• No results found

University of Groningen Endothelial cell transcriptional regulation in vascular disease Sol, Marloes

N/A
N/A
Protected

Academic year: 2021

Share "University of Groningen Endothelial cell transcriptional regulation in vascular disease Sol, Marloes"

Copied!
19
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

Endothelial cell transcriptional regulation in vascular disease

Sol, Marloes

DOI:

10.33612/diss.136419916

IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to cite from

it. Please check the document version below.

Document Version

Publisher's PDF, also known as Version of record

Publication date:

2020

Link to publication in University of Groningen/UMCG research database

Citation for published version (APA):

Sol, M. (2020). Endothelial cell transcriptional regulation in vascular disease. University of Groningen.

https://doi.org/10.33612/diss.136419916

Copyright

Other than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), unless the work is under an open content license (like Creative Commons).

Take-down policy

If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim.

Downloaded from the University of Groningen/UMCG research database (Pure): http://www.rug.nl/research/portal. For technical reasons the number of authors shown on this cover page is limited to 10 maximum.

(2)

Chapter 1

General Introduction

(3)

GENERAL INTRODUCTION

The blood vascular system in the human body consists of the heart and blood vessels. Blood vessels are important as the conduits for the transportation of blood and the regulation of exchange of substances in the perfused tissues. Blood vessels can be divided in five different types that differ in diameter, morphology and function: arteries,  arterioles, capillaries, venules, and veins. The arteries are responsible for transportation of blood from the heart into organs and tissues. The arteries enter organs and tissues and branch into smaller arterioles, which distribute blood into the capillaries. Capillaries enable the exchange of oxygen, nutrients and catabolites across the endothelium. Arterioles provide resistance to blood flow and consequently regulate blood pressure  and tissue perfusion [1]. Subsequently, the blood is collected in post-capillary venules  and then flows into veins that are essential for the vascular reserve capacity and take  care of the transportation of blood back to the heart.

THE ENDOTHELIUM

The endothelium constitutes the inner monolayer of cells that lines all blood vessels and covers a surface area of approximately 1000 m2 in an average 70-kg human

[2]. The endothelium is a dynamic tissue consisting of heterogenic populations of endothelial cells that differ in morphology and function, but together maintain circulatory homeostasis [3]. The endothelium is pivotal in the regulation of multiple processes, including the exchange of oxygen and nutrients, vascular tone, vascular permeability, inflammation, angiogenesis, and haemostasis. Via molecular cross-talk,  the  endothelium  also  influences  the  function  of  cells  in  the  underlying  tissues.  An  example of molecular cross-talk by endothelial cells is the regulation of vascular tone via smooth muscle cell relaxation by the production of endothelial nitric oxide (NO) [4-6]. Proper functioning of endothelial cells is essential to maintain vascular health. Dysfunction of the endothelium is a key initiating step in the pathophysiology of several cardiovascular and renal diseases, including glomerulosclerosis and atherosclerosis [3, 7-12]. In glomerulosclerosis and atherosclerosis, endothelial homeostasis is disturbed leading to endothelial cell dysfunction.

ENDOTHELIAL CELL FUNCTION IN GLOMERULOSCLEROSIS

Blood  filtration  and  urine  production  take  place  in  the  functional  units  of  the  kidney,  i.e.  the  nephrons.  Each  nephron  consists  of  a  glomerulus,  where  the  blood  filtration  takes place, and a tubule, a segmented compartment responsible for reabsorption of water, small molecules and ions, and the excretion of toxins. The glomerulus is a tuft of microcapillaries enclosed by the Bowman’s capsule lined with parietal epithelial cells.

(4)

In the glomeruli, the glomerular filtration barrier (GFB) is responsible for the size- and  charge-selective filtration of molecules from the blood. The GFB is composed of non-diaphragmed fenestrated glomerular endothelial cells (GEnCs), which are covered by the endothelial glycocalyx at the luminal side, the glomerular basement membrane (GBM), and podocytes with interdigitating foot processes wrapped around the exterior of glomerular capillaries (fig.1). Intercallating mesangial cells are present between the  capillaries and are in direct contact with the glomerular endothelium.

Figure 1. Schematic representation of the glomerular filtration barrier. The  glomerular  filtration  barrier 

(GFB) is composed of fenestrated glomerular endothelial cells (GEnCs), which are covered by the endothelial glycocalyx, the glomerular basement membrane (GBM), and podocytes with interdigitating foot processes. The GFB filters water and small molecules out of the blood into the urinary space. Mesangial cells are in direct contact  with the glomerular endothelium.

