Skip to main content
Top
Published in: Diabetologia 3/2019

01-03-2019 | Diabetic Retinopathy | Article

lncRNA H19 prevents endothelial–mesenchymal transition in diabetic retinopathy

Authors: Anu A. Thomas, Saumik Biswas, Biao Feng, Shali Chen, John Gonder, Subrata Chakrabarti

Published in: Diabetologia | Issue 3/2019

Login to get access

Abstract

Aims/hypothesis

The pathophysiology of diabetic retinopathy is linked to hyperglycaemia and its effect on retinal microvascular tissues. The resulting endothelial injury changes the endothelial cell phenotype to acquire mesenchymal properties (i.e. endothelial–mesenchymal transition [EndMT]). Such changes can be regulated by epigenetic mechanisms, including long non-coding RNAs (lncRNAs). lncRNA H19 may influence EndMT through TGF-β. We investigated the role of H19 in regulating EndMT during diabetic retinopathy.

Methods

H19 was overexpressed or silenced in human retinal endothelial cells exposed to various glucose levels. The cells were examined for H19, endothelial and mesenchymal markers. We then expanded the study to retinal tissues in a mouse model of diabetic retinopathy and also examined vitreous humour samples from individuals with proliferative diabetic retinopathy.

Results

Expression of H19 was downregulated in high glucose conditions (25 mmol/l). H19 overexpression prevented glucose-induced EndMT. Such changes appear to involve TGF-β through a Smad-independent mechanism. Diabetes caused downregulation of retinal H19. Using H19 knockout mice, we demonstrated similar EndMT in the retina. Examination of vitreous humour from individuals with proliferative diabetic retinopathy also reinforced the downregulation of H19 in diabetes.

Conclusions/interpretation

We therefore concluded that H19 regulates EndMT in diabetic retinopathy through specific mechanisms.

