<<

Downloaded by guest on September 26, 2021 www.pnas.org/cgi/doi/10.1073/pnas.1707863114 a Seref-Ferlengez Zeynep Lewis J. vivo Karl in frequency loading magnitude and strain encode signals calcium Osteocyte clfre n eemnsmc fhwotoye experience osteocytes how of that much determines mechan- layer transmits and fluid-gel loaded, forces which mechanically a ical layer, are fluid by bones This when spaces (LCS). in moves space these lacunar-canalicular reside in the processes fills matrix surrounded bone and are within respectively) bodies and canaliculi, and cell (lacunae Osteocyte spaces array. nected and transducing osteoblasts regulate and osteoclasts. which subjected signals, downstream is func- sens- into bone renal is load which this osteocytes on to of impact load functions mechanical an essential ing have most that the FGF23, Among produc- tions. as cells, such endocrine as factors function ing also They (Ca level. matrix calcium mineraliza- bone also local that diverse resorption, and factors of and signaling tion, formation orchestrators soluble bone local produce both They major regulate bone. the in within are buried functions matrix, osteocytes, that bone recognized the increasingly How- is osteoporosis. it and defects ever, and growth (respectively), to leads resorption dysfunction and their skeleton formation bone cells that out effector of the carry mechani- are that integrity osteoclasts its the and Osteoblasts to maintaining life. structure appropriate throughout for its skeleton and adapt a demands to growing cal bone for of ability essential The is children. straightening of from derives bones mid-1700s, “ortho- the the word organ the in first indeed, coined the regard; first among this pedics,” in was recognized it be that to bone systems for prominent so is ability B loading vivo in osteocytes the how loading. determine of to frequency technique and a magnitude provide encode as cells mechanical well to as respond vivo, cells in offer bone load critical studies signifi- these how Our change into magnitudes. glimpse not a loading physiological did with osteocytes cantly responding Ca However, within load- magnitude. intensity strain all osteocytes applied responding At with of strongly bone. number increased for the to examined, reported frequencies 250 conditions ing from physiological ranging of strains istic to osteocytes of 3,000 responses the vivo investigated in We microscopy. fluorescence multiphoton using encoded genetically Ca a using fluorescent by osteocytes individual Ca of intracellular of achieved responses the monitoring bones is simultaneously of metatarsal while This capable mice to living is vivo. loads which mechanical in device well-defined bending examined delivering three-point be a bone using to in by and yet herein vitro has in and osteocytes but in perceive, explants, studied widely process, been has vivo lation Intra- in (Ca understood. calcium poorly osteocytes cellular remains loading how mechanical Donahue) to cells J. but respond Henry mechanosensory and major bone, Alliston Tamara the by be of reviewed to 2017; 11, considered May are review Osteocytes for (sent 2017 14, September Weinbaum, Sheldon by Contributed and 10461; NY Bronx, Medicine, eateto imdclEgneig iyCleeo e ok e ok Y10031; NY York, New York, New of College City Engineering, Biomedical of Department sectsi iueiti ope,3,hgl intercon- highly 3D, complex, a in exist situ in Osteocytes dp t tutr nrsos.Ti ehncladaptation and mechanical load This mechanical response. sense in structure to its ability adapt remarkable a has one µ n rqece rm05t z hc r character- are which Hz, 2 to 0.5 from frequencies and | acu signaling calcium | a 2+ bone or Frikha-Benayed Dorra , niao.Otoyersossaeiae by imaged are responses Osteocyte indicator. 2+ inln eutn rmmcaia stimu- mechanical from resulting signaling ) c | oetJ Majeska J. Robert , mechanotransduction 2+ c eateto rhpei ugr,Abr isenCleeo eiie rn,N 10461 NY Bronx, Medicine, of College Einstein Albert Surgery, Orthopaedic of Department n hsht rnpr tthe at transport phosphate and ) a oc Louie Joyce , | a hlo Weinbaum Sheldon , a aulStephen Samuel , 2+ 2+ 1073/pnas.1707863114/-/DCSupplemental at online information supporting contains article This 1 the under Published interest. of Common- conflict no Virginia declare H.J.D., authors The and Francisco; San K.J.L., California, University. and wealth of data; University analyzed T.A., S.S. Reviewers: and paper. the J.L., wrote K.J.L., M.B.S. and research; R.J.M., performed Z.S.