). Differences between data obtained by western blot analysis, comparing ORP150 expression in MC3T3-E1 and MLO-Y4 cells was assessed using Student's t-test.

Results

Induction of hypoxia

MLO-Y4 cells cultured at 1% O2 exhibited a significantly increased level of Hypoxyprobe staining (Fig. 1A and B) compared with cells cultured in normoxic conditions at the same time points: 16, 24, 48 and 72 h (P=0.005, 0.005, 0.009 and 0.007, respectively). In addition, in hypoxic conditions, the number of Hypoxyprobe-positive cells significantly increased over time (hypoxic MLO-Y4 P=0001; Kruskal Wallis test), suggesting that the cells became more sensitive to hypoxia (Fig. 1B). Conversely, the time-dependent increase of Hypoxyprobe-positive cells was not observed in normoxic conditions and only following 72 h exposure was a marginal increase detected. A similar trend was observed for the MC3T3-E1 cells (Fig. 1C); the number of Hypoxyprobe-positive cells was significantly different at all time points under hypoxic conditions compared with normal conditions (8, 24, 48 and 72 h; P=0.001, 0.001, 0.009 and 0.001, respectively). In addition, a significant time-dependent increase in the number of MC3T3-E1 Hypoxyprobe-positive cells was observed under hypoxia (Kruskal Wallis test; P=0001), however was not observed under normoxia (Kruskal Wallis test; P=0515).

These results indicate that the two cell lines were sensitive to hypoxia under the conditions of oxygen deprivation.

Cell viability and apoptosis

Osteocyte viability, as determined by trypan blue staining, was compromised by oxygen deprivation and resulted in a significant increase in cell death at 8, 24 and 48 h (P=0.049; P=0.002; P=0.043, respectively), however was not significant at 16 and 72 h (P=0.122; P=0.069, respectively) (Fig. 2A). Cell counting observed an increase in total cell number over time, however, deprivation of oxygen significantly reduced total number of cells at 8 and 48 h compared with the same time points under normoxia (Fig. 2B). The number of apoptotic osteocytes, as determined by nuclear fragmentation, was increased by hypoxia and this increase was identified to be statistically significant at 16, 24 and 48 h, compared with the same time points cultured under normoxia. At 72 h, the number of apoptotic cells cultured under hypoxic conditions was reduced compared with normoxia, however this difference was identified to not be significant. The number of apoptotic cells in hypoxia at 72 h was additionally identified to be marginally reduced compared with the values observed at 48 h, however this difference was not statistically significant (Fig. 3).

ORP150 expression

MLO-Y4 ORP150 mRNA expression was significantly reduced in a time-dependent manner under hypoxic and normoxic conditions. No significant differences were observed in mRNA levels under hypoxic conditions compared with normoxia apart from at 72 h, where the difference was statistically significant with a smaller reduction under hypoxic conditions (Fig. 4). In particular, there were statistically significant differences under hypoxia identified between 72 and 16 h, 24 and 48 h (P=0.001, 0.009 and 0.007, respectively); similar results were obtained under normoxic conditions (P=0.000, 0.000 and 0.012, respectively). Western blot analysis conducted on MLO-Y4 indicated no statistically significant differences in ORP150 expression at a protein level at any time point under the same conditions of the mRNA analysis (Fig. 5). It is suggested that additional later time points would reflect the significant differences in the 72 h mRNA data.

Confirming the ORP150 expression data identified in MLO-Y4 cells, no differences were observed in ORP150 expression in MC3T3-E1 cells under normoxic and hypoxic conditions, similarly, there were no significant differences over time under normoxic and hypoxic conditions (data not shown). In addition, western blot analysis indicated that the expression of ORP150 was significantly reduced in normoxic MC3T3-E1 cells (ORP150/β-actin ratio=0.531±0.03-mean ± standard deviation-) compared with normoxic MLO-Y4 cells (ORP150/β-actin ratio=0.845±0.06-mean ± standard deviation-), with P=0.001, in agreement with a previous study (30).

Discussion

The objective of the current study was to determine whether the expression of ORP150 in osteocytes is regulated in response to oxygen deprivation.

