PreprintPDF Available

Accelerated Burn Healing in a Mouse Experimental Model by α-gal Nanoparticles

Authors:
Preprints and early-stage research may not have been peer reviewed yet.

Abstract and Figures

Macrophages play a pivotal role in the process of healing burns. One of the major risks in the course of burns healing, in the absence of regenerating epidermis, is infections which greatly contribute to morbidity and mortality in such patients. Therefore, it is widely agreed that accelerating recruitment of macrophages into burns may contribute to faster regeneration of the epidermis and thus, decreasing the risk of infections. This review describes a unique method for rapid recruitment of macrophages into burns and activation of these macrophages to mediate accelerated regrowth of the epidermis and healing of burns. The method is based on application of bio-degradable “alpha-gal” nanoparticles to burns. These nanoparticles present multiple alpha-gal epitopes (Gal alpha1-3Gal beta1-4GlcNAc-R) which bind the abundant natural anti-Gal antibody that constitutes ~1% of immunoglobulins in humans. Anti-Gal/alpha-gal nanoparticles interaction activates the complement system, resulting in localized production of the complement cleavage-peptides C5a and C3a that are highly effective chemotactic factors for monocytes derived macrophages. The macrophages recruited into the alpha-gal nanoparticles treated burns are activated following interaction between the Fc portion of anti-Gal coating the nanoparticles and the multiple Fc receptors on macrophages cell membranes. The activated macrophages secrete a variety of cytokines/growth factors that accelerate the regrowth of the epidermis and regeneration of the injured skin, thereby cutting the healing time by half. Studies on healing of thermal injuries in the skin of anti-Gal producing mice, demonstrated a much faster recruitment of macrophages into burns treated with alpha-gal nanoparticles than in control burns treated with saline and healing of the burns within 6 days, whereas healing of control burns takes ~12 days. alpha-Gal nanoparticles are non-toxic, and do not cause chronic granulomas or keloids. These findings suggest that alpha-gal nanoparticles treatment may harness anti-Gal for inducing similar accelerated burn healing effects also in humans.
Content may be subject to copyright.
Review Not peer-reviewed version
Accelerated Burn Healing in a
Mouse Experimental Model by
α
-
gal Nanoparticles
Uri Galili *
Posted Date: 6 September 2023
doi: 10.20944/preprints202309.0375.v1
Keywords: Burn healing; anti-Gal antibody; -gal epitope; -gal nanoparticles; macrophage migration; -gal
therapy; mesenchymal stem cells.
Preprints.org is a free multidiscipline platform providing preprint service that
is dedicated to making early versions of research outputs permanently
available and citable. Preprints posted at Preprints.org appear in Web of
Science, Crossref, Google Scholar, Scilit, Europe PMC.
Copyright: This is an open access article distributed under the Creative Commons
Attribution License which permits unrestricted use, distribution, and reproduction in any
medium, provided the original work is properly cited.
Review
Accelerated Burn Healing in a Mouse Experimental
Model by α-gal Nanoparticles
Uri Galili
Department of Medicine, Rush University Medical College, Chicago, IL 60612, USA; uri.galili@rcn.com;
Tel.: +001-312-753-5997
Abstract: Macrophages play a pivotal role in the process of healing burns. One of the major risks in
the course of burns healing, in the absence of regenerating epidermis, is infections which greatly
contribute to morbidity and mortality in such patients. Therefore, it is widely agreed that
accelerating recruitment of macrophages into burns may contribute to faster regeneration of the
epidermis and thus, decreasing the risk of infections. This review describes a unique method for
rapid recruitment of macrophages into burns and activation of these macrophages to mediate
accelerated regrowth of the epidermis and healing of burns. The method is based on application of
bio-degradable “α-gal” nanoparticles to burns. These nanoparticles present multiple α-gal epitopes
(Gal1-3Gal1-4GlcNAc-R) which bind the abundant natural anti-Gal antibody that constitutes ~1%
of immunoglobulins in humans. Anti-Gal/α-gal nanoparticles interaction activates the complement
system, resulting in localized production of the complement cleavage-peptides C5a and C3a that
are highly eective chemotactic factors for monocytes derived macrophages. The macrophages
recruited into the α-gal nanoparticles treated burns are activated following interaction between the
Fc portion of anti-Gal coating the nanoparticles and the multiple Fc receptors on macrophages cell
membranes. The activated macrophages secrete a variety of cytokines/growth factors that accelerate
the regrowth of the epidermis and regeneration of the injured skin, thereby cuing the healing time
by half. Studies on healing of thermal injuries in the skin of anti-Gal producing mice, demonstrated
a much faster recruitment of macrophages into burns treated with α-gal nanoparticles than in
control burns treated with saline and healing of the burns within 6 days, whereas healing of control
burns takes ~12 days. α-Gal nanoparticles are non-toxic, and do not cause chronic granulomas or
keloids. These ndings suggest that α-gal nanoparticles treatment may harness anti-Gal for
inducing similar accelerated burn healing eects also in humans.
Keywords: burn healing; anti-Gal antibody; α-gal epitope; α-gal nanoparticles; macrophage
migration; α-gal therapy; mesenchymal stem cells
1. Introduction
Macrophages play a pivotal role in the process of wound and burn healing [1–4]. In both types
of healing the M1 macrophages rst debride the injured skin of apoptotic and dead cells and of
intercellular cell matrix. Subsequently, M2 macrophages orchestrate the regeneration of the
epidermis, dermis and hypodermis of the injured skin [2–6]. This is mediated by a wide range of
cytokines/growth factors secreted by these macrophages, including vascular endothelial growth
factor (VEGF) mediating neo-vascularization, epidermal growth factor (EGF) inducing epidermal
re-growth, broblasts growth factor (FGF) recruiting broblasts, and factors recruiting mesenchymal
stem cells (MSC) which contribute to the regeneration of the injured skin [7]. In wounds, incisions,
and contusions, the macrophages mediating healing comprise of both residential macrophages and
monocytes derived macrophages that are recruited by chemotactic factors such as macrophage
inammatory protein-1 (MIP-1), monocyte chemoaractant protein-1 (MCP-1) and regulated on
activation, normal T cell expressed and secreted factor (RANTES) secreted by cells surrounding the
wound [8–11]. However, since in epidermis penetrating burns (burn degrees 2-4) the residential
Disclaimer/Publisher’s Note: The statements, opinions, and data contained in all publications are solely those of the individual author(s) and
contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting
from any ideas, methods, instructions, or products referred to in the content.
Preprints (www.preprints.org) | NOT PEER-REVIEWED | Posted: 6 September 2023 doi:10.20944/preprints202309.0375.v1
© 2023 by the author(s). Distributed under a Creative Commons CC BY license.
2
macrophages are inactivated or killed [1] and since the surface area size of the burns are in many
cases larger than that of wounds, inltration of macrophages into burns and healing of burns may
take longer time than in wounds and regeneration of epidermis in many burns may be slower than
in wounds. This slow re-epithelialization is a major risk factor because of microbial infections that
occur in the absence of intact epidermis. Such infections may result in high morbidity and mortality
following severe burn injuries [6,12–14].
Based on the pivotal role of macrophages in healing of burns, and in view of the immune
suppression of macrophages following burn injury [15,16], it has been suggested that the risk factors
due to slow re-epithelialization might be reduced by accelerating the regrowth of the epidermis over
the burned tissue [3,4,7,15–18]. Several methods of various degrees of diculty have been studied for
accelerating burn healing. These include topical application to burns of autologous MSC [19,20],
autologous cultured epidermal cell grafts [21,22], recombinant human granulocyte-macrophage
colony-stimulating factor (GM-CSF) [23,24], high-density lipoprotein nanoparticles [25], bioactive
molecules delivered in microbers [26,27] and the use of negative pressure wound therapy[28].
The present review oers an alternative method to those mentioned above, for accelerated
healing of burns by inducing rapid recruitment and activation of macrophages in treated burns, by
topical application of a-gal nanoparticles. This method recapitulates the physiologic healing
processes of burns, but the accelerated recruitment of macrophages into treated burns cuts the healing
time by half. The interaction of these nanoparticles with the natural anti-Gal antibody (one of the
most abundant natural antibodies in humans) within burns results in rapid and extensive recruitment
of monocytes derived macrophages into burns. Many of these macrophages polarize into M2
macrophages which orchestrate accelerated healing of burns by localized secretion of angiogenic
factors such as VEGF and growth factors recruiting MSC. This review describes studies that
characterized the anti-Gal antibody and a-gal nanoparticles, the simple production of these
nanoparticles from mammalian red blood cells and the great ecacy of the burn and wound
therapies with a-gal nanoparticles, as observed in the anti-Gal producing mouse experimental model.
