targeted extracellular delivery of indoleamine 2,3

162
TARGETED EXTRACELLULAR DELIVERY OF INDOLEAMINE 2,3 DIOXYGENASE VIA FUSION WITH GALECTIN 3 By EVELYN R. BRACHO SANCHEZ A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF FLORIDA IN PARTIAL FULFILLMENT OF THE REQUIREMENTS FOR THE DEGREE OF DOCTOR OF PHILOSOPHY UNIVERSITY OF FLORIDA 2017

Upload: others

Post on 27-Jan-2022

1 views

Category:

Documents


0 download

TRANSCRIPT

TARGETED EXTRACELLULAR DELIVERY OF INDOLEAMINE 2,3 DIOXYGENASE VIA FUSION WITH GALECTIN 3

By

EVELYN R. BRACHO SANCHEZ

A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF FLORIDA IN PARTIAL FULFILLMENT

OF THE REQUIREMENTS FOR THE DEGREE OF DOCTOR OF PHILOSOPHY

UNIVERSITY OF FLORIDA

2017

© 2017 Evelyn R. Bracho Sanchez

To my family, near and far, and their unconditional support

4

ACKNOWLEDGMENTS

I would like to express my deepest gratitude to my advisor, Dr. Benjamin

Keselowsky for his guidance, insight and influence during my development as a

scientist and engineer. To my committee and collaborators – Dr. Greg Hudalla, the

fusion proteins presented here were developed in partnership with him and his team,

Dr. Mark Wallet, Dr. Todd Brusko and Dr. Clayton Mathews for their support in

experimental design and guidance through the execution and analysis of immunological

assays, and Dr. Shannon Wallet for her contributions with the periodontal disease

model – your guidance through this process, countless suggestions and positive

feedback continues to be instrumental for my growth.

I would like to express my sincerest appreciation to all my lab mates and close

friends over the years. Thank you, Jamal and Matt for training me in my early career

and welcoming me into the Keselowsky Lab. Thank you, Maigan for explaining

immunology to me countless times, for entertaining my wildest theories, and teaching

me the joys of flow cytometry. Thank you, Antonietta, Maggie, Sabrina, Kevin and Josh

for cheering me on, for being my support system and safety net in the hardest and most

challenging times.

Lastly, I would like to thank my family. You have always been my source of

motivation, faith and energy and I share this accomplishment with you. To my parents,

Rosvel Bracho and Edith Sanchez, your unconditional love, sacrifices and example

continue to be the reason I thrive to be the best version of myself. Thank you for

believing in me even when I doubt myself, thank you for pushing me to always get back

up stronger and wiser. To my siblings, Edith and Rosvel, your courage to pursue your

dreams and remain authentic is inspiring, it is a blessing to have you on my corner. To

5

Jon, thank you for the countless ways you continue to love and support me and all my

goals, both personal and professional. To all my family in Venezuela, thank you for your

love through the distance and continuous encouragement, I am proud of my roots

because they always bring me back to you.

6

TABLE OF CONTENTS page

ACKNOWLEDGMENTS .................................................................................................. 4

LIST OF TABLES ............................................................................................................ 9

LIST OF FIGURES ........................................................................................................ 10

LIST OF ABBREVIATIONS ........................................................................................... 12

ABSTRACT ................................................................................................................... 15

CHAPTER

1 INTRODUCTION .................................................................................................... 17

Micro and Nano Material Carriers for Immunomodulation....................................... 17 Biomaterial-Carries ........................................................................................... 18

Polymeric ................................................................................................... 20 Micro- and nano-particles .......................................................................... 20

Micelles ...................................................................................................... 23 Dendrimers ................................................................................................ 24 Hydrogels ................................................................................................... 25

Lipids ......................................................................................................... 26 Metallic and inorganic ................................................................................ 27

Biologics..................................................................................................... 30

Fusion proteins .......................................................................................... 31

Carrier Properties Influencing Immune Responses .......................................... 33 Biomaterials-Based Immunomodulation of Dendritic Cells ..................................... 35

Innate and Adaptive Immunity Overview .......................................................... 37 Dendritic Cell Subsets ...................................................................................... 41

Inflammatory dendritic cell phenotype ........................................................ 42 Suppressive dendritic cell phenotype ......................................................... 43

Biomaterials-based Immunomodulation ........................................................... 44 Biomaterials-based DC Manipulation Toward an Inflammatory Phenotype ...... 44

Biomaterials as antigen carriers ................................................................. 45 Biomaterials as adjuvants and adjuvant carriers ........................................ 47

Biomaterials-based DC Manipulation Toward a Suppressive Phenotype ......... 50

2 EXTRACELLULAR INDOLEAMINE 2,3 DIOXYGENASE-TREATED DENDRITIC CELLS MAINTAIN AN IMMATURE PHENOTYPE AND SUPPRESS ANTIGEN-SPECIFIC T CELL PROLIFERATION ............................... 59

Background ............................................................................................................. 59 Indoleamine 2,3 Dioxygenase .......................................................................... 59

Tryptophan catabolism and the kynurenine pathway ................................. 59 Tissue and cellular expression ................................................................... 60

7

Role in immune regulation ......................................................................... 61

Materials and Methods............................................................................................ 62

IDO Characteristics and Activity Assay ............................................................ 62 Dendritic Cell Culture and Extracellular Enzyme Treatment ............................. 63 Dendritic Cell Phenotype and Maturation Resistance ...................................... 64 T Cell Isolation and Proliferation Assay ............................................................ 65 Statistical Analysis ............................................................................................ 66

Results .................................................................................................................... 66 IDO-treated DCs Resist LPS-induced Maturation ............................................ 66 IDO-treated DCs Suppress Antigen Specific Proliferation, and Suppression

is Active Enzyme Dependent ........................................................................ 68 Discussion .............................................................................................................. 70

3 FUSION OF GALECTIN 3 WITH A MODEL ENZYME AT THE N-TERMINUS PROLONGS RETENTION TIME IN VIVO .............................................................. 80

Background ............................................................................................................. 80 Galectin 3 ......................................................................................................... 80

NanoLuciferase ................................................................................................ 81 Materials and Methods............................................................................................ 82

Cloning of Galectin 3, Nanoluciferase and NanoLuc-Gal3 ............................... 82 Protein Expression and Purification .................................................................. 83 Mice and Cell Lines .......................................................................................... 84

Galectin 3 and NanoLucGal-3 Binding Affinity .................................................. 84 NanoLuc-Gal3 Binding Affinity to ECM Proteins ............................................... 84

NanoLuc-Gal3 Binding Affinity to Jurkat T Cells ............................................... 85

Jurkat T Cell Agglutination and Viability ........................................................... 85

Quantitative Precipitation Analysis of Galectin 3 and NanoLuc-Gal3 ............... 86 In Vivo Bioluminescence and Imaging .............................................................. 86

NanoLuc-Gal3 Tissue Distribution from Hock Injection .................................... 86 Alternative Injection Sites ................................................................................. 86 Statistical Analysis ............................................................................................ 87

Results .................................................................................................................... 87

NanoLuc-Galectin 3 Retains Binding Affinity for Sugar Moiety when Compared to WT-Gal3 .................................................................................. 87

Galectin 3 Retains Binding Affinity to Sugar Moiety on ECM and Serum Proteins When Fused to an Enzyme on the N-terminus ............................... 87

Galectin 3 Is Not Able to Self-assemble Into Pentamer and Precipitate Asialofetuin When Fused to NanoLuc on the N-terminus .............................. 88

Galectin 3 Binds to Jurkat T Cells but Does Not Induce Agglutination or Apoptosis ...................................................................................................... 88

NanoLuc-Gal3 Prevents Galectin 3-induced Cell Death ................................... 89 NanoLuc-Gal3 Binds Primary Splenocytes in a CRD Dependent Manner,

Binding Does Not Act as a Damage Associated Molecular Pattern .............. 90 Galectin 3 Targeting in NanoLuc-Gal3 Provides Prolonged Retention Time

When Administered Subcutaneously, Intraperitoneally and Intramuscularly .............................................................................................. 90

8

Galectin 3 Targeting in NanoLuc-Gal3 Provides Prolonged Retention Time When Administered Into the Hock with Minimal Drainage to Adjacent Tissues .......................................................................................................... 90

Discussion .............................................................................................................. 91

4 FUSION OF INDOLEAMINE 2,3 DIOXYGENASE WITH GALECTIN 3 RETAINS ENZYME AT INJECTION SITE AND HALTS INFLAMMATION IN VIVO ............. 103

Background ........................................................................................................... 103

Subcutaneous LPS-induced Inflammation...................................................... 103 Periodontal Disease ....................................................................................... 103

Immunopathogenesis ............................................................................... 104 Current treatments ................................................................................... 105

Materials and Methods.......................................................................................... 106

Cloning of IDO-Galectin 3 ............................................................................... 106

Protein Expression and Purification ................................................................ 106 IDO Enzymatic Activity Assay ........................................................................ 107 Galectin 3 and IDO-Gal3 Binding Affinity........................................................ 108

Antibodies ....................................................................................................... 108 Mice and Cell Lines ........................................................................................ 109

DC Maturation and Cytokine Release Profile ................................................. 109 Antigen Specific Co-cultures .......................................................................... 109 LPS Administration at the Hock as a Model of Inflammation .......................... 110

Quantitative PCR ............................................................................................ 110 Mass Spectrometry ........................................................................................ 110

Hock Infiltration ............................................................................................... 111

In vivo Bioluminescence and Imaging ............................................................ 111

Mucosal Induction of Inflammation ................................................................. 111 Subgingival Cytokine Production .................................................................... 112

Results .................................................................................................................. 112 IDO Activity and Gal3 Affinity Are Not Altered Upon Fusion ........................... 112 IDO Retains Immunomodulatory Properties When Fused to Galectin 3 ......... 112 IDO-Gal3 Reduces Inflammatory Cytokine Gene Expression at Various

Time Points. ................................................................................................ 114 IDO-Gal3 Modulates Local Tryptophan Catabolism ....................................... 115 IDO-Gal3 Modulates LPS-induced Inflammation Locally ................................ 116 Gal3 Provides Retention of a Model Enzyme at Subgingival Tissue,

Retention Time Is Not Altered by Infection State ......................................... 117

IDO-Gal3 Suppresses Inflammatory Cytokine and Chemokine Production .... 118

Discussion ............................................................................................................ 119

5 CONCLUSIONS AND FUTURE DIRECTIONS .................................................... 133

LIST OF REFERENCES ............................................................................................. 140

BIOGRAPHICAL SKETCH .......................................................................................... 162

9

LIST OF TABLES

Table page 1-1 Advantages and disadvantages of biomaterials carriers for

immunomodulation ............................................................................................. 19

10

LIST OF FIGURES

Figure page 1-1 Interaction of nano- and micro-scale biomaterial carriers with key members of

the innate and adaptive immune system ............................................................ 55

1-2 Simplified immune response to foreign pathogen ............................................... 56

1-3 Dendritic cell phenotype dictates T cell function. ................................................ 57

1-4 Overview of biomaterials-based dendritic cell modulation. ................................. 58

2-1 Tryptophan metabolism through the kynurenine pathway .................................. 74

2-2 Dendritic cells treated with IDO maintain an immature morphology even after LPS challenge .................................................................................................... 75

2-3 Treatment with soluble IDO does not induce dendritic cell death ....................... 76

2-4 Dendritic cells treated with soluble IDO resist LPS maturation. .......................... 77

2-5 Treatment of dendritic cells with exogenous IDO inhibits their IL-12 secretion and maintains IL-10 production. ......................................................................... 77

2-6 IDO-treated DCs suppress antigen specific T cell proliferation, and suppression is active enzyme dependent ........................................................... 78

2-7 Tryptophan depletion and kynurenine accumulation are needed to suppress antigen specific proliferation. .............................................................................. 79

3-1 Classification of galectins ................................................................................... 96

3-2 Fusion protein construct. .................................................................................... 96

3-3 Galectin 3 retains binding affinity to sugar moiety when fused to an enzyme on the N-terminus ............................................................................................... 97

3-4 Biology associated with Galectin 3 is disrupted when fused to an enzyme on the N-terminus .................................................................................................... 98

3-5 NanoLuc-Gal3 fusion prevents wild type Galectin 3-mediated agglutination and apoptosis of Jurkat T cells. .......................................................................... 99

3-6 NanoLuc-Gal3 fusion binds primary splenocytes in a CRD dependent manner, binding to DCs does not act as a damage associated molecular pattern .............................................................................................................. 100

11

3-7 Galectin 3 targeting in NanoLuc-Gal3 provides prolonged retention when administered subcutaneously, intraperitoneally, and intramuscularly. .............. 101

3-8 Galectin 3 targeting provides retention of a model enzyme at the injection site with minimal drainage to adjacent tissues. ....................................................... 102

4-1 Enzymatic activity of IDO is not altered upon fusion with Galectin 3, Galectin 3 retains binding affinity to sugar moiety upon fusion with IDO at N-terminus .. 124

4-2 Indoleamine 2,3-dioxygenase maintains an immature dendritic cell phenotype when fused to Galectin 3. ............................................................... 125

4-3 IDO-Gal3-treated DCs attenuate antigen specific T cell proliferation, suppression is active enzyme dependant. ........................................................ 126

4-4 IDO-Gal3 blocks subcutaneous LPS-induced inflammatory cytokine gene expression. ....................................................................................................... 127

4-5 IDO-Gal3 modulates tryptophan metabolism at the injection site. .................... 128

4-6 IDO-Gal3 blocks subcutaneous LPS-induced inflammatory response, suppression is not systemic .............................................................................. 129

4-7 Galectin 3 provides retention of a model enzyme in the subgingival space; retention profile is not altered by disease state................................................. 130

4-8 Schematic representation of infection model used to induce periodontal disease in mice. ................................................................................................ 131

4-9 Inflammatory cytokine and chemokine production in the subgingival space is significantly reduced by administration of IDO-Gal3 both prophylactically and therapeutically. ................................................................................................. 132

5-1 Proposed mechanism for IDO-related suppression of inflammation by blockage of NF-κβ pathway initiated through binding of LPS to TLR4 on surface of immune cell. ..................................................................................... 138

5-2 Proposed in vivo cytokine network affected by the introduction of IDO. ........... 139

12

LIST OF ABBREVIATIONS

AhR Aryl hydrocarbon receptor

APC Antigen presenting cells

B6 C57BL/6

B-PER Bacterial protein extraction reagent

CD Cluster differentiation

CFSE Carboxyfluorescein succinimidyl ester

CRD Carbohydrate recognition domain

CREB cAMP response element binding protein

DCs Dendritic cells

DNAse Deoxyribonuclease

EDTA Ethylenediaminetetraacetic acid

ELISA Enzyme linked immunosorbent assay

Gal3 Galectin 3

GM-CSF Granulocyte-macrophage colony stimulating factor

H&E Haemotoxylin and Eosin

IACUC Institutional Animal Care and Use Committee

IDO Indoleamine 2,3 dioxygenase

IFNγ Interferon gamma

IΚΚ IκB Kinase

IL-10 Interleukin 10

IL-12 Interleukin 12

IL-1β Interleukin 1 beta

IL-33 Interleukin 33

IL-6 Interleukin 6

13

IM Intramuscular

IP Intraperitoneal

IP10 Interferon gamma-induced protein 10

IPTG isopropyl β-D-1-thiogalactopyranoside

IRAK IL-1 Receptor-Associated Kinases

IV Intravenous

KC Keratinocyte chemoattractant

LacNAc N-Acetyl-D-lactosamine

LATs L-amino acid transporters

LPS Lipopolysaccharide

MAPK Mitogen-Activated Protein Kinase

MCP1 Monocyte chemotactic protein 1

MHC I/II Major histocompatibility complex class I and II

MIP2 Macrophage inflammatory protein 2

MT 1-methyl tryptophan

MyD88 Myeloid Differentiation Primary Response Gene 88

NanoLuc Nanoluciferase

NanoLuc-Gal3 Nanoluciferase-Galectin 3 fusion protein

NF-κβ nuclear factor kappa-light-chain-enhancer of activated B cell

NK cells Natural killer cells

OT-I C57BL/6-Tg(TcraTcrb)1100Mjb/J mice

OT-II B6.Cg-Tg(TcraTcrb)425Cbn/J mice

OVA Ovalbumin

PBS Phosphate-buffered saline

PD Periodontal disease

14

SDS-PAGE sodium dodecyl sulfate polyacrylamide gel electrophoresis

Subq Subcutaneous

TAK1 Transforming growth factor-β-Activated Kinase 1

Teff Effector T cell

TLR4 Toll-like receptor 4

TNFα Tumor necrosis factor alpha

TRAF6 TNF Receptor-Associated Factor

Treg Regulatory T cell

15

Abstract of Dissertation Presented to the Graduate School of the University of Florida in Partial Fulfillment of the Requirements for the Degree of Doctor of Philosophy

TARGETED EXTRACELLULAR DELIVERY OF INDOLEAMINE 2,3 DIOXYGENASE

VIA FUSION WITH GALECTIN 3

By

Evelyn R. Bracho Sanchez

December 2017

Chair: Benjamin Keselowsky Major: Biomedical Engineering

Suppression of inflammatory processes would be beneficial for the treatment of

autoimmune and inflammatory diseases as well as whole organ transplantation. Current

treatments are limited to non-specific systemic immunosuppressant drugs which carry

harmful off-target effects and the potential for opportunistic infections. To address these

limitations, scientists have focused on reprogramming patients own immune systems to

take advantage of naturally occurring tolerance mechanisms such as the upregulation of

indoleamine 2,3 dioxygenase (IDO). Indoleamine 2,3 dioxygenase is an intracellular

enzyme responsible for initiating tryptophan catabolism to kynurenine and downstream

metabolites, and is primarily expressed by lymphoid organs and the placenta.

Overexpression of IDO has been shown to induce T cell tolerance in transplant and

pregnancy. Thus, efforts have primarily focused on increasing intracellular enzyme

expression in antigen presenting cells using pharmacological agents, cytokines and

genetic manipulation. These approaches are constrained by several factors, including

the potentially deleterious biological effects of IDO-inducing agents such as

lipopolysaccharide (LPS) or interferon gamma (IFNγ), or malignancies and even

lethality associated with systemic overexpression. Here we report a new approach for

16

the metabolic programming of immune environments through the local delivery and

retention of IDO, a potent immunomodulator, through fusion with the carbohydrate

binding lectin Galectin 3 (Gal3). This study establishes IDO as an immunomodulator in

the extracellular space and uniquely employs its enzymatic activity for the suppression

of excessive inflammation. Galectin 3 is a member of the lectin family with affinity for N-

Acetyl-D-lactosamine (LacNAc) glycosylation patterns present, for example, on proteins

of the extracellular matrix, such as laminin, and immune cell surfaces. A fusion chimera,

IDO-Gal3, was developed, characterized and demonstrated to maintain function for both

the targeting and enzymatic moieties, in vitro and in vivo. Galectin 3 confers

subcutaneous and oral submucosal retention for 7 and 10 days, respectively, and IDO-

Gal3 suppresses LPS-induced inflammation in a subcutaneous model as well as in a

polymicrobial model of periodontal disease. The findings from this research project will

provide valuable information regarding IDO-Gal3 as a treatment for chronic

inflammation, autoimmune diseases and transplant tolerance.

17

CHAPTER 1 INTRODUCTION

Micro and Nano Material Carriers for Immunomodulation

The immune system is intricately organized, composed of multiple layers that

work in unison to protect the host against foreign invaders and provides homeostatic

regulation of self and non-self. Appropriate immune recognition initiates isolation and

elimination of pathogens, and tolerance to self or benign antigens, such as food

proteins1. Due to its crucial role in health and disease, manipulation of the immune

system by therapeutic interventions is of great interest for the amelioration of

malignancies. Immunostimulation may be sought, as in the case of vaccines and

adjuvants for infectious diseases and cancer. Other times, immunosuppression, or

diminished immune potency, is desired. While systemic immunosuppressants lower the

body’s ability to fight foreign invaders systemic treatments continue to be required for

allergies, autoimmune diseases and transplant rejection.

Cell and whole-organ transplantation has become a standard procedure for the

treatment of numerous conditions including cardiac, hepatic and renal failure2. Donor

tissue is normally derived from an allogenic source, which upon introduction to the

recipient activates a cascade of immune responses. Much progress has been made due

to immunosuppressant therapies, however chronic rejection and dysfunction persist,

with only 47-61% of grafts surviving to the 10 year mark3. Therefore, the induction of

antigen specific tolerance to transplanted tissues remains a primary objective. Antigen

specific therapies aim at preventing the host from rejecting cells or whole-organs while

maintaining complete and functional activity to fight foreign invaders. To achieve

tolerance, key cellular players must be engaged and re-programmed, including antigen

18

presenting cells (APCs,) such as dendritic cells (DCs) and macrophages, T and B

lymphocytes, a strategy currently been explored through the use of nano- and micro-

technologies.

Biomaterials offer unique opportunities to modulate the immune system either

toward a suppressive or stimulatory state by engaging components of the innate and

adaptive immune system (Figure 1-1). Biomaterials are synthetic or naturally-derived

materials suitable for incorporation into the human body, and are meant to perform,

enhance or replace physiological functions4. Given the variety and complexity of signals

that must work together to achieve an immunological outcome, the type of biomaterial,

its structure and properties should be considered when designing nano- and micro-

technologies to ameliorate health concerns. In this chapter, we focus on recent

advancements of biomaterials-based nano- and micro-technologies for

immunomodulation.

Biomaterial-Carries

Many nano and micro-scale biomaterial systems have been described as

platforms for targeting and delivery of therapeutic agents and effective

immunomodulation. These agents can be encapsulated, conjugated, fused or adsorbed

onto the material system and co-delivered with excipients or stabilizers to achieve ideal

release profiles and immunological responses5. Many carriers have been proposed and

can be sorted into polymeric, lipids, metals and inorganics. Recent advances and

limitations of each carrier type are highlighted below and summarized (Table 1).

19

Table 1-1. Advantages and disadvantages of biomaterials carriers for immunomodulation

ADVANTAGE DISADVANTAGE

POLYMERIC CARRIERS

MICRO AND NANO PARTICLES

High stability High loading capacity of hydrophilic and hydrophobic molecules Tunable properties Feasibility of various routes of administration

Aggregation leads to difficult handling Fabrication processes are harsh for proteins and enzymes

MICELLES Tunable properties Controlled release Incorporation of poorly soluble molecules

Require stabilizers at low concentrations Difficult polymer synthesis

DENDRIMERS Dimensional length scaling and narrow size distribution Controlled conjugations

Low biocompatibility Material selection may result in increased toxicity High manufacturing cost

HYDROGELS Controlled release Highly biocompatible

Low tensile strength Limited quantity and homogeneity of loads of hydrophobic molecules

LIPID CARRIERS LIPOSOMES Stable encapsulation of both

hydrophilic and hydrophobic molecules Biocompatible, biodegradable Ability to cross lipid bilayer Non-immunogenic

High manufacturing costs Difficult to sterilize Short shelf life and stability

METAL AND INORGANIC

GOLD Controllable synthesis Biocompatible

Non-biodegradable Biodistribution

CARBON Controllable synthesis Multivalent surface conjugation

Toxicity Non-biodegradable

SILICA Ease of fabrication Tunable properties

Non-biodegradable Require surface functionalization for biocompatibility

BIOLOGICS ANTIBODIES High affinity

High bioactivity High doses required High production and distribution cost

PROTEINS Naturally derived Low immunogenicity

Short half-life in vivo Formulation changes upon packaging

20

Polymeric

Polymeric biomaterials have been extensively investigated for the delivery of

drugs, biomolecules and genes. Biocompatibility, low toxicity and biodegradability have

promoted their use as a promising strategy. Additionally, chemical structures and

compositions can be easily tuned to achieve desirable properties such as controlled

release profiles. Examples of polymers widely used for delivery applications include

polyesters (e.g., poly(lactic acid), poly(glycolic acid) and their copolymers),

polyorthoesters, polyanhydrides and polycarbonates. These materials can be fabricated

in the form of particles, micelles, dendrimers and hydrogels, and have each been

extensively studied.

Micro- and nano-particles

The most common form of polymeric carriers are micro and nanoparticles, which

are highly stable can effectively entrap and adsorb hydrophobic as well as hydrophilic

molecules, and are easily administered through various routes. Particle sizes ranging

from nanometers to micrometers can be transported through cellular and subcellular

barriers making them amenable for site specific targeting. In the context of

immunomodulation, for example, polymeric particles can be designed to display

proteins commonly expressed by DCs, thus mimicking APCs and dictating T cell

activation and differentiation. These have been referred to as artificial APCs (aAPCs),

the most widely used form consists of polystyrene beads surface coated with anti-

CD3/CD28 antibodies allowing the delivery of antigen-independent signal to polyclonal

T cells. For antigen-specificity, polystyrene beads have also been coated with MHC-

peptide single chain construct dimers or tetramers and have been used for the ex vivo

expansion of tumor specific T cells.

21

In addition to stimulation of the T cell receptor signaling pathway through

CD3/CD28, co-stimulation and inhibitory signals can be provided by covalently binding

agonistic or antagonistic ligands to the beads. In a recent study, Hippen et al.,

demonstrated anti-CD3 antibody-loaded, aAPCs that displayed CD64 and CD86 on

their surface were able to expand human natural regulatory T cells (nTregs) 6. Upon

original contact nTregs numbers increased 80-fold and a single re-stimulation increased

expansion to 3,000 fold while maintaining Foxp3 expression and suppressor function.

These cells were then infused into an immune-deficient mice and significantly reduced

graft-vs-host lethality, a disease most commonly seen following a bone marrow

transplantation. In another study, Clemente-Casares et al., showed that systemic

delivery of nanoparticles coated with autoimmune-disease relevant peptides bound to

MHC II molecules triggers the generation and expansion of antigen-specific regulatory

CD4+ T cells type 1 (Tr1)-like cells in vivo preventing and reversing type 1 diabetes7.

Tolerance can be introduced not only by transplanting regulatory T cells but by

manipulating existing ones. Low dose IL-2 treatment has been shown to increase the

counts of regulatory T cells ameliorating graft vs host disease and a number of other

autoimmune and inflammatory conditions 8. Biomaterials engineering has incorporated

these findings and increased the functionality of aAPCs by also providing controlled

release of encapsulated cytokines. For example, Steenblock et al., fabricated poly

(lactic co-glycolic) acid (PLGA) microparticles surface-modified with anti-mouse CD3

and CD28 antibodies, and encapsulating IL-2. The authors demonstrated aAPCs

stimulated T cells more strongly than particles with surface ligands alone, and 10-fold

22

higher than soluble supplementation of the cytokine9. Furthermore, they showed the

response of T cells was dependent on the sustained release of IL-2.

In a different approach, polymeric particles can be designed for interactions with

phagocytes and lymphocytes in mind either for the detection or prevention of transplant

rejection 10-12. Early studies aimed to limit immune detection by avoiding protein

adsorption and opsonization, and the use of polyethylene glycol coatings on particles

rapidly emerged, as the polyethylene glycol layer sterically resists protein interactions13.

However, recent efforts have instead focused on actively directing phenotype and

function of immune cells14,15. For example, Shirali et al., fabricated mycophenolic acid

loaded PLGA nanoparticles to prolong murine skin allograft survival by upregulating PD-

L1 on dendritic cells 16; Hlavaty et al., developed PLG antigen-loaded nanoparticles to

promote bone marrow transplant tolerance in sex-mismatched C57BL/6 mice by

interactions with CD4+ and CD8+ T cells17 and Pan Q et al., administered corticosteroid-

loaded PLGA nanoparticles weekly for the prevention of corneal allograft rejection in

rats18. These studies represent an advancement in transplant therapies without the use

of a systemic immunosuppressant. Additionally, Lewis et al. developed a dual

microparticle delivery system using PLGA to encapsulate combinations of immuno-

suppressive factors to condition DCs toward a tolerance-inducing phenotype19.

Following up on this approach, Lewis et al., subcutaneously administered

phagocytosable (encapsulating Vitamin D3 and insulin peptide as antigen) and non-

phagocytosable microparticles (encapsulating TGF-β1 and GM-CSF) to non-obese

diabetic mice and demonstrated 40% protection from Type 1 Diabetes development 20,

representing one of the few microparticle vaccine system to successfully prevent

23

autoimmune diabetes. Furthermore, combinatorial approaches for loading adjuvants

into polymeric carriers are also being investigated, and tested using cellular based

microarrays 21-23

Micelles

Micelles are colloidal particles consisting of self-assembled aggregates of

amphiphilic molecules or surfactants. In aqueous solutions and at low concentrations,

amphiphiles exist as monomers. However, as their concentration increases,

thermodynamic processes drive the formation of aggregates sequestering hydrophobic

regions into a core like structure surrounded by hydrophilic shell24. Micelles have been

used to contain hydrophobic or poorly soluble drugs within its core. Following

administration, dilution occurs rapidly and if the concentration drops below the critical

micelle concentration, the stability can be compromised. However, with the addition of

stabilizers, micelle carriers have successfully been employed by various groups in the

context of immunomodulation. For example, Dane et al., delivered drug-loaded micelles

to lymph nodes and prolonged allograft survival. Specifically, the authors used

poly(ethylene glycol)-bl-poly(propylene sulfide) block copolymers 50 nm micelles to

encapsulate rapamycin and tacrolimus and showed a 2-fold improvement in survival of

MHC-mismatched tail skin allograft in a BALB/c mouse model 25. In another study, Miki

et al., supplemented a dendritic cell vaccine with polymeric nano-micelles comprised of

PEG-polyGlutamate block co-polymer carrying IL-2, and demonstrated enhanced intra-

tumoral accumulation of antigen-specific cytotoxic T lymphocytes in EG7 tumor bearing

mice. Furthermore, this micelle system was able to prolong IL-2 retention in blood

circulation, significantly increasing DC vaccine efficacy against tumors 26.

24

Dendrimers

Dendrimers can be linear, cross-linked, or branched macromolecules forming a

star-like structure. They have been referred to as artificial proteins based on their

dimensional length scaling and narrow size distribution. Dendrimers offer a robust,

covalently fixed, three dimensional structure that can be divided into three domains: (i)

the multivalent surface, containing a large number of potentially reactive sites, (ii) the

interior shell consisting of branches referred to as dendrons, and (iii) the core 27. The

first and most extensively studied dendrimer, poly(amido amine) (PAMAM), is

synthesized by using a step-wise fashion. This results in precisely defined structures

with large functional groups at the surface providing opportunities for controlled

conjugations of drugs and targeting moieties, separating dendrimers from other carriers

by virtue of their well-controlled chemistry. However, dendrimers have shown low

biocompatibility and the material selection may lead to increased toxicity.

Much of the work with dendrimers has focused on the encapsulation in its core

and the covalent attachment of drugs to its surface. For example, carbohydrates

constitute an important class of biological recognition molecules, displaying a wide

variety of spatial structures due to their branching and various occurring isomers. In

order to achieve high binding with cell surfaces, carbohydrates must be presented in a

multivalent or cluster fashion28, and functionalization of dendrimers provides an

excellent platform for such multivalent presentation. For example, Heimburg et al.,

created a PAMAM dendrimer bearing the Thomsen-Friedenreich carbohydrate antigen

(a well-documented antigen for the detection and therapy of carcinomas, particularly

relevant to breast cancer) that was able to raise IgG antibodies against the antigen. This

successfully impeded binding of malignant cells to vascular endothelium, blocking a

25

metastatic step and providing a survival advantage29,30. Although progress has been

made to understand the capability of dendrimers as therapeutics in immune

applications, their clinical translation has been limited by some concerns over

biocompatibility and toxicity. Dendrimers can have affinity for metal ions, lipids, proteins

and nucleic acids sometimes resulting in the disruption of biological processes31.

Additionally, the expense associated with the multi-step synthesis of dendrimers has

also been a concern for translation.

