You are on page 1of 16

International Reviews of Immunology

ISSN: 0883-0185 (Print) 1563-5244 (Online) Journal homepage: https://www.tandfonline.com/loi/iiri20

Triggering receptor expressed on myeloid cells-1


(TREM-1) as a therapeutic target in infectious and
noninfectious disease: a critical review

Pedro Henrique dos Santos Dantas, Amanda de Oliveira Matos, Ernandes da


Silva Filho, Marcelle Silva-Sales & Helioswilton Sales-Campos

To cite this article: Pedro Henrique dos Santos Dantas, Amanda de Oliveira Matos,
Ernandes da Silva Filho, Marcelle Silva-Sales & Helioswilton Sales-Campos (2020): Triggering
receptor expressed on myeloid cells-1 (TREM-1) as a therapeutic target in infectious
and noninfectious disease: a critical review, International Reviews of Immunology, DOI:
10.1080/08830185.2020.1762597

To link to this article: https://doi.org/10.1080/08830185.2020.1762597

Published online: 07 May 2020.

Submit your article to this journal

Article views: 50

View related articles

View Crossmark data

Full Terms & Conditions of access and use can be found at


https://www.tandfonline.com/action/journalInformation?journalCode=iiri20
INTERNATIONAL REVIEWS OF IMMUNOLOGY
https://doi.org/10.1080/08830185.2020.1762597

REVIEW

Triggering receptor expressed on myeloid cells-1 (TREM-1) as a therapeutic


target in infectious and noninfectious disease: a critical review
Pedro Henrique dos Santos Dantas , Amanda de Oliveira Matos , Ernandes da Silva Filho ,
Marcelle Silva-Sales , and Helioswilton Sales-Campos
Institute of Tropical Pathology and Public Health, Federal University of Goias, Goi^ania, Goias, Brazil

ABSTRACT ARTICLE HISTORY


The triggering receptor expressed on myeloid cells-1 (TREM-1) is an innate immune receptor Received 23 December 2019
found in the surface of several immune and non-immune cells. Since its first description in Accepted 23 April 2020
2000, this molecule and its soluble form (sTREM-1) have been implicated in many diseases
KEYWORDS
with infectious and noninfectious origins. As an amplifier of inflammation, the membrane-
Anti-inflammatory drugs;
associated TREM-1 (mTREM-1) isoform induces the production of pro-inflammatory media- inflammation; TREM-1
tors, thus contributing to the pathogenesis of diseases such as sepsis, arthritis, colitis and
infections. In this context, many studies have used molecules capable of inhibiting TREM-1
activity as anti-inflammatory drugs. In this regard, a few peptides have been showing prom-
ising results in the amelioration of detrimental immune responses. Some commercially avail-
able drugs, including corticosteroids and antibiotics, with known anti-inflammatory effects,
have also shown activity in TREM-1 signaling. Therefore, considering the potential of this
receptor as a therapeutic target, the present review encompasses the main compounds
explored so far in TREM-1 modulation, highlighting and critically discussing its effects and
major drawbacks of such approaches.

Introduction endothelial cells and gastric epithelial cells, especially


The immune system is constantly challenged by sev- during inflammatory processes [5]. In these contexts,
eral infectious and noninfectious inflammatory trig- TREM-1 was proposed to act as an amplifier of sig-
gers [1]. These molecules are initially recognized by naling by other PRRs, such as Toll-like receptor 4
pattern recognition receptors (PRRs), that are located (TLR-4), thus helping to increase the inflammatory
on the surface and intracellularly of immune and response [6]. However, the amplification of inflamma-
non-immune cells recognizing damaged-associated tion may result in undesirable side effects, such as
molecular patterns (DAMPs), and microbial-associated those observed in sepsis and in other immune medi-
molecular patterns (MAMPs) [2]. Among these PRRs, ated diseases with infectious and noninfectious origins
the family of the triggering receptor expressed on [4,7,8]. This role as an amplifier of inflammation was
myeloid cells (TREM) has attracted considerable reinforced in TREM-1 knockout mice exposed to
attention in the last decades. Composed of TREM-1, inflammatory and infectious agents [9]. After dextran
TREM-2, TREM-3 and the ITIM-bearing TREM-like
sulfate sodium-induced colitis, the receptor deficiency
transcripts (TLT-1, 2, 3, 4, 5) [3], these recep-
was associated with reduced expression of inflamma-
tors were first described by Bouchon and colleagues
tory cytokines and inflammatory infiltrate in colon,
in 2000 [4].
In the pioneer study, TREM-1 was reported to be thus resulting in a better clinical outcome [9]. Despite
expressed on the surface of monocytes and neutro- the similarity regarding the reduction in inflammatory
phils activated by lipopolysaccharide (LPS), and was infiltrate and morbidity after parasite and viral infec-
implicated in acute inflammation [4]. In addition, tion, TREM-1 deficiency had no impact on pathogen
TREM-1 has also been shown to be expressed on the clearance [9]. These data suggest that targeting this
surface of non-immune cells, such as hepatic receptor may represent an advantageous and safe

CONTACT Helioswilton Sales-Campos tonsales@ufg.br Instituto de Patologia Tropical e Sa ublica – IPTSP, Universidade Federal de Goias –
ude P
UFG, Rua 235 S/N Setor Leste Universitario, Goi^ania, Goias 746050-050, Brazil
These authors equally collaborated to this manuscript.
ß 2020 Taylor & Francis Group, LLC
2 P. H. DOSSANTOSDANTAS ET AL.

Figure 1. TREM-1 signaling pathway and compounds that modulate its activity. The triggering receptor expressed on myeloid
cells-1 (TREM-1) can be observed in two isoforms: membrane bound (mTREM-1) and soluble (sTREM-1). After activation, immunore-
ceptor tyrosine-based activation motifs (ITAM) are phosphorylated, and anchors Zeta-associated protein of 70 kDa (ZAP70) and tyro-
sine-protein kinase (SYK), which results in the activation of the intracellular pathways janus kinase (JAK), extracellular signal-
regulated kinase (ERK), phosphoinositide 3-kinases (PI3K) and phospholipase C-gamma (PLCe). These pathways activate the tran-
scription factors NFAT, AP-1, STAT3/STAT5 and NF-jB, involved in the production of inflammatory mediators. The activity of
mTREM-1 may be inhibited by numerous compounds: LR12 and LP17 act as decoy receptors, competing for the ligands of
mTREM-1 and blocking the oligomerization of the receptor; GF9 and sneaking ligand construct to triggering receptor expressed on
myeloid cells-1 (SLC-TREM-1) seems to interfere with the interaction between mTREM-1 and DAP12 in cell membrane; Vasoactive
intestinal peptide (VIP), Corticosteroids and statins inhibit the activity of NF-jB; Rapamycin inhibits mTOR and ERK1/ERK2 path-
ways, besides of inhibiting the activity of the transcription factor AP-1. The inhibitory mechanisms attributed to LL-37, Doxycycline,
Clarithromycin, Azithromycin, Imipenem and Tacrolimus, are still unclear. The pattern recognition receptors depicted in the figure
includes: toll-like receptors 2 (TLR2) and 4 (TLR4), nucleotide-binding oligomerization domain-like receptors (NLRs), nucleotide-bind-
ing oligomerization domain-like (NOD) and NOD-like receptor pyrin domain-containing protein 3 (NLRP3). Arrow – Activation or
agonist. Blunted arrow: Inhibition or antagonist.

approach in microbial control, which is not observed glycoproteins [10,11]. So far, two isoforms of this
when other inflammatory mediators are targeted. molecule were identified: the membrane-bound
Therefore, due to the importance of TREM-1 on (mTREM-1) and soluble (sTREM-1) forms [12,13].
the development of inflammation and pathogenesis of The mTREM-1 ( 30 kDa) has an extracellular
several diseases, this review aims to evaluate the role immune globulin-like V-type domain; a transmem-
of peptides and other immune modulatory drugs in brane region and a short cytoplasmic tail [10,11].
the modulation of this PRR. In the following sections, Because the receptor doesn’t present immunoreceptor
TREM-1 structure, signaling pathways, activation mol- tyrosine-based activation motifs (ITAMs) in its intra-
ecules and role as a pharmacological target will be cellular portion, it needs to be associated with an
critically addressed. adaptor protein, DAP12, for signaling transduction
[14]. This association occurs through a positively
Structure, ligands, expression, signaling and charged lysine in the receptor and a negatively
functions of TREM-1 charged aspartic acid in DAP12 [14]. Studies regard-
ing its structure have generated conflicting results in
Structure and ligands the past years, but recently, the receptor was proposed
The genes encoding the TREM family of receptors are to display both monomeric and oligomeric forms
located on chromosome 6p21 in humans and are [10,11,15]. The former is described as the inactive
INTERNATIONAL REVIEWS OF IMMUNOLOGY 3

