Corneal Mesenchymal Stromal Cells Are Directly Antiangiogenic via PEDF and sFLT-1

Medi Eslani, Ilham Putra, Xiang Shen, Judy Hamouie, Neda Afsharkhamseh, Soroush Besharat, Mark I Rosenblatt, Reza Dana, Peiman Hematti, Ali R Djalilian, Medi Eslani, Ilham Putra, Xiang Shen, Judy Hamouie, Neda Afsharkhamseh, Soroush Besharat, Mark I Rosenblatt, Reza Dana, Peiman Hematti, Ali R Djalilian

Abstract

Purpose: To evaluate the angiogenic properties of corneal derived mesenchymal stromal cells (Co-MSC).

Methods: Co-MSCs were extracted from human cadaver, and wild-type (C57BL/6J) and SERPINF1-/- mice corneas. The MSC secretome was collected in a serum-free medium. Human umbilical vein endothelial cell (HUVEC) tube formation and fibrin gel bead assay (FIBA) sprout formation were used to assess the angiogenic properties of Co-MSC secretome. Complete corneal epithelial debridement was used to induce corneal neovascularization in wild-type mice. Co-MSCs embedded in fibrin gel was applied over the debrided cornea to evaluate the angiogenic effects of Co-MSCs in vivo. Immunoprecipitation was used to remove soluble fms-like tyrosine kinase-1 (sFLT-1) and pigment epithelium-derived factor (PEDF, SERPINF1 gene) from the Co-MSC secretome.

Results: Co-MSC secretome significantly inhibited HUVECs tube and sprout formation. Co-MSCs from different donors consistently contained high levels of antiangiogenic factors including sFLT-1 and PEDF; and low levels of the angiogenic factor VEGF-A. In vivo, application of Co-MSCs to mouse corneas after injury prevented the development of corneal neovascularization. Removing PEDF or sFLT-1 from the secretome significantly diminished the antiangiogenic effects of Co-MSCs. Co-MSCs isolated from SERPINF1-/- mice had significantly reduced antiangiogenic effects compared to SERPINF1+/+ (wild-type) Co-MSCs.

Conclusions: These results illustrate the direct antiangiogenic properties of Co-MSCs, the importance of sFLT-1 and PEDF, and their potential clinical application for preventing pathologic corneal neovascularization.

Figures

Figure 1
Figure 1
Human and mouse corneal stromal cells demonstrate mesenchymal stromal cells (MSC) features. (A) Bright-field image of passage-4 human corneal MSCs. (B) Flow cytometry analysis demonstrated a homogenous MSC population. More than 95% of the cells were positive for cell surface markers CD73, CD90, CD105, and negative for CD19, CD45, HLA-DR, CD11b, and CD34 (n = 10). (C) Differentiation into the three mesenchymal lineages: I: Osteogenesis: calcium deposition stained with Alizarin Red; II: Adipogenesis: lipid formation stained with LipidTOX; III: Chondrogenesis: Glycosaminoglycans stained with Alcian Blue. (D) Bright-field microscopy image of passage-4 mouse corneal MSCs. (E) Flow cytometry analysis demonstrated a homogenous MSC population. More than 95% of the cells are positive for cell surface markers CD29, Sca-1, CD105, CD44 and CD106 and negative for CD11b, and CD45. (F) Differentiation into the three mesenchymal lineages: I: Calcium deposition stained with Alizarin Red; II: Lipid formation stained with LipidTOX; III: Glycosaminoglycans stained with Alcian Blue.
Figure 2
Figure 2
Corneal MSCs (Co-MSCs) are directly antiangiogenic in vitro. (A) Co-MSC secretome significantly inhibited mean sprout count per bead and mean sprout length in a fibrin-induced bead assay (n = 5). *P S, secretome.
Figure 3
Figure 3
Co-MSCs have an antiangiogenic profile by protein expression. (A) Human angiogenesis array was used to profile pro- and antiangiogenesis mediators in the Co-MSC secretome. It revealed it has high levels of antiangiogenic factors, whereas low levels of proangiogenic factors. (B) The secretome from Co-MSCs contains high levels of sFLT-1 (1875 ± 677 pg/mL); PEDF (4829 ± 2342 pg/mL); TSG-6 (643.3 ± 149.1 pg/mL); and low amount of VEGF-A (106.9 ±103.3 pg/mL; n = 5). The values shown are mean ± SD (error bars).
Figure 4
Figure 4
Co-MSCs inhibit pathologic corneal neovascularization in vivo. (A) Schematic illustration shows the steps for debriding the epithelium, MSC delivery using fibrin gel and temporarily closing the lids. (B) In vivo microscopy confirmed the presence of fluorescent-labeled MSCs in the cornea three days after the procedure. Scale bars: 200 μm. (C) The slit lamp photos and CD31 staining demonstrated both Co-MSCs significantly prevented corneal neovascularization compared to fibrin gel alone 14 days after injury. The values shown are mean ± SD (error bars). *P

