Pancreatic islet transplantation (PIT) represents a potential therapy to circumvent the need for exogenous insulin in type 1 diabetes. However, PIT remains limited by lack of donor islets and the need for long-term multidrug immunosuppression to prevent alloimmune islet rejection. Our goal was to evaluate a local immunoregulatory strategy that sustains islet allograft survival and restores glucose homeostasis in the absence of systemic immunosuppression. Nanogram quantities of murine CTLA4/Fc fusion protein were controllably delivered within human acellular dermal matrix scaffolds using an inkjet-based biopatterning technology and cotransplanted with allogeneic islets under the renal capsule to create an immunoregulatory microenvironment around the islet allograft. We achieved long-term engraftment of small loads of allogeneic islet cells with 40% of MHC-mismatched mouse recipients maintaining sustained normoglycemia following pancreatic β-cell ablation by streptozotocin. Biopatterned CTLA4/Fc local therapy was associated with expansion of Foxp3+ regulatory T cells and shifts in cytokine production and gene expression from proinflammatory to regulatory profiles, thus substantially benefiting islet allografts survival and function. This study is a new paradigm for targeted therapies in PIT that demonstrates the favorable effects of immune alterations in the transplant milieu and suggests a unique strategy for minimizing systemic immunosuppression and promoting islet allograft survival.

There is an immediate need for pancreatic islet transplantation (PIT) strategies that increase functional efficacy of small cell loads by optimizing engraftment and eliminating the need for chronic immunosuppression for graft survival. Costimulation blockade with cytotoxic T lymphocyte antigen 4 fusion protein (CTLA4-Ig) has been effective in downregulating allo- and autoimmune responses, leading to enhancement of islet allograft survival and inhibition of type 1 diabetes development (14). However, systemic administration of large doses of CTLA4-Ig not only has long-term side effects but also has failed to completely inhibit alloimmune destruction of islet β-cells and to induce tolerance (5). Site-specific immunomodulation may be a promising strategy to prevent acute rejection and minimize indiscriminate damage to essential cells and organs by systemic immunosuppression (68).

Our team has developed a cutting-edge biopatterning technology that can spatially control the distribution of physiologically relevant, pico-to-nanogram level doses of signaling molecules immobilized as “solid-phase” patterns to extracellular matrix (ECM) constructs via native binding affinities to direct cell behavior in vitro and tissue formation in vivo (911). Our a priori hypothesis is based on successful prior work where biopatterning of signaling molecules in solid phase significantly improved outcomes across multiple applications (12,13). Here, we fabricated immunoregulatory microenvironments using murine CTLA4/Fcγ2a heavy-chain chimeric fusion protein (CTLA4/Fc) constructs biopatterned within immunoneutral human-derived acellular dermal matrix (ADM). We investigated the effects of this CTLA4/Fc microenvironment on inhibiting alloimmune responses after PIT and promoting islet allograft survival without additional immunosuppression. Our work is the first to demonstrate that targeted, spatially controlled delivery of extremely small doses of solid-phase proregulatory agents, such as biopatterned CTLA4/Fc microenvironments, can promote engraftment, function, and long-term survival of islet allografts.

Creation of Biopatterned Murine CTLA4/Fc Microenvironment in PIT

Uniform patterns of murine CTLA4/Fc bioink (200 μg/mL) were produced in our laboratory and printed onto 200-μm thick, 5 × 5 mm pieces of ADM (Synthes, West Chester, PA) using our inkjet-based biopatterning system (9,10,14). The printed bioink absorbs into the open-pore structure of the dermal surface and binds within the matrix to produce immobilized 3-D patterns that persist for sufficient periods of time needed to elicit the desired biological control (9). Binding retention of CTLA4/Fc was estimated by printing 125I-CTLA4/Fc onto ADM using the method similar to other hormone bioinks (14,15), which demonstrated that biopatterned CTLA4/Fc persist stabilized release with 50% of bound remaining for 21 days under simulated in vitro conditions.