2. Anti-Gal and the a-gal Epitope
The method described in this review for accelerating burn healing harnesses the immunologic
potential of the natural anti-Gal antibody which is one of the most abundant natural antibodies in
humans, constituting 1% of serum immunoglobulins [29]. The immune system in humans produces
anti-Gal throughout life in response to antigenic stimulation by some carbohydrate antigens
presented on gastrointestinal bacteria [30,31]. The mammalian antigen recognized by the anti-Gal is
the a-gal epitope (Gala1- 3Galb1-4GlcNAc-R) [32–34]. The a-gal epitope is abundantly expressed on
glycolipids and glycoproteins of nonprimate mammals, lemurs, and New-World monkeys (monkeys
of South America), therefore, these mammals cannot produce anti-Gal [35–37]. In contrast, humans,
apes, and Old-World monkeys (monkeys of Asia and Africa) all lack a-gal epitopes but produce the
natural anti-Gal antibody [35–38]. Incubation of cells presenting a-gal epitope in human serum results
in eective activation of the complement cascade in the serum, because of the binding of serum anti-
Gal to these a-gal epitopes. The ecacy of this complement activation was demonstrated in
xenotransplantation studies. Interaction between anti-Gal and a-gal epitopes on endothelial cells of
pig xenografts was found to result in the activation of the complement system, causing cytolysis of
these cells, collapse of the vascular bed and rapid (hyperacute) rejection of such xenografts in
monkeys or humans [39–42]. Similarly, incubation of enveloped viruses presenting a-gal epitopes in
human serum was found to result in binding of anti-Gal to these epitopes and in activation of the
complement system which leads to complement mediated destruction of such viruses [43–46]. Since
the very potent macrophage directing chemotactic factors C5a and C3a are produced as complement
cleavage peptide byproducts during complement activation, we assumed that binding of serum anti-
Gal to multiple a-gal epitopes on a-gal nanoparticles applied to burns and wounds may result in
extensive recruitment of monocyte derived macrophages to treated skin injury sites.
3. Hypothesis
Preprints (www.preprints.org) | NOT PEER-REVIEWED | Posted: 6 September 2023 doi:10.20944/preprints202309.0375.v1
3
The eective complement activation by anti-Gal binding to a-gal epitopes led us to hypothesize
that topical application of nanoparticles presenting multiple a-gal epitopes (called a-gal nanoparticles
and previously called a-gal liposomes) during the early stages of hemostasis in burns, results in
binding of the natural anti-Gal antibody to these nanoparticles [17,47]. Anti-Gal is present in the uid
lm on the surface of burns together with the complement system proteins, as well as with other
serum proteins which leak from injured capillaries. As detailed below, a-gal nanoparticles are small
size liposomes (~200-300 nm) constructed of a-gal presenting glycolipids that are anchored in the
nanoparticles wall that is comprised of phospholipids and cholesterol (Figure 1A).
The binding of anti-Gal to the multiple a-gal epitopes on the nanoparticles results in activation
of the complement cascade and thus in generation of chemotactic complement cleavage peptides C5a
and C3a (Step 1 in Figure 1B). These chemotactic factors induce extensive migration of neutrophils
and monocytes derived macrophages into the burn area (Step 2 in Figure 1B). In addition, it was
further hypothesized that whereas the neutrophils survive only for few hours in the burn, the
recruited macrophages are long-lived, and they bind the a-gal nanoparticles as a result of interaction
between the Fcg “tail” of anti-Gal bound to the nanoparticles and Fcg receptors (FcgR) on the
macrophages (Step 3 in Figure 1B). It was further hypothesized that the multiple Fcg/FcgR
interactions between anti-Gal coated a-gal nanoparticles and macrophages may activate the recruited
macrophages to secrete various cytokines/growth factors that mediate accelerated migration of
broblasts and MSC into the treated burn, as well as neo-vascularization of the healing burn (Step
4 in Figure 1B), and rapid re-epithelialization. Ultimately, these multiple cytokines/growth factors
secreted by the recruited and activated macrophages may accelerated healing of the a-gal
nanoparticles treated burns in comparison to non-treated burns.
Figure 1. Illustration of an α-gal nanoparticle (A) and hypothesized immune processes induced by α-
gal nanoparticles applied to burns (B). A. The α-gal nanoparticles present multiple α-gal epitopes
(rectangles) on glycolipids that are anchored in the phospholipid bilayer that forms the wall of the
nanoparticle. The nanoparticle wall may also contain cholesterol which stabilizes the wall. The natural
anti-Gal antibody readily binds to these α-gal epitopes (illustrated as rectangles). B. The steps
hypothesized to occur in burns after application of α-gal nanoparticles: Step 1- Anti-Gal binding to α-
gal nanoparticles activates the complement system. Step 2- The complement cleavage peptides C5a
and C3a formed as a result of complement activation, function as chemotactic factors that direct
extensive and rapid recruitment of monocytes derived macrophages into the treated burns. Step 3-
The recruited macrophages interact via their Fcγ receptors (FcγR) with the Fcγ portion (tail) of anti-
Gal coating the α-gal nanoparticles. Step 4- The Fcγ/FcγR interactions activate the macrophages to
Preprints (www.preprints.org) | NOT PEER-REVIEWED | Posted: 6 September 2023 doi:10.20944/preprints202309.0375.v1
4
secrete cytokines/growth factors that induce accelerated healing of the treated burns. Reproduced
from ref. 47 with permission.
4. Preparation of a-gal Nanoparticles
A relatively simple way for preparing a-gal nanoparticles is by extraction of their components
from the membranes (ghosts) of rabbit red blood cells (RBC) in a mixture of chloroform and methanol
[17]. The reason for using these RBC is that they present as many as 2x106 a-gal epitopes per RBC, an
amount that is several folds higher than any other mammalian RBC studied [35]. After the rabbit RBC
are lysed in water and their membranes are washed for the removal of hemoglobin, the RBC
membranes are incubated overnight in a solution of chloroform:methanol 1:2 with constant stirring,
resulting in the extraction of phospholipids, glycolipids and cholesterol from these membranes,
whereas all proteins are denatured and removed from the solution by ltration. The solution
containing the extracted molecules is dried and the mixture of phospholipids, glycolipids and
cholesterol is resuspended in saline by extensive sonication. This sonication results in formation of a
suspension of submicroscopic liposomes (~100-300 nm) with walls comprised of phospholipid and
cholesterol and studded with multiple a-gal epitopes in the form of anchored a-gal glycolipids
(Figures 1A). These submicroscopic liposomes originally called a-gal liposomes [17,48] have been
subsequently referred to as a-gal nanoparticles [18,47] to indicate that they do not contain any
substance in their lumen.
The a-gal nanoparticles were found to present ~1014 a-gal epitopes/mg nanoparticles [18].
Processed 500 ml packed rabbit RBC were found to yield ~6 grams of a-gal nanoparticles. Because of
their small size, a-gal nanoparticles suspensions can be sterilized by ltration through a 0.4 mm lter.
It is of note that a-gal nanoparticles may be prepared also from synthetic a-gal glycolipids,
phospholipids and cholesterol by similar mixing and sonication processes. The a-gal nanoparticles
are highly stable and can be kept as frozen suspensions for >10 years, at 4oC for >2 years and as dried
nanoparticles on wound dressings kept at room temp. for >1 year (unpublished observations).
Stability of the stored a-gal nanoparticles could be conrmed by their ability to bind anti-Gal in
amounts like those measured immediately after production. Topical application of a-gal
nanoparticles to burns and wounds can be performed by various methods including the use of
nanoparticles suspensions in saline or PBS, nanoparticles dried on wound dressings, aerosol
suspensions and suspensions in hydrogels.
5. Experimental Animal Models
Studies of anti-Gal associated therapies cannot be performed in standard animal experimental
models such as mice, rats, rabbits, pigs or guinea pigs because these animals, like other non-primate
mammals, synthesize a-gal epitopes [35,36]. Therefore, such mammals are immunotolerant to the a-
gal epitope and cannot produce the anti-Gal antibody [35]. However, the two experimental non-
primate mammalian models available for studying anti-Gal associated therapies have been mice
[49,50] and pigs [51,52] in which the GGTA1 gene coding for the glycosyltransferase synthesizing a-
gal epitope “a1,3galactosyltransferase”, was disrupted (i.e., knocked out). These
a1,3galactosyltransferase knockout (GT-KO) mice [17,18] and pigs [53–55] lack the ability to
synthesize a-gal epitopes and thus, can produce the anti-Gal antibody. Whereas GT-KO pigs produce
the natural anti-Gal antibody, as humans do, GT-KO mice do not naturally produce this antibody
since they do not develop gastrointestinal bacterial ora that may immunize them, because they are
kept in a sterile environment and receive sterile food. Nevertheless, GT-KO mice readily produce the
anti-Gal antibody following several immunizations with xenogeneic cells or tissues presenting a-gal
epitopes, such as pig kidney membranes (PKM) homogenate [56].
