Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Feb 1.
Published in final edited form as: Hepatology. 2012 Feb;55(2):594–608. doi: 10.1002/hep.24701

OSTEOPONTIN, AN OXIDANT STRESS-SENSITIVE CYTOKINE, UP-REGULATES COLLAGEN-I VIA INTEGRIN αVβ3 ENGAGEMENT AND PI3K-pAkt-NFκB SIGNALING

Raquel Urtasun *, Aritz Lopategi *, Joseph George, Tung-Ming Leung, Yongke Lu, Xiaodong Wang, Xiaodong Ge, Maria Isabel Fiel (1), Natalia Nieto
PMCID: PMC3561739  NIHMSID: NIHMS326651  PMID: 21953216

Abstract

Background & Aim

A key feature in the pathogenesis of liver fibrosis is fibrillar collagen-I deposition; yet, mediators that could be key therapeutic targets remain elusive. We hypothesized that osteopontin (OPN), an extracellular matrix (ECM) cytokine expressed in hepatic stellate cells (HSC), could drive fibrogenesis by modulating the HSC profibrogenic phenotype and collagen-I expression.

Results

rOPN up-regulated collagen-I protein in primary HSC in a TGFβ-independent fashion whereas it down-regulated matrix metalloprotease-13 (MMP13) thus favoring scarring. rOPN activated primary HSC -confirmed by increased α-smooth muscle actin (α-SMA) expression- and enhanced their invasive and wound-healing potential. HSC isolated from wild type (WT) mice were more profibrogenic than those from Opn-/- mice and infection of primary HSC with an Ad-OPN increased collagen-I, indicating correlation between both proteins. The OPN induction of collagen-I occurred via integrin αvβ3 engagement and activation of the PI3K-pAkt-NFκB signaling pathway, while CD44-binding and mTOR-p70S6K were not involved. Neutralization of integrin αvβ3 prevented the OPN-mediated activation of the PI3K-pAkt-NFκB signaling cascade and collagen-I up-regulation. Likewise, inhibition of PI3K and NFκB blocked the OPN-mediated collagen-I increase. HCV-cirrhotic patients showed co-induction of collagen-I and cleaved OPN compared to healthy individuals. Acute and chronic liver injury by carbon tetrachloride (CCl4)-injection or thioacetamide (TAA)-treatment elevated OPN expression. Reactive oxygen species up-regulated OPN in vitro and in vivo and antioxidants prevented this effect. OpnHEP Tg mice developed spontaneous liver fibrosis compared to WT mice. Lastly, chronic CCl4-injection and TAA-treatment caused more liver fibrosis to WT than to Opn-/- mice and the reverse occurred in OpnHEP Tg mice.

Conclusion

OPN emerges as a cytokine within the ECM protein network driving the increase in collagen-I protein contributing to scarring and liver fibrosis.

Keywords: Extracellular matrix, fibrosis, hepatic stellate cells, oxidant stress


Fibrogenesis, or activation of the wound-healing response to persistent liver injury, is characterized by changes in the composition and quantity of the ECM deposits distorting the normal hepatic architecture by forming fibrotic scars. Failure to degrade accumulated ECM is a major reason why fibrosis progresses to cirrhosis. Emerging antifibrogenic therapies aim at inhibiting the activation of profibrogenic cells to prevent fibrillar collagen-I deposition, at degrading the excessive ECM to recover the normal liver architecture and at restoring functional liver mass.

Although different cell types contribute to the increase in fibrillar collagen-I during hepatic fibrogenesis, they all undergo a common process of differentiation and acquisition of a classical myofibroblast-like phenotype. HSC are considered a central ECM-producing cell within the injured liver (1), playing a significant role in collagen-I deposition when hepatocellular injury is concentrated within the liver lobule and sinusoids. In normal liver, they reside in the sinusoidal space of Disse; however, during chronic injury, they activate while acquiring motile, proinflammatory and profibrogenic properties (2). Activated HSC migrate and accumulate at the sites of tissue repair, secreting large amounts of ECM, mostly collagen-I, and regulating ECM remodeling. Up-regulation of fibrillar collagen-I is thus a key event leading to scarring, the pathophysiological hallmark of liver fibrosis.

While some current therapies have proven beneficial, dissecting key profibrogenic mechanisms, pathways and mediators of disease progression is vital. Several studies have identified OPN as significantly up-regulated during liver injury and in HSC (3-6). OPN is a soluble cytokine and a matrix-bound protein that can remain intracellular or is secreted; hence, allowing autocrine and paracrine signaling (7-8). OPN, as a matricellular phosphoglycoprotein, functions as adaptor and modulator of cell-matrix interactions (8). Among its many roles, it regulates cell migration, ECM-invasion and cell adhesion due to its ability to bind integrins -via its RGD motif- or CD44 -via a cryptic site (SVVYGLR)-exposed after cleavage by thrombin, plasminogen, plasmin, cathepsin-B and some MMPs (5, 9). OPN expression increases in tumorigenesis, angiogenesis and in response to inflammation, cellular stress and injury (10-14). OPN plays an important role in regulating tissue remodeling and cell survival as well as in chemoattracting inflammatory cells (15). Moreover, Opn-/- mice show matrix disarrangement and alteration of collagen fibrillogenesis in cartilage compared to their WT littermates (16).

There is limited information on the contribution of OPN to the HSC profibrogenic behavior and the molecular mechanisms and signaling pathways involved in governing collagen-I protein expression during the fibrogenic response to liver injury (3-6, 17). Since OPN is expressed in HSC (3-6), we hypothesized that OPN could trigger signals capable of up-regulating collagen-I per se; hence, acting as a feed-forward mechanism promoting scarring. Therefore, the major goal of this work was to determine how OPN could become a profibrogenic ‘switch’ and to characterize the underlying cellular mechanism for this effect. In the present study, we identified a role for intracellular OPN in increasing collagen-I, the HSC membrane proteins engaged by extracellular OPN, the proximal signaling molecules/stress-sensitive kinases activated upon binding that trigger the profibrogenic cascade, the ability of OPN to respond to oxidant stress and the effect of Opn ablation or overexpression on collagen-I deposition in vivo.

