Human VE-Cadherin Fusion Protein as an Artificial Extracellular Matrix

Feb 9, 2016 - ACS Editors' Choice - This is an open access article published under ... vascular endothelial-cadherin extracellular domain and immunogl...
0 downloads 0 Views 26MB Size
Subscriber access provided by UNIV OSNABRUECK

Article

Human VE-cadherin fusion protein as an artificial extracellular matrix enhancing the proliferation and differentiation functions of endothelial cell Ke Xu, Qizhi Shuai, Xiaoning Li, Yan Zhang, Chao Gao, Lei Cao, Feifei Hu, Toshihiro Akaike, Jianxi Wang, Zhongwei Gu, and Jun Yang Biomacromolecules, Just Accepted Manuscript • DOI: 10.1021/acs.biomac.5b01467 • Publication Date (Web): 09 Feb 2016 Downloaded from http://pubs.acs.org on February 11, 2016

Just Accepted “Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted online prior to technical editing, formatting for publication and author proofing. The American Chemical Society provides “Just Accepted” as a free service to the research community to expedite the dissemination of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts appear in full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been fully peer reviewed, but should not be considered the official version of record. They are accessible to all readers and citable by the Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered to authors. Therefore, the “Just Accepted” Web site may not include all articles that will be published in the journal. After a manuscript is technically edited and formatted, it will be removed from the “Just Accepted” Web site and published as an ASAP article. Note that technical editing may introduce minor changes to the manuscript text and/or graphics which could affect content, and all legal disclaimers and ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors or consequences arising from the use of information contained in these “Just Accepted” manuscripts.

Biomacromolecules is published by the American Chemical Society. 1155 Sixteenth Street N.W., Washington, DC 20036 Published by American Chemical Society. Copyright © American Chemical Society. However, no copyright claim is made to original U.S. Government works, or works produced by employees of any Commonwealth realm Crown government in the course of their duties.

Page 1 of 36

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biomacromolecules

1

5

Human VE-cadherin fusion protein as an artificial extracellular matrix enhancing the proliferation and differentiation functions of endothelial cell

6

Ke Xu1, Qizhi Shuai1, Xiaoning Li1, Yan Zhang1, Chao Gao1, Lei Cao1, Feifei Hu1,Toshihiro

7

Akaike2, Jian-xi Wang3, Zhongwei Gu3, Jun Yang1#

2 3 4

8 9

(1The Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science,

10

Nankai University, Tianjin, 300071, China;

11

2Biomaterials Center for Regenerative Medical Engineering, Foundation for Advancement of

12

International Science, Tsukuba, Japan;

13

3National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610064,

14

China ;)

15 16 17 18 19 20 21 22 23 24

#Corresponding author.

25

Prof. Jun Yang, The Key Laboratory of Bioactive Materials, Ministry of Education, College of life

26

science, Nankai University, Tianjin, 300071, China

27

94 Weijin Road, Nankai District, Tianjin, China

28

E-mail address: [email protected]

29

Tel& Fax: +0086 22 23498038 1

ACS Paragon Plus Environment

Biomacromolecules

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

1

ABSTRACT

2

In an attempt to enhance endothelial cell (EC) capture and promote the

3

vascularization of engineered tissue, we biosynthesized and characterized the

4

recombinant fusion protein consisting of human vascular endothelial-cadherin

5

extracellular domain and immunoglobulin IgG Fc region (hVE-cad-Fc) to serve as a

6

bioartificial extracellular matrix (ECM). The hVE-cad-Fc protein naturally formed

7

homodimers and was used to construct hVE-cad-Fc matrix by stably adsorbing on

8

polystyrene (PS) plates. Atomic Force Microscop (AFM) assay showed uniform

9

hVE-cad-Fc distribution with nano-rod topography. The hVE-cad-Fc matrix markedly

10

promoted human umbilical vein endothelial cells (HUVECs) adhesion and

11

proliferation, with fibroblastoid morphology. Additionally, the hVE-cad-Fc matrix

12

improved HUVECs migration, vWF expression and NO release, which are closely

13

related to vascularization. Furthermore, the hVE-cad-Fc matrix activated endogenous

14

VE-cadherin/β-catenin proteins, and effectively triggered the intracellular signals such

15

as F-actin stress fiber, p-FAK, AKT and Bcl-2. Taken together, hVE-cad-Fc could be a

16

promising bioartificial matrix to promote vascularization in tissue engineering.

17 18 19 20 21 22

KEYWORDS: bioartificial extracellular matrix; fusion protein; VE-cadherin;

23

vascularization.

