ArticlePDF AvailableLiterature Review

Macrophages in xenotransplantation

Authors:

Abstract and Figures

Xenotransplantation refers to organ transplantation across species. Immune rejection of xenografts is stronger and faster than that of allografts because of significant molecular differences between species. Recent studies have revealed the involvement of macrophages in xenograft and allograft rejections. Macrophages have been shown to play a critical role in inflammation, coagulation, and phagocytosis in xenograft rejection. This review presents a recent understanding of the role of macrophages in xenograft rejection and possible strategies to control macrophage-mediated xenograft rejection.
Content may be subject to copyright.
2019 The Korean Society for Transplantation
This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/
licenses/by-nc/4.0/) whi ch permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly
cited.
Macrophages in xenotransplantation
Jae Young Kim
Department of Life Science, Gachon University, Seongnam, Korea
Xenotransplantation refers to organ transplantation across species. Immune rejection of xenografts is stronger and faster
than that of allografts because of significant molecular differences between species. Recent studies have revealed the involve-
ment of macrophages in xenograft and allograft rejections. Macrophages have been shown to play a critical role in inflammation,
coagulation, and phagocytosis in xenograft rejection. This re vi ew presents a recent understanding of the role of macrophages
in xenograft rejection and possible strategies to control macrophage-mediated xenograft rejection.
Keywords: Xenotransplantation; Macrophages; Inflammation; Coagulation
Received December 9, 2019
Revised December 19, 2019
Accepted December 19, 2019
Correspondence to: Jae Young Kim
Department of Life Science, Gachon University, 1342 Seongnam-
daero, Sujeong-gu, Seongnam 13120, Korea
Tel: +82-31-750-4762
E-mail: jykim85@gachon.ac.kr
Review Article
INTRODUCTION
Xenotransplantation is the transplantation of organs, tis-
sues, or cells across species. Pigs are considered to be
an ideal organ source for xenotransplantation due to their
physiological similarity to humans and the feasibility of
pig breeding. However, immune rejection of xenografts
is believed to be stronger and faster than that of
allografts. This can be attributed to the significant mo-
lecular differences between species. Xenograft rejection
can be temporally divided into four interlinked immune
rejection types: hyperacute rejection (HAR), acute vas-
cular and cellular rejections, and chronic rejection [1].
HAR is mediated by human antibodies against carbohy-
drate moieties, such as galactose 1,3-galactose (-gal)
and N-glycolylneuraminic acid, present on the surfaces
of pig endothelial cells [2]. Binding of pre-existing anti-
bodies to these antigens and subsequent complement ac-
tivation destroy pig endothelial cells, resulting in xeno-
graft rejection within a few minutes [3]. During the last
two decades, outcomes of pig-to-non-human primate
organ transplantation have been markedly improved ow-
ing to development of genetically-engineered pigs;
transplants from such pigs help avoid HAR [4].
However, many aspects of immune responses to xeno-
grafts still pose a critical problem for successful
transplantation.
Macrophages are phagocytic innate immune cells that
play a crucial role in host defense. Recent studies have
revealed the involvement of macrophages in immune re-
jection of organ transplants. In animal allotransplant
models, macrophages recognize allogeneic antigens, in-
duce immune responses, and thus contribute to graft re-
jection [5,6]. In addition, they are able to kill allogeneic
cells by phagocytosis [7]. Clinical studies exhibit a pos-
itive correlation between macrophage infiltration and
graft rejection [8-11]. The recently arising role of mac-
rophages in allograft rejection must be one of the inter-
esting topics in the field of transplant immunology.
Therefore, the purpose of this paper is to review the re-
cent understanding of the role of macrophages in xeno-
graft rejection and possible strategies to control macro-
phage-mediated xenograft rejection.
Korean J Transplant 2019;33:74-82
https://doi.org/10.4285/jkstn.2019.33.4.74
75
Kim JY. Macrophages in xenotransplantation
HIGHLIGHTS
Damage-associated molecular pattern (DAMP) re-
lease during ischemia reperfusion injury is one of the
main causes of activation of macrophages, which play
a critical role in inflammation and coagulation in xeno -
graft rejection.
Cross-talk between macrophages, hepatocytes, and
vascular endothelial cells by producing immune medi-
ators, such as monocyte chemoattractant protein 1
(MCP-1), interl eu ki n ( IL)-6, and creac ti ve protein
(CRP) may play a critical role in inflammatory re-
sponses and coagulation in pig-to-baboon organ
transplantation.
Early generation of MCP-1, IL-6, and CRP as well
as DAMPs needs to be controlled to avoid inflammation
and coagulation.
ROLE OF MACROPHAGES IN
XENOGRAFT REJECTION
Inflammation and Coagulation
Inflammation, triggered by innate immune cells as a de-
fense mechanism against infectious agents or tissue dam-
age, is a major problem in organ transplantation.
Damaged host cells release or secrete various dam-
age-associated molecular patterns (DAMPs) [12]. In or-
gan transplantation, DAMP release from injured tissues
is inevitable during ischemia reperfusion injury (IRI) and
levels of the released DAMPs increase following IRI.
DAMPs are recognized by cellular receptors of various
cell types including, the Toll-like receptors (TLRs) of
macrophages, and the inflammasomes, which trigger in-
tracellular signaling that leads to inflammatory responses
and subsequent initiation of rejection of the transplanted
organ [5,6]. As shown in Table 1, IRI-induced release
of various DAMPs, such as high-mobility group box 1
(HMGB1) [13-16], nuclear DNAs or mitochondrial DNAs
[17-19], heat shock proteins (HSPs) [20-23], and his-
tones [24], was detected in different types of solid or-
gans after transplantation.
Unlike other DAMPs, adenosine triphosphate (ATP) is
a relatively small molecule and is recognized by specific
cell surface receptors, such as P2X and P2Y. Binding of
extracellular ATP to these receptors induces in-
flammatory responses of macrophages [25]. A study us-
ing a murine liver allotransplantation model has suggested
extracellular ATP involvement in increased graft dys-
function and the involvement of the reduction of regu-
latory T cell frequency in overall graft survival [26].