Glomerulosclerosis  is  the  scarring  of  the  glomeruli  (fig.  2).  Glomerulosclerosis  is  an  end-stage chronic pathologic condition that is often observed in kidney diseases, such as focal segmental glomerulosclerosis (FSGS) and diabetic nephropathy (DN). Glomerulosclerosis is a common pathway leading to the development of renal failure and end-stage renal disease (ESRD), irrespective of the underlying primary renal disease. Glomerulosclerosis is characterized by damage to podocytes and dysfunction of glomerular endothelial cells [10, 13-15], which culminate into proteinuria (the loss of blood-derived proteins into the urine). Furthermore, glomerulosclerosis is characterized by  activation  of  inflammatory  pathways  in  the  glomerulus  [16,  17],  mesangial  cell proliferation, and the accumulation of extracellular matrix in the glomerulus.

(5)

Glomerulosclerosis ultimately leads to obliteration of glomerular capillaries, due to the intracapillary accumulation of proteinaceous material (hyalinosis), extracellular matrix, and occasionally inflammatory cells [18, 19]. 

Figure 2. Healthy glomerulus and glomerulosclerosis. Light microscopy photomicrographs of glomeruli,

stained with hematoxylin-eosin. Nuclei are stained in blue. Cytoplasm, cell membranes and extracellular matrix are stained in pink. (A) In a healthy glomerulus, multiple capillary loops are observed (red arrows). (B) A glomerulus from a diabetic nephropathy patient presenting glomerulosclerosis, representing an increase in extracellular matrix in the glomerulus, by which capillary loops are obliterated. Scale bars represent 50µm. Courtesy of Dr. M. C. van den Heuvel.

GEnCs are the first cells of the GFB in contact with the blood. At the luminal side, GEnCs  are covered by the endothelial glycocalyx, a gel-like layer consisting of proteoglycans with covalently bound glycosaminoglycan (GAG) side chains [20-22]. The endothelial glycocalyx is negatively charged and provides a barrier to proteins and hereby contributes to a large extent to the charge and size-selective filtration of blood by the GFB [20, 21,  23-25]. Furthermore, the endothelial glycocalyx is essential for vascular protection, attenuation of blood cell-vessel wall interactions, mechanotransduction as it serves as the primary sensor of shear stress, and signaling processes such as NO production. In glomerulosclerosis, the endothelial glycocalyx is degraded. Degradation of the glomerular endothelial glycocalyx has been shown to be important in the development of proteinuria and kidney failure [26, 27]. Different types of GAGs are located in the endothelial glycocalyx, including heparan sulphate (HS). HS plays an important role in regulating the specific  permeability of the GFB [28, 29]. Heparanase (HPSE), the only mammalian enzyme that can cleave HS, is involved in the pathogenesis of several glomerular diseases by reducing HS in the endothelial glycocalyx and other compartments of the GFB, including the GBM

(6)

[20, 27, 30, 31]. Next to HPSE, matrix metalloproteases are known to be involved in the degradation of the endothelial glycocalyx as well [32, 33]. Other mechanisms involved in endothelial glycocalyx degradation remain largely unknown.

ENDOTHELIAL CELL FUNCTION IN ATHEROSCLEROSIS

Under physiological conditions, the arterial endothelium, together with some subendothelial connective tissue and an elastic membrane (i.e. the internal elastic lamina),  form  the  tunica  intima.  Vascular  smooth  muscle  cells,  elastic  lamellae  and  collagen comprise the tunica media. The outer layer of arteries is composed of fibroblasts  and collagens, called the tunica externa (or tunica adventitia) (fig. 3). 

Figure 3. Schematic representation of a normal artery. The endothelium lines the inner layer of the artery

and together with elastic tissue (the internal elastic lamina) and some subendothelial connective tissue forms the tunica intima. Smooth muscle cells are present in the middle layer, and together with elastin lamellae and collagens form the tunica media. Fibroblasts are found on the outer layer of the artery, and together with collagens form the tunica externa (or tunica adventitia).

Atherosclerosis is a chronic cardiovascular disease. Atherogenesis comprises a process in which a new intimal layer is formed (neo-intima) [34, 35]. Atherogenesis is characterized by accumulation of oxidized low-density lipoprotein cholesterol (ox-LDL) in  the  vessel  wall  [36],  the  infiltration  of  inflammatory  cells  [37],  smooth  muscle  cell  migration and proliferation in the neo-intima [38], and the accumulation of extracellular matrix proteins in the neo-intima. Together, these processes result in intimal thickening [39] that ultimately restricts blood flow through the artery (fig. 4). 

(7)

Figure 4. Healthy (A) and atherosclerotic (B) coronary artery. Light microscopy photomicrographs of arteries,

stained with hematoxylin-eosin. Nuclei are stained in blue. Cytoplasm, cell membranes and extracellular matrix are stained in pink. (A) In a healthy artery, the vessel wall normal layers of tunica intima (I), media (II) and externa (III).  (B)  In  atherosclerosis,  a  new  intimal  layer  is  formed  (neo-intima,  IV)  and  restricts  the  artery.  Scale  bars  represent 50µm.

