Skip to main content
Top
Published in: Arthritis Research & Therapy 1/2017

Open Access 01-12-2017 | Research article

Proangiogenic effects of soluble α-Klotho on systemic sclerosis dermal microvascular endothelial cells

Authors: Celestina Mazzotta, Mirko Manetti, Irene Rosa, Eloisa Romano, Jelena Blagojevic, Silvia Bellando-Randone, Cosimo Bruni, Gemma Lepri, Serena Guiducci, Lidia Ibba-Manneschi, Marco Matucci-Cerinic

Published in: Arthritis Research & Therapy | Issue 1/2017

Login to get access

Abstract

Background

Systemic sclerosis (SSc) is characterized by endothelial cell (EC) apoptosis, impaired angiogenesis and peripheral microvasculopathy. Soluble α-Klotho (sKl) is a pleiotropic molecule with multiple effects on ECs, including antioxidant and vasculoprotective activities. On the EC surface, sKl interacts with vascular endothelial growth factor (VEGF) receptor-2 (VEGFR-2) and transient receptor potential canonical-1 (TRPC-1) cation channel to control EC homeostasis. Here, we investigated whether sKl might act as a protective factor to improve angiogenesis in dermal microvascular endothelial cells (MVECs) from SSc patients (SSc-MVECs).

Methods

Wound healing assay was performed on healthy dermal MVECs (H-MVECs) challenged with sera from healthy controls or SSc patients with or without the addition of sKl. Capillary morphogenesis on Matrigel was assessed in H-MVECs and SSc-MVECs at basal conditions and treated with sKl, as well as in H-MVECs challenged with healthy or SSc sera in presence or absence of sKl. The expression of α-Klotho, VEGF165b, VEGFR-2, TRPC-1, Ki67 and active caspase-3 in H-MVECs and SSc-MVECs was investigated by western blotting. Immunostaining for α-Klotho was performed in H-MVECs and SSc-MVECs, and in healthy and SSc skin sections.

Results

Treatment with sKl effectively counteracted the inihibitory effects of SSc sera on wound healing ability and angiogenic performance of H-MVECs. The addition of sKl significantly improved angiogenesis and maintained over time capillary-like tube formation in vitro by SSc-MVECs. Stimulation of SSc-MVECs with sKl resulted in the upregulation of the proliferation marker Ki67 in parallel with the downregulation of proapoptotic active caspase-3. The expression of α-Klotho was significantly lower in SSc-MVECs than in H-MVECs. The expression of TRPC-1 was also significantly decreased, while that of VEGFR-2 and VEGF165b was significantly increased, in SSc-MVECs compared with H-MVECs. Challenge with sKl either significantly increased TRPC-1 or decreased VEGF165b in SSc-MVECs. Ex vivo analyses revealed that α-Klotho immunostaining was almost absent in the dermal microvascular network of SSc skin compared with control skin.