24

2

ACS Paragon Plus Environment

Page 2 of 36

Page 3 of 36

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biomacromolecules

1

1. Introduction

2

Tissue engineering holds enormous potential to replace or restore the function of

3

damaged or diseased tissue.1 While significant progress has been achieved in seeding

4

cells on polymer scaffolds, the inability to timely develop a sufficient and functional

5

blood vessel system mainly limits the growing of thick, 3-dimensional viable

6

engineered tissue in vitro survival and in vivo integration.2 Different tissue

7

engineering vascularization strategies have included material-based and cell-based

8

vascularization. These approaches have shown that scaffold surface modifications can

9

resist nonspecific protein or cell adhesion and improve endothelial-specific adhesion

10

to form an endothelial monolayer on the scaffolds. Consequently, there has been an

11

increasing interest in exploring biomaterials as bioartificial ECM for modifying the

12

surfaces of tissue engineering scaffolds and rapidly inducing endothelialization and

13

vascularization.3

14

In the last decades, ECM proteins (collagen, fibronectin and laminin),4

15

glycosaminoglycans (hyaluronic acid),5 ECM-derived peptides (RGDS, YIGSR and

16

REDV)6,7,8 and soluble signaling proteins (EGF, VEGF and FGF)9-11 have been

17

frequently used to coat the surfaces of various biomaterials by chemical conjugating

18

or physical adsorbing to augment their interaction with endothelial cells and

19

accelerate endothelialization on the scaffolds. More recently, with large scale

20

purification at high yields, new biological materials composed of recombinant

21

proteins are now being developed to create well-defined, multifunctional matrix using

22

common genetic engineering.12 Several recombinant proteins have been employed to

23

engineer bioartificial ECM, including structural proteins, growth factors and adhesion

24

proteins that are fused with ECM protein binding domain or immunoglobin G (IgG)

25

Fc domain.13-15 Fc-fusion proteins, which comprise an IgG Fc region linked

26

genetically to the target protein, have shown great promise as chimeric proteins with

27

both functional integrity and long-term stability.16,17 In a Fc-fusion protein, the Fc

28

domain can bind directly to the scaffold surface via hydrophobic interactions,

29

allowing the fused target protein to stretch out and interact with a suitable partner.18 In

30

our previous work, multiple Fc-fusion proteins, including epithelial (E)-cadherin-Fc, 3

ACS Paragon Plus Environment

Biomacromolecules

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

1

neuronal (N)-cadherin-Fc and cell growth factor-Fc, were established and used as

2

bioartificial ECM for biomaterial surface modification and shown to successfully

3

provide a biofunctional extracellular microenvironment for regulating cell

4

behavior.14,19-22

5

In an attempt to improve endothelialization and vascularization rates within

6

tissue engineering scaffolds, this study focused on designing a novel vascular

7

endothelial (VE)-cadherin-Fc fusion protein to enable the construction of the

8

bioactive interface via biomaterial surface modification. Cadherins are a large family

9

of transmembrane proteins with more than thirty members. VE-cadherin, an

10

endothelial specific cell-cell adhesion molecule, stabilizes endothelial junctions

11

through the homophilic binding of VE-cadherin extracellular regions.23 The adherens

12

junctions play a pivotal role in endothelial cell adhesion, proliferation, migration and

13

apoptosis, as well as the formation and maturation of the endothelium during

14

vascularization.23,24 Additionally, the adherens junctions, which are protein complexes,

15

contain large numbers of intracellular proteins that anchor to the actin cytoskeleton

16

and activate a series of intercellular signals.25 Given our previous work on E/N-cad-Fc

17

fusion proteins for culturing stem cells, we hypothesized that the extracellular domain

18

of VE-cadherin would be an ideal candidate enabling the development of a novel

19

multifunctional bioartificial ECM able to mimic adherens junctions and regulate

20

endothelial cell-to-cell, cell-ECM and cell-cell growth factor interactions.

21

In this study, hVE-cad-Fc was biosynthesized and applied to the surface

22

modification of biomaterials as a novel ECM. To begin, the hVE-cad-Fc protein was

23

identified and characterized via immobilization on polystyrene (PS) plates. Next, we

24

investigated the abilities of the hVE-cad-Fc matrix to promote adhesion, proliferation,

25

migration and differentiation in human umbilical vein endothelial cells (HUVECs) in

26

vitro.

27 28

2. Materials and methods

29

2.1. Construction and expression of the fusion protein hVE-cad-Fc

30

VE-cadherin extracellular domains from HUVECs were amplified by PCR to 4

ACS Paragon Plus Environment

Page 4 of 36

Page 5 of 36

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biomacromolecules

1

generate cDNA. The specific primer pair used for amplification was as follows:

2

5’-CCGGATATCATGCAGAGGCTCATGATGCTCC-3’

3

GCCGCTCTGGGCGGCCATATC-3’. The amplified VE-cadherin cDNA was

4

inserted into the EcoRV-NotӀ site of the pcDNA3.1 vector, which contained the

5

human IgG Fc domain (a gift from the Akaike research group). 293-6E cells were

6

grown in serum-free FreeStyleTM 293 Expression Medium (Life Technologies,

7

Carlsbad, CA, USA). The recombinant plasmid encoding hVE-cad-Fc was transiently

8

transfected into 293-6E cell suspension. At day 6, cell culture supernatants were

9

collected and used for purification, with target proteins captured on a Protein A CIP 5

10

ml column (GenScript, Cat.No.L00433), followed by a second SEC purification step.

