ArticlePDF Available

Biological characterization of dehydrated amniotic membrane allograft: Mechanisms of action and implications for wound care

Authors:

Abstract and Figures

There is a growing clinical demand in the wound care market to treat chronic wounds such as diabetic foot ulcers. Advanced cell and tissue‐based products (CTPs) are often used to address challenging chronic wounds where healing has stalled. These products contain active biologics such as growth factors and cytokines as well as structural components that support and stimulate cell growth and assist in tissue regeneration. This study addresses the in vitro biologic effects of a clinically available dehydrated amniotic membrane allograft (DAMA). The broad mechanism of action results from DAMA's biologic composition that leads to stimulation of cell migration cell proliferation, and reduction of pro‐inflammatory cytokines. Results show that DAMA possesses growth factors and cytokines such as EGF, FGF, PDGFs, VEGF, TGF‐β, IL‐8, and TIMPs 1 and 2. Furthermore, in vitro experiments demonstrate that DAMA stimulates cell proliferation, cell migration, secretion of collagen type I, and the reduction of pro‐inflammatory cytokines IL‐1β, IL‐6, and TNF‐α. This study findings are consistent with the clinical benefits previously published for DAMA and other CTPs in chronic wounds suggesting that the introduction of DAMA to non‐healing, complex wounds helps to improve the wound milieu by providing essential structural components, cytokines, and growth factors to create an appropriate environment for wound healing.
This content is subject to copyright. Terms and conditions apply.
ORIGINAL RESEARCH REPORT
Biological characterization of dehydrated amniotic membrane
allograft: Mechanisms of action and implications for
wound care
Marc C. Moore
1
| Paul P. Bonvallet
2
| Sita M. Damaraju
2
| Heli N. Modi
2
|
Ankur Gandhi
2
| Peter S. McFetridge
3,4
1
Stephenson School of Biomedical
Engineering, University of Oklahoma, Norman,
Oklahoma
2
Product Development, Integra LifeSciences,
Princeton, New Jersey, 08540
3
J. Crayton Pruitt Family Department of
Biomedical Engineering , University of Florida,
Gainesville, Florida
4
Department of Molecular Genetics and
Microbiology, University of Florida,
Gainesville, Florida
Correspondence
Peter McFetridge, J. Crayton Pruitt Family
Department of Biomedical Engineering,
University of Florida, Gainesville, FL, USA.
Email: pmcfetridge@bme.ufl.edu
Funding information
Integra LifeSciences, Grant/Award Number: -
Abstract
There is a growing clinical demand in the wound care market to treat chronic wounds
such as diabetic foot ulcers. Advanced cell and tissue-based products (CTPs) are
often used to address challenging chronic wounds where healing has stalled. These
products contain active biologics such as growth factors and cytokines as well as
structural components that support and stimulate cell growth and assist in tissue
regeneration. This study addresses the in vitro biologic effects of a clinically available
dehydrated amniotic membrane allograft (DAMA). The broad mechanism of action
results from DAMA's biologic composition that leads to stimulation of cell migration
cell proliferation, and reduction of pro-inflammatory cytokines. Results show that
DAMA possesses growth factors and cytokines such as EGF, FGF, PDGFs, VEGF,
TGF-β, IL-8, and TIMPs 1 and 2. Furthermore, in vitro experiments demonstrate that
DAMA stimulates cell proliferation, cell migration, secretion of collagen type I, and
the reduction of pro-inflammatory cytokines IL-1β, IL-6, and TNF-α. This study find-
ings are consistent with the clinical benefits previously published for DAMA and
other CTPs in chronic wounds suggesting that the introduction of DAMA to non-
healing, complex wounds helps to improve the wound milieu by providing essential
structural components, cytokines, and growth factors to create an appropriate envi-
ronment for wound healing.
KEYWORDS
cytokines, growth factors, wound healing
1|INTRODUCTION
Wound healing is a complex process that is characterized by three dis-
tinct phases: inflammation, proliferation, and remodeling (Broughton
et al., 2006; Enoch & Leaper, 2005; Falanga, 2005; Kirsner &
Eaglstein, 1993; Velnar et al., 2009). During the initial stage of wound
healing, platelets flood the wound site to begin the clotting process as
well as release multiple chemical signals to begin a cellular signaling
cascade. In this phase, the inflammatory response results in phagocy-
tosis of debris and dead or injured cells. During the phase transition
from inflammation to proliferation, growth factors and cytokines such
as epidermal growth factor (EGF), transforming growth factor beta
(TGF-β), and platelet-derived growth factor (PDGF) are released
(Barrientos et al., 2008; Stojanovic et al., 1996; Werner &
Grose, 2017). These molecules signal cellular migration, recruitment
and proliferation, processes that are essential for effective tissue rem-
odeling, vascularization, and epithelialization (Ellis et al., 2018). In
addition, extracellular matrix (ECM) components, including structural
proteins, glycoproteins, and glycosaminoglycans (GAGs), play a critical
role in maintaining the physiological microenvironment and contribute
Received: 28 October 2019 Revised: 31 March 2020 Accepted: 29 April 2020
DOI: 10.1002/jbm.b.34635
3076 © 2020 Wiley Periodicals, Inc. J Biomed Mater Res. 2020;108B:30763083.wileyonlinelibrary.com/journal/jbmb
building blocks for the healing process (Cornwell et al., 2009; Schultz
et al., 2011). During the proliferative phase, new ECM forms as cells
respond to signals from fibroblasts, myofibroblasts, keratinocytes, and
inflammatory cells which leads to angiogenesis, extracellular matrix
deposition, and finally re-epithelialization (Hutton et al., 2009; Koob
et al., 2014a; Olczyk et al., 2014; Sheikh et al., 2014; Tseng
et al., 1999). Once re-epithelialization is complete, the remodeling
phase begins where the disorganized ECM proteins such as collagen
are remodeled and organized allowing for an increase in strength.
During the wound healing process, cells and ECM interact in a
bidirectional manner known as dynamic reciprocity, where cells syn-
thesize and reconstruct the ECM and in turn the ECM provides a
favorable environment for cell attachment and proliferation
(McQuilling et al., 2017; Schultz et al., 2011). This interaction is
heavily regulated by cytokines and growth factors. In normal wound
healing, the transition between phases is orderly and the signaling
molecules regulate this process; however, in chronic wounds, progres-
sion stalls at the inflammatory phase (Ellis et al., 2018). During this
stalled state, there is an elevated inflammatory response along with
sustained levels of proteases such as matrix metalloproteinases
(MMPs) which have been shown to regulate the growth factor and
cytokine response (Zhang, 2010) as well as degrade existing and new
ECM, further damaging tissue and preventing the wound from pro-
gressing to the proliferative stage (Schultz et al., 2011; Cutting &
Tong, 2003). In these cases, the clinician must intervene with more
advanced wound care solutions. Studies have demonstrated a clinical
benefit from interventions such as cell and tissue-based products
(CTPs), which incorporate both ECM and growth factors to stimulate
the healing process (Davis, 1910; Fetterolf & Snyder, 2012; Regulski
et al., 2013). In one such study, Snyder et al. (2016) demonstrated that
when patients are treated with either the standard of care or a
dehydrated amniotic membrane allograft (DAMA) in addition to stan-
dard of care that a higher incidence of wound closure occurs in
patients treated with DAMA.
Available CTPs range from decellularized collagen-based sub-
strates to those derived by harvesting tissue from donor humans (allo-
graft/autograft) or animals (xenograft) (Cornwell et al., 2009; Jones
et al., 2002). Allografts of interest are derived from human placental
tissue because of their relative ease of sourcing and composition that
includes a range of functional biological molecules that aid in wound
healing and host-tissue integration (Akle et al., 1981; Bailo
et al., 2004; Erickson & Couchman, 2000; Koob et al., 2013). These
attributes have made these membranes a highly valued tissue source
for use in wound management. Furthermore, these placental derived
membranes have shown favorable biocompatibility due to their low
immunogenicity (Ueta et al., 2002), anti-inflammatory functions (Hao
et al., 2000), and minimal processing. Furthermore, these tissues are
harvested without the ethical concerns associated with human embry-
onic tissues and use of animal sources (Chen & Tofe, 2010; Liu
et al., 2010; Toda et al., 2007). Human placental membranes provide
an abundance of useful components including membrane constitu-
ents, ECM components such as collagen (Parolini et al., 2012), and
growth factors and cytokines which are all critical to support and
stimulate cell growth. When used in chronic wounds such as diabetic
foot ulcers, membranes such as dehydrated amniotic allografts have
been shown to have a positive stimulatory effect on wounds stalled in
the inflammatory phase (Abdo, 2016; Barr, 2014; Lintzeris
et al., 2015; Rosenblum, 2016; Snyder et al., 2016). They contain key
interleukins (ILs), IL-4, 6, 8, and 10, which regulate cell function, act as
chemokines, and stimulate cell proliferation (Kindt et al., 2006). IL-10
further serves an anti-inflammatory role by down-regulating MHC-II
antigens and counteracting other inflammatory factors in the process
of tissue repair (Mosser & Zhang, 2008; Weber & Iacono, 1996).