-F. and M.M.T., simultaneously a while created mice live also in We use environment. for vivo approach loading in bone authentic genetically their osteocyte-targeted in an cytes with Ca model gen- encoded We mouse loading. a mechanical during erated imaged individually be can Ca 19). osteocyte 18, explore (8, the pressure in vascular loss vivo to in pressure of subject fluid loss as from are resulting well explants LCS as bone tissue changes, localized while tissue limita- attendant flow, postmortem have and fluid which LCS and 17), the architecture lack (10, cultures bones Cell explanted tions. in studies or most cultures Finally, cell (16). Ca cells Ca osteocyte other whole-cell in of typical seen a that like initiated response then and body Ca cell triggered vitro forces in fluid cesses that cells with the (12–15). demonstrated on stimuli al. acting biochemical et Thi and particular, mechanical In Ca both cytoplasmic to stim- local increases response Mechanical the osteocytes (8–11). dominates of situ LCS in ulation osteocytes evidence the of to through body input flow growing mechanical a fluid Indeed, that (5–7). LCS indicates the gonadal in flow in fluid changes stim- as other (1–4). such as microdamage tissue remodeling, well local and as bone steroids load initiate is mechanical that molecules in uli signaling changes key and to these RANKL, sensitive of sclerostin, expression as Osteocyte osteocyte- such OPG. and key molecules as to well signaling transport as produced solute metabolites for including accounts osteocytes, from also It environment. their uhrcnrbtos ...... n ...dsge eerh ... D.F.-B., K.J.L., research; designed M.B.S. and S.W., D.C.S., K.J.L., contributions: Author owo orsodnemyb drse.Eal enamcn.uyeuor [email protected] Email: addressed. be may correspondence whom To mschaffl[email protected]. leigtenme frsodn el n htti effect this that and frequency. loading cells on by responding dependent is of signals number mechanical the integrate demonstrate altering results populations Our tissue-level osteocyte loading. that to bone with whole events respond signaling directsimultaneous calcium the osteocyte of for acutely observation technique vivo a in vivo here report about We bone in loading. known of mechanical is regulator osteocytes little major how However, the be events. to mechanosensation thought are Osteocytes Significance ntepeetsuis erpr ni ioapoc to approach vivo in an report we studies, present the In to translated are level tissue bone the at loads Mechanical a,1 n icelB Schaffler B. Mitchell and , a b ai .Spray C. David , eateto ersine letEnti olg of College Einstein Albert Neuroscience, of Department PNAS 2+ α niao GC)t td Ca study to (GECI) indicator NSlicense. PNAS v 2+ β | 3 coe 1 2017 31, October inln aebe odce ihri 2D in either conducted been have signaling nernahso ie notoyecl pro- cell osteocyte on sites adhesion integrin 2+ inln nwoebn hr h cells the where bone whole in signaling b i .Thi M. Mia , 2+ . epne htsra oadthe toward spread that responses | a,1 o.114 vol. www.pnas.org/lookup/suppl/doi:10. c , | 2+ o 44 no. inln nosteo- in signaling 2+ ubiquitous a , | 2+ 11775–11780 signaling

PHYSIOLOGY ENGINEERING ing off-axis stresses. MT3 bones from OtGP3 mice had larger cortical bone areas, but slightly lower tissue mineral density than WT bones (Table 1; P < 0.05), resulting in similar cross-sectional moduli and diaphyseal bending rigidities.

Osteocyte Response to Mechanical Loading in Vivo. We used a custom-designed device to deliver controlled mechanical strains to MT3 bones of living OtGP3 mice while simultaneously moni- toring intracellular Ca2+ responses with MPM. Ca2+ fluorescence in responding cells increased very rapidly, within 200 ms of load- ing onset (Fig. 2), and oscillations followed the applied loading (Movie S1). Osteocytes were examined in bones loaded cycli- cally to habitual physiological strain magnitudes [250, 500, 1,000, and 2,000 microstrain(µ)] as well as a nonhabitual high strain level (3,000 µ). Loading frequency effects were examined as well Fig. 1. Multiphoton z-stack image of OtGP3 MT3 osteocytes in vivo. Note (0.5, 1, or 2 Hz; n = 6 animals per loading frequency group). that all cells in the field of view exhibit GCaMP3 signal, indicating highly Once activated, Ca2+ response intensity remained consistent efficient expression in osteocytes. for each osteocyte for loading up to 2,000 µ. Osteocyte Ca2+ intensity increased only with loading to 3,000 µ (Fig. 3; P < 0.05, ANOVA). In contrast, the number of Ca2+ responding osteo- observing the intracellular Ca2+ responses of individual osteo- 2+ cytes increased markedly with increasing strain magnitude for cytes with multiphoton microscopy (MPM). We report how Ca all loading frequencies tested (Fig. 4). At 0.5- and 1-Hz loading, signaling of osteocytes in vivo, both individually and as networks, the load–response curves (i.e., relationship between responding responds to mechanical loading. We used this approach to test osteocytes and strain magnitude) were linear (P < 0.01). How- the hypothesis that encoding of mechanical strain and frequency ever, osteocyte “recruitment rate” (i.e., the number of respond- information by osteocytes in vivo is based on recruitment of ing osteocytes per strain level) was approximately twofold responding cells (i.e., numerical encoding) rather than on graded greater at 1- vs. 0.5-Hz loading (0.023 vs. 0.012; P < 0.001, anal- 2+ Ca responses by individual cells. ysis of covariance). The load–response curve was fundamentally different at 2 Hz. At this frequency, the number of Ca2+ respond- Results ing osteocytes was near zero for strains of <1,000 µ. Above this Osteocyte GCaMP3 Expression. Mice expressing the osteocyte- strain level, osteocyte recruitment increased steeply. The rela- targeted GECI GCaMP3, which are here termed OtGP3, were tionship between applied strain levels and responding osteocytes created. Baseline GCaMP3 fluorescence was visible in osteo- at 2-Hz loading appeared nominally exponential, but was better cytes throughout the diaphyseal cortex (Fig. 1). In pilot