The Human Amniotic Membrane: A Versatile Scaffold for Tissue

May 14, 2018 - Over the past decade, the development of tissue engineering and regenerative medicine, along with new decellularization protocols, has ...
2 downloads 0 Views 2MB Size
Subscriber access provided by UNIVERSITY OF CALGARY

Review

The Human Amniotic Membrane: A Versatile Scaffold for Tissue Engineering Julien H Arrizabalaga, and Matthias U Nollert ACS Biomater. Sci. Eng., Just Accepted Manuscript • DOI: 10.1021/acsbiomaterials.8b00015 • Publication Date (Web): 14 May 2018 Downloaded from http://pubs.acs.org on May 14, 2018

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 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 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.

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 35 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

The Human Amniotic Membrane: A Versatile Scaffold for Tissue Engineering Julien H. Arrizabalaga1 and Matthias U. Nollert*,1,2

1

Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, United

States 2

School of Chemical, Biological & Materials Engineering, University of Oklahoma, Norman,

OK, United States *Author to whom correspondence should be addressed to. Email: [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 35

ABSTRACT The human amniotic membrane (hAM) is a collagen-based extracellular matrix derived from the human placenta. It is a readily-available, inexpensive, and naturally biocompatible material. Over the past decade, the development of tissue engineering and regenerative medicine, along with new decellularization protocols, has recast this simple biomaterial as a tunable matrix for cellularized tissue engineered constructs. Thanks to its biocompatibility, decellularized hAM is now commonly used in a broad range of medical fields. New preparation techniques and composite scaffold strategies have also emerged as ways to tune the properties of this scaffold. The current state of understanding about the hAM as a biomaterial is summarized in this review. We examine the processing techniques available for the hAM, addressing their effect on the mechanical properties, biodegradation, and cellular response of processed scaffolds. The latest in vitro applications, in vivo studies, clinical trials, and commercially-available products based on the hAM are reported, organized by medical field. We also look at the possible alterations to the hAM to tune its properties, either through composite materials incorporating decellularized hAM, chemical crosslinking, or innovative layering and tissue preparation strategies. Overall, this review compiles the current literature about the myriad capabilities of the human amniotic membrane, providing a much-needed update on this biomaterial.

KEYWORDS Human amniotic membrane, tissue engineering, biomaterials, extracellular matrix, regenerative medicine.

ACS Paragon Plus Environment

2

Page 3 of 35 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

INTRODUCTION Anatomy and Components The human amniotic membrane (hAM), also known as the amnion, is the innermost layer of the fetal membranes. It is loosely attached to the chorionic membrane, or chorion. Together, these fetal membranes form the amniotic sac, which contains the embryo and amniotic fluid during pregnancy. Both membranes are loosely connected via a spongy collagen layer, making them easy to peel and separate from each other.1 The hAM is a translucent and avascular biomaterial with a thickness of 20 to 50µm.1,2 As seen in Figure 01, the hAM consists of an epithelium monolayer in contact with the amniotic fluid, a basement membrane, a compact layer, a fibroblast layer, and then a spongy layer connected to the chorion.3 In a majority of tissue engineering applications, what is mentioned as the hAM actually refers to the basement membrane, decellularized and isolated from its surrounding layers to produce a thin and homogeneous biomaterial. Collagens type III, IV, and V are the major components of the basement membrane, providing structural integrity and mechanical strength to the tissue.4,5 Non collagenous glycoproteins such as laminin, fibronectin, and nidogen are also present.3,6 Amniotic Fluid Epithelium Basement membrane Compact layer Fibroblast layer Spongy layer Chorion

Figure 01: Schematic representation of the structure of the human fetal membranes. The amnion is composed of 5 distinct layers: epithelium, basement membrane, compact layer,

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 35

fibroblast layer, and spongy layer. The chorion (not depicted) consists of a reticular layer, a basement membrane, and an outer trophoblasts layer.

Biological Properties The hAM possesses anti-microbial, anti-fibrosis, and anti-inflammatory properties.7,8 When implanted into the human body it effortlessly adheres to the wound surface, reduces scars, decreases pain, promotes wound healing, and has a low risk of immunogenicity.7,9–12 Another critical property of the hAM for tissue engineering applications is its ability to support cell attachment, proliferation, and differentiation.7,11,12 Various cell lines have been reported to adhere and proliferate on decellularized hAM. Figure 02 presents a specific example of the type of results numerous investigators have obtained, demonstrating that the culture of human mesenchymal stem cells on the hAM does not affect their immunophenotype or differentiation abilities.13 This unique set of biological and biochemical properties combined with its reduced cost and unlimited availability have shaped the hAM into a biomaterial of choice for clinical and tissue engineering applications.

ACS Paragon Plus Environment

4

Page 5 of 35 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

Figure 02: Characterization of human mesenchymal stem cells derived from bone marrow (BM-dMSC) or from adipose tissue (ASCs) previously seeded on hAM scaffolds. A: Immunophenotype of the cells as determined by flow cytometric analysis. B: Chondrogenic (Alcian Blue staining), osteogenic (Alizarin Red staining), and adipogenic (Oil Red O staining) differentiation of the cells. The culture of both BM-dMSC and ASCs on hAM scaffolds did not affect their immunophenotype or differentiation abilities. Reproduced with permission from ref 13.13 Copyright 2015 Mary Ann Liebert, Inc.

Amniotic Membrane-Derived Cells The hAM has also been reported as a source of 2 populations of stem cells: human amniotic epithelial cells (hAECs) and human amniotic mesenchymal stromal cells (hAMSCs).14,15 hAECs are present on the epithelium monolayer facing the amniotic fluid. hAMSCs are part of the stromal layer found between the basement amniotic membrane and the loose spongy layer connected to the chorion, as depicted in Figure 01. Overall, hAECs and hAMSCs represent valuable sources of multipotent stem cells for tissue engineering and regenerative medicine applications. The possibility of obtaining these cells from the hAM adds even more value to this tissue, which is usually considered to be medical waste. The isolation, characterization, differentiation, and applications of amniotic membrane-derived cells have already been thoroughly reviewed.11,12,16–18 Consequently, the present article will only focus on the prospects offered by the human amniotic membrane.

Historical perspective The hAM was first used for skin reconstruction by Dr. Davis in 1910.19 Subsequently, the hAM was employed for a broad range of applications: surgical dressings, the treatment of burns and ulcers, the reconstruction of the oral cavity, bladder, and vagina.20,21 In the 1940s the first ophthalmic applications of the hAM to treat conjunctival defects and chemical burns were reported.22–24 The initial interest for this biomaterial then faded away, most likely due to more

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 35

stringent regulations regarding tissue transplantations, and the hAM almost disappeared from the literature for about 50 years. The resurgence of the hAM happened in the 1990s when it was reported by Kim et al. as an effective scaffold for ocular surface reconstruction.25,26 New preparation, preservation, and storage techniques were also developed over the same time period, assisting the hAM to gain significance for ophthalmic and dermatologic applications without the worry of disease transmission from fresh unprocessed tissue. In 2001, processed hAM received approval from the United Stated Food and Drug Administration (FDA) for ocular surface reconstruction, increasing its use even further and triggering the development of several commercial hAM derived products.27 Nowadays, the hAM has been widely adopted for burn wound dressing and ocular surface reconstruction. New clinical applications are also currently explored such as nerve wrapping,28–30 reconstruction of the oral cavity,31 flexor tendon repair,32 and the treatment of limbal stem cell deficiency.33

The development of tissue engineering diversified the potential uses of the hAM as a scaffolding material from traditional fields such as ophthalmology and dermatology to new disciplines. Over the last two decades, novel hAM modifications have been incorporating cells and utilizing the hAM as a scaffold to recreate complex 3D structures as opposed to last century uses as an acellular dressing.26 An ideal scaffold for tissue engineering applications is non-immunogenic and supports the attachment and proliferation of cells without affecting their immunophenotype or differentiation abilities. It also has comparable mechanical properties, biodegradation rate permeability, and flexibility to the tissue it intends to replace. Decellularized hAM is a versatile biomaterial that can be used to engineer several types of tissues from the human body.7

ACS Paragon Plus Environment

6

Page 7 of 35 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 hAM is currently being investigated for a broad range of applications such as blood vessel tissue engineering,34 cartilage regeneration,35 and urothelium tissue engineering.36

PRESERVATION AND DECELLULARIZATION TECHNIQUES Tissue Processing and Preservation The hAM can be used two ways, either intact with its epithelium layer or decellularized with its epithelium removed. An intact hAM would simply be detached from the chorion, cut to size, and preserved for upcoming applications. The epithelium monolayer and extracellular matrix would remain untouched. Intact membranes are typically used for wound dressing in clinical applications, while decellularized hAM are ideal scaffolds for tissue engineering constructs. Epithelial cells need to be removed from the hAM in order to not interfere with new cells seeded on the scaffold. The goal of the decellularization process is to remove all epithelial cells and obtain a uniform and smooth basement membrane.37–39 Figure 03 illustrates the removal of the epithelium layer after decellularization.40 Intact hAM for clinical applications are processed, sterilized, and preserved prior to implantation in humans. Tissue processing simply consists of mechanically separating the amnion from the chorion, cutting it into smaller pieces, and flattening it onto nitrocellulose paper. Tissue preservation helps provide a steady supply of scaffolds for clinical applications. Several protocols have been developed for the preservation of intact hAM, including cryopreservation in glycerol, air-drying, freeze-drying, and incubation in trehalose.38,41

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 35

Figure 03: Histological comparison of (A) an intact hAM with (B) a hAM decellularized with 0.1% trypsin-EDTA. The letters, e, b, and s respectively designate the epithelium, basement membrane, and stroma. Adapted with permission from ref 40.40 Copyright 2007 Mary Ann Liebert, Inc.