The objective of this study was to characterize the in vitro cellular
response underlying the observed clinical effects of DAMA in chronic
wounds. Specifically, biochemical and biophysical analyses were per-
formed to verify the retention of structural and biochemical compo-
nents of amniotic membranes post-processing, including the
quantification and identification of growth factors and cytokines. Fur-
thermore, the modulatory effect of these membranes on cell migra-
tion, proliferation, ECM deposition, and inflammation was evaluated
in vitro using human adult dermal fibroblasts.
2|MATERIALS AND METHODS
2.1 |Dehydrated amniotic membrane allograft
DAMA (AmnioExcel, Integra LifeSciences, Princeton, NJ) single layer
amniotic membranes were donated from Integra LifeSciences for
characterization studies. Placental tissues were donated by consenting
women and retrieved at the time of planned Cesarean section during
childbirth. In accordance with the US Food and Drug Administration
(FDA) and the American Association of Tissue Banks (AATB) guide-
lines, placental tissue donors were screened for communicable dis-
eases before processing. DAMA processing and dehydration steps
were completed within a few hours to maintain the tissues distinct
biochemical composition and handling properties.
2.2 |ELISA array
Growth factor content and concentrations were quantified for DAMA
membranes using enzyme-linked immunosorbent assay (ELISA,
RayBiotech Inc.). Membrane samples were weighed, homogenized in
lysis buffer containing protease inhibitors for 24 h at 4C, homoge-
nized again and then centrifuged to remove residual tissue fragments.
Supernatant extracts were used as samples in the ELISA kit assays.
2.3 |Histology
DAMA membranes were rehydrated in 1×PBS for 1 h. The samples
were subsequently fixed in formalin for 24 h, paraffin embedded, sec-
tioned, and stained with Hematoxylin and Eosin (H&E). Imaging was
performed on a Leica Confocal microscope at magnifications of 20×.
MOORE ET AL.3077
15524981, 2020, 8, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/jbm.b.34635 by University Of Florida, Wiley Online Library on [13/12/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Collagen type I, collagen type III, fibronectin, and laminin were all sta-
ined by immunohistochemistry (IHC) using formalin fixed paraffin
embedded sections. Briefly, slides were dried overnight, baked at
60C for 1 h, deparaffinized in xylene, rinsed in alcohol, rehydrated in
water, and equilibrated in wash buffer (TRIS buffered saline with
0.05% Tween 20; Dako, K8007). Collagen type I stain was carried out
using a rabbit polyclonal antibody directed against Collagen type I
from human placenta (Bio-Rad, 2,1500020). Collagen type III stain
was carried out using a rabbit polyclonal antibody directed against
Collagen type III from human placenta (Bio-Rad, 2,1500100). Fibro-
nectin stain was carried out using a rabbit polyclonal antibody raised
against Fibronectin isolated human plasma (Abcam, ab6584). Laminin
stain was carried out using a rabbit polyclonal antibody isolated from
the rat yolk sac tumor cell line L2 (Agilent, Z0097). Next a serum free
protein block was applied for 5 min (Dako, X0909) followed by anti-
Rabbit labelled Polymer-HRP (Dako, K4003) for 30 min and DAB+
Chromogen solution (Dako, K4011) for 5 min. Tissues were counter-
stained in a modified Harris hematoxylin (Dako, S3301) for 5 min.
Also, for collagen type I, collagen type III, laminin, and fibronectin,
human skin tissue was stained (not shown) to validate the specificity
of the antibodies and staining protocols.
2.4 |Extraction protocol
Briefly, 5 cm
2
of DAMA was homogenized (Precellys, Bertin Instru-
ments) in 1 ml of Fibroblast basal media (ATCC) for 4 min. This solu-
tion was then incubated at 37C for 72 h shaking at 300 rpm. Extract
was centrifuged and sterile filtered using a 0.22 μm PVDF filter
(Millipore). In these experiments, 5 cm
2
of DAMA membrane in 1 ml
of media correlates to 100% extract.
2.5 |Cell culture and ECM stain
Passage 4 human adult dermal fibroblasts (hDF, ATCC) were seeded at
5×10
3
cellsperwellin24-wellplatesandculturedin(a)assaymedia
with 1% fetal bovine serum (FBS) for the negative () control or (b) 15%
DAMA extract in assay media for 7 days or (c) complete growth media
with 10% FBS as a positive (+) control. Samples were fixed in 4% parafor-
maldehyde for 20 min, nuclei were stained blue with Hoescht 33342
(Thermo Fisher Scientific), and collagen type I was stained with a mono-
clonal rabbit collagen type I antibody (Invitrogen PA1-26204) and Alexa
Fluor 488 goat anti-rabbit secondary antibody (Thermo Fisher Scientific).
2.6 |Cellular migration assays
2.6.1 |Boyden chamber assay
Passage 5 hDF were seeded at a density of 2 ×10
4
cells per well in the
top insert of a 24-well Transwell plate (pore size 8.0 μm, Corning
Costar) and were serum starved overnight. The bottom wells contained
basal media (control), treatment groups in basal media: 15 or 25%
DAMA extract, or complete growth media with 10% FBS (+ control).
The cells were then incubated for 24 h. After incubation, the bottom
side of the Transwell inserts were washed with PBS and fixed in 4%
paraformaldehyde for 20 min and stained with Hoechst 33342. Tile
scan images of 200 ×200 μm were acquired and migrated cells were
counted using ImageJ software (National Institutes of Health).
2.6.2 |Wound healing scratch assay
A wound healing scratch assay was performed to visualize cell migra-
tion. Passage 4 hDF were seeded at 7 ×10
4
cells per well into a
culture-insert 2 Well24 well plate (Ibidi) and allowed to attach over-
night. 24 h later, the inserts were removed, cells were stained with
Hoescht 33342, and treatments were applied to the wells. Treatment
groups included basal media (control), 15% DAMA extract in basal
media, 25% DAMA extract in basal media, and complete growth
media containing 10% FBS (+ control). Images were then taken at 0, 6,
20, and 27 h and analyzed with ImageJ software.
2.7 |Cellular proliferation assays
2.7.1 |BrDU cell proliferation assay
Passage 3 hDF were seeded at a density of 2.5 ×10
3
cells per cm
2
in
a 96-well plate. Treatment groups included: basal media with 1% FBS
(control), 15 and 25% DAMA extract in basal media with 1% FBS, or
complete media with 10% FBS (+ control). After 24 h incubation with
treatment groups, BrdU reagent (BrdU Cell Proliferation ELISA, Roche)
was added to the wells and incubated for 24 h. Following incubation,
BrdU ELISA was performed according to the manufacturer's protocol
and plates were assayed in a plate reader (SpectraMax M3, Molecular
Device) at an absorbance of 450 and 690 nm.
2.7.2 |CyQuant cell proliferation assay
Passage 4 hDF (2 ×10
3
cells per cm
2
) were seeded in a 96-well plate
along with treatment groups: basal media with 1% FBS (control), 15%
DAMA extract in basal media with 1% FBS or complete media with
10% FBS (+ control). Cells were cultured for up to 7 days with media
changes every 2 days. Timepoints were harvested at days 1, 3, and
7. CyQuant reagent (Thermo Fisher Scientific) was added to each well,
and the plates were read on a plate reader (SpectraMax M3) with fluo-
rescence setting at 480 nm excitation and 520 nm emission.
2.8 |Cell attachment and adhesion
Passage 4 hDF (5 ×10
4
cells per well) were seeded directly on DAMA
membranes (12 mm in diameter) and were cultured in basal media
3078 MOORE ET AL.
15524981, 2020, 8, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/jbm.b.34635 by University Of Florida, Wiley Online Library on [13/12/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
with 1% FBS for 7 days. Cell seeded membranes were harvested at
days 1, 3, and 7 for image analysis. The membranes were washed with
PBS, fixed in 4% paraformaldehyde for 20 min and stained with
Hoechst 33342 and Alexa Fluor 488 Phalloidin for actin cytoskeleton
(Thermo Fisher Scientific).
2.9 |Inflammation inhibition assay
Human Peripheral blood mononuclear cells (PBMCS) were seeded at
1×10
5
cells per well in a 96-well plate and incubated for 1 h. Treat-
ment groups were incubated with PBMCS for 1 h and included 1 μM
Dexamethasone in RPMI media (control), RPMI media with 10%
FBS (+ control), and 25% DAMA extract in RPMI media. PBMCs were
then stimulated with 100 pg/ml lipopolysaccharide (LPS from E. coli
bacteria) for 24 h. After 24 h, supernatants were collected for quanti-
fying TNF-α, IL-1β, and IL-6 levels using Luminex ELISA assay
(Millipore).
3|RESULTS
Growth factors, TIMPs, and cytokine concentrations in single layer
amniotic membranes were evaluated by use of a multiplex ELISA kit.
