The role of endothelial cells in the vasculopathy of systemic sclerosis: A systematic review

The role of endothelial cells in the vasculopathy of systemic sclerosis: A systematic review

    The role of endothelial cells in the vasculopathy of systemic sclerosis: A systematic review Y. Mostmans, M. Cutolo, C. Giddelo, S. D...

1MB Sizes 0 Downloads 38 Views

    The role of endothelial cells in the vasculopathy of systemic sclerosis: A systematic review Y. Mostmans, M. Cutolo, C. Giddelo, S. Decuman, K. Melsens, H. Declercq, E. Vandecasteele, F. De Keyser, O. Distler, J. Gutermuth, V. Smith PII: DOI: Reference:

S1568-9972(17)30148-9 doi:10.1016/j.autrev.2017.05.024 AUTREV 2028

To appear in:

Autoimmunity Reviews

Received date: Accepted date:

8 April 2017 13 April 2017

Please cite this article as: Mostmans Y, Cutolo M, Giddelo C, Decuman S, Melsens K, Declercq H, Vandecasteele E, De Keyser F, Distler O, Gutermuth J, Smith V, The role of endothelial cells in the vasculopathy of systemic sclerosis: A systematic review, Autoimmunity Reviews (2017), doi:10.1016/j.autrev.2017.05.024

This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

ACCEPTED MANUSCRIPT The role of endothelial cells in the vasculopathy of systemic sclerosis: a systematic review SSc vasculopathy: the role of endothelial cells

SC R

IP

T

Y. Mostmans1, M. Cutolo2, C. Giddelo1, S. Decuman3, K. Melsens4, H. Declercq5, E. Vandecasteele6, F. De Keyser4, O. Distler7, J. Gutermuth1, V. Smith4

1. Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Department of

4. 5.

CE P

TE

D

6. 7.

NU

3.

Dermatology, Laarbeeklaan 101, 1090 Brussels, Belgium; Research Laboratory and Academic Unit of Clinical Rheumatology, Department of Internal Medicine, University of Genova, Genova, Italy Department of Internal Medicine, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium Department of Internal Medicine, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium; and Department of Rheumatology, Ghent University, Ghent, Belgium Department of Basic Medical Sciences, Tissue Engineering and Biomaterials Group, Ghent University, Ghent, Belgium Department of Cardiology, Ghent University Hospital, Ghent, Belgium Department of Rheumatology, University Hospital Zurich, Zurich, Switzerland

MA

2.

AC

Corresponding author:

Dr. Yora Mostmans* Department of Dermatology/Universitary Hospital Brussels (UZ Brussel) Vrije Universiteit Brussel (VUB) Laarbeeklaan 101 1090 Brussels, Belgium Tel. +32 2 477 63 55; Fax +32 2 477 63 57 Email: [email protected] *

Present address: Department of Immunology and Allergology (CIA) Centre Hospitalier Universitaire (CHU) Brugmann Université Libre de Bruxelles (ULB) Van Gehuchtenplein 4 1020 Brussels, Belgium Tel. +32 2 477 22 94; Fax +32 2 477 22 76 Email: [email protected] 1

ACCEPTED MANUSCRIPT Statement of author contribution:

T

Yora Mostmans: substantial contributions to design of the study, acquisition of data and analysis and interpretation of data, drafting of the article, final approval of the version to be published.

SC R

IP

Maurizio Cutolo: interpretation of data, critical revising of the article for important intellectual content and final approval of the version to be published.

NU

Christina Giddelo: acquisition of data and analysis and interpretation of data, critical revising of the article for important intellectual content, final approval of the version to be published.

MA

Saskia Decuman: interpretation of data, critical revising of the article for important intellectual content and final approval of the version to be published.

TE

D

Karin Melsens: interpretation of data, critical revising of the article for important intellectual content and final approval of the version to be published.

CE P

Heidi De Clercq: interpretation of data, critical revising of the article for important intellectual content and final approval of the version to be published.

AC

Els Vandecasteele: interpretation of data, critical revising of the article for important intellectual content and final approval of the version to be published. Filip De Keyser: interpretation of data, critical revising of the article for important intellectual content and final approval of the version to be published. Oliver Distler: interpretation of data, critical revising of the article for important intellectual content and final approval of the version to be published. Jan Gutermuth: analysis and interpretation of data, critical revising of the article for important intellectual content and final approval of the version to be published. Vanessa Smith: substantial contributions to design of the study, acquisition of data and analysis and interpretation of data, drafting of the article, final approval of the version to be published. 2

ACCEPTED MANUSCRIPT Statement of conflict of interest:

AC

CE P

TE

D

MA

NU

SC R

IP

T

Yora Mostmans: no conflicts of interest to declare Maurizio Cutolo: no conflicts of interest to declare Christina Giddelo: no conflicts of interest to declare Saskia Decuman: no conflicts of interest to declare Karin Melsens: no conflicts of interest to declare Heidi De Clercq: no conflicts of interest to declare Els Vandecasteele: no conflicts of interest to declare Filip De Keyser: no conflicts of interest to declare Oliver Distler: has/had consultancy relationship and/or has received research funding from 4 D Science, Actelion, Active Biotec, Bayer, Biogen Idec, Boehringer Ingelheim Pharma, BMS, ChemomAb, EpiPharm, Ergonex, espeRare foundation, GSK,Roche-Genentech, Inventiva, Lilly, medac, MedImmune, Mitsubishi Tanabe, Pharmacyclics, Pfizer, Sanofi, Serodapharm and Sinoxa in the area of potential treatments of scleroderma and its complications. He has a patent on mir-29 for the treatment of systemic sclerosis licensed. Jan Gutermuth: no conflicts of interest to declare Vanessa Smith: no conflicts of interest to declare

3

ACCEPTED MANUSCRIPT ABSTRACT

IP

T

Introduction: Systemic sclerosis (SSc) is an autoimmune connective tissue disorder characterized by fibroproliferative vasculopathy, immunological abnormalities and progressive fibrosis of multiple organs including the skin. In this study, all English speaking articles concerning the role of endothelial cells (ECs) in SSc vasculopathy and representing biomarkers are systematically reviewed and categorized according to endothelial cell (EC) (dys)function in SSc.

NU

SC R

Methods: A sensitive search on behalf of the EULAR study group on microcirculation in Rheumatic Diseases was developed in Pubmed, The Cochrane Library and Web of Science to identify articles on SSc vasculopathy and the role of ECs using the following Mesh terms: (systemic sclerosis OR scleroderma) AND pathogenesis AND (endothelial cells OR marker). All selected papers were read and discussed by two independent reviewers. The selection process was based on title, abstract and full text level. Additionally, both reviewers further searched the reference lists of the articles selected for reading on full text level for supplementary papers. These additional articles went through the same selection process.

CE P

TE

D

MA

Results: In total 193 resulting articles were selected and the identified biomarkers were categorized according to description of EC (dys)function in SSc. The most representing and reliable biomarkers described by the selected articles were adhesion molecules for EC activation, anti-endothelial cell antibodies for EC apoptosis, vascular endothelial growth factor (VEGF), its receptor VEGFR-2 and endostatin for disturbed angiogenesis, endothelial progenitors cells for defective vasculogenesis, endothelin-1 for disturbed vascular tone control, Von Willebrand factor for coagulopathy and interleukin (IL)-33 for EC- immune system communication. Emerging, relatively new discovered biomarkers described in the selected articles, are VEGF165b, IL-17A and the adipocytokines. Finally, myofibroblasts involved in tissue fibrosis in SSc can derive from ECs or epithelial cells through a process known as endothelial-to-mesenchymal transition.

AC

Conclusion: This systematic review emphasizes the growing evidence that SSc is primarily a vascular disease where EC dysfunction is present and prominent in different aspects of cell survival (activation and apoptosis), angiogenesis and vasculogenesis and where disturbed interactions between ECs and various other cells contribute to SSc vasculopathy.

KEYWORDS biomarker, endothelial cells, systemic sclerosis, systematic review, vasculopathy, EULAR study group on microcirculation in Rheumatic Diseases 4

ACCEPTED MANUSCRIPT Abbreviations Ab: antibodies ACAs: anti-centromere antibodies

IP

T

ACE: angiotensin converting enzyme ADCC: antibody dependent cell mediated cytotoxicity

SC R

ADMA: asymmetric dimethylarginine ADP: adenosine diphosphate

NU

AECA: anti-endothelial cell antibodies Ag: antigen

MA

AIF-1: allograft inflammatory factor 1 Ang I: angiotensin I

D

Ang II: angiotensin II

Ang-2: angiopoietin 2

TE

Ang-1: angiopoietin 1

CE P

Angptl3: angiopoietin-like protein 3 Anti-topo I: anti-topoisomerase ATIII: antithrombin III

AC

BALF: broncho-alveolar lavage fluid BCGF: B cell growth factor bFGF: basic fibroblast growth factor-2 BM: bone marrow BM-MSCs: bone marrow mesenchymal stem cells BMPRII: bone morphogenetic protein receptor II CACs: circulating angiogenic cells cAMP: cyclic adenosine monophosphate CCL: chemokine ligand CCN-1: cysteine-rich 61 matrix protein

5

ACCEPTED MANUSCRIPT CD: cluster of differentiation CD44R: cluster of differentiation 44 receptor CEC: circulating endothelial cells

IP

T

CFU: colony-forming unit cGMP: cyclic guanosine monophosphate

SC R

CIC: circulating immune complexes CKO: knock out

NU

CRP: C-reactive protein CTSB: cathepsin B

CTSV: cathepsin V

D

CXCL: chemokine (C-X-C motif) ligand

MA

CTSL: cathepsin L

TE

CX3CR1: fractalkine receptor DAF: decay accelerating factor

DD: d-dimers

CE P

dcSSc: diffuse cutaneous systemic sclerosis

DLCO: diffusing capacity for carbon monoxide

AC

DMVEC: dermal microvascular endothelial cells DRC3f: complement C3f-des-arginine DS: dermatansulphate DU: digital ulcers EC: endothelial cell ECA: endothelial cytotoxic activity ECs: endothelial cells EGFL7: epidermal growth factor-like protein 7 EMP: endothelial cell-derived microparticles ENA-78: epithelial-neutrophil activating peptide-78

6

ACCEPTED MANUSCRIPT EndoMT: endothelial-to-mesenchymal transition eNOS: endothelial nitric oxide synthase EPC: endothelial progenitor cells

IP

T

ERK: extracellular signal-regulated kinase EScSG: the European Scleroderma Study Group

SC R

ESR: erythrocyte sedimentation rate

EUSTAR: The European Scleroderma Trials and Research group

NU

F-AnxV(-): Annexin V non-binding

FASSc: fibrosing alveolitis associated with systemic sclerosis

MA

FEV: forced expiratory volume FKN: fractalkine

D

FNG: fibrinogen

Fra-2: fos-related antigen-2

CE P

FVC: forced vital capacity

TE

Fli1: friend leukemia integration factor 1

F2IP-M: tetranor-dicarboxylic acid metabolite of F2-isoprostanes F1+2: prothrombin fragments 1+2

AC

fVIII/vWF Ag: factor VIII/von Willebrand factor antigen Gal3: galectin 3

GLUT-1: glucose transporter 1 GPA: granulomatosis with polyangiitis GRO-α : growth-regulated oncogene-alpha GVHD: graft-versus-host disease HC: healthy controls HDEC: human dermal endothelial cells HMVEC: human microvascular endothelial cells HGF: hepatocyte growth factor

7

ACCEPTED MANUSCRIPT HRC: hypertensive renal crisis HUVEC: human umbilical vein endothelial cells ICAM: intercellular adhesion molecule

IP

T

IF: immunofluorescence IFI16: gamma-interferon-inducible protein IFI-16

SC R

IFN-γ: interferon gamma Ig: immunoglobulin

NU

IL: interleukin IP: interstitial pneumonia

MA

iNOS: inducible NO synthase JAM: junctional adhesion molecule

D

KDR: kinase insert domain receptor

TE

KLK: human tissue kallikrein

LMP: leukocyte-derived microparticle

CE P

Lp(a): lipoprotein (a)

MAC: membrane attack complex of complement MC: mast cells

AC

MCP: membrane cofactor protein

MDMP: monocyte-derived microparticle Mesh: medical subject heading MIF: macrophage migration inhibitory factor MMP: matrix metalloproteinases MMT: mesenchymal-to-mesenchymal transition MPs: microparticles mRNA: messenger ribonucleic acid MRSS: modified Rodnan skin thickness score MVD: microvessel density

8

ACCEPTED MANUSCRIPT MVEC: microvascular endothelial cells Nb: number NEP: neutral endopeptidase

IP

T

NIH: national institute of health NO: nitric oxide

SC R

NOS: nitric oxide synthase NOx: total nitrate and nitrite

NU

NVC: nail fold videocapillaroscopy OA: osteoarthritis

MA

OPG: osteoprotegerin OSM: oncostatin M

TE

PAP: pulmonary artery pressure

D

PAI: plasminogen activator inhibitor

PCs: pericytes

CE P

PDGF (-BB): platelet derived growth factor (-BB) PDMP: platelet-derived microparticle PEDF: pigment epithelium-derived factor

AC

PECAM: platelet endothelial cell adhesion molecule PH: pulmonary hypertension PI3K: phosphatidylinositol-4,5-bisphosphate 3-kinase PL: phospholipids PlGF: placental growth factor PRP: primary raynaud phenomenon PS: phosphatidylserine PSS: progressive systemic sclerosis PTX3: pentraxin 3 RA: rheumatoid arthritis

9

ACCEPTED MANUSCRIPT RANTES: regulated on activation normally T cell expressed and secreted RAS: renin-angiotensin system RBP4: retinol binding protein-4

IP

T

RF: rheumatoid factor RhoA: GTPase Ras homolog gene family-member Ab

SC R

RNAse: ribonuclease ROCKS: rho-associated creatine kinases

NU

ROS: reactive oxygen species RP: raynaud phenomenon

RVSP: right ventricular systolic pressure

MA

RT-PCR: reverse-transcriptase polymerase chain reaction

D

sCD40L: soluble cluster of differentiation-40 ligand

TE

SCF: stem cell factor SDF-1: stromal cell-derived factor 1

CE P

sENG: soluble endoglin sFKN: soluble fractalkine

sGP130: soluble glycoprotein 130

AC

sIL: soluble interleukin

sIL-2R: soluble IL-2 receptor sIL-6R: soluble IL-6 receptor siRNA: small interfering RNA SLE: systemic lupus erythematosus α-SMA: alpha smooth muscle actin SM22α: transgelin SNAIL-1: transcriptional repressor zinc finger protein SPARC: secreted protein, acidic and rich in cysteine SRC: scleroderma renal crisis

