Potential Synergistic Effects of Stem Cells and ... - ACS Publications

Jul 18, 2017 - damaged tissues. These biomaterials are prepared by decellularization ... components. The goal of the decellularization process is to r...
0 downloads 0 Views 7MB Size
Subscriber access provided by UNIV OF DURHAM

Review

Potential Synergistic Effects of Stem Cells and Extracellular Matrix Scaffolds Lewis Gaffney, Emily Ann Wrona, and Donald O. Freytes ACS Biomater. Sci. Eng., Just Accepted Manuscript • DOI: 10.1021/acsbiomaterials.7b00083 • Publication Date (Web): 18 Jul 2017 Downloaded from http://pubs.acs.org on July 19, 2017

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

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

Page 1 of 47

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

ACS Biomaterials Science & Engineering

Potential Synergistic Effects of Stem Cells and Extracellular Matrix Scaffolds

Lewis Gaffney1,2, Emily A. Wrona1,2 and Donald O. Freytes Ph.D1,2,*

1) Joint Department of Biomedical Engineering, North Carolina State University/ University of North Carolina-Chapel Hill, Raleigh, NC 2) Comparative Medicine Institute, North Carolina State University, Raleigh, NC

Contact Author:

**Donald O. Freytes, Ph.D. Joint Department of Biomedical Engineering North Carolina State University University of North Carolina-Chapel Hill 4208D Engineering Building III Campus Box 7115 Raleigh, NC 27695 [email protected] [email protected] Office: 919-513-7933 Lewis Gaffney Joint Department of Biomedical Engineering North Carolina State University University of North Carolina-Chapel Hill 4208 Engineering Building III Raleigh, NC 27695 [email protected] Emily A. Wrona Joint Department of Biomedical Engineering North Carolina State University University of North Carolina-Chapel Hill 4208 Engineering Building III Raleigh, NC 27695 [email protected]

ACS Paragon Plus Environment

1

ACS Biomaterials Science & Engineering

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

Page 2 of 47

ABSTRACT In recent years, extracellular matrix (ECM)-derived biomaterials have been used as scaffolds to help regenerate diseased or damaged tissues. These biomaterials are prepared by decellularization of a tissue of interest by chemical or physical removal of the cellular components. The goal of the decellularization process is to remove cells without disturbing tissue specific composition, growth factor content and, in some cases, the mechanical properties. As decellularization can be achieved without significantly affecting the native architecture of the tissue or organ of interest, it provides a scaffold material with native-like composition and structure. ECM scaffolds promote constructive remodeling through several mechanisms that include chemotactic properties, growth factor release and modulation of the immune response. Constructive remodeling by ECM scaffolds relies, in part, on the recruitment of neighboring or circulating cells to the wound site. However, this is a relatively lengthy process and the cells recruited may not be sufficient for complete tissue repair, suggesting that there might be applications that would benefit from the combination of a cellular component with an ECM biomaterial. Stem cells represent a potential therapeutic cell source with clinical evidence of potential beneficial effects. This review will describe how ECM scaffolds can help localize stem cells to the site of injury while directing stem cell differentiation and modulating the response towards tissue regeneration. As the field of tissue engineering takes its next steps, recellularized ECM scaffolds could become the next class of promising bioengineered constructs. This review covers the advantages of acellular ECM scaffolds, and the potential beneficial effect that seeding stem cells in the scaffold has on tissue regeneration.

Key words: Extracellular Matrix (ECM), Stem Cells, Pluripotent Stem Cells (PSCs), Wound Healing, Macrophage Polarization

ACS Paragon Plus Environment

2

Page 3 of 47

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

ACS Biomaterials Science & Engineering

1. INTRODUCTION The mammalian wound healing response is a highly complex process involving orchestrated interactions between cells at the site of injury and the activation and recruitment of circulating cells with the eventual goal of promoting tissue repair while minimizing bleeding and infection. The ultimate goal from a tissue engineering or regenerative medicine perspective is to restore damaged or diseased tissue to a healthy, functional structure. Functional restoration can be achieved via two interconnected mechanisms: tissue repair and tissue regeneration. Adult tissue regeneration does not occur in some human tissue, but does occur more regularly in other species and during fetal wound healing1. Regeneration can be defined as the repair of a tissue to nearly identical function and structure of uninjured tissue through proliferation and activation of resident stem cells or progenitor cells and production of new matrix. Tissue repair is the coordinated mechanisms that restore tissue barriers and homeostasis. However, in the case of most wounds, this process results in unorganized matrix production and a higher density of cells, mostly fibroblasts. If the integrity of the extracellular matrix (ECM) is compromised, which occurs in wounds with large defects, cells produce unorganized matrix to close the wound quickly, resulting in the formation of scar tissue and a significant mismatch in morphology and appearance between the native tissue and the repaired tissue. In cases in which there is a pathological alteration of the wound healing response, there could be chronic deficiencies and inadequate healing that could lead to a poor quality of life. Around 6-7 million patients suffer from chronic wounds in the US with healthcare costs of managing such conditions totaling $25 billion2. Chronic or difficult to heal wounds have limited healing capacity that may be due to changes in the healthy cellular response that often leads to incomplete repair of the tissue and loss of function. Regardless of the type of wound, interventions through tissue engineering strategies aim to harness the inherent mammalian healing process to promote constructive tissue remodeling by providing one or a combination of: mechanical

ACS Paragon Plus Environment

3

ACS Biomaterials Science & Engineering

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

Page 4 of 47

support, bioactive signals, and cellular components1. The ultimate goal is the restoration of the native composition and function through the enhancement of the local wound healing process3. In recent years, there has been exciting research focused on the regenerative properties of naturally derived biomaterials obtained from the ECM of tissues. We define ECM scaffolds in this review as biomaterials prepared via the decellularization of target tissues using a combination of detergents and solutions that remove the cellular components while maintaining as much of the native structure of the ECM as possible4-7. ECM scaffolds are also characterized by the retention of similar mechanical properties to the tissue of origin and the retention of bioactive components (e.g. growth factors, peptides, etc.) capable of tissue regeneration via the recruitment and/or activation of surrounding cells8. Retention of mechanical properties and bioactive components after decellularization allow these materials to be tissue specific, which could have effects on tissue specific cell proliferation and differentiation9. One of the most attractive properties of ECM scaffolds is their ability to degrade during the host tissue response, shifting the normal mammalian wound healing response towards constructive remodeling and helping to avoid scar tissue formation10 (Figure 1). During the degradation process, ECM scaffolds release bioactive proteins and peptides that help activate and recruit local and circulating cells, beginning with the inherent wound healing process that ultimately leads to the generation of more organized and functional tissue10-12. Recent research has also emphasized the role of macrophage polarization and T-cell modulation as a result of ECM scaffold implantation13-15. For these reasons, ECM scaffolds have been used in a variety of applications5, 16 and have been adopted in clinical settings, resulting in an increasing number of medical devices composed of decellularized ECMs (reviewed elsewhere)17-19. However, there are situations in which there is incomplete remodeling when using only an acellular ECM scaffold. For example, an early study using a tubular form of an ECM scaffold for esophageal reconstruction benefited from the retention of native muscle layers during implantation to avoid stricture and improve the remodeling response20.

Another example is the use of an ECM

ACS Paragon Plus Environment

4

Page 5 of 47

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

ACS Biomaterials Science & Engineering

scaffold to repair ventricular wall defects in which studies show promising results towards remodeling of the infarcted ventricular wall but could not restore significant cardiac muscle21-23. In the case of the ECM scaffolds alone, there appears to be a passive mechanical effect that improves regional contractility of the tissue24. However, once the material is degraded, there is limited mechanical assistance and the muscle component is not completely replenished. This is not surprising given the large number of cells lost (~1 billion) after myocardial infarction25. For cardiac applications, it might be necessary to use therapeutic cells such as cardiac progenitor cells or cardiomyocytes derived from pluripotent stem cell sources to help regain the active cellular mass lost. For example, myocardial ECM scaffolds combined with bone marrow-derived mesenchymal stem cells (MSCs) show increased vascularization and potential healing when compared to the ECM alone26. Clinical studies have also shown the efficacy of c-kit

+

cardiac

progenitor cells and bone marrow-derived mesenchymal stem cells at promoting cardiac healing. In both cases, the amount of scar tissue was reduced, neovascularization increased and ventricular function was improved in the presence of therapeutic cells at the site of myocardial infarction27, 28. These studies show the importance of other passive components that should be considered in the case of ventricular remodeling, such as passive mechanics provided by the scaffold, and cells that may not contribute active forces but modulate the development of new tissue. In addition to supplementing cells that support the active component of functional cardiac tissue, it may also be important to supplement the passive cell population, such as myocardial fibroblasts, which regulate matrix production and aid in tissue regeneration29. This highlights the beneficial effects of a cellular component while harnessing the favorable effects of the ECM material (mechanically and bioactively). At this time, it is unclear how much cellular component is needed to achieve the best possible reparative outcome or stimulate regeneration, but it is most likely dependent on the target tissue being replaced. Therefore, current research should also focus on determining the need and/or benefit

ACS Paragon Plus Environment

5

ACS Biomaterials Science & Engineering

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

Page 6 of 47

of using a cellular component while weighing it against the safety and economic hurdles posed by such addition. While the use of multipotent stem cells has been established for years now, the targeted use of stem cells for tissue repair and regeneration have come into focus in the last decade as a potential cell treatment as multiple new multipotent cell types have been discovered and new technologies make the expansion of stem cells more efficient30-32. As previously stated, it has been thought that multipotent stem cells could help replenish some of the cellular components lost due to injury via in situ differentiation of recruited or delivered cells. However, most studies suggest that paracrine effects are the main mode of action by supporting angiogenesis and cell survival when using current therapeutic cells33, 34. Some studies have shown improvements in tissue function via the addition of specialized cells lost during injury that can only be obtained in sufficient numbers using pluripotent cell sources (i.e. myocardium)35-37. In addition, stem cells have been shown to promote a pro-healing macrophage phenotype by helping modulate macrophage activation, further supporting the notion of a regulatory function during wound healing38-42. Although stem cell therapy shows promise, there are still difficulties with its implementation and can benefit from further improvements on their retention and survival after implantation43. ECM scaffolds represent an ideal delivery biomaterial by providing a physiologically relevant substrate to the cells while helping modulate the site of healing, thus providing an ideal environment for stem cells to engraft and support reparative and regenerative processes. ECM scaffolds can also promote and aid in the differentiation of stem cells. In other words, therapies that include both stem cells and ECM components may have several advantages over acellular ECM scaffolds such as the ability to promote a pro-healing environment vital for tissue repair and regeneration5, 16, 44, 45. This review will provide a brief background on the composition and derivation of ECM scaffolds and their potential use as a stem cell delivery vehicle. We summarize the current efforts to harness this class of biomaterial and their inherent bioactivity to further develop new

ACS Paragon Plus Environment

6

Page 7 of 47

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

ACS Biomaterials Science & Engineering

tissue engineering strategies in combination with stem cells that can result in superior tissue remodeling for applications that may necessitate a cellular component. 2. Production and Processing of ECM Scaffolds One unique aspect of ECM scaffolds as biomaterials is the fact that they retain important material properties (e.g. mechanical properties, porosity, etc.) and biochemical information (e.g. growth factors, peptides, structural proteins, etc.) from the source tissue4,

46, 47

. In addition to

material properties, ECM scaffolds often retain many bioactive peptides and growth factors that, when released during the normal degradation process, result in the modulation of the host tissue response by affecting key cellular players such as macrophages, T-cells and resident progenitor cells. Therefore, it is important to briefly review the most common components found in these ECM scaffolds and how they relate to the overall properties of the biomaterial.

2.1. Composition of ECM Scaffolds The native ECM is an organized, acellular, three-dimensional network that fills the intercellular space and provides the necessary support for cells in tissues throughout the body48. Specifically, the ECM is composed of a variety of macromolecules in tissue-specific amounts that include collagen, elastin, laminin, soluble growth factors, and glycosaminoglycans49. The main fibrous ECM proteins are collagens, elastins, fibronectins and laminins. Adhesive proteins, such as fibronectin and laminins, play important roles in organization of the matrix and promoting cell attachment. Depending on the tissue of origin, the ECM scaffold may also retain the basement membrane50, 51 which provides a supportive layer for cells and aids in cell growth, migration, and differentiation52. Collagen is the main structural protein component of the ECM and is mostly secreted by fibroblasts49. Collagen fibrils bundle together to form fibers that provide most of the mechanical strength within the tissue. There are at least 29 types of collagens 53 with collagen type IV being the most abundant in the basement membrane and collagen type I the most abundant in the

ACS Paragon Plus Environment

7

ACS Biomaterials Science & Engineering

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

Page 8 of 47

interstitial matrix. Tissues such as tendons, ligaments, bone, skin, and cartilage consist largely of collagen. Due to collagen’s uniaxial mechanical properties, the orientation of collagen is extremely important in providing the overall and tissue-specific mechanical properties of ECM scaffolds54, 55. Other important structural components of the ECM include laminins, elastins, and glycosaminogylans.