Some of the more notable factors present in the DAMA membranes
are bFGF, EGF, PDGF-AA, PDGF-BB, TGF-β, IL-6, IL-8, TIMP-1, and
TIMP-2 (Table 1).
Histological analysis was performed to visualize the membranes
as shown in Figure 1. An H&E stain reveals a single layer of amnion
with flattened epithelial cells. The ECM fibrous proteins appear dense
and compact since the membrane undergoes a dehydration step
during manufacturing. Immunostaining of DAMA membranes highlight
the presence of extracellular matrix proteins collagen type I, collagen
type III, fibronectin, and laminin.
Wound healing progression relies on the recruitment and prolifer-
ation of cells. Cellular recruitment and function are essential for the
deposition of new ECM proteins, signaling the recruitment of other
cells, angiogenesis, and regulation of inflammation. As shown in
Figure 2, fibroblast migration through a Transwell culture system
(Figure 2a,b) increases with the increasing concentration of DAMA
extracts (p< .05 for 25% DAMA vs () control). Similarly, the scratch
assay (Figure 2c,d), visually demonstrates increasing fibroblast migra-
tion in the wells containing DAMA extract compared to the () control
group (p< .05 for both 15 and 25% DAMA).
When cells migrate into a wound bed, it is important for them to
be able to proliferate to enable new tissue development. Figure 3
demonstrates that DAMA extracts influence fibroblast proliferation.
Furthermore, with increasing concentrations of extract, fibroblast pro-
liferation responds accordingly. Treatment with 15 or 25% DAMA sig-
nificantly increases fibroblast proliferation when compared to the ()
control group (p< .0001) as detected by the BrDU cell proliferation
assay (Figure 3a). In addition, DAMA supports fibroblasts viability and
growth over 7 days in culture as shown in Figure 3b. Immunostained
images of cell-seeded DAMA membranes confirmed fibroblast attach-
ment onto the membranes and proliferation.
Cells and ECM interact in a synergistic manner with the matrix
providing a favorable environment for cell attachment, growth, prolif-
eration, and differentiation; in turn, the cells are able to secrete and
TABLE 1 Relative concentrations of growth factors present in
DAMA membranes as detected by ELISA
Concentration (pg/mg)
bFGF 1,113 ± 845
EGF 42.1 ± 7.1
G-CSF 1.99 ± 1.75
PDGF-AA 234.1 ± 15.2
PDGF-BB 38.3 ± 1.7
PLGF 16.9 ± 5.8
TGF-α1.9 ± 0.4
TGF-β1 41.5 ± 10.4
IL-4 0.7 ± 1.1
IL-6 1.71 ± 0.9
IL-8 10.7 ± 0.09
IL-10 0.33 ± 0.08
TIMP-1 4,746 ± 369
TIMP-2 841 ± 19
TIMP-4 16.1 ± 2.3 FIGURE 1 DAMA membrane was fixed and stained with H&E,
collagen type I, collagen type III, fibronectin, and laminin
MOORE ET AL.3079
15524981, 2020, 8, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/jbm.b.34635 by University Of Florida, Wiley Online Library on [13/12/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
remodel the ECM. Figure 4 demonstrates that, in the presence of
DAMA extract, cells proliferate and secrete collagen type I. The cell
proliferation for groups with DAMA extract significantly increased as
compared to the () control group over 7 days in culture (p< .05). Col-
lagen type I levels increased by 7 days as confirmed by immuno-
staining (Figure 4a).
FIGURE 2 Evaluation of DAMA effect on fibroblast cell migration. (a, b) Transwell migration analysis of hDF stained with Hoechst after
migrating for 24 h (One-way ANOVA, *p< .05, ***p< .0002). (c) A scratch assay showing analysis of hDF migration into a void space at 0, 6,
20, and 27 h (Two-way ANOVA *p< .05)
FIGURE 3 Treatment of hDF
with 15 and 25% DAMA
increases cell proliferation as
detected by BrDU Cell
Proliferation Assay (One-way
ANOVA, ****p< .0001). (b) hDF
seeded on the surface of DAMA
membranes and cultured in basal
media for 1, 3, and 7 days and
stained for actin cytoskeleton
(green) and nuclei (blue)
FIGURE 4 Treatment of hDF with 15% DAMA increases ECM secretion and cell proliferation as detected by (a) Hoescht for nuclei (blue) and
collagen type I (green) stains at 7 days. (b) A DNA CyQUANT assay shows the cell number increases over time (Two-way ANOVA, **p< .01,
***p< .0002, ****p< .0001)
3080 MOORE ET AL.
15524981, 2020, 8, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/jbm.b.34635 by University Of Florida, Wiley Online Library on [13/12/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Chronic wounds persist with no wound resolution due to an ele-
vated inflammatory response within and surrounding the wound bed.
As demonstrated in Figure 5, a 25% DAMA extract significantly
decreased the level of pro-inflammatory cytokines secreted by LPS
stimulated PBMCs: TNF-α, IL-6 and IL-1β(p< .05 for cytokines). Spe-
cifically, IL-6 and TNF-αlevels were reduced by 83% and IL-1βlevels
were reduced by 70% when normalized to the (+) control group. It has
been shown that when these markers are down regulated with pla-
cental membranes, chronic wounds have a higher degree of wound
closure (Berven et al., 2010).
4|DISCUSSION
Chronic wounds, such as diabetic foot ulcers, that persist in the
inflammatory phase of healing can lead to morbidity and mortality if
not addressed (Ellis et al., 2018). Clinical application of DAMA was
previously shown to be beneficial for managing chronic diabetic foot
ulcers. Furthermore, one case series showed that wounds treated with
DAMA have a 60% decrease in wound size after the first application
(Boyar & Galiczewski, 2018). As previously presented by Snyder
et al. (2016), DAMA accelerates diabetic foot ulcer wound closure
with 45% of patients achieved complete wound closure as compared
to 0% of the wounds treated with the standard of care having closure
by 6 weeks. We hypothesize that DAMA aides in the wound healing
process by releasing growth factors, which then regulates cell behav-
ior in the wound microenvironment. The objective of this study was
to characterize growth factor content in DAMA and their ability to
influence in vitro cell functions specific to wound healing. The results
demonstrate that DAMA consisted of multiple growth factors and
cytokines, which are known to participate in the wound healing pro-
cess. In addition, DAMA is a bioactive membrane that positively
impacts hDF activity in vitro.
In normal wound healing, there is an interplay between several
cell types including fibroblasts, keratinocytes, and inflammatory cells
along with the surrounding ECM, also known as dynamic reciprocity.
These cells interact by secreting growth factors, cytokines, and ECM
that guide tissue remodeling through several different phases. DAMA
is a rich source of cytokines and growth factors as demonstrated by
ELISA results. Some of the factors present in these membranes
providing clinical benefits include EGF, bFGF, PDGFs, TGF-α, and
TIMPs 1 and 2. EGF has a role in the stimulation, proliferation, and
migration of many different cell types including hDF, keratinocytes,
and endothelial cells and has been shown to accelerate healing
(Brown et al., 2010). bFGF has multiple biochemical and biological
roles including angiogenesis, proliferation and migration of many cell
types, stimulation of collagen synthesis, and epithelialization (Grazul-
Bilska et al., 2003). PDGF is involved in all stages of wound healing
and has a role in the chemotaxis of hDF and smooth muscle cells,
stimulation of collagen and glycosaminoglycans production from fibro-
blasts, and signals to other cells including the keratinocytes in the epi-
thelial layer (Koob et al., 2014b; Lynch et al., 1987). TIMP 1 and
2 both have a role in regulating several matrix metalloproteinases
(MMPs). MMPs are involved in many phases of wound healing such
as epithelialization and inflammatory responses and can act in both a
positive and negative manner during wound healing (Gill &
Parks, 2008).
When wounds remain in the inflammatory phase, high levels of
upregulated cytokines persist in the wound bed such as: IL-6, IL-8,
IL-1 β, and TNF-α. IL-6 promotes angiogenesis, however at high levels,
IL-6 has been known to delay re-epithelialization (Lin et al., 2003). In a
chronic wound TNF-αand IL-1βhave a synergistic effect in promoting
the release of MMPs and suppressing the synthesis of TIMPs and
ECM forming proteins (Barrientos et al., 2008; Xu et al., 2013). IL-8
expression aides in the stimulation of neutrophil migration which
induces an inflammatory response at the wound site. However, over
simulation of IL-8 can lead to destruction of healthy ECM by neutro-
phils, preventing wounds from entering the proliferative stage (Ellis
et al., 2018).
A reduction in pro-inflammatory cytokines has been linked to the
progression of chronic wounds through the healing process and ulti-
mately wound closure. High levels of pro-inflammatory cytokines
inhibit healing capabilities; thus, the management of these cytokines
is essential to the treatment of stalled wounds.