MATERIAL AND METHODS

Please see Supplementary Experimental Procedures

RESULTS

rOPN slightly increases HSC proliferation rates and promotes HSC migration

rOPN did not alter HSC viability but slightly induced proliferation rates both in rat and in human HSC (Supplementary Figure 1); however, rOPN caused a 2-fold increase in the invasive potential or chemotaxis (Supplementary Figure 2A-2B) and enhanced the wound closure ability of rat HSC (Supplementary Figure 2C), important functions gained by HSC during their activation that contribute to their profibrogenic ability. Neutralizing Abs to αvβ3 integrin and OPN blocked the effects on HSC invasion (not shown) and on closure ability (Supplementary Figure 2C).

rOPN induces profibrogenic effects in primary HSC

Upon stimulation with rOPN, rat HSC up-regulated intra- and extracellular collagen-I in a time-dependent fashion (Figure 1A, left). Denatured rOPN did not elevate collagen-I; thus, confirming the specificity of the rOPN effect on collagen-I in HSC (not shown). rOPN lowered extracellular MMP13 protein by 50%, contributing to extracellular collagen-I accumulation. Reciprocal modulation of MMP13 and collagen-I has been previously described in rat HSC (18). Extracellular pro-, intermediate and active MMP2 and MMP9 remained unchanged (Figure 1A, left). Likewise, tissue inhibitor of MMP1 was comparable (not shown). rOPN induced rat HSC activation as shown by the increase in collagen-I and α-SMA proteins (Figure 1A, right). Analogous results were observed in human HSC (Figure 1B).

Figure 1. Profibrogenic effects of rOPN in primary HSC.

Figure 1

Primary rat HSC cultured for 7 days were treated with 0-50 nM rOPN for 6 and 24 hours. Western blot analysis of intra- and extracellular collagen-I, extracellular MMP13 and β-tubulin. Gelatine zymography showing extracellular pro-, intermediate and active MMP2 and MMP9 (A, left). Western blot analysis of intracellular collagen-I, α-SMA and actin in rat HSC treated with 0-50 nM rOPN for 24 hours (A, right). Human HSC cultured for 7 days were treated with 0-100 nM rOPN for 1 and 24 hours. Western blot analysis of intra- and extracellular collagen-I, extracellular MMP13 and MMP1 and β-tubulin. Pro-MMP2 activity was measured by gelatine zymography. Extracellular MMP9 activity was undetectable (not shown) (B). Western blot analysis of intracellular collagen-I and actin in HSC from WT and Opn-/- mice. Extracellular collagen-I was undetectable (C). Western blot analysis of intracellular collagen-I and OPN as well as extracellular OPN in rat HSC infected with Ad-LacZ or with Ad-OPN for 48 hours. Extracellular collagen-I was undetectable (D). Results are expressed as average values. Experiments were performed in triplicates four times. **p<0.01 and ***p<0.001 for rOPN, Opn-/- and Ad-OPN vs control, WT and Ad-LacZ, respectively.

Because of the ability of HSC to secrete TGFβ (19) along with its well-known profibrogenic effect (20), rat HSC were treated with anti-TGFβ Ab. Neutralization of TGFβ did not alter the rOPN-mediated induction of collagen-I; thus, implying a mechanism independent of TGFβ production by HSC (Supplementary Figure 3).

To dissect whether intracellular OPN could play an autocrine role in modulating collagen-I expression, HSC were isolated from WT and Opn-/- mice. WT HSC appeared more profibrogenic than Opn-/- HSC since intracellular collagen-I expression at 7 days of culture was higher in WT HSC than in Opn-/- HSC (Figure 1C). Infection of rat HSC with an Ad-OPN increased intracellular collagen-I and intra- and extracellular OPN compared to HSC infected with Ad-LacZ (Figure 1D). Therefore, a novel autocrine role for intracellular OPN in modulating collagen-I deposition was identified.

Anti-αvβ3 integrin Ab blocks the rOPN-driven collagen-I increase

Since OPN is also a soluble cytokine and a matrix-bound protein, next we evaluated the role of extracellular OPN-mediated signaling (paracrine role) on collagen-I induction in HSC. OPN signals via integrins - mostly integrin αvβ3 highly expressed in HSC- (21-23) and via CD44 -also expressed in HSC- (24). Incubation with anti-αvβ3 integrin blocked the rOPN-driven total collagen-I (intra-plus extracellular) increase in rat HSC, while no major effect was observed by anti-CD44 (Figure 2A). Neutralization of other integrins (i.e. β1, β5 and β6) failed to prevent the increase in collagen-I by rOPN (not shown). Similar results were observed in human HSC (not shown).

Figure 2. Role of αvβ3 integrin and the PI3K-pAkt-NFκB signaling pathway in the rOPN-mediated effects on collagen-I.

Figure 2

Primary rat HSC cultured for 7 days were incubated with 0-50 nM rOPN plus 5 μg/ml of non-immune IgG, anti-αvβ3 or anti-CD44 for 6 hours. Western blot analysis of intra- and extracellular collagen-I and actin (A). Western blot analysis of PI3K, pAkt 473Ser, Akt and β-tubulin up to 3 hours of 0-50 nM rOPN treatment in rat HSC (B). Western blot analysis of pIKKα,β 176/180Ser, IKKα,β, pIκBα 32Ser, IκBα, nuclear and cytosolic p65 and actin up to 30 min of 0-50 nM rOPN treatment in rat HSC (C). Western blot analysis of mTOR, p70S6K and actin up to 1 hour of 0-50 nM rOPN treatment in rat HSC (D). Results are expressed as average values. Experiments were performed in triplicates four times. *p<0.05, **p<0.01 and ***p<0.001 for rOPN-treated vs control at any given time-point. •p<0.05, ••p<0.01 and •••p<0.001 for co-treated vs Ab-treated.

rOPN increases collagen-I protein via activation of the PI3K-pAkt-NFκB signaling pathway

Given that collagen-I protein is highly responsive to oxidant stress-sensitive kinases, we analyzed the expression of protein kinases involved in regulating collagen-I expression such as pp38 (25-26), pERK1/2 (27), pJNK (28-29), PI3K and pAkt (26, 30). Only PI3K and the ratio pAkt 473Ser/Akt were elevated time-dependently by rOPN up to 3 hours in rat HSC (Figure 2B) and up to 1 hour in human HSC (Supplementary Figure 4A).