Figure 5

Direct antiangiogenic properties of Co-MSCs…

Figure 5

Direct antiangiogenic properties of Co-MSCs depend on PEDF and sFLT-1. (A) Removing sFLT-1…

Figure 5
Direct antiangiogenic properties of Co-MSCs depend on PEDF and sFLT-1. (A) Removing sFLT-1 and PEDF with neutralizing antibody capturing and immunoprecipitation abrogated the antiangiogenic property of Co-MSC secretome in a HUVEC assay (n = 5 1-way ANOVA and then 2-sided t-test were used to compare the groups). *P

Figure 6

Direct antiangiogenic properties of Co-MSCs…

Figure 6

Direct antiangiogenic properties of Co-MSCs are in part due to PEDF. Knocking down…

Figure 6
Direct antiangiogenic properties of Co-MSCs are in part due to PEDF. Knocking down PEDF in Co-MSCs by siRNA obviates the antiangiogenic effects of their secretome and increases tubule formation in a HUVEC assay (n = 4, 1-way ANOVA, and then 2-sided t-test were used to compare the groups). *P

Figure 7

PEDF plays a key role…

Figure 7

PEDF plays a key role on the antiangiogenic properties of Co-MSCs in vivo.…

Figure 7
PEDF plays a key role on the antiangiogenic properties of Co-MSCs in vivo. (A) Western blot shows Co-MSCs isolated from SERPINF1−/− mice do not express PEDF. (B) Co-MSCs isolated from SERPINF1−/− mice had lower antiangiogenic effects, compared to wild-type (SERPINF1+/+) Co-MSCs, when placed in fibrin gels after corneal epithelial injury in wild-type mice (same model as in Fig. 4A; n = 5, 2-sided t-test: *P = 0.0001). Scale bars: 500 μm. The values shown are mean ± SD (error bars). Co-mMSCSF1, corneal mouse MSCSERPINF1; NV, neovascularization.

Figure 8

Schematic illustration summarizes the mechanisms…

Figure 8

Schematic illustration summarizes the mechanisms by which Co-MSCs confer their antiangiogenic properties. (A)…

Figure 8
Schematic illustration summarizes the mechanisms by which Co-MSCs confer their antiangiogenic properties. (A) After corneal epithelial injury, DAMPS, chemokines, and cytokines released by the injured and resident cells directly induce angiogenesis while also recruiting inflammatory cells that further promote angiogenesis. The net effect is shifting the antiangiogenic balance of the cornea toward angiogenesis. (B) Co-MSCs applied to the cornea after injury modulate neovascularization directly through the secretion of antiangiogenic factors including PEDF and sFLT-1 and indirectly by suppressing inflammation via TSG6 and other anti-inflammatory factors. DAMP, damage-associated molecular pattern.
All figures (8)
Similar articles
References
    1. Basu S, Hertsenberg AJ, Funderburgh ML,et al. . Human limbal biopsy-derived stromal stem cells prevent corneal scarring. Sci Transl Med. 2014; 6: 266ra172. - PMC - PubMed
    1. Mittal SK, Omoto M, Amouzegar A,et al. . Restoration of corneal transparency by mesenchymal stem cells. Stem cell reports. 2016; 7: 583– 590. - PMC - PubMed
    1. Amouzegar A, Mittal SK, Sahu A, Sahu SK, Chauhan SK. . Mesenchymal stem cells modulate differentiation of myeloid progenitor cells during inflammation. Stem cells. 2017; 35: 1532– 1541. - PMC - PubMed
    1. Prockop DJ. . Repair of tissues by adult stem/progenitor cells (MSCs): controversies, myths, and changing paradigms. Mol Ther. 2009; 17: 939– 946. - PMC - PubMed
    1. Premer C, Blum A, Bellio MA,et al. . Allogeneic mesenchymal stem cells restore endothelial function in heart failure by stimulating endothelial progenitor cells. EBioMedicine. 2015; 2: 467– 475. - PMC - PubMed
Show all 76 references
MeSH terms
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM

