S-nitrosoglutathione reductase (GSNOR) enhances ...S-nitrosoglutathione reductase (GSNOR) enhances...

6
S-nitrosoglutathione reductase (GSNOR) enhances vasculogenesis by mesenchymal stem cells Samirah A. Gomes a , Erika B. Rangel a , Courtney Premer a , Raul A. Dulce a , Yenong Cao a , Victoria Florea a , Wayne Balkan a , Claudia O. Rodrigues a,b , Andrew V. Schally c,d,e,1 , and Joshua M. Hare a,f,1 a Interdisciplinary Stem Cell Institute, b Department of Molecular and Cellular Pharmacology, c Department of Pathology, d Division of Hematology/Oncology, and f Division of Cardiology, Department of Medicine, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136; and e Endocrine, Polypeptide and Cancer Institute, Veterans Affairs Medical Center, Miami, FL 33136 Contributed by Andrew V. Schally, November 27, 2012 (sent for review October 31, 2012) Although nitric oxide (NO) signaling promotes differentiation and maturation of endothelial progenitor cells, its role in the differen- tiation of mesenchymal stem cells (MSCs) into endothelial cells remains controversial. We tested the role of NO signaling in MSCs derived from WT mice and mice homozygous for a deletion of S-nitrosoglutathione reductase (GSNOR /), a denitrosylase that regulates S-nitrosylation. GSNOR /MSCs exhibited markedly diminished capacity for vasculogenesis in an in vitro Matrigel tubeforming assay and in vivo relative to WT MSCs. This decrease was associated with down-regulation of the PDGF receptorα (PDGFRα) in GSNOR /MSCs, a receptor essential for VEGF-A action in MSCs. Pharmacologic inhibition of NO synthase with L-N G -nitroarginine methyl ester (L-NAME) and stimulation of growth hormonereleasing hormone receptor (GHRHR) with GHRH agonists augmented VEGF-A production and normalized tube formation in GSNOR /MSCs, whereas NO donors or PDGFR antagonist re- duced tube formation 50% by murine and human MSCs. The antagonist also blocked the rescue of tube formation in GSNOR /MSCs by L-NAME or the GHRH agonists JI-38, MR-409, and MR-356. Therefore, GSNOR /MSCs have a decient capacity for endothelial differentiation due to downregulation of PDGFRα related to NO/GSNOR imbalance. These ndings unravel important aspects of modulation of MSCs by VEGF-A activation of the PDGFR and illustrate a paradoxical inhibitory role of S-nitrosylation sig- naling in MSC vasculogenesis. Accordingly, disease states charac- terized by NO deciency may trigger MSC-mediated vasculogenesis. These ndings have important implications for therapeutic applica- tion of GHRH agonists to ischemic disorders. angiogenesis | nitrosoredox imbalance N itric oxide (NO) and VEGF signaling promotes vasculogenesis by endothelial progenitor cells (EPCs) (15). For example, mice decient in endothelial NO synthase (NOS3 /) show re- duced VEGF-induced mobilization of bone marrow progenitor cells to sites of injury (4). In EPC-mediated vasculogenesis, VEGF-A activates its major receptor [VEGF receptor 2 (VEGFR2)], trig- gering cell differentiation into mature endothelial cells (ECs) and enhancing angiogenesis (6, 7). Mesenchymal stem cells (MSCs) also participate in postnatal angiogenesis (8, 9), and vascular pericytes, which are crucial for maintaining vascular integrity, share similar phenotypic features with MSCs (10). Exogenously administered, MSCs readily form new capillaries and medium-sized arteries (11, 12), properties important for the tissue regenerative capacity of MSCs (13). We and others have shown that MSCs differentiate into endothelial cells in vitro (14) and in vivo and contribute to neovascularization, particularly during tissue ischemia and tumor vascularization (8, 11, 12, 15). As with EPCs, VEGF also plays an important role in stimulating MSC differentiation, but does so by activating the PDGF receptor (PDGFR) as opposed to the VEGFR2, which is absent on MSCs (16). However, the impact of NO signaling in the differentiation of MSCs into endothelial cells has not been pre- viously tested. Given the similar signaling involved in endothelial differentiation of EPCs and MSCs, we reasoned that NO plays an equivalent role in this process. Accordingly, we tested the hypothesis that NO signaling, me- diated by small molecular weight thiols (molecular weight < 500), promotes MSC differentiation into endothelial cells. To test our hypothesis, we assessed the functional consequences of deletion of S-nitrosoglutathione reductase (GSNOR), which in turn increases S-nitrosothiols (17, 18), on MSC-mediated vasculo- genesis. We report that paradoxically, MSCs from GSNOR /mice exhibit diminished endothelial differentiation, thereby demonstrating an inhibitory effect of S-nitrosylation on vascu- logenesis mediated by MSCs. Results Murine Bone Marrow MSC Characterization. Both WT and GSNOR /- derived MSCs were spindle shaped, adherent to plastic tissue culture dishes (Fig. S1A), and negative for Lineage, a mixture of hematopoietic markers, CD34 and CD45, and positive for stem cell antigens SCA-1, CD73, CD90.2, and CD105 (Fig. S1 B and C). NO Signaling and MSCs. Denitrosylation is controlled in signicant part by GSNOR, a unique and specic enzyme for which S-nitro- soglutathione (GSNO) is the substrate (18) (Fig. S2A). Neither MSCs (Fig. S2 B and C) nor liver (Fig. S2D) from GSNOR /mice expresses GSNOR. NOS1 and NOS2 were constitutively expressed in both strains of MSCs (Fig. S2 E and F), whereas NOS3 expres- sion was absent from both strains (Fig. S2G). Interestingly, NOS1 mRNA was 100-fold higher in GSNOR /mice (2.7 × 10 3 ± 3 × 10 2 absolute number of transcripts, ΔCt) compared with WT MSCs (2 × 10 4 ± 2 × 10 3 , P < 0.05; Fig. S2E). Despite the up-regulation of NOS1, actual NO production, measured by 4,5-diamino-uores- cein diacetate (DAF-2DA), was nearly identical between the strains (Fig. S2H). Thus, GSNOR deciency up-regulates NOS1 but does not lead to increased NO production. GSNOR /MSCs Exhibit Impaired Formation of Capillary TubeLike Structures in Vitro. Next we used a Matrigel assay to analyze the ability of MSCs to form tube-like structures in vitro. Cells were grown in endothelial medium (EGM-2; Lonza) for 1 wk before plating on Matrigel. Surprisingly, GSNOR /-derived MSCs formed signicantly fewer (29.9 ± 11.75 vs. 50.2 ± 14.04, P < 0.001) and shorter (62.8 ± 14.0 vs. 124.2 ± 49.0 μm, P < 0.001) Author contributions: S.A.G., W.B., and J.M.H. designed research; S.A.G., E.B.R., C.P., R.A.D., Y.C., V.F., and C.O.R. performed research; S.A.G. and J.M.H. analyzed data; A.V.S. and J.M.H. contributed new reagents/analytic tools; and S.A.G., E.B.R., C.O.R., A.V.S., and J.M.H. wrote the paper. The authors declare no conict of interest. Freely available online through the PNAS open access option. See Commentary on page 2695. 1 To whom correspondence may be addressed. E-mail: [email protected] or jhare@ med.miami.edu. This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10. 1073/pnas.1220185110/-/DCSupplemental. 28342839 | PNAS | February 19, 2013 | vol. 110 | no. 8 www.pnas.org/cgi/doi/10.1073/pnas.1220185110 Downloaded by guest on July 3, 2021