stud- described by a polynomial relationship (r2 = 0.55 vs. 0.85, respec- ies, we found that >95% of osteocytes in OtGP3 mice exhib- tively). Responding osteocytes were distributed within regions ited detectable fluorescence. Fluorescence intensity level varied of interest (ROIs) without an apparent spatial pattern. We among cells, and resting osteocytes exhibited low-level fluctua- 2+ observed instances in which loading at a given strain level trig- tions in Ca fluorescence intensity (Fig. 2). gered a Ca2+ response in one osteocyte, but none in an adjacent osteocyte. Bone Structure in OtGP3 Mice. Our studies used the third meta- To address the importance of vascular pressure to maintaining tarsal (MT3) of the mouse foot. Previous studies in our group the fluid pressure of the LCS, MT3s from an additional group of demonstrated that this bone is well-suited for direct visualiza- mice (n = 3) were cyclically loaded to a single test strain (2,000 tion of cortical bone osteocytes in living animals by using MPM µ, 1 Hz) for 60 s while animals were alive and then again at (20). Its tubular geometry lends itself to bending without creat- 15 min postmortem (no vascular pressure but cells viable) and 60 min postmortem (severe mitochondrial stress) (20). Ampli- tude of osteocyte Ca2+ response was attenuated markedly at 15 min after death compared with in vivo loading (Fig. 5). These small Ca2+ oscillations in response to loading became irregular and did not track the applied loading frequency. The situation further decayed by 60 min. Discussion This study provides a report of in vivo osteocyte Ca2+ responses to mechanical loading of bone in living animals and provides

Table 1. Bone structure, mineralization, stiffness, and osteocyte density in WT and OtGP3 MT3 bone Parameter B6-WT OtGP3

Bone length, mm 7.75 ± 0.07 7.59 ± 0.14* Total bone area, mm2 0.208 ± 0.25 E-2 0.205 ± 0.12 E-2 Fig. 2. A representative Ca2+ trace for a single osteocyte before (dashed TMD, g/cm3 1.15 ± 0.02 1.05 ± 0.02* line) and after the start of cyclical loading (solid line) at 1 Hz to 3,000 µ. Sectional stiffness, N/m 7.58 ± 0.36 7.82 ± 1.03 2+ Before mechanical loading, all osteocytes exhibited low-level Ca fluctu- Ot.N, no./mm2 491 ± 38 448 ± 39* ations. Immediately following the start of mechanical loading, responsive cells showed cyclic increases in Ca2+ fluorescence intensity coinciding with Values are mean ± SD. n = 6 per group. Ot.N, osteocyte number; TMD, the applied loading frequency. tissue mineral density. *P < 0.05.

11776 | www.pnas.org/cgi/doi/10.1073/pnas.1707863114 Lewis et al. Downloaded by guest on September 26, 2021 Downloaded by guest on September 26, 2021 ag fmcaia hlegs(2 52) oevr this Moreover, 25–28). to (12, due challenges mechanical stimulation Ca of mechanical cytoplas- range to Increased a response 22). initial 21, 12, established (10, Ca body Ca cytoplasmic mic cell the the triggers in turn observed in that process Ca release a cell causes ATP then release allowing ATP channels This process (16). opens cell a that on attachment stretch through integrin triggered is osteocytes in vation loading by influenced strongly Ca 4). (Fig. of also frequencies number was the osteocytes Finally, magnitude. responding propor- strain strongly applied was to osteocytes of tional OFF– number (i.e., the fashion contrast, all-or-none In phys- an ON). habitual in of respond that typical activities magnitudes discovered iological strain we to osteocytes, loaded bone osteocytes cortical fluores- from visualizing signals per- simultaneously cence that while technology loading a skeletal bone Using mitted govern loading. that mechanical to information adaptation frequency and strain Ca mechanical osteocyte how about information critical SD, represent bars Error group. loading 1-Hz uvsicesdwt nraigsri n eeefcieylna t05 n -zlaig h -zlaigrsos uv a olna.Errbars Error nonlinear. was curve response loading 2-Hz al. The et loading. Lewis 1-Hz and 0.5- at linear effectively were and strain SD. represent increasing with increased curves 4. Fig. Ca 2,000 to up levels strain increasing 3. Fig. 2+ epeiul hwdta ehnclysiuae Ca stimulated mechanically that showed previously We

2+ Normalized Intensity 0 1 2 3 4 eentda 3,000 at noted were inln sipiae ncnrlo xrsino PGE2, of expression of control in implicated is signaling Mean Intensity In hw stepretg frsodn sectsa ucino ple tanmgiuefr05,1,ad2H odn rqece.Alresponse All frequencies. loading 2-Hz and 1-, 0.5-, for magnitude strain applied of function a as osteocytes responding of percentage the is Shown 2+ nest fCa of Intensity 250 Osteocytes Responding To Load notols n sect nvto(3 4 sawell- a is 24) (23, vitro in osteocyte and osteoblast in n e rqec group. frequency per 6 = 500 2+ (a) µ. inln e sectswsntcagdwith changed not was osteocytes per signaling -Strain 1000 P µ < .5 -zlaiggop (b) group; loading 2-Hz 0.05; nallaigrt rus nrae in Increases groups. rate loading all in 2000 n 2+ e group. per 6 = nu oteosteocyte the to influx 2+ 3000 a rninsencode transients b 2+ increase P 2Hz 1Hz 0.5Hz 2+ < ele- 0.05 2+ i,maigta hybcm tfe slaigrt increases. rate loading viscoelas- as osteocyte are stiffer significant Cells become range. no they strain is same that there this meaning for contrast, tic, Hz In 2 Hz. at shown 1 response results and experimental our 0.5 1,000 with for to consistent 250 from is increased behavior is This strain tissue whole the as frequen- strain loading At (5). pre- an process cies theoretical at osteocyte an membrane shows on cell which site the attachment 6, in strain Fig. axial in for dictions exemplified is 8, (5, This might mechanically frequency 44). experience loading osteocytes that what way influence a directly suggesting exam- study, frequencies this loading of in range ined situ the in over processes sharply osteocyte increase on demon- can stresses previously flow fluid we local Alternatively, that and strated similar. 