Abstract
The human amniotic membrane (hAM) is the innermost layer of the placenta. Its distinctive structure and the biological and physical characteristics make it a highly biocompatible material in a variety of regenerative medicine applications. It also acts as a supply of bioactive factors and cells, which indicate the advantages over other tissues. In this review, we firstly discussed the biological properties of hAM-derived cells in vivo or in vitro, along with their stemness of markers, pointing out a promising source of stem cells for regenerative medicine. Then, we systematically summarized current knowledge on the collection, preparation, preservation, and decellularization of hAM, as well as their characteristics helping to improve the understanding of applications in tissue engineering. Finally, we highlighted the recent advances in which hAM has undergone additional modifications to achieve an adequate perspective of regenerative medicine applications. More investigations are required in utilizing appropriate modifications to enhance the therapeutic effectiveness of hAM in the future.
Keywords: Human amniotic membrane, Properties, Modifications, Tissue engineering, Regenerative medicine
Graphical abstract
The schematic diagram highlights the biological importance of hAM or hAM-derived stem cells, along with their additional modifications, contributing to an outstanding perspective in clinical applications.
1. Introduction
The human amniotic membrane (hAM) is the deepest layer of the placenta and is characterized by being a rough, thin (∼0.02–0.5 mm), and semi-transparent membrane without blood vessels, lymphatics, and nerves [1,2]. Like the placenta, umbilical cord, and amniotic fluid, hAM is readily available and regarded as a significant source of stem and progenitor cells. Two main cell types can be found in the hAM, amniotic mesenchymal stromal cells (AMSCs) and amniotic epithelial cells (AECs) [3]. Specifically, AECs is towards the fetus and in direct contact with the amniotic fluid. Meanwhile, the AMSC population is inserted in the extracellular matrix (ECM) of the hAM [4]. The hAM-derived stem cells have multiple characteristics, including immunomodulation, inflammation inhibition, fibrosis and scarring reduction, angiogenesis facilitation, and oxidative stress remission [5,6]. Additionally, hAM can be divided into the following five layers, as Fig. 1A shown; a single epithelial layer toward the fetus, the basement membrane, a compact layer, a fibroblast layer, and a spongy outer layer [7]. These tissues are usually applied as a biological matrix as they fulfill the three main components of the tissue engineering concept: cells, scaffolds, and growth factors.
Fig. 1.
Human amniotic membrane. (A) Schematic illustration of hAM anatomy: an epithelial layer, the basement membrane, a compact layer, a fibroblast layer, and a spongy outer layer. The components of hAM fulfill the requirements of tissue engineering, which contains cells, growth factors, and scaffolds. (B) A timeline of hAM for clinical applications over the past century.
The epithelial layer, enclosed by amniotic fluid, comprises a single columnar or cuboidal AECs generated from ectoderm [8]. The AECs adhere to the basement membrane, separating them from the compact layer connected with the stromal side [9]. The AECs can generate several growth factors, including epidermal growth factor (EGF), basic fibroblast growth factor (bFGF), vascular endothelial growth factor (VEGF), transforming growth factor (TGF-α/β1-3), and platelet-derived growth factor (PDGF) [10]. The intermediate layer is a thick basement membrane that divides the epithelium and stromal layers, which is morphologically similar to human skin due to their comparable composition like collagen I/II/III/IV, laminin, and fibronectin [2]. It is one of the thickest membranes in the human body, offering tensile strength, structural integrity and mechanical characteristics. It indicates that these characteristics pertain to the stromal layer of the membrane [8,11]. Moreover, the basement membrane contains some proteins, including collagen I/II/III/IV, laminin-1/5, and fibronectin [12], besides comprising hyaluronic acid and critical ECM proteins, forming and functioning as an ECM [13].
The stromal layer (the mesenchymal layer) is the outer hAM layer and comprises connective tissues. It is the thickest layer in hAM, including three sub-layers from the inside to the outside: an acellular compact layer composed of reticular fibers, including collagen I/III secreted by AMSCs, a fibroblast layer composed of a loose reticular network containing dispersed fibroblast-like mesenchymal stromal cells and sporadic macrophages, and a spongy layer that is in touch with the chorion membrane and has a complicated network of adequate fibrils wrapped in mucus [14,15]. This stromal layer is transparent due to its lack of vascularization and innervation [9].
The ECM of hAM comprises various collagens, laminin, proteoglycans, glycoproteins, elastin, and fibronectin, besides various significant growth factors and cytokines [16]. It has been used as a natural biomaterial and as a possible cell source for various therapeutic purposes [17,18]. For example, Sabklla N et al. [19] and Stern M et al. [20] advocated its application in skin transplantation in early 1910. In 1940, De Rötth et al. [21] advocated its utilization to replace injured conjunctiva instead of the mouth mucous membrane due to its thin, smooth, and translucent structure that matches the original conjunctiva tissue. Chao et al. [22] successfully rebuilt the dura in an experimental severe head injury using hAM as the matrix. Since then, hAM has been gained momentum as a new biomaterial in many other fields owing to its significant capabilities as a scaffold for blood vessels or other tissue regeneration [23]. Moreover, it can be improved by adding growth factors and cytokines in distinct applications in regenerative medicine, three-dimensional (3D) scaffolds for tissue regeneration, and drug administration [24]. Fig. 1B presents a timeline of the hAM development for research throughout the past century.
Therefore, many beneficial features of hAM, including availability, low cost, and isolation simplicity, make it a suitable biomaterial in tissue engineering researches, particularly in the engineered skin and other soft tissues [25,26]. Moreover, hAM has excellent biological properties, including antibacterial, anti-scarring, immunomodulatory, site-dependent angiogenic functions, and low immunogenicity [27,28]. Thereby, it has been extensively exploited in tissue engineering and regenerative medicine. This review aims to summarize the biological and mechanical properties of hAM and the characteristics of hAM-derived stem cells. In addition, the modifications of hAM for the applications in regenerative medicine were discussed.
2. hAM-derived stem cells
Stem cells are essential in regenerative medicine due to their self-renewal, proliferation, and differentiation abilities. Many investigations have shown that the hAM includes cells having stem cell properties. The primary stem cell types are AECs which rest on a basement membrane, and AMSCs, which reside in the fibroblast layer of the membrane [29,30]. Both cell types have embryological origin characteristics and generate before the three germ layers formation in the pre-gastrulation stages [31]. The AECs are formed from the pluripotent epiblast on the eighth day of conception, while the AMSCs are generated from the extraembryonic mesoderm of the primitive streak [32].
Additionally, hAM-derived stem cells are simple to isolate, where AECs present a cobblestone-like morphology, and AMSCs display a fibroblast-like morphology in vitro [[33], [34], [35]]. Compared with other stem cell sources, hAM-derived stem cells have several advantages such as easy availability, non-tumorigenicity, low immunogenicity, high histocompatibility, effective paracrine function and the minimal ethical concerns. These advantages enable the hAM-derived stem cells to be highly appealed to people, and thus achieve the opportunity to be used as cells in cell therapy and regenerative medicine in the clinic [6,36,37].
When exposed to appropriate culture conditions, hAM-derived stem cells can differentiate into cell lineages of all three primary germ layers [38]. Both AMSCs and AECs display the conventional embryonic stem cell markers, including SSEA-3/4, TRA1-60/81, particularly the pluripotent markers of SOX-2, OCT-4, and NANOG, revealing their potential for regenerative medicine [34,39]. Most hAM-generated cells express embryonic stem cell markers like TRA1-60/81 and SSEA-3/4 in the early second trimester [40]. Similarly, most AECs and AMSCs display certain embryonic stem cells associated pluripotent markers in the early stage, including SSEA-3/4, OCT-4, NANOG, and TRA1-60/80 [41].
Depending on the profiling of different stem cell markers, hAM-derived stem cells represent a heterogeneous cell population [42]. For instance, the transcription factor NANOG, which is considered the most stringent marker for stem cells, is expressed in only 1–3% of AECs, while ∼50% of AECs are SSEA-4-positive at gestation [41]. Only 4% of hAM-derived cells co-express the stem cell markers SSEA-4 and TRA1-60/81 [43]. Stem marker-positive AECs are distributed indiscriminately over the whole hAM rather than residing in clusters and seem surrounded by marker-negative cells [44,45]. The pluripotent marker expressions and proliferative capacity of AECs from distinct hAM regions are very different [46]. This indicates that the AECs encompass the whole epithelial layer at different gestational stages with distinct gene expressions and stemness degrees. Additionally, most primary hAM-derived stem cells may be oriented toward a particular differentiation in vitro, despite the absence of stem cell markers [47].
Besides, the typical mesenchymal stem cells (MSCs) markers CD44/73/90/105 are expressed within AMSCs and AECs, whereas CD34/45/80/86 and HLA-CR are not expressed [31]. MSC markers in AMSCs suggest that the cells have immune privilege and immunomodulatory properties, desirable therapeutic advantages in the medical field [48]. This indicates that the immune protection of AMSCs may contribute to immune system suppression or modulation. Table 1 lists the stem cell markers of AECs and AMSCs.
Table 1.
Stem cell markers of AECs and AMSCs generated from the hAM.
Cell type | MSCs markers | Cell surface markers | Transcription factors | Reference |
---|---|---|---|---|
AECs | CD29/73/105 | SSEA-4 | OCT-4 and NANOG | [35] |
CD9/24 | SSEA-3/4 and TRA1-60/81 | OCT-4 and NANOG | [42] | |
CD29/90/166 | TRA1-60 | OCT-4, NANOG, and SOX-2 | [56] | |
CD29/44/73/90/105 | SSEA-4 | OCT-4 and SOX-2 | [57] | |
CD29/44/90 | SSEA-4 | OCT-4 and SOX-2 | [58] | |
CD29/73 | SSEA-4 | OCT-3/4 and SOX-2 | [59] | |
AMSCs | CD29/73/90/105 | SSEA-4 | OCT-4 and NANOG | [34] |
CD29/44/73/90/105 | SSEA-4 | OCT-4 and SOX2 | [57] | |
CD44/29/73/90/105/271 | SSEA-4 and TRA1-60 | OCT-3/4, SOX-2, and NANOG | [59] | |
CD29/44/73/90/105 | SSEA-4 | OCT-4 | [60] | |
CD29/44/73/90/105166 | SSEA-4 | STRO-1 | [61] | |
CD29/105 | SSEA-4 and REX-1 | OCT-3/4, SOX-2, and NANOG | [62] | |
CD44/73/90/105 | SSEA-3/4 | OCT-3/4, SOX-2, and NANOG | [63] |
The lack of immunogenicity is widely recognized as a prominent characteristic of hAM-derived cells, resulting in their state of immunological tolerance [49]. Regarding innate immune suppression, AECs and their conditioned medium (CM) substantially reduce T-cell proliferation with or without direct contact [50]. Moreover, AECs and AMSCs have indicated their ability to inhibit T-cell proliferation in a dose-dependent manner, which was observed after T-cell exposure to alloantigen in the presence of hAM-derived stem cells, either after CD3/28 stimulation or in typical mixed lymphocyte response models [51,52]. The tolerogenic HLA-G expression is a crucial factor in the immunomodulatory abilities of hAM-derived stem cells. HLA-G interacts with the appropriate immune cell receptors (ILT2/4 and KIR2DL4) to exert immunomodulatory effects [53]. Furthermore, the immunological checkpoint proteins like programmed death ligands 1 and 2 (PD-L1/2), are associated with immune tolerance induction by hAM-generated cells [54]. These molecules are present on AMSCs and syncytiotrophoblasts in the placenta. However, they can be synthesized by AECs without being exposed to IFN-γ [55].
2.1. hAM-derived AECs
AECs' migration to damaged tissues to replace the injured cell is essential to ameliorate acute or chronic wounds. Nevertheless, there are several challenges to stem cell transplantation, including low survival and poor repair capacity. Recently, AECs may offer a favorable microenvironment for cell survival and trigger endogenous tissue regeneration by secreting bioactive cytokines, growth factors, and exosomes [64]. AECs could synthesize and release brain-derived neurotrophic factor (BDNF), ciliary neurotrophic factor (CNTF), and forskolin. These factors are involved in embryonic neural development at early stages and have a neuroprotective function [65].
Additionally, literature [66] revealed that AECs increased the lifespan of dopamine (DA) neurons by generating biologically active neurotrophins, including BDNF and neurotrophin-3 (NT-3), preventing DA neurons loss in rats with Parkinson's disease. Regarding corneal injury, AECs-CM injection enhanced wound healing by lowering inflammatory cell infiltration and neovascularization [67]. The AECs-CM injection also improved the ovarian microenvironment, defended the ovaries against the harm caused by chemotherapy, and promoted ovarian angiogenesis [68]. Furthermore, AECs effectively reduced chemotherapy-induced apoptosis and activated the TGF-β signaling pathway in primary granulosa-lutein cells in a paracrine manner [69]. The AECs-CM injection also promoted angiogenesis in mice with premature ovarian failure/insufficiency in the injured ovaries.
On the other hand, AECs reduced collagen synthesis in human hepatic stellate cells with paracrine effect, and some soluble factors like MMP-2/9 were secreted by AECs, enhancing ECM remodeling, reducing liver fibrosis, and altering macrophage polarization, particularly in liver fibrosis models [70]. Administrating AECs and AECs-CM decreased hepatic inflammation and fibrosis significantly in a diet-induced non-alcoholic steatohepatitis model, indicating the anti-fibrotic effects of AECs [71]. Under hypoxic conditions, AECs secrete higher human-origin proangiogenic cytokine levels, including angiogenin (ANG), EGF, interleukin (IL)-6, and monocyte chemoattractant protein (MCP)-1, contributing to myocardial tissue regeneration in a myocardial infarction rat model [72].
Extracellular vesicles, specifically exosomes, have received much attention as they are essential components of AECs paracrine effects. Exosomes are nano-sized microvesicles containing multiple bioactive molecules, including proteins, nucleic acids, lipids, and organelles. These vesicles are involved in intercellular communication and regulate many intracellular biological activities [73]. Internalization of AECs-derived exosomes enhanced fibroblast migration and proliferation, partly abolished ECM deposition, and improved cutaneous wound healing within in vivo experiments [74]. In idiopathic pulmonary fibrosis, AECs-derived exosomes polarized and enhanced macrophage phagocytosis, reduced neutrophil myeloperoxidase activity, and directly suppressed T-cell proliferation, eventually improving lung fibrosis [75]. Moreover, AECs-derived exosomes increased the number of follicles and enhanced ovarian function in a chemotherapy-induced premature ovarian failure mice model. Another in vitro study showed that AECs-derived exosomes prevented chemotherapy-induced apoptosis in granulosa cells by transmitting miR-1246 [76]. In brief, there has been a systematic review of the biological functions of AECs secreted exosomes or extracellular vesicles in regenerative medicine [77].