Under physiological conditions, endothelial cells are in a quiescent state, and they have  a vasodilatory function, and an anti-inflammatory, anti-thrombotic, and anti-proliferative  phenotype [40]. Fluid shear stress (FSS), the frictional force induced by blood flow to  which endothelium is continuously exposed, is a major determinant of vascular health [41].  Areas  in  arteries  exposed  to  laminar  fluid  shear  stress  (i.e.  unidirectional  flow)  are protected from atherosclerosis. Areas exposed to non-unidirectional (disturbed) or oscillatory flow are more prone to the development of atherosclerotic lesions [42, 43]. FSS  is important for endothelial homeostasis via mechanotransduction within the endothelial cell. Laminar shear stress protects the endothelial cells via mechanotransduction and the  subsequent  initiation  of  essential  signalling  pathways,  including  the  MAP2K5-MAPK7 signalling pathway, which results in the expression of the transcription factors Krüppel-like factor-2 and 4 (KLF2 and KLF4) [44, 45]. KLF2 and KLF4 drive the expression of downstream atheroprotective genes such as endothelial nitric oxide synthase (eNOS) [46]. Contrary, upon non-unidirectional or oscillatory shear stress mechanotransduction and signalling are lost. Subsequent endothelial homeostasis is  disturbed, resulting in endothelial cell dysfunction. Endothelial dysfunction in these atherosusceptible regions is characterized by a reduced production of NO leading to impaired vasorelaxation and platelet adhesion [7], the expression of pro-inflammatory  genes, increased adhesiveness for inflammatory cells that consequently migrate into  the arterial wall, increased permeability to plasma particles such as ox-LDL, and a proliferative  phenotype.  Additionally,  endothelial  cells  can  obtain  a  fibro-proliferative  phenotype via the process of endothelial-to-mesenchymal transition (EndMT) [42, 47].

(8)

EndMT is a process wherein endothelial cells lose their endothelial-specific phenotype,  including  the  expression  of  endothelial  cell-specific  markers  such  as  CD31  and  VE-cadherin.  Endothelial  cells  conversely  acquire  a  mesenchymal  phenotype,  including  the expression of mesenchymal markers such as alpha smooth muscle actin (αSMA), smooth muscle protein 22 alpha (SM22α) and calponin, and start to display contractile behaviour. Additionally, EndMT is associated with the increased production of extracellular matrix proteins [48, 49]. EndMT is important during embryonic cardiac valve formation [50], but in adulthood EndMT contributes to the development of various fibrotic diseases [51, 52]. In atherosclerosis, EndMT has been shown to play a pivotal  role in the formation of neointimal lesions [53]. Notably, our lab previously established a pivotal role for FSS-responsive MAPK7 signalling in the inhibition of EndMT [54].

MOLECULAR MECHANISMS INVOLVED IN ENDOTHELIAL HOMEOSTASIS: REGULATION OF THE ENDOTHELIAL TRANSCRIPTOME

A quiescent endothelial phenotype is secured by the tight regulation of the endothelial  transcriptome, i.e. the full array of RNA transcripts produced [55-57]. The transcriptome is  regulated  at  multiple  levels,  such  as  activation  of  specific  signal-transduction  pathways, binding of transcription factors, chromatin packaging of the DNA, and post-transcriptional regulation via microRNAs [58]. Dysregulation of any of these aforementioned factors might drastically change the endothelial transcriptome and culminate in endothelial dysfunction and concurrently the pathogenesis of atherosclerosis and glomerulosclerosis [59]. In this thesis, we focussed on three players of regulation of the transcriptome, i.e. epigenetic processes including histone modifications, binding of  transcription factors, and microRNAs, which are further detailed below.

Nuclear DNA is wrapped around an octamer of histones, consisting of two molecules of each of the four histone proteins (H2A, H2B, H3 and H4), and a linker histone (H1). The DNA and histones together form the nucleosome, the basic building block of chromatin. All nucleosomes together are called chromatin. Histone proteins contain flexible tails that  protrude from the nucleosome and are prone to a variety of post-translational modifications,  called  histone  modifications.  The  best  characterized  histone  modifications  involve  methylation,  acetylation,  and  phosphorylation.  Histone  modifications  drive  epigenetic  processes and dictate the conformation of chromatin, and thereby the accessibility of the DNA for proteins, including transcription factors. Consequently, histone modifications  modulate the initiation of gene transcription, but without altering the DNA sequence of the  respective gene [60]. Histone modifications result either in accessible chromatin (“open”  euchromatin) or not accessible chromatin (“condensed” or “closed” heterochromatin) for  transcription-associated proteins in order to facilitate or prevent gene expression [61].