10

ACCEPTED MANUSCRIPT SSc: systemic sclerosis sTie1: soluble tyrosine kinase 1 sTie2: soluble tyrosine kinase 2

IP

T

ST2: IL-1 receptor-related protein sVEGF: soluble vascular endothelial growth factor

SC R

TAT: thrombin-antithrombin TBARS: thiobarbituric acid reactive substances

TGF-β: tumor growth factor beta

MA

TIMP: tissue inhibitor of metalloproteinases

NU

β-TG: beta-thromboglobulin

TLR: toll-like receptors

D

TNF-α: tumor necrosis factor alpha

TE

TM: thrombomodulin

tPA: tissue type plasminogen activator

CE P

TRAIL: tumor necrosis factor related apoptosis-inducing ligand TSLP: thymic stromal lymphopoietin TSP-1: thrombospondin 1

AC

TXAR: thromboxane A2 receptor

uPAR: urokinase-type plasminogen activator receptor U-II: urotensin-II VA: alveolar volume VC: vital capacity VCAM: vascular cell adhesion molecule VE-cadherin: vascular endothelial cadherin VEGF: vascular endothelial growth factor VEGFR: vascular endothelial growth factor receptor VSMC: vascular smooth muscle cells

11

ACCEPTED MANUSCRIPT

AC

CE P

TE

D

MA

NU

SC R

IP

T

vWF: von willebrand factor

12

ACCEPTED MANUSCRIPT The role of endothelial cells in the vasculopathy of systemic sclerosis: a systematic review 1. INTRODUCTION

NU

SC R

IP

T

Systemic sclerosis (SSc) is an autoimmune connective tissue disorder characterized by fibroproliferative vasculopathy, immunological abnormalities and progressive fibrosis of multiple organs such as skin and lung (1). Dysregulation of endothelial cell (EC) function within the vascular wall plays an important role in vascular remodeling associated with the fibroproliferative vasculopathy observed in SSc. Endothelial cell injury is proposed as a crucial initiating event leading to vascular remodeling with intimal proliferation of arterioles and capillary breakdown and finally, blood vessel occlusion (2-4). Ongoing research continues to focus on the role of endothelial cells (ECs) in SSc vasculopathy and on identification of related biomarkers. This study, performed on behalf of the EULAR study group on microcirculation in Rheumatic Diseases, gives an overview on the large body of data of current research in this domain, obtained after systematically reviewing the literature.

MA

2. METHODS 2.1 Search strategy

CE P

TE

D

A structured search on Pubmed, The Cochrane Library and Web of Science was performed without limitation on publication date to identify articles on SSc vasculopathy and the role of ECs. The Medical subject heading (Mesh) term for “systemic sclerosis” and its synonym “scleroderma” were used in combination with other groups of search terms: (systemic sclerosis OR scleroderma) AND pathogenesis AND (endothelial cells OR marker). The structured search was last updated on the 24th of November 2016. Details on search strategy are shown in Supplementary file 1. 2.2 Searching process

AC

All articles were screened by two independent reviewers (YM and CG). This selection process was based on title, abstract and full text level. All studies elaborating on the role of ECs in the pathogenesis of SSc vasculopathy were included in the systematic review. All study designs with the exception of reviews, editorials and case studies were included. Languages other than English were excluded. Titles and abstracts selected by either one of the reviewers were included for further screening. The final articles were withheld after reading and judgement of the full text. When different opinions existed among the two reviewers on full text level, consensus was reached. Additionally, both reviewers further searched the reference lists of the articles selected for reading on full text level for supplementary papers. These additional articles went through the same selection process performed by the same two reviewers. Supplementary file 2 depicts the flow of the searching process performed in this systematic review (5). The majority of articles were observational studies of which most were evaluated to be of fair quality using the quality assessment tool for observational studies adapted from the national institute of health (NIH) (supplementary file 3)(6). 3. RESULTS Of 2442 articles screened (on title, abstract and full text level) as potentially relevant to the topic of this study, 127 articles were included. From these articles, reviewers further searched the reference 13

ACCEPTED MANUSCRIPT

NU

SC R

IP

T

lists for more relevant articles not present in the database search, and identified 115 articles for additional screening. The flow of the searching and selection process is shown in supplementary file 2. Supplementary file 4 summarizes all 193 selected manuscripts on biomarkers reflecting EC dysfunction described in SSc vasculopathy. The majority of articles were observational studies, of which most were evaluated to be of fair quality according to the quality assessment tool for observational studies adapted from the national institute of health (NIH) (supplementary file 3)(6). The 193 selected articles were categorized according to description of EC (dys) function in SSc, as shown in Table 1 and Figure 1: EC-activation, EC-apoptosis, disturbed angiogenesis, endothelial-tomesenchymal transition (EndoMT), disturbed vasculogenesis, defective vascular tone control, coagulation cascade impairment, immune dysregulation, mast cell dysregulation, dysregulation of the vascular system through pericytes/ECs crosstalk, fibrosis and dysregulation of adipocytokines. Since many of these functional pathways interact, some markers were therefore categorized in more than one function group. 3.1 Markers of EC-activation

AC

CE P

TE

D

MA

In its early stages, SSc is characterized by EC injury and perivascular mononuclear cell infiltration with an accumulation of T and B lymphocytes and monocytes in the affected tissues (7). Interaction between these leukocytes and ECs is of major importance in the induction of EC permeability contributing to SSc vasculopathy. These interactions are highly dependent on the expression and function of cell adhesion molecules for the maintenance of transendothelial leukocyte migration. In total 38 articles concerning markers of EC-activation were selected (Table 1). The majority of selected articles covered adhesion molecules as biomarker of EC-activation: E-selectin (8-19), intercellular adhesion molecule (ICAM)(8, 9, 11, 12, 15-17, 20-24), junctional adhesion molecule (JAM)(25-27), platelet endothelial cell adhesion molecule (PECAM)(28), P-selectin (8, 9, 14, 15, 19, 29) and vascular cell adhesion molecule (VCAM)(9-12, 15, 17, 29-31). Nevertheless also other biomarkers in EC activation are described but less frequent: allograft inflammatory factor 1 (AIF-1)(32), asymetric dimethylarginine (ADMA)(33), fractalkine (FKN)(34, 35), fractalkine receptor (CX3CR1)(34), soluble endoglin (sENG)(31, 33, 36), Β1/β2/β4 integrins (16), von Willebrand factor (vWF) (37-44), counter adhesive proteins thrombospondin 1 (TSP-1) and secreted protein, acidic and rich in cysteine (SPARC)(13). The majority of discussed markers of EC activation are elevated in the sera of SSc or certain SSc subtypes (supplementary file 4). 3.2 EC-apoptosis Endothelial injury is regarded as a crucial initiating event leading to vascular remodeling with intimal proliferation of arterioles and capillary breakdown and blood vessel occlusion. EC apoptosis is recognized as an important component of the response to the injury. 29 articles investigating EC apoptosis and possible biomarkers were selected for this topic. Described biomarkers are: antiendothelial cell antibodies (AECA) (through antibody dependent cell mediated cytotoxicity (ADCC))(3, 37, 45-57), immunoglobulin (Ig) G anti-caspase-3 antibodies (Ab)(58), caspase-3 (59), gammainterferon-inducible protein (IFI)-16 Ab (60, 61), bone morphogenetic protein receptor II (BMPRII)(62), circulating endothelial cells (CEC)(31, 63), osteoprotegerin (OPG) and TNF-related apoptosis-inducing ligand (TRAIL)(64), membrane attack complex of complement (MAC)(65), collagen V (66), microparticles (MPs)(14, 67, 68) and Fos-related antigen-2 (Fra-2)(69). 14

ACCEPTED MANUSCRIPT Most markers of EC apoptosis were elevated in the sera of SSc patients (Table 2). 3.3 Disturbed angiogenesis

CE P

TE

D

MA

NU

SC R

IP

T

A loss of vasculature in SSc results in tissue hypoxia, which normally promotes angiogenesis through the production of pro-angiogenic factors. Angiogenesis implicates endothelium sprouting from preexisting ECs and involves the proliferation and migration of mature ECs. However in SSc, angiogenesis is disturbed through expression of inefficient pro-angiogenic mediators, upregulation of powerful inhibitors of angiogenesis and by alteration of transcripts involved in signal transduction pathways (70). In total 56 articles on mechanisms of dysfunctional angiogenesis in SSc were selected (Table 1). Biomarkers described in these selected articles demonstrating this disturbance, shown in Table 2, are: vascular endothelial growth factor (VEGF)(28, 31, 33, 71-82), VEGF165b (83), vascular endothelial growth factor receptor (VEGFR) 1 (74, 78, 84, 85), VEGFR 2 (72, 74, 78, 85, 86), VEGFR 3 (78, 85), platelet-derived growth factor (PDGF)(77, 87), PDGF-BB (28), basic fibroblast growth factor-2 (bFGF, also known as FGF-2 or FGF-β)(73, 77, 79, 88), hepatocyte growth factor (HGF)(28, 77, 79), soluble tyrosine kinase 1 (sTie1)(89), sTie2 (31, 90), angiopoietin (Ang)-1 (Ang-1)(91), Ang-2 (28, 31, 91), angiopoietin-like protein 3 (Angptl3)(92), placental growth factor (PlGF) (31, 77, 93, 94), endostatin (31, 73, 75, 77, 81, 82, 95), tissue inhibitor of metalloproteinase 2 (TIMP-2)(75), growth-regulated oncogene-α (GRO-α)(96), glucose transporter 1 (GLUT-1)(72), pigment epithelium-derived factor (PEDF)(76), matrix metalloproteinases (MMP)-9 (77), MMP-12 (97-100) and pro-MMP-1 (77), urokinase-type plasminogen activator receptor (uPAR)(98, 99, 101), human tissue kallikreins (KLK) 1 (102, 103), KLK9 (102), KLK11 (102), KLK 12 (102), cathepsin B (CTSB)(104), cathepsin V (CTSV)(105), cathepsin L (CTSL)(106), friend leukemia integration factor 1 (Fli1)(107-109), galectin-3 (Gal3)(110), apelin (111), CCN1 (112) and chemokines epithelial-neutrophil activating peptide (ENA-78)(15), regulated on activation normally T-cell expressed and secreted (RANTES) (15), chemokine ligand (CXCL)1 (96), CXCL4 (113), CXCL5 (108), CXCL8 (15, 28-30, 96), CXCL9 (114), CXCL10 (114, 115), CXCL12 (116), CXCL16 (114, 117) and chemokine ligands (CCL)2 (19, 30), CCL13 (118), CCL23 (119).

AC

Most articles showed upregulation of proangiogenic and/or angiostatic biomarker(s) in SSc as shown in Table 2. 3.4 Endothelial-to-mesenchymal transition (EndoMT) In the pathogenesis of SSc, an accumulation of fibroblasts and myofibroblasts occurs and production of interstitial collagens and extracellular matrix components is excessive. Numerous studies have shown that myofibroblasts involved in tissue fibrosis can derive from ECs through a process known as EndoMT (120-127). It is a nonmalignant phenomenon of cellular transdifferentiation by which ECs undergo a phenotypical conversion where they lose vascular EC markers and gain mesenchymal cell markers (120-122). Seven articles concerning this topic were selected (127-133). Most included studies (127-131, 133) are in vitro studies (mostly using mouse models) suggestive of a role for EndoMT in SSc progressive fibrosis. 3.5 Disturbed vasculogenesis Vasculogenesis involves recruitment, mobilization and in situ differentiation of endothelial progenitor cells (EPC) from the bone marrow (BM). Recent data show that the morphology of SSc BM is normal but that the number of microvessels is severely reduced despite striking increase of VEGF 15

ACCEPTED MANUSCRIPT

SC R

IP

T

(80). Furthermore the angiogenic potential of EC-like mesenchymal stromal cells was reduced after being stimulated with VEGF and stromal cell-derived factor-1 in vitro, suggesting that endothelial repair may be affected in SSc starting from the BM (134). EPC normally have the ability to develop into fully mature EC and contribute to neovascularization by targeting sites of endothelial injury (135). They represent a heterogeneous cell population originated from a single multipotent progenitor within the BM and consist of cells at different stages of maturation, ranging from early CD133+/VEGFR2+ to more mature CD34+/VEGFR2+ phenotypes (136). Impaired functioning of the EPC has been thought to be involved in the pathogenesis of SSc.