2.2. hAM-derived AMSCs
Currently, AMSCs originate from the hAM and are regarded as highly intriguing stem cells in regenerative medicine. AMSCs contribute to treating skin wounds due to their multilineage differentiation, immunosuppressive, and anti-inflammatory capabilities [78]. AMSCs and AMSCs-CM prevented heat stress-induced apoptosis in human keratinocytes and dermal fibroblasts and enhanced their proliferation with the paracrine effect. Moreover, AMSCs could secrete many bioactive cytokines, including PAI-1, C-GSF, TIMP-1, and uPAR, which may activate the PI3K/AKT signaling pathway [34]. Additionally, AMSCs-CM significantly accelerated wound closure by secreting IGF-1, EGF, and IL-8 in a diabetic mice model [79].
The AMSCs exhibit unique characteristics that render them valuable in tissue regeneration applications. For example, AMSCs combined with PPCNg (thermoresponsive biomaterial composed of Poly (polyethylene glycol citrate-co-N-isopropyl acrylamide) (PPCN) mixed with gelatin) intrauterine transplantation could promote damaged endometrium regeneration, restoring reproductive function in an intrauterine adhesions rat model [80]. PPCNg has been used as a scaffold for stem cell transplantation. Additionally, AMSCs injection significantly restored ovarian function in an age-related diminished ovarian reserve (AR-DOR) mice model while dramatically suppressing granulosa and stromal cell apoptosis in the ovaries [81]. In a carbon tetrachloride-induced liver fibrosis mice model, IGFBP-3 and DKK1/3 generated from AMSCs prevented the activation of hepatic stellate cells by suppressing the Wnt/β-catenin signaling pathway, decreasing liver fibrosis in mice [82]. These findings suggested that AMSCs or AMSC-CM could provide an alternative therapeutic strategy for treating hepatic fibrosis. Moreover, AMSCs transplantation might reverse Bmi-1 deficiency-induced mandibular osteoporosis by promoting osteoblastic bone formation and preventing bone resorption [83].
The proangiogenic potential of AMSCs has been investigated in many publications due to exhibiting high levels of the proangiogenic genes VEGF-A, angiopoietin-1, HGF, FGF-2, and the antiapoptotic protein AKT-1. Administrating AMSCs increased blood flow and capillary density in hindlimb ischemia of mice, demonstrating that AMSCs significantly enhanced neovascularization [84]. Similarly, AMSCs-CM significantly recovered the blood flow in a murine hindlimb ischemia model [85]. Intramyocardial injection of AMSCs might restore cardiac function and suppress inflammation at the damage site by regulating the inflammatory MAPK/NF-κB pathway in a myocardial injury rat model [86]. Furthermore, superparamagnetic iron oxide nanoparticles-labeled AMSCs dramatically enhanced cardiac function and decreased fibrosis and tissue injury in a magnetic field by inhibiting inflammation in an NF-κB/MAPK-dependent manner [87].
Moreover, in vivo and in vitro results revealed that the derivatives and protein extracts of the AMSCs present potential anti-cancer effects. For instance, DKK-1/3 and IGFBP-3 generated from AMSCs suppressed hepatoma cell proliferation and induced apoptosis by inhibiting specific signaling pathways [33]. The anti-tumor effects of AMSCs on prostate cancer cells were also identified by activating apoptosis, inhibiting the epithelial-mesenchymal transition process, and down-regulating EGFR using 2D and 3D cell culture methods [88]. The immunomodulatory characteristics of AMSCs secreted exosomes or extracellular vesicles enable them to promote tissue regeneration and produce therapeutic benefits in preclinical disease models. AMSCs performed immunoregulatory functions via the paracrine effect activated by factors in their extracellular vesicles [89]. This suggests that extracellular vesicles generated from AMSCs inhibited peripheral blood mononuclear cell proliferation and T-cell polarization toward inflammatory Th subtypes, besides stimulating regulatory T-cell activation. The biological characteristics of AECs and AMSCs are summarized in Table 2, and the applications in tissue engineering are diagrammatically displayed in Fig. 2.
Table 2.
hAM-derived stem cells display biological functions in various disease treatments.
Stem cells | Secreted factors/cytokines | Biological functions | Experimental cells or animals | Involved diseases | Reference |
---|---|---|---|---|---|
AECs | BDNF, CNTF and forskolin | Neuroprotective effect | Rat retinal ganglion cells | N/A | [65] |
AECs | BDNF and NT-3 | Enhance DA neurons survival | Rats | Parkinson's disease | [66] |
AECs-CM | N/A | Anti-inflammatory effect | Rabbits | Corneal injury | [67] |
AECs-CM | VEGF | Promote angiogenesis | Mice | Chemotherapy-induced primary ovarian insufficiency | [68] |
AECs and AECs-CM | TGF-β | Anti-apoptosis and promote angiogenesis | Primary granulosa-lutein cells, mice | Premature ovarian failure/insufficiency | [69] |
AECs and AECs-CM | MMP-2/9 | Enhance ECM remodeling, anti-fibrosis, and alter macrophage polarization | Human hepatic stellate cells | Liver fibrosis | [70] |
AECs and AECs-CM | MMP-2/9 | Anti-inflammation and anti-fibrosis | Mice | Non-alcoholic steatohepatitis | [71] |
AECs | ANG, EGF, IL-6, and MCP-1 | Promote angiogenesis | Rats | Myocardial infarction | [72] |
AECs-derived exosomes | MMP-1 | Enhance fibroblast migration and proliferation, and downregulate collagen expression | Fibroblasts and rats | Skin injury | [74] |
AECs-derived exosomes | miRNA | Enhance macrophage phagocytosis, reduce neutrophil myeloperoxidases activity, and suppress T cell proliferation | Mice | Idiopathic pulmonary fibrosis | [75] |
AECs-derived exosomes | miR-1246 | Anti-apoptosis | Granulosa cells and Mice | Chemotherapy-induced premature ovarian failure | [76] |
AMSCs and AMSCs-CM | PAI-1, C-GSF, TIMP-1, and uPAR | Anti-apoptosis and pro-proliferation | Keratinocytes, dermal fibroblasts, and Mice | Skin wound | [34] |
AMSCs | IGF-1, EGF, and IL-8 | Promote angiogenesis | Mice | Diabetic skin wound | [79] |
AMSCs | VEGF and Ki-67 | Promote angiogenesis and pro-proliferation | Rats | Intrauterine adhesions | [80] |
AMSCs | N/A | Anti-apoptosis | Granulosa cells and Mice | Age-related diminished ovarian reserve | [81] |
AMSCs and AMSCs-CM | IGFBP-3 and DKK-1/3 | Inhibit the collagen deposition and anti-fibrosis | Hepatic stellate cells and Mice | Carbon tetrachloride-induced liver fibrosis | [82] |
AMSCs | N/A | Stimulate the osteoblastic bone formation | Mice | Mandibular osteoporosis | [83] |
AMSCs | VEGF-A, angiopoietin-1, and FGF-2 | Promote angiogenesis | Mice | Hindlimb ischemia | [84] |
AMSCs-CM | N/A | Promote angiogenesis | Mice | Hindlimb ischemia | [85] |
AMSCs | N/A | Anti-inflammation | Rats | Myocardial injury | [86] |
AMSCs | N/A | Anti-fibrosis | Rats | Isoproterenol-induced myocardial injury | [87] |
AMSCs | DKK-1/3 and IGFBP-3 | Anti-tumor effect | Hepatoma cells | Liver cancer | [33] |
AMSCs | N/A | Anti-tumor effect | Prostate cancer cells | Prostate cancer | [88] |
AMSCs-derived extracellular vesicles | N/A | Immunomodulatory effect | Immune cells | N/A | [89] |
Fig. 2.
The biological effects of hAM-derived AECs and AMSCs in various disease models. AECs and AMSCs display similar therapeutic efficiency in animal models of reproductive system disease, liver disease, myocardial infarction, corneal injury, hindlimb ischemia, and skin injury.
3. Processing and properties of hAM-derived materials
The interdisciplinary fields of tissue engineering include process engineering, material science, biology, chemistry, physics, and medicine. It focuses on fabricating tissue analogs to support cellular responses in successfully regenerating lost or injured tissues and organs [28]. Tissue engineering consists of cells, scaffolds, and growth factors or biomolecules that stimulate tissue regeneration. The primary concept underlying tissue engineering is creating a new effective scaffold or scaffold-based tissue structure in tissue-engineered constructions or products. Tissue-engineered constructions are created by colonizing a scaffold with cells that can proliferate, differentiate, and duplicate the functions of cells or/and tissues [90]. The scaffold is a cell adhesion and infiltration platform to produce functional tissue. Consequently, the cells need a biocompatible scaffold made of natural, synthetic, or a combination of both materials that resemble the ECM [91]. The optimal scaffolds should be biodegradable, meaning they should degrade non-toxic when implanted into the impaired tissue microenvironment and be replaced by native components after tissue repair is finished [92].
The hAM is a biomaterial with multiple appealing properties. There are no ethical issues since it is considered biological waste after delivery. The availability, affordability, and simplicity of isolation make hAM a suitable biomaterial for tissue engineering [93]. Additionally, the hAM can be extensively employed in tissue engineering due to its favorable biological characteristics. The properties of hAM with different forms are summarized in Table 3.
Table 3.
Properties of hAM with different forms.
Characteristics | Specific features | Forms | Mechanism | Study mode | Reference |
---|---|---|---|---|---|
Mechanical properties | Elasticity | Intact hAM | Young's modulus (Full term hAM: 2.29 MPa; preterm hAM: 3.6 MPa) | In vitro | [13] |
Stiffness | Intact hAM | Elastic modulus: 4.048 ± 1.702 MPa | In vitro | [110] | |
Tensile strength | Dehydrated hAM | 5.475 ± 0.135 MPa | In vitro | [111] | |
Stiffness | Decellularized hAM | Elastic modulus higher than native hAM | In vitro | [28] | |
Biological properties | Antimicrobial effect | Intact hAM | Expression of elafin, HBD-2/3, and cathelicidin LL-37 | In vivo | [112] |
Antimicrobial effect | Cryopreserved hAM | Expression of HBD-2/3 | In vivo | [113] | |
Antimicrobial effect | hAM extract | Secretion of antimicrobial proteins and peptides | In vitro and in vitro | [114] | |
Antimicrobial effect | hAM homogenate | Expression of β-defenses | In vitro | [115] | |
Antiviral effect | Intact hAM | Expression of IL-1RA | Clinical cases | [117] | |
Antiviral effect | Intact hAM | Inhibition of inflammatory response | Clinical cases | [118] | |
Anti-fibrotic effect | hAM extract | Reversion to the fibroblast phenotype | In vitro | [122] | |
Anti-scarring effect | Intact hAM | Antagonizing the effect of TGF-β | In vitro | [123] | |
Anti-inflammatory effect | Decellularized hAM | Suppression of IL-1α/1β | In vitro | [125] | |
Anti-inflammatory effect | Intact hAM | Trapping inflammatory cells | Clinical cases | [126] | |
Low immunogenicity | Intact hAM | Expression of HLA-G and Fas ligand | In vivo | [127] | |
Immunomodulatory effect | Intact hAM | Proliferation and induction of immune cells | In vitro | [128] | |
Anti-angiogenesis effect | Flesh hAM | Secretion of collagen IV, laminin, and integrins 4/6 | In vivo | [130] | |
Anti-angiogenesis effect | Intact hAM | Secretion of endostatin, thrombospondin-1, and TIMP-1/2/3/4 | Clinical trial | [131,132] | |
Angiogenesis effect | Intact hAM | Secretion of fibronectin | In vitro | [133] | |
Angiogenesis effect | Decellularized hAM | Enhancing PECAM-1 and VE-cadherin expression | In vitro | [134] | |
Support cell growth and adhesion | Decellularized hAM | Expression fibronectin, laminin, collagens, and proteoglycans | In vitro | [135] |
3.1. Collection, preparation, preservation, and decellularization
Generally, hAM is acquired from the human placenta and taken by medical faculty following a cesarean with informed consent from expectant mothers. hAM could also be obtained from a natural delivery. The source of hAM collection, whether after vaginal delivery or via cesarean section, influence its physiology, integrity, availability, and growth factor content and has a contamination risk. The hAM donors should be screened to minimize infectious disease transmission, particularly after the COVID-19 pandemic owing to the concerns about vertical transmission [94]. After collection, the placenta is packaged in a sterile container and transferred to the laboratory for processing. The transport duration should be instant, within 24 h at 4 °C or 2 h at room temperature [28]. It should be immersed in sterile saline solution or phosphate-buffered saline to prevent the hAM from dehydration [95].
The preservation of hAM has been developed over the last century benefiting from technical advances, preparation methods, and sterilizing procedures. In 1940, Chao et al. [22] advocated drying hAM before usage to reduce discomfort caused by fresh hAM. Dry hAM marked a turning point when used in tissue reconstruction since it proved safe and addressed the shortcoming of fresh hAM. Later, Kim et al. [96] suggested cryopreservation of the hAM by storing it at −80 °C using a storage medium composed of glycerol and Dulbecco's Modified Eagle Medium (DMEM). However, cryopreservation altered the composition and distribution of ECM, leading to poor cell viability in the hAM. Moreover, cryopreservation reduces the number of growth factors or cytokines produced by hAM, endangering its structural and mechanical features [2]. In 2004, Nakamura et al. [97] presented a novel preservation method, freeze-dried hAM as a substrate for ocular surface reconstruction, to confirm the feasibility. Both cryopreservation and lyophilization have drawbacks since they could influence hAM characteristics. Current efforts to preserve the hAM in suspension have shown the promise in reducing the invasiveness, comparing with the grafting process of suturing the hAM, particularly for corneal ulcers [24]. This suspension has been commercialized as a cryopreserved amniotic membrane extract (AME) or as AME-eye drops. The cryopreserved AME has been shown to alleviate the clinical manifestations of dry eye disease [98].
Collectively, the hAM is currently utilized in various forms, including fresh, air-dried, freeze-dried, cryopreserved, and suspension. Due to the rejection response of hAM allografts after transplantation, intact hAM immunogenicity remains a serious problem [15]. The decellularization of hAM before transplantation is considered a highly effective method.