PIT

PIT was performed from donor DBA/2(H-2d) mice to streptozotocin-induced diabetic C57BL/6(H-2b) recipient mice as previously described (16). After prerinsing, ADM squares were cut into 1 × 5 mm strips and inserted below the renal capsule. Approximately 300 islet allografts were injected between strips, so that the biopatterned CTLA4/Fc could be in direct contact with islet allografts (Fig. 1). Other groups included single dose CTLA4/Fc (20 mg/kg) intraperitoneally (i.p.), CTLA4/Fc ADM implanted under the contralateral renal capsule, nonprinted ADM, and untreated recipients. Allograft function was assessed by monitoring blood glucose levels (BGL), and rejection was defined as BGL of >300 mg/dL on two consecutive measurements (17).

Figure 1

Creation of biopatterned CTLA4/Fc microenvironment in the islet transplant site. Bioinks consisting of murine CTLA4/Fc were printed in a uniform pattern onto the 200-μm thick, 5 × 5 mm ADM. The printed dose of CTLA4/Fc was controlled by a fixed number of 50 overprints per each square. Printed side (dermal side) is facing up when the notch and semicircle are oriented at the 6:00 and 4:00 position. After prerinsing, ADM biopatterned with CTLA4/Fc was cut into 5 pieces, 1 mm × 5 mm × 0.2 mm, and inserted into the pouch of the renal capsule, and islet allografts were injected between strips. The photo (lower right corner) and immunohistochemical insulin staining (upper right corner) show the positional relationship of the transplanted islets with respect to the ADM and kidney. Scale bar = 100 μm.

Figure 1

Creation of biopatterned CTLA4/Fc microenvironment in the islet transplant site. Bioinks consisting of murine CTLA4/Fc were printed in a uniform pattern onto the 200-μm thick, 5 × 5 mm ADM. The printed dose of CTLA4/Fc was controlled by a fixed number of 50 overprints per each square. Printed side (dermal side) is facing up when the notch and semicircle are oriented at the 6:00 and 4:00 position. After prerinsing, ADM biopatterned with CTLA4/Fc was cut into 5 pieces, 1 mm × 5 mm × 0.2 mm, and inserted into the pouch of the renal capsule, and islet allografts were injected between strips. The photo (lower right corner) and immunohistochemical insulin staining (upper right corner) show the positional relationship of the transplanted islets with respect to the ADM and kidney. Scale bar = 100 μm.

Close modal

Quantification of Cytokines and Genes

The production of IL-4, IFN-γ, IL-6, and IL-17 in serum samples obtained at 2 and 8 weeks posttransplantation were assessed by enzyme-linked immunosorbent assay (ELISA). Total RNA was extracted from draining lymph nodes or islet allografts. Levels of specific mRNA IL-4, IFN-γ, Gzmb, IL-17, and Foxp3 were quantified by real-time PCR.

Assessment of T-Cell Subsets

Cells were harvested from lymph nodes, spleens, or peripheral blood of long-term survival recipients at day 150 after transplantation. The expression of cell surface and intracellular markers (CD3, CD4, CD25, and Foxp3) were analyzed by flow cytometric analysis. To test regulatory T-cell (Treg) suppression, flow-sorted CD4+CD25 responder cells from naive C57BL/6 mice were cultured with CD4+CD25hi T cells from long-term islet allograft survival recipients or age-matched naive mice in 96-well plates, using anti-CD3 and anti-CD28 monoclonal antibody (mAb) or irradiated splenocytes from donor (DBA/2) or the third party (C3H) as stimulators. Cultures were pulsed for the last 8 h of the incubation with 1 μCi/well 3H-thymidine, and 3H-thymidine incorporation was measured using standard scintillation procedures and expressed as percent inhibition of proliferation.

Histological Analysis

The kidney bearing the islet graft was removed from recipients and processed for hematoxylin-eosin (H&E) staining and immune-peroxidase histochemistry with anti-mouse Foxp3 mAb or anti-insulin mAb. Samples were evaluated in a blinded fashion at two different levels of sectioning.

Statistical Analysis

Data are expressed as mean ± SD. Comparisons between two samples were performed using the Student t test. Multiple groups were compared by one-way ANOVA with Tukey multiple comparisons test. Allograft survival was analyzed using a log-rank test. Differences are considered significant if P < 0.05.