Laminins are large glycoproteins found within the ECM and basement

membrane of ECM scaffolds51, 56, 57. Laminin is important for cellular adhesion and is often a desired component to be retained in the decellularized ECM. Elastin is another structural component that can improve elasticity of tissues. Elastin is especially abundant in skin and large elastic blood vessels, such as the aorta, and is key to the function of other mechanically active tissues such as: the lungs, the urinary bladder, and elastic cartilage. Glycosaminoglycans (GAGs) are long-chain polysaccharide based polymers and occur naturally in the ECM and connective tissue. GAGs swell in aqueous environments, providing shock absorption in reaction to compressive forces and act as lubricant in various soft tissues. GAGs also promote growth factor retention and modulate critical cell processes.58 Other components present in ECM that affect wound healing and tissue regeneration include: Hyaluronic Acid/Hyaluronan, Chondroitin Sulfate, Keratin Sulfate, Heparin Sulfate, and proteoglycans. The composition and role of all these components have been reviewed elsewhere5, 59, 60. However, given the diversity of ECM constituents among different tissues and their individual contribution to the inherent properties found within ECM scaffolds, it is important to understand the composition of each constituent and to determine how to maximize their retention during the derivation of ECM scaffolds to preserve tissue-specific signals. Another critical factor in the effectiveness of ECM-derived materials is the age of the animal from which the tissue for decellularization is obtained61-64. In vitro studies by Williams et al concluded that cardiac ECM derived from fetal rats had higher proliferation of neonatal cardiomyocytes when compared to ECM derived from neonatal and adult rats61. The possible

ACS Paragon Plus Environment

8

Page 9 of 47

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

ACS Biomaterials Science & Engineering

mechanism for this finding was described by the same group in a later study that examined the effect of partially solubilizing adult ECM. The partially solubilized ECM contained higher amounts of soluble matrix proteins, similar to fetal ECM, and led to the same increase in proliferation of neonatal cardiomyocytes62. Sicari et al also found that ECM materials derived from younger pigs (3 and 12 weeks old) showed a more constructive tissue response when compared to ECM derived from older pigs (26 and 52 weeks old), which was characterized by a lower collagen content after 180 days and lower M1/M2 macrophage ratio63. Tottey et al compared small intestine submucosa derived ECM (SIS-ECM) from different age source animals, and examined mechanical, compositional and structural characteristics of the derived scaffolds. This study found several differences, namely that stiffness of the ECM material was highest when the ECM was derived from 12 and 26 week old pigs. In addition, all groups showed chemotactic properties towards perivascular stem cells, with the highest migration in the scaffolds derived from 12 week old pigs. More proliferation was observed in the ECM of 52 week old pigs, and growth factor content was lower in scaffolds from 3 week old pigs, when compared to all of the other scaffold groups64. These studies highlight the importance of age and ECM composition on bioactivity and structure and re-enforces the need for further investigation to determine the optimal ECM source. While previous studies have shown that there may be differences in the mechanical properties of ECM based on the age of the donor animal64, these changes may not be as significant as other effects such as chemotactic properties and bioactivity, which are more inherent in ECM materials derived from younger sources.61, 62 The source will also be dictated by availability and cost which would make market weight animals more attractive as a tissue source. Furthermore, as mechanical and bioactive properties are direct functions of the decellularization process, the optimization of ECM material derivation should continue to be a topic of research. Ultimately, the most cost effective scaffolding material will most likely succeed in the clinical setting.

ACS Paragon Plus Environment

9

ACS Biomaterials Science & Engineering

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

Page 10 of 47

Growth and chemotactic factors are important parts of the signaling mechanisms present during the wound healing response. An inherent advantage of using ECM scaffolds is the presence of growth factors and chemotactic agents following decellularization has been shown to modulate the healing response by avoiding scar tissue formation. Growth factors control several processes, such as monocyte recruitment, macrophage polarization, cell differentiation and matrix production. In tissue-specific ECM scaffolds, these growth factors can be harnessed to direct local cell recruitment and differentiation, ultimately improving the regenerative response. The retention of growth factors is highly dependent on the processing and decellularization protocol used65 and may be correlated to the type of tissue that is being decellularized. In addition, degradation products of ECM have been found to affect progenitor cells but not differentiated cells. Reing et al used physiologically relevant enzymes pepsin and papain to degrade a decellularized urinary bladder matrix (UBM) to determine the chemotactic effects on blastema cells, human aortic endothelial cells and human microvascular endothelial cells10. The blastema cells had a significantly stronger chemotactic response over a control with no degraded ECM. Depending on the intended application, it is crucial to assess whether these factors are necessary and have remained in a sufficiently active conformation after decellularization and further processing.

2.2. ECM Scaffold Preparation Decellularization is the removal of cellular content from a tissue, that otherwise may cause an immune response when implanted in vivo such as cellular wall debris and DNA content46,

66, 67

. Keane et al demonstrated the requisite of near complete cellular removal by

decellularizing SIS-ECM at varying degrees and found that decellularized ECM with lower DNA content was better for promoting a pro-healing response rather than an inflammatory response when compared to materials with higher DNA content68. The decellularization process is characterized by a series of steps that may include multiple water or buffer washes, sonication,

ACS Paragon Plus Environment

10

Page 11 of 47

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

ACS Biomaterials Science & Engineering

electroporation, freeze/thaw cycles, enzymatic digestion, acid/base exposure or a combination of these4, 5, 9. A summary of decellularization processes is presented in Figure 2. A successful decellularization protocol will remove most the cellular content without damaging the native architecture of the ECM and can be performed on tissue sheets or on perfused organs9, 69-71. Methods for processing ECM scaffolds have been reviewed elsewhere 5, and include combining with polymers, lyophilized to make sheets, powdered, digested or made into hydrogels, (summarized in Figure 3). Overall, xenogenic ECM scaffolds are optimal due to their availability46 and reduced immunogenicity when properly decellularized46. Given the complexity of each tissue and its unique composition, decellularization protocols are customized and optimized for each type of tissue. For example, lungs can be best decellularized via the ionic detergent

3-((3-Cholamidopropyl)dimethylammonio)-1-propanesulfonate

(CHAPS),

urinary

bladders via peracetic acid, and vocal folds via a combination of enzymatic and ionic detergent treatments72-74. Cytotoxicity and cell proliferation assays such as DNA quantification and MTT assays are commonly used during the optimization of decellularization protocols to test for detrimental residues.

Ultimately, the objective is to determine which protocol adequately removes the

cellular material while maintaining the rest of the ECM intact so that the efficacy of the ECM material is maximized73-76. It has been suggested that sufficient decellularization is achieved when the DNA content found is no more than 200 base pairs and contains less than 50 ng of dsDNA per mg of dried ECM9. Histological staining can be used in combination with DNA quantification to determine the retention and distribution of common ECM components: Hematoxylin and Eosin (for nuclear and protein content), Movat’s Pentachrome (to determine the arrangement of collagen, elastic fibers, mucins, fibrin, and muscle fibers), and Alcian blue (for GAG content). During histological evaluation, decellularized ECM should have no visible nuclei present while showing visible retention of ECM components. Additional collagen

ACS Paragon Plus Environment

11

ACS Biomaterials Science & Engineering

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

Page 12 of 47

quantification and GAG colorimetric assays and lipid quantification can further examine the retention of these important ECM components. Recently mass spectrometry and proteomics have been used as additional methods to assess ECM quality and content77, 78, and have been reviewed by other groups79, 80. Recently, Li et al used proteomics to analyze several different ECM derived materials: Rat Tail Collagen Type 1, Growth Factor Reduced Matrigel®, decellularized rat liver, and decellularized human lung. They also analyzed the decellularized rat liver at several points throughout the decellularization process. While the decellularization techniques used were adequate for removal of nucleic content and disruption of cellular membranes, several plasma membrane and intermediate filament proteins remained. While the effects of these proteins are unclear, proteomics may be necessary to fully characterize the cellular environment of ECM materials77. Welham et al. characterized vocal fold mucosa scaffolds with 2D electrophoresis, and mass spectrometry. While the presence of structural and organizational proteins was observed, cellular proteins that are known to be bioactive and possibly antigenic were also present, even though there was no evidence of cellular proteins after histological and immuno-histochemical analysis78. These examples highlight the need for new methodologies to further characterize ECM scaffolds, leading to less variability in the final products and more stringent quality control for clinical translation. Hill et al. used a new methodology for proteomics analysis that was targeted at identifying specific proteins that were present within the ECM scaffold rather than a discovery based proteomic approach. Using ECM specific labeled peptides and chemical digestion to include insoluble proteins, this method improves repeatability and accuracy during the characterization of ECM materials81. If proteomic analysis could be included during the initial characterization alongside bioactivity, it could help researchers place priorities on specific components that are needed for optimal ECM derivation. Furthermore, proteomic analysis coupled with chemotactic and bioactive studies may highlight mechanisms that make one source material more advantageous, which could direct the manufacturing process81.

ACS Paragon Plus Environment

12

Page 13 of 47

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

ACS Biomaterials Science & Engineering

Decellularization can be performed at the whole organ level or on dissected pieces of tissues. If the decellularization is performed via perfusion of an organ, the ECM scaffold can be maintained with the native three-dimensional structure of the target organ9, 82-84. Alternatively, the decellularization process can be performed on pre-sliced tissue sections resulting in 2-D sheets that can be sutured onto the site of injury. There are extra steps that can be taken after the decellularization step such as dehydration and terminal sterilization to prepare the ECM scaffold for cell seeding or for implantation85-87. In addition, ECM scaffolds can be processed in a way that produces a hydrogel that can be used to encapsulate cells and self-assemble in situ88. Hydrogels can create a 3D environment during the delivery of cells via minimally invasive methods, possibly improving cellular function and survival. The production of ECM hydrogels largely affects the native ultrastructure of ECM proteins due to the digestion step. While this affects the quaternary and tertiary structures of ECM proteins, which affects the tissue’s mechanical properties and topographical features, matricryptic sites that are exposed during degradation could maintain some of the inherent bioactivity89. For instance, while studying the metabolic activity of kidney stem cells cultured on different forms of kidney papilla ECM (sheets, hydrogels, and solubilized ECM), similar metabolic activity was observed across all three forms of the ECM scaffolds, suggesting that composition of the stem cell niche was sufficient to affect cell metabolism when compared to ECM derived from the medulla and cortex90. These results show that the versatility of ECM scaffold spans from organ level to a highly processed level such as hydrogels. Large decellularized organs have the added advantage that they can be recellularized via perfusion of a cell suspension using the same approach used during the decellularization process making it an ideal closed loop system

70, 91-93

. Two dimensional sheets

or smaller tissues can be surface seeded in traditional culture conditions and used for in vitro testing or implantation

83

. Figure 3 summarizes various methods for processing 2D ECM

materials to provide different functions.

ACS Paragon Plus Environment

13

ACS Biomaterials Science & Engineering

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

Page 14 of 47

In summary, decellularized ECM materials have several advantages with respect to wound healing. Due to the removal of cellular content, there is almost no rejection of the material by the body’s immune system. As ECM scaffolds degrade, rather than altering the natural chemical content of the tissue, they have naturally occurring cryptic peptides and growth factors that enhance the overall host tissue response (Figure 1). This bioactivity can also be harnessed by combining with synthetic biomaterials to create bioactive hybrid scaffolds94.

3. ECM scaffolds and Tissue Healing There are a variety of commercial ECMs that are currently available for clinical use. Some of these examples include ECMs derived from the small intestine, urinary bladder (UBM), skin, pericardium and fasciae latae muscle, to name a few5. However, there is a lack of preclinical and clinical studies that address the necessity for the combination of an ECM scaffold with cells, let alone constructs with ECM and stem cells. The first tissue engineered construct with any living cells that was FDA approved for clinical use is Apligraf®, a “living skin equivalent” composed of a fibroblast dermal matrix layer supporting basal, spinous, granular and stratum corneum layers made of keratinocytes

95

. While other living tissue-engineered constructs have

been added to the market since then, the combination of extracellular matrix and stem cellbased therapies is relatively new and there are multiple efforts to harness the constructive remodeling found in ECM scaffolds with the therapeutic potential of stem cells. Obstacles to clinical translation of an ECM-stem cell product that should be considered include the safety and regulatory hurdles that will be encountered during FDA approval.

In addition, there are

important issues regarding safety and efficacy of the cellular component and the current cost of cell culture in a GMP environment that needs to be considered as well96. Although the manufacturing infrastructure is established for both components individually, combining the ECM and stem cell will pose new challenges that will need to be addressed and could influence its market readiness. The time required for the product to get to the market could also be

ACS Paragon Plus Environment

14

Page 15 of 47

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

ACS Biomaterials Science & Engineering

affected by the form of ECM chosen (whole organ vs sheets vs hydrogels). While recognizing the manufacturing and regulatory difficulties that pose the use of cells, the combination of ECM scaffolds and stem cells could result in improved regeneration in specific applications.