Hydrogels

Hydrogels are three-dimensional, cross-linked networks of highly water-soluble

polymers with high porosity encompassing a wide range of chemical compositions and

bulk properties. They can be formulated in a variety of different forms including micro-

and nano- particles, scaffolds, coatings and films32. Hydrogels can be tuned by

controlling the density of cross-links in the gel matrix and drugs can be loaded with

factors for short-term release at a rate dependent on the diffusion coefficient of the

molecule through the network. Hydrogels can also be highly biocompatible, with a high

water content and similar mechanical properties as the extracellular environment of soft

tissues.

The utilization of hydrogels in transplant applications dates back to the 1980s

when encapsulation of pancreatic islets restricted contact between the donor islets and

the recipients immune cells in diabetic rats33. The pore size of the hydrogel was large

enough to allow small molecules and signaling proteins, including insulin, through but

small enough to block cells and the complement system which resulted in a delayed

rejection. In a more recent study conducted by Neufeld et al., encapsulation of rat islets

within polymer film membranes restored normoglycemia in chemically-induced diabetic

26

pigs for three months with no additional immunosuppression required 34. In principle,

encapsulation decreases the need for systemic immunosuppression, however eventual

loss of glycemic control continues to be observed 35

Hydrogels also have limitations. For instance, low tensile strengths limits their

use in load bearing applications, and can cause premature dissolution. Additionally, due

to their high-water content the quantity and homogeneity of agents loaded into

hydrogels may be limited, particularly in the case of hydrophobic drugs. Their large

water content and high porosity often results in a quick release profile of only a few

hours to a few days, maximum. Clinical administration may also be a concern. Although

many hydrogels can be injected, some must be implanted surgically, giving rise to a

new set of complications.

Lipids

Liposomes are typically assemblies composed of one or more bilayers of

amphipathic lipid molecules enclosing aqueous compartments. Their structure allows

hydrophilic molecules to be incorporated within the inner compartment, while the

hydrophobic compounds will be entrapped in the hydrophobic bilayer. Considering their

biocompatibility, biodegradability and ability to cross lipid bilayer and cell membranes,

liposomes have been widely demonstrated for various delivery platforms for vaccines36,

cancer treatments37, gene therapy38 and transplant39 in various forms including single

layer liposomes, solid lipid nanoparticles and phospholipid micelles. Although lipids tend

to be non-immunogenic, this feature may be intentionally altered by the incorporation of

antigens, and surface ligands.

Liposomes have shown much promise in advanced clinical trials for many years,

with at least two adjuvant systems currently approved for human use: Inflexal®V and

27

Epaxal® both marketed by Crucell (Leiden, Netherlands). More specifically, both

vaccines uses virosomes (unilamellar phospholipid membrane vesicle incorporating

virus derived proteins) to deliver either influenza (Inflexal®V) or hepatitis A (Epaxal®)

antigens to APCs in order to stimulate strong immune responses40,41. Through these

studies it has been hypothesized that the inherent ability of APCs to sequester

nanoscale liposomes more efficiently than larger-sized liposome counterparts may be a

key to enhanced immune responses observed with nano or micro liposome

formulations. More recently, liposomes have been investigated as a novel approach to

induce long-term tolerance in organ transplantation without continuous administration of

immunosuppressants. Hirai et al., at REGiMMUNE Corporation (Santa Clara, CA),

demonstrated donor-specific tolerance can be achieved by induction of mixed

chimerism in various animal models of bone transplantation 42. The authors describe a

novel approach using a ligand (alpha-GalCer) for invariant Natural Killer T cells and

antibody for CD40-CD40L blockage. Treatment resulted in complete acceptance of

transplanted bone marrow as well as cardiac allograft from the same donor.

As with any carrier discussed here, liposomes also have limitations. They are

highly susceptible to chemical and physical degradation resulting in extremely high cost

of manufacturing as conventional cost-effective sterilization techniques may not be

employed. Currently, filtration and aseptic technique are recommended for the

preparation of liposomes to be used in clinical settings. Liposomes also display low

stability decreasing their shelf-life and limiting their widespread application.

Metallic and inorganic

Many inorganic materials have been studied for their use in vaccine and various

immunology related applications. A well-established example is particulates of calcium

28

phosphate, aluminum hydroxide and aluminum phosphate, collectively referred to as

alum. This inorganic material causes antigen aggregation providing a depot and serving

as an adjuvant for many vaccines. Interestingly, alum remains the only material

approved by the U.S. Food and Drug Administration as an adjuvant in human vaccines.

Although inorganic materials may be non-biodegradable, their advantage lies in their

rigid structure and controllable synthesis43. Gold, carbon, and silica particles have all

been studied for their use in vaccine development.

Gold was one of the first metals to be discovered and its use in medical

applications can be traced back to the seventeenth century. Gold particles can easily be

fabricated into different shapes (spherical, rod, cubic, shell), with a size range of 2-150

nm, and can be surface-conjugated to achieve desired outcomes44. Gold nanorods in

particular, have been used as carriers for antigens derived from various viruses such as

influenza45, or as DNA adjuvants for human immunodeficiency virus (HIV)46.

Furthermore, gold nanoparticles have been conjugated with dye-oligonucleotide ( 5′-

Cy5-GAG CTG CAC GCT GCC GTC AAA AAA AAA A-SH-3′) to investigate a non-viral

transfection delivery system for pancreatic islet cell transplantation 47. The authors

demonstrated transfected islets maintained normal mitochondrial function, calcium influx

and insulin release when stimulated by glucose both in vitro and in vivo. This technology

has the potential to facilitate a wide variety of applications, such as the direct

manipulation of factors following transplantation using gold nanoparticles complexed

with siRNA, functional proteins, and pharmacological agents that have previously been

shown to improve outcomes of pancreatic islet transplants. Although widely used in

29

experimental models, the biodistribution, circulation time and toxicity of gold particles

continue to raise concerns and limit their application in clinical settings.

Carbon nanoparticles are another inorganic composition for drug and vaccine

delivery. Carbon nanoparticles are easily synthesized into a variety of shapes

(nanotubes, mesoporous spheres, etc), and can be made to be biocompatible. In

particular, carbon nanotubes offer the possibility of multivalent surface conjugation of

peptide antigens. For example, Villa et al. investigated the delivery of single-wall carbon

nanotubes as antigen carriers to APCs to promote responses to human tumor antigens.

The authors covalently attached a large number of peptide ligands on to carbon

nanotubes. Immunization of mice with the construct along with adjuvant induced specific

IgG responses against the peptide, while in comparison, the peptide with adjuvant

without the vehicle did not induce such a result48.

Although carbon based carriers have attracted much attention because of their

unique physical, chemical and mechanical properties, toxicity data at the molecular,

cellular and whole animal level is often conflicting. During large-scale preparation and

purification procedures impurities, mainly metal catalysts residues, are introduced and

virtually impossible to remove without destroying the structural integrity of the carrier.

These impurities are often released from carbon particles leading to increased oxidative

stress, inflammatory responses, malignant transformation and DNA damage or

mutation49.

Lastly, of the inorganic family, a promising material for immunomodulation is

silica. Silica nanoparticles have properties amenable for various applications including

tumor targeting, real time imaging, and vaccine delivery. They can be prepared with

30

tunable properties including size, shape and porosity which can alter interaction with

immune cells 50. Furthermore, its abundant surface silanol makes silica unique as it

allows for further conjugation and introduction of modulation of cell recognition,

absorption or uptake. For instance, Xia et al. demonstrated that polyethyleneimine

coating of mesoporous silica nanoparticles enhanced pancreatic cancer cellular uptake

and safely delivered siRNA and DNA constructs 51.

Biologics

Given their ubiquitous presence, diverse roles, and importance in the body, it

should not be surprising that proteins dominate the growing list of more than 200

approved biotherapeutic agents used in medicine today52. Protein therapies include the

abundantly present albumin important for osmolarity and volume of blood, vaccines,

enzymes, targeted antibodies, with monoclonal antibodies dominating the market,

fusion proteins and receptors, factors involved in blood clotting, homeostasis and

thrombosis, the potent botulinum neurotoxin, and hormones and cytokines as signaling

and immunomodulating agents, respectively. Protein-based therapies and carriers offer

several advantages over small molecules such as specificity of action and potent

therapeutic efficacy. Additionally, they have predictable behavior following

administration resulting in fewer side effects (e.g. decreased immunogenicity) and faster

regulatory approval time.

Protein-based carriers are often prepared by recombinant DNA technology and

produced in bacterial, yeast, insect or mammalian cells as they are more efficient and

cost-effective means of production. However, the complexity of proteins poses a

challenge as the possibility for heterogeneity due to changes in amino acid sequence,

presence and degree of glycosylation and folding conformation may affect the

31

properties mentioned above. Some proteins have short half-life in serum requiring

frequent parenteral administration leading to poor patient compliance.

Fusion proteins

Chimera fusion proteins, produced through genetic engineering, consist of a

carefully selected and usually short-lived effector domain, generally a peptide, coupled

with a carrier, usually a protein or peptide, that also contributes to the functional

properties of resultant fusion proteins. The linked effector domain can have a wide

variety of functions for example, contributing to recognition, binding and toxicity while its

fused partner can aid in stability or targeting. Effector moieties employed to date have

been limited to the ligand-binding portions of receptors of a few cytokines and growth

factors53,54, extracellular domains of lymphocyte antigens55, coagulation factors56 and

fragments of a toxin57. Three overarching categories drive the successful development

of chimeric proteins: (i) stability or extended half-life (ii) effective targeting and

subsequent binding (iii) capacity to inhibit deleterious processes underlying treated

condition. Most peptides likely to be considered as effectors have a short half-life due to

proteolytic degradation and are usually rapidly cleared, minutes to hours, through the

kidneys. Conjugation to polyethylene glycol (PEG), or pegylation, can extend half-life by

increasing the hydrodynamic radius and decreasing filtration in the kidneys. However,

safety concerns regarding the lack of biodegradability have given pegylation a bad

reputation and have inspired scientists to seek alternative methods.

The Fc region of the human IgG antibody has been the most commonly

employed fusion partner with multiple applications approved for clinical use53,58. The Fc

portion of IgG consist of the CH2-CH3 domains of the immunoglobulin heavy chain, the

hinge region and the two disulfide bridges connecting the heavy chains. In most fusion

32

proteins, the C-terminus of the effector molecule is fused to the N-terminus of the hinge

region59. Receptor-mediated recycling via interaction with the salvage neonatal FcRn

receptor protects Fc-containing molecules from lysosomal degradation. At low pH, FcRn

salvages the Fc fragment by binding, recycling, and releasing the protein in the blood at

neutral pH thus extending the Fc half-life. Furthermore, Fc fusion proteins can also

interact directly with immune cells, dictating the overall outcome of the immunological

response. Although 10 of the 12 currently FDA-approved fusion proteins are Fc derived,

multiple disadvantages have been observed including increased risk for infection,

neurological side effects, and allergic reaction (reviewed in depth in 59) inspiring the

search for alternative fusion constructs to continue.

Human serum albumin fusions have also been employed as partners in

therapeutic chimeric proteins. Albumin, the protein present in the greatest concentration

in plasma, has relative long half-life (14 d) and is normally considered safe due to its low

immunogenic potential. Effector molecules can be attached at either the N- or C-

terminus, and like the Fc fusion proteins, albumin can also engage the FcRn receptor

resulting in recycling by binding at low pH and release at neutral pH. The most

commonly occurring drawback seeing with albumin, however, is the interference of the

carrier with the specific activity of the effector molecule and its target. This has occurred

despite the use of linkers and efforts to orient the relevant domains60. Despite

drawbacks, albumin-based fusions have been successfully approved and implemented

in clinical settings with Eperzan® (dipeptidyl peptidase-4-resistant dimer fused to

albumin) employed for the treatment of type 2 diabetes61, and Idelvion® (factor IX –

albumin fusion protein) for the treatment of hemophilia B62.

33

Carrier Properties Influencing Immune Responses

Pre-clinical and clinical evaluations of many types of carriers have demonstrated

that material properties are related to their biological outcome. Much can be learned

from nature as immune cells have evolved to respond to pathogens displaying many

sizes, shapes and surface charges. These same properties are important

considerations when designing carriers for immunomodulation. Carrier size appears to

be a major influence in the cellular uptake and further endocytic pathway directing their

intracellular fate and thus overall biological effect. Carriers may be assimilated by

receptor-mediated endocytosis, which relies on the specific recognition of surface

receptors and their ligands; by receptor-independent endocytosis (pinocytosis) referring

to the invagination of the cell membrane encapsulating liquids from the extracellular

environment; or phagocytosis in which solid factors are engulfed by the cell membrane.

Carriers with diameters larger than 0.5 µm tend to be assimilated through

phagocytosis63, which is carried out by members of the innate immune system (e.g.,

DCs, macrophages, neutrophils and mast cells), and leads to cargo degradation in

lysosomes and presentation on the cell surface for recognition by the adaptive immune

system. Smaller carriers, less than 150 nm, are generally taken by cells via receptor-

mediated endocytosis or pinocytosis which are involved in the uptake of essential

nutrients, downregulation of cell signaling by internalization and degradation of

receptors, and maintaining cellular homeostasis64.

It has also been reported that geometrical shape of particulate carriers influences

cellular uptake and trafficking. While spherical polymeric carriers are quickly

internalized, anisotropic systems are poorly phagocytosed thus increasing their

circulation time and systemic delivery of their cargo65. To demonstrate this concept,

34

Champion et al., used polystyrene particles of various sizes and shapes to study the

phagocytosis of alveolar macrophages. The authors report that all shapes were able to

initiate phagocytosis in at least one direction. However, it was reported that the point of

contact dictated whether macrophages phagocytosed or simply spread on the particles,

concluding this effect is based on the actin structure that must form around the particle

to be internalized66. Further studies elucidating the role of biomaterials shape will not

only allow researchers to understand immune cells interactions with pathogens further

but could inform the design of micro and nano carrier-based therapeutics.

Also important for the design on new technologies to modulate the immune

system, is consideration of carriers’ surface properties, which plays a role in interactions

with innate immune cells. For example, charged gold particles are reported to be more

toxic than their neutral counter parts67, cytotoxicity of PAMAM dendrimers is correlated

with the number of primary amino groups 68, and DCs and macrophages preferentially

interact with cationic molecules69. The surface of biomaterials can also be modified by

protein adsorption, which can direct subsequent cell-protein-material interactions 70-74.

For example, Acharya et al., demonstrated that DC morphology and production of

cytokines is differentially dependent upon adhesive substrates 70. Specifically, DCs

cultured on albumin and serum coated surfaces maintained low levels of stimulatory

and co-stimulatory molecules and produce increased levels of IL-10. Conversely, DCs

cultured on collagen and vitronectin substrates expressed higher stimulatory and co-

stimulatory molecules and generated higher levels of IL-12p40 indicating a suppressive

and inflammatory DC phenotype respectively.

35

Technologies that target the immune system through the use of materials as

nano- and micro carriers have gained traction in recent years. Such biomaterials are

contributing to translation of basic immunology discoveries into therapies for transplant

rejection, autoimmune and infectious diseases, and cancer. They offer many

advantages over current clinical approaches including targeted delivery, controlled

release, and stability. Expanding the implementation of materials-based technologies in

clinical settings is expected to have broad impact.

Biomaterials-Based Immunomodulation of Dendritic Cells

The immune system provides a formidable barrier to the clinical application of

many biomaterials, and controlling interactions with components of the immune system

has recently become a major focus in the field5. The immune system is intricately

organized, composed of multiple layers of protection that work in unison for the defense

of the host against would-be invaders. Historically, biomaterials scientists have

developed materials that simply try to limit chronic aggravation of the immune system.

However, recent biomaterials approaches to actively engage and modulate immune

responses to achieve specific outcomes hold great promise. Key cellular players in

immunological defense and homeostasis, are dendritic cells (DCs), professional antigen

presenting cells that have a crucial role in dictating T cell-mediated immunity. Dendritic

cells naturally encounter, uptake, process and present antigen to naïve T cells, and

appropriately shape the resulting T and B cell responses. These features make DCs a

major target of strategies to manipulate the immune system. Pioneering cellular-based

therapies are ongoing using DCs, where DCs are generated from peripheral blood cells,

exposed to antigen and stimuli ex vivo, and then reintroduced into the patient’s

circulatory system. Successes from these cellular therapy approaches highlight the

36

importance of DC modulation. However, widespread application of DC cellular therapies

is limited for most treatments, as the costs are tremendous. Recent accomplishments

using implanted/injected biomaterials to direct DC function while remaining in situ, are

encouraging, and also address the high costs of cell-based therapies.

Biomaterial based methodologies of modulating DCs can be broadly categorized

as directing DC responses toward either inflammatory or suppressive phenotypes.

Numerous approaches have aimed to augment the inflammatory response of DCs for

their use in infectious diseases and cancer applications. Fewer, but a growing number

of approaches are pursuing suppressive DC phenotypes, which is of interest for

autoimmunity and transplant rejection therapies. Strategies currently employed to direct

DCs through the introduction of biomaterials into the body include the use of both

implanted scaffolds and injected particulates. The modulation of material properties

such as hydrophobicity, surface chemistry and degradation rate, as well as the

biomaterial based delivery of proteins, nucleic acids and small drug molecules, are

approaches actively being investigated for directing immune responses. Suitable

outcomes for strategies promoting inflammatory DCs are typically increased expression

of stimulatory76 and co-stimulatory molecules76, and release of pro-inflammatory

cytokines77. Conversely, desired outcomes of strategies promoting suppressive DCs

are often decreased expression of stimulatory and co-stimulatory molecules19, release

of regulatory cytokines77, and increased expression of inhibitory molecules and

tolerogenic mediators78. The ability to manipulate DC function via biomaterials is proving

to be enabling for numerous immune-related applications.

37

Here, we discuss tools of biomaterials-based DC immunomodulation currently

being developed, and suggest that they are broadly applicable to regenerative

medicine. Given that regenerative medicine applications often require careful balance

between inflammatory and suppressive immunological processes involved in wound

healing and regeneration, the field may substantially benefit from the tools and

principles uncovered from such biomaterial based immunomodulation approaches. To

begin, major principles of immunology will first be introduced, and relevant specifics of

DC biology briefly described.

Innate and Adaptive Immunity Overview

The immune system represents a major hurdle to successful incorporation of

many biomaterial. This complex network of cellular interactions and processes has

evolved as an organized and lethal defense mechanism against invaders, as well as

providing homeostatic regulation of self and non-self. Recognition of foreign entities

initiates a series of events that can lead to isolation and antigen-specific elimination in

the case of a pathogen, or tolerance, as in the case of food proteins. In vertebrates,

immunity can be divided into two main categories: the innate and the adaptive immune

system79.

Innate immunity is considered the first line of defense, consisting of anatomical

barriers such as the skin and mucosal membranes, soluble factors, and key cellular

players80. Members of this system include the complement cascade (made up of a large

number of distinct plasma proteins reacting with one another to opsonize pathogens

and induce a series of inflammatory responses), neutrophils, natural killer (NK) cells,

macrophages and DCs. If a pathogen has penetrated beyond physical skin and

mucosal barriers, then the cellular elements of innate immunity are typically engaged.

38

Innate cellular defense mechanism are triggered by signaling through pattern

recognition receptors (PRRs), which are encoded in the germline and can be secreted

or expressed on the host cellular surfaces. These PRRs can recognize and rapidly

respond to a wide variety of common molecular features known as pathogen associated

molecular patterns79,80, facilitating uptake and clearance of the foreign material. Upon

uptake and degradation of a pathogen, cellular machinery presents peptides from

degraded proteins presented on the cell surface bound to specialized stimulatory

molecules known as human leukocyte antigens (HLA) in humans, and major

histocompatibility complex (MHC) in mice79. This process is known as antigen

presentation, and can be carried out by a number of cells collectively known as antigen

presenting cells (APCs).

The adaptive arm of the immune system provides specific responses to newly

encountered antigens, and is mainly driven by the interactions between APCs and

lymphocytes81. Two major classes of lymphocytes exist, B and T cells, both originating

from a common progenitor in the bone marrow. These lymphocytes patrol the body,

routing through the bloodstream and lymphatic system to travel between peripheral and

lymphatic tissues. The basic functions of B cells include antigen presentation, antibody

production and immunological memory. B lymphocytes are equipped with antigen-

recognizing, membrane-bound immunoglobulins, termed B cell receptors that allow it to

interact with and uptake a vast array of antigens. When a naïve B cell encounters an

antigen with affinity for its B cell receptor, the B cell will then differentiate into either a

plasma or a memory B cell, which proliferate to generate a pool of these antigen-

specific cells to combat the invasion. This process is known as clonal selection and

39

expansion. Plasma cells produce soluble antibodies, which is the secreted form of the

B cell receptor. Antibodies that bind to surface antigens on pathogens will attract the

first components of the complement cascade leading to complement activation. Clonally

expanded memory B cells possess the same antigen-specific B cell receptor and will

persist in vivo for an extended period following the resolution of the infection. These

cells form the basis of an immunological memory response in the event of secondary

invasion by the same pathogen.

Much like B lymphocytes, T lymphocytes express a unique, membrane bound

protein termed the T cell receptor, which recognizes antigenic peptides that are bound

to stimulatory molecules on the surface of APCs. T cells also express associated co-

receptors that coordinate with the T cell receptor. Different types of these co-receptors

(e.g., CD4 or CD8) will correspond to the function of a T cell. Lymphocytes expressing

CD4 are generally categorized as helper T cells. The CD4 molecule recognizes

stimulatory molecules HLA-D/MHC II on the surface of a professional APCs. Main

subtypes of CD4+ helper T cells include Th1, Th2 and Th17. The Th1 T cell subset

supports pro-inflammatory cell-mediated immunity, which is intended to eliminate

intracellular pathogens and inducing B cell production of opsonizing IgG82. Th1 T cells

are characterized by the release of effector cytokines such as IFN-γ, TNF-α and IL-2.

Th2 T cells on the other hand, are associated with less inflammatory immune responses

and B cell production of IgG, IgA, and IgE82. More recently, the Th17 subset, was found

to abundantly produce the pro-inflammatory cytokine IL-1783 and is associated with

antimicrobial responses in epithelial and mucosal tissues. T lymphocytes expressing

CD8 receptors are generally categorized as cytotoxic T cells and recognize intracellular

40

antigen presented on stimulatory molecules HLA-A/B/C (human) or MHC I (mouse),

which are expressed by all nucleated cells including both professional APCs and non-

professional APCs. CD8+ T cells directly bind to infected cells and eliminate them by

releasing cytotoxins.

Professional APCs (DCs, macrophages, B cells) serve as the bridge between the

innate and adaptive immune system. These cells patrol the body and can capture

pathogens in their immature state via several uptake mechanisms. Once engulfed by an

APC, antigen is loaded into acidic vesicles where proteins are degraded into peptide

fragments, and presented in the context of the stimulatory HLA/MHC molecules. The

binding of a T cell receptor to antigen-loaded HLA/MHC molecules is commonly termed

signal 1 of the T cell-APC interaction. Once bound to a T cell receptor, APCs can also

provide signal 2, by presenting co-stimulatory molecules, which bind cognate T cell

receptors that activate the T cell and trigger clonal T cell expansion. Furthermore, APCs

can produce a signal 3, which refers to the cytokines released by APCs. Through these

mechanisms APCs shape T cell responses. Antigen presenting cells can also

sometimes engage in the secondary mechanism of cross presentation. During this

process, an extracellularly-supplied antigen is loaded on to HLA-A/B/C or MHC I

molecules (instead of the primary route via HLA-D/MHC II) and presented to CD8+ T

cells. The role of macrophages as APCs is primarily to stimulate immunity to previously

encountered antigens, and the role of B cells as APCs is primarily to engage helper T

cell support to cognate antigen. Dendritic cells, in contrast, are of particular interest due

to their unique ability to initiate and tune naïve lymphocyte responses to new antigens.

As such, DCs are a central regulator of immunity and have become a major target for

41

immunomodulation. In summary, Figure 1-2 depicts a simplified schematic of the typical

immune response to a foreign pathogen.

The successful application of immunological principles has been most widely

implemented through the use of vaccines. Vaccination engages innate and adaptive

immunity and has been able to manipulate immunological responses for the treatment

of numerous pathologies, particularly infectious diseases, and more recently cancer.

Vaccines have been primarily successful in eliciting humoral (antibody-mediated)

responses, but have had limited ability to induce cellular immunity (which provides

clearance of intracellular pathogens in infected cells). Incorporation of biomaterials tools

into vaccine strategies holds potential to address this and other limitations, and will be

discussed further.

Dendritic Cell Subsets

Two main subsets of dendritic cells are plasmacytoid dendritic cells (pDCs) and

conventional DCs. Both cell types express low levels of stimulatory HLA-D/MHC II and

co-stimulatory molecules (e.g., CD40, CD80 and CD86) at homeostatic resting state,

which are upregulated upon activation, and are distinguished by high expression of the

integrin receptor αXβ2 (the alpha subunit of which is also known as CD11c).

Plasmacytoid dendritic cells arise from lymphoid committed precursors and constitute a

small subset of DCs mainly found in the blood and lymphoid tissue, and enter the lymph

nodes through peripheral blood circulation79. Upon activation, pDCs are characterized

by the copious release of type 1 interferon (IFN-α and IFN-β), particularly in response to

viral infections. Plasmacytoid DCs, however, do not play a crucial role in the activation

of naïve T lymphocytes, which is in contrast to conventional DCs. Conventional DCs

originate from myeloid precursors and form a small subset of hematopoietic cells that

42

populate most lymphatic and non-lymphatic tissue. Conventional DCs are the most

potent antigen presenting cells of the immune system as they have an enhanced ability

to sense injury, capture antigen, process and present it to T lymphocytes. Conventional

DCs can also further differentiate into phenotypes that are either inflammatory or

tolerogenic84 (Figure 1-3). Another major type of DC, the Langerhans cell, resides in the

skin and mucosa, and unlike conventional DCs, during homeostasis they originate from

a unique pool of skin-localized precursors85. While these (and more) distinct DC

subsets have been identified, the field of biomaterials based immune modulation has

primarily focused on conventional DCs due to their ability to initiate de novo antigen

response and their relative abundance.

Inflammatory dendritic cell phenotype

In the immature state, DCs are constantly monitoring their microenvironment,

sampling, processing and presenting antigens, and ready to respond to signals of

danger. They capture antigens through several mechanisms including pinocytosis79 (an

actin-driven mechanism in which the plasma membrane ruffles to form a vesicle

incorporating foreign material), receptor mediated endocytosis79 (either via C-type lectin

receptors, or CD64 and CD32), and phagocytosis79 (internalization of larger particulates

usually aided by specific membrane receptors). Once immunogenic antigen is captured

in conjunction with activation signals (e.g., from PRRs), DCs undergo phenotypical

changes rendering their mature state. These changes are characterized by morphology

alteration (e.g., loss of cell surface adhesion molecules, cytoskeleton reorganization and

increased cellular motility79), increased expression of HLA/MHC stimulatory molecules,

upregulation of co-stimulatory molecules (such as CD40, CD80 and CD86), and release

of pro-inflammatory cytokines (e.g., IL-12). Dendritic cell maturation is closely linked

43

with migration from peripheral tissue to the secondary lymphoid organs (spleen, lymph

nodes, tonsils, appendix and Peyer’s patches) where they interact with T cells and

influence their function.

Suppressive dendritic cell phenotype

In the periphery, DCs encounter self-derived antigens and present them to T

cells as a normal process of homeostasis. Rarely, this presentation results in the

pathological activation of the adaptive immune system and can result in autoimmunity

against self-antigen. Typically, the normal homeostatic reaction involves DC initiation of

suppressive immune networks that can lead to T cell anergy, deletion, or expansion of a

regulatory phenotype. For instance, transforming growth factor-β1 (TGF-β1) and/or

interleukin-10 (IL-10) secreted by suppressive DCs can induce regulatory T cells

(Tregs). These Tregs suppress inflammatory immune responses, and are characterized

by the expression of surface receptors CD4 and CD25, and the transcription factor,

forkhead box P3 (Foxp3). Subset of Tregs can also secrete IL-10 and have suppressive

roles when in contact with effector T cells. Generally, characteristics of tolerance-

promoting DCs have been reported to include low expression of stimulatory and co-

stimulatory molecules, low production of inflammatory cytokines (e.g., IL-12), increase

production of regulatory cytokines (e.g., IL-10, TGF-β1), high levels of inhibitory

molecules (e.g., programmed death ligand 1; PD-L1), as well as high expression of

tolerogenic mediators such as indoleamine 2,3-dioxygenase (IDO)84,86. There is

tremendous potential in targeting DCs for therapeutic applications across the spectrum

of immune diseases.

44

Biomaterials-based Immunomodulation

Numerous synthetic polymers, have been investigated for their use as

biomaterials in regenerative medicine applications, including polyesters (e.g., poly(lactic

acid), poly(glycolic acid) and their copolymers), polyorthoesters, polyanhydrides and

polycarbonates. Of these, polymers fabricated using biodegradable copolymer

poly(lactic co-glycolic) acid (PLGA) have been the most researched for the delivery and

controlled release of immunotherapeutics, and their interaction with DCs well

characterized10. By altering the composition of the PLGA copolymer, microparticles can

be fabricated to provide an initial burst of encapsulated immunomodulatory molecule

followed by sustained release. This feature has notably been identified as enabling the

development of therapeutics which can administer both prime and boost doses in a

single injection19. This section details efforts in the manipulation of immune responses

through the use of polymeric systems as controlled release carriers of

immunomodulatory molecules, as well as the potential for biomaterials to provide direct

interactions with DCs, for example, through proteins adsorbed onto the biomaterial

surface. A conceptual summary of biomaterials-based immunomodulation of DCs is

illustrated in Figure 1-4.

Biomaterials-based DC Manipulation Toward an Inflammatory Phenotype

The quintessential scenario in which an inflammatory DC phenotype would be

desired is vaccine applications. Vaccines are generally composed of an antigen,

adjuvant, and a delivery system (e.g., a liquid solvent able to be injected). Adjuvants

can be materials or molecules which stimulate innate immunity by activating APCs. This

engagement of the innate branch of immunity results in the development of long-lasting,

antigen-specific immune responses toward the co-delivered antigen87. The only FDA

45

approved agents for use as adjuvants in the US, interestingly, are materials consisting

of inorganic particulates of calcium phosphate, aluminum hydroxide and aluminum

phosphate, which are collectively referred to as alum87. Alum causes aggregation of

antigen onto the particulates, providing an antigen depot, and eliciting a strong

inflammatory response87. Although these materials have been used for decades, their

mechanism of action is still being uncovered. Recent reports have shown that alum

engages the NALP (NACHT, LRR and pyrin domain-containing protein) -3

inflammasome, resulting in the production of pro-inflammatory cytokines IL-1 and IL-

1888. However, alum has limitations, in that it is not optimal for different types of

antigens, and is ineffective at inducing cellular immunity. This motivates development of

new adjuvant technologies where biomaterials can be implemented as both antigen

carriers, and serve as adjuvants or adjuvant carriers.

Biomaterials as antigen carriers

The use of biomaterials is an attractive approach as they can be formulated to

provide an antigen depot with long term protection, persistence, and controlled release

of antigens, potentially eliminating the need for booster doses, while increasing the

likelihood for antigen uptake and processing by APCs. In contrast to alum, synthetic

biodegradable polymers can be used to provide a depot for more antigens than just

proteins, including, for example, small antigenic peptides. Antigens can be loaded into,

or conjugated onto biomaterial particles or scaffolds, protecting the cargo from

enzymatic degradation. For example, studies conducted by Ali et. al, sought to create

an inflammatory microenvironment through the use of implanted porous PLGA scaffolds

as a cancer vaccine89. This approach aimed at not only using biomaterials as a delivery

vehicle but also as a structural support in which DCs are attracted through release of a

46

recruiting molecule, granulocyte macrophage colony stimulating factor (GM-CSF), and

in which the DCs can reside while receiving co-encapsulated antigen (tumor lysate) and

activation signal (CpG). The investigators reported that after 14 days in vivo, the cellular

infiltration of the scaffolds containing GM-CSF was significantly higher when compared

to the matrix containing no recruiting agent. Not only were the cells primed in a

controlled microenvironment, activated DCs were also able to leave the PLGA matrix

and travel to draining lymph nodes for further interaction with T cells89. Notably, these

immune modulating materials were able to develop specific and protective anti-tumor

immunity reporting a 90% survival rate in melanoma mouse models.