Transcript of S-nitrosoglutathione reductase (GSNOR) enhances ...S-nitrosoglutathione reductase (GSNOR) enhances...

  • S-nitrosoglutathione reductase (GSNOR) enhancesvasculogenesis by mesenchymal stem cellsSamirah A. Gomesa, Erika B. Rangela, Courtney Premera, Raul A. Dulcea, Yenong Caoa, Victoria Floreaa, Wayne Balkana,Claudia O. Rodriguesa,b, Andrew V. Schallyc,d,e,1, and Joshua M. Harea,f,1

    aInterdisciplinary Stem Cell Institute, bDepartment of Molecular and Cellular Pharmacology, cDepartment of Pathology, dDivision of Hematology/Oncology,and fDivision of Cardiology, Department of Medicine, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136; and eEndocrine,Polypeptide and Cancer Institute, Veterans Affairs Medical Center, Miami, FL 33136

    Contributed by Andrew V. Schally, November 27, 2012 (sent for review October 31, 2012)

    Although nitric oxide (NO) signaling promotes differentiation andmaturation of endothelial progenitor cells, its role in the differen-tiation of mesenchymal stem cells (MSCs) into endothelial cellsremains controversial. We tested the role of NO signaling in MSCsderived from WT mice and mice homozygous for a deletion ofS-nitrosoglutathione reductase (GSNOR−/−), a denitrosylase thatregulates S-nitrosylation. GSNOR−/− MSCs exhibited markedlydiminished capacity for vasculogenesis in an in vitro Matrigeltube–forming assay and in vivo relative to WT MSCs. This decreasewas associated with down-regulation of the PDGF receptorα(PDGFRα) in GSNOR−/− MSCs, a receptor essential for VEGF-Aaction in MSCs. Pharmacologic inhibition of NO synthase withL-NG-nitroarginine methyl ester (L-NAME) and stimulation of growthhormone–releasing hormone receptor (GHRHR) with GHRH agonistsaugmented VEGF-A production and normalized tube formation inGSNOR−/− MSCs, whereas NO donors or PDGFR antagonist re-duced tube formation ∼50% by murine and human MSCs. Theantagonist also blocked the rescue of tube formation inGSNOR−/− MSCs by L-NAME or the GHRH agonists JI-38, MR-409,and MR-356. Therefore, GSNOR−/− MSCs have a deficient capacityfor endothelial differentiation due to downregulation of PDGFRαrelated to NO/GSNOR imbalance. These findings unravel importantaspects of modulation of MSCs by VEGF-A activation of the PDGFRand illustrate a paradoxical inhibitory role of S-nitrosylation sig-naling in MSC vasculogenesis. Accordingly, disease states charac-terized by NO deficiency may triggerMSC-mediated vasculogenesis.These findings have important implications for therapeutic applica-tion of GHRH agonists to ischemic disorders.

    angiogenesis | nitroso–redox imbalance

    Nitric oxide (NO) andVEGF signaling promotes vasculogenesisby endothelial progenitor cells (EPCs) (1–5). For example,mice deficient in endothelial NO synthase (NOS3−/−) show re-duced VEGF-induced mobilization of bone marrow progenitorcells to sites of injury (4). In EPC-mediated vasculogenesis, VEGF-Aactivates its major receptor [VEGF receptor 2 (VEGFR2)], trig-gering cell differentiation into mature endothelial cells (ECs) andenhancing angiogenesis (6, 7).Mesenchymal stem cells (MSCs) also participate in postnatal

    angiogenesis (8, 9), and vascular pericytes, which are crucial formaintaining vascular integrity, share similar phenotypic featureswith MSCs (10). Exogenously administered, MSCs readily formnew capillaries and medium-sized arteries (11, 12), propertiesimportant for the tissue regenerative capacity of MSCs (13). Weand others have shown that MSCs differentiate into endothelialcells in vitro (14) and in vivo and contribute to neovascularization,particularly during tissue ischemia and tumor vascularization (8,11, 12, 15). As with EPCs, VEGF also plays an important role instimulating MSC differentiation, but does so by activating thePDGF receptor (PDGFR) as opposed to the VEGFR2, which isabsent on MSCs (16). However, the impact of NO signaling in thedifferentiation of MSCs into endothelial cells has not been pre-viously tested. Given the similar signaling involved in endothelial

    differentiation of EPCs and MSCs, we reasoned that NO playsan equivalent role in this process.Accordingly, we tested the hypothesis that NO signaling, me-

    diated by small molecular weight thiols (molecular weight < 500),promotes MSC differentiation into endothelial cells. To test ourhypothesis, we assessed the functional consequences of deletionof S-nitrosoglutathione reductase (GSNOR), which in turnincreases S-nitrosothiols (17, 18), on MSC-mediated vasculo-genesis. We report that paradoxically, MSCs from GSNOR−/−

    mice exhibit diminished endothelial differentiation, therebydemonstrating an inhibitory effect of S-nitrosylation on vascu-logenesis mediated by MSCs.