NU

In total 22 articles on the role of ECs in defective SSc vasculogenesis were selected. The most described biomarker in this selection are EPC (31, 63, 79, 80, 84, 88, 94, 116, 135, 137-147). Biomarkers other than EPC for this disturbed process, described in the selected articles, are: circulating angiogenic cells (CACs) (139), epidermal growth factor-like domain 7 (EGFL7) (148) and pentraxin 3 (PTX3) (88, 100, 146). 3.6 Defective vascular tone control and the nitric oxide (NO) paradox in SSc

AC

CE P

TE

D

MA

Endothelin-1 (ET-1) plays a prominent role in vascular tone regulation through its receptors ETA and ETB. ETA receptor predominates on vascular smooth muscle and mediates vasoconstriction, whereas the ETB receptor subtype, when present on vascular endothelium, mediates vasodilation through NO release (149-151). ET-1 is shown overexpressed in skin, lung tissue and plasma of SSc patients suggesting a role for this mediator in SSc vasculopathy (31, 38, 87, 152-158). In total 32 articles on the role of EC in vascular tone mediation were selected (Table 1). Suggested biomarkers, other than ET-1, of (direct or indirect) vascular tone dysregulation, described by the selected articles, are: urotensin-II (U-II)(159), angiotensin (Ang) I (160), Ang II (160), heptapeptide Ang (1-7)(160), angiotensin converting enzyme (ACE) (37-39, 160), 8-isoprostane (161-165), nitric oxide (NO)(11, 166-177) and inducible nitric oxide synthase (iNOS)(166). The latter is the result of ischemia caused by vascular obliteration known to occur in proliferative vasculopathy. As a result fibroblasts and ECs in skin and lung tissue produce reactive oxygen species (ROS), including NO, that selectively activates ECs or trigger fibroblasts to produce collagen (178). Synthesized ROS (i.e. superoxide anions, hydrogen peroxide, hydroxyl radicals and/or peroxynitrite) selectively activate ECs, leading to vascular inflammation (179, 180). 3.7 Coagulation cascade and complement system impairment The interaction between ECs and platelets plays an important role in vascular tone regulation in SSc. Damaged ECs release several molecular substances into the circulation that interfere with coagulation homeostasis (181). In a recent in vitro study, it is shown that activated platelets induce thymic stromal lymphopoietin (TSLP) production in human dermal microvascular ECs inducing profibrotic and EC-activating factors such as interleukin (IL)-13 (182). Abnormalities and impairment of the coagulation system, such as presence of circulating supranormal multimers of von Willebrand factor (vWF) or elevated levels of vWF, factor VIII antigen, fibrinogen (FNG), thrombomodulin (TM), tissue plasminogen activator (t-PA), plasminogen activator inhibitor 1 (PAI-1) have been described in SSc in multiple studies (19, 37-44, 87, 156, 181, 183). However whether or not impairment of fibrinolysis is present is still under debate since both depressed fibrinolytic activity (41) and a normal plasma fibrinolytic profile (40) has been reported. In total 14 articles have been selected for this topic. 16

ACCEPTED MANUSCRIPT

SC R

IP

T

The role of the complement system (CS) in the pathogenesis of SSc vasculopathy has not been exhaustively investigated. The CS classically functions as a recruiter of inflammatory cells and regulates coagulation cascade and angiogenesis by damaging EC integrity (184). On human ECs, a group of membrane-bound complement regulators, including membrane cofactor protein (MCP or CD46) and decay accelerating factor (DAF or CD55), participates in protection from activation of both alternative and classic CS. An impaired expression of these regulators is shown in SSc skin, suggesting that a defective endothelial protection might be mediated by reduced expression of the complement regulatory proteins. This could potentially lead to endothelium-bound membrane attack complex of complement (MAC or C5B-9) depositions which are known to cause EC apoptosis (65, 185). Furthermore comprehensive peptidomics analysis revealed the predominance of complement C3fdes-arginine (DRC3f), derived from C3b, in the sera of patients with SSc and the level of DRC3f was related to vascular involvement in SSc and to SSc disease activity (186).

NU

3.8 ECs and their immune signature on SSc vasculopathy

CE P

TE

D

MA

Although articles on vascular abnormalities in SSc were reviewed and selected, also the (direct or indirect) interaction between ECs with the immune system can contribute to the emergence or maintenance of SSc vasculopathy. In total 27 articles suggesting and discussing this interaction were selected. The selected immunological biomarkers, mostly cytokines, important in this vascular ECimmune system communication, are: macrophage migration inhibitory factor (MIF)(187, 188), IL-1α (189-191) and IL-1β (189, 191), IL-1 receptor-related protein (ST2)(192, 193), IL-2 (189, 194-196), soluble IL-2 receptor (21, 22, 194, 195), B cell growth factor (BCGF)(194), IL-4 (189, 197), IL-6 (18, 29, 189, 198, 199), soluble IL-6 receptor (198), oncostatin M (OSM)(198), soluble glycoprotein 130 (sGP130)(198), interferon γ (IFN-γ)(189), tumor necrosis factor alpha (TNF-α)(29, 189), IL-8 (19, 29, 30, 200), IL-10 (197), IL-13 (30, 197, 201), IL-15 (202, 203), IL-17A (204) and IL-33 (30, 191-193, 205207). All cytokines described in the selected clinical studies are shown upregulated in SSc plasma or (late stage) SSc skin as shown in supplementary file 4 and Table 2.





AC

3.9 EC interaction with other cells

Macrophages: They exist of diverse phenotypes, from "classically activated" M1 to "alternatively activated" M2 macrophages. M2 macrophages, including the subsets M2a and M2c, are considered to promote angiogenesis and tissue regeneration, while M1 macrophages are considered to be anti-angiogenic. M2a macrophages secrete the highest levels of PDGF-BB, a chemoattractant for stabilizing pericytes, and also promote anastomosis of sprouting ECs in vitro whereas M2c macrophages secrete the highest levels of MMP-9, a protease involved in vascular remodeling (208). A crosstalk between ECs and macrophages has been shown in cardiac microvascular inflammation (209). Furthermore ET-1 seems to induce the M2 phenotype in cultured human macrophages (210). Therefore similar intercellular interaction is suggested but not yet deeply investigated in SSc (211). Mast cells (MC): It is known for ECs to interact with mast cells through the production of Stem Cell Factor (SCF; c-kit ligand) to influence mast cell proliferation and differentiation (212). MC are increased in number in the lesional skin of SSc patients and are a valid source of TGF-β when degranulation occurs (213). Additionally SCF is shown to be strongly expressed on ECs in the SSc skin (214).

17

ACCEPTED MANUSCRIPT

T

4. DISCUSSION

CE P

TE

D



MA

NU

SC R



Pericytes (PCs): Among other cell types, ECs secrete PDGF-BB which recruits and induces proliferation of PCs progenitors (215). PDGF-BB serum levels are shown elevated in SSc patients (28). There is increasing evidence that PCs exhibit a phenotype similar to that of BM mesenchymal stem cells (BM-MSCs)(215). In a recent in vitro study SSc BM-MSCs are cocultured with healthy controls (HC) MVEC and cell reprogramming towards a pro-angiogenic behavior is observed (216). Fibroblasts: There is considerable evidence that EC products can modulate fibroblast properties in vivo. It is shown that SSc fibroblasts are particularly sensitive to EC-induced phenotypic modulation through IL-1 and bFGF and that damaging the EC vascular monolayer alters fibroblast responses (217). Furthermore it is shown that EC apoptosis plays a very important role in activating fibrogenic pathways through mediators that induce resistance to apoptosis in fibroblasts. These mediators in turn increase proliferation and inhibit apoptosis of EC and vascular smooth muscle cells (VSMC)(218, 219). A more recent study shows that SSc MVEC recruit and activate dermal fibroblasts by induction of a CCN2/TGF-β-dependent mesenchymal-to-mesenchymal transition this way clarifying the possible vascular connection of SSc-associated fibrosis (220). Furthermore evidence is provided that fibroblasts from SSc patients over-express MMP-12, which cleaves uPAR of normal MVEC, thus contributing to the failure of SSc-ECs to induce an efficient angiogenic program (99). Adipocytes: Recent studies have been focusing on the role of adipocytokines in the pathogenesis of SSc. Described markers that emphasize this emerging new interaction are: resistin (221), leptin (28, 221), adiponectin (221), chemerin (109) and retinol binding protein4 (RBP4)(222). Four articles have been selected concerning this interaction. Serum levels of possible biomarkers, shown in supplementary file 4, were mostly normal although correlations with the presence of digital ulcers (DU) were shown for elevated resistin and chemerin levels.

IP



AC

In this systematic review on the role of EC in SSc vasculopathy, 193 selected articles present one or more aspects of EC dysfunction (Figure 1). Multiple significantly elevated activation markers, but mostly adhesion molecules, in the sera of SSc patients underline the significant involvement of EC activation in SSc pathogenesis. Dysregulation of apoptosis and proliferation within the vascular wall plays an important role in vascular remodeling associated with fibroproliferative vasculopathy observed in SSc. Apoptosis of ECs is recognized as an important component of the response to the injury phenomenon. Multiple selected studies have shown that AECA might be key players responsible for mediating this EC injury through ADCC. It is suggested that this ADCC is induced via the Fas pathway (46, 48). Though an earlier study not only failed to show Fas receptor binding by apoptosis-inducing AECA, it also showed that purified IgG AECA from SSc patients can induce apoptosis in human umbilical vein EC (HUVEC) through a membrane phospholipid reassembly with anionic phospholipid exposure and phosphatidylserine reaching the surface from its intracellular position (47). Additionally the apoptotic phenotype of endothelial progenitor cells (EPC) was investigated for possible mechanisms inducing apoptosis. Bone marrow studies showed significant titers of AECA, correlating with the number of apoptotic progenitors and in vitro assays on normal progenitors confirmed these results through induction of apoptosis by addition of AECA+purified IgG (55). Furthermore up-regulation of adhesion 18

ACCEPTED MANUSCRIPT

T

molecule expression (E-selectin, ICAM-1 and VCAM-1) and increased leukocyte adhesion (U937 cells) following AECA binding has also been shown to be an AECA binding effect (56). Because of these contradictory underlying mechanisms (activation versus apoptosis), more studies are needed to define the pathogenic role of AECA and its various subtypes before considering it as a useful biomarker.

NU

SC R

IP

MPs are released from a variety of cells during activation and apoptosis via an exocytic budding process. Although long considered as inert debris, they are now appreciated as important mediators of cellular cross-talk regulating inflammation, coagulation, vascular functions, apoptosis and cell proliferation (223). Increased (67) as well as decreased (14) numbers of this EC apoptosis biomarker have been described in SSc. This contrast could be explained by a discrepancy in definition of examined MPs (according to size, Annexin V binding/non-binding MPs, CD144+/CD146+ EMP), isolation procedures and dissimilar use of fluorochromes and cell-specific, monoclonal antibodies. This implicates the need of a clear cut consensus on the exact characterization of MPs.

AC

CE P

TE

D

MA

Furthermore, it is shown in SSc that ECs lose their potential to participate in a normal angiogenesis process in response to EC injury. Mainly biomarkers suggestive for dysfunctional angiogenesis in relation to overexpression of pro-angiogenic mediators are described. The most suggestive ones in the selected articles are VEGF, VEGFR-2 and CXCL8. Despite elevation of VEGF in plasma and skin, adaptive angiogenesis is absent and a progressive loss of capillaries is observed (71). This paradox is further supported by the knowledge that short-time upregulation of VEGF is a strong inducer of angiogenesis, while chronic and uncontrolled overexpression of VEGF, which occurs in SSc, leads to the formation of irregularly shaped sac-like vessels with reduced blood flow in the newly formed vessels (74). However, most studies could not make the distinction between proangiogenic and antiangiogenic VEGF splice variants, which have been uncovered only recently. Indeed, although VEGF had conventionally been considered to be a family of proangiogenic mediators, the VEGF premRNA is shown to be differentially spliced in its terminal exon to form two distinct subfamilies of proteins, the so-called proangiogenic VEGFxxx isoforms and antiangiogenic VEGFxxxb isoforms (mainly VEGF165 and VEGF165b)(224). In a recent study, an increase in VEGF in SSc patients appears to be the result of a significant increase in the anti-angiogenic VEGF165b isoform instead of VEGF165. Furthermore in this study elevated circulating levels of VEGF165b in SSc patients are shown associated with early disease. Additionally by using blocking antibodies, severe impairment of in vitro angiogenesis is ameliorated in their study. Therefore this switch from pro-angiogenic to antiangiogenic VEGF isoforms may have a crucial role in the insufficient angiogenic response to chronic ischemia and needs further investigation (83). The most often described upregulated angiogenesis inhibitor in the selected articles is endostatin (31, 73, 75, 77, 81, 82, 95). Furthermore also alteration of transcripts involved in signal transduction pathways, i.e. the Ang/Tie2 signalling pathway (90) and the mechanism of uPAR truncation (98, 99, 101), is described. Also downregulation of the critical proangiogenic transcript Fli1 is mentioned in several articles (107-109) to play a part in the disturbed angiogenesis known in SSc vasculopathy. In seven selected articles, the role of EndoMT in SSc is also emphasized (127-133). In vitro studies using MVEC obtained from SSc patients confirmed a role for ET-1 and TGFβ as enhancers for EndoMT (127, 133). Nevertheless the mechanisms behind this transition are still not yet fully understood.

19

ACCEPTED MANUSCRIPT While EndoMT is a known center stage in the fibrotic processes of many diseases (120-126), its role in the pathogenesis of SSc vasculopathy still needs clarification.

MA

NU

SC R

IP

T

The most often described biomarker of disturbed vasculogenesis are EPC. Nevertheless whether or not they can be used as biomarker in practice is still under debate since both decreased (79, 94, 137, 141, 143, 145) and increased (31, 63, 84, 138, 140, 142, 144, 147) levels of circulating EPC have been described in the selected literature. These contradictory results, displayed in supplementary file 5, could be due to the use of different protocols to quantify EPC, the use of different combinations of surface markers and differences in subsets of patients, used medications and disease durations between the studies. EPC can be defined by colony-forming unit (CFU) assays that use culture-based methods and by flow cytometry where expression of cell-surface antigens including CD34, CD133 and VEGFR2 is assayed. However, protocols for isolating and counting EPC are not yet standardized therefore causing discrepancy in results between studies. Recently the European League Against Rheumatism Scleroderma Trials and Research group (EUSTAR) proposed recommendations for the identification and measurement of EPC (225). Ever since these recommendations were published, mostly decreasing levels of EPC have been reported.

CE P

TE

D

In the dysregulation of endothelial dependent control of arteriolar/venular tone, NO is the most investigated biomarker in SSc vasculopathy. Nevertheless the exact status of NO production as biomarker in SSc (vasculopathy) is still controversial since both increased (11, 166-173, 175) and decreased (172, 174, 175) total nitrate (metabolites) levels in SSc patients have been reported (Supplementary file 6). Decreased NO levels could be explained by the rapid reaction of NO and superoxide anions to generate the reactive intermediate peroxynitrite (11). ET-1 is the second most described elevated biomarker in dysfunctional vascular tone control known to occur in SSc vasculopathy. Furthermore, multiple selected articles also emphasized the role of the reninangiotensin system (RAS) and its products in the integrated regulation of vascular tone.