Decellularization hAM intended for tissue engineering applications are de-epithelialized. Rapid freezing is often used as a preliminary step to introduce ice crystals into the intracellular space and cause cell lysis.42 Enzymatic treatments and chemical treatments are then needed to solubilize the remaining cytoplasmic and nuclear cellular membranes.43 Enzymatic treatments for decellularization include protease digestion, calcium chelating agents, and nucleases.44 Most protocols commonly use EDTA, dispase, trypsin, or thermolysin.38,43,45 Saghizadeh et al. also reported using NaOH to efficiently de-epithelialize the hAM.46 Nucleases such as DNases and RNases are also advantageous for removing nucleotides after an initial cell lysis.44 Finally, sodium dodecyl sulfate is one of the most popular and attractive detergents used for treating the hAM since it yields a complete removal of cellular remnants without damaging the collagen from the tissue.44,47,48

ACS Paragon Plus Environment

8

Page 9 of 35 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

Gamma irradiation and chemical solutions (peracetic acid and ethanol) are the main techniques used for the sterilization of the hAM.48 Supercritical carbon dioxide has also been described as a preparation technique to obtain a sterile hAM without affecting its composition.49

Commercially Available Products The advances realized these past decades for the stabilization, preservation, and storage of tissues also lead to the development of several commercially available products derived from the hAM.35,50 These products have the advantages of being stable, as well as easily transported and stored. A majority of them consist of either cryopreserved or dehydrated intact hAM and have been recommended for the treatment of ophthalmic and dermal wounds. For instance, BioTissue AmnioGraft (cryopreserved intact hAM on a carrier paper) and IOP Ophthalmics AmbioDisk (dehydrated intact hAM) are both recommended for ophthalmic surgery. MiMedx AmnioFix, also made of dehydrated intact hAM, is endorsed for soft tissue surgery. Detailed information about commercially available hAM products can be found in the review written by Gindraux et al.50

TISSUE MODIFICATIONS Despite its attractive biological and biochemical properties, the hAM has been limited by its mechanical properties and biodegradation rate for some tissue engineering or regenerative medicine applications. To overcome these limitations, alterations to the hAM have been made to

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 35

tune its properties, either through incorporating composite materials, chemical crosslinking, or innovative layering and tissue preparation strategies into the hAM.

Crosslinking Chemical crosslinking of the hAM has been reported to improve its mechanical properties and biodegradation rate without affecting affect the viability, proliferation, and immunophenotype of cells cultured on top of crosslinked hAM.51–57 Figure 04 presents an innovative technique developed by Hariya et al. for the production of transparent and resilient crosslinked hAM tissue laminates.58 Carbodiimide was used for crosslinking and up to 8 layers were assembled to produce a tougher and optically clearer graft for corneal transplantation.

Figure 04: Fabrication of transparent and resilient crosslinked hAM tissue laminates. For the first lamination stage, hAM are repeatedly dried and stacked up to produce a multilayer tissue laminate. Then, tissue constructs are optically clarified after successive carbodiimide crosslinking, washing, drying, and quenching steps. The final laminates contain up to 8 hAM layers and benefit from improved light transmittance and mechanical properties. Reproduced with permission from ref 58.58 Copyright 2016 Elsevier Ltd.

Composites

ACS Paragon Plus Environment

10

Page 11 of 35 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

New preparation techniques and composite scaffold strategies have also emerged as ways to tune the properties of the hAM. Recent publications describe applications of the hAM for osteochondral tissue engineering by coating it with poly(lactic-co-glycolic acid).59 The hAM was mixed with fibrin to produce a tissue engineered blood vessel and was combined with collagenglycosaminoglycan scaffolds for tendon tissue engineering.60,61 A similar composite material approach was used by Adamowicz et al. who layered denuded hAM with electrospun poly(Llactide-co-ε-caprolactone) (PLCL) for reconstructive urology.62 These composite scaffolds, presented in Figure 05, were successfully implanted in rats and promoted bladder augmentation.62

Figure 05: Scanning electron microscopy images of hAM-PLCL composite scaffolds. A: Cross-section image of the composite scaffold. B: Delaminated composite. Frozen hAM was sandwiched and covered on both sides with two layers of electrospun PLCL. Adapted with permission from ref 62.62 Copyright 2016 Adamowicz et al.

Several more composite and preparation techniques have been used to functionally augment the hAM for tissue regeneration purposes.63 A decellularized hAM was assembled with electrospun silk fibroin to produce a 3D bilayer of artificial skin.64 Denuded hAM was coated with

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 35

poly(ester) urethane on both sides to improve its mechanical strength and produce a biocompatible surgical mesh.65 Another popular preparation method is the combination of several hAM scaffolds to produce multilayer constructs such as the hAM tissue laminates presented in Figure 04. Multilayer constructs have been reported for the fabrication of tissue engineered blood vessels,60,66,67 surgical patches,68 applications in ophthalmology,58,69,70 and oral and maxillofacial surgery.71,72 Skin defects have also been treated with solubilized hAM combined with hyaluronic acid hydrogel.73 Finally, micronized hAM (300 to 600µm microparticles) combined with epidermal stem cells successfully repaired full-thickness skin defects when transplanted in nude mice.74

CLINICAL APPLICATIONS Ophthalmology The hAM is widely used for a variety of ocular surface diseases thanks to its attractive biological properties, transparency, thinness, and composition akin to the one of the conjunctiva.75 The hAM can be applied on the ocular surface either as a temporary dressing or as a permanent graft.27 When used as a temporary biological bandage, the hAM acts as barrier to protect the healing epithelium from the movement of the eyelids. A hAM dressing aims to repel inflammation of the host tissue and reduce scarring.27 A permanent hAM graft transplanted on the ocular surface is used to promote the proliferation of the epithelial cells present on the ocular surface.76 The patient’s cells will grow over the hAM and the scaffold will be integrated into the patient’s tissue. For deep corneal ulcerations or perforations, the hAM can also be stuffed into the defects prior to patching or grafting.77

ACS Paragon Plus Environment

12

Page 13 of 35 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

Today the hAM is widely used for the management of several ocular ailments of the cornea or the conjuctiva. Applications of the hAM for corneal surface reconstruction include the treatment of chemical burns, bullous keratopathy, persistent epithelial defects, ulcers, and lesions.77 The hAM has been adopted for a variety of conjunctival pathologies including symblepharon, pterygium, chemical burns, and scleral thinning, for the covering and reconstruction of conjunctival lesions, and for the management of Stevens-Johnson syndrome.77–79

The hAM has also been applied to the treatment of limbal stem cell deficiency. Unlike the ophthalmic applications previously described, this recent method uses the hAM to produce a cellularized graft.80–82 Limbal stem cells are critical for the constant maintenance and regeneration of the corneal epithelium. Damage to the limbus would lead to the deficiency of these stem cells, epithelial breakdown, chronic inflammation, corneal conjunctivalization, and a loss of corneal clarity.83 Limbal stem cell deficiency can be treated with the transplantation of limbal epithelial cells expanded ex vivo. Cells can be obtained from a small biopsy of the patient’s other eye if healthy, or from another living individual or a cadaveric donor if both of the patient’s eyes are affected by limbal stem cell deficiency.83 In the case of donor shortages, oral mucosal epithelial cells and bone marrow mesenchymal stem cells could also be used as replacement cell sources to reconstruct the corneal surface.84–87 Decellularized hAM have been successfully used as substrates for the ex vivo expansion of limbal epithelial cells. Transplantation of these cellularized grafts resulted in the repopulation of the corneal epithelium and integration of the hAM with the corneal tissue.77,88,89 Figure 06 illustrates the transplantation process of limbal epithelial cells using the hAM as a carrier membrane.

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 35

Limbal epithelial cells Biopsy Transplantation Healthy donor eye Cell seeding Diseased eye hAM graft

Decellularized hAM

Figure 06: Illustration of the transplantation process of autologous limbal epithelial cells for the treatment of a unilateral limbal stem cell deficiency. A small biopsy (about 2 by 2 mm of tissue) is performed on the patient’s healthy donor eye. Limbal epithelial cells are then isolated and seeded onto a sheet of decellularized hAM. After 2 to 3 weeks of culture, the cellularized hAM graft is transplanted onto the patient’s diseased corneal surface.

Other recent innovations have been finding new ways to attach the hAM to the ocular surface. The hAM is a thin material that is typically attached to the corneal surface with sutures. However, hAM bandages need to be replaced every week and repeated interventions at the diseased corneal surface is not recommended. Possible side effects of sutures also include conjunctival bleeding or scarring.90 The strategies developed for the sutureless fixation of the hAM include the use of fibrin,91,92 chemically defined bioadhesives,93,94 light-initiated bonding with Rose Bengal dye95,96 and medical devices (ProKera and AmnioClip) that use a dual ring system to mount the hAM and apply it like a contact lens.90,97 Dermatology The hAM has been described as an effective therapeutic option for burn wound dressing. Indeed, the hAM presents several advantages for skin applications such as its anti-inflammatory

ACS Paragon Plus Environment

14

Page 15 of 35 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

properties, antimicrobial effects, low immunogenicity, and good wound adherence. When used to cover burn wounds, the hAM reduced pain and scars, prevented infections, and promoted reepithelialization. 98–100 The hAM has been used to treat both superficial and partial-thickness burns,100 as an overlay for skin autografts,

101,102

and as a dressing for skin graft donor sites.103,104 Besides burn wound

dressing, the hAM proved effective for the treatment of chronic leg ulcers, pressure sores, and for patients with epidermolysis bullosa.105–107 These long-established dermatology applications of the hAM involve using it as an acellular biomaterial, while the most recent studies report using the hAM for the attachment and proliferation of various cell lines. Patients with stable vitiligo were transplanted with autologous melanocytes seeded on a decellularized hAM, resulting in over 90% skin repigmentation.108,109 Skin equivalents were constructed by combining a hAM seeded with fibroblasts and normal keratinocytes.55,110,111 Biological wound dressings were designed to promote tissue regeneration for patients with full-thickness skin defects, by combining the hAM with mesenchymal stem cells.13,74,112

Oral and Maxillofacial Surgery After oral surgery, bone surfaces are often exposed to the oral cavity which can lead to possible infections and scar formation. Thanks to its biological properties, the hAM has been described as an effective scaffold for the reconstruction of the oral cavity.31 Consequently, the hAM has been used for a broad range of applications: treatment of oral mucosa defects after removal or cancerous legions,113 flap surgery,

114

oronasal fistula reconstruction in cleft palate surgery,71,72

and temporomandibular joint surgery.115

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 35

Recent tissue engineering approaches involve the fabrication of a myomucosal flap using oral keratinocytes cultured on hAMs,116 and the autologous transplantation of oral mucosal epithelial cell sheets cultured on hAMs for intraoral mucosal defects.117–119 Figure 07 shows that oral mucosal epithelial cells cultured on the hAMs present comparable keratin expression to cells present in the native oral mucosa.118

ACS Paragon Plus Environment

16

Page 17 of 35 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

Figure 07: Comparison of hAM-cultured oral mucosal epithelial cells with the oral mucosa. A-F: hematoxylin and eosin staining. B-G, C-H, D-I, E-J: Expression of keratins 4, 13, 1, and 10 in green with the cell nuclei stained in red with propidium iodide. Keratin expression was similar between the hAM cultured oral mucosal epithelial cells and the oral mucosa. Reproduced with permission from ref 118.118 Copyright 2015 Amemiya et al.