Localized Delivery of a Biopatterned CTLA4/Fc Immunoregulatory Microenvironment Promotes Islet Allograft Survival Across a Full MHC Barrier in Mice

In the MHC-mismatched DBA/2 to C57BL/6 mice PIT model, treatment with local biopatterned CTLA4/Fc ADM significantly prolonged islet allograft survival with median survival time (MST) of 71 days (n = 10; P < 0.001 vs. untreated group; P < 0.05 vs. CTLA4/Fc i.p. group). All those recipients maintained normoglycemia for >28 days, 40% of which demonstrated long-term (>150 days) euglycemia. Contralaterally implanted CTLA4/Fc ADM did not induce engraftment of islet allografts, indicating that the biopatterned CTLA4/Fc functioned at the transplant site but not remotely (Fig. 2A). Furthermore, biopatterned CTLA4/Fc maintains persistence of detectable low serum levels longer than systemically delivered CTLA4/Fc as detected by ELISA over time after PIT (Fig. 2B).

Figure 2

Pancreatic islets from DBA/2 mice were transplanted under the renal capsule of diabetic C57BL/6 recipients, which were treated with either localized delivery of biopatterned CTLA4/Fc ADM (n = 10), a single i.p. injection of CTLA4/Fc (n = 4), biopatterned CTLA4/Fc ADM implanted under the contralateral renal capsule (n = 4), nonprinted ADM (n = 6), or no treatment (n = 6). A: Survival of islet allografts. Untreated: MST 15.5 days; nonprinted ADM: MST 15.0 days; CTLA4/Fc i.p.: MST 36.5 days; contralateral CTLA4/Fc ADM: MST 23 days; local biopatterned CTLA4/Fc ADM: MST 71 days. *P < 0.05, **P < 0.01, ***P < 0.001. B: Mean CTLA4/Fc serum concentrations in islet allograft recipient mice given either a single i.p. injection of CTLA4/Fc (20 mg/kg) or implantation of biopatterned CTLA4/Fc ADM (3 mice in each group) immediately after transplantation. Serum samples were collected from recipients at a serial time points. Concentrations of CTLA4/Fc were determined by a sandwich ELISA using anti-mouse CTLA4 mAb as the capture antibody and horseradish peroxidase–conjugated rat anti-mouse IgG2a mAb as the detection antibody. Serum CTLA4/Fc demonstrates a gradual elevation to 1.5 μg/mL and maintained low detectable levels for up to 8 weeks until it was completely undetectable at 12 weeks.

Figure 2

Pancreatic islets from DBA/2 mice were transplanted under the renal capsule of diabetic C57BL/6 recipients, which were treated with either localized delivery of biopatterned CTLA4/Fc ADM (n = 10), a single i.p. injection of CTLA4/Fc (n = 4), biopatterned CTLA4/Fc ADM implanted under the contralateral renal capsule (n = 4), nonprinted ADM (n = 6), or no treatment (n = 6). A: Survival of islet allografts. Untreated: MST 15.5 days; nonprinted ADM: MST 15.0 days; CTLA4/Fc i.p.: MST 36.5 days; contralateral CTLA4/Fc ADM: MST 23 days; local biopatterned CTLA4/Fc ADM: MST 71 days. *P < 0.05, **P < 0.01, ***P < 0.001. B: Mean CTLA4/Fc serum concentrations in islet allograft recipient mice given either a single i.p. injection of CTLA4/Fc (20 mg/kg) or implantation of biopatterned CTLA4/Fc ADM (3 mice in each group) immediately after transplantation. Serum samples were collected from recipients at a serial time points. Concentrations of CTLA4/Fc were determined by a sandwich ELISA using anti-mouse CTLA4 mAb as the capture antibody and horseradish peroxidase–conjugated rat anti-mouse IgG2a mAb as the detection antibody. Serum CTLA4/Fc demonstrates a gradual elevation to 1.5 μg/mL and maintained low detectable levels for up to 8 weeks until it was completely undetectable at 12 weeks.