43), (42, be ear inner may the ‘frequency-tuned’ as osteocytes such of systems remains other examples in frequency, effects are cells excitatory and There frequency 39–41). rate for (37, loading basis unknown on physiological strongly the depend physio- remodeling although to and of known formation range Bone are the examined. over frequencies recruitment logical osteocyte on frequency as signaling cell in bone-formation proliferative transition to a response. controlled signaling from reflects osteocyte moves level seems bone in strain it the change Thus, high the 38). this (37, that at healing postulate fracture orga- to in poorly reasonable seen of as proliferation tissue bone nized rapid with bone associated which response system at “emergency” level formation—the a bone the to woven is apposition dysregulated bone strain lamellar high controlled from this investigation. switches that formation further noting requires worth and is time It this (35, at stimuli obscure mechanical remains Ca osteo- local adjacent Why same that the 36). such experience in LCS, not differences the do local through cytes are from patterns responses arise flow cell where could fluid system this a Alternatively, and in cells activa- binary. expected the to mechanical be intrinsic would for be indeed may thresholds differences strains that threshold different Such tissue observed tion. have identical we to skele- experiencing Interestingly, and appeared osteocytes glia (34). including adjacent muscles networks, some cardiac and types and cell tal of range Ca of a typical in are none). responses or all “OFF–ON” (i.e., Such binary effectively Ca is of loading to magnitude osteocytes vidual or intensity range, strain of regulators 29–33). critical (2, all resorption Wnt, and as formation well bone as sclerostin, and RANKL, h rsn tde eeldapoon feto loading of effect profound a revealed studies present The physiological habitual the within that establish findings Our z hr saruhylna nraei xa membrane axial in increase linear roughly a is there Hz, <2 2+ PNAS inln nest a lvtda 3,000 at elevated was intensity signaling | coe 1 2017 31, October | o.114 vol. 2+ 2+ epnefrindi- for response inln events signaling | o 44 no. α v β 3 integrin | 11777 µ. µ

PHYSIOLOGY ENGINEERING Downloaded by guest on September 26, 2021 inln tl rce h ple odn rqec.I otat t6 i ototm h epneo sectst odn a o n reua,with irregular, and low was loading to osteocytes of response the postmortem, min 60 at contrast, In frequency. loading Ca applied the tracked still signaling 11778 (10). Ca magnitude that load reported on further They dependent magnitude and displayed tibiae frequency mouse explanted Ca in repetitive osteocytes found that showed found al. Ca needle al. of a et et increase using Adachi prolonged by and 17). rapid deformed (10, a calvaria bones in explanted osteocytes using that by situ in actu- bone. signal adapt would to it osteoclasts) strain, or appropriate (osteoblasts the cells “mechanos- by ator or on mechanosensor, turned set- the is Once the tat,” thermostat. a to a had analogous of osteocyte) activation, point then-unknown the more for be the set-point Frost to strain that known by mechanical posited (now He forth bone (50). in put con- mechanosensor ago concept are decades results two “set-point” responses. experimental than the cell our with and individual model sistent of popu- Pereira modulation signaling the is by the Both adaptation than to rather bone osteocytes exper- more lation that our recruiting suggest with by results regulated consistent These manner, results. strain-dependent inten- imental response osteocyte a vary in while to allowed dys- sity level, were was strain adaptation osteocytes Bone when threshold load. regulated with given varied a number recruitment OFF above (i.e., ON) responders achieved binary be or as could functioned adaptation osteocytes forma- bone when bone on of only normal predictions based that and loading rates found anabolic tion They to (49). tibiae responses mouse osteocyte of response the dict signals systems. stretch receptors. muscle output control and baroreceptors and transducer include measurement examples input. Physiological precise and frequency of input mechanical characteristic are between of remark- encoding are coupling their networks Tight in osteocyte precise that ably indicated further strain Ca results of applied number the between and magnitudes correlations we high However, surprisingly unexpected. not observed was observed we Ca magnitude of strain asso- number an the Thus, prostaglandin between 46–48). (33, ciation activity, sclerostin and GP6DH c-fos, of IGF-1, synthase, expression including in numer- markers, changes from molecular strain-dependent inferred showing be studies can ous osteocytes responding of number con- is (45). as study loads, this of low results at the evident with most sistent cell be reduced would see that would transduction we threshold, that, frequency/rate posit reasonably some can above strains we thus, membrane sites; reduced mechanotransduction experience at would osteocyte stiffer A (Right min 60 and 5. Fig. 2+

