You are on page 1of 8

ORIGINAL ARTICLE

Journal of
1044
Cellular
Physiology
Blockade of TNFR1 Signaling:
A Role of Oscillatory Fluid Shear
Stress in Osteoblasts
HAIFANG WANG,1,2 SUZANNE R. YOUNG,1 RITA GERARD-O’RILEY,1 JULIA M. HUM,1
ZHOUQI YANG,1,2 JOSEPH P. BIDWELL,3 AND FREDRICK M. PAVALKO1*
1
Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana
2
Key Laboratory for Space Bioscience and Biotechnology, Faculty of Life Sciences, Northwestern Polytechnical University,
Shaanxi, China
3
Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana

Fluid shear stress protects cells from TNF-a-induced apoptosis. Oscillatory fluid shear stress (OFSS) is generally perceived as
physiologically relevant biophysical signal for bone cells. Here we identify several cellular mechanisms responsible for mediating the
protective effects of OFSS against TNF-a-induced apoptosis in vitro. We found that exposure of MC3T3-E1 osteoblast-like cells to as little
as 5 min of OFSS suppressed TNF-a-induced activation of caspase-3, cleavage of PARP and phosphorylation of histone. In contrast, H2O2-
induced apoptosis was not inhibited by OFSS suggesting that OFSS might not be protecting cells from TNF-a-induced apoptosis via
stimulation of global pro-survival signaling pathways. In support of this speculation, OFSS inhibition of TNF-a-induced apoptosis was
unaffected by inhibitors of several pro-survival signaling pathways including pI3-kinase (LY294002), MAPK/ERK kinase (PD98059 or
U0126), intracellular Ca2þ release (U73122), NO production (L-NAME), or protein synthesis (cycloheximide) that were applied to cells
during exposure to OFSS and during TNF-a treatment. However, TNF-a-induced phosphorylation and degradation of IkBa was blocked
by pre-exposure of cells to OFSS suggesting a more specific effect of OFSS on TNF-a signaling. We therefore focused on the mechanism of
OFSS regulation of TNF-receptor 1 (TNFR1) signaling and found that OFSS (1) reduced the amount of receptor on the cell surface, (2)
prevented the association of ubiquitinated RIP in TNFR1 complexes with TRADD and TRAF2, and (3) reduced TNF-a-induced IL-8
promoter activity in the nucleus. We conclude that the anti-apoptotic effect of OFSS is not mediated by activation of universal pro-survival
signaling pathways. Rather, OFSS inhibits TNF-a-induced pro-apoptotic signaling which can be explained by the down-regulation of
TNFR1 on the cell surface and blockade of TNFR1 downstream signaling by OFSS.
J. Cell. Physiol. 226: 1044–1051, 2011. ß 2010 Wiley-Liss, Inc.

Mechanical loading of the skeleton plays a key role in regulating linked to the cytoskeleton and can be mobilized from
the health of bone tissue by maintaining a proper balance membrane structures such cadherin-mediated cell–cell
between resorption of existing bone and deposition of new junctions or integrin-containing focal adhesions (Pavalko et al.,
bone (Turner and Pavalko, 1998). A critical component in 1998; Ponik and Pavalko, 2004) and may involve kinases such as
mechanoregulation of bone is the movement of interstitial fluid focal adhesion kinase (FAK) (Young et al., 2009),
through the spaces within bone that generates fluid shear stress nucleocytoplasmic shuttling molecules, and transcriptional
(FSS) across the surfaces of bone cells (Piekarski and Munro, regulators such as Nmp4, NFkB, and b-catenin (Chen et al.,
1977). FSS functions as an external mechanical stimulus that can 2003; Norvell et al., 2004; Yang et al., 2010; Young et al., 2010).
activate cytoplasmic signaling molecules to produce changes in The in vitro application of FSS induces a number of responses in
bone metabolism through a process known as cellular osteoblastic cells, including intracellular calcium mobilization
mechanotransduction, recently reviewed in (Huang and (Hung et al., 1995; Jacobs et al., 1998; Donahue et al., 2001) and
Ogawa, 2010). Many in vitro models use unidirectional or activation of MAPKs (You et al., 2001), PI3K/AKT activation
steady fluid shear stress (SFSS) to study the response of cells to (Pavalko et al., 2003a), NF-kB activation (Chen et al., 2003;
fluid movement. While this is appropriate for endothelial cells Young et al., 2010), production of nitric oxide (Klein-Nulend
that normally experience unidirectional flow in blood vessels, et al., 1995; Johnson et al., 1996), and cytoskeleton
given the cyclic nature of normal skeletal loading, we and others reorganization (Pavalko et al., 1998; Chen et al., 2000). These
have suggested that oscillatory fluid shear stress (OFSS) may be are all generally associated with anti-apoptotic signaling. Our
a more physiological biophysical signal than SFSS for bone cells studies demonstrate for the first time that these traditional
(Jacobs et al., 1998; Kurokouchi et al., 2001; Ponik et al., 2007). anti-apoptotic pathways are not responsible for mediating the
Our previous work indicated that exposure of osteoblasts to beneficial effects of OFSS against TNF-a-induced apoptosis.
SFSS protected cells from TNF-a-induced apoptosis (Pavalko
et al., 2003a). To better understand the molecular mechanisms
responsible for the anti-apoptotic effects of FSS, we have Contract grant sponsor: NIH;
extended these studies to include consideration of the effect of Contract grant numbers: R01AR052682, AR056188, DK053796.
OFSS on TNF-a-induced apoptosis in osteoblasts. *Correspondence to: Fredrick M. Pavalko, Department of Cellular
We have proposed that mechanical loading of cells regulates and Integrative Physiology, Indiana University School of Medicine,
the formation and function of membrane-associated multi- 635 Barnhill Dr., MS346A, Indianapolis, IN 46202. E-mail:
protein complexes, referred to as mechanosomes, that affect fpavalko@iupui.edu
cellular responses to external mechanical stimuli (Pavalko et al., Received 27 August 2010; Accepted 31 August 2010
2003b). This hypothesis predicts the integration of diffusion- Published online in Wiley Online Library
controlled signaling pathways with a solid-state scaffold directly (wileyonlinelibrary.com), 20 September 2010.
linking extracellular mechanical stimuli at the cell membrane to DOI: 10.1002/jcp.22427
the genes. These multi-protein membrane complexes may be

