Vous êtes sur la page 1sur 10

Acta Biomaterialia 44 (2016) 178–187

Contents lists available at ScienceDirect

Acta Biomaterialia
journal homepage: www.elsevier.com/locate/actabiomat

Full length article

Macroporous biohybrid cryogels for co-housing pancreatic islets with


mesenchymal stromal cells
Danielle J. Borg a,1, Petra B. Welzel b,1, Milauscha Grimmer b, Jens Friedrichs b, Marc Weigelt a,
Carmen Wilhelm a, Marina Prewitz b, Aline Stißel b, Angela Hommel a, Thomas Kurth c,
Uwe Freudenberg b, Ezio Bonifacio a,⇑, Carsten Werner b,⇑
a
Preclinical Approaches to Stem Cell Therapy, DFG-Center for Regenerative Therapies Dresden, Dresden University of Technology, Fetscherstrasse 105, Dresden 01307, Germany
b
Leibniz Institute of Polymer Research Dresden, Max Bergmann Center of Biomaterials Dresden, Hohe Str. 6, 01069 Dresden, Germany
c
Electron Microscopy Facility, DFG-Center for Regenerative Therapies Dresden, Dresden University of Technology, Fetscherstrasse 105, Dresden, 01307, Germany

a r t i c l e i n f o a b s t r a c t

Article history: Intrahepatic transplantation of allogeneic pancreatic islets offers a promising therapy for type 1 diabetes.
Received 11 March 2016 However, long-term insulin independency is often not achieved due to severe islet loss shortly after
Received in revised form 19 July 2016 transplantation. To improve islet survival and function, extrahepatic biomaterial-assisted transplantation
Accepted 5 August 2016
of pancreatic islets to alternative sites has been suggested. Herein, we present macroporous, star-shaped
Available online 6 August 2016
poly(ethylene glycol) (starPEG)-heparin cryogel scaffolds, covalently modified with adhesion peptides,
for the housing of pancreatic islets in three-dimensional (3D) co-culture with adherent mesenchymal
Keywords:
stromal cells (MSC) as accessory cells. The implantable biohybrid scaffolds provide efficient transport
Pancreatic islet
Mesenchymal stromal cell
properties, mechanical protection, and a supportive extracellular environment as a desirable niche for
Cryogel the islets. MSC colonized the cryogel scaffolds and produced extracellular matrix proteins that are impor-
Heparin tant components of the natural islet microenvironment known to facilitate matrix-cell interactions and to
Poly(ethylene glycol) prevent cellular stress. Islets survived the seeding procedure into the cryogel scaffolds and secreted insu-
lin after glucose stimulation in vitro. In a rodent model, intact islets and MSC could be visualized within
the scaffolds seven days after subcutaneous transplantation. Overall, this demonstrates the potential of
customized macroporous starPEG-heparin cryogel scaffolds in combination with MSC to serve as a mul-
tifunctional islet supportive carrier for transplantation applications.

Statement of Significance

Diabetes results in the insufficient production of insulin by the pancreatic b-cells in the islets of
Langerhans. Transplantation of pancreatic islets offers valuable options for treating the disease; however,
many transplanted islets often do not survive the transplantation or die shortly thereafter. Co-
transplanted, supporting cells and biomaterials can be instrumental for improving islet survival, function
and protection from the immune system. In the present study, islet supportive hydrogel sponges were
explored for the co-transplantation of islets and mesenchymal stromal cells. Survival and continued func-
tion of the supported islets were demonstrated in vitro. The in vivo feasibility of the approach was shown
by transplantation in a mouse model.
Ó 2016 Acta Materialia Inc. Published by Elsevier Ltd. All rights reserved.

1. Introduction
⇑ Corresponding authors.
E-mail addresses: danielle.borg@mater.uq.edu.au (D.J. Borg), welzel@ipfdd.de
(P.B. Welzel), grimmer@ipfdd.de (M. Grimmer), friedrichs@ipfdd.de (J. Friedrichs), Pancreatic islet transplantation therapy is a viable option for
marc.weigelt@crt-dresden.de (M. Weigelt), carmenzitawilhelm@web.de type 1 diabetic individuals [1]. However, maintaining long-term
(C. Wilhelm), mc.prewitz@googlemail.com (M. Prewitz), alinestissel@web.de graft efficacy remains challenging since the current intrahepatic
(A. Stißel), angela.hommel@crt-dresden.de (A. Hommel), thomas.kurth@ portal vein transplant site is prone to mechanical stress and
crt-dresden.de (T. Kurth), freudenberg@ipfdd.de (U. Freudenberg), ezio.bonifacio@
crt-dresden.de (E. Bonifacio), werner@ipfdd.de (C. Werner).
inflammation associated with the risk of islet capsule rupture
1
Danielle J. Borg and Petra B. Welzel share first authorship. and prolonged ischemia [2–5]. Furthermore, islet endocrine cells

http://dx.doi.org/10.1016/j.actbio.2016.08.007
1742-7061/Ó 2016 Acta Materialia Inc. Published by Elsevier Ltd. All rights reserved.
D.J. Borg et al. / Acta Biomaterialia 44 (2016) 178–187 179

frequently undergo anoikis during islet isolation due to detach- and heparin (starPEG-heparin cryogels) [31–33] with adhesion-
ment from their natural extracellular matrix (ECM) in the pancreas mediating peptide ligands to house islets and MSC together in a
[6–8]. Thus, transplantation of islets into alternate sites and inhibi- defined, multifunctional microenvironment. The architecture of
tion of inflammation and anoikis during and after transplantation this cryogel system with interconnected macropores, allows for
are of particular interest to improve survival and function. unhindered exchange of nutrients, and affords mechanical protec-
Related efforts mostly focus on the immunoisolation of islets in tion, 3D spatial distribution and retention of MSC and islets. MSC
diabetic animal models by encapsulation of single insulin- attach to the adhesion peptides bound to the cryogel matrix and
producing beta cells or whole islets in natural or synthetic bioma- further improve the presented artificial 3D niche by secretion of
terials [9]. However, encapsulation of islets for protection against ECM proteins, enabling proper cell-matrix interactions (Fig. 1).
the immune response was found to hinder their revascularization Our results show that islets remain as functional units in the cryo-
after transplantation, resulting in graft hypoxia and attrition gel scaffolds verified by the secretion of insulin in response to glu-
[10,11]. Macroporous cell scaffolds are used to overcome these cose stimulation in vitro. Subcutaneous transplantation into mice
limitations by providing mechanical protection, enabling exchange confirmed the principal feasibility of the approach.
of oxygen, nutrients, and metabolites, as well as release of insulin
from the pancreatic beta cells [12]. In addition, macroporous mate-
rials facilitate cell infiltration from the surrounding tissue includ- 2. Materials and methods
ing neovascularization [13]. Scaffolds prepared from natural
[14,15], synthetic [12,16–18] and mixtures of natural and synthetic 2.1. Preparation and characterization of biofunctional starPEG-
precursors [19–21], have been successfully applied in housing heparin cryogel scaffolds
islets for periods of up to 60 days ex vivo without affecting cell
survival. The fabrication of starPEG-heparin cryogel scaffolds with inter-
Beyond immunoisolation or scaffolding, biomaterials can sup- connected macropores has been described elsewhere [31,32].
port transplanted islets with the localized provision of bioactive Briefly, network formation via chemical crosslinking (EDC/sulfo-
molecules, such as peptide sequences and ECM proteins, and/or NHS chemistry) of amino terminated 4-arm poly(ethylene glycol
accessory cells [22–25]. Mesenchymal stromal cells (MSC) define (starPEG, Mw 10,000 Da, JenKem Technology, USA) and heparin
a promising target, since these accessory cells have been shown (Porcine intestinal heparin sodium salt; Mw 14,000 Da; Cal-
to exert beneficial immunomodulatory, pro-angiogenic, and anti- biochem, Darmstadt, Germany) was combined with cryogelation
apoptotic effects which improved diabetes outcome when co- technology [34–37]. Architectural and mechanical scaffold proper-
transplanted with islets in animal models [26–28]. Moreover, ties can be modulated by altering the concentrations of either poly-
MSC are known to secrete ECM proteins in response to environ- mer precursors or their molar ratio and also freezing temperature
mental stimulation [29,30]. [31,32]. starPEG-heparin cryogel materials were adjusted to com-
Here, we customized a recently introduced macroporous hydro- bine high porosity and appropriate pore size with suitable mechan-
gel system based on star-shaped poly(ethylene glycol) (starPEG) ical properties by varying cryogelation processing conditions