Hypoxia is a normal physiological condition for osteocytes. A concentration of approximately 5% O2 is more likely to be physiological for osteocytes than a concentration of 20%O2, as osteocytes are embedded deep inside the mineralized bone matrix and their nutrient availability is greatly dependent on diffusion (4345). The actual concentration of oxygen inside of the osteocyte lacunae during skeletal unloading remains unknown. A previous study has measured pO2 values of approximately 6% in normal human bone marrow aspirates (46). On the basis of this information, 1% O2 was selected as the hypoxic condition and 20% O2 as the normoxic condition (representing the atmospheric oxygen levels); although it is widely accepted that a pO2 of 20% corresponds to hyperoxia (44).

On the basis of this assumption, the data of the present study indicate that 1% O2 induces hypoxia and significantly compromises cellular viability. The results obtained additionally demonstrate that apoptosis serves a role in oxygen deprivation-mediated induction of cell death. Despite the fact that osteocytes were sensitive to hypoxia from 16 to 72 h, as demonstrated by Hypoxyprobe staining (Fig. 1), the difference in the rate of apoptosis in hypoxia compared with normoxia, remained significant until the 48 h time point. Following that, the apoptotic stimuli induced by hypoxia appear to be inhibited at 72 h, where the percentage of apoptotic cells was not identified to be significantly different in hypoxia compared with normoxia. It is hypothesized that, in the early stages, additional mechanisms of protection against oxygen deprivation-induced cell death may occur (47,48).

No significant differences were observed in ORP150 mRNA levels until the 72 h time point, and protein levels at any time point, however, it is suggested that the protein levels may have been greater at later time points. This suggests that ORP150 mRNA expression is delayed, as based on the Hypoxyprobe results, the cells become hypoxic from 16 h. Notably, ORP150 expression reduced over time under normoxic and hypoxic conditions. Unlike additionally cell types including Hela cells (49), neuronal cells (5052), renal cells (53) and cardiomyocytes (54,55), in MLO-Y4 cells ORP150 does not appear to be regulated by hypoxia at early time points. Similar results were identified for the ORP150 protein expression in pre-osteoblasts grown under hypoxic conditions compared with normoxic conditions. The ORP150 protein levels were increased in MLO-Y4 osteocytes compared with MC3T3-E1 pre-osteoblasts grown under normoxic conditions. These observations are in agreement with previous studies demonstrating that low oxygen tension serves an important role in the differentiation of osteoblasts into osteocytes (45,56). Guo et al (30) demonstrated that a greater amount of hypoxia-associated proteins in osteocytes (such as ORP150), compared with in osteoblasts. The higher level of ORP150 protein may be a possible explanation for the capacity of osteocytes to adapt to their hypoxic environment.

In conclusion, ORP150 is commonly highly expressed in osteocytes as compared with osteoblasts, and mRNA expression is not elevated compared with the control until late stages of hypoxic culture conditions. Significant cell death and apoptosis precede the significant differences in ORP150 mRNA in hypoxia compared with normoxia. It is hypothesized that the amount of ORP150 in MLO-Y4 is sufficient to protect the cells from hypoxia up to 48 h, however longer exposure to oxygen deprivation may lead to upregulation of ORP150 expression. After 72 h, the significant increase in ORP150 mRNA, identified in hypoxia compared with normoxia, strongly support the hypothesis that longer exposure to hypoxia may additionally induce an increase of ORP150 protein levels. It is notable that at present the actual concentration of oxygen inside of the osteocyte lacunae remains unknown, and this should be considered a limitation. However, in order to understand the behavior of osteocytes in response to the oxygen deprivation and to increase knowledge of this complex pathway, evaluation of the hypoxic condition at early time points is scientifically valuable. It is thus important to further investigate the role of ORP150 in the apoptotic process under hypoxic conditions for longer time periods, and to block ORP150 expression, using ORP150 small interfering RNA or knockout mice, to more fully elucidate the protective role of this protein.

Acknowledgements

The authors would like to thank Dr Luca Dalle Carbonare, of the Policlinico G.B. Rossi (Verona, Italy), for providing the MLO-Y4 cell line. The authors would additionally like to thank Dr Lucia Mancini for helping with the revision of the manuscript, and Mr. Luigi Lena for the illustrations.

