ArticlePDF Available

Comparison of cryopreserved amniotic membrane and umbilical cord tissue with dehydrated amniotic membrane/chorion tissue

Authors:

Abstract and Figures

Objective: To evaluate how the different processing methods cryopreservation and dehydration affect the structural integrity and biological composition of key signalling molecules within amniotic membrane and umbilical cord tissues. Method: We directly compared cryopreserved amniotic membrane (AM) and umbilical cord (UC) tissues with dehydrated amniotic membrane/chorion (dHACM) tissue using biochemical and functional assays including histological and histochemical staining, BCA, agarose gel electrophoresis, western blot, ELISA, and proliferation and cell death assays. Results: Cryopreservation retains the native architecture of the AM/UC extracellular matrix and maintains the quantity and activity of key biological signals present in fresh AM/UC, including high molecular weight hyaluronic acid, heavy chain-HA complex, and pentraxin 3. In contrast, dehydrated tissues were structurally compromised and almost completely lacked these crucial components. Conclusion: The results presented here indicate that cryopreservation better preserves the structural and biological signaling molecules of foetal tissues.
Content may be subject to copyright.
THIS ARTICLE IS REPRINTED FROM THE JOURNAL OF WOUND CARE VOL 23, NO 10, OCTOBER 2014
M. Cooke,1 BS, Associate Product Manager; E.K. Tan,2, 3 MS Director of Product Development;
C. Mandrycky,4 BSE, Laboratory Technician; H. He, 2, 3 PhD, is a Senior Scientist; J. O’Connell,1 PhD, Senior Director of
Research and Development; S.C.G. Tseng,1,2,3 MD, PhD, Chief Scientic Ofcer;
1 Amniox Medical, Atlanta, GA, 30339, USA; 2 TissueTech, Inc., Miami, FL, 33173, USA;
3 Ocular Surface Center, Miami, FL, 33173, USA; 4 Wallace H. Coulter Department of Biomedical Engineering
at Georgia Institute of Technology, Atlanta, GA, 30332, USA.
Email: stseng@ocularsurface.com
journal of wound care
? ?
C
W
C
? ? ?
volume 23. number 10. october 2014
Comparison of cryopreserved
amniotic membrane and umbilical
cord tissue with dehydrated
amniotic membrane/chorion tissue
research
s
THIS ARTICLE IS REPRINTED FROM THE JOURNAL OF WOUND CARE VOL 23, NO 10, OCTOBER 2014
© 2014 MA HeAltHcAre ltd
Comparison of cryopreserved amniotic
membrane and umbilical cord tissue
with dehydrated amniotic membrane/
chorion tissue
l Objective: To evaluate how the different processing methods cryopreservation and dehydration affect
the structural integrity and biological composition of key signalling molecules within amniotic membrane
and umbilical cord tissues.
l Method: We directly compared cryopreserved amniotic membrane (AM) and umbilical cord (UC)
tissues with dehydrated amniotic membrane/chorion (dHACM) tissue using biochemical and functional
assays including histological and histochemical staining, BCA, agarose gel electrophoresis, western blot,
ELISA, and proliferation and cell death assays.
lResults: Cryopreservation retains the native architecture of the AM/UC extracellular matrix and
maintains the quantity and activity of key biological signals present in fresh AM/UC, including high
molecular weight hyaluronic acid, heavy chain-HA complex, and pentraxin 3. In contrast, dehydrated
tissues were structurally compromised and almost completely lacked these crucial components.
lConclusion: The results presented here indicate that cryopreservation better preserves the
structural and biological signaling molecules of foetal tissues.
lDeclaration of interest: S.C.G. Tseng and his family are more than 5% shareholders of TissueTech,
Inc. TissueTech, Inc. and its Subsidiaries (Bio-Tissue and Amniox Medical) own US Patents Nos. 6,152,142;
6,326,019; and PCT/US2010/046675 on the method of preparation and clinical uses of human amniotic
membrane with the CryoTek method distributed by Bio-Tissue, Inc. and Amniox Medical. S.C.G. Tseng,
E.K. Tan, and H. He are employees of TissueTech, Inc. J. O’Connell and M. Cooke are employees and
shareholders of Amniox Medical. C. Mandrycky has no nancial conict. This research was supported by
a Venture Lab Grant #398 from the Georgia Research Alliance, Atlanta, GA, and a research grant from
TissueTech, Inc., Miami, FL.
amniotic membrane, umbilical cord, cryopreserved, dehydrated, anti-inflammatory, anti-scarring
A
mniotic membrane (AM) consists of a
monolayer of simple epithelium
attached to a thick basement mem-
brane and an underlying avascular stro-
mal region that can be further subdi-
vided into compact, broblast and spongy layers.
The stroma of the umbilical cord is primarily com-
posed of a viscous connective material called Whar-
ton’s jelly surrounded by an outer layer of AM. While
the umbilical cord supplies the foetus with oxygen-
ated, nutrient-rich blood, the AM protects the foetus
from maternal insults during development.1,2
Clinically, AM has been used as an allograft across
multiple disciplines. Particularly in ophthalmology,
transplantation of AM has become a standard surgi-
cal procedure for ocular surface reconstruction to
promote epithelialisation and reduce inammation
and scarring.3 Elsewhere, cryopreserved amniotic
tissue has been used to treat wounds in different
types of tissue including tendon4,5 and nerve
repair.6,7 AM was used to cover open wounds as early
as 1910,8 and has also been used on wounds caused
by many other aetiologies such as venous leg
ulcers,9–12 pressure ulcers9,13–15, diabetes mellitus,15
trauma15,16 and burns.17–22
While the clinical use of AM is well documented,
the use of umbilical cord (UC) tissue in this setting
is relatively new. Comprised of AM with additional
components unique to the foetal environment, it is
likely that UC would have comparable, if not
increased, effectiveness to AM in both biological
and functional assays and is therefore included in
the analyses presented here.
The successful commercialisation of any tissue
product relies on the effective preservation of key
biological components essential to maintaining the
intended therapeutic action of the tissue. Differ-
ences in processing methods can dramatically alter
both the structural and biochemical composition of
the tissue, and impair the activity of vital signalling
M. Cooke,1 BS, Associate
Product Manager;
E.K. Tan,2, 3 MS Director
of Product Development;
C. Mandrycky,4 BSE,
Laboratory Technician; H.
He, 2, 3 PhD, is a Senior
Scientist; J. O’Connell,1
PhD, Senior Director of
Research and
Development; S.C.G.
Tseng,1,2,3 MD, PhD,
Chief Scientic Ofcer;
1 Amniox Medical,
Atlanta, GA, 30339, USA;
2 TissueTech, Inc., Miami,
FL, 33173, USA; 3 Ocular
Surface Center, Miami, FL,
33173, USA; 4 Wallace H.
Coulter Department of
Biomedical Engineering at
Georgia Institute of
Technology, Atlanta, GA,
30332, USA.
Email: stseng@
ocularsurface.com
research
THIS ARTICLE IS REPRINTED FROM THE JOURNAL OF WOUND CARE VOL 23, NO 10, OCTOBER 2014
© 2014 MA HeAltHcAre ltd
molecules such as cytokines, proteoglycans and
growth factors, which are critical to the intended
use of the product.23 From AM, we have successfully
puried and characterised ‘HC-HA/PTX3’ as a
unique matrix component responsible for its thera-
peutic actions.24–28,30 HC-HA/PTX3 is formed by
tight association between pentraxin 3 (PTX3), a
member of the pentraxin family of proteins, known
to be involved in innate immunity, and HC-HA, a
high molecular weight (HMW) hyaluronic acid (HA)
covalently linked to heavy chain 1 (HC1) of inter-
α-trypsin inhibitor (IαI).24–28,30 In vitro, soluble HC-
HA/PTX3 suppresses the proliferation and promotes
apoptosis of lipopolysaccharide (LPS)-induced mac-
rophages.27 Additionally, immobilised HC-HA/PTX3
upregulates IL-10 and downregulates IL-12 upon
stimulation of IFN-α and LPS to polarise macro-
phages toward the M2 (activated macrophage) phe-
notype which plays an integral role in promoting
the healing response.27-29 Therefore, measurement
of the presence and function of HC-HA/PTX3 can
be used to judge how well different processing
methods may preserve the therapeutic actions of
AM and UC.
While several processing methods exist, the most
widely employed are cryopreservation and dehydra-
tion via heat drying. In order to judge the effects of
cryopreservation on AM and UC tissues, we recently
reported that cryopreserved AM/UC is comparable to
fresh AM/UC based on the retention of key architec-
tural and biochemical components essential for the
therapeutic actions of the tissues.31 Here, we extend
our study to compare cryopreserved AM/UC tissues
with dehydrated amnion/chorion tissues in order to
verify whether different processing methods might
affect tissue integrity and therapeutic potential.
Materials and methods
Tissue preparation
Cryopreserved human AM (CT-AM) and UC (CT-
UC) tissue processed by the CryoTek (CT) method
(US 6,326,019, US 6,152,142 and PCT/
US2010/046675) was provided by TissueTech, Inc.
(Miami, FL) and compared with dehydrated tissue
(dHACM), processed by the PURION method (US
8,323,701).To prepare cryopreserved AM and UC tis-
sues, donated full-term human placentas with the
umbilical cord were recovered after cesarean-section
delivery in compliance with American Association
of Tissue Banks (AATB) standards and immediately
stored at -80°C for up to one year. Prior to process-
ing, the frozen placenta and UC were thawed at
room temperature for 8 hours in a Good Manufac-
turing Practice (GMP) facility before being placed at
8°C for an additional 16 hours. Under aseptic condi-
tions, the placenta and UC were rst cleaned of
blood clots with phaosphate-buffered saline (PBS)
prior to separation of AM and UC by blunt dissec-
tion. The chorion was separated from AM and blood
vessels were stripped from UC to generate a at graft
before gentle rinsing in PBS until all blood colora-
tion was removed. AM was afxed on a lter mem-
brane and cut to 6 x 6cm while UC was cut to 6 x
3cm. The AM or UC tissue was nally packaged in a
pouch containing 1:1 v/v Dulbecco Modied Eagle
Medium (DMEM) and glycerol before storage at
-80°C for up to two years.