Fig. 1. Representative SEM image of the dry cryogel (center) with schematic representation of the underlying starPEG-heparin network (bottom left) and representative
digital image of dry cryogel cylinder and discs (top left). The scaffold was designed to offer mechanical protection and allow 3D distribution and attachment of islets and MSC
accessory cells via bioactive surfaces (RGD peptide) within pores to support continued cell survival. MSC further biofunctionalized the cryogel scaffold by secretion of ECM
proteins. The interconnected pore system allows for the exchange of nutrients, oxygen and waste as well as for the transport of insulin released from the pancreatic beta cells
via convection.
180 D.J. Borg et al. / Acta Biomaterialia 44 (2016) 178–187

[31,32]. For the present study a molar ratio of starPEG to heparin of Information) using Image J software for image processing. The
c = 3 and a total precursor concentration of 4.3% (w/w) was used, mean pore size and the pore size distribution were obtained by
resulting in a material with appropriate architecture and stiffness quantifying the dimensions of more than 1,200 pores using discs
for housing of islets and MSC and subcutaneous transplantation. from 3 different cylinders. In addition, cryogel scaffolds were infil-
Gels were fluorescently labeled by mixing heparin with 1% (w/w) trated with poly(L-lysine) coated, Alexa FluorÒ 633-conjugated
of Alexa FluorÒ 488- or Alexa FluorÒ 405-labeled heparin (prepared pancreatic islet-sized glass beads (£ 125 and 250 lm, MO-SCI Spe-
from Alexa FluorÒ 488 or Alexa FluorÒ 405, respectively, Gibco, cialty Products, Rolla, MO, provided by Dr. Sophie Pautot, Center
UK). Specifically, starPEG and heparin were each dissolved in for Regenerative Therapies Dresden, Germany) to test their appli-
one-third of the total volume of deionized, decarbonized water cability for housing islets. Uniaxial compression tests were utilized
(Milli-Q) on ice. After treating the solutions in an ultrasonic bath to achieve the global (bulk) mechanical properties of the material.
filled with ice-water for approximately 3 5 min, they were kept Moreover, the local mechanical properties (stiffness of the cryogel
on ice. Applying the same procedure, EDC and sulfo-NHS (molar struts) were characterized by an AFM nanoindentation approach
ratio of EDC/sulfo-NHS of 2:1) were each dissolved in one-sixth on thin cryogel sections (see Supplementary Information).
of the total volume of ice-cold Milli-Q. Based on the amount of Cryogel scaffolds used for in vitro and in vivo studies were 2 mm
NH2 groups of starPEG, a 2-fold molar excess of EDC was used. in height and 8 mm in diameter in the PBS swollen state. To
The EDC and sulfo-NHS solutions were mixed with the heparin improve cell adhesion, the starPEG-heparin cryogel scaffolds were
solution, and incubated for 10 min on ice to activate the heparin biofunctionalized with a RGD (cyclo(Arg-Gly-Asp-D-Tyr-Lys)) con-
carboxylic groups. Finally, the starPEG solution was added to the taining peptide sequence (Peptides International, USA) as
activated heparin and mixed for 15 s on a vortexer (Minishaker described [32]. The PBS swollen scaffolds were first sterilized in
MS2, IKA, Germany). The resulting reaction mixtures were added Proclin/0.02% PBS (Supelco, USA) overnight and washed three
to the cavities of 96-well plates (350 ll per well); frozen at times with fresh PBS. After another washing step (three times)
20 °C overnight, and lyophilized for 24 h. The produced cryogel with 67 mM phosphate buffer (pH 5) at 4 °C, carboxylic acid groups
cylinders were cut into discs, punched to the desired diameter of heparin were activated with EDC/sulfo-NHS solution (50 mM
using a punching tool (Hoffmann GmbH Qualitätswerkzeuge, Ger- EDC, 25 mM sulfo-NHS in 67 mM phosphate buffer (pH 5)) for
many), and were finally swollen and repeatedly washed in phos- 1 h. Subsequently, the scaffolds were washed three times in borate
phate buffered saline (PBS, pH 7.4). Determination of swelling buffer (100 mM, pH 8, 4 °C) to remove unbound EDC/sulfo-NHS
properties, and 3D architecture of the cryogel is reported else- and then incubated in 500 lL cyclo(Arg-Gly-Asp-D-Tyr-Lys)-
where [31,32]. Pore sizes were determined from cross-sectional solution (160 lg/mL; dissolved in borate buffer) for 3 h at room
confocal images of Alexa FluorÒ 405-fluorescently labeled PBS temperature. Finally, all scaffolds were washed in PBS three times,
swollen cryogels (Fig. 2A; methods described in the Supplementary and maintained in cell culture medium until use.

Fig. 2. Properties of the PBS swollen cryogel scaffolds. (A) Representative scanning confocal image of an Alexa FluorÒ 405-labeled cryogel section (blue). Scale bar = 500 lm
(B) Representative uniaxial compression stress strain curve. (C) Pore size distribution (2426 pore dimensions were measured from n = 3 scaffolds). Refer to Supplementary
Information for more details. (D) Representative digital image of a PBS swollen cryogel disc. (For interpretation of the references to colour in this figure legend, the reader is
referred to the web version of this article.)
D.J. Borg et al. / Acta Biomaterialia 44 (2016) 178–187 181