Decellularization involves removing the cells and cell debris from the hAM and leaving the extracellular structural proteins unchanged to avoid immunogenic reactions and reduce the risk of infection [99]. The acellular hAM has been shown to retain ECM components, including collagen I/III/IV/V/VII, laminin, fibronectin, elastin, hyaluronic acid, and several bioactive factors. The decellularized hAM produces multiple growth factors such as NGF, EGF, HGF, KGF, and bFGF accumulating in the stromal layer of the membrane [100]. Each of these growth factors can significantly accelerate tissue regeneration. Besides providing a supporting scaffold with minimal immunogenicity, the decellularized hAM stimulates cell growth, adhesion, and proliferation [101]. Consequently, an acellularized hAM may represent a more suitable and biocompatible alternative for regenerative medicine applications than intact hAM.
Successful decellularization of the hAM while minimizing any adverse impact on the ECM components is vital to prevent immunogenic response after transplantation. The effectiveness of the decellularized procedure is assessed using optical microscopy, immunohistochemistry analyses, fluorescent staining, or DNA quantitative analysis [28]. Generally, most manufacturing methods of decellularized hAM involve two steps under sterile conditions. Briefly, hAM is treated with an enzymatic or chemical reagent, followed by mechanical scraping under light microscopy to remove loosened cells. There are different reagents for hAM decellularization, including sodium dodecyl sulfate, urea, methanol, chloroform, EDTA, and trypsin [102]. However, mechanical scraping may severely damage the integrity of the basement membrane and stroma layer. Therefore, recent research has proposed other strategies that obviate further mechanical scraping [9]. Additionally, decellularization removes the stem cell components from the hAM. Residual non-nuclear cellular components may be found in the ECM; however, their presence is not expected to prevent ECM therapeutic usage.
3.2. Mechanical properties
The mechanical properties of hAM, including elasticity, stiffness, and tensile strength, are related to its ECM components. Elastic deformation is determined by the presence of elastin fibers, laminin, hyaluronic acid, and glycosaminoglycan in the ECM. Meanwhile, the collagen fibrils composition determines tensile strength [103]. The hAM displays viscoelastic characteristics and time-dependent mechanical properties depending on its term stage. For instance, as compared to full-term hAM (36–40 weeks), the preterm (26–36 weeks) hAM was shown to have better mechanical integrity [104]. A study further confirmed that Young's modulus of the full-term and preterm hAM were 2.29 and 3.36 Mpa, respectively [13]. Young's modulus is characterized by the elasticity of hAM, defined as a biomaterial's capacity to endure a force that causes distortion and regains its initial form and size when the force is withdrawn.
In addition, it has been proven that the stiffness decreases with the increase of hAM thickness. According to its anatomical structure, hAM thickness varies from 0.02 to 0.5 mm and can be divided into different subregions, known as the placental, reflected and umbilical amnion which differ distinctively in morphology [18,105,106]. A specific area within the reflected amnion is found at the lower uterine pole and cervix, known as the cervical amnion [107,108]. The thickest subregion is adjacent to the placental disk (placental amnion), while the distal subregion (cervical amnion) is thinner and more transparent [109]. Consistently, the placental amnion has a lower elastic modulus (stiffness of biomaterial) than the cervical amnion, which could be attributed to collagen fiber arrangement [110]. Besides differences in stiffness between hAM subregions, differences in tensile strength have also been studied. The placental amnion was significantly more substantial and stretchable than the reflected amnion by an average of 82% ± 45% and 19% ± 6%, respectively [111]. The differences in the elastic modulus of hAM are caused by the various hAM forms utilized. Decellularization of hAM displays a higher elastic modulus than the native one, a possible explanation may be attributed to the denudation process dehydrating the membrane and then decreasing its thickness [28].
3.3. Biological properties
As mentioned above, the hAM has shown promising biological properties that enhance various applications in tissue engineering and regenerative medicine. Regarding the antibacterial capabilities of hAM, a study demonstrated that hAM secreted more antimicrobial peptides, including elafin, HBD-2/3, and cathelicidin LL-37, when exposed to IL-1β [112]. This confirms the antibacterial activity, which inflammatory signal inducers can augment. Recently, it has shown that a cryopreserved hAM may efficiently promote wound closure and prevent wound-related infections [113]. Another study declared that the hAM extract suppressed S. pneumoniae proliferation in the planktonic and biofilm states by secreting antimicrobial proteins and peptides [114]. In the same direction, Taja et al. [115] showed that the hAM homogenate exhibited an antimicrobial effect against 7 of 11 tested multidrug-resistant pathogens, with methicillin-resistant S. aureus having the most significant impact.
There are currently a few studies on the antimicrobial and antiviral effects of hAM. Notably, hAM can exert antiviral properties by restricting either virus colonization and reproduction or severe consequences caused by an abnormal host response to infection, such as sepsis [116]. A study including 18 patients with herpetic keratitis-related corneal ulcers or non-healing epithelial abnormalities was treated with hAM transplantation. The findings indicated that in the individuals with herpes simplex virus type 1 (HSV-1) keratitis, hAM transplantation enhanced rapid epithelial healing and inhibited stromal inflammation and ulceration. The advantages of hAM in those individuals may be attributed to hAM secreting IL-1RA, which is IL-1α′s natural antagonist [117]. In another study, transplantation of multilayer hAM was employed to treat 15 patients' eyes with herpes necrotizing stromal keratitis that had not healed using topical and systemic aciclovir therapy. After 14 days post-transplantation, all patients presented with an intact epithelial corneal surface and a clinically significant decrease in inflammation [118].
Fibrosis is a pathological characteristic of most chronic inflammatory diseases. Fibrotic tissue is characterized by an abnormal deposition of ECM components such as collagen and fibronectin, which disrupts normal tissue architecture and function [119]. Damage to epithelial or endothelial cells triggered by diverse stressors initially activates the coagulation pathway, followed by acute inflammation and activation of innate immune mediators including resident macrophages, neutrophils, and dendritic cells. Inflammatory and immune mediators cooperate to diminish the cytokines or chemokines and then the inactive fibroblasts were transferred into myofibroblasts [120]. Myofibroblasts may generate additional ECM quantities and exert tractional stresses across the ECM, distorting tissue architecture and eventually leading to fibrosis [120]. hAM could inhibit the abnormal development of fibrotic tissue at various levels, resulting in scarring reduction or absence and maintenance of tissue architecture and functions [121]. A study revealed that hAM extract can transfer differentiated myofibroblasts back into fibroblasts in addition to maintaining the initial fibroblastic phenotype in vitro [122]. Moreover, hAM could selectively counteract the effect of TGF-β and promote cell proliferation in keratinocytes by modulating the genetic program [123]. TGF-β can overexpress ECM elements, including metalloprotease inhibitors, collagen, and proteoglycans, which leads to fibrosis in the tissue repair process.
The hAM can induce immunomodulatory properties by inhibiting inflammatory cytokines, decreasing oxidative burst, restricting chemotaxis, and improving phagocytic function [124]. For example, a study found that human limbal epithelial cells cultivated on the ECM of hAM significantly exhibited lower IL-1α/1β mRNA and protein expressions in contrast to plastic cultures, regardless of whether lipopolysaccharide was added or not [125]. In a clinical study, the hAM patches of the ocular surface were applied to 20 eyes of 20 patients with persistent corneal epithelial abnormalities. The results showed that hAM patches attracted and trapped inflammatory cells infiltrating the ocular surface [126]. Therefore, the anti-inflammatory effects of the hAM are speculated to be cytokine-mediated as well as mechanical. In addition, the hAM seems to be immune-privileged tissue containing immunoregulatory factors such as HLA-G and Fas ligand [127]. This low immunogenicity of hAM was also related to low expression of HLA IA, together with negative expressions of immunogenic markers CD40/80/86 and HLA-DR [37]. Consistently, a study conducted in vitro demonstrated that CM containing immunoregulatory factors from the intact hAM has immunomodulatory properties in terms of human peripheral blood mononuclear cells and T-cell proliferation, T-cell subset polarization, and T-regulatory cell induction [128].
There are challenging studies about the pro- or anti-angiogenic effects of the hAM. However, a few studies indicated that hAM has a dual-effects on angiogenesis, which is side-dependent [121,129]. In a dorsal skinfold chamber rats model, the number of formed vessels and their lengths increased when hAM was positioned epithelial side, whereas angiogenesis reduced when it was placed stromal side [129]. On the one hand, the hAM contains collagen IV, laminin, and integrins 4/6 that inhibit neovascularization after transplantation, which has been related to angiogenesis suppression in severely damaged rabbit corneas [130]. The hAM also generates other valuable anti-angiogenic proteins, including endostatin, thrombospondin-1, and four tissue inhibitors of metalloproteases (TIMP-1/2/3/4) [131]. When hAM was utilized in post-pterygium operations, it inhibited the blood formation where the fibrous pterygium lesions were located [132]. On the other hand, the fibronectin proteins in the ECM of hAM interact with the growth factors PDGF, EGF, and b-FGF to activate the ERK pathway and then exert its angiogenesis effect in vitro [133]. Another study proposed that the hAM might be regarded as an excellent matrix for establishing mature vascular constructions [134]. This is owing to its potential for increasing the expression of platelet-endothelial cell adhesion molecule-1 (PECAM-1) and adhesion molecule VE-cadherin in porcine vascular endothelial cells. In addition, the ECM of hAM that contains hyaluronic acid and proteins, including fibronectin, laminin, collagens, and proteoglycans. This ECM acts as a support cultivation system for cell growth and adhesion [135]. These biological properties of hAM with different forms are diagrammatically displayed in Fig. 3.
Fig. 3.
Schematic representation of distinct features of hAM. The features include antimicrobial and antiviral effects, anti-fibrotic or anti-scarring activity, immunomodulatory function with low immunogenicity, and pro- or anti-angiogenesis effect.
4. Applications of hAM and its composites in tissue engineering
hAM has been utilized extensively in tissue regeneration and has shown positive outcomes as a reconstruction material. Since the beginning of the 20th century, hAM has shown advantages as a therapeutic agent in clinical applications, primarily in the wound healing of soft tissues like skin and cornea. To date, the hAM has been broadly applied in clinical procedures, including gynecology [136], ophthalmology [137], neurosurgery [138], plastic surgery [139], gastrointestinal surgery [140], and orthopedics [141], as well as wound dressing for burns, or the acute and chronic wounds [121,142]. The clinical applications of hAM also have been reviewed in many publications [[143], [144], [145], [146]]. These articles have investigated unmodified hAM applications. Collectively, hAM, regardless of its state (intact, dried, decellularized, or frozen), has distinct qualities that render it highly suitable for various applications.
However, the fast biodegradability of hAM is considered a severe limitation in its usage for regenerative medicine [15]. Additionally, the biological or mechanical properties of hAM are sometimes not efficient enough for some applications. Therefore, to enhance these characteristics, modifications on hAM with various methods are needed for tissue regeneration. The following section overviews the different modifications based on hAM, both in the intact and decellularized form, as well as their applications in tissue engineering and regenerative medicine.
4.1. Eyes
The usage of hAM in treating ophthalmologic diseases has become broadly acknowledged. However, its intrinsic limitations, including weak mechanical characteristics, short-term therapeutic efficiency, difficult suturing during operations, and ineffective adhesion, underline the need for hAM modifications in ocular surface reconstruction [147]. Polyvinyl alcohol (PVA) is a transparent hydrogel that is biocompatible and physically strong, rendering it a viable material for an artificial cornea. Uchino et al. [148] fabricated a hAM-immobilized PVA hydrogel (PVA-AM) to substitute cornea. The results showed that the corneal epithelialization on PVA-AM was improved in rabbit corneas, indicating that PVA-AM could be used as a biocompatible hybrid scaffold for keratoprosthesis. Another study investigated the biodegradability of fibrin glue-coated and freeze-dried hAM (FG-AM) implanted into a rabbit scleral surface without suturing [149]. This FG-AM remained for at least 12 weeks after transplantation. It was verified to be safe, convenient, and effective for ocular surface reconstruction. Cai et al. [147] confirmed that a composite structure of FG-AM transplantation is effective and valuable for ocular reconstruction.
Additionally, Zhou et al. [150] fabricated another composite membrane by electrospinning a layer of poly(-caprolactone) (PCL) nanofiber mesh to strengthen the decellularized hAM sheet by covalent interfacial bonding while preserving the special bioactivity of the decellularized hAM. The authors demonstrated the superior ability of this PCL-AM composite for repairing the damaged cornea in rabbits with limbal stem cell (LSC) deficiency caused by alkaline burn. Notably, biological or chemical components could be coated on the membrane by applying diverse procedures including peptide self-assembly and deposition. These modifications have mainly enhanced the antibacterial property of hAM. For instance, Singh et al. [151] applied a coating of clavanin-A to self-assembled acellular hAM to inhibit biofilm formation on the membrane. The results revealed that the clavanin A-coated hAM was more biocompatible than the decellularized AM alone and exhibited lower levels of fungal and cell colonization. Furthermore, this composite based on hAM had outstanding physical, morphological, and antifungal properties, rendering it appropriate for the controlling ocular surface infections. Our group administered severe corneal injury by AME-derived composites in the form of in situ gels or tablets. The results showed that AME could promote wound healing, with the in situ gels having the most significant therapeutic efficiency [152].
4.2. Skin
Due to the intrinsic bioactive and biocompatible characteristics of hAM, it is believed to have significant therapeutic potential for skin wound healing. However, it is anticipated that hAM reinforcement with a biodegradable polymer would give acceptable mechanical characteristics because it is too soft and thin to handle. Literature [153] reported a fibrin-coated silver nanoparticle-infused poly-[lactide-co-glycolide-co-caprolactone] terpolymers (PLGC) to strengthen the hAM membrane, together with bioactive chemicals or factors to repair the wound. This hybrid scaffold demonstrated favorable qualities and significant biocompatibility for possible usage in skin wound healing. The ability of the hAM composites as a wound dressing could shield the wound from outside harm and provide an ideal microenvironment for tissue regeneration. Recently, Yang et al. [154] produced a double-layer membrane comprised of decellularized hAM and chitosan sponge as a wound dressing. The results revealed that the porous structure of the composites accelerated blood coagulation and swelling functions, while the bilayer structure exhibited acceptable biocompatibility, particularly for the attachment and proliferation of human foreskin fibroblast cells (HFF-1). After eight-day treatments, the full-thickness skin lesions in the diabetic mice model exhibited more than 80% closure.