    ResultsMurine BoneMarrowMSCCharacterization.Both WT and GSNOR−/−-derived MSCs were spindle shaped, adherent to plastic tissueculture dishes (Fig. S1A), and negative for Lineage, a mixture ofhematopoietic markers, CD34 and CD45, and positive for stemcell antigens SCA-1, CD73, CD90.2, and CD105 (Fig. S1 B and C).

    NO Signaling and MSCs. Denitrosylation is controlled in significantpart by GSNOR, a unique and specific enzyme for which S-nitro-soglutathione (GSNO) is the substrate (18) (Fig. S2A). NeitherMSCs (Fig. S2B andC) nor liver (Fig. S2D) fromGSNOR−/−miceexpresses GSNOR. NOS1 and NOS2 were constitutively expressedin both strains of MSCs (Fig. S2 E and F), whereas NOS3 expres-sion was absent from both strains (Fig. S2G). Interestingly, NOS1mRNA was ∼100-fold higher in GSNOR−/− mice (2.7 × 103 ± 3 ×102 absolute number of transcripts,ΔCt) comparedwithWTMSCs(2× 104± 2 × 103, P< 0.05; Fig. S2E). Despite the up-regulation ofNOS1, actual NO production, measured by 4,5-diamino-fluores-cein diacetate (DAF-2DA), was nearly identical between thestrains (Fig. S2H). Thus, GSNOR deficiency up-regulates NOS1but does not lead to increased NO production.

    GSNOR−/− MSCs Exhibit Impaired Formation of Capillary Tube–LikeStructures in Vitro. Next we used a Matrigel assay to analyze theability of MSCs to form tube-like structures in vitro. Cells weregrown in endothelial medium (EGM-2; Lonza) for 1 wk beforeplating on Matrigel. Surprisingly, GSNOR−/−-derived MSCsformed significantly fewer (29.9 ± 11.75 vs. 50.2 ± 14.04, P <0.001) and shorter (62.8 ± 14.0 vs. 124.2 ± 49.0 μm, P < 0.001)

    Author contributions: S.A.G., W.B., and J.M.H. designed research; S.A.G., E.B.R., C.P., R.A.D.,Y.C., V.F., and C.O.R. performed research; S.A.G. and J.M.H. analyzed data; A.V.S. and J.M.H.contributed new reagents/analytic tools; and S.A.G., E.B.R., C.O.R., A.V.S., and J.M.H. wrotethe paper.

    The authors declare no conflict of interest.

    Freely available online through the PNAS open access option.

    See Commentary on page 2695.1To whom correspondence may be addressed. E-mail: [email protected] or [email protected].

    This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10.1073/pnas.1220185110/-/DCSupplemental.

    2834–2839 | PNAS | February 19, 2013 | vol. 110 | no. 8 www.pnas.org/cgi/doi/10.1073/pnas.1220185110

    Dow

    nloa

    ded

    by g

    uest

    on

    July

    3, 2

    021

    http://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1220185110/-/DCSupplemental/pnas.201220185SI.pdf?targetid=nameddest=SF1http://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1220185110/-/DCSupplemental/pnas.201220185SI.pdf?targetid=nameddest=SF1http://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1220185110/-/DCSupplemental/pnas.201220185SI.pdf?targetid=nameddest=SF2http://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1220185110/-/DCSupplemental/pnas.201220185SI.pdf?targetid=nameddest=SF2http://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1220185110/-/DCSupplemental/pnas.201220185SI.pdf?targetid=nameddest=SF2http://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1220185110/-/DCSupplemental/pnas.201220185SI.pdf?targetid=nameddest=SF2http://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1220185110/-/DCSupplemental/pnas.201220185SI.pdf?targetid=nameddest=SF2http://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1220185110/-/DCSupplemental/pnas.201220185SI.pdf?targetid=nameddest=SF2http://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1220185110/-/DCSupplemental/pnas.201220185SI.pdf?targetid=nameddest=SF2mailto:[email protected]:[email protected]:[email protected]://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1220185110/-/DCSupplementalhttp://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1220185110/-/DCSupplementalwww.pnas.org/cgi/doi/10.1073/pnas.1220185110

  • tubes than those derived from WT mice (Fig. 1 A–C). To testwhether this impairment was NO-mediated, we inhibited NOproduction in GSNOR−/− MSCs with the NO synthase inhibitorL-NG-nitroarginine methyl ester (L-NAME). This treatmentcompletely normalized the number (49.9 ± 14.8, P < 0.001) andlength (82.8 ± 20.5 μm, P < 0.001) of GSNOR−/− tubes (Fig. 1A–C), indicating that inhibition of NO production restores net-work formation by MSCs. L-NAME had no effect on WT MSCtube formation; however, treatment with the NO donors,S-nitrosoglutathione (GSNO) and S-nitroso-N-acetyl-D,L-penicil-lamine (SNAP), impaired tube formation (15.6 ± 9.0 and 31.8 ±9.9, P < 0.001) and decreased tube length (98.4 ± 56.6 and 57.1 ±24.9 μm, P < 0.001) by WTMSCs, confirming that small molecularweight S-nitrosothiols inhibit the vasculogenic potential of MSCs(Fig. S3 A–C).