References

1 

Bianconi E, Piovesan A, Facchin F, Beraudi A, Casadei R, Frabetti F, Vitale L, Pelleri MC, Tassani S, Piva F, et al: An estimation of the number of cells in the human body. Ann Hum Biol. 40:463–471. 2013. View Article : Google Scholar : PubMed/NCBI

2 

Bonewald LF: Osteocytes as dynamic multifunctional cells. Ann N Y Acad Sci. 1116:281–290. 2007. View Article : Google Scholar : PubMed/NCBI

3 

Busse B, Djonic D, Milovanovic P, Hahn M, Püschel K, Ritchie RO, Djuric M and Amling M: Decrease in the osteocyte lacunar density accompanied by hypermineralized lacunar occlusion reveals failure and delay of remodeling in aged human bone. Aging Cell. 9:1065–1075. 2010. View Article : Google Scholar : PubMed/NCBI

4 

Bonewald LF: The amazing osteocyte. J Bone Miner Res. 26:229–238. 2011. View Article : Google Scholar : PubMed/NCBI

5 

Nakashima T, Hayashi M, Fukunaga T, Kurata K, Oh-Hora M, Feng JQ, Bonewald LF, Kodama T, Wutz A, Wagner EF, et al: Evidence for osteocyte regulation of bone homeostasis through RANKL expression. Nat Med. 17:1231–1234. 2011. View Article : Google Scholar : PubMed/NCBI

6 

Baron R and Kneissel M: WNT signaling in bone homeostasis and disease: From human mutations to treatments. Nat Med. 19:179–192. 2013. View Article : Google Scholar : PubMed/NCBI

7 

Javaheri B, Stern AR, Lara N, Dallas M, Zhao H, Liu Y, Bonewald LF and Johnson ML: Deletion of a single b-catenin allele in osteocytes abolishes the bone anabolic response to loading. J Bone Miner Res. 29:705–715. 2014. View Article : Google Scholar : PubMed/NCBI

8 

Robling AG, Niziolek PJ, Baldridge LA, Condon KW, Allen MR, Alam I, Mantila SM, Gluhak-Heinrich J, Bellido TM, Harris SE and Turner CH: Mechanical stimulation of bone in vivo reduces osteocyte expression of Sost/sclerostin. J Biol Chem. 283:5866–5875. 2008. View Article : Google Scholar : PubMed/NCBI

9 

Paszty C, Turner CH and Robinson MK: Sclerostin: A gem from the genome leads to bone-building antibodies. J Bone Miner Res. 25:1897–1904. 2010. View Article : Google Scholar : PubMed/NCBI

10 

Xiong J and O'Brien CA: Osteocyte RANKL: New insights into the control of bone remodeling. J Bone Miner Res. 27:499–505. 2012. View Article : Google Scholar : PubMed/NCBI

11 

Burgers TA and Williams BO: Regulation of Wnt/b-catenin signaling within and from osteocytes. Bone. 54:244–249. 2013. View Article : Google Scholar : PubMed/NCBI

12 

Tate ML Knothe, Niederer P and Knothe U: In vivo tracer transport through the lacunocanalicular system of rat bone in an environment devoid of mechanical loading. Bone. 22:107–117. 1998. View Article : Google Scholar : PubMed/NCBI

13 

Gortazar AR, Martin-Millan M, Bravo B, Plotkin LI and Bellido T: Crosstalk between caveolin-1/extracellular signal-regulated kinase (ERK) and b-catenin survival pathways in osteocyte mechanotransduction. J Biol Chem. 288:8168–8175. 2013. View Article : Google Scholar : PubMed/NCBI

14 

Tate ML Knothe: ‘Whither flows the fluid in bone?’ An osteocyte's perspective. J Biomech. 36:1409–1424. 2003. View Article : Google Scholar : PubMed/NCBI

15 

Dufour C, Holy X and Marie PJ: Transforming growth factor-beta prevents osteoblast apoptosis induced by skeletal unloading via PI3K/Akt, Bcl-2 and phospho-Bad signaling. Am J Physiol Endocrinol Metab. 294:E794–E801. 2008. View Article : Google Scholar : PubMed/NCBI

16 

Jilka RL, Noble B and Weinstein RS: Osteocyte apoptosis. Bone. 54:264–271. 2013. View Article : Google Scholar : PubMed/NCBI

17 

Bikle DD and Halloran BP: The response of bone to unloading. J Bone Miner Metab. 17:233–244. 1999. View Article : Google Scholar : PubMed/NCBI

18 

Ontiveros C, Irwin R, Wiseman RW and McCabe LR: Hypoxia suppresses runx2 independent of modeled microgravity. J Cell Physiol. 200:169–176. 2004. View Article : Google Scholar : PubMed/NCBI