Close modal

Biopatterned CTLA4/Fc Microenvironment Alters T-Cell–Derived Cytokine Production, Favors Treg Expansion, and Maintains Treg Suppression After PIT

The levels of IL-4 increased and IFN-γ and IL-6 decreased in the local biopatterned CTLA4/Fc treatment group compared with the nonprinted ADM group at 2 weeks posttransplantation. After 8 weeks, the alteration of IL-4 and IL-6 remained in biopatterned CTLA4/Fc–treated recipients, when a significant decrease of IL-17 was detected (Fig. 3A). Additionally, there was marked upregulation of IL-4 and Foxp3 mRNA and downregulation of IFN-γ and Gzmb mRNA expression in both islet allografts and draining lymph nodes of biopatterned CTLA4/Fc–treated recipients at 2 weeks posttransplantation (Fig. 3B). T cells from biopatterned CTLA4/Fc–treated, long-term surviving recipients demonstrated an increase of CD4+Foxp3+ Treg in peripheral blood (6.0 ± 0.3%), spleen (8.8 ± 0.5%), and lymph nodes (9.5 ± 0.4%) compared with the naive mice (Fig. 3C). Furthermore, CD4+CD25hi Treg isolated from these long-term surviving recipients significantly inhibited the proliferation of CD4+CD25 T cells activated by the same donor antigen in a mixed lymphocyte reaction (71.2 ± 2.7% vs. 31.6 ± 2.1% inhibition; P < 0.01) but could not suppress third-party antigen-induced alloreactivity. These Treg also exhibited more potent suppressive ability than Treg from naive mice (67.9 ± 3.1% vs. 24.7 ± 4.0% inhibition; P < 0.01) under nonspecific stimulation (Fig. 3D).

Figure 3

Cytokine production, gene expression, and Treg assessment in islet allograft recipients. A: Serum samples were obtained from recipients at 2 and 8 weeks posttransplantation. Serum levels of IL-4, IFN-γ, IL-6, and IL-17 were determined by ELISA. B: Draining lymph nodes and islet allografts were harvested from recipients at 2 weeks posttransplantation. The mRNA expression of IL-4, IFN-γ, Gzmb, IL-17, and Foxp3 (relative to that of GAPDH) in draining lymph nodes and islet allografts was determined by quantitative real-time PCR. C: Peripheral blood, spleen, and draining lymph nodes (DLn) were harvested from biopatterned CTLA4/Fc–treated long-term survival recipients at 150 days after islet transplantation or from age-matched naive C57BL/6 mice. CD3+CD4+Foxp3+ cells were analyzed by flow cytometry. The percentages are presented by the scatter plot. Bar represents mean value of four samples each group. D: Flow-sorted CD4+CD25hi Treg (>98% purity) from naive or biopatterned CTLA4/Fc–treated long-term survival recipients were cultured with naive CD4+CD25 T cells in the presence of anti-CD3/CD28 mAb (each 2 μg/mL) or irradiated (3,000 rad) splenocytes from DBA/2 or C3H mice. T-cell proliferation was measured by 3H-thymidine incorporation after 72 h or 6 days of culture. Data are expressed as the mean percent inhibition of T-cell proliferation and are representative of three independent experiments, with triplicates in each experiment. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001; ns, not significant.

Figure 3

Cytokine production, gene expression, and Treg assessment in islet allograft recipients. A: Serum samples were obtained from recipients at 2 and 8 weeks posttransplantation. Serum levels of IL-4, IFN-γ, IL-6, and IL-17 were determined by ELISA. B: Draining lymph nodes and islet allografts were harvested from recipients at 2 weeks posttransplantation. The mRNA expression of IL-4, IFN-γ, Gzmb, IL-17, and Foxp3 (relative to that of GAPDH) in draining lymph nodes and islet allografts was determined by quantitative real-time PCR. C: Peripheral blood, spleen, and draining lymph nodes (DLn) were harvested from biopatterned CTLA4/Fc–treated long-term survival recipients at 150 days after islet transplantation or from age-matched naive C57BL/6 mice. CD3+CD4+Foxp3+ cells were analyzed by flow cytometry. The percentages are presented by the scatter plot. Bar represents mean value of four samples each group. D: Flow-sorted CD4+CD25hi Treg (>98% purity) from naive or biopatterned CTLA4/Fc–treated long-term survival recipients were cultured with naive CD4+CD25 T cells in the presence of anti-CD3/CD28 mAb (each 2 μg/mL) or irradiated (3,000 rad) splenocytes from DBA/2 or C3H mice. T-cell proliferation was measured by 3H-thymidine incorporation after 72 h or 6 days of culture. Data are expressed as the mean percent inhibition of T-cell proliferation and are representative of three independent experiments, with triplicates in each experiment. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001; ns, not significant.