2.2. Experimental animals Karnovsky (2% paraformaldehyde/2% glutaraldehyde in 50 mM


HEPES) followed by 1% osmium tetroxide in PBS, dehydrated in
C57BL/6 J and B6.Cg-Tg(Ins1-EGFP)1Hara/J mice were pur- a graded series of ethanol, critically point dried using a CPD
chased from Jackson Laboratories (Jackson Laboratories, USA) and 030 (Leica Microsystems, Austria), and coated with gold using a
maintained and sacrificed for MSC or islet isolation, respectively, sputter coater device (SCD 050; Leica Microsystems, Austria).
or transplanted and sacrificed 7 days after transplantation in accor- Samples were analyzed with a TM 1000 tabletop SEM (Hitachi
dance with the institutional animal ethics guidelines (24D- High-Technologies Europe GmbH, Germany) or a JSM 7500F cold
9168.11-1/2012-50). field emission SEM (Jeol GmbH, Germany). To determine whether
MSC and islets penetrated the whole scaffolds, cryogels were
2.3. MSC isolation and expansion fixed as described for SEM analysis and were dehydrated in etha-
nol, infiltrated and embedded in the glycol methacrylate resin
MSC were isolated from bone marrow aspirates from the femur TechnovitÒ 7100 (Heraeus-Kulzer, Germany). Sections (2 lm)
and tibia of 9–10 week old C57BL/6 J donor mice. Passage 0 MSC were stained with 1% toluidine blue/0.5% borax and multiple
were cultured for 3–4 weeks in MSC medium (IMDM medium sup- images were captured using a Biozero 8000 light microscope
plemented with 10% fetal calf serum (Gibco, USA), 10% equine and tile images were stitched into larger mosaics using the image
serum (Hyclone Laboratories, USA), 1% L-glutamine (Lonza, merge function of the associated BZ Analyzer software (Keyence,
Switzerland), 1% antibiotic-antimycotic (Gibco, USA)) at 37 °C/5% Germany).
CO2 with a medium change after 24 h of culture and every 3–
4 days thereafter. MSC were subcultured using 0.25% trypsin/1 mM
EDTA (Gibco, USA) when 60–80% confluent until passage 10, at a 2.7. Characterization of extracellular matrix deposition within cell-
cell density of 2,500 cells/cm2. MSC were used for experiments at laden cryogels in vitro
passage 8–10. MSC were characterized by flow cytometry as previ-
ously reported [27]. MSC were able to differentiate in vitro into Seeded scaffolds were washed in PBS, fixed in cold 4% forma-
chondrocytes, osteoblasts and adipocytes as determined by aggre- lin, submerged in cold 30% sucrose overnight, and frozen in liquid
can, osteopontin and BODIPY staining, respectively [27]. nitrogen. Cryogels were sectioned (5 lm; Cryostat) from the top
seeding surface until cells were observed under a light micro-
2.4. Islet isolation scope. Serial cryosections (5 lm) throughout the cryogels were
stained with anti-laminin IgG (Sigma-Aldrich, USA) and anti-
Islets were isolated from the pancreas of 8–9 week old B6.Cg-Tg fibronectin IgG (Rockland Immunochemicals Inc, USA) in
(Ins1-EGFP)1Hara/J or 9–10 week old C57BL/6 J donor mice by col- PBS/0.1% Tween for 1 h at room temperature and thereafter
lagenase digestion (0.7 mg/mL; Sigma-Aldrich, USA) and purified labeled with respective secondary antibodies (Alexa FluorÒ 546;
using a discontinuous Ficoll density gradient, as described previ- Invitrogen, USA) for 1 h at room temperature. Cells were stained
ously [27]. Islets were washed with islet medium: RPMI-1640 for actin using Phalloidin-Alexa FluorÒ 633 (Invitrogen, USA) for
medium supplemented with 1% (v/v) L-glutamine (Sigma-Aldrich, 30 min at room temperature. Images were acquired using a SP5
USA), 1% penicillin-streptomycin (Sigma-Aldrich, USA), 5.5 mM laser scanning confocal microscope (Leica Microsystems,
glucose (Sigma-Aldrich, USA) and 5% fetal calf serum (Gibco, Germany).
USA). Isolated, free-floating islets were rested 18–24 h prior to
seeding into the cryogel scaffolds at 37 °C/5% CO2. 2.8. Analysis of MSC and islet viability within cell-laden scaffolds
in vitro
2.5. Seeding of MSC and islets into cryogel scaffolds
Cryogel scaffolds were washed with PBS and submerged in
Islets were seeded into cryogels as follows. The cryogel scaffolds 1 lM of Calcein-AM and EthD-1, provided in the LIVE/DEADÒ Via-
were placed on sterile filter paper (to remove culture medium). bility/Cytotoxicity Kit (Invitrogen, USA) and incubated in the dark,
One hundred islets in 250 lL of islet medium were seeded on top for 30 min at 37 °C/5% CO2. Cryogels were washed in PBS and its
of the scaffold that was then placed in a 24-well plate to allow top region and bottom site imaged within 30 min 1 h after stain-
the islets to recover at 37 °C/5% CO2, for 30 min, prior to adding ing using a Leica SP5 multi-photon laser scanning confocal micro-
islet medium. scope (Leica Microsystems, Germany). For the top region (93–
MSC were seeded into cryogel scaffolds in a two-step process. 195 lm from the top seeding surface), z stacks (approximately
The cryogels were placed on sterile filter paper and 250,000 MSC every 10 lm) were deconvoluted using Imaris software and were
in 500 lL MSC medium were seeded on top of the scaffold. Cryo- reported as maximum projections or 3D projections (see Supple-
gels were then placed in a 24-well plate to allow MSC to attach mentary Information).
at 37 °C/5% CO2, for 1 h. After this time, a second suspension of
250,000 MSC in 500 lL medium was added to the top of the cryo-
gel. Newly seeded MSC were allowed to attach at 37 °C/5% CO2, for 2.9. In vitro islet function: glucose-stimulated insulin secretion
1 h, before filling the well with MSC medium.
Cryogels containing MSC and islets were prepared as follows. The glucose stimulated insulin secretion assay involved the
First, MSC were seeded into the scaffolds as described above. Cryo- static resting of islets ± MSC inside cryogels, or freely sus-
gels were cultured for 3 days with daily medium changes. There- pended, in resting buffer containing a glucose concentration of
after, islets were seeded as described above and were analyzed 2.8 mM for 2 h and subsequent stimulation of islets in a buffer
or transplanted 2 h after islet seeding. containing a high glucose concentration (25 mM) as previously
described [38]. Secreted insulin was detected from supernatants
2.6. Analysis of MSC and islet distribution within cell-laden scaffolds using a commercial mouse insulin ELISA kit (Crystal Chem Inc.,
in vitro USA) according to the manufacturers’ instructions. A stimulation
index was calculated as the ratio of the insulin released during
For scanning electron microscopy (SEM), scaffolds were first high glucose to the insulin release during low glucose
fixed with 4% formalin and subsequently post-fixed in modified treatment.
182 D.J. Borg et al. / Acta Biomaterialia 44 (2016) 178–187