11

The purified protein was analyzed by SDS-PAGE and Western blotting using 20 µL of

12

sample, with primary goat anti-human IgG-HRP utilized (GenScript, Cat. No.

13

A00166).

and

5’-AAGCG

14 15

2.2 Preparation of hVE-cad-Fc-, Collagen- and Poly-L-lysine- coated plates

16

The purified hVE-cad-Fc protein was diluted in PBS at different concentrations

17

and incubated on polystyrene (PS) plates. After 2 h incubation at 37 °C,the surface

18

was washed three times with PBS and incubated with 1.0% bovine serum albumin

19

(BSA) for 1 h to block nonspecific interactions. To prepare the control groups, PS

20

plates were treated with 1% Collagen Ӏ (BD, USA), 5µg/ml of poly-L-lysine (PLL)

21

(ScienCell, USA) or tissue culture treated polystyrene (TC-PS) plates (Corning

22

Corporation) for 2 h at 37 °C.

23 24

2.3 Assessment of hVE-cad-Fc immobilization onto PS plates

25

The quantities of immobilized hVE-cad-Fc on the plates were determined by

26

ELISA assay. Briefly, the coated plates were first incubated with 1.0 % (w/v) BSA for

27

1 h to block nonspecific interactions and then incubated with rabbit anti-VE-cadherin

28

antibody (1:1000, Cell Signaling Technology) for 2 h at 37 °C. After washing with

29

PBS, the plates were incubated with HRP-labeled goat anti-rabbit IgG (H+L)

30

antibody for an additional 2 h, followed by the addition of TMB solution, which was 5

ACS Paragon Plus Environment

Biomacromolecules

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

1

added for 15 min at 37 °C in darkness. Stop buffer was added, and the absorbance was

2

measured at 450 nm.

3

The amount of immobilized hVE-cad-Fc was calculated basing on the

4

quantification of the hVE-cad-Fc in the solution before and after immobilization with

5

a VE-cadherin ELISA kit (BD, USA).

6

For the water contact angle (WCA) analysis, 5 µl water was dripped onto the

7

hVE-cad-Fc-coated plates (5µg/ml) and other plates and measurements were obtained

8

using an optical contact angle meter (Dataphysics Inc, OCA20). For the adsorption

9

stability evaluation, PBS and culture medium were added to the hVE-cad-Fc-coated

10

plates (5µg/ml) for one week and then immobilized hVE-cad-Fc was measured by

11

ELISA assay. To examine the topography of the hVE-cad-Fc-coated (5µg/ml) surface,

12

the samples were characterized by atomic force microscope (AFM; NTEGRA Prima,

13

NT-MDT). The modified surface was dried with nitrogen for 1 h and the results were

14

analyzed using AFM nasoscope V7.2 version.

15 16 17

2.4 Cell culture HUVECs and endothelial cell medium were obtained from the ScienCell

18

Corporation (USA). Human vascular smooth muscle cells (HVSMCs; ScienCell, USA)

19

were cultured in DMEM-F12 supplemented with 10 % (v/v) fetal bovine serum.

20 21

2.5 HUVECs adhesion and proliferation assay

22

The HUVECs were seeded into a 96-well plate at a density of 1×104 cells/well.

23

After incubation for 4 h, 24 h or 48 h, the supernatants were removed and the cells

24

were washed three times with PBS. Cells were incubated with CCK-8 at 37 °C for 4 h

25

and read on a micro-plate reader (Biotech, MQX200) at an absorbance of 450nm. For

26

inhibition experiments, cells were pre-cultured with anti-VE-cadherin antibody (Cell

27

Signaling Technology) at 37 °C for 30 min and seeded onto Collagen, PLL or

28

hVE-cad-Fc-coated (5µg/ml) plates. Non-adherent cells were removed after 4 h by

29

rinsing with PBS and the remaining HUVECs were quantified using a CCK-8 assay.

30

Calcium-dependent adhesion was evaluated by incubating cells in EDTA, EGTA and 6

ACS Paragon Plus Environment

Page 6 of 36

Page 7 of 36

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biomacromolecules

1

MgSO4 at final concentrations of 5 mM for 30 min prior to the CCK-8 assay.