Close modal

Biopatterned CTLA4/Fc Microenvironment Improves Allograft Engraftment and Survival by Local Regulation and Immunoprotection

A decreased inflammatory cell infiltration and minimal damage of islet allografts were observed by H&E at day 14 posttransplantation in the biopatterned CTLA4/Fc–treated recipients compared with the nonprinted ADM–treated recipients (Fig. 4A). The low degree of inflammatory infiltration and significant preservation of islet allografts were further characterized in the biopatterned CTLA4/Fc–treated long-term surviving recipients at 150 days posttransplantation by H&E and immunohistochemical analysis. The strong insulin staining demonstrated the potent function of allogeneic islets with insulin-producing cells. An increase of Foxp3-expressing cells suggests that local immune regulation is dominant in the protection of islet allografts from alloimmune destruction (Fig. 4B).

Figure 4

Histological analysis of islet allografts from recipients with biopatterned CTLA4/Fc treatment. A: H&E staining of islet allograft sections at day 14 posttransplantation showed significant reduction of inflammatory cell infiltrates and minimal damage of islet allografts in the biopatterned CTLA4/Fc–treated recipients. B: A low degree of inflammatory infiltration and significant preservation of islet allografts were observed in biopatterned CTLA4/Fc–treated long-term survival recipients at 150 days posttransplantation (H&E). Immunohistochemical insulin staining demonstrated intact islets positive for insulin granules (brown stain) and confirmed the allogeneic islets are functional. Significant Foxp3+ expression was observed in the long-term surviving islet allograft by immunohistochemical Foxp3 staining, suggesting the dominant immune regulation. Scale bars = 100 µm.

Figure 4

Histological analysis of islet allografts from recipients with biopatterned CTLA4/Fc treatment. A: H&E staining of islet allograft sections at day 14 posttransplantation showed significant reduction of inflammatory cell infiltrates and minimal damage of islet allografts in the biopatterned CTLA4/Fc–treated recipients. B: A low degree of inflammatory infiltration and significant preservation of islet allografts were observed in biopatterned CTLA4/Fc–treated long-term survival recipients at 150 days posttransplantation (H&E). Immunohistochemical insulin staining demonstrated intact islets positive for insulin granules (brown stain) and confirmed the allogeneic islets are functional. Significant Foxp3+ expression was observed in the long-term surviving islet allograft by immunohistochemical Foxp3 staining, suggesting the dominant immune regulation. Scale bars = 100 µm.

Close modal

Local or spatial control of the transplant microenvironment can modulate inflammation, immune cell infiltration, and activation associated alloimmune responses (18). Prior work has shown that T-cell–mediated cytotoxicity may be mitigated through local delivery of CTLA4-Ig (19). In this study, we sought to improve the efficiency of local immune regulation by targeting delivery of CTLA4/Fc to the transplant microenvironment of engrafted β-cells. Our goal was to eliminate systemic nonspecific immunosuppression to minimize islet toxicity, thereby reducing the islet cell load needed for PIT and supporting long-term engraftment, survival, and endocrine function.

We printed bioinks of CTLA4/Fc on a biodegradable ADM with defined surface concentrations and characterized spatial organization to create a tissue-engineered microenvironment using our established inkjet biopatterning technology, which is scalable and readily adaptable to other signaling molecules and/or ECMs (12,14). This technique allows for the solid-phase delivery of therapeutic proteins in extremely low dosages, with increased bioactivity relative to liquid-phase delivery. Our results show that a biopatterned CTLA4/Fc microenvironment implanted along with a low number of islet cells under the renal capsule achieved significantly prolonged survival, whereas a single i.p. injection of CTLA4/Fc or contralaterally implanted CTLA4/Fc ADM was less effective, suggesting that the immunoregulatory effect is exerted locally by CTLA4/Fc at the site of islet transplantation. Furthermore, prolonged low levels of serum CTLA4/Fc were observed following implantation of biopatterned CTLA4/Fc, indicating that high levels of CTLA4/Fc in the systemic circulation were not necessary. This would suggest that a persistent presence of solid-phase CTLA4/Fc might be optimal for immunoregulation.