The role of the coagulation pathway in IRI and its
crosstalk with the inflammatory pathway have been re-
cently proposed [27]. Tissue factor (TF), which is the
primary initiator of coagulation and is expressed on both,
monocytes/macrophages and endothelial cells, is a central
player in providing a bridge between these pathways
[28]. Macrophages play a critical role in coagulation as
well as inflammation in xenograft rejection (Fig. 1).
During IRI, DAMPs activate monocytes/macrophages,
which then, produce proinflammatory cytokines. In re-
sponse to DAMPs and the pro-inflammatory cytokines,
TF is rapidly induced by these cells in the graft recipi-
ents, and becomes exposed to blood [29,30]. Cell surface
TF can complex with factor VIIa, and thereby trigger co-
agulation by activating factor X and subsequent coagulat-
ing factors. TF can also induce protease-activated re-
ceptor-mediated signaling, which leads to the production
of more inflammatory cytokines, upregulation of adhesion
molecules, and suppression of thrombomodulin (Fig. 1A)
[31].
One of the reasons for the stronger immune rejection
of xenografts than that of allografts is associated with
vascular endothelial cells. In the early phase of organ
xenotransplantation, xenoantigen-specific innate anti-
bodies can bind to and activate the endothelial cells,
causing the release of proinflammatory immune media-
tors, as well as the activation of the complement medi-
ated destruction of the endothelial layer. Monokines,
such as interleukin (IL)-1, IL-6, and tumor necrosis
factor-, secreted by monocytes/macrophages, can also
activate vascular endothelial cells across pig and human
species [32]. Activated endothelial cells release CD62,
von Willebrand Factor, platelet factor 4, and CD40 ligand
(Fig. 1A) in addition to proinflammatory cytokines. All
these mediators eventually lead to the recruitment of
more inflammatory cells and activation of coagulation
[31]. The interplay between the inflammatory responses
76
Korean J TransplantㆍDecember 2019ㆍVolume 33ㆍIssue 4
Table 1. DAMPs identified in inflammatory diseases and upon clinical organ transplantation [39,40]
Origin DAMP Receptor Organa)
Extracellular matrix Biglycan
Decorin
Heparan sulfate
Hyaluronan
Fibrinogen
Fibronectin
Tenascin C
Versican
TLR2, 4, NLRP3
TLR2, 4
TLR4
TLR2, 4, NLRP3
TLR4
TLR4
TLR4
TLR2, 6, CD14
K
H, Lu
H, K
H, K, Lu
Intracellular compartment
cytosol ATP
-amyloid
Cyclophilin A
F-actin
HSP
S100 proteins
Uric acid
P2X7, P2Y2
TLR2, NLRP1, 3, CD36, RAGE CD147
DNGR-1
TLR2, 4, CD91, LOX-1
TLR2, 4, RAGE
NLRP3, P2X7
H, Li
H, K, Li, Lu
Nuclei DNA
Histones
HMGB1
HMGN1
IL-1
IL-33
RNA
SAP130
TLR9, AIM2
TLR2, 4
TLR2, 4, RAGE
TLR4
IL-1R
ST2
TLR3, 7, 8, RIG-I, MDA5, Mincle
H, K, Li, Lu
H, K, Li, Lu
Mitochondria Formyl peptide
mtDNA
mtROS
mtTFA
FPR1
TLR9
NLRP3
RAGE
H, Li, Lu
Granule Cathelicidin
Defensins
EDN
Granulysin
P2X7, FPR2
TLR4
TLR2
TLR4
Plasma membrane Glypicans
Syndecans
TLR4
TLR4
Endoplasmic reticulum Calreticulin CD91
DAMP, damage-associated molecular pattern; K, kidney; H, heart; Lu, lun g ; ATP , a deno s i n e tri p h osph a t e; Li , l i v er; HS P , hea t s h ock
protein;
IL, interleukin; mtDNA, mitochondrial DNA; mtROS, mitochondrial reactive oxygen species; mtTFA, mitochondrial transcription
factor
A; EDN, eosinophil-derived neurotoxin.
a)Designated organs where indicated DAMPs have been identified and studied in clinical solid organ transplantation.
and the coagulation system plays a significant role in
xenograft rejection [33-36].
Phagocytosis
The mechanism by which macrophages distinguish be-
tween self and allogeneic non-self organs, tissues, cells,
or antigens and promote organ rejection has been re-
cently clarified [37]. Mice that lack T, B, and natural
killer cells could distinguish allogeneic antigens from
those of self-tissues and induce an innate response. This
innate allo-activation is triggered by mismatch between
donor and recipient signal regulatory protein (SIRP),
which is a cell surface molecule interacting with CD47.
Similarly, macrophages are able to recognize and destroy
xenografts through their cell surface interactions between
CD47 and SIRP 1 (Fig. 1B). Due to their molecular in-
compatibility, an impaired interaction between pig CD47
and human SIRP 1 can result in the phagocytic killing
of pig endothelial cells by human macrophages [38].
However, pig hematopoietic cells expressing human
77
Kim JY. Macrophages in xenotransplantation
Fig. 1. (A-C) Central role of activ-
ated macrophages in
inflammation,
coagulation, phagocytosis, and ant-
igen presentation [31]. TNF,
tumor
necros; IL, interleukin; MCP-1, mo
-
nocyte chemoattractant protein
DAMP, damage-associated
molecular
pattern; TLR, Toll-like receptor;
TF,
tissue factor; PLT, platelet;
VWF,
von Willebrand factor; PF4,
platelet
factor 4; TM, thrombomodulin; PAR
,
protease-activated receptor; ICA-
M-1, intercellular adhes io n mole-
cule-1; VCAM-1, vascular cell adhe-
sion molecule-1; SIRP, signal regul-
atory protein .
Fig. 2. Possible cross-talk between macrophages, hepatocytes,
and
vascular endothelial cells through the production of
interleukin
(IL)-6, monocyte chemoattractant protein 1 (MCP-1), and
C-
react i ve p r o t e i n ( C RP) in inf l am m a t o r y r e spons e s a n d
coagulation
in pig-to-baboon organ transplantation. TF, tissue factor.
CD47 could be protected from phagocytic killing by hu-
man macrophages in hematopoietic cell engraftment ex-
periments [41].