6. In vitro Eects of a-gal Nanoparticles on Macrophages
Some of the steps of anti-Gal/a-gal nanoparticles interaction, described in the hypothesis
illustrated in Figure 1B, could be demonstrated in vitro in studies described in Figure 2. Step 1 of anti-
Gal binding to a-gal epitopes on a-gal nanoparticles was demonstrated by the specic binding of
Preprints (www.preprints.org) | NOT PEER-REVIEWED | Posted: 6 September 2023 doi:10.20944/preprints202309.0375.v1
5
monoclonal anti-Gal antibody to these nanoparticles (Figure 2A). A similar binding was observed
with serum anti-Gal from anti-Gal producing GT-KO mice that interacts woth a-gal nanoparticles
(Figure 2B). In the absence of a-gal epitopes on the nanoparticles, no binding of the monoclonal anti-
Gal antibody was observed [17].
Figure 2. In vitro demonstration of anti-Gal binding to α-gal nanoparticles and the resulting eects
on macrophages. A. Binding of monoclonal anti-Gal IgM antibody to α-gal epitopes on α-gal
nanoparticles. The thin line represents the IgM isotype control. B. As in Figure 2A, using anti-Gal
IgG in serum of α1,3galactosyltransferase knockout (GT-KO) mouse producing anti-Gal. The thin line
represents the IgG isotype control. C and D. Binding of anti-Gal-coated α-gal nanoparticles to
adherent GT-KO pig macrophages as shown by scanning electron microscopy (SEM), after 2 hours
incubation of anti-Gal coated nanoparticles with the macrophages at room temp., followed by
washings to remove nonadherent nanoparticles. The surfaces of representative macrophages are
covered with α-gal nanoparticles that have the shape of small spheres. The size of the α-gal
nanoparticles is 100-300 nm. E. GT-KO mouse peritoneal macrophages secretion of VEGF following
incubation with anti-Gal-coated α-gal nanoparticles (closed columns), α-gal nanoparticles without
anti-Gal (grey columns), or as macrophages alone (open columns, background levels). VEGF secretion
by the macrophages was measured by ELISA in culture media after 24 or 48 hours. Data with
macrophages from 4 GT-KO mice and their means+S.D. Modied from ref. 47 with permission.
Step 3 in the hypothesis in Figure 1B predicts binding of anti-Gal coated α-gal nanoparticles to
macrophages via Fcg/FcgR interaction. This binding is demonstrated by scanning electron
microscopy (SEM) in Figure 2C,D. Macrphages lacking a-gal epitopes were generated in vitro by
culturing of monocytes obtained from blood of GT-KO pigs. These macrophages were co-incubated
for 2 hours at room temp. with anti-Gal-coated α-gal nanoparticles. Such incubation resulted in
extensive binding of α-gal nanoparticles to the macrophages, shown as the multiple small spheres
Preprints (www.preprints.org) | NOT PEER-REVIEWED | Posted: 6 September 2023 doi:10.20944/preprints202309.0375.v1
6
(size of 100–300nm) covering the surface of the two macrophages (Figure 2C,D). In the absence of
anti-Gal on the α-gal nanoparticles, no binding of these nanoparticles to macrophages was observed
[47]. Step 4 in the hypothesis in Figure 1B predicted that the binding of anti-Gal coated a-gal
nanoparticles to recruited macrophages via Fcγ/FcγR interaction may generate signals that activate
a variety of cytokines/growth factors producing genes that orchestrate accelerated healing of a-gal
nanoparticles treated burns. Possible activation of macrophages by anti-Gal coated a-gal
nanoparticles was studied with GT-KO mouse macrophages incubated for 24-48 hours at 37°C, alone
or with α-gal nanoparticles coated with anti-Gal or lacking the antibody. The secretion of VEGF by
the macrophages was measured in the tissue culture medium after 24 and 48 hours of co-incubation.
Macrophages co-incubated with α-gal nanoparticles lacking anti-Gal, secreted only background level
of VEGF, as determined by ELISA measuring this cytokine (Figure 2E). However, co-incubation of
the macrophages with anti-Gal-coated α-gal nanoparticles for 24 and 48 hours resulted in elevated
secretion of VEGF by the activated macrophages, at levels that were signicantly higher than the
background levels (Figure 2E) [48]. These ndings indicated that anti-Gal mediated binding of a-gal
nanoparticles to cultured macrophages indeed induces these macrophages to secrete VEGF.
7. In vivo Eects of a-gal Nanoparticles on Macrophages
A crucial step in the hypothesis in Figure 1B is Step 2 which predicts that anti-Gal binding to a-
gal nanoparticles applied to burns activates the complement system, resulting in the formation of
complement cleavage chemotactic peptides C5a and C3a. These peptides direct a rapid and extensive
recruitment of monocytes derived macrophages to the treated burn. The occurrence of Step 2 was
studied by intradermal injection of 10 mg a-gal nanoparticles in anti-Gal producing GT-KO mice (i.e.,
GT-KO mice immunized with PKM) and microscopic evaluation of macrophages in the injection
site at various time points. The rst eect of such injection was the accumulation of many neutrophils,
observed at the injection site within 12 hours post injection [48]. These neutrophils are also
chemotactically recruited by C5a and C3a generated by anti-Gal/a-gal nanoparticles interaction.
However, after 24 hours, most neutrophils disappeared, and multiple mononuclear cells were
observed migrating to the injection site (Figure 3A). The number of macrophages increased after 4
days and as expected, they all were immunostained by the macrophage specic antibody F4/80
(Figure 3B). Quantitative real time PCR (qRT-PCR) of skin specimen containing the recruited
macrophages displayed activation of genes encoding for broblast growth factor (FGF), interleukin
1 (IL1), platelet derived growth factor (PDGF), and colony stimulating factor (CSF) [48].
The number of recruited macrophages further increased by Day 7 (Figure 3C). These
macrophages had large size and ample cytoplasm, characteristic of activated macrophages (Figure
3C,D) [48]. Large numbers of recruited macrophages were observed at the injection site, even on Day
14. However, by Day 21 post injection of the a-gal nanoparticles, all macrophages disappeared from
the injection site and the skin in that area displayed normal structure with no granuloma, chronic
inammatory response, or keloid formation [48]. Intradermal injection of a-gal nanoparticles together
with cobra venom factor (a complement activation inhibitor), with saline, or with nanoparticles
lacking a-gal epitopes (i.e., nanoparticles produced from GT-KO pig RBC) all resulted in no
signicant recruitment of macrophages to the injection site [48]. Similarly, intradermal injection of a-
gal nanoparticles into wild-type (WT) mice (i.e., mice lacking the anti-Gal antibody) resulted in no
macrophage recruitment. These observations clearly demonstrated the ability of a-gal nanoparticles
to induce extensive and rapid recruitment of macrophages by binding of the anti-Gal antibody and
activation of the complement system which generates the potent chemotactic complement cleavage
chemotactic peptides C5a and C3a.
Preprints (www.preprints.org) | NOT PEER-REVIEWED | Posted: 6 September 2023 doi:10.20944/preprints202309.0375.v1
7
Figure 3. Macrophages recruitment by intradermally injected α-gal nanoparticles (10 mg), in anti-Gal
producing GT-KO mice. A. Macrophage recruitment 24 hours following injection of α-gal
nanoparticles. The empty oval area is the space formed by the injection of α-gal nanoparticles. The
nanoparticles were dissolved by alcohol during staining with hematoxylin & eosin staining (H&E
×100). B. Macrophages at 4 days post injection, identied by immunostaining with the F4/80 antibody
coupled to peroxidase (HRP) (×200). C. The injection area after 7 days. The site is full of many large
macrophages containing vacuoles with morphology characteristic of activated macrophages (H&E ×
400). D. Individual macrophages, similar to those in (C) were harvested from polyvinyl alcohol (PVA)
sponge disc containing α-gal nanoparticles. The PVA sponge discs were explanted 6 days post
subcutaneous implantation into anti-Gal producing GT-KO mice. The multiple vacuoles observed in
the macrophages are of internalized anti-Gal coated α-gal nanoparticles (Wright staining, ×1000).
Modied from ref. 47 with permission.
8. Macrophages Recruited by a-gal Nanoparticles are M2 Further Recruiting MSC
Analysis of the characteristics of macrophages recruited by a-gal nanoparticles in anti-Gal
producing GT-KO mice could be further performed by subcutaneous implantation of biologically
inert sponge discs (made of polyvinyl alcohol- PVA, 10 mm diameter, 3 mm thickness) that contained
10 mg α-gal nanoparticles. The PVA sponge discs were explanted on Day 6 or Day 9. The cells
harvested from these sponge discs had the morphology of large macrophages as those presented in
Figure 3D. Each of the PVA sponge discs contained at those time points ~0.4x10
6
and ~0.6x10
6
inltrating cells, respectively, whereas sponge discs with only saline contained ~0.02x10
6
and
~0.04x10
6
cells, respectively [17]. Immunostaining and ow cytometry analysis of the cells recruited
by a-gal nanoparticles indicated that most of them (>90%) expressed the macrophage markers CD11b
and CD14 (Figure 4A). In contrast, no signicant proportion of the inltrating cells displayed surface
markers of CD4+ T cells, CD8+ T cells, or of B cells (i.e., lymphocytes presenting CD20+ cell marker)
[17].