form and the latter is observed after activation (Figure (PI3K) and Phosphoinositide phospholipase C gamma
1) [15]. (PLCc) pathways [41](Figure 1). These pathways acti-
The molecules that are able to bind and activate vate the transcription factors Elk-1, NFAT, AP1, c-Fos,
mTREM-1 are still a matter of strong investigation. c-Jun, STAT3/STAT5 and NF-jB, all involved in the
Among the putative ligands, it is possible to highlight production of inflammatory mediators such as IL-6,
actin in platelets [16,17], PGLYRP1 [18], HMGB1, IL-8, IL-1b and TNF-a (Figure 1), besides inducing cal-
Hsp70 [5,19] and the recently described extracellular cium mobilization and changes in the actin cytoskel-
cold-inducible RNA-binding protein (eCIRP) [20]. eton [42]. The activation of mTREM-1 also inhibits
Moreover, LPS, observed in the membrane of gram- pro-apoptotic molecules (BID, BAD and BAX) and the
negative bacteria, may act as a ligand because it is release of cytochrome C by mitochondria, thus main-
involved in TREM-1 activation [4,15]. However, it is taining mitochondrial integrity and prolonged cell sur-
still not clear if LPS activates TREM-1 directly or vival [5,35].
indirectly through TLR-4 activation [7]. Other pos- On the other hand, the expression of mTREM-1 is
sible MAMPs recognized by this receptor are zymo- downregulated by PU.1, which is a transcription factor
san, HIV gp120, Schistosoma mansoni egg antigens present in myeloid and lymphoid cells [38,43,44], and
and Marburg virus glycoprotein [21–25]. anti-inflammatory cytokines such as IL-10 [32,45] and
The sTREM-1 has only the Ig-like domain that is TGF-b [45]. In addition to cytokines, other com-
important for antigen recognition and to compete pounds capable of decreasing mTREM-1 activity
with mTREM-1 for the same ligands [26], thus it include cathelicidin LL-37 and CpG-ODN, that are
is assumed to play an anti-inflammatory role in both inhibitors of the activation induced by
different contexts. The origin of sTREM-1 is not LPS [46,47].
fully understood, and it is most likely to occur due
to proteolytic cleavage of mTREM-1 by metallopro-
Interaction between TLR and TREM-1
teinases (MMPs), such as MMP-9 [27]. Therefore,
the release of sTREM-1 depends on the activation The mTREM-1 increases and amplifies the TLR4 sig-
of TREM-1 in the membrane followed by its cleav- naling pathway, and this in turns leads to the
age [27,28]. increased expression of mTREM-1 [7], suggesting a
possible mechanism of mutual cooperation between
TREM-1 and TLRs. The relationship between
Expression and signaling of TREM-1
mTREM-1 and TLR is critical for enhancing the
Different situations and molecules have been impli- immune response, as together they are able to amplify
cated in the up-regulation of mTREM-1, including: both innate and adaptive immune responses through
hypoxia in dendritic cells [29], vitamin D3 [30], oxi- the increase of pro-inflammatory molecules [5,35].
dized low-density lipoprotein [31], Tumor necrosis The interaction between TLR4 and TREM-1 relies on
factor alpha (TNFa), Interleukin 1 beta (IL-1b), GM- the adaptor molecule MyD88 that is critical for the
CSF [32,33], PGE2 [34], cAMP [35] and compounds onset of their intracellular signaling [5,35]. In add-
derived from microorganisms, such as LPS and lipo- ition, MyD88 activity is linked to the activation of
teichoic acid (LTA) [36]. So far, these effects have transcription factors such as NF-jB and AP-1 that
been associated with the activity of transcription fac- regulate TREM-1 expression [4,48,49]. On the other
tors including the activator protein 1 (AP-1) and the hand, the inhibition of mTREM-1 seems to not dir-
nuclear-factor kappa B (NF-jB) [37,38]. ectly interfere with TLR4 expression [5,50]. However,
As mentioned above, mTREM-1 activity depends the inhibition of mTREM-1 downregulates several
on DAP12 for activation and intracellular signaling genes involved in the TLR-4 intracellular signaling,
[14] (Figure 1). After activation, ITAM motifs are such as MyD88, CD14 and IkBa [5,50], thus suggest-
phosphorylated by Src kinases [39,40]. Then, these ing that TLR4 has a greater and direct influence on
domains anchor zeta-chain-associated protein kinase TREM-1 activity, and the latter has mainly an indirect
70 (ZAP70) and spleen tyrosine kinase (SYK) [40]. role over TLR4 functionality.
After that, SYK phosphorylates casitas b-lineage lymph- Aside from the role of TLR4 in mTREM-1 activa-
oma (CBL), son of sevenless protein (SOS) and growth tion, other TLRs may also contribute to the activity of
factor receptor binding protein-2 (GRB2), leading to mTREM-1 in different contexts. The binding of LTA
the activation of Jakus kinase (JAK), Extracellular sig- to TLR2 enhances TREM-1 mRNA expression [36,51].
nal-regulated kinases (ERK), Phosphoinositide 3-kinase The mTREM-1 is able to amplify cellular signaling
4 P. H. DOSSANTOSDANTAS ET AL.

Table 1. Peptides and drugs that modulate TREM-1 activity.


Compound Mechanism Model/Disease/Effects Reference
LR12 Decoy Receptor Mice/Septic shock/#IL-6, IL-8, TNFa, IL-10;#complications [54]
Mice/Septic shock/#complications [59]
Pigs/Septic shock/#TNFa and IL-6;#complications [60]
Monkey/Sepsis/#IL-6, IL-8, TNFa;#complications [61]
Human primary monocytes/P.gingivalis/#IL-1b, TNFa, IL-8 [62]
Mice/IR/#TNFa [6]
Mice/AMI/#TREM-1;#IL-6, TNFa;#complications [63]
Pigs/AMI/#complications of disease [64]
Mice/Atherosclerosis/#TNFa, IL-12, IFN-c;#complications [65]
Mice/ALI/#IL-1b, TNFa, IL-6;"IL-10;#complications [66]
Mice/ALI/#IL-1b, TNFa, IL-6;"IL-10;#complications [58]
Mice/Colitis/IL-6, IL-1b, TNF-a;#complications [67]
LP17 Decoy receptor Mice/Sepsis/#TNF-a, IL-1b, IL-6;"sTREM-1;#complications [12]
Mice/Sepsis/#complications [68]
Rats/Sepsis/#TNF-a, IL-1b, IL-6;#sTREM-1;#complications [69]
Leucocytes/neonatal sepsis/#TNFa, IL-8, IL-6;#complications [70]
MonoMac-6/P.gingivalis/#TNFa, IL-6;#complications [71]
Mice/Periodontitis/#IL-17;#complications of disease [72]
Mice/Pneumonia/#IL-1b, TNFa, IL-6;#complications [73]
Mice/Empyema/#IL-1b, TNFa, IL-6;#complications [74]
Zebrafish and rats/PD/#TREM-1;#complications [75]
Rats/Early Brain Injury/#complications [76]
Mice/Ischemic shock/#IL-1b, IL-6;#complications [77]
Mice/Nephritis/#complications [78]
Mice/IR/#CXCL1 [6]
Mice/Colitis/#TNF-a, IL-6, IL-1b;#complications [79]
Rats/Acute Pancreatitis/#TNFa, IL-1b;#complications [80]
Rats/Acute Pancreatitis/#complications [81]
VSMCs/In-stent restenosis/#TREM-1;#complications [82]
Macrophages/Atherosclerosis/#IL-6 [31]
Rats/Hemorrhagic shock/#TNFa, IL-6;#complications [83]
Mice/Pneumonia/#TNF-a, IL-1b, IL-6;#complications [26]
Mice/ARDS/#TNF-a, IL-1b;#complications [84]
GF9 Blockade of mTREM-1 and DAP12 interaction Mice/Tumor and Sepsis/#TNF-a, IL-6, IL-1b;#complications [85]
Mice/Arthritis/#TREM-1;#IL-1, TNF-a, IL6;#complications [86]
Mice/Alcoholic Liver Disease/TNF-a, IL-1b;#TREM-1;#complications [87]
SLC-TREM-1 Blockade of mTREM-1 and DAP12 interaction Mice / Sepsis /#IL-8;#complications;#sTREM-1 [88]
LL-37 n.d. Mice/sepsis/#IL-1b, IL-6,sTREM-;#complications [89]
PBMCs/LPS/#mTREM-1 [46]
VIP n.d. Mice/ALI/#TREM-1 [90]
Dexamethasone Inhibition of NF-jB Mice and in vitro/P. aeruginosa / #TREM-1; #TNF-a; [91]

Methylprednisolone Inhibition of NF-jB Human patients / ARDS / #sTREM-1 ;#complications [92]


Azithromycin n.d. Mice/Sepsis /#TREM-1; #IL-6, TNF-a, IL-1;b #sTREM-1; #complications [93]
Clarithromycin n.d. Humans/Sepsis/"TREM-1;"sTREM-1;#complications [94]
Rapamycin Inhibition of mTOR Mice/IPA/#IL-6, IFN-c, "sTREM-1; #complications [95]
Tacrolimus n.d. Mice / Fungal keratitis /#TREM-1; #TNF-a, IL-1b [96]
Doxycicline n.d. Monocytes/P. gingivalis /#IL-8;#sTREM-1;#TREM-1 [97]
Iminipem n.d. Mice/Sepsis/#TNF-a, IL-6;#sTREM-1 [98]
Pravastatin Inhibition of NF-jB Mice/Artherosclerosis/#TREM-1;#IL-1, TNF-a;#TREM-1 [99]
PBMCs/LPS/#TREM-1;#IL-6, TNF-a;#sTREM-1 [100]
n.d: not determined; #:decrease;":increase; IR:Renal Ischemia Reperfusion; AMI: Acute Myocardial Infarction; ALI: Acute Lung Injury; P.D: Parkinsons disease;
VSMCs: Vascular Smooth Muscle Cells; ARDS: Acute respiratory distress syndrome; PBMCs:Peripheral Blood Cells; mTREM-1: Triggering expressed on mye-
loid cells 1 (membrane); sTREM-1: Triggering expressed on myeloid Cells 1 (Soluble); IL: Interleukine; NF-jB: nuclear-factor kappa B; mTOR: mammalian
target of rapamycin; TNF-a: Tumor necrosis factor alpha.