DAMA is shown to reduce pro-inflammatory cytokines IL-1β,
IL-6, and TNF-αand when reduced will allow the wound to progress
into the proliferation stage. In the proliferative stage, hDF will migrate
and proliferate, becoming the predominant cell type in the wound
(Stadelmann et al., 1998). Once in the wound bed, hDF secrete differ-
ent types of ECM such as collagens and GAGs to fill the wound bed
FIGURE 5 Reduction in pro-inflammatory cytokines secreted from PBMCs in the presence of 25% DAMA as compared to an LPS-treated
positive control (One-way ANOVA, **p< .002, ***p< .001, ****p< .0001)
MOORE ET AL.3081
15524981, 2020, 8, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/jbm.b.34635 by University Of Florida, Wiley Online Library on [13/12/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
while supporting epithelialization and angiogenesis (Dobaczewski
et al., 2010). Furthermore, as demonstrated in vitro, DAMA promotes
migration and cell proliferation of hDF which further supports the
wound transition from the inflammatory to proliferative phase. This
cascade of wound resolution occurs once the appropriate environ-
ment for wound healing is created and DAMA membranes have the
ability to restore favorable cellular responses for proper wound
healing.
Placental tissue membranes have gained traction in the chronic
wound space because of their ability to progress and resolve difficult
to heal wounds. As many studies have shown, using amniotic mem-
branes decreases scar tissue, reduces anti-inflammatory markers,
increases expression levels of angiogenic proteins, and can assist with
re-epithelialization (Adzick & Longaker, 1992; Hao et al., 2000; Mer-
met et al., 2007). These are all factors that have an influence in creat-
ing the proper balance of factors for proper wound healing (Tseng
et al., 1999). Unlike traditional collagen matrices, DAMA membranes
have a role in regulating the inflammatory response and assisting in
the progression of chronic wounds. While many factors that contrib-
ute to the wound healing benefits of amniotic membranes are dis-
cussed, future studies will investigate other possible factors of pro-
inflammatory cytokine reduction as well as differences with traditional
ECM scaffolds in a wound healing model.
CONFLICT OF INTEREST
Funding for this project was provided by Integra LifeSciences.
REFERENCES
Abdo, R. J. (2016). Treatment of diabetic foot ulcers with dehydrated
amniotic membrane allograft: A prospective case series. Journal of
Wound Care,25(Suppl. 7), S4S9. https://doi.org/10.12968/jowc.
2016.25.Sup7.S4
Adzick, N. S., & Longaker, M. T. (1992). Scarless fetal healing therapeutic
implications. Annals of Surgery,215(1), 330. https://doi.org/10.1097/
00000658-199201000-00004
Akle, C. A., Adinolfi, M., Welsh, K. I., Leibowitz, S., & McColl, I. (1981).
Immunogenicity of human amniotic epithelial cells after transplanta-
tion into volunteers. Lancet (London, England),2(8254), 10031005.
https://doi.org/10.1016/s0140-6736(81)91212-5
Bailo, M., Soncini, M., Vertua, E., Signoroni, P. B., Sanzone, S.,
Lombardi, G., Parolini, O. (2004). Engraftment potential of human
amnion and chorion cells derived from term placenta. Transplantation,
78(10), 14391448. https://doi.org/10.1097/01.TP.0000144606.
84234.49
Barr, S. M. (2014). Dehydrated amniotic membrane allograft for treatment
of chronic leg ulcers in patients with multiple comorbidities: A case
series. Journal of the American College of Clinical Wound Specialists,6(3),
3845. https://doi.org/10.1016/j.jccw.2016.01.002
Barrientos, S., Stojadinovic, O., Golinko, M. S., Brem, H., & Tomic-
Canic, M. (2008). Growth factors and cytokines in wound healing.
Wound Repair and Regeneration,16(5), 585601. https://doi.org/10.
1111/j.1524-475x.2008.00410.x
Berven, E., Freberg, A., Evangelista, M., & Soncini, M. (2010). Human pla-
centa: Structure and development, circulation, and functions. New York:
Nova Biomedical Books.
Boyar, V., & Galiczewski, C. (2018). Efficacy of dehydrated human amniotic
membrane allograft for the treatment of severe extravasation injuries
in preterm neonates. Wounds,30(8), 224228.
Broughton, G., Janis, J. E., & Attinger, C. E. (2006). The basic science of
wound healing. Plastic and Reconstructive Surgery,117(7S), 12S34S.
https://doi.org/10.1097/01.prs.0000225430.42531.c2
Brown, G. L., Nanney, L. B., Griffen, J., Cramer, A. B., Yancey, J. M.,
Curtsinger, L. J., III, Lynch, J. B. (2010). Enhancement of wound
healing by topical treatment with epidermal growth factor. The New
England Journal of Medicine,321(2), 7679. https://doi.org/10.1056/
nejm198907133210203
Chen, E., & Tofe, A. (2010). A literature review of the safety and biocom-
patibility of amnion tissue. Journal of Implant and Advanced Clinical
Dentistry,2(3), 6775. https://doi.org/10.1111/iwj.12097
Cornwell, K. G., Landsman, A., & James, K. S. (2009). Extracellular matrix
biomaterials for soft tissue repair. Clinics in Podiatric Medicine and Sur-
gery,26(4), 507523. https://doi.org/10.1016/j.cpm.2009.08.001
Cutting, K., & Tong, A. (2003). Wound physiology and moist wound healing.
Holsworthy: Medical Communication. https://doi.org/10.12968/indn.
2012.5.3.90107
Davis, J. (1910). Skin transplantation with a review of 550 cases at the
Johns Hopkins Hospital. The Johns Hopkins Medical Journal,15,
307396.
Dobaczewski, M., Gonzalez-Quesada, C., & Frangogiannis, N. G. (2010).
The extracellular matrix as a modulator of the inflammatory and repar-
ative response following myocardial infarction. Journal of Molecular
and Cellular Cardiology,48(3), 504511. https://doi.org/10.1016/j.
yjmcc.2009.07.015
Ellis, S., Lin, E. J., & Tartar, D. (2018). Immunology of wound healing. Cur-
rent Dermatology Report,7(4), 350358. https://doi.org/10.1007/
s13671-018-0234-9
Enoch, S., & Leaper, D. J. (2005). Basic science of wound healing. Surgery,
23(2), 3742. https://doi.org/10.1383/SURG.23.2.37.60352
Erickson, A. C., & Couchman, J. R. (2000). Still more complexity in mamma-
lian basement membranes. The Journal of Histochemistry and Cyto-
chemistry,48(10), 12911306. https://doi.org/10.1177/0022155400
04801001
Falanga, V. (2005). Wound healing and its impairment in the diabetic foot.
Lancet,366(9498), 17361743. https://doi.org/10.1016/S0140-6736
(05)67700-8
Fetterolf, D., & Snyder, R. (2012). Scientific and clinical support for the use
of dehydrated amniotic membrane in wound management. Wounds,
24(10), 114.
Gill, S., & Parks, W. (2008). Metalloproteinases and their inhibitors: Regula-
tors of wound healing. The International Journal of Biochemistry and Cell
Biology,40(67), 13341347. https://doi.org/10.1038/mp.2011.182.doi
Grazul-Bilska, A. T., Johnson, M. L., Bilski, J. J., Redmer, D. A.,
Reynolds, L. P., Abdullah, A., & Abdullah, K. M. (2003). Wound healing:
The role of growth factors. Drugs of Today,39(10), 787800. https://
doi.org/10.1358/dot.2003.39.10.799472
Hao, Y., Ma, D. H.-K., Hwang, D. G., Kim, W.-S., & Zhang, F. (2000). Identi-
fication of antiangiogenic and Antiinflammatory proteins in human
amniotic membrane. Cornea,19(3), 348352. https://doi.org/10.
1097/00003226-200005000-00018
Hutton M, Selvarajah D, Tesfaye S, Wilkinson I. Diffusion tensor imaging
tractography of the corticosomatosensory pathway in painless diabetic
neuropathy. Paper presented at the Proceedings of the 17th Scientific
Meeting on International Society of Magnetic Resonance in Medicine,
Honolulu. 2009;37(Suppl. 1):3401. doi:https://doi.org/10.1002/eji.
200737772.Historical
Jones, I., Currie, L., & Martin, R. (2002). A guide to biological skin substi-
tutes the function of normal skin. British Journal of Plastic Surgery,55,
185193. https://doi.org/10.1054/hips.2002.3800
Kindt, T. J., Goldsby, R. A., Osborne, B. A., & Kuby, J. (2006). Kuby immu-
nology (6th ed.). New York: W.H. Freeman.
Kirsner, R. S., & Eaglstein, W. H. (1993). The wound healing process. Der-
matologic Clinics,11(4), 629640. https://doi.org/10.1016/S0733-
8635(18)30216-X
3082 MOORE ET AL.
15524981, 2020, 8, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/jbm.b.34635 by University Of Florida, Wiley Online Library on [13/12/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
Koob, T. J., Rennert, R., Zabek, N., Massee, M., Lim, J. J., Temenoff, J. S.,
Gurtner, G. (2013). Biological properties of dehydrated human
amnion/chorion composite graft: Implications for chronic wound
healing. International Wound Journal,10(5), 493500. https://doi.org/
10.1111/iwj.12140
Koob, T. J., Lim, J. J., Massee, M., Zabek, N., Rennert, R., Gurtner, G., &
Li, W. W. (2014a). Angiogenic properties of dehydrated human
amnion/chorion allografts: Therapeutic potential for soft tissue repair
and regeneration. Vascular Cell,6(1), 110. https://doi.org/10.1186/
2045-824X-6-10
Koob, T. J., Lim, J. J., Massee, M., Zabek, N., & Denozière, G. (2014b).