(9)

Histone modifications are regulated by enzymes that place or remove histone marks.  For example, histone acetyltransferases and histone deacetyl transferases (HDAC) can respectively add and remove acetyl-modifications from histone tails. Acetylation of  histones generally results in euchromatin formation and thus enhances gene expression. Histone methyltransferases and demethylases add and remove methyl groups on histone tail residues, respectively. Methylation of lysine residues of histone tails can either enhance or inhibit gene transcriptional activity, depending on the position of the  modified  residue  within  the  histone  tail  [62].  Polycomb  proteins  form  chromatin-modifying complexes and are important epigenetic regulators. Two main families of Polycomb complexes exist, called Polycomb Repressive Complex 1 (PRC1) and PRC2. PRC2 contains the catalytic subunit Enhancer of Zeste Homolog 2 (EZH2), a highly conserved methyltransferase. EZH2 methylates histone 3 on lysine 27 (H3K27me3). H3K27me3 associates with gene repression [63, 64]. EZH2 is previously identified as  essential player in developmental processes, and has recently also been shown to play an important role in endothelial homeostasis. EZH2 is a modulator of a number of endothelial cell functions, such as endothelial-leukocyte interactions and angiogenesis [65, 66] (fig. 5). 

Figure 5. Schematic representation of EZH2-mediated H3K27me3 and concomitant gene repression in endothelial cells. In loosely packed chromatin (euchromatin), genes that are essential for endothelial cells, are

accessible for transcription. Enhancer of Zeste Homolog 2 (EZH2) methylates lysine 27 on histone 3 (H3K27me3) which leads to densely packed chromatin (heterochromatin) thereby preventing transcription. Histone modifications are reversible and H3K27me3 can be removed by Jumonji demethylase 3 (JMJD3) or Ubiquitously  Transcribed X Chromosome Tetratricopeptide Repeat Protein (UTX).

(10)

Next to histone modifications, epigenetic modifications also include DNA methylation.  DNA methylation is in general associated with gene repression due to changes in the biophysical characteristics of the DNA [61, 67].

When the chromatin has an open structure (euchromatin), transcription factors can bind to a transcription factor binding site in the promoter region of a gene [68]. Once a transcription factor is bound to this specific site in the DNA, a co-activator complex is  recruited and RNA polymerase II can start gene transcription (fig. 6) [69]. 

However, the presence of euchromatin does not necessarily imply that transcription occurs. Instead of the recruitment of co-activator complexes, also co-repressor complexes can be recruited to DNA-bound transcription factors [70]. Co-repressor complexes, at their turn, recruit HDACs, which remove acetyl groups from histone tails leading to a more compact structure of the chromatin and a reduced accessibility of the DNA, ultimately leading to gene repression [71].

Once a gene is transcribed into messenger RNA (mRNA), the mRNA transcript can be translated into the encoded protein on the ribosome complex (polysome) in the cytoplasm. However, translation can be inhibited by post-translational regulation. More specifically,  a  mRNA  molecule  can  be  degraded  prior  to  translation,  a  process  that  is initiated by the binding of microRNAs (miRNAs) to the mRNA transcript. MiRNAs are small single-stranded non-coding RNAs with an average length of 22 nucleotides. MiRNAs mostly interact with the 3’ UTR of a target mRNA to induce translational repression by inhibition of ribosome binding. Multiple different mRNA transcripts can be targeted by one miRNA sequence, as long as the miRNA binding sequence matches  with the 3’ UTR of the mRNAs [72-74] (fig. 6).

(11)

Figure 6. Three levels of regulation of the transcriptome: histone modifications, binding of transcription factors, and microRNAs. [1] The first level of regulation of the transcriptome is formed by histone modifications. 

Histone modifications stably alter the confirmation of the chromatin, and thereby the accessibility of the DNA  for transcription-associated proteins. Repressive histone modifications (marks) reduce the accessibility of the  chromatin, whereas activating marks lead to an open chromatin structure (euchromatin). [2] In an open chromatin structure, transcription factors can bind the DNA and recruit co-activators and RNA polymerase II to initiate transcription of the DNA into mRNA. To inhibit transcription, transcription factors recruit co-repressors, which at their turn recruit histone deacetylases (HDACs), to remove activating acetyl groups on the histones leading to a closed chromatin structure (heterochromatin) and transcriptional repression (indicated by the lock). Binding of transcription factors leading to transcriptional activation or repression is a second level of regulation of the transcriptome. [3] In case mRNA is transcribed from the DNA, a third layer of regulation exists. The mRNA can be translated into protein in the ribosome, or degraded by microRNAs (miRNAs) to inhibit protein expression.

SCOPE AND OUTLINE OF THIS THESIS

Significant  advancements  have  been  made  in  the  elucidation  of  the  pathogenetic  pathways underlying atherosclerosis and glomerulosclerosis. However, there are still no definite preventative or curative treatments available for these pathologies. To halt  the development and progression of glomerulosclerosis and atherosclerosis, a deeper understanding of the pathogenetic mechanisms involved as well as the identification  of novel therapeutic targets is needed. Dysfunction of endothelial cells is a critical initiating process in both the pathogenesis of glomerulosclerosis and atherosclerosis. Differential gene and protein expression patterns lie at the basis of endothelial dysfunction and the concomitant phenotypic changes of endothelial cells. Aberrant

(12)

epigenetic, transcriptional, and translational regulation of genes therefore underlies the development of endothelial dysfunction during pathogenesis. The overall aim of this thesis is to identify epigenetic, transcriptional and translational mechanisms involved in the modulation of the endothelial transcriptome underlying endothelial dysfunction in glomerulosclerosis and atherosclerosis.