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 35

Otolaryngology The human amniotic membrane has shown therapeutic promises as a scaffold in otolaryngology. Patches of hyperdry hAM were used as dressing substitutes for fascia grafts of the temporal muscle in canal wall down tympanoplasty.120,121 The hAM grafts were completely epithelialized faster than the classic fascia grafts.

TISSUE ENGINEERING APPLICATIONS The hAM has been investigated as a scaffold for tissue engineering and regenerative medicine applications in a broad range of disciplines. Several of these studies were already reported in previous paragraphs dedicated to tissue modifications of the hAM, along with novel cellularized applications of the hAM in ophthalmology, dermatology, and oral and maxillofacial surgery. Articular cartilage has minimal self-healing capability and often advances to osteoarthritis once damaged.122,123 The hAM has been described as an ideal substrate for the culture of chondrocytes.124–127 Consequently, chondrocytes have been seeded on the HAM in order to obtain a delivery vehicle for cell transplantation that could promote cartilage regeneration in vivo. For instance, Jin et al. seeded rabbit articular chondrocytes on decellularized hAM and used these constructs in vivo to cover rabbit osteochondral defects. After 8 weeks of implantation the defect area was successfully regenerated.40 The hAM has been reported as a potential osteoinductive biomaterial for bone regeneration.128,129 Decellularized hAM helped induce the osteogenic differentiation of human dental apical papilla cells seeded on top of it, opening potential applications for the hAM in bone and tooth tissue engineering.130 Intact hAM simply exposed to osteogenic media also promoted the differentiation of the stem cells contained in the hAM towards osteogenic pathways.131

ACS Paragon Plus Environment

18

Page 19 of 35 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

In the past years, the hAM has been investigated as a scaffold for tissue engineering of the urinary tract system. The hAM was described as a viable scaffold for the culture of urothelial cells and the reconstruction of damaged urothelium tissue.36,132–134 The hAM has been characterized as blood compatible and as an adequate cell carrier matrix for endothelial and smooth muscle cells.66,135,136 Consequently, several studies have reported rolling the hAM to engineer small diameter blood vessels.34,60,66,67,137 Finally, the hAM has been used as a scaffold for salivary gland regeneration,138 for esophageal wall tissue engineering,139 for the treatment of pelvic floor dysfunction,140 for liver regeneration, 141,142

and as a support of in vitro ovarian follicular culture.143

LIMITATIONS Risk of disease transmission Due to its human origin, the hAM carries a risk of disease transmission from its donor. Consequently, the use of fresh untreated hAM is not recommended. The collection, processing, and transplantation of the hAM have to follow the proper laws and regulations of the country where they would be performed.35 To avoid bacterial contamination, it is recommended to collect hAM tissue after caesarean section under sterile conditions. Typically, serological testing for syphilis, hepatitis, and HIV are performed on the donor at the time of tissue collection.77 Serological screening is repeated 6 months after birth to confirm that there were no false negatives during the first round of testing. In the meantime, the hAM is quarantined until both results are negative. Then, the hAM needs to be processed under aseptic conditions and stored in a sterile environment.77,88

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 35

Donor variation The hAM is a biological human tissue and its composition is affected by intra and inter donor variations.76,144 The fetal sex, gestational age, donor age, health, and diet are all parameters that affect the properties of the hAM. Between donors, the thickness of the hAM varies from 20 to 50µm.1,2 From the same donor, the hAM will also be thinner away from the placenta and thicker closer to the umbilical cord.145 The transparency of the hAM will also change depending on its original location in the amniotic sac.146 One possibility to overcome these donor variations would be the development of pooling and screening methods to sort out different hAMs according to previously defined characteristics.

Effects of decellularization and storage techniques The preservation, decellularization, and sterilization protocols used to process the hAM will affect its mechanical properties, transparency, chemical composition, and cell adhesion properties.38,147–150 Characterizing how a treatment influences the morphology and composition of the hAM helps verify that no undesirable or unwanted property changes were induced. For instance, it is critical to ensure that the cell adhesion properties of the hAM are maintained after processing if the hAM is intended for tissue engineering applications. Standardization of processing techniques helps reduce variability. Standard protocols developed for commercially available hAM products and for tissue banks help produce consistent hAM allografts for clinical applications.50,151–153

ACS Paragon Plus Environment

20

Page 21 of 35 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

CONCLUSION The hAM is a readily available and inexpensive biomaterial that can efficiently be processed, preserved, and de-epithelialized. Thanks to its unique set of biological properties, the hAM is now a material of choice for several clinical applications in ophthalmology, dermatology, and oral and maxillofacial surgery. The development of standard processing protocols and tissue banks, along with commercially available hAM derived products, will further expand the clinical applications of the hAM. Moreover, decellularized hAM has proven worthwhile for a broad range of tissue engineering applications due to its ability to support cell attachment, proliferation, and differentiation. Modifications of the hAM have been developed to tune its properties, either through composite materials incorporating the hAM, chemical crosslinking, or innovative layering and tissue preparation strategies. Such advantageous techniques have the potential to become increasingly popular for the preparation of hAM based scaffolds for tissue engineering applications. Additionally, the hAM has been identified as a source of human amniotic epithelial cells and human amniotic mesenchymal stromal cells. These two multipotent cell lines are valuable for tissue engineering and regenerative medicine applications. Overall, the hAM is truly a versatile biomaterial and its foreseeable future as a scaffold for tissue engineering applications looks very promising.

SUPPORTING INFORMATION This manuscript is not accompanied by any supporting information for publication.

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 35

AUTHOR INFORMATION Corresponding Author *Author to whom correspondence should be addressed to. Email: [email protected]

ACKNOWLEDGMENTS This work was supported by the Vice President for Research of the University of Oklahoma.

REFERENCES (1) (2)

(3) (4)

(5)

(6)

(7)

(8)

(9)

(10)

Bourne, G. L. The Microscopic Anatomy of the Human Amnion and Chorion. Am. J. Obstet. Gynecol. 1960, 79 (6), 1070–1073 DOI: 10.1016/0002-9378(60)90512-3. Danforth, D. N.; Hull, R. W. The Microscopic Anatomy of the Fetal Membranes with Particular Reference to the Detailed Structure of the Amnion. Am. J. Obstet. Gynecol. 1958, 75 (3), 536–550 DOI: 10.1016/0002-9378(58)90610-0. Parry, S.; Strauss, J. F. Premature Rupture of the Fetal Membranes. N. Engl. J. Med. 1998, 338 (10), 663–670 DOI: 10.1056/NEJM199803053381006. Malak, T. M.; Ockleford, C. D.; Bell, S. C.; Dalgleish, R.; Bright, N.; Macvicar, J. Confocal Immunofluorescence Localization of Collagen Types I, III, IV, V and VI and Their Ultrastructural Organization in Term Human Fetal Membranes. Placenta 1993, 14 (4), 385–406 DOI: 10.1016/S0143-4004(05)80460-6. Uchide, N.; Ohyama, K.; Bessho, T.; Takeichi, M.; Toyoda, H. Possible Roles of Proinflammatory and Chemoattractive Cytokines Produced by Human Fetal Membrane Cells in the Pathology of Adverse Pregnancy Outcomes Associated with Influenza Virus Infection. Mediators Inflamm. 2012, 2012, 1–32 DOI: 10.1155/2012/270670. Takashima, S.; Yasuo, M.; Sanzen, N.; Sekiguchi, K.; Okabe, M.; Yoshida, T.; Toda, A.; Nikaido, T. Characterization of Laminin Isoforms in Human Amnion. Tissue Cell 2008, 40 (2), 75–81 DOI: 10.1016/j.tice.2007.09.001. Niknejad, H.; Jorjani, M.; Ahmadiani, A.; Ghanavi, J.; Seifalian, A. Properties of the Amniotic Membrane for Potential Use in Tissue Engineering. Eur. Cell. Mater. 2008, 7, 88–99 DOI: 10.22203/eCM.v015a07. Hortensius, R. A.; Harley, B. A. Naturally Derived Biomaterials for Addressing Inflammation in Tissue Regeneration. Exp. Biol. Med. 2016, 241 (10), 1015 DOI: 10.1177/1535370216648022. Inge, E.; Talmi, Y. P.; Sigler, L.; Finkelstein, Y.; Zohar, Y. Antibacterial Properties of Human Amniotic Membranes. Placenta 1991, 12 (3), 285–288 DOI: 10.1016/01434004(91)90010-D. Hao, Y.; Ma, D. H.-K.; Hwang, D. G.; Kim, W.-S.; Zhang, F. Identification of Antiangiogenic and Antiinflammatory Proteins in Human Amniotic Membrane. Cornea 2000, 19 (3), 348–352 DOI: 10.1097/00003226-200005000-00018.

ACS Paragon Plus Environment

22

Page 23 of 35 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

(11)

(12)

(13)

(14)

(15)

(16)

(17) (18)

(19) (20) (21)

(22) (23) (24)