ß 2 0 1 0 W I L E Y - L I S S , I N C .
SHEAR STRESS INDUCED BLOCK OF TNFR1 SIGNALING 1045

Instead, OFSS, rather than simply generating pro-survival washed twice with ice-cold PBS and incubated with 1 mg/ml of EZ
signals, appears to inhibit several processes specifically involved link NHS-Sulfo-SS-biotin in PBS for 30 min at 48C. The reaction was
in TNF-a signaling via TNF receptor 1 (TNFR1) including down- quenched by two washes with cold quenching buffer. The cells
regulation of surface expression of TNFR1 and inhibition of were then lysed in 800 ml of lysis buffer (50 mM Tris–HCl, pH 7.4,
signaling downstream of TNFR1 including activation of the 500 mM NaCl, 1 mM EDTA/EGTA, 0.5% SDS, 1% Triton X-100,
TNF-a target gene, IL-8. Thus, these results suggest blockade of and 0.1% NP-40) containing fresh protease inhibitors (1 mM
death-induced signaling via TNFR1 by OFSS may represent pepstatin A, 250 mM phenylmethylsulfonyl fluoride, 1 mg/ml
another example of the ability of a mechanical stimulus to affect leupeptin, and 1 mg/ml aprotinin) and disrupted by sonicating for
the assembly and function of an important cell surface- 40 s on ice. Lysates were centrifuged at 10,000g for 10 min at 48C,
associated multi-protein signaling complex, or mechanosome, and supernatants were incubated with Neutravidin beads (100 ml)
which ultimately results in modulation of genes. for 1.5 h at 48C. Beads were washed three times with fresh lysis
buffer, and bound proteins were eluted with 80 ml of 2 Western
Materials and Methods blotting sample buffer for 6 min at 1008C. Aliquots from total cell
Cell culture lysates (40 ml) and Neutravidin-bound samples (40 ml) were
analyzed by Western blotting analysis. Biotinylated TNFR1 protein
The mouse osteoblast-like MC3T3 cell line was obtained from the (representing the surface TNFR1) was normalized using levels of
American Type Culture Collection and cultured in minimal cadherin on the cell surface, and values were averaged across three
essential media alpha (a-MEM, Gibco, Life Technologies, Carlsbad, experiments. To analyze TNF-a-induced TNFR1 internalization,
CA) supplemented with 10% fetal calf serum (FCS) and 1% cells were subjected to 2 h of static culture or flow condition and
penicillin/streptomycin (Gibco, Life Technologies). Cells were then treated with 10 ng/ml TNF-a for 1 and 5 min at 378C, or 4 and
maintained in 5% CO2 at 378C. For the induction of apoptosis, cells 10 min in 48C cold media at room temperature to allow for
were treated with 10 ng/ml of TNF-a (Calbiochem, San Diega, CA) internalization of TNFR1. Then cells were cooled rapidly to 48C to
and 10 mg/ml of cyclohexamide (CHX, Sigma–Aldrich, St. Louis, inhibit additional internalization, washed twice with ice-cold PBS
MO) for 4 h. Apoptosis was evaluated by observing the cellular and subjected to surface protein isolation.
morphologic changes under light microscope and Western blotting
analyses of caspase-3 processing, PARP cleavage, and histone Analysis of TNFR1–TRADD–RIP complex by co-
phosphorylation. immunoprecipitation