Conclusions

Our findings provide the first evidence that α-Klotho is significantly decreased in the microvasculature in SSc skin and that sKl administration may effectively improve SSc-MVEC functions in vitro by acting as a powerful proangiogenic factor.
Literature
1.
go back to reference Balbir-Gurman A, Braun-Moscovici Y. Scleroderma - new aspects in pathogenesis and treatment. Best Pract Res Clin Rheumatol. 2012;26:13–24.CrossRefPubMed Balbir-Gurman A, Braun-Moscovici Y. Scleroderma - new aspects in pathogenesis and treatment. Best Pract Res Clin Rheumatol. 2012;26:13–24.CrossRefPubMed
2.
go back to reference LeRoy EC, Black C, Fleischmajer R, Jablonska S, Krieg T, Medsger Jr TA, et al. Scleroderma (systemic sclerosis): classification, subsets and pathogenesis. J Rheumatol. 1988;15:202–5.PubMed LeRoy EC, Black C, Fleischmajer R, Jablonska S, Krieg T, Medsger Jr TA, et al. Scleroderma (systemic sclerosis): classification, subsets and pathogenesis. J Rheumatol. 1988;15:202–5.PubMed
3.
go back to reference van den Hoogen F, Khanna D, Fransen J, Johnson SR, Baron M, Tyndall A, et al. 2013 Classification criteria for systemic sclerosis: an American College of Rheumatology/European League Against Rheumatism Collaborative initiative. Arthritis Rheum. 2013;65:2737–47.CrossRefPubMedPubMedCentral van den Hoogen F, Khanna D, Fransen J, Johnson SR, Baron M, Tyndall A, et al. 2013 Classification criteria for systemic sclerosis: an American College of Rheumatology/European League Against Rheumatism Collaborative initiative. Arthritis Rheum. 2013;65:2737–47.CrossRefPubMedPubMedCentral
4.
go back to reference Manetti M, Guiducci S, Ibba-Manneschi L, Matucci-Cerinic M. Impaired angiogenesis in systemic sclerosis: the emerging role of the antiangiogenic VEGF165b splice variant. Trends Cardiovasc Med. 2011;21:204–10.CrossRefPubMed Manetti M, Guiducci S, Ibba-Manneschi L, Matucci-Cerinic M. Impaired angiogenesis in systemic sclerosis: the emerging role of the antiangiogenic VEGF165b splice variant. Trends Cardiovasc Med. 2011;21:204–10.CrossRefPubMed
6.
go back to reference Folkman J. Angiogenesis in cancer, vascular, rheumatoid and other disease. Nat Med. 1995;1:27–31.CrossRefPubMed Folkman J. Angiogenesis in cancer, vascular, rheumatoid and other disease. Nat Med. 1995;1:27–31.CrossRefPubMed
8.
9.
go back to reference Asahara T, Murohara T, Sullivan A, Silver M, van der Zee R, Li T, et al. Isolation of putative progenitor endothelial cells for angiogenesis. Science. 1997;275:965–7.CrossRef Asahara T, Murohara T, Sullivan A, Silver M, van der Zee R, Li T, et al. Isolation of putative progenitor endothelial cells for angiogenesis. Science. 1997;275:965–7.CrossRef
11.
go back to reference De Santis M, Ceribelli A, Cavaciocchi F, Crotti C, Massarotti M, Belloli L, et al. Nailfold videocapillaroscopy and serum VEGF levels in scleroderma are associated with internal organ involvement. Auto Immun Highlights. 2016;7:5.CrossRefPubMedPubMedCentral De Santis M, Ceribelli A, Cavaciocchi F, Crotti C, Massarotti M, Belloli L, et al. Nailfold videocapillaroscopy and serum VEGF levels in scleroderma are associated with internal organ involvement. Auto Immun Highlights. 2016;7:5.CrossRefPubMedPubMedCentral
12.