2 3

2.6 Nitric oxide release assay

4

Supernatants were collected from different substrate surfaces after HUVECs

5

culturing and nitric oxide (NO) concentrations were determined using Total Nitric

6

Oxide Assay Kit (Beyotime, China) according to the manufacturer's instructions;

7

absorbance was measured at 540 nm (n=3).

8 9

2.7 Cell migration assay

10

Cell suspensions were seeded onto 24 well AggreWellTM plates (Stem Cell, USA)

11

and then centrifuged at 1000 rpm for 5 min. The cells were subsequently cultured for

12

16 h to form 3D HUVECs aggregates. The cell aggregates were then quickly

13

transferred onto hVE-cad-Fc, Collagen, PLL or TC-PS matrices. After 4 h, cellular

14

spreading and crawling from the aggregates were observed by microscopy.

15 16

2.7 Western blotting analysis

17

Total protein was extracted by adding lysis buffer [10 mM Tris (pH 7.4), 150 mM

18

NaCl, 1% Triton X -100, 1% sodium deoxycholate, 0.1% SDS, 10 mM EDTA and

19

protease inhibitor cocktail] to the cell cultures while on ice. The cells were disrupted

20

with the assistance of a cell scraper and lysates were centrifuged at 13,000 rpm for 10

21

min at 4 °C. The supernatants were collected and protein concentrations were

22

determined using BCA assay (Beyotime, China). Samples were then separated by

23

electrophoresis using ~8 – 12% SDS-polyacrylamide gels, transferred onto

24

polyvinylidene difluoride membranes (Roche, USA) and incubated with one of the

25

following primary antibodies: rabbit anti-VE-cadherin antibody (1:1000, Cell

26

Signaling Technology), anti-rabbit von Willebrand factor antibody (1:2000, Abcam),

27

rabbit anti-Phospho-Akt (Ser473) antibody (1:1000, Cell Signaling Technology),

28

rabbit anti-Bcl-2 antibody (1:1000, Gene Tex), rabbit anti-Phospho-FAK antibody

29

(Tyr397) (1:1000, Cell Signaling Technology), rabbit anti-β-actin antibody (1:1000,

30

Cell Signaling Technology) or mouse anti-α/β/p120-catenin antibody (1:1000, BD 7

ACS Paragon Plus Environment

Biomacromolecules

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

1

Transduction Laboratories). The membranes were then incubated with HRP-labeled

2

goat anti-rabbit IgG (H+L) antibody (1:1000, Beyotime) and washed with PBS. All

3

bands were quantified using Quantity One v.4.62 software.

4 5

2.8 Cell cytoskeleton staining

6

After 4h, 24h or 48h of culture, the cells were fixed with 4 % (v/v)

7

paraformaldehyde for 10 min and incubated with 0.1% (v/v) Triton X-100 for 10 min

8

at 37 °C. Cells were then blocked with 1.0% (w/v) BSA for 30 min at 37 °C, stained

9

with FITC-phalloidin/propidium iodide and the nuclei were counterstained with DAPI.

10

Staining was assessed using a confocal laser scanning microscope (TCS SP8, Leica).

11 12

2.9 Immunofluorescence staining

13

Cells were fixed with 4% (v/v) paraformaldehyde for 10 min and then blocked

14

with 1% (w/v) BSA for 30 min at 37 °C. The samples were then incubated with

15

primary rabbit VE-cadherin antibody (1:100, Abcam), washed with PBS and

16

subsequently incubated with goat anti-rabbit antibody conjugated with Alexa

17

Fluorophore 488 and 594 (Invitrogen) at room temperature for 1 h. All samples were

18

examined by confocal laser scanning microscopy (TCS SP8, Leica).

19 20

2.10 Immunoprecipitation

21

HUVECs were lysed in lysis buffer [1% Triton X-100, Tris-HCl (pH 7.5), 100

22

mM NaF and 1.0 % NP-40] and cleared by centrifugation at 12,000 rpm for 10 min at

23

4 °C. The supernatants were collected, incubated with FAK-specific antibody (1:100,

24

Abcam) for 6 h and then protein A/G plus agarose beads (Beyotime, China) for 2 h at

25

4 °C. The immunocomplexes were washed three times, boiled in 1x sample loading

26

buffer and electrophoresed on sodium dodecyl sulfate-polyacrylamide gels for protein

27

analysis.

28 29 30

2.11 Statistical analyses GraphPad Prism Version 5.0 software for windows (GraphPad, San Diego, CA) 8

ACS Paragon Plus Environment

Page 8 of 36

Page 9 of 36

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Biomacromolecules

1

was used for statistical analysis. Data analysis was performed by one-way analysis of

2

variance (ANOVA). The minimum significance level was set at * p < 0.05 and **p