Chemotaxis and Acute Phase Responses
Macrophages are the major cells infiltrating into an allog-
raft during severe rejection [42]. Similarly, macrophage
infiltration occurs just after IRI of a xenograft and per-
sists until graft rejection [43,44]. The infiltration level
of macrophages was significantly higher in -gal knock-
out xenogeneic islets than in allogeneic islets [45]. The
mechanism of monocyte accumulation within a xenograft
is thought to be associated with the production of chemo-
kines, such as monocyte chemoattractant protein 1
(MCP-1), in the graft [35,46].
In pig-to-baboon heart and kidney transplantation, it
was observed that early elevated serum levels of MCP-1,
IL-6, and C-reactive protein (CRP), which is an acute
phase protein synthesized by hepatocytes in response to
proinflammatory cytokines [47], precede consumptive
coagulopathy [35]. In addition, increased numbers of
monocytes were associated with enhanced expression of
TF [35]. These results, taken together with those of
previous reports indicate that IL-6 provokes liver cells
to produce CRP [48], which stimulates endothelial cells
to produce MCP-1 [49], and both IL-6 [50] and CRP
[51] promote TF expression, suggesting the occurrence
of cross-talk between macrophages, hepatocytes, and
vascular endothelial cells through the production of these
mediators. MCP-1, produced by the activated vascular
endothelial cells, recruit monocytes/macrophages, which
are eventually activated to produce IL-6, which stim-
ulates liver cells to release CRP, which in turn activates
vascular endothelial cells. This positive feedback loop
may play a critical role in inflammatory responses and
coagulation in pig-to-baboon organ transplantation (Fig.
2). Therefore, early upregulation of CRP, IL-6, and
MCP-1 levels needs to be suppressed to avoid in-
flammation-induced coagulopathy.
78
Korean J TransplantㆍDecember 2019ㆍVolume 33ㆍIssue 4
STRATEGIES TO CONTROL
MACROPHAGE-MEDIATED
XENOGRAFT REJECTION
A recent study has suggested that there is increasing
evidence for sustained inflammatory response in
pig-to-baboon xenograft recipients, and this systemic
inflammation is a critical hurdle for successful xeno-
transplantation [52]. Therefore, therapeutic prevention
of inflammation is necessary to achieve successful pig or-
gan xenotransplantation.
Anti-inflammatory Drugs
A commonly used immunosuppressive regimen in recent
studies of pig-to-non-human primate xenotrans-
plantation includes antithymocyte globulin, cobra venom
factor (CVF), corticosteroids, and an immunophilin in-
hibitor, such as cyclosporine A, tacrolimus, or rap-
amycin. In addition to this regimen, several other se-
lective biological drugs aimed at suppressing immune re-
sponses after xenotransplantation have been tested in
separate studies, such as anti-CD154 monoclonal anti-
body (mAb), CTLA-4 fusion proteins, anti-CD40 mAb,
anti-CD20 mAb, anti-CD25 mAb, anti-IL-6 receptor
mAb, and anti-LFA1 mAb [45,53]. Among them, mAbs
for blocking the interaction between CD40 and CD154,
and CTLA-4 fusion protein appear to be essential for
achieving long term xenograft survival. CD40CD154 in-
teractions stimulate the inflammatory response. CD40
expression is induced in activated monocytes/macro-
phages while CD154 is also expressed on monocytes/
macrophages during inflammation [54]. Blockade of the
IL-6 receptor with the anti-IL-6 receptor mAb, tocili-
zumab, resulted in a reduction in the levels of CRP [36]
and serum histones [55] upon pig-to-non-human pri-
mate xenotransplantation.
Other inflammatory drugs were also tested. Nuclear
factor kappa B inhibitor, parthenolide, significantly sup-
pressed histone-induced pig endothelial cell death in in
vitro study [55]. CVF, which had been originally used
to deplete complements causing HAR following xeno-
transplantation, was found to reverse the increased IL-6
and MCP-1 levels in pig-to-baboon heart and artery
patch transplantation [56].
Since the effective treatment of an established in-
flammatory response to DAMPs is relatively difficult, se-
lective and rapid blocking or scavenging of released
DAMPs would be a more promising therapeutic strategy.
Indeed, anti-histone therapy was found to prevent his-
tone-induced inflammation in xenotransplantation [55].
Mice treated with HMGB1 antibody were protected
against pulmonary dysfunction and had improved lung al-
lograft outcomes [16]. Blockade of HMGB1 secretion by
small molecule inhibitor was found to be beneficial to pre-
vent the loss of islet grafts and to reverse diabetes in
murine syngeneic islet transplantation [57]. Administr-
ation of ATP antagonist to a recipient mouse for 2 weeks
led to prolonged survival of the transplanted allogeneic
heart [58].
Laboratory studies have suggested possible manipu-
lation of the inflammatory response by using a DAMP an-
tigen rather than using an antibody or antagonist.
Ischemic preconditioning with HMGB1 protected grafts
from IRI through TLR4 signaling in renal and hepatic al-
lotransplantation [59,60]. In addition, genetic over-
expression of HSP27 could reduce IRI-induced apoptosis
of graft cells and delay the onset of acute rejection in
murine heart allotransplantation [61].
Targeting Macrophages
Deletion or inhibition of macrophages can attenuate graft
injury and prolong graft survival [62]. In recent animal
and clinical studies, some macrophage subsets have been
reported to act as regulatory cells, and the adoptive
transfer of these macrophages significantly prolonged
graft survival. A subset of macrophages was found to
suppress allogeneic T cell proliferation and inhibit den-
dritic cell maturation [63,64]. Furthermore, adoptive
transfer of these macrophages promotes graft survival
and minimizes immunosuppression [64,65].
Although immunological memory has long been thought
to be driven exclusively by adaptive immunity, new evi-
dence suggests that various tissue-derived factors can
induce epigenetic changes, leading to the formation of in-
nate memory of macrophages [66]. Further under-
standing of the mechanisms of innate memory could allow
79
Kim JY. Macrophages in xenotransplantation
us to dampen the response to DAMPs induced during the
xenograft rejection.
CONCLUSIONS
Although T cells are major players of organ transplant
rejection, early inflammation after transplantation is
critical for T-cell mediated immune rejection. Thus,
prevention of inflammation following transplantation
might help avoid acute graft rejection. Monocytes/mac-
rophages are critical immune mediators of inflammation,
and thus are important targets for immune modulation.