3.1 ECM Scaffold-Macrophage Interactions Recent studies have highlighted the role of macrophage polarization as a predictor of constructive remodeling and wound healing as a result of ECM scaffold implantation13, 46, 47, 97, 98. These studies suggest that macrophages are responsible, in part, for orchestrating the wound healing response. There is a large amount of heterogeneity in macrophage phenotypes, but they can be generally described by their activating signals and can mostly be sorted into two broad categories: pro-inflammatory and pro-healing3, reviewed elsewhere

46, 98-101

. These activation states are

99, 100, 102-104

. M1-like or pro-inflammatory macrophages make up most of

the initial macrophage population at the wound site and can be activated by interferon-gamma (IFN-γ) and bacterial wall debris

13

. These cells typically release pro-inflammatory signals such

as interlukin-1β (IL-1β) and tumor necrosis factor- α (TNF-α) in humans and nitric oxide synthase (iNOS) in mice

105

. In addition to playing a crucial role during inflammation, M1-like

macrophages recruit other cells to the wound, and play a role during matrix degradation. Over time, the macrophage population changes to a predominantly M2-like or pro-wound healing macrophage phenotype. There are different subtypes of M2-like macrophages that are classified depending on their activation signal99, 106. For example, M(IL-4, IL-13) macrophages (also referred to as M2a) are activated in vitro by interleukin-4 (IL-4) and interleukin-13 (IL-13) while M(IL-10) macrophages (also referred to as M2c) are activated in vitro by interleukin-10 (IL10). M2-like macrophages promote proliferation and differentiation of progenitor cells, ameliorate inflammatory processes, and stimulate matrix production107. It should be noted that the role of macrophages is dependent on the local environment and is described more as a spectrum of activation rather than a binary state108. In wound healing, both populations are

ACS Paragon Plus Environment

15

ACS Biomaterials Science & Engineering

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

Page 16 of 47

necessary and the timing of the transition is extremely important. For instance, while M1-like macrophages are necessary at the beginning of host tissue response, it is clear that with prolonged exposure there could be excess ECM degradation with impaired healing 109, 110 It is important to note, however, that there are exceptions to the biphasic appearances of M1-like and M2-like macrophages and that this is a general and highly simplified model

111, 112

.

ECM scaffolds have been reported to shift the macrophage polarization at the site of implantation towards an M2-like phenotype12,

113, 114

(Figure 1). The constructive remodeling

observed after the implantation of ECM scaffolds is in part attributed to this M2-like macrophage shift during pre-clinical studies46, 115. When ECM scaffolds are chemically cross-linked, resulting in diminished degradation and bioactivity when implanted, an M1-like macrophage population predominates, possibly leading to fibrous encapsulation and scar tissue formation116. The prohealing properties of minimally processed ECM scaffolds make them very attractive biomaterials and a focus of research using macrophages to screen ECM scaffold materials117. Synthetic materials, including biocompatible materials, often prolong the inflammatory process. This may be due to M1-like macrophages attempting to break down the material, which doesn’t occur as naturally as compared to ECM scaffold, and in some cases, results in encapsulation of the material

46

. This prolonged inflammatory response has an inhibitory effect

on tissue healing and regeneration. ECM scaffolds on the other hand, typically break down via secreted enzymes facilitating the transition from M1-like to M2-like macrophages and therefore improving the wound healing response. Sicari et al showed that solubilized SIS-ECM combined with perivascular stem cells (PVSCs) affected the chemotactic response of macrophages115. The study found that the macrophages cultured within the ECM hydrogel were biased towards the M2-like phenotype and the ECM scaffold could promote the migration and myogenesis of the PVSCs. Another study demonstrated that polypropylene mesh coated with porcine dermis and UBM hydrogels significantly reduced the M1 response in a rodent model when compared to polypropylene alone98. Dearth et al recently described a possible explanation of this dominant

ACS Paragon Plus Environment

16

Page 17 of 47

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

ACS Biomaterials Science & Engineering

M2-like response with ECM when exploring the effect of Aspirin, a common cyclooxygenase 1 and 2 (COX1/2) inhibitor118. The application of the COX 2 inhibitor to UBM hydrogel-treated in a rat model of skeletal muscle injury caused an increase in M1-like macrophage presence. This suggests that the COX2 pathway promotes a M2-like response when using an ECM scaffold. Interestingly, Dziki et al examined the response of bone marrow-derived macrophages towards ECM derived from various sources119 and found that macrophages exposed to dermal ECM were dominated by a M1-like phenotype, while brain, bladder, colon, small intestine and esophagus ECM had a M2-like phenotype. These studies further highlight the diversity of ECM compositions of different organs and tissues and the need to select appropriate scaffolds depending on the needs of the site of damaged tissue. Other immune cells are also affected by the components and/or degradation products of ECM. For instance, Bayrak et. al. investigated the gene expression of T-cells in response to various biomaterials. Native porcine tissue had the highest increase in cytotoxic T-cell proliferation and activation, while porcine ECMs with elastin still present had a significantly lower T-cell activation14. When comparing xenogenic tissue implants in mice with SIS implants, Thelper cells had more of a regenerative phenotype (Th-2) than mice who received other implants120. The modulation of T-helper cells by biomaterials has been reviewed elsewhere121. In models of volumetric muscle loss, ECM biomaterials skewed the ratio of CD4+:CD8+ towards the CD4+ regenerative phenotype when compared to control groups. In addition to skewing of the T-cell population, the ECM biomaterial also promoted release of IL-4 which is implicated with Th-2 and M2 macrophage responses. Sadtler et al concluded that the ECM promoted the T- Helper cell regenerative phenotype which further plays a role in the immune microenvironment, supporting the M2-like regenerative macrophage phenotype.15 This suggests that multiple mechanisms exist by which ECM materials promote a regenerative immune microenvironment.

ACS Paragon Plus Environment

17

ACS Biomaterials Science & Engineering

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

Page 18 of 47

3.2. Stem Cell-Macrophage Interactions An understanding of the inflammatory environment and how therapeutic cells interact with such environment is critical for a successful implantation of tissue-engineered construct comprised of an ECM scaffold and stem cells122. One potential therapeutic cell source are mesenchymal stromal cells or MSCs.

These cells can be harvested in a clinical setting,

commonly from the bone marrow or adipose tissue, and have been shown to have beneficial effects in clinical trials123,

124

. The immunomodulatory properties of MSCs has been reviewed

elsewhere123. However, in vivo and in vitro experiments using cells of the immune system and other repair cells remain necessary to gain a complete understanding of the potential interactions between a tissue-engineered construct and the inflammatory environment after injury. As previously stated, adult stem cells have been shown to modulate macrophage polarization by promoting more pro-healing phenotypes125-127. Recruitment of macrophage cells is one mechanism by which MSCs promote tissue regeneration. In vitro and in vivo studies showed that MSC pre-conditioned media had a higher chemotactic effect on macrophage cells, resulting in a larger population at the wound site, as well as promoting angiogenesis128. In response to the wound environment, MSCs can secrete cytokines that promote either pro-inflammatory or pro-healing macrophages.

For example,

when exposed to low concentrations of interleukin-6 (IL-6), MSCs produce interferon-gamma (IFN-γ) and interleukin-1 (IL-1) which promotes classically activated macrophages123. However, when MSCs are exposed to an environment with a high concentration of inflammatory cytokines, there is an increase in expression of pro-healing factors. Nemeth et. al. observed that MSCs promote a pro-healing response in vivo and in vitro through E2 prosteaglandin secretion which binds to receptors on macrophages to promote production of IL-10, cyclooxygenase 2, and TNF-α in a sepsis model126. MSCs are also implicated in polarization of macrophages to M2-like phenotypes125. Another study showed that macrophage associated cytokines (IFN-γ, TNFα, IL-1β and IL-6 for M1-like macrophages and IL10, TGFβ, VEGF for M2-like

ACS Paragon Plus Environment

18

Page 19 of 47

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

ACS Biomaterials Science & Engineering

macrophages) had different effect on the survival and viability of MSCs in vitro

41

. M2-like

macrophages associated cytokines were able to support MSC growth while M1-like macrophages and cytokines inhibited growth. When delivered to the site of myocardial infarct, MSCs promote an M2-like phenotype and the secretion of pro-healing cytokines40. In summary, while ECM scaffolds have immune modulating effects, stem cells also have similar effects on their own, re-enforcing the premise for stem cell seeded scaffolds. Another potential source for therapeutic cells are induced pluripotent stem cells (iPSCs). iPSCs are derived from somatic cells that have been reprogrammed to a pluripotent stem cell state via the expression of key transcription factors and have been reviewed elsewhere129. iPSCs could be derived from each patient making them an autologous therapy and thus avoiding immune-rejection (although this is still a subject of debate)130. Alternatively, iPSCs or human embryonic stem cells (hESCs) could be manufactured and stored in haplobanks and closely matched to the recipient131.

Regardless, the interactions between these potential

therapeutic cells and the host tissue is relatively unknown and a subject that should be explored. Recently, Pallotta et al investigated the effect of polarized human blood-derived macrophages on hESC-derived cardiomyocytes (hESC-CMs)132. The study showed that in a 3D invasion assay, hESC-CM conditioned media recruited pro-inflammatory M1-like macrophages due, in part, to hESC-CM-derived bone morphologic protein 4 (BMP4). Furthermore, the proinflammatory M1-like macrophages affected a subset of hESC-CM progenitor-like cells’ proliferation and maturation due to, in part, to macrophage-derived BMP proteins. These results highlight the potential to harness stem cells and inflammation to improve the host tissue response via macrophage modulation and tissue regeneration via stem cell differentiation. This then can be coupled with the beneficial effects of extracellular matrix scaffolds to create superior tissue engineered constructs.

3.3 ECM Scaffolds as a Delivery Vehicle for Stem Cells

ACS Paragon Plus Environment

19

ACS Biomaterials Science & Engineering

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

Page 20 of 47

Although ECM scaffolds have shown positive effects on the host tissue response when used as an implantable biomaterial on its own, there are applications that may result in superior tissue repair and regeneration if combined with a stem cell or stem cell derived-cell. Extracellular matrix components can have numerous effects on the differentiation of stem cells (see Table 1 for examples). Conversely, stem cells can also influence matrix production at the site of wound healing, making them both equally important for the goal of modulating the host tissue response. In chronic and difficult to heal wounds, several factors inhibit normal tissue regeneration, including: increased activity of matrix metallo-proteinases (MMPs) that break down the ECM, inhibition of ECM production, and modulation of activity in cells such as fibroblasts, keratinocytes, and leukocytes16. In addition, the components of the ECM are crucial in maintaining the stem cell niche in vivo, being an important target in directing stem cell recruitment3, 10. Integrin binding is highly specific to cell types and components of ECM, varies between tissue and helps to direct cell migration and differentiation. Minimally processed ECM materials can maintain these integrin binding properties and thus preserve their biological effects on the cells at the wound site58. If ECM scaffolds are used in combination with stem cells, there are several advantages over acellular materials such as the initial delivery of a cellular component that can help modulate the local wound healing environment without the need to wait for the recruitment of cells from neighboring tissues or from circulation. If designed properly, there can be a synergistic effect between the bioactivity inherent within the ECM scaffold and the modulating effects of the delivered stem cell. The choice of stem cell to use is still up for debate and will likely be dictated by safety and applicability concerns. Currently, MSCs derived from various sources and progenitor cells are popular choices since, for the most part, they can be harvested under clinical conditions and can often be used in an autologous fashion. For example, MSCs have shown modest improvements in some clinical studies mostly related to positive effects on the wound and host tissue response123. However, these adult stem cells fail to replenish the

ACS Paragon Plus Environment

20

Page 21 of 47

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

ACS Biomaterials Science & Engineering

musculature lost after a myocardial infarction. Pluripotent stem cells on the other hand have been shown to efficiently produce significant numbers of cells such as cardiomyocytes but are met with safety and feasibility limitations. Delivery of pluripotent stem cell-derived cells may benefit from the combination with extracellular matrix scaffold as a vehicle to improve their survival and maintain their differentiation state after implantation. Ultimately, the most efficient and safest cell should be used. ECM can also be used as a cell delivery system to provide structural support and to maintain the cells at the site of injection. Choi et al injected fluorescently labeled rabbit bone marrow-derived MSCs combined with SIS in a digested form as a delivery medium into rabbits with scars on the right vocal fold

133

. After 8 weeks, it was found that the MSCs injected within

the decellularized ECM showed higher engraftment and the vocal folds that received the MSC/ECM gels resulted in improved vibratory motions when compared to injections of MSCs or ECM alone. In this example, the MSCs were delivered to the injury site with a gelled ECM and were found to stay at the site and exhibit regenerative effects of the damaged vocal fold tissue. There have been several interesting studies where ECM scaffolds have been seeded with stem cells or progenitor cells. As discussed earlier, the composition and structure of ECMs varies from tissue to tissue, so studies have focused on these differences and on any impact on cell behavior. Heart ECM hydrogels, for example, have been shown to support the differentiation of hESC-CMs. Duan et al decellullarized porcine heart tissue and transformed decellularized myocardial slices into a hydrogel composed of digested myocardium and collagen type I

134

. hESCs were encapsulated within these hydrogels as embryoid bodies and

differentiated with or without supplemental growth factors. It was found that the cardiac ECM hydrogel without growth factors was sufficient to direct the differentiation of hESC-derived cardiac progenitor cells into hESC-CMs. This concept was expanded with the use of decellularized fetal and adult porcine heart ECM to aid in the maturation of iPSC-derived cardiomyocytes. ECM derived from cardiac tissue led to a higher expression in calcium pathway

ACS Paragon Plus Environment

21

ACS Biomaterials Science & Engineering

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

Page 22 of 47

genes, and promoted the proliferation of the cardiomyocytes derived from pluripotent stem cells135. Current methods to derive cardiomyocytes from iPSC tend to make CMs that are characteristically more fetal than adult. However, these studies are a promising step towards using ECM scaffolds to help maintain the differentiation of cells during the tissue healing process. In vitro applications for skeletal tissue have also shown benefits from using ECM materials for differentiation of progenitor and stem cells. Alom et al studied demineralized bovine tibias using 1:1 chloroform and methanol and subsequently decellularized via a 24 hour treatment of 0.05% Trypsin, 0.02% ethylenediaminetetraacetic acid (EDTA) and 1% pencillin/streptomycin in PBS136. The bone ECM (bECM) was digested, gelled and seeded with C2C12 mouse myoblasts. After 7 days of in vitro culture, there was a significant difference in the expression of osteogenic markers such as osteopontin and osteocalcin. The results suggested that bECM hydrogels have osteogenic-inducing properties. Ning et al decellularized canine Achilles tendons using repetitive freeze-thaw cycles and enzymatic digestions with RNase and DNase137. The study used two types of cells to test the ECM: rat bone marrow-derived stem cells (brMSCs) and rat tendon-derived stem cells (TDSCs). Both cell types were seeded onto lyophilized slices of tendon for 3, 7 and 14 days and assessed for cell viability, proliferation and possible differentiation. BMSCs seeded on tendons demonstrated an increase in the gene expression of tendon-specific genes such as scleraxis (SCX), tenomodulin (TNMD) and thrombospondin-4 TBS4 when compared to BMSCs seeded on tissue culture plastic. TDSCs seeded on tendons showed an increase in gene expression of TNMD, TBS4 and collagen type 1 (COL1) when compared to TDSCs seeded on tissue culture plastic further supporting the tissue specific effect of ECM scaffolds. As an additional example, Yuan et al used tissue specific hydrogels to inject therapeutic hMSCs into rat models of meniscus injury. Bovine meniscus tissue was minced, lyophilized and washed in SDS for decellularization, then digested to form a gel. The gels supported hMSC integration and the formation of fibrocartilaginous tissue138.