19 

Stevens HY, Meays DR and Frangos JA: Pressure gradients and transport in the murine femur upon hindlimb suspension. Bone. 39:565–572. 2006. View Article : Google Scholar : PubMed/NCBI

20 

Dodd JS, Raleigh JA and Gross TS: Osteocyte hypoxia: A novel mechanotransduction pathway. Am J Physiol. 277:C598–C602. 1999.PubMed/NCBI

21 

Hinoi E, Ochi H, Takarada T, Nakatani E, Iezaki T, Nakajima H, Fujita H, Takahata Y, Hidano S, Kobayashi T, et al: Positive regulation of osteoclastic differentiation by growth differentiation factor 15 upregulated in osteocytic cells under hypoxia. J Bone Miner Res. 27:938–949. 2012. View Article : Google Scholar : PubMed/NCBI

22 

Plotkin LI, Mathov I, Aguirre JI, Parfitt AM, Manolagas SC and Bellido T: Mechanical stimulation prevents osteocyte apoptosis: Requirement of integrins, Src kinases, and ERKs. Am J Physiol Cell Physiol. 289:C633–C643. 2005. View Article : Google Scholar : PubMed/NCBI

23 

Wang H, Ji B, Liu XS, Nakatani E, Iezaki T, Nakajima H, Fujita H, Takahata Y, Hidano S and Kobayashi T: Osteocyte-viability-based simulations of trabecular bone loss and recovery in disuse and reloading. Biomech Model Mechanobiol. 13:153–166. 2014. View Article : Google Scholar : PubMed/NCBI

24 

Aguirre JI, Plotkin LI, Stewart SA, Weinstein RS, Parfitt AM, Manolagas SC and Bellido T: Osteocyte apoptosis is induced by weightlessness in mice and precedes osteoclast recruitment and bone loss. J Bone Miner Res. 21:605–615. 2006. View Article : Google Scholar : PubMed/NCBI

25 

Bellido T: Osteocyte-driven bone remodeling. Calcif Tissue Int. 94:25–34. 2014. View Article : Google Scholar : PubMed/NCBI

26 

Gross TS, Akeno N, Clemens TL, Komarova S, Srinivasan S, Weimer DA and Mayorov S: Selected contribution: Osteocytes upregulate HIF-1alpha in response to acute disuse and oxygen deprivation. J Appl Physiol (1985). 90:2514–2519. 2001.PubMed/NCBI

27 

Noble BS, Peet N, Stevens HY, Brabbs A, Mosley JR, Reilly GC, Reeve J, Skerry TM and Lanyon LE: Mechanical loading: Biphasic osteocyte survival and targeting of osteoclasts for bone destruction in rat cortical bone. Am J Physiol Cell Physiol. 284:C934–C943. 2003. View Article : Google Scholar : PubMed/NCBI

28 

Jung TW, Lee KT, Lee MW and Ka KH: SIRT1 attenuates palmitate-induced endoplasmic reticulum stress and insulin resistance in HepG2 cells via induction of oxygen-regulated protein 150. Biochem Biophys Res Commun. 422:229–232. 2012. View Article : Google Scholar : PubMed/NCBI

29 

Kuwabara K, Matsumoto M, Ikeda J, Hori O, Ogawa S, Maeda Y, Kitagawa K, Imuta N, Kinoshita T, Stern DM, et al: Purification and characterization of a novel stress protein, the 150-kDa oxygen-regulated protein (ORP150), from cultured rat astrocytes and its expression in ischemic mouse brain. J Biol Chem. 271:5025–5032. 1996. View Article : Google Scholar : PubMed/NCBI

30 

Guo D, Keightley A, Guthrie J, Veno PA, Harris SE and Bonewald LF: Identification of osteocyte-selective proteins. Proteomics. 10:3688–3698. 2010. View Article : Google Scholar : PubMed/NCBI

31 

Boraldi F, Annovi G, Carraro F, Naldini A, Tiozzo R, Sommer P and Quaglino D: Hypoxia influences the cellular cross-talk of human dermal fibroblasts. A proteomic approach. Biochim Biophys Acta. 1774:1402–1413. 2007. View Article : Google Scholar : PubMed/NCBI