Properties of dehydrated human amnion/chorion composite grafts:
Implications for wound repair and soft tissue regeneration. Journal of
Biomedical Materials Research Part B: Applied Biomaterials,102(6),
13531362. https://doi.org/10.1002/jbm.b.33141
Lin, Z.-Q., Kondo, T., Ishida, Y., Takayasu, T., & Mukaida, N. (2003). Essen-
tial involvement of IL-6 in the skin wound-healing process as
evidenced by delayed wound healing in IL-6-deficient mice. Journal of
Leukocyte Biology,73(6), 713721. https://doi.org/10.1189/jlb.
0802397.Journal
Lintzeris, D., Yarrow, K., Johnson, L., White, A., Hampton, A., Strickland, A.,
Cook, A. (2015). Use of a dehydrated amniotic membrane allograft
on lower extremity ulcers in patients with challenging wounds: A ret-
rospective case series. Ostomy Wound Management,61(10), 3036.
Liu, J., Sheha, H., Fu, Y., Liang, L., & Tseng, S. C. G. (2010). Update on amni-
otic membrane transplantation. Expert Review of Ophthalmology,5(5),
645661.
Lynch, S. E., Nixontt, J. O. N. C., Colvin, R. B., & Antoniades, H. N. (1987).
Role of platlet-derived growth factor in wound healing: Synergistic
effects with other growth factors. Proceedings of the National Academy
of Sciences USA,84(21), 76967700.
McQuilling, J. P., Vines, J. B., & Mowry, K. C. (2017). In vitro assessment of
a novel, hypothermically stored amniotic membrane for use in a
chronic wound environment. International Wound Journal,14(6),
9931005. https://doi.org/10.1111/iwj.12748
Mermet, I., Pottier, N., Sainthillier, J. M., Malugani, C., Cairey-
Remonnay, S., Maddens, S., Aubin, F. (2007). Use of amniotic mem-
brane transplantation in the treatment of venous leg ulcers. Wound
Repair and Regeneration,15(4), 459464. https://doi.org/10.1111/j.
1524-475X.2007.00252.x
Mosser, D. M., & Zhang, X. (2008). Interleukin-10: New perspectives on an
old cytokine. Immunological Reviews,226, 205218. https://doi.org/
10.1111/j.1600-065X.2008.00706.x.Interleukin-10
Olczyk, P., Mencner, Ł., & Komosinska-Vassev, K. (2014). The role of the
extracellular matrix components in cutaneous wound healing. BioMed
Research International,2014,18. https://doi.org/10.1155/2014/
747584
Parolini, O., Solomon, A., Evangelista, M., & Soncini, M. (2012). Human
term placenta as a therapeutic agent: From the first clinical applications
to future perspectives. Hauppauge, NY: Nova Biomedical Books.
Regulski, M., Jacobstein, D. A., Petranto, R. D., Migliori, V. J., Nair, G., &
Pfeiffer, D. (2013). A retrospective analysis of a human cellular repair
matrix for the treatment of chronic wounds. Ostomy/Wound Manage-
ment,59(12), 3848.
Rosenblum, B. I. (2016). A retrospective case series of a dehydrated amni-
otic membrane allograft for treatment of unresolved diabetic foot
ulcers. Journal of the American Podiatric Medical Association,106(5),
328337. https://doi.org/10.7547/15-139
Schultz, G. S., Davidson, J. M., Kirsner, R. S., Bornstein, P., & Herman, I. M.
(2011). Dynamic reciprocity in the wound microenvironment. Wound
Repair and Regeneration,19(2), 134148. https://doi.org/10.1111/j.
1524-475X.2011.00673.x
Sheikh, E. S., Sheikh, E. S., & Fetterolf, D. E. (2014). Use of dehydrated
human amniotic membrane allografts to promote healing in patients
with refractory non healing wounds. International Wound Journal,11
(6), 711717. https://doi.org/10.1111/iwj.12035
Snyder, R. J., Shimozaki, K., Tallis, A., Kerzner, M., Reyzelman, A.,
Lintzeris, D., Rosenblum, B. (2016). A prospective, randomized, mul-
ticenter, controlled evaluation of the use of dehydrated amniotic
membrane allograft compared to standard of care for the closure of
chronic diabetic foot ulcer. Wounds: A Compendium of Clinical Research
and Practice,28(3), 7077.
Stadelmann, K., Digenis, A., & Tobin, G. (1998). Physiology and healing
dynamics of chronic cutaneous wounds. American Journal of Surgery,
176(2), 26S38S. https://doi.org/10.1016/S0002-9610(98)00183-4
Stojanovic, S., Tijanic, M., Jovanovic, G., Spasic, M., Stojkovic, B.,
Petrovic, M., B., & Dencic, T. (1996). The role of growth factors in
wound healing. The Journal of Trauma Injury Infection and Critical Care,
41(1), 159167. https://doi.org/10.5937/asn1572524s
Toda, A., Okabe, M., Yoshida, T., & Nikaido, T. (2007). The potential of
amniotic membrane/amnion-derived cells for regeneration of various
tissues. Journal of Pharmacological Sciences,105(3), 215228. https://
doi.org/10.1254/jphs.cr0070034
Tseng, S. C. G., Li, D.-Q., & Ma, X. (1999). Suppression of transforming
growth factor-beta isoforms, TGF-beta receptor type II, and
myofibroblast differentiation in cultured human corneal and limbal
fibroblasts by amniotic membrane matrix. Journal of Cellular Physiology,
179(3), 325335. https://doi.org/10.1002/(SICI)1097-4652(199906)
179:3<325::AID-JCP10>3.0.CO;2-X
Ueta, M., Kweon, M. N., Sano, Y., Sotozono, C., Yamada, J., Koizumi, N.,
Kinoshita, S. (2002). Immunosuppressive properties of human amniotic
membrane for mixed lymphocyte reaction. Clinical and Experimental
Immunology,129(3), 464470. https://doi.org/10.1046/j.1365-2249.
2002.01945.x
Velnar, T., Bailey, T., & Smrkolj, V. (2009). The wound healing process: An
overview of the cellular and molecular mechanisms. The Journal of
International Medical Research,37(5), 15281542. https://doi.org/10.
1177/147323000903700531
Weber, R. L., & Iacono, V. J. (1996). The cytokines: A review of interleu-
kins. Periodontal Clinical Investigations,19(1), 1722.
Werner, S., & Grose, R. (2017). Regulation of wound healing by growth
factors and cytokines. Physiological Reviews,83(3), 835870. https://
doi.org/10.1152/physrev.2003.83.3.835
Xu, F., Zhang, C., & Graves, D. T. (2013). Abnormal cell responses and role
of TNF-αin impaired diabetic wound healing. BioMed Research Interna-
tional,2013,19. https://doi.org/10.1155/2013/754802
Zhang, L. (2010). Glycosaminoglycan (GAG) biosynthesis and GAG-binding
proteins. Progress in Molecular Biology and Translational Science,93,
117. https://doi.org/10.1016/S1877-1173(10)93001-9
How to cite this article: Moore MC, Bonvallet PP,
Damaraju SM, Modi HN, Gandhi A, PS McFetridge. Biological
characterization of dehydrated amniotic membrane allograft:
Mechanisms of action and implications for wound care.
J Biomed Mater Res. 2020;108B:30763083. https://doi.org/
10.1002/jbm.b.34635
MOORE ET AL.3083
15524981, 2020, 8, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/jbm.b.34635 by University Of Florida, Wiley Online Library on [13/12/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
... [9][10][11][12] Some of the marked features of these growth factors are their ability to influence proliferation, migration, and ECM secretion from cells such as fibroblasts, in addition to support angiogenic, anti-inflammatory, and immunomodulatory functions. [13][14][15][16] Clinical case studies utilizing placental membranes for up to 8 weeks in burns and complex wounds as a biological dressing have demonstrated decreased in-flammation, reduced pain and scarring, and enhanced re-epithelialization. 17,18 The inherent bioactive properties and favorable clinical findings further reinforce the therapeutic potential of placental membranes as a therapy for chronic wound healing and tissue repair. ...
... 24 Other studies have investigated the in vitro biological efficacy of amnion alone and amnion/chorion membranes using different cell lines to demonstrate wound healing applications. 9,16,25,26 However, the biophysical properties of these various membranes can limit their clinical utility. When hydrated, a placental membrane often becomes self-adherent and lacks mechanical rigidity that results in compromised handling properties and is difficult to position on a wound bed. ...
... Histological procedures have been described previously in detail. 16 Briefly, three different lots of TPAM were rehydrated in phosphate buffered saline (PBS) before histology processing. TPAM was then embedded in paraffin and sectioned. ...