Since PI3K/pAkt are upstream of IKK and the IKK complex is central for the activation of NFκB to regulate collagen-I (26, 31), we focused on analyzing this signaling pathway. There was up-regulation of the ratios pIKKα,β 176/180Ser/IKKα,β and pIκBα 32Ser/IκBα as well as of nuclear/cytosolic p65 in OPN-treated rat HSC (Figure 2C). However, involvement of the mTOR-p70S6K cascade, a translational regulatory mechanism downstream of PI3K and pAkt 473Ser for regulating collagen-I (32), was precluded since rOPN neither altered mTOR and p706SK expression (Figure 2D) nor induced mTOR phosphorylation at 2448Ser or 2481Ser in rat HSC (undetectable).

To define further the molecular mechanism for the collagen-I induction under rOPN challenge, we evaluated the potential role of the activation of these two stress-sensitive kinases (i.e. PI3K and pAkt) and of the NFκB signaling pathway. Wortmannin, a PI3K inhibitor, neither altered rat HSC viability (100% by the MTT assay), morphology nor proliferation rates (Supplementary Figure 4B and not shown); however, three different doses of wortmannin down-regulated total collagen-I expression in rat HSC co-treated with rOPN (Figure 3A, top). Similar effects were observed by co-incubation with LY294002, a second PI3K inhibitor (Figure 3A, bottom); thus, linking OPN, PI3K-pAkt activation and collagen-I up-regulation in rat HSC. Comparable results were observed in human HSC (Supplementary Figure 4C). Lastly, inhibitors of pp38, pERK1/2 and pJNK signaling did not prevent the increase in collagen-I by rOPN (not shown).

Figure 3. Blocking αvβ3 integrin, PI3K-pAkt activation and the NFκB signaling pathway prevents the rOPN-mediated effects on collagen-I.

Figure 3

Primary rat HSC cultured for 7 days were treated with 0-50 nM rOPN or co-treated with 0-10 μM wortmannin, 0-10 μM LY294002, 0-10 μM PDTC, 0-5 μM CAY10512 or with 5 μg/ml of non-immune IgG or a neutralizing Ab to integrin αvβ3. Western blot analysis showing that the rOPN-mediated induction of collagen-I in HSC was blunted by 0.1, 1 and 10 μM wortmannin (A, top), LY294002 (A, bottom), PDTC (B, top) and CAY10512 (B, middle). Infection of HSC with Ad-NFκB-Luc for 48 hours and treatment with 50 nM rOPN for 24 hours increased luciferase activity over that of non-treated Ad-NFκB-Luc-infected cells (B, bottom). Both, an integrin αvβ3 Ab and wortmannin blunted the rOPN-mediated induction of the ratios pIKKα,β 176/180Ser/IKKα,β, pIκBα 32Ser/IκBα and nuclear/cytosolic p65 (C). A neutralizing Ab to integrin αvβ3 prevented the induction of PI3K, the ratio pAkt 473Ser/Akt and collagen-I by rOPN in HSC (D). Results are expressed as average values. Experiments were performed in triplicates four times. **p<0.01 and ***p<0.001 for rOPN-treated vs control. •p<0.05, ••p<0.01 and •••p<0.001 for inhibitor or Ab-cotreated vs rOPN-treated or control.

Addition of pyrrolidine dithiocarbamate (PDTC) to block NFκB signaling prevented the rOPN-driven increase in collagen-I in rat HSC (Figure 3B, top). Analogous effects were observed by co-incubation with CAY10512 -a second inhibitor of NFκB signaling- (Figure 3B, middle). Moreover, HSC infected with Ad-NFκB-Luc and treated with rOPN for 24 hours showed a 2-fold increase in luciferase activity compared to non-rOPN-treated Ad-NFκB-Luc-infected cells (Figure 3B, bottom). Both, wortmannin and an αvβ3 integrin neutralizing Ab, blunted the rOPN-mediated effect on the ratios pIKKα,β 176/180Ser/IKKα,β and pIκBα 32Ser/IκBα as well as on nuclear/cytosolic p65 (Figure 3C), suggesting engagement of OPN with integrin αvβ3, PI3K-pAktactivation and NFκB signaling to up-regulate collagen-I expression in rat HSC. Lastly, blocking αvβ3 integrin prevented the increase in PI3K, the ratio pAkt 473Ser/Akt and collagen-I by rOPN in rat HSC (Figure 3D). In summary, these results established a connection among rOPN, αvβ3 integrin, PI3K-pAkt activation and the NFκB signaling pathway to drive collagen-I up-regulation in rat HSC in a paracrine manner.

OPN expression is up-regulated in liver fibrosis

Samples from stage-3 HCV-cirrhotic patients displayed correlation between elevated collagen-I and cleaved OPN protein (~55, ~42 and ~25 kDa isoforms) compared to healthy individuals. Fully modified (glycosylated and phosphorylated) monomeric OPN, typically running at ~75 kDa, was not detectable (Figure 4A).

Figure 4. OPN expression is induced during liver injury and under oxidant stress conditions.

Figure 4

Western blot analysis of cleaved OPN, total collagen-I and actin in livers from control and from patients with stage-3 HCV-induced cirrhosis (A). Western blot analysis of cleaved OPN and actin in livers of mice injected with CCl4 for 24 hours (acute liver injury), with CCl4 for 1 month or with TAA for 4 months (chronic liver injury) (B). In (A) and (B) fully modified (glycosylated and phosphorylated) monomeric OPN was not detected. Immunocytochemistry for OPN in primary HSC isolated from WT mice and cultured for 6 days (C, left). IHC depicting significant OPN expression in HSC, biliary epithelial cells and hepatocytes at 1 month of CCl4-injection (C, middle) and in HSC, biliary epithelial cells, oval cells and hepatocytes after 4 months of TAA-treatment (C, right). The insets show OPN+ HSC in both models (Inline graphic). Immunofluorescence showing co-localization of OPN+ with α-SMA+ (a HSC activation marker) after 4 months of TAA-treatment (D). Western blot analysis of intracellular OPN and β-tubulin in HSC in the presence of two prooxidants (H2O2 and BSO) and an antioxidant (GSH-EE: glutathione-ethyl ester) (E). Results are expressed as average values. Experiments were performed in triplicates four times. ***p<0.001 for HCV, CCl4, TAA or prooxidant treated vs control, mineral oil (MO) or water, respectively. •••p<0.001 for BSO + GSH-EE co-treated vs BSO-treated.