NCBI Literature Resources

MeSH PMC Bookshelf Disclaimer

The PubMed wordmark and PubMed logo are registered trademarks of the U.S. Department of Health and Human Services (HHS). Unauthorized use of these marks is strictly prohibited.

Follow NCBI
Figure 5
Figure 5
Direct antiangiogenic properties of Co-MSCs depend on PEDF and sFLT-1. (A) Removing sFLT-1 and PEDF with neutralizing antibody capturing and immunoprecipitation abrogated the antiangiogenic property of Co-MSC secretome in a HUVEC assay (n = 5 1-way ANOVA and then 2-sided t-test were used to compare the groups). *P

Figure 6

Direct antiangiogenic properties of Co-MSCs…

Figure 6

Direct antiangiogenic properties of Co-MSCs are in part due to PEDF. Knocking down…

Figure 6
Direct antiangiogenic properties of Co-MSCs are in part due to PEDF. Knocking down PEDF in Co-MSCs by siRNA obviates the antiangiogenic effects of their secretome and increases tubule formation in a HUVEC assay (n = 4, 1-way ANOVA, and then 2-sided t-test were used to compare the groups). *P

Figure 7

PEDF plays a key role…

Figure 7

PEDF plays a key role on the antiangiogenic properties of Co-MSCs in vivo.…

Figure 7
PEDF plays a key role on the antiangiogenic properties of Co-MSCs in vivo. (A) Western blot shows Co-MSCs isolated from SERPINF1−/− mice do not express PEDF. (B) Co-MSCs isolated from SERPINF1−/− mice had lower antiangiogenic effects, compared to wild-type (SERPINF1+/+) Co-MSCs, when placed in fibrin gels after corneal epithelial injury in wild-type mice (same model as in Fig. 4A; n = 5, 2-sided t-test: *P = 0.0001). Scale bars: 500 μm. The values shown are mean ± SD (error bars). Co-mMSCSF1, corneal mouse MSCSERPINF1; NV, neovascularization.

Figure 8

Schematic illustration summarizes the mechanisms…

Figure 8

Schematic illustration summarizes the mechanisms by which Co-MSCs confer their antiangiogenic properties. (A)…

Figure 8
Schematic illustration summarizes the mechanisms by which Co-MSCs confer their antiangiogenic properties. (A) After corneal epithelial injury, DAMPS, chemokines, and cytokines released by the injured and resident cells directly induce angiogenesis while also recruiting inflammatory cells that further promote angiogenesis. The net effect is shifting the antiangiogenic balance of the cornea toward angiogenesis. (B) Co-MSCs applied to the cornea after injury modulate neovascularization directly through the secretion of antiangiogenic factors including PEDF and sFLT-1 and indirectly by suppressing inflammation via TSG6 and other anti-inflammatory factors. DAMP, damage-associated molecular pattern.
All figures (8)
Similar articles
References
    1. Basu S, Hertsenberg AJ, Funderburgh ML,et al. . Human limbal biopsy-derived stromal stem cells prevent corneal scarring. Sci Transl Med. 2014; 6: 266ra172. - PMC - PubMed
    1. Mittal SK, Omoto M, Amouzegar A,et al. . Restoration of corneal transparency by mesenchymal stem cells. Stem cell reports. 2016; 7: 583– 590. - PMC - PubMed
    1. Amouzegar A, Mittal SK, Sahu A, Sahu SK, Chauhan SK. . Mesenchymal stem cells modulate differentiation of myeloid progenitor cells during inflammation. Stem cells. 2017; 35: 1532– 1541. - PMC - PubMed
    1. Prockop DJ. . Repair of tissues by adult stem/progenitor cells (MSCs): controversies, myths, and changing paradigms. Mol Ther. 2009; 17: 939– 946. - PMC - PubMed
    1. Premer C, Blum A, Bellio MA,et al. . Allogeneic mesenchymal stem cells restore endothelial function in heart failure by stimulating endothelial progenitor cells. EBioMedicine. 2015; 2: 467– 475. - PMC - PubMed
Show all 76 references
MeSH terms
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM
Figure 6
Figure 6
Direct antiangiogenic properties of Co-MSCs are in part due to PEDF. Knocking down PEDF in Co-MSCs by siRNA obviates the antiangiogenic effects of their secretome and increases tubule formation in a HUVEC assay (n = 4, 1-way ANOVA, and then 2-sided t-test were used to compare the groups). *P