Preprints (www.preprints.org) | NOT PEER-REVIEWED | Posted: 6 September 2023 doi:10.20944/preprints202309.0375.v1
8
Figure 4. Analysis of cells migrating into PVA sponge discs following anti-Gal/α-gal nanoparticles
interaction. A. Flow cytometry analysis of the recruited cells, retrieved from PVA sponge discs
containing 10 mg α-gal nanoparticles, 6 days post-subcutaneous implantation. Most of the recruited
cells were macrophages expressing CD11b and CD14 cell markers, whereas no signicant inltration
of T cells, or B cells was observed (representative data of ve mice). B. Analysis of recruited
macrophages polarization. The large size macrophages (CD11b
pos
/F4/80
pos
)
were positive also for IL-
10 and Arginase-1 but were negative for IL-12. This implied that the majority of the recruited cells
were M2 macrophages. C and D. Cell colonies formed by what seemed to be MSC recruited by
macrophages migrating into PVA sponge discs containing α-gal nanoparticles and harvested 6 days
post subcutaneous implantation. The colonies were observed on Day 5 post culturing. E and F.
Expression of MSC markers Sca-1 and CD-29 respectively, by cells harvested from colonies as those
in Figure 4C,D. Isotype controls are blue curves. Modied from ref. 58 with permission.
Further analysis of the polarization state of macrophages recruited by a-gal nanoparticles
indicated that they were M2 macrophages since they were positively immunostained for M2 markers
IL-10 and Arginase-1 and were negatively immunostained for IL-12, a marker which characterizes
M1 macrophages (Figure 4B) [57]. When these inltrating macrophages were cultured in vitro for 5
days, the culture wells were found to contain cell colonies at a frequency of one colony per 50,000 to
100,000 cultured macrophages. These cell colonies had the morphological characteristics of colonies
formed by MSC (Figure 4C,D). Accordingly, the majority of the cells retrieved from these colonies
presented the stem cell markers Sca-1 and CD-29 (Figure 4E,F). These colonies contained 300-1000
cells per colony, suggesting that the cells forming them proliferated at an average cell-cycle time of
~12 hours. Overall, the observations in Figure 4B–F suggest that the majority of macrophages
recruited and activated by a-gal nanoparticles polarized into M2 macrophages and further directed
migration of MSC into the implanted PVA sponge discs [58].
9. Accelerated Healing of Burns by Topical Application of a-gal Nanoparticles
The above in vitro and in vivo studies on the eects of a-gal nanoparticles on macrophages
(Figures 3 and 4, respectively) prompted the analysis of eects of these nanoparticles on burns healing
in anti-Gal producing GT-KO mice [17]. For this purpose, 10 mg a-gal nanoparticles from a
suspension containing 100 mg/ml, were dried under sterile conditions on 1x1cm pads of small “spot”
Preprints (www.preprints.org) | NOT PEER-REVIEWED | Posted: 6 September 2023 doi:10.20944/preprints202309.0375.v1
9
bandages. Pads with dried 0.1ml saline served as controls. Two thermal injuries were performed on
shaved two abdominal anks of anesthetized mice by a brief contact of heated end of a metal spatula
(~2x3 mm), resulting in a second degree burn aecting epidermis and dermis, but not the
hypodermis. The right side burns were covered with α-gal nanoparticles coated spot bandages, and
the left side burns with control saline containing spot bandages (Figure 5A). Removal of the bandages
by the mice was prevented by covering them with Tegaderm
TM
and with Transpore
TM
adhesive tape.
The dressings were removed at various time points, the extent of covering the burn by re-
epithelialization and macrophage inltration were measured and the burn areas were sectioned and
subjected to histologic staining by hematoxylin & eosin (H&E) (Figures 5B,C and 6) and by Mason
trichrome that stains collagen blue (Figure 7). The extent of macrophages inltrates into burns and of
re-epithelialization (i.e., covering of the burn injury by regenerating epidermis) are presented in
Figures 8A and 8B, respectively.
Figure 5. Demonstration of an α-gal nanoparticles treated burn and saline control burn in a
representative anti-Gal producing GT-KO mouse, 6 days post thermal injuries and treatment. A. Gross
morphology. Note the big dierence in the healing of the two burns. B. Histology of the saline treated
burn presented in A. No epidermis growth is observed over the dermis which is covered by the eschar.
C. Histology of α-gal nanoparticles treated burn presented in A. The burn is covered by the
regenerating epidermis including stratum corneum and the eschar is observed above the regenerating
stratum corneum. (H&E, x100). Based on observations in ref. 17.
Preprints (www.preprints.org) | NOT PEER-REVIEWED | Posted: 6 September 2023 doi:10.20944/preprints202309.0375.v1
10
Figure 6. Histology of the accelerated healing of representative burns in anti-Gal producing GT-KO
mice treated with α-gal nanoparticles, at various days post treatment. A. Normal mouse skin. B. A
burn, 24 hours post injury, displaying histology similar to second-degree burns in humans in that
epidermis and dermis are destroyed, but not the hypodermis. C. Saline-treated burn on Day 3. D. α-
Gal nanoparticles treated burn on Day 3, characterized by extensive recruitment of macrophages and
neutrophils in the injured dermis. E. Saline-treated burn on Day 6 demonstrating migration of
macrophages and neutrophils toward the surface of the burn. F. α-Gal nanoparticles treated burn on
Day 6 displaying complete regeneration of the epidermis, including stratum corneum. Most of the
recruited macrophages demonstrated on Day 3 in the dermis are observed on Day 6 above and within
the apical part of the stratum corneum. G and H. Day 12 demonstrates complete regrowth of the
regenerative epidermis in healing burns treated with saline and α-gal nanoparticles, respectively.
Specimens are presented in pairs obtained from the same mouse and are representative of ve mice
at each time point (H&E, x100). Reproduced with permission from ref. 17.
Preprints (www.preprints.org) | NOT PEER-REVIEWED | Posted: 6 September 2023 doi:10.20944/preprints202309.0375.v1
11
Figure 7. Determination of dermis regeneration as evaluated by Mason trichrome staining blue of de
novo formed collagen in saline treated (A and C) and in α-gal nanoparticles treated burns (B, D and
E). Normal uninjured skin is presented in (F). Specimens A-D are presented in pairs obtained from
the same mouse and are representative of ve mice at each time point (x100).
Figure 8. Quantication of macrophage inltration (A) and burn healing determined by % of
epidermal regeneration (B) in burns treated with α-gal nanoparticles or with saline. A. The number
of inltrating macrophages at various time points was determined in histological sections by counting
cells within a rectangular area marked in a microscope lens at magnication of x400. B. The proportion
(%) of epidermis regeneration was determined histologically by the proportion of the burn surface
Preprints (www.preprints.org) | NOT PEER-REVIEWED | Posted: 6 September 2023 doi:10.20944/preprints202309.0375.v1
12
covered with the newly formed epidermis. Mean + S.E. from ve mice per group. Adapted based on
data in ref. 17.
The histology of a representative normal mouse skin is presented in Figures 6A and 7F. The
epidermis in such skins is comprised of 2-3 layers of epithelial cells, the underlying dermis is stained
pink by H&E and blue by Mason trichrome and the hypodermis contained mostly fat tissue,
characterized by multiple adipocytes. The thermal injuries in the mouse skin resulted in destruction
of both the epidermis and the dermis, as observed 24 hours post injury (Figure 6B). This damage is
similar to second degree burns in humans, in that both epidermis and dermis are destroyed, whereas
damage to the hypodermis is minimal. No dierences have been observed 24 hours post injury in a-
gal nanoparticles treated burns (Figure 6B) and in burns treated with saline (not shown).
A major dierence was observed between treated and control burns, inspected 3 days post
injury. Whereas no signicant number of macrophages was observed in control burns, as many as 40
macrophages were detected in same size eld in a-gal nanoparticles treated burns (Figures 6C,D and
8A). In addition, control burns displayed some degree of neutrophils inltration, but a-gal
nanoparticles treated burns displayed ~5 fold higher number of neutrophils (Figure 6C,D).
The most dramatic dierence between the two burn treatments was observed 6 days post
thermal injury. At that time point, a-gal nanoparticles treated burns displayed extensive regeneration
of epidermis as 50-100% re-epithelialization of the surface areas (mean of ~70%) (Figure 8B). The
newly formed epidermis also included formation of stratum corneum (Figures 5C and 6D). Many of
the macrophages and neutrophils were found to be removed to the surface of the regenerating
epidermis, above the stratum corneum and were mixed with remnants of the eschar. This accelerated
healing was found to be dose depended since 1 mg of a-gal nanoparticles induced an average of 23%
healing after 6 days and 0.1 mg elicited not measurable healing [17]. No signicant epidermis
regeneration was observed on Day 6 in control burns (Figures 5B, 6C and 8B). Nevertheless, the
dermis displayed increasing numbers of macrophages in a state of migration to the apical area of the
injured dermis.