Glomerular endothelial cell (GEnC) dysfunction is critical in the development of glomerulosclerosis and comprises multiple facets. In chapter 2 we postulate that GEnC dysfunction is a pivotal and early factor in the development of glomerulosclerosis and at the basis of the podocyte dysfunction and mesangial expansion, which is observed later during disease progression. We review the importance of the cross-talk between GEnCs and podocytes, and GEnCs and mesangial cells, and further describe that GEnC dysfunction comprises multiple facets, precedes podocyte damage, and is sufficient for the development of proteinuria and glomerulosclerosis in Focal Segmental  Glomerulosclerosis (FSGS) and diabetic nephropathy (DN).

Endothelial cell function is hampered by aberrant changes to the endothelial transcriptome, some of which involve the epigenetic silencing of genes. The glomerular endothelial glycocalyx is reduced in glomerular sclerotic diseases, including DN. In

chapter 3, we investigated whether the expression of the histone methyltransferase

Enhancer  of  Zeste  Homolog  2  (EZH2)  and  its  concomitant  histone  modification  H3K27me3 are changed in GEnCs, and associate with a reduced glomerular endothelial glycocalyx in experimental DN in mice. In vitro, we investigated whether, and if so, how EZH2 reduces the endothelial glycocalyx. Furthermore, in vitro and in vivo findings are  translated to the clinical setting by studying renal biopsies obtained from DN patients. In chapter 3, it is shown that EZH2-mediated H3K27me3 is increased in human and experimental DN and associated with a compromised endothelial glycocalyx. In chapter

4, it is evaluated whether downregulation of (glomerular) endothelial EZH2, i.e. reduced

H3K27me3 activity, can be achieved in vivo, and if so whether this results in ameliorated DN. To decrease EZH2 and H3K27me3, an activated endothelial cell-specific siRNA-based targeting approach is used in which cationic liposomes, called SAINT-O-Somes (SOS), containing siRNA against EZH2 are administered to diabetic BTBRob/ob mice. The

delivery of the siRNA-encapsulating SOS to GEnCs, and the decrease of EZH2 and H3K27me3 by the encapsulated siRNA are assessed. Proteinuria, plasma creatinine, the endothelial glycocalyx, and renal histology are evaluated.

(13)

The heparan sulphate cleaving enzyme heparanase (HPSE) is involved in the pathogenesis of glomerular sclerotic diseases. Aberrant transcriptional regulation of the HPSE gene could be involved in an increased expression of HPSE, culminating in glomerular damage. In chapter 5, we investigated in GEnC and podocytes whether an increased expression of HPSE results from decreased transcriptional inhibition of the HPSE gene by the transcription factor peroxisome proliferator-activated receptor gamma (PPARɣ). It is investigated whether, and if so, how PPARɣ agonists reduce the expression of HPSE at the transcriptional level, and modulate glomerular disease development in adriamycin-induced glomerulopathy, an experimental model for FSGS. Unidirectional flow is a major determinant for endothelial health and activates Mitogen-Activated Protein Kinase 7 (MAPK7) signalling, which suppresses endothelial dysfunction and mesenchymal transition (i.e. EndMT). Conversely, loss of MAPK7 signalling leads to endothelial dysfunction and EndMT. Currently, it is unknown how MAPK7 activity is regulated in coronary artery disease. The histone methyltransferase EZH2, responsible for H3K27 methylation (H3K27me3), silences gene expression and plays a pivotal role in endothelial dysfunction. It was previously found that uniform flow  reduces the expression of EZH2, whereas repression of EZH2 reciprocally activates MAPK7 signalling. In chapter 6, the reciprocal cross-talk between MAPK7 and EZH2 in endothelial cells and the process of EndMT is studied. To translate in vitro findings to  the clinical setting, it is investigated whether the balance between MAPK7 and EZH2 is disturbed in human coronary artery stenosis.

In chapter 7, the findings of the studies presented in this thesis are summarized, the  translational aspects are discussed and directions of future research are proposed.

(14)

REFERENCES

1. Martinez-Lemus, L.A., The dynamic structure of arterioles. Basic Clin Pharmacol Toxicol, 2012. 110(1): p. 5-11.

2. Jaffe, E.A., Cell biology of endothelial cells. Hum Pathol, 1987. 18(3): p. 234-9.

3. Cahill, P.A. and E.M. Redmond, Vascular endothelium - Gatekeeper of vessel health. Atherosclerosis, 2016. 248: p. 97-109.