Histology and histochemistry
Cryopreserved tissues were allowed to thaw for 10
minutes at room temperature and dehydrated sam-
ples were rehydrated according to package instruc-
tions. All tissues were xed with 10% formalin for
one hour, washed three times with PBS for ve min-
utes each, and cut with a 15mm biopsy punch to
obtain equal-sized tissue samples. Tissues were sub-
sequently embedded in histogel, processed, embed-
ded into parafn blocks, and cut into 5μm sections.
Histological sections were then stained with either
hematoxylin & eosin (H&E), Masson’s trichrome
(MAS), or Safranin O (SafO) with Fast Green FCF
counterstain. For HA histochemistry, de-parafn-
ised sections were incubated with biotinylated
Hyaluronic Acid Binding Protein (HABP) followed
by Alexa Fluor 488 streptavidin. Nuclei were coun-
terstained with Hoechst-33342 and images were
photographed by laser confocal microscopy
(LSM700, Axio Observer.Z1, Zeiss, Germany).
Tissue-extract preparation
The preparation of tissue extracts for CT-AM, CT-UC
and dHACM was carried out aseptically as previ-
ously reported.32 To assess the retention of key bio-
chemical molecules, homogenised tissues were
extracted by 4M guanidine hydrochloride and the
supernatant dialysed against PBS for 30 hours to
obtain water-soluble extracts from each tissue. Pro-
tein content was quantied using a standard BCA
assay (Pierce) while quantication of HA was per-
formed by HABP-coated microwell kit (Corgenix).
Determination of HA sizes by agarose-gel
electrophoresis
The molecular weight of HA in tissue extracts was
analysed by agarose gel electrophoresis as previous-
ly reported.33 Tissue extracts were loaded at the same
equivalent (15µg) of HA per lane with or without
pretreatment (one hour, 37°C) with 9 units HAase
per μg. The samples were separated on a 0.5% agar-
ose gel at 20V for the rst 30 minutes and then 40V
for 4 hours. The gel was subsequently stained with
0.005% Stains-all dye in 50% ethanol overnight at
25°C in the dark before destaining in water and
exposing to ambient light for 6 hours. The molecu-
lar weight range of HA samples, which appear as a
research
s
THIS ARTICLE IS REPRINTED FROM THE JOURNAL OF WOUND CARE VOL 23, NO 10, OCTOBER 2014
© 2014 MA HeAltHcAre ltd
bluish smear on the agarose gel, was estimated by
comparison to the Select-HA HiLadder and HMW
HA (Healon).
Western blot
Tissue-extract samples were loaded with equivalent
(20µg) protein per lane with or without pretreat-
ment (one hour, 37°C) with two units of HAase per
μg HA before denaturation in Laemmli Buffer (1:1
dilution with sample) at 95°C for ve minutes. Sam-
ples were then electrophoresed and electrophoreti-
cally transferred on to a 0.45µm nitrocellulose
membrane. The membrane was then blocked with
5% fat-free milk in tris-buffered saline and tween 20
(TBST) and sequentially incubated with mouse anti-
human primary antibodies against HC1 (1:1000) in
5% fat-free milk in TBST (16 hours, 4°C) followed by
rabbit anti-mouse HRP-conjugated secondary anti-
body (1:1000) in 5% fat-free milk in TBST (two
hours, room temperature). Immunoreactive protein
bands were detected with Western Lightning
Chemiluminesence Reagent and imaged by a Lumi-
nescent Image Analyzer (ImageQuant LAS 4000,
GE). The same membrane was rinsed and stripped
with Restore Plus Western Blot Stripping Buffer and
then re-blocked with 5% fat-free milk in TBST and
sequentially incubated with rat anti-human prima-
ry antibodies against PTX3 (1:1000) in 5% fat-free
milk in TBST (16 hours, 4°C) followed by goat anti-
rat HRP-conjugated secondary antibody (1:1000) in
5% fat-free milk in TBST (2 hours, room tempera-
ture). Immunoreactive protein bands were detected
with Western Lighting Chemiluminesence Reagent
and imaged by a Luminescent Image Analyzer.
Macrophage proliferation assay
RAW264.7 cells were seeded at a density of 156 cells/
mm2 on a 96-well plate in DMEM/10% foetal bovine
serum (FBS) (t=0) and treated at t=2 hours with
either CT-AM, CT-UC, or dHACM tissue extracts
containing 100μg protein/ml, or PBS vehicle con-
trol before activation with IFN-α (200units/ml) and
LPS (1μg/ml) at t=3 hours. At t=25 hours, cells were
labelled with 10µM BrdU for 2 hours and were sub-
sequently xed with FixDenat (provided in BrdU
ELISA kit, Roche) at 25°C for 30 minutes, followed
by incubation with anti-BrdU-peroxidase conjugate
at 25°C for 2 hours. The colour was developed for 30
minutes by adding the substrate tetramethylbenzi-
dine (TMB) and stopped by adding 1M H2SO4.
Colorimetric measurements were performed at
450nm with a reference wavelength at 690nm.
Macrophage cell death assay
RAW264.7 cells were seeded and treated as above for
the rst three hours. At t=27 hours, cell lysates were
collected using lysis buffer (provided in Cell Death
ELISA kit, Roche). The cell lysate (20μl) was trans-
ferred to a microplate and 80μl of Immunoreagent
was added before incubation at 300rpm (2 hours,
25°C). After incubation, wells were rinsed three
times with 300μl incubation buffer. The colour was
developed for 20 min by adding 100μl of ABTS solu-
tion and stopped by adding 100μl ABTS Stop Solu-
tion. Colorimetric measurements were performed at
405nm with a reference.
ELISA analysis
RAW264.7 cells were seeded at a density of 250 cells/
mm2 on a 24-well plate in DMEM/10% FBS (t=0)
and treated at t=2 hours with either CT-AM, CT-UC,
or dHACM tissue extracts containing 100μg pro-
tein/ml, or PBS vehicle control before activation
with IFN-α (200 units/ml) and LPS (1μg/ml) at t=3
hours. Culture media from each well was collected
at t=27 hours for IL-10 and IL-12 ELISA analysis.
Briey, a capture antibody was adsorbed onto a
96-well plate, followed by a blocking step, incuba-
tion with 100µL sample, and binding of analyte to a
biotinylated detection antibody. Concentrations of
capture and detection antibodies were used accord-
ing to the manufacturer’s protocol (BioLegend).
IL-10 or IL-12 in the cell supernatants was assessed
using the colorimetric reaction of peroxidase TMB
at an absorbance reading of 450nm.
Statistical analysis
Unless otherwise indicated, data are represented as
mean±standard error with a sample size of three or
more for each condition. A Student’s t-test was per-
formed to test for statistical signicance in protein
levels with Microsoft Ofce Excel 2007. An analysis
of variance (ANOVA) coupled with Tukey’s post-hoc
analysis was performed to test statistical signicance
for HA quantication, macrophage cell prolifera-
tion, macrophage cell death and ELISA analysis with
SPSS Statistics 20 (IBM). Where p<0.05 the results
were considered statistically signicant.
Results
Histological and immunohistochemical staining
To compare the effects of cryopreservation and
dehydration on tissue morphology and extracellular
matrix (ECM) components, sections were examined
by histochemical staining We have previously
shown that cryopreserved AM/UC tissues contain
no signicant structural differences compared with
fresh tissues.31 As seen previously, H&E staining
revealed that the structural morphology of CT-AM
(Fig 1a) closely resembled that of fresh AM tissue,
which contains a thin basement membrane sand-
wiched between a simple epithelium and an avascu-
lar stroma. As expected, CT-UC mirrored this mor-
phology, with an expanded stromal layer (Fig 1b). In
contrast, dHACM contained a layer of chorion in
research
THIS ARTICLE IS REPRINTED FROM THE JOURNAL OF WOUND CARE VOL 23, NO 10, OCTOBER 2014
© 2014 MA HeAltHcAre ltd
addition to the AM layer, and was dramatically
compacted in comparison to the cryopreserved tis-
sue (Fig 1c). Both Masson’s Trichrome (Fig 1 d–f)
and Safranin O staining (Fig 1 g–i), used to indicate
ECM collagen content and sulfated proteoglycans,
respectively, corroborated these results. Collectively,
these ndings support the notion that cryopreserva-
tion did not cause notable changes in the histologi-
cal properties of thin or thick AM tissue, while
dehydration drastically altered the structural integ-
rity of the tissue.
Biochemical analyses of HA and HC-HA
Recent studies have indicated HC-HA/PTX3 is a key
signalling proteoglycan present in AM/UC.25–28,30,31
Histochemistry with HABP was performed to visual-
ise the density and distribution of HA within the
ECM of cryopreserved AM/UC and dHACM. HA
staining was positive in the AM stroma but absent
in the amniotic epithelium for all tissues. Of note,
HA was only weakly present in the subjacent chori-
on of dHACM. Cryopreserved tissues presented
robust uniform staining across the stromal layer,
especially apparent in the thick stromal layer of the
CT-UC (Figs 2 a and b,). dHACM exhibited only
modest HA staining that was sporadically distribut-
ed throughout the ECM, indicating the disruption
and removal of HA originally present in the mem-
brane by this processing method (Fig 2c). The
absence of staining following pretreatment of histo-
logic sections with HAase conrmed the specicity
of HA staining (Fig. 2 d–f).
To directly quantify the differences in ECM HA
content, tissue extracts were analysed using an
HABP assay. While both cryopreserved samples
exhibited an increased HA/total protein ratio com-
pared to dHACM, the results only reached signi-
cance in the CT-UC sample (Fig 3). Consistent with
HA staining, the CT-UC sample contained signi-
cantly more (~50X) HA than dHACM. Of note,
while not signicantly different, CT-AM contained
three times more HA than the dehydrated tissue,
likely due to the chorion layer contributing to an
increase in total protein while appearing to have
comparatively little HA.