go back to reference Bielecki M, Kowal K, Lapinska A, Chwiesko-Minarowska S, Chyczewski L, Kowal-Bielecka O. Peripheral blood mononuclear cells from patients with systemic sclerosis spontaneously secrete increased amounts of vascular endothelial growth factor (VEGF) already in the early stage of the disease. Adv Med Sci. 2011;56:255–63.CrossRefPubMed Bielecki M, Kowal K, Lapinska A, Chwiesko-Minarowska S, Chyczewski L, Kowal-Bielecka O. Peripheral blood mononuclear cells from patients with systemic sclerosis spontaneously secrete increased amounts of vascular endothelial growth factor (VEGF) already in the early stage of the disease. Adv Med Sci. 2011;56:255–63.CrossRefPubMed
13.
go back to reference Manetti M, Guiducci S, Romano E, Ceccarelli C, Bellando-Randone S, Conforti ML, et al. Overexpression of VEGF165b, an inhibitory splice variant of vascular endothelial growth factor, leads to insufficient angiogenesis in patients with systemic sclerosis. Circ Res. 2011;109:e14–26.CrossRefPubMed Manetti M, Guiducci S, Romano E, Ceccarelli C, Bellando-Randone S, Conforti ML, et al. Overexpression of VEGF165b, an inhibitory splice variant of vascular endothelial growth factor, leads to insufficient angiogenesis in patients with systemic sclerosis. Circ Res. 2011;109:e14–26.CrossRefPubMed
14.
go back to reference Choi JJ, Min DJ, Cho ML, Min SY, Kim SJ, Lee SS, et al. Elevated vascular endothelial growth factor in systemic sclerosis. J Rheumatol. 2003;30:1529–33.PubMed Choi JJ, Min DJ, Cho ML, Min SY, Kim SJ, Lee SS, et al. Elevated vascular endothelial growth factor in systemic sclerosis. J Rheumatol. 2003;30:1529–33.PubMed
15.
go back to reference Distler O, Distler JH, Scheid A, Acker T, Hirth A, Rethage J, et al. Uncontrolled expression of vascular endothelial growth factor and its receptors leads to insufficient skin angiogenesis in patients with systemic sclerosis. Circ Res. 2004;95:109–16.CrossRefPubMed Distler O, Distler JH, Scheid A, Acker T, Hirth A, Rethage J, et al. Uncontrolled expression of vascular endothelial growth factor and its receptors leads to insufficient skin angiogenesis in patients with systemic sclerosis. Circ Res. 2004;95:109–16.CrossRefPubMed
16.
go back to reference Kusaba T, Okigaki M, Matui A, Murakami M, Ishikawa K, Kimura T, et al. Klotho is associated with VEGF receptor-2 and the transient receptor potential canonical-1 Ca2+ channel to maintain endothelial integrity. Proc Natl Acad Sci U S A. 2010;107:19308–13.CrossRefPubMedPubMedCentral Kusaba T, Okigaki M, Matui A, Murakami M, Ishikawa K, Kimura T, et al. Klotho is associated with VEGF receptor-2 and the transient receptor potential canonical-1 Ca2+ channel to maintain endothelial integrity. Proc Natl Acad Sci U S A. 2010;107:19308–13.CrossRefPubMedPubMedCentral
17.
go back to reference Matsumura Y, Aizawa H, Shiraki-Iida T, Nagai R, Kuro-o M, Nabeshima Y. Identification of the human Klotho gene and its two transcripts encoding membrane and secreted Klotho protein. Biochem Biophys Res Commun. 1998;242:626–30.CrossRefPubMed Matsumura Y, Aizawa H, Shiraki-Iida T, Nagai R, Kuro-o M, Nabeshima Y. Identification of the human Klotho gene and its two transcripts encoding membrane and secreted Klotho protein. Biochem Biophys Res Commun. 1998;242:626–30.CrossRefPubMed
18.
go back to reference Shiraki-Iida T, Aizawa H, Matsumura Y, Sekine S, Iida A, Anazawa H, et al. Structure of the mouse klotho gene and its two transcripts encoding membrane and secreted protein. FEBS Lett. 1998;424:6–10.CrossRefPubMed Shiraki-Iida T, Aizawa H, Matsumura Y, Sekine S, Iida A, Anazawa H, et al. Structure of the mouse klotho gene and its two transcripts encoding membrane and secreted protein. FEBS Lett. 1998;424:6–10.CrossRefPubMed
19.