2.10. Co-transplantation of cell-laden cryogel scaffolds into a rodent demonstrated by scanning electron microscopy (SEM, Fig. 1). The
model scaffolds prepared from a reaction mixture with a molar ratio of star-
PEG to heparin of c = 3 and a total precursor concentration of 4.3%
Nine week old anesthetized (100 mg/kg Ketamine and 10 mg/kg (w/w) using a freezing temperature of 20 °C, exhibited a high total
Xylazine) C57BL6/J mice were transplanted subcutaneously with porosity of approximately 98%, determined from mass and volume
Alexa FluorÒ 405-labeled cryogels containing no cells (n = 3), 100 swelling data [31]. The mean pore size in the PBS swollen state, mea-
green fluorescent protein (GFP) positive islets (n = 3) or 100 GFP+ sured from cross-sectional confocal images of fluorescently labeled
islets plus 500,000 MSC (passage 8–10) labeled with a cell tracker cryogels (Fig. 2A), was 228 ± 130 lm (pore size distribution is shown
dye (CM-DiI; Invitrogen, USA) (n = 3). Mice were monitored daily in Fig. 2C). Uptake of fluorescently labeled model glass beads with
and 7 days post-transplant, mice were sacrificed and cryogel diameters (125 and 215 lm) comparable to murine islets (50–
implants were removed. Scaffolds were washed, fixed, frozen and 250 lm) was demonstrated (Fig. S1), suggesting that the scaffolds
sectioned as described above. Fluorescent images were acquired should allow for infiltration and housing of whole islets.
randomly throughout the cryogel on an upright Leica TCS SP5 con- A global (bulk) Young’s modulus of 0.6 ± 0.1 kPa was calculated
focal system (Leica Microsystems, Germany). from the linear elastic part of stress-strain curves that were
obtained by uniaxial compression of the whole PBS swollen cryogel
2.11. Statistical analysis scaffolds (Fig. 2B). The local Young’s modulus of the pore walls
(struts) was determined by an AFM-based nanoindentation
Statistical analyses were performed using GraphPad Prism soft- approach [31,34] and was found to be 68 ± 38 kPa (a representative
ware (La Jolla, USA). Data with a Gaussian normal distribution are force-distance curve is given in Fig. S2).
reported as mean values with standard deviation. P values were As previously demonstrated, the starPEG-heparin hydrogel plat-
determined by the unpaired Student’s t-test or one-way ANOVA. form can be readily functionalized with a variety of adhesion-
Comparison of insulin secretion under multiple conditions was mediating peptides and glycosaminoglycan-binding growth factors
performed using the general linear model. A two-tailed p value [34,35]. Herein, the scaffolds (Fig. 2D) were additionally modified
of <0.05 was considered significant. Changes in weight post- by conjugating a RGD (Arg-Gly-Asp)-containing peptide sequence
transplant were analyzed using linear regression and slope com- (Fig. 1) via carbodiimide chemistry to remaining carboxylic acid
parison. Overall slopes were pooled if no significant differences functionalities of the heparin component of the gels for anchoring
were observed. MSC to the macropore walls prior to islet loading.

3. Results 3.2. Co-housing of islets and MSC within cryogel scaffolds in vitro

3.1. Engineering and characterization of macroporous starPEG- The potential of the cryogel scaffolds to co-house islets and MSC
heparin cryogel scaffolds accessory cells was tested in a series of in vitro experiments. Mouse
bone marrow-derived MSC were seeded on RGD-functionalized,
Macroporous starPEG-heparin cryogel scaffolds were prepared starPEG-heparin cryogel scaffolds to investigate cell attachment
using chemical crosslinking (EDC/sulfo-NHS chemistry) of amino and survival. Subsequently, the infiltration of mouse pancreatic
terminated starPEG and heparin in aqueous solution at sub-zero islets into empty scaffolds and scaffolds pre-cultivated with MSC
temperatures as previously described [31,32,34,35]. Lyophilization for 3 days was studied.
of the incompletely frozen gel (Fig. 1) resulted in sponge-like When seeded into the cryogel scaffolds, MSC maintained a
materials with a system of interconnected macropores as fibroblast-like morphology and remained viable after 3 days of

Fig. 3. Islets and MSC survive in cryogels in vitro. Live (green; Calcein AM+) and dead (red; Ethidium homodimer-1(EthD-1)+) staining reveals minimal cell death after (A)
3 days of MSC culture, (D) 2 h of islet culture or (G) 2 h of islet and 3 day cultured-MSC co-culture in cryogels (blue); maximum intensity confocal images (deconvoluted
maximum projections of Z stacks ranging from 93 lm to 195 lm from the top seeding surface of the cryogel); (scale bar = 100 lm; n = 3). Representative scanning electron
images on the periphery of the cryogels (n = 3) show flat multilayered MSC (B; scale bar = 10 lm) with several protrusions and deposition of extracellular matrix (C; scale
bar = 10 lm), whole islets (E; scale bar = 10 lm) with intact microvilli and cilia indicated by the black arrowheads (F; scale bar = 10 lm) and islets surrounded by MSC (H;
scale bar = 10 lm) with intact microvilli after seeding (I; scale bar = 10 lm). A representative scanning electron image of a cryogel seeded with islets and MSC demonstrates
the presence of islets (asterisks) which are surrounded by MSC (dotted line), within the pores of the cryogel (J, scale bar = 100 lm). (For interpretation of the references to
colour in this figure legend, the reader is referred to the web version of this article.)
D.J. Borg et al. / Acta Biomaterialia 44 (2016) 178–187 183

in vitro culture as demonstrated by the large proportion of Calcein When seeded in scaffolds pre-cultivated with MSC, islets were
AM+ cells (Fig. 3A, S3A and S4A). SEM revealed multiple layers of found within the macropores adjacent to the adherent MSC
MSC exhibiting a flattened morphology within the macropores (Fig. 3H and J), with villi and microvilli present on the endocrine
(Fig. 3B). cells of the islet (Fig. 3I) as demonstrated by SEM images. Scanning
Deposition of extracellular matrix in the MSC-seeded cryogel confocal images of the stained cell-laden scaffolds (LIVE/DEADÒ
scaffolds was observed by SEM (Fig. 3C) and the matrix structures Viability/Cytotoxicity kit) illustrate that most islets and MSC sur-
were shown to contain laminin and fibronectin as visualized by vived and maintained their spheroid or fibroblast-like morphology,
immunochemistry staining (Fig. 5A and 5B). respectively, within the macropores of the cryogel (Fig. 3G, S3C and
SEM and scanning confocal images revealed that intact islets S4C). The results suggest that the cryogel scaffolds may keep the
survived the seeding procedure, entered the macropores, and spa- accessory MSC in close proximity to transplanted islets.
tially distributed within the sponge-like cryogel material (Fig. 3, To better observe how far MSC and islets could penetrate into
S3B and S4B). The cryogel-supported islets exhibited minimal the cryogel, co-seeded cryogels were embedded in TechnovitÒ gly-
rounding of individual endocrine cells (Fig. 3E) with villi and col methacrylate resin and thin sections (2 lm) were stained with
microvilli present, supporting contact between neighbouring toluidine blue. MSC attached homogenously throughout the entire
endocrine cells (Fig. 3F) and remained viable with little cell death thickness of the 2 mm thick cryogel scaffold (Fig. 4A). Even after
(Fig. 3D, G, S3B and S4B). seeding of only 100 islets onto the rather large scaffold (2 mm

Fig. 4. Islets and MSC distribute throughout the scaffold. Representative light microscopy images of islet/MSC co-cultures in starPEG-heparin cryogels scaffolds (purple)
stained with toluidine blue after embedding in resin and sectioning (2 lm, n = 1 cryogel). (A) MSC (grey; spindle) are attached throughout an entire cross-section after 3 days
of culture within the scaffold. Asterisk represents the top of the cryogel. Scale bar = 50 lm. In (B) and (C) islets are visible (grey-blue; indicated by asterisk) in a depth of
550 lm (B) and 625 lm (C) from the top seeding surface. Scale bar = 20 lm. (For interpretation of the references to colour in this figure legend, the reader is referred to the
web version of this article.)