4.  Vita, J.A., Endothelial function. Circulation, 2011. 124(25): p. e906-12.

5. Fu, J., et al., Glomerular endothelial cell injury and cross talk in diabetic kidney disease. Am J Physiol Renal Physiol, 2015. 308(4): p. F287-97.

6. Jin, R.C. and J. Loscalzo, Vascular Nitric Oxide: Formation and Function. J Blood Med, 2010. 2010(1): p. 147-162.

7. Gimbrone, M.A., Jr. and G. Garcia-Cardena, Endothelial Cell Dysfunction and the Pathobiology of

Atherosclerosis. Circ Res, 2016. 118(4): p. 620-36.

8. Maezawa, Y., M. Takemoto, and K. Yokote, Cell biology of diabetic nephropathy: Roles of endothelial cells,

tubulointerstitial cells and podocytes. J Diabetes Investig, 2015. 6(1): p. 3-15.

9. Cheng, H. and R.C. Harris, Renal endothelial dysfunction in diabetic nephropathy. Cardiovasc Hematol Disord Drug Targets, 2014. 14(1): p. 22-33.

10. Morita, M., et al., Glomerular endothelial cell injury and focal segmental glomerulosclerosis lesion in

idiopathic membranous nephropathy. PLoS One, 2015. 10(4): p. e0116700.

11. Daehn, I., et al., Endothelial mitochondrial oxidative stress determines podocyte depletion in segmental

glomerulosclerosis. J Clin Invest, 2014. 124(4): p. 1608-21.

12. Sun, Y.B., et al., Glomerular endothelial cell injury and damage precedes that of podocytes in

adriamycin-induced nephropathy. PLoS One, 2013. 8(1): p. e55027.

13. Taneda, S., et al., Podocyte and endothelial injury in focal segmental glomerulosclerosis: an ultrastructural

analysis. Virchows Arch, 2015. 467(4): p. 449-58.

14. Weil, E.J., et al., Podocyte detachment and reduced glomerular capillary endothelial fenestration promote

kidney disease in type 2 diabetic nephropathy. Kidney Int, 2012. 82(9): p. 1010-7.

15. Boels, M.G., et al., Atrasentan Reduces Albuminuria by Restoring the Glomerular Endothelial Glycocalyx

Barrier in Diabetic Nephropathy. Diabetes, 2016. 65(8): p. 2429-39.

16. Toth-Manikowski, S. and M.G. Atta, Diabetic Kidney Disease: Pathophysiology and Therapeutic Targets. J Diabetes Res, 2015. 2015: p. 697010.

17.  D’Agati, V.D., F.J. Kaskel, and R.J. Falk, Focal segmental glomerulosclerosis. N Engl J Med, 2011. 365(25): p. 2398-411.

18.  De Vriese, A.S., et al., Differentiating Primary, Genetic, and Secondary FSGS in Adults: A Clinicopathologic

Approach. J Am Soc Nephrol, 2018. 29(3): p. 759-774.

19. Fioretto, P. and M. Mauer, Histopathology of diabetic nephropathy. Semin Nephrol, 2007. 27(2): p. 195-207. 20. Garsen, M., et al., The role of heparanase and the endothelial glycocalyx in the development of proteinuria.

(15)

Nephrol Dial Transplant, 2014. 29(1): p. 49-55.

21. Dane, M.J., et al., A microscopic view on the renal endothelial glycocalyx. Am J Physiol Renal Physiol, 2015. 308(9): p. F956-66.

22. Reitsma, S., et al., The endothelial glycocalyx: composition, functions, and visualization.  Pflugers  Arch,  2007. 454(3): p. 345-59.

23. Singh, A., et al., Glomerular endothelial glycocalyx constitutes a barrier to protein permeability. J Am Soc Nephrol, 2007. 18(11): p. 2885-93.

24.  Friden, V., et al., The glomerular endothelial cell coat is essential for glomerular filtration. Kidney Int, 2011. 79(12): p. 1322-30.

25. Weinbaum, S., J.M. Tarbell, and E.R. Damiano, The structure and function of the endothelial glycocalyx

layer. Annu Rev Biomed Eng, 2007. 9: p. 121-67.

26. Dane, M.J., et al., Glomerular endothelial surface layer acts as a barrier against albumin filtration. Am J Pathol, 2013. 182(5): p. 1532-40.

27. Garsen, M., et al., Heparanase Is Essential for the Development of Acute Experimental Glomerulonephritis. Am J Pathol, 2016. 186(4): p. 805-15.

28. van den Born, J., et al., A monoclonal antibody against GBM heparan sulfate induces an acute selective

proteinuria in rats. Kidney Int, 1992. 41(1): p. 115-23.

29.  Kanwar, Y.S., A. Linker, and M.G. Farquhar, Increased permeability of the glomerular basement membrane

to ferritin after removal of glycosaminoglycans (heparan sulfate) by enzyme digestion. J Cell Biol, 1980.

86(2): p. 688-93.

30.  Levidiotis, V., et al., Increased expression of heparanase in puromycin aminonucleoside nephrosis. Kidney Int, 2001. 60(4): p. 1287-96.