Biomaterials in particulate form can promote targeted delivery of antigen to DCs

and intracellular DC compartments, facilitating specific processing and presentation on

either MHC I or MHC II complexes. For example, Lewis et. al. demonstrated that DC

uptake was markedly increased when PLGA microparticles were surface conjugated

with either anti-DEC-205 antibody, anti-CD11c antibody or the CD11c-binding peptide,

P-D272. Notably, the PD-2 peptide conjugation greatly improved DC MHC II-antigen

presentation compared to controls, by prolonging presentation to a 4 day time span, in

vitro. It was reported that none of the modified or unmodified PLGA particles (50:50

composition) activated DCs(activating factors were not included). The P-D2 peptide

surface modification also greatly promoted in vivo DC uptake and trafficking of

microparticles to lymph nodes72. In contrast to delivering exogenous antigen for MHC II

loading, promotion of cross-presentation, the exogenous delivery of antigen to induce

immunity through the MHC I pathway, is known to be challenging. This is due to the fact

that internalized particulates are quickly trafficked to acidic endosomes where they are

47

enzymatically degraded. However, several groups have made the attempt, with varying

degrees of success. For example, Sneh-Edri et. al, investigated PLGA nanoparticles

decorated with peptides with an endoplasmic reticulum localizing motif and loaded with

antigenic materials. Although enhanced localization to the endoplasmic reticulum was

not achieved, the particle modifications did increase endocytosis, and influenced

intracellular trafficking, which provided low levels of cross-presentation of the antigenic

peptide, for a modestly prolonged time of 2 day, in vitro90. More successful strategies

have overcome the challenge of inducing cross-presentation through the development

of pH-responsive polymers91 that disrupt endosomal membranes, thereby delivering

antigen to the cytoplasm (where processing occurs for MHC I loading). For example,

Flanary et. al, investigated the use of poly(propylacrylic acid; PPPA) as a protein

vaccine carrier, and reported enhanced delivery of ovalbumin (as a model antigen) into

the MHC I pathway92. In the ionized state PPAA maintains aqueous solubility, but when

the environment becomes acidic, as is the case of endosomes, the pendant carboxyl

groups become protonated and the polymer transitions to an insoluble state that is

membrane destabilizing, delivering cargo to the cytosol. This cytosolic delivery resulted

in an increased MHC I presentation and antigen specific CD8+ lymphocyte activation,

promoting cellular immunity92. These studies illustrate the use of biomaterials for the

delivery of antigen to cellular and subcellular compartments.

Biomaterials as adjuvants and adjuvant carriers

In addition to being able to provide an antigen depot, biomaterials can also serve

adjuvant functions by either delivering adjuvant molecules93, or by providing direct

activation through cell-material interactions94. Several experimental adjuvant molecules,

such as unmethylated bacterial CpG DNA (CpG), polyinosinic:polycytidylic acid

48

(PolyI:C), lipopolysaccharide (LPS), and monophosphoryl lipid A (MPLA) are known to

activate APCs directly. The mechanism involves the binding of toll-like receptors on

DCs, resulting in the upreguation of MHC II, co-stimulatory molecules CD80, CD86 and

CD40 and secretion of pro-inflammatory cytokines such as IL-6. Studies by Schlosser

et. al showed that CpG and PolyI:C encapsulated along with ovalbumin (model antigen)

in a PLGA microparticle stimulated a potent cytotoxic T lymphocyte response. A single

immunization was provided and significant levels of antigen-specific tetramer

(SIINFEKL/H-2K(b)) -positive cytotoxic T lymphocytes were detected, as was production

of IFN-α, efficient cytolysis, and protection from vaccinia virus infection93. Similarly,

Beaudette et. al, reported that the co-encapsulation of ovalbumin and CpG resulted in

higher cytotoxic T lymphocyte activity when compared to particles co-administered with

the CpG adjuvant in the free form95. In another example, Elamanchili et. al,

demonstrated that DCs pulsed with MPLA-encapsulating PLGA nanoparticles showed

upregulation of MHC II and CD86 molecules resulting in the activation of naïve T

lymphocytes96. Extending upon their previous work89 (described above), Mooney and

colleagues further experimented with CpG and tumor lysate loaded PLGA scaffold

materials, implanted as a cancer vaccine, this time comparing co-encapsulation with

different recruiting factors GM-CSF, Flt3L, or CCL2097. The authors demonstrated a

significant increase in recruited and activated DC numbers (with high levels of MHC II

and CD86) infiltrated into the scaffold, leading to potent anti-tumor T cell responses.

These vaccine formulations resulted in long-term survival in melanoma mice models97.

Combination approaches such as the co-delivery of adjuvants, particularly of PRR-

activating molecules, along with the immune modulating nucleotides such as silencing

49

RNA (siRNA) are promising98,99. Specifically, studies conducted by Pradhan et. al,

conjugated IL-10 siRNA and CpG onto the surface of a PLGA microparticle loaded with

pDNA antigen. Their studies demonstrated uptake, high expression of stimulatory and

co-stimulatory molecules (CD80, CD86 and MHC II), low expression of inhibitory

cytokines (IL-10) and high expression of pro-inflammatory cytokines (IL-12 and TNF-α)

by DCs treated with microparticles when compared to its soluble formulation. This DC

profile shifted the T cell activation to a Th1 subset which resulted in increased survival

of animal models of lymphoma99.

In contrast to approaches where biomaterials serve as an adjuvant carrier,

another strategy is designing materials that can directly act as adjuvants themselves.

Studies conducted by Hudalla et. al, in the Collier group developed peptides bearing p-

nitrophenyl phosphonate ligands that self-assembled into nanofibers displaying properly

folded, biologically active protein antigens100. These nanofibers were able to

successfully present controlled amount of a model protein antigen (green fluorescent

protein) and acted as a self-adjuvanting vaccine material, in mice100. Additionally,

Narasimhan and colleagues demonstrated the ability of a family of polyanhydride

nanoparticles to differentially act as self-adjuvants, depending on the polymer

composition, inducing potent immunomodulatory activity101-103. Along these lines, it is

has been observed that various PLGA compositions influence DC activation differently.

Studies conducted by Fischer et. al have shown microparticles with a 50:50 ratio of

lactide to glycolide maintain DC immaturity104 while the more hydrophobic ratio of 75:25

has been reported to induce DC activation94. Guiding principles for materials chemistry-

based immune modulation are still being explored, but surface immobilized adhesive

50

proteins may play a role by directing DC adhesion71,73,105. Additionally, like alum and

other particulate materials, PLGA microparticles are capable of engaging the NALP3

inflammasome88.Altogether, this literature supports the concept that biomaterials have

tremendous potential to improve current adjuvant technologies by either direct activation

of APCs, or encapsulation of stimulating agents.

Biomaterials-based DC Manipulation Toward a Suppressive Phenotype

So far, discussion has centered on the modulation of DCs to initiate an

inflammatory immunological response, and this has been the largest focus in the field.

However, biomaterials immune modulation of DCs for suppressive applications is

gaining momentum, and hinges on the fact that DCs can also support suppressive

pathways that maintain homeostasis and dictate key regulators of peripheral tolerance.

It has been reported that suppressive DCs can elicit T cell anergy, expansion of

regulatory T cells and repression of B cell differentiation and expansion106. T cell anergy

is a tolerogenic mechanism in which lymphocytes become hypo-responsive following

the incomplete delivery of co-stimulatory signals107. In addition to inducing T cell anergy,

DCs can also expand the formation of CD4+CD25+FoxP3+ regulatory T cells. This Treg

subset can directly halt effector T cells, and by inhibiting antigen presentation from

DCs108. Therefore, guidance of DC phenotype toward a suppressive or tolerogenic state

holds great promise, particularly for the mitigation of autoimmune diseases and

transplant rejection. While the list of works to date is shorter compared to inflammatory

applications, biomaterial approaches also offer opportunities for targeting, controlled

release and payload protection to deliver modulatory molecules and nucleotides for

suppressive applications. These biomaterial strategies can thereby overcome barriers of

51

limited bioavailability that often accompanies administration of these types of

therapeutic molecules.

Numerous pharmacological methods have been reported to inhibit DC

maturation, including the use of anti-inflammatory (e.g., corticosteroids109, salicylates110)

or immunosuppressive (e.g., mycophenolate mofetil111, 1α,25-dihydroxyvitamin D3112,113)

agents, as well as certain cytokines (transforming growth factor β [TGF-β]114, IL-

10115).The application of pharmacological agents is often limited when administered

systemically in soluble form. Targeted delivery of these agents to specific cells in vivo

can be critical, given the pleiotropic nature of many of them possess, with potential off

target effects and toxicity on bystander cells and tissues. This limitation provides

another window of opportunity for the use of biomaterials to provide targeted delivery

and controlled release of these types of factors. For example, the systemic

administration of the widely used immunosuppressant rapamycin, in vivo, decreases DC

surface expression of CD40, CD80, CD86 and MHC II molecules116. Taking advantage

of this, Bryant et. al, investigated the use of PLGA particles for the delivery of donor

antigens to induce transplant tolerance in allogeneic islet transplantation model117.

Infusion of the particles mediated tolerance in approximately 20% of recipients, and

combination with a low dose rapamycin at the time of transplant significantly improved

tolerance to approximately 60% efficacy. This method offered a biomaterials-based

approach to inducing long term donor graft-specific tolerance117. However, long term

systemic administration of rapamycin and other such immunosuppressants at normal

therapeutic levels results in systemic immunosuppression, by affecting off-target

lymphocytes118. This has motivated the use rapamycin encapsulating particles. Work by

52

the Little group examined the effects of rapamycin loaded PLGA microparticles on

DCs113 This study demonstrated low expression of stimulatory and co-stimulatory

molecules (MHC II, CD86, CD40) and decreased T cell activation113. Additionally,

Maldonado et. al, demonstrated that biodegradable PLGA nanoparticles carrying either

protein or peptide antigen, co-formulated with rapamycin induced a durable antigen-

specific tolerogenic effect119. Specifically, treated animals showed reduced allergic

hypersensitivity disorders, protection from disease relapse in a multiple sclerosis model,

and prevention of antidrug antibodies responses in a hemophilia A model. Treatment

with these nanoparticles resulted in the inhibition of CD4+ and CD8+ T cell activation, an

increase in regulatory T cells and durable B cell tolerance resistant to multiple

immunogenic challenges with protein or peptide antigen. Importantly, the authors

demonstrated that only the encapsulated rapamycin, and not the soluble form could

induce immunological tolerance at the doses used119. Altogether, these works show

how immunomodulation can be improved through the use of biomaterials encapsulating

small drug molecules.

Dendritic cell adhesive interactions with biomaterials and biomaterials-adsorbed

proteins have also been investigated, and these interactions have been reported to be

able differentially promote suppressive DC phenotypes71,120. For example, studies

performed by Acharya et. al, indicate that DC morphology and production of cytokines is

differentially dependent upon adhesive substrates70. Specifically, DCs cultured on

albumin and serum-coated tissue cultured-treated substrates maintained low levels of

CD80, CD86 and MHC II, and produced increased levels of IL-1070. Using a different

approach, Leclerc et. al, demonstrated that DCs can receive biological cues from

53

tethered signaling molecules121. The authors reported covalently immobilized GM-CSF

onto planar silane and polystyrene surfaces resulted in increased DC differentiation in

response to GM-CSF tethering121.

Delivery of immune modulating nucleotides also has the potential to modulate

DC responses and further dictate the outcome of an immunological response.

Biomaterials can serve as delivery vehicles for silencing RNA99, gene vectors122,

messenger RNA123, and antisense oligonucleotides124. Biomaterial carriers can provide

a vehicle to protect nucleotides from systemic elimination, and provide intact delivery to

subcellular compartments125. For example, Giannoukakis and colleagues demonstrated

that non-obese diabetic mouse (NOD; a well-established animal model for type 1

diabetes)-derived DCs treated ex vivo with antisense oligonucleotides resulted in

diminished expression of costimulatory molecules (CD40, CD80 and CD86) and a

significant delay in the incidence of autoimmune diabetes in NOD mice124. Furthermore,

phase I safety clinical trials concluded positively, where antisense-treated autologous

DCs were manipulated ex vivo and administered as a cellular therapy to established

type 1 diabetes adult patients126. Nevertheless, the limiting high cost of cellular

therapies has motivated the group to pursue a biomaterials microparticle-based

approach. Along this line, encapsulation of antisense oligonucleotides in a polymeric

microparticle was able to effect gene knockdown in DCs127. The subcutaneously

injected microparticles were trafficked to pancreatic draining lymph nodes, augmented

Tregs and provoked hypo-responsiveness to β-cell antigen without compromising global

immunity127. The studies presented in this section highlight the delivery of small

molecules and nucleotides through the use of biomaterials as well as surface

54

interactions specifically targeting DCs for suppressive applications, and are directly

relevant to regenerative medicine.

Biomaterials can be engineered to fulfill many functions in the context of

regenerative medicine. They have long been studied to perform, augment or replace a

natural function, historically attempting to avoid provoking the immune system.

However, more recently biomaterials have gained much interest as a tool with which to

actively engage the immune system, particularly through the targeting of DCs. Dendritic

cells play a crucial role in dictating the fate of an immune response and maintaining

homeostasis and have therefore become the focus of numerous experimental

approaches. This chapter has outlined key concepts for the understanding of immune

responses, described relevant DC subsets and functions, and detailed illustrative

biomaterials-based technologies directing DC phenotype toward either an inflammatory

or a suppressive role. Techniques being developed include the use of biomaterials as

antigen carriers, biomaterials as adjuvants and adjuvant carriers, the delivery of small

molecules and nucleotides, and adhesive interactions with biomaterials. The field of

biomaterials-driven immunotherapies will continue to grow as critical mechanisms are

uncovered and more advanced material designs emerge. Notably, the concepts

encompassing the field of regenerative medicine have grown over the years to include

fields such as tissue engineering and wound healing. Looking forward, through the use

of new tools such as biomaterials-based immunomodulation of dendritic cells, it is

conceivable that the range of regenerative medicine will be further expanded to include

related disciplines such as cancer, transplant rejection, autoimmune diseases, and

infectious diseases.

55

Figure 1-1. Interaction of nano- and micro-scale biomaterial carriers with key members

of the innate and adaptive immune system. Polymeric, lipid, metallic/inorganic and biologic materials have been useful as carriers of bioactive molecules or direct immunomodulators to induce either stimulation (e.g., vaccine or cancer applications) or suppression (e.g. transplant tolerance and autoimmunity)

56

Figure 1-2. Simplified immune response to foreign pathogen. 1) Pathogen encounters

epithelial barrier which serves as first line of defense. 2) If the barrier is broken, the innate immune system will attempt to clear it by attacks mediated largely by macrophages, neutrophils and NK cells. 3) To initiate adaptive immune system, dendritic cells (DCs) encounter antigen, undergo maturation during the antigen processing and present it on their surface for recognition by T cells. 4) CD8+ T cells then undergo clonal expansion and become antigen specific cytotoxic T cells. 5) CD4+ T cells also undergo clonal expansion and interact with B cells leading to, 6) the production of pathogen specific antibodies.

57

Figure 1-3. Dendritic cell phenotype dictates T cell function. Immature dendritic cells can

be derived from monocytes in the presence of IL-4 and GM-CSF. Dendritic cells can then become tolerogenic or inflammatory depending on the environmental stimulus, and subsequently direct T cell polarization. Inflammatory danger signals and pathogen associated molecular patterns (PAMPs) can activate iDCs into classically mature (inflammatory) DCs which express high levels of MHC class II and co-stimulatory molecules, release pro-inflammatory cytokines, migrate to lymph nodes and stimulate naïve T cells to differentiate into Th1, Th2 or Th17 effector T cells. Suppressive DCs can be induced in response to signals such as apoptotic cells and TGF-β1. Suppressive DCs can induce antigen-specific T cell apoptosis, anergy and Treg expansion.

58

Figure 1-4 Overview of biomaterials-based dendritic cell modulation. Common

strategies can be broadly divided into those employing biomaterials as antigen carriers, for targeted delivery of small molecules and nucleotides, and controlled release of adjuvants. Biomaterials can furthermore directly function as an adjuvant, possibly through dendritic cell (DC) interactions with surface-adsorbed proteins.

59

CHAPTER 2 EXTRACELLULAR INDOLEAMINE 2,3 DIOXYGENASE-TREATED DENDRITIC CELLS MAINTAIN AN IMMATURE PHENOTYPE AND SUPPRESS ANTIGEN-

SPECIFIC T CELL PROLIFERATION

Background

Indoleamine 2,3 Dioxygenase

Tryptophan catabolism and the kynurenine pathway

Indoleamine 2,3-dioxygenase (IDO) is a non-secreted heme containing enzyme

responsible for the catalysis of the least-abundant amino acid, tryptophan, along the

kynurenine pathway128. IDO catalyzes the conversation of L-tryptophan into N-formyl

kynurenine, the rate limiting step, by oxidative cleavage of the indole ring present in

tryptophan. N-formyl-kynurenine is further converted into formic acid and the stable end-

product kynurenine either spontaneously under acidotic conditions or enzymatically via

formidase129,130. Depending on the cell type and expression level of IDO and other

kynurenine pathway enzymes, a cascade of reactions occurs yielding various

biologically active metabolites collectively known as kynurenines including kynunrenine,

formic acid, anthranilic acid, kynurenic acid, xanthurenic acid, 3-hydroxykynurenine, 3-

hydroxyanthralinic acid, quinolinic acid and picolinic acid (Figure 2-1). Approximately

95% of free tryptophan is metabolized through the kynurenine pathway with the

remaining 4% consumed through the methoxyindole pathway for conversion into

serotonin and melatonin, and the remainder used for protein synthesis. Intracellular

expression of IDO can act as a sink for extracellular tryptophan resulting in a reduction

of local levels of the amino acid. Early studies suggest tryptophan is transported into

IDO-expressing cells from the extracellular space via system L amino acid transporters

(LATs) which have affinity for l-tryptophan and other larger neutral amino acids131. Other

60

studies have demonstrated uptake of tryptophan can also be enhanced by stimulation

with interferon gamma (IFNγ)132-135, lipopolysaccharide (LPS)136 and aryl hydrocarbon

receptor (AhR) agonists, which include kynurenine128,130,137,138. Metabolites generated

during catabolism of tryptophan can be released into the extracellular environment for

uptake by nearby cells via LATs or binding to the AhR. This represents a positive

feedback loop for cell signaling which can influence the function and phenotype of

surrounding cells that do not necessarily express IDO themselves.

Tissue and cellular expression

Indoleamine 2,3-dioxygenase is expressed in a wide range of tissues and cell

types either constitutively or upon stimulation. IDO expression under normal

physiological conditions in mice occurs primarily in mucosal tissues including the

placenta, epidermis, prostate, adipose tissue, in lymphoid organs such as the thymus,

spleen, the central nervous system, kidney, pancreas, lungs and regions of the eye.

Expression of IDO in the small and large intestines, as well as lymph nodes is

dependent on IFNγ, and is thought to play a role in the homeostasis of mucosal

immunity in response to the local microbiota128

In many cell types, IDO is not expressed under normal physiological conditions

but instead requires inflammatory stimuli. IDO expression is induced in antigen

presenting cells, such as monocyte-derived macrophages and dendritic cell subsets,

neutrophils and eosinophils, and non-haematopoietic including endothelial cells,

epithelial cells, fibroblasts and smooth muscle cells. IDO expression has been shown to

be induced by several cytokines such as interlukin 1 (IL-1), tumor necrosis factor alpha

(TNFα), IFNγ and cytotoxic T-lymphocyte associated protein 4 (CTLA-4)132,139,140

61

Role in immune regulation

Early studies suggested the main role of IDO was to halt microbial growth by

limiting their supply of tryptophan. Although some microorganisms have evolved to

synthesize tryptophan de novo, it is energetically unfavorable128 and therefore many

microbes take up tryptophan from the host microenvironment. This requirement for

external sources also makes them susceptible to its depletion, particularly those that are

closely reliant on the host cell for replication and survival. More recent literature,

however, suggests that IDO also participates in modulation of T cell responses,

particularly toward a tolerogenic lineage 130,135,141-144 by initiation of apoptosis, induction

of T cell anergy and limitations on the activity of effector T cells (Teff) by regulatory T

cells (Tregs) 135,143-147. IDO was first established in 1998 by Munn and Mellor to be

present at the maternal/fetal interface of pregnant mice. By inhibiting IDO activity

through the administration of 1-methyl tryptophan (MT), an IDO inhibitor, the authors

demonstrated IDO prevents T-cell mediated destruction of the allogeneic fetus148. Since

then, efforts to elucidate the mechanism of IDO-mediated T cell suppression have

focused on antigen presenting cells, mainly DCs and macrophages.

The immunomodulatory action of IDO involves various biochemical and

metabolic processes affecting an array of immune cells, but the focus remains primarily

on T cells. Two proposed mechanisms for IDO-mediated tolerance have emerged: (i)

depletion of tryptophan suppresses T cell proliferation by activating the general control

nonderepressible 2 (GCN2) stress-response kinase which functions to control

transcriptional and translational processes that couple cell growth to amino acid

availability 149-151 and (ii) downstream metabolites (collectively referred to as

kynurenines) directly interact with immune cells through the aryl hydrocarbon receptor

62

(AhR) 137,138 144 or inhibit IL-2 signaling, crucial T cell survival 152. Dendritic cells

expressing IDO can dictate T cell differentiation into a regulatory phenotype, thus

indirectly suppressing effector T cells. Thus, much of the literature describing IDO as an

tolerance-inducing immunomodulator has focused on overexpression of the gene in

transplant cells/tissues, shown to prolong graft survival in a number of models 145,153-156,

or inhibition of its enzymatic activity for the treatment of cancer 157-160.

The effects of IDO-expressing cells have been well characterized and

documented 86,130,143,148,161, however, little is known about any direct effect on DCs or

the effect of extracellular enzyme activity. In this study, we demonstrate that murine

DCs treated with exogenous human recombinant IDO maintain an immature phenotype

and provide robust suppression of antigen-specific T cell proliferation in vitro. The

mechanism of this suppression involves tryptophan depletion as well as kynurenine

accumulation. These results establish that IDO maintains its immunomodulatory

capacity in the extracellular environment and that extracellular IDO acts directly upon

DCs.

Materials and Methods

IDO Characteristics and Activity Assay

Recombinant human IDO (IDO) expressed in Escherichia coli was purchased

from R&D Systems (Minneapolis, MN) with a predicted molecular mass of 46 kDa,

purity >95% by SDS-PAGE, endotoxin levels <1 EU/µg of protein by the LAL method

and a specific activity of >500pmoles/min/µg as measured by its ability to oxidize L-

tryptophan to N-formyl-kynurenine. The specific activity was measured before

experiments to ensure maximal effect at the beginning of the assay following an

adapted procedure from Valladares et al. 162. The reaction substrate contained 200 µM

63

tryptophan, 20 mM ascorbic acid, 10 µM methylene blue, 225 U catalase and 50 mM

MES buffer (pH 6.5). Recombinant human IDO at 16 ng/mL was loaded onto a flat

bottom 96-well plate and the reaction started by mixture in 1:1 ratio with reaction

substrate. Absorbance was measured in kinetic mode for 5 minutes at 321 nm.

Dendritic Cell Culture and Extracellular Enzyme Treatment

Dendritic cells were generated by isolating the bone marrow from femurs and

tibias of 8-12-week-old C57BL6/j female mice euthanized by CO2 asphyxiation followed

by cervical dislocation in accordance with approved protocols by the University of

Florida. The marrow cells were obtained by flushing the shaft of the bones with a

25G5/8 needle using RPMI 1640 (Corning, Corning, NY) containing 10% fetal bovine

serum (Lonza, Walkersville, MD) and 1% penicillin-streptomycin (Hyclone, Logan, UT)

and mixed homogenously. The cell suspension was strained using a 70 µm cell strainer

(Becton, Dickinson, NJ, USA) and collected after centrifugation at 1600 rpm for 5

minutes at 4°C. The red blood cells were then removed by lysing with ACK Lysing buffer

(Lonza, Walkersville, MD) followed by centrifugation at 1600 rpm for 5 minutes at 4°C to

recover leukocyte progenitors. Remaining cells were re-suspended in DC media:

DMEM/F-12 with L-glutamine (Cellgro, Herndon, VA) containing 10% fetal bovine

serum, 1% sodium pyruvate (Lonza, Walkersville, MD), 1% non-essential amino acids

(Lonza, Walkersville, MD), 1% penicillin-streptomycin, 20 ng/ml of GM-CSF (R&D

Systems, MN, USA) and were plated on tissue culture flasks for 2 d in order to remove

adherent cells. After 2 d, floating cells were transferred to low attachment tissue culture

plates with fresh DC media for the expansion of DC precursors. Half media change was

performed on day 4 of isolation. On day 6 floating cells were carefully removed and

plated on to tissue culture plates at an appropriate cell density for the adhesion and

64

proliferation of DCs. On day 8 media was removed, cells washed with PBS and fresh

media provided. On day 10 dendritic cells were either treated with 15 µg/mL of human

IDO (R&D Systems, Minneapolis, MN) cultured in tryptophan free media supplemented

with 500 µM L-kynurenine (Sigma-Aldrich, St. Louis, MO) for 24 h at 37°C.

Dendritic Cell Phenotype and Maturation Resistance

Following IDO incubation, DCs were challenged with 1 µg/mL of

lipopolysaccharide (LPS) (Sigma-Aldrich, St. Louis, MO). DCs were then imaged to

determine physical characteristics using a Zeiss Axiovert 200M microscope and phase

contrast. DC viability was determined through microscopy using LIVE/DEAD imaging kit

(ThermoFisher Scientific, Waltham, VA) and confirmed via flow cytometry using

LIVE/DEAD Fixable Near-IR Dead Cell Stain Kit (ThermoFisher Scientific, Waltham,

VA) both techniques were used according to manufacturer’s instructions. DC maturation

was evaluated by the surface expression of stimulatory and co-stimulatory markers

(MHC II, CD80 and CD86) as well as cytokine release (IL-10 and IL-12p70) by flow

cytometry and enzyme-linked immunosorbent assay (ELISA), respectively. Following

enzyme incubation and LPS challenge, supernatant was collected and stored at -20°C

for subsequent analysis using BD OptEIA ELISA kits (BD Biosciences, Franklin Lakes,

NJ) following manufacturer’s instructions. Adherent cells were incubated with 5 mM

Na2EDTA in PBS solution at 37°C for 30 minutes and lifted using a cell scrapper. Cells

were then washed with 1% fetal bovine serum in PBS and incubated with LIVE/DEAD

Fixable Near-IR Dead Cell Stain kit for 10 mins at room temperature followed by

washing and incubation with antibodies (BD Bioscience, San Jose, CA) against

CD16/32 (Fcγ III/II receptor) (clone 2.4G2) for 30 mins on ice to block Fcγ receptors on

65

DCs. Cells were washed and stained with antibodies against CD11c (clone HL3) CD80

(clone 16-10A1), CD86 (clone GL1) and MHC II (clone M5/114.15.2).

T Cell Isolation and Proliferation Assay

T cells were isolated from splenocytes of OT-I and OT-II female mice (Jackson

Laboratories, Bar Harbor, ME) 8-12 weeks of age. The OT-I mouse model carries a

transgene insert for rearranged TCR α and β genes on CD8+ T cells, that assemble

allow recognition of ovalbumin peptide residue 257-264 in the context of H2kb. The OT-

II mouse model carries a transgene insert for rearranged TCR α and β genes on CD4+ T

cells, that are specific for ovalbumin 323-339 in the context of I-Ab. Animals were

euthanized by CO2 asphyxiation followed by cervical dislocation as a secondary method

in accordance with guidelines approved by the University of Florida. Spleens were

excised and homogenized with RPMI 1640 medium (MP Biomedicals, OH, USA)

containing 10% fetal bovine serum (Lonza, Walkersville, MD) and 1% penicillin-

streptomycin (Hyclone, Logan, UT). The cell suspension was then filtered through a 70

µm cell strainer and centrifuged at 1600 rpm for 5 minutes at 4°C. Red blood cells were

lysed with ACK Lysing buffer (Lonza, Walkersville, MD) followed by centrifugation to

recover lymphocytes. CD4+ and CD8+ T cells were purified using a negative selection

isolation kit (MACS Miltenyi Biotec, Bergisch Gladbach, Germany) following the

manufacturer’s instructions. Resulting T cells were then labeled with 10 µM

carboxyfluorescein succinimidyl ester (CFSE) to track proliferation. For antigen specific

proliferation, DCs were treated with extracellular IDO either in the presence or absence

of 1 mM 1-methyl-tryptophan (MT) for 24 h then pulsed with 1 µg/mL ovalbumin peptide,

323-339 for CD4+ T cell assays and 257-264 for CD8+ assays, for 3 h or cultured in

tryptophan free media supplemented with kynurenine. DCs and T cells were co-cultured

66

for 4 d. After day 4, cells were centrifuged and washed with 1% fetal bovine serum in

PBS and incubated with LIVE/DEAD Fixable Near-IR Dead Cell Stain kit for 10 mins at

room temperature. Dye was then removed and cells incubated with antibodies against

CD16/32 (Fcγ III/II receptor) (clone 2.4G2) for 30 mins on ice. Cells were washed and

stained with antibodies against either CD4 (clone RM4-5) or CD8 (clone 53-6.7) for 30

mins on ice.

Statistical Analysis

Statistical analyses were performed using ANOVA followed by Tukey’s

significance test to make pair-wise comparisons. Differences were considered

significant when p<0.05 using GraphPad Prism.

Results

IDO-treated DCs Resist LPS-induced Maturation

Murine bone marrow derived DCs were treated with recombinant human IDO at

15 µg/mL for 24 h. At this concentration with a specific activity of 500 pmoles/min/µg, it

was calculated that IDO would catabolize all tryptophan present in the cell culture media

within 18 hours. After 24 h DCs were challenged with LPS, a known DC stimulant,

overnight (>16 h). Following LPS challenge, DCs were imaged to characterize their

morphology and representative phase contrast images are shown in Figure 2-2.

Untreated DCs displayed characteristics of an immature phenotype marked by microvilli

(Figure 2-2A, arrow) which become veils upon maturation in response to LPS (Figure 2-

2B, arrow). When cells were treated with IDO (Figure 2-2C), immature morphology was

maintained, suggesting DCs did not recognize the enzyme as a damage associated

molecular pattern or a pathogen associated molecular pattern. Similar morphologies

were observed when DCs were cultured in tryptophan free media (Figure 2-2D), DC

67

media supplemented with kynurenine (Figure 2-2E) or tryptophan free media

supplemented with kynurenine (Figure 2-2F). Finally, when DCs were treated with IDO

for 24 h and then challenged with LPS overnight, an immature phenotype was observed

(Figure 2-2G) demonstrating that IDO-treated DCs resist LPS-induced maturation.

The effects of IDO supplementation on DC viability were inspected using

microscopy and quantified through flow cytometry, shown in Figure 2-3 (results an

average of three experiments with three replicates, total n=9). DCs were cultured in DC

media or with Triton X-100, to serve as an untreated and dead cell control respectively,

with IDO, in tryptophan free media, in DC media supplemented with kynurenine, or in

tryptophan free media supplemented with kynurenine (Figure 2-3A). Green staining in

Figure 2A represent viable cells, whereas the red color signifies cell death. Viability was

further quantified through flow cytometry (Figure 2-3B) where all groups, apart from

Triton X-100, maintained 60% viability after 24 h, equivalent to untreated controls. This

indicates that IDO, tryptophan depletion, and kynurenine formation do not induce

dendritic cell death.