Fig. 5. Representative images taken throughout Alexa FluorÒ 405-labeled cryogels (blue) seeded with MSC secreting (A) laminin (green) or (B) fibronectin (green) after three
days of in vitro culture. The actin cytoskeleton of the MSC is labeled in red (scale bar = 50 lm; n = 3; sections: 5 lm). (For interpretation of the references to colour in this
figure legend, the reader is referred to the web version of this article.)
184 D.J. Borg et al. / Acta Biomaterialia 44 (2016) 178–187

thickness, 8 mm diameter), we were able to detect islets at a depth 4. Discussion


of 550 and 625 lm from the top seeding surface of the cryogel
(Fig. 4B and C, respectively), which is 3- to 4-fold of an islet average Transplantation of pancreatic islets for type 1 diabetic individu-
size or one-third to one-fourth of the total cryogel thickness. als can delay or stabilize diabetic complications by providing
appropriate glycemic control [1]. However, the poor long-term
3.3. Islet function within cryogel scaffolds in vitro effectiveness of transplanted islets directs current research
towards engineering superior transplant microenvironments
Insulin secretion was assessed by glucose stimulation of the [10,12] in order to overcome central problems like the lack of vas-
islets 2 h after cryogel housing (Fig. 6A). Islets maintained basal culature and ECM attachment/integrin ligation [25].
insulin secretion in low glucose either freely suspended Here, we applied a recently developed macroporous, starPEG-
(32.6 ± 26.6 ng/mL) or housed in cryogels (59.2 ± 42.2 ng/mL, heparin cryogel material [31–33] and MSC as a synergistic strategy
p = 0.25). Islets were able to secrete insulin in stimulating condi- to design multifunctional 3D islet-supportive carriers. Since pan-
tions in cryogels, similar to that of freely suspended islets creatic islets of a given species are not uniform in size, the broad
(84.2 ± 42 ng/mL; suspension vs 158.7 ± 105.3 ng/mL; cryogel). pore size distribution (Fig. 2C) of the cryogel scaffolds was consid-
Co-culture of MSC did not impede basal (69.9 ± 64.9 ng/mL) or ered advantageous for an optimal uptake. Figs. 2–4 as well as
stimulated (155.4 ± 73.8 ng/mL) insulin secretion in suspension, Figs. S1, S3 and S4 clearly demonstrate that the size of the macro-
and insulin secretion was similar when islet and MSC were co- pores is large enough and the interconnectivity is sufficient to
housed in cryogels (basal, 42.7 ± 29.5 ng/mL; stimulated, allow for 3D distribution and housing of living MSC and whole
168.8 ± 27 ng/mL). Islets alone or islets plus MSC cultured within islets within the cryogel scaffolds. Although fewer islets were
cryogels revealed a similar insulin secretion as islets in suspension observed on the bottom site of the cryogel (Fig. S3B and S3C), a
(resting; p = 0.81, stimulation; p = 0.34; general linear regression). number of islets were detected throughout the cryogel
No differences were observed in the stimulation index between (Fig. 4B and C). The difficulty in islet location may be attributed
groups (p = 0.185; one-way ANOVA, Fig. 6B). Thus, the viability to the small number of seeded islets (100), relative to the large
and functionality of islets loaded within the cryogel scaffold was cryogel scaffold (diameter 8 mm, thickness 2 mm).
similar to controls, which were cultured in suspension. Due to their high porosity (approximately 98%) and their thin
but stiff struts (Fig. S2), the cryogel scaffolds are soft but robust
upon compression (Fig. 2B), and can withstand large deformation
3.4. Graft assessment of islet and MSC laden cryogel scaffolds after
(approximately 90%) without losing integrity [31,32]. The local
syngeneic transplantation in rodents
stiffness of the cryogel pore walls (struts) was found to be orders
of magnitudes higher in comparison to the low global (bulk) stiff-
In order to test the transplantation of islets in starPEG-heparin
ness of the whole cryogel scaffold. This enables both mechanical
cryogel scaffolds and the co-transplantation of MSC and islets,
protection of the MSC and islets in the macropores, and at the same
mice were transplanted subcutaneously with empty, islet (GFP+
time matches the tissue stiffness at potential extra-hepatic trans-
islets) laden or islet plus MSC (CM-DiI labeled) laden cryogels.
plantation sites. The global Young’s modulus of the cryogel scaf-
No adverse behavioral effects were observed in the recipients
folds (0.6 kPa) is comparable to the Young’s modulus of muscle
post-transplant, and mice maintained their weight 7 days post-
[39] and normal adipose tissue [40], and thus should be advanta-
transplant with no differences in weight change over time between
geous for subcutaneous implantation applications. Adaptation to
groups (pooled slope = 0.14, p = 0.85; linear regression, Fig. 7B). A
the tissue stiffness of other transplantation sites is similarly possi-
thin fibrous capsule surrounding the cryogel was observed in all
ble by choosing different experimental conditions during scaffold
cryogels removed. Sequential sectioning and imaging of the cryo-
preparation. This may further allow integration into alternate pan-
gels by means of fluorescence microscopy revealed the presence
creatic transplantation sites such as brachioradialis muscle of the
of intact islets and MSC in the scaffolds after 7 days in vivo
forearm [41], or the omentum pouch [42]. Despite its low bulk
throughout the scaffold (Fig. 7A) as well as the presence of host
stiffness, the scaffold provides a stiff, local microenvironment that
F4/80+ macrophages (Fig. S5).

Fig. 6. In vitro insulin secretion after glucose stimulation (A) Islets with or without MSC in suspension (n = 5–6) or cryogels (n = 6) were rested in Krebs buffer containing
2.8 mM glucose (open symbols) and stimulated in Krebs buffer containing 25 mM glucose (closed symbols) for 2 h. Mean insulin concentrations ± SD are shown. (B)
Stimulation index calculated from insulin secreted under resting and stimulating conditions in (A). Scatter dot plots with the mean ± SD are shown.
D.J. Borg et al. / Acta Biomaterialia 44 (2016) 178–187 185

Fig. 7. Performance of Alexa FluorÒ 405 cryogels as islet carriers 1 week post-subcutaneous transplantation in C57BL6/J mouse hosts. A cryogel (n = 1/mouse) contained
either 100 GFP+ islets alone or together with 500,000 CM-DiI MSC (n = 3 mice/group). Empty Alexa FlourÒ 405-conjugated cryogels were also transplanted (n = 3).
Representative fluorescence microscopy images of cryogel slices (5 lm) were acquired with a laser scanning microscope throughout the cryogel. (A) GFP+ islets (green) and
CM-DiI labeled MSC (red) located within the cryogel 7 days post-transplant (blue). Empty cryogels (blue) seeded without cells are shown in the right panel. Scale bar = 50 lm.
(B) Mean weight of mice from the time of transplant to sacrifice. Error bars are shown as SEM. (For interpretation of the references to colour in this figure legend, the reader is
referred to the web version of this article.)