Oscillatory fluid flow The complexes initiated upon TNF-a stimulation were analyzed by
use of a rabbit TRADD antibody (Santa Cruz) or a mouse TNFR1
Before flow experiments, MC3T3 cells were passed onto sterile antibody. MC3T3 cells (1  107 cells) grown in culture dishes or on
glass slides at a density of 2.5  105 cells per slide, grown for 24 h glass slides were subjected to 2 h static or flow condition,
and serum-starved in a-MEM media supplemented with 0.1% FCS stimulated for the indicated times with 10 ng/ml TNF-a and then
and 1% penicillin/streptomycin for 18–22 h. The a-MEM media lysed in 1 ml co-IP lysis buffer (50 mM Tris–HCl, pH 7.4, 150 mM
supplemented with 0.1% FCS and 1% penicillin/streptomycin was NaCl, 0.5% NP40, 10% glycerol, 1 mM EDTA/EGTA) containing
also used for flow experiments. Oscillatory fluid flow was fresh protease inhibitors for 25 min on ice. Detergent-insoluble
performed in a parallel plate flow chamber at 378C using a material was removed by centrifugation at 10,000g for 10 min at
previously described fluid flow device. This system subjected cells 48C. The supernatants were pre-cleared with 40 ml Protein A
to 10–12 dyn/cm2 of OFSS at a frequency of 1 Hz. Hard-walled PVC beads (Sigma) for 1 h at 48C and incubated with rabbit anti-TRADD
tubing was used to connect the chamber to the pump. Static or mouse anti-TNFR1 antibody for 2 h. The immune complexes
control cells were held in a-MEM containing 0.1% serum at 378C were then precipitated with 40 ml Protein A beads (Sigma) for
with 5% CO2. 3 h at 48C. Beads were recovered by centrifuging at 10,000g
for 3 min and washed four times with 500 ml of lysis buffer. The
Western blotting analysis and antibodies beads-bound proteins were eluted with 80 ml of Western blotting
Cells extracts were collected directly in SDS sample buffer and sample buffer for 6 min at 1008C and subjected to immunoblotting
protein concentrations were determined using the amido black analysis.
method. Equal amounts of protein (20 mg) were loaded onto SDS– IL-8 reporter assay
PAGE gels for separation and transferred to nitrocellulose for
immunoblotting. The following primary antibodies were used: Osteoblasts grown to 80% confluency in 10 cm dishes were
caspase-3/cleaved caspase-3 (Cell Signaling Technology, Danvers, transfected with 6 mg of the IL-8 promoter-firefly luciferase
MA), Parp (Santa Cruz Biotechnology, Santa Cruz, CA), ph-histone construct using FuGENE per manufacture’s protocol. Transfected
and vinculin (Sigma–Aldrich), ph-IkBa/IkBa (Cell Signaling cells were incubated over night and the next day were plated for
Technology), TNFR1 (Abcam, Cambridge, MA), cadherin-11 (Life TNF-a treatment or exposure to OFFS. Luciferase activities were
Technologies, Carlsbad, CA), receptor-interacting protein (RIP, determined using at Luciferase Assay Kit (Promega, Madison, WI)
BD Biosciences, San Jose, CA), TRADD (Santa Cruz according to the manufacture’s protocol. Data are presented as
Biotechnology), ph-Erk1/2/Erk1/2 (Santa Cruz Biotechnology). fold change in activity relative to untreated static controls.
The appropriate anti-mouse Ig and anti-rabbit Ig peroxidase- Graphical data is representative of three independent trials.
conjugated secondary antibodies (Jackson Immunoresearch
Laboratories, West Grove, PA) and anti-goat Ig (Santa Cruz Statistical analysis
Biotechnology) were used. The second antibody signals were
detected and quantified using a Luminescent Image Analyzer LAS- Statistical significance was assessed by one-tailed t-tests or analysis
3000 system (Fujifilm Life Science, Stamford, CT). of variance as indicated and a P-value of <0.05 was interpreted as
statistically significant.
Analysis of cell surface TNFR1 protein
Results
Cell surface protein biotinylation and purification were performed Oscillatory fluid shear stress inhibits TNF-a-induced, but
using the Pierce Cell Surface Protein Isolation Kit, and the amount not H2O2-induced apoptosis
of TNFR1 protein was quantified by Western blotting analysis.
MC3T3 cells grown in 10 cm dishes or on glass slides were To evaluate the relevant molecular mechanisms involved in
subjected to static or flow condition for 2 h, then the cells were mediating the effect of OFSS on TNF-a-induced apoptosis in

JOURNAL OF CELLULAR PHYSIOLOGY


1046 WANG ET AL.

osteoblasts in vitro, MC3T3 cells were subjected to OFSS for a activation of caspase-8, which is the initiator of TNFR1-
very brief period (5 min) to longer periods (2 h), followed by mediated apoptosis and is upstream of caspase-3. We found
treatment with 10 ng/ml TNF-a and 10 mg/ml CHX for 4 h. that cleavage of caspase-8 was almost completely abolished by
Apoptosis was evaluated by observing the cellular morphologic application of OFSS suggesting that initiation of TNFR1 signaling
changes under light microscope and Western blotting analyses might be blocked by OFSS (Fig. 1C).
of caspase-3 activation, PARP cleavage, and histone
phosphorylation. This OFSS time-course study indicated that Inhibition of MEK/ERKs pathway, pI3K/AKT pathway,
OFSS inhibits TNF-a-induced caspase-3 activation in a time- cytoskeleton reorganization, intracellular calcium
dependent manner. A significant inhibitory effect was achieved mobilization, new protein synthesis, or nitric oxide
with very brief exposure to OFSS (5 min) and almost complete production did not attenuate OFSS-induced anti-
inhibition was achieved with 2 h of flow (Fig. 1A). We found that apoptosis
typical apoptotic morphologic signs induced by TNF-a,
including cell shrinkage and rounding, surface blebbing, and To evaluate the potential role of several well-established pro-
apoptotic bodies were completely abolished by pre-exposing survival signaling pathways in mediating the protective effect of
cells to 2 h of OFSS (Fig. 1B). Western blotting analyses showed OFSS against TNF-a-induced apoptosis, MC3T3 cells were
that cleavage of caspase-3 and PARP were almost completely treated with an effective dose of selective inhibitors of ERK
abolished and phosphorylation of histone H2A.X was (50 mM PD98059 or 20 mM U1026), or PI3K/AKT signaling
significantly reduced by 2 h of OFSS (Fig. 1C). In contrast, when (50 mM LY294002), cytoskeleton reorganization (10 mM
apoptosis was induced by 250 mM H2O2, 2 h of OFSS had no colchicine or nocodazole for tubulin cytoskeleton and 4 mM
inhibitory effect on apoptosis (Fig. 1D). These results suggested cytochalasin B or D for actin filaments), phospholipase C (PLC)-
a protective effect of OFSS specifically against TNF-a-induced mediated intracellular Ca2þ release (5 mM U73122), new
cell apoptosis. We also examined the effect of OFSS on protein synthesis (10 mg/ml CHX), or nitric oxide production

Fig. 1. Comparison of effects of OFSS on TNF-a (A–C) and H2O2 (D)-induced apoptosis in MC3T3 cells. A: Time-dependent inhibition of OFSS on
TNF-a-induced activation of caspase-3. B: Prevention of TNF-a-induced apoptotic morphological changes by 2 h of OFSS. C: Inhibition of TNF-a-
induced activation of caspase-3, cleavage of Parp, phosphorylation of histone, and activation of caspase-8 by 2 h of OFSS. D: H2O2-induced histone
phosphorylation was not affected by 2 hof OFSS. MC3T3 cells under static condition or exposed to 2 hof OFSS were treated with 10 ng/ml TNF-a for
4 h or 250 mM H2O2 for 2 h, cell pictures were taken and then samples were collected and subjected to Western blot analysis (aP < 0.05, bP < 0.01 vs.
static plus TNF-a treatment).