Mamede, A. C.; Carvalho, M. J.; Abrantes, A. M.; Laranjo, M.; Maia, C. J.; Botelho, M. F. Amniotic Membrane: From Structure and Functions to Clinical Applications. Cell Tissue Res. 2012, 349 (2), 447–458 DOI: 10.1007/s00441-012-1424-6. Lim, R. Concise Review: Fetal Membranes in Regenerative Medicine: New Tricks from an Old Dog? STEM CELLS Transl. Med. 2017, 6 (9), 1767–1776 DOI: 10.1002/sctm.160447. Roux, S.; Bodivit, G.; Bartis, W.; Lebouvier, A.; Chevallier, N.; Fialaire-Legendre, A.; Bierling, P.; Rouard, H. In Vitro Characterization of Patches of Human Mesenchymal Stromal Cells. Tissue Eng. Part A 2015, 21 (3–4), 417–425 DOI: 10.1089/ten.tea.2013.0615. Toda, A.; Okabe, M.; Yoshida, T.; Nikaido, T. The Potential of Amniotic Membrane/Amnion-Derived Cells for Regeneration of Various Tissues. J. Pharmacol. Sci. 2007, 105 (3), 215–228 DOI: 10.1254/jphs.CR0070034. Parolini, O.; Alviano, F.; Bagnara, G. P.; Bilic, G.; Bühring, H.-J.; Evangelista, M.; Hennerbichler, S.; Liu, B.; Magatti, M.; Mao, N.; Miki, T.; Marongiu, F.; Nakajima, H.; Nikaido, T.; Portmann-Lanz, C. B.; Sankar, V.; Soncini, M.; Stadler, G.; Surbek, D.; Takahashi, T. A.; Redl, H.; Sakuragawa, N.; Wolbank, S.; Zeisberger, S.; Zisch, A.; Strom, S. C. Concise Review: Isolation and Characterization of Cells from Human Term Placenta: Outcome of the First International Workshop on Placenta Derived Stem Cells. Stem Cells 2008, 26 (2), 300–311 DOI: 10.1634/stemcells.2007-0594. Díaz-Prado, S.; Muiños-López, E.; Hermida-Gómez, T.; Cicione, C.; Rendal-Vázquez, M. E.; Fuentes-Boquete, I.; de Toro, F. J.; Blanco, F. J. Human Amniotic Membrane as an Alternative Source of Stem Cells for Regenerative Medicine. Differentiation 2011, 81 (3), 162–171 DOI: 10.1016/j.diff.2011.01.005. Miki, T. Amnion-Derived Stem Cells: In Quest of Clinical Applications. Stem Cell Res. Ther. 2011, 2, 25 DOI: 10.1186/scrt66. Insausti, C. L.; Blanquer, M.; García-Hernández, A. M.; Castellanos, G.; Moraleda, J. M. Amniotic Membrane-Derived Stem Cells: Immunomodulatory Properties and Potential Clinical Application. Stem Cells Cloning Adv. Appl. 2014, 7, 53–63 DOI: 10.2147/SCCAA.S58696. Davis, J. W. Skin Transplantation with a Review of 550 Cases at the Johns Hopkins Hospital. Johns Hopkins Med J 1910, 15, 307–396. Trelford, J. D.; Trelford-Sauder, M. The Amnion in Surgery, Past and Present. Am. J. Obstet. Gynecol. 1979, 134 (7), 833–845 DOI: 10.1016/0002-9378(79)90957-8. Dua, H. S.; Gomes, J. A. .; King, A. J.; Maharajan, V. S. The Amniotic Membrane in Ophthalmology. Surv. Ophthalmol. 2004, 49 (1), 51–77 DOI: 10.1016/j.survophthal.2003.10.004. Rötth, A. de. Plastic Repair of Conjunctival Defects with Fetal Membranes. Arch. Ophthalmol. 1940, 23 (3), 522–525 DOI: 10.1001/archopht.1940.00860130586006. Sorsby, A.; Symons, H. Amniotic Membrane Grafts in Caustic Burns of the Eye - Burns of the 2nd Degree. Br. J. Ophthalmol. 1946, 30 (6), 337–345 DOI: 10.1136/bjo.30.6.337. Sorsby, A.; Haythorne, J.; Reed, H. Further Experience with Amniotic Membrane Grafts in Caustic Burns of the Eye. Br. J. Ophthalmol. 1947, 31 (7), 409–418 DOI: 10.1136/bjo.31.7.409.

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

(25)

(26)

(27)

(28)

(29)

(30)

(31)

(32)

(33)

(34)

(35)

(36)

(37)

(38)

Page 24 of 35

Kim, J. C.; Tseng, S. C. Transplantation of Preserved Human Amniotic Membrane for Surface Reconstruction in Severely Damaged Rabbit Corneas. Cornea 1995, 14 (5), 473– 484 DOI: 10.1097/00003226-199509000-00006. Silini, A. R.; Cargnoni, A.; Magatti, M.; Pianta, S.; Parolini, O. The Long Path of Human Placenta, and Its Derivatives, in Regenerative Medicine. Front. Bioeng. Biotechnol. 2015, 3 DOI: 10.3389/fbioe.2015.00162. Tseng, S. C. G.; Espana, E. M.; Kawakita, T.; Di Pascuale, M. A.; Li, W.; He, H.; Liu, T.-S.; Cho, T.-H.; Gao, Y.-Y.; Yeh, L.-K.; Liu, C.-Y. How Does Amniotic Membrane Work? Ocul. Surf. 2004, 2 (3), 177–187 DOI: 10.1016/S1542-0124(12)70059-9. Mohammad, J.; Shenaq, J.; Rabinovsky, E.; Shenaq, S. Modulation of Peripheral Nerve Regeneration: A Tissue-Engineering Approach. The Role of Amnion Tube Nerve Conduit across a 1-Centimeter Nerve Gap. Plast. Reconstr. Surg. 2000, 105 (2), 660–666 DOI: 10.1097/00006534-200002000-00027. Kim, S. S.; Sohn, S. K.; Lee, K. Y.; Lee, M. J.; Roh, M. S.; Kim, C. H. Use of Human Amniotic Membrane Wrap in Reducing Perineural Adhesions in a Rabbit Model of Ulnar Nerve Neurorrhaphy. J. Hand Surg. Eur. Vol. 2010, 35 (3), 214–219 DOI: 10.1177/1753193409352410. Meng, H.; Li, M.; You, F.; Du, J.; Luo, Z. Assessment of Processed Human Amniotic Membrane as a Protective Barrier in Rat Model of Sciatic Nerve Injury. Neurosci. Lett. 2011, 496 (1), 48–53 DOI: 10.1016/j.neulet.2011.03.090. Kesting, M. R.; Wolff, K.-D.; Nobis, C. P.; Rohleder, N. H. Amniotic Membrane in Oral and Maxillofacial Surgery. Oral Maxillofac. Surg. 2014, 18 (2), 153–164 DOI: 10.1007/s10006-012-0382-1. Demirkan, F.; Colakoglu, N.; Herek, O.; Erkula, G. The Use of Amniotic Membrane in Flexor Tendon Repair: An Experimental Model. Arch. Orthop. Trauma Surg. 2002, 122 (7), 396–399 DOI: 10.1007/s00402-002-0418-3. Jirsova, K.; Jones, G. L. A. Amniotic Membrane in Ophthalmology: Properties, Preparation, Storage and Indications for Grafting—a Review. Cell Tissue Bank. 2017, 18 (2), 193–204 DOI: 10.1007/s10561-017-9618-5. Lee, P.-H.; Tsai, S.-H.; Kuo, L.; Hwang, C.-Y.; Kuo, C.-Y.; Yang, V. C.; Chen, J.-K. A Prototype Tissue Engineered Blood Vessel Using Amniotic Membrane as Scaffold. Acta Biomater. 2012, 8 (9), 3342–3348 DOI: 10.1016/j.actbio.2012.05.012. Keeley, R.; Topoluk, N.; Mercuri, J. Tissues Reborn: Fetal Membrane-Derived Matrices and Stem Cells in Orthopedic Regenerative Medicine. Crit. Rev. Biomed. Eng. 2014, 42 (3–4), 249–270 DOI: 10.1615/CritRevBiomedEng.2014011591. Jerman, U. D.; Veranič, P.; Kreft, M. E. Amniotic Membrane Scaffolds Enable the Development of Tissue-Engineered Urothelium with Molecular and Ultrastructural Properties Comparable to that of Native Urothelium. Tissue Eng. Part C Methods 2014, 20 (4), 317–327 DOI: 10.1089/ten.tec.2013.0298. Niknejad, H.; Deihim, T.; Peirovi, H.; Abolghasemi, H. Serum-Free Cryopreservation of Human Amniotic Epithelial Cells before and after Isolation from Their Natural Scaffold. Cryobiology 2013, 67 (1), 56–63 DOI: 10.1016/j.cryobiol.2013.05.001. Riau, A. K.; Beuerman, R. W.; Lim, L. S.; Mehta, J. S. Preservation, Sterilization and deEpithelialization of Human Amniotic Membrane for Use in Ocular Surface Reconstruction. Biomaterials 2010, 31 (2), 216–225 DOI: 10.1016/j.biomaterials.2009.09.034.

ACS Paragon Plus Environment

24

Page 25 of 35 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

(39)

(40)

(41)

(42) (43)

(44)

(45)

(46)

(47)

(48)

(49)

(50)

(51)

Tehrani, F. A.; Ahmadiani, A.; Niknejad, H. The Effects of Preservation Procedures on Antibacterial Property of Amniotic Membrane. Cryobiology 2013, 67 (3), 293–298 DOI: 10.1016/j.cryobiol.2013.08.010. Jin, C. Z.; Park, S. R.; Choi, B. H.; Lee, K.-Y.; Kang, C. K.; Min, B.-H. Human Amniotic Membrane as a Delivery Matrix for Articular Cartilage Repair. Tissue Eng. 2007, 13 (4), 693–702 DOI: 10.1089/ten.2006.0184. Nakamura, T.; Sekiyama, E.; Takaoka, M.; Bentley, A. J.; Yokoi, N.; Fullwood, N. J.; Kinoshita, S. The Use of Trehalose-Treated Freeze-Dried Amniotic Membrane for Ocular Surface Reconstruction. Biomaterials 2008, 29 (27), 3729–3737 DOI: 10.1016/j.biomaterials.2008.05.023. Gilbert, T. W.; Sellaro, T. L.; Badylak, S. F. Decellularization of Tissues and Organs. Biomaterials 2006, 27 (19), 3675–3683 DOI: 10.1016/j.biomaterials.2006.02.014. 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 (Supplement C), 25–34 DOI: 10.1016/j.ymeth.2015.03.005. Crapo, P. M.; Gilbert, T. W.; Badylak, S. F. An Overview of Tissue and Whole Organ Decellularization Processes. Biomaterials 2011, 32 (12), 3233–3243 DOI: 10.1016/j.biomaterials.2011.01.057. Hopkinson, A.; Shanmuganathan, V. A.; Gray, T.; Yeung, A. M.; Lowe, J.; James, D. K.; Dua, H. S. Optimization of Amniotic Membrane (AM) Denuding for Tissue Engineering. Tissue Eng. Part C Methods 2008, 14 (4), 371–381 DOI: 10.1089/ten.tec.2008.0315. Saghizadeh, M.; Winkler, M. A.; Kramerov, A. A.; Hemmati, D. M.; Ghiam, C. A.; Dimitrijevich, S. D.; Sareen, D.; Ornelas, L.; Ghiasi, H.; Brunken, W. J.; Maguen, E.; Rabinowitz, Y. S.; Svendsen, C. N.; Jirsova, K.; Ljubimov, A. V. A Simple Alkaline Method for Decellularizing Human Amniotic Membrane for Cell Culture. PLoS ONE 2013, 8 (11), e79632 DOI: 10.1371/journal.pone.0079632. Wilshaw, S.-P.; Kearney, J. N.; Fisher, J.; Ingham, E. Production of an Acellular Amniotic Membrane Matrix for Use in Tissue Engineering. Tissue Eng. 2006, 12 (8), 2117–2129 DOI: 10.1089/ten.2006.12.2117. Figueiredo, G. S.; Bojic, S.; Rooney, P.; Wilshaw, S.-P.; Connon, C. J.; Gouveia, R. M.; Paterson, C.; Lepert, G.; Mudhar, H. S.; Figueiredo, F. C.; Lako, M. Gamma-Irradiated Human Amniotic Membrane Decellularised with Sodium Dodecyl Sulfate Is a More Efficient Substrate for the Ex Vivo Expansion of Limbal Stem Cells. Acta Biomater. 2017, 61, 124–133 DOI: 10.1016/j.actbio.2017.07.041. Wehmeyer, J. L.; Natesan, S.; Christy, R. J. Development of a Sterile Amniotic Membrane Tissue Graft Using Supercritical Carbon Dioxide. Tissue Eng. Part C Methods 2015, 21 (7), 649–659 DOI: 10.1089/ten.tec.2014.0304. Gindraux, F.; Laurent, R.; Nicod, L.; de Billy, B.; Meyer, C.; Zwetyenga, N.; Wajszczak, L.; Garbuio, P.; Obert, L. Human Amniotic Membrane: Clinical Uses, Patents and Marketed Products. Recent Pat. Regen. Med. 2013, 3 (3), 193–214 DOI: 10.2174/22102965113039990021. Spoerl, E.; Wollensak, G.; Reber, F.; Pillunat, L. Cross-Linking of Human Amniotic Membrane by Glutaraldehyde. Ophthalmic Res. 2004, 36 (2), 71–77 DOI: 10.1159/000076884.