shields the cells in the macropores from mechanical stress, as elas- three days prior to islet seeding. In addition, the MSC, previously
tic energy is adsorbed by the cryogel struts [43]. shown to be beneficial for islet priming as well as during trans-
The heparin component of the underlying hydrogel matrix plantation [26–28], remained in the cryogel scaffold after islet
offers the possibility to introduce adhesion-mediating peptide loading. Additional in vitro time-course studies following the pro-
ligands (here: RGD). The architecture and bioactive motifs of the gression of ECM protein deposition by MSC may optimize this
cryogel system were found to support adhesion of MSC within cell-based modification of the cryogel scaffold.
the pores of the cryogel. The flattened cell morphology suggests Our in vitro experiments demonstrated islet anchorage near
tight interaction with the RGD-modified gel matrix of the cryogel MSC, suggesting a favorable local niche at the struts for islet
struts whilst maintaining cell-cell contact with other MSC attachment. SEM images revealed that endocrine cells of the islets
(Fig. 3B). The scaffolds were fully compatible to both cell types as maintained cell-to-cell contact and maintained the presence of
proven by live/dead staining (Fig. 3, S3 and S4). microvilli. Microvilli are found on native islets [48] and the loss
MSC survived the seeding procedure (Fig. 3A, S3A and S4A) and of microvilli in a beta cell line has been reported as an early feature
maintained their ability to secrete ECM proteins (Fig. 5), which are of apoptosis [49]. Thus, starPEG-heparin cryogel scaffolds support
important cues for survival, function and proliferation of pancre- pancreatic islets by mechanical, architectural and biomolecular
atic islets [44–46]. The significance of islet-ECM interactions for cues and enable the localized interaction of islets with accessory
recapitulating the native environment of functional pancreatic cells and cell-secreted ECM proteins (Fig. 1). Further, islets
islets in order to further minimize anoikis has been highlighted remained responsive to glucose suggesting their function remained
in literature. Within their native environment, functional pancre- short-term in vitro (Fig. 6). The co-housing of MSC with islets how-
atic islets are supported by a basement membrane rich in ECM pro- ever did not significantly enhance insulin secretion in contrast to
teins, which is destroyed during islet isolation by collagenase previous encapsulation studies [22]. Future in vitro studies would
treatment [7,8]. Thus, there have been many attempts to coat or benefit from glucose challenges over long-term culture, to contin-
functionalize 2D and 3D biomaterials for islet cell culture with var- ually monitor islet function within the scaffold, and to determine
ious ECM proteins, such as laminin 5 and collagen type IV [6,47]. In additional support provided by the artificial 3D microenvironment
our approach, we used pre-seeded scaffold-bound MSC for secre- and/or the MSC, which may have been masked due to the short-
tion and deposition of ECM proteins in the cryogel scaffolds for term culture period in the present study.
186 D.J. Borg et al. / Acta Biomaterialia 44 (2016) 178–187