The molecular weight of HA has been tied to dif-
ferent biological outcomes in vivo with critical dis-
tinction between low molecular weight HA (LMW
HA) and high molecular weight (HMW) HA.33 To
determine the size distribution of HA in the tissue
extracts, we used agarose gel electrophoresis fol-
lowed by Stains-all dye as previously described.25
Staining was shown to be specic for HA by the dis-
appearance of the HMW HA fraction with HAase
digestion (Fig 4, lanes 6–8). HA from CT-UC (Fig 4,
lane 4) exhibited a similar HMW distribution as the
Fig 2. HA histochemistry of cryopreserved and dehydrated tissues. Tissues
were incubated with HA binding protein with (d,e,f) or without (a,b,c)
HAase digestion. All images are 10x magnication, scale bar=200µM.
a
d
b
e
c
f
CT-AM CT-UC dHACM
HAase +
HAase -
HA=hyaluronic acid, CT-AM=cryopreserved amniotic membrane, CT-UC=cryopreserved umbilical cord,
dHACM=dehydrated human amnion/chorion membrane
Fig 3. HA content quantied using a HABP
microwell kit and normalised to total protein. The
amount of HA retained in CT-UC was signicantly
higher than all other tissues.
HA (g)/Protein (g)
CT-AM CT-UC dHACM
1.8
1.6
1.4
1.2
1.0
0.8
0.6
0.4
0.2
0.0
*
* Indicates p<0.01. CT-AM=cr yopreserved amniotic membrane,
CT-UC=cryopreserved umbilical cord, dHACM=dehydrated human amnion/
chorion membrane
Fig 1. Histological staining of cryopreserved and dehydrated amniotic
tissues. CT-AM (a, d ,g), CT-UC (b, e, h) and dHACM (c, f, i). All images are
20x magnication.
a
d
g
b
e
h
c
f
i
CT-AM CT-UC dHACM
CT-AM=cryopreserved amniotic membrane, CT-UC=cyropreserved umbilical cord, dHACM=dehydrated human
amnion/chorion membrane
Hematoxylin and
eosin
Masson’s Trichrome
Safranin O
research
s
THIS ARTICLE IS REPRINTED FROM THE JOURNAL OF WOUND CARE VOL 23, NO 10, OCTOBER 2014
© 2014 MA HeAltHcAre ltd
Healon control (Fig 4, lane 2) in the range of 1000 to
6000 kDa, indicating that UC preferentially con-
tained HMW HA. In contrast, dHACM contained no
HMW HA, instead displaying a LMW HA smear at
the bottom of the gel (Fig 4, lane 5) with a molecular
weight below 500 kDa. Interestingly, a HMW band
was detected in the loading wells of both CT-AM and
CT-UC samples (Fig 4, lanes 3 and 4) which was abol-
ished after HAase digestion (Fig 4, lanes 6 and 7), sug-
gesting the presence of a HMW HA species in cryop-
reserved AM/UC that was larger than that of Healon.
In contrast, no band from the dehydrated sample
existed in the loading well (Fig 4, lane 5), corroborat-
ing the LMW smear within the lane.
Previous research has shown that the HA present
in AM/UC exists as a HC-HA/PTX3 complex.25–28,30,31
To determine if this complex was altered or
destroyed by the dehydration processing method,
we subjected tissue extracts from cryopreserved and
dehydrated tissues to Western blot using an HC1-
specic antibody. Additionally, to ensure that HC1
was indeed associated with HA, tissue-extract sam-
ples were examined in parallel by rst treating one
replicate with NaOH, which hydrolyses the ester
bond between HA and HC1. As expected, IαI puri-
ed from human plasma yielded a major band at
~250kDa (Fig 5, lane 2) and treatment of IαI with
50mM NaOH cleaved the ester bonds linking the
HCs to IαI to yield a 75kDa HC1 fragment (Fig 5,
lane 3). As seen in the agarose gel, both cryopre-
served tissues contained a band that remained in
the well (Fig 5, lanes 5 and 7) most likely HC-HA,
because digestion with NaOH partially cleaved the
complex to yield an increase in the intensity of the
75kDa HC1 fragment (Fig. 5, lanes 6 and 8) indicat-
ing that the HC-HA complex is present in both CT-
AM and CT-UC tissues. Interestingly, a band
remained in the CT-UC well even after digestion
with NaOH, possibly due to the large amount of HA
within the CT-UC extract. The dehydrated sample
did not contain any HMW bands and exhibited the
75kDa HC1 band before and after digestion, sug-
gesting the lack of the HC-HA complex due to the
degradation and removal of HMW HA, leaving only
unbound LMW HA.
PTX3 content
PTX3 is an oligomeric protein shown to be consti-
tutively expressed in the AM that serves to stabilise
the HC-HA complex. 30,34 In order to determine the
effects of processing methods on PTX3 protein lev-
els, tissue extracts were analysed via Western blot.
PTX3 exists as two forms: a ~45kDa monomer and
~90kDa dimer (Fig 6, lane 4). Cryopreserved tissue
extracts had strong PTX3 bands at 45 and 90kDa
(Fig 6, lanes 5–8). In addition, the CT-UC sample
contained another band that remained in the well
due to its large size (Fig 6, lanes 7 and 8), similar to
Fig 4. Agarose gel electrophoresis of CT-AM, CT-UC and dHACM tissue
extracts was performed to determine the size of HA present. Select-HA
HiLadder (M, Lane 1) and HMW HA control (Lane 2). The specicity of HA
was conrmed by HAase digestion (Lanes 6–8)
M
1
HA
2
CT-AM
3
CT-UC
4
dHACM
5
HAase (–) HAase (+)
CT-AM
6
CT-UC
7
dHACM
8
Lane
kDa
6100
4570
3050
1520
1090
966
572
495
HA=hyaluronic acid, HMW=high molecular weight, CT-AM=cryopreserved amniotic membrane
CT-UC=cryopreserved umbilical cord, dHACM=dehydrated human amnion/chorion membrane
Fig 5. Heavy chain hyaluronic acid complex detected by Western Blot.
HC-HA
IαI
HC1
kDa
200
140
100
80
60
50
40
30
20
10
HC1 NaOH
IαI
– +
2 3
CT-AM
– +
5 6
CT-UC
– +
7 8
dHACM
– +
9 10
PTX3
4
M
1
CT-AM=cryopreserved amniotic membrane CT-UC=cryopreserved umbilical cord, dHACM=dehydrated human
amnion/chorion membrane, PTX3= pentraxin 3, HC-HA= Heavy chain hyaluronic acid, HC1=heavy chain 1
research
THIS ARTICLE IS REPRINTED FROM THE JOURNAL OF WOUND CARE VOL 23, NO 10, OCTOBER 2014
© 2014 MA HeAltHcAre ltd
the agarose gel, indicating the presence of PTX3 as
part of the HC-HA complex. In comparison, the
dehydrated sample was devoid of PTX3 (Fig 6,
lanes 9 and 10), supporting the idea that the dehy-
dration process degrades and removes PTX3 natu-
rally present in the ECM of AM while cryopreserva-
tion preserves the integrity of this critical HC-HA
stabilising protein.
Functional analyses
AM’s anti-inammatory activity has been demon-
strated by the suppression of viability and prolifera-
tion of macrophages by both AM tissue35,36 and AM
extracts.3,32 To explore the effect of processing on
innate activity, tissue extracts containing 100µg
protein/ml from either cryopreserved or dehydrated
samples were applied to activated RAW264.7 cells,
(a macrophage cell line) and both macrophage pro-
liferation (Fig 7a) and cell death (Fig 7b) were meas-
ured in separate assays. Compared with cells in the
PBS vehicle control, RAW264.7 macrophages treat-
ed with cryopreserved tissue extracts showed signi-
cantly inhibited proliferation (p<0.01; ANOVA fol-
lowed by Turkey’s post-hoc test). However, tissue
extract from dehydrated samples was comparable to
the control and displayed no signicant effect. Fur-
thermore, only CT-UC was signicantly (p<0.01;
ANOVA followed by Turkey’s post-hoc test) more
effective than the control in promoting macrophage
apoptosis. In fact, CT-UC-stimulated apoptosis was
approximately 14 times more greater than that seen
with all other tissue extracts.
ELISA quantication of cytokines
We have previously reported that the HC-HA/PTX3
complex downregulates pro-inammatory cytokines
while upregulating anti-inammatory
cytokines.27,28,30 ELISA analysis of two critical
cytokines IL-10 (anti-inammatory) and IL-12 (pro-
inammatory) was performed to elucidate what
effects, if any, processing techniques had on HC-
HA/PTX3 ability to regulate cytokine expression.
CT-UC extract dramatically increased IL-10 expres-
sion compared to all other samples, with IL-10 lev-
els approximately 8-fold higher than dehydrated
samples (Fig 8a). When assessing IL-12 expression,
all samples were signicantly different to control,
with CT-AM and dHACM exhibiting similar expres-
sion levels and CT-UC reducing IL-12 expression 7–
fold more than dHACM (Fig 8b).
Discussion
Fresh AM has been proven efcacious in clinical
applications.11,37,38 However, its use presents a seri-
ous risk of disease transmission.39,40 Therefore,
processing methods that ensure the safety of the tis-
sue while preserving its innate biological effective-
ness become critical. With several processing proto-
cols commonly used, techniques differ dramatically
and can have varying impacts on both the structur-
al and biochemical composition of the tissue, as
well as the activity of critical signalling molecules
(cytokines, proteoglycans, etc), essential to the ther-
apeutic actions of the tissue.23 Specic to the meth-
ods compared here, the cryopreservation process
involves quickly freezing the tissue and was speci-
cally developed to maintain the structural integrity
of the extracellular matrix and the endogenous bio-
chemical functions of the native AM and UC tis-
sues. In contrast, dehydration is much harsher and
has been shown to cause protein denaturation, loss
of function, and irreparable damage to the
ultrastructure and material properties of the tissue,
resulting in loss of cell attachment and decreased
cell inltration.23,41–43 This process creates the poten-
tial for compromising the survival and permanence
of key anti-inammatory and anti-scarring factors,
suggesting the clinical efcacy of tissue processed by
this method may be lessened.