go back to reference Kuro-o M, Matsumura Y, Aizawa H, Kawaguchi H, Suga T, Utsugi T, et al. Mutation of the mouse Klotho gene leads to a syndrome resembling ageing. Nature. 1997;390:45–51.CrossRefPubMed Kuro-o M, Matsumura Y, Aizawa H, Kawaguchi H, Suga T, Utsugi T, et al. Mutation of the mouse Klotho gene leads to a syndrome resembling ageing. Nature. 1997;390:45–51.CrossRefPubMed
20.
21.
go back to reference Imura A, Iwano A, Tohyama O, Tsuji Y, Nozaki K, Hashimoto N, et al. Secreted Klotho protein in sera and CSF: implication for post-translational cleavage in release of Klotho protein from cell membrane. FEBS Lett. 2004;565:143–7.CrossRefPubMed Imura A, Iwano A, Tohyama O, Tsuji Y, Nozaki K, Hashimoto N, et al. Secreted Klotho protein in sera and CSF: implication for post-translational cleavage in release of Klotho protein from cell membrane. FEBS Lett. 2004;565:143–7.CrossRefPubMed
22.
go back to reference Li SA, Watanabe M, Yamada H, Nagai A, Kinuta M, Takei K. Immunohistochemical localization of Klotho protein in brain, kidney, and reproductive organs of mice. Cell Struct Funct. 2004;29:91–9.CrossRefPubMed Li SA, Watanabe M, Yamada H, Nagai A, Kinuta M, Takei K. Immunohistochemical localization of Klotho protein in brain, kidney, and reproductive organs of mice. Cell Struct Funct. 2004;29:91–9.CrossRefPubMed
23.
go back to reference Kurosu H, Ogawa Y, Miyoshi M, Yamamoto M, Nandi A, Rosenblatt KP, et al. Regulation of fibroblast growth factor-23 signaling by klotho. J Biol Chem. 2006;281:6120–3.CrossRefPubMedPubMedCentral Kurosu H, Ogawa Y, Miyoshi M, Yamamoto M, Nandi A, Rosenblatt KP, et al. Regulation of fibroblast growth factor-23 signaling by klotho. J Biol Chem. 2006;281:6120–3.CrossRefPubMedPubMedCentral
24.
go back to reference Kuro-o M. Klotho as a regulator of fibroblast growth factor signaling and phosphate/calcium metabolism. Curr Opin Nephrol Hypertens. 2006;15:437–41.CrossRefPubMed Kuro-o M. Klotho as a regulator of fibroblast growth factor signaling and phosphate/calcium metabolism. Curr Opin Nephrol Hypertens. 2006;15:437–41.CrossRefPubMed
26.
go back to reference Yamamoto M, Clark JD, Pastor JV, Gurnani P, Nandi A, Kurosu H, et al. Regulation of oxidative stress by the anti-aging hormone klotho. J Biol Chem. 2005;280:38029–34.CrossRefPubMedPubMedCentral Yamamoto M, Clark JD, Pastor JV, Gurnani P, Nandi A, Kurosu H, et al. Regulation of oxidative stress by the anti-aging hormone klotho. J Biol Chem. 2005;280:38029–34.CrossRefPubMedPubMedCentral
27.
go back to reference Hsieh CC, Kuro-o M, Rosenblatt KP, Brobey R, Papaconstantinou J. The ASK1-Signalosome regulates p38 MAPK activity in response to levels of endogenous oxidative stress in the Klotho mouse models of aging. Aging (Albany NY). 2010;2:597–611.CrossRef Hsieh CC, Kuro-o M, Rosenblatt KP, Brobey R, Papaconstantinou J. The ASK1-Signalosome regulates p38 MAPK activity in response to levels of endogenous oxidative stress in the Klotho mouse models of aging. Aging (Albany NY). 2010;2:597–611.CrossRef
29.
go back to reference Chen CD, Podvin S, Gillespie E, Leeman SE, Abraham CR. Insulin stimulates the cleavage and release of the extracellular domain of Klotho by ADAM10 and ADAM17. Proc Natl Acad Sci U S A. 2007;104:19796–801.CrossRefPubMedPubMedCentral Chen CD, Podvin S, Gillespie E, Leeman SE, Abraham CR. Insulin stimulates the cleavage and release of the extracellular domain of Klotho by ADAM10 and ADAM17. Proc Natl Acad Sci U S A. 2007;104:19796–801.CrossRefPubMedPubMedCentral
30.