Subcutaneous transplantation of MSC/islet-seeded cryogel scaf- Glycation and Diabetes and Inflammatory Diseases and Therapeu-
folds confirmed the principal feasibility of the approach in a mouse tics Group, Mater Research Institute- University of Queensland,
model. Our provided data indicate that implantation of the cell- Translational Research Institute, Brisbane, Australia.
seeded cryogels did not interfere with the localization of islet
and MSC. GFP+ islets and cell membrane-labeled MSC could be
Appendix A. Supplementary data
visualized within the cryogel 7 days post-transplantation
(Fig. 7A). The cryogel material did induce the production of a
Supplementary data associated with this article can be found, in
fibrous capsule around the implant and macrophage infiltration
the online version, at http://dx.doi.org/10.1016/j.actbio.2016.08.
(Fig. S5). Similar host responses were reported in pancreatic islet
007.
transplantation experiments when using PLGA scaffolds [50],
alginate-encapsulated islets [51] and islet containing devices
[52]. Nevertheless, mice maintained their weight throughout the References
transplantation period (Fig. 7B). Despite host responses, short-
[1] B. Ludwig, S. Ludwig, A. Steffen, H.-D. Saeger, S.R. Bornstein, Islet versus
term islet engraftment was demonstrated after 7 days post- pancreas transplantation in type 1 diabetes: competitive or complementary?,
transplantation. In this pilot study however, we were unable to Curr Diab. Rep. 10 (2010) 506–511.
confirm islet function, namely insulin secretion in vivo, by [2] R.P. Robertson, Medical progress: islet transplantation as a treatment for
diabetes – a work in progress, N. Engl. J. Med. 350 (2004) 694–705.
immunofluorescence, due to technical limitations caused by fixa- [3] A.M. Davalli, Y. Ogawa, L. Scaglia, Y.-J. Wu, J. Hollister, S. Bonner-Weir, G.C.
tion. Thus, to assist in the translation of this approach in clinical Weir, Function, mass, and replication of porcine and rat islets transplanted into
islet transplantation, long-term experiments in a type 1 diabetes diabetic nude mice, Diabetes 44 (1995) 104–111.
[4] S. Merani, A. Shapiro, Current status of pancreatic islet transplantation, Clin.
animal model under autogenic and allogenic settings to assess islet Sci. 110 (2006) 611–625.
function and survival, are still required. It would also be useful to [5] C. Hellerström, A method for the microdissection of intact pancreatic islets of
further examine the beneficial aspects of MSC as accessory cells mammals, Acta Endocrinol. 45 (1964) 122–132.
[6] G.G.M. Pinkse, W.P. Bouwman, R. Jiawan-Lalai, O.T. Terpstra, J.A. Bruijn, E. de
within the cryogel such as subsequent immunomodulatory or neo- Heer, Integrin signaling via RGD peptides and anti-beta1 antibodies confers
vascularization effects, which was not the subject of this pilot resistance to apoptosis in islets of Langerhans, Diabetes 55 (2006) 312–317.
study. Indeed, ongoing research aims at further adjusting the [7] F. Thomas, J. Wu, J.L. Contreras, C. Smyth, G. Bilbao, J. He, J. Thomas, A tripartite
anoikis-like mechanism causes early isolated islet apoptosis, Surgery 130
cryogel-based islet carriers with respect to their revascularization (2015) 333–338.
potential utilizing the heparin component of the underlying biohy- [8] F.T. Thomas, J.L. Contreras, G. Bilbao, C. Ricordi, D. Curiel, J.M. Thomas, Anoikis,
brid material for additional administration of pro-angiogenic extracellular matrix, and apoptosis factors in isolated cell transplantation,
Surgery 126 (1999) 299–304.
growth factors [33,53].
[9] P. de Vos, M.M. Faas, B. Strand, R. Calafiore, Alginate-based microcapsules for
immunoisolation of pancreatic islets, Biomaterials 27 (2006) 5603–5617.
[10] V. Vaithilingam, B.E. Tuch, Islet transplantation and encapsulation: an update
5. Conclusion on recent developments, Rev. Diabet. Stud. 8 (2011) 51–67.
[11] M. de Groot, T.A. Schuurs, R. Van Schilfgaarde, Causes of limited survival of
microencapsulated pancreatic islet grafts, J. Surg. Res. 121 (2004) 141–150.
An islet-supportive carrier was designed by combining the
[12] H. Blomeier, X. Zhang, C. Rives, M. Brissova, E. Hughes, M. Baker, A.C. Powers,
advantageous properties of macroporous starPEG-heparin cryogels D.B. Kaufman, L.D. Shea, W.L.J. Lowe, Polymer scaffolds as synthetic
with benefits of bone marrow-derived MSC accessory cells. The microenvironments for extrahepatic islet transplantation, Transplantation 82
interconnectivity and size distribution of the macropores enabled (2006) 452–459.
[13] Y. Tabata, Y. Ikada, Vascularization effect of basic fibroblast growth factor
MSC and whole islets to enter the cryogel scaffolds with their stiff released from gelatin hydrogels with different biodegradabilities, Biomaterials
pore walls that shield the cells from mechanical stress. RGD- 20 (1999) 2169–2175.
modification mediated adhesion of pre-seeded MSC which [14] N. Aloysious, P.D. Nair, Enhanced survival and function of islet-like clusters
differentiated from adipose stem cells on a three-dimensional natural
secreted extracellular matrix proteins to provide further attach- polymeric scaffold: an in vitro study, Tissue Eng. Part A 20 (2014) 1508–1522.
ment support for the pancreatic islets in the cryogel and to prevent [15] K. Bloch, V.I. Lozinsky, I.Y. Galaev, K. Yavriyanz, M. Vorobeychik, D. Azarov, L.G.
their anoikis. The survival and continued function of cryogel- Damshkaln, B. Mattiasson, P. Vardi, Functional activity of insulinoma cells
(INS-1E) and pancreatic islets cultured in agarose cryogel sponges, J. Biomed.
housed islets was demonstrated short-term in vitro with no Mater. Res. 75A (2005) 802–809.
adverse effect on insulin secretion, and short-term implantation [16] J.M. Dufour, R.V. Rajotte, M. Zimmerman, A. Rezania, T. Kin, D.E. Dixon, G.S.
in vivo in a mouse model confirmed the feasibility of the approach. Korbutt, Development of an ectopic site for islet transplantation, using
biodegradable scaffolds, Tissue Eng. 11 (2005) 1323–1331.
Thus, starPEG-heparin cryogel scaffolds tuneable in architecture, [17] E. Pedraza, A.-C. Brady, C.A. Fraker, R.D. Molano, S. Sukert, D.M. Berman, N.S.
mechanical characteristics and biomolecular functionalization, Kenyon, A. Pileggi, C. Ricordi, C.L. Stabler, Macroporous three-dimensional
with the ability to load accessory cells, are highly promising as PDMS scaffolds for extrahepatic islet transplantation, Cell Transplant. 22
(2013) 1123–1135.
supportive carriers for pancreatic islets in the context of transplan-
[18] R.F. Gibly, X. Zhang, M.L. Graham, B.J. Hering, D.B. Kaufman, W.L.J. Lowe, L.D.
tation in various alternate sites. Shea, Extrahepatic islet transplantation with microporous polymer scaffolds in
syngeneic mouse and allogeneic porcine models, Biomaterials 32 (2011)
9677–9684.
Acknowledgements [19] J.T. Daoud, M.S. Petropavlovskaia, J.M. Patapas, C.E. Degrandpre, R.W. DiRaddo,
L. Rosenberg, M. Tabrizian, Long-term in vitro human pancreatic islet culture
using three-dimensional microfabricated scaffolds, Biomaterials 32 (2011)
Financial support was provided by the German Research Foun- 1536–1542.
dation (Deutsche Forschungsgemeinschaft) through Grant num- [20] S. Muthyala, R.R. Bhonde, P.D. Nair, Cytocompatibility studies of mouse
bers: SFB-TR 67, WE 2539-7 and FOR/EXC999, by the Leibniz pancreatic islets on gelatin-PVP semi IPN scaffolds in vitro potential
implication towards pancreatic tissue engineering, Islets 2 (2010) 357–366.
Association (SAW-2011-IPF-2 68) and by the European Union [21] A.-C. Brady, M.M. Martino, E. Pedraza, S. Sukert, A. Pileggi, C. Ricordi, J.A.
through the Integrated Project ANGIOSCAFF (Seventh Framework Hubbell, C.L. Stabler, Proangiogenic Hydrogels within macroporous scaffolds
Program). Moreover, this work was supported by the DFG-Center enhance islet engraftment in an extrahepatic site, Tissue Eng. Part A 19 (2013)
2544–2552.
for Regenerative Therapies Dresden, Cluster of Excellence (FZT
[22] N.E. Davis, L.N. Beenken-Rothkopf, A. Mirsoian, N. Kojic, D.L. Kaplan, A.E.
111) and the Federal Ministry of Education and Research, Germany Barron, M.J. Fontaine, Enhanced function of pancreatic islets co-encapsulated
(01GN0945). D. J. Borg was enrolled in the Regenerative Medicine with ECM proteins and mesenchymal stromal cells in a silk hydrogel,
Ph.D. Program of the Dresden International Graduate School for Biomaterials 33 (2012) 6691–6697.
[23] J.C. Stendahl, L.-J. Wang, L.W. Chow, D.B. Kaufman, S.I. Stupp, Growth factor
Biomedicine & Bioengineering (DIGS-BB) as part of the Technical delivery from self-assembling nanofibers to facilitate islet transplantation,
University of Dresden. D. J. Borg’s current affiliation is the Transplantation 86 (2008) 478–481.
D.J. Borg et al. / Acta Biomaterialia 44 (2016) 178–187 187