Evaluation of dermis regeneration was performed by Mason trichrome which stained blue de
novo synthesized collagen. Near complete regeneration of the dermis was observed in a-gal
nanoparticles treated burns after 6 days (Figure 7D), whereas in control burns much of the dermis
was stained red, characteristic of thermal damage of the dermis (Figures 7A,C). Initial indication of
re-epithelialization of control burns was observed on Day 9, in which ~20% of the burn surfaces were
covered by regenerating epidermis (Figure 8B). In contrast, 100% of the a-gal nanoparticles treated
burns were healed by that time point. By Day 12, all control burns displayed complete healing as that
observed in a-gal nanoparticles treated burns (Figures 6G, 6H, 7E and 8B). All treated and control
healed burns did not develop keloids. These ndings imply that topical application of a-gal
nanoparticles to burns accelerates burns healing and cuts the healing time by ~40%-50%. It is of note
that in the absence of anti-Gal (e.g., in wild-type mice), no dierence in the healing process was
observed between control burns and burns treated with a-gal nanoparticles. Both were similar to the
healing of control burns in anti-Gal producing GT-KO mice [17].
10. α-.Gal Nanoparticles Mediated Accelerated Healing of Wounds
Since both healing processes of burns and wounds are mediated by macrophages, it was of
interest to determine whether a-gal nanoparticles treatment has similar accelerating eects on wound
healing in anti-Gal producing GT-KO mice, as the eects described above in burns healing. Oval
shaped full thickness wounds (~6x9 mm) were performed in anesthetized anti-Gal producing GT-KO
mice. The wounds were covered with spot bandage dressings containing 10 mg dried a-gal
nanoparticles or with control spot bandage dressing containing saline. The healing of the wounds
was evaluated by re-epithelialization at various time points. As with treated burns, wounds treated
with a-gal nanoparticles completely healed by Day 6 post treatment, whereas control wounds healed
only after 12-14 days [18,48]. Studies on completely healed treated and control wounds, 28 days post
injury, indicated that, healing of saline treated control wounds resulted in brosis and scar formation,
Preprints (www.preprints.org) | NOT PEER-REVIEWED | Posted: 6 September 2023 doi:10.20944/preprints202309.0375.v1
13
characteristic to the physiologic default healing of untreated wounds. In contrast, healing of a-gal
nanoparticles treated wounds resulted in restoration of the normal structure of skin including re-
appearance of skin appendages such as hair, sebaceous glands, and hypodermal adipocytes [48]. It
was suggested that the accelerated recruitment and activation of macrophages resulted in
regeneration of the normal skin structure prior to the initiation of the default brosis and scar
formation processes, thereby avoiding the laer processes [18,47]. It should be noted that a similar
healing that includes restoration of the original structure and function was observed in anti-Gal
producing GT-KO mice following myocardial infarction (MI) which was treated by injections of a-
gal nanoparticles [59]. In contrast, post infarction ischemic myocardium injected with saline
displayed brosis and scar formation, similar to the pathology observed in post-MI injured
myocardium in humans.
11. Concluding Remarks
Burn healing can be accelerated by the use of a-gal nanoparticles, which harness the
immunologic potential of the natural anti-Gal antibody, an abundant antibody in humans
constituting ~1% of immunoglobulins. Application of a-gal nanoparticles to burns results in binding
of anti-Gal to the a-gal epitopes on these nanoparticles. This interaction activates the complement
system, resulting in formation of complement cleavage chemotactic peptides, that direct rapid and
extensive migration of monocytes derived macrophages into the treated burns. These recruited
macrophages bind via their Fcg receptors the Fcg “tails” of anti-Gal coating the a-gal nanoparticles
and are activated to into an M2 polarization state. The activated macrophages further produce a
variety of cytokines/growth factors that mediate accelerated regrowth of the epidermis and
regeneration of the injured dermis. In anti-Gal producing mice the accelerated epidermal regrowth
results in covering of burn with intact epidermis, twice as fast as physiologic regrowth. Similarly,
healing of a-gal nanoparticles treated burns in these mice is 40–60% faster than physiologic burn
healing. The a-gal nanoparticles are non-toxic, and do not induce chronic granulomas or keloids. In
addition, the a-gal nanoparticles are highly stable for long periods at various temperatures, and in
view of their accelerated healing eects they may be considered for treatment of human burns.
Accelerated healing by a-gal nanoparticles is also observed in treated wounds of anti-Gal producing
mice. Application of a-gal nanoparticles to burns and wounds may be feasible in the form of dried
nanoparticles on wound dressings and as suspensions, aerosols, hydrogels, or incorporated into
sheets of biodegradable scaold materials such as collagen sheets.
References
1. Mahdavian Delavary, B.; van der Veer, W M.; van Egmond, M.; Niessen, F.B.; Beelen, R.H. Macrophages in
skin injury and repair. Immunobiology. 2011, 216, 753–762.
2. DiPietro, L.A.; Koh, T.J. Macrophages and wound healing. Adv. Wound Care 2016, 2, 71–75.
3. Suda, S.; Williams, H.; Medbury, H.J.; Holland. A.J. A Review of Monocytes and Monocyte-Derived Cells
in Hypertrophic Scarring Post Burn. J. Burn Care Res. 2016, 37, 26572.
4. Penaer JA, Srinivas S, Thakkar RK. The role of macrophages in thermal injury. Int J Burns Trauma. 2022
Feb 15;12(1):1-12.
5. Italiani, P.; Boraschi, D. From Monocytes to M1/M2 macrophages: phenotypical vs. functional
dierentiation. Front. Immunol. 2014, 5, 514.
6. Singer, A.J; Clark, R.A. Cutaneous wound healing. N. Engl. J. Med. 1999, 341,738–746.
7. Jeschke, M.G.; van Baar, M.E.; Choudhry, M.A.; Chung, K.K.; Gibran, N.S.; Logsey, S. Burn injury. Nat.
Rev. Dis. Primer. 2020, 6, 11
8. Piccolo, M.T.; Wang, Y.; Sannomiya, P.; Piccolo, N.S.; Piccolo, M.S.; Hugli, T.E.; Ward, P.A.; Till, G.O.
Chemotactic mediator requirements in lung injury following skin burns in rats. Exp. Mol. Pathol. 1999, 66,
220–226.
9. Shukaliak, J.; Dorovini-Zis, K. Expression of the [β]-Chemokines RANTES and MIP-1β by Human Brain
Microvessel Endothelial Cells in Primary Culture. J. Neuropathol. Exp. Neurol. 2000, 59, 339–352.
Preprints (www.preprints.org) | NOT PEER-REVIEWED | Posted: 6 September 2023 doi:10.20944/preprints202309.0375.v1
14
10. Shallo, H.; Placke, T.P.; Heinrich, S.A.; Kovacs, E.J. Monocyte chemoaractant protein-1 (MCP-1) and
macrophage inltration into the skin after burn injury in aged mice. Burns 2003, 29, 641–647.
11. Heinrich, S.A.; Messingham, K.A.; Gregory, M.S.; Colantoni, A.; Ferreira, A.M.; Dipietro, L.A.; Kovacs, E.J.
Elevated monocyte chemoaractant protein-1 levels following thermal injury precede monocyte
recruitment to the wound site and are controlled, in part, by tumor necrosis factor-α. Wound Repair Regen.
2003, 11, 110–119.
12. Tyler MP, Was AM, Perry ME, Roberts AH, McGrouther DA. Dermal cellular inammation in burns. An
insight into the function of dermal microvascular anatomy. Burns 2001, 27, 433-438.
13. Martin, P.; Leibovich, S. J. Inammatory cells during wound repair: the good, the bad and the ugly. Trends
. Biol. 2005, 15, 599–607.
14. Franz, M.G.; Steed, D.L.; Robson, M.C. Optimizing healing of the acute wound by minimizing
complications. Curr. Probl. Surg. 2007, 4, 691–763.
15. Alexander, M.; Chaudry, I.H.; Schwacha, M.G. Relationships between burn size, immunosuppression, and
macrophage hyperactivity in a murine model of thermal injury. Cell. Immunol. 2002, 220, 63–69.
16. Schwacha, M.G. Macrophages and post-burn immune dysfunction. Burns 2003, 29, 1–14.
17. Galili, U.; Wigglesworth, K.; Abdel-Motal, U.M. Accelerated healing of skin burns by anti-Gal/α-gal
liposomes interaction. Burns 2010, 36, 239–251.