Owing to phenotypical and functional heterogeneity of
macrophages, the identification and/or isolation of dis-
tinct subsets of macrophages involved in graft rejection
or tolerance is essential to develop macrophage-based
therapeutic strategies. Compared to allotransplantation,
xenotransplantation has some advantages, including fea-
sibility of genetic modification and preconditioning for
transplantation. Thus, understanding precise mecha-
nisms of macrophage-mediated immune responses in or-
gan xenotransplantation will enable establishment of
strategies to modulate macrophage function, which can
improve the outcomes of xenotransplantation in future
clinical practice.
ACKNOWLEDGMENTS
Conflict of Interest
No potential conflict of interest relevant to this article
was reported.
Funding/Support
This study was supported by research grant from the Korean
Society for Transplantation (2019-04-03001-002).
ORCID
Jae Young Kim https://orcid.org/0000-0001-9323-3580
REFERENCES
1. Samstein B, Platt JL. Physiologic and immunologic hurdles
to xenotransplantation. J Am Soc Nephrol 2001;12:
182-93.
2. Galili U. The alpha-gal epitope and the anti-Gal antibody
in xenotransplantation and in cancer immunotherapy.
Immunol Cell Biol 2005;83:674-86.
3. Cooper DK, Ekser B, Tector AJ. Immunobiological bar-
riers to xenotransplantation. Int J Surg 2015;23(Pt B):
211-6.
4. Denner J. Xenotransplantation-progress and problems:
a review. J Transplant Technol Res 2014;4:1000133.
5. Zecher D, van Rooijen N, Rothstein DM, Shlomchik WD,
Lakkis FG. An innate response to allogeneic nonself medi-
ated by monocytes. J Immunol 2009;183:7810-6.
6. Oberbarnscheidt MH, Zeng Q, Li Q, Dai H, Williams AL,
Shlomchik WD, et al. Non-self recognition by monocytes
initiates allograft rejection. J Clin Invest 2014;124:
3579-89.
7. Liu W, Xiao X, Demirci G, Madsen J, Li XC. Innate
NK cells and macrophages recognize and reject allogeneic
nonself in vivo via different mechanisms. J Immunol
2012;188:2703-11.
8. Matheson PJ, Dittmer ID, Beaumont BW, Merrilees MJ,
Pilmore HL. The macrophage is the predominant in-
flammatory cell in renal allograft intimal arteritis.
Transplantation 2005;79:1658-62.
9. Xu L, Collins J, Drachenberg C, Kukuruga D, Burke A.
Increased macrophage density of cardiac allograft biopsies
is associated with antibody-mediated rejection and alloan-
tibodies to HLA antigens. Clin Transplant 2014;28:
554-60.
10. Bergler T, Jung B, Bourier F, Kühne L, Banas MC,
Rümmele P, et al. Infiltration of macrophages correlates
with severity of allograft rejection and outcome in human
kidney transplantation. PLoS One 2016;11:e0156900.
11. Bräsen JH, Khalifa A, Schmitz J, Dai W, Einecke G,
Schwarz A, et al. Macrophage density in early surveillance
biopsies predicts future renal transplant function. Kidney
Int 2017;92:479-89.
12. Matzinger P. Tolerance, danger, and the extended family.
Annu Rev Immunol 1994;12:991-1045.
13. Wu H, Chen G, Wyburn KR, Yin J, Bertolino P, Eris
JM, et al. TLR4 activation mediates kidney ischemia/re-
perfusion injury. J Clin Invest 2007;117:2847-59.
14. Kaczorowski DJ, Nakao A, Vallabhaneni R, Mollen KP,
Sugimoto R, Kohmoto J, et al. Mechanisms of Toll-like
receptor 4 (TLR4)-mediated inflammation after cold is-
chemia/reperfusion in the heart. Transplantation 2009;
87:1455-63.
15. Yang Z, Deng Y, Su D, Tian J, Gao Y, He Z, et al.
80
Korean J TransplantㆍDecember 2019ㆍVolume 33ㆍIssue 4
TLR4 as receptor for HMGB1-mediated acute lung injury
after liver ischemia/reperfusion injury. Lab Invest 2013;
93:792-800.
16. Weber DJ, Gracon AS, Ripsch MS, Fisher AJ, Cheon
BM, Pandya PH, et al. The HMGB1-RAGE axis mediates
traumatic brain injury-induced pulmonary dysfunction in
lung transplantation. Sci Transl Med 2014;6:252ra124.
17. Hu Q, Wood CR, Cimen S, Venkatachalam AB, Alwayn
IP. Mitochondrial damage-associated molecular patterns
(MTDs) are released during hepatic ischemia reperfusion
and induce inflammatory responses. PLoS One 2015;
10:e0140105.
18. De Vlaminck I, Martin L, Kertesz M, Patel K, Kowarsky
M, Strehl C, et al. Noninvasive monitoring of infection
and rejection after lung transplantation. Proc Natl Acad
Sci U S A 2015;112:13336-41.
19. Gielis EM, Ledeganck KJ, De Winter BY, Del Favero
J, Bosmans JL, Claas FH, et al. Cell-free DNA: an up-
coming biomarker in transplantation. Am J Transplant
2015;15:2541-51.
20. Baba HA, Schmid KW, Schmid C, Blasius S, Heinecke
A, Kerber S, et al. Possible relationship between heat
shock protein 70, cardiac hemodynamics, and survival
in the early period after heart transplantation. Transplan-
tation 1998;65:799-804.
21. Schimke I, Lutsch G, Schernes U, Kruse I, Dübel HP,
Pregla R, et al. Increased level of HSP27 but not of HSP72
in human heart allografts in relation to acute rejection.
Transplantation 2000;70:1694-7.
22. Sarri S, Shaw SM, Gieschen-Krische MA, Archer L,
Yonan N, Fildes JE. Myocardial heat shock protein 60
expression is upregulated following acute cardiac
rejection. Transpl Immunol 2009;21:140-2.
23. Maehana T, Tanaka T, Kitamura H, Fukuzawa N, Ishida
H, Harada H, et al. Heat shock protein 90 is a potential
serological biomarker of acute rejection after renal
transplantation. PLoS One 2016;11:e0162942.