To quantitatively asses the suppressive capacity of DC treatment with IDO, the

expression level of stimulatory (MHC II) and co-stimulatory (CD80, CD86) molecules

was analyzed by flow cytometry, shown in Figure 2-4 (results an average of three

experiments with three replicates, total n=9). Untreated DCs expressed low frequency of

the maturation markers CD80 (Figure 2-4A, 9% + 0.6), CD86 (Figure 2-4B, 3% + 0.2)

and MHC II (Fiure. 2-4C, 7% + 0.8). In contrast, upon LPS treatment expression of all

three activation markers significantly increased to 47% + 0.5 (CD80), 18% + 0.2 (CD86)

and 33% + 0.8 (MHCII) of cells. When DCs were treated with IDO for 24 h, maturation

68

markers remained comparable to immature or untreated cells with 10% + 0.5 (CD80),

3% + 0.2 (CD86) and 7% + 0.7 (MHC II). However, when DCs were cultured with IDO

followed by LPS challenge, maturation markers were lower than LPS alone, with 24% +

0.5 (CD80), 5% + 0.2 (CD86) and 13% + 0.7 (MHC II), confirming cells treated with IDO

resist LPS-induced maturation. Next, the release of inflammatory and anti-inflammatory

cytokines, IL-12p70 and IL-10, was evaluated through ELISA (Figure 2-5, results an

average of three experiments with three replicates, total n=9). IL-12p70 secretion levels

were determined at 38 + 8 pg/mL (untreated) and 1920 + 7 pg/mL (LPS treated). When

IDO was introduced to the culture IL-12p70 secretion remained low (61 + 8 pg/mL).

However, when cells were treated with IDO and challenged with LPS, IL-12p70

secretion significantly diminished compared to the LPS alone group (65 pg/mL, p < 0.05

(Figure 2-5A)). Conversely, IL-10 levels remained consistent, with significant changes

from untreated cells observed only in the LPS treated group at 52 + 13 pg/mL (Figure 2-

5B).

IDO-treated DCs Suppress Antigen Specific Proliferation, and Suppression is Active Enzyme Dependent

To evaluate whether IDO-treated DCs can attenuate antigen specific T cell

proliferation, we co-cultured these DCs with CFSE labeled T cells isolated from

splenocytes of T cell receptor (TCR) transgenic mice, OT-I and OT-II. The CD4+ and

CD8+ T cells of these mice proliferate in response to OVA peptide 323-339 and 257-

264, respectively. The data shown in Figure 2-6 (results an average of three

experiments with three replicates, total n=9) represent the percent proliferation of viable

CD4+ or CD8+ T cells, with the gating scheme used to quantify proliferation (Figure 2-

6A-D). In co-cultures where only DCs and T cells were present, or when IDO alone was

69

introduced, minimal proliferation was observed (11% + 1 and 4% + 1 for CD4+ T cells

and 17% + 1 and 35% + 3 for CD8+ T cells, respectively). When DCs were pulsed with

the corresponding peptide and co-cultured with T cells, T cells were activated and

proliferation was 95% + 1 for CD4+ and 83% + 0.8 for CD8+ after 4 d. However, when

DCs were cultured with IDO for 24 h, washed, and pulsed with peptide, T cell

proliferation was reduced to basal levels, 16% + 1 for CD4+ T cells and 36% + 2 for

CD8+ T cells. T cells only were also cultured and included as a control with proliferation

at 8% + 1 for CD4+ T cells and 11% + 2 for CD8+ T cells (Figure 2-6E-F). The

experiment was repeated, with the introduction of 1-methyl tryptophan (MT), a potent

IDO inhibitor 163, as a DC treatment (Figure. 2-6G). In the presence of MT-treated DCs,

both CD4+ and CD8+ T cell proliferation were restored to over 70%, indicating active

enzyme is required for the attenuation of proliferation. Lastly, the proposed mechanism

of action for IDO-induced suppression, tryptophan starvation and kynurenine

accumulation, were evaluated (Figure 2-7, results an average of three experiments with

three replicates, total n=9). The same co-culture was used, with variations in relevant

components of the culture media. DCs and T cells were co-cultured and percent

proliferating cells assessed in tryptophan containing media (31% + 0.5), in tryptophan

depleted media (21% + 2.3), or in tryptophan depleted media supplemented with

kynurenine (30% + 2.3) respectively. T cells cultured with OVA-pulsed DCs exhibited

proliferating cell frequencies of 81% + 2 in tryptophan containing media, 31% + 2.3 in

tryptophan free media, and 39% + 3 in tryptophan containing media supplemented with

kynurenine. Notably, the largest suppression of T cell proliferation, at 15% + 3, was

observed when DCs were pulsed in OVA and cultured in tryptophan free media

70

supplemented with kynurenine, suggesting the most dramatic response would be

provided by the active enzyme. T cells alone were cultured in all media conditions

mentioned (data not shown), and did not differ significantly from control T cells in

tryptophan containing media (5% + 2 proliferation).

Discussion

Suppression of inflammatory processes would be beneficial for the treatment of

autoimmune and inflammatory diseases as well as whole organ transplantation. Current

treatments are limited to non-specific systemic immunosuppressant drugs which carry

harmful off-target effects and the potential for opportunistic infections 164. To address

these limitations, scientists have focused on reprogramming patients own immune

systems to take advantage of naturally occurring tolerance mechanisms such as the

upregulation of IDO. Since Munn and Mellor demonstrated that the presence of IDO at

the maternal-fetal interface played a pivotal role in maternal T cell tolerance to fetal

tissue in mammals, 148 IDO is considered a potent immunomodulator and promoter of

tolerance.

Subsequent studies by Munn and Mellor indicated that suppression of T cell

proliferation was driven by depletion of the essential amino acid. T cells stimulated in

the presence of tryptophan activated normally, whereas T cells stimulated without

tryptophan experienced growth arrest at the G1 phase 142. The tryptophan starvation

theory was later challenged by Terness et al. who infected human DCs with

recombinant adenovirus harboring the human IDO gene and co-cultured them with

allogeneic T cells 144. The authors concluded suppression of T cell proliferation was

driven by the accumulation of tryptophan metabolites, particularly kynurenine, 3-

hydroxykunureine, 3-hydroxyanthranilic and quinolinic acid which largely induced cell

71

death. However, no studies to date have addressed the immunomodulatory properties

of IDO in the extracellular space or indicated a potential mechanism of action. This

study demonstrates IDO administered in the extracellular milieu metabolically

reprograms immune cells and maintains its potent suppressive capacity: inhibits the

maturation of DCs, even in the presence of inflammatory stimulus, and subsequently

modulates their immunostimulatory functions in vitro. Here, we demonstrate

suppression of T cell proliferation is mediated by a combination of both hypotheses

mentioned above. Tryptophan starvation and formation of kynurenines combine to

abrogate T cell responses in vitro.

Dendritic cells are key regulators of the immune system and although they play a

crucial role in the initiation of inflammatory responses they are also able to induce anti-

inflammatory outcomes. The potential for DCs to activate tolerance-inducing

mechanisms has been shown previously to be closely related to their maturation state.

T cell tolerance can be induced by immature DCs, which are characterized by low

expression of stimulatory and co-stimulatory molecules, and secretion of suppressive

cytokines 84,165,166. Multiple approaches have been used to generate tolerogenic DCs in

vitro and in vivo primarily by pharmacological agents 14,116,167-169 as well as gene

silencing of pro-inflammatory molecules and cytokines 170. This is the first study to

deliver extracellular IDO to DCs and induce a suppressive phenotype. Dendritic cells

treated with IDO showed significant resistance to LPS-induced upregulation of MHC II

and CD80/86 molecules. Additionally, IL-12p70 production was significantly diminished

while IL-10 was maintained in IDO + LPS conditions. While we did not define the

mechanism by which IDO acts upon DCs, kynurenines can engage the DC AhR and

72

induce the production of intracellular IDO through a positive feedback loop 171. Previous

reports showed AhR signaling is required for expression of IDO in DCs 137,138, which in

turn mediates DC maturation status through generalized reduction in cellular energetics,

generated by tryptophan starvation and lack of NAD production 172. Multiple reports

have shown IDO-mediated T cell arrest also induces apoptosis 143,173,174. However, in

this study no significant cell death was observed in either population. Dendritic cells

were cultured without tryptophan or in the presence of kynurenine to simulate conditions

generated by the enzyme and cell viability was on par with control cells, consistent with

in vitro extracellular IDO cultures.

Following maturation studies, IDO-treated DCs were co-cultured with CD4+ or

CD8+ T cells isolated from OT-I and OT-II mice, respectively, to evaluate the potential of

IDO-treated DCs to dictate immune responses. Suppression of antigen-induced T cell

proliferation was observed only in the groups where DCs were exposed to extracellular

IDO. Additionally, suppression of proliferation was confirmed to be mediated by the

active enzyme through use of MT. 1-Methyl tryptophan inhibits conversion of tryptophan

to kynurenines, likely preventing activation of the AhR and halting further IDO synthesis.

To further confirm depletion is mediated by the active enzyme, co-cultures were

performed in simulated enzymatic conditions. We demonstrated both tryptophan

depletion and kynurenine production are required for maximum T cell suppression

indicating future studies implementing IDO as a biological therapeutic must deliver it in

the active form.

In this study, we provided evidence to establish IDO as a potent

immunomodulator in the extracellular space. In recent years, therapeutic application of

73

IDO has primarily focused on overexpression of the gene in transplant cells/tissues,

shown to prolong graft survival in a number of models 145,153-156, or inhibition of its

enzymatic activity for the treatment of cancer 157-160. However, genetic overexpression

of IDO through the use of viral and non-viral vectors faces several obstacles, such as

viral vector-induced inflammation and malignancies175,176, which may hinder its clinical

application 177. For example, Tan et al. demonstrated DCs transduced with commonly

used high efficiency viral-vectors upregulate expression of stimulatory co-stimulatory

molecules and in the case of adenovirus, induced the production of Th1 and pro-

inflammatory cytokines 178. Additionally, infected cells demonstrated altered function

and inability to properly stimulate mixed lymphocytes reactions. The data presented

here lays the foundation for the use of extracellular IDO as a biologically active

therapeutic recombinant protein, as the enzyme enacts T cell suppression in vitro

utilizing the mechanisms established in studies of its native intracellular functions. By

demonstrating IDO can function as a potent immunomodulator in the extracellular

space, new delivery strategies and tissue targeting can be designed and used to induce

anti-inflammatory responses in autoimmunity, chronic inflammation and transplant

models.

74

Figure 2-1. Tryptophan metabolism through the kynurenine pathway

75

Figure 2-2. Dendritic cells treated with IDO maintain an immature morphology even after

LPS challenge. Murine bone marrow derived DCs were cultured A) untreated, B) in the presence of LPS, C) with human recombinant IDO, D) in tryptophan free media, E) in tryptophan-containing media supplemented with kynurenine, F) in tryptophan-free media supplemented with kynurenine, or G) with human recombinant IDO for 24 h then challenged with LPS overnight. Arrows in panels A and B denote microvilli and veils characteristic of immature and mature DCs, respectively. Scale bar = 50 µm.

76

Figure 2-3. Treatment with soluble IDO does not induce dendritic cell death. Dendritic

cells were cultured in DC media or Triton X to serve as an untreated and dead cell control, respectively, in the presence of IDO, in tryptophan free media, in tryptophan-containing media supplemented with kynurenine or in tryptophan free media supplemented with kynurenine for 24 h. Following treatment, cells were A) stained with cell-permeant (green) and cell-impermeant dye (red) to denote live and dead cells, respectively, and imaged using AxioVision 200M. Scale bars = 50 µm or B) stained with live/dead fixable NIR dye and viability assessed through flowcytometry. n =3, mean + SEM with pair-wise significant difference from all other groups, ANOVA with Tukey’s post-hoc, denoted by * where p < 0.05.

77

Figure 2-4. Dendritic cells treated with soluble IDO resist LPS maturation. Dendritic cells

were incubated with soluble IDO for 24 h and challenged with LPS overnight. Untreated and LPS groups were included for comparison. Cells were immunostained for maturation markers A) CD80, B) CD86 and C) MHC II. Viable cells expressing CD11c and the marker of interest were assed via flow cytometry and shown as percent positive. n =3, mean + SEM with pair-wise significant difference from all other groups, ANOVA with Tukey’s post-hoc, denoted by * where p < 0.05.

Figure 2-5. Treatment of dendritic cells with exogenous IDO inhibits their IL-12 secretion

and maintains IL-10 production. Dendritic cells were incubated with IDO for 24 h and challenged with LPS overnight. Untreated and LPS groups were included for comparison. A) IL-12p70 secretion and B) IL-10 secretion were evaluated via ELISA of the supernatant for each condition. n =3, mean + SEM with pair-wise significant difference from all other groups, ANOVA with Tukey’s post-hoc, denoted by * where p < 0.05.

78

Figure 2-6. IDO-treated DCs suppress antigen specific T cell proliferation, and

suppression is active enzyme dependent. Dendritic cells were incubated with IDO in the presence or absence of 1-methyl tryptophan (MT) for 24 h, pulsed with the corresponding ovalbumin peptide (OVA) for 3 h. Dendritic cells were then co-cultured with CD4+ or CD8+ CFSE labeled T cells for 4 d. T cell proliferation was quantified through CFSE dilution via flowcytometry. Representative histograms for live T cells stimulated with A) non-treated DCs, B) DCs pulsed with OVA, C) IDO-treated DCs pulsed with OVA, D) IDO-MT-treated DCs pulsed with OVA. T cell proliferation was further quantified for E) CD4+ and F) CD8+ population where + and – represent presence or absence of specific component. G) CD4+ T cell proliferation assay with the introduction of MT during IDO treatment of DCs. n =3, mean + SEM with pair-wise significant difference from all other groups, ANOVA with Tukey’s post-hoc, denoted by * where p < 0.05.

79

Figure 2-7. Tryptophan depletion and kynurenine accumulation are needed to suppress

antigen specific proliferation. Dendritic cells were cultured in tryptophan (tryp) free media supplemented with kynurenine (kyn) or relevant controls for 24 h then pulsed with ovalbumin peptide (OVA) for an additional 3 h. Cells were washed and co-cultured with CD4+ CFSE labeled T cells isolated from OT-II mice for 4 d. + and – denote the presence or absence of a particular component during assay. Proliferation of live T cells was quantified through CFSE dilution via flowcytometry. n =3, mean + SEM with pair-wise significant difference from all other groups, ANOVA with Tukey’s post-hoc, denoted by * where p < 0.05.

80

CHAPTER 3 FUSION OF GALECTIN 3 WITH A MODEL ENZYME AT THE N-TERMINUS

PROLONGS RETENTION TIME IN VIVO

Background

Galectin 3

Galectins are evolutionarily conserved carbohydrate-binding proteins that share a

common structural fold and at least one carbohydrate recognition domain (CRD) of

approximately 130 amino acids179. The classification of all 15 galectins identified up to

date is based on structural similarities and includes: prototype galectins (galectin 1,

galectin 2, galectin 5, galectin 7, galectin 10, galectin 11, galectin 13, galectin 14 and

galectin 15), which have one CRD and exist as monomers or dimers; tandem repeat-

type galectins (galectin 4, galectin 6, galectin 8, galectin 9, galectin 12), which contain

two CRDs separated by a linker; and the chimera type galectin 3 (Gal3), which contains

a CRD connected to a non-lectin amino-terminal region179 (Figure 3-1). Within the

immune system, galectins are expressed by virtually all cells, either at a basal level or in

an inducible manner, and are significantly upregulated by activated B cells, T cells,

inflammatory macrophages, natural killer cells and CD4+CD25+ Tregs180. Particularly,

galectin 3 can be found within the nucleus, in the cytosol, on the cell surface and in the

extracellular compartment. It is well documented that Gal3 acts as an adhesion

molecule by cross-linking adjacent cells181. However, intracellularly, Gal3 is engaged in

several processes including pre-mRNA splicing182, regulation of cell growth183, cell cycle

progression and apoptosis184. Additionally, work by Demetriou et. al. has shown that

multivalent Gal3-N-glycan complexes can limit TCR clustering by restricting lateral TCR

movement within the plane of the membrane hindering T cell signaling184.

81

Galectin 3 as a retention strategy. Galectin 3 is member of the carbohydrate-

binding soluble lectin family and has the ability to oligomerize into a pentamer through

its flexible collagen-like domain on the N-terminus upon binding through the CRD to a

carbohydrate ligand181. Specifically, Gal3 has a strong affinity for N-Acetyl-D-

lactosamine (LacNAc) present in proteins of the extracellular matrix and certain surface

receptors on various immune cells184-187. Its pentameric architecture allows for cross-

linkage of cell surfaces and extracellular matrix proteins, triggering signals that dictate

the outcome of events such as cell adhesion, differentiation, and apoptosis188,189.

Galectin 3 has also been linked to tumor metastasis by triggering angiogenesis through

cross-linking of LacNAc presenting vascular endothelial growth factor (VEGF) receptor

2 and prolonging its retention at the cell surface190-194. Therefore, Gal3 has been

considered a target for cancer therapy with fusions of Gal3 proposed as drug delivery

tools in the context of tumor angiogenesis195-197. In this study, we show fusion of Gal3

with a model enzyme or IDO through the N-terminus alters its physiological

characteristics while conserving its binding affinity through the CRD.

NanoLuciferase

Bioluminescence methodologies are extraordinarily useful, particularly as

reporter molecules, due to their high sensitivity, broad dynamic range, and operational

simplicity198. These characteristics have incrementally been improved through

adaptations of native enzymes and substrates, originating from luminous organisms of

diverse evolutionary lineage199. Promega engineered both the enzyme: Nanoluciferase

(NanoLuc), from deep sea shrimp (Oplophorus gracilirostris), and the substrate:

furimazine, an imidazopyrazinone analog, to create a superior reporter system with

more efficient light emission200. The new enzyme produces signal (half-life > 2 h) with a

82

specific activity 150-fold greater than that of either firefly (Photinus pylaris) or Renilla

luciferases. Additionally, NanoLuc exhibits greater physical stability, retaining activity up

to incubations at 55°C or in culture medium for more than 15 h at 37°C. In this study

NanoLuc was used as a model enzyme to evaluate the ability of Gal3 to serve as a

tissue-targeting strategy.

Materials and Methods

Cloning of Galectin 3, Nanoluciferase and NanoLuc-Gal3

Genes encoding recombinant human Gal3 (Origene) and NanoLuc-Gal3 (IDT)

were inserted into pET-21d+ vectors between the NcoI and XhoI sites (Figure 3-2) by

Margaret Fettis, a PhD candidate in the laboratory of Dr. Greg Hudalla. The modified

plasmids were transformed into Top10 E.coli (Thermo Fisher) and selected on 100

μg/mL ampicillin-doped LB/agar plates overnight at 37°C. Positive clones were

sampled and used to inoculate 5 mL 100 μg/mL ampicillin-containing LB broth. Cultures

were grown overnight at 37°C, 220 rpm on an orbital shaker. Plasmids were isolated

from cultures via a plasmid mini-prep kit (Qiagen), according to manufacturer’s

instructions, and sequenced at the Interdisciplinary Center for Biotechnology Research

at the University of Florida. Using PCR, Nanoluciferase was amplified from the

NanoLuc-Gal3 gene and mutated to have NcoI and XhoI flank the gene using the

primers, 5’-CGC CTC GAG CGC CAG AAT GCG TT-3’ and 5’-GCT TAG CCA TGG

CGG TCT TCA CAC TCG AAG-3’. The PCR product was digested using NcoI and XhoI

and re-inserted into pET21d+. Plasmids were screened and sequenced as described

above.

83

Protein Expression and Purification

Origami B (DE3) E.coli (Novagen) were transformed with pET-21d-Gal3, pET-

21d-NanoLuc, or pET-21d-NanoLuc-Gal3 vectors and selected on 100 μg/mL ampicillin-

and 50 μg/mL kanamycin A-doped LB/agar plates overnight at 37°C. Positive clones

were picked and used to inoculate 10 mL 100 μg/mL ampicillin- and 50 μg/mL

kanamycin A-containing LB broth. Cultures were grown overnight at 37°C, 220 rpm on

an orbital shaker. Cultures were expanded into 1 L 100 μg/mL ampicillin- and 50 μg/mL

kanamycin A-containing 2xTY broth and grown at 37°C, 225 rpm on an orbital shaker

until an optical density of 0.6-0.8 (λ=600 nm) was reached. Protein expression was

induced with 0.5 mM isopropyl β-D-1-thiogalactopyranoside (IPTG) and incubated for 18

h at 18°C, 225 rpm in an orbital shaker. Bacteria were pelleted by centrifugation, and

washed with PBS. Bacteria were lysed with B-PER (Thermo Fisher), a protease inhibitor

tablet (Thermo Fisher), 300 units DNAse I from bovine pancreas (Sigma), and 100 μg

lysozyme (Sigma) for 20 min. Lysed bacteria was cleared by centrifugation, and

supernatant containing recombinant proteins were loaded onto columns containing

HisPur cobalt resin (Thermo Fisher) equilibrated with PBS. Columns were washed with

20-30 column volumes and proteins were eluted with increasing concentration of

imidazole. Imidazole was removed by centrifugation, using Amicon filter tubes (MWCO

10kDa) (Millipore). For IDO-Gal3, a second purification step was required using size

exclusion chromatography in an AKTA pure chromatography system (GE Life

Sciences). Protein molecular weight and purity were analyzed with SDS-PAGE.

Endotoxin contaminants were removed by Detoxi-gel endotoxin removing columns

(Thermo Fisher) following manufacturer’s instructions. Endotoxin content was analyzed

84

using Pierce™ LAL Chromogenic Endotoxin Quantitation Kit (Thermo Fisher), and was

below 0.1 EU/mL in all stocks.

Mice and Cell Lines

C57BL/6 (B6) and B6.Cg-Tg(TcraTcrb)425Cbn/J (OT-II) mice were purchased

from Jackson Laboratory. Age-matched (6-12 weeks) female animals were used

throughout all experiments. All studies were performed in accordance with protocols

approved by the Institutional Animal Care and Use Committee (IACUC) at the University

of Florida. Experimental groups and sizes were approved by authorities for animal

welfare after being defined to balance statistical power, feasibility and ethical aspects.

All animals were cared for/monitored/inspected daily either by a member of the research

team or with an agreement by Animal Care Services personnel. Jurkat T cells were a

generous gift from the laboratory of Dr. Mark Atkinson purchased from the American

Type Culture Collection number TIB-152 clone E6-1.

Galectin 3 and NanoLucGal-3 Binding Affinity

Affinity of Gal3 and NanoLucGal3 for lactose was determined using affinity

chromatography in an AKTA Pure chromatography system (GE Life Sciences) equipped

with an α-lactose agarose column (Sigma-Aldrich) by Antonietta Restuccia, a PhD

candidate in the laboratory of Dr. Greg Hudalla. Proteins were eluted with a linear

gradient of β-lactose (Sigma-Aldrich) in phosphate buffer.

NanoLuc-Gal3 Binding Affinity to ECM Proteins

Non-tissue culture treated polystyrene plates were coated with either natural

mouse laminin (Invitrogen), collagen I from rat tail (Life Technologies), vitronectin from

human plasma (Corning Cellgro), IgG from mouse serum (Sigma-Aldrich) or α2-

macroglobulin from human plasma (Sigma-Aldrich) at various concentrations. Plates

85

were washed 3 times with PBS (ThermoFisher Scientific) and blocked with 1% heat

denatured bovine serum albumin (BSA) (ThermoFisher Scientific) for 30 min at room

temperature. Plates were washed as described earlier and incubated with 2nM

NanoLuc-Gal3 in the presence or absence of excess β-lactose (100mM) for 45 mins at

room temperature. Plates were washed and furimazine (Promega) added according to

manufacturer’s instructions. Bioluminescence was read using a Biotek Flx Fluorescent

Microplate Reader and compared to a standard curve to determine concentration of

bound protein.

NanoLuc-Gal3 Binding Affinity to Jurkat T Cells

Jurkat T cells were seeded at a density of 100,000 per well and incubated with

2nM NanoLuc-Gal3 in the presence or absence of excess β-lactose (100mM) for 1 hour,

washed 3 times with PBS and incubated with furimazine according to the

manufacturer’s instructions. Bioluminescence was read using a Biotek Flx Fluorescent

Microplate Reader. Results were compared to standard curve to determine

concentration of bound protein.

Jurkat T Cell Agglutination and Viability

Jurkat T cells were seeded at a density of 20,000 per well and incubated with 5

µM NanoLuc-Gal3 or 5 µM Gal3. After 1 hour, cells were imaged using a Zeiss Axiovert

200M with a 20X objective lens (Carl Zeiss Microscopy) to determine agglutination.

After 24 hours, viability was assessed using the LIVE/DEAD Fixable NIR Dead Cell

Stain Kit for 633 or 635 nm excitation (Life Technologies) according to the

manufacturer’s instructions and analyzed using flow cytometry.

86

Quantitative Precipitation Analysis of Galectin 3 and NanoLuc-Gal3

Precipitation analysis of galectin-3 and NanoLucGal-3 were completed following

protocols established elsewhere201 by Antonietta Restuccia. Briefly, 30 uM of galectin-3

or NanoLuc-Gal3 were incubated with asialofetuin (ASF, Sigma-Aldrich) at different

molar ratios in a 20 mM sodium phosphate buffer, pH 7.2, containing 10 mM β-

mercaptoethanol and 0.15 M sodium chloride. Proteins were incubated at room

temperature for 30 min, and light scattering was measured at 420 nm using

SpectraMax® M3 Multi-Mode Microplate Reader (Molecular Devices).

In Vivo Bioluminescence and Imaging

Bioluminescence images were acquired using an IVIS Spectrum In Vivo System

(IVIS). Living Image Software version 4.3.1 (Perkin Elmer, Waltham, MA) was used to

acquire the data immediately after furimazine administration. Exposure time for the

bioluminescence imaging was 1 second. Regions of interest were quantified as the

average radiance (photons/second/cm2/sr).

NanoLuc-Gal3 Tissue Distribution from Hock Injection

164 picomoles of NanoLuc-Gal3 in 40 µL of PBS were injected subcutaneously

into the hock of B6 mice. At the prescribed time points, animals were euthanized in

accordance with approved protocols. Organs and tissues of interest were harvested,

weighed, processed, incubated with furimazine and bioluminescence quantified by a

luminometer. Specific amount of protein in tissue was determined by comparison with a

standard curve of NanoLuc-Gal3 activity.

Alternative Injection Sites

164 ρmoles of NanoLuc-Gal3 or NanoLuc in 10 µL (subgingival), 100 µL

(intraperitoneal), 40 µL (intramuscular), 40 µL (subcutaneous – scruff of the neck) or 40

87

µL (subcutaneous – abdominal region) of PBS were administered. Furimazine (1:50

dilution in PBS at the same sites and volumes listed above) was administered daily until

no signal was observed for two consecutive days.

Statistical Analysis

Statistical analyses were performed using ANOVA followed by Tukey’s

significance test to make pair-wise comparisons. Differences were considered

significant when p<0.05 using GraphPad Prism.

Results

NanoLuc-Galectin 3 Retains Binding Affinity for Sugar Moiety when Compared to WT-Gal3

Equal concentrations of NanoLuc-Gal3 (red line) and wildtype galectin 3 (black

line) were injected into an α-lactose-agarose resin column and eluted with an increasing

β lactose gradient (Figure 3-3A). Wildtype and fusion proteins elute at the same β

lactose concentration (20 mM) indicating equivalent binding affinity for β lactose.

Galectin 3 Retains Binding Affinity to Sugar Moiety on ECM and Serum Proteins When Fused to an Enzyme on the N-terminus

Non-tissue culture treated polystyrene plates were coated with various

concentrations of proteins of the extracellular matrix known to bind Gal3 with various

degrees of affinity202-204. Namely, laminin, collagen type I, vitronectin, IgG and α2

macroglobulin were incubated with NanoLuc-Gal3 in the presence or absence of excess

β-lactose (Figure 3-3B-F). Plates were then washed and incubated with furimazine to

determine NanoLuc activity and correlated to specific protein mass through a standard

curve. Binding to all proteins is mediated by the carbohydrate recognition domain (CRD)

as determined by the incorporation of β-lactose which competes with immobilized

protein for the CRD. Binding follows a trend associated with the concentration of

88

immobilized protein, less protein present represents less binding of NanoLuc-Gal3,

consistent with expected LacNAc content. Additionally, binding to laminin is much

higher than collagen type I, vitronectin and both serum proteins, again consistent with

expected binding sites.

Galectin 3 Is Not Able to Self-assemble Into Pentamer and Precipitate Asialofetuin When Fused to NanoLuc on the N-terminus

Asialofetuin (ASF) is a glycoprotein containing complex carbohydrate chains

with terminal galactose and N-acetyl lactosamine (LacNAc) residues. As found

previously201, galectin-3 binds to terminal LacNAc, crosslinking and precipitating ASF as

seen by the increased light scatter. NanoLucGal-3, on the other hand, fails to crosslink

and precipitate ASF at any of the ratios of lectin to glycoprotein studied (Figure 3-4A).

Galectin 3 Binds to Jurkat T Cells but Does Not Induce Agglutination or Apoptosis

It has been well documented that WT Galectin 3 induces T cell agglutination and

apoptosis by binding CD7 and CD29 and inhibiting TCR clustering186,205. In this study,

Jurkat T cells were incubated with NanoLuc-Gal3 in the presence or absence of excess

β-lactose. First, binding was determined by washing cells and incubating them with

furimazine and a specific mass was determined by comparison to a standard curve (Fig.

2-3B). Jurkat T cells incubated with NanoLuc-Gal3 showed binding of 0.56 + 0.2 ng of

protein whereas Jurkat T cells incubated with NanoLuc-Gal3 and excess β-lactose

showed significantly decreased binding (p <0.05, 0.07 + 0.04 ng). Jurkat T cells were

then cultured untreated, with Gal3 or NanoLuc-Gal3 imaged and stained with Live/Dead

dye to assess agglutination and viability, respectively (Fig. 2-3C,D). Untreated cells

shown no agglutination and 53% + 4 viability, cells treated with Gal3 induced

agglutination determined by cells clusters and showed only 0.6% + 0.2 viability.

89

Conversely, cells treated with NanoLuc-Gal3 showed no appreciable clusters and 61%

+ 3 viability, comparable with untreated cultures.

NanoLuc-Gal3 Prevents Galectin 3-induced Cell Death

Jurkat T cells were incubated with various ratios of NanoLuc-Gal3 and Gal3

(Figure 3-5) to assess whether NanoLuc-Gal3 acts as an inhibitor of wild type Gal3.

Representative phase contrast images demonstrating Gal3-mediated agglutination of T

cells is shown in Fig. 3-5A. Untreated cells or cell treated with NanoLuc-Gal3 do not

exhibit appreciable clusters of cells, whereas cells treated with Gal3 severely

agglutinate after 4 h of culture. Remarkably, agglutination is alleviated upon incubation

with a 1:1 or a 2:1 ratio of NanoLuc-Gal3 to Gal3. Upon treatment with 3:1 ratio,

agglutination resembles that observed with untreated cells, trend is maintained at 4:1

and 5:1 ratios. Furthermore, viability following agglutination was evaluated (Fig. 3-5B).

Cultures of untreated cells showed 66% + 3 and 33% + 3 live and dead cells,

respectively. Upon treated with NanoLuc-Gal3 levels remained comparable to untreated

group with 63% + 4 live and 36% + 4 dead cells. Following incubation with Gal3, cell

cultures decreased to 30% + 0.5 live and increased to 68% + 1 dead. Treatment with

NanoLuc-Gal3 and Gal3 at a 1:1 ratio, increased viability to 54% + 2 and decreased cell

death to 44% + 3. An upward trend was observed in cell viability as the concentration of

NanoLuc-Gal3 was increased, with 63% + 6 live cells and 35% + 6 dead cells for the 2:1

ratio, 64% + 5 live cells and 29% + 3 dead cells for the 3:1 ratio, 69% + 1 live cells and

27% + 0.5 dead cells for the 4:1 ratio and 72% + 2 live cells and 25% + 2 dead cells for

the 5:1 ratio.