AC

Additionally, EC also interact with the coagulation system by upregulating multiple markers of coagulation (most importantly vWF), this way contributing to the prothrombotic state of SSc patients (41). However whether or not impairment of fibrinolysis is present, is still under debate since both depressed fibrinolytic activity (41) and a normal plasma fibrinolytic profile (40) has been reported. The importance of the interaction of EC with the complement system in SSc vasculopathy has not been deeply investigated. Only three articles were selected concerning this topic. Although the immune system plays its own particular part in SSc pathogenesis, its interaction with EC and therefore its role in SSc vasculopathy, is not negligible. IL-33, one of the most described cytokines in the selected articles is abnormally expressed in early stage SSc skin (193) and is elevated in the sera of early SSc patients (30, 205, 206). Furthermore it is shown that the IL-33/ST2 axis increases proliferation, migration and morphologic differentiation of human ECs with increased endothelial permeability, consistently with increased angiogenesis in vivo (193). These findings support the hypothesis that IL-33 might mediate very early pathogenic events of SSc through recruitment and stimulation of ST2-expressing cells (immune cells and fibroblast/myofibroblast), this way modulating inflammatory and fibrotic processes in SSc. IL-2 is known to increase binding of natural killer cells to the endothelium (226). Needleman et al. show significantly higher IL-2 levels in SSc patients compared to HC using a CTLL bioassay (189). However when the same authors use an ELISA to determine both levels, no statistical significance between both groups was shown. Their 20

ACCEPTED MANUSCRIPT

SC R

IP

T

ELISA findings suggest similar IL-2 levels in SSc patients and HC but increased IL-2 biologic activity in SSc patient sera. The latter is supported by the hypothesis of Kahaleh and LeRoy (196) that an IL-2 inhibitor is present in sera from HC but reduced in SSc sera. Famularo et al (194) also found an increase in mean IL-2 serum levels in SSc patients compared to controls. In contrast, Degiannis et al (195) found comparable levels of IL-2 in plasma from SSc patients and controls. Both authors use an ELISA detection method for IL-2 quantification. More recently IL-17A is investigated and authors observe that this cytokine induces the expression of adhesion molecules and chemokines in HUVEC. Furthermore it mediates vascular inflammation through ERK phosphorylation (204) clarifying the link between IL-17 producing T cells and endothelial injury in SSc vasculopathy.

MA

NU

Finally the selected articles also provide limited evidence of EC interaction with other inflammatory (MC), vascular (PCs) or adipose cells. The latter is a relatively new discovery, which involves a new type of cytokines, the adipocytokines, this way expanding the spectrum of SSc vasculopathy. Although recent data suggest a role for macrophages in angiogenesis (208-211), there is currently no scientific evidence (in the form of (randomized) control trials, observational and/or in vitro studies) proving that EC-macrophage interaction contributes to SSc vasculopathy.

5. CONCLUSION

AC

CE P

TE

D

Systemic sclerosis (SSc) is an autoimmune connective tissue disorder characterized by proliferative vasculopathy, immunological abnormalities and progressive fibrosis of multiple organs including the skin. This systematic review focuses on the role of ECs in SSc vasculopathy. The most representing and reliable biomarkers described by the selected articles were adhesion molecules (ICAM1, VCAM1, E-selectin, P-selectin) for EC activation, AECA for EC apoptosis, VEGF, its receptor VEGFR-2 and endostatin for disturbed angiogenesis, circulating EPC for defective vasculogenesis, ET-1 for disturbed vascular tone control, vWF for coagulopathy and IL-33 for EC- immune system communication. Emerging, relatively new discovered biomarkers described in the selected articles are VEGF165b, IL-17A and the adipocytokines. Although the etiology of SSc remains unclear, this systematic review summarizes the growing evidence that SSc is primarily a vascular disease where EC dysfunction is present and prominent in different aspects of cell survival (activation and apoptosis), angiogenesis and vasculogenesis and where disturbed interactions between ECs and various other cells contribute to SSc vasculopathy.

21

ACCEPTED MANUSCRIPT APPENDICES

Figure 1. Endothelial cell dysfunction in SSc vasculopathy: a multifunctional approach

T

Figure legend: this figure summarizes the different dysfunctional aspects of EC in SSc vasculopathy.

IP

Table 1. Number of selected articles according to EC (dys)function in SSc vasculopathy

SC R

Table legend: data are given as number of articles selected in the systematic review, categorized according to EC (dys)function. Some articles (n=33) covered more than one topic and were therefore categorized in more than one function group. This way the grand total exceeds 193 articles.

Table 2. Regulation of EC apoptosis markers described by the selected articles

MA

NU

Table legend: data are given as number of selected articles describing significant upregulation, significant downregulation or normal regulation (i.e. no significant difference in regulation between controls and SSc population) of markers of EC apoptosis, disturbed angiogenesis and EC-immune system communication in SSc or certain SSc subtypes. Selected basic science articles (in vitro experiments, animal studies) involving the process of EC apoptosis, disturbed angiogenesis and EC-immune system communication in SSc are not included in this table.

Supplementary file 1. Details on search strategy using the Prisma 2009 checklist Supplementary file 2. Flow diagram of search process according to PRISMA 2009 guidelines

TE

D

Supplementary file 3. Quality assessment of the selected articles Supplementary file 4. Summary of all 193 selected articles

CE P

Supplementary file 5. Discrepancy in selected literature concerning levels of circulating EPC in SSc patients

AC

Supplementary file 6. Discrepancy in literature concerning levels of NO (metabolites)

22

ACCEPTED MANUSCRIPT REFERENCES

AC

CE P

TE

D

MA

NU

SC R

IP

T

[1] Desbois AC, Cacoub P. Systemic sclerosis: An update in 2016. Autoimmun Rev 2016; 15(5): 417-26. [2] Matucci-Cerinic M, Kahaleh B, Wigley FM. Review: evidence that systemic sclerosis is a vascular disease. Arthritis Rheum 2013; 65(8): 1953-62. [3] Sgonc R, Gruschwitz MS, Dietrich H, Recheis H, Gershwin ME, Wick G. Endothelial cell apoptosis is a primary pathogenetic event underlying skin lesions in avian and human scleroderma. J Clin Invest 1996; 98(3): 785-92. [4] Cabral-Marques O, Riemekasten G. Vascular hypothesis revisited: Role of stimulating antibodies against angiotensin and endothelin receptors in the pathogenesis of systemic sclerosis. Autoimmun Rev 2016; 15(7): 690-4. [5] Moher D, Liberati A, Tetzlaff J, Altman DG, Group P. Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement. PLoS Med 2009; 6(7): e1000097. [6] Quality Assessment Tool for Observational Cohort and Cross-Sectional Studies, adapted from the national institute of Health (NIH), available from: URL: https://www.nhlbi.nih.gov/healthpro/guidelines/in-develop/cardiovascular-risk-reduction/tools/cohort; last updated March 2014 [accessed 28.11.16]. [7] Roumm AD, Whiteside TL, Medsger TA, Rodnan GP. Lymphocytes in the skin of patients with progressive systemic sclerosis. Quantification, subtyping, and clinical correlations. Arthritis Rheum 1984; 27(6): 645-53. [8] Gruschwitz M, von den Driesch P, Kellner I, Hornstein OP, Sterry W. Expression of adhesion proteins involved in cell-cell and cell-matrix interactions in the skin of patients with progressive systemic sclerosis. J Am Acad Dermatol 1992; 27(2 Pt 1): 169-77. [9] Gruschwitz MS, Hornstein OP, von Den Driesch P. Correlation of soluble adhesion molecules in the peripheral blood of scleroderma patients with their in situ expression and with disease activity. Arthritis Rheum 1995; 38(2): 184-9. [10] Ihn H, Sato S, Fujimoto M, Takehara K, Tamaki K. Increased serum levels of soluble vascular cell adhesion molecule-1 and E-selectin in patients with systemic sclerosis. Br J Rheumatol 1998; 37(11): 1188-92. [11] Andersen GN, Caidahl K, Kazzam E, Petersson AS, Waldenström A, Mincheva-Nilsson L, et al. Correlation between increased nitric oxide production and markers of endothelial activation in systemic sclerosis: findings with the soluble adhesion molecules E-selectin, intercellular adhesion molecule 1, and vascular cell adhesion molecule 1. Arthritis Rheum 2000; 43(5): 1085-93. [12] Cerinic MM, Valentini G, Sorano GG, D'Angelo S, Cuomo G, Fenu L, et al. Blood coagulation, fibrinolysis, and markers of endothelial dysfunction in systemic sclerosis. Semin Arthritis Rheum 2003; 32(5): 285-95. [13] Macko RF, Gelber AC, Young BA, Lowitt MH, White B, Wigley FM, et al. Increased circulating concentrations of the counteradhesive proteins SPARC and thrombospondin-1 in systemic sclerosis (scleroderma). Relationship to platelet and endothelial cell activation. J Rheumatol 2002; 29(12): 2565-70. [14] Iversen LV, Østergaard O, Ullman S, Nielsen CT, Halberg P, Karlsmark T, et al. Circulating microparticles and plasma levels of soluble E- and P-selectins in patients with systemic sclerosis. Scand J Rheumatol 2013; 42(6): 473-82. [15] Nomura S, Inami N, Ozaki Y, Kagawa H, Fukuhara S. Significance of microparticles in progressive systemic sclerosis with interstitial pneumonia. Platelets 2008; 19(3): 192-8. [16] Sollberg S, Peltonen J, Uitto J, Jimenez SA. Elevated expression of beta 1 and beta 2 integrins, intercellular adhesion molecule 1, and endothelial leukocyte adhesion molecule 1 in the skin of patients with systemic sclerosis of recent onset. Arthritis Rheum 1992; 35(3): 290-8.

23

ACCEPTED MANUSCRIPT

AC

CE P

TE

D

MA

NU

SC R

IP

T

[17] Kawano-Dourado L, Ab'Saber AM, Capelozzi VL, Valeri C, Barbas CS. In situ evidence of pulmonary endothelial activation in patients with granulomatosis with polyangiitis and systemic sclerosis. Lung 2015; 193(3): 355-9. [18] Barnes TC, Spiller DG, Anderson ME, Edwards SW, Moots RJ. Endothelial activation and apoptosis mediated by neutrophil-dependent interleukin 6 trans-signalling: a novel target for systemic sclerosis? Ann Rheum Dis 2011; 70(2): 366-72. [19] Yalçınkaya Y, Adın-Çınar S, Artim-Esen B, Kamalı S, Pehlivan Ö, Öcal L, et al. Capillaroscopic findings and vascular biomarkers in systemic sclerosis: Association of low CD40L levels with late scleroderma pattern. Microvasc Res 2016; 108: 17-21. [20] Gruschwitz MS, Vieth G. Up-regulation of class II major histocompatibility complex and intercellular adhesion molecule 1 expression on scleroderma fibroblasts and endothelial cells by interferon-gamma and tumor necrosis factor alpha in the early disease stage. Arthritis Rheum 1997; 40(3): 540-50. [21] Sfikakis PP, Tesar J, Baraf H, Lipnick R, Klipple G, Tsokos GC. Circulating intercellular adhesion molecule-1 in patients with systemic sclerosis. Clin Immunol Immunopathol 1993; 68(1): 88-92. [22] Søndergaard K, Stengaard-Pedersen K, Zachariae H, Heickendorff L, Deleuran M, Deleuran B. Soluble intercellular adhesion molecule-1 (sICAM-1) and soluble interleukin-2 receptors (sIL-2R) in scleroderma skin. Br J Rheumatol 1998; 37(3): 304-10. [23] Kiener H, Graninger W, Machold K, Aringer M, Graninger WB. Increased levels of circulating intercellular adhesion molecule-1 in patients with systemic sclerosis. Clin Exp Rheumatol 1994; 12(5): 483-7. [24] Ihn H, Sato S, Fujimoto M, Kikuchi K, Kadono T, Tamaki K, et al. Circulating intercellular adhesion molecule-1 in the sera of patients with systemic sclerosis: enhancement by inflammatory cytokines. Br J Rheumatol 1997; 36(12): 1270-5. [25] Manetti M, Guiducci S, Romano E, Rosa I, Ceccarelli C, Mello T, et al. Differential expression of junctional adhesion molecules in different stages of systemic sclerosis. Arthritis Rheum 2013; 65(1): 247-57. [26] Hou Y, Rabquer BJ, Gerber ML, Del Galdo F, Jimenez SA, Haines GK, et al. Junctional adhesion molecule-A is abnormally expressed in diffuse cutaneous systemic sclerosis skin and mediates myeloid cell adhesion. Ann Rheum Dis 2010; 69(1): 249-54. [27] Rabquer BJ, Hou Y, Del Galdo F, Kenneth Haines G, Gerber ML, Jimenez SA, et al. The proadhesive phenotype of systemic sclerosis skin promotes myeloid cell adhesion via ICAM-1 and VCAM-1. Rheumatology (Oxford) 2009; 48(7): 734-40. [28] Riccieri V, Stefanantoni K, Vasile M, Macrì V, Sciarra I, Iannace N, et al. Abnormal plasma levels of different angiogenic molecules are associated with different clinical manifestations in patients with systemic sclerosis. Clin Exp Rheumatol 2011; 29(2 Suppl 65): S46-52. [29] Koch AE, Kronfeld-Harrington LB, Szekanecz Z, Cho MM, Haines GK, Harlow LA, et al. In situ expression of cytokines and cellular adhesion molecules in the skin of patients with systemic sclerosis. Their role in early and late disease. Pathobiology 1993; 61(5-6): 239-46. [30] Vettori S, Cuomo G, Iudici M, D'Abrosca V, Giacco V, Barra G, et al. Early systemic sclerosis: serum profiling of factors involved in endothelial, T-cell, and fibroblast interplay is marked by elevated interleukin-33 levels. J Clin Immunol 2014; 34(6): 663-8. [31] Avouac J, Vallucci M, Smith V, Senet P, Ruiz B, Sulli A, et al. Correlations between angiogenic factors and capillaroscopic patterns in systemic sclerosis. Arthritis Res Ther 2013; 15(2): R55. [32] Del Galdo F, Maul GG, Jiménez SA, Artlett CM. Expression of allograft inflammatory factor 1 in tissues from patients with systemic sclerosis and in vitro differential expression of its isoforms in response to transforming growth factor beta. Arthritis Rheum 2006; 54(8): 2616-25. [33] Wipff J, Avouac J, Borderie D, Zerkak D, Lemarechal H, Kahan A, et al. Disturbed angiogenesis in systemic sclerosis: high levels of soluble endoglin. Rheumatology (Oxford) 2008; 47(7): 972-5. [34] Hasegawa M, Sato S, Echigo T, Hamaguchi Y, Yasui M, Takehara K. Up regulated expression of fractalkine/CX3CL1 and CX3CR1 in patients with systemic sclerosis. Ann Rheum Dis 2005; 64(1): 21-8. 24