Comparable research was conducted to determine whether hAM and collagen-based hydrogels worked to repair cutaneous burn wounds in rats with wound dressing membranes. The results found that when a wound dressing membrane was applied, a hydrogel consisting of hAM and collagen markedly displayed rapid wound healing, driven by full re-epithelialization and closure by wound contraction [155]. Murphy et al. [156] also studied the composites of hAM-based hydrogel, proving the effectiveness of the solubilized hAM combined with the carrier hyaluronic acid hydrogel (HA-SAM) as a wound therapy in a full-thickness wound mice model. The findings showed that HA-SAM dramatically expedited wound closure by re-epithelialization and inhibited wound contraction.
Hypertrophic scarring is a skin disease caused by the deposition of excessive collagen and other ECM proteins after deep trauma, severe burn, and surgical wounds [157,158]. A 3D bilayered decellularized hAM/electrospun silk fibroin membrane was produced and investigated for hypertrophic scarring applications [159]. The results showed a notable reduction in collagen deposition and increased MMP1 expression and deposition in the wound bed in a rabbit ear model, indicating that the manufactured construct can be an effective anti-scarring wound recovery.
4.3. Abdominal wall
Laparotomy is a typical surgical technique that may result in complications, such as abdominal infection, adhesions, and hernias [160]. There are 4 million abdominal surgeries are performed annually, and 10–20% of the patients are predicted to develop abdominal hernias at the incision site, which could be regarded as a failure in abdominal repair [161]. Currently, polypropylene (PP) mesh is a synthetic material most frequently utilized to treat incisional hernias and repair abdominal wall defects. However, recurrences and adhesions are frequent complications of abdominal wall reconstruction with a PP mesh [162]. In some cases, hAM is utilized as a coating to improve the therapeutic efficacy of PP mesh. For example, in an abdominal wall defect rat model, PP mesh was covered by fresh hAM to reduce postoperative abdominal adhesions and inflammation while increasing epithelialization after abdominal hernias repair surgery [163]. In another laparotomy rabbit model, the effectiveness of PP mesh coated with hAM could potentially minimize adhesions in the abdominal viscera after laparotomy was performed, compared to PP mesh alone [164]. In addition, Rashid et al. [165] performed a similar study in the abdominal hernias rat model, revealing that the combined bovine amniotic membrane (bAM) and 5% polyethylene glycol (PEG)-4000 effectively prevented the complications generated by PP mesh.
4.4. Cardiovascular system
Globally, ischemic heart disease is the leading cause of patient mortality. Myocardial infarction (MI)-related heart failure is linked to severe cardiac remodeling and fibrosis production [166]. A tissue engineering strategy of hAM-based hydrogels has been developed to treat MI. For example, Henry et al. [167] fabricated a thermoresponsive, injectable hAM hydrogel to accelerate myocardial regeneration. They infused hAM-based hydrogel into rat MI hearts via ultrasound-guided injection and examined its effectiveness compared to phosphate-buffered saline. The findings revealed that this hybrid hydrogel markedly increased heart contractility and reduced fibrosis. Another research used an acellular hAM hydrogel modified with polyacrylamide-alginate (AlgSr/PAM-AM) as a vascular biomaterial [168]. The results demonstrated that this vascular graft exhibited the superior mechanical strength, resistance to enzymatic degradation, and anti-calcification properties. Furthermore, it could drastically reduce platelet adhesion, aggregation, activation, and hemolysis. Moreover, this composite hydrogel could substantially stimulate the adherence and growth of endothelial cells as well as vascular tissue regeneration. Similarly, REDV (ArgGlu-Asp-Val) polypeptides were grafted onto double-cross-linked decellularized hAM hydrogel, which demonstrated exceptional mechanical strength, resistance to enzymatic degradation and prevented hemolysis and coagulation [169].
Research is still ongoing for engineered vascular grafts. Aslani et al. [170] successfully produced aligned and random electrospun poly-l-lactic acid (PLLA) scaffolds with hAM lysate surface-coated on them, along with acetylsalicylic acid (ASA) as the anticoagulant. The AM lysate-coated ASA-PLLA-aligned scaffold has shown appropriate tensile strength and proved to support endothelial differentiation, indicating that the scaffolds could significantly develop a biocompatible small-diameter vascular graft.
4.5. Orthopedics
Tendon wound is characterized by disordered collagen fibers development, reducing its mechanical characteristics and causing scar production. Instructive scaffolds for tendon regeneration are frequently intended to give mechanical and cellular support. In this case, Hortensius et al. [171,172] fabricated a hAM-based composite structure for tendon regeneration. Collagen-glycosaminoglycan (CG) scaffolds coupled with decellularized hAM was employed to improve tendon wound healing. The scaffolds containing ECM of hAM had dramatically enhanced mechanical qualities, in which the tenocytes had higher metabolic activity even when the medium contained the pro-inflammatory cytokine IL-1β [171]. The fabricated scaffolds impacted the early responses of MSCs to inflammatory stimuli, suggesting that they could be used as a scaffold biomaterial to improve tendon repair [172]. Dewey et al. [173] have developed another composite structure based on decellularized hAM and mineralized collagen scaffold for healing bone wounds. Their results implied that a mineralized collagen-hAM scaffold might be a helpful setting to support craniomaxillofacial bone regeneration, particularly when bone defects displayed severe inflammatory responses.
Besides repairing tendons or bone injury of the composite scaffolds based on hAM, they can also support chondrocyte cultivation. For example, Hussin et al. [174] fabricated a scaffold resembling hyaline cartilage in structure and comprised fibrin and hAM, besides two other biodegradable and biocompatible polymers. The findings demonstrated that this structure had a sufficient biodegradation rate, as well as secreting cartilage-specific ECM and GAGs to preserve cellular phenotype. Furthermore, hAM-based hydrogels may provide sufficient mechanical characteristics for articular cartilage tissue engineering. Toniato et al. [175] have developed a hybrid hydrogel with a high swelling capacity and elastic modulus using hAM, collagen, and chitosan.
4.6. Other organs or tissues
The applications of hAM in urology reconstruction are limited due to its poor mechanical characteristics. The combination of electrospun nanofibers and hAM provides a novel method for enhancing mechanical resistance while preserving the distinctive bioactivity characteristics of hAM. A sandwich-structured biocomposite material constructed from frozen hAM and the electrospun poly-(l-lactide-co-E-caprolactone) (PLCL) was developed to promote bladder wall regeneration [176]. An application of hAM composites for oral mucosal defects is also available. Zhang et al. [177] developed a composite of methacrylate gelatin (GelMA) hydrogel combined with decellularized hAM particles (dAMP) as an oral mucosal substitute to promote mucosa regeneration. The composite substitute (GelMA-dAMP) was easy to synthesize and store for oral mucosal defects repair. The findings showed that neovascularization levels in chick chorioallantoic membranes were significantly promoted, effectively treating oral mucosal defect repair. Another interesting application of hAM composites is the treatment of amniotic membrane defects during pregnancy [178]. The 3D printing technology was used to produce a biocompatible amnion-analogous medical device (AMED) to repair amniotic membrane defects. This construct consists of a polycaprolactone framework and hydrogel generated from decellularized hAM to prevent amniotic fluid leakage and protect fetal survival. In addition, we have conducted a study using decellularized hAM composited with clinical PP mesh to investigate the efficiency of the surgical mesh. The results of in vivo tests revealed that the decellularized hAM composited mesh performed excellent biocompatibility, better than the clinical PP mesh in Gynecology surgery [179]. All these applications of hAM-based biomaterials were summarized in Table 4 and vividly exhibited in Fig. 4.
Table 4.
Applications of hAM-based materials in tissue engineering.
Organs/tissues | Research purpose | hAM state | Composites | Experimental type | Outcomes | Reference |
---|---|---|---|---|---|---|
Cornea | Artificial cornea material | Decellularized hAM | PVA-AM | In vitro (rabbit corneal epithelial cells) | A biocompatible hybrid scaffold for keratoprosthesis | [148] |
Sclera | Ocular surface reconstruction | Freeze-dried hAM | FG-AM | In vivo (rabbits) | FG-AM with ideal biodegradability is useful for ocular surface reconstruction | [149] |
Cornea | Corneal alkali burns repair | Intact hAM | FG-AM | In vivo (rabbits) | FG-AM has more ideal biomechanical properties for ocular surface reconstruction | [147] |
Cornea | Corneal alkali burns repair | Decellularized hAM | PCL-AM | In vivo (rabbits) | PCL-AM composite can improve re-epithelialization, and reduce inflammation and neovascularization | [150] |
Cornea | Corneal infection control | Decellularized hAM | Clavanin A-coated hAM | In vivo (rabbits) | Clavanin A-coated hAM with better biocompatibility is appropriate to control ocular surface infection | [151] |
Cornea | Corneal injury repair | AME | AME-in situ gels or -tablets | In vivo (rabbits) | AME with in situ gels shows the best treatment efficiency | [152] |
Skin | Wound healing | Decellularized hAM | PLGC + silver nanoparticle + fibrin + hAM | In vitro (dermal fibroblasts) | hAM, combined with fibrin-silver nanoparticle-PLGC, has excellent biocompatibility for skin regeneration | [153] |
Skin | Diabetic wound healing | Decellularized hAM | Double-layer of chitosan sponge + hAM | In vitro (HFF-1) and In vivo (mice) | Double-layer dressing based on hAM and chitosan has favorable biocompatibility for diabetic wound healing | [154] |
Skin | Burn wound healing | Intact hAM | hAM and collagen-based hydrogel | In vivo (rats) | Hydrogel based on hAM and collagen accelerates wound healing by covering membranes | [155] |
Skin | Wound healing | Solubilized hAM | HA-SAM | In vivo (mice) | HA-SAM significantly accelerates wound healing by re-epithelialization | [156] |
Skin | Scar minimization | Decellularized hAM | hAM/electrospun silk fibroin | In vivo (rabbits) | hAM/electrospun silk fibroin has the potential as an efficient anti-scarring wound dressing | [159] |
Abdominal wall | Abdominal wall repair | Fresh hAM | PP mesh covered with hAM | In vivo (rats) | PP mesh coated with hAM reduces the abdominal adhesions postoperatively | [163] |
Abdominal wall | Abdominal adhesion prevention | Intact hAM | PP mesh covered with hAM | In vivo (rabbits) | PP mesh coated with hAM reduces the abdominal adhesions after laparotomy | [164] |
Abdominal wall | Abdominal adhesion prevention | bAM | PEG + PP mesh + bAM | In vivo (rats) | PP mesh coated with bAM and 5% PEG prevents the abdominal adhesions postoperatively | [165] |
Heart | Myocardial infarction repair | Injectable hAM | hAM-based hydrogel | In vivo (rats) | hAM-based hydrogel increases heart contractility and reduces fibrosis | [167] |
Blood vessel | Vascular graft | Decellularized hAM | AlgSr/PAM-AM | In vitro (HUVECs) and In vivo (rabbits) | AlgSr/PAM-AM enhances vascular repair | [168] |
Blood vessel | Vascular graft | Decellularized hAM | Hydrogel + REDV + hAM | In vitro (HUVECs) and In vivo (rabbits) | The graft enhances mechanical strength and resistance to enzymic degradation and prevents hemolysis and coagulation | [169] |
Blood vessel | Small-diameter vascular graft | hAM lysate | PLLA + ASA + hAM | In vitro (HUVECs) | The hybrid scaffold supports endothelial differentiation and provides appropriate biocompatibility | [170] |
Tendon | Tendon regeneration | Decellularized hAM | CG + hAM | In vitro (tenocytes) | CG combined with hAM enhances mechanical properties and improves tenocytes activity | [171] |
Tendon | Tendon injury repair | Decellularized hAM | CG + hAM | In vitro (MSCs) | CG combined with hAM improves the inflammatory response of MSCs | [172] |
Bone | Bone defect repair | Decellularized hAM | Collagen-hAM | In vitro (porcine adipose-derived stem cells) | Collagen-hAM composite scaffold promotes bone repair | [173] |
Cartilage | Cartilage tissue engineering | Decellularized hAM | hAM based on fibrin | In vitro (chondrocytes) | hAM based on fibrin scaffold can secrete cartilage-specific ECM and GAGs | [174] |
Cartilage | Cartilage tissue engineering | Decellularized hAM | Hydrogel (chitosan + collagen + hAM) | In vitro | Hybrid hydrogel based on hAM and chitosan, and collagen has a high swelling capacity and elastic modulus | [175] |
Bladder | Bladder wall reconstruction | Frozen hAM | PLCL + hAM | In vivo (rats) | Frozen hAM sandwiched between two layers of electrospun PLCL promotes bladder wall regeneration | [176] |
Oral mucosa | Oral mucosal defect repair | dHAP | GelMA-dAMP | In vitro (human fibroblasts) and In vivo (rabbits) | GelMA-dAMP is effective for oral mucosal defect repair | [177] |
Amniotic membrane | Amniotic membrane defect restoration | Decellularized hAM | AMED | In vivo (pig) | AMED prevents amniotic fluid leakage and protects fetal survival | [178] |
Uterus | Biocompatibility investigation | Decellularized hAM | PP mesh covered with hAM | In vivo (rabbits) | hAM coated with PP mesh shows good biocompatibility | [179] |
Fig. 4.
Therapeutic effects of hAM composites in multiple preclinical studies. Modifications based on hAM have potential in the eyes, skin, abdominal wall, cardiovascular system, orthopedics, and some other organs.
5. Conclusions and perspectives
The research of the hAM has become a prevalent topic for decades. The hAM contains most of tissue engineering components because it can provide the appropriate ECM, stem cells, and other growth factors or cytokines. The unique characteristics of hAM are responsible for its broad applications in clinics in the future. Nowadays, several commercial products are available for clinical applications, especially in ophthalmology and skin wound healing. The hAM-related products will be expanded to various medical fields as high-performance tissue engineering material, benefiting hundreds of millions of patients.
hAM-derived stem cells, including AMSCs and AECs, have significant benefits over other stem cells in abundant sources, easy isolation, minimal ethical issues, no tumorigenicity, and low immunogenicity. Moreover, the biological characteristics of hAM-derived stem cells, especially for the paracrine effects, significantly improved their importance in the experimental study and clinical practice. These characteristics make them a prospective source of stem cells for cell therapy and regenerative medicine. However, more studies are needed to further explore their pluripotent features and the mechanisms of paracrine effects. Further preclinical studies of AMSCs and AECs in treating various disorders should be performed to fully understand their therapeutic prospect.