90

NanoLuc-Gal3 Binds Primary Splenocytes in a CRD Dependent Manner, Binding Does Not Act as a Damage Associated Molecular Pattern

NanoLuc-Gal3 was labeled with Fluorescein (FITC) and incubated with

splenocytes isolated from B6 mice in the presence or absence of excess β-lactose,

untreated cells were used as a negative control (Figure 3-6A). Approximately 30% of

CD3+CD4+ cells were bound to NanoLuc-Gal3 which significantly reduced to 10% when

treated with β-lactose; 25% of CD3+CD8+ decreased to 10%, 20% CD11c+ decreased to

8%, 40% B220 decreased to 10% and 25% of F4/80 decreased to 10%.

Galectin 3 Targeting in NanoLuc-Gal3 Provides Prolonged Retention Time When Administered Subcutaneously, Intraperitoneally and Intramuscularly

NanoLuc-Gal3 or NanoLuc was administered to B6 mice through various pre-

clinical relevant routes and imaged using an IVIS system (Figure 3-7). When

administered subcutaneously at the scruff of the neck, NanoLuc-Gal3 remained at the

injection site for 1 d, when introduced intraperitoneally 2 d, subcutaneously at the

abdominal region for 5 d, intramuscularly for 1 d, and subcutaneously at the hock for 7

d. Conversely, untargeted NanoLuc cleared from all injection sites within 24 h after

administration (Fig. 3-7A-E). Furthermore, the average radiance emitted was quantified

and as shown in the images, NanoLuc-Gal3 (black line) had a prolong local activity

when compared to untargeted NanoLuc (blue line) (Fig. 3-7F-J), demonstrating Gal3

serves as a retention strategy in vivo.

Galectin 3 Targeting in NanoLuc-Gal3 Provides Prolonged Retention Time When Administered Into the Hock with Minimal Drainage to Adjacent Tissues

NanoLuc-Gal3 was administered subcutaneously at the hock, mice euthanized

at various time points and tissues of interest harvested, processed, incubated with

furimazine and bioluminescence quantified by luminometer. The highest quantity of

91

protein was detected in the hock for 7 days with minimal drainage to the spleen,

bladder, kidneys and blood.

Discussion

Proteins have the most dynamic and diverse roles of any macromolecule in the

body, catalyzing biochemical reactions, forming receptors and channels on cell

membranes, transporting molecules within cells or organs, or aiding in cell to cell

communications 206. Because of their versatility, proteins are often used for replacement

therapies, to augment an existing pathway or to provide a novel function. For example,

enzyme replacement therapy, is a common practice for the treatment Goucher’s

disease, a genetic lysosomal storage disease characterized by deficiency of the

enzyme glucocerabrosidase 207. Deficit of the enzyme leads to accumulation of its

substrate, glucocerebroside, within the lysosomes of macrophages resulting in anemia,

thrombocytopenia, bone disease, hepatomegaly, and splenomegaly 208,209. In

Goucher’s disease the target cells are macrophages, which can efficiently uptake the

recombinant enzyme, imiglucerase, through the mannose-6-phosphate receptor

providing a unique targeting strategy 209. However, most enzymes do not make optimal

therapeutics as they are rapidly cleared from circulation by renal filtration and have half-

lives of minutes to a few hours leading to poor bioavailability and decreased activity 52,

which in many cases can render them marginal or unsuitable for clinical applications

210,211.

Fusion protein technologies aim at addressing rapid clearance limitation often

observed with enzyme therapeutic candidates. They exploit different properties of

protein domains to generate new molecular entities with desirable or enhanced

properties. The most clinically and commercially successful fusion therapeutics to date

92

contain the Fc fragment of immunoglobulins 212. Such Fc fusion can increase proteins

and peptides serum half-life to days and even weeks by binding to the salvage receptor,

FcRn 53,54. However, these technologies and platforms do not provide a tissue-retention

strategy and can often elicit an immune response213. In this study, we developed and

characterized a new fusion chimera, NanoLuc-Gal3, which demonstrated maintenance

of function for both the targeting and enzymatic moieties in vitro and prolonged retention

time of model enzyme in vivo.

Following expression and purification of the fusion, NanoLuc-Gal3, the activity of

both molecules was tested. Analysis of binding affinity for α-lactose agarose column

revealed fusion construct behaves almost identical to wild type Gal3 consistent with

published literature demonstrating proteins fused to Gal3 through the N-terminus have

no effect on Gal3 binding to its sugar ligand193,194. Additionally, NanoLuc-Gal3 binding to

immobilized proteins was also observed to be consistent with published works. Gal3

fusion binds components of the extracellular matrix with carbohydrate specificity, as

determined by the competitive assay with β-lactose, and binding to laminin is much

higher than collagen type I, vitronectin and serum proteins consistent with expected

LacNAc content 181,202,204,214.

Following ligand binding, wild type Gal3 undergoes conformational changes,

which enable its oligomerization into higher order structures 179. Therefore, we sought to

evaluate NanoLuc-Gal3 oligomerization capacity compared to Gal3 in collaboration with

Dr. Greg Hudalla and his student Antonietta Restuccia. Asialofetuin (ASF) is a

glycoprotein containing complex carbohydrate chains with terminal galactose and

LacNAc residues. As previously established 201, Gal3 binds to terminal LacNAc,

93

crosslinking and precipitating ASF as seen by the increased light scatter. NanoLucGal-

3, on the other hand, fails to crosslink and precipitate ASF at any of the ratios of lectin to

glycoprotein studied. Galectin 3 crosslinking of ASF molecules is driven by the

oligomerization of the lectin via the N-terminus. In the case of NanoLucGal-3, fusion is

achieved through the N-terminus likely making it unavailable for self-assembly into

higher ordered structures.

Much of the physiological processes involving Gal3 are due to its ability to

oligomerize into pentamers. With respect to immune cells, Gal3 acts in a dual manner,

either protecting T cells from apoptosis or promoting cell death, depending on whether it

functions intracellularly or is added exogenously to T cell cultures 179,186,215. Extracellular

Gal3, induces cell death in human T cells by binding to CD7 and CD29 186 or to CD45

and CD71 205 and mobilizing intracellular Ca2+ to promote the exposure of

phosphatidylserine on the cell surface, thus preparing the cell for phagocytic recognition

and subsequent removal 216. Additionally, Gal3-ligand lattices can limit T cell receptor

(TCR) clustering, thereby increasing the threshold for TCR signaling. In this study,

NanoLuc-Gal3 was exogenously administered to Jurkat T and determined to bind

specifically through the CRD to the surface of the cell. Whereas wild type Gal3

promotes Jurkat T cell agglutination and apoptosis driven by the N-terminal self-

association domain, fusion of NanoLuc at the N-terminus domain abrogates this activity,

presumably by blocking oligomerization. Additionally, we demonstrated NanoLuc-Gal3

can bind primary splenocytes in a CRD-dependent manner and binding to DCs

increases expression levels of stimulatory and co-stimulatory molecules. The data

presented here demonstrated NanoLuc-Gal3 does not act as a damage associated or

94

molecular associated pattern and does not initiate an inflammatory response, an

important characteristic for any therapeutic aiming to suppress inflammation.

Previous studies have demonstrated Gal3 with a truncated N-terminus can act as

an inhibitor of wild type Gal3 due to higher affinity for carbohydrate ligand 194. Although

no higher binding affinity was observed by fusion with NanoLuc, the ability of NanoLuc-

Gal3 to inhibit wild type Gal3 was evaluated. NanoLuc-Gal3 and Gal3 were

administered at various ratios to Jurkat T cells and agglutination and apoptosis

measured. Interestingly, the fusion construct competes with wild type Gal3 and even at

a 1:1 ration apoptosis is significantly reduced. This effect is attributed again to the

inability of the fusion protein to oligomerize into higher structured lattices often

associated with T cell apoptosis. Although not explored here, the data presented could

serve a new line of research for the development of Gal3 inhibitors sought after in

cancer metastasis applications 217.

With the work presented here, demonstrating fusion of Gal3 with a model

enzyme does not alter the binding affinity or activate immunological responses, we

sought to establish Gal3 as a tissue retention platform. In this study, we demonstrated

the Gal3 targeting domain in the NanoLuc-Gal3 fusion provides a prolonged retention

time for all sites tested whereas non-targeted NanoLuc is undetectable at all injection

sites after 24 h. Although an improvement is observed for all sites tested, a significantly

shorter retention time is observed for the intramuscular (IM) and intraperitoneal (IP)

groups. We hypothesize in the IM group this is due to injections of furimazine not

penetrating the tissue far enough limiting the necessary physical proximity between

enzyme and substrate, or the signal emitted is too deep into the tissue and below the

95

detection limit of the equipment used. For the IP group, although not tested here,

retention time is likely decreased due to the highly hydrodynamic environment and fast

turnover of proteins in the peritoneal fluid 218. Lastly, we established drainage of

NanoLuc-Gal3 administered at the hock to be minimal to major organs and clearance

through the renal system consistent with approved enzyme and fusion therapeutics.

96

Figure 3-1 Classification of galectins

Figure 3-2. Fusion protein construct. A) Schematic representation of wild-type galectin 3

(WT Gal3) and NanoLuciferase-Galectin 3 fusion protein. WT Gal3 forms a pentamer through self-assembly of the N-terminus domain. Upon fusion of NanoLuciferase at the N-terminus, self-assembly is disrupted while binding affinity for N-acetyl-D-lactosamine (LacNAc), present on proteins of the extracellular matrix and surface receptors on immune cells, is maintained. B) Gene construct for WT Gal3, NanoLuciferase, and NanoLuc-Gal3.

97

Figure 3-3. Galectin 3 retains binding affinity to sugar moiety when fused to an enzyme

on the N-terminus. A) Equal concentrations of NanoLuc-Gal3 (red line) and wildtype galectin 3 (black line) were injected into an α-lactose-agarose resin column and eluted with an increasing β lactose gradient. Wildtype and fusion proteins elute almost identically, indicating equivalent binding affinity. B) Polystyrene plates were coated at the specified concentrations with laminin, C) collagen type I, D) vitronectin, E) IgG, and F) α2 macroglobulin. NanoLuc-Gal3 was added either in the presence or absence of excess β lactose (to compete for Gal3 binding) and incubated 45 min. Plates were then washed, furimazine (NanoLuciferase substrate) added and bioluminescence quantified. Gal3 fusion binds extracellular matrix proteins with carbohydrate specificity, as β lactose blocks CRD activity; binding to laminin is much higher than collagen type I, vitronectin, and serum proteins consistent with expected LacNAc content.

98

Figure 3-4. Biology associated with Galectin 3 is disrupted when fused to an enzyme on

the N-terminus. A) Incubation of Galectin 3 and NanoLuc-Gal3 (30 uM) with increasing concentrations of asialofetuin (ASF), a glycoprotein containing carbohydrate chains with terminal LacNAc residues. Galectin 3 crosslinks and precipitates ASF molecules even at low ASF concentrations as determined by light scattering at 420 nm. NanoLuc-Gal3 fails to precipitate ASF, likely due to lack of the self-association domain that joins multiple CRDs and lead to the formation of large aggregates. B) Jurkat T cells were cultured with NanoLuc-Gal3 1 h in the presence or absence of excess β lactose. Cells were washed and incubated with furimazine to detect bioluminescence. NanoLuc-Gal3 binds to Jurkat through the CRD as demonstrated by inhibition with excess β lactose. T cells were cultured with NanoLuc-Gal3 1 h, C) imaged via phase contrast (scale bar = 50 µm) to evaluate agglutination, or D) live/dead stained and analyzed by flow cytometry to determine viability. Whereas wildtype galectin 3 promotes Jurkat T cell agglutination and apoptosis driven by the N-terminal self-association domain, fusion of NanoLuc to galectin 3 at the N-terminus abrogates this activity, presumably by blocking access to the self-association domain. N=3, mean + SEM with pair-wise significant difference (by ANOVA and Tukey’s post hoc) denoted by * where p < 0.05.

99

Figure 3-5. NanoLuc-Gal3 fusion prevents wild type Galectin 3-mediated agglutination

and apoptosis of Jurkat T cells. Jurkat T cells were cultured with NanoLuc-Gal3 (x µM), Galectin 3 (x µM) or different ratios of the two proteins for 4 h, A) imaged via phase contrast (scale bar = 50 µm) to evaluate agglutination, or B) live/dead stained and analyzed by flow cytometry to determine viability. Whereas wildtype galectin 3 promotes Jurkat T cell agglutination and apoptosis, NanoLuc-Gal3 competes with WT-Galectin 3 and prevents this activity in a concentration dependent manner. N=3, mean + SEM with pair-wise significant difference (by ANOVA and Tukey’s post hoc) denoted by * where p < 0.05.

100

Figure 3-6. NanoLuc-Gal3 fusion binds primary splenocytes in a CRD dependent

manner, binding to DCs does not act as a damage associated molecular pattern. A. Splenocytes were incubated with FITC-labeled NanoLuc-Gal3 fusion 45 min in the presence or absence of excess β lactose. Cells were immunostained and binding was assessed by flow cytometry of T cells (CD3, CD4 & CD8), dendritic cells (CD11c), B cells (B220) and macrophages (F4/80). Gal3 fusion binds leukocyte proteins with carbohydrate specificity. B. Murine bone marrow derived DCs were incubated with NanoLuc-Gal3 fusion in the presence or absence of excess β lactose. Binding to dendritic cells is CRD-mediated. C. Murine bone marrow derived DCs were treated with NanoLuc-Gal3 for 24 h. Cells were immunostained for activation markers (CD80, CD86 MHC II). Viable CD11c+ cells and the marker of interest were assed via flow cytometry, shown as % positive. NanoLuc-Gal3 does not activate DCs, suggesting foreign protein does not act as a damage associated molecular pattern. N=3, mean + SEM with pair-wise significant difference (by ANOVA and Tukey’s post hoc) denoted by * where p < 0.05.

101

Figure 3-7. Galectin 3 targeting in NanoLuc-Gal3 provides prolonged retention when

administered subcutaneously, intraperitoneally, and intramuscularly. NanoLuc or NanoLuc-Gal3 was administered to 8-week-old C57Bl/6 female mice. At specified times, furimazine was injected and animals imaged using in vivo imaging system (IVIS). Representative images of NanoLuc (left panel) and NanoLuc-Gal3 (right panel) administered A. subcutaneously into the scruff of the neck, B. intraperitoneally, C. subcutaneously into the abdominal region, D. intramuscularly or E. subcutaneously into the hock. F-J. Average radiance (p/s/cm2/sr) for each injection site was quantified. NanoLuc-Gal3 is retained subcutaneously for up to 7 d, intraperitoneally up to 3 d and intramuscularly up to 1 d whereas non-targeted NanoLuc is undetectable at 1 d for all administration routes. n=5 mice per administration route per protein, mean + SEM, with pair-wise significance (by ANOVA and Tukey’s post hoc) between both proteins denoted by ^, compared to 0 d for NanoLuc denoted by + and compared to 0 d for NanoLuc-Gal3 denoted by *, where p<0.05.

102

Figure 3-8. Galectin 3 targeting provides retention of a model enzyme at the injection

site with minimal drainage to adjacent tissues. NanoLuc-Gal3 was injected subcutaneous at the hock of 8-week-old C57BL/6 female mice and tissue distribution assessed over a 7 d period. To measure localized active NanoLuciferase, organs and tissues were excised, processed, incubated with furimazine and bioluminescence quantified by luminometer. n=5 mice per time point. NanoLuciferase remains active and at the injection site for up to 7 d, with minimal drainage to adjacent tissue and clearance through the renal system.

Sple

en

Liver

Lungs

Kid

ney

Inte

stin

e

Bla

dder

Inj.

hock

inj.

tibia

inj.

fem

ur

Con. h

ock

Con. t

ibia

Con. f

emur

Pla

sma

RBC

Urin

e

Feces

0.0

0.2

0.4

0.6

0.8

1 day 2 days 4 days 5 days 6 days 7 days

ug

pro

tein

/g t

issu

e

103

CHAPTER 4 FUSION OF INDOLEAMINE 2,3 DIOXYGENASE WITH GALECTIN 3 RETAINS

ENZYME AT INJECTION SITE AND HALTS INFLAMMATION IN VIVO

Background

Subcutaneous LPS-induced Inflammation

Lipopolysaccharide (LPS), found on the outer layer of gram-negative bacteria, is

often used to induce inflammation in vitro and in vivo in an array of pre-clinical

models219-222. Most studies have focused on systemic, either intravenous (IV), or

intraperitoneal (IP), administration for the induction of inflammation223. The dose

required to induce detectable cytokine levels in serum is 2 ng/kg in humans and 500

ng/kg in mice with peak concentrations at 2 h post injection224. In this study, we

administered a dose of 2 ng/g of body weight of a mouse subcutaneously to induce a

localized inflammatory response with no adverse systemic distribution.

Periodontal Disease

Periodontal diseases (PD) are a class of chronic inflammatory conditions of the

soft and hard tissues found in the oral cavity. Periodontal disease is highly prevalent

and begins with gingivitis, localized inflammation of the gingiva. If left untreated,

gingivitis can result in progressive loss of gingival tissue, the periodontal ligament and

eventually the adjacent supporting alveolar bone225. Chronic inflammation is initiated by

complex subgingival biofilms generally containing a portion of the gram negative

anaerobic commensal oral microbiota (dental plaque) as well as opportunistic

pathogens, including Porphyromonas gingivalis226. While microbes are key agents of

periodontal disease, they do not directly cause disease, but rather induce harmful

inflammatory responses in a susceptible host227-230. For example, in response to

pathogens, polymorphonuclear cells release reactive oxygen species and other factors

104

that can damage the gingival tissue. The secreted agents also enhance the production

of numerous proinflammatory cytokines that contribute to the disease, including IL-1β,

IL-6, and TNFα231. Periodontal disease may also worsen other inflammatory conditions

such as type 1 diabetes and atherosclerosis232,233.

Immunopathogenesis

Under conditions of health, gingival tissues contain low number of T lymphocytes

that assist in the maintenance of homeostasis between the host tissue and the bacterial

plaque. Disease occurs when the balance between the microbial biofilm and the host

are lost. This imbalance is still not fully understood but there are significant changes

observed to both the dental plaque and the host immune profile that results in a

heighted inflammatory state231.

The first line of defense to pathogens is provided by epithelial cells that function

as a physical barrier against microbes and elicit innate and adaptive immune

responses. Langerhans cells within the epithelium take up antigenic materials, transport

it to lymphoid organs and present it to lymphocytes. Tissue infiltration by neutrophils,

granulocytes and lymphocytes follows: neutrophils try to engulf and eliminate bacteria,

but in the case of periodontal disease, are overwhelmed by the persistence of the

microbial biofilm. Early lesions follow an acute neutrophil response and a slight increase

in lymphocyte infiltration. In chronic disease, an increase in DCs and macrophages as

well as lymphocytes is observed with higher presence of CD8+ T cells than CD4+ T

cells. The relevant importance and timing of Th cell subset involvement is unclear. Th1

cells might be important during the early stages of chronic periodontitis, whereas Th2

cell involvement appears to occur at later stages234. Lastly, this severe chronic

105

inflammatory response leads to alveolar bone resorption by osteoclasts, degradation of

ligament fibers and the formation of granulated tissue235.

Current treatments

Current treatment modalities for PD focus on mechanical approaches to reduce

the number of pathogens through non-surgical and surgical therapies. Non-surgical

approaches are used as a first line of treatment and consist of removal of supragingival

and subgingival dental plaque and calculus with scaling and root planing231. Non-

surgical therapies can often be combined with adjunctive therapies such as the local

and systemic delivery of antibiotics, or the systemic modulation of the host’s immune

responses. However, non-surgical approaches can be ineffective on their own,

particularly when moderate (4-6 mm) to deep (> 6mm) pockets have formed. In these

cases, several surgical approaches are available. Pocket reduction surgery includes

resection of soft and hard tissue using various techniques236,237, regenerative guided

surgery uses membranes to direct the growth of new periodontium preventing the

epithelium and connective tissue from growing in areas desired for bone and

ligament238, grafting239 and more recently laser-assisted attachment as a more

conservative approach240.

A more favorable approach to the treatment of PD should reduce the bacterial

challenge and favorably modulate the host response locally and in a non-invasive

fashion. Because of the multifactorial nature of the host immunological responses in

periodontal disease, a multimodal approach controlling the host response would be

advantageous. A key aspect of PD pathogenesis commonly overlooked is its localized

pathology, whereby the nature of the inflammation and tissue breakdown is not

localized to the oral cavity, but to the specific teeth and some cases to specific sites of

106

these teeth. Thus, an adjunctive therapeutic approach would be most appropriate if it

can achieve sustained localized efficacy. In this study we develop, characterize and

utilize a chimera fusion protein containing an immunomodulatory moiety, indoleamine

2,3 dioxygenase (IDO), linked to a tissue-targeting molecule, Galectin 3 (Gal3), IDO-

Gal3, for the amelioration of inflammation in an LPS-induced model of inflammation and

a murine model of PD.

Materials and Methods

Cloning of IDO-Galectin 3

For the IDO-Gal3 gene, the IDO sequence was generously provided by the

laboratory of Dr. Carlos Gonzalez. Amplification and insertion of the BamHI restriction

site on the 3’, was done using the primers, 5’-CAG CTA CCA TGG CAC ACG CTA TGG

AAA-3’ and GAG AAC 5’-GGA TCC ACC TTC CTT CAA AAG-3’ by Antonietta

Restuccia. The NanoLuc-Gal3 gene was digested with NcoI and BamHI to remove

nanoluciferase gene and insert gal3 gene. Plasmids were screened and sequenced as

described in Chapter 3.

Protein Expression and Purification

Origami B (DE3) E.coli (Novagen) were transformed with pET-21d-galectin-3,

pET-21d-NanoLuc, pET-21d-NanoLuc-Gal3, or pET-21d-IDO-Gal3 vectors and selected

on 100 μg/mL ampicillin- and 50 μg/mL kanamycin A-doped LB/agar plates overnight at

37°C. Positive clones were picked and used to inoculate 10 mL 100 μg/mL ampicillin-

and 50 μg/mL kanamycin A-containing LB broth. Cultures were grown overnight at

37°C, 220 rpm on an orbital shaker. Plasmids were isolated from cultures via a plasmid

mini-prep kit (Qiagen), according to manufacturer’s instructions, and sequenced at the

Interdisciplinary Center for Biotechnology Research at the University of Florida. Cultures

107

of positive clones were then sub-cultured into 1 L 100 μg/mL ampicillin- and 50 μg/mL

kanamycin A-containing LB broth and grown at 37°C, 225 rpm on an orbital shaker until

an optical density of 0.6-0.8 (λ=600 nm) was reached. Protein expression was induced

with 0.5 mM isopropyl β-D-1-thiogalactopyranoside (IPTG) and incubated for 18 h at

18°C, 225 rpm in an orbital shaker. Bacteria were pelleted by centrifugation, and

washed with PBS. Bacteria were lysed with B-PER (Thermo Fisher), a protease inhibitor

tablet (Thermo Fisher), 300 units DNAse I from bovine pancreas (Sigma), and 100 μg

lysozyme (Sigma) for 20 min. Lysed bacteria was cleared by centrifugation, and

supernatant containing recombinant proteins were loaded onto columns containing

HisPur cobalt resin (Thermo Fisher) equilibrated with PBS. Columns were washed with

20-30 column volumes and proteins were eluted with increasing concentration of

imidazole. Imidazole was removed by centrifugation, using Amicon filter tubes (MWCO

10kDa) (Millipore). A second purification step was required using size exclusion

chromatography in an AKTA pure chromatography system (GE Life Sciences). Protein

molecular weight and purity were analyzed with SDS-PAGE. Endotoxin contaminants

were removed by Detoxi-gel endotoxin removing columns (Thermo Fisher) following

manufacturer’s instructions. Endotoxin content was analyzed using Pierce™ LAL

Chromogenic Endotoxin Quantitation Kit (Thermo Fisher), and determined to be below

0.1 EU/mL in all stocks.

IDO Enzymatic Activity Assay

Recombinant human IDO (IDO) expressed in Escherichia coli was purchased

from R&D Systems (Minneapolis, MN) with a predicted molecular mass of 46 kDa,

purity >95% by SDS-PAGE, endotoxin levels <1 EU/µg of protein by the LAL method

and a specific activity of >500pmoles/min/µg as measured by its ability to oxidize L-

108

tryptophan to N-formyl-kynurenine. The specific activity of both proteins, IDO and IDO-

Gal3, was measured before experiments to ensure maximal effect at the beginning of

the assay following an adapted procedure from Valladares et al.162. The reaction

substrate contained 200 µM tryptophan, 20 mM ascorbic acid, 10 µM methylene blue,

225 U catalase and 50 mM MES buffer (pH 6.5). Equal moles of IDO and IDO-Gal3

were loaded onto a flat bottom 96-well plate and the reaction started by mixture in 1:1

ratio with reaction substrate. Absorbance was measured at 480 nm.

Galectin 3 and IDO-Gal3 Binding Affinity

Affinity of galectin-3 and NanoLucGal-3 for lactose was determined using affinity

chromatography in an AKTA Pure chromatography system (GE Life Sciences) equipped

with an alpha-lactose agarose column (Sigma-Aldrich) by Antonietta Restuccia. Proteins

were eluted with a linear gradient of beta-lactose (Sigma-Aldrich) in phosphate buffer.

Antibodies

The CD11c (PE-Cy7 hamster anti-mouse clone: HL3), CD80 (APC hamster anti-

mouse clone: 16-10A1), CD86 (FITC rat anti-mouse clone: GL1), MHC II (PE rat anti-

mouse clone: M5/114.15.2) and CD16/32 (Fc block purified rat anti-mouse clone:

2.4G2) antibodies used to define DCs and their maturation status were purchased from

BD Bioscience. The CD4 ( PE-Cy7 hamster anti-mouse clone: RM4-5) and CD8 (PE rat

anti-mouse clone: 53-6.7) antibodies were used to define T cells and also purchased

from BD Bioscience. The stained cells were analyzed using a Guava easyCyte 8HT

Benchtop Flow Cytometer (EMD Millipore), using the InCyte Software (version 3.1) for

analysis.

109

Mice and Cell Lines

C57BL/6 (B6) and B6.Cg-Tg(TcraTcrb)425Cbn/J (OT-II) mice were purchased

from Jackson Laboratory. Age-matched (6-12 weeks) female animals were used

throughout all experiments. All studies were performed in accordance with protocols

approved by the Institutional Animal Care and Use Committee (IACUC) at the University

of Florida. Experimental groups and sizes were approved by authorities for animal

welfare after being defined to balance statistical power, feasibility and ethical aspects.

All animals were cared for/monitored/inspected daily either by a member of the research

team or with an agreement by Animal Care Services personnel.

DC Maturation and Cytokine Release Profile

Dendritic cells were generated from murine, bone marrow isolated from the

femurs and tibias of B6 mice in the presence of granulocyte-macrophage colony

stimulating factor (GM-CSF) (R&D Systems) according to previously described

protocols72. Cells were cultured untreated, with 1 µg/mL LPS (Sigma-Aldrich) overnight,

0.32 µM IDO-Gal3 for 24 h, or with IDO-Gal3 for 24 h, washed, and challenged with

LPS overnight. Cells were stained for viability using LIVE/DEAD Fixable NIR Dead Cell

Stain Kit for 633 or 635 nm excitation, stimulatory (MHC II) and co-stimulatory

(CD80/86) markers and assessed via flowcytometry. Supernatants were collected and

analyzed for IL-10 and IL-12p70 cytokine expression using BD OptEIA ELISA kits (BD

Biosciences) following the manufacturer’s instructions.

Antigen Specific Co-cultures

Dendritic cells were generated as mentioned above and treated with IDO-Gal3 in

the presence or absence of 1 mM 1-methyl-DL-tryptophan (Sigma-Aldrich). T cells were

isolated from OT-II splenocytes and labeled with 1 µM Carboxyfluorescein succinimidyl

110

ester (CFSE) according to previously described methods (Chapter 2). Cells were co-

cultured at a 1:10 DC to T cell ratio and proliferation assessed by CFSE dilution after 4

days via flow cytometry.

LPS Administration at the Hock as a Model of Inflammation

B6 mice were administered 2.2 µg of IDO-Gal3 in 40 µL of PBS subcutaneously

at the region of the hock and challenged with 2 ng/g of lipopolysaccharide (LPS) in 40

µL 1 d, 3 d, or 5 d post IDO-Gal3 treatment. 2 h after LPS administration animals were

euthanized and the injection site collected. Soft tissue was macroscopically separated

from bone and submerged in in RNAlater RNA Stabilization Reagent (Qiagen) in

preparation for qPCR.

Quantitative PCR

Soft tissues were homogenized and RNA purified using RNeasy Protect Mini Kit

(Qiagen). cDNA was synthesized from RNA using the High-Capacity cDNA Reverse

Transcriptase Kit (ThermoFisher) for use in qPCR in accordance with manufacturers

instructions. qPCR analysis was ran with primers specific for pro-inflammatory

cytokines (I12a, Il12b, Il1b, Ifng and Il6). Results are presented as the ratio of gene

expression to Gapdh expression determined by the relative quantification method.

Treatment groups were normalized to PBS only group.

Mass Spectrometry

2.2µg of IDO-Gal3 in 40 µL of PBS or 40 µL of PBS alone was injected into the

hock site of B6 mice (n=3). 30 min after injection, the mice were sacrificed, the hock and

tibia tissue regions excised and flash frozen using liquid nitrogen. These samples were

then submitted to the Southeast Center for Integrated Metabolomics at the University of

for mass spectrometric analysis of kynurenine and tryptophan levels.

111

Hock Infiltration

B6 mice were injected into the hock with IDO-Gal3 and 1 or 5 d later challenged

with LPS (ipsilateral and contralateral) to assess modulation of inflammation locally and

systemically. 2 h after LPS challenge animals were euthanized and the injection sites

harvested. Tissues were fixed with 10% Neutral Formalin pH 7.4 overnight. After

fixation, tissues were washed in diH2O and decalcified by storing in 10%

Ethylenediaminetetraacetic acid (EDTA) at 4°C for 3 weeks. Samples were assessed

every 2-3 days for stiffness and EDTA solution replenished. Tissues were submitted in

70% ethanol to the University of Florida Molecular Pathology Core for processing,

paraffin embedding, sectioning, mounting and staining with Haemotoxylin and Eosin

(H&E). Tissues were imaged using a Zeiss Axiovert 200M with a 20X objective lens

through the multidimensional acquisition module. 11 images were taken per tissue and

scored by two blinded independent individuals based on cellular infiltration (0: absent,

1:mild, 2:moderate, 3:severe) and epidermis hypertrophy (0:0-20 µm in thickness, 1: 21-

40 µm, 2: 41-60 µm, 3: 61µm or above).

In vivo Bioluminescence and Imaging

Bioluminescence images were acquired using an IVIS Spectrum In Vivo System

(IVIS). Living Image Software version 4.3.1 (Perkin Elmer, Waltham, MA) was used to

acquire the data immediately after furimazine administration. Exposure time for the

bioluminescence imaging was 1 second. Regions of interest were quantified as the

average radiance (photons/second/cm2/sr).

Mucosal Induction of Inflammation

6-week-old B6 mice were lavaged with 0.12% chlorhexidine gluconate for 3 d

followed by an infection consisting of an oral lavage with 2.5x109 Porphorymonas

112

gingivalis strain 381 and 2.5x109 Aggregatibacter actinomycetemcomitans strain 29522

resuspended in 2% low viscosity carboxy-methyl-cellulose on 4 consecutive d, for 5

straight weeks. Mice received a dose of IDO-Gal3 each week either prophylactically or

therapeutically during the development of periodontal disease.

Subgingival Cytokine Production

The local immunological soluble mediator milieu was evaluated using

multiplexing technology by Fernanda Rocha, DDS, PhD in the laboratory of Dr.

Shannon Wallet. Briefly, the mandible and maxillae were subjected to ‘bead beating’ in

a cell-lysis buffer containing protease inhibitors. The resulting homogenates were

analyzed for various soluble mediators using multiplex technology.