ACS Paragon Plus Environment

22

Page 23 of 47

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

ACS Biomaterials Science & Engineering

Decellularized tendon was altered using ultrasound to create a random-aligned-random structure to mimic a gradient found in native, healthy ligament. The biomimetic tendon material promoted the healing of the bone ligament junction which was characterized by higher expression of osteogenic and chondrogenic markers, and higher osteoinductivity with rabbit bone marrow stromal cells compared to the control139. Repair of skeletal tissue is a broad field where ECM scaffolds have great function due to these tissues consisting mostly of ECM in their mature form. Other organs have shown similar trends, such as liver and kidney. Studies by O’Neill et al90 and Batchelder et al140 highlight the potential influence of ECM scaffold on the phenotype of stem cells in renal tissue engineering. Since the kidney is comprised of three regions (cortex, medulla and papilla), O’Neill et al studied the effects of porcine kidney ECM derived from these three regions on kidney stem cell growth and behavior. This group showed that the ECM derived from these three regions affected cell growth and metabolic activity of mouse kidney stem cells (KSCs) and mouse MSCs. After decellularizing all three regions of the kidney, KSCs and MSCs were seeded on 2D sheets or on hydrogels. It was found that the papilla ECM, the location where KSCs normally reside, had a distinct effect on the metabolism and growth of KSCs but not on MSCs. Batchelder et al obtained monkey kidneys and used perfused solutions of heparin, PBS and 1% sodium dodecyl sulfate (SDS) for several days to the remove cellular content

140

. The sections of decellularized kidneys, as well as whole organs, were then

reseeded using hESCs in endothelial growth media and compared hESCs grown on embryoid bodies. Early renal developmental markers such as Wilms tumor 1 (WT1) and protein box 2 (PAX2) were expressed significantly higher on hESCs cultured on the renal ECM then on embryoid bodies, suggesting that renal ECM encourages hESCs to express early renal lineage markers. Additional studies have used iPSC-derived cells have also been coupled with ECM scaffolds. Wang et al

141

examined the effect of liver-derived ECM on the maturation of iPSC-

derived hepatocytes. Hepatocytes cultured in ECM were shown to have higher mRNA

ACS Paragon Plus Environment

23

ACS Biomaterials Science & Engineering

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

Page 24 of 47

expression of P450 and increased metabolic enzyme activity when compared to cells cultured on a 3D printed poly-L-lactic acid (PLLA) scaffolds infused with collagen I and cells cultured on collagen I alone. Both scaffolds were biocompatible and could promote bile canaliculi-like structures. These studies highlight how ECM structure can influence cell growth and behavior and the importance of selection the appropriate ECM depending on the applications. There are a few, but valuable studies that use iPSC-derived cells and ECM in an in vivo setting. Porcine iPSC-derived hepatocytes have been supplemented with decellularized porcine liver ECM containing liver-derived growth factors and transplanted into rats142. The cells attached well to the grafts and expressed hepatocyte markers such as albumin (ALB) and αfetoprotein (AFP). iPSC-derived lung epithelial progenitor cells have also been recently investigated in vivo. Decellularized whole human and rat lungs were seeded with lung epithelial progenitor cells generated from hiPSCs143. Cells cultured on decellularized lungs proliferated and expressed NK2 homeobox 1 (NKX2.1) when cultured in a bioreactor. Left lung transplants were performed and showed red blood cell perfusion throughout the alveolar capillary network with some hemorrhages. The same group used iPSC-derived endothelial cells and pericytes and efficiently repopulated the vasculature in decellularized lung ECM144. While there are not many studies that have explored use of iPSCs and ECM delivery vehicles, it remains a promising direction for the field. These studies will be more feasible as technologies advance and the culture of iPSCs becomes more efficient. Another technique for cell delivery with ECM scaffolds is 3D printing. Advances in 3D printing technology have allowed for the development of bio-inks that contain materials that form hydrogels in the right environment, and cells specific to the intended application. The use of ECM-derived materials in bio-inks has the inherent advantages of using tissue specific ECM and the ability to form 3D structures145. For instance, bioprinting of cardiac progenitor cells and MSCs in a bio-ink made from decellularized porcine heart supported improved cardiac function and tissue repair when compared to bio-ink made from collagen. Moreover, the ability to

ACS Paragon Plus Environment

24

Page 25 of 47

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

ACS Biomaterials Science & Engineering

engineer channels that promote vascularization in the patch also had beneficial effects for the delivered cells146. Structural control of the printed construct has also been used in the printing of skeletal muscle tissue using a bio-ink derived from decellularized muscle tissue147. Future directions and recent developments with 3D printing and ECM bio-inks has been reviewed elsewhere, and offers an promising area of research in the area of delivery of therapeutic cells145. Current challenges of using ECM scaffolds in combination with seeded stem cells include perfusion, especially with larger and whole organ scaffolds, and adjusting material stiffness to direct stem cell lineage. Whole organ perfusion and its challenges have been reviewed elsewhere9. For example, human hearts have been recently decellularized via perfusion and recellularized using iPSC-derived cardiomyocytes148. Material stiffness and composition is known to play a role in stem cell differentiation149 and how it applies to decellularized tissue remains to be investigated. In addition to the studies mentioned above, which focus on the use of ECM materials derived from decellularized tissue to deliver stem cells or progenitor cells, there are studies that use ECM to deliver and support primary cell lines such as cardiomyocytes150 and hepatocytes151, 152. In addition, studies have also been conducted that focus on the differentiation and the use of pluripotent cells with materials that mimic ECM, like fibrin cardiac patches153,

154

, hyaluronic acid materials150,

151, 155

and nanofiber scaffolds that

incorporate gradients156 . Future research should focus on optimizing these delivery vehicles and should be designed to drive tissue healing at the target site using the optimal combination of scaffolding material, therapeutic cells (if needed) and assembly paradigm.

4.0 Conclusions and Future Directions The advances in tissue engineering constructs using ECM scaffolds and stem cells remains promising, even at the face of the difficulties that hinder the progress of a combined ECM/cell-based regenerative approach to wound repair. Adult stem cells remain a viable clinical

ACS Paragon Plus Environment

25

ACS Biomaterials Science & Engineering

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

Page 26 of 47

choice as therapeutic cells. With regards to pluripotent stem cells, a major hurdle has been overcome in recent years by the discovery of iPSCs, which avoids ethical concerns raised with the use of hESCs and cells derived from somatic cell nuclear transfer. However, our limited understanding of their differentiation potential and cellular abnormalities as a result of the reprogramming step and prolonged culture times still represent a challenge. Recent publications have highlighted the potential to generate these cells under GMP conditions, making them clinically viable96. However, long term safety studies are still needed to understand the risks associated with pluripotent stem cells and to determine quality control strategies that would ensure the removal of non-differentiated cells that could form teratomas and ectopic tissues. Nevertheless, the recent use of pluripotent stem cells during clinical trials of macular degeneration and other diseases show promising results on their safety (reviewed elsewhere157). Other challenges faced by the clinical use of stem cells include immature cell differentiation, poor cell engraftment, incorporation of the construct with native tissues, and vascularization of the construct before and after implantation. For example, pluripotent stem cells can be differentiated very efficiently to cardiomyocytes and delivered to the infarct. However, these cells are still immature when compared to adult cardiomyocytes and the cells do not seem to mature as expected when injected into the heart25. Interestingly, beta cells derived from pluripotent stem cell sources were previously only matured in vivo can now be derived in vitro157. The limitation of each cell type and how ECM can be harnessed, will need to be determined to address these limitations. In addition, it is imperative to test biocompatibility of the scaffold material and it is necessary for any tissue-engineered construct to be tested in an in vivo environment to ensure no negative host tissue response. The inclusion of ECM with stem cells is a promising approach to tissue repair and regeneration. Progress has been made in regard to processing technologies and the methods are becoming more efficient. As a rapidly evolving and defining field, tissue engineering and the

ACS Paragon Plus Environment

26

Page 27 of 47

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

ACS Biomaterials Science & Engineering

constructs created with tissue engineering fundamentals, will advance the field of regenerative medicine through the enhancement of the local wound healing response.

5.0 Acknowledgements We thank our current funding sources: UNC/NCSU Joint Department of Biomedical Engineering, the Comparative Medicine Institute and NC TraCS 550KR141616. We also acknowledge our undergraduate research assistants, Andrew Baldwin and Brady Trevisan, for their help and contributions during the preparation of this review.

ACS Paragon Plus Environment

27

ACS Biomaterials Science & Engineering

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

Page 28 of 47

FIGURE CAPTIONS

A. Chemotaxis

B. Growth Factors

C. Cryptic Peptides Interactions with Cells

VEGF TGF-ß

Th-1 Inflammatory Lympohocytes

Th-2 Pro-regenerative Lympohocytes

D. Lymphocyte Response

M1-Like, Pro-inflammatory M2-Like, Pro-healing Macrophages Macrophages

E. Macrophage Response

Figure 1: Diagrammatic representation of how ECM Scaffolds promote tissue regeneration through several mechanisms. A) ECM scaffolds have chemotactic effect on progenitor cells. B) Growth Factors such as Transforming Growth Factor- ß and Vascular Endothelial Growth Factor are released from ECM scaffolds during degradation. These growth factors promote matrix production and angiogenesis, respectively. C) Cryptic, bioactive peptides of proteoglycans contained in the ECM scaffold are released as the scaffold degrades and can promote tissue regeneration processes. D) Lymphocytes are shifted to T-Helper 2 Lympohocytes which have more of a regenerative phenotype. E) ECM scaffolds promote a transition of macrophages from pro-inflammatory phenotype to the pro-healing phenotype. The pro-healing phenotype promotes cell differentiation and matrix production. Art adapted from Servier Medical Arts.

ACS Paragon Plus Environment

28

Page 29 of 47

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

ACS Biomaterials Science & Engineering

- 80°C +2 5 °C

+2 5 °C

- 80°C

Electroporation

Freeze/Thaw Cycles

Decellularization

Sterilization and washes

Enzymatic treatment

Sonication

Hypotonic or hypertonic solution Ionic and nonionic detergents

Figure 2: The decellularization process is characterized by a series of steps that may include multiple water or buffer washes, sonication, electroporation, freeze/thaw cycles, enzymatic digestion, acid/base exposure. These methods are usually used in combination for an effective decellularization.

ACS Paragon Plus Environment

29

ACS Biomaterials Science & Engineering

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

Hydrated Scaffold Cell seeding, structural support, implantation

Lyophilized Scaffold Cell Seeding, implantation

Hydrogel Cell seeding, encapsulation, implantation

Page 30 of 47

Lyophilized Powder Media supplementation

Digested ECM Vehicle for injection

Figure 3: ECM processing methods allow for these materials to be used in several applications. Hydrated scaffolds are the first product after decellularization of a 2D sheet and are used for cell seeding and implantation. After lyophilization, scaffolds have a much longer shelf-life and can be used similarly as hydrated scaffolds. Grinding the lyophilized sheet yields an ECM powder that can be used for media supplementation. Powder can also be digested and used for injection or in a mixture that gels at physiologic temperature creating a 3D microenvironment for cell seeding.

ACS Paragon Plus Environment

30

Page 31 of 47

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

ACS Biomaterials Science & Engineering

Table 1: Examples of studies that used ECM scaffolds with stem cells and showed differentiation specific to the tissue.