Histological analysis revealed that the ECM archi-
tectural structure within CT-AM and CT-UC was not
altered by cryopreservation but was compromised
by dehydration. This is a critical nding, as the pro-
teins within the ECM regulate the functions of cells
and small molecules, and act as a reservoir and
modulator of cytokines and growth factors.44,45
Moreover, the healing potential of AM is mediated
by the complex assembly of these components,46–48
further indicating the importance of maintaining
the integrity of the ECM after preservation. Of note,
cryopreserved tissues were comparable in structure
Fig 6. Pentraxin 3 identied by Western Blot.
HC-HA
PTX3
(dimer)
PTX3
(monomer)
kDa
200
140
100
80
60
50
40
30
20
10
PTX3 NaOH
IαI
– +
2 3
CT-AM
– +
5 6
CT-UC
– +
7 8
dHACM
– +
9 10
PTX3
4
M
1
CT-AM=cryopreserved amniotic membrane, CT-UC=cryopreserved umbilical cord, dHACM=dehydrated human
amnion/chorion membrane, PTX3= pentraxin 3, HC-HA= heavy chain hyaluronic acid
research
s
THIS ARTICLE IS REPRINTED FROM THE JOURNAL OF WOUND CARE VOL 23, NO 10, OCTOBER 2014
© 2014 MA HeAltHcAre ltd
to fresh AM31 while dehydrated tissues were notably
impaired, indicated by the compact appearance of
the stromal and chorion layers.
The clinical success of AM/UC as a potent anti-
inammatory and anti-scarring agent has prompted
examination into the physiological mechanism of
its therapeutic actions. Previous studies revealed
AM’s regenerative potential may be mediated by
complex arrays of cytokines, chemokines, and
growth factors which have been linked to the pro-
motion of wound healing via suppression of host
immune cells.49,50 Recent studies have implicated HA
complex as a critical biological component that con-
tributes to the anti-inammatory and anti-scarring
properties of AM.25,28,30,32 Cryopreservation retained
high levels of HA within both CT-AM and CT-UC
samples and also preserved the distribution of HA
across the stromal layer of the tissues. In contrast,
while HA was still present in the dHACM sample,
the distribution was radically altered, with sporadic
pockets of HA randomly dispersed throughout the
tissue and total levels of HA much lower, compara-
tively. Moreover, the HA present within cryopre-
served tissues, particularly CT-UC, was HMW HA,
while the HA contained in dHACM was LMW HA.
This is of particular interest because certain studies
have suggested HMW HA is the key isoform of HA
responsible for the therapeutic properties mentioned
above while LMW HA contributes to the inamma-
tory response and is immune-stimulatory.33,51
Fig 7. Effects of tissue extracts on macrophage proliferation (a) and cell death (b).
Absorbance/Protein
CTL CT-AM CT-UC dHACM
0.08
0.07
0.06
0.05
0.04
0.03
0.02
0.01
0.00
*
*
Absorbance/Protein
CTL CT-AM CT-UC dHACM
0.08
0.07
0.06
0.05
0.04
0.03
0.02
0.01
0.00
*
a b
* Indicates p<0.01 compared to all other groups. CT-AM=cryopreserved amniotic membrane, CT-UC=cr yopreserved umbilical cord, dHACM=dehydrated
human amnion/chorion membrane, CTL=control.
Fig 8. Macrophage cytokine expression in response to tissue extracts. IL-10, an anti-inammatory cytokine (a)
and IL-12, a pro-inammatory cytokine (b).
Absorbance/Protein
CTL CT-AM CT-UC dHACM
6.0
5.0
4.0
3.0
2.0
1.0
0.00
*
Absorbance/Protein
CTL CT-AM CT-UC dHACM
0.10
0.09
0.08
0.07
0.06
0.05
0.04
0.03
0.02
0.01
0.00
*
p=0.010
ab
* Indicates p<0.01 compared to all other groups. CT-AM=cryopreserved amniotic membrane, CT-UC=cr yopreserved umbilical cord, dHACM=dehydrated human
amnion/chorion membrane, CTL=control.
research
THIS ARTICLE IS REPRINTED FROM THE JOURNAL OF WOUND CARE VOL 23, NO 10, OCTOBER 2014
© 2014 MA HeAltHcAre ltd
We have previously shown that HMW HA in AM
exists within the HC-HA complex that is stabilised
by the multimeric protein PTX325,26,28,30 and that this
larger complex exerts a more powerful therapeutic
effect than HMW HA alone.27,28 Western blot analy-
ses clearly showed the presence of this crucial matrix
component in both CT-AM and CT-UC samples, fur-
ther supporting that the cryopreservation process
does not disturb the innate biological components
of the ECM with AM tissue. On the contrary, the
dHACM sample contained neither HC-HA nor
PTX3, only displaying LMW HA and HC1 frag-
ments. While the presence of these smaller proteins
may impart some transient benecial effects to the
dHACM, the lack of the key ECM components HC-
HA and PTX3 suggests reduced longevity of the
active components within the tissue, following the
dehydration process.
While macrophage activation during inamma-
tion is critical to the body’s response to insult, persist-
ence at the site of injury can result in chronic inam-
mation and inability to heal properly, often leading
to tissue damage.52–54 Recent studies have shown the
unique ability of HC-HA/PTX3 to promote the death
of activated macrophages while downregulating pro-
inammatory cytokines and upregulating anti-
inammatory cytokines.25,27,28,30 The results here
demonstrate the ability of CT-AM and CT-UC to sig-
nicantly reduce activated macrophage proliferation
and the substantial ability of CT-UC to enhance acti-
vated macrophage apoptosis when compared to
dHACM. Due to the aforementioned retention of
high levels of HC-HA and PTX3 within the cryopre-
served tissues, the ability of CT-AM and CT-UC to
modulate macrophage cell survival and growth is not
surprising, and is imperative to their success in
wound healing. Similarly, the lack of HC-HA and
PTX3 within dHACM may contribute to the decreased
effectiveness of the dehydrated tissue in reducing
inammation and brosis in vivo.55 Additionally,
cytokines are known to play critical roles in wound
healing.56,57 IL-10 and IL-12 are well studied cytokines
and are known to be anti- and pro-inammatory,
respectively.58–60 Similar to results seen with pure HC-
HA/PTX3,27,28,30 CT-UC signicantly increased IL-10
expression while decreasing IL-12 expression, most
likely due to the substantially higher levels of HC-HA
within CT-UC tissue compared to AM alone.
While the data presented here adequately tests
and describes the differences in structure, func-
tion, and biochemical make-up of both cryopre-
served and dehydrated tissues, the study does have
its limitations. Only a single cryopreservation and
dehydration processing method were analysed and
future experiments including tissues processed by
varying cryopreservation and dehydration proto-
cols should be performed to see if the differences
outlined here extend to other tissues with similar
processing techniques. Additionally, while the
IL-10/IL-12 ratio is indicative of M1 to M2 macro-
phage polarisation, an additional experiments
should include analysis of a larger panel of
cytokines to more fully understand the immu-
nomodulatory actions of the tissue. Finally, to tru-
ly understand the efcacy of the tissue, in vivo
experiments should be performed in various mod-
els of inammation and disease.
Conclusion
Overall, the results of this comparative study bring
to light considerable differences in the structural
and biochemical properties of cryopreserved and
dehydrated foetal tissues. Histological analysis
revealed that while cryopreservation did not dam-
age the delicate architecture of the ECM with the
tissue, dehydration resulted in a compacted AM
morphology and altered the distribution of struc-
tural matrix components. Furthermore, the key
molecules of HC-HA and PTX3 were absent in the
dehydrated tissue and only LMW HA and HC1 pro-
teins were identied, most likely byproducts of
ECM compaction and alteration after preservation.
Not surprisingly, the lack of these critical mole-
cules lessened the biological activity of the dHACM,
as evidenced in the functional assays. Cryopre-
served tissues retained elevated levels of crucial
biological elements and exhibited high functional-
ity in modulating macrophage viability and
cytokine expression. Taken together, this data
strongly suggests that cryopreservation effectively
preserves the structural and biochemical integrity
of AM and UC matrix components, essential for
the anti-inammatory and anti-scarring effects
observed clinically. n
References
1. Gray, H. Anatomy of the Human
Body. (1918).
2. Mast, B.A., Diegelmann, R.F.,
Krummel, T.M., Cohen, I.K. Scarless
wound healing in the mammalian
foetus. Surg Gynecol Obstet 1992;
174: 5, 441–451.
3. Liu, J., Hosam, S., Fu, Y., Liang, L.,
Tseng, S.C. Update on amniotic
membrane transplantation. Expert
Rev Ophthalmol 2010; 5: 5,
645–661.
4. Gao, M., Zhoa, H., Tian, D. et al.
Experimental Study on Human
Amniotic Membrane for Repairing
Tendon Sheath Defect. Chinese J
Reparative Reconstr Surg 2013; 27:
3, 335-339.
5. Demirkan, F., Colakoglu, N.,
Herek, O., Erkula, G. The use of
amniotic membrane in exor
tendon repair: an experimental
model. Arch Orthop Trauma Surg
2002; 122: 7, 396–399.
6. Kim, S. S., Sohn, S.K., Lee, K.Y. et
al. Use of human amniotic
membrane wrap in reducing
perineural adhesions in a rabbit
model of ulnar nerve
neurorrhaphy. J Hand Surg Eur
2010; 35: 3, 214–219.
7. Meng, H., Li, M., You, F. et al.
Assessment of processed human
amniotic membrane as a
protective barrier in rat model of
sciatic nerve injury. Neurosci Lett
2011; 496: 1, 48–53.
8. Davis, J. S. Skin Grafting at the
Johns Hopkins Hopital. Ann Surg
1909; 50: 3, 542–549.
9. Boc, S., Chairman, E.L., Freed, E.L.
Implications for the use of amnion
and chorion in podiatric medicine
and surgery. J Foot Surg 1985; 24: 4,
236–242.
10. Mermet, I., Pottier, N.,
Sainthiller, J.M. et al. Use of
amniotic membrane
transplantation in the treatment of
venous leg ulcers. Wound Repair
Regen 2007; 15: 4, 459–464.
research
s
THIS ARTICLE IS REPRINTED FROM THE JOURNAL OF WOUND CARE VOL 23, NO 10, OCTOBER 2014
© 2014 MA HeAltHcAre ltd
11. Troensegaard-Hansen, E.
Amniotic Grafts in Skin Ulceration.