Results

IDO Activity and Gal3 Affinity Are Not Altered Upon Fusion

The enzymatic activity of IDO and the binding affinity of Gal3 were determined

for both fused and unfused proteins (Figure 4-1). Unfused IDO was able to generate 0.1

µg/mL of kynurenine whereas fusion protein, IDO-Gal3, produced 0.3 µg/mL of

kynurenine demonstrating comparable activity regardless of fusion state. Wild type Gal3

eluded from α-lactose-agarose column at 15% β-lactose whereas IDO-Gal3 eluted at

65% indicative of stronger affinity.

IDO Retains Immunomodulatory Properties When Fused to Galectin 3

Murine bone marrow derived dendritic cells were incubated with IDO-Gal3 at 2

nM for 24 h and their maturation status determined (Figure 4-2). Expression of

stimulatory (MHC II) and co-stimulatory (CD80/86) molecules was determined through

flow cytometry (presented as mean + SEM of three experiments, total n=9) (Fig. 4-2A).

The percent of untreated DCs expressing CD80, CD86 and MHC II was 13% + 0.3, 6%

113

+ 0.1 and 23% + 0.4, respectively. In contrast, upon LPS treatment expression of three

activation markers significantly increased (p < 0.05) to 30% + 1, 33% + 3, and 27% +

0.3 of cells. When DCs were treated with IDO-Gal3 for 24 h, maturation markers

remained comparable to immature or untreated cells with 11% + 0.4, 5% + 0.2, and

14% + 2 of cells expressing CD80, CD86 and MHC II, respectively. However, when DCs

were cultured with IDO-Gal3 followed by LPS challenge maturation markers were lower

than LPS alone, with 12% + 0.5, 5% + 0.2 and 19% + 0.6 expressing CD80, CD86 and

MHC II, confirming cells treated with IDO-Gal3 resist LPS-induced maturation. Next, the

release profile of inflammatory and anti-inflammatory cytokines, IL-12p70 and IL-10

were analyzed via ELISA (Fig. 4-2B). IL-12p70 secretion levels were established at 17 +

9 pg/mL and 219 + 20 pg/mL for untreated and LPS-treated cells, respectively. When

IDO-Gal3 was introduced to the DC culture, IL-12p70 levels were determined at 12 + 5

pg/mL. Remarkably, when DCs were treated with IDO-Gal3 and challenged with LPS,

IL-12p70 levels were established at 3 + 1 pg/mL, comparable to untreated group.

Conversely, IL-10 production remained consistent, with no significant changes observed

between groups.

To evaluate whether IDO-Gal3 treated DCs can attenuate antigen specific CD4+

T cell proliferation, CFSE labeled T cells isolated from splenocytes of OT-II transgenic

mice were co-cultured with treated DCs. T cells from these mice proliferate in response

to OVA peptide 323-339 (discussed in detail in Chapter 2). The data shown in Figure 4-

3 represent the percent proliferation of viable CD4+ T cells stimulated with IDO-Gal3-

treated DCs. In co-cultures where only DCs and T cells were present, or when IDO-

Gal3 was introduced, minimal proliferation was observed at 33% + 0.9 and 35% + 1,

114

respectively. When DCs were pulsed with OVA323-339 and co-cultured with T cells, T

cells became activated and proliferation measured at 95% + 1 after 4 d. However, when

DCs were cultured with IDO-Gal3 for 24 h, washed, pulsed with OVA323-339, and co-

cultured with T cells, proliferation was reduced to basal levels measured at 33% + 2. T

cells only were also cultured for 4 d and included as a negative control with proliferation

at 5% + 0.7. (Fig. 4-3A) The experiment was repeated, with the introduction of the IDO

inhibitor MT as a DC treatment (Fig. 4-3B). In the presence of MT-treated DCs, CD4+ T

cell proliferation was restored to 58% + 2, indicating the active enzyme is required for

the attenuation of proliferation. A representative flow cytometry histogram for

proliferation of T cells stimulated with IDO-Gal3 treated DCs or IDO-Gal3 and MT

treated DCs is shown in Fig. 4-2C. Herein, we demonstrate fused IDO maintains its

metabolic reprogramming potential comparable to unfused enzyme as seen in Chapter

2.

IDO-Gal3 Reduces Inflammatory Cytokine Gene Expression at Various Time Points.

B6 mice were subcutaneously injected with IDO-Gal3 at the hock of the left hind

limb. 1 d, 3 d or 5 d after treatment mice were challenged with LPS as a model of local

inflammation and gene expression of pro-inflammatory cytokines (IL-12p35, IL-12p40,

IL-1β, IFN-γ, and IL-6) analyzed through qPCR. The data presented in Figure 4-4 is

representative of 5 animals per group normalized to cohort that received a PBS sham

injection. A schematic representation of the experimental schedule is provided in Fig. 4-

3A. Groups that showed no significant contribution to the modulation of inflammation at

early time points were dropped from further analysis. Treatment with LPS significantly

increased compared to PBS sham for IL-12p35 (4-fold), IL-12p40 (28-fold), IL-1β (2860-

115

fold), IFNγ (860-fold) and IL-6 (25400-fold). Treatment with untargeted IDO did not

increase IL-12p35 gene expression compared to sham PBS but increased IL-12p40

(14-fold), IL-1β (420-fold), INFγ (185-fold) and IL-6 (5556-fold). Challenge of untargeted

IDO with LPS injection did not increase IL-12p35 gene expression when compared to

sham but increased IL-12p40 (17-fold), IL-1β (1370-fold), IFNγ (370-fold) and IL-6

(15600-fold). Treatment with IDO-Gal3 only decreased IL-12p35 gene expression when

compared to sham (0.5-fold), and increased IL-12p40 (15-fold), IL-1β (1453-fold), IFNγ

(178-fold) and IL-6 (1400-fold). Remarkably, treatment with IDO-Gal3 challenged with

LPS 1 d later, decreased IL-12p35 expression compared to sham injection (0.2-fold),

and increased IL-12p40 (16-fold), IL-1β (1370-fold), IFNγ (156-fold) and IL-6 (6757-

fold). A different cohort of animals was again treated with IDO-Gal3 at D1, but the LPS

challenged was administered 3 d after (Fig. 4-3C). Similarly to 1 d data, large fold

changed were observed for all pro-inflammatory cytokines measured when mice were

treated with LPS only as a positive control. Following treatment with IDO-Gal3 at D1,

and LPS challenge at D3, all cytokines significantly decreased compared to LPS only

group. Lastly, a third cohort was administered IDO-Gal3 at D1 and challenged with LPS

at D5. Again, mice that received LPS only injections showed significant increase in

proinflammatory cytokine gene expression whereas mice that were prophylactically

treated with IDO-Gal3 and challenged with LPS showed a decreased in cytokine gene

expression when compared to LPS only group. Prophylactic treatment with IDO-Gal3

decreases LPS-mediated inflammatory cytokine gene expression at the hock.

IDO-Gal3 Modulates Local Tryptophan Catabolism

IDO-Gal3 or PBS was injected into the hock of B6 mice. Either 30 mins or 24 h

after injections animals were euthanized, the injection site and adjacent tissues excised

116

and flash frozen for mass spectrometry analysis of kynurenine and tryptophan levels

(Figure 4-5). 30 min after treatment (PBS) the kynurenine levels at the hock were 55 +

15 ng/g of protein while levels at the tibia were 48 + 4 ng/g of protein. For animals

injected with IDO-Gal3, kynurenine levels increased at the hock (1534 + 400 ng/g) and

the tibia (273 + 83 ng/g (Fig. 4-5A)). For the same cohort of mice, the tryptophan levels

in the PBS group were 3627 + 2000 ng/g of protein at the hock and 1714 + 400 ng/g

protein at the tibia. For the IDO-Gal3 injected groups, at the hock tryptophan levels were

4416 + 680 ng/g of protein and 2134 + 60 ng/g of protein at the region of the tibia (Fig.

4-5B). At 24 h post treatment, animals treated with PBS showed kynurenine levels at 14

+ 5 ng/g of protein at the hock, 13 + 5 ng/g of protein at the region of the tibia, and 23 +

9 ng/g of protein at the draining (popliteal) lymph node. When animals were injected

with IDO-Gal3, kynurenine levels were measured at 76 + 30 ng/g of protein at the hock,

73 + 30 ng/g of protein at the tibia and 57 + 25 ng/g of protein at the popliteal lymph

node (Figure 4-5C). Lastly, tryptophan levels were measured 24 h post treatment.

Tryptophan levels were established at 2153 + 929 ng/g of protein at the hock, 1608 +

900 ng/g of protein at the tibia, and 2609 + 600 ng/g of protein at the popliteal lymph

node. The IDO-Gal3 group showed Tryptophan levels for the IDO-Gal3 injected group

were 2244 + 700 ng/g of protein at the hock, 1884 + 600 ng/g of protein at the tibia and

1903 + 600 ng/g of protein at the popliteal lymph node (Fig. 4-5D).

IDO-Gal3 Modulates LPS-induced Inflammation Locally

To test the efficacy of the localization strategy in combating inflammation, B6

mice were injected with IDO-Gal3 or relevant controls as before and challenged with

LPS 1 d or 5 d after, in the ipsilateral (previously treated hind limb hock) and

contralateral (untreated opposite hind limb) site. 2 h post LPS challenge, animals were

117

euthanized and injection site excised (Figure 4-6). A schematic representation of the

injection schedule is shown in Fig. 4-6A. H&E stained tissues were scored based on

cellular infiltration and epidermal hypertrophy (enlargement of epidermis layer) following

LPS challenge either 1 d or 5 d post-treatment (Fig. 4-6B-E). Animals that were

administered PBS on both sides and received no LPS challenged showed low cellular

infiltration and average epidermal thickness (Fig 4-6panel). Animals that received PBS

injection followed by LPS challenge on both sides, demonstrated high cellular infiltration

and increased epidermal thickness (Fig 4-6panel). Animals that were treated with

untargeted IDO and received no LPS challenged showed characteristics comparable to

PBS group. When animals were treated with untargeted IDO and challenged with LPS,

H&E slides show cellular infiltration and epidermal hypertrophy similar to LPS group.

When IDO-Gal3 was administered with no LPS challenge, tissue resembled the PBS

only group (Fig 4-6panel). Remarkably, when IDO-Gal3 was administered and

challenged with LPS ipsilaterally, cellular infiltration and epidermis hypertrophy were

significantly reduced. Conversely, when LPS was administered contralaterally to IDO-

Gal3 treatment, tissue resembled the LPS only group with significant inflammation (Fig.

4-6F).

Gal3 Provides Retention of a Model Enzyme at Subgingival Tissue, Retention Time Is Not Altered by Infection State

NanoLuc or NanoLuc-Gal3 was administered to B6 mice subgingivally on 1 d

followed by daily administration of furimazine. Mice were sedated and imaged daily via

IVIS until no signal was detected for two consecutive days (Figure 4-7). NanoLuc alone

cleared from the injection site within 48 h. NanoLuc-Gal3 however, remained at the

injection site with detectable levels for 10 d. Quantification of signal obtained for both

118

proteins is shown as the average radiance (p/s/cm2/sr) over 10 d (Fig. 4-7A).

Representative images from animals injected with NanoLuc (left panel) and NanoLuc-

Gal3 (right panel) are shown in Fig. 4-7B. Furthermore, the retention time of NanoLuc-

Gal3 was evaluated in tissues with varying levels of infection (Fig. 4-7C). NanoLuc-Gal3

was administered to uninfected (black line), acutely infected (blue), or chronically

infected (red line) animals. No significant differences were observed.

IDO-Gal3 Suppresses Inflammatory Cytokine and Chemokine Production

IDO-Gal3 was administered to B6 mice, prophylactically or therapeutically to

polymicrobial infection (Figure 4-8), and the soluble mediators in the soft tissue

analyzed (Figure 4-9). Infected animals showed consistently high expression of IL-1β

(72 + 2 ρg/mL), IL-12p70 (26 + 1 ρg/mL), IL-33 (2505 + 271 ρg/mL), KC (101 + 9

ρg/mL), and IL-6 (24 + 1 ρg/mL) cytokines (Fig. 4-9A). Levels of IL-10 for infected group

were established at 8 + 0.7 ρg/mL. For animals treated prophylactically, IL-1β (66 + 1

ρg/mL), IL-12p70 (21 + 0.6 ρg/mL), IL-33 (1159 + 65 ρg/mL), KC (75 + 3 ρg/mL), and

IL-6 (21 + 0.4 ρg/mL) secretion levels significantly decreased, While IL-10 levels were

detected at 16 + 0.7 ρg/mL. When animals were treated therapeutically, significant

reduction was observed in the production of IL-1β (60 + 1 ρg/mL), IL-12p70 (13 + 0.8

ρg/mL), IL-33 (908 + 40 ρg/mL), KC (59 + 2 ρg/mL), and IL-6 (14 + 0.8 ρg/mL) and an

increase in IL-10 (23 + 0.8 ρg/mL) production. Uninfected animals were also treated

with IDO-Gal3 to determine any inflammation associated with administration of foreign

protein, IL-1β (56 + 0.5 ρg/mL), IL-12p70 (7 + 1 ρg/mL), IL-33 (752 + 29 ρg/mL), KC (51

+ 2 ρg/mL), IL-6 (8 + 0.7 ρg/mL), IL-10 (31 + 1 ρg/mL), levels were measured and

determined to not have major contributions to inflammatory response. Lastly, uninfected

animals were evaluated and levels of of IL-1β (51 + 1 ρg/mL), IL-12p70 (5 + 0.6 ρg/mL),

119

IL-33 (462 + 98 ρg/mL), KC (35 + 3 ρg/mL), IL-6 (5 + 0.6 ρg/mL) and IL-10 (25 + 0.6

ρg/mL) cytokines established. A similar trend was observed when chemokines (IP10,

MCP1 and MIP2) were evaluated (Fig. 4-9B) where statistically significant differences in

MCP1 and MIP2 were observed in animals treated therapeutically vs prophylactically.

Herein, we demonstrated treatment with IDO-Gal3 ameliorates local inflammation either

in an LPS-induced subcutaneous model of inflammation or in a polymicrobial murine

model of periodontal disease by decreasing cytokine gene expression and production

subsequently preventing cellular infiltration of the affected tissue.

Discussion

Inflammation is the protective reaction orchestrated by the immune system in

response to infection and/or injury241. Local and systemic inflammatory responses aim

to eliminate the inciting stimulus, promote tissue repair and healing, and in the case of

infection, establish immune memory such that the host mounts a faster response upon

repeated insult242. However, complications arise when the inflammatory response is

chronic and the balance between protective and destructive immunity favors

destruction. The inflammatory response is a complex but highly coordinated sequence

of events, involving molecular, cellular and physiological alterations. It begins with the

production of soluble mediators (e.g. complement, cytokines and chemokines) by

resident cells, including macrophages and DCs, in the injured or infected tissue. At the

same time, cell adhesion molecules are upregulated on circulating leukocytes and

endothelial cells, promoting the exudation of proteins and influx of granulocytes from the

blood243.This well-characterized phase of the inflammatory response is routinely

targeted using drugs such as non-steroidal anti-inflammatory drugs (NSAIDs)244-249 and

pro-inflammatory cytokine-blocking antibodies29,53,250-252. These drugs currently lead the

120

industry for the treatment of inflammatory diseases253. However, strategies that achieve

sustained, localized efficacy are still needed. In the present work, a fusion chimera,

IDO-Gal3, was developed, characterized and employed to suppress inflammation by

reprogramming local metabolism. This study demonstrates Gal3 fusion confers oral

submucosal retention for 10 days and IDO-Gal3 provides robust suppression of

inflammation, both in vitro and in vivo.

IDO-Gal3 was first characterized to guarantee no significant changes occurred

during fusion of both proteins. Previous published work by others as well as our analysis

of NanoLuc-Gal3 have demonstrated that fusion of a model enzyme at the N-terminus

has no effect on the carbohydrate affinity profile of Gal3194. Indeed, IDO-Gal3 bound an

α-lactose agarose column more tightly than wild type Gal3. Further studies will

investigate any conformational changes that may be responsible for this favorable

increased affinity. Chapter 2 established IDO as an immunomodulator in the

extracellular space by suppressing DC maturation and subsequent T cell responses.

The IDO-Gal3 fusion maintained the immunomodulatory capacity of unfused IDO. DCs

treated with exogenous IDO-Gal3 did not upregulate expression levels of stimulatory

and co-stimulatory molecules, exhibiting diminished IL-12p70 secretion while

maintaining IL-10 production. This study further solidifies the notion that IDO enacts an

anti-inflammatory response in the extracellular environment.

Following in vitro characterization, mice were treated with IDO-Gal3 at the hock

and challenged with LPS either 1 d, 3 d or 5 d post-treatment. The hock is a suitable

alternative to the more common footpad injection, and minimizes animal distress254.

Moreover, LPS-induced inflammation serves as a proof of concept platform and

121

establishes the fusion protein as a mediator of multiple inflammatory conditions.

Analysis of the injection site revealed IDO-Gal3 modulated innate and adaptive

immunity as determined by the low-expression of all pro-inflammatory cytokines tested.

IL-6 responds to immediate tissue damage while IL-12, expressed by macrophages and

DCs, works synergistically with IL-1β, produced by neutrophils, to regulate T cell

production of IFNγ and differentiation into Th1 T cells, key players in autoimmune

diseases such as type 1 diabetes, rheumatoid arthritis and psoriasis. Treatment with

IDO-Gal3 before an insult lessens potential for auto inflammatory damage and

establishes Gal3 as a retention strategy at the site of inflammation. Additionally, given

the complexity of the inflammatory milieu, technologies that simply target one soluble

mediator are often inefficient. Demonstrated here, IDO-Gal3 can alter genetic

expression of multiple soluble mediators involved in immunological responses with a

single dose.

As gene expression does not always translate to protein production and

subsequent cellular changes255, we performed experiments to verify that administration

of IDO-Gal3 was ideal for application in disease models of microbial inflammation. Upon

histological analysis of relevant tissues, it was determined IDO-Gal3 halts LPS-induced

inflammation by inhibiting cellular infiltration of the affected tissue. Anti-inflammatory

effects can only be observed in the tissue treated with IDO-Gal3 (ipsilateral to injection

site), indicating treatment is local and not systemic. Localized suppression is crucial for

decreasing susceptibility of the host to opportunistic infections, abnormal course of

disease and increased risk of lymphomas and cancers256 often observed with systemic

immunosuppression.

122

Periodontal disease (PD) is an ideal candidate for application of our novel fusion

protein. The initial response to polymicrobial insult, characteristic of PD, is an acute

inflammatory response with increased vascular dilation and blood flow. Then, an

increase in neutrophil and macrophage migration toward the lesion, due to chemokines

such as IP10, MCP1 and MIP2, originating from the microbial cells as well as host-

derived inflammatory mediators is observed. These cells then release pro-inflammatory

cytokines, IL-1β, IL-12 and IL-6, and KC (IL-8 murine homologue), at the site of injury

and recruit members of adaptive immunity. The character and intensity of the

inflammatory response determines whether the initial lesion resolves or becomes

chronic, leading to PD. Overproduction of IL-33, for example, has been shown to

significantly exacerbate periodontitis in mice leading to alveolar bone loss257.

In this study, Gal3 was established as a retention strategy in the subgingival

space. Studies using NanoLuc-Gal3 demonstrated a retention time of 10 d was

observed with no differences between healthy and infected tissues. Furthermore, the

state of disease did not alter retention time mediated by Gal3. These findings are of

extreme importance for the treatment of PD as the state of inflammation varies with the

patient. A larger clinical impact can be appreciated when treatment can be used across

an entire population.

Surprisingly, when compared to infected groups, or prophylactically treated

animals, subjects that received IDO-Gal3 therapeutically showed significantly less

production of pro-inflammatory cytokines and chemokines and an increase in the anti-

inflammatory cytokine, IL-10. Work with NanoLuc-Gal3 revealed a decrease in enzyme

activity over time. Under the assumption that IDO-Gal3 behaves similarly, animals

123

treated prophylactically received a decreased effective dose resulting in higher

production of pro-inflammatory cytokines. Further studies will need to be conducted to

determine the specific mechanism by which IDO-Gal3 is able to halt inflammation in a

polymicrobial model of periodontal disease.

124

Figure 4-1. Enzymatic activity of IDO is not altered upon fusion with Galectin 3, Galectin

3 retains binding affinity to sugar moiety upon fusion with IDO at N-terminus. A) Equal molar quantities of IDO-Gal3 and IDO were incubated with tryptophan and its conversion to kynurenine measured by addition of Ehrlich reagent. IDO-Gal3 shows comparable production of kynurenine to unfused IDO. B) Equal concentration of IDO-Gal3 (red line) and wild type Gal3 (black line) were injected into an α-lactose-agarose resin column and eluded with an increasing gradient of β-lactose. Fusion protein elutes at a higher concentration of β-lactose indicative of stronger binding affinity.

125

Figure 4-2. Indoleamine 2,3-dioxygenase maintains an immature dendritic cell

phenotype when fused to Galectin 3. Murine bone marrow derived dendritic cells were treated with IDO-Gal3 for 24 h, washed and and challenged with LPS overnight. A) Cells were co-stained for viability, CD11c and maturation markers: CD80, CD86 and MHC II and analyzed through flow cytometry. IDO-Gal3-treated dendritic cells resist LPS-induced maturation. B) Supernatants were collected and analyzed for the secretion of IL-12p70 and IL-10 through ELISA. Dendritic cells treated with IDO-Gal3 do not release IL-12p70 even when stimulated with LPS and maintain a steady IL-10 secretion. Values shown as percent positive. n =3, mean + SEM with pair-wise significant difference from all other groups, ANOVA with Tukey’s post-hoc, denoted by * where p < 0.05.

126

Figure 4-3. IDO-Gal3-treated DCs attenuate antigen specific T cell proliferation,

suppression is active enzyme dependant. Murine bone marrow derived DCs were cultured with IDO-Gal3 for 24 h, washed and pulsed with OVA peptide. OVA specific T cells were isolated from transgenic OT-II mice, labeled with CFSE and co-cultured with DCs for 4 d. A) Proliferation of viable CD4+ was assessed via CFSE dilution. IDO-Gal3-treated DCs attenuate antigen-specific T cell proliferation to levels comparable to no-antigen control. B) DCs were cultured with IDO-Gal3 in the presence or absence of a potent IDO inhibitor, 1-methyl tryptophan (MT), and co-cultured with T cells as described above. When IDO activity is inhibited T cell proliferation is re-stored to levels compared to antigen control. C) Representative flow plots of T cell proliferation with IDO-Gal3-treated DCs (left panel) and T cell proliferation with IDO-Gal3 and methyl-tryptophan-treated DCs (right panel). Values shown as mean + SEM, pair-wise significant difference (by ANOVA and Tukey’s significant test) is denoted by * where p < 0.05 for n=3.

127

Figure 4-4. IDO-Gal3 blocks subcutaneous LPS-induced inflammatory cytokine gene

expression. IDO-Gal3 or controls were injected in the hock of 8-week-old C57BL/6 female mice. A) Schematic representation of injection schedule. B) On day 1, C) day 3 and D) day 5, subcutaneous hock injection of LPS was administered as a local inflammatory challenge. Two hours post LPS challenge, soft tissue at the injection site was harvested, and inflammatory gene expression analyzed through real-time qPCR. Pair-wise significant difference (by ANOVA and Tukey’s post hoc) is denoted by * where p < 0.05; n=5 mice per group. IDO-Gal3 modulates inflammation subcutaneously at the hock by halting gene expression of inflammatory cytokines for at least 5 d. Values shown are normalized to PBS group. n =5, mean + SEM with pair-wise significant difference from all other groups, ANOVA with Tukey’s post-hoc, denoted by * where p < 0.05.

128

Figure 4-5. IDO-Gal3 modulates tryptophan metabolism at the injection site. IDO-Gal3

or PBS was administered subcutaneously at the hock, the injection site and adjacent tissues excised, flash frozen and submitted to the Biomedical Mass Spectrometry Core for analysis of kynurenine and tryptophan levels. A) 30 min post IDO-Gal3 treatment high amounts of kynurenine accumulate at the hock with minimal detection at the region of the tibia. B) Tryptophan levels at the hock and tibia do not change between groups, C) 24 h post treatment kynurenine levels are elevated compared to PBS group indicative of active IDO-Gal3 and metabolic reprogramming. D) Tryptophan levels between groups and tissues does not change. IDO-Gal3 promotes tryptophan metabolism and induces kynurenine production, but kynurenine likely diffuses away from site of injection making difference undetectable at 24 h. Values shown as percent positive. n =2, mean + SEM with significant difference from IDO-Gal3, t-test, denoted by * where p < 0.05, and significant difference from hock region, ANOVA, denote by + where p < 0.05.

129

Figure 4-6. IDO-Gal3 blocks subcutaneous LPS-induced inflammatory response,

suppression is not systemic. IDO-Gal3, or relevant controls, were injected into the hock of 8-week-old C57BL/6 female mice. On day 1 and day 5, a subcutaneous hock injection of LPS was administered as a local inflammatory challenge ipsilateral or contralateral to treatment. A) Schematic representation of injection schedule and experimental design. 2 h post LPS administration, the injection site was excised, demineralized, paraffin embedded and stained with Hematoxylin and Eosin (H&E). Sections were scored (0-3) based on B) cellular infiltration on day 1 or C) epidermis hypertrophy on day 1 AND D) cellular infiltration on day 5 or E) epidermis hypertrophy on day 5. F) Representative H&E images of injection sites IDO-Gal3 halts inflammation subcutaneously at the hock by preventing cellular infiltration locally (scale bar = 50 µm). n =5, mean + SEM with pair-wise significance (ANOVA with Tukey’s post hoc) denoted by * when compared to contralateral side, by + when compared to PBS + LPS ipsilateral side, and ^ when compared to PBS + LPS contralateral side.

130

Figure 4-7. Galectin 3 provides retention of a model enzyme in the subgingival space;

retention profile is not altered by disease state. NanoLuc or NanoLuc-Gal3 was administered to 8-week-old C57BL/6 female mice in the subgingival space. At specified times, furimazine was injected and animals imaged using in vivo imaging system (IVIS) A) Maximum relative luminescence was quantified over time. B) Representative images of NanoLuc (top row) and NanoLuc-Gal3 (bottom row) over time. C) NanoLuc-Gal3 administration into the subgingival space over time for no-infection (black line) early infection (blue line) and late infection (red line) cohorts. Galectin 3 provides retention of a model enzyme in the subgingival space for at least 10 days and retention profile is not altered by disease state. Values shown as percent positive. n =5, mean + SEM with pair-wise significant difference from all other groups, ANOVA with Tukey’s post-hoc.

131

Figure 4-8. Schematic representation of infection model used to induce periodontal

disease in mice. Three different infection models were used to study IDO-Gal3-mediated modulation of periodontal disease. A) C57BL/6 mice were lavaged with equal amounts of porphorymonas 381 and aggregatibacter actinomycetmcomitans 29522 for 4 consecutive days followed by 3 days of no infection. B) Mice were prophylactically treated with IDO-Gal3 on day 1, followed by 4 consecutive days of infection and 2 days of no infection. C) Mice were infected for 4 consecutive days, treated therapeutically on day 5 and allowed to recover for 2 days. All models were carried out for 5 consecutive weeks. Non-infected, healthy mice were used as negative control.

132

Figure 4-9. Inflammatory cytokine and chemokine production in the subgingival space is

significantly reduced by administration of IDO-Gal3 both prophylactically and therapeutically. C57BL/6 mice were either infected with porphorymonas 381 and aggregatibacter actinomycetmcomitans 29522 (Infect) and treated with IDO-Gal3 prophylactically (IDO-Gal3-P) or therapeutically (IDO-Gal3-T); treated without presence of infection (IDO-Gal3-U) or left uninfected and untreated (unifect). After the 5 week study, animals were euthanized, mandibles and maxillae extracted and subjected to bead beating. The resulting homogenates were analyzed for A) cytokines and B) chemokines. * p value < 0.05 as indicated, ^ p value <0.05 IDO-Gal3-P vs IDO-Gal3-T, + p value < 0.05 IDO-Gal3-U vs uninfected as determined by oneway ANOVA with Bonferoni’s correction, n=7. Administration of IDO-Gal3 therapeutically significantly halts pro-inflammatory cytokine production in a polymicrobial model of periodontal disease.

133

CHAPTER 5 CONCLUSIONS AND FUTURE DIRECTIONS

Enzyme therapeutics hold great promise for the treatment of inflammatory

conditions as they offer multiple advantages over small molecules. Enzymes serve a

highly specific and complex set of functions that cannot be mimicked by single chemical

compounds. Additionally, since the action of enzymes is so specific, there is less

potential for deleterious effects or interference with biological processes if administered

locally. The focused of this thesis was the development of a chimera protein with a

tissue-binding moiety fused to the enzyme indoleamine 2,3 dioxygenase (IDO) to modify

the immune microenvironment and ameliorate local inflammation.

The first set of studies conducted here established IDO as an

immunomodulator in the extracellular space. To date, therapeutic application of IDO has

primary focused on overexpression of the gene for transplant tolerance. However,

techniques used to induce IDO gene expression, such as treatment of APCs with IFNγ

or LPS and gene transfection have demonstrated harmful effects. Additionally, the cost

associated with gene therapy or ex vivo approaches prevent their worldwide application.

Herein, we demonstrate extracellular IDO prevents DC maturation in the presence of a

potent inflammatory stimuli, and subsequently suppress antigen specific T cell

proliferation. Although the effects of IDO on T cells is well known, future studies will

determine the specific mechanism by which proliferation is halted. DCs can induce

anergy or aid in the differentiation of naïve T cells into regulatory T cells (Tregs) causing

suppression of anti-inflammatory responses. Additionally, the role of the aryl

hydrocarbon receptor (AhR) and its activation through generated kynurenines should be

further explored. Here, we hypothesize the catabolism of tryptophan in the extracellular

134

space by IDO yield kynurenines that are transported into the cell by the Large

Amino Acid Transporter 1 (LAT-1) and bind the AhR inducing translocation into the

nucleus and binding to the dioxin-responsive enhancer (DRE) for the synthesis of

intracellular IDO. This potential mechanism is shown in Figure 5-1.

IDO was determined to be most efficacious when administered in the active form

which limits its delivery through commonly used biomaterials platforms such as particles

or hydrogels due to the harsh organic solvents used during the fabrication processes

and IDO’s susceptibility to denaturation. Additionally, it is well known bolus

administration of proteins often result in clearance through the renal system before any

therapeutic effect can be obtained. Therefore, a fusion protein, with Galectin 3 (Gal3)

was developed and characterized with Gal3 motif serving as a tissue-retention strategy.

As a complimentary reporter fusion, NanoLuciferase-Galectin 3 (NanoLuc-Gal3) was

also produced. Nanoluciferase reacts with its substrate, furimazine, to produce light

easily detectable during in vitro and in vivo assays. Characterization of the fusion

construct revealed Gal3 retains binding affinity to sugar moiety when fused through the

N-terminus but its biological activity is disrupted, likely due to its inability to oligomerize

into a higher-ordered structure. Furthermore, we established NanoLuc-Gal3 can bind

immune cells without triggering a DC-mediated inflammatory response. Retention was

significantly improved at multiple injection sites with renal filtration as a potential

clearance path for excess or unbound fusion administered. Future studies will

investigate clearance mechanism for sites other than the hock and processes in vivo

leading to protein degradation and inactivation, such as proteolytic degradation and

immunogenicity.