To determine whether OPN also increased during liver injury in mice, we used well-established in vivo models to induce liver fibrosis such as CCl4-injection and TAA-treatment (33). These drugs undergo cytochrome P450 metabolism leading to significant oxidant stress, inflammation and hepatocyte necrosis within hours. The ~25 kDa OPN form was markedly induced in acute and chronic models of liver injury, while the ~55 kDa OPN form was elevated only under chronic CCl4-injection and TAA-treatment (Figure 4B). Hence, there was association between OPN induction, OPN proteolytic processing and the extent of liver fibrosis both in humans and in mice.

Next, we evaluated the specific localization of the OPN induction in the liver. Non-treated livers showed OPN+ biliary epithelial cells (not shown). Primary HSC isolated from WT mice and cultured for 6 days were OPN+ (Figure 4C, left). Immunohistochemical (IHC) analysis revealed OPN expression in HSC (30), biliary epithelial cells (4, 6, 34), oval cells and mostly in damaged hepatocytes in WT mice injected CCl4 for 1 month (Figure 4C, middle). Similar results were observed under TAA-treatment, although hepatocytes showed punctated staining (Figure 4C, right). The insets show OPN+ HSC in both models. In the early stages of CCl4- and TAA-mediated liver injury, Kupffer cells were also OPN+ (not shown); however, the staining faded with disease progression. Of note, granular OPN+ staining -typical of secreted proteins-appeared in focal-septal hepatocytes (Figure 4C, middle). There was co-localization of OPN+ staining with α-SMA+ (a HSC activation marker) under TAA-treatment (Figure 4D) and by CCl4-injection (not shown).

Since liver fibrosis is associated with significant oxidant stress, to dissect whether OPN was responsive to reactive oxygen species, HSC were challenged with H2O2 -a prooxidant typically generated during CCl4 metabolism- or with L-buthionine sulfoximine (BSO) -which depletes glutathione. Both treatments increased OPN expression in HSC, whereas co-treatment with glutathione-ethyl ester to restore glutathione levels, blunted this effect (Figure 4E). To validate the induction of OPN by oxidant stress in vivo, WT mice were CCl4-injected for 1 month in the presence or absence of S-adenosylmethionine (SAM), an antioxidant known to restore glutathione levels. Co-injection with SAM lowered OPN protein (Figure 5A-5B) and the extent of liver fibrosis (Figure 5C-5D) by 50% when compared to mice injected CCl4 alone. In summary, these data proved the ability of OPN to respond to drug-induced liver injury and to oxidant stress.

Figure 5. SAM protects WT mice from CCl4-induced chronic liver injury.

Figure 5

C57BL/6J WT mice were injected MO, SAM plus MO, CCl4 or CCl4 plus SAM for 1 month. Co-treated mice showed decreased OPN expression (A), which was quantified by morphometry analysis (B). Likewise, fibrosis was less apparent in co-treated mice than in CCl4-injected mice as shown by Sirius red/fast green staining (C) and morphometry analysis (D). Results are expressed as mean values ± SEM. n=8/group; ***p<0.001 for CCl4 or CCl4 + SAM vs MO or SAM; ••p<0.01 for CCl4 + SAM vs CCl4.

WT show more CCl4-induced chronic liver injury and fibrosis than Opn-/- mice

Fibrosis typically develops due to chronic liver injury. To decipher the role of OPN in the progression of liver disease, we tested whether chronic CCl4-injection could lead to differences in the extent of liver fibrosis. CCl4-injected C57BL/6J WT showed greater alanine aminotransferase (ALT) activity and more inflammation, hepatocyte ballooning degeneration and necrosis than Opn-/- mice (Figure 6A-6E). Cytochrome P450 2E1 (CYP2E1) expression was similar in WT and Opn-/- mice, indicating that the extent of liver injury in these mice was not due to different CCl4metabolism (Figure 6F).

Figure 6. WT mice show more CCl4-induced chronic liver injury than Opn-/- mice.

Figure 6

C57BL/6J WT and Opn-/- mice were injected CCl4 or MO for 1 month. H&E staining revealed more centrilobular necrosis (Inline graphic), centrilobular inflammation (Inline graphic) and hepatocyte ballooning degeneration (Inline graphic) in CCl4-injected WT than in Opn-/- mice (A). ALT activity (B), centrilobular and parenchymal inflammation scores (C), hepatocyte ballooning degeneration score (D) and centrilobular and parenchymal necrosis scores (E). A Western blot analysis showing similar CYP2E1 expression in WT and Opn-/- mice (F). Results are expressed as mean values ± SEM. n=8/group; ***p<0.001 for CCl4 vs MO; •p<0.05, ••p<0.01 and •••p<0.001 for Opn-/- + CCl4 vs WT + CCl4.

In addition, CCl4-injected WT mice presented elevated collagenous proteins, portal fibrosis, bridging fibrosis, scar thickness, Brunt fibrosis score and Sirius red and collagen-I morphometry compared to Opn-/- mice (Figure 7A-7E). The above results were validated in WT and Opn-/- 129sv mice (Supplementary Figures 5-6). Transgenic mice overexpressing OPN in hepatocytes (OpnHEP Tg) and injected CCl4 for 1 month showed similar ALT activity, necrosis and inflammation but significant periportal, bridging and sinusoidal fibrosis along with increased collagen-I scar thickness compared to WT mice (Figure 8). Moreover, OpnHEP Tg mice developed spontaneous perivenular, perisinusoidal and portal fibrosis over time (1 yr) in the absence of any profibrogenic treatment (Supplementary Figure 7). In aggregate, the data suggest that OPN plays a major role in chronic CCl4-induced hepatic fibrosis by regulating scar formation.

Figure 7. WT mice show more CCl4-induced liver fibrosis than Opn-/- mice.

Figure 7

C57BL/6J WT and Opn-/- mice were injected CCl4 or MO for 1 month. Sirius red/fast green staining indicated fibrosis stage ~3 in CCl4-injected WT and ~1-2 in CCl -injected Opn-/- 4 mice (portal Inline graphic and bridging Inline graphic fibrosis) as well as greater scar thickness in WT compared to Opn-/- mice (Inline graphic) (A). Collagen-I IHC confirmed the extent of portal fibrosis (Inline graphic), bridging fibrosis (Inline graphic) and scar thickness (Inline graphic) in CCl4-injected mice (B). Brunt fibrosis score (C), Sirius red morphometry (D) and collagen-I morphometry analysis (E). Results are expressed as mean values ± SEM. n=8/group; **p<0.01 and ***p<0.001 for CCl4 vs MO; ••p<0.01 and •••p<0.001 for Opn-/- + CCl4 vs WT + CCl4.