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

(52)

(53)

(54)

(55)

(56) (57)

(58)

(59)

(60)

(61)

(62)

(63)

(64)

Page 26 of 35

Ma, D. H.-K.; Lai, J.-Y.; Cheng, H.-Y.; Tsai, C.-C.; Yeh, L.-K. Carbodiimide CrossLinked Amniotic Membranes for Cultivation of Limbal Epithelial Cells. Biomaterials 2010, 31 (25), 6647–6658 DOI: 10.1016/j.biomaterials.2010.05.034. Chau, D. Y. S.; Brown, S. V.; Mather, M. L.; Hutter, V.; Tint, N. L.; Dua, H. S.; Rose, F. R. A. J.; Ghaemmaghami, A. M. Tissue Transglutaminase (TG-2) Modified Amniotic Membrane: A Novel Scaffold for Biomedical Applications. Biomed. Mater. 2012, 7 (4), 45011 DOI: 10.1088/1748-6041/7/4/045011. Tanaka, Y.; Kubota, A.; Yokokura, S.; Uematsu, M.; Shi, D.; Yamato, M.; Okano, T.; Quantock, A. J.; Nishida, K. Optical Mechanical Refinement of Human Amniotic Membrane by Dehydration and Cross-Linking. J. Tissue Eng. Regen. Med. 2012, 6 (9), 731–737 DOI: 10.1002/term.479. Huang, G.; Ji, S.; Luo, P.; Liu, H.; Zhu, S.; Wang, G.; Zhou, P.; Xiao, S.; Xia, Z. Accelerated Expansion of Epidermal Keratinocyte and Improved Dermal Reconstruction Achieved by Engineered Amniotic Membrane. Cell Transplant. 2013, 22 (10), 1831– 1844 DOI: 10.3727/096368912X657945. Lai, J.-Y. Photo-Cross-Linking of Amniotic Membranes for Limbal Epithelial Cell Cultivation. Mater. Sci. Eng. C 2014, 45, 313–319 DOI: 10.1016/j.msec.2014.09.001. Ma, D. H.-K.; Chen, H.-C.; Ma, K. S.-K.; Lai, J.-Y.; Yang, U.; Yeh, L.-K.; Hsueh, Y.-J.; Chu, W.-K.; Lai, C.-H.; Chen, J.-K. Preservation of Human Limbal Epithelial Progenitor Cells on Carbodiimide Cross-Linked Amniotic Membrane via Integrin-Linked KinaseMediated Wnt Activation. Acta Biomater. 2016, 31, 144–155 DOI: 10.1016/j.actbio.2015.11.042. Hariya, T.; Tanaka, Y.; Yokokura, S.; Nakazawa, T. Transparent, Resilient Human Amniotic Membrane Laminates for Corneal Transplantation. Biomaterials 2016, 101, 76–85 DOI: 10.1016/j.biomaterials.2016.05.038. Nogami, M.; Kimura, T.; Seki, S.; Matsui, Y.; Yoshida, T.; Koike-Soko, C.; Okabe, M.; Motomura, H.; Gejo, R.; Nikaido, T. A Human Amnion-Derived Extracellular MatrixCoated Cell-Free Scaffold for Cartilage Repair: In Vitro and In Vivo Studies. Tissue Eng. Part A 2016, 22 (7–8), 680–688 DOI: 10.1089/ten.tea.2015.0285. Amensag, S.; Goldberg, L. A.; O’Malley, K. A.; Rush, D. S.; Berceli, S. A.; McFetridge, P. S. Pilot Assessment of a Human Extracellular Matrix-Based Vascular Graft in a Rabbit Model. J. Vasc. Surg. 2016 DOI: 10.1016/j.jvs.2016.02.046. Hortensius, R. A.; Ebens, J. H.; Harley, B. A. C. Immunomodulatory Effects of Amniotic Membrane Matrix Incorporated into Collagen Scaffolds. J. Biomed. Mater. Res. A 2016, 104 (6), 1332–1342 DOI: 10.1002/jbm.a.35663. Adamowicz, J.; Pokrywczyńska, M.; Tworkiewicz, J.; Kowalczyk, T.; Breda, S. V. van; Tyloch, D.; Kloskowski, T.; Bodnar, M.; Skopinska-Wisniewska, J.; Marszałek, A.; Frontczak-Baniewicz, M.; Kowalewski, T. A.; Drewa, T. New Amniotic Membrane Based Biocomposite for Future Application in Reconstructive Urology. PLOS ONE 2016, 11 (1), e0146012 DOI: 10.1371/journal.pone.0146012. Allen, A. B.; Priddy, L. B.; Li, M.-T. A.; Guldberg, R. E. Functional Augmentation of Naturally-Derived Materials for Tissue Regeneration. Ann. Biomed. Eng. 2015, 43 (3), 555–567 DOI: 10.1007/s10439-014-1192-4. Gholipourmalekabadi, M.; Samadikuchaksaraei, A.; Seifalian, A. M.; Urbanska, A.; Ghanbarian, H.; Hardy, J. G.; Omrani, M.; Mozafari, M.; Reis, R. L.; Kundu, S. C. Silk

ACS Paragon Plus Environment

26

Page 27 of 35 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

(65)

(66)

(67)

(68)

(69)

(70)

(71)

(72)

(73)

(74)

(75)

(76)

Fibroin/amniotic Membrane 3D Bi-Layered Artificial Skin. Biomed. Mater. 2017 DOI: 10.1088/1748-605X/aa999b. Shi, P.; Gao, M.; Shen, Q.; Hou, L.; Zhu, Y.; Wang, J. Biocompatible Surgical Meshes Based on Decellularized Human Amniotic Membrane. Mater. Sci. Eng. C 2015, 54, 112– 119 DOI: 10.1016/j.msec.2015.05.008. Brennan, J. A.; Arrizabalaga, J. H.; Nollert, M. U. Development of a Small Diameter Vascular Graft Using the Human Amniotic Membrane. Cardiovasc. Eng. Technol. 2014, 5 (1), 96–109 DOI: 10.1007/s13239-013-0170-6. Amensag, S.; McFetridge, P. S. Rolling the Human Amnion to Engineer Laminated Vascular Tissues. Tissue Eng. Part C Methods 2012, 18 (11), 903–912 DOI: 10.1089/ten.tec.2012.0119. Kesting, M. R.; Wolff, K.-D.; Mücke, T.; Demtroeder, C.; Kreutzer, K.; Schulte, M.; Jacobsen, F.; Hirsch, T.; Loeffelbein, D. J.; Steinstraesser, L. A Bioartificial Surgical Patch from Multilayered Human Amniotic membrane—In Vivo Investigations in a Rat Model. J. Biomed. Mater. Res. B Appl. Biomater. 2009, 90B (2), 930–938 DOI: 10.1002/jbm.b.31365. Prabhasawat, P.; Tesavibul, N.; Komolsuradej, W. Single and Multilayer Amniotic Membrane Transplantation for Persistent Corneal Epithelial Defect with and without Stromal Thinning and Perforation. Br. J. Ophthalmol. 2001, 85 (12), 1455–1463 DOI: 10.1136/bjo.85.12.1455. Sridhar, M. S.; Bansal, A. K.; Rao, G. N. Multilayered Amniotic Membrane Transplantation for Partial Thickness Scleral Thinning Following Pterygium Surgery. Eye 2002, 16 (5), 639–642 DOI: 10.1038/sj.eye.6700123. Kesting, M. R.; Loeffelbein, D. J.; Classen, M.; Slotta-Huspenina, J.; Hasler, R. J.; Jacobsen, F.; Kreutzer, K.; Al-Benna, S.; Wolff, K.-D.; Steinstraesser, L. Repair of Oronasal Fistulas with Human Amniotic Membrane in Minipigs. Br. J. Oral Maxillofac. Surg. 2010, 48 (2), 131–135 DOI: 10.1016/j.bjoms.2009.04.025. Rohleder, N. H.; Loeffelbein, D. J.; Feistl, W.; Eddicks, M.; Wolff, K.-D.; Gulati, A.; Steinstraesser, L.; Kesting, M. R. Repair of Oronasal Fistulae by Interposition of Multilayered Amniotic Membrane Allograft. Plast. Reconstr. Surg. 2013, 132 (1), 172– 181 DOI: 10.1097/PRS.0b013e3182910b50. Murphy, S. V.; Skardal, A.; Song, L.; Sutton, K.; Haug, R.; Mack, D. L.; Jackson, J.; Soker, S.; Atala, A. Solubilized Amnion Membrane Hyaluronic Acid Hydrogel Accelerates Full-Thickness Wound Healing. STEM CELLS Transl. Med. 2017, 6 (11), 2020–2032 DOI: 10.1002/sctm.17-0053. Ji, S.; Xiao, S.; Luo, P.; Huang, G.; Wang, G.; Zhu, S.; Wu, M.; Xia, Z. An Epidermal Stem Cells Niche Microenvironment Created by Engineered Human Amniotic Membrane. Biomaterials 2011, 32 (31), 7801–7811 DOI: 10.1016/j.biomaterials.2011.06.076. Fukuda, K.; Chikama, T.; Nakamura, M.; Nishida, T. Differential Distribution of Subchains of the Basement Membrane Components Type IV Collagen and Laminin among the Amniotic Membrane, Cornea, and Conjunctiva. Cornea 1999, 18 (1), 73–79 DOI: 10.1097/00003226-199901000-00013. Rahman, I.; Said, D. G.; Maharajan, V. S.; Dua, H. S. Amniotic Membrane in Ophthalmology: Indications and Limitations. Eye 2009, 23 (10), 1954–1961 DOI: 10.1038/eye.2008.410.