go back to reference Ohyama Y, Kurabayashi M, Masuda H, Nakamura T, Aihara Y, Kaname T, et al. Molecular cloning of rat klotho cDNA: markedly decreased expression of klotho by acute inflammatory stress. Biochem Biophys Res Commun. 1998;251:920–5.CrossRefPubMed Ohyama Y, Kurabayashi M, Masuda H, Nakamura T, Aihara Y, Kaname T, et al. Molecular cloning of rat klotho cDNA: markedly decreased expression of klotho by acute inflammatory stress. Biochem Biophys Res Commun. 1998;251:920–5.CrossRefPubMed
31.
go back to reference Vervloet MG, Larsson TE. Fibroblast growth factor-23 and Klotho in chronic kidney disease. Kidney Int. 2011;1:130–5.CrossRef Vervloet MG, Larsson TE. Fibroblast growth factor-23 and Klotho in chronic kidney disease. Kidney Int. 2011;1:130–5.CrossRef
32.
go back to reference Wang Y, Kuro-o M, Sun Z. Klotho gene delivery suppresses Nox2 expression and attenuates oxidative stress in rat aortic smooth muscle cells via the cAMP-PKA pathway. Aging Cell. 2012;11:410–7.CrossRefPubMedPubMedCentral Wang Y, Kuro-o M, Sun Z. Klotho gene delivery suppresses Nox2 expression and attenuates oxidative stress in rat aortic smooth muscle cells via the cAMP-PKA pathway. Aging Cell. 2012;11:410–7.CrossRefPubMedPubMedCentral
33.
go back to reference Scott PA, Bicknell R. The isolation and culture of microvascular endothelium. J Cell Sci. 1993;105:269–73.PubMed Scott PA, Bicknell R. The isolation and culture of microvascular endothelium. J Cell Sci. 1993;105:269–73.PubMed
34.
go back to reference D’Alessio S, Fibbi G, Cinelli M, Guiducci S, Del Rosso A, Margheri F, et al. Matrix metalloproteinase 12-dependent cleavage of urokinase receptor in systemic sclerosis microvascular endothelial cells results in impaired angiogenesis. Arthritis Rheum. 2004;50:3275–85.CrossRefPubMed D’Alessio S, Fibbi G, Cinelli M, Guiducci S, Del Rosso A, Margheri F, et al. Matrix metalloproteinase 12-dependent cleavage of urokinase receptor in systemic sclerosis microvascular endothelial cells results in impaired angiogenesis. Arthritis Rheum. 2004;50:3275–85.CrossRefPubMed
35.
go back to reference Margheri F, Manetti M, Serratì S, Nosi D, Pucci M, Matucci-Cerinic M, et al. Domain 1 of the urokinase-type plasminogen activator receptor is required for its morphologic and functional, beta2 integrin-mediated connection with actin cytoskeleton in human microvascular endothelial cells: failure of association in systemic sclerosis endothelial cells. Arthritis Rheum. 2006;54:3926–38.CrossRefPubMed Margheri F, Manetti M, Serratì S, Nosi D, Pucci M, Matucci-Cerinic M, et al. Domain 1 of the urokinase-type plasminogen activator receptor is required for its morphologic and functional, beta2 integrin-mediated connection with actin cytoskeleton in human microvascular endothelial cells: failure of association in systemic sclerosis endothelial cells. Arthritis Rheum. 2006;54:3926–38.CrossRefPubMed
36.
go back to reference Ikushima M, Rakugi H, Ishikawa K, Maekawa Y, Yamamoto K, Ohta J, et al. Anti-apoptotic and anti-senescence effects of klotho on vascular endothelial cells. Biochem Res Commun. 2006;339:827–32.CrossRef Ikushima M, Rakugi H, Ishikawa K, Maekawa Y, Yamamoto K, Ohta J, et al. Anti-apoptotic and anti-senescence effects of klotho on vascular endothelial cells. Biochem Res Commun. 2006;339:827–32.CrossRef
37.
go back to reference Maekawa Y, Ohishi M, Ikushima M, Yamamoto K, Yasuda O, Oguro R, et al. Klotho protein diminishes endothelial apoptosis and senescence via a mitogen-activated kinase pathway. Geriatric Gerontol Int. 2011;11:510–6.CrossRef Maekawa Y, Ohishi M, Ikushima M, Yamamoto K, Yasuda O, Oguro R, et al. Klotho protein diminishes endothelial apoptosis and senescence via a mitogen-activated kinase pathway. Geriatric Gerontol Int. 2011;11:510–6.CrossRef