and the immune response triggered by TLR2 and activation of TLR9 by CpG-ODN, increases the pro-
TLR4 besides inhibiting the expression of negative duction of sTREM-1 [47], thus inhibiting the activity
regulators of these pathways, such as Tollip, ST2, of mTREM-1.
SIGIRR and IRAK-M [52,53]. The abrogation of In general, the interaction between TLRs and
TREM-1 activity decreases the production of the pro- TREM-1 seems to rely on a mutual contribution, and
inflammatory cytokines TNF-a and IL-6, which are is mainly dependent on the modulation of proteins
commonly produced after TLR4-activation [7,31]. The and pathways related to the activity of both family of
activity of mTREM-1 seems also to be dependent on receptors. Further, the outcome of the mutual inter-
TLR since the triggering receptor is apparently unable action between TREM-1 and TLR seems to rely on
to generate a sustained immune response without the nature of the ligands and the localization (mem-
TLR activation [36,54]. On the other hand, the brane or intracellular) of the TLR.
INTERNATIONAL REVIEWS OF IMMUNOLOGY 5

Interaction between NLR and TREM-1 IL-6, IL-8 and TNF-a [61] (Table 1). These effects
were mainly attributed to the ability of LR12 to com-
The family of nucleotide-binding oligomerization
pete with TREM-1 for the same ligands (thus acting
domain like receptors (NLR) are involved in the regu-
as a decoy receptor) [54] (Figure 1), and to impair
lation of inflammation and apoptotic responses [55]
TREM-1 multimerization, which is crucial for TREM-1
and, similarly to TLR, it cooperates with mTREM-1
activation [15].
functionality [56,57]. The interaction between NLRs
The role of LR12 has also been addressed in peri-
and mTREM-1 leads to the activation of monocytes,
odontal disease [62], that is also triggered by microor-
which triggers and amplifies the phosphorylation of
ganisms [101]. In human primary monocytes infected
Akt, besides of inducing the production of MAPK
with Porphyromonas gingivalis, the main species asso-
and pro-inflammatory mediators such as IL-1b and
ciated with periodontal disease, treatment with LR12
IL-6 [56,57].
reduced the expression of TREM-1 and production of
The activation of mTREM-1 is capable of inducing
pro-inflammatory cytokines such as IL-1b, TNF-a and
the expression of NOD2 and NF-jB, resulting in the
IL-8 (Table 1) [62]. Despite the results suggesting
production of IL-1b and IL-6 [56]. TREM-1 seems
LR12 as a promising candidate to treat periodontal
also to interact with NLRP3, one of the main compo-
diseases, in vivo studies are required to better under-
nents of the inflammasome [58] whose downstream
stand its role in this scenario.
activation promotes cleavage of pro-IL-1b and pro-
The mTREM-1 has been implicated in the onset
IL18 by caspase 1, leading to the production of the
and worsening of noninfectious diseases, including
active forms of these cytokines [55]. This mutual
renal ischemia reperfusion (IR) [6], acute myocardial
interaction is reinforced by the inhibition of mTREM-
infarction (AMI) [64], acute lung injury (ALI) [58],
1 by the peptide LR12, which reduced the expression
atherosclerosis [65] and colitis [67].
and activation of NLRP3 and the production of IL-1b
In IR, treatment with LR12 was able to reduce the
in C57BL/6J mice with acute lung injury [58]. These
expression of TNF-a in C57BL/6 mice, but failed to
results suggest that the interaction between NLRs and
inhibit disease progression (Table 1) [6], suggesting
TREM-1 is important for the amplification and pro-
that mTREM-1 may not play a critical role in the
duction of inflammatory mediators [56]. However, the
clinical development of IR.
mechanisms of this cooperation and the nature of the
In AMI, LR12 reduced IL-6 and TNF-a expression
ligands with reciprocal ability to activate both
and improved left ventricular function and survival in
mTREM-1 and NLRs are still unclear.
C57BL/6 mice [63]. LR12 also reduced heart dysfunc-
tion and led to a reduction in infarcted area in pigs
Modulation of TREM-1 by peptides with AMI [64]. Atherosclerosis is a cause of blood
vessel obstruction that leads to AMI, and treatment
LR12
with LR12 attenuated the production of TNF-a, IL-12
The peptide LR12 (LQEEDAGEYGCM) is based in a and IFN-c and development of atherosclerosis in
conserved sequence of mTREM-1 and TLT-1 [54]. C57BL/6 ApoE-/- mice [65].
LR12 was initially described as LR17, and was shown In an experimental model of ALI in C57BL/6 mice,
to inhibit mTREM-1 in Balb/c and C57BL/6 mice LR12 increased production of IL-10 and reduced pro-
with sepsis, reducing the production of IL-6, IL-8, duction of IL-1b, TNFa and IL-6, besides of diminish-
TNF-a and IL-10 and disease-associated complica- ing histopathological changes, leukocytes infiltration,
tions, leading to improved survival of mice [54,59] oxidative stress and disease worsening, mainly due to
(Table 1). Subsequently, 5 amino acids were removed a reduction in NF-jB and NLRP3 activity [58,66].
from LR17, resulting in the 12 amino-acids peptide LR12 also ameliorated colitis, leading to improve-
(LR12) [60], that is apparently the sequence respon- ment in histopathological features and clinical status
sible for the anti-inflammatory effects [60]. Thus, in C57BL/6 mice due to attenuation of mTOR activ-
LR12 was also used in treatment of septic shock in ity [67].
minipigs, where it attenuated sepsis-related disorders These results suggest LR12 as a prominent mol-
such as hypotension, multiple organ failure, cardiovas- ecule in the treatment of inflammatory diseases in
cular complications and production of TNFa and IL-6 which TREM-1 plays a pivotal role. Interestingly,
[60] (Table 1). LR12 mitigated sepsis in monkeys pharmacokinetics and different formulations of LR12
(Macaca fascicularis) with diminution of leukopenia have been tested in vivo [102]. LR12 was administered
and production of pro-inflammatory cytokines such as in rats by in situ implants of poly-lactide-co-glycolide
6 P. H. DOSSANTOSDANTAS ET AL.

and poly-lactide (that provided sustained release of In lungs, LP17 inhibited the deleterious effects
the peptide) for seven days, and presented good bio- caused by Staphylococcus aureus and Pseudomonas
availability [102]. This peptide is now commercially aeruginosa infection in Wistar rats, leading to
known as NangibotideV R , and was tested through con- increased survival [73,74]. The beneficial effect of
tinuous intravenous administration in humans, show- LP17 in lungs were mainly attributed to a reduction
ing to be safe and well tolerated even at high doses (6 in the production of pro-inflammatory cytokines
milligrams/kg, uninterruptedly during 7 h and (IL-1b, TNFa and IL-6) that prevented tissue damaged
45 minutes) [103]. Furthermore, patients treated with and improved disease outcome [73,74] (Table 1).
NangibotideV R displayed few adverse events, and did Interestingly, LP17 has no antimicrobial role, thus
not develop any side effects 28 days after its adminis- these beneficial effects are most likely due to the
tration [103]. Hence, LR12 may be safely used in downregulation of TREM-1-related inflamma-
humans due to good pharmacokinetic parameters, tol- tion [72,73].
erability and stability. Pharmacological inhibition of mTREM-1 by LP17
has been also explored in experimental models of dis-
eases affecting the brain, kidneys, intestine, blood ves-
LP17
sels and lungs [5]. In brain, LP17 was used to treat
The peptide LP17 (LQVTDSGLYRCVIYHPP) is com- neuroinflammation using in vitro and in vivo
posed of highly conserved regions of the extracellular approaches, especially in disturbances in which
portion of murine and human mTREM-1 [12]. mTREM-1 has been intrinsically related to, such as
Initially, LP17 was explored in septic mice, in which it Parkinsons disease (PD), ischemic stroke and sub-
reduced TNFa, IL-1b and IL-6, and increased arachnoid hemorrhage [75–77]. In PD, LP17 reduced
sTREM-1, leading to a reduction in complications and TREM-1 expression and activity of nitric oxide syn-
lethality in Balb/c and C3H/HeN mice [12,68]. These thase (iNOS), cyclooxygenase-2 and NF-jB, all related
effects were also observed in rats with sepsis, where to inflammatory responses in microglia [75]. In zebra-
LP17 also inhibited the increase of sTREM-1 [69]. fish and rats, treatment using LP17 increased the
Interestingly, LP17 diminished the secretion of IL-8, expression of proteins favoring autophagy, thus
TNF-a and IL-6 in leukocytes stimulated with LPS, decreasing the locomotor deficiency observed in PD
obtained from the umbilical cord of newborns [70], caused by chemical compounds [75].
suggesting a putative protective role of LP17 in neo- In situations of neurological bleeding conditions,
natal sepsis [70]. The effects of LP17 in sepsis can be such as subarachnoid hemorrhage and ischemic shock,
at least partly attributed to the inhibition of NF-jB a strong participation of mTREM-1 in the develop-
[12]. The mechanisms concerning LP17 activity on ment of inflammation in rats and C57BL/6J mice has
mTREM-1 still need further clarification, however, been reported [76,77]. In rats, the inhibition of this
two hypotheses have been postulated in this regard. receptor by LP17 was associated with attenuation of
The most accepted theory is that LP17, as LR12, acts early brain injury after subarachnoid hemorrhage by
as a decoy receptor, competing with mTREM-1 for its inhibiting the activity of p38 mitogen protein kinase
natural ligands [68,83,104,105] (Figure 1 and Table 1). and MMP-9, which are critical for disease develop-
Also, similarly to LR12, it is hypothesized that LP17 ment [76]. In C57BL/6J mice with ischemic shock,
may block mTREM-1 dimerization [15,104]. treatment with LP17 ameliorated neuronal infarction
LP17 has been reported attenuating mTREM-1- and injury and promoted cell proliferation, synaptic
mediated inflammation caused by microorganisms in plasticity in hippocampus, inhibition of M1-microglia
periodontal disease, periodontitis, pneumonia and pul- polarization, decreased neutrophil recruitment and
monary empyema [71–74]. In periodontal disease, expression of pro-inflammatory mediators such as IL-
LP17 inhibited the secretion of TNF-a and IL-6 1b and IL-6 [77]. The beneficial activity attributed to
stimulated by P. gingivalis in the myelomonocytic cell LP17 was associated to a reduced activation of
line MonoMac-6 [71], showing that LP17 is an inter- CARD9/NF-jB and NLRP3/caspase 1 that induced
esting approach to treat periodontal diseases. Further, the expression of spleen tyrosine kinase (SYK), which
LP17 attenuated in C57BL/6 mice, periodontitis- is a promoter of neuroinflammation in post-ischemic
related complications and decreased the expression of shock [77]. Based on these data, LP17 is a promising
IL-17, that is associated with worsened periodon- candidate to treat brain disorders in which mTREM-1
titis [72]. plays a central role, and such modulation may favor
INTERNATIONAL REVIEWS OF IMMUNOLOGY 7