Figure 8. OpnHEP Tg mice in C57BL/6J genetic background show more CCl4-induced fibrosis than WT mice.

Figure 8

Figure 8

WT and OpnHEP Tg mice were injected MO or CCl4 for 1 month. H&E staining revealed similar centrilobular necrosis (Inline graphic) and inflammation (Inline graphic) in CCl4-injected OpnHEP Tg and in WT mice (A). ALT activity (B). Necrosis and inflammation scores (C). Sirius red/fast green staining and IHC for collagen-I demonstrated more portal fibrosis (Inline graphic), bridging fibrosis (Inline graphic) and sinusoidal fibrosis (Inline graphic) in CCl -injected OpnHEP 4 Tg than in WT mice (D-E). Brunt fibrosis score, collagen-I and Sirius red/fast green morphometry (F). Results are expressed as mean values ± SEM. n=8/group; **p<0.01 and ***p<0.001 for CCl4 vs MO; ••p<0.01 for OpnHEP Tg + CCl4 vs WT + CCl4.

WT mice show significant liver fibrosis under chronic TAA-treatment compared to Opn-/- mice

To confirm the results obtained under chronic CCl4-injection, we used the TAA-treatment as a second model of chronic drug-induced liver fibrosis. Sirius red/fast green staining and collagen-I IHC showed stage >3 fibrosis in TAA-treated WT and ~1-2 in Opn-/- mice with clear induction of collagen-I deposition in TAA-treated WT compared to Opn-/- mice, extensive portal fibrosis, bridging fibrosis and a ~3-fold increase in scar thickness (Supplementary Figure 8A-8B). Thus, fibrosis was more distinct in TAA-treated WT than in Opn-/- mice as quantified by the Brunt fibrosis score and by the Sirius red and collagen-I morphometry (Supplementary Figure 8C-8E).

Collectively, these results suggest that increased OPN expression per se or following chronic liver injury and oxidant stress can stimulate collagen-I deposition in vivo. In addition, the in vitrostudies demonstrate that intracellular OPN plays an autocrine role in regulating collagen-I expression in HSC. Moreover, treatment with rOPN to resemble the paracrine actions of secreted OPN, increases HSC invasion, chemotaxis and wound healing potential and up-regulates collagen-I via integrin αvβ3 engagement, activation of PI3K-pAkt and NFκB signaling (Supplementary Figure 9).

DISCUSSION

It is becoming clearer that OPN is significantly induced during liver injury both in humans and in rodents (4-6, 17). In the past few years, work from several groups (4-6, 17, 34) studied the potential role of OPN in liver fibrosis albeit with inconclusive results. Studies by Lee et al (5) demonstrated an OPN increase in the culture medium from culture-activated HSC and under oral CCl4-administration; however, no mechanistic studies were performed to dissect how OPN regulates collagen-I protein deposition. Lorena et al (6) suggested increased susceptibility to CCl4-injection in Opn-/- mice. Although the authors claimed that the protection observed in WT mice was due to enhancement of hepatocyte survival and reduction in NOS2 expression; yet, they neither provided IHC for cell survival markers nor measured the concentration of NO· or ONOO- to support their conclusions, and no studies on collagen-I regulation were performed. Lastly, a recent publication from Syn et al (4) proposes a role for the Hedgehog signaling pathway in activating OPN and promoting fibrosis progression in non-alcoholic steatohepatitis; however, it is not clear which OPN isoform the authors are referring to and it is Gli1, and not Gli2, expression that it is widely considered the most reliable readout for cells undergoing active Hedgehog signaling.

Thus, there is a well timed need for dissecting the molecular mechanism on how this matricellular protein could regulate the fibrogenic response to liver injury, and specifically collagen-I protein expression by HSC. Currently, there are many unresolved questions on how OPN could act as a feed-forward mechanism to promote scarring, among these are: Does extracellular OPN have the ability to increase the HSC profibrogenic potential and HSC-derived collagen-I protein? Which HSC receptors are involved in the profibrogenic cascade triggered by OPN? What intracellular signals are activated upon OPN-receptor binding that drive the collagen-I increase in HSC? Is autocrine OPN signaling involved in regulating collagen-I in HSC? Is OPN sensitive to oxidant stress? Which OPN isoforms appear during the course of liver fibrosis? Does OPN induce sinusoidal fibrosis?

Thus, the overall goal of this work was to address these questions, identify the mechanism for the OPN-driven collagen-I up-regulation in HSC and determine the functional role of OPN in the pathogenesis of liver fibrosis. The idea that OPN mediates liver fibrosis is relevant for several reasons. First, because the observation that OPN is up-regulated in HSC during hepatic injury provides an excellent conceptual advance in our understanding of liver fibrogenesis, as it appears that OPN up-regulates HSC collagen-I protein in an autocrine and paracrine fashion. Second, it supports the clinicopathological finding that injury occurring in the central region is accompanied by fibrosis. Third, it opens the possibility of linking a soluble cytokine/matricellular protein with fibrogenesis. Lastly, the identification of the mechanism and mediators involved in the profibrogenic actions of OPN could help in devising strategies for therapeutic targeting.

Our in vitro experiments validated the hypothesis of the profibrogenic and proinvasive actions of OPN in HSC. Mechanistic studies identified the HSC membrane proteins engaged by OPN and the proximal signaling molecules/oxidant stress-sensitive kinases activated upon OPN binding that trigger the fibrogenic cascade. The experimental data identified integrin αvβ3 as an efficient conveyor of the OPN-mediated profibrogenic actions in HSC and pointed at the PI3K-pAkt activation and the NFκB signaling pathway as highly involved in this process.

Since OPN signals via integrins and CD44, it is feasible that following liver injury, a ligand for αvβ3 integrin such as OPN accumulates in the space of Disse and acts in a αvβ3 integrin-dependent manner to maintain collagen-I induction, HSC activation, invasion and migration. Because OPN binds ECM proteins (35-36), this binding ability may enhance HSC activation,migration and invasion, key HSC features for the development of fibrosis. The finding that blocking CD44 did not prevent the effect of rOPN on collagen-I may be related to the ability of hyaluronic acid -a glycosaminglycan synthesized during HSC activation (24, 37)- to bind CD44; thus, competitive inhibition between hyaluronic acid and rOPN for CD44 binding could occur in HSC, although this possibility needs further investigation.