ADM, as a native biological scaffold maintaining the original dermal ECM architecture and low immunogenicity, was determined to be a suitable surrogate material to sequester biologically active components (9). It has also been shown to support cell growth in vitro and to support neovascularization following implantation in vivo (20). Therefore, biopatterning technology combined with the features of ADM enables prolonged retention and an extended bioavailability of biopatterned CTLA4/Fc, thus allowing it to be sequestered at nanogram levels. This establishes a microenvironment that supports transplanted islet β-cells and exerts local immunomodulation by CTLA4/Fc to promote islet allograft engraftment and long-term function.

Type 1 T-helper cells (Th1) and type 2 T-helper cells (Th2) are the principal regulators of alloimmunity in transplantation. The induction of Th2 cytokines can inhibit Th1-mediated rejection responses, thereby promoting allograft acceptance (21). The cytokine profiles from biopatterned CTLA4/Fc–treated recipients revealed increased Th2 cytokine (IL-4) and decreased Th1 cytokines (IFN-γ) production in serum, with changes being more significant in the late posttransplantation period (8 weeks). In addition, the corresponding upregulation of IL-4 gene and downregulation of IFN-γ gene expression were detected in both islet allografts and draining lymph nodes. This finding suggests that biopatterned CTLA4/Fc delivered locally acts efficiently to modulate Th1/Th2 cytokines and to promote an immune deviation toward Th2 responses. Furthermore, histological examination showed decreased lymphocyte infiltration and long-term preserved islet allograft with increased Foxp3 staining in the biopatterned CTLA4/Fc–treated recipients, indicating the protective effect of biopatterned CTLA4/Fc in situ against alloreactive effector T-cell responses.

Active immune suppression by Treg is a prominent periphery tolerance mechanism. Nevertheless, inflammation of local tissue during transplantation not only limits Treg suppression but also promotes proinflammatory Th17 responses (22,23). Our data demonstrated that localized biopatterned CTLA4/Fc treatment reduced serum levels of IL-6 and IL-17 and decreased Gzmb gene and enhanced Foxp3 gene expression in both islet allografts and draining lymph nodes, accompanied by increased Treg cells. These findings further support the multifaceted immunomodulation conferred by localized biopatterned CTLA4/Fc. They may provide microenvironments that favor Treg generation in the hostile environment of an alloimmune response through suppressing cytotoxic T lymphocytes and inhibiting IL-6–mediated Th17 responses. In this study, the expansion of Treg and maintenance of their suppressive potency on effector T-cell proliferation against donor antigen in a mixed lymphocyte reaction may at least in part underlie the long-term allograft survival induced by the local biopatterned CTLA4/Fc therapy. A continuing presence of CTLA4/Fc in the islet allograft could especially benefit the recruitment of Treg to the allograft and ensure Treg effective control of antidonor reactivity. These factors encourage the further investigation of local immunoregulation for the induction of transplantation tolerance.

In conclusion, for the first time, we demonstrated that biopatterning of native matrices, such as ADM, with immunoregulatory agents, such as CTLA4/Fc, facilitates highly localized delivery of these agents without diminution of efficacy. Our study confirms that this technology promotes long-term engraftment of minimal loads of allogeneic islet cells and leads to sustained glucose homeostasis. The CTLA4/Fc microenvironment promoted expansion of Treg and shifted cytokine production and gene expression from proinflammatory to regulatory profiles. Our findings suggest that the targeted delivery of biopatterned CTLA4/Fc microenvironments could become a safe and effective strategy for minimizing systemic immunosuppression and promoting islet allograft survival in PIT.