18. Galili, U. α-Gal Nanoparticles in Wound and Burn Healing Acceleration. Adv. Wound Care (New
Rochelle). 2017, 6, 81–92.
19. Hocking, A.M. Mesenchymal Stem Cell Therapy for Cutaneous Wounds. Adv. Wound Care (New
Rochelle). 2012 1, 166-171.
20. Jo, H., Brito, S.; Kwak, B.M.; Park, S.; Lee, M.G.; Bin, B.H. Applications of Mesenchymal Stem Cells in Skin
Regeneration and Rejuvenation. Int. J. Mol. Sci. 2021, 22, 2410.
21. Hernon, C,A.; Dawson, R.A.; Freedlander, E.; Short, R.; Haddow, D.B.; Brotherston, M.; MacNeil, S. Clinical
experience using cultured epithelial autografts leads to an alternative methodology for transferring skin
cells from the laboratory to the patient. Regen. Med. 2006 1, 809–821.
22. Lee, H. Outcomes of sprayed cultured epithelial autografts for full-thickness wounds: a single-centre
experience. Burns 2012, 38, 931–936.
23. Yuan, L.; Minghua, C.; Feifei, D.; Runxiu, W.; Ziqiang, L.; Chengyue, M., Wenbo, J. Study of the use of
recombinant human granulocyte-macrophage colony-stimulating factor hydrogel externally to treat
residual wounds of extensive deep partial-thickness burn. Burns 2015, 41, 1086–1091.
24. Yan, D.; Liu, S.; Zhao, X.; Bian, H.; Yao. X.; Xing, J.; Sun, W.; Chen, X. Recombinant human granulocyte
macrophage colony stimulating factor in deep second-degree burn wound healing. Medicine (Baltimore)
2017, 96, e6881.
25. Lavker, R.M.; Kaplan, N.; McMahon, K.M.; Calvert, A.E.; Henrich, S.E.; Onay, U.V.; Lu, K.Q.; Peng, H.;
Thaxton, C.S. Synthetic high-density lipoprotein nanoparticles: Good things in small packages. Ocul. Surf.
2021, 21,19-26.
26. Hermosilla, J.; Pastene-Navarrete, E.; Acevedo, F. Electrospun Fibers Loaded with Natural Bioactive
Compounds as a Biomedical System for Skin Burn Treatment. A Review. Pharmaceutics 2021,13, 2054;
27. Weng, T.; Wang, J.; Yang, M.; Zhang, W.; Wu, P.; You, C.; Han, C.; Wang, X. Nanomaterials for the delivery
of bioactive factors to enhance angiogenesis of dermal substitutes during wound healing. Burns Trauma
2022, 10, tkab049.
28. Frear, C.C.; Grin, B.R.; Cule, L.; Kimble, R.M.; McPhail, S.M. Cost-eectiveness of adjunctive negative
pressure wound therapy in pediatric burn care: evidence from the SONATA in C randomized controlled
trial. Sci. Rep. 2021, 11, 16650.
29. Galili, U.; Rachmilewi, E.A.; Peleg, A.; Flechner, I. A unique natural human IgG antibody with anti-α-
galactosyl specicity. J. Exp. Med. 1984, 160, 1519–1531.
30. Galili, U.; Mandrell, R.E.; Hamadeh, R.M.; Shohet, S.B.; Griss, J.M. Interaction between human natural
anti-α-galactosyl immunoglobulin G and bacteria of the human ora. Infect. Immun. 1988, 56, 1730–1737.
31. Mañez, R.; Blanco, F.J.; Díaz, I.; Centeno, A.; Lopez-Pelaez, E.; Hermida, M.; Davies, H.F.; Katopodis, A.
Removal of bowel aerobic gram-negative bacteria is more eective than immunosuppression with
cyclophosphamide and steroids to decrease natural α-galactosyl IgG antibodies. Xenotransplantation 2001,
8, 15–23.
Preprints (www.preprints.org) | NOT PEER-REVIEWED | Posted: 6 September 2023 doi:10.20944/preprints202309.0375.v1
15
32. Galili, U.; Macher, B.A.; Buehler, J.; Shohet, S.B. Human natural anti-α-galactosyl IgG. II. The specic
recognition of α[1,3]-linked galactose residues. J. Exp. Med. 1985, 162, 573–582.
33. Towbin, H.; Rosenfelder, G.; Wieslander, J.; Avila, J.L.; Rojas, M.; Szarfman, A.; Esser, K.; Nowack, H.;
Timpl, R. Circulating antibodies to mouse laminin in Chagas disease, American cutaneous leishmaniasis,
and normal individuals recognize terminal galactosyl [α1-3]-galactose epitopes. J. Exp. Med. 1987, 166, 419–
432.
34. Teneberg, S.; Lönnroth, I.; Torres Lopez, J.F.; Galili, U.; Olwegard Halvarsson, M.; Angstrom, J.; Karlsson,
K.A. Molecular mimicry in the recognition of glycosphingolipids by Galα3Galß4GlcNAcß-binding
Clostridium dicile toxin A, human natural anti-α-galactosyl IgG and the monoclonal antibody Gal-13:
Characterization of a binding-active human glycosphingolipid, non-identical with the animal receptor.
Glycobiology 1996, 6, 599–609.
35. Galili, U.; Clark, M.R.; Shohet, S.B.; Buehler, J.; Macher, B.A. Evolutionary relationship between the anti-
Gal antibody and the Galα1-3Gal epitope in primates. Proc. Natl. Acad. Sci. USA 1987, 84, 1369–1373.
36. Galili, U.; Shohet, S.B.; Kobrin, E.; Stults, C.L.M.; Macher, B.A. Man, apes, and Old-World monkeys dier
from other mammals in the expression of α-galactosyl epitopes on nucleated cells. J. Biol. Chem. 1988, 263,
17755–17762.
37. Thall, A.; Galili, U. Distribution of Gal alpha 1----3Gal beta 1----4GlcNAc residues on secreted mammalian
glycoproteins (thyroglobulin, brinogen, and immunoglobulin G) as measured by a sensitive solid-phase
radioimmunoassay. Biochemistry 1990, 29, 3959–3965.
38. Oriol, R.; Candelier, J.J.; Taniguchi, S.; Balanzino, L.; Peters, L.; Niekrasz, M.; Hammer, C.; Cooper, D.K.
Major carbohydrate epitopes in tissues of domestic and African wild animals of potential interest for
xenotransplantation research. Xenotransplantation 1999, 6, 79–89.
39. Cooper, D.K.C.; Good, A.H.; Koren, E.; Oriol, R.; Malcolm, A.J.; Ippolito, R.M.; Neethling, F.A.; Ye, Y.;
Romano, E.; Zuhdi, N. Identication of α-galactosyl and other carbohydrate epitopes that are bound by
human anti-pig antibodies: Relevance to discordant xenografting in man. Transpl. Immunol. 1993, 1, 198–
205.
40. Galili, U. Interaction of the natural anti-Gal antibody with α-galactosyl epitopes: A major obstacle for
xenotransplantation in humans. Immunol. Today 1993, 14, 480–482
41. Sandrin, M.S.; Vaughan, H.A.; Dabkowski, P.L.; McKenzie, I.F.C. Anti-pig IgM antibodies in human serum
react predominantly with Gal (αl-3)Gal epitopes. Proc. Natl. Acad. Sci. USA 1993, 90, 11391–11395.
42. Collins, B.H.; Coerell, A.H.; McCurry, K.R.; Alvarado, C.G.; Magee, J.C.; Parker, W.; Pla, J.L. Cardiac
xenografts between primate species provide evidence for the importance of the α-galactosyl. determinant
in hyperacute rejection. J. Immunol. 1995, 154, 5500–5510.
43. Repik, P.M.; Strizki, M.; Galili, U. Dierential host dependent expression of α-galactosyl epitopes on viral
glycoproteins: A study of Eastern equine encephalitis virus as a model. J. Gen. Virol. 1994, 75, 1177–1181.
44. Pipperger, L.; Koske, I.; Wild, N.; Müllauer, B.; Krenn, D.; Stoiber, H.; Wollmann, G.; Kimpel, J.; von Laer,
D.; Bánki, Z. Xenoantigen-dependent complement-mediated neutralization of LCMV glycoprotein
pseudotyped VSV in human serum. J. Virol. 2019, 93, e00567-19.
45. Takeuchi, Y.; Porter, C.D.; Strahan, K.M.; Preece, A.F.; Gustafsson, K.; Cosset, F.L.; Weiss, R.A.; Collins,
M.K. Sensitization of cells and retroviruses to human serum by [α1-3] galactosyltransferase. Nature 1996,
379, 85–88.
46. Preece, A.F.; Strahan, K.M.; Devi, J.; Yamamoto, F.; Gustafsson, K. Expression of ABO or related antigenic
carbohydrates on viral envelopes leads to neutralization in the presence of serum containing specic
natural antibodies and complement. Blood 2002, 99, 2477–2482.