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

(77)

(78)

(79)

(80)

(81)

(82)

(83) (84)

(85)

(86)

(87)

(88)

(89)

(90)

Page 28 of 35

Malhotra, C.; Jain, A. K. Human Amniotic Membrane Transplantation: Different Modalities of Its Use in Ophthalmology. World J. Transplant. 2014, 4 (2), 111–121 DOI: 10.5500/wjt.v4.i2.111. John, T.; Foulks, G. N.; John, M. E.; Cheng, K.; Hu, D. Amniotic Membrane in the Surgical Management of Acute Toxic Epidermal Necrolysis. Ophthalmology 2002, 109 (2), 351–360 DOI: 10.1016/S0161-6420(01)00900-9. Hsu, M.; Jayaram, A.; Verner, R.; Lin, A.; Bouchard, C. Indications and Outcomes of Amniotic Membrane Transplantation in the Management of Acute Stevens–johnson Syndrome and Toxic Epidermal Necrolysis: A Case–control Study. Cornea 2012, 31 (12), 1394–1402 DOI: 10.1097/ICO.0b013e31823d02a8. Schwab, I. R.; Reyes, M.; Isseroff, R. R. Successful Transplantation of Bioengineered Tissue Replacements in Patients with Ocular Surface Disease. Cornea 2000, 19 (4), 421– 426 DOI: 10.1016/S0002-9394(00)00747-9. Tsai, R. J.; Li, L. M.; Chen, J. K. Reconstruction of Damaged Corneas by Transplantation of Autologous Limbal Epithelial Cells. N. Engl. J. Med. 2000, 343 (2), 86–93 DOI: 10.1056/NEJM200007133430202. Koizumi, N.; Inatomi, T.; Suzuki, T.; Sotozono, C.; Kinoshita, S. Cultivated Corneal Epithelial Stem Cell Transplantation in Ocular Surface Disorders. Ophthalmology 2001, 108 (9), 1569–1574 DOI: 10.1016/S0161-6420(01)00694-7. Ahmad, S. Concise Review: Limbal Stem Cell Deficiency, Dysfunction, and Distress. Stem Cells Transl. Med. 2012, 1 (2), 110–115 DOI: 10.5966/sctm.2011-0037. Nakamura, T.; Inatomi, T.; Sotozono, C.; Amemiya, T.; Kanamura, N.; Kinoshita, S. Transplantation of Cultivated Autologous Oral Mucosal Epithelial Cells in Patients with Severe Ocular Surface Disorders. Br. J. Ophthalmol. 2004, 88 (10), 1280–1284 DOI: 10.1136/bjo.2003.038497. Nishida, K.; Yamato, M.; Hayashida, Y.; Watanabe, K.; Yamamoto, K.; Adachi, E.; Nagai, S.; Kikuchi, A.; Maeda, N.; Watanabe, H.; Okano, T.; Tano, Y. Corneal Reconstruction with Tissue-Engineered Cell Sheets Composed of Autologous Oral Mucosal Epithelium. N. Engl. J. Med. 2004, 351 (12), 1187–1196 DOI: 10.1056/NEJMoa040455. Ma, Y.; Xu, Y.; Xiao, Z.; Yang, W.; Zhang, C.; Song, E.; Du, Y.; Li, L. Reconstruction of Chemically Burned Rat Corneal Surface by Bone Marrow–Derived Human Mesenchymal Stem Cells. STEM CELLS 2006, 24 (2), 315–321 DOI: 10.1634/stemcells.2005-0046. Rohaina, C. M.; Then, K. Y.; Ng, A. M. H.; Wan Abdul Halim, W. H.; Zahidin, A. Z. M.; Saim, A.; Idrus, R. B. H. Reconstruction of Limbal Stem Cell Deficient Corneal Surface with Induced Human Bone Marrow Mesenchymal Stem Cells on Amniotic Membrane. Transl. Res. 2014, 163 (3), 200–210 DOI: 10.1016/j.trsl.2013.11.004. Fernandes, M.; Sridhar, M. S.; Sangwan, V. S.; Rao, G. N. Amniotic Membrane Transplantation for Ocular Surface Reconstruction. Cornea 2005, 24 (6), 643–653 DOI: 10.1097/01.ico.0000151501.80952.c5. Nakamura, T.; Inatomi, T.; Sotozono, C.; Koizumi, N.; Kinoshita, S. Ocular Surface Reconstruction Using Stem Cell and Tissue Engineering. Prog. Retin. Eye Res. 2016, 51, 187–207 DOI: 10.1016/j.preteyeres.2015.07.003. Kotomin, I.; Valtink, M.; Hofmann, K.; Frenzel, A.; Morawietz, H.; Werner, C.; Funk, R. H. W.; Engelmann, K. Sutureless Fixation of Amniotic Membrane for Therapy of Ocular

ACS Paragon Plus Environment

28

Page 29 of 35 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

(91)

(92)

(93)

(94)

(95)

(96)

(97)

(98)

(99) (100)

(101)

(102)

(103)

Surface Disorders. PloS One 2015, 10 (5), e0125035 DOI: 10.1371/journal.pone.0125035. Duchesne, B.; Tahi, H.; Galand, A. Use of Human Fibrin Glue and Amniotic Membrane Transplant in Corneal Perforation. Cornea 2001, 20 (2), 230–232 DOI: 10.1097/00003226-200103000-00027. Hick, S.; Demers, P. E.; Brunette, I.; La, C.; Mabon, M.; Duchesne, B. Amniotic Membrane Transplantation and Fibrin Glue in the Management of Corneal Ulcers and Perforations: A Review of 33 Cases. Cornea 2005, 24 (4), 369–377 DOI: 10.1097/01.ico.0000151547.08113.d1. Takaoka, M.; Nakamura, T.; Sugai, H.; Bentley, A. J.; Nakajima, N.; Fullwood, N. J.; Yokoi, N.; Hyon, S.-H.; Kinoshita, S. Sutureless Amniotic Membrane Transplantation for Ocular Surface Reconstruction with a Chemically Defined Bioadhesive. Biomaterials 2008, 29 (19), 2923–2931 DOI: 10.1016/j.biomaterials.2008.03.027. Takaoka, M.; Nakamura, T.; Sugai, H.; Bentley, A. J.; Nakajima, N.; Yokoi, N.; Fullwood, N. J.; Hyon, S.-H.; Kinoshita, S. Novel Sutureless Keratoplasty with a Chemically Defined Bioadhesive. Invest. Ophthalmol. Vis. Sci. 2009, 50 (6), 2679–2685 DOI: 10.1167/iovs.08-2944. Verter, E. E.; Gisel, T. E.; Yang, P.; Johnson, A. J.; Redmond, R. W.; Kochevar, I. E. Light-Initiated Bonding of Amniotic Membrane to Cornea. Invest. Ophthalmol. Vis. Sci. 2011, 52 (13), 9470–9477 DOI: 10.1167/iovs.11-7248. Soeken, T. A.; Zhu, H.; DeMartelaere, S.; Davies, B. W.; Kim, M.; Wang, H.-C.; Aden, J.; Grimm, R.; Alt, C.; Kochevar, I. E.; Johnson, A. J. Sealing of Corneal Lacerations Using Photoactivated Rose Bengal Dye and Amniotic Membrane. Cornea 2018, 37 (2), 211–217 DOI: 10.1097/ICO.0000000000001389. Sheha, H.; Liang, L.; Li, J.; Tseng, S. C. G. Sutureless Amniotic Membrane Transplantation for Severe Bacterial Keratitis. Cornea 2009, 28 (10), 1118–1123 DOI: 10.1097/ICO.0b013e3181a2abad. Kesting, M. R.; Wolff, K.-D.; Hohlweg-Majert, B.; Steinstraesser, L. The Role of Allogenic Amniotic Membrane in Burn Treatment. J. Burn Care Res. 2008, 29 (6), 907– 916 DOI: 10.1097/BCR.0b013e31818b9e40. Lo, V.; Pope, E. Amniotic Membrane Use in Dermatology. Int. J. Dermatol. 2009, 48 (9), 935–940 DOI: 10.1111/j.1365-4632.2009.04173.x. Fairbairn, N. G.; Randolph, M. A.; Redmond, R. W. The Clinical Applications of Human Amnion in Plastic Surgery. J. Plast. Reconstr. Aesthet. Surg. 2014, 67 (5), 662–675 DOI: 10.1016/j.bjps.2014.01.031. Mohammadi, A. A.; Seyed Jafari, S. M.; Kiasat, M.; Tavakkolian, A. R.; Imani, M. T.; Ayaz, M.; Tolide-ie, H. R. Effect of Fresh Human Amniotic Membrane Dressing on Graft Take in Patients with Chronic Burn Wounds Compared with Conventional Methods. Burns 2013, 39 (2), 349–353 DOI: 10.1016/j.burns.2012.07.010. Mohammadi, A. A.; Johari, H. G.; Eskandari, S. Effect of Amniotic Membrane on Graft Take in Extremity Burns. Burns 2013, 39 (6), 1137–1141 DOI: 10.1016/j.burns.2013.01.017. Singh, R.; Kumar, A.; Singh, D.; Malviya, A. Use of Gamma-Irradiated Amniotic Membrane for the Healing of Split Skin Graft Donor Site. Tissue Eng. Regen. Med. 2013, 10 (3), 110–114 DOI: 10.1007/s13770-013-0004-5.