JOURNAL OF CELLULAR PHYSIOLOGY


SHEAR STRESS INDUCED BLOCK OF TNFR1 SIGNALING 1047

(250 mM L-NAME) during the 2 h period of time cells were MC3T3 cells were first subjected to 2 h of OFSS, and then
exposed to OFSS and during subsequent treatment with TNF-a treated with TNF-a for 5, 10, 15, and 30 min. Phosphorylation
for 4 h. Surprisingly, none of these inhibitors showed any ability and degradation of IkBa, which represent activation of NF-kB
to block the protective effect of OFSS against TNF-a-induced signaling pathway, were analyzed by Western blotting. Under
apoptosis as assessed by morphological changes (not shown) or static culture conditions, phosphorylation, and degradation of
as measured by activation of caspase-3 (Fig. 2). IkBa induced by TNF-a was observed as early as 5 min post-
stimulation. In contrast, phosphorylation of IkBa and
Blockade of TNF-a-induced IkBa signaling by 2 h of degradation of IkBa were inhibited by pre-exposure of the
OFSS MC3T3 cells to 2 h of OFSS (Fig. 3). These results suggest that
In addition to activating a pro-apoptotic signals, TNF-a also OFSS may inhibit the initiation and progression of TNF-a-
stimulates NF-kB signaling which is mediated by induced TNFR1-mediated signaling.
phosphorylation and subsequent degradation of inhibitor-kB Two hours of OFSS reduced expression level of TNFR1
(IkBa) (Chen and Goeddel, 2002). As expected, and as we have on cell surface but did not prevent TNF-a-induced
shown previously (Young et al., 2010), application of OFSS internalization of TNFR1
resulted in IkBa degradation within 30 min of the onset of flow,
with IkBa levels recovering to pre-treatment values after 2 h of We next investigated the effect of 2 h of OFSS on upstream
continuous flow (not shown). Because IkBa levels returned to initiation of TNFR1 signaling by TNF-a by measuring cell surface
normal after 2 h of OFSS, we were able to assess the effect of expression of TNFR1 in response to OFSS and by assessing
OFSS on TNF-a-activation of the NF-kB signaling pathway. TNF-a-induced receptor internalization. First, MC3T3 cells

Fig. 2. Inhibitory effect of 2 h OFSS on TNF-a-induced caspase-3 activation was not reduced by treating cells with inhibitors of ph-ERKs pathway
(50 mM PD98059 or 20 mM U1026, A), PI3K/ph-AKT pathway (50 mM LY294002, B), or cytoskeleton reorganization (10 mM colchicine or
nocodazole for tubulin cytoskeleton and 4 mM cytochalasin B or D for actin filaments, C), intracellular calcium mobilization (5 mM U73122 for
effective inhibition of phospholipase C, D), new protein synthesis (10 mg/ml cyclohexymide, E), or NO synthesis (250 mM L-NAME, E) during the
whole flow period. MC3T3 cells were treated with individual inhibitor under static condition or during the 2 h of flow experiment, then cells were
treated with 10 ng/ml TNF-a for 4 h and samples were subjected to Western blot analysis.

JOURNAL OF CELLULAR PHYSIOLOGY


1048 WANG ET AL.

maintained in static culture and subjected to OFSS, the level of


TNFR1 on the cell surface was significantly reduced (35%) by
OFSS (Fig. 4A). We next treated cells at 378C with TNF-a to
induce receptor internalization. As expected, treatment with
TNF-a resulted in rapid loss of TNFR1 from the cell surface that
was significant within 5 min as the receptor was rapidly
internalized. Importantly, TNFR1 surface expression did not
recover during the following 60 min (Fig. 4B). Because TNF-a
induced such a rapid internalization of TNFR1 receptors, we
attempted a similar experiment at 48C. We found that TNF-a-
induced internalization of TNFR1 was completely blocked at
10 min when cells were maintained at 48C demonstrating that
the rate of internalization of TNFR1 could be effectively
reduced by treating cells with TNF-a in cold media (Fig. 4B). We
Fig. 3. Blockade of TNF-a-induced phosphorylation and took advantage of this fact to measure the effect of OFSS on
degradation of IkBa by 2 h of OFSS. MC3T3 cells under static
condition or subjected to 2 h of OFSS were treated with 10 ng/ml
TNF-a-induced receptor internalization as shown in
TNF-a for 5, 10, 15, or 30 min. IkBa and phosphorylated IkBa were Figure 4C. The upper part (378C) shows that, at 1 and 5 min
analyzed by Western blot. Vinculin was analyzed as a loading control. post-TNF-a treatment, surface expression of TNFR1 appeared
lower in cells pre-exposed to OFSS than in cells held in static
culture, although this difference was not statistically significant.
To better demonstrate this difference, we slowed down the
were exposed to 2-h OFSS, or maintained in static culture. The receptor internalization by treating MC3T3 cells with TNF-a in
amount of total TNFR1 in cell lysates and the amount of TNFR1 cold (48C) media. As shown in the bottom part of
specifically expressed on the cell surface was analyzed by Figure 4C (48C), a trend towards an increased rate of TNF-a-
Western blotting. While we found no significant difference in induced TNFR1 internalization between the two groups (static
the total levels of TNFR1 in cell lysates between cells vs. OFSS) was seen at both 5 and 10 min post-treatment,

Fig. 4. Effect of 2 h of OFSS on TNFR1 protein level at cell surface. A: Down-regulation of TNFR1 level on cell surface by 2 h of OFSS. MC3T3 cells
under static condition or exposed to 2 h of OFSS were immediately subjected to cell surface protein extraction and Western blot analysis for
TNFR1. B: TNF-a-induced internalization of TNFR1. Cells treated with 10 ng/ml TNF-a for 5, 10, 15, 30, or 60 min were subjected to cell surface
protein extraction and Western blot analysis. C: Western blot analysis of effects of OFSS on TNF-a-stimulated internalization of TNFR1. Cells
under static condition or pre-exposed to 2 h of OFSS were treated with 10 ng/ml TNF-a in warm (37-C, upper bands) or ice-cold (4-C) media at 37-C
as indicated time and then subjected to cell surface protein extraction and Western blot analysis. D: Effects of OFSS on TNF-a-induced
internalization of TNFR1. Cells under static condition or pre-exposed to 2 h of OFSS were treated with 10 ng/ml TNF-a in ice-cold (4-C) media at 37-
C, and then subjected to cell surface protein extraction and Western blot analysis. Cadherin-11 expressed on cell surface was used as a loading
control (aP < 0.05 vs. static plus TNF-a treatment).