    MSCs from NOS2−/− Mice Produce Enhanced Capillary Tube–LikeFormation in Vitro. We also tested the impact of reducing in-tracellular NO production by assessing the vasculogenic potentialof MSCs isolated from mice lacking NOS2 (NOS2−/−). Signifi-cantly, MSCs from NOS2−/− mice formed more tubes (92.8 ± 26.0vs. 50.0 ± 15.4, P < 0.001) than WT MSCs, and the NOS2−/−

    tubes were longer (121.0 ± 52.0 vs. 62.8 ± 34.6 μm, P < 0.001)than tubes from GSNOR−/− MSCs but were similar (121.0 ± 52.0vs. 124.2 ± 76.0 μm) to WT tubes (Fig. S4 A–C), further con-firming the inhibitory role of NO on MSC vasculogenesis. Wewere unable to generate MSCs from NOS1−/− mice, suggestingan indispensable role for this enzyme in MSC biology.

    NO/GSNOR Modulates VEGF-A/PDGFR Signaling. Accordingly we ex-amined the expression of VEGF-A, VEGFR2, PDGFRα, andPDGFRβ in MSCs (Fig. 2 A–J). Neither WT nor GSNOR−/−MSCs expressed VEGFR2, although its ligand VEGF-A was pro-duced at similar levels by both strains (Fig. 2 A–D). PDGFRαexpression was diminished by ∼50% in GSNOR−/− MSCs as mea-sured by FACS (Fig. 2 E–G), qRT-PCR (Fig. 2H), and Westernblotting (Fig. 2 I and J). The expression of PDGFRβ did not change(Fig. 2H). Under physiologic conditions, MSCs express high levelsof PDGFRα but not VEGFR2 (16). Differentiation of MSC intoendothelial cells requires activation of the PDGFRα by VEGF-Afollowing a switch of receptors where VEGFR2 increases andPDGFRα decreases (19). Together these findings support theparadigm that environments rich in bioavailable NO or geneticmodifications (deletion of GSNOR) inhibit PDGFRα expres-sion by MSCs resulting in impaired endothelial differentiation

    (Fig. 2K). Furthermore, incubation with GSNO down-regu-lated PDGFRα by approximately twofold and up-regulatedVEGF-A by∼2.5-fold inWTMSCs, supporting the actions of low-molecular-weight thiols in mediating this phenotype (Fig. S5A).Finally, in GSNOR−/− MSCs, inhibition of NOS with L-NAMEaugmented VEGF-A production ∼4.5-fold (Fig. 3A), demon-strating that NO levels modulate VEGF-A production. Activa-tion of the GHRH receptor, with JI-38, a synthetic agonist, alsoincreased VEGF-A production by 2.8-fold in GSNOR−/− MSCs(Fig. 3B).

    VEGF-A/PDGFR Signaling Is a Key in MSC-Mediated Vasculogenesis.We next examined whether inhibition of PDGFRα, using a spe-cific PDGFR IV antagonist (PIV), could reduce tube formationby MSCs. Treatment with PIV (0.1 μmol/L) impaired tube for-mation in both WT (∼2.6-fold; Fig. 3 C and D; Fig. S6A) andGSNOR−/− MSCs (∼4.1 fold; Fig. 3 E and F; Fig. S6B). How-ever, the impact of PIV was much greater on GSNOR−/− tubesthan WT. In addition, while treatment with L-NAME rescuedtube formation by GSNOR−/− MSCs, PIV counteracted this ef-fect, confirming that PDGFRα activation is required for tubeformation by MSCs (Fig. 3 E and F; Fig. S6B). Furthermore,when GSNOR−/− MSCs were treated with L-NAME, VEGF-Aexpression was significantly increased (Fig. 3A), leading to en-hanced tube formation through PDGFRα activation (Fig. 3 Eand F), although mRNA expression of PDGFRα or β did notchange following NOS inhibition.

    Activation of VEGF-A Production with GHRH Agonists. As an alter-native to NOS inhibition, we sought to augment VEGF-A pro-duction by activating the growth hormone–releasing hormone(GHRH) receptor, which was shown to be present on MSCs (Fig.S7 A–C). Stimulation of the GHRH receptor with the syntheticgrowth hormone–releasing hormone GHRH agonist JI-38 (20) orother potent GHRH agonists, MR-409 and MR-356 (Fig. S7 D–F), also normalized the impaired tube formation by GSNOR−/−

    MSCs and did so to a similar extent as L-NAME (69 ± 11 tubesand 73 ± 33-μm tube length, P < 0.001; Fig. 3). Treatment withJI-38 and L-NAME in WT MSCs did not affect tube formation(Fig. 3 C and D; Fig. S3 A–C), although stimulation of WT MSCswith JI-38 up-regulated VEGF-A expression by ∼2.5 fold (Fig.S5B). As with L-NAME, blockade of PDGFRα abolished theimpact of this GHRH agonist (Fig. 3 E and F).

    Fig. 1. Impaired capillary tube–like formation from GSNOR−/− MSCs in vitro. (A) Representative images of tube-like formation by WT and GSNOR−/− MSCsplated on Matrigel-coated plates for 0, 6, and 24 h in the presence of vehicle (rows 1 and 3) or 15 μmol/L L-NAME (rows 2 and 4) (magnification: 10×).Quantification of the number (B) and length (μm) (C) of tubes at 24 h (n ≥ 3, *P < 0.001 vs. WT, †P < 0.001 vs. GSNOR−/−).