31. Gil, N., et al., Heparanase is essential for the development of diabetic nephropathy in mice. Diabetes, 2012. 61(1): p. 208-16.

32. Ramnath, R.D., et al., Blocking matrix metalloproteinase-mediated syndecan-4 shedding restores the

endothelial glycocalyx and glomerular filtration barrier function in early diabetic kidney disease. Kidney Int,

2020. 97(5): p. 951-965.

33. Butler, M.J., et al., Aldosterone induces albuminuria via matrix metalloproteinase-dependent damage of the

endothelial glycocalyx. Kidney Int, 2019. 95(1): p. 94-107.

34. Bergheanu, S.C., M.C. Bodde, and J.W. Jukema, Pathophysiology and treatment of atherosclerosis :

Current view and future perspective on lipoprotein modification treatment. Neth Heart J, 2017. 25(4): p.

231-242.

35. Sakakura, K., et al., Pathophysiology of atherosclerosis plaque progression. Heart Lung Circ, 2013. 22(6): p. 399-411.

36. Holvoet, P., et al., LDL hypercholesterolemia is associated with accumulation of oxidized LDL, atherosclerotic

plaque growth, and compensatory vessel enlargement in coronary arteries of miniature pigs. Arterioscler

Thromb Vasc Biol, 1998. 18(3): p. 415-22.

37. Jones, D.P., H.D. True, and J. Patel, Leukocyte Trafficking in Cardiovascular Disease: Insights from

(16)

Experimental Models. Mediators Inflamm, 2017. 2017: p. 9746169.

38. Rudijanto, A., The role of vascular smooth muscle cells on the pathogenesis of atherosclerosis. Acta Med Indones, 2007. 39(2): p. 86-93.

39. Nakashima, Y., T.N. Wight, and K. Sueishi, Early atherosclerosis in humans: role of diffuse intimal thickening

and extracellular matrix proteoglycans. Cardiovasc Res, 2008. 79(1): p. 14-23.

40.  Deanfield, J.E., J.P. Halcox, and T.J. Rabelink, Endothelial function and dysfunction: testing and clinical

relevance. Circulation, 2007. 115(10): p. 1285-95.

41. Garcia-Cardena, G. and M.A. Gimbrone, Jr., Biomechanical modulation of endothelial phenotype:

implications for health and disease. Handb Exp Pharmacol, 2006(176 Pt 2): p. 79-95.

42. Krenning, G., et al., Endothelial Plasticity: Shifting Phenotypes through Force Feedback. Stem Cells Int, 2016. 2016: p. 9762959.

43. Kwak, B.R., et al., Biomechanical factors in atherosclerosis: mechanisms and clinical implications. Eur Heart J, 2014. 35(43): p. 3013-20, 3020a-3020d.

44. Ohnesorge, N., et al., Erk5 activation elicits a vasoprotective endothelial phenotype via induction of

Kruppel-like factor 4 (KLF4). J Biol Chem, 2010. 285(34): p. 26199-210.

45. Dekker, R.J., et al., KLF2 provokes a gene expression pattern that establishes functional quiescent

differentiation of the endothelium. Blood, 2006. 107(11): p. 4354-63.

46. Nayak, L., Z. Lin, and M.K. Jain, “Go with the flow”: how Kruppel-like factor 2 regulates the vasoprotective

effects of shear stress. Antioxid Redox Signal, 2011. 15(5): p. 1449-61.

47. Sitia, S., et al., From endothelial dysfunction to atherosclerosis. Autoimmun Rev, 2010. 9(12): p. 830-4. 48. Krenning, G., et al., Vascular smooth muscle cells for use in vascular tissue engineering obtained by

endothelial-to-mesenchymal transdifferentiation (EnMT) on collagen matrices. Biomaterials, 2008. 29(27):

p. 3703-11.

49. Moonen, J.R., et al., Endothelial progenitor cells give rise to pro-angiogenic smooth muscle-like progeny. Cardiovasc Res, 2010. 86(3): p. 506-15.

50. Markwald, R.R., T.P. Fitzharris, and F.J. Manasek, Structural development of endocardial cushions. Am J Anat, 1977. 148(1): p. 85-119.

51. Zeisberg, E.M., et al., Endothelial-to-mesenchymal transition contributes to cardiac fibrosis. Nat Med, 2007. 13(8): p. 952-61.

52. Zeisberg, E.M., et al., Fibroblasts in kidney fibrosis emerge via endothelial-to-mesenchymal transition. J Am Soc Nephrol, 2008. 19(12): p. 2282-7.

53. Chen, P.Y., et al., Endothelial-to-mesenchymal transition drives atherosclerosis progression. J Clin Invest, 2015. 125(12): p. 4514-28.

54. Moonen, J.R., et al., Endothelial-to-mesenchymal transition contributes to fibro-proliferative vascular

disease and is modulated by fluid shear stress. Cardiovasc Res, 2015. 108(3): p. 377-86.