32 

Ozawa K, Kuwabara K, Tamatani M, Takatsuji K, Tsukamoto Y, Kaneda S, Yanagi H, Stern DM, Eguchi Y, Tsujimoto Y, et al: 150-kDa oxygen-regulated protein (ORP150) suppresses hypoxia-induced apoptotic cell death. J Biol Chem. 274:6397–6404. 1999. View Article : Google Scholar : PubMed/NCBI

33 

Kretowski R, Borzym-Kluczyk M and Cechowska-Pasko M: Hypoxia enhances the senescence effect of bortezomib-the proteasome inhibitor-on human skin fibroblasts. Biomed Res Int. 2014:1962492014. View Article : Google Scholar : PubMed/NCBI

34 

Kretowski R, Stypulkowska A and Cechowska-Pasko M: Low-glucose medium induces ORP150 expression and exerts inhibitory effect on apoptosis and senescence of human breast MCF7 cells. Acta Biochim Pol. 60:167–173. 2013.PubMed/NCBI

35 

Tamatani M, Matsuyama T, Yamaguchi A, Mitsuda N, Tsukamoto Y, Taniguchi M, Che YH, Ozawa K, Hori O, Nishimura H, et al: ORP150 protects against hypoxia/ischemia-induced neuronal death. Nat Med. 7:317–323. 2001. View Article : Google Scholar : PubMed/NCBI

36 

Bonewald LF: Establishment and characterization of an osteocyte-like cell line, MLO-Y4. J Bone Miner Metab. 17:61–65. 1999. View Article : Google Scholar : PubMed/NCBI

37 

Kato Y, Windle JJ, Koop BA, Mundy GR and Bonewald LF: Establishment of an osteocyte-like cell line, MLO-Y4. J Bone Miner Res. 12:2014–2023. 1997. View Article : Google Scholar : PubMed/NCBI

38 

Raleigh JA, Chou SC, Bono EL, Thrall DE and Varia MA: Semiquantitative immunohistochemical analysis for hypoxia in human tumors. Int J Radiat Oncol Biol Phys. 49:569–574. 2001. View Article : Google Scholar : PubMed/NCBI

39 

Ahuja SS, Zhao S, Bellido T, Plotkin LI, Jimenez F and Bonewald LF: CD40 ligand blocks apoptosis induced by tumor necrosis factor alpha, glucocorticoids, and etoposide in osteoblasts and the osteocyte-like cell line murine long bone osteocyte-Y4. Endocrinology. 144:1761–1769. 2003. View Article : Google Scholar : PubMed/NCBI

40 

Jilka RL, Weinstein RS, Bellido T, Parfitt AM and Manolagas SC: Osteoblast programmed cell death (apoptosis): Modulation by growth factors and cytokines. J Bone Miner Res. 13:793–802. 1998. View Article : Google Scholar : PubMed/NCBI

41 

Plotkin LI, Weinstein RS, Parfitt AM, Roberson PK, Manolagas SC and Bellido T: Prevention of osteocyte and osteoblast apoptosis by bisphosphonates and calcitonin. J Clin Invest. 104:1363–1374. 1999. View Article : Google Scholar : PubMed/NCBI

42 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2 (−Delta Delta C (T)) Method. Methods. 25:402–408. 2001. View Article : Google Scholar : PubMed/NCBI

43 

Al Hadi H, Smerdon GR and Fox SW: Hyperbaric oxygen therapy suppresses osteoclast formation and bone resorption. J Orthop Res. 31:1839–1844. 2013.PubMed/NCBI

44 

Arnett TR: Acidosis, hypoxia and bone. Arch Biochem Biophys. 503:103–109. 2010. View Article : Google Scholar : PubMed/NCBI

45 

Hirao M, Hashimoto J, Yamasaki N, Ando W, Tsuboi H, Myoui A and Yoshikawa H: Oxygen tension is an important mediator of the transformation of osteoblasts to osteocytes. J Bone Miner Metab. 25:266–276. 2007. View Article : Google Scholar : PubMed/NCBI

46 

Harrison JS, Rameshwar P, Chang V and Bandari P: Oxygen saturation in the bone marrow of healthy volunteers. Blood. 99:3942002. View Article : Google Scholar : PubMed/NCBI

47 

Hou M, Liu J, Liu F, Liu K and Yu B: C1q tumor necrosis factor-related protein-3 protects mesenchymal stem cells against hypoxia- and serum deprivation-induced apoptosis through the phosphoinositide 3-kinase/Akt pathway. Int J Mol Med. 33:97–104. 2014.PubMed/NCBI