Lancet 1950; 1: 859–860.
12. Ward, D.J., Bennett, J.P., Burgos,
H., Fabre, J. The healing of chronic
venous leg ulcers with prepared
human amnion. Br J Plast Surg
1989; 42: 4, 463–467 (1989).
13. Gajiwala, K., Gajiwala, A.L.
Evaluation of lyophilised,
gamma-irradiated amnion as a
biological dressing. Cell Tissue
Bank 2004; 5: 2, 73–80.
14. Gajiwala, A., Sharma, V. Use of
irradiated amnion as a biological
dressing in the treatment of
radiation induced ulcers. Cell
Tissue Bank 2003; 4: 2-4, 147–150.
15. Singh, R., Chouhan, U.S.,
Purohit, S. et al. Radiation
processed amniotic membranes
in the treatment of non-healing
ulcers of different etiologies. Cell
Tissue Bank 2004; 5: 2, 129–134.
16. Gruss, J.S., Jirsch, D.W. Human
amniotic membrane: a versatile
wound dressing. Can Med Assoc J
1978;118: 10, 1237–1246.
17. Bose, B. Burn wound dressing
with human amniotic membrane.
Ann R Coll Surg Engl 1979; 61: 6,
444–447.
18. Matthews, R., Bennett, J.,
Faulk, W. Wound healing using
amniotic membranes. Br J Plast
Surg 1981; 34: 1, 76–78.
19. Ramakrishnan, K.M.,
Jayaraman, V. Management of
partial-thickness burn wounds by
amniotic membrane: a
cost-effective treatment in
developing countries. Burns 1997;
23 Suppl 1: S33–S36.
20. Ravishanker, R., Bath, A.S., Roy,
R. “Amnion Bank”—the use of
long term glycerol preserved
amniotic membranes in the
management of supercial and
supercial partial thickness burns.
Burns 2003; 29: 4, 369–374.
21. Singh, R., Purohit, S.,
Chacharkar, M. P. et al.
Microbiological safety and clinical
efcacy of radiation sterilized
amniotic membranes for
treatment of second-degree
burns. Burns 2007; 33: 4, 505–510.
22. Branski, L. K. Herndon, D.N.
Cells, M.M. et al. Amnion in the
treatment of pediatric
partial-thickness facial burns.
Burns 2008; 34: 3, 393–39.
23. Badylak, S. F. Regenerative
medicine and developmental
biology: the role of the
extracellular matrix. Anat Rec B
New Anat 2005; 287: 1, 36–41.
24. Shay, E., He, H., Sakurai, S.,
Tseng, S.C. Inhibition of
angiogenesis by HC·HA, a
complex of hyaluronan and the
heavy chain of inter-α-inhibitor,
puried from human amniotic
membrane. Invest Ophthalmol Vis
Sci 2011; 52: 5, 2669–2678.
25. He, H. Li, W., Tseng, D.Y. et al.
Biochemical characterization and
function of complexes formed by
hyaluronan and the heavy chains
of inter-alpha-inhibitor (HC*HA)
puried from extracts of human
amniotic membrane. J. Biol Chem
2009; 284: 30, 20136–20146.
26. Zhang, S., He, H., Day, A.J.,
Tseng, S.C. Constitutive
expression of inter-α-inhibitor
(IαI) family proteins and tumor
necrosis factor-stimulated gene-6
(TSG-6) by human amniotic
membrane epithelial and stromal
cells supporting formation of the
heavy chain-hyaluronan (HC-HA)
complex. J Biol Chem 2012; 287:
15, 12433–12444.
27. He, H., Zhang,S., Tighe,S. et
al.Immobilized heavy chain-
hyaluronic acid polarizes
lipopolysaccharide-activated
macrophages toward M2
phenotype. J Biol Chem 2013;
288: 36, 25792–25803.
28. He, H., Tan, Y., Duffort, S. et al.
In vivo downregulation of innate
and adaptive immune responses
in corneal allograft rejection by
HC-HA/PTX3 complex puried
from amniotic membrane. Invest.
Ophthalmol Vis Sci 2014; 55: 3,
1647–1656.
29 Koh, T.J., DiPietro, L.A.
Inammation and wound healing:
the role of the macrophage.
Exper Rev Mol Med 2011; 13: e23.
30. Zhang, S., Zhu, Y.T., Chen, S.Y.
et al. Constitutive Expression of
Pentraxin 3 (PTX3) Protein by
Human Amniotic Membrane Cells
Leads to Formation of the Heavy
Chain (HC)-Hyaluronan
(HA)-PTX3 Complex. 2014 J Biol
Chem 2014; 289: 19, 13531–
13542.
31. Tan, E. K., Cooke, M.,
Mandrycky, C. et al. Structual and
Biological Comparison of
Cryopreserved and Fresh
Amniotic Membrane Tissues. J.
Biomater Tissue Eng 2014; 4: 5,
379–388.
32. He, H. Li, W., Chen, Y.T. et al.
Suppression of activation and
induction of apoptosis in
RAW264.7 cells by amniotic
membrane extract. Invest.
Ophthalmol Vis Sci 2008; 49: 10,
4468–4475.
33. McKee, C.M., Penno, M.B.,
Cowman., M. et al. Hyaluronan
(HA) fragments induce
chemokine gene expression in
alveolar macrophages. The role of
HA size and CD44. J Clin Invest
1996; 98: 10, 2403–2413.
34. Salustri, A., Garlanda, C .,
Hirsch, E. et al. PTX3 plays a key
role in the organization of the
cumulus oophorus extracellular
matrix and in in vivo fertilization.
Development 2004; 131: 7,
1577–1586.
35. Yoneda, M., Suzuki, S., Kimata,
K. Hyaluronic acid associated
with the surfaces of cultured
broblasts is linked to a
serum-derived 85-kDa protein. J
Biol Chem 1990; 265: 9,
5247–5257.
36. Li, W., He, H., Kawakita, T. et al.
Amniotic membrane induces
apoptosis of interferon-gamma
activated macrophages in vitro.
Exp Eye Res 2006; 82: 2, 282–292.
37. Sawhney, C.P. Amniotic
membrane as a biological dressing
in the management of burns.
Burns 1989; 15: 5, 339–42.
38. Troensegaard-Hansen, E.
Amnion implantation in
peripheral vascular disease.
Lancet 1963; 1, 327–328.
39. Maral, T., Borman, H., Arslan,
H. et al. Effectiveness of human
amnion preserved long-term in
glycerol as a temporary biological
dressing. Burns 1999; 25: 7,
625–635.
40. Adds, P.J., Hunt, C.J., Dart, J.K.
Amniotic membrane grafts,
“fresh” or frozen? A clinical and
in vitro comparison. Br J
Ophthalmol 2001; 85: 8, 905–907.
41. Badylak, S.F., Freytes, D.O.,
Gilbert, T.W. Extracellular matrix
as a biological scaffold material:
Structure and function. Acta
Biomater. 2009; 5: 1, 1–13.
42. Freytes, D.O., Tullius, R.S.,
Valentin, J.E. et al. Hydrated
versus lyophilized forms of
porcine extracellular matrix
derived from the urinary bladder.
J Biomed Mater Res A 2008; 87: 4,
862–872.
43. Lepock, J.R., Frey, H.E., Ritchie,
K.P. Protein denaturation in intact
hepatocytes and isolated cellular
organelles during heat shock. J
Cell Biol 1993; 122: 6, 1267–1276.
44. Agren, M. S., Werthén, M. The
extracellular matrix in wound
healing: a closer look at
therapeutics for chronic wounds.
Int J Low Extrem Wounds 2007; 6:
2, 82–97.
45. Olczyk, P., Mencner, Ł.,
Komosinska-Vassev, K. The role of
the extracellular matrix
components in cutaneous wound
healing. Biomed Res Int 2014,
747584.
46. Hopkinson,A., McIntosh, R.S.,
Tighe, P.J. et al. Amniotic
membrane for ocular surface
reconstruction: donor variations
and the effect of handling on
TGF-beta content. Invest
Ophthalmol Vis Sci 2006; 47: 10,
4316–4322.
47. Wolbank, S., Hinder, F., Redl,
H. et al. Impact of human
amniotic membrane preparation
on release of angiogenic factors. J
Tissue Eng Regen Med 2009; 3: 8,
651–654.
48. Schulze, U. Hampel, U., Sel.et
al. Fresh and cryopreserved
amniotic membrane secrete the
trefoil factor family peptide 3 that
is well known to promote wound
healing. Histochem Cell Biol
2012;138: 2, 243–250.
49. Tseng, S.C ., Espana, E.M.,
Kawakita, T.et al. How does
amniotic membrane work? Ocul
Surf 2004; 2: 3, 177–187.
50. Bauer, D., Hennig, M., Baehler,
H. et al. Amniotic membrane
induces peroxisome proliferator-
activated receptor-α positive
alternatively activated
macrophages. Invest Ophthalmol
Vis Sci 2012; 53: 2, 799–810.
51. Girish, K.S., Kemparaju, K. The
magic glue hyaluronan and its
eraser hyaluronidase: A biological
overview. Life Sci 2007; 80: 1,
1921–1943.
52. Koh, T.J., DiPietro, L.A.
Inammation and wound healing:
the role of the macrophage.
Expert Rev Mol Med 2011; 13,
e23-e35.
53. Khanna, S., Biswas, S., Shang, Y.
et al. Macrophage dysfunction
impairs resolution of
inammation in the wounds of
diabetic mice. PLoS One 2010; 5:
3, e9539.
54. Sindrilaru, A., Peters, T.,
Wieschalka, S.et al. An
unrestrained proinammatory M1
macrophage population induced
by iron impairs wound healing in
humans and mice. Jounal Clin
Investig 2011; 121: 3, 985–997.
55. Chun, B.Y., Kim, H.K., Shin, J.P.
Dried human amniotic membrane
does not alleviate inammation
and brosis in experimental
strabismus surgery. J Ophthalmol.
2013; 2013: 369126.