Several observations support the role for the PI3K-pAkt activation and the NFκB signaling pathway in the effects mediated by rOPN on collagen-I. First, rOPN rapidly increased PI3K, the ratios pAkt 473Ser/Akt, pIKKα,β 176/180Ser/IKKα,β and pIκBα 32Ser/IκBα as well as nuclear translocation of p65. Second, inhibitors of PI3K activation and NFκB signaling blunted the rOPN-mediated increase in intra- and extracellular collagen-I protein. Third, blockade of αvβ3 integrin signaling with a neutralizing Ab and incubation with wortmannin or LY294002 prevented the induction of PI3K, the increase in the ratios pAkt 473Ser/Akt, pIKKα,β 176/180Ser/IKKα,β and pIκBα 32Ser/IκBα, nuclear translocation of p65 and the up-regulation of collagen-I protein by rOPN. Involvement of the mTOR cascade was ruled out since rOPN altered neither mTOR-p706SK expression nor mTOR phosphorylation. Therefore, this study linked extracellular and/or secreted OPN (i.e. paracrine effect) with αvβ3 integrin binding, PI3K-pAkt activation, NFκB signaling and scarring.

Work from several laboratories (3-6), including our own, suggests that HSC are an important source of OPN during liver injury. To date, OPN was believed to exert its effects by binding the RGD motif in integrins and the cell-surface receptor CD44; however, an intracellular function of OPN in liver fibrosis was largely unknown. Because HSC isolated from Opn-/- mice were less profibrogenic than those from WT mice and infection of HSC with Ad-OPN increased intracellular collagen-I, these results suggested a novel autocrine mechanism whereby intracellular OPN could modulate collagen-I deposition in HSC. Alternatively, extracellular OPNeither from HSC or from neighboring cells may activate HSC through its receptor (αvβ3 integrin), as suggested above; thus, creating a positive feedback loop.

To further validate our hypothesis, we then assessed whether OPN contributed to the fibrogenic response in vivo using two mouse models of drug-induced liver injury. The data from human samples and from the mouse models showed that most of the OPN found in liver injury appears to be cleaved at least at the end-point of the experiments. The role of each cleaved isoform in regulating the fibrogenic response to liver injury as well as the identification of the proteases that cleave hepatic OPN is currently under active investigation in our laboratory since additional integrin binding sites, other than αvβ3 integrin, are likely to be uncovered by proteolytic processing of the protein.

Upon the onset of liver injury in mice, the increase in OPN likely results from oxidant stress because CCl4 and TAA metabolism via cytochrome P450s generates a considerable amount of free radicals (33), and the in vitro data demonstrated the OPN-responsiveness to oxidant stress, which was blocked by antioxidant treatment. Furthermore, co-treatment with SAM, known to elevate glutathione levels, prevented the increase in OPN and the fibrogenic response in WT mice injected CCl4 for 1 month.

Although it is possible that the chronic effects of Opn ablation could be secondary to its effects on liver injury itself (i.e. inflammation and ductular reaction, unpublished observations), the data clearly reveal a direct action of OPN on collagen-I protein expression, a key event in liver fibrosis. Hence, OPN appears to induce scarring per se. This is indeed also supported by the finding that while ALT activity and the necrosis and inflammation scores were similar, there was increased portal, bridging and sinusoidal fibrosis along with enhanced width of the collagenous septa in CCl4-injected OpnHEP Tg mice compared to their WT littermates. Notably, OpnHEP Tg mice developed spontaneous fibrosis over time while WT mice did not. Lastly, in line with the results using OpnHEP Tg mice and the in vitro data, fibrilar collagen-I content and scar thickness was significantly lowered by OPN ablation in vivo. It is likely that secreted OPN allows paracrine signaling to HSC, while endogenous OPN expression in HSC signals in an autocrine fashion amplifying the fibrogenic response. The cell- and matrix-binding ability of OPN may also facilitate a proper stromal and fibrillar collagen network organization. Overall, it is reasonable to propose that OPN may drive the fibrogenic response, among others, by directly regulating collagen-I deposition. Thus, OPN emerges as a key soluble cytokine and ECM-bound molecule promoting liver fibrosis.

Supplementary Material

Supp Figure S1
Supplementary Legends&Supp Table S1
Supp Figure S2
Supp Figure S3
Supp Figure S4
Supp Figure S5
Supp Figure S6
Supp Figure S7
Supp Figure S8
Supp Figure S9

ACKNOWLEDGEMENT

The authors are very grateful to the following investigators: David T. Denhardt (Rutgers University, NJ) for his generous gift of the 2A1 Ab and for the Opn-/- mice in 129sv background; Satoshi Mochida (Saitama Medical University, Japan) for providing the OpnHEP Tg mice; Andrea D. Branch (Mount Sinai School of Medicine, NY) for donating the human liver protein lysates; Toshimitsu Uede (Hokkaido University, Japan) for the Ad-OPN and Ad-LacZ; John Engelhardt (University of Iowa, IA) for the recombinant Ad expressing the NFκB-Luc reporter; and Feng Hong (Mount Sinai School of Medicine, NY) for supplying the primary human HSC isolated from normal liver margin of patients undergoing hepatic tumor resection.

We are also very thankful to all former and current members from the Nieto Laboratory for their helpful comments and suggestions throughout this project as well as for their critical review of the manuscript. Special thanks go to Marcos Rojkind, Arthur I. Cederbaum and David T. Denhardt for their constant support and for their very helpful insight throughout the course of this project.

Grant Support: Postdoctoral Fellowships from the Government of Navarre (Spain) (R. U.) and from the Basque Government (Spain) (A. L.). US Public Health Service Grants 5R01 DK069286 and 2R56 DK069286 from the National Institute of Diabetes and Digestive and Kidney Diseases and 5P20 AA017067 from the National Institute on Alcohol Abuse and Alcoholism (N. N.).