47. Galili, U. book “The natural anti-Gal antibody as foe turned friend in medicine” Academic Press/Elsevier,
London, New York, 2018, pp. 207-228.
48. Wigglesworth, K.M.; Racki, W.J.; Mishra, R.; Szomolanyi-Tsuda, E.; Greiner, D.L.; Galili. U. Rapid
recruitment and activation of macrophages by anti-Gal/α-Gal liposome interaction accelerates wound
healing. J. Immunol. 2011, 186, 4422-4432.
49. Thall, A.D.; Maly, P.; Lowe, J.B. Oocyte Gal1,3Gal epitopes implicated in sperm adhesion to the zona
pellucida glycoprotein ZP3 are not required for fertilization in the mouse. J. Biol. Chem. 1995, 270, 21437-
21440.
Preprints (www.preprints.org) | NOT PEER-REVIEWED | Posted: 6 September 2023 doi:10.20944/preprints202309.0375.v1
16
50. Tearle, R.G.; Tange, M.J.; Zaneino, Z.L.; Katerelos, M.; Shinkel, T.A.; Van Denderen, B.J.; Lonie, A.J.;
Lyonsm, I., Nole, M.B.; Cox, T. et al. The alpha-1,3-galactosyltransferase knockout mouse. Implications
for xenotransplantation. Transplantation 1996, 61, 13-19.
51. Lai, L.; Kolber-Simonds, D.; Park, K.W.; Cheong, H.T.; Greenstein, J.L.; Im, G.S.; Samuel, M.; Bonk, A.;
Rieke, A.; Day, B.N.; et al. Production of α-1,3-galactosyltransferase knockout pigs by nuclear transfer
cloning. Science 2002, 295, 1089–1092.
52. Phelps, C.J.; Koike, C.; Vaught, T.D.; Boone, J.; Wells, K.D.; Chen, S.H.; Ball, S.; Specht, S.M.; Polejaeva, I.A.;
Monahan, J.A.; et al. Production of α1,3-galactosyltransferase-decient pigs. Science 2003, 299, 411–414.
53. Dor, F.J.; Tseng, Y.L.; Cheng, J.; Moran, K.; Sanderson, T.M.; Lancos, C.J.; Shimizu, A.; Yamada, K.; Awwad,
M.; Sachs, D.H.; et al. α1,3-Galactosyltransferase gene-knockout miniature swine produce natural cytotoxic
anti-Gal antibodies. Transplantation 2004, 78, 15–20.
54. Fang, J.; Walters, A.; Hara, H.; Long, C.; Yeh, P.; Ayares, D.; Cooper, D.K.; Bianchi, J. Anti-gal antibodies in
α1,3- galactosyltransferase gene knockout pigs. Xenotransplantation 2012, 19, 305–310.
55. Galili, U. The natural anti-Gal antibody and simulate the evolutionary appearance of this antibody in
primates. Xenotransplantation 2013, 20, 267–276
56. Tanemura, M.; Yin, D.; Chong, A.S.; Galili, U. Dierential immune responses to α-gal epitopes on
xenografts and allografts: Implications for accommodation in xenotransplantation. J. Clin. Investig. 2000,
105, 301–310.
57. Kaymakcalan, O.E.; Abadeer, A.; Goldufsky, J.W.; Galili, U.; Karinja, S.J.; Dong, X.; Jin, J.L.; Samadi, A.;
Spector, J.A. Topical α-gal nanoparticles accelerate diabetic wound healing. Exp. Dermatol. 2020, 29, 404–
413.
58. Galili, U.; Goldufsky, J.W.; Schaer, G.L. α-Gal Nanoparticles Mediated Homing of Endogenous Stem Cells
for Repair and Regeneration of External and Internal Injuries by Localized Complement Activation and
Macrophage Recruitment. Int. J. Mol. Sci. 2022, 23, 11490.
59. Galili, U.; Zhu, Z.; Chen, J.; Goldufsky, J.W.; Schaer, G.L. Near Complete Repair After Myocardial Infarction
in Adult Mice by Altering the Inammatory Response With Intramyocardial Injection of α-Gal
Nanoparticles. Frontiers Cardiovasc. Med. 2021, 8, 719160.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those
of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s)
disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or
products referred to in the content.
Preprints (www.preprints.org) | NOT PEER-REVIEWED | Posted: 6 September 2023 doi:10.20944/preprints202309.0375.v1
Article
Full-text available
The healing of skin wounds, myocardial, and spinal cord injuries in salamander, newt, and axolotl amphibians, and in mouse neonates, results in scar-free regeneration, whereas injuries in adult mice heal by fibrosis and scar formation. Although both types of healing are mediated by macrophages, regeneration in these amphibians and in mouse neonates also involves innate activation of the complement system. These differences suggest that localized complement activation in adult mouse injuries might induce regeneration instead of the default fibrosis and scar formation. Localized complement activation is feasible by antigen/antibody interaction between biodegradable nanoparticles presenting α-gal epitopes (α-gal nanoparticles) and the natural anti-Gal antibody which is abundant in humans. Administration of α-gal nanoparticles into injuries of anti-Gal-producing adult mice results in localized complement activation which induces rapid and extensive macrophage recruitment. These macrophages bind anti-Gal-coated α-gal nanoparticles and polarize into M2 pro-regenerative macrophages that orchestrate accelerated scar-free regeneration of skin wounds and regeneration of myocardium injured by myocardial infarction (MI). Furthermore, injection of α-gal nanoparticles into spinal cord injuries of anti-Gal-producing adult mice induces recruitment of M2 macrophages, that mediate extensive angiogenesis and axonal sprouting, which reconnects between proximal and distal severed axons. Thus, α-gal nanoparticle treatment in adult mice mimics physiologic regeneration in amphibians. These studies further suggest that α-gal nanoparticles may be of significance in the treatment of human injuries.
Article
Full-text available
This review discusses a novel experimental approach for the regeneration of original tissue structure by recruitment of endogenous stem-cells to injured sites following administration of α-gal nanoparticles, which harness the natural anti-Gal antibody. Anti-Gal is produced in large amounts in all humans, and it binds the multiple α-gal epitopes (Galα1-3Galβ1-4GlcNAc-R) presented on α-gal nanoparticles. In situ binding of anti-Gal to α-gal nanoparticles activates the complement system and generates complement cleavage chemotactic-peptides that rapidly recruit macrophages. Macrophages reaching anti-Gal coated α-gal nanoparticles bind them via Fc/Fc receptor interaction and polarize into M2 pro-reparative macrophages. These macrophages secrete various cytokines that orchestrate regeneration of the injured tissue, including VEGF inducing neo-vascularization and cytokines directing homing of stem-cells to injury sites. Homing of stem-cells is also directed by interaction of complement cleavage peptides with their corresponding receptors on the stem-cells. Application of α-gal nanoparticles to skin wounds of anti-Gal producing mice results in decrease in healing time by half. Furthermore, α-gal nanoparticles treated wounds restore the normal structure of the injured skin without fibrosis or scar formation. Similarly, in a mouse model of occlusion/reperfusion myocardial-infarction, near complete regeneration after intramyocardial injection of α-gal nanoparticles was demonstrated, whereas hearts injected with saline display ~20% fibrosis and scar formation of the left ventricular wall. It is suggested that recruitment of stem-cells following anti-Gal/α-gal nanoparticles interaction in injured tissues may result in induction of localized regeneration facilitated by conducive microenvironments generated by pro-reparative macrophage secretions and “cues” provided by the extracellular matrix in the injury site.
Article
Full-text available
Dermal substitutes provide a template for dermal regeneration and reconstruction. They constitutes an ideal clinical treatment for deep skin defects. However, rapid vascularization remains as a major hurdle to the development and application of dermal substitutes. Several bioactive factors play an important regulatory role in the process of angiogenesis and an understanding of the mechanism of achieving their effective delivery and sustained function is vital. Nanomaterials have great potential for tissue engineering. Effective delivery of bioactive factors (including growth factors, peptides and nucleic acids) by nanomaterials is of increasing research interest. This paper discusses the process of dermal substitute angiogenesis and the roles of related bioactive factors in this process. The application of nanomaterials for the delivery of bioactive factors to enhance angiogenesis and accelerate wound healing is also reviewed. We focus on new systems and approaches for delivering bioactive factors for enhancing angiogenesis in dermal substitutes. Highlights • The role of bioactive factors in the process of dermal substitute angiogenesis is discussed. • Recent progress in the use of nanomaterials for delivery of bioactive factors to enhance angiogenesis and accelerate wound healing are systematically reviewed. • New systems and approaches for delivering nucleic acids for enhancing angiogenesis in dermal substitutes are described and summarized.