Acknowledgments. The authors thank Kia M. Washington and Maxine Miller (Department of Plastic Surgery, University of Pittsburgh School of Medicine) for providing input and advice to our work.

Funding. This study was supported by a JDRF Innovative Grant (5-2012-308 to M.G.S. [principal investigator], X.X.Z., V.S.G., and P.G.C. [co-principal investigator]) and in part by the Plastic Surgery Foundation and Musculoskeletal Transplant Foundation Dermal Tissue Research Grant (349234 to W.Z. [principal investigator]).

Duality of Interest. No potential conflicts of interest relevant to this article were reported.

Author Contributions. W.Z., V.S.G., P.G.C., L.E.W., and X.X.Z. generated the original study conception. W.Z., V.S.G., P.G.C., L.E.W., X.X.Z., and M.G.S. designed experiments, analyzed data, and interpreted results. W.Z., V.S.G., and P.G.C. wrote the manuscript and prepared the figures. W.Z., V.S.G., P.G.C., L.E.W., X.X.Z., and M.G.S. made critical scientific input and revisions for intellectual content. W.Z., Y.L., Y.Y., and C.K. performed animal experiments, immunological assays, and data acquisition. W.Z. and X.X.Z. constructed the CTLA4/Fc fusion protein. P.G.C. and L.E.W. made the biopatterned constructs. M.G.S. coordinated and directed the project. M.G.S. is the guarantor of this work and, as such, had full access to all the data in the study and takes responsibility for the integrity of the data and the accuracy of the data analysis.