56. Gharaee-Kermani, M., Phan,
S.H. Role of cytokines and
cytokine therapy in wound
healing and brotic diseases. Curr
Pharm Des 2001; 7: 11,
1083–1103.
57. Slavin, J. The role of cytokines
in wound healing. 1996; 178: 1,
5–10.
58. Fiorentino, D.F., Zlotnik, A.,
Mosmann, T.R. et al. IL-10 inhibits
cytokine production by activated
macrophages. J Immunol 1991;
147: 11, 3815–3822.
59. De Waal Malefyt, R., Abrams,
J., Bennett, B. et al. Interleukin 10
(IL-10) inhibits cytokine synthesis
by human monocytes: An
autoregulatory role of IL-10
produced by monocytes. J Exp
Med 1991; 174: 5, 1209–1220.
60. Trinchieri, G. Interleukin-12: A
Proinammatory Cytokine with
Immunoregulatory Functions that
Bridge Innate Resistance and
Antigen-Specic Adaptive
Immunity. Annu Rev Immunol
1995; 13, 251–276.
... In particular, over the past ten years, amniotic membrane and umbilical cord (AM/UC) particulate has demonstrated safety and effectiveness in treatment of a wide range of musculoskeletal indications including knee osteoarthritis (5), wrist osteoarthritis (6), spine facet osteoarthritis (7), and discogenic pain (8). The rationale of using AM/UC in musculoskeletal indications manifesting pain is based on its antiinflammatory, anti-scarring, and pro-regenerative properties which are thought to reduce inflammation and promote restorative healing in the local environment (9)(10)(11)(12). Such effects have not only provided a rapid pain relief, but also longstanding relief for 6 to 12 months after a single injection. ...
... During the processing of the AM/UC tissue, the tissue is cleansed, lyophilized via freeze drying, micronized into particulates, and sterilized via gamma irradiation. According to the manufacturer, the product preserves the natural biological characteristics innate to fresh AM/UC tissue but not any living cells (12). ...
Article
Full-text available
Background Radiculopathy can be a debilitating condition. Amniotic membrane/umbilical cord (AM/UC) particulate is a relatively new injectable treatment modality. Herein we report the outcomes of epidural injection of AM/UC particulate in managing lumbar radiculopathy. Methods Consecutive patients with lumbar radiculopathy who received epidural injection of AM/UC particulate for lumbar radiculopathy were included. Primary outcome was change in pain as measured by the 11-point numerical rating scale. Safety was assessed by AM/UC- and procedure-related complications. Paired t-tests were used to determine statistical significance. Results A total of 12 patients with a mean age of 56.7 ± 21.0 years were included in the analysis. The patients were previously treated with physical therapy (91.7%), chiropractic corrective measures (16.7%), epidural steroid injection (83.3%), and radiofrequency ablation (8.3%). Two patients (16.7%) were taking opioids for chronic pain syndrome. After AM/UC injection, the average pain score decreased from 6.6 ± 1.5 to 5.2 ± 1.9 at 1–3 months, 2.0 ± 1.4 at 6 months, and 2.9 ± 1.4 at last mean follow-up of 21.3 ± 11.1 months (p < 0.001). No patients required subsequent treatment or surgery. There were also no complications. Conclusion This case series supports the preliminary safety and shows potential benefit of epidural AM/UC particulate injection in this cohort of patients with lumbar radiculopathy pain.
... 24,25 AMUC tissue use may be advantageous in these cases because of its compositional components that modulate inflammation and promote healing. 26 Placental tissues are processed to retain key biologic and structural components, including many growth factors, cytokines, and proteins such as HC-HA/PTX3 that promote regenerative healing. 10,26,27 Placental tissue contrasts with PRP, a biologic that is theoretically more inclined to incite a proinflammatory environment with growth factors at supraphysiologic levels and may lead to poorer long-term outcomes, as seen in our study. ...
... 26 Placental tissues are processed to retain key biologic and structural components, including many growth factors, cytokines, and proteins such as HC-HA/PTX3 that promote regenerative healing. 10,26,27 Placental tissue contrasts with PRP, a biologic that is theoretically more inclined to incite a proinflammatory environment with growth factors at supraphysiologic levels and may lead to poorer long-term outcomes, as seen in our study. ...
Article
Full-text available
Background: Meniscal tears are one of the most frequent injuries to the knee, with an estimated incidence of 222 per 100,000 individuals aged 18 to 55 years based on magnetic resonance imaging. Poor outcomes following meniscal surgical interventions are common and have led many surgeons to use biologic augmentation strategies to enhance the healing. Methods: We conducted a single-center, retrospective, observational study of patients who underwent arthroscopic meniscectomy with and without adjunctive platelet-rich plasma (PRP) or the particulate form of amniotic umbilical cord (AMUC) tissue. We evaluated patient-reported outcomes on the visual analog scale for pain, International Knee Documentation Committee (IKDC) Subjective Knee Evaluation Form, Lysholm Knee Scoring Scale, 12-Item Short Form Survey, and Knee Injury and Osteoarthritis Outcome Score (KOOS) during a 1-year postoperative period. Complications and follow-up procedures were also evaluated. Results: We evaluated 113 patients who underwent meniscectomies from November 2010 to March 2017. Pain severity was significantly decreased only in the AMUC group at 6 months (P=0.0143). Patients in the AMUC group demonstrated significant improvement in functional recovery based on the IKDC and the KOOS subscales of pain, symptoms, activities of daily living, and sport and recreation function at 6 months. Patients in the PRP group had a significant benefit in the KOOS subscales of pain, symptoms, sport and recreation function, and knee-related quality of life at 3 months. Improvement in the control group was less substantial. Patients in the PRP group had more complications and follow-up procedures (30.0%) than patients in the AMUC group (8.3%). Conclusion: In our study population, arthroscopic meniscectomy with adjunctive use of AMUC tissue improved patient-reported outcomes and reduced the reoperation rate compared to conventional technique or adjunctive use of PRP.
... These are considered to be the key properties of AM responsible for its anti-inflammatory, anti-scarring, and anti-angiogenic effects [15,16]. Anophthalmic patients are monitored in ophthalmology clinics for extended periods with various issues. ...
Article
Full-text available
Purpose This report presents the results of using cryopreserved umbilical amniotic membrane (cUAM) as an alternative mucosal graft for ocular surface reconstruction in cases of anophthalmic socket contracture (ASC), cicatricial entropion (CE), and conjunctival-scleral defects. Methods The study included patients who underwent non-commercial implantation of cUAM grafts (prepared by corneal banking methods) for ASC, CE, conjunctival defect, and scleral melting. The main success criteria for this study were the comfortable fitting of the ocular prosthesis in ASC patients, the natural eyelid position in CE patients, and the degree of conjunctivalisation in melting patients. Results cUAM transplantation was performed in 2 patients who could not use a prosthetic eye due to conjunctival contracture, 2 patients with CE, and 1 patient with conjunctival defect and 1 patient with conjunctival-scleral melting. The primary outcome was achieved in 83.3% (5/6) of patients. In one patient with CE, partial healing was achieved due to the persistence of CE in the medial upper eyelid. Conclusions cUAM is a viable alternative to mucosal grafting for reconstructing the bulbar and palpebral conjunctival surface, fornix, and orbit, with reduced donor morbidity and shorter surgical time. Its regenerative ability allows for tissue defect healing and improves cosmetic appearance through epithelialization within weeks.
... In a comparison study with dHACMs, vCPMs revealed greater clinical effectiveness, notably significantly higher closure rates. This enhanced performance can be attributed, in part, to vCPMs preservation of the native matrix, which conserves its intrinsic functionality, including structural integrity and the biological composition of essential signaling molecules [50]. ...
Article
Full-text available
Purpose of This Review Skin grafting is a surgical procedure that involves replacing damaged or missing skin with healthy skin. This technique helps protect wounds, promotes healing, and enhances functionality and appearance. Skin grafting can be beneficial in treating burns, traumatic injuries, chronic ulcers, surgical wounds, and congenital defects, among others. Recent Findings A range of cellular and tissue-based products (CTPs) can be employed, either in conjunction with autologous skin grafts or independently, to facilitate wound healing. Human skin allografts, sourced from donated human skin, often obtained from cadavers, serve as a valuable resource for wound protection. Allogeneic matrices, comprising neonatal fibroblasts or membranes, alongside chorion, amnion, and other placental products, provide a means to accelerate the wound healing process. Composite matrices, which combine human keratinocytes, fibroblasts, and xenogeneic collagen, provide a solution to replicate the complexity of natural skin. Moreover, acellular matrices derived from xenogeneic collagen or tissue offer a versatile platform for tissue regeneration. Conclusion Skin grafting is a complex procedure that requires careful planning and postoperative care. Success depends on factors like the type of graft, wound management, and overall health of the patient. Skin grafting has evolved with advancements in surgery, anesthesia, and wound care and remains a crucial technique for restoring function and appearance.
... Existing treatment modalities for managing low back pain include non-pharmacological conservative modalities, such as physical therapy, acupuncture, and chiropractic care; pharmacological agents, such as oral narcotics and non-steroidal antiinflammatory drugs, and injections for medial nerve blocks consisting of local anesthetics and steroids; and minimally invasive procedures, such as radiofrequency ablation [2,6,15,16]. However, these modalities above have shortcomings, including limited long-term amelioration of symptoms and side effects [2,6,17]. ...
Article
Full-text available
Low back pain affects millions of people, creating an enormous financial burden on the global healthcare system. Traditional treatment modalities are short-lived and have shortcomings. Recently, orthobiologics, including extracellular vesicles or exosomes derived from mesenchymal stem cells, have markedly increased for managing musculoskeletal conditions. Here, the primary aim is to review the outcomes of clinical studies using extracellular vesicles or exosomes for treating low back pain. Numerous databases (Scopus, PubMed, Web of Science, Embase, and Google Scholar) were searched using terms for the intervention ‘exosomes’ and the treatment ‘low back pain’ for studies published in English to March 18, 2024. Articles utilizing exosomes for the management of low back pain were included. Articles not utilizing exosomes, not explicitly stating the presence of exosomes in their formulation, or not targeting low back pain were excluded. Two articles that met our pre-defined criteria were included in this review. The results showed that administering extracellular vesicles or exosomes is safe and potentially effective in patients suffering from low back pain. Yet, more sufficiently powered, multi-center, prospective, randomized, and non-randomized trials with longer follow-up are essential to assess the long-term safety and efficacy of extracellular vesicles or exosomes derived from various sources and to support its routine clinical use for managing low back pain.