ACS Paragon Plus Environment

31

ACS Biomaterials Science & Engineering

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

Page 32 of 47

6. REFERENCES 1. Gurtner, G. C.; Werner, S.; Barrandon, Y.; Longaker, M. T., Wound repair and regeneration. Nature 2008, 453 (7193), 314-321. 2. Wong, V. W.; Gurtner, G. C., Tissue engineering for the management of chronic wounds: current concepts and future perspectives. Exp Dermatol 2012, 21 (10), 729-34. 3. Chen, F. M.; Liu, X., Advancing biomaterials of human origin for tissue engineering. Prog Polym Sci 2016, 53, 86-168. 4. Gilbert, T. W.; Sellaro, T. L.; Badylak, S. F., Decellularization of tissues and organs. Biomaterials 2006, 27 (19), 3675-83. 5. Badylak, S. F.; Freytes, D. O.; Gilbert, T. W., Extracellular matrix as a biological scaffold material: Structure and function. Acta Biomater 2009, 5 (1), 1-13. 6. White, L. J.; Taylor, A. J.; Faulk, D. M.; Keane, T. J.; Saldin, L. T.; Reing, J. E.; Swinehart, I. T.; Turner, N. J.; Ratner, B. D.; Badylak, S. F., The impact of detergents on the tissue decellularization process: A ToF-SIMS study. Acta Biomater 2016. 7. Starnecker, F.; Koenig, F.; Hagl, C.; Thierfelder, N., Tissue-engineering acellular scaffolds-The significant influence of physical and procedural decellularization factors. J Biomed Mater Res B Appl Biomater 2016. 8. Keane, T. J.; Swinehart, I. T.; Badylak, S. F., Methods of tissue decellularization used for preparation of biologic scaffolds and in vivo relevance. Methods 2015, 84, 25-34. 9. Crapo, P. M.; Gilbert, T. W.; Badylak, S. F., An overview of tissue and whole organ decellularization processes. Biomaterials 2011, 32 (12), 3233-43. 10. Reing, J. E.; Zhang, L.; Myers-Irvin, J.; Cordero, K. E.; Freytes, D. O.; Heber-Katz, E.; Bedelbaeva, K.; McIntosh, D.; Dewilde, A.; Braunhut, S. J.; Badylak, S. F., Degradation products of extracellular matrix affect cell migration and proliferation. Tissue Eng Part A 2009, 15 (3), 605-14. 11. Tottey, S.; Corselli, M.; Jeffries, E. M.; Londono, R.; Peault, B.; Badylak, S. F., Extracellular matrix degradation products and low-oxygen conditions enhance the regenerative potential of perivascular stem cells. Tissue Eng Part A 2011, 17 (1-2), 37-44. 12. Valentin, J. E.; Stewart-Akers, A. M.; Gilbert, T. W.; Badylak, S. F., Macrophage participation in the degradation and remodeling of extracellular matrix scaffolds. Tissue Eng Part A 2009, 15 (7), 1687-94. 13. Brown, B. N.; Ratner, B. D.; Goodman, S. B.; Amar, S.; Badylak, S. F., Macrophage polarization: an opportunity for improved outcomes in biomaterials and regenerative medicine. Biomaterials 2012, 33 (15), 3792-802. 14. Bayrak, A.; Tyralla, M.; Ladhoff, J.; Schleicher, M.; Stock, U. A.; Volk, H. D.; Seifert, M., Human immune responses to porcine xenogeneic matrices and their extracellular matrix constituents in vitro. Biomaterials 2010, 31 (14), 3793-3803. 15. Sadtler, K.; Estrellas, K.; Allen, B. W.; Wolf, M. T.; Fan, H. N.; Tam, A. J.; Patel, C. H.; Luber, B. S.; Wang, H.; Wagner, K. R.; Powell, J. D.; Housseau, F.; Pardoll, D. M.; Elisseeff, J. H., Developing a pro-regenerative biomaterial scaffold microenvironment requires T helper 2 cells. Science 2016, 352 (6283), 366-370. 16. Eweida, A. M.; Marei, M. K., Naturally Occurring Extracellular Matrix Scaffolds for Dermal Regeneration: Do They Really Need Cells? Biomed Res Int 2015, 2015, 839694. 17. Parmaksiz, M.; Dogan, A.; Odabas, S.; Elcin, A. E.; Elcin, Y. M., Clinical applications of decellularized extracellular matrices for tissue engineering and regenerative medicine. Biomed Mater 2016, 11 (2), 022003. 18. Song, L.; Murphy, S. V.; Yang, B.; Xu, Y.; Zhang, Y.; Atala, A., Bladder acellular matrix and its application in bladder augmentation. Tissue Eng Part B Rev 2014, 20 (2), 163-72. 19. Corona, B. T.; Greising, S. M., Challenges to acellular biological scaffold mediated skeletal muscle tissue regeneration. Biomaterials 2016, 104, 238-46.

ACS Paragon Plus Environment

32

Page 33 of 47

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

ACS Biomaterials Science & Engineering

20. Badylak, S. F.; Vorp, D. A.; Spievack, A. R.; Simmons-Byrd, A.; Hanke, J.; Freytes, D. O.; Thapa, A.; Gilbert, T. W.; Nieponice, A., Esophageal reconstruction with ECM and muscle tissue in a dog model. J Surg Res 2005, 128 (1), 87-97. 21. Badylak, S. F.; Kochupura, P. V.; Cohen, I. S.; Doronin, S. V.; Saltman, A. E.; Gilbert, T. W.; Kelly, D. J.; Ignotz, R. A.; Gaudette, G. R., The use of extracellular matrix as an inductive scaffold for the partial replacement of functional myocardium. Cell Transplant 2006, 15 Suppl 1, S29-40. 22. D'Amore, A.; Yoshizumi, T.; Luketich, S. K.; Wolf, M. T.; Gu, X.; Cammarata, M.; Hoff, R.; Badylak, S. F.; Wagner, W. R., Bi-layered polyurethane - Extracellular matrix cardiac patch improves ischemic ventricular wall remodeling in a rat model. Biomaterials 2016, 107, 1-14. 23. Kelly, D. J.; Rosen, A. B.; Schuldt, A. J.; Kochupura, P. V.; Doronin, S. V.; Potapova, I. A.; Azeloglu, E. U.; Badylak, S. F.; Brink, P. R.; Cohen, I. S.; Gaudette, G. R., Increased myocyte content and mechanical function within a tissue-engineered myocardial patch following implantation. Tissue Eng Part A 2009, 15 (8), 2189-201. 24. Kochupura, P. V.; Azeloglu, E. U.; Kelly, D. J.; Doronin, S. V.; Badylak, S. F.; Krukenkamp, I. B.; Cohen, I. S.; Gaudette, G. R., Tissue-engineered myocardial patch derived from extracellular matrix provides regional mechanical function. Circulation 2005, 112 (9), I144I149. 25. Chong, J. J.; Yang, X.; Don, C. W.; Minami, E.; Liu, Y. W.; Weyers, J. J.; Mahoney, W. M.; Van Biber, B.; Cook, S. M.; Palpant, N. J.; Gantz, J. A.; Fugate, J. A.; Muskheli, V.; Gough, G. M.; Vogel, K. W.; Astley, C. A.; Hotchkiss, C. E.; Baldessari, A.; Pabon, L.; Reinecke, H.; Gill, E. A.; Nelson, V.; Kiem, H. P.; Laflamme, M. A.; Murry, C. E., Human embryonic-stem-cellderived cardiomyocytes regenerate non-human primate hearts. Nature 2014, 510 (7504), 273-7. 26. Godier-Furnemont, A. F.; Martens, T. P.; Koeckert, M. S.; Wan, L.; Parks, J.; Arai, K.; Zhang, G.; Hudson, B.; Homma, S.; Vunjak-Novakovic, G., Composite scaffold provides a cell delivery platform for cardiovascular repair. Proc Natl Acad Sci U S A 2011, 108 (19), 7974-9. 27. Fazel, S.; Cimini, M.; Chen, L. W.; Li, S. H.; Angoulvant, D.; Fedak, P.; Verma, S.; Weisel, R. D.; Keating, A.; Li, R. K., Cardioprotective c-kit(+) cells are from the bone marrow and regulate the myocardial balance of angiogenic cytokines. Journal of Clinical Investigation 2006, 116 (7), 1865-1877. 28. Hare, J. M.; Traverse, J. H.; Henry, T. D.; Dib, N.; Strumpf, R. K.; Schulman, S. P.; Gerstenblith, G.; DeMaria, A. N.; Denktas, A. E.; Gammon, R. S.; Hermiller, J. B.; Reisman, M. A.; Schaer, G. L.; Sherman, W., A Randomized, Double-Blind, Placebo-Controlled, DoseEscalation Study of Intravenous Adult Human Mesenchymal Stem Cells (Prochymal) After Acute Myocardial Infarction. Journal of the American College of Cardiology 2009, 54 (24), 22772286. 29. Sullivan, K. E.; Black, L. D., The Role of Cardiac Fibroblasts in Extracellular MatrixMediated Signaling During Normal and Pathological Cardiac Development. Journal of Biomechanical Engineering-Transactions of the Asme 2013, 135 (7). 30. Mandai, M.; Fujii, M.; Hashiguchi, T.; Sunagawa, G. A.; Ito, S.; Sun, J.; Kaneko, J.; Sho, J.; Yamada, C.; Takahashi, M., iPSC-Derived Retina Transplants Improve Vision in rd1 EndStage Retinal-Degeneration Mice. Stem Cell Reports 2017, 8 (1), 69-83. 31. Hu, J.; Wang, Y.; Jiao, J.; Liu, Z.; Zhao, C.; Zhou, Z.; Zhang, Z.; Forde, K.; Wang, L.; Wang, J.; Baylink, D. J.; Zhang, X. B.; Gao, S.; Yang, B.; Chen, Y. E.; Ma, P. X., Patient-specific cardiovascular progenitor cells derived from integration-free induced pluripotent stem cells for vascular tissue regeneration. Biomaterials 2015, 73, 51-9. 32. Kurmann, A. A.; Serra, M.; Hawkins, F.; Rankin, S. A.; Mori, M.; Astapova, I.; Ullas, S.; Lin, S.; Bilodeau, M.; Rossant, J.; Jean, J. C.; Ikonomou, L.; Deterding, R. R.; Shannon, J. M.; Zorn, A. M.; Hollenberg, A. N.; Kotton, D. N., Regeneration of Thyroid Function by Transplantation of Differentiated Pluripotent Stem Cells. Cell Stem Cell 2015, 17 (5), 527-42.

ACS Paragon Plus Environment

33

ACS Biomaterials Science & Engineering

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

Page 34 of 47

33. Tilokee, E. L.; Latham, N.; Jackson, R.; Mayfield, A. E.; Ye, B.; Mount, S.; Lam, B. K.; Suuronen, E. J.; Ruel, M.; Stewart, D. J.; Davis, D. R., Paracrine Engineering of Human Explant-Derived Cardiac Stem Cells to Over-Express Stromal-Cell Derived Factor 1alpha Enhances Myocardial Repair. Stem Cells 2016, 34 (7), 1826-35. 34. Figeac, F.; Lesault, P. F.; Le Coz, O.; Damy, T.; Souktani, R.; Trebeau, C.; Schmitt, A.; Ribot, J.; Mounier, R.; Guguin, A.; Manier, C.; Surenaud, M.; Hittinger, L.; Dubois-Rande, J. L.; Rodriguez, A. M., Nanotubular crosstalk with distressed cardiomyocytes stimulates the paracrine repair function of mesenchymal stem cells. Stem Cells 2014, 32 (1), 216-30. 35. Abu-Hassan, D. W.; Li, X.; Ryan, E. I.; Acott, T. S.; Kelley, M. J., Induced pluripotent stem cells restore function in a human cell loss model of open-angle glaucoma. Stem Cells 2015, 33 (3), 751-61. 36. Gouadon, E.; Moore-Morris, T.; Smit, N. W.; Chatenoud, L.; Coronel, R.; Harding, S. E.; Jourdon, P.; Lambert, V.; Rucker-Martin, C.; Puceat, M., Concise Review: Pluripotent Stem Cell-Derived Cardiac Cells, A Promising Cell Source for Therapy of Heart Failure: Where Do We Stand? Stem Cells 2016, 34 (1), 34-43. 37. El Khatib, M. M.; Ohmine, S.; Jacobus, E. J.; Tonne, J. M.; Morsy, S. G.; Holditch, S. J.; Schreiber, C. A.; Uetsuka, K.; Fusaki, N.; Wigle, D. A.; Terzic, A.; Kudva, Y. C.; Ikeda, Y., Tumor-Free Transplantation of Patient-Derived Induced Pluripotent Stem Cell Progeny for Customized Islet Regeneration. Stem Cells Transl Med 2016, 5 (5), 694-702. 38. Adutler-Lieber, S.; Ben-Mordechai, T.; Naftali-Shani, N.; Asher, E.; Loberman, D.; Raanani, E.; Leor, J., Human macrophage regulation via interaction with cardiac adipose tissuederived mesenchymal stromal cells. J Cardiovasc Pharmacol Ther 2013, 18 (1), 78-86. 39. Cho, D. I.; Kim, M. R.; Jeong, H. Y.; Jeong, H. C.; Jeong, M. H.; Yoon, S. H.; Kim, Y. S.; Ahn, Y., Mesenchymal stem cells reciprocally regulate the M1/M2 balance in mouse bone marrow-derived macrophages. Exp Mol Med 2014, 46, e70. 40. Dayan, V.; Yannarelli, G.; Billia, F.; Filomeno, P.; Wang, X. H.; Davies, J. E.; Keating, A., Mesenchymal stromal cells mediate a switch to alternatively activated monocytes/macrophages after acute myocardial infarction. Basic Res Cardiol 2011, 106 (6), 1299-310. 41. Freytes, D. O.; Kang, J. W.; Marcos-Campos, I.; Vunjak-Novakovic, G., Macrophages modulate the viability and growth of human mesenchymal stem cells. J Cell Biochem 2013, 114 (1), 220-9. 42. Bartosh, T. J.; Ylostalo, J. H.; Mohammadipoor, A.; Bazhanov, N.; Coble, K.; Claypool, K.; Lee, R. H.; Choi, H.; Prockop, D. J., Aggregation of human mesenchymal stromal cells (MSCs) into 3D spheroids enhances their antiinflammatory properties. Proc Natl Acad Sci U S A 2010, 107 (31), 13724-9. 43. Roche, E. T.; Hastings, C. L.; Lewin, S. A.; Shvartsman, D. E.; Brudno, Y.; Vasilyev, N. V.; O'Brien, F. J.; Walsh, C. J.; Duffy, G. P.; Mooney, D. J., Comparison of biomaterial delivery vehicles for improving acute retention of stem cells in the infarcted heart. Biomaterials 2014, 35 (25), 6850-8. 44. Bhowmick, S.; Scharnweber, D.; Koul, V., Co-cultivation of keratinocyte-human mesenchymal stem cell (hMSC) on sericin loaded electrospun nanofibrous composite scaffold (cationic gelatin/hyaluronan/chondroitin sulfate) stimulates epithelial differentiation in hMSCs: In vitro study. Biomaterials 2016, 88, 83-96. 45. Brown, B. N.; Sicari, B. M.; Badylak, S. F., Rethinking regenerative medicine: a macrophage-centered approach. Front Immunol 2014, 5, 510. 46. Badylak, S. F., Decellularized allogeneic and xenogeneic tissue as a bioscaffold for regenerative medicine: factors that influence the host response. Ann Biomed Eng 2014, 42 (7), 1517-27. 47. Brown, B. N.; Badylak, S. F., Expanded applications, shifting paradigms and an improved understanding of host-biomaterial interactions. Acta Biomater 2013, 9 (2), 4948-55.