the reduction in inflammation and a better outcome cardiovascular collapse, lactic acidosis, lung permeabil-
(Table 1). ity and organ dysfunction, thus improving survival of
In kidneys, mTREM-1 upregulation has been asso- Wistar rats [83]. These results suggest LP17 as a puta-
ciated with IR and nephritis in C57BL/6J mice [6,78]. tive candidate to treat blood vessel-related diseases
Treatment with LP17 in C57BL/6J mice with nephritis and their complications. However, the lack of transla-
induced a reduction in proteinuria, inflammatory cell tional studies exploring the role of LP17 in humans in
infiltrates, thus leading to a better clinical outcome such conditions, make difficult any further observa-
[78]. Despite the effects toward the inhibition of tion (Table 1).
CXCL1 and Myd88 in IR, LP17 was not able to pre- Because of the role of mTREM-1 in lung diseases
vent the development of nephritis [6]. Thus, due to such as pneumonia and acute respiratory distress syn-
conflicting results and the reduced number of studies drome (ARDS) [73,84], the role of LP17 in these scen-
exploring the role of LP17 on kidney diseases, it is arios have also been explored [73,84]. In Wistar rats
difficult to address the potential of this peptide in this with pneumonia, LP17 was effective in attenuating
scenario. However, the beneficial role of LP17 cannot disease-associated complications and reducing inflam-
be underestimated in renal conditions (Table 1). mation by decreasing the production of TNF-a, IL-6
The mTREM-1 has been implicated in the develop- and IL-1b, thus leading to increased survival in rats
ment of diseases affecting the intestine [106]. In this [73]. In ARDS, LP17 reduced TREM-1 expression,
context, LP17 was used to inhibit mTREM-1 in colitis production of TNF-a and IL-1b and lung injury in
and colitis-associated tumorigenesis in C57BL/6 mice C57BL/6 mice [84] (Table 1). Based on this, a formu-
[79]. Treatment with the peptide reduced mortality, lation of LP17 in nanomicelles was tested in ARDS,
tumor size and production of TNF-a, IL-6 and IL-1b and showed higher efficacy in decreasing inflamma-
[79]. Further, LP17 attenuated the proliferation of epi- tion and tissue injury [84] (Table 1). These effects of
thelial cells [79], thus suggesting that LP17 can be LP17 on inflammation in ARDS were partially attrib-
used to treat intestinal inflammation and to attenuate uted to a downregulation of miR-155. This microRNA
inflammation-related tumorigenesis in gut. positively influences mTREM-1 activity mainly
The beneficial role of LP17 was also investigated in through the blockade of inhibitory molecules such as
mesenteric ischemia-reperfusion. In this context, LP17 SOCS-1, thus favoring the expression of pro-inflam-
attenuated inflammation and disease severity mainly matory cytokines (TNF-a and IL-1b) [84,107].
by reducing TNF-a, IL-6, and NF-jB activity, thus Therefore, the role of LP17 in this context is appar-
resulting in diminished mortality in diseased rats ently indirect, but further studies may elucidate these
[105] (Table 1). effects [84,107].
Pancreatitis is another inflammatory condition in Altogether, these findings suggest the potential of
which the role of LP17 has been addressed [80,81]. LP17 to treat a myriad of diseases that are aggravated
Treatment with the peptide in Wistar rats inhibited by mTREM-1 activity. However, the pharmacokinetic
the expression of TNF-a and IL-1b, and attenuated and pharmacodynamics parameters of this peptide,
tissue damage in this organ [80]. Interestingly, LP17 besides of its toxicity, are aspects that still need fur-
also mitigated the hepatic and renal dysfunction ther clarification.
observed in Wistar rats with acute pancreatitis [81]
(Table 1).
GF9
The mTREM-1 is also involved with inflammation
that affects the blood vessels such as in-stent resten- GF9 is a peptide (GLLSKSLVF) based on mTREM-1
osis, atherosclerosis and hemorrhagic shock [5]. In the transmembrane region, designed to interfere with the
in-stent restenosis, LP17 attenuated disease progres- interaction between the receptor and its adaptor pro-
sion by reducing TREM-1 expression, and through tein, DAP12, in the cell membrane [85] (Figure 1).
the inhibition of cell proliferation and migration in GF9 was shown to inhibit the signaling pathway of
experiments with vascular smooth muscle cells [82]. mTREM-1 through DAP12, with a concomitant
In atherosclerosis, LP17 mitigated foam cell formation reduction of inflammatory mediators in murine mod-
in oxLDL-treated macrophages and drastically inhib- els of lung tumor, sepsis, arthritis and alcoholic liver
ited IL-6 production, leading to an improvement in disease [85–87]. Treatment with GF9 retarded lung
inflammation [31]. In hemorrhagic shock, treatment tumor growth in NU/J mice injected with the tumor
using LP17 reduced the levels of TNF-a and IL-6, and cell line H292 and A549 [85] (Table 1). These effects
inhibited disease-associated complications such as were must probably a consequence of reduced release
8 P. H. DOSSANTOSDANTAS ET AL.