Although hAM is a fantastic material for regenerative medicine due to its structural, physical, and biological characteristics, it has some drawbacks. These include a high degradation rate, sometimes graft rejection, and relatively poor mechanical characteristics. Therefore, appropriate addressing of these drawbacks can significantly improve hAM applications. Consequently, some modifications are needed to improve its properties combined with other biological, synthetic, or hybrid materials. Limited evidence has confirmed the effectiveness of some modifications in the fields, including ocular surface reconstruction, skin wound repair, abdominal wall reconstruction, and several other applications mentioned in this review. Eventually, more studies are needed to further investigate the effectiveness and importance of hAM-based modified structure.
Funding
This work was financially supported by National Natural Science Foundation of China (U20A20121, 32201139) Major Project of 2025 Sci & Tech Innovation of Ningbo (2020Z096), and One Health Interdisciplinary Research Project of Ningbo University (HY202210), Ningbo.
Declaration of competing interest
The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.
Acknowledgements
We acknowledge the support from K.C.Wang Magna/Education Fund of Ningbo University.
Data availability
No data was used for the research described in the article.
References
- 1.Pereira P.N., Dobreva M.P., Graham L., Huylebroeck D., Lawson K.A., Zwijsen A.N. Amnion formation in the mouse embryo: the single amniochorionic fold model. BMC Dev. Biol. 2011;11:48. doi: 10.1186/1471-213X-11-48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.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. [DOI] [PubMed] [Google Scholar]
- 3.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. [DOI] [PubMed] [Google Scholar]
- 4.Silini A.R., Di Pietro R., Lang-Olip I., Alviano F., Banerjee A., Basile M., Borutinskaite V., Eissner G., Gellhaus A., Giebel B., Huang Y.C., Janev A., Kreft M.E., Kupper N., Abadía-Molina A.C., Olivares E.G., Pandolfi A., Papait A., Pozzobon M., Ruiz-Ruiz C., Soritau O., Susman S., Szukiewicz D., Weidinger A., Wolbank S., Huppertz B., Parolini O. Perinatal derivatives: where Do we stand? A roadmap of the human placenta and consensus for tissue and cell nomenclature. Front. Bioeng. Biotechnol. 2020;8 doi: 10.3389/fbioe.2020.610544. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.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. [DOI] [PubMed] [Google Scholar]
- 6.Abbasi-Kangevari M., Ghamari S.H., Safaeinejad F., Bahrami S., Niknejad H. Potential therapeutic features of human amniotic mesenchymal stem cells in multiple sclerosis: immunomodulation, inflammation suppression, angiogenesis promotion, oxidative stress inhibition, neurogenesis induction, MMPs regulation, and remyelination stimulation. Front. Immunol. 2019;10:238. doi: 10.3389/fimmu.2019.00238. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Manuelpillai U., Moodley Y., Borlongan C.V., Parolini O. Amniotic membrane and amniotic cells: potential therapeutic tools to combat tissue inflammation and fibrosis? Placenta. 2011;32(Suppl 4):S320–S325. doi: 10.1016/j.placenta.2011.04.010. [DOI] [PubMed] [Google Scholar]
- 8.Wilshaw S.P., Kearney J., Fisher J., Ingham E. Biocompatibility and potential of acellular human amniotic membrane to support the attachment and proliferation of allogeneic cells. Tissue Eng. 2008;14(4):463–472. doi: 10.1089/tea.2007.0145. [DOI] [PubMed] [Google Scholar]
- 9.Fenelon M., Maurel D.B., Siadous R., Gremare A., Delmond S., Durand M., Brun S., Catros S., Gindraux F., L'Heureux N., Fricain J.C. Comparison of the impact of preservation methods on amniotic membrane properties for tissue engineering applications. Mater. Sci. Eng., C. 2019;104 doi: 10.1016/j.msec.2019.109903. [DOI] [PubMed] [Google Scholar]
- 10.Nouri M., Ebrahimi M., Bagheri T., Fatemi M.J., Najafbeygi A., Araghi S., Molaee M. Healing effects of dried and acellular human amniotic membrane and mepitelas for coverage of skin graft donor areas; A randomized clinical trial. Bull Emerg Trauma. 2018;6(3):195–200. doi: 10.29252/beat-060302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Abazari M.F., Soleimanifar F., Enderami S.E., Nasiri N., Nejati F., Mousavi S.A., Soleimani M., Kiani J., Ghoraeian P., Kehtari M. Decellularized amniotic membrane Scaffolds improve differentiation of iPSCs to functional hepatocyte-like cells. J. Cell. Biochem. 2020;121(2):1169–1181. doi: 10.1002/jcb.29351. [DOI] [PubMed] [Google Scholar]
- 12.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. [DOI] [PubMed] [Google Scholar]
- 13.Niknejad H., Peirovi H., Jorjani M., Ahmadiani A., Ghanavi J., Seifalian A.M. Properties of the amniotic membrane for potential use in tissue engineering. Eur. Cell. Mater. 2008;15:88–99. doi: 10.22203/ecm.v015a07. [DOI] [PubMed] [Google Scholar]
- 14.Arrizabalaga J.H., Nollert M.U. Human amniotic membrane: a versatile scaffold for tissue engineering. ACS Biomater. Sci. Eng. 2018;4(7):2226–2236. doi: 10.1021/acsbiomaterials.8b00015. [DOI] [PubMed] [Google Scholar]
- 15.Gholipourmalekabadi M., Mozafari M., Salehi M., Seifalian A., Bandehpour M., Ghanbarian H., Urbanska A.M., Sameni M., Samadikuchaksaraei A., Seifalian A.M. Development of a cost-effective and simple protocol for decellularization and preservation of human amniotic membrane as a soft tissue replacement and delivery system for bone marrow stromal cells. Adv. Healthcare Mater. 2015;4(6):918–926. doi: 10.1002/adhm.201400704. [DOI] [PubMed] [Google Scholar]
- 16.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. [DOI] [PubMed] [Google Scholar]
- 17.Khosravimelal S., Momeni M., Gholipur M., Kundu S.C., Gholipourmalekabadi M. Protocols for decellularization of human amniotic membrane. Methods Cell Biol. 2020;157:37–47. doi: 10.1016/bs.mcb.2019.11.004. [DOI] [PubMed] [Google Scholar]
- 18.Weidinger A., Poženel L., Wolbank S., Banerjee A. Sub-regional differences of the human amniotic membrane and their potential impact on tissue regeneration application. Front. Bioeng. Biotechnol. 2020;8 doi: 10.3389/fbioe.2020.613804. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Sabklla N. Use of the fetal membranes in skin grafting. Medical Record. 1866-1922;83(11):478. 1913. [Google Scholar]
- 20.Stern M. The grafting of preserved amniotic membrane to burned and ulcerated surfaces, substituing skin grafts: a preliminary report. J. Am. Med. Assoc. 1913;60(13):973–974. [Google Scholar]
- 21.De Rotth A. Plastic repair of conjunctival defects with fetal membrane. Arch. Ophthalmol. 1940;23:522–525. [Google Scholar]
- 22.Chao Y.C., Humphreys S., Penfield W. A new method of preventing adhesions. The use of amnioplastin after craniotomy. Br. Med. J. 1940;1(4134) doi: 10.1136/bmj.1.4134.517. 517-538.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Francisco J.C., Correa Cunha R., Cardoso M.A., Baggio Simeoni R., Mogharbel B.F., Picharski G.L., Silva Moreira Dziedzic D., Guarita-Souza L.C., Carvalho K.A. Decellularized amniotic membrane scaffold as a pericardial substitute: an in vivo study. Transplant. Proc. 2016;48(8):2845–2849. doi: 10.1016/j.transproceed.2016.07.026. [DOI] [PubMed] [Google Scholar]
- 24.Elkhenany H., El-Derby A., Abd Elkodous M., Salah R.A., Lotfy A., El-Badri N. Applications of the amniotic membrane in tissue engineering and regeneration: the hundred-year challenge. Stem Cell Res. Ther. 2022;13(1):8. doi: 10.1186/s13287-021-02684-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Xiao S., Xiao C., Miao Y., Wang J., Chen R., Fan Z., Hu Z. Human acellular amniotic membrane incorporating exosomes from adipose-derived mesenchymal stem cells promotes diabetic wound healing. Stem Cell Res. Ther. 2021;12(1):255. doi: 10.1186/s13287-021-02333-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Orman S., Yol S., Uzun H., Ceyran A.B., Eyüboğlu F. Effect of acellular amniotic membrane matrix patch on healing of cut surface after sleeve gastrectomy in rats. J. Invest. Surg. 2020;33(1):97–105. doi: 10.1080/08941939.2018.1473902. [DOI] [PubMed] [Google Scholar]
- 27.Gholipourmalekabadi M., Sameni M., Radenkovic D., Mozafari M., Mossahebi-Mohammadi M., Seifalian A. Decellularized human amniotic membrane: how viable is it as a delivery system for human adipose tissue-derived stromal cells? Cell Prolif. 2016;49(1):115–121. doi: 10.1111/cpr.12240. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Leal-Marin S., Kern T., Hofmann N., Pogozhykh O., Framme C., Börgel M., Figueiredo C., Glasmacher B., Gryshkov O. Human Amniotic Membrane: a review on tissue engineering, application, and storage. J. Biomed. Mater. Res. B Appl. Biomater. 2021;109(8):1198–1215. doi: 10.1002/jbm.b.34782. [DOI] [PubMed] [Google Scholar]
- 29.Cai J., Li W., Su H., Qin D., Yang J., Zhu F., Xu J., He W., Guo X., Labuda K., Peterbauer A., Wolbank S., Zhong M., Li Z., Wu W., So K.F., Redl H., Zeng L., Esteban M.A., Pei D. Generation of human induced pluripotent stem cells from umbilical cord matrix and amniotic membrane mesenchymal cells. J. Biol. Chem. 2010;285(15):11227–11234. doi: 10.1074/jbc.M109.086389. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Evangelista M., Soncini M., Parolini O. Placenta-derived stem cells: new hope for cell therapy? Cytotechnology. 2008;58(1):33–42. doi: 10.1007/s10616-008-9162-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.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 Cell. 2008;26(2):300–311. doi: 10.1634/stemcells.2007-0594. [DOI] [PubMed] [Google Scholar]
- 32.Sakuragawa N., Kakinuma K., Kikuchi A., Okano H., Uchida S., Kamo I., Kobayashi M., Yokoyama Y. Human amnion mesenchyme cells express phenotypes of neuroglial progenitor cells. J. Neurosci. Res. 2004;78(2):208–214. doi: 10.1002/jnr.20257. [DOI] [PubMed] [Google Scholar]
- 33.Liu Q.W., Li J.Y., Zhang X.C., Liu Y., Liu Q.Y., Xiao L., Zhang W.J., Wu H.Y., Deng K.Y., Xin H.B. Human amniotic mesenchymal stem cells inhibit hepatocellular carcinoma in tumour-bearing mice. J. Cell Mol. Med. 2020;24(18):10525–10541. doi: 10.1111/jcmm.15668. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Li J.Y., Ren K.K., Zhang W.J., Xiao L., Wu H.Y., Liu Q.Y., Ding T., Zhang X.C., Nie W.J., Ke Y., Deng K.Y., Liu Q.W., Xin H.B. Human amniotic mesenchymal stem cells and their paracrine factors promote wound healing by inhibiting heat stress-induced skin cell apoptosis and enhancing their proliferation through activating PI3K/AKT signaling pathway. Stem Cell Res. Ther. 2019;10(1):247. doi: 10.1186/s13287-019-1366-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Liu Q.W., Liu Q.Y., Li J.Y., Wei L., Ren K.K., Zhang X.C., Ding T., Xiao L., Zhang W.J., Wu H.Y., Xin H.B. Therapeutic efficiency of human amniotic epithelial stem cell-derived functional hepatocyte-like cells in mice with acute hepatic failure. Stem Cell Res. Ther. 2018;9(1):321. doi: 10.1186/s13287-018-1063-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Muttini A., Barboni B., Valbonetti L., Russo V., Maffulli N. Amniotic epithelial stem cells: salient features and possible therapeutic role. Sports Med. Arthrosc. Rev. 2018;26(2):70–74. doi: 10.1097/JSA.0000000000000189. [DOI] [PubMed] [Google Scholar]
- 37.Magatti M., Vertua E., Cargnoni A., Silini A., Parolini O. The immunomodulatory properties of amniotic cells: the two sides of the coin. Cell Transplant. 2018;27(1):31–44. doi: 10.1177/0963689717742819. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Ilancheran S., Michalska A., Peh G., Wallace E.M., Pera M., Manuelpillai U. Stem cells derived from human fetal membranes display multilineage differentiation potential. Biol. Reprod. 2007;77(3):577–588. doi: 10.1095/biolreprod.106.055244. [DOI] [PubMed] [Google Scholar]
- 39.Díaz-Prado S., Muiños-López E., Hermida-Gómez T., Rendal-Vázquez M.E., Fuentes-Boquete I., de Toro F.J., Blanco F.J. Isolation and characterization of mesenchymal stem cells from human amniotic membrane. Tissue Eng. C Methods. 2011;17(1):49–59. doi: 10.1089/ten.TEC.2010.0136. [DOI] [PubMed] [Google Scholar]
- 40.Izumi M., Pazin B.J., Minervini C.F., Gerlach J., Ross M.A., Stolz D.B., Turner M.E., Thompson R.L., Miki T. Quantitative comparison of stem cell marker-positive cells in fetal and term human amnion. J. Reprod. Immunol. 2009;81(1):39–43. doi: 10.1016/j.jri.2009.02.007. [DOI] [PubMed] [Google Scholar]
- 41.Miki T., Lehmann T., Cai H., Stolz D.B., Strom S.C. Stem cell characteristics of amniotic epithelial cells. Stem Cell. 2005;23(10):1549–1559. doi: 10.1634/stemcells.2004-0357. [DOI] [PubMed] [Google Scholar]
- 42.Miki T., Strom S.C. Amnion-derived pluripotent/multipotent stem cells. Stem Cell Rev. 2006;2(2):133–142. doi: 10.1007/s12015-006-0020-0. [DOI] [PubMed] [Google Scholar]