ACS Paragon Plus Environment

34

Page 35 of 47

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

ACS Biomaterials Science & Engineering

48. Geckil, H.; Xu, F.; Zhang, X.; Moon, S.; Demirci, U., Engineering hydrogels as extracellular matrix mimics. Nanomedicine (Lond) 2010, 5 (3), 469-84. 49. Frantz, C.; Stewart, K. M.; Weaver, V. M., The extracellular matrix at a glance. J Cell Sci 2010, 123 (Pt 24), 4195-200. 50. Faulk, D. M.; Carruthers, C. A.; Warner, H. J.; Kramer, C. R.; Reing, J. E.; Zhang, L.; D'Amore, A.; Badylak, S. F., The effect of detergents on the basement membrane complex of a biologic scaffold material. Acta Biomater 2014, 10 (1), 183-93. 51. Brown, B.; Lindberg, K.; Reing, J.; Stolz, D. B.; Badylak, S. F., The basement membrane component of biologic scaffolds derived from extracellular matrix. Tissue Eng 2006, 12 (3), 51926. 52. Kruegel, J.; Miosge, N., Basement membrane components are key players in specialized extracellular matrices. Cell Mol Life Sci 2010, 67 (17), 2879-95. 53. Chattopadhyay, S.; Raines, R. T., Review collagen-based biomaterials for wound healing. Biopolymers 2014, 101 (8), 821-33. 54. Gilbert, T. W.; Wognum, S.; Joyce, E. M.; Freytes, D. O.; Sacks, M. S.; Badylak, S. F., Collagen fiber alignment and biaxial mechanical behavior of porcine urinary bladder derived extracellular matrix. Biomaterials 2008, 29 (36), 4775-82. 55. Gilbert, T. W.; Sacks, M. S.; Grashow, J. S.; Woo, S. L.; Badylak, S. F.; Chancellor, M. B., Fiber kinematics of small intestinal submucosa under biaxial and uniaxial stretch. J Biomech Eng 2006, 128 (6), 890-8. 56. Halper, J.; Kjaer, M., Basic components of connective tissues and extracellular matrix: elastin, fibrillin, fibulins, fibrinogen, fibronectin, laminin, tenascins and thrombospondins. Adv Exp Med Biol 2014, 802, 31-47. 57. Barnes, C. A.; Brison, J.; Michel, R.; Brown, B. N.; Castner, D. G.; Badylak, S. F.; Ratner, B. D., The surface molecular functionality of decellularized extracellular matrices. Biomaterials 2011, 32 (1), 137-43. 58. Pellowe, A. S.; Gonzalez, A. L., Extracellular matrix biomimicry for the creation of investigational and therapeutic devices. Wiley Interdiscip Rev Nanomed Nanobiotechnol 2016, 8 (1), 5-22. 59. Kim, S. H.; Turnbull, J.; Guimond, S., Extracellular matrix and cell signalling: the dynamic cooperation of integrin, proteoglycan and growth factor receptor. J Endocrinol 2011, 209 (2), 139-51. 60. Bonnans, C.; Chou, J.; Werb, Z., Remodelling the extracellular matrix in development and disease. Nat Rev Mol Cell Biol 2014, 15 (12), 786-801. 61. Williams, C.; Quinn, K. P.; Georgakoudi, I.; Black, L. D., Young developmental age cardiac extracellular matrix promotes the expansion of neonatal cardiomyocytes in vitro. Acta Biomaterialia 2014, 10 (1), 194-204. 62. Williams, C.; Sullivan, K.; Black, L. D., Partially Digested Adult Cardiac Extracellular Matrix Promotes Cardiomyocyte Proliferation In Vitro. Advanced Healthcare Materials 2015, 4 (10), 1545-1554. 63. Sicari, B. M.; Johnson, S. A.; Siu, B. F.; Crapo, P. M.; Daly, K. A.; Jiang, H. B.; Medberry, C. J.; Tottey, S.; Turner, N. J.; Badylak, S. E., The effect of source animal age upon the in vivo remodeling characteristics of an extracellular matrix scaffold. Biomaterials 2012, 33 (22), 5524-5533. 64. Tottey, S.; Johnson, S. A.; Crapo, P. M.; Reing, J. E.; Zhang, L.; Jiang, H.; Medberry, C. J.; Reines, B.; Badylak, S. F., The effect of source animal age upon extracellular matrix scaffold properties. Biomaterials 2011, 32 (1), 128-36. 65. Reing, J. E.; Brown, B. N.; Daly, K. A.; Freund, J. M.; Gilbert, T. W.; Hsiong, S. X.; Huber, A.; Kullas, K. E.; Tottey, S.; Wolf, M. T.; Badylak, S. F., The effects of processing methods upon mechanical and biologic properties of porcine dermal extracellular matrix scaffolds. Biomaterials 2010, 31 (33), 8626-33.

ACS Paragon Plus Environment

35

ACS Biomaterials Science & Engineering

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

Page 36 of 47

66. Keane, T. J.; Badylak, S. F., The host response to allogeneic and xenogeneic biological scaffold materials. J Tissue Eng Regen Med 2015, 9 (5), 504-11. 67. Wong, M. L.; Griffiths, L. G., Immunogenicity in xenogeneic scaffold generation: antigen removal vs. decellularization. Acta Biomater 2014, 10 (5), 1806-16. 68. Keane, T. J.; Londono, R.; Turner, N. J.; Badylak, S. F., Consequences of ineffective decellularization of biologic scaffolds on the host response. Biomaterials 2012, 33 (6), 1771-81. 69. Guyette, J. P.; Gilpin, S. E.; Charest, J. M.; Tapias, L. F.; Ren, X.; Ott, H. C., Perfusion decellularization of whole organs. Nat Protoc 2014, 9 (6), 1451-68. 70. Goh, S. K.; Bertera, S.; Olsen, P.; Candiello, J. E.; Halfter, W.; Uechi, G.; Balasubramani, M.; Johnson, S. A.; Sicari, B. M.; Kollar, E.; Badylak, S. F.; Banerjee, I., Perfusion-decellularized pancreas as a natural 3D scaffold for pancreatic tissue and whole organ engineering. Biomaterials 2013, 34 (28), 6760-72. 71. Hou, N.; Cui, P.; Luo, J.; Ma, R.; Zhu, L., Tissue-engineered larynx using perfusiondecellularized technique and mesenchymal stem cells in a rabbit model. Acta Otolaryngol 2011, 131 (6), 645-52. 72. O'Neill, J. D.; Anfang, R.; Anandappa, A.; Costa, J.; Javidfar, J.; Wobma, H. M.; Singh, G.; Freytes, D. O.; Bacchetta, M. D.; Sonett, J. R.; Vunjak-Novakovic, G., Decellularization of human and porcine lung tissues for pulmonary tissue engineering. Ann Thorac Surg 2013, 96 (3), 1046-55; discussion 1055-6. 73. Wrona, E. A.; Peng, R.; Born, H.; Amin, M. R.; Branski, R. C.; Freytes, D. O., Derivation and characterization of porcine vocal fold extracellular matrix scaffold. Laryngoscope 2016, 126 (4), 928-35. 74. Freytes, D. O.; Martin, J.; Velankar, S. S.; Lee, A. S.; Badylak, S. F., Preparation and rheological characterization of a gel form of the porcine urinary bladder matrix. Biomaterials 2008, 29 (11), 1630-7. 75. Wolf, M. T.; Daly, K. A.; Reing, J. E.; Badylak, S. F., Biologic scaffold composed of skeletal muscle extracellular matrix. Biomaterials 2012, 33 (10), 2916-25. 76. Perea-Gil, I.; Uriarte, J. J.; Prat-Vidal, C.; Galvez-Monton, C.; Roura, S.; LluciaValldeperas, A.; Soler-Botija, C.; Farre, R.; Navajas, D.; Bayes-Genis, A., In vitro comparative study of two decellularization protocols in search of an optimal myocardial scaffold for recellularization. Am J Transl Res 2015, 7 (3), 558-73. 77. Li, Q. Y.; Uygun, B. E.; Geerts, S.; Ozer, S.; Scalf, M.; Gilpin, S. E.; Ott, H. C.; Yarmush, M. L.; Smith, L. M.; Welham, N. V.; Frey, B. L., Proteomic analysis of naturally-sourced biological scaffolds. Biomaterials 2016, 75, 37-46. 78. Welham, N. V.; Chang, Z.; Smith, L. M.; Frey, B. L., Proteomic analysis of a decellularized human vocal fold mucosa scaffold using 2D electrophoresis and high-resolution mass spectrometry. Biomaterials 2013, 34 (3), 669-676. 79. Naba, A.; Clauser, K. R.; Hynes, R. O., Enrichment of Extracellular Matrix Proteins from Tissues and Digestion into Peptides for Mass Spectrometry Analysis. Jove-Journal of Visualized Experiments 2015, (101). 80. Barallobre-Barreiro, J.; Lynch, M.; Yin, X. K.; Mayr, M., Systems biology-opportunities and challenges: the application of proteomics to study the cardiovascular extracellular matrix. Cardiovascular Research 2016, 112 (3), 626-636. 81. Hill, R. C.; Calle, E. A.; Dzieciatkowska, M.; Niklason, L. E.; Hansen, K. C., Quantification of Extracellular Matrix Proteins from a Rat Lung Scaffold to Provide a Molecular Readout for Tissue Engineering. Molecular & Cellular Proteomics 2015, 14 (4), 961-973. 82. Faulk, D. M.; Wildemann, J. D.; Badylak, S. F., Decellularization and cell seeding of whole liver biologic scaffolds composed of extracellular matrix. J Clin Exp Hepatol 2015, 5 (1), 69-80. 83. Faulk, D. M.; Johnson, S. A.; Zhang, L.; Badylak, S. F., Role of the extracellular matrix in whole organ engineering. J Cell Physiol 2014, 229 (8), 984-9.