ubro tde aeeaie sect Ca osteocyte examined have studies of number A pre- to model computational a reported recently al. et Pereira and levels loading mechanical between association loose A Normalized Intensity inln olne ossetwt h ple odn frequency. loading applied the with consistent longer no signaling 0.5 1.5 | 0 1 2 yia Ca Typical www.pnas.org/cgi/doi/10.1073/pnas.1707863114 56 57 75 70 65 60 55 0 2+ ototm sect Ca Osteocyte postmortem. ) 2+ pksi epnet ylclaig ihspike with loading, cyclic to response in spikes 5 rcsfrtorpeettv sectsbfr n fe h ne fmcaia odn nlv oe(Left bone live in loading mechanical of onset the after and before osteocytes representative two for traces Live Live 10 2+ 15 epne eedpneton dependent were responses 2+ 2+ epnigotoye.Our osteocytes. responding 2+ epnigotoye and osteocytes responding 2+ ocnrto 5) Jing (51). concentration inlapiueaueyatrdahwsmrel teutdcmae ihi io lhuhteCa the although vivo, in with compared attenuated markedly was death after acutely amplitude signal 20 0.5 1.5 0 1 2 56 57 75 70 65 60 55 2+ 0 signaling 15 min post-mortem 5 n e group. per 3 = Time (s) Time fe yia fdsae.W pclt hthroa rcell or hormonal that speculate We where diseases. are system, behaviors of to encoding nervous typical coupled parallels the often tightly suggests normal in from also in encoding deviations frequency both information and That stimuli sensory magnitude systems. loading of controls track terms precisely engineering populations precision indispens- or osteocyte of an sensors is feature input having track able precisely Indeed, that essential loading. ultimately, systems an and, measurement mechanical mechanosensing is bone to osteocytes of adaptation control responding the between of of coupling feature tight number that and indicate loading data load- These that upon and frequency. dependent magnitude ing also strain were to curves correlated load–response respond- strongly osteocyte osteocytes was of load number to that ing found We vivo. in responses vascular once markedly vivo removed. is and in to pressure rapidly of responses change osteocyte study loading that our observed mechanical in we oxida- Indeed, effects, and postmortem (20). transport, vs. osteocytes solute on 19), will 18, stresses which (8, tive environment, pressure of vascular fluid removal normal LCS include osteo- alter its likely on from is factors bone occur studies contributing explant differences but our these clear, of Why yet substantially. mechan- results not differ the The vivo than in rate (10). cytes slower stimulus much input a at ical and duration longer of vrtepyilgclfeunyrneeaie xeietlyi the process in osteocyte experimentally examined on exponen- the range expected highlight studies. frequency strains present to physiological membrane added the been focal over has in region increases various shaded at tial The frequency (5)]. Wang loading of al. model of et mathematical function the from a [adapted as magnitudes tissue-loading membrane cell process cyte 6. Fig. that found al. et Jing (16). osteocytes isolated for Ca al. by reported et mechanism the Thi with consistent signaling, purinergic 10

Osteocyte Process Ca osteocyte real-time about information here report We 2+ 15 Axial Strain10 (%) ek fmcaial oddotoye nepat were explants in osteocytes loaded mechanically of peaks 0 5 010 203040 physiological loading physiological rdce xa tana h rndcinstso h osteo- the on sites transduction the at strain axial Predicted 15 Loading frequencyLoading (Hz) 20 0.5 1.5 0 1 2 56 570 65 60 55 0 60 min post-mortem 5 (0.025%) 5 MPa 10 MPa 20 MPa (0.1%) n t1 i (Center min 15 at and ) (0.05%) 10 520 15 magnitude loading tissue-level Applied ei tal. et Lewis 2+ 2+ ) Downloaded by guest on September 26, 2021 0 igD ta.(04 nst nrclua acu siltosi sectsin osteocytes Mechanosensa- in (2012) S Weinbaum oscillations A, Vatsa calcium RG, Bacabac AD, intracellular Bakker J, situ Klein-Nulend 11. In (2014) al. et D, Jing 10. 2 uX,HoB akM u E(02 acu epnei sectcntok under networks osteocytic in response Calcium (2012) XE Guo M, Park B, Huo XL, Lu 12. oe oeapsto 3) odn rqec fet eeeaie as examined were effects frequency Loading (38). periosteal apposition on turning bone for woven reported (3,000 threshold examined the level approximates and strain activities highest running to The walking and 56). moderate (55, 2,000, from activities 1,000, physiological 500, normal of 250, teristic of magnitudes strain 3,000 to Ca in cyclically alter positioned not loaded was does were foot Isoflurane the and apparatus. accessed, loading was the surface Vivo. dorsal in MT3 Loading the thesia, Mechanical to Response Osteocyte control. were displacement studies under Loading region). performed (tensile points contact loading upper between in Loading multi- vivo. simultaneous in ( osteocytes during details of MT3 apparatus Instruments) of Bruker (Ultima; bending imaging