135

Upon establishment of Galectin 3 as a tissue-retention strategy it was fused with

IDO, IDO-Gal3, and their immunomodulatory and binding properties combined to target

inflammatory conditions. Using an LPS-induced subcutaneous inflammation model,

IDO-Gal3 was shown to inhibit cellular infiltration by decreased cytokine gene

expression at a localized tissue. Anti-inflammatory effects were observed for 5 days

post treatment at a single high dose. A proposed mechanism of action is depicted in

Figures 5-1. LPS binds to the toll-like receptor 4 (TLR4), resulting in conformational

changes and recruitment of the Myeloid Differentiation Primary Response Gene 88

(MyD88) and TIR Domain-Containing Adaptor Protein (TIRAP). The MyD88-TIRAP

complex regulates early nuclear factor kappa-light-chain-enhancer of activated B cell

(NF-κβ) activation and regulation of proinflammatory cytokines such as IL-12 and IL-1β

and anti-inflammatory cytokines such as IL-10. This is through the activation of the IL-1

Receptor-Associated Kinases (IRAKs) and the adaptor molecules TNF Receptor-

Associated Factor 6 (TRAF6). TRAF6 induces the activation of Transforming growth

factor-β-Activated Kinase 1 (TAK1) that leads to the activation of p38 Mitogen-Activated

Protein Kinase (p38 MAPK) and IκB Kinase (IΚΚ). IΚΚ signaling pathway leads to the

induction of the transcription factor NF-κβ whereas p38 leads to the activation of cAMP

response element binding protein (CREB). Figure 5-2 demonstrates the cytokine

network proposed in vivo and all the different cellular interactions IDO is believed to

alter. An initial injury is produced which results in the release of MCP1 and IP10 by

epithelial cells recruiting monocytes and NK cells. Monocytes then release MIP2 which

recruits polymorphonuclear leukocytes initiating the innate response. NK cells on the

other hand, release IFN-γ recruiting DCs and macrophages which play a crucial role in

136

the initiation of the adaptive immune system. IDO likely acts on NK cells in vivo given

the inflammatory signal is only provided for 2 h. Once IFN-γ production is diminished by

IDO, DCs do not release IL-12 halting the differentiation of naïve T cells into Th1 cells.

Furthermore, without the production of IL-1β, IL-6 or TNFα, Th17 or Th22 differentiation

is also not possible limiting the inflammation observed in the tissues evaluated. Future

work should focus on elucidating the specific mechanism and cytokine networks

involved in the suppression of inflammation.

Furthermore, work will be performed to determine minimal dose required to

induce desired conditions at early (1-5 d) and late time points (10-14 d). Additionally,

immunostaining of histological samples will be performed to correlate tissue retention

time and distribution of IDO-Gal3 with that of NanoLuc-Gal3. Lastly, antigenicity toward

the fusion protein should be evaluated as it might inhibit longevity and efficacy in vivo.

In terms of application to clinically relevant models, in this thesis IDO-Gal3 was

employed to halt inflammation in a polymicrobial model of periodontal disease.

NanoLuc-Gal3 confers an oral submucosal retention time of 10 d. Therapeutic

administration of IDO-Gal3 halts inflammation by decreasing the production of

proinflammatory cytokines and chemokines. Future studies will stain histological

samples with H&E to determine degree of cellular infiltration and correlate mechanism

of action to LPS-induced inflammation model. Furthermore, bone density analysis will

demonstrate whether treatment with IDO-Gal3 can prevent alveolar bone loss, a critical

component of new PD treatments.

137

Moving forward we hope to utilize the platformed engineered here for the

treatment of additional inflammatory conditions. We hope to introduce IDO-Gal3 into a

rat model of osteoarthritis for the modulation of inflammation in the intra articular space.

Efforts have already been dedicated to this space and an active collaboration with Dr.

Kyle Allen has shown Gal3 acts as a retention motif in an arthritic joint. Additionally, we

would like to test the efficacy of IDO-Gal3 in a murine model of psoriasis. NanoLuc-Gal3

administered subcutaneously remained at the injection site far longer than any of the

other spaces investigated (IP, IM). Psoriasis is a skin related autoimmune disorder with

cytokine release profiles shown here to be suppressed.

The work presented in this thesis shows IDO maintains its immunomodulatory

properties when administered to the extracellular space and can be retained at the site

of injection through fusion with Gal3 for 10 d. Subcutaneous and oral submucosal

administration of IDO-Gal3 halted inflammation by altering release profile of pro-

inflammatory cytokines preventing cellular infiltration.

138

Figure 5-1. Proposed mechanism for IDO-related suppression of inflammation by blockage of NF-κβ pathway initiated through binding of LPS to TLR4 on surface of immune cell.

139

Figure 5-2. Proposed in vivo cytokine network affected by the introduction of IDO. IDO likely primarily affects NK cells in vivo which halts initiation of adaptive immune system by cross-talk with DCs.

140

LIST OF REFERENCES

1 Khan, T. A. & Reddy, S. T. Immunological principles regulating

immunomodulation with biomaterials. Acta Biomaterialia 10, 1720-1727, doi:http://dx.doi.org/10.1016/j.actbio.2013.12.011 (2014).

2 Hlavaty, K. A., Luo, X., Shea, L. D. & Miller, S. D. Cellular and molecular

targeting for nanotherapeutics in transplantation tolerance. Clinical Immunology 160, 14-23, doi:http://dx.doi.org/10.1016/j.clim.2015.03.013 (2015).

3 Page, E. K., Dar, W. A. & Knechtle, S. J. Tolerogenic therapies in transplantation.

Front Immunol 3, 198, doi:10.3389/fimmu.2012.00198 (2012). 4 Bracho-Sanchez, E., Lewis, J. S. & Keselowsky, B. G. in Biomaterials in

Regenerative Medicine and the Immune System (ed Laura Santambrogio) Ch. 8, 139-156 (Springer International Publishing, 2015).

5 Lewis, J. S., Roy, K. & Keselowksy, B. G. Vol. 39 25-34 (Cambridge University

Press, MRS Bulletin, 2014). 6 Hippen, K. L. et al. Massive ex vivo expansion of human natural regulatory T

cells (T(regs)) with minimal loss of in vivo functional activity. Sci Transl Med 3, 83ra41, doi:10.1126/scitranslmed.3001809 (2011).

7 Clemente-Casares, X. et al. Expanding antigen-specific regulatory networks to

treat autoimmunity. Nature 530, 434-440, doi:10.1038/nature16962 (2016). 8 Klatzmann, D. & Abbas, A. K. The promise of low-dose interleukin-2 therapy for

autoimmune and inflammatory diseases. Nat Rev Immunol 15, 283-294, doi:10.1038/nri3823 http://www.nature.com/nri/journal/v15/n5/abs/nri3823.html#supplementary-information (2015).

9 Steenblock, E. R., Fadel, T., Labowsky, M., Pober, J. S. & Fahmy, T. M. An artificial antigen-presenting cell with paracrine delivery of IL-2 impacts the magnitude and direction of the T cell response. J Biol Chem 286, 34883-34892, doi:10.1074/jbc.M111.276329 (2011).

10 Keselowsky, B. G., Xia, C. Q. & Clare-Salzler, M. Multifunctional dendritic cell-

targeting polymeric microparticles: engineering new vaccines for type 1 diabetes. Hum Vaccin 7, 37-44 (2011).

141

11 Butler, K. S. et al. Development of antibody-tagged nanoparticles for detection of transplant rejection using biomagnetic sensors. Cell Transplant 22, 1943-1954, doi:10.3727/096368912X657963 (2013).

12 Penno, E., Johnsson, C., Johansson, L. & Ahlström, H. Macrophage uptake of ultra-small iron oxide particles for magnetic resonance imaging in experimental acute cardiac transplant rejection. Acta Radiol 47, 264-271 (2006).

13 Alcantar, N. A., Aydil, E. S. & Israelachvili, J. N. Polyethylene glycol-coated

biocompatible surfaces. J Biomed Mater Res 51, 343-351 (2000). 14 Azzi, J. et al. Polylactide-cyclosporin A nanoparticles for targeted

immunosuppression. FASEB J 24, 3927-3938, doi:10.1096/fj.10-154690 (2010). 15 Azzi, J. et al. Targeted Delivery of Immunomodulators to Lymph Nodes. Cell

Rep, doi:10.1016/j.celrep.2016.04.007 (2016). 16 Shirali, A. C. et al. Nanoparticle delivery of mycophenolic acid upregulates PD-L1

on dendritic cells to prolong murine allograft survival. Am J Transplant 11, 2582-2592, doi:10.1111/j.1600-6143.2011.03725.x (2011).

17 Hlavaty, K. A. et al. Tolerance induction using nanoparticles bearing HY peptides

in bone marrow transplantation. Biomaterials 76, 1-10, doi:http://dx.doi.org/10.1016/j.biomaterials.2015.10.041 (2016).

18 Pan, Q. et al. Corticosteroid-loaded biodegradable nanoparticles for prevention of

corneal allograft rejection in rats. J Control Release 201, 32-40, doi:10.1016/j.jconrel.2015.01.009 (2015).

19 Lewis, J. S. et al. Combinatorial delivery of immunosuppressive factors to

dendritic cells using dual-sized microspheres. J Mater Chem B Mater Biol Med 2, 2562-2574, doi:10.1039/c3tb21460e (2014).

20 Lewis, J. S. et al. A combination dual-sized microparticle system modulates

dendritic cells and prevents type 1 diabetes in prediabetic NOD mice. Clin Immunol 160, 90-102, doi:10.1016/j.clim.2015.03.023 (2015).

21 Acharya, A. P., Clare-Salzler, M. J. & Keselowsky, B. G. A high-throughput

microparticle microarray platform for dendritic cell-targeting vaccines. Biomaterials 30, 4168-4177, doi:10.1016/j.biomaterials.2009.04.032 (2009).

22 Acharya, A. P. et al. A cell-based microarray to investigate combinatorial effects

of microparticle-encapsulated adjuvants on dendritic cell activation. Journal of Materials Chemistry B, doi:10.1039/C5TB01754H (2016).

142

23 Acharya, A. P., Lewis, J. S. & Keselowsky, B. G. Combinatorial co-encapsulation of hydrophobic molecules in poly(lactide-co-glycolide) microparticles. Biomaterials 34, 3422-3430, doi:10.1016/j.biomaterials.2013.01.032 (2013).

24 Larson, N. & Ghandehari, H. Polymeric conjugates for drug delivery. Chemistry of materials : a publication of the American Chemical Society 24, 840-853, doi:10.1021/cm2031569 (2012).

25 Dane, K. Y. et al. Nano-sized drug-loaded micelles deliver payload to lymph node

immune cells and prolong allograft survival. Journal of Controlled Release 156, 154-160, doi:http://dx.doi.org/10.1016/j.jconrel.2011.08.009 (2011).

26 Miki, K. et al. Combination therapy with dendritic cell vaccine and IL-2

encapsulating polymeric micelles enhances intra-tumoral accumulation of antigen-specific CTLs. Int Immunopharmacol 23, 499-504, doi:10.1016/j.intimp.2014.09.025 (2014).

27 Svenson, S. & Tomalia, D. A. Dendrimers in biomedical applications--reflections

on the field. Adv Drug Deliv Rev 57, 2106-2129, doi:10.1016/j.addr.2005.09.018 (2005).

28 Lundquist, J. J. & Toone, E. J. The cluster glycoside effect. Chem Rev 102, 555-

578 (2002). 29 Heimburg, J. et al. Inhibition of spontaneous breast cancer metastasis by anti-

Thomsen-Friedenreich antigen monoclonal antibody JAA-F11. Neoplasia 8, 939-948, doi:10.1593/neo.06493 (2006).

30 Baek, M.-G. & Roy, R. Synthesis and protein binding properties of T-antigen

containing GlycoPAMAM dendrimers. Bioorganic & Medicinal Chemistry 10, 11-17, doi:http://dx.doi.org/10.1016/S0968-0896(01)00248-6 (2002).

31 Cheng, Y., Zhao, L., Li, Y. & Xu, T. Design of biocompatible dendrimers for

cancer diagnosis and therapy: current status and future perspectives. Chem Soc Rev 40, 2673-2703, doi:10.1039/c0cs00097c (2011).

32 Ahmed, E. M. Hydrogel: Preparation, characterization, and applications: A

review. Journal of Advanced Research 6, 105-121, doi:http://dx.doi.org/10.1016/j.jare.2013.07.006 (2015).

33 Lim, F. & Sun, A. M. Microencapsulated islets as bioartificial endocrine pancreas.

Science 210, 908-910 (1980). 34 Neufeld, T. et al. The efficacy of an immunoisolating membrane system for islet

xenotransplantation in minipigs. PLoS One 8, e70150, doi:10.1371/journal.pone.0070150 (2013).

143

35 Zakrzewski, J. L., van den Brink, M. R. & Hubbell, J. A. Overcoming immunological barriers in regenerative medicine. Nat Biotechnol 32, 786-794, doi:10.1038/nbt.2960 (2014).

36 Schwendener, R. A. Liposomes as vaccine delivery systems: a review of the recent advances. Ther Adv Vaccines 2, 159-182, doi:10.1177/2051013614541440 (2014).

37 Brown, S. & Khan, D. R. The treatment of breast cancer using liposome

technology. J Drug Deliv 2012, 212965, doi:10.1155/2012/212965 (2012). 38 Xu, H., Li, Z. & Si, J. Nanocarriers in gene therapy: a review. J Biomed

Nanotechnol 10, 3483-3507 (2014). 39 de Kleer, I. et al. Autologous stem cell transplantation for autoimmunity induces

immunologic self-tolerance by reprogramming autoreactive T cells and restoring the CD4+CD25+ immune regulatory network. Blood 107, 1696-1702, doi:10.1182/blood-2005-07-2800 (2006).

40 Mischler, R. & Metcalfe, I. C. Inflexal®V a trivalent virosome subunit influenza

vaccine: production. Vaccine 20, Supplement 5, B17-B23, doi:http://dx.doi.org/10.1016/S0264-410X(02)00512-1 (2002).

41 Bovier, P. A. Epaxal: a virosomal vaccine to prevent hepatitis A infection. Expert

Rev Vaccines 7, 1141-1150, doi:10.1586/14760584.7.8.1141 (2008). 42 Hirai, T. et al. A novel approach inducing transplant tolerance by activated

invariant natural killer T cells with costimulatory blockade. Am J Transplant 14, 554-567, doi:10.1111/ajt.12606 (2014).

43 Jiao, Q., Li, L., Mu, Q. & Zhang, Q. Immunomodulation of Nanoparticles in

Nanomedicine Applications. BioMed Research International 2014, 426028, doi:10.1155/2014/426028 (2014).

44 Zhao, L. et al. Nanoparticle vaccines. Vaccine 32, 327-337,

doi:http://dx.doi.org/10.1016/j.vaccine.2013.11.069 (2014). 45 Tao, W., Ziemer, K. S. & Gill, H. S. Gold nanoparticle-M2e conjugate

coformulated with CpG induces protective immunity against influenza A virus. Nanomedicine (Lond) 9, 237-251, doi:10.2217/nnm.13.58 (2014).

46 Xu, L. et al. Surface-engineered gold nanorods: promising DNA vaccine adjuvant

for HIV-1 treatment. Nano Lett 12, 2003-2012, doi:10.1021/nl300027p (2012). 47 Vega, R. A. et al. Modified gold nanoparticle vectors: a biocompatible intracellular

delivery system for pancreatic islet cell transplantation. Surgery 148, 858-865; discussion 865-856, doi:10.1016/j.surg.2010.07.036 (2010).

144

48 Villa, C. H. et al. Single-walled carbon nanotubes deliver peptide antigen into

dendritic cells and enhance IgG responses to tumor-associated antigens. ACS Nano 5, 5300-5311, doi:10.1021/nn200182x (2011).

49 Liu, Y., Zhao, Y., Sun, B. & Chen, C. Understanding the Toxicity of Carbon

Nanotubes. Accounts of Chemical Research 46, 702-713, doi:10.1021/ar300028m (2013).

50 Niut, Y. et al. Recent advances in the rational design of silica-based

nanoparticles for gene therapy. Ther Deliv 3, 1217-1237 (2012). 51 Xia, T. et al. Polyethyleneimine coating enhances the cellular uptake of

mesoporous silica nanoparticles and allows safe delivery of siRNA and DNA constructs. ACS Nano 3, 3273-3286, doi:10.1021/nn900918w (2009).

52 Carter, P. J. Introduction to current and future protein therapeutics: a protein

engineering perspective. Exp Cell Res 317, 1261-1269, doi:10.1016/j.yexcr.2011.02.013 (2011).

53 Huang, C. Receptor-Fc fusion therapeutics, traps, and MIMETIBODY technology.

Curr Opin Biotechnol 20, 692-699, doi:10.1016/j.copbio.2009.10.010 (2009). 54 Jazayeri, J. A. & Carroll, G. J. Fc-based cytokines : prospects for engineering

superior therapeutics. BioDrugs 22, 11-26 (2008). 55 Liu, F. et al. Fusion with extracellular domain of cytotoxic T-lymphocyte-

associated-antigen 4 leads to enhancement of immunogenicity of Hantaan virus DNA vaccines in C57BL/6 mice. Virology Journal 8, 448-448, doi:10.1186/1743-422X-8-448 (2011).

56 Santagostino, E. et al. Long-acting recombinant coagulation factor IX albumin

fusion protein (rIX-FP) in hemophilia B: results of a phase 3 trial. Blood 127, 1761 (2016).

57 Fahrer, J., Rausch, J. & Barth, H. A cell-permeable fusion protein based on

Clostridium botulinum C2 toxin for delivery of p53 tumorsuppressor into cancer cells. PLoS One 8, e72455, doi:10.1371/journal.pone.0072455 (2013).

58 Tan, Q. et al. Characterization and comparison of commercially available TNF

receptor 2-Fc fusion protein products. mAbs 4, 761-774, doi:10.4161/mabs.22276 (2012).

59 Mahler, S. Safety of biologics therapy: Monoclonal antibodies, cytokines, fusion

proteins, hormones, enzymes, coagulation proteins, vaccines, botulinum toxins. mAbs 9, 885-888, doi:10.1080/19420862.2017.1343709 (2017).

145

60 Rogers, B., Dong, D. & Li, Z. Recombinant human serum albumin fusion proteins and novel applications in drug delivery and therapy. Curr Pharm Des 21, 1899-1907 (2015).

61 Blair, H. A. & Keating, G. M. Albiglutide: a review of its use in patients with type 2

diabetes mellitus. Drugs 75, 651-663, doi:10.1007/s40265-015-0370-5 (2015). 62 Lyseng-Williamson, K. A. Coagulation Factor IX (Recombinant), Albumin Fusion

Protein (Albutrepenonacog Alfa; Idelvion(R)): A Review of Its Use in Haemophilia B. Drugs 77, 97-106, doi:10.1007/s40265-016-0679-8 (2017).

63 Foged, C., Brodin, B., Frokjaer, S. & Sundblad, A. Particle size and surface

charge affect particle uptake by human dendritic cells in an in vitro model. Int J Pharm 298, 315-322, doi:10.1016/j.ijpharm.2005.03.035 (2005).

64 Sahay, G., Alakhova, D. Y. & Kabanov, A. V. Endocytosis of Nanomedicines.

Journal of controlled release : official journal of the Controlled Release Society 145, 182-195, doi:10.1016/j.jconrel.2010.01.036 (2010).

65 Champion, J. A., Katare, Y. K. & Mitragotri, S. Particle shape: a new design

parameter for micro- and nanoscale drug delivery carriers. J Control Release 121, 3-9, doi:10.1016/j.jconrel.2007.03.022 (2007).

66 Champion, J. A. & Mitragotri, S. Role of target geometry in phagocytosis. Proc

Natl Acad Sci U S A 103, 4930-4934, doi:10.1073/pnas.0600997103 (2006). 67 Schaeublin, N. M. et al. Surface charge of gold nanoparticles mediates

mechanism of toxicity. Nanoscale 3, 410-420, doi:10.1039/c0nr00478b (2011). 68 Naha, P. C., Davoren, M., Lyng, F. M. & Byrne, H. J. Reactive oxygen species

(ROS) induced cytokine production and cytotoxicity of PAMAM dendrimers in J774A.1 cells. Toxicol Appl Pharmacol 246, 91-99, doi:10.1016/j.taap.2010.04.014 (2010).

69 Kwon, Y. J., Standley, S. M., Goh, S. L. & Fréchet, J. M. J. Enhanced antigen

presentation and immunostimulation of dendritic cells using acid-degradable cationic nanoparticles. Journal of Controlled Release 105, 199-212, doi:http://dx.doi.org/10.1016/j.jconrel.2005.02.027 (2005).

70 Acharya, A. P., Dolgova, N. V., Clare-Salzler, M. J. & Keselowsky, B. G.

Adhesive substrate-modulation of adaptive immune responses. Biomaterials 29, 4736-4750, doi:10.1016/j.biomaterials.2008.08.040 (2008).

146

71 Acharya, A. P., Dolgova, N. V., Xia, C. Q., Clare-Salzler, M. J. & Keselowsky, B. G. Adhesive substrates modulate the activation and stimulatory capacity of non-obese diabetic mouse-derived dendritic cells. Acta Biomater 7, 180-192, doi:10.1016/j.actbio.2010.08.026 (2011).

72 Lewis, J. S., Zaveri, T. D., Crooks, C. P., 2nd & Keselowsky, B. G. Microparticle

surface modifications targeting dendritic cells for non-activating applications. Biomaterials 33, 7221-7232, doi:10.1016/j.biomaterials.2012.06.049 (2012).

73 Acharya, A. P. et al. The modulation of dendritic cell integrin binding and

activation by RGD-peptide density gradient substrates. Biomaterials 31, 7444-7454, doi:10.1016/j.biomaterials.2010.06.025 (2010).

74 Zaveri, T. D., Lewis, J. S., Dolgova, N. V., Clare-Salzler, M. J. & Keselowsky, B.

G. Integrin-directed modulation of macrophage responses to biomaterials. Biomaterials 35, 3504-3515, doi:10.1016/j.biomaterials.2014.01.007 (2014).

75 Sionkowska, A. Current research on the blends of natural and synthetic polymers

as new biomaterials: Review. Progress in Polymer Science 36, 1254-1276, doi:http://dx.doi.org/10.1016/j.progpolymsci.2011.05.003 (2011).

76 Winter, M. et al. Activation of the inflammasome by amorphous silica and TiO2

nanoparticles in murine dendritic cells. Nanotoxicology 5, 326-340, doi:10.3109/17435390.2010.506957 (2011).

77 Lutz, M. B. & Schuler, G. Immature, semi-mature and fully mature dendritic cells:

which signals induce tolerance or immunity? Trends Immunol 23, 445-449 (2002).

78 Hwu, P. et al. Indoleamine 2,3-dioxygenase production by human dendritic cells

results in the inhibition of T cell proliferation. J Immunol 164, 3596-3599 (2000). 79 Banchereau, J. et al. Immunobiology of dendritic cells. Annu Rev Immunol 18,

767-811, doi:10.1146/annurev.immunol.18.1.767 (2000). 80 Akira, S., Uematsu, S. & Takeuchi, O. Pathogen Recognition and Innate

Immunity. Cell 124, 783-801, doi:http://dx.doi.org/10.1016/j.cell.2006.02.015 (2006).

81 Iwasaki, A. & Medzhitov, R. Toll-like receptor control of the adaptive immune

responses. Nat Immunol 5, 987-995, doi:10.1038/ni1112 (2004). 82 Broere, F., Apasov, S. G., Sitkovsky, M. V. & Eden, W. v. Principles of

Immunopharmacology: 3rd revised and extended edition. (Springer, 2011).

147

83 Korn, T., Bettelli, E., Oukka, M. & Kuchroo, V. K. IL-17 and Th17 Cells. Annu Rev Immunol 27, 485-517, doi:10.1146/annurev.immunol.021908.132710 (2009).

84 Steinman, R. M., Hawiger, D. & Nussenzweig, M. C. Tolerogenic dendritic cells.

Annu Rev Immunol 21, 685-711, doi:10.1146/annurev.immunol.21.120601.141040 (2003).

85 Hoeffel, G. et al. Adult Langerhans cells derive predominantly from embryonic

fetal liver monocytes with a minor contribution of yolk sac-derived macrophages. J Exp Med 209, 1167-1181, doi:10.1084/jem.20120340 (2012).

86 Huang, L., Baban, B., Johnson, B. A., 3rd & Mellor, A. L. Dendritic cells,

indoleamine 2,3 dioxygenase and acquired immune privilege. Int Rev Immunol 29, 133-155, doi:10.3109/08830180903349669 (2010).

87 Jones, K. S. Biomaterials as vaccine adjuvants. Biotechnol Prog 24, 807-814,

doi:10.1002/btpr.10 (2008). 88 Sharp, F. A. et al. Uptake of particulate vaccine adjuvants by dendritic cells

activates the NALP3 inflammasome. Proc Natl Acad Sci U S A 106, 870-875, doi:10.1073/pnas.0804897106 (2009).

89 Ali, O. A., Huebsch, N., Cao, L., Dranoff, G. & Mooney, D. J. Infection-mimicking

materials to program dendritic cells in situ. Nat Mater 8, 151-158, doi:10.1038/nmat2357 (2009).

90 Sneh-Edri, H., Likhtenshtein, D. & Stepensky, D. Intracellular targeting of PLGA

nanoparticles encapsulating antigenic peptide to the endoplasmic reticulum of dendritic cells and its effect on antigen cross-presentation in vitro. Mol Pharm 8, 1266-1275, doi:10.1021/mp200198c (2011).

91 El-Sayed, M. E., Hoffman, A. S. & Stayton, P. S. Smart polymeric carriers for

enhanced intracellular delivery of therapeutic macromolecules. Expert Opin Biol Ther 5, 23-32, doi:10.1517/14712598.5.1.23 (2005).

92 Flanary, S., Hoffman, A. S. & Stayton, P. S. Antigen Delivery with

Poly(Propylacrylic Acid) Conjugation Enhances MHC-1 Presentation and T-Cell Activation. Bioconjug Chem 20, 241-248, doi:10.1021/bc800317a (2009).

93 Schlosser, E. et al. TLR ligands and antigen need to be coencapsulated into the

same biodegradable microsphere for the generation of potent cytotoxic T lymphocyte responses. Vaccine 26, 1626-1637, doi:10.1016/j.vaccine.2008.01.030 (2008).

148

94 Yoshida, M., Mata, J. & Babensee, J. E. Effect of poly(lactic-co-glycolic acid) contact on maturation of murine bone marrow-derived dendritic cells. J Biomed Mater Res A 80, 7-12, doi:10.1002/jbm.a.30832 (2007).

95 Beaudette, T. T. et al. In vivo studies on the effect of co-encapsulation of CpG

DNA and antigen in acid-degradable microparticle vaccines. Mol Pharm 6, 1160-1169, doi:10.1021/mp900038e (2009).

96 Elamanchili, P., Diwan, M., Cao, M. & Samuel, J. Characterization of poly(D,L-

lactic-co-glycolic acid) based nanoparticulate system for enhanced delivery of antigens to dendritic cells. Vaccine 22, 2406-2412, doi:10.1016/j.vaccine.2003.12.032 (2004).

97 Ali, O. A., Tayalia, P., Shvartsman, D., Lewin, S. & Mooney, D. J. Inflammatory

cytokines presented from polymer matrices differentially generate and activate DCs. Adv Funct Mater 23, 4621-4628, doi:10.1002/adfm.201203859 (2013).

98 Singh, A., Suri, S. & Roy, K. In-situ crosslinking hydrogels for combinatorial

delivery of chemokines and siRNA-DNA carrying microparticles to dendritic cells. Biomaterials 30, 5187-5200, doi:10.1016/j.biomaterials.2009.06.001 (2009).

99 Pradhan, P. et al. The effect of combined IL10 siRNA and CpG ODN as

pathogen-mimicking microparticles on Th1/Th2 cytokine balance in dendritic cells and protective immunity against B cell lymphoma. Biomaterials 35, 5491-5504, doi:10.1016/j.biomaterials.2014.03.039 (2014).

100 Hudalla, G. A. et al. A self-adjuvanting supramolecular vaccine carrying a folded

protein antigen. Adv Healthc Mater 2, 1114-1119, doi:10.1002/adhm.201200435 (2013).

101 Goodman, J. et al. Vol. Volume 31, issue 12 1269-1280 (Particle & Particle

Systems Characterization, 2014). 102 Haughney, S. L., Ross, K. A., Boggiatto, P. M., Wannemuehler, M. J. &

Narasimhan, B. Effect of nanovaccine chemistry on humoral immune response kinetics and maturation. Nanoscale 6, 13770-13778, doi:10.1039/c4nr03724c (2014).

103 Vela-Ramirez, J. E. et al. Safety and biocompatibility of carbohydrate-

functionalized polyanhydride nanoparticles. Aaps j 17, 256-267, doi:10.1208/s12248-014-9699-z (2015).

104 Fischer, S. et al. The preservation of phenotype and functionality of dendritic

cells upon phagocytosis of polyelectrolyte-coated PLGA microparticles. Biomaterials 28, 994-1004, doi:10.1016/j.biomaterials.2006.10.034 (2007).

149

105 Rogers, T. H. & Babensee, J. E. The role of integrins in the recognition and response of dendritic cells to biomaterials. Biomaterials 32, 1270-1279, doi:10.1016/j.biomaterials.2010.10.014 (2011).

106 Volchenkov, R., Karlsen, M., Jonsson, R. & Appel, S. Type 1 regulatory T cells

and regulatory B cells induced by tolerogenic dendritic cells. Scand J Immunol 77, 246-254, doi:10.1111/sji.12039 (2013).

107 Schwartz, R. H. T cell anergy. Annu Rev Immunol 21, 305-334,

doi:10.1146/annurev.immunol.21.120601.141110 (2003). 108 Sakaguchi, S. et al. Immunologic tolerance maintained by CD25+ CD4+

regulatory T cells: their common role in controlling autoimmunity, tumor immunity, and transplantation tolerance. Immunol Rev 182, 18-32 (2001).

109 Woltman, A. M. et al. The effect of calcineurin inhibitors and corticosteroids on

the differentiation of human dendritic cells. Eur J Immunol 30, 1807-1812, doi:10.1002/1521-4141(200007)30:7&#60;1807::aid-immu1807&#62;3.0.co;2-n (2000).

110 Hackstein, H. et al. Aspirin inhibits in vitro maturation and in vivo

immunostimulatory function of murine myeloid dendritic cells. J Immunol 166, 7053-7062 (2001).

111 Mehling, A. et al. Mycophenolate mofetil impairs the maturation and function of

murine dendritic cells. J Immunol 165, 2374-2381 (2000). 112 Penna, G. & Adorini, L. 1 Alpha,25-dihydroxyvitamin D3 inhibits differentiation,

maturation, activation, and survival of dendritic cells leading to impaired alloreactive T cell activation. J Immunol 164, 2405-2411 (2000).

113 Jhunjhunwala, S., Raimondi, G., Thomson, A. W. & Little, S. R. Delivery of

rapamycin to dendritic cells using degradable microparticles. J Control Release 133, 191-197, doi:10.1016/j.jconrel.2008.10.011 (2009).

114 Yamaguchi, Y., Tsumura, H., Miwa, M. & Inaba, K. Contrasting effects of TGF-

beta 1 and TNF-alpha on the development of dendritic cells from progenitors in mouse bone marrow. Stem Cells 15, 144-153, doi:10.1002/stem.150144 (1997).

115 Steinbrink, K., Wolfl, M., Jonuleit, H., Knop, J. & Enk, A. H. Induction of tolerance

by IL-10-treated dendritic cells. J Immunol 159, 4772-4780 (1997). 116 Hackstein, H. et al. Rapamycin inhibits IL-4--induced dendritic cell maturation in

vitro and dendritic cell mobilization and function in vivo. Blood 101, 4457-4463, doi:10.1182/blood-2002-11-3370 (2003).

150

117 Bryant, J. et al. Nanoparticle delivery of donor antigens for transplant tolerance in allogeneic islet transplantation. Biomaterials 35, 8887-8894, doi:10.1016/j.biomaterials.2014.06.044 (2014).

118 Powell, J. D., Pollizzi, K. N., Heikamp, E. B. & Horton, M. R. Regulation of

immune responses by mTOR. Annu Rev Immunol 30, 39-68, doi:10.1146/annurev-immunol-020711-075024 (2012).

119 Maldonado, R. A. et al. Polymeric synthetic nanoparticles for the induction of

antigen-specific immunological tolerance. Proc Natl Acad Sci U S A, doi:10.1073/pnas.1408686111 (2014).