- 43.Bryzek A., Czekaj P., Plewka D., Komarska H., Tomsia M., Lesiak M., Sieroń A.L., Sikora J., Kopaczka K. Expression and co-expression of surface markers of pluripotency on human amniotic cells cultured in different growth media. Ginekol. Pol. 2013;84(12):1012–1024. doi: 10.17772/gp/1673. [DOI] [PubMed] [Google Scholar]
- 44.Han Y.M., Romero R., Kim J.S., Tarca A.L., Kim S.K., Draghici S., Kusanovic J.P., Gotsch F., Mittal P., Hassan S.S., Kim C.J. Region-specific gene expression profiling: novel evidence for biological heterogeneity of the human amnion. Biol. Reprod. 2008;79(5):954–961. doi: 10.1095/biolreprod.108.069260. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Miki T., Mitamura K., Ross M.A., Stolz D.B., Strom S.C. Identification of stem cell marker-positive cells by immunofluorescence in term human amnion. J. Reprod. Immunol. 2007;75(2):91–96. doi: 10.1016/j.jri.2007.03.017. [DOI] [PubMed] [Google Scholar]
- 46.Centurione L., Passaretta F., Centurione M.A., Munari S., Vertua E., Silini A., Liberati M., Parolini O., Di Pietro R. Mapping of the human placenta: experimental evidence of amniotic epithelial cell heterogeneity. Cell Transplant. 2018;27(1):12–22. doi: 10.1177/0963689717725078. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Moodley Y., Ilancheran S., Samuel C., Vaghjiani V., Atienza D., Williams E.D., Jenkin G., Wallace E., Trounson A., Manuelpillai U. Human amnion epithelial cell transplantation abrogates lung fibrosis and augments repair. Am. J. Respir. Crit. Care Med. 2010;182(5):643–651. doi: 10.1164/rccm.201001-0014OC. [DOI] [PubMed] [Google Scholar]
- 48.Liu Q.W., Huang Q.M., Wu H.Y., Zuo G.S., Gu H.C., Deng K.Y., Xin H.B. Characteristics and therapeutic potential of human amnion-derived stem cells. Int. J. Mol. Sci. 2021;22(2) doi: 10.3390/ijms22020970. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Wassmer C.H., Berishvili E. Immunomodulatory properties of amniotic membrane derivatives and their potential in regenerative medicine. Curr. Diabetes Rep. 2020;20(8):31. doi: 10.1007/s11892-020-01316-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Pratama G., Vaghjiani V., Tee J.Y., Liu Y.H., Chan J., Tan C., Murthi P., Gargett C., Manuelpillai U. Changes in culture expanded human amniotic epithelial cells: implications for potential therapeutic applications. PLoS One. 2011;6(11) doi: 10.1371/journal.pone.0026136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Wolbank S., Peterbauer A., Fahrner M., Hennerbichler S., van Griensven M., Stadler G., Redl H., Gabriel C. Dose-dependent immunomodulatory effect of human stem cells from amniotic membrane: a comparison with human mesenchymal stem cells from adipose tissue. Tissue Eng. 2007;13(6):1173–1183. doi: 10.1089/ten.2006.0313. [DOI] [PubMed] [Google Scholar]
- 52.Magatti M., De Munari S., Vertua E., Gibelli L., Wengler G.S., Parolini O. Human amnion mesenchyme harbors cells with allogeneic T-cell suppression and stimulation capabilities. Stem Cell. 2008;26(1):182–192. doi: 10.1634/stemcells.2007-0491. [DOI] [PubMed] [Google Scholar]
- 53.Ugurel S., Rebmann V., Ferrone S., Tilgen W., Grosse-Wilde H. U. Reinhold, Soluble human leukocyte antigen--G serum level is elevated in melanoma patients and is further increased by interferon-alpha immunotherapy. Cancer. 2001;92(2):369–376. doi: 10.1002/1097-0142(20010715)92:2<369::aid-cncr1332>3.0.co;2-u. [DOI] [PubMed] [Google Scholar]
- 54.Wu W., Lan Q., Lu H., Xu J., Zhu A., Fang W., Ge F., Hui G. Human amnion mesenchymal cells negative co-stimulatory molecules PD-L1 expression and its capacity of modulating microglial activation of CNS. Cell Biochem. Biophys. 2014;69(1):35–45. doi: 10.1007/s12013-013-9763-9. [DOI] [PubMed] [Google Scholar]
- 55.Kronsteiner B., Wolbank S., Peterbauer A., Hackl C., Redl H., van Griensven M., Gabriel C. Human mesenchymal stem cells from adipose tissue and amnion influence T-cells depending on stimulation method and presence of other immune cells. Stem Cell. Dev. 2011;20(12):2115–2126. doi: 10.1089/scd.2011.0031. [DOI] [PubMed] [Google Scholar]
- 56.Yang P.J., Yuan W.X., Liu J., Li J.Y., Tan B., Qiu C., Zhu X.L., Qiu C., Lai D.M., Guo L.H., Yu L.Y. Biological characterization of human amniotic epithelial cells in a serum-free system and their safety evaluation. Acta Pharmacol. Sin. 2018;39(8):1305–1316. doi: 10.1038/aps.2018.22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Wu Q., Fang T., Lang H., Chen M., Shi P., Pang X., Qi G. Comparison of the proliferation, migration and angiogenic properties of human amniotic epithelial and mesenchymal stem cells and their effects on endothelial cells. Int. J. Mol. Med. 2017;39(4):918–926. doi: 10.3892/ijmm.2017.2897. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Wu X., Gao F., Wu Y., Sun R., Guan W., Tian X. Isolation and biological characteristics of sheep amniotic epithelial cells. Cytotechnology. 2019;71(2):539–551. doi: 10.1007/s10616-019-00299-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Koike C., Zhou K., Takeda Y., Fathy M., Okabe M., Yoshida T., Nakamura Y., Kato Y., Nikaido T. Characterization of amniotic stem cells. Cell. Reprogr. 2014;16(4):298–305. doi: 10.1089/cell.2013.0090. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.De Coppi P., Bartsch G., Jr., Siddiqui M.M., Xu T., Santos C.C., Perin L., Mostoslavsky G., Serre A.C., Snyder E.Y., Yoo J.J., Furth M.E., Soker S., Atala A. Isolation of amniotic stem cell lines with potential for therapy. Nat. Biotechnol. 2007;25(1):100–106. doi: 10.1038/nbt1274. [DOI] [PubMed] [Google Scholar]
- 61.Díaz-Prado S., Muiños-López E., Hermida-Gómez T., Rendal-Vázquez M.E., Fuentes-Boquete I., de Toro F.J., Blanco F.J. Multilineage differentiation potential of cells isolated from the human amniotic membrane. J. Cell. Biochem. 2010;111(4):846–857. doi: 10.1002/jcb.22769. [DOI] [PubMed] [Google Scholar]
- 62.Mihu C.M., Rus Ciucă D., Soritău O., Suşman S., Mihu D. Isolation and characterization of mesenchymal stem cells from the amniotic membrane. Rom. J. Morphol. Embryol. 2009;50(1):73–77. [PubMed] [Google Scholar]
- 63.Nogami M., Tsuno H., Koike C., Okabe M., Yoshida T., Seki S., Matsui Y., Kimura T., Nikaido T. Isolation and characterization of human amniotic mesenchymal stem cells and their chondrogenic differentiation. Transplantation. 2012;93(12):1221–1228. doi: 10.1097/TP.0b013e3182529b76. [DOI] [PubMed] [Google Scholar]
- 64.Zhang Q., Lai D. Application of human amniotic epithelial cells in regenerative medicine: a systematic review. Stem Cell Res. Ther. 2020;11(1):439. doi: 10.1186/s13287-020-01951-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Uchida S., Suzuki Y., Araie M., Kashiwagi K., Otori Y., Sakuragawa N. Factors secreted by human amniotic epithelial cells promote the survival of rat retinal ganglion cells. Neurosci. Lett. 2003;341(1):1–4. doi: 10.1016/s0304-3940(02)01454-4. [DOI] [PubMed] [Google Scholar]
- 66.Kakishita K., Nakao N., Sakuragawa N., Itakura T. Implantation of human amniotic epithelial cells prevents the degeneration of nigral dopamine neurons in rats with 6-hydroxydopamine lesions. Brain Res. 2003;980(1):48–56. doi: 10.1016/s0006-8993(03)02875-0. [DOI] [PubMed] [Google Scholar]
- 67.Kim T.H., Park Y.W., Ahn J.S., Ahn J.T., Kim S.E., Jeong M.B., Seo M.S., Kang K.S., Seo K.M. Effects of conditioned media from human amniotic epithelial cells on corneal alkali injuries in rabbits. J. Vet. Sci. 2013;14(1):61–67. doi: 10.4142/jvs.2013.14.1.61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Yao X., Guo Y., Wang Q., Xu M., Zhang Q., Li T., Lai D. The paracrine effect of transplanted human amniotic epithelial cells on ovarian function improvement in a mouse model of chemotherapy-induced primary ovarian insufficiency. Stem Cell. Int. 2016;2016 doi: 10.1155/2016/4148923. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Zhang Q., Bu S., Sun J., Xu M., Yao X., He K., Lai D. Paracrine effects of human amniotic epithelial cells protect against chemotherapy-induced ovarian damage. Stem Cell Res. Ther. 2017;8(1):270. doi: 10.1186/s13287-017-0721-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Alhomrani M., Correia J., Zavou M., Leaw B., Kuk N., Xu R., Saad M.I., Hodge A., Greening D.W., Lim R., Sievert W. The human amnion epithelial cell secretome decreases hepatic fibrosis in mice with chronic liver fibrosis. Front. Pharmacol. 2017;8:748. doi: 10.3389/fphar.2017.00748. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Kuk N., Hodge A., Sun Y., Correia J., Alhomrani M., Samuel C., Moore G., Lim R., Sievert W. Human amnion epithelial cells and their soluble factors reduce liver fibrosis in murine non-alcoholic steatohepatitis. J. Gastroenterol. Hepatol. 2019;34(8):1441–1449. doi: 10.1111/jgh.14643. [DOI] [PubMed] [Google Scholar]
- 72.Song Y.S., Joo H.W., Park I.H., Shen G.Y., Lee Y., Shin J.H., Kim H., Shin I.S., Kim K.S. Transplanted human amniotic epithelial cells secrete paracrine proangiogenic cytokines in rat model of myocardial infarction. Cell Transplant. 2015;24(10):2055–2064. doi: 10.3727/096368914X685609. [DOI] [PubMed] [Google Scholar]
- 73.LeBleu V.S., Kalluri R. Exosomes exercise inhibition of anti-tumor immunity during chemotherapy. Immunity. 2019;50(3):547–549. doi: 10.1016/j.immuni.2019.02.019. [DOI] [PubMed] [Google Scholar]
- 74.Zhao B., Zhang Y., Han S., Zhang W., Zhou Q., Guan H., Liu J., Shi J., Su L., Hu D. Exosomes derived from human amniotic epithelial cells accelerate wound healing and inhibit scar formation. J. Mol. Histol. 2017;48(2):121–132. doi: 10.1007/s10735-017-9711-x. [DOI] [PubMed] [Google Scholar]
- 75.Tan J.L., Lau S.N., Leaw B., Nguyen H.P.T., Salamonsen L.A., Saad M.I., Chan S.T., Zhu D., Krause M., Kim C., Sievert W., Wallace E.M., Lim R. Amnion epithelial cell-derived exosomes restrict lung injury and enhance endogenous lung repair. Stem Cells Transl Med. 2018;7(2):180–196. doi: 10.1002/sctm.17-0185. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Zhang Q., Sun J., Huang Y., Bu S., Guo Y., Gu T., Li B., Wang C., Lai D. Human amniotic epithelial cell-derived exosomes restore ovarian function by transferring MicroRNAs against apoptosis. Mol. Ther. Nucleic Acids. 2019;16:407–418. doi: 10.1016/j.omtn.2019.03.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Pozzobon M., D'Agostino S., Roubelakis M.G., Cargnoni A., Gramignoli R., Wolbank S., Gindraux F., Bollini S., Kerdjoudj H., Fenelon M., Di Pietro R., Basile M., Borutinskaitė V., Piva R., Schoeberlein A., Eissner G., Giebel B., Ponsaerts P. General consensus on multimodal functions and validation analysis of perinatal derivatives for regenerative medicine applications. Front. Bioeng. Biotechnol. 2022;10 doi: 10.3389/fbioe.2022.961987. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Arno A., Smith A.H., Blit P.H., Shehab M.A., Gauglitz G.G., Jeschke M.G. Stem cell therapy: a new treatment for burns? Pharmaceuticals. 2011;4(10):1355–1380. doi: 10.3390/ph4101355. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Kim S.W., Zhang H.Z., Guo L., Kim J.M., Kim M.H. Amniotic mesenchymal stem cells enhance wound healing in diabetic NOD/SCID mice through high angiogenic and engraftment capabilities. PLoS One. 2012;7(7) doi: 10.1371/journal.pone.0041105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Huang J., Zhang W., Yu J., Gou Y., Liu N., Wang T., Sun C., Wu B., Li C., Chen X., Mao Y., Zhang Y., Wang J. Human amniotic mesenchymal stem cells combined with PPCNg facilitate injured endometrial regeneration. Stem Cell Res. Ther. 2022;13(1):17. doi: 10.1186/s13287-021-02682-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Liu H., Jiang C., La B., Cao M., Ning S., Zhou J., Yan Z., Li C., Cui Y., Ma X., Wang M., Chen L., Yu Y., Chen F., Zhang Y., Wu H., Liu J., Qin L. Human amnion-derived mesenchymal stem cells improved the reproductive function of age-related diminished ovarian reserve in mice through Ampk/FoxO3a signaling pathway. Stem Cell Res. Ther. 2021;12(1):317. doi: 10.1186/s13287-021-02382-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Liu Q.W., Ying Y.M., Zhou J.X., Zhang W.J., Liu Z.X., Jia B.B., Gu H.C., Zhao C.Y., Guan X.H., Deng K.Y., Xin H.B. Human amniotic mesenchymal stem cells-derived IGFBP-3, DKK-3, and DKK-1 attenuate liver fibrosis through inhibiting hepatic stellate cell activation by blocking Wnt/β-catenin signaling pathway in mice. Stem Cell Res. Ther. 2022;13(1):224. doi: 10.1186/s13287-022-02906-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Yin Y., Wang Q., Xie C., Chen H., Jin J., Miao D. Amniotic membrane mesenchymal stem cells-based therapy improves Bmi-1-deficient mandible osteoporosis through stimulating osteoblastic bone formation and inhibiting osteoclastic bone resorption. J Tissue Eng Regen Med. 2022;16(6):538–549. doi: 10.1002/term.3300. [DOI] [PubMed] [Google Scholar]
- 84.Kim S.W., Zhang H.Z., Kim C.E., An H.S., Kim J.M., Kim M.H. Amniotic mesenchymal stem cells have robust angiogenic properties and are effective in treating hindlimb ischaemia. Cardiovasc. Res. 2012;93(3):525–534. doi: 10.1093/cvr/cvr328. [DOI] [PubMed] [Google Scholar]