    Gomes et al. PNAS | February 19, 2013 | vol. 110 | no. 8 | 2835

    CELL

    BIOLO

    GY

    SEECO

    MMEN

    TARY

    Dow

    nloa

    ded

    by g

    uest

    on

    July

    3, 2

    021

    http://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1220185110/-/DCSupplemental/pnas.201220185SI.pdf?targetid=nameddest=SF3http://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1220185110/-/DCSupplemental/pnas.201220185SI.pdf?targetid=nameddest=SF3http://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1220185110/-/DCSupplemental/pnas.201220185SI.pdf?targetid=nameddest=SF3http://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1220185110/-/DCSupplemental/pnas.201220185SI.pdf?targetid=nameddest=SF4http://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1220185110/-/DCSupplemental/pnas.201220185SI.pdf?targetid=nameddest=SF4http://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1220185110/-/DCSupplemental/pnas.201220185SI.pdf?targetid=nameddest=SF4http://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1220185110/-/DCSupplemental/pnas.201220185SI.pdf?targetid=nameddest=SF5http://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1220185110/-/DCSupplemental/pnas.201220185SI.pdf?targetid=nameddest=SF6http://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1220185110/-/DCSupplemental/pnas.201220185SI.pdf?targetid=nameddest=SF6http://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1220185110/-/DCSupplemental/pnas.201220185SI.pdf?targetid=nameddest=SF6http://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1220185110/-/DCSupplemental/pnas.201220185SI.pdf?targetid=nameddest=SF7http://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1220185110/-/DCSupplemental/pnas.201220185SI.pdf?targetid=nameddest=SF7http://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1220185110/-/DCSupplemental/pnas.201220185SI.pdf?targetid=nameddest=SF7http://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1220185110/-/DCSupplemental/pnas.201220185SI.pdf?targetid=nameddest=SF7http://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1220185110/-/DCSupplemental/pnas.201220185SI.pdf?targetid=nameddest=SF7http://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1220185110/-/DCSupplemental/pnas.201220185SI.pdf?targetid=nameddest=SF7http://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1220185110/-/DCSupplemental/pnas.201220185SI.pdf?targetid=nameddest=SF7http://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1220185110/-/DCSupplemental/pnas.201220185SI.pdf?targetid=nameddest=SF3http://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1220185110/-/DCSupplemental/pnas.201220185SI.pdf?targetid=nameddest=SF3http://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1220185110/-/DCSupplemental/pnas.201220185SI.pdf?targetid=nameddest=SF3http://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1220185110/-/DCSupplemental/pnas.201220185SI.pdf?targetid=nameddest=SF5http://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1220185110/-/DCSupplemental/pnas.201220185SI.pdf?targetid=nameddest=SF5

  • NO Signaling Impaired Vasculogenesis in Vivo. To confirm our invitro data, we examined vasculogenesis in vivo by performing aMatrigel plug assay. GFP-labeledMSCs fromWT andGSNOR−/−

    mice were injected s.c. into immunocompromised mice (NOD-SCID). Two weeks later, Matrigel plugs were harvested andassayed for immunofluorescence staining and determination ofcapillary formation (Fig. 4A). MSCs from GSNOR−/− mice hadimpaired blood vessel formation compared withWTMSCs (1.84±1.3 vs. 10.2 ± 2.7/mm2, P < 0.01; Fig. 4 A and B). Furthermore, wedetermined the percentage of GFP-transduced MSCs that differ-entiated into endothelial cells and also formed blood vessels (Fig.4 C and D). GSNOR−/− MSCs exhibited diminished endothelialdifferentiation (7.7 ± 1.64% vs. 12.5 ± 1.0%, P < 0.02) as assessedby isolectin staining (in orange) colocalized with GFP and alsoreduced the number of GFP+ blood vessels (0.4 ± 0.2 vs. 1.6 ± 0.4mm2) compared with WT MSCs . These results agree with our invitro data and support the negative effect of NO signaling on vas-culogenesis by MSCs.

    NO Donors Impaired the Ability of Human Bone Marrow–DerivedMSCs to Form Capillary Tube–Like Structures in Vitro. We nexttested whether human and mouse MSCs responded similarly toNO donors and observed that NO donors also reduced tubeformation by hMSCs. Whereas the human cells formed a net-work more rapidly than murine cells (Fig. S8), hMSCs treatedwith NO donors GSNO or SNAP produced half as many tubes asvehicle-treated cells. Treatment with 10, 40, and 100 μmol/L

    GSNO reduced the number (26.6 ± 7.1, 24.8 ± 4.3, and 27.6 ±4.0, respectively, vs. 49.2.0 ± 8.1, P < 0.001) and length (94.0 ±34.3, 78.0 ± 41.0 and 85.0 ± 34.4, respectively, vs. 125.0 ± 52.6,P < 0.001) of tubes (Fig. 5 A and B). Similarly, treatment with 10and 100 μmol/L SNAP reduced tube number (34.0 ± 6.1 and 25.0 ±4.9 vs. 49.2 ± 8.1, P < 0.001) and length (82.0 ± 29.2 and 91.0 ±32.2 vs. 128 ± 52 μm, P < 0.001) compared with untreated hMSCs.Similar to murine MSCs, inhibition of NOS with L-NAME had noeffect on hMSC tube formation (Fig. 5 A and B).Finally, to test whetherMSCs respond differently to endothelial

    cells, we used the Matrigel assay with human umbilical vein en-dothelial cells (HUVECs), and in contrast to mouse and humanMSCs, HUVECs treated with NO donors (GSNO and SNAP)exhibited enhanced network formation, but NOS inhibition with15 μmol/L L-NAMEwas not sufficient to reduce tube-like capillarystructure formation (Fig. S9 A–C). These experiments revealedthat NO signaling has a negative impact on endothelial differen-tiation byMSCs in contrast to (human) endothelial cells whereNOfavors angiogenesis.

    DiscussionThe major finding shown in this work is that GSNOR signalingcontributes to MSC-mediated vasculogenesis. We demonstratedthis principle in a variety of ways. First, MSCs from GSNOR-de-ficient mice exhibited attenuated vasculogenesis both in vitro andin vivo. Similarly, S-nitrosothiol (SNO) donors diminished vascu-logenesis in human MSCs. Pharmacological inhibition of NO in

    Fig. 2. Down-regulation of PDGFRα in GSNOR−/− MSCs. (A–C) Representative FACS analysis depicting absence of VEGFR2 (