three-point photon vivo in Apparatus. for Loading device Mechanical Vivo In tolui- with den- counterstained were osteocyte blue. (n dine which and (53) cross-sections, area middiaphyseal elsewhere bone thick detailed woven mm as per vs. (number bones Lamellar sity C57Bl/6 genotype). (µCT) WT micro-CT per using vs. bones by OtGP3 performed assess Bones. were Metatarsal to studies B6 mineralization and and OtGP3 of tural of Properties College Material and City Structural and Medicine of College Com- Use Einstein all York. and Albert and New Care both mice Animal female Institutional at the 16-wk-old mittees by used approved Studies were bred is background. were procedures which Mice promoter, C57BL/6 (52). Cre DMP1 a odontoblasts the and onto possess of osteocytes in fragment which expressed 10-kb dominantly a Labs], by driven JAX recombinase [B6N.FVB-Tg(Dmp1-cre)1Jqfe/BwdJ; mice Ai38 mice mating DMP1/Cre by with Labs] obtained JAX were [B6;129S-Gt(ROSA)26Sortm38(CAG-GCaMP3)Hze/J; OtGP3) (designated GCaMP3. GCaMP3 targeted Osteocyte-Targeted Expressing Mice in found be can Methods. methods and study Materials and SI development device regarding Details Methods and Materials osteoporosis. as such predic- diseases even or skeletal characteristic of be tive may recruit- input osteocyte mechanical between and couplings ment the disrupt that alterations ei tal. et Lewis .Vrrge W aga J caaaL 21)Fudflwi h osteocyte the in flow Fluid (2013) Flow-induced LM bone: McNamara in TJ, transport Vaughan solute SW, and Verbruggen Fluid 9. (2009) S Weinbaum orches- SP, Master Fritton Osteocytes: (2013) 8. O Kennedy RJ, signaling. Majeska WY, Wnt Cheung and MB, Schaffler mechanosensing Osteocytes, 7. (2008) ML of Johnson role LF, the Bonewald for model 6. A (2007) S Weinbaum MB, Schaffler LM, McNamara Y, involves Wang bone 5. fatigue-loaded in resorption of Activation (2012) al. et follow- OD, resorption Kennedy bone of control 4. and apoptosis Osteocyte (2010) al. et KB, in Emerton increase 3. the prevents apoptosis osteocyte of Inhibition (2015) al. et LI, Plotkin 2. unload- Hindlimb by caused apoptosis Osteocyte (2016) al. et P, Cabahug-Zuckerman 1. sectswr iulzdi middiaphyses in visualized were Osteocytes Methods. and Materials SI inadtasuto nosteocytes. in transduction and tion loading. mechanical dynamic under bones 1592. long mouse intact approach. interaction fluid–structure Mechanobiol A environment: mechanical mechanotransduction. bone. of trators Bone osteocytes. in 104:15941–15946. mechanotransduction induced flow in integrins popu- osteocyte distinct by signaling lations. pro-osteoclastogenic active and apoptosis both mice. in ovariectomy ing unloading. by induced bone of loss 18942. the or resorption factor bone nuclear of activator receptor osteocytic of resorption femurs. mice subsequent in and bone production Trabecular RANKL and osteocyte cortical trigger to required is ing tayadoclaoyfli flow. fluid oscillatory and steady tan pto up Strains µ. 42:606–615. Bone 50:1115–1122. 13:85–97. acfTsu Int Tissue Calcif r presented are S3 ) (Fig. data calibrations strain and S2) Fig. 2 eemaue t400× at measured were ) nuRvFudMech Fluid Rev Annu ∼2,000 Bone 46:577–583. 94:5–24. µ Bone Bone cu ndahssi ioadaecharac- are and vivo in diaphyses in occur 51:466–473. 54:182–190. ecetdamcaia loading mechanical a created We 41:347–374. oeMnrRes Miner Bone J iad(AK)btde o stop not does but (RANKL) ligand κB ieehbtn osteocyte- exhibiting Mice anfiainfo 5-µm- from magnification stpclo extreme of typical is µ) 2+ ne sflrn anes- isoflurane Under inln 5) Bones (54). signaling ilChem Biol J rcNt cdSiUSA Sci Acad Natl Proc 31:1356–1365. AE J FASEB imc Model Biomech 290:18934– 28:1582– Struc- 6 = 3 aosC,Tmysti ,Csil B(00 sect ehnbooyadpericel- and signal- mechanobiology Osteocyte calcium (2010) AB Castillo intracellular S, Temiyasathit of CR, Jacobs properties 23. Spatiotemporal (2013) al. et D, Jing 22. mediates mechanism ATP-dependent An (2010) XE Guo Q, Xu KD, Costa XL, Lu B, differ- Huo Regional 21. (2016) MB Schaffler RJ, Majeska space J, pore Basta-Pljakic hierarchical D, the in Frikha-Benayed flow interstitial 20. and Blood (2015) L venous Cardoso to induced due SC, adaptation Cowin flow bone On 19. fluid (2003) SC Dynamic Cowin S, (2015) Weinbaum SP, YX Fritton L, Qin Wang J, 18. Jiao DE, Gibbons GW, Tian M, Hu Mechanosen- 17. (2013) DC Spray S, Weinbaum MB, and Schaffler SO, oxide Suadicani nitric MM, of Thi production 16. The (2001) J Klein-Nulend K, Soejima by AD, production prostaglandin Bakker increases 15. flow fluid Pulsating (1996) al. et NE, Ajubi extra- 14. and stores Ca2+ Intracellular (1996) CT Brighton SR, Pollack FD, Allen CT, Hung 13. ihFg .Ti okspotdb I rnsA011,AR057139, AR041210, Grants NIH by supported work assistance for This DK081435. and Fritton 6. DK091466, Susanah Dr. Fig. and with microscopy multiphoton with tance ACKNOWLEDGMENTS. IBM). 