Figure 7

PEDF plays a key role…

Figure 7

PEDF plays a key role on the antiangiogenic properties of Co-MSCs in vivo.…

Figure 7
PEDF plays a key role on the antiangiogenic properties of Co-MSCs in vivo. (A) Western blot shows Co-MSCs isolated from SERPINF1−/− mice do not express PEDF. (B) Co-MSCs isolated from SERPINF1−/− mice had lower antiangiogenic effects, compared to wild-type (SERPINF1+/+) Co-MSCs, when placed in fibrin gels after corneal epithelial injury in wild-type mice (same model as in Fig. 4A; n = 5, 2-sided t-test: *P = 0.0001). Scale bars: 500 μm. The values shown are mean ± SD (error bars). Co-mMSCSF1, corneal mouse MSCSERPINF1; NV, neovascularization.

Figure 8

Schematic illustration summarizes the mechanisms…

Figure 8

Schematic illustration summarizes the mechanisms by which Co-MSCs confer their antiangiogenic properties. (A)…

Figure 8
Schematic illustration summarizes the mechanisms by which Co-MSCs confer their antiangiogenic properties. (A) After corneal epithelial injury, DAMPS, chemokines, and cytokines released by the injured and resident cells directly induce angiogenesis while also recruiting inflammatory cells that further promote angiogenesis. The net effect is shifting the antiangiogenic balance of the cornea toward angiogenesis. (B) Co-MSCs applied to the cornea after injury modulate neovascularization directly through the secretion of antiangiogenic factors including PEDF and sFLT-1 and indirectly by suppressing inflammation via TSG6 and other anti-inflammatory factors. DAMP, damage-associated molecular pattern.
All figures (8)
Figure 7
Figure 7
PEDF plays a key role on the antiangiogenic properties of Co-MSCs in vivo. (A) Western blot shows Co-MSCs isolated from SERPINF1−/− mice do not express PEDF. (B) Co-MSCs isolated from SERPINF1−/− mice had lower antiangiogenic effects, compared to wild-type (SERPINF1+/+) Co-MSCs, when placed in fibrin gels after corneal epithelial injury in wild-type mice (same model as in Fig. 4A; n = 5, 2-sided t-test: *P = 0.0001). Scale bars: 500 μm. The values shown are mean ± SD (error bars). Co-mMSCSF1, corneal mouse MSCSERPINF1; NV, neovascularization.
Figure 8
Figure 8
Schematic illustration summarizes the mechanisms by which Co-MSCs confer their antiangiogenic properties. (A) After corneal epithelial injury, DAMPS, chemokines, and cytokines released by the injured and resident cells directly induce angiogenesis while also recruiting inflammatory cells that further promote angiogenesis. The net effect is shifting the antiangiogenic balance of the cornea toward angiogenesis. (B) Co-MSCs applied to the cornea after injury modulate neovascularization directly through the secretion of antiangiogenic factors including PEDF and sFLT-1 and indirectly by suppressing inflammation via TSG6 and other anti-inflammatory factors. DAMP, damage-associated molecular pattern.