24. Esmon CT. Molecular circuits in thrombosis and inflammation.
Thromb Haemost 2013;109:416-20.
25. Idzko M, Ferrari D, Eltzschig HK. Nucleotide signalling
during inflammation. Nature 2014;509:310-7.
26. Yoshida O, Dou L, Kimura S, Yokota S, Isse K, Robson
SC, et al. CD39 deficiency in murine liver allografts pro-
motes inflammatory injury and immune-mediated rejection.
Transpl Immunol 2015;32:76-83.
27. Jennewein C, Paulus P, Zacharowski K. Linking in-
flammation and coagulation: novel drug targets to treat
organ ischemia. Curr Opin Anaesthesiol 2011;24:375-80.
28. Osterud B, Olsen JO, Bjørklid E. What is blood borne
tissue factor? Thromb Res 2009;124:640-1.
29. Ezzelarab M, Garcia B, Azimzadeh A, Sun H, Lin CC,
Hara H, et al. The innate immune response and activation
of coagulation in alpha1,3-galactosyltransferase gene-
knockout xenograft recipients. Transplantation 2009;87:
805-12.
30. Lin CC, Ezzelarab M, Shapiro R, Ekser B, Long C, Hara
H, et al. Recipient tissue factor expression is associated
with consumptive coagulopathy in pig-to-primate kidney
xenotransplantation. Am J Transplant 2010;10:1556-68.
31. Foley JH, Conway EM. Cross talk pathways between co-
agulation and inflammation. Circ Res 2016;118:1392-
408.
32. Gao H, Liu L, Zhao Y, Hara H, Chen P, Xu J, et al.
Human IL-6, IL-17, IL-1, and TNF- differently regu-
late the expression of pro-inflammatory related genes,
tissue factor, and swine leukocyte antigen class I in porcine
aortic endothelial cells. Xenotransplantation 2017;24:
e12291.
33. Iwase H, Ekser B, Zhou H, Dons EM, Cooper DK,
Ezzelarab MB. Platelet aggregation in humans and nonhu-
man primates: relevance to xenotransplantation. Xeno-
transplantation 2012;19:233-43.
34. Ezzelarab MB, Cooper DK. Systemic inflammation in xen-
ograft recipients (SIXR): a new paradigm in pig- to-pri-
mate xenotransplantation? Int J Surg 2015;23(Pt
B):301-5.
35. Ezzelarab MB, Ekser B, Azimzadeh A, Lin CC, Zhao
Y, Rodriguez R, et al. Systemic inflammation in xenograft
recipients precedes activation of coagulation. Xenotrans-
plantation 2015;22:32-47.
36. Iwase H, Ekser B, Zhou H, Liu H, Satyananda V, Humar
R, et al. Further evidence for sustained systemic in-
flammation in xenograft recipients (SIXR). Xenotrans-
plantation 2015;22:399-405.
37. Dai H, Friday AJ, Abou-Daya KI, Williams AL, Mortin-
Toth S, Nicotra ML, et al. Donor SIRPa polymorphism
modulates the innate immune response to allogeneic
grafts. Sci Immunol 2017;2:eaam6202.
38. Ide K, Wang H, Tahara H, Liu J, Wang X, Asahara T,
et al. Role for CD47-SIRPalpha signaling in xenograft
rejection by macrophages. Proc Natl Acad Sci U S A
2007;104:5062-6.
39. Todd JL, Palmer SM. Danger signals in regulating the
immune response to solid organ transplantation. J Clin
Invest 2017;127:2464-72.
40. Roh JS, Sohn DH. Damage-associated molecular patterns
81
Kim JY. Macrophages in xenotransplantation
in inflammatory diseases. Immune Netw 2018;18:e27.
41. Tena A, Kurtz J, Leonard DA, Dobrinsky JR, Terlouw
SL, Mtango N, et al. Transgenic expression of human
CD47 markedly increases engraftment in a murine model
of pig-to-human hematopoietic cell transplantation. Am
J Transplant 2014;14:2713-22.
42. Hancock WW, Thomson NM, Atkins RC. Composition
of interstitial cellular infiltrate identified by monoclonal
antibodies in renal biopsies of rejecting human renal
allografts. Transplantation 1983;35:458-63.
43. Candinas D, Lesnikoski BA, Robson SC, Miyatake T,
Scesney SM, Marsh HC Jr, et al. Effect of repetitive
high-dose treatment with soluble complement receptor
type 1 and cobra venom factor on discordant xenograft
survival. Transplantation 1996;62:336-42.
44. Lin Y, Vandeputte M, Waer M. Natural killer cell- and
macrophage-mediated rejection of concordant xenografts
in the absence of T and B cell responses. J Immunol 1997;
158:5658-67.
45. Samy KP, Davis RP, Gao Q, Martin BM, Song M, Cano
J, et al. Early barriers to neonatal porcine islet engraft-
ment in a dual transplant model. Am J Transplant 2018;
18:998-1006.
46. Ehrnfelt C, Kumagai-Braesch M, Uzunel M, Holgersson
J. Adult porcine islets produce MCP-1 and recruit human
monocytes in vitro. Xenotransplantation 2004;11:184-94.
47. An HJ, Jang JW, Bae SH, Choi JY, Yoon SK, Lee MA,
et al. Serum C-reactive protein is a useful biomarker
for predicting outcomes after liver transplantation in pa-
tients with hepatocellular carcinoma. Liver Transpl
2012;18:1406-14.
48. Boras E, Slevin M, Alexander MY, Aljohi A, Gilmore
W, Ashworth J, et al. Monomeric C-reactive protein and
Notch-3 co-operatively increase angiogenesis through
PI3K signalling pathway. Cytokine 2014;69:165-79.
49. Han KH, Hong KH, Park JH, Ko J, Kang DH, Choi KJ,
et al. C-reactive protein promotes monocyte chemo-
attractant protein-1—mediated chemotaxis through upre-
gulating CC chemokine receptor 2 expression in human
monocytes. Circulation 2004;109:2566-71.
50. Kruithof EK, Mestries JC, Gascon MP, Ythier A. The
coagulation and fibrinolytic responses of baboons after
in vivo thrombin generation: effect of interleukin 6.