by macrophages of compounds that stimulate the LL-37


growth of neoplastic cells such as the vascular endo-
Cathelicidins are peptides that demonstrated activity
thelial growth factor (VEGF), Platelet-derived growth against fungi, viruses, bacteria and parasites [110,111].
factor (PDGF) and TGF-b, which production is indir- The most important among them is the LL-37
ectly influenced by mTREM-1 signalization [108,109]. (LLGDFFRKSKEKIGKEFRIVQRIKDFLRNLVPRTES),
The beneficial effects of GF9 were further explored in a cationic molecule capable of modulating inflamma-
C57BL/6 mice with LPS-induced sepsis, leading to tory and immune responses [110,111]. However, the
prolonged survival and reduction in TNF-a, IL-6 and immunomodulatory features attributed to LL-37 are
IL-1b [85] (Table 1). quite controversial and may have either inhibitory or
In collagen-induced arthritis in DBA/1 mice, GF9 stimulatory activity, depending on the context [110].
reduced the levels of IL-1, TNF-a and IL-6, thus low- As an anti-endotoxin molecule, LL-37 inhibits the
ering the arthritis-associated complications [86]. production of inflammatory mediators, such as TNF-
Similarly, GF9 attenuated TREM-1 expression and a, produced after LPS stimulation in monocytes [112].
diminished production of pro-inflammatory cyto- LL-37 inhibited the production of TNF-a and expres-
kines (TNF-a IL-1b) in C57BL/6 mice with alcoholic sion of mTREM-1 in PBMCs stimulated by LPS [46],
liver disease, resulting in decreased steatosis and tis- thus evidencing the influence of LL-37 on mTREM-1
sue damage in liver [87]. Importantly, when GF9 activity. In other study, LL-37 constrained the effects
was delivered by a HDL-mimicking nanoparticle of LPS-induced sepsis in Balb/c mice also through the
system, its therapeutic effects and pharmacokinetic inhibition of mTREM-1 signaling, thus resulting in
parameters were drastically increased, probably due diminished production of IL-6 and IL-1b, alongside a
to macrophage recognition of the peptide-nanopar- decreased expression of mTREM-1 in monocytes and
ticle molecule via scavenger receptor, thus favoring sTREM-1 in plasma and peritoneal fluids [89].
the endocytosis and consequently the action of GF9 LL-37 also showed antibacterial proprieties,
[85–87]. Therefore, GF9 seems to be a promising decreasing the number of bacteria in plasma and peri-
candidate to treat complications in lung tumors, toneal fluid, besides of improving survival in septic
sepsis, arthritis and alcoholic liver disease (Table 1). Balb/c mice [89] (Table 1 and Figure 1). The peptide
Besides, these results reinforce the beneficial role of has a high potential to be used alone or in combin-
nanosystems as key carriers of TREM-1-inhibi- ation to treat sepsis, acting in the blockade of
tory peptides. mTREM-1 activity and expression, which has been
associated to clinical worsening in septic subjects
[46,89]. However, before the onset of clinical studies,
SLC-TREM-1 several aspects still need further clarification, includ-
The sneaking ligand construct (SLC-TREM-1) is com- ing those concerning its interaction and modulatory
activity toward mTREM-1, its toxicity, and the mecha-
posed by 3 portions: E-selectin, exotoxin A of
nisms regarding its interaction with other drugs fre-
Pseudomonas aeruginosa and 7 amino-acids
quently used to treat sepsis.
(LSKSLVF) of the transmembrane region of mTREM-
1 [88]. The E-selectin and exotoxin A portions act
facilitating the entrance of the construct into the cyto- Vasoactive intestinal peptide
sol of targeted endothelial cells, and the TREM-1- The vasoactive intestinal peptide (VIP) is a 28 amino
derived peptide interacts with DAP12, similarly to acids pleiotropic neuropeptide
GF9, interrupting the interaction between mTREM-1 (HSDAVFTDNYTRLRLQMAVKKYLNSILN), mainly
and DAP12 thus, inhibiting the downstream signaliza- produced in brain, peripheral nervous system,
tion [88] (Figure 1). Initially, SLC-TREM-1 was tested immune and endocrine cells, that possess modulatory
in endothelial cells, where it inhibited TREM-1 effects in macrophages, lymphocytes, mast cells and
expression and production of IL-8 [88]. Then, the neutrophils [113]. In innate immunity cells, VIP was
construct was administered to C57BL/6 mice with shown to have inhibitory activities impairing the pro-
sepsis, leading to reduced neutrophil transmigration duction of pro-inflammatory mediators and stimulat-
and increased mice survival [88] (Table 1). Despite ing the production of IL-10 in LPS-stimulated
the limited availability of data regarding the role of macrophages [113]. Further, VIP reduced the recruit-
SLC-TREM-1 to treat inflammation, it may become a ment of phagocytes and the upregulation of TLRs,
promising drug to treat inflammatory disorders. thus preventing macrophages and neutrophils from
INTERNATIONAL REVIEWS OF IMMUNOLOGY 9

being stimulated by exogenous and endogenous another study, treatment with methylprednisolone in
ligands [113]. Thus, because of its anti-inflammatory acute respiratory distress syndrome, reduced the levels
effects [90], it is reasonable to assume that VIP may of sTREM-1 in the bronchoalveolar lavage and
have inhibitory activity over mTREM-1. Indeed, VIP improved lung injury score when treated and
inhibited the expression of TREM-1 in lung cells and untreated patients were compared [92]. The activity of
increased the expression of the anti-inflammatory iso- corticosteroids on TREM-1 expression may be
form TREM-2, in kunming mice with LPS-induced explained by the inhibitory activity over NF-jB, which
acute lung injury (ALI), on the other hand, in ALI- is one of the key molecules in the transcription of
non treated mice, the expression of TREM-1 was TREM-1 [115] (Figure 1), however, further studies are
increased [90] (Table 1). These effects seems to be needed to better elucidate this interaction.
related to the downregulation, mediated by VIP recep-
tor, of the transcription factors AP-1 and NF-jB,
Antibiotics
which are directly involved in TREM-1 expression
[90,113] (Figure 1). Taken together, these findings The relationship between antibiotics and inflammation
suggest that VIP may become a new therapeutic is widely explored [116,117]. To better illustrate this,
approach in the modulation of TREM-1 in the future. it is possible to highlight the role of macrolide antibi-
otics in inhibiting oxidative burst, transmigration of
neutrophils and production of inflammatory cyto-
Classic drugs with activity over TREM-1 kines, such as IL-6 and TNF-a [116,117].
As mentioned beforehand, TREM-1 has a central role Macrolides are lactones widely used to treat bacter-
in the onset and amplification of inflammation and, ial infections, targeting the bacterial 50S ribosomal
for this reason, this receptor has been implicated in subunit, thus preventing protein synthesis and leading
the pathology and worsening of different diseases to the death of the pathogen or preventing their
[5,35]. Therefore, it is reasonable to assume that multiplication [116,117]. Moreover, these drugs have
pharmacological treatments with known activity anti-inflammatory properties as mentioned beforehand
toward other players in inflammation and infection, [116,117]. Therefore, the role of some macrolides such
such as antibiotics [93], corticosteroids [92] and sta- as azithromycin, rapamycin and tacrolimus, was
tins [99], may also interfere with mTREM-1 explored in the modulation of TREM-1. Azithromycin
functionality. inhibited the activity and expression of mTREM-1,
besides reducing the levels of TNF-a, IL-6, sTREM-1
and IL-1b, thus resulting in a higher survival of septic
Corticosteroids
Balb/c mice [93]. In fungal infections, rapamycin and
Corticosteroids are suppressors of the immune tacrolimus also inhibited mTREM-1 activity [95,96].
response, used in the treatment of allergies, auto- For invasive pulmonary aspergillosis (IPA), treatment
immune and uncontrolled inflammatory diseases with rapamycin inhibited the release of sTREM-1, IL-
[114]. Unfortunately, prolonged use and high doses of 6 and IFN-c, and induced the production of IL-10
this class of drug can promote osteoporosis, skin atro- [95] (Table 1). The mechanism behind the modulation
phy, diabetes, abdominal obesity, hypertension among of rapamycin on sTREM-1 activity is mainly attrib-
others disturbances [114]. Despite these undesirable uted to the inhibition of the mTOR pathway, which is
effects, corticosteroids are widely used and have their a known inducer of mTREM-1 expression [95]
role in TREM-mediated diseases already (Figure 1). Another macrolide with activity on
explored [91,92]. mTREM-1 in fungal infections is tacrolimus [96]. This
Treatment with dexamethasone decreased the antimicrobial drug reduced the transcription and
expression of TREM-1, the release of sTREM-1 and expression of mTREM-1, TNF-a and IL-1b in fungal
the production of TNF-a in neutrophils and mono- keratitis induced by Aspergillus fumigatus, leading to
cytes stimulated by Pseudomonas aeruginosa [91] reduced corneal damage in C57BL/6 mice [96] (Table
(Table 1). The effects of this drug on the transcription 1 and Figure 1). Treatment with clarithromycin in
of TREM-1 seems to be dependent on TNF-a activity, 200 septic patients, increased the expression of
as in C57BL/6 TNF-/- mice, dexamethasone had no TREM-1 and release of sTREM-1 [94] (Table 1 and
effect on the transcription of TREM-1 [91] (Table 1). Figure 1). Furthermore, clarithromycin decreased the
However, the mechanism that dictates the influence of IL-10/TNF-a ratio in serum, thus suggesting that
TNF-a on TREM-1 functionality is unclear. In treatment with this antibiotic reestablished the
10 P. H. DOSSANTOSDANTAS ET AL.