20; (Version SPSS anal- with regression performed or ANOVA testing were either hoc Analyses post using yses. for by difference examined squared were least Fisher’s groups with strain among level the strain using vs. by assessed were strains Analyses. Statistical exhibited they loading. during if intensity responding fluorescence normalized as Accord- in 60). considered increase (59, intensity were fluorescent osteocytes in signal- change ingly, lower fold a potential have lower a GECIs thus, other Ca period and exogenous 30-s than GCaMP3 a ratio loading. for to-noise intensity of mean start its the to normalized before was Fluorescence cell (NIH). each ImageJ for using intensity images postprocessing by performed were Ca ROI. the within located tion plane 6 optical single at a acquisition in ∼20 imaged filter were bandpass 40× Osteocytes 560-nm second. a per to frames using 490- 920-nm By with and performed MPM. excitation was wavelength using imaging objective, by immersion visualized water nification was loading bone whole death; studies Imaging. after explant min bone 60 in and reported 15 times 58). 17, at incubation (10, repeated approximates were min imaging 60 and loading and Ca additional from cyte MT3 vivo. in (n response mice osteocyte to contributes pressure lar study. loading the of euthanized end were imag- the Mice from observed. at adjacent response was the osteocyte effect in sequential one at No offset at strain sequence. or loading conditioning lowest strain potential to highest assess to highest to site from lowest ing then from was and going procedure site level, This imaging (57). strain cells each bone min, for 15 for for repeated period rest refractory to allowed the and at exceeds unloaded period then which loading was 60-s bone The a strain. for test continuously each collected were strain (see Images measurable studies details). no for calibration effectively strain had on they based because gradients middiaphysis. chosen the of were side ROIs either on These (n immediately Hz located 2 ROIs two or in 1, acquired 0.5, at tested groups with well, nadto,w sdti xeietlsse oass hte vascu- whether assess to system experimental this used we addition, In n notoyi n sebatccl ewrsudrfli flow. fluid mechanics. lular under networks cell osteoblastic and 540. osteocytic in ing nanoindenta- cell single under network cell bone tion. in signaling calcium intercellular osteo- bone cortical among activity cytes. mitochondrial and metabolism oxidative in ences tissue. bone of architecture stasis. oscillations. calcium Biophys osteocyte situ and in of modulation processes mechanobiological along occur require forces and physiological 21017. body to cell osteocytes not of responses dependent. stress sory shear is cells bone primary by 671–677. 2 E prostaglandin Commun process. cytoskeleton-dependent osteocytes–a chicken cultured experiencing cells bone of response Ca2+ real-time flow. the fluid in required are Ca2+ cellular eo h eiselsraet ananuiomsri distribu- strain uniform maintain to surface periosteal the below µm elCalcium Cell 2+ ae)wr oddt igets tan(2,000 strain test single a to loaded were males) 3 = Bone Biomech J sect Ca Osteocyte 579:55–61. epnewsmaue.Mc eete uhnzdi place, in euthanized then were Mice measured. was response 225:62–68. Biomech J 90:15–22. nuRvBoe Eng Biomed Rev Annu 36:1439–1451. 47:234–241. PNAS The 29:1411–1417. 2+ 2+ etakD.Ara orge-otea o assis- for Rodriguez-Contreras Adrian Dr. thank We Tadhsooia ifrnebtenmouse between difference histological and µCT nest esrmnsfo sectsobserved osteocytes from measurements intensity | epnei T oe fOG3mc during mice OtGP3 of bones MT3 in response Biomech J α coe 1 2017 31, October Vβ integrin. 3 2+ t esn ys( s 2 epciey,and, respectively), 12, vs. (5 dyes sensing 48:842–854. et ifrne notoyeresponse osteocyte in Differences test. 12:369–400. rcNt cdSiUSA Sci Acad Natl Proc e rqec) mgswere Images frequency). per 6 = | o.114 vol. IMtrasadMethods and Materials SI | ice ipy Res Biophys Biochem o 44 no. ,adosteo- and µ), Biomech J Bone rhBiochem Arch 110:21012– | 53:531– >25% 11779 mag- 34:

PHYSIOLOGY ENGINEERING 24. Thi MM, Urban-Maldonado M, Spray DC, Suadicani SO (2010) Characterization of 45. Nawaz S, et al. (2012) Cell visco-elasticity measured with AFM and optical trapping at hTERT-immortalized osteoblast cell lines generated from wild-type and connexin43- sub-micrometer deformations. PLoS One 7:e45297–e45299. null mouse calvaria. Am J Physiol-Cell Physiol 299:C994–C1006. 46. Skerry TM, Bitensky L, Chayen J, Lanyon LE (1989) Early strain-related changes in 25. Zaman G, et al. (1999) Mechanical strain stimulates nitric oxide production by rapid enzyme activity in osteocytes following bone loading in vivo. J Bone Miner Res 4: activation of endothelial nitric oxide synthase in osteocytes. J Bone Miner Res 783–788. 14:1123–1131. 47. Inaoka T, et al. (1995) Sequential analysis of expression after an osteogenic 26. Xu H, et al. (2012) Oscillatory fluid flow elicits changes in morphology, cytoskeleton stimulus: c-fos expression is induced in osteocytes. Biochem Biophysical Res Commun and integrin-associated molecules in MLO-Y4 cells, but not in MC3T3-E1 cells. Biol Res 217:264–270. 45:163–169. 48. Rawlinson SC, Wheeler-Jones CP, Lanyon LE (2000) Arachidonic acid for loading 27. Lu XL, Huo B, Chiang V, Guo XE (2012) Osteocytic network is more responsive in induced prostacyclin and prostaglandin E(2) release from osteoblasts and osteocytes calcium signaling than osteoblastic network under fluid flow. J Bone Miner Res 27: is derived from the activities of different forms of phospholipase A(2). Bone 27: 563–574. 241–247. 