[24] A. Kerby, E.S. Jones, P.M. Jones, A.J. King, Co-transplantation of islets with [39] E.J. Chen, J. Novakofski, W.K. Jenkins, W.D. O’Brien, Young’s modulus
mesenchymal stem cells in microcapsules demonstrates graft outcome can be measurements of soft tissues with application to elasticity imaging, IEEE
improved in an isolated-graft model of islet transplantation in mice, Trans. Ultrason. Ferroelect. Freq. Contr. 43 (1996) 191–194.
Cytotherapy 15 (2013) 192–200. [40] A. Samani, J. Zubovits, D. Plewes, Elastic moduli of normal and pathological
[25] M.M. Coronel, C.L. Stabler, Engineering a local microenvironment for human breast tissues: an inversion-technique-based investigation of 169
pancreatic islet replacement, Curr. Opin. Biotechnol. 24 (2013) 900–908. samples, Phys. Med. Biol. 52 (2007) 1565–1576.
[26] T. Ito, S. Itakura, I. Todorov, J. Rawson, S. Asari, J. Shintaku, I. Nair, K. Ferreri, F. [41] G. Christoffersson, P.-O. Carlsson, M. Phillipson, Intramuscular islet
Kandeel, Y. Mullen, Mesenchymal stem cell and islet co-transplantation transplantation promotes restored islet vascularity, Islets 3 (2011) 69–71.
promotes graft revascularization and function, Transplantation 89 (2010) [42] D.M. Berman, J.J. O’Neil, L.C.K. Coffey, P.C.J. Chaffanjon, N.M. Kenyon, P.J. Ruiz,
1438–1445. A. Pileggi, C. Ricordi, N.S. Kenyon, Long-term survival of nonhuman primate
[27] D.J. Borg, M. Weigelt, C. Wilhelm, M. Gerlach, M. Bickle, S. Speier, E. Bonifacio, islets implanted in an omental pouch on a biodegradable scaffold, Am. J.
A. Hommel, Mesenchymal stromal cells improve transplanted islet survival Transplant. 9 (2009) 91–104.
and islet function in a syngeneic mouse model, Diabetologia 57 (2014) 522– [43] A. Béduer, T. Braschler, O. Peric, G.E. Fantner, S. Mosser, P.C. Fraering, S.
531. Benchérif, D.J. Mooney, P. Renaud, A compressible scaffold for minimally
[28] D.M. Berman, M.A. Willman, D. Han, G. Kleiner, N.M. Kenyon, O. Cabrera, J.A. invasive delivery of large intact neuronal networks, Adv. Healthcare Mater. 4
Karl, R.W. Wiseman, D.H. O’Connor, A.M. Bartholomew, N.S. Kenyon, (2015) 301–312.
Mesenchymal stem cells enhance allogeneic islet engraftment in nonhuman [44] E. Hammar, G. Parnaud, D. Bosco, N. Perriraz, K. Maedler, M. Donath, D.G.
primates, Diabetes 59 (2010) 2558–2568. Rouiller, P.A. Halban, Extracellular matrix protects pancreatic beta-cells
[29] M.C. Prewitz, F.P. Seib, M. von Bonin, J. Friedrichs, A. Stissel, C. Niehage, K. against apoptosis: role of short- and long-term signaling pathways, Diabetes
Müller, K. Anastassiadis, C. Waskow, B. Hoflack, M. Bornhauser, C. Werner, 53 (2004) 2034–2041.
Tightly anchored tissue-mimetic matrices as instructive stem cell [45] D. Bosco, P. Meda, P.A. Halban, D.G. Rouiller, Importance of cell-matrix
microenvironments, Nat. Methods 10 (2013) 788–794. interactions in rat islet beta-cell secretion in vitro: role of alpha6beta1
[30] F.G. Teixeira, M.M. Carvalho, N. Sousa, A.J. Salgado, Mesenchymal stem cells integrin, Diabetes 49 (2000) 233–243.
secretome: a new paradigm for central nervous system regeneration?, Cell [46] G. Nikolova, N. Jabs, I. Konstantinova, A. Domogatskaya, K. Tryggvason, L.
Mol. Life Sci. 70 (2013) 3871–3882. Sorokin, R. Fässler, G.Q. Gu, H.P. Gerber, N. Ferrara, D.A. Melton, E. Lammert,
[31] P.B. Welzel, J. Friedrichs, M. Grimmer, S. Vogler, U. Freudenberg, C. Werner, The vascular basement membrane: a niche for insulin gene expression and
Cryogel micromechanics unraveled by atomic force microscopy-based beta cell proliferation, Dev. Cell 10 (2006) 397–405.
nanoindentation, Adv. Healthcare Mater 3 (2014) 1849–1853. [47] L.M. Weber, K.S. Anseth, Hydrogel encapsulation environments functionalized
[32] P.B. Welzel, M. Grimmer, C. Renneberg, L. Naujox, S. Zschoche, U. Freudenberg, with extracellular matrix interactions increase islet insulin secretion, Matrix
C. Werner, Macroporous starPEG-heparin cryogels, Biomacromolecules 13 Biol. 27 (2008) 667–673.
(2012) 2349–2358. [48] L. Orci, B. Thorens, M. Ravazzola, H.F. Lodish, Localization of the pancreatic
[33] B. Newland, P.B. Welzel, H. Newland, C. Renneberg, P. Kolar, M. Tsurkan, A. beta cell glucose transporter to specific plasma membrane domains, Science
Rosser, U. Freudenberg, C. Werner, Tackling cell transplantation anoikis: an 245 (1989) 295–297.
injectable, shape memory cryogel microcarrier platform material for stem cell [49] E.L. Saafi, B. Konarkowska, S. Zhang, J. Kistler, G.J. Cooper, Ultrastructural
and neuronal cell growth, in: Small 11 (2015) 5047–5053. evidence that apoptosis is the mechanism by which human amylin evokes
[34] U. Freudenberg, A. Hermann, P.B. Welzel, K. Stirl, S.C. Schwarz, M. Grimmer, A. death in RINm5F pancreatic islet beta-cells, Cell Biol. Int. 25 (2001) 339–350.
Zieris, W. Panyanuwat, S. Zschoche, D. Meinhold, A. Storch, C. Werner, A star- [50] T. Kheradmand, S. Wang, R.F. Gibly, X. Zhang, S. Holland, J. Tasch, J.G. Graham,
PEG–heparin hydrogel platform to aid cell replacement therapies for D.B. Kaufman, S.D. Miller, L.D. Shea, X. Luo, Permanent protection of PLG
neurodegenerative diseases, Biomaterials 30 (2009) 5049–5060. scaffold transplanted allogeneic islet grafts in diabetic mice treated with ECDI-
[35] U. Freudenberg, J.-U. Sommer, K.R. Levental, P.B. Welzel, A. Zieris, K. Chwalek, fixed donor splenocyte infusions, Biomaterials 32 (2011) 4517–4524.
K. Schneider, S. Prokoph, M. Prewitz, R. Dockhorn, C. Werner, Using mean field [51] S. Veriter, J. Mergen, R.-M. Goebbels, N. Aouassar, C. Gregoire, B. Jordan, P.
theory to guide biofunctional materials design, Adv. Funct. Mater. 22 (2012) Leveque, B. Gallez, P. Gianello, D. Dufrane, In vivo selection of biocompatible
1391–1398. alginates for islet encapsulation and subcutaneous transplantation, Tissue Eng.
[36] A. Kumar, A. Srivastava, Cell separation using cryogel-based affinity Part A 16 (2010) 1503–1513.
chromatography, Nat Protoc. 5 (2010) 1737–1747. [52] K. Suzuki, S. Bonner-Weir, N. Trivedi, K.H. Yoon, J. Hollister-Lock, C.K. Colton, G.
[37] V.I. Lozinsky, Cryogels on the basis of natural and synthetic polymers: C. Weir, Function and survival of microencapsulated syngeneic islets
preparation, properties and application, Russ. Chem. Rev. 71 (2007) 489–511. transplanted into streptozocin-diabetic mice, Transplantation 66 (1998) 21–
[38] K.P. Knoch, R. Meisterfeld, S. Kersting, H. Bergert, A. Altkruger, C. Wegbrod, M. 28.
Jager, H.D. Saeger, M. Solimena, CAMP-dependent phosphorylation of PTB1 [53] A. Zieris, K. Chwalek, S. Prokoph, K.R. Levental, P.B. Welzel, U. Freudenberg, C.
promotes the expression of insulin secretory granule proteins in beta cells, Cell Werner, Dual independent delivery of pro-angiogenic growth factors from
Metab. 3 (2006) 123–134. starPEG-heparin hydrogels, J. Controlled Release 156 (2011) 28–36.

Vous aimerez peut-être aussi