JOURNAL OF CELLULAR PHYSIOLOGY


SHEAR STRESS INDUCED BLOCK OF TNFR1 SIGNALING 1049

although we were unable to demonstrate that this difference Inhibition of TNF-a-induced IL-8 promoter activity by
was statistically significant. Taken together, these results OFSS
indicated that TNF-a-induced TNFR1 internalization was
certainly not inhibited by pre-exposure to OFSS (Fig. 4D) and, The results described above predict that OFSS protects cells
although not statistically significant, actually trended towards a from TNF-a-induced apoptosis by down-regulating TRNR1
more rapid rate of internalization following pre-exposure to signaling. To test this directly, we used an IL-8 promoter activity
OFSS. assay to determine the effect of OFSS on TNF-a-induced IL-8
transcriptional activity in the nucleus of MC3T3-E1 cells. We
found that pre-exposure of cells to OFSS significantly reduced
Inhibition of ubiquitinated RIP in TNF-a-induced TNF-a-induced IL-8 activation compared to cells that were
membrane complexes by 2 h of OFSS maintained in static culture (Fig. 6). As expected, and as we have
reported previously (Young et al., 2010), OFSS alone induced a
Next we evaluated the effect of OFSS on TNF-a-induction of significant increase in IL-8 reporter activity. This result strongly
NFkB-mediated signaling by assessing association of supports the hypothesis that OFSS regulates the assembly and
ubiquitinated RIP, an important component of the TNF-a- activity of the multi-protein TNFR1 complex and that this
induced pro-survival signaling complex I that forms with regulation by OFSS at the cell membrane ultimately affects gene
TNFR1, TRADD, and TNF-associated factor 2 (TRAF2). We regulatory activity in the nucleus.
used co-immunoprecipitation experiments to pull down
signaling complex I components using either an anti-TRADD
antibody or an anti-TNFR1 antibody and then blotted for RIP Discussion
and ubiquitinated RIP, which migrates more slowly on SDS–
PAGE. MC3T3 cells were first subjected to 2 h of OFSS or In this study we have made several novel observations and
maintained in static culture and then stimulated with TNF-a identified multiple specific mechanisms through which
(10 ng/ml) for 5 min or left untreated. Cells were then lysed, mechanical stimulation of osteoblast-like MC3T3-E1 cells by
TRADD or TNFR1 were immunoprecipitated, and the OFSS may promote cell survival. These specific mechanisms
presence of RIP and ubiquitinated RIP in the immune complex include (1) down-regulation of surface expression of TNFR1,
was assayed by Western blotting. As shown in Figure 5, TNFa- (2) inhibition of the association of ubiquitinted RIP in TRNR1/
induced association of ubiquitinated RIP in complexes pulled TRADD/TRAF2 complexes, (3) inhibition of NFkB signaling by
down with either TRADD or TNFR1 antibody was observed at blockade of TNF-aIkBa phosphorylation and subsequent
5 min post-TNF-a treatment in cells maintained in static degradation, and (4) inhibition of caspase-8 activation
culture. However, the amount of ubiquitinated RIP that was downstream of DISC formation (summarized in Fig. 7). Among
pulled down in co-immunoprecipitated complexes was several novel observations, we show for the first time the
significantly reduced in cells pre-exposed to 2 h of OFSS. Taken protective effect of a brief period of oscillatory (OFSS), as
together with the observation that the amount of non- opposed to continuous steady (SFSS) or unidirectional shear
ubiquitinated RIP co-immunoprecipitating in complexes stress, against TNF-a-induced apoptosis in osteoblastic cells.
following pre-exposure to 2 h of OFSS was unchanged, these Furthermore, and we think very importantly, we show that
results suggest that OFSS specifically inhibited the association of multiple classical pro-survival signaling pathways, including
ubiquitinated RIP with TNF-a-induced TNFR1/TRADD/ MAPK, AKT, PLC, and NO-dependent pathways, are
TRAF2 complexes. TRAF2-mediated ubiquitination of RIP has apparently not involved in mediating the anti-apoptotic effect of
been shown to be required for TNF-a-induced complex I OFSS. Further, we found that inhibition of TNF-a-induced
formation and NF-kB activation (Li et al., 2006). Therefore,
these results suggest that OFSS may inhibit TNF-a-induced
complex I formation and thereby prevent recruitment of Fas-
associated DD protein (FADD) and caspase-8 which would
ultimately prevent the formation of the death-inducing signaling
complex (DISC).

Fig. 6. Quantification of IL-8 promoter reporter assay expressed as


fold activation. MC3T3-E1 osteoblasts transfected with an IL-8
promoter-firefly luciferase construct were maintained in static
Fig. 5. Prevention of ubiquitination RIP by 2 h of OFSS. MC3T3 cells culture conditions (static) or exposed to 2 h of OFFS followed by a 4-h
under static condition or exposed to 2 h of OFSS were left untreated post-flow incubation period during which either TNF-a or vehicle
or stimulated with TNF-a for 10 min. Cells were lyzed, TRADD or alone was added. aSignificant difference OFFS versus static;
b
TNFR1 was immunoprecipitated and the presence of RIP and its significant difference static versus static R TNF-a; csignificant
modified form in the immune complex was detected by Western difference static RTNF-a versus OFSS R TNF-a by ANOVA
blotting analysis. ( P < 0.05). Error bars represent SEM.