Data availability

The results from our previous microarray can be found online using the GEO accession number GSE122189.
Appendix
Available only for authorised users
Literature
4.
go back to reference Wong TY, Cheung CM, Larsen M, Sharma S, Simó R (2016) Diabetic retinopathy. Nat Rev Dis Primers 2:1–16CrossRef Wong TY, Cheung CM, Larsen M, Sharma S, Simó R (2016) Diabetic retinopathy. Nat Rev Dis Primers 2:1–16CrossRef
5.
go back to reference Balakumar P, Jindal S, Shah DI, Singh M (2007) Experimental models for vascular endothelial dysfunction. Trends Med Res 2:12–20CrossRef Balakumar P, Jindal S, Shah DI, Singh M (2007) Experimental models for vascular endothelial dysfunction. Trends Med Res 2:12–20CrossRef
7.
22.
go back to reference Raveh E, Matouk IJ, Gilon M, Hochberg A (2015) The H19 long non-coding RNA in cancer intiation, progression and metastasis-a proposed unifying theory. Mol Cancer 14:1–14CrossRef Raveh E, Matouk IJ, Gilon M, Hochberg A (2015) The H19 long non-coding RNA in cancer intiation, progression and metastasis-a proposed unifying theory. Mol Cancer 14:1–14CrossRef
27.
go back to reference Feng B, Cao Y, Chen S et al (2016) miR-200b mediates endothelial-to- mesenchymal transition in diabetic cardiomyopathy. Diabetes 65:1–12CrossRef Feng B, Cao Y, Chen S et al (2016) miR-200b mediates endothelial-to- mesenchymal transition in diabetic cardiomyopathy. Diabetes 65:1–12CrossRef
28.
go back to reference Zhu GH, Li R, Zeng Y, Zhou T, Xiong F, Zhu M (2018) MicroRNA-142-3p inhibits high-glucose-induced endothelial-to-mesenchymal transition through targeting TGF-β1/Smad pathway in primary human aortic endothelial cells. Int J Clin Exp Pathol 11:1208–1217PubMedPubMedCentral Zhu GH, Li R, Zeng Y, Zhou T, Xiong F, Zhu M (2018) MicroRNA-142-3p inhibits high-glucose-induced endothelial-to-mesenchymal transition through targeting TGF-β1/Smad pathway in primary human aortic endothelial cells. Int J Clin Exp Pathol 11:1208–1217PubMedPubMedCentral
32.
go back to reference Biswas S, Thomas AA, Chen S et al (2018) MALAT1: an epigenetic regulator of inflammation in diabetic retinopathy. Sci Rep 8:1–15CrossRef Biswas S, Thomas AA, Chen S et al (2018) MALAT1: an epigenetic regulator of inflammation in diabetic retinopathy. Sci Rep 8:1–15CrossRef
35.
go back to reference Gordon AD, Biswas SS, Feng B, Chakrabarti S (2018) MALAT1: a regulator of inflammatory cytokines in diabetic retinopathy. Endocrinol Diabetes Metab 1:1–11CrossRef Gordon AD, Biswas SS, Feng B, Chakrabarti S (2018) MALAT1: a regulator of inflammatory cytokines in diabetic retinopathy. Endocrinol Diabetes Metab 1:1–11CrossRef
36.
go back to reference Wu Y, Zuo Y, Chakrabarti R, Feng B, Chen S, Chakrbarti S (2010) ERK5 contributes to VEGF alteration in diabetic retinopathy. J Ophthalmol 2010:1–11 Wu Y, Zuo Y, Chakrabarti R, Feng B, Chen S, Chakrbarti S (2010) ERK5 contributes to VEGF alteration in diabetic retinopathy. J Ophthalmol 2010:1–11
38.
go back to reference Tang R, Li Q, Lv L et al (2010) Angiotensin II mediates the high-glucose-induced endothelial-to-mesenchymal transition in human aortic endothelial cells. Cardiovasc Diabetol 9:1–7 Tang R, Li Q, Lv L et al (2010) Angiotensin II mediates the high-glucose-induced endothelial-to-mesenchymal transition in human aortic endothelial cells. Cardiovasc Diabetol 9:1–7
40.
go back to reference Xu Q, Deng F, Qin Y et al (2016) Long non-coding RNA regulation of epithelial–mesenchymal transition in cancer metastasis. Cell Death Dis 7:1–10 Xu Q, Deng F, Qin Y et al (2016) Long non-coding RNA regulation of epithelial–mesenchymal transition in cancer metastasis. Cell Death Dis 7:1–10
42.
go back to reference Neumann P, Jae N, Knau A et al (2018) The lncRNA GATA6-AS epigenetically regulates endothelial gene expression via interaction with LOXL2. Nat Commun 9:1–12CrossRef Neumann P, Jae N, Knau A et al (2018) The lncRNA GATA6-AS epigenetically regulates endothelial gene expression via interaction with LOXL2. Nat Commun 9:1–12CrossRef
45.
go back to reference Popov D (2020) Endothelial cell dysfunction in hyperglycemia: phenotypic change, intracellular signaling modification, ultrastructural alteration, and potential clinical outcomes. In J Diabetes Mellitus 2:189–195CrossRef Popov D (2020) Endothelial cell dysfunction in hyperglycemia: phenotypic change, intracellular signaling modification, ultrastructural alteration, and potential clinical outcomes. In J Diabetes Mellitus 2:189–195CrossRef
46.
go back to reference Biswas S, Chakrabarti S (2017) Pathogenetic mechanisms in diabetic retinopathy: from molecules to cells to tissues. In: Kartha CC, Ramachandran S, Pillai RM (eds) Mechanisms of Vascular Defects in Diabetes Mellitus, Advances in Biochemistry in Health and Disease, 1st edn. Springer, Cham, pp 209–247 Biswas S, Chakrabarti S (2017) Pathogenetic mechanisms in diabetic retinopathy: from molecules to cells to tissues. In: Kartha CC, Ramachandran S, Pillai RM (eds) Mechanisms of Vascular Defects in Diabetes Mellitus, Advances in Biochemistry in Health and Disease, 1st edn. Springer, Cham, pp 209–247
49.
go back to reference Abu El-Asrar AM, Struyf S, Kangave D, Van Damme J (2006) Chemokines in proliferative diabetic retinopathy and proliferative vitreoretinopathy. Eur Cytokine Netw 17(3):155–165PubMed Abu El-Asrar AM, Struyf S, Kangave D, Van Damme J (2006) Chemokines in proliferative diabetic retinopathy and proliferative vitreoretinopathy. Eur Cytokine Netw 17(3):155–165PubMed
50.
go back to reference Peng H, Li Y, Wang C et al (2016) ROCK1 induces endothelial-to-mesenchymal transition in glomeruli to aggravate albuminuria in diabetic nephropathy. Sci Rep 6:1–10CrossRef Peng H, Li Y, Wang C et al (2016) ROCK1 induces endothelial-to-mesenchymal transition in glomeruli to aggravate albuminuria in diabetic nephropathy. Sci Rep 6:1–10CrossRef
52.
go back to reference Yan XC, Cao J, Liang L et al (2016) miR-342-5p Is a Notch downstream molecule and regulates multiple angiogenic pathways including Notch, vascular endothelial growth factor and transforming growth factor signaling. J Am Heart Assoc 5:1–15 Yan XC, Cao J, Liang L et al (2016) miR-342-5p Is a Notch downstream molecule and regulates multiple angiogenic pathways including Notch, vascular endothelial growth factor and transforming growth factor signaling. J Am Heart Assoc 5:1–15
53.
go back to reference Pardali E, Sanchez-Duffhues G, Gomez-Puerto MC, Dijke P (2017) TGF-induced endothelial-mesenchymal transition in fibrotic diseases. Int J Mol Sci 18:1–22CrossRef Pardali E, Sanchez-Duffhues G, Gomez-Puerto MC, Dijke P (2017) TGF-induced endothelial-mesenchymal transition in fibrotic diseases. Int J Mol Sci 18:1–22CrossRef
54.
go back to reference Biswas S, Thomas AA, Chakrabarti S (2018) LncRNAs: proverbial genomic junk or key epigenetic regulators during cardiac fibrosis in diabetes? Front Cardiovasc Med 2018:1–13 Biswas S, Thomas AA, Chakrabarti S (2018) LncRNAs: proverbial genomic junk or key epigenetic regulators during cardiac fibrosis in diabetes? Front Cardiovasc Med 2018:1–13
59.
go back to reference Cruz-Solbes AS (2017) Epithelial to mesenchymal transition (EMT) and endothelial to mesenchymal transition (EndMT): role and implications in kidney fibrosis. Kidney Dev Disease 60:345–372CrossRef Cruz-Solbes AS (2017) Epithelial to mesenchymal transition (EMT) and endothelial to mesenchymal transition (EndMT): role and implications in kidney fibrosis. Kidney Dev Disease 60:345–372CrossRef
Metadata
Title
lncRNA H19 prevents endothelial–mesenchymal transition in diabetic retinopathy
Authors
Anu A. Thomas
Saumik Biswas
Biao Feng
Shali Chen
John Gonder
Subrata Chakrabarti
Publication date
01-03-2019
Publisher
Springer Berlin Heidelberg
Published in
Diabetologia / Issue 3/2019
Print ISSN: 0012-186X
Electronic ISSN: 1432-0428
DOI
https://doi.org/10.1007/s00125-018-4797-6

Other articles of this Issue 3/2019

Diabetologia 3/2019 Go to the issue