- 85.Kim H.G., Choi O.H. Neovascularization in a mouse model via stem cells derived from human fetal amniotic membranes. Heart Ves. 2011;26(2):196–205. doi: 10.1007/s00380-010-0064-6. [DOI] [PubMed] [Google Scholar]
- 86.Naseroleslami M., Aboutaleb N. Human amniotic membrane mesenchymal stem cells exert cardioprotective effects against isoproterenol (ISO)-induced myocardial injury through suppression of inflammation and modulation of inflammatory MAPK/NF-κB pathway. Cell Tissue Bank. 2022;23(1):67–77. doi: 10.1007/s10561-021-09915-x. [DOI] [PubMed] [Google Scholar]
- 87.Naseroleslami M., Aboutaleb N., Mokhtari B. Amniotic membrane mesenchymal stem cells labeled by iron oxide nanoparticles exert cardioprotective effects against isoproterenol (ISO)-induced myocardial damage by targeting inflammatory MAPK/NF-κB pathway. Drug Deliv Transl Res. 2021;11(1):242–254. doi: 10.1007/s13346-020-00788-3. [DOI] [PubMed] [Google Scholar]
- 88.Safari F., Shakery T., Sayadamin N. Evaluating the effect of secretome of human amniotic mesenchymal stromal cells on apoptosis induction and epithelial-mesenchymal transition inhibition in LNCaP prostate cancer cells based on 2D and 3D cell culture models. Cell Biochem. Funct. 2021;39(6):813–820. doi: 10.1002/cbf.3654. [DOI] [PubMed] [Google Scholar]
- 89.Papait A., Ragni E., Cargnoni A., Vertua E., Romele P., Masserdotti A., Perucca Orfei C., Signoroni P.B., Magatti M., Silini A.R., De Girolamo L., Parolini O. Comparison of EV-free fraction, EVs, and total secretome of amniotic mesenchymal stromal cells for their immunomodulatory potential: a translational perspective. Front. Immunol. 2022;13 doi: 10.3389/fimmu.2022.960909. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Webber M.J., Khan O.F., Sydlik S.A., Tang B.C., Langer R. A perspective on the clinical translation of scaffolds for tissue engineering. Ann. Biomed. Eng. 2015;43(3):641–656. doi: 10.1007/s10439-014-1104-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Ngadiman N.H.A., Noordin M.Y., Idris A., Kurniawan D. A review of evolution of electrospun tissue engineering scaffold: from two dimensions to three dimensions. Proc. Inst. Mech. Eng. H. 2017;231(7):597–616. doi: 10.1177/0954411917699021. [DOI] [PubMed] [Google Scholar]
- 92.Hollister S.J. Porous scaffold design for tissue engineering. Nat. Mater. 2005;4(7):518–524. doi: 10.1038/nmat1421. [DOI] [PubMed] [Google Scholar]
- 93.Guo Q., Lu X., Xue Y., Zheng H., Zhao X., Zhao H. A new candidate substrate for cell-matrix adhesion study: the acellular human amniotic matrix. J. Biomed. Biotechnol. 2012;2012 doi: 10.1155/2012/306083. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Yeung T.L.M., Liu S., Li B.C.Y., Mok K.M., Li K.K.W. Amniotic membrane harvesting during COVID-19 pandemic. Eye (Lond) 2021;35(3):1019. doi: 10.1038/s41433-020-0947-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.E.D.f.t.Q.o . European Directorate for the Quality of Medicines & HealthCare; 2019. Medicines, C. d'Europa, Guide to the Quality and Safety of Tissues and Cells for Human Application. [Google Scholar]
- 96.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. [PubMed] [Google Scholar]
- 97.Nakamura T., Yoshitani M., Rigby H., Fullwood N.J., Ito W., Inatomi T., Sotozono C., Nakamura T., Shimizu Y., Kinoshita S. Sterilized, freeze-dried amniotic membrane: a useful substrate for ocular surface reconstruction. Invest. Ophthalmol. Vis. Sci. 2004;45(1):93–99. doi: 10.1167/iovs.03-0752. [DOI] [PubMed] [Google Scholar]
- 98.Yeu E., Goldberg D.F., Mah F.S., Beckman K.A., Luchs J.I., Solomon J.D., White D.E., Gupta P.K. Safety and efficacy of amniotic cytokine extract in the treatment of dry eye disease. Clin. Ophthalmol. 2019;13:887–894. doi: 10.2147/OPTH.S203510. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Xue S.L., Liu K., Parolini O., Wang Y., Deng L., Huang Y.C. Human acellular amniotic membrane implantation for lower third nasal reconstruction: a promising therapy to promote wound healing. Burns Trauma. 2018;6:34. doi: 10.1186/s41038-018-0136-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Wu Z., Liu X., Yuan D., Zhao J. Human acellular amniotic membrane is adopted to treat venous ulcers. Exp. Ther. Med. 2018;16(2):1285–1289. doi: 10.3892/etm.2018.6331. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Eng D.L., Gordon T.R., Kocsis J.D., Waxman S.G. Development of 4-AP and TEA sensitivities in mammalian myelinated nerve fibers. J. Neurophysiol. 1988;60(6):2168–2179. doi: 10.1152/jn.1988.60.6.2168. [DOI] [PubMed] [Google Scholar]
- 102.Doudi S., Barzegar M., Taghavi E.A., Eini M., Ehterami A., Stokes K., Alexander J.S., Salehi M. Applications of acellular human amniotic membrane in regenerative medicine. Life Sci. 2022;310 doi: 10.1016/j.lfs.2022.121032. [DOI] [PubMed] [Google Scholar]
- 103.Gholipourmalekabadi M., Samadikuchaksaraei A., Seifalian A.M., Urbanska A.M., Ghanbarian H., Hardy J.G., Omrani M.D., Mozafari M., Reis R.L., Kundu S.C. Silk fibroin/amniotic membrane 3D bi-layered artificial skin. Biomed. Mater. 2018;13(3) doi: 10.1088/1748-605X/aa999b. [DOI] [PubMed] [Google Scholar]
- 104.Jin H., Yang Q., Ji F., Zhang Y.J., Zhao Y., Luo M. Human amniotic epithelial cell transplantation for the repair of injured brachial plexus nerve: evaluation of nerve viscoelastic properties. Neural Regen Res. 2015;10(2):260–265. doi: 10.4103/1673-5374.152380. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Banerjee A., Weidinger A., Hofer M., Steinborn R., Lindenmair A., Hennerbichler-Lugscheider S., Eibl J., Redl H., Kozlov A.V., Wolbank S. Different metabolic activity in placental and reflected regions of the human amniotic membrane. Placenta. 2015;36(11):1329–1332. doi: 10.1016/j.placenta.2015.08.015. [DOI] [PubMed] [Google Scholar]
- 106.Horvath V., Svobodova A., Cabral J.V., Fiala R., Burkert J., Stadler P., Lindner J., Bednar J., Zemlickova M., Jirsova K. Cell Tissue Bank; 2023. Inter-placental Variability Is Not a Major Factor Affecting the Healing Efficiency of Amniotic Membrane when Used for Treating Chronic Non-healing Wounds. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.McLaren J., Malak T.M., Bell S.C. Structural characteristics of term human fetal membranes prior to labour: identification of an area of altered morphology overlying the cervix. Hum. Reprod. 1999;14(1):237–241. doi: 10.1093/humrep/14.1.237. [DOI] [PubMed] [Google Scholar]
- 108.Richardson L., Menon R. Fetal membrane at the feto-maternal interface: an underappreciated and understudied intrauterine tissue. Placenta Reprod Med. 2022;1 doi: 10.54844/prm.2022.0104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Dua H.S., Maharajan V.S., Hopkinson A. Controversies and limitations of amniotic membrane in ophthalmic surgery. Cornea and external eye disease. 2006:21–33. [Google Scholar]
- 110.Chen B., Jones R.R., Mi S., Foster J., Alcock S.G., Hamley I.W., Connon C.J. The mechanical properties of amniotic membrane influence its effect as a biomaterial for ocular surface repair. Soft Matter. 2012;8(32):8379–8387. [Google Scholar]
- 111.Grémare A., Jean-Gilles S., Musqui P., Magnan L., Torres Y., Fénelon M., Brun S., Fricain J.C., L'Heureux N. Cartography of the mechanical properties of the human amniotic membrane. J. Mech. Behav. Biomed. Mater. 2019;99:18–26. doi: 10.1016/j.jmbbm.2019.07.007. [DOI] [PubMed] [Google Scholar]
- 112.Tehrani F.A., Modaresifar K., Azizian S., Niknejad H. Induction of antimicrobial peptides secretion by IL-1β enhances human amniotic membrane for regenerative medicine. Sci. Rep. 2017;7(1) doi: 10.1038/s41598-017-17210-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Mao Y., Hoffman T., Singh-Varma A., Duan-Arnold Y., Moorman M., Danilkovitch A., Kohn J. Antimicrobial peptides secreted from human cryopreserved viable amniotic membrane contribute to its antibacterial activity. Sci. Rep. 2017;7(1) doi: 10.1038/s41598-017-13310-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Yadav M.K., Go Y.Y., Kim S.H., Chae S.W., Song J.J. Antimicrobial and antibiofilm effects of human amniotic/chorionic membrane extract on Streptococcus pneumoniae. Front. Microbiol. 2017;8:1948. doi: 10.3389/fmicb.2017.01948. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Ramuta T., Tratnjek L., Janev A., Seme K., Starčič Erjavec M., Kreft M.E. The antibacterial activity of human amniotic membrane against multidrug-resistant bacteria associated with urinary tract infections: new insights from normal and cancerous urothelial models. Biomedicines. 2021;9(2) doi: 10.3390/biomedicines9020218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Papait A., Cargnoni A., Sheleg M., Silini A.R., Kunis G., Ofir R., Parolini O. Perinatal cells: a promising COVID-19 therapy? Front. Bioeng. Biotechnol. 2020;8 doi: 10.3389/fbioe.2020.619980. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Brijacak N., Dekaris I., Gagro A., Gabrić N. Therapeutic effect of amniotic membrane in persistent epithelial defects and corneal ulcers in herpetic keratitis. Coll. Antropol. 2008;32(Suppl 2):21–25. [PubMed] [Google Scholar]
- 118.Shi W., Chen M., Xie L. Amniotic membrane transplantation combined with antiviral and steroid therapy for herpes necrotizing stromal keratitis. Ophthalmology. 2007;114(8):1476–1481. doi: 10.1016/j.ophtha.2006.11.027. [DOI] [PubMed] [Google Scholar]
- 119.Eming S.A., Martin P., Tomic-Canic M. Wound repair and regeneration: mechanisms, signaling, and translation. Sci. Transl. Med. 2014;6(265):265sr6. doi: 10.1126/scitranslmed.3009337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Wynn T.A., Ramalingam T.R. Mechanisms of fibrosis: therapeutic translation for fibrotic disease. Nat. Med. 2012;18(7):1028–1040. doi: 10.1038/nm.2807. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Munoz-Torres J.R., Martínez-González S.B., Lozano-Luján A.D., Martínez-Vázquez M.C., Velasco-Elizondo P., Garza-Veloz I., Martinez-Fierro M.L. Biological properties and surgical applications of the human amniotic membrane. Front. Bioeng. Biotechnol. 2022;10 doi: 10.3389/fbioe.2022.1067480. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Li W., He H., Chen Y.T., Hayashida Y., Tseng S.C. Reversal of myofibroblasts by amniotic membrane stromal extract. J. Cell. Physiol. 2008;215(3):657–664. doi: 10.1002/jcp.21345. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Castellanos G., Bernabé-García Á., Moraleda J.M., Nicolás F.J. Amniotic membrane application for the healing of chronic wounds and ulcers. Placenta. 2017;59:146–153. doi: 10.1016/j.placenta.2017.04.005. [DOI] [PubMed] [Google Scholar]
- 124.Li J., Qiu C., Zhang Z., Yuan W., Ge Z., Tan B., Yang P., Liu J., Zhu X., Qiu C., Lai D., Guo L., Yu L. Subretinal transplantation of human amniotic epithelial cells in the treatment of autoimmune uveitis in rats. Cell Transplant. 2018;27(10):1504–1514. doi: 10.1177/0963689718796196. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Solomon A., Rosenblatt M., Monroy D., Ji Z., Pflugfelder S.C., Tseng S.C. Suppression of interleukin 1alpha and interleukin 1beta in human limbal epithelial cells cultured on the amniotic membrane stromal matrix. Br. J. Ophthalmol. 2001;85(4):444–449. doi: 10.1136/bjo.85.4.444. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Shimmura S., Shimazaki J., Ohashi Y., Tsubota K. Antiinflammatory effects of amniotic membrane transplantation in ocular surface disorders. Cornea. 2001;20(4):408–413. doi: 10.1097/00003226-200105000-00015. [DOI] [PubMed] [Google Scholar]
- 127.Kubo M., Sonoda Y., Muramatsu R., Usui M. Immunogenicity of human amniotic membrane in experimental xenotransplantation. Invest. Ophthalmol. Vis. Sci. 2001;42(7):1539–1546. [PubMed] [Google Scholar]
- 128.Silini A.R., Papait A., Cargnoni A., Vertua E., Romele P., Bonassi Signoroni P., Magatti M., De Munari S., Masserdotti A., Pasotti A., Rota Nodari S., Pagani G., Bignardi M., Parolini O. CM from intact hAM: an easily obtained product with relevant implications for translation in regenerative medicine. Stem Cell Res. Ther. 2021;12(1):540. doi: 10.1186/s13287-021-02607-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Niknejad H., Paeini-Vayghan G., Tehrani F.A., Khayat-Khoei M., Peirovi H. Side dependent effects of the human amnion on angiogenesis. Placenta. 2013;34(4):340–345. doi: 10.1016/j.placenta.2013.02.001. [DOI] [PubMed] [Google Scholar]
- 130.Kim J.C., Tseng S.C. The effects on inhibition of corneal neovascularization after human amniotic membrane transplantation in severely damaged rabbit corneas. Kor. J. Ophthalmol. 1995;9(1):32–46. doi: 10.3341/kjo.1995.9.1.32. [DOI] [PubMed] [Google Scholar]
- 131.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Küçükerdönmez C., Akova Y.A., Altinörs D.D. Vascularization is more delayed in amniotic membrane graft than conjunctival autograft after pterygium excision. Am. J. Ophthalmol. 2007;143(2):245–249. doi: 10.1016/j.ajo.2006.10.032. [DOI] [PubMed] [Google Scholar]