JOURNAL OF CELLULAR PHYSIOLOGY


1050 WANG ET AL.

(Pavalko et al., 2003b). Interestingly, down-regulation of growth


factor/cytokine receptor signaling activity does not appear to
be a universal response to FSS. In fact, our group has recently
reported up-regulation of IGF-1 receptor signaling in response
to SFSS in MC3T3-E1 cells (Triplett et al., 2007) indicating that
the response of membrane receptors can differ depending on
the specific receptor, and perhaps, based on the type of
mechanical stimulus applied. This raises the question as to
whether there are ‘‘stop’’ mechanosomes that carry
information for attenuating bone formation, ‘‘go’’
mechanosomes that enhance bone growth, and finally whether
there is cross-talk between them within a network.
Studies using TRADD-deficient mice (Ermolaeva et al., 2008;
Pobezinskaya et al., 2008) indicate that, as predicted by
biochemical data, TRADD is absolutely necessary for TNF-
induced apoptosis and is also required for the recruitment of
TRAF2 to TNFR1 (Pobezinskaya et al., 2008).
Upon TNF-a stimulation, TRADD, RIP, and TRAF2 were
sequentially recruited to activated TNFR1 and formed a plasma
membrane bound complex (complex I), leading to activation of
NF-kB (Micheau and Tschopp, 2003). RIP is a serine/thronine
kinase that plays an essential role in TNF-a-induced NF-kB
Fig. 7. Model of blockade of TNFR1-mediated signaling by OFSS. activation through interaction with downstream signaling
components such as NEMO, the regulator subunit of the IKK
complex, and other molecules (Devin et al., 2000). TRAF2
catalyzed ubiquitination of RIP is required for TNF-a-induced
apoptosis did not require an intact microtubule or actin NF-kB activation (Li et al., 2006). Therefore, the failure of TNF-
cytoskeleton. a-induced NF-kB activation can be caused by the inhibition of
It is generally accepted that binding of TNF-a to TNFR1 on RIP ubiquitination. The loss of uniquitinated RIP from TNFR1
the plasma membrane rapidly triggers the recruitment of the complexes (complex I) following OFSS suggests that fluid shear
intracellular adapter protein TNFR1-associated death domain may be affecting the ability of TRAF2 to associate with the
containing protein (TRADD), RIP and TRAF2 to form an active complex. Previous studies in endothelial cells showed that FSS
complex I and signals activation of NF-kB. This is followed by inhibited TNF-a-induced activation of MAPK by destroying
dissociation of TRADD from TNFR1 and recruitment of FADD association of TRAF2 with TNFR1 (Yamawaki et al., 2003).
and caspase-8 to form a complex II, or the DISC which leads to Our group showed that the PI3-kinase inhibitor LY294002
cell apoptosis if new gene transcription induced by NF-kB is suppressed SFSS-induced inhibition of caspase-3 activation and
inhibited (Micheau and Tschopp, 2003). blocked the ability of SFSS to rescue osteoblasts from apoptosis
TNF-a-induced apoptosis is inhibited if the interactions (Pavalko et al., 2003a). However, LY294002 treatment did not
between TNFR1 and its downstream molecules are blocked affect inhibition of caspase-3 activation and anti-apoptosis
(He and Ting, 2002). Yang et al. (2005) reported that the induced by OFSS in MC3T3 cells. Notably, AKT phosphorylation
activation of TNF-a/IkBa/NF-kB signaling pathway was induced by OFSS was effectively inhibited by LY294002 but
inhibited by cardiac glycosides through a mechanism blocking SFSS-induced phosphorylation of Akt was not affected,
recruitment of TRADD, RIP, and TRAF2 to the TNFR1. suggesting different mechanisms may be involved in activation of
Gururaja et al. (2007) showed that a class of small-molecule signaling pathways by the two patterns of fluid flow.
triazoloquinoxalines were able to block ligand-dependent Importantly, OFSS inhibits TNF-a-induced IKK activation,
internalization of the TNFR1 and the association of TNFR1 with leading to a decrease in phosphorylation and degradation of
TRADD and RIP1, thus inhibiting both survival and death IkBa, which in turn resulted in decreased TNF-a-induced NF-
pathways induced by TNFa. Yamawaki et al. (2003) reported kB activation (Kurokouchi et al., 2001).
the inhibition of TNF-a-induced MAPKs (JNK, p38, and ERK) by Plotkin et al. (2005) have reported that mechanical
chronic physiological FSS in rabbit aorta endothelial cells stimulation by stretching rescued osteocytes from etoposide-
through blocking the association of TRAF2 with TNFR1. Here, or glucocorticoid-induced apoptosis via activation of ERKs.
we show that OFSS down-regulates the signaling capability of ERKs promote cell survival by phosphorylating transcription
TNFR1 membrane complexes and that this ultimately affects factors that regulate the expression of apoptosis-related genes
transcriptional regulation of the IL-8 promoter, a well- (Kousteni et al., 2003; Huang et al., 2007). OFSS also activates
characterized target of TNF-a signaling in the nucleus. This ERKs (You et al., 2001; Young et al., 2009) and that may result in
result is consistent with a previous report in which OFSS synthesis of new proteins, leading to the pro-survival effect. In
inhibited TNF-a-induced NF-kB activation and intercellular this study we found that these signaling pathways were not
adhesion molecule-1 (ICAM-1) mRNA expression involved in the anti-apoptotic effects of OFSS. Taken together,
(Kurokouchi et al., 2001). This study, using UMR106 cells, also our results suggest that OFSS protected MC3T3 cells from
found that inhibition was shear stress-dependent indicating that TNF-a-induced apoptosis using mechanisms different from
OFSS has potent effects on mechanotransduction pathways. those involved in the anti-apoptotic response of stretching.
Our results provide further evidence that TNF-a-induced We evaluated whether OFSS altered the amount of TNFR1
apoptosis is inhibited by OFSS in osteoblasts via blockade of expressed on the cell surface and whether OFSS affected the
signaling through membrane-associated protein complexes. ability of TNF-a to induce internalization of the receptor. Since
These findings are consistent with the mechanosome concept TNF-a binds TNFR1 and triggers a signaling cascade leading to
in that mechanical signals detected at the cell surface are the activation of NFkB transcription factors and the activation
translated into biochemical responses inside the cell through of caspases, the effect of OFSS on TNFR1 surface expression
regulation of membrane-associated multi-protein signaling and internalization could impact cell survival. Previous studies
complexes that ultimately affect gene expression in the nucleus showed that the TNFR1-mediated death pathway is