120 Babensee, J. E. & Paranjpe, A. Differential levels of dendritic cell maturation on

different biomaterials used in combination products. J Biomed Mater Res A 74, 503-510 (2005).

121 Leclerc, C. et al. Immobilized cytokines as biomaterials for manufacturing

immune cell based vaccines. J Biomed Mater Res A 86, 1033-1040, doi:10.1002/jbm.a.31751 (2008).

122 Yuba, E. et al. Gene delivery to dendritic cells mediated by complexes of

lipoplexes and pH-sensitive fusogenic polymer-modified liposomes. J Control Release 130, 77-83, doi:10.1016/j.jconrel.2008.05.007 (2008).

123 Palama, I. E., Cortese, B., D'Amone, S. & Gigli, G. Vol. 3 144 (Biomaterials

Science, 2015). 124 Machen, J. et al. Antisense oligonucleotides down-regulating costimulation

confer diabetes-preventive properties to nonobese diabetic mouse dendritic cells. J Immunol 173, 4331-4341 (2004).

125 Wang, J., Lu, Z., Wientjes, M. G. & Au, J. L. S. in Aaps j Vol. 12 492-503

(2010). 126 Giannoukakis, N., Phillips, B., Finegold, D., Harnaha, J. & Trucco, M. Phase I

(safety) study of autologous tolerogenic dendritic cells in type 1 diabetic patients. Diabetes Care 34, 2026-2032, doi:10.2337/dc11-0472 (2011).

127 Phillips, B. et al. A microsphere-based vaccine prevents and reverses new-onset

autoimmune diabetes. Diabetes 57, 1544-1555, doi:10.2337/db07-0507 (2008). 128 Yeung, A. W., Terentis, A. C., King, N. J. & Thomas, S. R. Role of indoleamine

2,3-dioxygenase in health and disease. Clin Sci (Lond) 129, 601-672, doi:10.1042/cs20140392 (2015).

151

129 Takikawa, O. Biochemical and medical aspects of the indoleamine 2,3-dioxygenase-initiated l-tryptophan metabolism. Biochemical and Biophysical Research Communications 338, 12-19, doi:https://doi.org/10.1016/j.bbrc.2005.09.032 (2005).

130 Fallarino, F., Grohmann, U. & Puccetti, P. Indoleamine 2,3-dioxygenase: from

catalyst to signaling function. Eur J Immunol 42, 1932-1937, doi:10.1002/eji.201242572 (2012).

131 Yanagida, O. et al. Human L-type amino acid transporter 1 (LAT1):

characterization of function and expression in tumor cell lines. Biochim Biophys Acta 1514, 291-302 (2001).

132 Babcock, T. A. & Carlin, J. M. Transcriptional activation of indoleamine

dioxygenase by interleukin 1 and tumor necrosis factor alpha in interferon-treated epithelial cells. Cytokine 12, 588-594, doi:10.1006/cyto.1999.0661 (2000).

133 Carlin, J. M., Borden, E. C., Sondel, P. M. & Byrne, G. I. Interferon-induced

indoleamine 2,3-dioxygenase activity in human mononuclear phagocytes. J Leukoc Biol 45, 29-34 (1989).

134 Guillemin, G. J., Smythe, G., Takikawa, O. & Brew, B. J. Expression of

indoleamine 2,3-dioxygenase and production of quinolinic acid by human microglia, astrocytes, and neurons. Glia 49, 15-23, doi:10.1002/glia.20090 (2005).

135 Pallotta, M. T. et al. Indoleamine 2,3-dioxygenase is a signaling protein in long-

term tolerance by dendritic cells. Nat Immunol 12, 870-878, doi:10.1038/ni.2077 (2011).

136 Efron, P. A. et al. Differential maturation of murine bone-marrow derived dendritic

cells with lipopolysaccharide and tumor necrosis factor-alpha. J Endotoxin Res 11, 145-160, doi:10.1179/096805105x46583 (2005).

137 Mezrich, J. D. et al. An interaction between kynurenine and the aryl hydrocarbon

receptor can generate regulatory T cells. J Immunol 185, 3190-3198, doi:10.4049/jimmunol.0903670 (2010).

138 Nguyen, N. T. et al. Aryl hydrocarbon receptor negatively regulates dendritic cell

immunogenicity via a kynurenine-dependent mechanism. Proc Natl Acad Sci U S A 107, 19961-19966, doi:10.1073/pnas.1014465107 (2010).

139 Grohmann, U. et al. CTLA-4-Ig regulates tryptophan catabolism in vivo. Nat

Immunol 3, 1097-1101, doi:10.1038/ni846 (2002).

152

140 Braun, D., Longman, R. S. & Albert, M. L. A two-step induction of indoleamine 2,3 dioxygenase (IDO) activity during dendritic-cell maturation. Blood 106, 2375-2381, doi:10.1182/blood-2005-03-0979 (2005).

141 Lim, J. Y., Lee, S. E., Park, G., Choi, E. Y. & Min, C. K. Inhibition of indoleamine

2,3-dioxygenase (IDO) by stereoisomers of 1-methyl tryptophan in an experimental graft-versus-tumor model. Exp Hematol, doi:10.1016/j.exphem.2014.06.006 (2014).

142 Munn, D. H. et al. Inhibition of T cell proliferation by macrophage tryptophan

catabolism. J Exp Med 189, 1363-1372 (1999). 143 Sun, J. et al. Upregulated expression of indoleamine 2, 3-dioxygenase in CHO

cells induces apoptosis of competent T cells and increases proportion of Treg cells. J Exp Clin Cancer Res 30, 82, doi:10.1186/1756-9966-30-82 (2011).

144 Terness, P. et al. Inhibition of allogeneic T cell proliferation by indoleamine 2,3-

dioxygenase-expressing dendritic cells: mediation of suppression by tryptophan metabolites. J Exp Med 196, 447-457 (2002).

145 do Prado, K. M. et al. Indoleamine 2,3-dioxygenase (IDO) activity in placental

compartments of renal-transplanted pregnant women. Am J Reprod Immunol 72, 45-56, doi:10.1111/aji.12233 (2014).

146 Johnson, T. S. & Munn, D. H. Host indoleamine 2,3-dioxygenase: contribution to

systemic acquired tumor tolerance. Immunol Invest 41, 765-797, doi:10.3109/08820139.2012.689405 (2012).

147 Stubbendorff, M. et al. Immunological properties of extraembryonic human

mesenchymal stromal cells derived from gestational tissue. Stem Cells Dev 22, 2619-2629, doi:10.1089/scd.2013.0043 (2013).

148 Munn, D. H. et al. Prevention of allogeneic fetal rejection by tryptophan

catabolism. Science 281, 1191-1193 (1998). 149 Zhang, P. et al. The GCN2 eIF2α Kinase Is Required for Adaptation to Amino

Acid Deprivation in Mice. Molecular and Cellular Biology 22, 6681-6688, doi:10.1128/MCB.22.19.6681-6688.2002 (2002).

150 Fallarino, F. et al. The combined effects of tryptophan starvation and tryptophan

catabolites down-regulate T cell receptor zeta-chain and induce a regulatory phenotype in naive T cells. J Immunol 176, 6752-6761 (2006).

151 Mellor, A. L. & Munn, D. H. Tryptophan catabolism and T-cell tolerance:

immunosuppression by starvation? Immunol Today 20, 469-473 (1999).

153

152 Dagenais-Lussier, X. et al. Kynurenine Reduces Memory CD4 T-Cell Survival by Interfering with Interleukin-2 Signaling Early during HIV-1 Infection. J Virol 90, 7967-7979, doi:10.1128/jvi.00994-16 (2016).

153 Brandacher, G. et al. Non-invasive monitoring of kidney allograft rejection

through IDO metabolism evaluation. Kidney Int 71, 60-67, doi:10.1038/sj.ki.5002023 (2007).

154 Cook, C. H. et al. Spontaneous renal allograft acceptance associated with

"regulatory" dendritic cells and IDO. J Immunol 180, 3103-3112 (2008). 155 Miki, T. et al. Blockade of tryptophan catabolism prevents spontaneous

tolerogenicity of liver allografts. Transplant Proc 33, 129-130 (2001). 156 Guillonneau, C. et al. CD40Ig treatment results in allograft acceptance mediated

by CD8CD45RC T cells, IFN-gamma, and indoleamine 2,3-dioxygenase. J Clin Invest 117, 1096-1106, doi:10.1172/jci28801 (2007).

157 Friberg, M. et al. Indoleamine 2,3-dioxygenase contributes to tumor cell evasion

of T cell-mediated rejection. Int J Cancer 101, 151-155, doi:10.1002/ijc.10645 (2002).

158 Johnson, B. A., 3rd, Baban, B. & Mellor, A. L. Targeting the immunoregulatory

indoleamine 2,3 dioxygenase pathway in immunotherapy. Immunotherapy 1, 645-661, doi:10.2217/imt.09.21 (2009).

159 Muller, A. J. & Prendergast, G. C. Marrying immunotherapy with chemotherapy:

why say IDO? Cancer Res 65, 8065-8068, doi:10.1158/0008-5472.can-05-2213 (2005).

160 Munn, D. H. & Mellor, A. L. Indoleamine 2,3-dioxygenase and tumor-induced

tolerance. Journal of Clinical Investigation 117, 1147-1154, doi:10.1172/JCI31178 (2007).

161 Funeshima, N. et al. Inhibition of allogeneic T-cell responses by dendritic cells

expressing transduced indoleamine 2,3-dioxygenase. J Gene Med 7, 565-575, doi:10.1002/jgm.698 (2005).

162 Valladares, R. et al. Lactobacillus johnsonii inhibits indoleamine 2,3-dioxygenase

and alters tryptophan metabolite levels in BioBreeding rats. Faseb j 27, 1711-1720, doi:10.1096/fj.12-223339 (2013).

163 Cady, S. G. & Sono, M. 1-Methyl-DL-tryptophan, beta-(3-benzofuranyl)-DL-

alanine (the oxygen analog of tryptophan), and beta-[3-benzo(b)thienyl]-DL-alanine (the sulfur analog of tryptophan) are competitive inhibitors for indoleamine 2,3-dioxygenase. Arch Biochem Biophys 291, 326-333 (1991).

154

164 Wiseman, A. C. Immunosuppressive Medications. Clinical Journal of the American Society of Nephrology : CJASN 11, 332-343, doi:10.2215/CJN.08570814 (2016).

165 Barratt-Boyes, S. M. & Thomson, A. W. Dendritic cells: tools and targets for

transplant tolerance. Am J Transplant 5, 2807-2813, doi:10.1111/j.1600-6143.2005.01116.x (2005).

166 Li, H. & Shi, B. Tolerogenic dendritic cells and their applications in

transplantation. Cell Mol Immunol 12, 24-30, doi:10.1038/cmi.2014.52 (2015). 167 Sauma, D. et al. Cyclosporine preconditions dendritic cells during differentiation

and reduces IL-2 and IL-12 production following activation: a potential tolerogenic effect. Transplant Proc 35, 2515-2517 (2003).

168 Fischer, R., Turnquist, H. R., Taner, T. & Thomson, A. W. Use of rapamycin in

the induction of tolerogenic dendritic cells. Handb Exp Pharmacol, 215-232, doi:10.1007/978-3-540-71029-5_10 (2009).

169 Barragan, M., Good, M. & Kolls, J. K. Regulation of Dendritic Cell Function by

Vitamin D. Nutrients 7, 8127-8151, doi:10.3390/nu7095383 (2015). 170 Li, M. et al. Immune modulation and tolerance induction by RelB-silenced

dendritic cells through RNA interference. J Immunol 178, 5480-5487 (2007). 171 Li, Q., Harden, J. L., Anderson, C. D. & Egilmez, N. K. Tolerogenic Phenotype of

IFN-gamma-Induced IDO+ Dendritic Cells Is Maintained via an Autocrine IDO-Kynurenine/AhR-IDO Loop. J Immunol 197, 962-970, doi:10.4049/jimmunol.1502615 (2016).

172 Mbongue, J. C. et al. Induction of Indoleamine 2, 3-Dioxygenase in Human

Dendritic Cells by a Cholera Toxin B Subunit—Proinsulin Vaccine. PLoS ONE 10, e0118562, doi:10.1371/journal.pone.0118562 (2015).

173 Fallarino, F. et al. T cell apoptosis by tryptophan catabolism. Cell Death Differ 9,

1069-1077, doi:10.1038/sj.cdd.4401073 (2002). 174 Hayashi, T. et al. Inhibition of experimental asthma by indoleamine 2,3-

dioxygenase. J Clin Invest 114, 270-279, doi:10.1172/jci21275 (2004). 175 Hacein-Bey-Abina, S. et al. in N Engl J Med Vol. 348 255-256 (2003). 176 Couzin, J. & Kaiser, J. in Science Vol. 307 1028 (2005).

155

177 Rezakhanlou, A. M. et al. Highly Efficient Stable Expression of Indoleamine 2,3 Dioxygenase Gene in Primary Fibroblasts. Biological Procedures Online 12, 107-112, doi:10.1007/s12575-010-9028-6 (2010).

178 Tan, P. H. et al. Modulation of human dendritic-cell function following

transduction with viral vectors: implications for gene therapy. Blood 105, 3824-3832, doi:10.1182/blood-2004-10-3880 (2005).

179 Rabinovich, G. A. & Toscano, M. A. Turning 'sweet' on immunity: galectin-glycan

interactions in immune tolerance and inflammation. Nat Rev Immunol 9, 338-352, doi:10.1038/nri2536 (2009).

180 van Kooyk, Y. & Rabinovich, G. A. Protein-glycan interactions in the control of

innate and adaptive immune responses. Nat Immunol 9, 593-601, doi:10.1038/ni.f.203 (2008).

181 Dumic, J., Dabelic, S. & Flogel, M. Galectin-3: an open-ended story. Biochim

Biophys Acta 1760, 616-635, doi:10.1016/j.bbagen.2005.12.020 (2006). 182 Dagher, S. F., Wang, J. L. & Patterson, R. J. Identification of galectin-3 as a

factor in pre-mRNA splicing. Proc Natl Acad Sci U S A 92, 1213-1217 (1995). 183 Wang, J. L., Gray, R. M., Haudek, K. C. & Patterson, R. J. Nucleocytoplasmic

lectins. Biochim Biophys Acta 1673, 75-93, doi:10.1016/j.bbagen.2004.03.013 (2004).

184 Demetriou, M., Granovsky, M., Quaggin, S. & Dennis, J. W. Negative regulation

of T-cell activation and autoimmunity by Mgat5 N-glycosylation. Nature 409, 733-739, doi:10.1038/35055582 (2001).

185 Rapoport, E. M. & Bovin, N. V. Specificity of Human Galectins on Cell Surfaces.

Biochemistry (Mosc) 80, 846-856, doi:10.1134/s0006297915070056 (2015). 186 Fukumori, T. et al. CD29 and CD7 mediate galectin-3-induced type II T-cell

apoptosis. Cancer Res 63, 8302-8311 (2003). 187 MacKinnon, A. C. et al. Regulation of alternative macrophage activation by

galectin-3. J Immunol 180, 2650-2658 (2008). 188 Hernandez, J. D. & Baum, L. G. Ah, sweet mystery of death! Galectins and

control of cell fate. Glycobiology 12, 127r-136r (2002). 189 Swarte, V. V., Mebius, R. E., Joziasse, D. H., Van den Eijnden, D. H. & Kraal, G.

Lymphocyte triggering via L-selectin leads to enhanced galectin-3-mediated binding to dendritic cells. Eur J Immunol 28, 2864-2871, doi:10.1002/(sici)1521-4141(199809)28:09<2864::aid-immu2864>3.0.co;2-u (1998).

156

190 Markowska, A. I., Liu, F. T. & Panjwani, N. Galectin-3 is an important mediator of VEGF- and bFGF-mediated angiogenic response. J Exp Med 207, 1981-1993, doi:10.1084/jem.20090121 (2010).

191 Croci, D. O. et al. Glycosylation-dependent lectin-receptor interactions preserve

angiogenesis in anti-VEGF refractory tumors. Cell 156, 744-758, doi:10.1016/j.cell.2014.01.043 (2014).

192 Funasaka, T., Raz, A. & Nangia-Makker, P. Galectin-3 in angiogenesis and

metastasis. Glycobiology 24, 886-891, doi:10.1093/glycob/cwu086 (2014). 193 Kupper, C. E. et al. Fluorescent SNAP-tag galectin fusion proteins as novel tools

in glycobiology. Curr Pharm Des 19, 5457-5467 (2013). 194 Bocker, S. & Elling, L. Binding characteristics of galectin-3 fusion proteins.

Glycobiology 27, 457-468, doi:10.1093/glycob/cwx007 (2017). 195 John, C. M., Leffler, H., Kahl-Knutsson, B., Svensson, I. & Jarvis, G. A.

Truncated galectin-3 inhibits tumor growth and metastasis in orthotopic nude mouse model of human breast cancer. Clin Cancer Res 9, 2374-2383 (2003).

196 Johnson, K. D. et al. Galectin-3 as a Potential Therapeutic Target in Tumors

Arising from Malignant Endothelia. Neoplasia (New York, N.Y.) 9, 662-670 (2007).

197 Pieters, R. J. Inhibition and detection of galectins. Chembiochem 7, 721-728,

doi:10.1002/cbic.200600011 (2006). 198 He, S.-X., Song, G., Shi, J.-P., Guo, Y.-Q. & Guo, Z.-Y. Nanoluciferase as a

novel quantitative protein fusion tag: Application for overexpression and bioluminescent receptor-binding assays of human leukemia inhibitory factor. Biochimie 106, 140-148, doi:https://doi.org/10.1016/j.biochi.2014.08.012 (2014).

199 Thorne, N., Inglese, J. & Auld, D. S. Illuminating insights into firefly luciferase and

other bioluminescent reporters used in chemical biology. Chemistry & biology 17, 646-657, doi:10.1016/j.chembiol.2010.05.012 (2010).

200 Hall, M. P. et al. Engineered Luciferase Reporter from a Deep Sea Shrimp

Utilizing a Novel Imidazopyrazinone Substrate. ACS Chemical Biology 7, 1848-1857, doi:10.1021/cb3002478 (2012).

201 Ahmad, N. et al. Galectin-3 precipitates as a pentamer with synthetic multivalent

carbohydrates and forms heterogeneous cross-linked complexes. J Biol Chem 279, 10841-10847, doi:10.1074/jbc.M312834200 (2004).

157

202 Barboni, E. A., Bawumia, S. & Hughes, R. C. Kinetic measurements of binding of galectin 3 to a laminin substratum. Glycoconj J 16, 365-373 (1999).

203 Friedrichs, J., Manninen, A., Muller, D. J. & Helenius, J. Galectin-3 Regulates

Integrin α2β1-mediated Adhesion to Collagen-I and -IV. Journal of Biological Chemistry 283, 32264-32272, doi:10.1074/jbc.M803634200 (2008).

204 Cederfur, C. et al. Different affinity of galectins for human serum glycoproteins:

galectin-3 binds many protease inhibitors and acute phase proteins. Glycobiology 18, 384-394, doi:10.1093/glycob/cwn015 (2008).

205 Stillman, B. N. et al. Galectin-3 and galectin-1 bind distinct cell surface

glycoprotein receptors to induce T cell death. J Immunol 176, 778-789 (2006). 206 Leader, B., Baca, Q. J. & Golan, D. E. Protein therapeutics: a summary and

pharmacological classification. Nat Rev Drug Discov 7, 21-39 (2008). 207 Lachmann, R. H. Enzyme replacement therapy for lysosomal storage diseases.

Curr Opin Pediatr 23, 588-593, doi:10.1097/MOP.0b013e32834c20d9 (2011). 208 Barton, N. W. et al. Replacement Therapy for Inherited Enzyme Deficiency —

Macrophage-Targeted Glucocerebrosidase for Gaucher's Disease. New England Journal of Medicine 324, 1464-1470, doi:10.1056/NEJM199105233242104 (1991).

209 Rosenbloom, B. E. & Weinreb, N. J. Gaucher disease: a comprehensive review.

Crit Rev Oncog 18, 163-175 (2013). 210 Kontermann, R. E. Strategies to Extend Plasma Half-Lives of Recombinant

Antibodies. BioDrugs 23, 93-109, doi:10.2165/00063030-200923020-00003 (2009).

211 Kontermann, R. E. Strategies for extended serum half-life of protein therapeutics.

Current Opinion in Biotechnology 22, 868-876, doi:https://doi.org/10.1016/j.copbio.2011.06.012 (2011).

212 Walsh, G. Biopharmaceutical benchmarks 2014. Nat Biotech 32, 992-1000,

doi:10.1038/nbt.3040 http://www.nature.com/nbt/journal/v32/n10/abs/nbt.3040.html#supplementary-information (2014).

213 Golding, B., Jiang, H., Sauna, Z. & Levin, D. Immune responses to chimeric

human-FIX-mouse-Fc fusion protein in a hemophilia B mouse model (THER2P.963). The Journal of Immunology 194, 67.14 (2015).

158

214 Nangia-Makker, P., Balan, V. & Raz, A. Galectin-3-Binding and Metastasis. Methods in molecular biology (Clifton, N.J.) 878, 251-266, doi:10.1007/978-1-61779-854-2_17 (2012).

215 Yang, R. Y., Hsu, D. K. & Liu, F. T. Expression of galectin-3 modulates T-cell

growth and apoptosis. Proc Natl Acad Sci U S A 93, 6737-6742 (1996). 216 Stowell, S. R. et al. Differential roles of galectin-1 and galectin-3 in regulating

leukocyte viability and cytokine secretion. J Immunol 180, 3091-3102 (2008). 217 Ahmed, H. & AlSadek, D. M. M. Galectin-3 as a Potential Target to Prevent

Cancer Metastasis. Clinical Medicine Insights. Oncology 9, 113-121, doi:10.4137/CMO.S29462 (2015).

218 Edelstam, G. A., Laurent, U. B., Lundkvist, O. E., Fraser, J. R. & Laurent, T. C.

Concentration and turnover of intraperitoneal hyaluronan during inflammation. Inflammation 16, 459-469 (1992).

219 Catorce, M. N. & Gevorkian, G. LPS-induced Murine Neuroinflammation Model:

Main Features and Suitability for Pre-clinical Assessment of Nutraceuticals. Current Neuropharmacology 14, 155-164, doi:10.2174/1570159X14666151204122017 (2016).

220 Juskewitch, J. E. et al. LPS-induced murine systemic inflammation is driven by

parenchymal cell activation and exclusively predicted by early MCP-1 plasma levels. Am J Pathol 180, 32-40, doi:10.1016/j.ajpath.2011.10.001 (2012).

221 Ghorani, V., Boskabady, M. H., Khazdair, M. R. & Kianmeher, M. Experimental

animal models for COPD: a methodological review. Tob Induc Dis 15, 25, doi:10.1186/s12971-017-0130-2 (2017).

222 Kell, D. B. & Pretorius, E. On the translocation of bacteria and their

lipopolysaccharides between blood and peripheral locations in chronic, inflammatory diseases: the central roles of LPS and LPS-induced cell death. Integr Biol (Camb) 7, 1339-1377, doi:10.1039/c5ib00158g (2015).

223 Copeland, S. et al. Acute Inflammatory Response to Endotoxin in Mice and

Humans. Clinical and Diagnostic Laboratory Immunology 12, 60-67 (2005). 224 Ianaro, A., Tersigni, M. & D'Acquisto, F. New insight in LPS antagonist. Mini Rev

Med Chem 9, 306-317 (2009). 225 Oz, H. S. & Puleo, D. A. Animal models for periodontal disease. J Biomed

Biotechnol 2011, 754857, doi:10.1155/2011/754857 (2011).

159

226 Holt, S. C. & Ebersole, J. L. Porphyromonas gingivalis, Treponema denticola, and Tannerella forsythia: the "red complex", a prototype polybacterial pathogenic consortium in periodontitis. Periodontol 2000 38, 72-122, doi:10.1111/j.1600-0757.2005.00113.x (2005).

227 Blinkhorn, A. et al. Is there a role for triclosan/copolymer toothpaste in the

management of periodontal disease? Br Dent J 207, 117-125, doi:10.1038/sj.bdj.2009.669 (2009).

228 Page, R. C. & Kornman, K. S. The pathogenesis of human periodontitis: an

introduction. Periodontol 2000 14, 9-11 (1997). 229 Madianos, P. N., Bobetsis, Y. A. & Kinane, D. F. Generation of inflammatory

stimuli: how bacteria set up inflammatory responses in the gingiva. J Clin Periodontol 32 Suppl 6, 57-71, doi:10.1111/j.1600-051X.2005.00821.x (2005).

230 Gaffar, A., Scherl, D., Afflitto, J. & Coleman, E. J. The effect of triclosan on

mediators of gingival inflammation. J Clin Periodontol 22, 480-484 (1995). 231 Kinane, D. F., Stathopoulou, P. G. & Papapanou, P. N. Periodontal diseases. Nat

Rev Dis Primers 3, 17038, doi:10.1038/nrdp.2017.38 (2017). 232 Gotsman, I. et al. Periodontal destruction is associated with coronary artery

disease and periodontal infection with acute coronary syndrome. J Periodontol 78, 849-858, doi:10.1902/jop.2007.060301 (2007).

233 Khader, Y. S., Dauod, A. S., El-Qaderi, S. S., Alkafajei, A. & Batayha, W. Q.

Periodontal status of diabetics compared with nondiabetics: a meta-analysis. J Diabetes Complications 20, 59-68, doi:10.1016/j.jdiacomp.2005.05.006 (2006).

234 Gemmell, E. & Seymour, G. J. Immunoregulatory control of Th1/Th2 cytokine

profiles in periodontal disease. Periodontol 2000 35, 21-41, doi:10.1111/j.0906-6713.2004.003557.x (2004).

235 Gemmell, E., Marshall, R. I. & Seymour, G. J. Cytokines and prostaglandins in

immune homeostasis and tissue destruction in periodontal disease. Periodontol 2000 14, 112-143 (1997).

236 Everett, F. G., Waerhaug, J. & Widman, A. Leonard Widman: surgical treatment

of pyorrhea alveolaris. J Periodontol 42, 571-579, doi:10.1902/jop.1971.42.9.571 (1971).

237 Ramfjord, S. P. & Nissle, R. R. The modified widman flap. J Periodontol 45, 601-

607, doi:10.1902/jop.1974.45.8.2.601 (1974).

160

238 Nyman, S., Lindhe, J., Karring, T. & Rylander, H. New attachment following surgical treatment of human periodontal disease. J Clin Periodontol 9, 290-296 (1982).

239 Larsson, L. et al. Regenerative Medicine for Periodontal and Peri-implant

Diseases. J Dent Res 95, 255-266, doi:10.1177/0022034515618887 (2016). 240 Yukna, R. A., Carr, R. L. & Evans, G. H. Histologic evaluation of an Nd:YAG

laser-assisted new attachment procedure in humans. Int J Periodontics Restorative Dent 27, 577-587 (2007).

241 Weiss, U. Inflammation. Nature 454, 427-427 (2008). 242 Fullerton, J. N. & Gilroy, D. W. Resolution of inflammation: a new therapeutic

frontier. Nat Rev Drug Discov 15, 551-567, doi:10.1038/nrd.2016.39 (2016). 243 Gabay, C. & Kushner, I. Acute-Phase Proteins and Other Systemic Responses to

Inflammation. New England Journal of Medicine 340, 448-454, doi:10.1056/NEJM199902113400607 (1999).

244 Wang, L. et al. Efficacy evaluation of low-dose aspirin in IVF/ICSI patients

evidence from 13 RCTs: A systematic review and meta-analysis. Medicine (Baltimore) 96, e7720, doi:10.1097/md.0000000000007720 (2017).

245 Taneja, A., Della Pasqua, O. & Danhof, M. Challenges in translational drug

research in neuropathic and inflammatory pain: the prerequisites for a new paradigm. Eur J Clin Pharmacol, doi:10.1007/s00228-017-2301-8 (2017).

246 Munoz Olmo, L., Juan Armas, J. & Gomariz Garcia, J. J. [Risk of fatal/non-fatal

events in patients with previous coronary heart disease/acute myocardial infarction and treatment with non-steroidal anti-inflammatory drugs]. Semergen, doi:10.1016/j.semerg.2017.07.004 (2017).

247 Nimmo, S. M., Foo, I. T. H. & Paterson, H. M. Enhanced recovery after surgery:

Pain management. J Surg Oncol, doi:10.1002/jso.24814 (2017). 248 Patel, N. U., Vera, N. C., Shealy, E. R., Wetzel, M. & Feldman, S. R. A Review of

the Use of Secukinumab for Psoriatic Arthritis. Rheumatol Ther, doi:10.1007/s40744-017-0076-0 (2017).

249 Hayta, E. & Elden, H. Acute spinal cord injury: A review of pathophysiology and

potential of non-steroidal anti-inflammatory drugs for pharmacological intervention. J Chem Neuroanat, doi:10.1016/j.jchemneu.2017.08.001 (2017).

250 Herold, K. C. et al. Anti-CD3 monoclonal antibody in new-onset type 1 diabetes

mellitus. N Engl J Med 346, 1692-1698, doi:10.1056/NEJMoa012864 (2002).

161

251 Keymeulen, B. et al. Insulin needs after CD3-antibody therapy in new-onset type 1 diabetes. N Engl J Med 352, 2598-2608, doi:10.1056/NEJMoa043980 (2005).

252 Kotsovilis, S. & Andreakos, E. Therapeutic human monoclonal antibodies in

inflammatory diseases. Methods Mol Biol 1060, 37-59, doi:10.1007/978-1-62703-586-6_3 (2014).

253 Laveti, D. et al. Anti-inflammatory treatments for chronic diseases: a review.

Inflamm Allergy Drug Targets 12, 349-361 (2013). 254 Kamala, T. Hock immunization: a humane alternative to mouse footpad

injections. J Immunol Methods 328, 204-214, doi:10.1016/j.jim.2007.08.004 (2007).

255 Vogel, C. & Marcotte, E. M. Insights into the regulation of protein abundance

from proteomic and transcriptomic analyses. Nat Rev Genet 13, 227-232, doi:10.1038/nrg3185 (2012).

256 Kowal, A., Warminska, J. & Krasowska, D. Immunosuppressive drugs in

dermatology--benefits and threats. Ann Univ Mariae Curie Sklodowska Med 58, 14-21 (2003).

257 Malcolm, J. et al. IL-33 Exacerbates Periodontal Disease through Induction of

RANKL. Journal of Dental Research 94, 968-975, doi:10.1177/0022034515577815 (2015).

162

BIOGRAPHICAL SKETCH

Evelyn Bracho Sanchez was born in Caracas, Venezuela in 1989. She moved to

Titusville, FL in 2002 and attended middle and high school until 2007 when she returned

to Venezuela. Evelyn obtained her high school degree from Escuela Comunitaria and

soon thereafter returned to Gainesville, FL. In 2008, Evelyn graduated from Eastside

High School and in 2010 from Santa Fe College. Evelyn transferred to the University of

Florida where she obtained her Bachelor of Science in Materials Science and

Engineering. During her undergraduate tenure, Evelyn conducted research under the

mentorship of Dr. Brian Sorg, who introduced her to biomaterials for immune

applications. The work with Dr. Sorg, lead to an interest in researching potential cures

for complex immunological diseases which directed her interest to the laboratory of Dr.

Benjamin Keselowsky. In August 2012, Evelyn joined the doctoral program in

Biomedical Engineering at the University of Florida and received her PhD in December

2017 under the mentorship of Dr. Keselowsky.

During her doctoral studies, Evelyn worked on numerous projects, largely

focused on biomaterials for immunomodulation. She began with the production and

modification of synthetic microparticles which serve as a vaccine delivery system for

type 1 diabetes. Soon after, through collaboration with Dr. Greg Hudalla, her focus

shifted to engineering of a cell and tissue targeting fusion enzyme to direct localized

cellular metabolism for the treatment of inflammation.

Following her studies, Evelyn hopes to pursue a career as a Medical Science

Liaison in biotechnology where she can apply her skills developed at the University of

Florida to disseminate knowledge and expand strategies to combat immunological

diseases.