38.
go back to reference de Oliveira RM. Klotho RNAi induces premature senescence of human cells via a p53/p21 dependent pathway. FEBS Lett. 2006;580:5753–8.CrossRefPubMed de Oliveira RM. Klotho RNAi induces premature senescence of human cells via a p53/p21 dependent pathway. FEBS Lett. 2006;580:5753–8.CrossRefPubMed
39.
go back to reference Safirstein R, DiMari J, Megyesi J, Price P. Mechanisms of renal repair and survival following acute injury. Semin Nephrol. 1998;18:519–22. Safirstein R, DiMari J, Megyesi J, Price P. Mechanisms of renal repair and survival following acute injury. Semin Nephrol. 1998;18:519–22.
41.
go back to reference Imura A, Tsuji Y, Murata M, Maeda R, Kubota K, Iwano A, et al. Alpha-Klotho as a regulator of calcium homeostasis. Science. 2007;316:1615–8.CrossRefPubMed Imura A, Tsuji Y, Murata M, Maeda R, Kubota K, Iwano A, et al. Alpha-Klotho as a regulator of calcium homeostasis. Science. 2007;316:1615–8.CrossRefPubMed
42.
go back to reference Griffith TM, Edwards DH, Lewis MJ, Newby AC, Henderson AH. The nature of endothelium-derived vascular relaxant factor. Nature. 1984;308:645–7.CrossRefPubMed Griffith TM, Edwards DH, Lewis MJ, Newby AC, Henderson AH. The nature of endothelium-derived vascular relaxant factor. Nature. 1984;308:645–7.CrossRefPubMed
43.
go back to reference Palmer RM, Ferrige AG, Moncada S. Nitric oxide release accounts for the biological activity of endothelium-derived relaxing factor. Nature. 1987;327:524–6.CrossRefPubMed Palmer RM, Ferrige AG, Moncada S. Nitric oxide release accounts for the biological activity of endothelium-derived relaxing factor. Nature. 1987;327:524–6.CrossRefPubMed
44.
go back to reference Saito Y, Nakamura T, Ohyama Y, Suzuki T, Iida A, Shiraki-Iida T, et al. In vivo klotho gene delivery protects against endothelial dysfunction in multiple risk factor syndrome. Biochem Biophys Res Commun. 2000;276:767–72.CrossRefPubMed Saito Y, Nakamura T, Ohyama Y, Suzuki T, Iida A, Shiraki-Iida T, et al. In vivo klotho gene delivery protects against endothelial dysfunction in multiple risk factor syndrome. Biochem Biophys Res Commun. 2000;276:767–72.CrossRefPubMed
45.
go back to reference Rakugi H, Matsukawa N, Ishikawa K, Yang J, Imai M, Ikushima M, et al. Anti-oxidative effect of Klotho on endothelial cells through cAMP activation. Endocrine. 2007;31:82–7.CrossRefPubMed Rakugi H, Matsukawa N, Ishikawa K, Yang J, Imai M, Ikushima M, et al. Anti-oxidative effect of Klotho on endothelial cells through cAMP activation. Endocrine. 2007;31:82–7.CrossRefPubMed
46.
go back to reference Mukai Y, Shimokawa H, Higashi M, Morikawa K, Matoba T, Hiroki J, et al. Inhibition of renin-angiotensin system ameliorates endothelial dysfunction associated with aging in rats. Arterioscler Thromb Vasc Biol. 2002;22:1445–50.CrossRefPubMed Mukai Y, Shimokawa H, Higashi M, Morikawa K, Matoba T, Hiroki J, et al. Inhibition of renin-angiotensin system ameliorates endothelial dysfunction associated with aging in rats. Arterioscler Thromb Vasc Biol. 2002;22:1445–50.CrossRefPubMed
47.
go back to reference Okada S, Yoshida T, Hong Z, Ishii G, Hatano M. Kuro-OM, et al. Impairment of B lymphopoiesis in precocious aging (klotho) mice. Int Immunol. 2000;12:861–71.CrossRefPubMed Okada S, Yoshida T, Hong Z, Ishii G, Hatano M. Kuro-OM, et al. Impairment of B lymphopoiesis in precocious aging (klotho) mice. Int Immunol. 2000;12:861–71.CrossRefPubMed