cytokine balance during sepsis, constraining its devel- study, the modulation of mTREM-1 leads to the
opment and the risk of multiple organ dysfunc- inhibition of key pro-inflammatory mediators and
tions [94]. induction of anti-inflammatory molecules such as IL-
Other antibiotics like doxycycline and iminipem 10, which alone or in combination, can be explored to
also have modulatory activity over mTREM-1 [97,98]. treat inflammatory conditions. The mTREM-1 seems
In vitro experiments with monocytes infected by P. to be influenced by and to influence the functionality
gingivalis and treated with doxycycline, showed a of other PRRs in different ways, thus suggesting that
reduction in expression of mTREM-1, besides dimin- therapies aiming at modulating such receptors may
ishing levels of IL-8 and sTREM-1 [97]. Combined represent a promising strategy to treat infectious and
treatment of imipenem with cornuside (a secoiridoid noninfectious conditions in which these receptors play
glucoside derived from Cornus officinalis) in Sprague- a central role. Canonical transcription factors and
Dawley rats with sepsis, reduced the levels of sTREM- pathways generally associated to the onset of inflam-
1, TNF-a and IL-6, besides increasing the production matory response are also related to the activity of
of IL-10 [98]. Although promising in microbial infec- mTREM-1, which broads the possibility of drugs able
tions, the interactions between antimicrobial drugs to constrain its functionality. Despite these encourag-
and TREM-1 activity still needs further clarification, ing results, the majority of studies using mTREM-1 as
especially regarding toxicity, drug interaction and a therapeutic target were performed in experimental
metabolism (Table 1 and Figure 1). models using rodents, which compromise the extrapo-
lation to humans bearing similar conditions. Finally,
there is a lack of data concerning toxicity, pharmaco-
Statins
kinetics and interaction between regularly used drugs
Statins are a class of drugs that inhibits the enzyme 3- and other modulators of mTREM-1 activity, which
hydroxy-3-methylglutaryl-coenzime A (HMG-CoA) reinforces the necessity for more research in the field.
reductase involved in the synthesis of cholesterol
[118]. These drugs have pleiotropic effects including
Disclosure of interest
anti-inflammatory and immunomodulatory properties
based on their inhibition on the production of inflam- The authors report no conflict of interest.
matory mediators such as IL-6, IL-8, TNF-a, IL-1b
and IFN-c [118]. Interestingly, statins were reported
Funding
to be capable of inhibiting the activity and expression
of TREM-1 in myeloid cells [99,100,119]. The authors would like to thank Coordenaç~ao de
Aperfeiçoamento de Pessoal de Nıvel Superior (CAPES) for
Pravastatin inhibited the expression of TREM-1
A.O.M and P.H.S.D scholarships.
and the release of sTREM-1 in PBMCs stimulated
with LPS [100]. Furthermore, it reduced the produc-
tion of IL-6 and TNF-a in culture supernatant [100] ORCID
(Table 1). These effects seemed to be related to a Pedro Henrique dos Santos Dantas http://orcid.org/
blockade in NF-jB activity [100] (Figure 1), which 0000-0001-9413-0954
may explain the reduced expression of TREM-1. In a Amanda de Oliveira Matos http://orcid.org/0000-0001-
different study, pravastatin ameliorated atherosclerosis 5676-5959
Ernandes da Silva Filho http://orcid.org/0000-0002-
in C57BL/6 ApoE-/- mice, reducing the numbers of
2926-5120
lipid deposits and inflammatory cells in atherosclerotic Marcelle Silva-Sales http://orcid.org/0000-0002-
plaques [99]. Further, treatment with pravastatin also 4275-9627
decreased the expression of TREM-1, DAP12, TNF-a Helioswilton Sales-Campos http://orcid.org/0000-0003-
and IL-1 [99] (Table 1). Altogether, these results 3252-2834
suggest that the activity of TREM-1 is indirectly
constrained by statins mainly by the inhibition in References
NF- jB activity.
1. Janeway CA, Jr., Medzhitov R. Innate immune rec-
ognition. Annu Rev Immunol. 2002;20(1):197–216.
Conclusions and future perspectives doi:10.1146/annurev.immunol.20.083001.084359.
2. Gulati A, Kaur D, Krishna Prasad GVR,
In general, regardless the source or the biochemical Mukhopadhaya A. PRR Function of Innate Immune
nature of the compounds described in the present Receptors in Recognition of Bacteria or Bacterial
INTERNATIONAL REVIEWS OF IMMUNOLOGY 11

Ligands. Adv Exp Med Biol. 2018;1112:255–280. doi: Receptor Expressed on Myeloid Cells-1 Signaling.
10.1007/978-981-13-3065-0_18. Front Immunol. 2017;8:917doi:10.3389/fimmu.2017.
3. Allcock RJ, Barrow AD, Forbes S, Beck S, Trowsdale 00917.
J. The human TREM gene cluster at 6p21.1 encodes 17. Haselmayer P, Grosse-Hovest L, von Landenberg P,
both activating and inhibitory single IgV domain Schild H, Radsak MP. TREM-1 ligand expression on
receptors and includes NKp44. European J Immunol. platelets enhances neutrophil activation. Blood. 2007;
2003;33(2):567–577. doi:10.1002/immu.200310033. 110(3):1029–1035. doi:10.1182/blood-2007-01-069195.
4. Bouchon A, Dietrich J, Colonna M. Cutting edge: 18. Read CB, Kuijper JL, Hjorth SA, et al. Cutting Edge:
inflammatory responses can be triggered by TREM- identification of neutrophil PGLYRP1 as a ligand for
1, a novel receptor expressed on neutrophils and TREM-1. J Immunol. 2015;194(4):1417–1421. doi:10.
monocytes. J Immunol. 2000;164(10):4991–4995. doi: 4049/ji.mmunol.1402303.
10.4049/jimmunol.164.10.4991. 19. El Mezayen R, El Gazzar M, Seeds MC, McCall CE,
5. Tammaro A, Derive M, Gibot S, Leemans JC, Dreskin SC, Nicolls MR. Endogenous signals released
Florquin S, Dessing MC. TREM-1 and its potential from necrotic cells augment inflammatory responses
ligands in non-infectious diseases: from biology to to bacterial endotoxin. Immunol Lett. 2007;111(1):
clinical perspectives. Pharmacol Ther. 2017;177: 36–44. doi:10.1016/j.imlet.2007.04.011.
81–95. doi:10.1016/j.pharmthera.2017.02.043. 20. Denning NL, Aziz M, Murao A, et al. Extracellular
6. Tammaro A, Kers J, Emal D, et al. Effect of TREM-1 CIRP as an endogenous TREM-1 ligand to fuel
blockade and single nucleotide variants in experi- inflammation in sepsis. JCI Insight. 2020;5(5):1–16.
mental renal injury and kidney transplantation. Sci doi:10.1172/jci.insight.134172.
Rep. 2016;6(1):38275. doi:10.1038/srep38275. 21. Cheng PC, Lin CN, Chen YJ, Chang FS, Tsaihong
7. Arts RJ, Joosten LA, Dinarello CA, Kullberg BJ, van JC, Lee KM. Triggering receptor expressed on mye-
der Meer JW, Netea MG. TREM-1 interaction with loid cells (TREM)-1 participates in Schistosoma
the LPS/TLR4 receptor complex. European Cytokine mansoni inflammatory responses. Parasite Immunol.
Netw. 2011;22(1):11–14. doi:10.1684/ecn.2011.0274. 2011;33(5):276–286. doi:10.1111/j.1365-3024.2011.
8. Thankam FG, Dilisio MF, Dougherty KA, Dietz NE, 01284.x.
Agrawal DK. Triggering receptor expressed on mye- 22. Denner J, Eschricht M, Lauck M, et al. Modulation
loid cells and 5’adenosine monophosphate-activated of cytokine release and gene expression by the
protein kinase in the inflammatory response: a immunosuppressive domain of gp41 of HIV-1. PLoS
potential therapeutic target. Expert Rev Clin One. 2013;8(1):e55199. doi:10.1371/journal.pone.
Immunol. 2016;12(11):1239–1249. doi:10.1080/ 0055199.
1744666X.2016.1196138. 23. Hyun J, McMahon RS, Lang AL, et al. HIV and
9. Weber B, Schuster S, Zysset D, et al. TREM-1 defi- HCV augments inflammatory responses through
ciency can attenuate disease severity without affect- increased TREM-1 expression and signaling in
ing pathogen clearance. PLoS Pathog. 2014;10(1): Kupffer and Myeloid cells. PLoS Pathog. 2019;15(7):
e1003900. doi:10.1371/journal.ppat.1003900. e1007883. doi:10.1371/journal.ppat.1007883.
10. Kelker MS, Foss TR, Peti W, et al. Crystal structure 24. Mohamadzadeh M, Coberley SS, Olinger GG, et al.
of human triggering receptor expressed on myeloid Activation of triggering receptor expressed on mye-
cells 1 (TREM-1) at 1.47 A. J Mol Biol. 2004;342(4): loid cells-1 on human neutrophils by marburg and
1237–1248. doi:10.1016/j.jmb.2004.07.089. ebola viruses. JVI. 2006;80(14):7235–7244. doi:10.
11. Radaev S, Kattah M, Rostro B, Colonna M, Sun PD. 1128/JVI.00543-06.
Crystal structure of the human myeloid cell activat- 25. Nochi H, Aoki N, Oikawa K, et al. Modulation of
ing receptor TREM-1. Structure. 2003;11(12): hepatic granulomatous responses by transgene
1527–1535. doi:10.1016/j.str.2003.11.001. expression of DAP12 or TREM-1-Ig molecules. Am J
12. Gibot S, Cravoisy A, Levy B, Bene MC, Faure G, Pathol. 2003;162(4):1191–1201. doi:10.1016/S0002-
Bollaert PE. Soluble triggering receptor expressed on 9440(10)63915-6.
myeloid cells and the diagnosis of pneumonia. N 26. Gibot S, Massin F. Soluble form of the triggering
Engl J Med. 2004a;350(5):451–458. doi:10.1056/ receptor expressed on myeloid cells 1: an anti-
NEJMoa031544. inflammatory mediator?. Intensive Care Med. 2006;
13. Molloy EJ. Triggering Receptor Expressed on 32(2):185–187. doi:10.1007/s00134-005-0018-0.
Myeloid Cells (TREM) family and the application of 27. Gomez-Pina V, et al. Metalloproteinases shed
its antagonists. PRI. 2009;4(1):51–56. doi:10.2174/ TREM-1 ectodomain from lipopolysaccharide-stimu-
157489109787236292. lated human monocytes. J Immunol. 2007;179:
14. Tessarz AS, Cerwenka A. The TREM-1/DAP12 path- 4065–4073. doi:10.4049/jimmunol.179.6.4065.
way. Immunol Lett. 2008;116(2):111–116. doi:10. 28. Gingras MC, Lapillonne H, Margolin JF. TREM-1,
1016/j.imlet.2007.11.021. MDL-1, and DAP12 expression is associated with a
15. Carrasco K, Boufenzer A, Jolly L, et al. TREM-1 mature stage of myeloid development. Mol Immunol.
multimerization is essential for its activation on 2002;38(11):817–824. doi:10.1016/S0161-5890(02)00004-4.
monocytes and neutrophils. Cell Mol Immunol. 2019; 29. Bosco MC, Pierobon D, Blengio F, et al. Hypoxia
16(5):460–472. doi:10.1038/s41423-018-0003-5. modulates the gene expression profile of immunore-
16. Fu L, Han L, Xie C, et al. Identification of gulatory receptors in human mature dendritic cells:
Extracellular Actin As a Ligand for Triggering identification of TREM-1 as a novel hypoxic marker
12 P. H. DOSSANTOSDANTAS ET AL.