ACCEPTED MANUSCRIPT

AC

CE P

TE

D

MA

NU

SC R

IP

T

[35] Sicinska J, Gorska E, Cicha M, Kuklo-Kowalska A, Hamze V, Stepien K, et al. Increased serum fractalkine in systemic sclerosis. Down-regulation by prostaglandin E1. Clin Exp Rheumatol 2008; 26(4): 527-33. [36] Fujimoto M, Hasegawa M, Hamaguchi Y, Komura K, Matsushita T, Yanaba K, et al. A clue for telangiectasis in systemic sclerosis: elevated serum soluble endoglin levels in patients with the limited cutaneous form of the disease. Dermatology 2006; 213(2): 88-92. [37] Salojin KV, Le Tonquèze M, Saraux A, Nassonov EL, Dueymes M, Piette JC, et al. Antiendothelial cell antibodies: useful markers of systemic sclerosis. Am J Med 1997; 102(2): 178-85. [38] Vancheeswaran R, Magoulas T, Efrat G, Wheeler-Jones C, Olsen I, Penny R, et al. Circulating endothelin-1 levels in systemic sclerosis subsets--a marker of fibrosis or vascular dysfunction? J Rheumatol 1994; 21(10): 1838-44. [39] Blann AD, Illingworth K, Jayson MI. Mechanisms of endothelial cell damage in systemic sclerosis and Raynaud's phenomenon. J Rheumatol 1993; 20(8): 1325-30. [40] Herrick AL, Illingworth K, Blann A, Hay CR, Hollis S, Jayson MI. Von Willebrand factor, thrombomodulin, thromboxane, beta-thromboglobulin and markers of fibrinolysis in primary Raynaud's phenomenon and systemic sclerosis. Ann Rheum Dis 1996; 55(2): 122-7. [41] Ames PR, Lupoli S, Alves J, Atsumi T, Edwards C, Iannaccone L, et al. The coagulation/fibrinolysis balance in systemic sclerosis: evidence for a haematological stress syndrome. Br J Rheumatol 1997; 36(10): 1045-50. [42] Goodfield MJ, Orchard MA, Rowell NR. Whole blood platelet aggregation and coagulation factors in patients with systemic sclerosis. Br J Haematol 1993; 84(4): 675-80. [43] Kahaleh MB, Osborn I, LeRoy EC. Increased factor VIII/von Willebrand factor antigen and von Willebrand factor activity in scleroderma and in Raynaud's phenomenon. Ann Intern Med 1981; 94(4 pt 1): 482-4. [44] Mannucci PM, Lombardi R, Lattuada A, Perticucci E, Valsecchi R, Remuzzi G. Supranormal von Willebrand factor multimers in scleroderma. Blood 1989; 73(6): 1586-91. [45] Holt CM, Lindsey N, Moult J, Malia RG, Greaves M, Hume A, et al. Antibody-dependent cellular cytotoxicity of vascular endothelium: characterization and pathogenic associations in systemic sclerosis. Clin Exp Immunol 1989; 78(3): 359-65. [46] Sgonc R, Gruschwitz MS, Boeck G, Sepp N, Gruber J, Wick G. Endothelial cell apoptosis in systemic sclerosis is induced by antibody-dependent cell-mediated cytotoxicity via CD95. Arthritis Rheum 2000; 43(11): 2550-62. [47] Bordron A, Dueymes M, Levy Y, Jamin C, Leroy JP, Piette JC, et al. The binding of some human antiendothelial cell antibodies induces endothelial cell apoptosis. J Clin Invest 1998; 101(10): 202935. [48] Ahmed SS, Tan FK, Arnett FC, Jin L, Geng YJ. Induction of apoptosis and fibrillin 1 expression in human dermal endothelial cells by scleroderma sera containing anti-endothelial cell antibodies. Arthritis Rheum 2006; 54(7): 2250-62. [49] Drenk F, Deicher HR. Pathophysiological effects of endothelial cytotoxic activity derived from sera of patients with progressive systemic sclerosis. J Rheumatol 1988; 15(3): 468-74. [50] Nguyen VA, Sgonc R, Dietrich H, Wick G. Endothelial injury in internal organs of University of California at Davis line 200 (UCD 200) chickens, an animal model for systemic sclerosis (Scleroderma). J Autoimmun 2000; 14(2): 143-9. [51] Quadros NP, Roberts-Thomson PJ, Gallus AS. IgG and IgM anti-endothelial cell antibodies in patients with collagen-vascular disorders. Rheumatol Int 1990; 10(3): 113-9. [52] Pignone A, Scaletti C, Matucci-Cerinic M, Vázquez-Abad D, Meroni PL, Del Papa N, et al. Antiendothelial cell antibodies in systemic sclerosis: significant association with vascular involvement and alveolo-capillary impairment. Clin Exp Rheumatol 1998; 16(5): 527-32. [53] Marks RM, Czerniecki M, Andrews BS, Penny R. The effects of scleroderma serum on human microvascular endothelial cells. Induction of antibody-dependent cellular cytotoxicity. Arthritis Rheum 1988; 31(12): 1524-34. 25

ACCEPTED MANUSCRIPT

AC

CE P

TE

D

MA

NU

SC R

IP

T

[54] Penning CA, Cunningham J, French MA, Harrison G, Rowell NR, Hughes P. Antibodydependent cellular cytotoxicity of human vascular endothelium in systemic sclerosis. Clin Exp Immunol 1984; 57(3): 548-56. [55] Del Papa N, Quirici N, Scavullo C, Gianelli U, Corti L, Vitali C, et al. Antiendothelial cell antibodies induce apoptosis of bone marrow endothelial progenitors in systemic sclerosis. J Rheumatol 2010; 37(10): 2053-63. [56] Carvalho D, Savage CO, Black CM, Pearson JD. IgG antiendothelial cell autoantibodies from scleroderma patients induce leukocyte adhesion to human vascular endothelial cells in vitro. Induction of adhesion molecule expression and involvement of endothelium-derived cytokines. J Clin Invest 1996; 97(1): 111-9. [57] Negi VS, Tripathy NK, Misra R, Nityanand S. Antiendothelial cell antibodies in scleroderma correlate with severe digital ischemia and pulmonary arterial hypertension. J Rheumatol 1998; 25(3): 462-6. [58] Okazaki S, Ogawa F, Iwata Y, Hara T, Muroi E, Komura K, et al. Autoantibody against caspase3, an executioner of apoptosis, in patients with systemic sclerosis. Rheumatol Int 2010; 30(7): 871-8. [59] Yamamoto T, Nishioka K. Possible role of apoptosis in the pathogenesis of bleomycin-induced scleroderma. J Invest Dermatol 2004; 122(1): 44-50. [60] Costa S, Mondini M, Caneparo V, Afeltra A, Airò P, Bellisai F, et al. Detection of anti-IFI16 antibodies by ELISA: clinical and serological associations in systemic sclerosis. Rheumatology (Oxford) 2011; 50(4): 674-81. [61] Mondini M, Vidali M, De Andrea M, Azzimonti B, Airò P, D'Ambrosio R, et al. A novel autoantigen to differentiate limited cutaneous systemic sclerosis from diffuse cutaneous systemic sclerosis: the interferon-inducible gene IFI16. Arthritis Rheum 2006; 54(12): 3939-44. [62] Wang Y, Kahaleh B. Epigenetic repression of bone morphogenetic protein receptor II expression in scleroderma. J Cell Mol Med 2013; 17(10): 1291-9. [63] Del Papa N, Colombo G, Fracchiolla N, Moronetti LM, Ingegnoli F, Maglione W, et al. Circulating endothelial cells as a marker of ongoing vascular disease in systemic sclerosis. Arthritis Rheum 2004; 50(4): 1296-304. [64] Castellino G, Corallini F, Bortoluzzi A, La Corte R, Lo Monaco A, Secchiero P, et al. The tumour necrosis factor-related apoptosis-inducing ligand-osteoprotegerin system in limited systemic sclerosis: a new disease marker? Rheumatology (Oxford) 2010; 49(6): 1173-6. [65] Scambi C, Ugolini S, Jokiranta TS, De Franceschi L, Bortolami O, La Verde V, et al. The local complement activation on vascular bed of patients with systemic sclerosis: a hypothesis-generating study. PLoS One 2015; 10(2): e0114856. [66] Parra ER, Aguiar AC, Teodoro WR, de Souza R, Yoshinari NH, Capelozzi VL. Collagen V and vascular injury promote lung architectural changes in systemic sclerosis. Clin Respir J 2009; 3(3): 13542. [67] Guiducci S, Distler JH, Jüngel A, Huscher D, Huber LC, Michel BA, et al. The relationship between plasma microparticles and disease manifestations in patients with systemic sclerosis. Arthritis Rheum 2008; 58(9): 2845-53. [68] Iversen LV, Ullman S, Østergaard O, Nielsen CT, Halberg P, Karlsmark T, et al. Cross-sectional study of soluble selectins, fractions of circulating microparticles and their relationship to lung and skin involvement in systemic sclerosis. BMC Musculoskelet Disord 2015; 16: 191. [69] Maurer B, Busch N, Jüngel A, Pileckyte M, Gay RE, Michel BA, et al. Transcription factor fosrelated antigen-2 induces progressive peripheral vasculopathy in mice closely resembling human systemic sclerosis. Circulation 2009; 120(23): 2367-76. [70] Giusti B, Fibbi G, Margheri F, Serratì S, Rossi L, Poggi F, et al. A model of anti-angiogenesis: differential transcriptosome profiling of microvascular endothelial cells from diffuse systemic sclerosis patients. Arthritis Res Ther 2006; 8(4): R115. [71] 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(7): 1529-33. 26

ACCEPTED MANUSCRIPT

AC

CE P

TE

D

MA

NU

SC R

IP

T

[72] Davies CA, Jeziorska M, Freemont AJ, Herrick AL. The differential expression of VEGF, VEGFR2, and GLUT-1 proteins in disease subtypes of systemic sclerosis. Hum Pathol 2006; 37(2): 190-7. [73] Distler O, Del Rosso A, Giacomelli R, Cipriani P, Conforti ML, Guiducci S, et al. Angiogenic and angiostatic factors in systemic sclerosis: increased levels of vascular endothelial growth factor are a feature of the earliest disease stages and are associated with the absence of fingertip ulcers. Arthritis Res 2002; 4(6): R11. [74] 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(1): 109-16. [75] Dziankowska-Bartkowiak B, Waszczykowska E, Zalewska A, Sysa-Jedrzejowska A. Correlation of endostatin and tissue inhibitor of metalloproteinases 2 (TIMP2) serum levels with cardiovascular involvement in systemic sclerosis patients. Mediators Inflamm 2005; 2005(3): 144-9. [76] Głodkowska-Mrówka E, Górska E, Ciurzyński M, Stelmaszczyk-Emmel A, Bienias P, Irzyk K, et al. Pro- and antiangiogenic markers in patients with pulmonary complications of systemic scleroderma. Respir Physiol Neurobiol 2015; 209: 69-75. [77] Hummers LK, Hall A, Wigley FM, Simons M. Abnormalities in the regulators of angiogenesis in patients with scleroderma. J Rheumatol 2009; 36(3): 576-82. [78] Mackiewicz Z, Sukura A, Povilenaité D, Ceponis A, Virtanen I, Hukkanen M, et al. Increased but imbalanced expression of VEGF and its receptors has no positive effect on angiogenesis in systemic sclerosis skin. Clin Exp Rheumatol 2002; 20(5): 641-6. [79] Kuwana M, Okazaki Y, Yasuoka H, Kawakami Y, Ikeda Y. Defective vasculogenesis in systemic sclerosis. Lancet 2004; 364(9434): 603-10. [80] Carrai V, Miniati I, Guiducci S, Capaccioli G, Alterini R, Saccardi R, et al. Evidence for reduced angiogenesis in bone marrow in SSc: immunohistochemistry and multiparametric computerized imaging analysis. Rheumatology (Oxford) 2012; 51(6): 1042-8. [81] Farouk HM, Hamza SH, El Bakry SA, Youssef SS, Aly IM, Moustafa AA, et al. Dysregulation of angiogenic homeostasis in systemic sclerosis. Int J Rheum Dis 2013; 16(4): 448-54. [82] Silva I, Almeida C, Teixeira A, Oliveira J, Vasconcelos C. Impaired angiogenesis as a feature of digital ulcers in systemic sclerosis. Clin Rheumatol 2016; 35(7): 1743-51. [83] 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(3): e14-26. [84] Avouac J, Wipff J, Goldman O, Ruiz B, Couraud PO, Chiocchia G, et al. Angiogenesis in systemic sclerosis: impaired expression of vascular endothelial growth factor receptor 1 in endothelial progenitor-derived cells under hypoxic conditions. Arthritis Rheum 2008; 58(11): 355061. [85] Higashi-Kuwata N, Makino T, Inoue Y, Ihn H. Expression pattern of VEGFR-1, -2, -3 and D2-40 protein in the skin of patients with systemic sclerosis. Eur J Dermatol 2011; 21(4): 490-4. [86] Jinnin M, Makino T, Kajihara I, Honda N, Makino K, Ogata A, et al. Serum levels of soluble vascular endothelial growth factor receptor-2 in patients with systemic sclerosis. Br J Dermatol 2010; 162(4): 751-8. [87] Silveri F, De Angelis R, Poggi A, Muti S, Bonapace G, Argentati F, et al. Relative roles of endothelial cell damage and platelet activation in primary Raynaud's phenomenon (RP) and RP secondary to systemic sclerosis. Scand J Rheumatol 2001; 30(5): 290-6. [88] Shirai Y, Okazaki Y, Inoue Y, Tamura Y, Yasuoka H, Takeuchi T, et al. Elevated levels of pentraxin 3 in systemic sclerosis: associations with vascular manifestations and defective vasculogenesis. Arthritis Rheumatol 2015; 67(2): 498-507. [89] Noda S, Asano Y, Aozasa N, Akamata K, Taniguchi T, Takahashi T, et al. Clinical significance of serum soluble Tie1 levels in patients with systemic sclerosis. Arch Dermatol Res 2013; 305(4): 325-31. [90] Noda S, Asano Y, Aozasa N, Akamata K, Yamada D, Masui Y, et al. Serum Tie2 levels: clinical association with microangiopathies in patients with systemic sclerosis. J Eur Acad Dermatol Venereol 2011; 25(12): 1476-9. 27