ACS Paragon Plus Environment

36

Page 37 of 47

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

ACS Biomaterials Science & Engineering

84. Arenas-Herrera, J. E.; Ko, I. K.; Atala, A.; Yoo, J. J., Decellularization for whole organ bioengineering. Biomed Mater 2013, 8 (1), 014106. 85. Freytes, D. O.; Tullius, R. S.; Valentin, J. E.; Stewart-Akers, A. M.; Badylak, S. F., Hydrated versus lyophilized forms of porcine extracellular matrix derived from the urinary bladder. J Biomed Mater Res A 2008, 87 (4), 862-72. 86. Freytes, D. O.; Stoner, R. M.; Badylak, S. F., Uniaxial and biaxial properties of terminally sterilized porcine urinary bladder matrix scaffolds. J Biomed Mater Res B Appl Biomater 2008, 84 (2), 408-14. 87. Freytes, D. O.; Tullius, R. S.; Badylak, S. F., Effect of storage upon material properties of lyophilized porcine extracellular matrix derived from the urinary bladder. J Biomed Mater Res B Appl Biomater 2006, 78 (2), 327-33. 88. Saldin, L. T.; Cramer, M. C.; Velankar, S. S.; White, L. J.; Badylak, S. F., Extracellular matrix hydrogels from decellularized tissues: Structure and function. Acta Biomater 2017, 49, 115. 89. Davis, G. E.; Bayless, K. J.; Davis, M. J.; Meininger, G. A., Regulation of tissue injury responses by the exposure of matricryptic sites within extracellular matrix molecules. American Journal of Pathology 2000, 156 (5), 1489-1498. 90. O'Neill, J. D.; Freytes, D. O.; Anandappa, A. J.; Oliver, J. A.; Vunjak-Novakovic, G. V., The regulation of growth and metabolism of kidney stem cells with regional specificity using extracellular matrix derived from kidney. Biomaterials 2013, 34 (38), 9830-41. 91. Uygun, B. E.; Soto-Gutierrez, A.; Yagi, H.; Izamis, M. L.; Guzzardi, M. A.; Shulman, C.; Milwid, J.; Kobayashi, N.; Tilles, A.; Berthiaume, F.; Hertl, M.; Nahmias, Y.; Yarmush, M. L.; Uygun, K., Organ reengineering through development of a transplantable recellularized liver graft using decellularized liver matrix. Nat Med 2010, 16 (7), 814-20. 92. Ott, H. C.; Matthiesen, T. S.; Goh, S. K.; Black, L. D.; Kren, S. M.; Netoff, T. I.; Taylor, D. A., Perfusion-decellularized matrix: using nature's platform to engineer a bioartificial heart. Nat Med 2008, 14 (2), 213-21. 93. Gilpin, S. E.; Charest, J. M.; Ren, X.; Tapias, L. F.; Wu, T.; Evangelista-Leite, D.; Mathisen, D. J.; Ott, H. C., Regenerative potential of human airway stem cells in lung epithelial engineering. Biomaterials 2016, 108, 111-9. 94. Stankus, J. J.; Freytes, D. O.; Badylak, S. F.; Wagner, W. R., Hybrid nanofibrous scaffolds from electrospinning of a synthetic biodegradable elastomer and urinary bladder matrix. J Biomater Sci Polym Ed 2008, 19 (5), 635-52. 95. Falanga, V.; Sabolinski, M., A bilayered living skin construct (APLIGRAF) accelerates complete closure of hard-to-heal venous ulcers. Wound Repair Regen 1999, 7 (4), 201-7. 96. Baghbaderani, B. A.; Tian, X.; Neo, B. H.; Burkall, A.; Dimezzo, T.; Sierra, G.; Zeng, X.; Warren, K.; Kovarcik, D. P.; Fellner, T.; Rao, M. S., cGMP-Manufactured Human Induced Pluripotent Stem Cells Are Available for Pre-clinical and Clinical Applications. Stem Cell Reports 2015, 5 (4), 647-59. 97. Wolf, M. T.; Vodovotz, Y.; Tottey, S.; Brown, B. N.; Badylak, S. F., Predicting in vivo responses to biomaterials via combined in vitro and in silico analysis. Tissue Eng Part C Methods 2015, 21 (2), 148-59. 98. Wolf, M. T.; Dearth, C. L.; Ranallo, C. A.; LoPresti, S. T.; Carey, L. E.; Daly, K. A.; Brown, B. N.; Badylak, S. F., Macrophage polarization in response to ECM coated polypropylene mesh. Biomaterials 2014, 35 (25), 6838-49. 99. Murray, P. J.; Allen, J. E.; Biswas, S. K.; Fisher, E. A.; Gilroy, D. W.; Goerdt, S.; Gordon, S.; Hamilton, J. A.; Ivashkiv, L. B.; Lawrence, T.; Locati, M.; Mantovani, A.; Martinez, F. O.; Mege, J. L.; Mosser, D. M.; Natoli, G.; Saeij, J. P.; Schultze, J. L.; Shirey, K. A.; Sica, A.; Suttles, J.; Udalova, I.; van Ginderachter, J. A.; Vogel, S. N.; Wynn, T. A., Macrophage activation and polarization: nomenclature and experimental guidelines. Immunity 2014, 41 (1), 14-20.

ACS Paragon Plus Environment

37

ACS Biomaterials Science & Engineering

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

Page 38 of 47

100. Nahrendorf, M.; Swirski, F. K., Monocyte and macrophage heterogeneity in the heart. Circ Res 2013, 112 (12), 1624-33. 101. Gordon, S.; Taylor, P. R., Monocyte and macrophage heterogeneity. Nat Rev Immunol 2005, 5 (12), 953-64. 102. Mantovani, A.; Biswas, S. K.; Galdiero, M. R.; Sica, A.; Locati, M., Macrophage plasticity and polarization in tissue repair and remodelling. J Pathol 2013, 229 (2), 176-85. 103. Lawrence, T.; Natoli, G., Transcriptional regulation of macrophage polarization: enabling diversity with identity. Nat Rev Immunol 2011, 11 (11), 750-61. 104. Zhou, D.; Huang, C.; Lin, Z.; Zhan, S.; Kong, L.; Fang, C.; Li, J., Macrophage polarization and function with emphasis on the evolving roles of coordinated regulation of cellular signaling pathways. Cell Signal 2014, 26 (2), 192-7. 105. Spiller, K. L.; Wrona, E. A.; Romero-Torres, S.; Pallotta, I.; Graney, P. L.; Witherel, C. E.; Panicker, L. M.; Feldman, R. A.; Urbanska, A. M.; Santambrogio, L.; Vunjak-Novakovic, G.; Freytes, D. O., Differential gene expression in human, murine, and cell line-derived macrophages upon polarization. Exp Cell Res 2016, 347 (1), 1-13. 106. Mantovani, A.; Sica, A.; Sozzani, S.; Allavena, P.; Vecchi, A.; Locati, M., The chemokine system in diverse forms of macrophage activation and polarization. Trends Immunol 2004, 25 (12), 677-86. 107. Gong, D.; Shi, W.; Yi, S. J.; Chen, H.; Groffen, J.; Heisterkamp, N., TGFbeta signaling plays a critical role in promoting alternative macrophage activation. BMC Immunol 2012, 13, 31. 108. Mosser, D. M.; Edwards, J. P., Exploring the full spectrum of macrophage activation. Nat Rev Immunol 2008, 8 (12), 958-69. 109. Lucas, T.; Waisman, A.; Ranjan, R.; Roes, J.; Krieg, T.; Muller, W.; Roers, A.; Eming, S. A., Differential roles of macrophages in diverse phases of skin repair. J Immunol 2010, 184 (7), 3964-77. 110. Brancato, S. K.; Albina, J. E., Wound macrophages as key regulators of repair: origin, phenotype, and function. Am J Pathol 2011, 178 (1), 19-25. 111. Villalta, S. A.; Nguyen, H. X.; Deng, B.; Gotoh, T.; Tidball, J. G., Shifts in macrophage phenotypes and macrophage competition for arginine metabolism affect the severity of muscle pathology in muscular dystrophy. Hum Mol Genet 2009, 18 (3), 482-96. 112. Mahbub, S.; Deburghgraeve, C. R.; Kovacs, E. J., Advanced age impairs macrophage polarization. J Interferon Cytokine Res 2012, 32 (1), 18-26. 113. Brown, B. N.; Valentin, J. E.; Stewart-Akers, A. M.; McCabe, G. P.; Badylak, S. F., Macrophage phenotype and remodeling outcomes in response to biologic scaffolds with and without a cellular component. Biomaterials 2009, 30 (8), 1482-91. 114. Badylak, S. F.; Valentin, J. E.; Ravindra, A. K.; McCabe, G. P.; Stewart-Akers, A. M., Macrophage phenotype as a determinant of biologic scaffold remodeling. Tissue Eng Part A 2008, 14 (11), 1835-42. 115. Sicari, B. M.; Dziki, J. L.; Siu, B. F.; Medberry, C. J.; Dearth, C. L.; Badylak, S. F., The promotion of a constructive macrophage phenotype by solubilized extracellular matrix. Biomaterials 2014, 35 (30), 8605-12. 116. Daly, K. A.; Liu, S.; Agrawal, V.; Brown, B. N.; Johnson, S. A.; Medberry, C. J.; Badylak, S. F., Damage associated molecular patterns within xenogeneic biologic scaffolds and their effects on host remodeling. Biomaterials 2012, 33 (1), 91-101. 117. Witherel, C. E.; Graney, P. L.; Freytes, D. O.; Weingarten, M. S.; Spiller, K. L., Response of human macrophages to wound matrices in vitro. Wound Repair Regen 2016, 24 (3), 514-24. 118. Dearth, C. L.; Slivka, P. F.; Stewart, S. A.; Keane, T. J.; Tay, J. K.; Londono, R.; Goh, Q.; Pizza, F. X.; Badylak, S. F., Inhibition of COX1/2 alters the host response and reduces ECM scaffold mediated constructive tissue remodeling in a rodent model of skeletal muscle injury. Acta Biomater 2016, 31, 50-60.

ACS Paragon Plus Environment

38

Page 39 of 47

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

ACS Biomaterials Science & Engineering

119. Dziki, J. L.; Wang, D. S.; Pineda, C.; Sicari, B. M.; Rausch, T.; Badylak, S. F., Solubilized extracellular matrix bioscaffolds derived from diverse source tissues differentially influence macrophage phenotype. J Biomed Mater Res A 2017, 105 (1), 138-147. 120. Allman, A. J.; McPherson, T. B.; Badylak, S. F.; Merrill, L. C.; Kallakury, B.; Sheehan, C.; Raeder, R. H.; Metzger, D. W., Xenogeneic extracellular matrix grafts elicit a TH2-restricted immune response. Transplantation 2001, 71 (11), 1631-40. 121. Badylak, S. F.; Gilbert, T. W., Immune response to biologic scaffold materials. Semin Immunol 2008, 20 (2), 109-16. 122. Freytes, D. O.; Santambrogio, L.; Vunjak-Novakovic, G., Optimizing dynamic interactions between a cardiac patch and inflammatory host cells. Cells Tissues Organs 2012, 195 (1-2), 171-82. 123. Bernardo, M. E.; Fibbe, W. E., Mesenchymal stromal cells: sensors and switchers of inflammation. Cell Stem Cell 2013, 13 (4), 392-402. 124. Karpov, A. A.; Udalova, D. V.; Pliss, M. G.; Galagudza, M. M., Can the outcomes of mesenchymal stem cell-based therapy for myocardial infarction be improved? Providing weapons and armour to cells. Cell Prolif 2017, 50 (2). 125. Francois, M.; Romieu-Mourez, R.; Li, M.; Galipeau, J., Human MSC suppression correlates with cytokine induction of indoleamine 2,3-dioxygenase and bystander M2 macrophage differentiation. Mol Ther 2012, 20 (1), 187-95. 126. Nemeth, K.; Leelahavanichkul, A.; Yuen, P. S.; Mayer, B.; Parmelee, A.; Doi, K.; Robey, P. G.; Leelahavanichkul, K.; Koller, B. H.; Brown, J. M.; Hu, X.; Jelinek, I.; Star, R. A.; Mezey, E., Bone marrow stromal cells attenuate sepsis via prostaglandin E(2)-dependent reprogramming of host macrophages to increase their interleukin-10 production. Nat Med 2009, 15 (1), 42-9. 127. Eggenhofer, E.; Hoogduijn, M. J., Mesenchymal stem cell-educated macrophages. Transplant Res 2012, 1 (1), 12. 128. Chen, L.; Tredget, E. E.; Wu, P. Y.; Wu, Y., Paracrine factors of mesenchymal stem cells recruit macrophages and endothelial lineage cells and enhance wound healing. PLoS One 2008, 3 (4), e1886. 129. Bellin, M.; Marchetto, M. C.; Gage, F. H.; Mummery, C. L., Induced pluripotent stem cells: the new patient? Nat Rev Mol Cell Biol 2012, 13 (11), 713-26. 130. Zhao, T.; Zhang, Z. N.; Westenskow, P. D.; Todorova, D.; Hu, Z.; Lin, T.; Rong, Z.; Kim, J.; He, J.; Wang, M.; Clegg, D. O.; Yang, Y. G.; Zhang, K.; Friedlander, M.; Xu, Y., Humanized Mice Reveal Differential Immunogenicity of Cells Derived from Autologous Induced Pluripotent Stem Cells. Cell Stem Cell 2015, 17 (3), 353-9. 131. Gourraud, P. A.; Gilson, L.; Girard, M.; Peschanski, M., The role of human leukocyte antigen matching in the development of multiethnic "haplobank" of induced pluripotent stem cell lines. Stem Cells 2012, 30 (2), 180-6. 132. Pallotta, I.; Sun, B.; Wrona, E. A.; Freytes, D. O., BMP protein-mediated crosstalk between inflammatory cells and human pluripotent stem cell-derived cardiomyocytes. J Tissue Eng Regen Med 2015. 133. Choi, J. W.; Park, J. K.; Chang, J. W.; Kim, D. Y.; Kim, M. S.; Shin, Y. S.; Kim, C. H., Small intestine submucosa and mesenchymal stem cells composite gel for scarless vocal fold regeneration. Biomaterials 2014, 35 (18), 4911-8. 134. Duan, Y.; Liu, Z.; O'Neill, J.; Wan, L. Q.; Freytes, D. O.; Vunjak-Novakovic, G., Hybrid gel composed of native heart matrix and collagen induces cardiac differentiation of human embryonic stem cells without supplemental growth factors. J Cardiovasc Transl Res 2011, 4 (5), 605-15. 135. Fong, A. H.; Romero-Lopez, M.; Heylman, C. M.; Keating, M.; Tran, D.; Sobrino, A.; Tran, A. Q.; Pham, H. H.; Fimbres, C.; Gershon, P. D.; Botvinick, E. L.; George, S. C.; Hughes, C. C., Three-Dimensional Adult Cardiac Extracellular Matrix Promotes Maturation of Human