55. Brooks, A.R., P.I. Lelkes, and G.M. Rubanyi, Gene expression profiling of human aortic endothelial cells

exposed to disturbed flow and steady laminar flow. Physiol Genomics, 2002. 9(1): p. 27-41.

56. Passerini, A.G., et al., Coexisting proinflammatory and antioxidative endothelial transcription profiles in a

(17)

disturbed flow region of the adult porcine aorta. Proc Natl Acad Sci U S A, 2004. 101(8): p. 2482-7.

57. Gimbrone, M.A., Jr. and G. Garcia-Cardena, Vascular endothelium, hemodynamics, and the pathobiology

of atherosclerosis. Cardiovasc Pathol, 2013. 22(1): p. 9-15.

58. Eccleston, A., F. Cesari, and M. Skipper, Transcription and epigenetics. Nature, 2013. 502(7472): p. 461. 59. Xu, Y., Transcriptional regulation of endothelial dysfunction in atherosclerosis: an epigenetic perspective. J

Biomed Res, 2014. 28(1): p. 47-52.

60. Berger, S.L., et al., An operational definition of epigenetics. Genes Dev, 2009. 23(7): p. 781-3.

61. Gibney, E.R. and C.M. Nolan, Epigenetics and gene expression. Heredity (Edinb), 2010. 105(1): p. 4-13. 62. Kouzarides, T., Chromatin modifications and their function. Cell, 2007. 128(4): p. 693-705.

63. Tan, J.Z., et al., EZH2: biology, disease, and structure-based drug discovery. Acta Pharmacol Sin, 2014. 35(2): p. 161-74.

64. Simon, J.A. and R.E. Kingston, Mechanisms of polycomb gene silencing: knowns and unknowns. Nat Rev Mol Cell Biol, 2009. 10(10): p. 697-708.

65. Maleszewska, M., et al., The decrease in histone methyltransferase EZH2 in response to fluid shear stress

alters endothelial gene expression and promotes quiescence. Angiogenesis, 2016. 19(1): p. 9-24.

66. Dreger, H., et al., Epigenetic regulation of cell adhesion and communication by enhancer of zeste homolog

2 in human endothelial cells. Hypertension, 2012. 60(5): p. 1176-83.

67. Lu, Z., N. Liu, and F. Wang, Epigenetic Regulations in Diabetic Nephropathy. J Diabetes Res, 2017. 2017: p. 7805058.

68. Cheng, C., et al., Understanding transcriptional regulation by integrative analysis of transcription factor

binding data. Genome Res, 2012. 22(9): p. 1658-67.

69. Giraud, S., et al., Functional interaction of STAT3 transcription factor with the coactivator NcoA/SRC1a. J Biol Chem, 2002. 277(10): p. 8004-11.

70. Yu, C., et al., The nuclear receptor corepressors NCoR and SMRT decrease peroxisome

proliferator-activated receptor gamma transcriptional activity and repress 3T3-L1 adipogenesis. J Biol Chem, 2005.

280(14): p. 13600-5.

71. Lazar, M.A., Nuclear receptor corepressors. Nucl Recept Signal, 2003. 1: p. e001.

72. Felekkis, K., et al., microRNAs: a newly described class of encoded molecules that play a role in health and

disease. Hippokratia, 2010. 14(4): p. 236-40.

73. Bartel, D.P., MicroRNAs: target recognition and regulatory functions. Cell, 2009. 136(2): p. 215-33. 74. O’Brien, J., et al., Overview of MicroRNA Biogenesis, Mechanisms of Actions, and Circulation. Front

Endocrinol (Lausanne), 2018. 9: p. 402.

(18)
(19)

Referenties

GERELATEERDE DOCUMENTEN

Modulation of vascular remodeling : a role for the immune system, growth factors, and transcriptional regulation.. Retrieved

Revascularization by arterio- and angiogenesis requires vessel growth and vessel remodeling which requires complex involvement of growth factors, cytokines, and multiple

Blood flow recovery after a single femoral artery (distal anatomical level) electro-coagulation (n=3, green line) or a single iliac artery (proximal anatomical

Laser-doppler paw perfusion recovery was significantly decreased throughout the observation period of 4 weeks following femoral artery occlusion in

In the present study we expand these observations and demonstrate that the differences in Natural Killer cell gene locus between C57BL/6 and BALB/c mice can explain

Methods and Results: C57BL/6, BALB/c and BALB.B6-CMV1 r (CMV1 r ) mice, of which the latter is congenic for the C57BL/6 NKC, were used in two different models of vascular

Marked morphological features of skeletal muscle, characteristic for chronic ischemia, were observed in muscle biopsies derived from distal levels of the limb (most ischemic

A, Quantification of total surface (in µm 2 ) covered by endothelial outgrowth of aortic explants. B, Representative microscopic images of endothelial outgrowth from aortic