in vitro and in vivo. Blood. 2011;117(9):2625–2639. Biol Chem. 1998;273(49):32934–32942. doi:10.1074/


doi:10.1182/blood-2010-06-292136. jbc.273.49.32934.
30. McMahon L, Schwartz K, Yilmaz O, Brown E, Ryan 42. Radsak MP, Salih HR, Rammensee HG, Schild H.
LK, Diamond G. Vitamin D-mediated induction of Triggering receptor expressed on myeloid cells-1 in
innate immunity in gingival epithelial cells. Infect neutrophil inflammatory responses: differential regu-
Immun. 2011;79(6):2250–2256. doi:10.1128/IAI. lation of activation and survival. J Immunol. 2004;
00099-11. 172(8):4956–4963. doi:10.4049/jimmunol.172.8.4956.
31. Li H, Hong F, Pan S, Lei L, Yan F. Silencing trigger- 43. del Fresno C, G omez-Pi~na V, Lores V, et al.
ing receptors expressed on myeloid cells-1 impaired Monocytes from cystic fibrosis patients are locked in
the inflammatory response to oxidized low-density an LPS tolerance state: down-regulation of TREM-1
lipoprotein in macrophages. Inflammation. 2016; as putative underlying mechanism. PLoS One. 2008;
39(1):199–208. doi:10.1007/s10753-015-0239-5. 3(7):e2667. doi:10.1371/journal.pone.0002667.
32. Bleharski JR, Kiessler V, Buonsanti C, et al. A role 44. Turkistany SA, DeKoter RP. The transcription factor
for triggering receptor expressed on myeloid cells-1 PU.1 is a critical regulator of cellular communication
in host defense during the early-induced and adap- in the immune system. Arch Immunol Ther Exp.
tive phases of the immune response. J Immunol. 2011;59(6):431–440. doi:10.1007/s00005-011-0147-9.
2003;170(7):3812–3818. doi:10.4049/jimmunol.170.7. 45. Schenk M, Bouchon A, Birrer S, Colonna M,
3812. Mueller C. Macrophages expressing triggering recep-
33. Chen LC, Laskin JD, Gordon MK, Laskin DL. tor expressed on myeloid cells-1 are underrepre-
Regulation of TREM expression in hepatic macro- sented in the human intestine. J Immunol. 2005;
phages and endothelial cells during acute endotoxe- 174(1):517–524. doi:10.4049/jimmunol.174.1.517.
mia. Exp Mol Pathol. 2008;84(2):145–155. doi:10. 46. Amatngalim GD, Nijnik A, Hiemstra PS, Hancock
1016/j.yexmp.2007.11.004. RE. Cathelicidin peptide LL-37 modulates TREM-1
34. Murakami Y, Kohsaka H, Kitasato H, Akahoshi T. expression and inflammatory responses to microbial
Lipopolysaccharide-induced up-regulation of trigger- compounds. Inflammation. 2011;34(5):412–425. doi:
ing receptor expressed on myeloid cells-1 expression
10.1007/s10753-010-9248-6.
on macrophages is regulated by endogenous prosta- 47. Molad Y, Pokroy-Shapira E, Carmon V. CpG-oligo-
glandin E2. J Immunol. 2007;178(2):1144–1150. doi:
deoxynucleotide-induced TLR9 activation regulates
10.4049/jimmunol.178.2.1144.
macrophage TREM-1 expression and shedding.
35. Gao S, Yi Y, Xia G, et al. The characteristics and
Innate Immun. 2013;19(6):623–630. doi:10.1177/
pivotal roles of triggering receptor expressed on
1753425913476970.
myeloid cells-1 in autoimmune diseases. Autoimmun
48. Gibot S. Clinical review: role of triggering receptor
Rev. 2019;18(1):25–35. doi:10.1016/j.autrev.2018.07.
expressed on myeloid cells-1 during sepsis. Crit
008.
Care. 2005;9(5):485–489.
36. Bouchon A, Facchetti F, Weigand MA, Colonna M.
49. Knapp S, Gibot S, de Vos A, Versteeg HH, Colonna
TREM-1 amplifies inflammation and is a crucial
M, van der Poll T. Cutting edge: expression patterns
mediator of septic shock. Nature. 2001;410(6832):
1103–1107. doi:10.1038/35074114. of surface and soluble triggering receptor expressed
37. Hosoda H, Tamura H, Kida S, Nagaoka I. on myeloid cells-1 in human endotoxemia. J
Transcriptional regulation of mouse TREM-1 gene Immunol. 2004;173(12):7131–7134. doi:10.4049/jim-
in RAW264.7 macrophage-like cells. Life Sci. 2011; munol.173.12.7131.
89(3-4):115–122. doi:10.1016/j.lfs.2011.05.007. 50. Ornatowska M, Azim AC, Wang X, et al. Functional
38. Zeng H, Ornatowska M, Joo MS, Sadikot RT. genomics of silencing TREM-1 on TLR4 signaling in
TREM-1 expression in macrophages is regulated at macrophages. Am J Physiol Lung Cell Mol Physiol.
transcriptional level by NF-kappaB and PU.1. Eur J 2007;293(6):L1377–1384. doi:10.1152/ajplung.00140.
Immunol. 2007;37(8):2300–2308. doi:10.1002/eji. 2007.
200737270. 51. Zheng H, Heiderscheidt CA, Joo M, et al. MYD88-
39. Tessarz AS, Weiler S, Zanzinger K, Angelisova P, dependent and -independent activation of TREM-1
Horejsı V, Cerwenka A. Non-T cell activation linker via specific TLR ligands. Eur J Immunol. 2010;40(1):
(NTAL) negatively regulates TREM-1/DAP12- 162–171. doi:10.1002/eji.200839156.
induced inflammatory cytokine production in mye- 52. Lagler H, Sharif O, Haslinger I, et al. TREM-1 acti-
loid cells. J Immunol. 2007;178(4):1991–1999. doi:10. vation alters the dynamics of pulmonary IRAK-M
4049/jimmunol.178.4.1991. expression in vivo and improves host defense during
40. Zarbock A, Abram CL, Hundt M, Altman A, Lowell pneumococcal pneumonia. J Immunol. 2009;183(3):
CA, Ley K. PSGL-1 engagement by E-selectin signals 2027–2036. doi:10.4049/jimmunol.0803862.
through Src kinase Fgr and ITAM adapters DAP12 53. Wu M, Peng A, Sun M, et al. TREM-1 amplifies cor-
and FcR gamma to induce slow leukocyte rolling. J neal inflammation after Pseudomonas aeruginosa
Exp Med. 2008;205(10):2339–2347. doi:10.1084/jem. infection by modulating Toll-like receptor signaling
20072660. and Th1/Th2-type immune responses. Infect Immun.
41. McVicar DW, Taylor LS, Gosselin P, et al. DAP12- 2011;79(7):2709–2716. doi:10.1128/IAI.00144-11.
mediated signal transduction in natural killer cells. A 54. Derive M, Bouazza Y, Sennoun N, et al. Soluble
dominant role for the Syk protein-tyrosine kinase. J TREM-like transcript-1 regulates leukocyte activation
INTERNATIONAL REVIEWS OF IMMUNOLOGY 13