Article
Full-text available
Objective: Placental tissues, including membranes composed of amnion and chorion, are promising options for the treatment of chronic wounds. Amnion and chorion contain multiple extracellular matrix (ECM) proteins as well as a multitude of growth factors and cytokines that, when used clinically, assist in the progression of difficult to heal wounds through restoration of a normal healing process. The objective of this study was to characterize the in vitro physical and biological properties of a dehydrated tri-layer placental allograft membrane (TPAM) consisting of a chorion layer sandwiched between 2 layers of amnion. Approach: Mechanical properties were evaluated by mechanical strength and enzyme degradation assays. The ECM composition of TPAM membranes was evaluated by histological staining while growth factors and cytokine presence was evaluated by a multiplex ELISA. Proliferation, migration, and ECM secretion assays were performed with fibroblasts. Immunomodulatory properties were assessed by a pro-inflammatory cytokine reduction assay while the macrophage phenotype was determined by quantifying the ratio of M1 vs. M2 secreted factors. Results: The unique three-layer construction improves mechanical handling properties over single- and bi-layer membranes. Results demonstrate that TPAM is rich in ECM proteins, growth factors, cytokines, and tissue inhibitors of metalloproteinases, and favorably influences fibroblast migration, proliferation, and ECM secretion when compared to negative controls. Furthermore, after processing and preservation, these membranes maintain their intrinsic immunomodulatory properties with the ability to suppress pro-inflammatory processes as well as modulate the M1 and M2 macrophage phenotype towards a pro-regenerative profile when compared to a negative control. Innovation: This is the first study to characterize both the biophysical and biological properties of a tri-layer placental membrane. Conclusion: This work demonstrates that TPAM has improved handling characteristics over single- and bi-layer membranes, stimulates pro-healing cellular responses, and advantageously modulates inflammatory responses, altogether making this scaffold a promising option for treating wounds, especially those which are complex or difficult to heal.
... However, the specific structure and composition of dHACMs, including the presence of amnion or chorion only, or both, along with various matrix components, growth factors and cytokines, depend on the processing methods employed. [16][17][18][19][20][21][22] Generally, the amnion and chorion from donated human placentas are isolated through a cleansing proprietary process and either processed separately or laminated to form a two-layer graft, before being dehydrated under controlled drying conditions. 23 All of the dHACMs that have been utilised in clinical trials to date lack the spongy layer, which bridges the amnion and chorion sides and, thus, is also referred to as an intermediate layer. ...
Article
Full-text available
We investigated the healing effect of a new dehydrated amnion/chorion membrane with a spongy layer over a 30-month period in 32 patients with 53 chronic non-healing wounds of different aetiologies. Wounds with <40% surface reduction after 4 weeks of best wound treatment underwent weekly allograft application by a certified wound specialist based on national guidelines and a standardised protocol until complete healing or definite treatment interruption. The main outcome measure was the percentage of wound surface reduction from baseline calculated using digital planimetry follow-up photographs. Overall, 38 (71.7%) wounds presented a favourable outcome (70%-100% area reduction), with 35 (66%) completely healing over a median time of 77 days (range 29-350 days). Favourable outcomes were observed in 75% of traumatic wounds, surgical wounds, venous leg ulcers and pressure injuries, as well as in 50% of ischaemic wounds. Wounds being present <12 months were significantly more likely to have a favourable outcome than more long-standing wounds (χ 2 = 7.799; p = 0.005; OR = 3.378; 95% CI, 1.410-8.092). Thus, treatment with dehydrated amnion/chorion membrane with a spongy layer improves the outcome of non-healing wounds of different aetiologies and, therefore, has to be considered early in the management of refractory wounds. K E Y W O R D S amniotic membrane, chronic non-healing wounds, dehydrated amnion/chorion membrane, intermediate layer, spongy layer Key Messages • the clinical significance of utilising human dehydrated amnion/chorion membrane allograft with an intact spongy layer for the treatment of chronic wounds has not been thoroughly investigated • thirty-two patients with 53 chronic non-healing wounds of different aetiol-ogies underwent weekly application of a new dehydrated amnion/chorion
... Amnion is often used solely or together with chorion to increase the thickness and the bioactive contents of the graft (Koob et al., 2015). The possible inside mechanisms were indicated to be the bioactive factors contained within amnion and chorion tissues, including growth factors, cytokines, chemokines, and matrix metalloproteinases (MMPs) (Choi et al., 2009;Rodriguez-Ares et al., 2009;Koob et al., 2013;Koob et al., 2014;Koob et al., 2015;Moore et al., 2020). ...
Article
Full-text available
Allogeneic amnion tissues have been widely used in tissue repair and regeneration, especially a remarkable trend of clinical uses in chronic wound repair. The virus inactivation procedures are necessary and required to be verified for the clinical use and approval of biological products. Cobalt-60 (Co-60) or electron-beam (e-beam) is the common procedure for virus and bacterial reduction, but the excessive dose of irradiation was reported to be harmful to biological products. Herein, we present a riboflavin (RB)-ultraviolet light (UV) method for virus inactivation of amnion and chorion tissues. We used the standard in vitro limiting dilution assay to test the viral reduction capacity of the RB-UV method on amnion or chorion tissues loaded with four types of model viruses. We found RB-UV was a very effective procedure for inactivating viruses of amnion and chorion tissues, which could be used as a complementary method to Co-60 irradiation. In addition, we also screened the washing solutions and drying methods for the retention of growth factors.
... In this study, the PCL-amnion nanofibrous membrane consists of PCL in the outer layer and amniotic membrane in the inner layer. The amniotic membrane can slowly release TGF-β1, bFGF, PDGF, and NGF cytokines (52). These cytokines continuously diffuses through the pore structure of PCL to the nerve repair site and promotes the proliferation of Schwann cells and axon regeneration. ...
Article
Full-text available
Functional recovery after peripheral nerve injury repair is typically unsatisfactory. An anastomotically poor microenvironment and scarring at the repair site are important factors impeding nerve regeneration. In this study, an electrospun poly-e-caprolactone (PCL)-amnion nanofibrous membrane comprising an amnion membrane and nonwoven electrospun PCL was used to wrap the sciatic nerve repair site in the rat model of a sciatic nerve transection. The effect of the PCL-amnion nanofibrous membrane on improving nerve regeneration and preventing scarring at the repair site was evaluated by expression of the inflammatory cytokine, sciatic functional index (SFI), electrophysiology, and histological analyses. Four weeks after repair, the degree of nerve adhesion, collagen deposition, and intraneural macrophage invasion of the PCL-amnion nanofibrous membrane group were significantly decreased compared with those of the Control group. Moreover, the PCL-amnion nanofibrous membrane decreased the expression of pro-inflammatory cytokines such as interleukin(IL)-6, Tumor Necrosis Factor(TNF)-a and the number of pro-inflammatory M1 macrophages, and increased the expression of anti-inflammatory cytokine such as IL-10, IL-13 and anti-inflammatory M2 macrophages. At 16 weeks, the PCL-amnion nanofibrous membrane improved functional recovery, including promoting nerve Schwann cell proliferation, axon regeneration, and reducing the time of muscle denervation. In summary, the PCL-amnion nanofibrous membrane effectively improved nerve regeneration and prevent fibrosis after nerve repair, which has good clinical application prospect for tissue repair.
Article
Objective Hard-to-heal (chronic) wounds negatively impact patients and are a source of significant strain on the healthcare system and economy. These wounds are often resistant to standard of care (SoC) wound healing approaches due to a diversity of underlying pathologies. Cellular, acellular, and matrix-like products, such as amniotic membranes (AM), are a potential solution to these challenges. A growing body of evidence suggests that AM may be useful for treatment-resistant wounds; however, limited information is available regarding the efficacy of dehydrated amniotic membrane (DHAM) on multi-aetiology, hard-to-heal wounds. Therefore, we analysed the efficacy of DHAM treatment in reducing the size of hard-to-heal diabetic and venous leg ulcers (VLUs) that had failed to improve after SoC-based treatments. Method In this multicentre retrospective study, we analysed wound size during clinic visits for patients being treated for either diabetic or VLUs. During each visit, the treatment consisted of debridement followed by application of DHAM. Each wound was measured after debridement and prior to DHAM application, and wound volumes over time or number of DHAM applications were compared. Results A total of 18 wounds in 11 patients were analysed as part of this study. Wounds showed a significant reduction in volume after a single DHAM application, and a 50% reduction in wound size was observed after approximately two DHAM applications. These findings are consistent with reports investigating DHAM treatment of diabetic ulcers that were not necessarily resistant to treatment. Conclusion To our knowledge, this study is the first to directly compare the efficacy of standalone DHAM application to hard-to-heal diabetic and venous leg ulcers, and our findings indicate that DHAM is an effective intervention for resolving these types of wounds. This suggests that implementing this approach could lead to fewer clinic visits, cost savings and improved patient quality of life. Declaration of interest This research was supported in part by Merakris Therapeutics, US, and facilitated access to deidentified patient datasets, which may represent a perceived conflict of interest; however, the primary data analysis was performed by FSB who is unaffiliated with Merakris Therapeutics. TCB is a founder, employee of and shareholder in Merakris Therapeutics; WSF is a co-founder of, consultant for, and shareholder in Merakris Therapeutics, and was also supported by the National Institutes of Health National Center for Advancing Translational Sciences Clinical and Translational Science Awards Grant KL2 Scholars Program (KL2TR001441). The research was also supported through endowments to WSF from the University of Texas Medical Branch Mimmie and Hallie Smith Endowed Chair of Transplant Research and the John L Hern University Chair in Transplant Surgery.