List of Abbreviations

ALT

alanine aminotransferase

BSO

L-buthionine sulfoximine

CCl4

carbon tetrachloride

CYP2E1

cytochrome P450 2E1

ECM

extracellular matrix

IHC

immunohistochemistry

IOD

integrated optical density

HSC

hepatic stellate cells

MMP

matrix metalloprotease

MO

mineral oil

OPN

osteopontin

Opn-/-

osteopontin knockout mice

OpnHEP Tg

osteopontin transgenic mice in hepatocytes

PDTC

pyrrolidine dithiocarbamate

rOPN

recombinant OPN

SAM

S-adenosylmethionine

α-SMA

α-smooth muscle actin

TAA

thioacetamide

WT

wild-type

REFERENCES

  • 1.Gabele E, Brenner DA, Rippe RA. Liver fibrosis: signals leading to the amplification of the fibrogenic hepatic stellate cell. Front Biosci. 2003;8:d69–77. doi: 10.2741/887. [DOI] [PubMed] [Google Scholar]
  • 2.Milani S, Herbst H, Schuppan D, Kim KY, Riecken EO, Stein H. Procollagen expression by nonparenchymal rat liver cells in experimental biliary fibrosis. Gastroenterology. 1990;98:175–184. doi: 10.1016/0016-5085(90)91307-r. [DOI] [PubMed] [Google Scholar]
  • 3.De Minicis S, Seki E, Uchinami H, Kluwe J, Zhang Y, Brenner DA, Schwabe RF. Gene expression profiles during hepatic stellate cell activation in culture and in vivo. Gastroenterology. 2007;132:1937–1946. doi: 10.1053/j.gastro.2007.02.033. [DOI] [PubMed] [Google Scholar]
  • 4.Syn WK, Choi SS, Liaskou E, Karaca GF, Agboola KM, Oo YH, Mi Z, et al. Osteopontin is induced by hedgehog pathway activation and promotes fibrosis progression in nonalcoholic steatohepatitis. Hepatology. 2011;53:106–115. doi: 10.1002/hep.23998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Lee SH, Seo GS, Park YN, Yoo TM, Sohn DH. Effects and regulation of osteopontin in rat hepatic stellate cells. Biochem Pharmacol. 2004;68:2367–2378. doi: 10.1016/j.bcp.2004.08.022. [DOI] [PubMed] [Google Scholar]
  • 6.Lorena D, Darby IA, Gadeau AP, Leen LL, Rittling S, Porto LC, Rosenbaum J, et al. Osteopontin expression in normal and fibrotic liver. altered liver healing in osteopontin-deficient mice. J Hepatol. 2006;44:383–390. doi: 10.1016/j.jhep.2005.07.024. [DOI] [PubMed] [Google Scholar]
  • 7.Kazanecki CC, Uzwiak DJ, Denhardt DT. Control of osteopontin signaling and function by post-translational phosphorylation and protein folding. J Cell Biochem. 2007;102:912–924. doi: 10.1002/jcb.21558. [DOI] [PubMed] [Google Scholar]
  • 8.Uede T. Osteopontin, intrinsic tissue regulator of intractable inflammatory diseases. Pathol Int. 2011;61:265–280. doi: 10.1111/j.1440-1827.2011.02649.x. [DOI] [PubMed] [Google Scholar]
  • 9.Kazanecki CC, Kowalski AJ, Ding T, Rittling SR, Denhardt DT. Characterization of anti-osteopontin monoclonal antibodies: Binding sensitivity to post-translational modifications. J Cell Biochem. 2007;102:925–935. doi: 10.1002/jcb.21487. [DOI] [PubMed] [Google Scholar]
  • 10.El-Tanani MK, Campbell FC, Kurisetty V, Jin D, McCann M, Rudland PS. The regulation and role of osteopontin in malignant transformation and cancer. Cytokine Growth Factor Rev. 2006;17:463–474. doi: 10.1016/j.cytogfr.2006.09.010. [DOI] [PubMed] [Google Scholar]
  • 11.Apte UM, Banerjee A, McRee R, Wellberg E, Ramaiah SK. Role of osteopontin in hepatic neutrophil infiltration during alcoholic steatohepatitis. Toxicol Appl Pharmacol. 2005;207:25–38. doi: 10.1016/j.taap.2004.12.018. [DOI] [PubMed] [Google Scholar]
  • 12.Banerjee A, Apte UM, Smith R, Ramaiah SK. Higher neutrophil infiltration mediated by osteopontin is a likely contributing factor to the increased susceptibility of females to alcoholic liver disease. J Pathol. 2006;208:473–485. doi: 10.1002/path.1917. [DOI] [PubMed] [Google Scholar]
  • 13.Ramaiah SK, Rittling S. Pathophysiological role of osteopontin in hepatic inflammation, toxicity, and cancer. Toxicol Sci. 2008;103:4–13. doi: 10.1093/toxsci/kfm246. [DOI] [PubMed] [Google Scholar]
  • 14.Denhardt DT, Mistretta D, Chambers AF, Krishna S, Porter JF, Raghuram S, Rittling SR. Transcriptional regulation of osteopontin and the metastatic phenotype: evidence for a Ras-activated enhancer in the human OPN promoter. Clin Exp Metastasis. 2003;20:77–84. doi: 10.1023/a:1022550721404. [DOI] [PubMed] [Google Scholar]
  • 15.Denhardt DT, Noda M, O’Regan AW, Pavlin D, Berman JS. Osteopontin as a means to cope with environmental insults: regulation of inflammation, tissue remodeling, and cell survival. J Clin Invest. 2001;107:1055–1061. doi: 10.1172/JCI12980. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Liaw L, Birk DE, Ballas CB, Whitsitt JS, Davidson JM, Hogan BL. Altered wound healing in mice lacking a functional osteopontin gene (spp1) J Clin Invest. 1998;101:1468–1478. doi: 10.1172/JCI1122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Fickert P, Stoger U, Fuchsbichler A, Moustafa T, Marschall HU, Weiglein AH, Tsybrovskyy O, et al. A new xenobiotic-induced mouse model of sclerosing cholangitis and biliary fibrosis. Am J Pathol. 2007;171:525–536. doi: 10.2353/ajpath.2007.061133. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Schaefer B, Rivas-Estilla AM, Meraz-Cruz N, Reyes-Romero MA, Hernandez-Nazara ZH, Dominguez-Rosales JA, Schuppan D, et al. Reciprocal modulation of matrix metalloproteinase-13 and type I collagen genes in rat hepatic stellate cells. Am J Pathol. 2003;162:1771–1780. doi: 10.1016/S0002-9440(10)64312-X. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Cubero FJ, Nieto N. Ethanol and arachidonic acid synergize to activate Kupffer cells and modulate the fibrogenic response via tumor necrosis factor alpha, reduced glutathione, and transforming growth factor beta-dependent mechanisms. Hepatology. 2008;48:2027–2039. doi: 10.1002/hep.22592. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Garcia-Trevijano ER, Iraburu MJ, Fontana L, Dominguez-Rosales JA, Auster A, Covarrubias-Pinedo A, Rojkind M. Transforming growth factor beta1 induces the expression of alpha1(I) procollagen mRNA by a hydrogen peroxide-C/EBPbeta-dependent mechanism in rat hepatic stellate cells. Hepatology. 1999;29:960–970. doi: 10.1002/hep.510290346. [DOI] [PubMed] [Google Scholar]