References

    1. Basu S, Hertsenberg AJ, Funderburgh ML,et al. . Human limbal biopsy-derived stromal stem cells prevent corneal scarring. Sci Transl Med. 2014; 6: 266ra172.
    1. Mittal SK, Omoto M, Amouzegar A,et al. . Restoration of corneal transparency by mesenchymal stem cells. Stem cell reports. 2016; 7: 583– 590.
    1. Amouzegar A, Mittal SK, Sahu A, Sahu SK, Chauhan SK. . Mesenchymal stem cells modulate differentiation of myeloid progenitor cells during inflammation. Stem cells. 2017; 35: 1532– 1541.
    1. Prockop DJ. . Repair of tissues by adult stem/progenitor cells (MSCs): controversies, myths, and changing paradigms. Mol Ther. 2009; 17: 939– 946.
    1. Premer C, Blum A, Bellio MA,et al. . Allogeneic mesenchymal stem cells restore endothelial function in heart failure by stimulating endothelial progenitor cells. EBioMedicine. 2015; 2: 467– 475.
    1. Prockop DJ. . The exciting prospects of new therapies with mesenchymal stromal cells. Cytotherapy. 2017; 19: 1– 8.
    1. Hatzistergos KE, Hare JM. . Cell therapy: targeting endogenous repair versus remuscularization. Circulation Res. 2015; 117: 659– 661.
    1. Maria AT, Toupet K, Maumus M,et al. . Human adipose mesenchymal stem cells as potent anti-fibrosis therapy for systemic sclerosis. J Autoimmunity. 2016; 70: 31– 39.
    1. Li C, Zhen G, Chai Y,et al. . RhoA determines lineage fate of mesenchymal stem cells by modulating CTGF-VEGF complex in extracellular matrix. Nat Commun. 2016; 7: 11455.
    1. Perez VL, Caspi RR. . Immune mechanisms in inflammatory and degenerative eye disease. Trends in immunology. 2015; 36: 354– 363.
    1. Ambati BK, Nozaki M, Singh N,et al. . Corneal avascularity is due to soluble VEGF receptor-1. Nature. 2006; 443: 993– 997.
    1. Cursiefen C, Chen L, Saint-Geniez M,et al. . Nonvascular VEGF receptor 3 expression by corneal epithelium maintains avascularity and vision. Proc Natl Acad Sci U S A. 2006; 103: 11405– 11410.
    1. Clements JL, Dana R. . Inflammatory corneal neovascularization: etiopathogenesis. Semin Ophthalmol. 2011; 26: 235– 245.
    1. Byun YS, Tibrewal S, Kim E,et al. . Keratocytes derived from spheroid culture of corneal stromal cells resemble tissue resident keratocytes. PLoS One. 2014; 9: e112781.
    1. Polisetty N, Fatima A, Madhira SL, Sangwan VS, Vemuganti GK. . Mesenchymal cells from limbal stroma of human eye. Mol Vis. 2008; 14: 431– 442.
    1. Afsharkhamseh N, Movahedan A, Gidfar S,et al. . Stability of limbal stem cell deficiency after mechanical and thermal injuries in mice. Exp Eye Res. 2016; 145: 88– 92.
    1. Nakatsu MN, Davis J, Hughes CC. . Optimized fibrin gel bead assay for the study of angiogenesis. J Vis Exp. 2007; 186.
    1. Cao R, Lim S, Ji H,et al. . Mouse corneal lymphangiogenesis model. Nat Protoc. 2011; 6: 817– 826.
    1. Rabiolo A, Bignami F, Rama P, Ferrari G. . VesselJ: a new tool for semiautomatic measurement of corneal neovascularization. Invest Ophthalmol. Vis Sci 2015; 56: 8199– 8206.
    1. Uccelli A, de Rosbo NK. . The immunomodulatory function of mesenchymal stem cells: mode of action and pathways. Ann N Y Acad Sci. 2015; 1351: 114– 126.
    1. Coulson-Thomas VJ, Coulson-Thomas YM, Gesteira TF, Kao WW. . Extrinsic and intrinsic mechanisms by which mesenchymal stem cells suppress the immune system. Ocul Surf. 2016; 14: 121– 134.
    1. Wietecha MS, Krol MJ, Michalczyk ER, Chen L, Gettins PG, DiPietro LA. . Pigment epithelium-derived factor as a multifunctional regulator of wound healing. Am J Physiol Heart Circ Physiol. 2015; 309: H812– H826.