48 

Rouault-Pierre K, Lopez-Onieva L, Foster K, Anjos-Afonso F, Lamrissi-Garcia I, Serrano-Sanchez M, Mitter R, Ivanovic Z, de Verneuil H, Gribben J, et al: HIF-2α protects human hematopoietic stem/progenitors and acute myeloid leukemic cells from apoptosis induced by endoplasmic reticulum stress. Cell Stem Cell. 13:549–563. 2013. View Article : Google Scholar : PubMed/NCBI

49 

Cechowska-Pasko M, Bankowski E and Chene P: The effect of hypoxia on the expression of 150 kDa oxygen-regulated protein (ORP 150) in HeLa cells. Cell Physiol Biochem. 17:89–96. 2006. View Article : Google Scholar : PubMed/NCBI

50 

Kitano H, Nishimura H, Tachibana H, Yoshikawa H and Matsuyama T: ORP150 ameliorates ischemia/reperfusion injury from middle cerebral artery occlusion in mouse brain. Brain Res. 1015:122–128. 2004. View Article : Google Scholar : PubMed/NCBI

51 

Ogawa S: ORP150 (150 kDa oxygen regulated protein) suppressed neuronal cell death. Nihon Yakurigaku Zasshi. 121:43–48. 2003.(In Japanese). View Article : Google Scholar : PubMed/NCBI

52 

Riezzo I, Neri M, De Stefano F, Fulcheri E, Ventura F, Pomara C, Rabozzi R, Turillazzi E and Fineschi V: The timing of perinatal hypoxia/ischemia events in term neonates: A retrospective autopsy study. HSPs, ORP-150 and COX2 are reliable markers to classify acute, perinatal events. Diagn Pathol. 5:492010. View Article : Google Scholar : PubMed/NCBI

53 

Arrington DD and Schnellmann RG: Targeting of the molecular chaperone oxygen-regulated protein 150 (ORP150) to mitochondria and its induction by cellular stress. Am J Physiol Cell Physiol. 294:C641–C650. 2008. View Article : Google Scholar : PubMed/NCBI

54 

Aleshin AN, Sawa Y, Kitagawa-Sakakida S, Bando Y, Ono M, Memon IA, Tohyama M, Ogawa S and Matsuda H: 150-kDa oxygen-regulated protein attenuates myocardial ischemia-reperfusion injury in rat heart. J Mol Cell Cardiol. 38:517–525. 2005. View Article : Google Scholar : PubMed/NCBI

55 

Matsusue A, Hara K, Kageura M, Kashiwagi M, Lu W, Ishigami A, Gotohda T, Tokunaga I, Nisimura A, Sugimura T and Kubo S: An autopsy case of rhabdomyolysis related to vegetamin and genetic analysis of the rhabdomyolysis-associated genes. J Forensic Leg Med. 17:46–49. 2010. View Article : Google Scholar : PubMed/NCBI

56 

Dallas SL and Bonewald LF: Dynamics of the transition from osteoblast to osteocyte. Ann N Y Acad Sci. 1192:437–443. 2010. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2016
Volume 14 Issue 5

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Montesi M, Jähn K, Bonewald L, Stea S, Bordini B and Beraudi A: Hypoxia mediates osteocyte ORP150 expression and cell death in vitro. Mol Med Rep 14: 4248-4254, 2016
APA
Montesi, M., Jähn, K., Bonewald, L., Stea, S., Bordini, B., & Beraudi, A. (2016). Hypoxia mediates osteocyte ORP150 expression and cell death in vitro. Molecular Medicine Reports, 14, 4248-4254. https://doi.org/10.3892/mmr.2016.5790
MLA
Montesi, M., Jähn, K., Bonewald, L., Stea, S., Bordini, B., Beraudi, A."Hypoxia mediates osteocyte ORP150 expression and cell death in vitro". Molecular Medicine Reports 14.5 (2016): 4248-4254.
Chicago
Montesi, M., Jähn, K., Bonewald, L., Stea, S., Bordini, B., Beraudi, A."Hypoxia mediates osteocyte ORP150 expression and cell death in vitro". Molecular Medicine Reports 14, no. 5 (2016): 4248-4254. https://doi.org/10.3892/mmr.2016.5790