... Hypothermic storage resulted in a significant decrease in matrix thickness; however, this is expected as a result of processing. Dehydration and loss of water content in dCM resulted in an altered ECM structure and a substantial decrease in matrix thickness, and it been previously reported that dehydration results in a compromised ECM structure and a substantial loss in growth factor and cytokine concentrations [14,25]. Ultrastructural changes as a result of dehydration may result in the formation of physical bonds between collapsed ECM molecules, potentially altering degradation characteristics and the potential for cellular invasion [23]. ...
Article
Full-text available
Placental membranes have been widely studied and used clinically for wound care applications, but there is limited published information on the benefits of using the chorion membrane. The chorion membrane represents a promising source of placental-derived tissue to support wound healing, with its native composition of extracellular matrix (ECM) proteins and key regulatory proteins. This study examined the impact of hypothermic storage on the structure of chorion membrane, ECM content, and response to degradation in vitro. Hypothermically stored chorion membrane (HSCM) was further characterized for its proteomic content, and for its functionality as a scaffold for cell attachment and proliferation in vitro. HSCM retained the native ECM structure, composition, and integrity of native unprocessed chorion membrane and showed no differences in response to degradation in an in vitro wound model. HSCM retained key regulatory proteins previously shown to be present in placental membranes and promoted the attachment and proliferation of fibroblasts in vitro. These data support the fact that hypothermic storage does not significantly impact the structure and characteristics of the chorion membrane compared to unprocessed tissue or its functionality as a scaffold to support tissue growth.
... Hence, human amniotic membrane undergone preservation procedures retain its structural properties and effectiveness as a biological dressing (Maral et al. 1999). Cooke et al. showed that cryopreservation better preserves the structural and biochemical integrity of the amniotic membrane compared to dehydration (Cooke et al. 2014). Long-term cryopreservation of AM with 50% glycerol did not significantly impair sterility, histological properties, or biological properties, which are considered as necessary for its clinical efficacy . ...
Article
Full-text available
Amniotic membrane (AM), the innermost layer of the placenta, is an exceptionally effective biomaterial with divers applications in clinical medicine. It possesses various biological functions, including scar reduction, anti-inflammatory properties, support for epithelialization, as well as anti-microbial, anti-fibrotic and angio-modulatory effects. Furthermore, its abundant availability, cost-effectiveness, and ethical acceptability make it a compelling biomaterial in the field of medicine. Given the potential unavailability of fresh tissue when needed, the preservation of AM is crucial to ensure a readily accessible and continuous supply for clinical use. However, preserving the properties of AM presents a significant challenge. Therefore, the establishment of standardized protocols for the collection and preservation of AM is vital to ensure optimal tissue quality and enhance patient safety. Various preservation methods, such as cryopreservation, lyophilization, and air-drying, have been employed over the years. However, identifying a preservation method that effectively safeguards AM properties remains an ongoing endeavor. This article aims to review and discuss different sterilization and preservation procedures for AM, as well as their impacts on its histological, physical, and biochemical characteristics.
Article
Purpose The purpose of this study was to investigate the efficacy and indications of using dried gamma ray-sterilized amniotic membrane (AM) transplantation with bandage contact lenses for the in-office sutureless treatment of various ocular surface diseases. Methods This study retrospectively included 56 eyes of 52 patients with various ocular surface diseases treated with sutureless dehydrated AM-assisted therapeutic contact lenses. The patients were followed up and assessed 1, 2, and 4 weeks after the treatment, and the therapeutic contact lenses were removed after 1 or 2 weeks. The size of the corneal lesion and the degree of pain experienced by each patient before and after the procedure were measured. Corneal re-epithelization rate and clinical aspects related to re-epithelialization were evaluated. Results Complete corneal re-epithelialization of the epithelial defect was observed in 40 eyes. The complete re-epithelialization rates of persistent epithelial defect caused by infectious ulcers, neurotrophic ulcers, ulcers due to burn, toxic keratopathy, previous penetrating keratoplasty or other corneal surgeries, and severe dry eye disease were 69.2%, 80.0%, 77.8%, 100%, 75.0%, and 100%, respectively. Five patients with uncontrolled infectious ulcers, neurotrophic ulcer, bullous keratopathy, and ulcers associated with rheumatic disease did not show significant improvement. Patient pain scores significantly decreased after the procedure. Conclusions Dehydrated AM transplantation with a bandage contact lens is minimally invasive and easily applicable through in-office procedures. This approach is expected to be more actively applied in various ocular surface disorders, not only in severe corneal diseases but also in relatively mild corneal surface diseases.
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.
Article
Full-text available
In the present study we demonstrate that human monocytes activated by lipopolysaccharides (LPS) were able to produce high levels of interleukin 10 (IL-10), previously designated cytokine synthesis inhibitory factor (CSIF), in a dose dependent fashion. IL-10 was detectable 7 h after activation of the monocytes and maximal levels of IL-10 production were observed after 24-48 h. These kinetics indicated that the production of IL-10 by human monocytes was relatively late as compared to the production of IL-1α, IL-1β, IL-6, IL-8, tumor necrosis factor Oi(TNFα), and granulocyte colony-stimulating factor (G-CSF), which were all secreted at high levels 4-8 h after activation. The production of IL-10 by LPS activated monocytes was, similar to that of IL-1α, IL-1β, IL-6, IL-8, TNFα, granulocyte-macrophage colony-stimulating factor (GM-CSF), and G-CSF, inhibited by IL-4. Furthermore we demonstrate here that IL-10, added to monocytes, activated by interferon γ(IFN-γ), LPS, or combinations of LPS and IFN-γ at the onset of the cultures, strongly inhibited the production of IL-1α, IL-1β, IL-6, IL-8, TNFα, GM-CSF, and G-CSF at the transcriptional level. Viral-IL-10, which has similar biological activities on human cells, also inhibited the production of TNFα and GM-CSF by monocytes following LPS activation. Activation of monocytes by LPS in the presence of neutralizing anti-IL-10 monoclonal antibodies resulted in the production of higher amounts of cytokines relative to LPS treatment alone, indicating that endogenously produced IL-10 inhibited the production of IL-1α, IL-1β, IL-6, IL-8, TNFα, GM-CSF, and G-CSF. In addition, IL-10 had autoregulatory effects since it strongly inhibited IL-10 mRNA synthesis in LPS activated monocytes. Furthermore, endogenously produced IL-10 was found to be responsible for the reduction in class II major histocompatibility complex (MHC) expression following activation of monocytes with LPS. Taken together our results indicate that IL-10 has important regulatory effects on immunological and inflammatory responses because of its capacity to downregulate class II MHC expression and to inhibit the production of proinflammatory cytokines by monocytes.
Article
Full-text available
The use of amniotic membrane (AM) to modulate wound healing and promote regeneration is increasing, but to date there has been no comprehensive study directly comparing the structural and biological properties of fresh and cryopreserved AM. Thus, in this study we compared fresh AM and fresh amniochorion to cryopreserved tissues. Histochemical staining confirmed that the cryopreservation process did not dramatically alter the tissue architecture nor collagen and glycosaminoglycan density. Biochemically, cryopreservation reduced total protein and human serum albumin contents, but retained high molecular weight hyaluronic acid species including the heavy chain-hyaluronic acid complex that is known to exert anti-inflammatory and anti-scarring effects. Cryopreserved and fresh AM extracts similarly suppressed viability and proliferation of RAW264.7 macrophages, and inhibited the transforming growth factor beta 1 promoter activity in corneal fibroblasts. These results collectively indicate that cryopreservation effectively preserves histological, biochemical, and functional properties of the AM tissue.
Article
Full-text available
Wound healing is the physiologic response to tissue trauma proceeding as a complex pathway of biochemical reactions and cellular events, secreted growth factors, and cytokines. Extracellular matrix constituents are essential components of the wound repair phenomenon. Firstly, they create a provisional matrix, providing a structural integrity of matrix during each stage of healing process. Secondly, matrix molecules regulate cellular functions, mediate the cell-cell and cell-matrix interactions, and serve as a reservoir and modulator of cytokines and growth factors' action. Currently known mechanisms, by which extracellular matrix components modulate each stage of the process of soft tissue remodeling after injury, have been discussed.
Article
Full-text available
HC-HA, a complex formed by the covalent linkage between HC1 from inter-α-trypsin inhibitor (IαI) and hyaluronic acid (HA), purified from human amniotic membrane (AM) is responsible for AM anti-inflammatory, anti-scarring, and anti-angiogenic actions. This HC-HA complex is produced by constitutive expression of TNF-stimulated gene-6 and endogenous production of IαI by AM cells. Pentraxin 3 (PTX3), a prototypic long pentraxin that plays a non-redundant role in innate immunity against selected pathogens also helps stabilize HC-HA to ensure female fertility. Herein, we noted strong positive PTX3 staining in the AM epithelium and compact stroma. PTX3 was constitutively expressed and secreted by cultured AM epithelial and stromal cells and further greatly upregulated by TNF and IL-1β. Using an agarose overlay to trap the HA-containing matrix, HC-HA/PTX3 complex was formed analyzed by Westernblot by AM cells but not human skin fibroblasts, despite being cultured in the presence of serum and TNF. However, exogenous PTX3 helps human skin fibroblasts form HC-HA/PTX3 complex with an agarose overlay. Furthermore, PTX3 can be co-immuneprecipitated with HC-HA complex from agarose-overlaid AM cell extracts by anti-human IαI antibody. Such HC-HA/PTX3 complex can be reconstituted in vitro and exhibit similar effects reported for AM HC-HA/PTX3 on polarization of M2 macrophages. The tight binding between PTX3 and AM HC-HA withstands four runs of CsCl ultracentrifugation in the presence of 4 M GnHCl. These results indicate that PTX3 is constitutively expressed and secreted by AM cells as an integral component of AM HC-HA/PTX3 complex, and contributes to the biological function of AM HC-HA/PTX3.