Thromb Haemost 1997;77:905-10.
51. Hage FG, Szalai AJ. C-reactive protein gene poly-
morphisms, C-reactive protein blood levels, and car-
diovascular disease risk. J Am Coll Cardiol 2007;50:
1115-22.
52. Li J, Hara H, Wang Y, Esmon C, Cooper DK, Iwase
H. Evidence for the important role of inflammation in
xenotransplantation. J Inflamm (Lond) 2019;16:10.
53. Cooper DK, Dou KF, Tao KS, Yang ZX, Tector AJ, Ekser
B. Pig liver xenotransplantation: a review of progress
toward the clinic. Transplantation 2016;100:2039-47.
54. Engel D, Seijkens T, Poggi M, Sanati M, Thevissen L,
Beckers L, et al. The immunobiology of CD154-CD40-
TRAF interactions in atherosclerosis. Semin Immunol
2009;21:308-12.
55. Li T, Lee W, Hara H, Long C, Ezzelarab M, Ayares
D, et al. An investigation of extracellular histones in
pig-to-baboon organ xenotransplantation. Transplanta-
tion 2017;101:2330-9.
56. Iwase H, Liu H, Li T, Zhang Z, Gao B, Hara H, et
al. Therapeutic regulation of systemic inflammation in
xenograft recipients. Xenotransplantation 2017;24:e12296.
57. Chung H, Hong SJ, Choi SW, Koo JY, Kim M, Kim HJ,
et al. High mobility group box 1 secretion blockade results
in the reduction of early pancreatic islet graft loss.
Biochem Biophys Res Commun 2019;514:1081-6.
58. Vergani A, Tezza S, D'Addio F, Fotino C, Liu K, Niewczas
M, et al. Long-term heart transplant survival by targeting
the ionotropic purinergic receptor P2X7. Circulation
2013;127:463-75.
59. Wu H, Steenstra R, de Boer EC, Zhao CY, Ma J, van
der Stelt JM, et al. Preconditioning with recombinant
high-mobility group box 1 protein protects the kidney
against ischemia-reperfusion injury in mice. Kidney Int
2014;85:824-32.
60. Izuishi K, Tsung A, Jeyabalan G, Critchlow ND, Li J,
Tracey KJ, et al. Cutting edge: high-mobility group box
1 preconditioning protects against liver ischemia-re-
perfusion injury. J Immunol 2006;176:7154-8.
61. Seemampillai B, Germack R, Felkin LE, McCormack A,
Rose ML. Heat shock protein-27 delays acute rejection
after cardiac transplantation: an experimental model.
Transplantation 2014;98:29-38.
62. Qi F, Adair A, Ferenbach D, Vass DG, Mylonas KJ,
Kipari T, et al. Depletion of cells of monocyte lineage
prevents loss of renal microvasculature in murine kidney
transplantation. Transplantation 2008;86:1267-74.
63. Hutchinson JA, Riquelme P, Sawitzki B, Tomiuk S,
Miqueu P, Zuhayra M, et al. Cutting edge: immunological
consequences and trafficking of human regulatory macro-
phages administered to renal transplant recipients. J
Immunol 2011;187:2072-8.
64. Riquelme P, Haarer J, Kammler A, Walter L, Tomiuk
82
Korean J TransplantㆍDecember 2019ㆍVolume 33ㆍIssue 4
S, Ahrens N, et al. TIGIT(+) iTregs elicited by human
regulatory macrophages control T cell immunity. Nat
Commun 2018;9:2858.
65. Hutchinson JA, Brem-Exner BG, Riquelme P, Roelen
D, Schulze M, Ivens K, et al. A cell-based approach
to the minimization of immunosuppression in renal
transplantation. Transpl Int 2008;21:742-54.
66. Netea MG, Latz E, Mills KH, O'Neill LA. Innate immune
memory: a paradigm shift in understanding host defense.
Nat Immunol 2015;16:675-9.
... In contrast, the expression of pro-inflammatory mediators was significantly suppressed, whereas that of the immunosuppressive mediators IL-10 and IDO was obviously enhanced in cocultured Mregs and MPN3 cells ( Figure 8A). The coagulation-associated mediators fgl2, PAR-1, and TF play important roles in inflammation and coagulation during xenograft rejection [39]. We examined the mRNA expression of these factors in THP-1 cells, M1 macrophages, and Mregs cocultured with MPN3 cells. ...
Article
Full-text available
Regulatory macrophages (Mregs) are unique in that they have anti-inflammatory and immunosuppressive properties. Thus, treating inflammatory diseases using Mregs is an area of active research. Human Mregs are usually generated by culturing peripheral blood monocytes stimulated using a macrophage colony-stimulating factor with interferon (IFN)-γ. Herein, we generated Mregs with an elongated cell morphology from THP-1 cells that were stimulated with phorbol 12-myristate 13-acetate and cultured with both arginylglycylaspartic acid and vitamin D3. These Mregs regulated macrophage function, and respectively downregulated and upregulated the expression of pro-inflammatory and immunosuppressive mediators. They also expressed Mregs-specific markers, such as dehydrogenase/reductase 9, even when exposed to such inflammatory stimulants as IFN-γ, lipopolysaccharide, purified xenogeneic antigen, and xenogeneic cells. The Mregs also exerted anti-inflammatory and anticoagulatory actions in response to xenogeneic cells, as well as exerting immunosuppressive effects on mitogen-induced Jurkat T-cell proliferation. Our method of generating functional Mregs in vitro without cytokines is simple and cost-effective.
Article
Due to a critical organ shortage, pig organs are being explored for use in transplantation. Differences between species, particularly in cell surface glycans, can trigger elevated immune responses in xenotransplantation. To mitigate the risk of hyperacute rejection, genetically modified pigs have been developed that lack certain glycans and express human complement inhibitors. Nevertheless, organs from these pigs may still provoke stronger inflammatory and innate immune reactions than allotransplants. Dysregulation of coagulation and persistent inflammation remain obstacles in the transplantation of pig organs into primates. Regulatory macrophages (Mregs), known for their anti-inflammatory properties, could offer a potential solution. Mregs secrete interleukin 10 and transforming growth factor beta, thereby suppressing immune responses and promoting the development of regulatory T cells. These Mregs are typically induced via the stimulation of monocytes or macrophages with macrophage colony-stimulating factor and interferon gamma, and they conspicuously express the stable marker dehydrogenase/reductase 9. Consequently, understanding the precise mechanisms governing Mreg generation, stability, and immunomodulation could pave the way for the therapeutic use of Mregs generated in vitro. This approach has the potential to reduce the required dosages and durations of anti-inflammatory and immunosuppressive medications in preclinical and clinical settings.