ACS Paragon Plus Environment

39

ACS Biomaterials Science & Engineering

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

Page 40 of 47

Induced Pluripotent Stem Cell-Derived Cardiomyocytes. Tissue Eng Part A 2016, 22 (15-16), 1016-25. 136. Alom, N.; Peto, H.; Kirkham, G. R.; Shakesheff, K. M.; White, L. J., Bone extracellular matrix hydrogel enhances osteogenic differentiation of C2C12 myoblasts and mouse primary calvarial cells. J Biomed Mater Res B Appl Biomater 2017. 137. Ning, L. J.; Zhang, Y. J.; Zhang, Y.; Qing, Q.; Jiang, Y. L.; Yang, J. L.; Luo, J. C.; Qin, T. W., The utilization of decellularized tendon slices to provide an inductive microenvironment for the proliferation and tenogenic differentiation of stem cells. Biomaterials 2015, 52, 539-50. 138. Yuan, X. N.; Wei, Y. Y.; Villasante, A.; Ng, J. J. D.; Arkonac, D. E.; Chao, P. H. G.; Vunjak-Novakovic, G., Stem cell delivery in tissue-specific hydrogel enabled meniscal repair in an orthotopic rat model. Biomaterials 2017, 132, 59-71. 139. Liu, H. H.; Yang, L.; Zhang, E. C.; Zhang, R.; Cai, D. D.; Zhu, S. A.; Ran, J. S.; Bunpetch, V.; Cai, Y. Z.; Heng, B. C.; Hu, Y. J.; Dai, X. S.; Chen, X.; Ouyang, H. W., Biomimetic tendon extracellular matrix composite gradient scaffold enhances ligament-to-bone junction reconstruction. Acta Biomaterialia 2017, 56, 129-140. 140. Batchelder, C. A.; Martinez, M. L.; Tarantal, A. F., Natural Scaffolds for Renal Differentiation of Human Embryonic Stem Cells for Kidney Tissue Engineering. PLoS One 2015, 10 (12), e0143849. 141. Wang, B.; Jakus, A. E.; Baptista, P. M.; Soker, S.; Soto-Gutierrez, A.; Abecassis, M. M.; Shah, R. N.; Wertheim, J. A., Functional Maturation of Induced Pluripotent Stem Cell Hepatocytes in Extracellular Matrix-A Comparative Analysis of Bioartificial Liver Microenvironments. Stem Cells Transl Med 2016, 5 (9), 1257-67. 142. Park, K. M.; Hussein, K. H.; Hong, S. H.; Ahn, C.; Yang, S. R.; Park, S. M.; Kweon, O. K.; Kim, B. M.; Woo, H. M., Decellularized Liver Extracellular Matrix as Promising Tools for Transplantable Bioengineered Liver Promotes Hepatic Lineage Commitments of Induced Pluripotent Stem Cells. Tissue Eng Part A 2016, 22 (5-6), 449-60. 143. Gilpin, S. E.; Ren, X.; Okamoto, T.; Guyette, J. P.; Mou, H.; Rajagopal, J.; Mathisen, D. J.; Vacanti, J. P.; Ott, H. C., Enhanced lung epithelial specification of human induced pluripotent stem cells on decellularized lung matrix. Ann Thorac Surg 2014, 98 (5), 1721-9; discussion 1729. 144. Ren, X.; Moser, P. T.; Gilpin, S. E.; Okamoto, T.; Wu, T.; Tapias, L. F.; Mercier, F. E.; Xiong, L. J.; Ghawi, R.; Scadden, D. T.; Mathisen, D. J.; Ott, H. C., Engineering pulmonary vasculature in decellularized rat and human lungs. Nature Biotechnology 2015, 33 (10), 1097-+. 145. Garreta, E.; Oria, R.; Tarantino, C.; Pla-Roca, M.; Prado, P.; Fernández-Avilés, F.; Campistol, J. M.; Samitier, J.; Montserrat, N., Tissue engineering by decellularization and 3D bioprinting. Materials Today 2017, 20 (4), 166-178. 146. Jang, J.; Park, H. J.; Kim, S. W.; Kim, H.; Park, J. Y.; Na, S. J.; Kim, H. J.; Park, M. N.; Choi, S. H.; Park, S. H.; Kwon, S. M.; Kim, P. J.; Cho, D. W., 3D printed complex tissue construct using stem cell-laden decellularized extracellular matrix bioinks for cardiac repair. Biomaterials 2017, 112, 264-274. 147. Choi, Y. J.; Kim, T. G.; Jeong, J.; Yi, H. G.; Park, J. W.; Hwang, W.; Cho, D. W., 3D Cell Printing of Functional Skeletal Muscle Constructs Using Skeletal Muscle-Derived Bioink. Advanced Healthcare Materials 2016, 5 (20), 2636-2645. 148. Guyette, J. P.; Charest, J. M.; Mills, R. W.; Jank, B. J.; Moser, P. T.; Gilpin, S. E.; Gershlak, J. R.; Okamoto, T.; Gonzalez, G.; Milan, D. J.; Gaudette, G. R.; Ott, H. C., Bioengineering Human Myocardium on Native Extracellular Matrix. Circ Res 2016, 118 (1), 5672. 149. Dalby, M. J.; Gadegaard, N.; Oreffo, R. O., Harnessing nanotopography and integrinmatrix interactions to influence stem cell fate. Nat Mater 2014, 13 (6), 558-69. 150. Turner, W. S.; Wang, X. L.; Johnson, S.; Medberry, C.; Mendez, J.; Badylak, S. F.; McCord, M. G.; McCloskey, K. E., Cardiac tissue development for delivery of embryonic stem

ACS Paragon Plus Environment

40

Page 41 of 47

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

ACS Biomaterials Science & Engineering

cell-derived endothelial and cardiac cells in natural matrices. Journal of Biomedical Materials Research Part B-Applied Biomaterials 2012, 100B (8), 2060-2072. 151. Deegan, D. B.; Zimmerman, C.; Skardal, A.; Atala, A.; Shupe, T. D., Stiffness of hyaluronic acid gels containing liver extracellular matrix supports human hepatocyte function and alters cell morphology. Journal of the Mechanical Behavior of Biomedical Materials 2016, 55, 87-103. 152. Skardal, A.; Smith, L.; Bharadwaj, S.; Atala, A.; Soker, S.; Zhang, Y. Y., Tissue specific synthetic ECM hydrogels for 3-D in vitro maintenance of hepatocyte function. Biomaterials 2012, 33 (18), 4565-4575. 153. Liau, B.; Christoforou, N.; Leong, K. W.; Bursac, N., Pluripotent stem cell-derived cardiac tissue patch with advanced structure and function. Biomaterials 2011, 32 (35), 9180-9187. 154. Zhang, D. H.; Shadrin, I. Y.; Lam, J.; Xian, H. Q.; Snodgrass, H. R.; Bursac, N., Tissueengineered cardiac patch for advanced functional maturation of human ESC-derived cardiomyocytes. Biomaterials 2013, 34 (23), 5813-5820. 155. Wong, T. Y.; Chang, C. H.; Yu, C. H.; Huang, L. L. H., Hyaluronan keeps mesenchymal stem cells quiescent and maintains the differentiation potential over time. Aging Cell 2017, 16 (3), 451-460. 156. Liu, W. Y.; Lipner, J.; Xie, J. W.; Manning, C. N.; Thomopoulos, S.; Xia, Y. N., Nanofiber Scaffolds with Gradients in Mineral Content for Spatial Control of Osteogenesis. Acs Applied Materials & Interfaces 2014, 6 (4), 2842-2849. 157. Trounson, A.; DeWitt, N. D., Pluripotent stem cells progressing to the clinic. Nat Rev Mol Cell Biol 2016, 17 (3), 194-200.

ACS Paragon Plus Environment

41

ACS Biomaterials Science & Engineering

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

Page 42 of 47

Potential Synergistic Effects of Stem Cells and Extracellular Matrix Scaffolds

Lewis Gaffney, Emily A. Wrona and Donald O. Freytes Ph.D

FOR TOC USE ONLY:

Synergistic Interactions

Combination of stem cells and extracellular matrix scaffold

GOAL: Enhanced Tissue Repair - Improved cell engraftment - Constructive remodeling

Host tissue response

- Direction of stem cell differentiation

Macrophages T Lymphocytes

ACS Paragon Plus Environment

42

A. Chemotaxis

Page 43 of 47

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42

B. Growth Factors

ACS Biomaterials Science & Engineering

C. Cryptic Peptides Interactions with Cells

VEGF TGF-ß

Th-1 Inflammatory Lympohocytes

Th-2 Pro-regenerative Lympohocytes

D. Lymphocyte Response

M1-Like, Pro-inflammatory M2-Like, Pro-healing Macrophages Macrophages

E. Macrophage Response ACS Paragon Plus Environment

ACS Biomaterials Science & Engineering

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42

-80°C

+25°C

Page 44 of 47

+25°C

-80°C

Electroporation

Freeze/Thaw Cycles Decellularization

Sterilization and washes

Enzymatic treatment

Sonication

Hypotonic or hypertonic solution Ionic and nonionic detergents

ACS Paragon Plus Environment

Page 45 of 47

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37

ACS Biomaterials Science & Engineering

Hydrated Scaffold Cell seeding, structural support, implantation

Lyophilized Scaffold Cell Seeding, implantation

Lyophilized Powder Media supplementation

Hydrogel Digested ECM Cell seeding, encapsulation, Vehicle for injection ACS Paragon Plus Environment implantation

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

ECM Scaffold

Stem Cell Source

Outcome

REF

Duan et al 2011

ACS Biomaterials Science & Engineering

Page 46 of 47

Heart

Decellularized Porcine Heart Tissue was sliced and digested to make a hydrogel

Human Embryonic Stem Cells/Induced pluripotent stem cells

Differentiated with and without supplements in media

Heart

Decellularized fetal and adulte Porcine heart tissue

Induced Pluripotent Stem cells

Promoted the expression Fong et al of Calcium signalling and 2016 promoted proliferation of iPSC derived Cardiomyocytes

Heart

Bio-ink derived from decellularized cardiac tissue

Cardiac Progenitor cells and MSCs

Compared to Collagen, bio-ink derived from cardiac tissue supported more functional cardiac regeneration

Jang et al 2017

Liver

Liver ECM

Induced Pluripotent Stem cells

Higher expression of P450 mRNA, increased metabolic enzyme activity

Wang et al 2016

Liver

Porcine liver ECM containing liver derived growth factors

Induced Pluripotent Stem Cells

Higher Expression of Hepatocyte Markers

Park et al. 2016

Kidney

Kidney ECM manufactured into Mouse kidney stem 2D sheets or hydrogels cells, mouse Mesenchymal Stem Cells

Stem cells had different O’neill et al. expression depending on 2013 which region was used for the ECM

Kidney

Decellularized monkey kidneys Human Embryonic using perfusion of heparin, PBS Stem Cells and 1%SDS

Renal Development markers were significantly higher in renal ECM than cells cultured in embryoid bodies

Vocal Fold

Small Intestine Submucosa in a Mesenchymal Stem digested form Cells

There was higher Choi et al. engraftment of cells, and 2014 improved vibratory motions compared to groups that had MSCs or ECM alone

Lung

Decellularized whole rat and human lung

Lung epithelial cells Higher proliferation and generated from expression of lung human induced markers Nkx2.1 Pluripotent Stem Cells

Gilpin, 2014

Lung

Decellularized whole rat and human lung

iPSC derived endothelial cells and pericytes

Resulted in complete vascularization in rat model and in scale up model for a human lung

Ren et al, 2015

Bone

Demineralized Bovine Tibia, digested and gelled

C2C12 Mouse Myoblasts

After 7 Days in culture, muscle progenitor cells expressed osteogenic markers

Alom et al, 2017

Tendon

Decellularized Canine Achilles tendons

Rat bone marrow derived stem cells, rat tendon derived stem cells

BMSCs expressed tendon Ning et al specific genes, TDSCs had 2015 higher expression of tendon specific markers compared to cells cultured on tissue culture plastic.

Tendon

Decellularized tendon, modified with ultrasound to form a random-alignedrandom structure

Rabbit Bone Marrow stromal cells

Modified ECM enhanced Osteogenic and Chonrogenic gene expression, resulting in higher osteoinductivity.

Meniscus

Minced, Decellularized Bovine Meniscus Hydrogels

Human MSCs

Hydrogels supported Yuan et al integration and formation 2017 of fibrocartilagenous tissue in rat models of meniscus injury

ACS Paragon Plus Environment

Batchelder et al 2015

Liu et al 2017

Synergistic Interactions Page ACS 47 Biomaterials of 47 Science & Engineering

Combination of stem cells and extracellular matrix scaffold

1 2 3 4 5 6 7

GOAL: Enhanced Tissue Repair - Improved cell engraftment - Constructive remodeling

Host tissue response

- Direction of stem cell differentiation

ACS Paragon Plus Environment Macrophages T Lymphocytes