JOURNAL OF CELLULAR PHYSIOLOGY


SHEAR STRESS INDUCED BLOCK OF TNFR1 SIGNALING 1051

internalization-dependent (Micheau and Tschopp, 2003; Correlation with prostaglandin upregulation. Biochem Biophys Res Commun 217:640–
Schneider-Brachert et al., 2004) and inhibition of receptor 648.
Kousteni S, Han L, Chen JR, Almeida M, Plotkin LI, Bellido T, Manolagas SC. 2003. Kinase-
internalization selectively blocked TNF-mediated cytotoxicity mediated regulation of common transcription factors accounts for the bone-protective
but did not affect NF-kB activation (Schutze et al., 1999; effects of sex steroids. J Clin Invest 111:1651–1664.
Kurokouchi K, Jacobs CR, Donahue HJ. 2001. Oscillating fluid flow inhibits TNF-alpha -
Schneider-Brachert et al., 2006). induced NF-kappa B activation via an Ikappa B kinase pathway in osteoblast-like UMR106
In conclusion, our results indicate that OFSS, generally cells. J Biol Chem 276:13499–13504.
Li H, Kobayashi M, Blonska M, You Y, Lin X. 2006. Ubiquitination of RIP is required for tumor
accepted as the most physiologically relevant fluid flow profile necrosis factor alpha-induced NF-kappaB activation. J Biol Chem 281:13636–13643.
for bone cells, inhibits TNF-a-induced apoptosis via a Micheau O, Tschopp J. 2003. Induction of TNF receptor I-mediated apoptosis via two
mechanism that specifically blocks key steps in the TNF-a sequential signaling complexes. Cell 114:181–190.
Norvell SM, Alvarez M, Bidwell JP, Pavalko FM. 2004. Fluid shear stress induces beta-catenin
signaling pathway that are mediated by TNFR1, RIP, and signaling in osteoblasts. Calcif Tissue Int 75:396–404.
caspase-8. Surprisingly, we found that rescue of osteoblasts Pavalko FM, Chen NX, Turner CH, Burr DB, Atkinson S, Hsieh YF, Qiu J, Duncan RL. 1998.
Fluid shear-induced mechanical signaling in MC3T3-E1 osteoblasts requires cytoskeleton-
from TNF-a-induced apoptosis did not require an intact actin integrin interactions. Am J Physiol 275:C1591–C1601.
cytoskeleton or any of several traditional ‘‘cell survival’’ Pavalko FM, Gerard RL, Ponik SM, Gallagher PJ, Jin Y, Norvell SM. 2003a. Fluid shear
stress inhibits TNF-alpha-induced apoptosis in osteoblasts: A role for fluid shear stress-
pathways including MAPK, AKT, PLC, and NO. Finally, these induced activation of PI3-kinase and inhibition of caspase-3. J Cell Physiol 194:194–
results are consistent with the concept that cellular regulation 205.
of external mechanical signals is mediated by load bearing Pavalko FM, Norvell SM, Burr DB, Turner CH, Duncan RL, Bidwell JP. 2003b. A Model for
mechanotransduction in bone cells: The load-bearing mechanosomes. J Cell Biochem
mechanosomes which are regulated by OFSS at the cell 88:104–112.
membrane and ultimately transmit information directly into the Piekarski K, Munro M. 1977. Transport mechanism operating between blood supply and
osteocytes in long bones. Nature 269:80–82.
nucleus to affect cell function. Plotkin LI, Mathov I, Aguirre JI, Parfitt AM, Manolagas SC, Bellido T. 2005. Mechanical
stimulation prevents osteocyte apoptosis: Requirement of integrins, Src kinases, and ERKs.
Acknowledgments Am J Physiol Cell Physiol 289:C633–C643.
Pobezinskaya YL, Kim YS, Choksi S, Morgan MJ, Li T, Liu C, Liu Z. 2008. The function of
TRADD in signaling through tumor necrosis factor receptor 1 and TRIF-dependent Toll-
This work was supported by NIH grants R01AR052682 to like receptors. Nat Immunol 9:1047–1054.
F.M.P., DK053796 to J.P.B., and by post-doctoral fellowship Ponik SM, Pavalko FM. 2004. Formation of focal adhesions on fibronectin promotes fluid
shear stress induction of COX-2 and PGE2 release in MC3T3-E1 osteoblasts. J Appl Physiol
AR056188 to SRLY. 97:135–142.
Ponik SM, Triplett JW, Pavalko FM. 2007. Osteoblasts and osteocytes respond differently to
oscillatory and unidirectional fluid flow profiles. J Cell Biochem 100:794–807.
Literature Cited Schneider-Brachert W, Tchikov V, Neumeyer J, Jakob M, Winoto-Morbach S, Held-Feindt J,
Heinrich M, Merkel O, Ehrenschwender M, Adam D, Mentlein R, Kabelitz D, Schutze S.
Chen G, Goeddel DV. 2002. TNF-R1 signaling: A beautiful pathway. Science 296:1634–1635. 2004. Compartmentalization of TNF receptor 1 signaling: Internalized TNF receptosomes
Chen NX, Ryder KD, Pavalko FM, Turner CH, Burr DB, Qiu J, Duncan RL. 2000. Ca(2þ) as death signaling vesicles. Immunity 21:415–428.
regulates fluid shear-induced cytoskeletal reorganization and gene expression in Schneider-Brachert W, Tchikov V, Merkel O, Jakob M, Hallas C, Kruse ML, Groitl P, Lehn A,