Article
Full-text available
There is increasing evidence of a sustained state of systemic inflammation after pig-to-nonhuman primate (NHP) xenotransplantation (that has been termed systemic inflammation in xenograft recipients [SIXR]). Increases in inflammatory markers, e.g., C-reactive protein, histones, serum amyloid A, D-dimer, cytokines, chemokines, and a decrease in free triiodothyronine, have been demonstrated in the recipient NHPs. The complex interactions between inflammation, coagulation, and the immune response are well-recognized, but the role of inflammation in xenograft recipients is not fully understood. The evidence suggests that inflammation can promote the activation of coagulation and the adaptive immune response, but the exact mechanisms remain uncertain. If prolonged xenograft survival is to be achieved, anti-inflammatory strategies (e.g., the administration of anti-inflammatory agents, and/or the generation of genetically-engineered organ-source pigs that are protected from the effect of inflammation) may be necessary to prevent, control, or negate the effect of the systemic inflammation that develops in xenograft recipients. This may allow for a reduction in the intensity of exogenous immunosuppressive therapy. If immunological tolerance to a xenograft is to be obtained, then control of inflammation may be essential.
Article
Full-text available
Damage-associated molecular patterns (DAMPs) are endogenous danger molecules that are released from damaged or dying cells and activate the innate immune system by interacting with pattern recognition receptors (PRRs). Although DAMPs contribute to the host's defense, they promote pathological inflammatory responses. Recent studies have suggested that various DAMPs, such as high-mobility group box 1 (HMGB1), S100 proteins, and heat shock proteins (HSPs), are increased and considered to have a pathogenic role in inflammatory diseases. Here, we review current research on the role of DAMPs in inflammatory diseases, including rheumatoid arthritis, systemic lupus erythematosus, osteoarthritis, atherosclerosis, Alzheimer's disease, Parkinson's disease, and cancer. We also discuss the possibility of DAMPs as biomarkers and therapeutic targets for these diseases.
Article
Full-text available
Human regulatory macrophages (Mreg) have shown early clinical promise as a cell-based adjunct immunosuppressive therapy in solid organ transplantation. It is hypothesised that recipient CD4+ T cell responses are actively regulated through direct allorecognition of donor-derived Mregs. Here we show that human Mregs convert allogeneic CD4+ T cells to IL-10-producing, TIGIT+ FoxP3+-induced regulatory T cells that non-specifically suppress bystander T cells and inhibit dendritic cell maturation. Differentiation of Mreg-induced Tregs relies on multiple non-redundant mechanisms that are not exclusive to interaction of Mregs and T cells, including signals mediated by indoleamine 2,3-dioxygenase, TGF-β, retinoic acid, Notch and progestagen-associated endometrial protein. Preoperative administration of donor-derived Mregs to living-donor kidney transplant recipients results in an acute increase in circulating TIGIT+ Tregs. These results suggest a feed-forward mechanism by which Mreg treatment promotes allograft acceptance through rapid induction of direct-pathway Tregs.
Article
Full-text available
Inflammation impairs renal allograft survival but is difficult to quantify by eye at low densities. Here we measured leukocyte abundance in early surveillance biopsies by digital image analysis to test for a role of chemokine receptor genotypes and analyze the predictive value of leukocyte subsets to allograft function. In six-week surveillance biopsies, T-cell (CD3), B-cell (CD20), macrophage (CD68), and dendritic cell (CD209) densities were assessed in whole slide scans. Renal cortical CD3, CD20, and CD68 were significantly higher in histologic rejection. The CCR2 V64I genotype was associated with lower CD3 and CD209 densities. Above-median CD68 density was significantly associated with lower combined patient and graft survival with a hazard ratio of 3.5 (95% confidence interval 1.1-11.0). Both CD20 and CD68 densities inversely correlated with estimated glomerular filtration rate (eGFR) four years after transplantation. Additionally, CD68 correlated with eGFR loss. Among histological measurements including a complete Banff classification, only CD68 density was a significant predictor of an eGFR under 30ml/min after four years (odds ratio 7.4, 1.8-31.0) and part of the best eGFR prediction set in a multivariable linear regression analysis of multiple clinical and pathologic parameters. In a second independent cohort, the original CD68 median maintained its discriminative power for survival and eGFR. Thus, digital high-resolution assessment of CD68(+) leukocyte infiltration significantly improves prognostic value of early renal transplant biopsies.
Article
Pancreatic islet transplantation has been known as the best cure for patients suffering from severe type 1 diabetes mellitus (T1DM). Despite meaningful advances in human allogeneic islet transplantation field, significant amounts of islet loss in early post-transplantation periods is still a big concern for clinicians. One of the major factors determining the fate of the islets is the danger-associated molecular patterns (DAMPs) secreted by activated immune cells or islets themselves under hypoxic stress. High mobility group box 1 (HMGB1) protein is one of the best characterized DAMP molecules associated with islets. HMGB1 is known to be passively released by transplanted murine islet cells after taking damages from cytokines, reactive oxygen species, and other DAMPS, and the released HMGB1 harms neighboring islet cells by interacting with receptors expressed on murine islets such as toll-like receptor 2 (TLR2) and TLR4, thereby forming a vicious cycle. Here, we show that a small molecule inhibitor inflachromene (ICM) was capable of blocking the secretion of HMGB1 from murine islet cells during the normoxic and hypoxic post-isolation period. Notably, the treatment of ICM during the islet isolation process resulted in decreased HMGB1 levels during the subsequent cell culture. ICM's in vivo efficacy was evaluated in murine syngeneic islet transplantation model, and it significantly reduced the serum and graft level of HMGB1. Ultimately, the intraperitoneal administration of ICM prevented the loss of marginal-mass islet grafts and reversed the diabetes in mice.