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

Page 30 of 35

(104) Loeffelbein, D. J.; Rohleder, N. H.; Eddicks, M.; Baumann, C. M.; Stoeckelhuber, M.; Wolff, K.-D.; Drecoll, E.; Steinstraesser, L.; Hennerbichler, S.; Kesting, M. R. Evaluation of Human Amniotic Membrane as a Wound Dressing for Split-Thickness Skin-Graft Donor Sites. BioMed Res. Int. 2014, 2014, 572183 DOI: 10.1155/2014/572183. (105) Lo, V.; Lara-Corrales, I.; Stuparich, A.; Pope, E. Amniotic Membrane Grafting in Patients with Epidermolysis Bullosa with Chronic Wounds. J. Am. Acad. Dermatol. 2010, 62 (6), 1038–1044 DOI: 10.1016/j.jaad.2009.02.048. (106) Serena, T. E.; Carter, M. J.; Le, L. T.; Sabo, M. J.; DiMarco, D. T.; EpiFix VLU Study Group. A Multicenter, Randomized, Controlled Clinical Trial Evaluating the Use of Dehydrated Human Amnion/chorion Membrane Allografts and Multilayer Compression Therapy vs. Multilayer Compression Therapy Alone in the Treatment of Venous Leg Ulcers. Wound Repair Regen. 2014, 22 (6), 688–693 DOI: 10.1111/wrr.12227. (107) Song, M.; Wang, W.; Ye, Q.; Bu, S.; Shen, Z.; Zhu, Y. The Repairing of Full-Thickness Skin Deficiency and Its Biological Mechanism Using Decellularized Human Amniotic Membrane as the Wound Dressing. Mater. Sci. Eng. C 2017, 77, 739–747 DOI: 10.1016/j.msec.2017.03.232. (108) Redondo, P.; Del Olmo, J.; García-Guzman, M.; Guembe, L.; Prósper, F. Repigmentation of Vitiligo by Transplantation of Autologous Melanocyte Cells Cultured on Amniotic Membrane. Br. J. Dermatol. 2008, 158 (5), 1168–1171 DOI: 10.1111/j.1365-2133.2008.08521.x. (109) Redondo, P.; Giménez de Azcarate, A.; Marqués, L.; García-Guzman, M.; Andreu, E.; Prósper, F. Amniotic Membrane as a Scaffold for Melanocyte Transplantation in Patients with Stable Vitiligo. Dermatol. Res. Pract. 2011, 2011, 1–6 DOI: 10.1155/2011/532139. (110) Yang, L.; Shirakata, Y.; Shudou, M.; Dai, X.; Tokumaru, S.; Hirakawa, S.; Sayama, K.; Hamuro, J.; Hashimoto, K. New Skin-Equivalent Model from de-Epithelialized Amnion Membrane. Cell Tissue Res. 2006, 326 (1), 69–77 DOI: 10.1007/s00441-006-0208-2. (111) Yang, L.; Shirakata, Y.; Tokumaru, S.; Xiuju, D.; Tohyama, M.; Hanakawa, Y.; Hirakawa, S.; Sayama, K.; Hashimoto, K. Living Skin Equivalents Constructed Using Human Amnions as a Matrix. J. Dermatol. Sci. 2009, 56 (3), 188–195 DOI: 10.1016/j.jdermsci.2009.09.009. (112) Sánchez-Sánchez, R.; Brena-Molina, A.; Martínez-López, V.; Melgarejo-Ramírez, Y.; Tamay de Dios, L.; Gómez-García, R.; Reyes-Frías, M. de L.; Rodríguez-Rodríguez, L.; Garciadiego-Cázares, D.; Lugo-Martínez, H.; Ibarra, C.; Martínez-Pardo, M. E.; Velasquillo-Martínez, C. Generation of Two Biological Wound Dressings as a Potential Delivery System of Human Adipose-Derived Mesenchymal Stem Cells: ASAIO J. 2015, 61 (6), 718–725 DOI: 10.1097/MAT.0000000000000277. (113) Arai, N.; Tsuno, H.; Okabe, M.; Yoshida, T.; Koike, C.; Noguchi, M.; Nikaido, T. Clinical Application of a Hyperdry Amniotic Membrane on Surgical Defects of the Oral Mucosa. J. Oral Maxillofac. Surg. 2012, 70 (9), 2221–2228 DOI: 10.1016/j.joms.2011.09.033. (114) Mücke, T.; Loeffelbein, D. J.; Hölzle, F.; Slotta-Huspenina, J.; Borgmann, A.; Kanatas, A. N.; Mitchell, D. A.; Wagenpfeil, S.; Wolff, K.-D.; Kesting, M. R. Intraoral Defect Coverage with Prelaminated Epigastric Fat Flaps with Human Amniotic Membrane in Rats. J. Biomed. Mater. Res. B Appl. Biomater. 2010, 95B (2), 466–474 DOI: 10.1002/jbm.b.31738.

ACS Paragon Plus Environment

30

Page 31 of 35 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

(115) Tuncel, U.; Ozgenel, G. Y. Use of Human Amniotic Membrane as an Interpositional Material in Treatment of Temporomandibular Joint Ankylosis. J. Oral Maxillofac. Surg. 2011, 69 (6), e58–e66 DOI: 10.1016/j.joms.2010.12.044. (116) Ahn, K.-M.; Lee, J.-H.; Hwang, S.-J.; Choung, P.-H.; Kim, M.-J.; Park, H.-J.; Park, J.K.; Jahng, J.; Yang, E.-K. Fabrication of Myomucosal Flap Using Tissue-Engineered Bioartificial Mucosa Constructed With Oral Keratinocytes Cultured on Amniotic Membrane. Artif. Organs 2006, 30 (6), 411–423 DOI: 10.1111/j.15251594.2006.00236.x. (117) Amemiya, T.; Nakamura, T.; Yamamoto, T.; Kinoshita, S.; Kanamura, N. Immunohistochemical Study of Oral Epithelial Sheets Cultured on Amniotic Membrane for Oral Mucosal Reconstruction. Biomed. Mater. Eng. 2010, 20 (1), 37–45 DOI: 10.3233/BME-2010-0613. (118) Amemiya, T.; Nakamura, T.; Yamamoto, T.; Kinoshita, S.; Kanamura, N. Autologous Transplantation of Oral Mucosal Epithelial Cell Sheets Cultured on an Amniotic Membrane Substrate for Intraoral Mucosal Defects. PLoS ONE 2015, 10 (4), e0125391 DOI: 10.1371/journal.pone.0125391. (119) Hsueh, Y.-J.; Huang, S.-F.; Lai, J.-Y.; Ma, S.-C.; Chen, H.-C.; Wu, S.-E.; Wang, T.-K.; Sun, C.-C.; Ma, K. S.-K.; Chen, J.-K.; Lai, C.-H.; Ma, D. H.-K. Preservation of Epithelial Progenitor Cells from Collagenase-Digested Oral Mucosa during Ex Vivo Cultivation. Sci. Rep. 2016, 6, 36266 DOI: 10.1038/srep36266. (120) Shojaku, H.; Takakura, H.; Okabe, M.; Fujisaka, M.; Watanabe, Y.; Nikaido, T. Effect of Hyperdry Amniotic Membrane Patches Attached over the Bony Surface of Mastoid Cavities in Canal Wall down Tympanoplasty. The Laryngoscope 2011, 121 (9), 1953– 1957 DOI: 10.1002/lary.22082. (121) Kanazawa, Y.; Shojaku, H.; Okabe, M.; Fujisaka, M.; Takakura, H.; Tachino, H.; Tsubota, M.; Watanabe, Y.; Nikaido, T. Application of Hyperdry Amniotic Membrane Patches without Fibrin Glue over the Bony Surface of Mastoid Cavities in Canal Wall down Tympanoplasty. Acta Otolaryngol. (Stockh.) 2012, 132 (12), 1282–1287 DOI: 10.3109/00016489.2012.701329. (122) Zhang, L.; Hu, J.; Athanasiou, K. A. The Role of Tissue Engineering in Articular Cartilage Repair and Regeneration. Crit. Rev. Biomed. Eng. 2009, 37 (1–2), 1–57 DOI: 10.1615/CritRevBiomedEng.v37.i1-2.10. (123) Yan, L.-P.; Oliveira, J. M.; Oliveira, A. L.; Reis, R. L. Current Concepts and Challenges in Osteochondral Tissue Engineering and Regenerative Medicine. ACS Biomater. Sci. Eng. 2015, 1 (4), 183–200 DOI: 10.1021/ab500038y. (124) Díaz-Prado, S.; Rendal-Vázquez, M. E.; Muiños-López, E.; Hermida-Gómez, T.; Rodríguez-Cabarcos, M.; Fuentes-Boquete, I.; Toro, F. J. de; Blanco, F. J. Potential Use of the Human Amniotic Membrane as a Scaffold in Human Articular Cartilage Repair. Cell Tissue Bank. 2010, 11 (2), 183–195 DOI: 10.1007/s10561-009-9144-1. (125) Krishnamurithy, G.; Shilpa, P. N.; Ahmad, R. E.; Sulaiman, S.; Ng, C. L. L.; Kamarul, T. Human Amniotic Membrane as a Chondrocyte Carrier Vehicle/substrate: In Vitro Study. J. Biomed. Mater. Res. A 2011, 99A (3), 500–506 DOI: 10.1002/jbm.a.33184. (126) Tan, S.-L.; Sulaiman, S.; Pingguan-Murphy, B.; Selvaratnam, L.; Tai, C.-C.; Kamarul, T. Human Amnion as a Novel Cell Delivery Vehicle for Chondrogenic Mesenchymal Stem Cells. Cell Tissue Bank. 2011, 12 (1), 59–70 DOI: 10.1007/s10561-009-9164-x.