ACCEPTED MANUSCRIPT

AC

CE P

TE

D

MA

NU

SC R

IP

T

[91] Michalska-Jakubus M, Kowal-Bielecka O, Chodorowska G, Bielecki M, Krasowska D. Angiopoietins-1 and -2 are differentially expressed in the sera of patients with systemic sclerosis: high angiopoietin-2 levels are associated with greater severity and higher activity of the disease. Rheumatology (Oxford) 2011; 50(4): 746-55. [92] Hayashi Y, Jinnin M, Makino T, Kajihara I, Makino K, Honda N, et al. Serum angiopoietin-like protein 3 concentrations in rheumatic diseases. Eur J Dermatol 2012; 22(4): 500-4. [93] Hamaguchi Y, Hasegawa M, Tanaka C, Kumada S, Sato S, Takehara K, et al. Elevated serum placenta growth factor (PlGF) levels in patients with systemic sclerosis: a possible role in the development of skin but not lung fibrosis. J Dermatol Sci 2010; 58(3): 229-31. [94] Avouac J, Meune C, Ruiz B, Couraud PO, Uzan G, Boileau C, et al. Angiogenic biomarkers predict the occurrence of digital ulcers in systemic sclerosis. Ann Rheum Dis 2012; 71(3): 394-9. [95] Hebbar M, Peyrat JP, Hornez L, Hatron PY, Hachulla E, Devulder B. Increased concentrations of the circulating angiogenesis inhibitor endostatin in patients with systemic sclerosis. Arthritis Rheum 2000; 43(4): 889-93. [96] Furuse S, Fujii H, Kaburagi Y, Fujimoto M, Hasegawa M, Takehara K, et al. Serum concentrations of the CXC chemokines interleukin 8 and growth-regulated oncogene-alpha are elevated in patients with systemic sclerosis. J Rheumatol 2003; 30(7): 1524-8. [97] Manetti M, Guiducci S, Romano E, Bellando-Randone S, Conforti ML, Ibba-Manneschi L, et al. Increased serum levels and tissue expression of matrix metalloproteinase-12 in patients with systemic sclerosis: correlation with severity of skin and pulmonary fibrosis and vascular damage. Ann Rheum Dis 2012; 71(6): 1064-72. [98] 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(10): 3275-85. [99] Serratì S, Cinelli M, Margheri F, Guiducci S, Del Rosso A, Pucci M, et al. Systemic sclerosis fibroblasts inhibit in vitro angiogenesis by MMP-12-dependent cleavage of the endothelial cell urokinase receptor. J Pathol 2006; 210(2): 240-8. [100] Margheri F, Serratì S, Lapucci A, Chillà A, Bazzichi L, Bombardieri S, et al. Modulation of the angiogenic phenotype of normal and systemic sclerosis endothelial cells by gain-loss of function of pentraxin 3 and matrix metalloproteinase 12. Arthritis Rheum 2010; 62(8): 2488-98. [101] 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(12): 3926-38. [102] Giusti B, Serratì S, Margheri F, Papucci L, Rossi L, Poggi F, et al. The antiangiogenic tissue kallikrein pattern of endothelial cells in systemic sclerosis. Arthritis Rheum 2005; 52(11): 3618-28. [103] Milia AF, Del Rosso A, Pacini A, Manetti M, Marrelli A, Nosi D, et al. Differential expression of tissue kallikrein in the skin of systemic sclerosis. Histol Histopathol 2005; 20(2): 415-22. [104] Noda S, Asano Y, Akamata K, Aozasa N, Taniguchi T, Takahashi T, et al. A possible contribution of altered cathepsin B expression to the development of skin sclerosis and vasculopathy in systemic sclerosis. PLoS One 2012; 7(2): e32272. [105] Noda S, Asano Y, Takahashi T, Akamata K, Aozasa N, Taniguchi T, et al. Decreased cathepsin V expression due to Fli1 deficiency contributes to the development of dermal fibrosis and proliferative vasculopathy in systemic sclerosis. Rheumatology (Oxford) 2013; 52(5): 790-9. [106] Yamashita T, Asano Y, Taniguchi T, Nakamura K, Saigusa R, Takahashi T, et al. A potential contribution of altered cathepsin L expression to the development of dermal fibrosis and vasculopathy in systemic sclerosis. Exp Dermatol 2016; 25(4): 287-92. [107] Asano Y, Stawski L, Hant F, Highland K, Silver R, Szalai G, et al. Endothelial Fli1 deficiency impairs vascular homeostasis: a role in scleroderma vasculopathy. Am J Pathol 2010; 176(4): 1983-98. [108] Ichimura Y, Asano Y, Akamata K, Takahashi T, Noda S, Taniguchi T, et al. Fli1 deficiency contributes to the suppression of endothelial CXCL5 expression in systemic sclerosis. Arch Dermatol Res 2014; 306(4): 331-8. 28

ACCEPTED MANUSCRIPT

AC

CE P

TE

D

MA

NU

SC R

IP

T

[109] Akamata K, Asano Y, Taniguchi T, Yamashita T, Saigusa R, Nakamura K, et al. Increased expression of chemerin in endothelial cells due to Fli1 deficiency may contribute to the development of digital ulcers in systemic sclerosis. Rheumatology (Oxford) 2015; 54(7): 1308-16. [110] Taniguchi T, Asano Y, Akamata K, Noda S, Masui Y, Yamada D, et al. Serum levels of galectin3: possible association with fibrosis, aberrant angiogenesis, and immune activation in patients with systemic sclerosis. J Rheumatol 2012; 39(3): 539-44. [111] Aozasa N, Asano Y, Akamata K, Noda S, Masui Y, Yamada D, et al. Serum apelin levels: clinical association with vascular involvements in patients with systemic sclerosis. J Eur Acad Dermatol Venereol 2013; 27(1): 37-42. [112] Saigusa R, Asano Y, Taniguchi T, Yamashita T, Takahashi T, Ichimura Y, et al. A possible contribution of endothelial CCN1 downregulation due to Fli1 deficiency to the development of digital ulcers in systemic sclerosis. Exp Dermatol 2015; 24(2): 127-32. [113] van Bon L, Affandi AJ, Broen J, Christmann RB, Marijnissen RJ, Stawski L, et al. Proteome-wide analysis and CXCL4 as a biomarker in systemic sclerosis. N Engl J Med 2014; 370(5): 433-43. [114] Rabquer BJ, Tsou PS, Hou Y, Thirunavukkarasu E, Haines GK, Impens AJ, et al. Dysregulated expression of MIG/CXCL9, IP-10/CXCL10 and CXCL16 and their receptors in systemic sclerosis. Arthritis Res Ther 2011; 13(1): R18. [115] Antonelli A, Ferri C, Fallahi P, Ferrari SM, Giuggioli D, Colaci M, et al. CXCL10 (alpha) and CCL2 (beta) chemokines in systemic sclerosis--a longitudinal study. Rheumatology (Oxford) 2008; 47(1): 459. [116] Cipriani P, Franca Milia A, Liakouli V, Pacini A, Manetti M, Marrelli A, et al. Differential expression of stromal cell-derived factor 1 and its receptor CXCR4 in the skin and endothelial cells of systemic sclerosis patients: Pathogenetic implications. Arthritis Rheum 2006; 54(9): 3022-33. [117] Yanaba K, Muroi E, Yoshizaki A, Hara T, Ogawa F, Shimizu K, et al. Serum CXCL16 concentrations correlate with the extent of skin sclerosis in patients with systemic sclerosis. J Rheumatol 2009; 36(9): 1917-23. [118] Yanaba K, Yoshizaki A, Muroi E, Hara T, Ogawa F, Shimizu K, et al. CCL13 is a promising diagnostic marker for systemic sclerosis. Br J Dermatol 2010; 162(2): 332-6. [119] Yanaba K, Yoshizaki A, Muroi E, Ogawa F, Asano Y, Kadono T, et al. Serum CCL23 levels are increased in patients with systemic sclerosis. Arch Dermatol Res 2011; 303(1): 29-34. [120] Zeisberg EM, Tarnavski O, Zeisberg M, Dorfman AL, McMullen JR, Gustafsson E, et al. Endothelial-to-mesenchymal transition contributes to cardiac fibrosis. Nat Med 2007; 13(8): 952-61. [121] Chaudhuri V, Zhou L, Karasek M. Inflammatory cytokines induce the transformation of human dermal microvascular endothelial cells into myofibroblasts: a potential role in skin fibrogenesis. J Cutan Pathol 2007; 34(2): 146-53. [122] Arciniegas E, Frid MG, Douglas IS, Stenmark KR. Perspectives on endothelial-to-mesenchymal transition: potential contribution to vascular remodeling in chronic pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol 2007; 293(1): L1-8. [123] Zeisberg EM, Potenta SE, Sugimoto H, Zeisberg M, Kalluri R. Fibroblasts in kidney fibrosis emerge via endothelial-to-mesenchymal transition. J Am Soc Nephrol 2008; 19(12): 2282-7. [124] Kizu A, Medici D, Kalluri R. Endothelial-mesenchymal transition as a novel mechanism for generating myofibroblasts during diabetic nephropathy. Am J Pathol 2009; 175(4): 1371-3. [125] Hashimoto N, Phan SH, Imaizumi K, Matsuo M, Nakashima H, Kawabe T, et al. Endothelialmesenchymal transition in bleomycin-induced pulmonary fibrosis. Am J Respir Cell Mol Biol 2010; 43(2): 161-72. [126] Li J, Qu X, Bertram JF. Endothelial-myofibroblast transition contributes to the early development of diabetic renal interstitial fibrosis in streptozotocin-induced diabetic mice. Am J Pathol 2009; 175(4): 1380-8. [127] Corallo C, Cutolo M, Kahaleh B, Pecetti G, Montella A, Chirico C, et al. Bosentan and macitentan prevent the endothelial-to-mesenchymal transition (EndoMT) in systemic sclerosis: in vitro study. Arthritis Res Ther 2016; 18(1): 228. 29

ACCEPTED MANUSCRIPT

AC

CE P

TE

D

MA

NU

SC R

IP

T

[128] Li Z, Jimenez SA. Protein kinase Cδ and c-Abl kinase are required for transforming growth factor β induction of endothelial-mesenchymal transition in vitro. Arthritis Rheum 2011; 63(8): 247383. [129] Li Z, Wermuth PJ, Benn BS, Lisanti MP, Jimenez SA. Caveolin-1 deficiency induces spontaneous endothelial-to-mesenchymal transition in murine pulmonary endothelial cells in vitro. Am J Pathol 2013; 182(2): 325-31. [130] Xu H, Zaidi M, Struve J, Jones DW, Krolikowski JG, Nandedkar S, et al. Abnormal fibrillin-1 expression and chronic oxidative stress mediate endothelial mesenchymal transition in a murine model of systemic sclerosis. Am J Physiol Cell Physiol 2011; 300(3): C550-6. [131] Chrobak I, Lenna S, Stawski L, Trojanowska M. Interferon-γ promotes vascular remodeling in human microvascular endothelial cells by upregulating endothelin (ET)-1 and transforming growth factor (TGF) β2. J Cell Physiol 2013; 228(8): 1774-83. [132] Mendoza FA, Piera-Velazquez S, Farber JL, Feghali-Bostwick C, Jiménez SA. Endothelial Cells Expressing Endothelial and Mesenchymal Cell Gene Products in Lung Tissue From Patients With Systemic Sclerosis-Associated Interstitial Lung Disease. Arthritis Rheumatol 2016; 68(1): 210-7. [133] Wermuth PJ, Li Z, Mendoza FA, Jimenez SA. Stimulation of Transforming Growth Factor-β1Induced Endothelial-To-Mesenchymal Transition and Tissue Fibrosis by Endothelin-1 (ET-1): A Novel Profibrotic Effect of ET-1. PLoS One 2016; 11(9): e0161988. [134] 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(6): 1994-2004. [135] Kuwana M, Okazaki Y. Quantification of circulating endothelial progenitor cells in systemic sclerosis: a direct comparison of protocols. Ann Rheum Dis 2012; 71(4): 617-20. [136] Hristov M, Weber C. Endothelial progenitor cells: characterization, pathophysiology, and possible clinical relevance. J Cell Mol Med 2004; 8(4): 498-508. [137] Kuwana M, Okazaki Y. Brief report: impaired in vivo neovascularization capacity of endothelial progenitor cells in patients with systemic sclerosis. Arthritis Rheumatol 2014; 66(5): 1300-5. [138] Nevskaya T, Bykovskaia S, Lyssuk E, Shakhov I, Zaprjagaeva M, Mach E, et al. Circulating endothelial progenitor cells in systemic sclerosis: relation to impaired angiogenesis and cardiovascular manifestations. Clin Exp Rheumatol 2008; 26(3): 421-9. [139] Distler JH, Akhmetshina A, Dees C, Jüngel A, Stürzl M, Gay S, et al. Induction of apoptosis in circulating angiogenic cells by microparticles. Arthritis Rheum 2011; 63(7): 2067-77. [140] Del Papa N, Quirici N, Soligo D, Scavullo C, Cortiana M, Borsotti C, et al. Bone marrow endothelial progenitors are defective in systemic sclerosis. Arthritis Rheum 2006; 54(8): 2605-15. [141] Andrigueti FV, Arismendi MI, Ebbing PC, Kayser C. Decreased numbers of endothelial progenitor cells in patients in the early stages of systemic sclerosis. Microvasc Res 2015; 98: 82-7. [142] 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(10): 1455-60. [143] Mok MY, Yiu KH, Wong CY, Qiuwaxi J, Lai WH, Wong WS, et al. Low circulating level of CD133+KDR+cells in patients with systemic sclerosis. Clin Exp Rheumatol 2010; 28(5 Suppl 62): S1925. [144] Yamaguchi Y, Okazaki Y, Seta N, Satoh T, Takahashi K, Ikezawa Z, et al. Enhanced angiogenic potency of monocytic endothelial progenitor cells in patients with systemic sclerosis. Arthritis Res Ther 2010; 12(6): R205. [145] Zhu S, Evans S, Yan B, Povsic TJ, Tapson V, Goldschmidt-Clermont PJ, et al. Transcriptional regulation of Bim by FOXO3a and Akt mediates scleroderma serum-induced apoptosis in endothelial progenitor cells. Circulation 2008; 118(21): 2156-65. [146] Iwata Y, Yoshizaki A, Ogawa F, Komura K, Hara T, Muroi E, et al. Increased serum pentraxin 3 in patients with systemic sclerosis. J Rheumatol 2009; 36(5): 976-83. 30