Article
Porcine islet xenografts have the potential to provide an inexhaustible source of islets for β cell replacement. Proof-of-concept has been established in nonhuman primates. However, significant barriers to xenoislet transplantation remain, including, the poorly understood instant blood mediated inflammatory reaction (IBMIR), and a thorough understanding of early xeno-specific immune responses. A paucity of data exist comparing xeno-specific immune responses to allo-islet responses in primates. We recently developed a dual islet transplant model, which enables direct histological comparison of early engraftment immunobiology. In this study, we investigate early immune responses to neonatal porcine islet (NPI) xenografts compared to rhesus islet allografts at 1 hour, 24 hours, and 7 days. Within the first 24 hours after intraportal infusion, we identified greater apoptosis (caspase 3 activity and TUNEL positive cells) of NPIs when compared to alloislets (AIs). Macrophage infiltration was significantly greater at 24 hours compared with 1 hour in both NPI (WT) and AIs. At 7 days, IgM and macrophages were highly specific for NPIs (GTKO) compared to AIs. These findings demonstrate an augmented macrophage and antibody response towards xenografts compared to allografts. These data may inform future immune or genetic manipulations required to improve xenoislet engraftment.
Article
Mice devoid of T, B, and natural killer (NK) cells distinguish between self and allogeneic nonself despite the absence of an adaptive immune system. When challenged with an allograft, they mount an innate response characterized by accumulation of mature, monocyte-derived dendritic cells (DCs) that produce interleukin-12 and present antigen to T cells. However, the molecular mechanisms by which the innate immune system detects allogeneic nonself to generate these DCs are not known. To address this question, we studied the innate response of Rag2 -/- γc -/- mice, which lack T, B, and NK cells, to grafts from allogeneic donors. By positional cloning, we identified that donor polymorphism in the gene encoding signal regulatory protein α (SIRPα) is a key modulator of the recipient's innate allorecognition response. Donors that differed from the recipient in one or both Sirpa alleles elicited an innate alloresponse. The response was mediated by binding of donor SIRPα to recipient CD47 and was modulated by the strength of the SIRPα-CD47 interaction. Therefore, sensing SIRPα polymorphism by CD47 provides a molecular mechanism by which the innate immune system distinguishes between self and allogeneic nonself independently of T, B, and NK cells.
Article
Endogenous danger signals, or damage-associated molecular patterns (DAMPs), are generated in response to cell stress and activate innate immunity to provide a pivotal mechanism by which an organism can respond to damaged self. Accumulating experimental and clinical data have established the importance of DAMPs, which signal through innate pattern recognition receptors (PRRs) or DAMP-specific receptors, in regulating the alloresponse to solid organ transplantation (SOT). Moreover, DAMPs may incite distinct downstream cellular responses that could specifically contribute to the development of allograft fibrosis and chronic graft dysfunction. A growing understanding of the role of DAMPs in directing the immune response to transplantation has suggested novel avenues for the treatment or prevention of allograft rejection that complement contemporary immunosuppression and could lead to improved outcomes for solid organ recipients.
Article
Inflammation is known to preclude tolerance after transplantation. We have previously shown that systemic inflammation in xenograft recipients (SIXR) precedes activation of coagulation in the absence of T cell responses. Accordingly, SIXR may amplify innate and adaptive immune responses against xenografts after pig-to-primate xenotransplantation, even with efficient immunosuppressive therapy. We evaluated the impact of anti-inflammatory agents on pro-inflammatory cytokines and chemokines in pig artery patch and heart xenograft recipients. Baboons received an artery patch (Group1, n=8) or heart (Group2, n=4) from genetically engineered pigs. All baboons received lymphodepletion with thymoglobulin (ATG) and costimulation blockade-based immunosuppression (anti-CD40 and/or CTLA4Ig). In Group1, baboons received either (i) no anti-inflammatory agents (n=2), (ii) cobra venom factor (CVF, n=2), (iii) α1-antitrypsin (AAT, n=2), or (iv) interleukin (IL)-6 receptor antagonist (IL-6RA, n=2). In Group2, all baboon received corticosteroids, either without (n=2) or with (n=2) IL-6RA. Serum IFN-γ, TNF-α, IL-1β, IL-17, IL-6, IL-8, MCP-1, and sCD40L levels were measured by Luminex. Fibrinogen, D-dimers, and C-reactive protein (C-RP) were also measured. Recipient baboon T cell proliferation was evaluated by mixed lymphocyte reaction (MLR) before and after transplantation. Pig and baboon tissue factor (TF) mRNA levels in heart xenografts were measured by RT-PCR. In no recipient was a marked increase in T cell response to pig cells observed after transplantation. In Groups 1 and 2, post-transplantation levels of IFN-γ, TNF-α, IL-1β, and IL-17 remained comparable to or lower than pre-transplant levels, except in one heart recipient that succumbed to CMV infection. In Group1, when no anti-inflammatory agent was administered, post-transplant levels of IL-6, IL-8, and MCP-1 were elevated. After CVF, IL-6, IL-8, and MCP-1 remained low. After IL-6RA, IL-6 and MCP-1 were elevated. After AAT, IL-8 was elevated. sCD40L became elevated intermittently in most recipients irrespective of the administered anti-inflammatory agent. In Group2, IL-6 was transiently elevated, particularly after IL-6RA administration. MCP-1 gradually increased by 2 months in Group2 recipients. sCD40L generally remained low except in one recipient. In Group1 and Group2 recipients, C-RP levels were elevated except after IL-6RA administration, while D-dimers were elevated regardless of administration of anti-inflammatory agent. In Group2, pig TF mRNA levels were increased in heart xenografts compared to naive pig hearts, irrespective of IL-6 receptor antagonist administration. Additionally, baboon TF mRNA levels were detectable in heart xenografts, but not in naive pig hearts. Some pro-inflammatory cytokines and chemokines are elevated in xenograft recipients, even with efficient T cell-directed immunosuppressive therapy. Persistent elevation of D-dimers, and individual cytokines and chemokines suggest a continuous inflammatory response, despite administration of anti-inflammatory agents. Systemic administration of combined anti-inflammatory agents as well as complement regulation may be essential to prevent SIXR after xenotransplantation.