    1. Zamiri P, Masli S, Streilein JW, Taylor AW. . Pigment epithelial growth factor suppresses inflammation by modulating macrophage activation. Invest Ophthalmol Vis Sci. 2006; 47: 3912– 3918.
    1. Kao WW, Coulson-Thomas VJ. . Cell therapy of corneal diseases. Cornea. 2016; 35 suppl 1: S9– S19.
    1. Forrester JV, Steptoe RJ, Klaska IP,et al. . Cell-based therapies for ocular inflammation. Prog Retin Eye Res. 2013; 35: 82– 101.
    1. Choi H, Lee RH, Bazhanov N, Oh JY, Prockop DJ. . Anti-inflammatory protein TSG-6 secreted by activated MSCs attenuates zymosan-induced mouse peritonitis by decreasing TLR2/NF-kappaB signaling in resident macrophages. Blood. 2011; 118: 330– 338.
    1. Stagg J, Galipeau J. . Mechanisms of immune modulation by mesenchymal stromal cells and clinical translation. Curr Mol Med. 2013; 13: 856– 867.
    1. Ko JH, Lee HJ, Jeong HJ,et al. . Mesenchymal stem/stromal cells precondition lung monocytes/macrophages to produce tolerance against allo- and autoimmunity in the eye. Proc Natl Acad Sci U S A. 2016; 113: 158– 163.
    1. Espagnolle N, Balguerie A, Arnaud E, Sensebe L, Varin A. . CD54-Mediated Interaction with Pro-inflammatory Macrophages Increases the Immunosuppressive Function of Human Mesenchymal Stromal Cells. Stem cell reports. 2017; 8: 961– 976.
    1. Kellner J, Sivajothi S, McNiece I. . Differential properties of human stromal cells from bone marrow, adipose, liver and cardiac tissues. Cytotherapy. 2015; 17: 1514– 1523.
    1. Klimczak A, Kozlowska U. . Mesenchymal stromal cells and tissue-specific progenitor cells: their role in tissue homeostasis. Stem Cells Int. 2016; 2016: 4285215.
    1. Bortolotti F, Ukovich L, Razban V,et al. . In vivo therapeutic potential of mesenchymal stromal cells depends on the source and the isolation procedure. Stem Cell reports. 2015; 4: 332– 339.
    1. Sun K, Zhou Z, Ju X,et al. . Combined transplantation of mesenchymal stem cells and endothelial progenitor cells for tissue engineering: a systematic review and meta-analysis. Stem Cell Res Ther. 2016; 7: 151.
    1. Tao H, Han Z, Han ZC, Li Z. . Proangiogenic features of mesenchymal stem cells and their therapeutic applications. Stem Cells Int. 2016; 2016: 1314709.
    1. Bronckaers A, Hilkens P, Martens W,et al. . Mesenchymal stem/stromal cells as a pharmacological and therapeutic approach to accelerate angiogenesis. Pharmacol Ther. 2014; 143: 181– 196.
    1. Watt SM, Gullo F, van der Garde M,et al. . The angiogenic properties of mesenchymal stem/stromal cells and their therapeutic potential. Br Med Bull. 2013; 108: 25– 53.
    1. Oh JY, Kim MK, Shin MS,et al. . The anti-inflammatory and anti-angiogenic role of mesenchymal stem cells in corneal wound healing following chemical injury. Stem Cells. 2008; 26: 1047– 1055.
    1. Oh JY, Roddy GW, Choi H,et al. . Anti-inflammatory protein TSG-6 reduces inflammatory damage to the cornea following chemical and mechanical injury. Proc Natl Acad Sci U S A. 2010; 107: 16875– 16880.
    1. Clarke MR, Imhoff FM, Baird SK. . Mesenchymal stem cells inhibit breast cancer cell migration and invasion through secretion of tissue inhibitor of metalloproteinase-1 and -2. Molecular carcinogenesis. 2015; 54: 1214– 1219.
    1. Daltro PS, Barreto BC, Silva PG,et al. . Therapy with mesenchymal stromal cells or conditioned medium reverse cardiac alterations in a high-fat diet-induced obesity model. Cytotherapy. 2017; 19: 1176– 1188.