ACCEPTED MANUSCRIPT

AC

CE P

TE

D

MA

NU

SC R

IP

T

[147] Allanore Y, Batteux F, Avouac J, Assous N, Weill B, Kahan A. Levels of circulating endothelial progenitor cells in systemic sclerosis. Clin Exp Rheumatol 2007; 25(1): 60-6. [148] Manetti M, Guiducci S, Romano E, Avouac J, Rosa I, Ruiz B, et al. Decreased expression of the endothelial cell-derived factor EGFL7 in systemic sclerosis: potential contribution to impaired angiogenesis and vasculogenesis. Arthritis Res Ther 2013; 15(5): R165. [149] Martin ER, Brenner BM, Ballermann BJ. Heterogeneity of cell surface endothelin receptors. J Biol Chem 1990; 265(23): 14044-9. [150] Lin HY, Kaji EH, Winkel GK, Ives HE, Lodish HF. Cloning and functional expression of a vascular smooth muscle endothelin 1 receptor. Proc Natl Acad Sci U S A 1991; 88(8): 3185-9. [151] Takayanagi R, Kitazumi K, Takasaki C, Ohnaka K, Aimoto S, Tasaka K, et al. Presence of nonselective type of endothelin receptor on vascular endothelium and its linkage to vasodilation. FEBS Lett 1991; 282(1): 103-6. [152] Abraham DJ, Vancheeswaran R, Dashwood MR, Rajkumar VS, Pantelides P, Xu SW, et al. Increased levels of endothelin-1 and differential endothelin type A and B receptor expression in scleroderma-associated fibrotic lung disease. Am J Pathol 1997; 151(3): 831-41. [153] Yamane K, Kashiwagi H, Suzuki N, Miyauchi T, Yanagisawa M, Goto K, et al. Elevated plasma levels of endothelin-1 in systemic sclerosis. Arthritis Rheum 1991; 34(2): 243-4. [154] Cambrey AD, Harrison NK, Dawes KE, Southcott AM, Black CM, du Bois RM, et al. Increased levels of endothelin-1 in bronchoalveolar lavage fluid from patients with systemic sclerosis contribute to fibroblast mitogenic activity in vitro. Am J Respir Cell Mol Biol 1994; 11(4): 439-45. [155] Yamane K, Miyauchi T, Suzuki N, Yuhara T, Akama T, Suzuki H, et al. Significance of plasma endothelin-1 levels in patients with systemic sclerosis. J Rheumatol 1992; 19(10): 1566-71. [156] Kadono T, Kikuchi K, Sato S, Soma Y, Tamaki K, Takehara K. Elevated plasma endothelin levels in systemic sclerosis. Arch Dermatol Res 1995; 287(5): 439-42. [157] Vancheeswaran R, Azam A, Black C, Dashwood MR. Localization of endothelin-1 and its binding sites in scleroderma skin. J Rheumatol 1994; 21(7): 1268-76. [158] Tabata H, Yamakage A, Yamazaki S. Cutaneous localization of endothelin-1 in patients with systemic sclerosis: immunoelectron microscopic study. Int J Dermatol 1997; 36(4): 272-5. [159] Pehlivan Y, Onat AM, Comez G, Babacan T. Urotensin-II in systemic sclerosis: a new peptide in pathogenesis. Clin Rheumatol 2011; 30(6): 837-42. [160] Pignone A, Rosso AD, Brosnihan KB, Perfetto F, Livi R, Fiori G, et al. Reduced circulating levels of angiotensin-(1--7) in systemic sclerosis: a new pathway in the dysregulation of endothelialdependent vascular tone control. Ann Rheum Dis 2007; 66(10): 1305-10. [161] Tsou PS, Amin MA, Campbell PL, Zakhem G, Balogh B, Edhayan G, et al. Activation of the Thromboxane A2 Receptor by 8-Isoprostane Inhibits the Pro-Angiogenic Effect of Vascular Endothelial Growth Factor in Scleroderma. J Invest Dermatol 2015; 135(12): 3153-62. [162] Ogawa F, Shimizu K, Muroi E, Hara T, Hasegawa M, Takehara K, et al. Serum levels of 8isoprostane, a marker of oxidative stress, are elevated in patients with systemic sclerosis. Rheumatology (Oxford) 2006; 45(7): 815-8. [163] Stein CM, Tanner SB, Awad JA, Roberts LJ, Morrow JD. Evidence of free radical-mediated injury (isoprostane overproduction) in scleroderma. Arthritis Rheum 1996; 39(7): 1146-50. [164] Volpe A, Biasi D, Caramaschi P, Mantovani W, Bambara LM, Canestrini S, et al. Levels of F2isoprostanes in systemic sclerosis: correlation with clinical features. Rheumatology (Oxford) 2006; 45(3): 314-20. [165] Montuschi P, Ciabattoni G, Paredi P, Pantelidis P, du Bois RM, Kharitonov SA, et al. 8Isoprostane as a biomarker of oxidative stress in interstitial lung diseases. Am J Respir Crit Care Med 1998; 158(5 Pt 1): 1524-7. [166] Yamamoto T, Katayama I, Nishioka K. Nitric oxide production and inducible nitric oxide synthase expression in systemic sclerosis. J Rheumatol 1998; 25(2): 314-7. [167] Takagi K, Kawaguchi Y, Hara M, Sugiura T, Harigai M, Kamatani N. Serum nitric oxide (NO) levels in systemic sclerosis patients: correlation between NO levels and clinical features. Clin Exp Immunol 2003; 134(3): 538-44. 31

ACCEPTED MANUSCRIPT

AC

CE P

TE

D

MA

NU

SC R

IP

T

[168] Dooley A, Gao B, Bradley N, Abraham DJ, Black CM, Jacobs M, et al. Abnormal nitric oxide metabolism in systemic sclerosis: increased levels of nitrated proteins and asymmetric dimethylarginine. Rheumatology (Oxford) 2006; 45(6): 676-84. [169] Sud A, Khullar M, Wanchu A, Bambery P. Increased nitric oxide production in patients with systemic sclerosis. Nitric Oxide 2000; 4(6): 615-9. [170] Paredi P, Kharitonov SA, Loukides S, Pantelidis P, du Bois RM, Barnes PJ. Exhaled nitric oxide is increased in active fibrosing alveolitis. Chest 1999; 115(5): 1352-6. [171] Fajac I, Kahan A, Menkès CJ, Dessanges JF, Dall'Ava-Santucci J, Dinh-Xuan AT. Increased nitric oxide in exhaled air in patients with systemic sclerosis. Clin Exp Rheumatol 1998; 16(5): 547-52. [172] Rolla G, Colagrande P, Scappaticci E, Chiavassa G, Dutto L, Cannizzo S, et al. Exhaled nitric oxide in systemic sclerosis: relationships with lung involvement and pulmonary hypertension. J Rheumatol 2000; 27(7): 1693-8. [173] Moodley YP, Lalloo UG. Exhaled nitric oxide is elevated in patients with progressive systemic sclerosis without interstitial lung disease. Chest 2001; 119(5): 1449-54. [174] Allanore Y, Borderie D, Hilliquin P, Hernvann A, Levacher M, Lemaréchal H, et al. Low levels of nitric oxide (NO) in systemic sclerosis: inducible NO synthase production is decreased in cultured peripheral blood monocyte/macrophage cells. Rheumatology (Oxford) 2001; 40(10): 1089-96. [175] Kharitonov SA, Cailes JB, Black CM, du Bois RM, Barnes PJ. Decreased nitric oxide in the exhaled air of patients with systemic sclerosis with pulmonary hypertension. Thorax 1997; 52(12): 1051-5. [176] Cotton SA, Herrick AL, Jayson MI, Freemont AJ. Endothelial expression of nitric oxide synthases and nitrotyrosine in systemic sclerosis skin. J Pathol 1999; 189(2): 273-8. [177] Servettaz A, Guilpain P, Goulvestre C, Chéreau C, Hercend C, Nicco C, et al. Radical oxygen species production induced by advanced oxidation protein products predicts clinical evolution and response to treatment in systemic sclerosis. Ann Rheum Dis 2007; 66(9): 1202-9. [178] Sambo P, Baroni SS, Luchetti M, Paroncini P, Dusi S, Orlandini G, et al. Oxidative stress in scleroderma: maintenance of scleroderma fibroblast phenotype by the constitutive up-regulation of reactive oxygen species generation through the NADPH oxidase complex pathway. Arthritis Rheum 2001; 44(11): 2653-64. [179] Laurent A, Nicco C, Chéreau C, Goulvestre C, Alexandre J, Alves A, et al. Controlling tumor growth by modulating endogenous production of reactive oxygen species. Cancer Res 2005; 65(3): 948-56. [180] Nicco C, Laurent A, Chereau C, Weill B, Batteux F. Differential modulation of normal and tumor cell proliferation by reactive oxygen species. Biomed Pharmacother 2005; 59(4): 169-74. [181] Lee P, Norman CS, Sukenik S, Alderdice CA. The clinical significance of coagulation abnormalities in systemic sclerosis (scleroderma). J Rheumatol 1985; 12(3): 514-7. [182] Truchetet ME, Demoures B, Eduardo Guimaraes J, Bertrand A, Laurent P, Jolivel V, et al. Platelets Induce Thymic Stromal Lymphopoietin Production by Endothelial Cells: Contribution to Fibrosis in Human Systemic Sclerosis. Arthritis Rheumatol 2016; 68(11): 2784-94. [183] Lemaire R, Burwell T, Sun H, Delaney T, Bakken J, Cheng L, et al. Resolution of Skin Fibrosis by Neutralization of the Antifibrinolytic Function of Plasminogen Activator Inhibitor 1. Arthritis Rheumatol 2016; 68(2): 473-83. [184] Ricklin D, Hajishengallis G, Yang K, Lambris JD. Complement: a key system for immune surveillance and homeostasis. Nat Immunol 2010; 11(9): 785-97. [185] Venneker GT, van den Hoogen FH, Boerbooms AM, Bos JD, Asghar SS. Aberrant expression of membrane cofactor protein and decay-accelerating factor in the endothelium of patients with systemic sclerosis. A possible mechanism of vascular damage. Lab Invest 1994; 70(6): 830-5. [186] Xiang Y, Matsui T, Matsuo K, Shimada K, Tohma S, Nakamura H, et al. Comprehensive investigation of disease-specific short peptides in sera from patients with systemic sclerosis: complement C3f-des-arginine, detected predominantly in systemic sclerosis sera, enhances proliferation of vascular endothelial cells. Arthritis Rheum 2007; 56(6): 2018-30. 32

ACCEPTED MANUSCRIPT

AC

CE P

TE

D

MA

NU

SC R

IP

T

[187] Selvi E, Tripodi SA, Catenaccio M, Lorenzini S, Chindamo D, Manganelli S, et al. Expression of macrophage migration inhibitory factor in diffuse systemic sclerosis. Ann Rheum Dis 2003; 62(5): 460-4. [188] Corallo C, Paulesu L, Cutolo M, Ietta F, Carotenuto C, Mannelli C, et al. Serum levels, tissue expression and cellular secretion of macrophage migration inhibitory factor in limited and diffuse systemic sclerosis. Clin Exp Rheumatol 2015; 33(4 Suppl 91): S98-105. [189] Needleman BW, Wigley FM, Stair RW. Interleukin-1, interleukin-2, interleukin-4, interleukin6, tumor necrosis factor alpha, and interferon-gamma levels in sera from patients with scleroderma. Arthritis Rheum 1992; 35(1): 67-72. [190] Maekawa T, Jinnin M, Ohtsuki M, Ihn H. Serum levels of interleukin-1α in patients with systemic sclerosis. J Dermatol 2013; 40(2): 98-101. [191] Zhang YJ, Zhang Q, Yang GJ, Tao JH, Wu GC, Huang XL, et al. Elevated serum levels of interleukin-1β and interleukin-33 in patients with systemic sclerosis in Chinese population. Z Rheumatol 2016. [192] Manetti M, Ibba-Manneschi L, Liakouli V, Guiducci S, Milia AF, Benelli G, et al. The IL1-like cytokine IL33 and its receptor ST2 are abnormally expressed in the affected skin and visceral organs of patients with systemic sclerosis. Ann Rheum Dis 2010; 69(3): 598-605. [193] Choi YS, Choi HJ, Min JK, Pyun BJ, Maeng YS, Park H, et al. Interleukin-33 induces angiogenesis and vascular permeability through ST2/TRAF6-mediated endothelial nitric oxide production. Blood 2009; 114(14): 3117-26. [194] Famularo G, Procopio A, Giacomelli R, Danese C, Sacchetti S, Perego MA, et al. Soluble interleukin-2 receptor, interleukin-2 and interleukin-4 in sera and supernatants from patients with progressive systemic sclerosis. Clin Exp Immunol 1990; 81(3): 368-72. [195] Degiannis D, Seibold JR, Czarnecki M, Raskova J, Raska K. Soluble interleukin-2 receptors in patients with systemic sclerosis. Clinical and laboratory correlations. Arthritis Rheum 1990; 33(3): 375-80. [196] Kahaleh MB, LeRoy EC. Interleukin-2 in scleroderma: correlation of serum level with extent of skin involvement and disease duration. Ann Intern Med 1989; 110(6): 446-50. [197] Hasegawa M, Fujimoto M, Kikuchi K, Takehara K. Elevated serum levels of interleukin 4 (IL-4), IL-10, and IL-13 in patients with systemic sclerosis. J Rheumatol 1997; 24(2): 328-32. [198] Hasegawa M, Sato S, Fujimoto M, Ihn H, Kikuchi K, Takehara K. Serum levels of interleukin 6 (IL-6), oncostatin M, soluble IL-6 receptor, and soluble gp130 in patients with systemic sclerosis. J Rheumatol 1998; 25(2): 308-13. [199] Khan K, Xu S, Nihtyanova S, Derrett-Smith E, Abraham D, Denton CP, et al. Clinical and pathological significance of interleukin 6 overexpression in systemic sclerosis. Ann Rheum Dis 2012; 71(7): 1235-42. [200] Sakamoto N, Kakugawa T, Hara A, Nakashima S, Yura H, Harada T, et al. Association of elevated α-defensin levels with interstitial pneumonia in patients with systemic sclerosis. Respir Res 2015; 16: 148. [201] Riccieri V, Rinaldi T, Spadaro A, Scrivo R, Ceccarelli F, Franco MD, et al. Interleukin-13 in systemic sclerosis: relationship to nailfold capillaroscopy abnormalities. Clin Rheumatol 2003; 22(2): 102-6. [202] Suzuki J, Morimoto S, Amano H, Tokano Y, Takasaki Y, Hashimoto H. Serum levels of interleukin 15 in patients with rheumatic diseases. J Rheumatol 2001; 28(11): 2389-91. [203] Wuttge DM, Eriksson P, Sirsjö A, Hansson GK, Stemme S. Expression of interleukin-15 in mouse and human atherosclerotic lesions. Am J Pathol 2001; 159(2): 417-23. [204] Xing X, Yang J, Yang X, Wei Y, Zhu L, Gao D, et al. IL-17A induces endothelial inflammation in systemic sclerosis via the ERK signaling pathway. PLoS One 2013; 8(12): e85032. [205] Manetti M, Guiducci S, Ceccarelli C, Romano E, Bellando-Randone S, Conforti ML, et al. Increased circulating levels of interleukin 33 in systemic sclerosis correlate with early disease stage and microvascular involvement. Ann Rheum Dis 2011; 70(10): 1876-8. 33