  • GSNOR−/− MSCs, or genetic reduction of NO production in theNOS2−/−, enhanced vasculogenesis by MSCs. Importantly, theopposite effect was shown to be true for HUVECs in which NO

    enhanced vascular tube formation. Together, these findings reveala unique, paradoxical mode of vascular regulation between MSCsand endothelial cells and suggest that MSC-mediated vascularformation may increase in states of NO deficiency. Importantly,the simultaneous potentiation of EPC- and inhibition of MSC-mediated vasculogenesis by NO may represent a mechanism forpreserving MSC regulatory capacity in an environment of en-hanced EPC vascular formation.NO signaling in MSCs has heretofore not been examined. The

    present study revealed that GSNOR deficiency impairs MSC-mediated postnatal vasculogenesis. Previously, we and othersshowed that GSNOR is a key regulator of cardiovascular func-tion and vascular tone, regulating a dynamic nitrosylation/deni-trosylation cycle of proteins (21, 22). We chose to investigateS-nitrosylation as the primary signaling mode exerting NO bio-activity and took advantage of the fact that both excessive NOproduction by NOS activation or reduced SNO metabolism dueto GSNOR deficiency enhances S-nitrosylation (23). Accord-ingly, we used MSCs from the GSNOR−/− as the primary modeof investigating NO/SNO signaling in these stem cells.At the mechanistic level, our studies reveal that the underlying

    basis by whichNO signaling throughGSNORdirectly affectsMSCvasculogenesis appears to be regulation of PDGFRα abundance(Fig. 2K). Bone marrow MSCs, which do not express VEGFR2(16), respond to VEGF-A through a PDGFR–ligand interaction(16, 24, 25). Here we show that GSNOR−/− MSCs have reducedexpression of PDGFRα, which is causally linked to impaired en-dothelial differentiation. Importantly, treatment with L-NAMErescued capillary network formation from GSNOR−/− MSCs byincreasing VEGF-A production followed by activation ofPDGFRα. However, this effect was not observed in WT MSCswhen similarly treated with L-NAME, which may represent celltype differences in dose response. Human MSCs cocultured withendothelial cells differentiate into endothelial-like cells, and thisprocess can be inhibited by VEGF-A antisera (26), corroboratingour finding that VEGF-A is crucial for MSC-mediated vasculo-genesis. In our study, we demonstrated clearly that VEGF-A/PDGFRα directly affected MSC fate decisions, although we can-not discount that the interactions between MSCs and endothelialcells are crucial for vascular homeostasis and repair. In this re-spect, MSCs are thought to be intimately involved with vascularhomeostasis throughout the body, preserving vascular integrity bydifferentiating to pericytes (9, 27).

    Fig. 3. VEGF-A/PDGFR signaling in MSC-mediated vasculogenesis. (A and B)GSNORMSCs increase VEGF-A production when stimulated by L-NAME and JI-38. Quantification of tube formation on Matrigel shows that MSCs from bothWT (C and D) and GSNOR−/− (E and F) mice exhibit reduced tube number andlength (μm) when treated with PDGFRα inhibitor (PIV) either alone or in thepresence of 0.25 μmol/L JI-38 or 15 μmol/L L-NAME. (n ≥ 3, *P < 0.05).

    Fig. 4. MSCs from GSNOR−/− mice exhibit reduced endothelial differentiation and impaired blood vessel formation in vivo. (A and B) Matrigel plug (2 wkafter injection) containing GFP+ MSCs from WT and GSNOR−/−. (B) H&E staining with blood vessel formation indicated by the black arrows (C) MSCs fromGSNOR−/− mice following isolectin (red) and GFP (green) staining shows colocalization (orange) and exhibit diminished potential to differentiate into en-dothelial cells than WT MSCs. (D) Quantification of the number of GFP+ blood vessels containing autofluorescent red blood cells [white arrows, see (A)]. (E)Quantification of blood vessel formation 2 wk after injection. *P < 0.05.

    Gomes et al. PNAS | February 19, 2013 | vol. 110 | no. 8 | 2837

    CELL

    BIOLO

    GY

    SEECO

    MMEN

    TARY

    Dow

    nloa

    ded

    by g

    uest

    on

    July

    3, 2

    021

  • We also used a second strategy to test whether VEGF-A sig-naling rescued diminished vasculogenesis by GSNOR−/− MSCs.Based on our previous observations, that the activation of GHRHreceptors leads to an increased VEGF-A in vivo (28), we usedGHRH agonists. Indeed these agonists normalized vasculogenesisin a PDGFRα-dependent manner. These findings have importantimplications for the therapeutic application of GHRH agonists toischemic disorders. We previously showed that GHRH agonistsimprove cardiac structure and function after myocardial infarction(28, 29). These findings shed further light on the beneficial effectsof GHRH agonists on wound healing (30) and the maintenance ofpancreatic β islets (31).Our results confirmed that NO signaling through small-mo-

    lecular-weight thiols is an important regulator of vasculogenesisin MSCs. More specifically, we found that endothelial differen-tiation is impaired in GSNOR−/− MSCs in vivo, suggesting thatGSNOR represents a valuable pharmacologic target for regu-lation of neovascularization. Moreover, our results support thatS-nitrosylation plays an important role in MSC-mediated vas-culogenesis and may affect cell fate decisions. Importantly, Limaet al. (22) showed that myocardial infarction size is reduced inGSNOR−/− mice, an effect that was associated with enhancedneovascularization, suggesting that neovascularization in thatsetting is primarily a result of EPC activation.In addition, we demonstrated that decreased production of NO

    enhanced tube-like formation on Matrigel in vitro. This result wasillustrated by the increased tube formation by MSCs fromNOS2−/− mice, the normalization of tube formation by treatmentof GSNOR−/− MSCs with L-NAME, and ultimately, the re-duction in tube formation in WT mMSCs and in hMSCs trea-ted with NO donors, indicating a cross-species effect.Therefore, deficiency of NO has a positive impact on MSC-

    mediated vasculogenesis. In agreement with our data, Wanget al. (32) demonstrated that treatment of human MSCs withNO donors suppresses production of the proangiogenic factorsVEGF and hepatocyte growth factor. Moreover, bone marrow–derived MSCs isolated from patients with systemic sclerosis,a disease characterized by NO overproduction, vascular dys-function, and systemic fibrosis, have impaired endothelial celldifferentiation (33), consistent with our findings that environ-ments rich in NO impair postnatal vasculogenesis by MSCs.Together, these findings suggest a reduced regenerative capacityof MSCs in nitroso/redox unbalanced environments such as

    heart failure, sepsis, and neuronal degenerative diseases (34).Furthermore, NO may attenuate the protection by MSCs in is-chemic myocardium by serving as a natural braking mechanismfor MSC-induced neovascularization in a tissue recovering fromischemic injury.We showed that NO synthases, NOS1 and NOS2, but not