osteoblasts. Am J Physiol Cell Physiol 278:C989–C997. Hildt E, Held-Feindt J, Dobner T, Kabelitz D, Kronke M, Schutze S. 2006. Inhibition of TNF
Chen NX, Geist DJ, Genetos DC, Pavalko FM, Duncan RL. 2003. Fluid shear-induced receptor 1 internalization by adenovirus 14.7 K as a novel immune escape mechanism.
NFkappaB translocation in osteoblasts is mediated by intracellular calcium release. Bone J Clin Invest 116:2901–2913.
33:399–410. Schutze S, Machleidt T, Adam D, Schwandner R, Wiegmann K, Kruse ML, Heinrich M, Wickel
Devin A, Cook A, Lin Y, Rodriguez Y, Kelliher M, Liu Z. 2000. The distinct roles of TRAF2 and M, Kronke M. 1999. Inhibition of receptor internalization by monodansylcadaverine
RIP in IKK activation by TNF-R1: TRAF2 recruits IKK to TNF-R1 while RIP mediates IKK selectively blocks p55 tumor necrosis factor receptor death domain signaling. J Biol Chem
activation. Immunity 12:419–429. 274:10203–10212.
Donahue SW, Jacobs CR, Donahue HJ. 2001. Flow-induced calcium oscillations in rat Triplett JW, O’Riley R, Tekulve K, Norvell SM, Pavalko FM. 2007. Mechanical loading by fluid
osteoblasts are age, loading frequency, and shear stress dependent. Am J Physiol Cell shear stress enhances IGF-1 receptor signaling in osteoblasts in a PKCzeta-dependent
Physiol 281:C1635–C1641. manner. Mol Cell Biomech 4:13–25.
Ermolaeva MA, Michallet MC, Papadopoulou N, Utermohlen O, Kranidioti K, Kollias G, Turner CH, Pavalko FM. 1998. Mechanotransduction and functional response of the skeleton
Tschopp J, Pasparakis M. 2008. Function of TRADD in tumor necrosis factor receptor 1 to physical stress: The mechanisms and mechanics of bone adaptation. J Orthop Sci 3:346–
signaling and in TRIF-dependent inflammatory responses. Nat Immunol 9:1037–1046. 355.
Gururaja TL, Yung S, Ding R, Huang J, Zhou X, McLaughlin J, Daniel-Issakani S, Singh R, Yamawaki H, Lehoux S, Berk BC. 2003. Chronic physiological shear stress inhibits tumor
Cooper RD, Payan DG, Masuda ES, Kinoshita T. 2007. A class of small molecules that inhibit necrosis factor-induced proinflammatory responses in rabbit aorta perfused ex vivo.
TNFalpha-induced survival and death pathways via prevention of interactions between Circulation 108:1619–1625.
TNFalphaRI, TRADD, and RIP1. Chem Biol 14:1105–1118. Yang Q, Huang W, Jozwik C, Lin Y, Glasman M, Caohuy H, Srivastava M, Esposito D, Gillette
He KL, Ting AT. 2002. A20 inhibits tumor necrosis factor (TNF) alpha-induced apoptosis by W, Hartley J, Pollard HB. 2005. Cardiac glycosides inhibit TNF-alpha/NF-kappaB signaling
disrupting recruitment of TRADD and RIP to the TNF receptor 1 complex in Jurkat T cells. by blocking recruitment of TNF receptor-associated death domain to the TNF receptor.
Mol Cell Biol 22:6034–6045. Proc Natl Acad Sci USA 102:9631–9636.
Huang C, Ogawa R. 2010. Mechanotransduction in bone repair and regeneration. FASEB J Yang Z, Bidwell JP, Young SR, Gerard-O’Riley R, Wang H, Pavalko FM. 2010. Nmp4/CIZ
24:1–8. inhibits mechanically induced beta-catenin signaling activity in osteoblasts. J Cell Physiol
Huang D, Khoe M, Befekadu M, Chung S, Takata Y, Ilic D, Bryer-Ash M. 2007. Focal adhesion 223:435–441.
kinase mediates cell survival via NF-kappaB and ERK signaling pathways. Am J Physiol Cell You J, Reilly GC, Zhen X, Yellowley CE, Chen Q, Donahue HJ, Jacobs CR. 2001. Osteopontin
Physiol 292:C1339–C1352. gene regulation by oscillatory fluid flow via intracellular calcium mobilization and activation
Hung CT, Pollack SR, Reilly TM, Brighton CT. 1995. Real-time calcium response of cultured of mitogen-activated protein kinase in MC3T3-E1 osteoblasts. J Biol Chem 276:13365–
bone cells to fluid flow. Clin Orthop 313:256–269. 13371.
Jacobs CR, Yellowley CE, Davis BR, Zhou Z, Cimbala JM, Donahue HJ. 1998. Differential Young SR, Gerard-O’Riley R, Kim JB, Pavalko FM. 2009. Focal adhesion kinase is important for
effect of steady versus oscillating flow on bone cells. J Biomech 31:969–976. fluid shear stress-induced mechanotransduction in osteoblasts. J Bone Miner Res 24:411–
Johnson DL, McAllister TN, Frangos JA. 1996. Fluid flow stimulates rapid and continuous 424.
release of nitric oxide in osteoblasts. Am J Physiol 271:E205–E208. Young SR, Gerard-O’Riley R, Harrington M, Pavalko FM. 2010. Activation of NF-kappaB by
Klein-Nulend J, Semeins CM, Ajubi NE, Nijweide PJ, Burger EH. 1995. Pulsating fluid flow fluid shear stress, but not TNF-alpha, requires focal adhesion kinase in osteoblasts. Bone
increases nitric oxide (NO) synthesis by osteocytes but not periosteal fibroblasts— 47:74–82.

JOURNAL OF CELLULAR PHYSIOLOGY

You might also like