ACCEPTED MANUSCRIPT

AC

CE P

TE

D

MA

NU

SC R

IP

T

[206] Terras S, Opitz E, Moritz RK, Höxtermann S, Gambichler T, Kreuter A. Increased serum IL-33 levels may indicate vascular involvement in systemic sclerosis. Ann Rheum Dis 2013; 72(1): 144-5. [207] Yanaba K, Yoshizaki A, Asano Y, Kadono T, Sato S. Serum IL-33 levels are raised in patients with systemic sclerosis: association with extent of skin sclerosis and severity of pulmonary fibrosis. Clin Rheumatol 2011; 30(6): 825-30. [208] Spiller KL, Anfang RR, Spiller KJ, Ng J, Nakazawa KR, Daulton JW, et al. The role of macrophage phenotype in vascularization of tissue engineering scaffolds. Biomaterials 2014; 35(15): 4477-88. [209] Pabois A, Pagie S, Gérard N, Laboisse C, Pattier S, Hulin P, et al. Notch signaling mediates crosstalk between endothelial cells and macrophages via Dll4 and IL6 in cardiac microvascular inflammation. Biochem Pharmacol 2016; 104: 95-107. [210] Soldano S, Pizzorni C, Paolino S, Trombetta AC, Montagna P, Brizzolara R, et al. Alternatively Activated (M2) Macrophage Phenotype Is Inducible by Endothelin-1 in Cultured Human Macrophages. PLoS One 2016; 11(11): e0166433. [211] Manetti M. Deciphering the alternatively activated (M2) phenotype of macrophages in scleroderma. Exp Dermatol 2015; 24(8): 576-8. [212] Coleman JW, Holliday MR, Kimber I, Zsebo KM, Galli SJ. Regulation of mouse peritoneal mast cell secretory function by stem cell factor, IL-3 or IL-4. J Immunol 1993; 150(2): 556-62. [213] Hügle T, Hogan V, White KE, van Laar JM. Mast cells are a source of transforming growth factor β in systemic sclerosis. Arthritis Rheum 2011; 63(3): 795-9. [214] Yamamoto T, Katayama I, Nishioka K. Expression of stem cell factor in the lesional skin of systemic sclerosis. Dermatology 1998; 197(2): 109-14. [215] da Silva Meirelles L, Caplan AI, Nardi NB. In search of the in vivo identity of mesenchymal stem cells. Stem Cells 2008; 26(9): 2287-99. [216] Cipriani P, Marrelli A, Benedetto PD, Liakouli V, Carubbi F, Ruscitti P, et al. Scleroderma Mesenchymal Stem Cells display a different phenotype from healthy controls; implications for regenerative medicine. Angiogenesis 2013; 16(3): 595-607. [217] Denton CP, Xu S, Black CM, Pearson JD. Scleroderma fibroblasts show increased responsiveness to endothelial cell-derived IL-1 and bFGF. J Invest Dermatol 1997; 108(3): 269-74. [218] Raymond MA, Vigneault N, Luyckx V, Hébert MJ. Paracrine repercussions of preconditioning on angiogenesis and apoptosis of endothelial cells. Biochem Biophys Res Commun 2002; 291(2): 2619. [219] Laplante P, Raymond MA, Gagnon G, Vigneault N, Sasseville AM, Langelier Y, et al. Novel fibrogenic pathways are activated in response to endothelial apoptosis: implications in the pathophysiology of systemic sclerosis. J Immunol 2005; 174(9): 5740-9. [220] Serratì S, Chillà A, Laurenzana A, Margheri F, Giannoni E, Magnelli L, et al. Systemic sclerosis endothelial cells recruit and activate dermal fibroblasts by induction of a connective tissue growth factor (CCN2)/transforming growth factor β-dependent mesenchymal-to-mesenchymal transition. Arthritis Rheum 2013; 65(1): 258-69. [221] Olewicz-Gawlik A, Danczak-Pazdrowska A, Kuznar-Kaminska B, Batura-Gabryel H, Katulska K, Wojciech S, et al. Circulating adipokines and organ involvement in patients with systemic sclerosis. Acta Reumatol Port 2015; 40(2): 156-62. [222] Toyama T, Asano Y, Takahashi T, Aozasa N, Akamata K, Noda S, et al. Clinical significance of serum retinol binding protein-4 levels in patients with systemic sclerosis. J Eur Acad Dermatol Venereol 2013; 27(3): 337-44. [223] Distler JH, Jüngel A, Huber LC, Seemayer CA, Reich CF, Gay RE, et al. The induction of matrix metalloproteinase and cytokine expression in synovial fibroblasts stimulated with immune cell microparticles. Proc Natl Acad Sci U S A 2005; 102(8): 2892-7. [224] Qiu Y, Hoareau-Aveilla C, Oltean S, Harper SJ, Bates DO. The anti-angiogenic isoforms of VEGF in health and disease. Biochem Soc Trans 2009; 37(Pt 6): 1207-13. [225] Distler JH, Allanore Y, Avouac J, Giacomelli R, Guiducci S, Moritz F, et al. EULAR Scleroderma Trials and Research group statement and recommendations on endothelial precursor cells. Ann Rheum Dis 2009; 68(2): 163-8. 34

ACCEPTED MANUSCRIPT

AC

CE P

TE

D

MA

NU

SC R

IP

T

[226] Aronson FR, Libby P, Brandon EP, Janicka MW, Mier JW. IL-2 rapidly induces natural killer cell adhesion to human endothelial cells. A potential mechanism for endothelial injury. J Immunol 1988; 141(1): 158-63.

35

ACCEPTED MANUSCRIPT

T

Activation Apoptosis

SC R

IP

Fibrosis

Defective vascular tone control

MA

NU

Coagulopathy

Dysfunctional EC

Disturbed angiogenesis

Defective vasculogenesis

AC

CE P

MC dysregulation

TE

D

Dysregulation of adipocytokines

Dysregulation of vascular system through pericyte/EC interaction

Immune dysregulation EndoMT

Figure 1. Endothelial cell dysfunction in SSc vasculopathy: a multifunctional approach Figure 1 legend: this figure summarizes the different dysfunctional aspects of EC in SSc vasculopathy. Abbreviations used in figure 1: EC: endothelial cell; EndoMT: endothelial-mesenchymal transition; MC: mast cell; SSc: systemic sclerosis

36

ACCEPTED MANUSCRIPT Table 1. Number of selected articles according to EC (dys)function in SSc vasculopathy TOTAL

EC activation

38

EC apoptosis

29

Disturbed angiogenesis

56

EndoMT

7

Defective vasculogenesis

22

Defective vascular tone control

32

IP SC R

NU

Coagulation cascade and complement impairment

17

MA

Immune dysregulation

27

Mast cell dysregulation

1

TE

D

Dysregulation of the vascular system through pericytes/ECs crosstalk Fibrosis

T

EC function

2

5 4

GRAND TOTAL

240

CE P

Dysregulation of adipocytokines

AC

Table legend: data are given as number of articles selected in the systematic review, categorized according to EC (dys)function. Some articles (n=33) covered more than one topic and were therefore categorized in more than one function group. This way the grand total exceeds 193 articles. Abbreviations used in table: EC: endothelial cell, EndoMT: endothelial-mesenchymal transition, EPC: endothelial progenitor cells

37

ACCEPTED MANUSCRIPT Table 2. Regulation of markers of EC apoptosis, disturbed angiogenesis and EC-immune system communication described by the selected articles Norma Upregulat Downregula lly TOT ion tion regulat AL ed

Biomarkers

4

0

1

5

0

0

1

1

0

0

1

2

0

0

2

1

1

0

2

1

0

0

1

0

0

1

1

0

0

1

1

1

1

0

2

1

0

0

1

0

1

0

1

1

0

0

1

1

0

0

1

Collagen V

1

0

0

1

VEGF

9

1

1

11

VEGF165bVEGF165bVEGF165bVEGF165bVEGF 165bVEGF165b

1

0

0

1

VEGFR-1

0

1

0

1

VEGFR-2

1

0

0

1

VEGFR-3

0

0

1

1

PDGF-BB

1

0

0

1

PDGF

1

0

1

2

FGF-2 Markers of disturbed Serum βFGF angiogenesi HGF s

2

0

0

2

1

0

1

2

3

0

0

3

sTie1

0

0

1

1

sTie2

1

0

1

2

Ang-1

0

1

0

1

Ang-2

3

0

0

3

Angptl3

1

0

0

1

PlGF

4

0

0

4

PEDF

0

0

1

1

Endostatin

6

0

1

7

IP

AECA

T

Examin ed SSc tissue

1

SC R

IgG anti-caspase-3 Ab Caspase-3 IFI16 Ab Serum CEC Markers of EC apoptosis

NU

OPG TRAIL MAC (C5b9)

MA

MPs IFI16 Ab BMPRII MAC (C5b9)

AC

CE P

Lung

TE

Fra-2

D

Skin

38

ACCEPTED MANUSCRIPT 0

0

1

1

GRO-α

1

0

0

1

MMP-12

1

0

0

1

MMP-9

1

0

0

1

Pro-MMP-1

1

0

0

1

CTSB

1

0

0

1

1

0

1

1

0

0

1

1

0

0

1

0

0

1

1

1

0

0

1

1

0

0

1

1

0

0

1

0

1

0

1

4

0

0

4

1

0

0

1

2

0

0

2

2

0

0

2

3

0

0

3

1

0

0

1

1

0

0

1

VEGF

3

0

0

3

VEGF165bVEGF165bVEGF165bVEGF165b

1

0

0

1

VEGFR-1

2

0

1

3

VEGFR-2

5

0

0

5

VEGFR-3

2

0

0

2

GLUT-1

1

0

0

1

Tie1

0

1

0

1

MMP-12

3

0

0

3

uPAR

1

0

0

1

KLK1

0

1

1

2

KLK9

0

1

0

1

KLK11

0

1

0

1

KLK12

0

1

0

1

CTSB

1

0

0

1

CTSV

0

1

0

1

CTSL

1

0

0

1

CCN-1

0

1

0

1

IP 0

SC R

CTSV CTSL Galectin 3 CCN-1

NU

Apelin CXCL1 CXCL4

MA

CXCL5 CXCL8 CXCL9

D

CXCL10

CCL13

AC

CE P

CCL23

TE

CXCL16 CCL2

Skin

T

TIMP2

39

ACCEPTED MANUSCRIPT 1

0

0

1

CXCL5

0

1

0

1

CXCL8

1

0

0

1

CXCL9

1

0

0

1

CXCL10

1

0

0

1

CXCL16

1

0

0

1

1

0

1

1

0

0

1

1

0

0

1

1

0

0

1

1

0

0

1

1

0

0

1

2

0

0

2

3

0

1

4

2

0

0

2

1

0

0

1

2

0

0

2

3

0

0

3

1

0

0

1

0

0

1

1

1

0

1

2

IL-1α

1

0

2

3

TNF-α

0

0

1

1

IFNγ

0

0

1

1

IL-8

2

1

0

3

IL-10

1

0

0

1

IL-13

3

0

0

3

IL-15

2

0

0

2

IL-17A

1

0

0

1

IL-33

6

0

0

6

MIF

2

0

0

2

ST2

1

0

0

1

IL-6

2

0

0

2

IL-8

1

0

0

1

IL-17A

1

0

0

1

IL-33

1*

0*

0*

1

IP 0

SC R

CXCR3 CXCR6 CXCL12 CCL13

NU

CCL23 BM

VEGF MIF

MA

IL-2 sIL-2R BCGF

D

IL-4

OSM

CE P

AC

Serum Markers of disturbed EC-immune system communica tion

IL-1β

TE

IL-6 sIL-6R

Skin

T

CXCL4

* In early SSc, IL-33 protein was downregulated or absent in EC, while IL-33 mRNA was normally expressed or even upregulated. In late SSc, IL-33 was constitutively found in most EC.

40

ACCEPTED MANUSCRIPT

AC

CE P

TE

D

MA

NU

SC R

IP

T

Table legend: data are given as number of selected articles describing significant upregulation, significant downregulation or normal regulation (i.e. no significant difference in regulation between controls and SSc population) of markers of EC apoptosis, disturbed angiogenesis and EC-immune system communication in SSc or certain SSc subtypes. Selected basic science articles (in vitro experiments, animal studies) involving the process of EC apoptosis, disturbed angiogenesis and EC-immune system communication in SSc are not included in this table. Abbreviations used in table: Ab: antibodies; AECA: anti-endothelial cell antibodies; Ang-: Angiopoietin-; Angptl3: Angiopoietin-like protein 3; BCGF: B cell growth factor; BM: bone marrow; BMPRII: bone morphogenetic protein receptor II; CCL: chemokine ligand; CCN-1: cysteine-rich 61 matrix protein; CEC: circulating endothelial cells; CTSB: cathepsin B; CTSL: cathepsin L; CTSV: cathepsin V; CXCL: chemokine (C-X-C motif) ligand; EC: endothelial cell; FGF: fibroblast growth factor; Fli1: Friend leukemia integration factor 1; Fra-2: Fos-related antigen-2; GLUT-1: glucose transporter 1; GRO-α: Growth-Regulated Oncogene-α; HGF: hepatocyte growth factor; IFI16: Gamma-interferon-inducible protein IFI-16; IFNγ: interferon gamma; IL: interleukin-; KLK: human tissue kallikreins; MAC: membrane attack complex of complement; MIF: macrophage migration inhibitory factor; MMP: matrix metalloproteinases; MPs: microparticles; mRNA: messenger ribonucleic acid; OPG: osteoprotegerin; OSM: oncostatin M; PDGF: platelet-derived growth factor; PEDF: pigment epithelium-derived factor; PlGF: placental growth factor; sIL-2R: soluble IL-2 receptor; sIL-6R: sIL-6 receptor; SSc: systemic sclerosis; ST2: IL-1 receptor-related protein; Tie: soluble tyrosine kinase; TIMP2: tissue inhibitor of metalloproteinase 2; TNF-α: tumor necrosis factor alpha; TRAIL: TNF-related apoptosisinducing ligand; uPAR: urokinase-type plasminogen activator receptor; VEGF: vascular endothelial growth factor; VEGFR: vascular endothelial growth factor receptor

41