Article
Full-text available
Burns are a major threat to public health and the economy due to their costly and laborious treatment and high susceptibility to infection. Efforts have been made recently to investigate natural bioactive compounds with potential use in wound healing. The importance lies in the capacities that these compounds could possess both in infection control by common and resistant microorganisms, as well as in the regeneration of the affected tissues, having in both cases low adverse effects. However, some bioactive molecules are chemically unstable, poorly soluble, and susceptible to oxidative degradation or have low bioavailability. Therefore, developing new technologies for an efficient treatment of wound healing poses a real challenge. In this context, electrospun nanofibers have gained increasing research interest because bioactive molecules can be easily loaded within the nanofiber, resulting in optimal burst control and enhanced drug stability. Additionally, the nanofibers can mimic the extracellular collagen matrix, providing a suitable highly porous structural support for growing cells that facilitate and accelerate skin burns healing. This review gives an overview of the current state of electrospun fibers loaded with natural bioactive compounds as a biomedical system for skin burn treatment.
Article
Full-text available
Background: Neonatal mice, but not older mice, can regenerate their hearts after myocardial-infarction (MI), a process mediated by pro-reparative macrophages. α-Gal nanoparticles applied to skin wounds in adult-mice bind the anti-Gal antibody, activate the complement cascade and generate complement chemotactic peptides that recruit pro-reparative macrophages which are further activated by these nanoparticles. The recruited macrophages decrease wound healing time by ~50%, restore the normal skin structure and prevent fibrosis and scar formation in mice. Objectives: The objective of this study is to determine if α-gal nanoparticles injected into the reperfused myocardium after MI in adult-mice can induce myocardial repair that restores normal structure, similar to that observed in skin injuries. Methods and Results: MI was induced by occluding the mid-portion of the left anterior descending (LAD) coronary artery for 30 min. Immediately following reperfusion, each mouse received two 10 μl injections of 100 μg α-gal nanoparticles in saline into the LAD territory ( n = 20), or saline for controls ( n = 10). Myocardial infarct size was measured by planimetry following Trichrome staining and macrophage recruitment by hematoxylin-eosin staining. Left ventricular (LV) function was measured by echocardiography. Control mice displayed peak macrophage infiltration at 4-days, whereas treated mice had a delayed peak macrophage infiltration at 7-days. At 28-days, control mice demonstrated large transmural infarcts with extensive scar formation and poor contractile function. In contrast, mice treated with α-gal nanoparticles demonstrated after 28-days a marked reduction in infarct size (~10-fold smaller), restoration of normal myocardium structure and contractile function. Conclusions: Intramyocardial injection of α-gal nanoparticles post-MI in anti-Gal producing adult-mice results in near complete repair of the infarcted territory, with restoration of normal LV structure and contractile function. The mechanism responsible for this benefit likely involves alteration of the usual inflammatory response post-MI, as previously observed with regeneration of injured hearts in adult zebrafish, salamanders and neonatal mice.
Article
Full-text available
Negative pressure wound therapy (NPWT) has been shown to improve clinical outcomes for children with burns by accelerating wound re-epithelialisation. Its effects on healthcare costs, however, remain poorly understood. The aim of this study was to evaluate the cost-effectiveness of NPWT from a healthcare provider perspective using evidence from the SONATA in C randomised controlled trial, in which 101 children with small-area burns were allocated to either standard care (silver-impregnated dressings) or standard care in combination with adjunctive NPWT. The primary outcome, time to re-epithelialisation, was assessed through a blinded photographic review. Resource usage and costs were prospectively recorded for each participant for up to 6 months. Incremental cost-effectiveness ratios and dominance probabilities were estimated and uncertainty quantified using bootstrap resampling. Mean costs per participant—including dressings, labour, medication, scar management, and theatre operations—were lower in the NPWT group (AUD 903.69)relativetothecontrolgroup(AUD903.69) relative to the control group (AUD 1669.01). There was an 89% probability that NPWT was dominant, yielding both faster re-epithelialisation and lower overall costs. Findings remained robust to sensitivity analyses employing alternative theatre costs and time-to-re-epithelialisation estimates for grafted patients. In conclusion, adjunctive NPWT is likely to be a cost-effective and dominant treatment for small-area paediatric burns (ANZCTR.org.au:ACTRN12618000256279).
Article
Full-text available
Mesenchymal stem cells (MSCs) are multipotent stem cells derived from adult stem cells. Primary MSCs can be obtained from diverse sources, including bone marrow, adipose tissue, and umbilical cord blood. Recently, MSCs have been recognized as therapeutic agents for skin regeneration and rejuvenation. The skin can be damaged by wounds, caused by cutting or breaking of the tissue, and burns. Moreover, skin aging is a process that occurs naturally but can be worsened by environmental pollution, exposure to ultraviolet radiation, alcohol consumption, tobacco use, and undernourishment. MSCs have healing capacities that can be applied in damaged and aged skin. In skin regeneration, MSCs increase cell proliferation and neovascularization, and decrease inflammation in skin injury lesions. In skin rejuvenation, MSCs lead to production of collagen and elastic fibers, inhibition of metalloproteinase activation, and promote protection from ultraviolet radiation-induced senescence. In this review, we focus on how MSCs and MSC-derived molecules improve diseased and aged skin. Additionally, we emphasize that induced pluripotent stem cell (iPSC)-derived MSCs are potentially advanced MSCs, which are suitable for cell therapy.
Article
Full-text available
An inadequate response from macrophages, key orchestrators of the wound healing process, has been implicated in the pathophysiology of impaired healing in diabetes. This study explored the utility of nanoparticles presenting the α‐gal (Galα1‐3Galβ1‐4GlcNAc‐R) epitope to induce anti‐Gal antibody mediated local transient recruitment of macrophages to accelerate wound closure and healing in a diabetic murine model. α1,3galactosyltrasferase knockout mice were stimulated to produce anti‐Gal antibodies and subsequently diabetes was induced by streptozotocin‐induced ‐cell destruction. Six mm full thickness skin wounds were made and α‐gal nanoparticles (AGN) were topically applied on post‐wounding days 0 and 1. Wounds were analyzed histologically for macrophage invasion and markers of wound healing, including epithelialization, vascularization, and granulation tissue deposition through post‐operative day 12. We found that application of AGN transiently but significantly increased macrophage recruitment into the wounds of diabetic mice. Treated wounds demonstrated more rapid closure and enhanced wound healing as demonstrated by significantly accelerated rates of epithelialization, vascularization, and granulation tissue deposition. Thus, topical treatment of full thickness wounds in diabetic mice with α‐gal nanoparticles induced a transient but significant increase in macrophage recruitment resulting in an accelerated rate of wound healing. Using α‐gal nanoparticles as a topical wound healing adjunct is a simple, safe and effective means of augmenting dysregulated macrophage recruitment present in the diabetic state.
Article
Macrophages, first discovered for their phagocytic ability, are a complicated and heterogeneous cell type. The unique properties of macrophages allow them to perform a vast array of functions, including phagocytosis, cytokine production, antigen presentation, and wound healing. Some macrophage populations are derived from monocytes and are induced into specific phenotypes by the local tissue microenvironment, while other macrophages form during early embryonic development. The exposure of the host to local pathogens and/or traumatic injury alters the tissue microenvironment and, in turn, influences changes in macrophage phenotype and function. Perhaps the most significant change in the local tissue microenvironment and subsequent macrophage phenotype occurs after thermal injury, which causes localized tissue damage and a massive systemic inflammatory response. However, few studies have explored the influence of burn injury on the host macrophages and macrophage function in burn wounds. Furthermore, the literature is scant regarding the impact macrophage function has on outcomes in thermal injury. This review will focus on the current knowledge of macrophage function in burn wounds and the phenotypic changes in macrophages during thermal injury while identifying knowledge gaps.
Article
Medicine has been a great beneficiary of the nanotechnology revolution. Nanotechnology involves the synthesis of functional materials with at least one size dimension between 1 and 100 nm. Advances in the field have enabled the synthesis of bio-nanoparticles that can interface with physiological systems to modulate fundamental cellular processes. One example of a diverse acting nanoparticle-based therapeutic is synthetic high-density lipoprotein (HDL) nanoparticles (NP), which have great potential for treating diseases of the ocular surface. Our group has developed a spherical HDL NP using a gold nanoparticle core. HDL NPs: (i) closely mimic the physical and chemical features of natural HDLs; (ii) contain apoA-I; (iii) bind with high-affinity to SR-B1, which is the major receptor through which HDL modulates cell cholesterol metabolism and controls the selective uptake of HDL cargo into cells; (iv) are non-toxic to cells and tissues; and (v) can be chemically engineered to display nearly any surface or core composition desired. With respect to the ocular surface, topical application of HDL NPs accelerates re-epithelization of the cornea following wounding, attenuates inflammation resulting from chemical burns and/or other stresses, and effectively delivers microRNAs with biological activity to corneal cells and tissues. HDL NPs will be the foundation of a new class of topical eye drops with great translational potential and exemplify the impact that nanoparticles can have in medicine.