    1. Efimenko A, Dzhoyashvili N, Kalinina N,et al. . Adipose-derived mesenchymal stromal cells from aged patients with coronary artery disease keep mesenchymal stromal cell properties but exhibit characteristics of aging and have impaired angiogenic potential. Stem Cells Transl Med. 2014; 3: 32– 41.
    1. Li F, Armstrong GB, Tombran-Tink J, Niyibizi C. . Pigment epithelium derived factor upregulates expression of vascular endothelial growth factor by human mesenchymal stem cells: Possible role in PEDF regulated matrix mineralization. Biochem Biophys Res Commun. 2016; 478: 1106– 1110.
    1. Oh JY, Kim MK, Shin MS, Wee WR, Lee JH. . Cytokine secretion by human mesenchymal stem cells cocultured with damaged corneal epithelial cells. Cytokine. 2009; 46: 100– 103.
    1. Ostanin AA, Petrovskii YL, Shevela EY, Chernykh ER. . Multiplex analysis of cytokines, chemokines, growth factors, MMP-9 and TIMP-1 produced by human bone marrow, adipose tissue, and placental mesenchymal stromal cells. Bull Exp Biol Med. 2011; 151: 133– 141.
    1. Zolochevska O, Shearer J, Ellis J,et al. . Human adipose-derived mesenchymal stromal cell pigment epithelium-derived factor cytotherapy modifies genetic and epigenetic profiles of prostate cancer cells. Cytotherapy. 2014; 16: 346– 356.
    1. Cappuzzello C, Doni A, Dander E,et al. . Mesenchymal stromal cell-derived PTX3 promotes wound healing via fibrin remodeling. Invest Dermatol. 2016; 136: 293– 300.
    1. Park HW, Moon HE, Kim HS,et al. . Human umbilical cord blood-derived mesenchymal stem cells improve functional recovery through thrombospondin1, pantraxin3, and vascular endothelial growth factor in the ischemic rat brain. Neurosci Res. 2015; 93: 1814– 1825.
    1. Kendall RL, Thomas KA. . Inhibition of vascular endothelial cell growth factor activity by an endogenously encoded soluble receptor. Proc Natl Acad Sci U S A. 1993; 90: 10705– 10709.
    1. Cai J, Jiang WG, Grant MB, Boulton M. . Pigment epithelium-derived factor inhibits angiogenesis via regulated intracellular proteolysis of vascular endothelial growth factor receptor 1. J Biol Chem. 2006; 281: 3604– 3613.
    1. Ambati BK, Patterson E, Jani P,et al. . Soluble vascular endothelial growth factor receptor-1 contributes to the corneal antiangiogenic barrier. Br J Ophthalmol. 2007; 91: 505– 508.
    1. Wu FT, Stefanini MO, Mac Gabhann F, Kontos CD, Annex BH, Popel AS. . A systems biology perspective on sVEGFR1: its biological function, pathogenic role and therapeutic use. J Cell Mol Med. 2010; 14: 528– 552.
    1. Ahmad S, Ahmed A. . Elevated placental soluble vascular endothelial growth factor receptor-1 inhibits angiogenesis in preeclampsia. Circ Res. 2004; 95: 884– 891.
    1. Kendall RL, Wang G, Thomas KA. . Identification of a natural soluble form of the vascular endothelial growth factor receptor, FLT-1, and its heterodimerization with KDR. Biochemical Biophysical Res Communications 1996; 226: 324– 328.
    1. Kommareddy S, Amiji M. . Antiangiogenic gene therapy with systemically administered sFlt-1 plasmid DNA in engineered gelatin-based nanovectors. Cancer Gene Ther. 2007; 14: 488– 498.
    1. Olsson AK, Dimberg A, Kreuger J, Claesson-Welsh L. . VEGF receptor signalling - in control of vascular function. Nat Rev Mol Cell Biol. 2006; 7: 359– 371.
    1. Kumai Y, Ooboshi H, Ibayashi S,et al. . Postischemic gene transfer of soluble Flt-1 protects against brain ischemia with marked attenuation of blood-brain barrier permeability. J Cereb Blood Flow Metab. 2007; 27: 1152– 1160.
    1. Tsao PN, Chan FT, Wei SC,et al. . Soluble vascular endothelial growth factor receptor-1 protects mice in sepsis. Crit Care Med. 2007; 35: 1955– 1960.