1.
Lenschow
DJ
,
Zeng
Y
,
Thistlethwaite
JR
, et al
.
Long-term survival of xenogeneic pancreatic islet grafts induced by CTLA4lg
.
Science
1992
;
257
:
789
792
[PubMed]
2.
Perez
VL
,
Van Parijs
L
,
Biuckians
A
,
Zheng
XX
,
Strom
TB
,
Abbas
AK
.
Induction of peripheral T cell tolerance in vivo requires CTLA-4 engagement
.
Immunity
1997
;
6
:
411
417
[PubMed]
3.
Vergani
A
,
D’Addio
F
,
Jurewicz
M
, et al
.
A novel clinically relevant strategy to abrogate autoimmunity and regulate alloimmunity in NOD mice
.
Diabetes
2010
;
59
:
2253
2264
[PubMed]
4.
Orban
T
,
Bundy
B
,
Becker
DJ
, et al.;
Type 1 Diabetes TrialNet Abatacept Study Group
.
Co-stimulation modulation with abatacept in patients with recent-onset type 1 diabetes: a randomised, double-blind, placebo-controlled trial
.
Lancet
2011
;
378
:
412
419
[PubMed]
5.
Charbonnier
LM
,
Vokaer
B
,
Lemaître
PH
,
Field
KA
,
Leo
O
,
Le Moine
A
.
CTLA4-Ig restores rejection of MHC class-II mismatched allografts by disabling IL-2-expanded regulatory T cells
.
Am J Transplant
2012
;
12
:
2313
2321
[PubMed]
6.
Lau
HT
,
Yu
M
,
Fontana
A
,
Stoeckert
CJ
 Jr
.
Prevention of islet allograft rejection with engineered myoblasts expressing FasL in mice
.
Science
1996
;
273
:
109
112
[PubMed]
7.
Olthoff
KM
,
Judge
TA
,
Gelman
AE
, et al
.
Adenovirus-mediated gene transfer into cold-preserved liver allografts: survival pattern and unresponsiveness following transduction with CTLA4Ig
.
Nat Med
1998
;
4
:
194
200
[PubMed]
8.
Schnider
JT
,
Weinstock
M
,
Plock
JA
, et al
.
Site-specific immunosuppression in vascularized composite allotransplantation: prospects and potential
.
Clin Dev Immunol
2013
;
2013
:
495212
[PubMed]
9.
Cooper
GM
,
Miller
ED
,
Decesare
GE
, et al
.
Inkjet-based biopatterning of bone morphogenetic protein-2 to spatially control calvarial bone formation
.
Tissue Eng Part A
2010
;
16
:
1749
1759
[PubMed]
10.
Shakir
S
,
MacIsaac
ZM
,
Naran
S
, et al
.
Transforming growth factor beta 1 augments calvarial defect healing and promotes suture regeneration
.
Tissue Eng Part A
2015
;
21
:
939
947
[PubMed]
11.
Miller
ED
,
Fisher
GW
,
Weiss
LE
,
Walker
LM
,
Campbell
PG
.
Dose-dependent cell growth in response to concentration modulated patterns of FGF-2 printed on fibrin
.
Biomaterials
2006
;
27
:
2213
2221
[PubMed]
12.
Herberg
S
,
Kondrikova
G
,
Periyasamy-Thandavan
S
, et al
.
Inkjet-based biopatterning of SDF-1β augments BMP-2-induced repair of critical size calvarial bone defects in mice
.
Bone
2014
;
67
:
95
103
[PubMed]
13.
Smith
DM
,
Cray
JJ
 Jr
,
Weiss
LE
, et al
.
Precise control of osteogenesis for craniofacial defect repair: the role of direct osteoprogenitor contact in BMP-2-based bioprinting
.
Ann Plast Surg
2012
;
69
:
485
488
14.
Miller
ED
,
Phillippi
JA
,
Fisher
GW
,
Campbell
PG
,
Walker
LM
,
Weiss
LE
.
Inkjet printing of growth factor concentration gradients and combinatorial arrays immobilized on biologically-relevant substrates
.
Comb Chem High Throughput Screen
2009
;
12
:
604
618
[PubMed]
15.
Campbell
PG
,
Skaar
TC
,
Vega
JR
,
Baumrucker
CR
.
Secretion of insulin-like growth factor-I (IGF-I) and IGF-binding proteins from bovine mammary tissue in vitro
.
J Endocrinol
1991
;
128
:
219
228
[PubMed]
16.
Zheng
XX
,
Sayegh
MH
,
Zheng
XG
, et al
.
The role of donor and recipient B7-1 (CD80) in allograft rejection
.
J Immunol
1997
;
159
:
1169
1173
[PubMed]
17.
Zhang
W
,
Zhang
D
,
Shen
M
, et al
.
Combined administration of a mutant TGF-beta1/Fc and rapamycin promotes induction of regulatory T cells and islet allograft tolerance
.
J Immunol
2010
;
185
:
4750
4759
[PubMed]
18.
Kheradmand
T
,
Wang
S
,
Gibly
RF
, et al
.
Permanent protection of PLG scaffold transplanted allogeneic islet grafts in diabetic mice treated with ECDI-fixed donor splenocyte infusions
.
Biomaterials
2011
;
32
:
4517
4524
[PubMed]
19.
Kimura
F
,
Gotoh
M
,
Tanaka
T
, et al
.
Locally expressed CTLA4-Ig in a pancreatic beta-cell line suppresses accelerated graft rejection response induced by donor-specific transfusion
.
Diabetologia
2002
;
45
:
831
840
[PubMed]
20.
Becker
S
,
Saint-Cyr
M
,
Wong
C
, et al
.
AlloDerm versus DermaMatrix in immediate expander-based breast reconstruction: a preliminary comparison of complication profiles and material compliance
.
Plast Reconstr Surg
2009
;
123
:
1
6; discussion 107–108
[PubMed]
21.
Stinn
JL
,
Taylor
MK
,
Becker
G
, et al
.
Interferon-gamma-secreting T-cell populations in rejecting murine cardiac allografts: assessment by flow cytometry
.
Am J Pathol
1998
;
153
:
1383
1392
[PubMed]
22.
Sakaguchi
S
,
Yamaguchi
T
,
Nomura
T
,
Ono
M
.
Regulatory T cells and immune tolerance
.
Cell
2008
;
133
:
775
787
[PubMed]
23.
Mitchell
P
,
Afzali
B
,
Lombardi
G
,
Lechler
RI
.
The T helper 17-regulatory T cell axis in transplant rejection and tolerance
.
Curr Opin Organ Transplant
2009
;
14
:
326
331
[PubMed]
Readers may use this article as long as the work is properly cited, the use is educational and not for profit, and the work is not altered. More information is available at http://www.diabetesjournals.org/content/license.