28. Takai E, Mauck RL, Hung CT, Guo XE (2004) Osteocyte viability and regulation of 49. Pereira AF, Javaheri B, Pitsillides AA, Shefelbine SJ (2015) Predicting cortical osteoblast function in a 3D trabecular bone explant under dynamic hydrostatic pres- bone adaptation to axial loading in the mouse tibia. J R Soc Interface 12: sure. J Bone Miner Res 19:1403–1410. 0590. 29. Kamel MA, Picconi JL, Lara-Castillo N, Johnson ML (2010) Activation of β-catenin sig- 50. Frost HM (1983) A determinant of bone architecture. The minimum effective strain. naling in MLO-Y4 osteocytic cells versus 2T3 osteoblastic cells by fluid flow shear stress Clin Orthop Relat Res®:286–292. and PGE2: Implications for the study of mechanosensation in bone. Bone 47:872–881. 51. Adachi T, et al. (2009) Osteocyte calcium signaling response to bone matrix deforma- 30. Kitase Y, et al. (2010) Mechanical induction of PGE2 in osteocytes blocks tion. J Biomech 42:2507–2512. glucocorticoid-induced apoptosis through both the β-catenin and PKA pathways. J 52. Lu Y, et al. (2007) DMP1-targeted Cre expression in odontoblasts and osteocytes. J Bone Miner Res 25:2657–2668. Dental Res 86:320–325. 31. Kramer I, et al. (2010) Osteocyte Wnt/-catenin signaling is required for normal bone 53. Cheung WY, et al. (2016) Pannexin-1 and P2X7-receptor are required for apoptotic homeostasis. Mol Cell Biol 30:3071–3085. osteocytes in fatigued bone to trigger RANKL production in neighboring bystander 32. Bellido TM, et al. (2005) Chronic elevation of parathyroid hormone in mice reduces osteocytes. J Bone Miner Res 31:890–899. expression of sclerostin by osteocytes: A novel mechanism for hormonal control of 54. Kim YS, et al. (2016) Coupled activation of primary sensory neurons contributes to osteoblastogenesis. Endocrinology 146:4577–4583. chronic pain. Neuron 91:1085–1096. 33. Robling AG, et al. (2008) Mechanical stimulation of bone in vivo reduces osteocyte 55. Burr DB, et al. (1996) In vivo measurement of human tibial strains during vigorous expression of Sost/sclerostin. J Biol Chem 283:5866–5875. activity. Bone 18:405–410. 34. Gu X, Spitzer NC (1995) Distinct aspects of neuronal differentiation encoded by fre- 56. Rubin CT, Lanyon LE (1984) Dynamic strain similarity in vertebrates; an alternative to quency of spontaneous Ca2+ transients. Nature 375:784–787. allometric limb bone scaling. J Theor Biol 107:321–327. 35. Sharma D, et al. (2012) Alterations in the osteocyte lacunar–canalicular microenviron- 57. Donahue SW, Donahue HJ, Jacobs CR (2003) Osteoblastic cells have refractory peri- ment due to estrogen deficiency. Bone 51:488–497. ods for fluid-flow-induced intracellular calcium oscillations for short bouts of flow 36. Ciani C, Sharma D, Doty SB, Fritton SP (2014) Ovariectomy enhances mechanical load- and display multiple low-magnitude oscillations during long-term flow. J Biomech 36: induced solute transport around osteocytes in rat cancellous bone. Bone 59:229–234. 35–43. 37. Forwood MR, Turner CH (1995) Skeletal adaptations to mechanical usage: Results 58. Ishihara Y, et al. (2013) Ex vivo real-time observation of Ca2+ signaling in liv- from tibial loading studies in rats. Bone 17:197S–205S. ing bone in response to shear stress applied on the bone surface. Bone 53:204– 38. Turner CH, Forwood MR, Rho JY, Yoshikawa T (1994) Mechanical loading thresholds 215. for lamellar and woven bone formation. J Bone Miner Res 9:87–97. 59. Mank M, Griesbeck O (2008) Genetically encoded calcium indicators. Chem Rev 39. Hsieh YF, Turner CH (2001) Effects of loading frequency on mechanically induced bone 108:1550–1564. formation. J Bone Miner Res 16:918–924. 60. Hagen BM, Boyman L, Kao JPY, Lederer WJ (2012) A comparative assessment of fluo 40. Robling AG, Hinant FM, Burr DB, Turner CH (2002) Improved bone structure and Ca2+ indicators in rat ventricular myocytes. Cell Calcium 52:170–181. strength after long-term mechanical loading is greatest if loading is separated into 61. Nakai J, Ohkura M, Imoto K (2001) A high signal-to-noise Ca2+ probe composed of a short bouts. J Bone Miner Res 17:1545–1554. single green fluorescent . Nat Biotechnol 19:137–141. 41. Forwood MR, Burr DB (1993) Physical activity and bone mass: Exercises in futility? 62. Ohkura M, Matsuzaki M, Kasai H, Imoto K, Nakai J (2005) Genetically encoded bright Bone Miner 21:89–112. Ca 2+probe applicable for dynamic Ca 2+imaging of dendritic spines. Anal Chem 42. Salvi JD, Maoileidigh´ DO,´ Fabella BA (2015) Control of a hair bundle’s mechanosen- 77:5861–5869. sory function by its mechanical load. Proc Natl Acad Sci USA 112:E1000–E1009. 63. Tsien RY (2009) Indicators based on fluorescence resonance energy transfer (FRET). 43. Dick IE, et al. (2008) A modular switch for spatial Ca2+ selectivity in the Cold Spring Harb Protoc 2009:pdb.top57. regulation of CaV channels. Nature 451:830–834. 64. Schriefer JL, et al. (2005) A comparison of mechanical properties derived from multi- 44. You L, Cowin SC, Schaffler MB, Weinbaum S (2001) A model for strain amplification ple skeletal sites in mice. J Biomech 38:467–475. in the actin cytoskeleton of osteocytes due to fluid drag on pericellular matrix. J 65. Chen TW, et al. (2013) Ultrasensitive fluorescent for imaging neuronal activ- Biomech 34:1375–1386. ity. Nature 499:295–300.

11780 | www.pnas.org/cgi/doi/10.1073/pnas.1707863114 Lewis et al. Downloaded by guest on September 26, 2021