    1. Lai CM, Brankov M, Zaknich T,et al. . Inhibition of angiogenesis by adenovirus-mediated sFlt-1 expression in a rat model of corneal neovascularization. Human Gene Ther. 2001; 12: 1299– 1310.
    1. Cho YK, Uehara H, Young JR, Archer B, Zhang X, Ambati BK. . Vascular endothelial growth factor receptor 1 morpholino decreases angiogenesis in a murine corneal suture model. Invest Ophthalmol Vis Sci. 2012; 53: 685– 692.
    1. Kommineni VK, Nagineni CN, William A, Detrick B, Hooks JJ. . IFN-gamma acts as anti-angiogenic cytokine in the human cornea by regulating the expression of VEGF-A and sVEGF-R1. Biochem Biophys Res Commun. 2008; 374: 479– 484.
    1. Lai CM, Shen WY, Brankov M,et al. . Long-term evaluation of AAV-mediated sFlt-1 gene therapy for ocular neovascularization in mice and monkeys. Mol Ther. 2005; 12: 659– 668.
    1. Fitchev P, Chung C, Plunkett BA, Brendler CB, Crawford SE. . PEDF & stem cells: niche vs. nurture. Curr Drug Deliv. 2014; 11: 552– 560.
    1. Filleur S, Nelius T, de Riese W, Kennedy RC. . Characterization of PEDF: a multi-functional serpin family protein. J Cell Biochem. 2009; 106: 769– 775.
    1. Ek ET, Dass CR, Choong PF. . PEDF: a potential molecular therapeutic target with multiple anti-cancer activities. Trends Mol Med. 2006; 12: 497– 502.
    1. Cai J, Wu L, Qi X,et al. . PEDF regulates vascular permeability by a gamma-secretase-mediated pathway. PLoS One. 2011; 6: e21164.
    1. Zhang SX, Wang JJ, Gao G, Parke K, Ma JX. . Pigment epithelium-derived factor downregulates vascular endothelial growth factor (VEGF) expression and inhibits VEGF-VEGF receptor 2 binding in diabetic retinopathy. J Mol Endocrinol. 2006; 37: 1– 12.
    1. Matsui T, Nishino Y, Maeda S, Yamagishi S. . PEDF-derived peptide inhibits corneal angiogenesis by suppressing VEGF expression. Microvasc Res. 2012; 84: 105– 108.
    1. Ferrari G, Hajrasouliha AR, Sadrai Z, Ueno H, Chauhan SK, Dana R. . Nerves and neovessels inhibit each other in the cornea. Invest Ophthalmol Vis Sci. 2013; 54: 813– 820.
    1. Karakousis PC, John SK, Behling KC,et al. . Localization of pigment epithelium derived factor (PEDF) in developing and adult human ocular tissues. Mol Vis. 2001; 7: 154– 163.
    1. Ogata N, Wada M, Otsuji T, Jo N, Tombran-Tink J, Matsumura M. . Expression of pigment epithelium-derived factor in normal adult rat eye and experimental choroidal neovascularization. Invest Ophthalmol Vis Sci. 2002; 43: 1168– 1175.
    1. Chan CK, Pham LN, Chinn C,et al. . Mouse strain-dependent heterogeneity of resting limbal vasculature. Invest Ophthalmol Vis Sci. 2004; 45: 441– 447.
    1. Azar DT. . Corneal angiogenic privilege: angiogenic and antiangiogenic factors in corneal avascularity, vasculogenesis, and wound healing (an American Ophthalmological Society thesis). Trans Am Ophthalmol Soc. 2006; 104: 264– 302.
    1. Dawson DW, Volpert OV, Gillis P,et al. . Pigment epithelium-derived factor: a potent inhibitor of angiogenesis. Science. 1999; 285: 245– 248.
    1. Bouck N. . PEDF: anti-angiogenic guardian of ocular function. Trends Mol Med. 2002; 8: 330– 334.
    1. Kim J, Escalante LE, Dollar BA, Hanson SE, Hematti P. . Comparison of breast and abdominal adipose tissue mesenchymal stromal/stem cells in support of proliferation of breast cancer cells. Cancer Invest. 2013; 31: 550– 554.
    1. Kim J, Breunig MJ, Escalante LE,et al. . Biologic and immunomodulatory properties of mesenchymal stromal cells derived from human pancreatic islets. Cytotherapy. 2012; 14: 925– 935.

Source: PubMed

3
Sottoscrivi