- 133.Miyamoto S., Teramoto H., Gutkind J.S., Yamada K.M. Integrins can collaborate with growth factors for phosphorylation of receptor tyrosine kinases and MAP kinase activation: roles of integrin aggregation and occupancy of receptors. J. Cell Biol. 1996;135(6 Pt 1):1633–1642. doi: 10.1083/jcb.135.6.1633. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.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. [DOI] [PubMed] [Google Scholar]
- 135.Ilic D., Vicovac L., Nikolic M., Lazic Ilic E. Human amniotic membrane grafts in therapy of chronic non-healing wounds. Br. Med. Bull. 2016;117(1):59–67. doi: 10.1093/bmb/ldv053. [DOI] [PubMed] [Google Scholar]
- 136.Lau H.H., Jou Q.B., Huang W.C., Su T.H. Amniotic membrane graft in the management of complex vaginal mesh erosion. J. Clin. Med. 2020;9(2) doi: 10.3390/jcm9020356. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Nassim J.S., Karim S.A., Grenier P.O., Schmidt B., Jones K.M. Infantile toxic epidermal necrolysis: successful treatment of an 8-week-old with intravenous immunoglobulin and amniotic membrane transplant. Pediatr. Dermatol. 2021;38(1):202–205. doi: 10.1111/pde.14376. [DOI] [PubMed] [Google Scholar]
- 138.Marton E., Giordan E., Gallinaro P., Curzi C., Trojan D., Paolin A., Guerriero A., Rossi S., Bendini M., Longatti P., Canova G. Homologous amniotic membrane as a dural substitute in decompressive craniectomies. J. Clin. Neurosci. 2021;89:412–421. doi: 10.1016/j.jocn.2021.05.030. [DOI] [PubMed] [Google Scholar]
- 139.Lipový B., Hladík M., Štourač P., Forostyak S. Case report: wound closure acceleration in a patient with toxic epidermal necrolysis using a Lyophilised amniotic membrane. Front. Bioeng. Biotechnol. 2021;9 doi: 10.3389/fbioe.2021.649317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Ratto C., Parolini O., Marra A.A., Orticelli V., Parello A., Campennì P., De Simone V., Trojan D., Litta F. Human amniotic membrane for the treatment of cryptoglandular anal Fistulas. J. Clin. Med. 2022;11(5) doi: 10.3390/jcm11051350. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.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. [DOI] [PubMed] [Google Scholar]
- 142.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. [DOI] [PubMed] [Google Scholar]
- 143.Lacorzana J. Amniotic membrane, clinical applications and tissue engineering. Review of its ophthalmic use. Arch. Soc. Esp. Oftalmol. 2020;95(1):15–23. doi: 10.1016/j.oftal.2019.09.010. [DOI] [PubMed] [Google Scholar]
- 144.Puyana S., Ruiz S., Elkbuli A., Bernal E., McKenney M., Lim R., Askari M., Mir H. Using dehydrated amniotic membrane skin substitute in facial burns: is there a outcome difference between adult and pediatric patients? J. Craniofac. Surg. 2020;31(2):e145–e147. doi: 10.1097/SCS.0000000000006077. [DOI] [PubMed] [Google Scholar]
- 145.Liang X., Zhou L., Yan J. Amniotic membrane for treating skin graft donor sites: a systematic review and meta-analysis. Burns. 2020;46(3):621–629. doi: 10.1016/j.burns.2019.09.010. [DOI] [PubMed] [Google Scholar]
- 146.Hossain M.L., Rahman M.A., Siddika A., Adnan M., Rahman H., Diba F., Hasan M.Z., Asaduzzaman S. Burn and wound healing using radiation sterilized human amniotic membrane and centella asiatica derived gel: a review. Regenerative Engineering and Translational Medicine. 2020;6:347–357. [Google Scholar]
- 147.Cai M., Zhang J., Guan L., Zhao M. Novel implantable composite biomaterial by fibrin glue and amniotic membrane for ocular surface reconstruction. J. Mater. Sci. Mater. Med. 2015;26(3):149. doi: 10.1007/s10856-015-5491-6. [DOI] [PubMed] [Google Scholar]
- 148.Uchino Y., Shimmura S., Miyashita H., Taguchi T., Kobayashi H., Shimazaki J., Tanaka J., Tsubota K. Amniotic membrane immobilized poly(vinyl alcohol) hybrid polymer as an artificial cornea scaffold that supports a stratified and differentiated corneal epithelium. J. Biomed. Mater. Res. B Appl. Biomater. 2007;81(1):201–206. doi: 10.1002/jbm.b.30654. [DOI] [PubMed] [Google Scholar]
- 149.Sekiyama E., Nakamura T., Kurihara E., Cooper L.J., Fullwood N.J., Takaoka M., Hamuro J., Kinoshita S. Novel sutureless transplantation of bioadhesive-coated, freeze-dried amniotic membrane for ocular surface reconstruction. Invest. Ophthalmol. Vis. Sci. 2007;48(4):1528–1534. doi: 10.1167/iovs.06-1104. [DOI] [PubMed] [Google Scholar]
- 150.Zhou Z., Long D., Hsu C.C., Liu H., Chen L., Slavin B., Lin H., Li X., Tang J., Yiu S., Tuffaha S., Mao H.Q. Nanofiber-reinforced decellularized amniotic membrane improves limbal stem cell transplantation in a rabbit model of corneal epithelial defect. Acta Biomater. 2019;97:310–320. doi: 10.1016/j.actbio.2019.08.027. [DOI] [PubMed] [Google Scholar]
- 151.Singh R., Kumar D., Kumar P., Chacharkar M.P. Development and evaluation of silver-impregnated amniotic membrane as an antimicrobial burn dressing. J. Burn Care Res. 2008;29(1):64–72. doi: 10.1097/BCR.0b013e31815f5a0f. [DOI] [PubMed] [Google Scholar]
- 152.Luo Y., Shen M., Feng P., Qiu H., Wu X., Yang L., Zhu Y. Various administration forms of decellularized amniotic membrane extract towards improving corneal repair. J. Mater. Chem. B. 2021;9(45):9347–9357. doi: 10.1039/d1tb01848e. [DOI] [PubMed] [Google Scholar]
- 153.Ramakrishnan R., Krishnan L.K., Nair R.P., Kalliyana Krishnan V. Reinforcement of amniotic membrane with fibrin coated poly-[Lactide-co-Glycolide-co-Caprolactone] terpolymer containing silver nanoparticles for potential wound healing applications. International Journal of Polymeric Materials and Polymeric Biomaterials. 2020;69(12):810–819. [Google Scholar]
- 154.Yang Y., Zhang Y., Yan Y., Ji Q., Dai Y., Jin S., Liu Y., Chen J., Teng L. A sponge-like double-layer wound dressing with chitosan and decellularized bovine amniotic membrane for promoting diabetic wound healing. Polymers. 2020;12(3) doi: 10.3390/polym12030535. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Rana M.M., Rahman M.S., Ullah M.A., Siddika A., Hossain M.L., Akhter M.S., Hasan M.Z., Asaduzzaman S.M. Amnion and collagen-based blended hydrogel improves burn healing efficacy on a rat skin wound model in the presence of wound dressing biomembrane. Bio Med. Mater. Eng. 2020;31(1):1–17. doi: 10.3233/BME-201076. [DOI] [PubMed] [Google Scholar]
- 156.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Mohammadi A.A., Eskandari S., Johari H.G., Rajabnejad A. Using amniotic membrane as a novel method to reduce post-burn hypertrophic scar formation: a prospective Follow-up study. J. Cutan. Aesthetic Surg. 2017;10(1):13–17. doi: 10.4103/JCAS.JCAS_109_16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Zhu Z., Ding J., Shankowsky H.A., Tredget E.E. The molecular mechanism of hypertrophic scar. J Cell Commun Signal. 2013;7(4):239–252. doi: 10.1007/s12079-013-0195-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Gholipourmalekabadi M., Khosravimelal S., Nokhbedehghan Z., Sameni M., Jajarmi V., Urbanska A.M., Mirzaei H., Salimi M., Chauhan N.P.S., Mobaraki M., Reis R.L., Samadikuchaksaraei A., Kundu S.C. Modulation of hypertrophic scar formation using amniotic membrane/electrospun silk fibroin bilayer membrane in a rabbit ear model. ACS Biomater. Sci. Eng. 2019;5(3):1487–1496. doi: 10.1021/acsbiomaterials.8b01521. [DOI] [PubMed] [Google Scholar]
- 160.Castro P.M., Rabelato J.T., Monteiro G.G., del Guerra G.C., Mazzurana M., Alvarez G.A. Laparoscopy versus laparotomy in the repair of ventral hernias: systematic review and meta-analysis. Arq. Gastroenterol. 2014;51(3):205–211. doi: 10.1590/s0004-2803201400030008. [DOI] [PubMed] [Google Scholar]
- 161.Kolker A.R., Brown D.J., Redstone J.S., Scarpinato V.M., Wallack M.K. Multilayer reconstruction of abdominal wall defects with acellular dermal allograft (AlloDerm) and component separation. Ann. Plast. Surg. 2005;55(1):36–41. doi: 10.1097/01.sap.0000168248.83197.d4. discussion 41-2. [DOI] [PubMed] [Google Scholar]
- 162.Kingsnorth A. The management of incisional hernia. Ann. R. Coll. Surg. Engl. 2006;88(3):252–260. doi: 10.1308/003588406X106324. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Soylu S., Yildiz C., Bozkurt B., Karakus S., Begum K., Kurt A. Amniotic membrane-coated polypropylene mesh for the repair of incisional hernia: an experimental study in a rat model of abdominal wall defect. Iran. Red Crescent Med. J. 2018;20(3) [Google Scholar]
- 164.Najibpour N., Ahmed M.A.A.H., Bananzadeh A., Rezaianzadeh A., Kermani M.R., Gabash K.M., Tajali H., Hosseini S.V., Mehrabani D. The effect of human amniotic membrane as a covering layer on propylene mesh in decrease of adhesion after laparotomy in the rabbit. Comp. Clin. Pathol. 2016;25:131–135. [Google Scholar]
- 165.Rashid H.Q., Karabulut E., Cevik A. Effectiveness of polypropylene mesh coated bovine amniotic membrane with adhesion barrier (polyethylene glycol) in repair of abdominal wall hernias in rats. Indian J. Anim. Sci. 2018;88:1010–1014. [Google Scholar]
- 166.Kim G.H., Uriel N., Burkhoff D. Reverse remodelling and myocardial recovery in heart failure. Nat. Rev. Cardiol. 2018;15(2):83–96. doi: 10.1038/nrcardio.2017.139. [DOI] [PubMed] [Google Scholar]
- 167.Henry J.J.D., Delrosario L., Fang J., Wong S.Y., Fang Q., Sievers R., Kotha S., Wang A., Farmer D., Janaswamy P., Lee R.J., Li S. Development of injectable amniotic membrane matrix for postmyocardial infarction tissue repair. Adv. Healthcare Mater. 2020;9(2) doi: 10.1002/adhm.201900544. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Lei X., Wu Y., Peng X., Zhao Y., Zhou X., Yu X. Research on alginate-polyacrylamide enhanced amnion hydrogel, a potential vascular substitute material. Mater. Sci. Eng., C. 2020;115 doi: 10.1016/j.msec.2020.111145. [DOI] [PubMed] [Google Scholar]
- 169.Peng X., Wang X., Cheng C., Zhou X., Gu Z., Li L., Liu J., Yu X. Bioinspired, artificial, small-diameter vascular grafts with selective and rapid endothelialization based on an amniotic membrane-derived hydrogel. ACS Biomater. Sci. Eng. 2020;6(3):1603–1613. doi: 10.1021/acsbiomaterials.9b01493. [DOI] [PubMed] [Google Scholar]
- 170.Aslani S., Kabiri M., Kehtari M., Hanaee-Ahvaz H. Vascular tissue engineering: fabrication and characterization of acetylsalicylic acid-loaded electrospun scaffolds coated with amniotic membrane lysate. J. Cell. Physiol. 2019;234(9):16080–16096. doi: 10.1002/jcp.28266. [DOI] [PubMed] [Google Scholar]
- 171.Hortensius R.A., Ebens J.H., Harley B.A. Immunomodulatory effects of amniotic membrane matrix incorporated into collagen scaffolds. J. Biomed. Mater. Res. 2016;104(6):1332–1342. doi: 10.1002/jbm.a.35663. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Hortensius R.A., Ebens J.H., Dewey M.J., Harley B.A.C. Incorporation of the amniotic membrane as an immunomodulatory design element in collagen scaffolds for tendon repair. ACS Biomater. Sci. Eng. 2018;4(12):4367–4377. doi: 10.1021/acsbiomaterials.8b01154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Dewey M.J., Johnson E.M., Slater S.T., Milner D.J., Wheeler M.B., Harley B.A.C. Mineralized collagen scaffolds fabricated with amniotic membrane matrix increase osteogenesis under inflammatory conditions. Regen Biomater. 2020;7(3):247–258. doi: 10.1093/rb/rbaa005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Hussin I., Pingguan-Murphy B., Osman S. 5th Kuala Lumpur International Conference on Biomedical Engineering 2011: (BIOMED 2011) 20-23 June 2011, Kuala Lumpur. Springer; Malaysia: 2011. The fabrication of human amniotic membrane based hydrogel for cartilage tissue engineering applications: a preliminary study; pp. 841–844. [Google Scholar]
- 175.Toniato T.V., Stocco T.D., Martins D.d.S., Santanna L.B., Tim C.R., Marciano F.R., Silva-Filho E.C., Campana-Filho S.P., Lobo A.d.O. Hybrid chitosan/amniotic membrane-based hydrogels for articular cartilage tissue engineering application. International Journal of Polymeric Materials and Polymeric Biomaterials. 2020;69(15):961–970. [Google Scholar]
- 176.Adamowicz J., Pokrywczyńska M., Tworkiewicz J., Kowalczyk T., van Breda S.V., 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) doi: 10.1371/journal.pone.0146012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Zhang Q., Qian C., Xiao W., Zhu H., Guo J., Ge Z., Cui W. Development of a visible light, cross-linked GelMA hydrogel containing decellularized human amniotic particles as a soft tissue replacement for oral mucosa repair. RSC Adv. 2019;9(32):18344–18352. doi: 10.1039/c9ra03009c. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Lee J.Y., Kim H., Ha D.H., Shin J.C., Kim A., Ko H.S., Cho D.W. Amnion-analogous medical device for fetal membrane healing: a preclinical Long-term study. Adv. Healthcare Mater. 2018;7(18) doi: 10.1002/adhm.201800673. [DOI] [PubMed] [Google Scholar]
- 179.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. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
No data was used for the research described in the article.