    NOS3, were constitutively expressed in MSCs. Importantly,MSCs from WT and GSNOR−/− mice had equivalent NOproduction; however, NOS1 expression was up-regulated inGSNOR−/− MSCs, presumably to maintain NO production atphysiologic levels. Unlike MSCs, endothelial cells express theNOS3 isoform (35) shown to participate in proangiogenic sig-naling (36). Additionally, NOS3 and GSNOR play an importantrole in endothelial cell–mediated postnatal angiogenesis andvascular tone (21, 22). Moreover, GSNOR−/− mice have aug-mented myocardial capillary density, at baseline, as shown byCD31 staining (22), suggesting that S-nitrosylation enhances an-giogenesis by endothelial cells in contrast to the inhibitory effect onMSCs. We observed a similar effect in murine and human MSCscompared with endothelial cells (HUVECs), suggesting that NOpromotes angiogenesis by endothelial cells and inhibits vasculo-genesis by MSC. Moreover, pathological conditions such as endo-thelial dysfunction where there is reduced NO bioavailability (37)may trigger vasculogenesis by MSC, perhaps serving as a compen-satory mechanism.In summary, our findings offer insight regarding the role of

    NO in vascular biology in which environments that are NO de-ficient trigger the participation of MSCs in vasculogenesis. Thus,NO exerts a balanced effect on the different cellular precursorsparticipating in neo-angiogenesis, promoting that portion me-diated by EPCs while simultaneously inhibiting that originatingwith MSC-like cells. These findings offer exciting insights intothe pathophysiology of conditions characterized by exuberantneo-vascularization such as cancer and diabetic retinopathy and,as such, have therapeutic implications.

    Materials and MethodsA detailed description of the materials and methods can be found in SIMaterials and Methods. Briefly, GSNOR−/− mice were generated as described(18) and compared with age- and sex-matched NOS2−/− and WT mice (C57BL/6). Bone marrow–derived MSCs isolated and expanded from WT, GSNOR−/−,and NOS2−/− mice and humans (hMSCs) were grown in endothelial growthmedia (EGM-2; Lonza) followed by 24 h in Matrigel, in the presence of ve-hicle, L-NAME [an NO synthase (NOS) inhibitor], GSNO, and SNAP (NOdonors), and JI-38 (20), a GHRH agonist. GHRH agonists, JI-38, MR-409, andMR-356 were synthesized in the laboratory of A.V.S. NO production and NOSexpression by MSCs was assessed. Additionally, we used an allograft assay tostudy in vivo vasculogenesis by murine MSCs. All animal protocols and ex-perimental procedures were approved by the University of Miami In-stitutional Animal Care and Use Committee.

    Data were analyzed for significance using one-way ANOVA, the Tukey-Kramer multiple comparisons test, and Student t test. All analyses wereperformed using GraphPad Prism, version 4.03, and P < 0.05 was consideredsignificant. All data were presented as mean ± SE.

    ACKNOWLEDGMENTS. We thank Irene Margitich, Lauro M. Takeuchi, andMirella Figueroa for technical assistance; Carmen Perez for preparing thehistologic sections; Shannon Opiela, Jay Enten, and James Phillips for FACSanalysis; and Dr. Norman Block for editorial suggestions. This work wasfunded by National Heart, Lung, and Blood Institute Grants R01 HL-094849,R01 HL084275, RO1 HL107110, and R01 HL110737 (to J.M.H.) and a W. H.Coulter Center Award [Medical Research Service of Veterans Affairs andDepartments of Pathology and Medicine, Division of Hematology/Oncology,University of Miami Miller School of Medicine and South Florida VeteransAffairs Foundation for Research and Education (to A.V.S.)].

    1. Asahara T, et al. (1997) Isolation of putative progenitor endothelial cells for angio-

    genesis. Science 275(5302):964–967.2. Murohara T, et al. (1998) Nitric oxide synthase modulates angiogenesis in response to

    tissue ischemia. J Clin Invest 101(11):2567–2578.

    3. Aicher A, et al. (2009) cGMP-dependent protein kinase I is crucial for angiogenesis and

    postnatal vasculogenesis. PLoS ONE 4(3):e4879.4. Aicher A, et al. (2003) Essential role of endothelial nitric oxide synthase for mobili-

    zation of stem and progenitor cells. Nat Med 9(11):1370–1376.

    Fig. 5. Exposure of human MSCs to NO donors impairs the formation ofcapillary tube-like structures (A and B). Quantification of the (A) number and(B) length (μm) of tube-like structures following 6-h exposure to no treat-ment, vehicle, L-NAME, GSNO, or SNAP (n ≥ 3, *P < 0.05 vs. hMSCS ± vehicle).

    2838 | www.pnas.org/cgi/doi/10.1073/pnas.1220185110 Gomes et al.

    Dow

    nloa

    ded

    by g

    uest

    on

    July

    3, 2

    021

    http://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1220185110/-/DCSupplemental/pnas.201220185SI.pdf?targetid=nameddest=STXThttp://www.pnas.org/lookup/suppl/doi:10.1073/pnas.1220185110/-/DCSupplemental/pnas.201220185SI.pdf?targetid=nameddest=STXTwww.pnas.org/cgi/doi/10.1073/pnas.1220185110

  • 5. Asahara T, et al. (1999) Bone marrow origin of endothelial progenitor cells responsiblefor postnatal vasculogenesis in physiological and pathological neovascularization. CircRes 85(3):221–228.

    6. Ferrara N, Gerber HP, LeCouter J (2003) The biology of VEGF and its receptors. NatMed 9(6):669–676.

    7. Coultas L, Chawengsaksophak K, Rossant J (2005) Endothelial cells and VEGF in vas-cular development. Nature 438(7070):937–945.