Article
Full-text available
Purpose: Heavy chain-hyaluronic acid (HC-HA)/PTX3 purified from human amniotic membrane (AM) was previously observed to suppress inflammatory responses in vitro. We now examine whether HC-HA/PTX3 is able to exert a similar effect in vivo, using murine models for keratitis and corneal allograft rejection. Methods: The in vitro effect of HC-HA/PTX3 was tested using OTII ovalbumin (OVA) transgenic, purified CD4(+) T cells, or IFN-γ/lipopolysaccharide (LPS)-stimulated RAW264.7 cells. Cytokine production was measured by ELISA, while cell surface markers and cell proliferation were determined by flow cytometry. In vivo effects of HC-HA/PTX3 were analyzed by quantifying the recruitment of enhanced green fluorescence-labeled macrophages and by measuring the expression of arginase 1 (Arg-1), IL-10, and IL-12 in LPS-induced keratitis in the macrophage Fas-induced apoptosis (Mafia) mouse. The effect of corneal allograft survival in a complete major histocompatibility complex (MHC) mismatched mouse model was assessed by grading corneal opacification. Results: In vitro studies demonstrated that HC-HA/PTX3 significantly enhanced the expansion of FOXP3 T cells and suppressed cell proliferation and protein expression of IFN-γ, IL-2, CD25, and CD69 in activated CD4(+) T cells. Furthermore, immobilized HC-HA/PTX3 significantly upregulated IL-10 gene expression but downregulated that of IL-12 and IL-23 in activated RAW264.7 cells. Finally, in vivo subconjunctival injection of HC-HA/PTX3 significantly prolonged corneal allograft survival, suppressed macrophage infiltration, and promoted M2 polarization by upregulating Arg-1 and IL-10 but downregulating IL-12. Conclusions: HC-HA/PTX3 can suppress inflammatory responses in vivo by modulating both innate and adaptive immunity of macrophages and CD4(+) T cells.
Article
Full-text available
Despite the known anti-inflammatory effect of amniotic membrane, its action mechanism remains largely unknown. HC-HA complex (HC-HA) purified from human amniotic membrane consists of high molecular weight hyaluronic acid (HA) covalently linked to the heavy chain (HC) 1 of inter-α-trypsin inhibitor. In this study, we show that soluble HC-HA also contained pentraxin 3 and induced the apoptosis of both formyl-Met-Leu-Phe or LPS-activated neutrophils and LPS-activated macrophages while not affecting the resting cells. This enhanced apoptosis was caused by the inhibition of cell adhesion, spreading, and proliferation caused by HC-HA binding of LPS-activated macrophages and preventing adhesion to the plastic surface. Preferentially, soluble HC-HA promoted phagocytosis of apoptotic neutrophils in resting macrophages, whereas immobilized HC-HA promoted phagocytosis in LPS-activated macrophages. Upon concomitant LPS stimulation, immobilized HC-HA but not HA polarized macrophages toward the M2 phenotype by down-regulating IRF5 protein and preventing its nuclear localization and by down-regulating IL-12, TNF-α, and NO synthase 2. Additionally, IL-10, TGF-β1, peroxisome proliferator-activated receptor γ, LIGHT (TNF superfamily 14), and sphingosine kinase-1 were up-regulated, and such M2 polarization was dependent on TLR ligation. Collectively, these data suggest that HC-HA is a unique matrix component different from HA and uses multiple mechanisms to suppress M1 while promoting M2 phenotype. This anti-inflammatory action of HC-HA is highly desirable to promote wound healing in diseases heightened by unsuccessful transition from M1 to M2 phenotypes. Background: HC-HA is a unique anti-inflammatory matrix different from hyaluronic acid (HA). Results: Soluble HC-HA induces apoptosis of inflammatory neutrophils and macrophages, and immobilized HC-HA promotes M2 polarization upon LPS/TLR ligation while both enhancing macrophage phagocytosis. Conclusion: HC-HA exerts its anti-inflammatory action using multiple mechanisms. Significance: HC-HA is the first known matrix component to polarize M2b.
Article
Full-text available
Purpose. The purpose of this study was to evaluate the efficacy of dried human amniotic membrane (AM) in reducing the postoperative inflammatory response and scarring after strabismus surgery. Methods. The inflammatory response at the extraocular muscle reattachment site was analyzed after superior rectus (SR) resection in 12 rabbits. Dried human AM (Ambiodry2) was applied between the resected SR muscle plane and Tenon's capsule of the left eyes of rabbits. As a control, the right eyes of rabbits underwent SR resection only. The surgeon randomly ordered which eye gets operated first during the experiment. Two weeks later, enucleation was performed. Six sagittal sections were made for each eye at the insertion of the SR muscle. The grade of postoperative inflammation and the presence of fibrosis were evaluated in histological examinations. Results. There was no statistically significant difference in the intensity of inflammation and fibrous proliferation between the eyes treated with dried human AM after SR resection and those treated with SR resection only. Conclusions. The use of dried human AM was not effective in controlling the postoperative inflammation and scarring in rabbit eyes after extraocular muscle surgery. However, this may be due to the devitalized dry preparation of human AM (Ambiodry2), which may have lost the expected anti-inflammatory and anti-scarring properties, and further studies on humans may be necessary.
Article
There is circumstantial evidence that protein denaturation occurs in cells during heat shock at hyperthermic temperatures and that denatured or damaged protein is the primary inducer of the heat shock response. However, there is no direct evidence regarding the extent of denaturation of normal cellular proteins during heat shock. Differential scanning calorimetry (DSC) is the most direct method of monitoring protein denaturation or unfolding. Due to the fundamental parameter measured, heat flow, DSC can be used to detect and quantitate endothermic transitions in complex structures such as isolated organelles and even intact cells. DSC profiles with common features are obtained for isolated rat hepatocytes, liver homogenate, and Chinese hamster lung V79 fibroblasts. Five main transitions (A-E), several of which are resolvable into subcomponents, are observed with transition temperatures (Tm) of 45-98 degrees C. The onset temperature is approximately 40 degrees C, but some transitions may extend as low as 37-38 degrees C. In addition to acting as the primary signal for heat shock protein synthesis, the inactivation of critical proteins may lead to cell death. Critical target analysis implies that the rate limiting step of cell killing for V79 cells is the inactivation of a protein with Tm = 46 degrees C within the A transition. Isolated microsomal membranes, mitochondria, nuclei, and a cytosolic fraction from rat liver have distinct DSC profiles that contribute to different peaks in the profile for intact hepatocytes. Thus, the DSC profiles for intact cells appears to be the sum of the profiles of all subcellular organelles and components. The presence of endothermic transitions in the isolated organelles is strong evidence that they are due to protein denaturation. Each isolated organelle has an onset for denaturation near 40 degrees C and contains thermolabile proteins denaturing at the predicted Tm (46 degrees C) for the critical target. The extent of denaturation at any temperature can be approximately by the fractional calorimetric enthalpy. After scanning to 45 degrees C at 1 degree C/min and immediately cooling, a relatively mild heat shock, an estimated fraction denaturation of 4-7% is found in hepatocytes, V79 cells, and the isolated organelles other than nuclei, which undergo only 1% denaturation because of the high thermostability of chromatin. Thus, thermolabile proteins appear to be present in all cellular organelles and components, and protein denaturation is widespread and extensive after even mild heat shock.
Article
Interleukin-12 (IL-12) is a heterodimeric cytokine produced mostly by phagocytic cells in response to bacteria, bacterial products, and intracellular parasites, and to some degree by B lymphocytes. IL-12 induces cytokine production, primarily of IFN-gamma from NK and T cells, acts as a growth factor for activated NK and T cells, enhances the cytotoxic activity of NK cells, and favors cytotoxic T lymphocyte generation. In vivo IL-12 acts primarily at three stages during the innate resistance/adaptive immune response to infection: 1. Early in the infection, IL-12 is produced and induces production from NK and T cells of IFN-gamma, which contributes to phagocytic cell activation and inflammation; 2. IL-12 and IL-12-induced IFN-gamma favor Th1 cell differentiation by priming CD4(+) T cells for high IFN-gamma production; and 3. IL-12 contributes to optimal IFN-gamma production and to proliferation of differentiated Th1 cells in response to antigen. The early preference expressed in the immune response depends on the balance between IL-12, which favors Th1 responses, and IL-4, which favors Th2 responses. Thus, IL-12 represents a functional bridge between the early nonspecific innate resistance and the subsequent antigen-specific adaptive immunity.
Article
To investigate the feasibility and effect of human amniotic membrane in prevention of tendon adhension after tendon sheat defect repair. The amniotic membrane in size of 1.5 cm x 1.0 cm was harvested from human placenta which was voluntary donated from maternal after cesarean. Forty healthy male Leghorn chicken (aged 3-6 months) were selected, weighing (1.86 +/- 0.04) kg. The model of flexor digitorum profundus tendon and tendon sheath defects was established at the third toe. After repair of the flexor digitorum profundus tendon, the human amniotic membrane was used to repair the tendon sheath defect in the right foot (group A), but tendon sheath defect was not repaired in the left foot (group B) . At 1, 2, 4, and 6 weeks after operation, the gross and histological observations were done; the degree of tendon adhesions was graded according to Tang's tendon adhesion general observation grading standards; and the biomechanical properties (tendon slip length and total flexion angle) were tested. All animals survived after operation and incisions healed. Gross and histological observations showed that the new tendon sheath formed with time passing after operation in groups A and B; new tendon sheath was more maturer and smoother in group A than in group B. The degree of tendon adhesions in group A was significantly less than that in group B (P < 0.05) at 1 and 6 weeks after operation. The biomechanical test results showed there was no significant difference in the tendon slip length between 2 groups at 1 and 2 weeks after operation (P > 0.05), but the tendon slip length of group A was significantly longer than that of group B at 4 and 6 weeks after operation (P < 0.05). The total flexion angle of group A was significantly smaller than that of group B at 1, 2, 4, and 6 weeks after operation (P < 0.05). It is effective in the prevention of tendon adhesion to use the amniotic membrane for repairing the tendon sheath defect, which is beneficial to recovery of the tendon sliding function.