48.
go back to reference Shimada T, Takeshita Y, Murohara T, Sasaki K, Egami K, Shintani S, et al. Angiogenesis and vasculogenesis are impaired in the precocious-aging klotho mouse. Circulation. 2004;110:1148–55.CrossRefPubMed Shimada T, Takeshita Y, Murohara T, Sasaki K, Egami K, Shintani S, et al. Angiogenesis and vasculogenesis are impaired in the precocious-aging klotho mouse. Circulation. 2004;110:1148–55.CrossRefPubMed
49.
go back to reference Kuwana M, Okazaki Y, Yasuoka H, Kawakami Y, Ikeda Y. Defective vasculogenesis in systemic sclerosis. Lancet. 2004;364:603–10.CrossRefPubMed Kuwana M, Okazaki Y, Yasuoka H, Kawakami Y, Ikeda Y. Defective vasculogenesis in systemic sclerosis. Lancet. 2004;364:603–10.CrossRefPubMed
50.
go back to reference Avouac J, Juin F, Wipff J, Couraud PO, Chiocchia G, Kahan A, et al. Circulating endothelial progenitor cells in systemic sclerosis: association with disease severity. Ann Rheum Dis. 2008;67:1455–60.CrossRefPubMed Avouac J, Juin F, Wipff J, Couraud PO, Chiocchia G, Kahan A, et al. Circulating endothelial progenitor cells in systemic sclerosis: association with disease severity. Ann Rheum Dis. 2008;67:1455–60.CrossRefPubMed
51.
go back to reference Manetti M, Guiducci S, Ibba-Manneschi L, Matucci-Cerinic M. Mechanisms in the loss of capillaries in systemic sclerosis: angiogenesis versus vasculogenesis. J Cell Mol Med. 2010;14:1241–54.CrossRefPubMedPubMedCentral Manetti M, Guiducci S, Ibba-Manneschi L, Matucci-Cerinic M. Mechanisms in the loss of capillaries in systemic sclerosis: angiogenesis versus vasculogenesis. J Cell Mol Med. 2010;14:1241–54.CrossRefPubMedPubMedCentral
52.
go back to reference Cipriani P, Guiducci S, Miniati I, Cinelli M, Urbani S, Marrelli A, et al. Impairment of endothelial cell differentiation from bone marrow-derived mesenchymal stem cells: new insight into the pathogenesis of systemic sclerosis. Arthritis Rheum. 2007;56:1994–2004.CrossRefPubMed Cipriani P, Guiducci S, Miniati I, Cinelli M, Urbani S, Marrelli A, et al. Impairment of endothelial cell differentiation from bone marrow-derived mesenchymal stem cells: new insight into the pathogenesis of systemic sclerosis. Arthritis Rheum. 2007;56:1994–2004.CrossRefPubMed
54.
go back to reference Kahaleh MB. Vascular involvement in systemic sclerosis. Clin Exp Rheumatol. 2004;22:S19–23.PubMed Kahaleh MB. Vascular involvement in systemic sclerosis. Clin Exp Rheumatol. 2004;22:S19–23.PubMed
55.
go back to reference Manetti M, Guiducci S, Matucci-Cerinic M. The crowded crossroad to angiogenesis in systemic sclerosis: where is the key to the problem? Arthritis Res Ther. 2016;18:36.CrossRefPubMedPubMedCentral Manetti M, Guiducci S, Matucci-Cerinic M. The crowded crossroad to angiogenesis in systemic sclerosis: where is the key to the problem? Arthritis Res Ther. 2016;18:36.CrossRefPubMedPubMedCentral
Metadata
Title
Proangiogenic effects of soluble α-Klotho on systemic sclerosis dermal microvascular endothelial cells
Authors
Celestina Mazzotta
Mirko Manetti
Irene Rosa
Eloisa Romano
Jelena Blagojevic
Silvia Bellando-Randone
Cosimo Bruni
Gemma Lepri
Serena Guiducci
Lidia Ibba-Manneschi
Marco Matucci-Cerinic
Publication date
01-12-2017
Publisher
BioMed Central
Published in
Arthritis Research & Therapy / Issue 1/2017
Electronic ISSN: 1478-6362
DOI
https://doi.org/10.1186/s13075-017-1233-0

Other articles of this Issue 1/2017

Arthritis Research & Therapy 1/2017 Go to the issue