    8. Nagaya N, et al. (2004) Intravenous administration of mesenchymal stem cells im-proves cardiac function in rats with acute myocardial infarction through angiogenesisand myogenesis. Am J Physiol Heart Circ Physiol 287(6):H2670–H2676.

    9. Bautch VL (2011) Stem cells and the vasculature. Nat Med 17(11):1437–1443.10. Caplan AI (2008) All MSCs are pericytes? Cell Stem Cell 3(3):229–230.11. Chen J, et al. (2003) Intravenous administration of human bone marrow stromal cells

    induces angiogenesis in the ischemic boundary zone after stroke in rats. Circ Res 92(6):692–699.

    12. Quevedo HC, et al. (2009) Allogeneic mesenchymal stem cells restore cardiac functionin chronic ischemic cardiomyopathy via trilineage differentiating capacity. Proc NatlAcad Sci USA 106(33):14022–14027.

    13. Williams AR, et al. (2011) Intramyocardial stem cell injection in patients with ischemiccardiomyopathy: Functional recovery and reverse remodeling. Circ Res 108(7):792–796.

    14. Oswald J, et al. (2004) Mesenchymal stem cells can be differentiated into endothelialcells in vitro. Stem Cells 22(3):377–384.

    15. Sun B, et al. (2005) Correlation between melanoma angiogenesis and the mesen-chymal stem cells and endothelial progenitor cells derived from bone marrow. StemCells Dev 14(3):292–298.

    16. Ball SG, Shuttleworth CA, Kielty CM (2007) Vascular endothelial growth factor cansignal through platelet-derived growth factor receptors. J Cell Biol 177(3):489–500.

    17. Liu L, et al. (2001) A metabolic enzyme for S-nitrosothiol conserved from bacteria tohumans. Nature 410(6827):490–494.

    18. Liu L, et al. (2004) Essential roles of S-nitrosothiols in vascular homeostasis and en-dotoxic shock. Cell 116(4):617–628.

    19. Beitz JG, Kim IS, Calabresi P, Frackelton AR, Jr. (1991) Human microvascular endo-thelial cells express receptors for platelet-derived growth factor. Proc Natl Acad SciUSA 88(5):2021–2025.

    20. Izdebski J, et al. (1995) Synthesis and biological evaluation of superactive agonists ofgrowth hormone-releasing hormone. Proc Natl Acad Sci USA 92(11):4872–4876.

    21. Beigi F, et al. (2012) Dynamic denitrosylation via S-nitrosoglutathione reductaseregulates cardiovascular function. Proc Natl Acad Sci USA 109(11):4314–4319.

    22. Lima B, et al. (2009) Endogenous S-nitrosothiols protect against myocardial injury.Proc Natl Acad Sci USA 106(15):6297–6302.

    23. Anand P, Stamler JS (2012) Enzymatic mechanisms regulating protein S-nitrosylation:implications in health and disease. J Mol Med (Berl) 90(3):233–244.

    24. Ball SG, Shuttleworth CA, Kielty CM (2007) Mesenchymal stem cells and neo-vascularization: Role of platelet-derived growth factor receptors. J Cell Mol Med 11(5):1012–1030.

    25. Ball SG, Shuttleworth CA, Kielty CM (2010) Platelet-derived growth factor receptorsregulate mesenchymal stem cell fate: Implications for neovascularization. Expert OpinBiol Ther 10(1):57–71.

    26. Wu X, et al. (2005) Mesenchymal stem cells participating in ex vivo endothelium re-pair and its effect on vascular smooth muscle cells growth. Int J Cardiol 105(3):274–282.

    27. Hall AP (2006) Review of the pericyte during angiogenesis and its role in cancer anddiabetic retinopathy. Toxicol Pathol 34(6):763–775.

    28. Kanashiro-Takeuchi RM, et al. (2012) Activation of growth hormone releasing hor-mone (GHRH) receptor stimulates cardiac reverse remodeling after myocardial in-farction (MI). Proc Natl Acad Sci USA 109(2):559–563.

    29. Kanashiro-Takeuchi RM, et al. (2010) Cardioprotective effects of growth hormone-releasing hormone agonist after myocardial infarction. Proc Natl Acad Sci USA 107(6):2604–2609.

    30. Dioufa N, et al. (2010) Acceleration of wound healing by growth hormone-releasinghormone and its agonists. Proc Natl Acad Sci USA 107(43):18611–18615.

    31. Ludwig B, et al. (2010) Agonist of growth hormone-releasing hormone as a potentialeffector for survival and proliferation of pancreatic islets. Proc Natl Acad Sci USA 107(28):12623–12628.

    32. Wang Y, et al. (2008) Nitric oxide suppresses the secretion of vascular endothelialgrowth factor and hepatocyte growth factor from human mesenchymal stem cells.Shock 30(5):527–531.

    33. Cipriani P, et al. (2007) Impairment of endothelial cell differentiation from bonemarrow-derived mesenchymal stem cells: New insight into the pathogenesis of sys-temic sclerosis. Arthritis Rheum 56(6):1994–2004.

    34. Hare JM (2004) Nitroso-redox balance in the cardiovascular system. N Engl J Med 351(20):2112–2114.

    35. Kuhlencordt PJ, et al. (2004) Role of endothelial nitric oxide synthase in endothelialactivation: Insights from eNOS knockout endothelial cells. Am J Physiol Cell Physiol286(5):C1195–C1202.

    36. Huang PL (2003) Endothelial nitric oxide synthase and endothelial dysfunction. CurrHypertens Rep 5(6):473–480.

    37. Gupta TK, Toruner M, Chung MK, Groszmann RJ (1998) Endothelial dysfunction anddecreased production of nitric oxide in the intrahepatic microcirculation ofcirrhotic rats. Hepatology 28(4):926–931.

    Gomes et al. PNAS | February 19, 2013 | vol. 110 | no. 8 | 2839

    CELL

    BIOLO

    GY

    SEECO

    MMEN

    TARY

    Dow

    nloa

    ded

    by g

    uest

    on

    July

    3, 2

    021