  • 21.Huang G, Brigstock DR. Integrin expression and function in the response of primary culture hepatic stellate cells to connective tissue growth factor (CCN2) J Cell Mol Med. 2010 doi: 10.1111/j.1582-4934.2010.01072.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Patsenker E, Popov Y, Stickel F, Schneider V, Ledermann M, Sagesser H, Niedobitek G, et al. Pharmacological inhibition of integrin alphavbeta3 aggravates experimental liver fibrosis and suppresses hepatic angiogenesis. Hepatology. 2009;50:1501–1511. doi: 10.1002/hep.23144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Zhou X, Murphy FR, Gehdu N, Zhang J, Iredale JP, Benyon RC. Engagement of alphavbeta3 integrin regulates proliferation and apoptosis of hepatic stellate cells. J Biol Chem. 2004;279:23996–24006. doi: 10.1074/jbc.M311668200. [DOI] [PubMed] [Google Scholar]
  • 24.Kikuchi S, Griffin CT, Wang SS, Bissell DM. Role of CD44 in epithelial wound repair: migration of rat hepatic stellate cells utilizes hyaluronic acid and CD44v6. J Biol Chem. 2005;280:15398–15404. doi: 10.1074/jbc.M414048200. [DOI] [PubMed] [Google Scholar]
  • 25.Deng ZY, Li J, Jin Y, Chen XL, Lu XW. Effect of oxymatrine on the p38 mitogen-activated protein kinases signalling pathway in rats with CCl4 induced hepatic fibrosis. Chin Med J (Engl) 2009;122:1449–1454. [PubMed] [Google Scholar]
  • 26.Nieto N. Ethanol and fish oil induce NFkappaB transactivation of the collagen alpha2(I) promoter through lipid peroxidation-driven activation of the PKC-PI3K-Akt pathway. Hepatology. 2007;45:1433–1445. doi: 10.1002/hep.21659. [DOI] [PubMed] [Google Scholar]
  • 27.Yuantai W, Tiancai W, Qiu Z. PD98059 inhibits expression of pERK1 protein and collagen alpha1(I) mRNA in rat pancreatic stellate cells activated by platelet-derived growth factor. Indian J Gastroenterol. 2005;24:100–103. [PubMed] [Google Scholar]
  • 28.Anania FA, Womack L, Jiang M, Saxena NK. Aldehydes potentiate alpha(2)(I) collagen gene activity by JNK in hepatic stellate cells. Free Radic Biol Med. 2001;30:846–857. doi: 10.1016/s0891-5849(01)00470-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Chen A, Davis BH. The DNA binding protein BTEB mediates acetaldehyde-induced, jun N-terminal kinase-dependent alphaI(I) collagen gene expression in rat hepatic stellate cells. Mol Cell Biol. 2000;20:2818–2826. doi: 10.1128/mcb.20.8.2818-2826.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.De Minicis S, Candelaresi C, Marzioni M, Saccomano S, Roskams T, Casini A, Risaliti A, et al. Role of endogenous opioids in modulating HSC activity in vitro and liver fibrosis in vivo. Gut. 2008;57:352–364. doi: 10.1136/gut.2007.120303. [DOI] [PubMed] [Google Scholar]
  • 31.Buttner C, Skupin A, Rieber EP. Transcriptional activation of the type I collagen genes COL1A1 and COL1A2 in fibroblasts by interleukin-4: analysis of the functional collagen promoter sequences. J Cell Physiol. 2004;198:248–258. doi: 10.1002/jcp.10395. [DOI] [PubMed] [Google Scholar]
  • 32.de Obanos MP Perez, Lopez-Zabalza MJ, Arriazu E, Modol T, Prieto J, Herraiz MT, Iraburu MJ. Reactive oxygen species (ROS) mediate the effects of leucine on translation regulation and type I collagen production in hepatic stellate cells. Biochim Biophys Acta. 2007;1773:1681–1688. doi: 10.1016/j.bbamcr.2007.07.005. [DOI] [PubMed] [Google Scholar]
  • 33.Comporti M. Three models of free radical-induced cell injury. Chem Biol Interact. 1989;72:1–56. doi: 10.1016/0009-2797(89)90016-1. [DOI] [PubMed] [Google Scholar]
  • 34.Fickert P, Thueringer A, Moustafa T, Silbert D, Gumhold J, Tsybrovskyy O, Lebofsky M, et al. The role of osteopontin and tumor necrosis factor alpha receptor-1 in xenobiotic-induced cholangitis and biliary fibrosis in mice. Lab Invest. 2010;90:844–852. doi: 10.1038/labinvest.2010.61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Chen Y, Bal BS, Gorski JP. Calcium and collagen binding properties of osteopontin, bone sialoprotein, and bone acidic glycoprotein-75 from bone. J Biol Chem. 1992;267:24871–24878. [PubMed] [Google Scholar]
  • 36.Singh K, DeVouge MW, Mukherjee BB. Physiological properties and differential glycosylation of phosphorylated and nonphosphorylated forms of osteopontin secreted by normal rat kidney cells. J Biol Chem. 1990;265:18696–18701. [PubMed] [Google Scholar]
  • 37.Cho MK, Lee GH, Park EY, Kim SG. Hyaluronic acid inhibits adhesion of hepatic stellate cells in spite of its stimulation of DNA synthesis. Tissue Cell. 2004;36:293–305. doi: 10.1016/j.tice.2004.05.001. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supp Figure S1
Supplementary Legends&Supp Table S1
Supp Figure S2
Supp Figure S3
Supp Figure S4
Supp Figure S5
Supp Figure S6
Supp Figure S7
Supp Figure S8
Supp Figure S9

RESOURCES