Article
Full-text available
Biological tissues from various anatomical sources have been utilized for tissue transplantation and have developed into an important source of extracellular scaffolding material for regenerative medicine applications. Tissue scaffolds ideally integrate with host tissue and provide a homeostatic environment for cellular infiltration, growth, differentiation, and tissue resolution. The human amniotic membrane is considered an important source of scaffolding material due to its 3D structural architecture and function and as a source of growth factors and cytokines. This tissue source has been widely studied and used in various areas of tissue repair including intraoral reconstruction, corneal repair, tendon repair, microvascular reconstruction, nerve procedures, burns, and chronic wound treatment. The production of amniotic membrane allografts has not been standardized, resulting in a wide array of amniotic membrane products, including single, dual, and tri-layered products, such as amnion, chorion, amnion–chorion, amnion–amnion, and amnion–chorion–amnion allografts. Since these allografts are not processed using the same methods, they do not necessarily produce the same clinical responses. The aim of this review is to highlight the properties of different human allograft membranes, present the different processing and preservation methods, and discuss their use in tissue engineering and regenerative applications.
Preprint
Full-text available
Chronic wounds are associated with considerable patient morbidity and present a significant economic burden to the healthcare system. Often, chronic wounds are in a state of persistent in-flammation and unable to progress to the next phase of wound healing. Placental-derived bio-materials are recognized for their biocompatibility, biodegradability, angiogenic, an-ti-inflammatory, anti-microbial, anti-fibrotic, immunomodulatory, and immune privileged prop-erties. As such, placental-derived biomaterials have been used in wound management for more than a century. Placental-derived scaffolds are composed of an extracellular matrix (ECM) that can mimic the native tissue, creating a reparative environment to promote ECM remodeling, cell migration, proliferation, and differentiation. Reliable evidence exists throughout the literature to support the safety and effectiveness of placental-derived biomaterials in wound healing. How-ever, differences in source (i.e., anatomical regions of the placenta), preservation techniques, decellularization status, design, and clinical application have not been fully evaluated. This re-view provides an overview of wound healing and placental-derived biomaterials, summarizes the clinical results of placental-derived scaffolds in wound healing, and suggests directions for future work.
Article
Full-text available
Chronic wounds are associated with considerable patient morbidity and present a significant economic burden to the healthcare system. Often, chronic wounds are in a state of persistent inflammation and unable to progress to the next phase of wound healing. Placental-derived biomaterials are recognized for their biocompatibility, biodegradability, angiogenic, anti-inflammatory, antimicrobial, antifibrotic, immunomodulatory, and immune privileged properties. As such, placental-derived biomaterials have been used in wound management for more than a century. Placental-derived scaffolds are composed of extracellular matrix (ECM) that can mimic the native tissue, creating a reparative environment to promote ECM remodeling, cell migration, proliferation, and differentiation. Reliable evidence exists throughout the literature to support the safety and effectiveness of placental-derived biomaterials in wound healing. However, differences in source (i.e., anatomical regions of the placenta), preservation techniques, decellularization status, design, and clinical application have not been fully evaluated. This review provides an overview of wound healing and placental-derived biomaterials, summarizes the clinical results of placental-derived scaffolds in wound healing, and suggests directions for future work.
Article
Full-text available
Background: Phantom limb pain (PLP) complicates approximately 85% of patients who undergo major limb amputation. Studies consistently show that PLP significantly affects mobility, ability to perform tasks for daily living, and overall quality of life. Surgical treatments for PLP have been described in modern literature and do seem to offer promising results. In addition, recent studies have also described the use of placental connective tissue matrix (PCTM) to promote wound healing and nerve repairs. Methods: A retrospective chart review identified eights patients who underwent peripheral nerve reconstruction (PNR). This reconstruction included targeted muscle reinnervation (TMR) and wrapping of the nerve collections with PCTM, for chronic and debilitating PLP. Patient charts were reviewed for demographics, risk factors, current functional status, self-reported pre- and post-operative pain scale ratings, pre- and post-operative PROMIS scores for pain intensity, and vascular quality of life (QOL) scores. Results: Prior to PNR, each patient had undergone nerve block which relieved their PLP. Pre-operative and post-operative scores and values were recorded in each of the aforementioned categories. The numeric rating scale for pain assessment showed an average pre-operative pain score of 9.01 (range 8-10), post-operative score of 3.4 (range 1-6; p
Article
Full-text available
Because synthetic vascular prostheses perform poorly in small-diameter revascularization, biological vascular substitutes are being developed as an alternative. Although their in vivo results are promising, their production involves long, complex, and expensive tissue engineering methods. To overcome these limitations, we propose an innovative approach that combines the human amniotic membrane (HAM), which is a widely available and cost-effective biological raw material, with a rapid and robust textile-inspired assembly strategy. Fetal membranes were collected after cesarean deliveries at term. Once isolated by dissection, HAM sheets were cut into ribbons that could be further processed by twisting into threads. Characterization of the HAM yarns (both ribbons and threads) showed that their physical and mechanical properties could be easily tuned. Since our clinical strategy will be to provide an off-the-shelf allogeneic implant, we studied the effects of decellularization and/or gamma sterilization on the histological, mechanical, and biological properties of HAM ribbons. Gamma irradiation of hydrated HAMs, with or without decellularization, did not interfere with the ability of the matrix to support endothelium formation in vitro. Finally, our HAM-based, woven tissue-engineered vascular grafts (TEVGs) exhibited clinically relevant mechanical properties. Thus, this study demonstrates that human, completely biological, allogeneic, small-diameter TEVGs can be produced from HAM, thereby avoiding costly cell culture and bioreactors.
Article
Full-text available
Purpose of Review Chronic wounds are a tremendous burden on the healthcare system and lead to significant patient morbidity and mortality. Normal cutaneous wound healing occurs through an intricate and delicate interplay between the immune system, keratinocytes, and dermal cells. Each cell type contributes signals that drive the normal phases of wound healing: hemostasis, inflammation, proliferation, and remodeling. This paper reviews how various immunological cell types and signaling molecules influence the way wounds develop, persist, and heal. Recent Findings Concurrent with the achievement of hemostasis, neutrophils are the first cells to migrate to the wound bed, brought in by pro-inflammatory signals including IL-8. Their apoptosis and engulfment by macrophages (efferocytosis) provides a key signal to the local immune milieu, including macrophages, to transition to an anti-inflammatory, pro-repair state, where angiogenesis occurs and granulation tissue is laid down. Myofibroblasts, activated through contractile forces and signaling molecules, then drive remodeling, where granulation tissue becomes scar. Unchecked inflammation at this stage can result in abnormal scar formation. Summary Although the derangement of immune signals at any stage can result in impaired wound healing, recent research has shown that the key transition point lies between the inflammatory and the proliferative phases. This review summarizes the events that facilitate this transition and discusses how this process can be disrupted, leading to chronic, non-healing wounds.
Article
Full-text available
Chronic wounds require extensive healing time and place patients at risk of infection and amputation. Recently, a fresh hypothermically stored amniotic membrane (HSAM) was developed and has subsequently shown promise in its ability to effectively heal chronic wounds. The purpose of this study is to investigate the mechanisms of action that contribute to wound-healing responses observed with HSAM. A proteomic analysis was conducted on HSAM, measuring 25 growth factors specific to wound healing within the grafts. The rate of release of these cytokines from HSAMs was also measured. To model the effect of these cytokines and their role in wound healing, proliferation and migration assays with human fibroblasts and keratinocytes were conducted, along with tube formation assays measuring angiogenesis using media conditioned from HSAM. Additionally, the cell-matrix interactions between fibroblasts and HSAM were investigated. Conditioned media from HSAM significantly increased both fibroblast and keratinocyte proliferation and migration and induced more robust tube formation in angiogenesis assays. Fibroblasts cultured on HSAMs were found to migrate into and deposit matrix molecules within the HSAM graft. These collective results suggest that HSAM positively affects various critical pathways in chronic wound healing, lending further support to promising qualitative results seen clinically and providing further validation for ongoing clinical trials.