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 35

(127) Garcia, D.; Giuseppe Longo, U.; Vaquero, J.; Forriol, F.; Loppini, M.; S. Khan, W.; Denaro, V. Amniotic Membrane Transplant for Articular Cartilage Repair: An Experimental Study in Sheep. Curr. Stem Cell Res. Ther. 2015, 10 (1), 77–83 DOI: 10.2174/1574888X09666140710120012. (128) Li, W.; Ma, G.; Brazile, B.; Li, N.; Dai, W.; Butler, J. R.; Claude, A. A.; Wertheim, J. A.; Liao, J.; Wang, B. Investigating the Potential of Amnion-Based Scaffolds as a Barrier Membrane for Guided Bone Regeneration. Langmuir 2015, 31 (31), 8642–8653 DOI: 10.1021/acs.langmuir.5b02362. (129) Tang, K.; Wu, J.; Xiong, Z.; Ji, Y.; Sun, T.; Guo, X. Human Acellular Amniotic Membrane: A Potential Osteoinductive Biomaterial for Bone Regeneration. J. Biomater. Appl. 2018, 32 (6), 754–764 DOI: 10.1177/0885328217739753. (130) Chen, Y.-J.; Chung, M.-C.; Jane Yao, C.-C.; Huang, C.-H.; Chang, H.-H.; Jeng, J.-H.; Young, T.-H. The Effects of Acellular Amniotic Membrane Matrix on Osteogenic Differentiation and ERK1/2 Signaling in Human Dental Apical Papilla Cells. Biomaterials 2012, 33 (2), 455–463 DOI: 10.1016/j.biomaterials.2011.09.065. (131) Lindenmair, A.; Wolbank, S.; Stadler, G.; Meinl, A.; Peterbauer-Scherb, A.; Eibl, J.; Polin, H.; Gabriel, C.; van Griensven, M.; Redl, H. Osteogenic Differentiation of Intact Human Amniotic Membrane. Biomaterials 2010, 31 (33), 8659–8665 DOI: 10.1016/j.biomaterials.2010.07.090. (132) Wang, F.; Liu, T.; Yang, L.; Zhang, G.; Liu, H.; Yi, X.; Yang, X.; Lin, T.; Qin, W.; Yuan, J. Urethral Reconstruction with Tissue-Engineered Human Amniotic Scaffold in Rabbit Urethral Injury Models. Med. Sci. Monit. Int. Med. J. Exp. Clin. Res. 2014, 20, 2430–2438 DOI: 10.12659/MSM.891042. (133) Sharifiaghdas, F.; Hamzehiesfahani, N.; Moghadasali, R.; Ghaemimanesh, F.; Baharvand, H. Human Amniotic Membrane as a Suitable Matrix for Growth of Mouse Urothelial Cells in Comparison with Human Peritoneal and Omentum Membranes. Urol. J. 2009, 4 (2), 71–78 DOI: 10.22037/uj.v4i2.134. (134) Sharifiaghdas, F.; Moghadasali, R.; Baharvand, H.; Hosseini-Moghaddam, S. M.; Mahmoudnejad, N. Special Characteristics of Culturing Mature Human Bladder Smooth Muscle Cells on Human Amniotic Membrane as a Suitable Matrix. Urol. J. 2009, 6 (4), 283–288 DOI: 10.22037/uj.v6i4.505. (135) Tsai, S.-H.; Liu, Y.-W.; Tang, W.-C.; Zhou, Z.-W.; Hwang, C.-Y.; Hwang, G.-Y.; Ou, B.-R.; Hu, C.-P.; Yang, V. C.; Chen, J.-K. Characterization of Porcine Arterial Endothelial Cells Cultured on Amniotic Membrane, a Potential Matrix for Vascular Tissue Engineering. Biochem. Biophys. Res. Commun. 2007, 357 (4), 984–990 DOI: 10.1016/j.bbrc.2007.04.047. (136) Kakavand, M.; Yazdanpanah, G.; Ahmadiani, A.; Niknejad, H. Blood Compatibility of Human Amniotic Membrane Compared with Heparin‐coated ePTFE for Vascular Tissue Engineering. J. Tissue Eng. Regen. Med. 2017, 11 (6), 1701–1709 DOI: 10.1002/term.2064. (137) Peirovi, H.; Rezvani, N.; Hajinasrollah, M.; Mohammadi, S. S.; Niknejad, H. Implantation of Amniotic Membrane as a Vascular Substitute in the External Jugular Vein of Juvenile Sheep. J. Vasc. Surg. 2012, 56 (4), 1098–1104 DOI: 10.1016/j.jvs.2012.02.036. (138) Hsiao, Y.-C.; Lee, H.-W.; Chen, Y.-T.; Young, T.-H.; Yang, T.-L. The Impact of Compositional Topography of Amniotic Membrane Scaffold on Tissue Morphogenesis

ACS Paragon Plus Environment

32

Page 33 of 35 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

(139)

(140)

(141)

(142)

(143)

(144)

(145)

(146)

(147)

(148)

of Salivary Gland. Biomaterials 2011, 32 (19), 4424–4432 DOI: 10.1016/j.biomaterials.2011.02.057. Poghosyan, T.; Gaujoux, S.; Vanneaux, V.; Bruneval, P.; Domet, T.; Lecourt, S.; Jarraya, M.; Sfeir, R.; Larghero, J.; Cattan, P. In Vitro Development and Characterization of a Tissue-Engineered Conduit Resembling Esophageal Wall Using Human and Pig Skeletal Myoblast, Oral Epithelial Cells, and Biologic Scaffolds. Tissue Eng. Part A 2013, 19 (19–20), 2242–2252 DOI: 10.1089/ten.tea.2012.0565. Zhao, B.; Liang, L.; Wang, J.; Ren, C.; Hu, M.; Wu, H.; Chen, L.; Liu, X.; Xu, F.; Zheng, X.; Chen, J.; Cui, S. The Effect of a Human Acellular Amniotic Membrane Loaded with Mechanical Stretch-Stimulated Bone Marrow Mesenchymal Stem Cells for the Treatment of Pelvic Floor Dysfunction. RSC Adv. 2017, 7 (59), 37086–37094 DOI: 10.1039/C7RA02020A. Despeyroux, A.; Duret, C.; Gondeau, C.; Perez-Gracia, E.; Chuttoo, L.; de Boussac, H.; Briolotti, P.; Bony, C.; Noël, D.; Jorgensen, C.; Larrey, D.; Daujat-Chavanieu, M.; Herrero, A. Mesenchymal Stem Cells Seeded on a Human Amniotic Membrane Improve Liver Regeneration and Mouse Survival after Extended Hepatectomy. J. Tissue Eng. Regen. Med. 2018, 12 (4), 1062–1073 DOI: 10.1002/term.2607. Yuan, J.; Li, W.; Huang, J.; Guo, X.; Li, X.; Lu, X.; Huang, X.; Zhang, H. Transplantation of Human Adipose Stem Cell-Derived Hepatocyte-like Cells with Restricted Localization to Liver Using Acellular Amniotic Membrane. Stem Cell Res. Ther. 2015, 6 (1) DOI: 10.1186/s13287-015-0208-9. Motamed, M.; Sadr, Z.; Valojerdi, M. R.; Moini, A.; Oryan, S.; Totonchi, M.; Ebrahimi, B.; Maroufizadeh, S.; Taghiabadi, E.; Fathi, R. Tissue Engineered Human Amniotic Membrane Application in Mouse Ovarian Follicular Culture. Ann. Biomed. Eng. 2017, 45 (7), 1664–1675 DOI: 10.1007/s10439-017-1836-2. López-Valladares, M. J.; Teresa Rodríguez-Ares, M.; Touriño, R.; Gude, F.; Teresa Silva, M.; Couceiro, J. Donor Age and Gestational Age Influence on Growth Factor Levels in Human Amniotic Membrane. Acta Ophthalmol. (Copenh.) 2010, 88 (6), e211– e216 DOI: 10.1111/j.1755-3768.2010.01908.x. Hopkinson, A.; McIntosh, R. S.; Tighe, P. J.; James, D. K.; Dua, H. S. Amniotic Membrane for Ocular Surface Reconstruction: Donor Variations and the Effect of Handling on TGF-Beta Content. Invest. Ophthalmol. Vis. Sci. 2006, 47 (10), 4316–4322 DOI: 10.1167/iovs.05-1415. Connon, C. J.; Doutch, J.; Chen, B.; Hopkinson, A.; Mehta, J. S.; Nakamura, T.; Kinoshita, S.; Meek, K. M. The Variation in Transparency of Amniotic Membrane Used in Ocular Surface Regeneration. Br. J. Ophthalmol. 2010, 94 (8), 1057–1061 DOI: 10.1136/bjo.2008.153064. Shortt, A. J.; Secker, G. A.; Lomas, R. J.; Wilshaw, S. P.; Kearney, J. N.; Tuft, S. J.; Daniels, J. T. The Effect of Amniotic Membrane Preparation Method on Its Ability to Serve as a Substrate for the Ex-Vivo Expansion of Limbal Epithelial Cells. Biomaterials 2009, 30 (6), 1056–1065 DOI: 10.1016/j.biomaterials.2008.10.048. Niknejad, H.; Deihim, T.; Solati-Hashjin, M.; Peirovi, H. The Effects of Preservation Procedures on Amniotic Membrane’s Ability to Serve as a Substrate for Cultivation of Endothelial Cells. Cryobiology 2011, 63 (3), 145–151 DOI: 10.1016/j.cryobiol.2011.08.003.

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 35

(149) Zhang, T.; Yam, G. H.-F.; Riau, A. K.; Poh, R.; Allen, J. C.; Peh, G. S.; Beuerman, R. W.; Tan, D. T.; Mehta, J. S. The Effect of Amniotic Membrane De-Epithelialization Method on Its Biological Properties and Ability to Promote Limbal Epithelial Cell Culture. Investig. Opthalmology Vis. Sci. 2013, 54 (4), 3072 DOI: 10.1167/iovs.1210805. (150) Wolbank, S.; Hildner, F.; Redl, H.; van Griensven, M.; Gabriel, C.; Hennerbichler, S. Impact of Human Amniotic Membrane Preparation on Release of Angiogenic Factors. J. Tissue Eng. Regen. Med. 2009, 3 (8), 651–654 DOI: 10.1002/term.207. (151) Ravishanker, R.; Bath, A. S.; Roy, R. “Amnion Bank”—the Use of Long Term Glycerol Preserved Amniotic Membranes in the Management of Superficial and Superficial Partial Thickness Burns. Burns 2003, 29 (4), 369–374 DOI: 10.1016/S0305-4179(02)00304-2. (152) Laurent, R.; Nallet, A.; Obert, L.; Nicod, L.; Gindraux, F. Storage and Qualification of Viable Intact Human Amniotic Graft and Technology Transfer to a Tissue Bank. Cell Tissue Bank. 2014, 15 (2), 267–275 DOI: 10.1007/s10561-014-9437-x. (153) Marsit, N.; Dwejen, S.; Saad, I.; Abdalla, S.; Shaab, A.; Salem, S.; Khanfas, E.; Hasan, A.; Mansur, M.; Abdul Sammad, M. Substantiation of 25 kGy Radiation Sterilization Dose for Banked Air Dried Amniotic Membrane and Evaluation of Personnel Skill in Influencing Finished Product Bioburden. Cell Tissue Bank. 2014, 15 (4), 603–611 DOI: 10.1007/s10561-014-9433-1.

ACS Paragon Plus Environment

34

Page 35 of 35 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 OF CONTENTS GRAPHIC

Cellularized hAM graft

Isolation & Processing

Amniotic Fluid Epithelium

Basement membrane

Tissue Engineering applications

Compact layer

Tubular construct

Fibroblast layer Spongy layer

Decellularized hAM

Chorion

Fetal membranes

Cells

Tissue laminate

For Table of Contents Use Only. Manuscript Title: “The Human Amniotic Membrane: A Versatile Scaffold for Tissue Engineering” Authors: Julien H. Arrizabalaga and Matthias U. Nollert

ACS Paragon Plus Environment

35