Article
Introduction: A peripheral intravenous (PIV) catheter is placed in 60% to 70% of neonatal intensive care unit (NICU) infants. Extravasation injuries occur in 18% to 33%, with 70% in neonates < 27 weeks of gestational age. Despite such frequent use of PIV therapy, evidence on best practice, injury prevention, management, and treatment of extravasations is lacking. Objective: This case series of 4 neonatal patients describes the experience and efficacy of using a dehydrated human amniotic membrane allograft (dHAMA) in the treatment of severe extravasation injuries. Materials and methods: The 4 preterm, critically ill neonates, all with stage 4 extravasations, were treated with 1 to 2 applications of the dHAMA to facilitate the repair process. Prior to treatments, standard of care included either enzymatic (collagenase ointment) or autolytic debridement (active Leptospermum honey) followed by mechanical debridement prior to allograft placement. Results: The 4 full-thickness wounds exhibited recalcitrant healing. The dHAMA invigorated the wounds after standard management failed to induce repair. Application was easy and follow-up care was minimal. All wounds healed without contractures and with minimal soft scars and normal pigmentation at the 1- to 2-month follow-up visits. Conclusions: The dHAMA proved to be an effective, safe, and easy-to-apply treatment in this case series, leading to regeneration and healing of deep neonatal wounds associated with extravasations.
Article
Objective: A diabetic foot ulcer (DFU) is one of the many potential complications associated with diabetes. If not effectively and rapidly treated, DFUs can result in lower extremity amputations. This prospective case series aimed to assess the effectiveness of a dehydrated amniotic-derived tissue allograft (DAMA), with regards to time to wound closure and total number of applications. Method: Patients were recruited with a neuropathic non-healing DFU(s) despite standard care for at least 4 weeks before the study. The number of DAMA applications and time between applications was based on the physician's judgment. For the majority of patients (n=13/14), offloading, usually total contact casting (TCC), was used in conjunction with DAMA. Wounds were assessed, measured, and photographed every 1-2 weeks. Results: Cases included 14 patients (11 men, 3 women; mean age 56.7±9.1 years) with 15 non-healing neuropathic DFUs with a mean baseline wound area of 6.5±11.6cm2 (median: 2.2cm2; range: 0.1-44.2cm2) and mean volume of 4.3±10.9cm3 (median: 0.3cm3; range: 0-39.8cm3). All patients in this series achieved complete wound closure within a median time of 5 weeks (range: 1-14 weeks). Wound area was reduced by a median of 58.3% at week 1 and 74.1% at week 3, and volume by a median of 62.8% at week 1, 97.4% at week 3 and by a median of 100% at week 5 and all time points thereafter. Patients received a median of 2 DAMA applications (range: 1-11). In those that required more than 1 application (n=12), DAMA was applied at intervals of 1 week (n=3) or ≥2 weeks (n=9). Smaller wounds (areas <2.2cm2) closed rapidly (<1 month, 1-2 applications), whereas larger wounds (>2.2cm2) required >2 weekly/biweekly applications. Conclusion: The use of DAMA, particularly when coupled with TCC, led to wound closure of DFUs in all patients in this case series, including complex patients with DFUs of ≥1 year in duration, lack of prior response to conservative treatment measures, area >10cm2 and/or multiple comorbidities. Prospective randomised trials would help to elucidate the precise role of DAMA in these encouraging results. Declaration of interest: Derma Sciences, Inc. (Princeton, NJ) funded editorial support services.
Article
Werner, Sabine, and Richard Grose. Regulation of Wound Healing by Growth Factors and Cytokines. Physiol Rev 83: 835–870, 2003; 10.1152/physrev.00032.2002.—Cutaneous wound healing is a complex process involving blood clotting, inflammation, new tissue formation, and finally tissue remodeling. It is well described at the histological level, but the genes that regulate skin repair have only partially been identified. Many experimental and clinical studies have demonstrated varied, but in most cases beneficial, effects of exogenous growth factors on the healing process. However, the roles played by endogenous growth factors have remained largely unclear. Initial approaches at addressing this question focused on the expression analysis of various growth factors, cytokines, and their receptors in different wound models, with first functional data being obtained by applying neutralizing antibodies to wounds. During the past few years, the availability of genetically modified mice has allowed elucidation of the function of various genes in the healing process, and these studies have shed light onto the role of growth factors, cytokines, and their downstream effectors in wound repair. This review summarizes the results of expression studies that have been performed in rodents, pigs, and humans to localize growth factors and their receptors in skin wounds. Most importantly, we also report on genetic studies addressing the functions of endogenous growth factors in the wound repair process.
Article
Over the past decade much has been elucidated concerning the cellular and molecular basis for health and disease. This increase in information has created a large gap in knowledge between the research scientist and the clinician. The following review attempts to bridge this gap by describing each of the thirteen interleukins and relating their functions to the clinical presentation of periodontal disease.
Article
Wound healing is a complex process that includes hemostasis, inflammation, proliferation and tissue remodeling. Growth factors are natural biological mediators that regulate crucial cellular processes involved in the tissue repair, such as DNA synthesis, angiogenesis, metabolic activity, migration, chemotaxis, proliferation, differentiation and matrix synthesis. The most important growth factors that play a part in the extraction-wound healing process and bone tissue regeneration are: platelet - derived growth factor-PDGF, transforming growth factor Beta-TGF β, insulin- like growth factor-IGF, bone morfogenetic protein-BMP-2, BMP-7, vascular endothelial growth factor- VEGF, fibroblast growth factor-FGF. Growth factors appear at various concentrations at different times, so that the wound age may be estimated by their age. Execpt wound healing growth factors may be used in better oseointegration of imlants, alveolar ridge augmentation, alveolit etc. Studies of physiological processes in which growth factors have a regulatory role indicate that these molecules rarely act in biological isolation. The study of the interaction between the growth factors in the alveolar bone can explain tissue ability to heal even under adverse conditions, such as infection and radiation. © 2015 Faculty of Medicine in Niš. Clinic of Dentistry in Niš. All rights reserved.
Article
Background: Foot ulcers are among the most serious complications of diabetes and can lead to amputation. Diabetic foot ulcers (DFUs) often fail to heal with standard wound care, thereby making new treatments necessary. This case series describes the addition of a dehydrated amniotic membrane allograft (DAMA) to standard care in unresolved DFUs. Methods: This is a single-center retrospective chart review of eight patients who had one to three applications of DAMA to nine DFUs that had failed to resolve despite offloading, other standard care, and adjuvant therapies. Following initial DAMA placement, wound size (length, width, depth) was measured every 1 to 2 weeks until closure. The principal outcome assessed was mean time to wound closure; other outcomes included mean percent reduction from baseline in wound area and volume at weeks 2 to 8. Results: All wounds were closed a mean of 9.2 weeks after the first DAMA application (range, 3.0-13.5 weeks). Compared with baseline, wound area and volume, respectively, were reduced by a mean of 48% and 60% at week 2 and by 89% and 91% at week 8. Time to closure was shorter among four patients who had three DAMA applications (mean, 8.3 weeks; range, 4.0-11.0 weeks) than among three patients who had only one application (mean, 12.1 weeks; range, 9.5-13.5 weeks). Conclusions: Chronic, unresolved DFUs treated with DAMA rapidly improved and reached closure in an average of 9.2 weeks. These cases suggest that DAMA can facilitate healing of DFUs that have failed to respond to standard treatments.
Article
Objective: A diabetic foot ulcer (DFU) is one of the many potential complications associated with diabetes. If not effectively and rapidly treated, DFUs can result in lower extremity amputations. This prospective case series aimed to assess the effectiveness of a dehydrated amniotic-derived tissue allograft (DAMA), with regards to time to wound closure and total number of applications. Method: Patients were recruited with a neuropathic non-healing DFU(s) despite standard care for at least 4 weeks before the study. The number of DAMA applications and time between applications was based on the physician's judgment. For the majority of patients (n=13/14), offloading, usually total contact casting (TCC), was used in conjunction with DAMA. Wounds were assessed, measured, and photographed every 1-2 weeks. Results: Cases included 14 patients (11 men, 3 women; mean age 56.7±9.1 years) with 15 non-healing neuropathic DFUs with a mean baseline wound area of 6.5±11.6cm2 (median: 2.2cm(2); range: 0.1-44.2cm(2)) and mean volume of 4.3±10.9cm(3) (median: 0.3cm(3); range: 0-39.8cm3). All patients in this series achieved complete wound closure within a median time of 5 weeks (range: 1-14 weeks). Wound area was reduced by a median of 58.3% at week 1 and 74.1% at week 3, and volume by a median of 62.8% at week 1, 97.4% at week 3 and by a median of 100% at week 5 and all time points thereafter. Patients received a median of 2 DAMA applications (range: 1-11). In those that required more than 1 application (n=12), DAMA was applied at intervals of 1 week (n=3) or ≥2 weeks (n=9). Smaller wounds (areas <2.2cm2) closed rapidly (<1 month, 1-2 applications), whereas larger wounds (>2.2cm(2)) required >2 weekly/biweekly applications. Conclusion: The use of DAMA, particularly when coupled with TCC, led to wound closure of DFUs in all patients in this case series, including complex patients with DFUs of ≥1 year in duration, lack of prior response to conservative treatment measures, area >10cm(2) and/or multiple comorbidities. Prospective randomised trials would help to elucidate the precise role of DAMA in these encouraging results.