and controls microbial sepsis. J Immunol. 2012; 67. Kokten T. TREM-1 Inhibition Restores Impaired
188(11):5585–5592. doi:10.4049/jimmunol.1102674. Autophagy Activity and Reduces Colitis in Mice. J
55. Chen G, Shaw MH, Kim YG, Nunez G. NOD-like Crohns Colitis. 2018;12:230–244. doi:10.1093/ecco-
receptors: role in innate immunity and inflammatory jcc/jjx129.
disease. Annu Rev Pathol Mech Dis. 2009;4(1): 68. Horst SA, Linner A, Beineke A, et al. Prognostic
365–398. doi:10.1146/annurev.pathol.4.110807. value and therapeutic potential of TREM-1 in
092239. Streptococcus pyogenes- induced sepsis. J Innate
56. Netea MG, Azam T, Ferwerda G, Girardin SE, Kim Immun. 2013;5(6):581–590. doi:10.1159/000348283.
SH, Dinarello CA. Triggering receptor expressed on 69. Shi X, Zhang Y, Wang H, Zeng S. Effect of
myeloid cells-1 (TREM-1) amplifies the signals Triggering Receptor Expressed on Myeloid Cells 1
induced by the NACHT-LRR (NLR) pattern recogni- (TREM-1) Blockade in Rats with Cecal Ligation and
tion receptors. J Leukoc Biol. 2006;80(6):1454–1461. Puncture (CLP)-Induced Sepsis. Med Sci Monit.
doi:10.1189/jlb.1205758. 2017;23:5049–5055. doi:10.12659/MSM.904386.
57. Prufer S, et al. Distinct signaling cascades of TREM- 70. Qian L, Weng XW, Chen W, Sun CH, Wu J.
1, TLR and NLR in neutrophils and monocytic cells. TREM-1 as a potential therapeutic target in neonatal
J Innate Immun. 2014;6:339–352. doi:10.1159/ sepsis. Int J Clin Exp Med. 2014;7:1650–1658.
000355892. 71. Bostanci N, Thurnheer T, Belibasakis GN.
58. Liu T, Zhou Y, Li P, et al. Blocking triggering recep- Involvement of the TREM-1/DAP12 pathway in the
tor expressed on myeloid cells-1 attenuates lipopoly- innate immune responses to Porphyromonas gingi-
saccharide-induced acute lung injury via inhibiting valis. Mol Immunol. 2011;49(1-2):387–394. doi:10.
NLRP3 inflammasome activation. Sci Rep. 2016;6(1): 1016/j.molimm.2011.09.012.
39473. doi:10.1038/srep39473. 72. Bostanci N, Abe T, Belibasakis GN, Hajishengallis G.
59. Jolly L, Carrasco K, Derive M, Lemarie J, Boufenzer TREM-1 Is upregulated in experimental periodon-
A, Gibot S. Targeted endothelial gene deletion of titis, and its blockade inhibits IL-17A and RANKL
triggering receptor expressed on myeloid cells-1 pro- expression and suppresses bone loss. J Clin Med.
tects mice during septic shock. Cardiovasc Res. 2018;
2019;8:1–14. doi:10.3390/jcm8101579.
114(6):907–918. doi:10.1093/cvr/cvy018. 73. Gibot S, Alauzet C, Massin F, et al. Modulation of
60. Derive M, Boufenzer A, Bouazza Y, et al. Effects of a
the triggering receptor expressed on myeloid cells-1
TREM-like transcript 1-derived peptide during hypo-
pathway during pneumonia in rats. J Infect Dis.
dynamic septic shock in pigs. Shock. 2013;39(2):
2006;194(7):975–983. doi:10.1086/506950.
176–182. doi:10.1097/SHK.0b013e31827bcdfb.
74. Luo L, Zhou Q, Chen XJ, Qin SM, Ma WL, Shi HZ.
61. Derive M, Boufenzer A, Gibot S. Attenuation of
Effects of the TREM-1 pathway modulation during
responses to endotoxin by the triggering receptor
empyema in rats. Chin. Med. J. 2010;123(12):
expressed on myeloid cells-1 inhibitor LR12 in non-
1561–1565.
human primate. Anesthesiology. 2014;120(4):935–942.
75. Feng C-W, Chen N-F, Sung C-S, et al. Therapeutic
doi:10.1097/ALN.0000000000000078.
62. Dubar M, Carrasco K, Gibot S, Bisson C. Effects of Effect of Modulating TREM-1 via Anti-inflammation
Porphyromonas gingivalis LPS and LR12 peptide on and Autophagy in Parkinson’s Disease. Front
TREM-1 expression by monocytes. J Clin Neurosci. 2019;13:769doi:10.3389/fnins.2019.00769.
Periodontol. 2018;45(7):799–805. doi:10.1111/jcpe. 76. Sun XG, Duan H, Jing G, Wang G, Hou Y, Zhang
12925. M. Inhibition of TREM-1 attenuates early brain
63. Boufenzer A, Lemarie J, Simon T, et al. TREM-1 injury after subarachnoid hemorrhage via downregu-
mediates inflammatory injury and cardiac remodel- lation of p38MAPK/MMP-9 and preservation of ZO-
ing following myocardial infarction. Circ Res. 2015; 1. Neuroscience. 2019;406:369–375. doi:10.1016/j.
116(11):1772–1782. doi:10.1161/CIRCRESAHA.116. neuroscience.2019.03.032.
305628. 77. Xu P, Zhang X, Liu Q, et al. Microglial TREM-1
64. Lemarie J, et al. Pharmacological inhibition of the receptor mediates neuroinflammatory injury via
triggering receptor expressed on myeloid cells-1 lim- interaction with SYK in experimental ischemic
its reperfusion injury in a porcine model of myocar- stroke. Cell Death Dis. 2019;10(8):555. doi:10.1038/
dial infarction. ESC Heart Fail. 2015;2:90–99. doi:10. s41419-019-1777-9.
1002/ehf2.12029. 78. Du C, Peng L, Kou G, Wang P, Lu L, Li Y.
65. Joffre J, Potteaux S, Zeboudj L, et al. Genetic and Assessment of Serum sTREM-1 as a Marker of
Pharmacological Inhibition of TREM-1 Limits the Subclinical Inflammation in Diarrhea-Predominant
Development of Experimental. Atherosclerosis J Am Patients with Irritable Bowel Syndrome. Dig Dis Sci.
Coll Cardiol. 2016;68(25):2776–2793. doi:10.1016/j. 2018;63(5):1182–1191. doi:10.1007/s10620-018-5002-
jacc.2016.10.015. y.
66. Shi R, Zhang J, Peng Z, et al. Expression level of 12- 79. Zhou J, Chai F, Lu G, et al. TREM-1 inhibition
amino acid triggering receptor on myeloid cells-like attenuates inflammation and tumor within the colon.
transcript 1 derived peptide alleviates lipopolysac- Int Immunopharmacol. 2013;17(2):155–161. doi:10.
charide-induced acute lung injury in mice. Int J Mol 1016/j.intimp.2013.06.009.
Med. 2018;41:2159–2168. doi:10.3892/ijmm.2018. 80. Dang S, Shen Y, Yin K, Zhang J. TREM-1 promotes
3443. pancreatitis-associated intestinal barrier dysfunction.
14 P. H. DOSSANTOSDANTAS ET AL.

Gastroenterol Res Pract. 2012;2012:1–8. doi:10.1155/ 93. Tong J, Liu ZC, Wang DX. Azithromycin acts as an
2012/720865. immunomodulatory agent to suppress the expression
81. Kamei K, Yasuda T, Ueda T, Qiang F, Takeyama Y, of TREM-1 in Bacillus pyocyaneus-induced sepsis.
Shiozaki H. Role of triggering receptor expressed on Immunol Lett. 2011;138(2):137–143. doi:10.1016/j.
myeloid cells-1 in experimental severe acute pancrea- imlet.2011.04.001.
titis. J Hepatobiliary Pancreat Sci. 2010;17(3): 94. Spyridaki A, Raftogiannis M, Antonopoulou A, et al.
305–312. doi:10.1007/s00534-009-0191-6. Effect of clarithromycin in inflammatory markers of
82. Wang F, Li C, Ding FH, et al. Increased serum patients with ventilator-associated pneumonia and
TREM-1 level is associated with in-stent restenosis, sepsis caused by Gram-negative bacteria: results
and activation of TREM-1 promotes inflammation, from a randomized clinical study. Antimicrob Agents
proliferation and migration in vascular smooth Chemother. 2012;56(7):3819–3825. doi:10.1128/AAC.
muscle cells. Atherosclerosis. 2017;267:10–18. doi:10. 05798-11.
1016/j.atherosclerosis.2017.10.015. 95. Cui N, Wang H, Su LX, Zhang JH, Long Y, Liu
83. Gibot S, Massin F, Alauzet C, et al. Effects of the DW. Role of triggering receptor expressed on mye-
TREM 1 pathway modulation during hemorrhagic loid cell-1 expression in mammalian target of rapa-
shock in rats. Shock. 2009;32(6):633–637. doi:10. mycin modulation of CD8(þ) T-cell differentiation
1097/SHK.0b013e3181a53842. during the immune response to invasive pulmonary
84. Yuan Z, Syed M, Panchal D, et al. TREM-1-accen- aspergillosis. Chin Med J (Engl). 2017;130:1211–1217.
tuated lung injury via miR-155 is inhibited by LP17 doi:10.4103/0366-6999.205850.
nanomedicine. Am J Physiol Lung Cell Mol Physiol. 96. Huang W, Ling S, Jia X, et al. Tacrolimus (FK506)
2016;310(5):L426–438. doi:10.1152/ajplung.00195. suppresses TREM-1 expression at an early but not at
2015. a late stage in a murine model of fungal keratitis.
85. Sigalov AB. A novel ligand-independent peptide PLoS One. 2014;9(12):e114386. doi:10.1371/journal.
inhibitor of TREM-1 suppresses tumor growth in pone.0114386.
human lung cancer xenografts and prolongs survival 97. Bostanci N, Belibasakis GN. Doxycycline inhibits
of mice with lipopolysaccharide-induced septic
TREM-1 induction by Porphyromonas gingivalis.
shock. Int Immunopharmacol. 2014;21(1):208–219.
FEMS Immunol Med Microbiol. 2012;66(1):37–44.
doi:10.1016/j.intimp.2014.05.001.
doi:10.1111/j.1574-695X.2012.00982.x.
86. Shen ZT, Sigalov AB. Rationally designed ligand-
98. Jiang WL, Chen XG, Zhu HB, Tian JW. Effect of
independent peptide inhibitors of TREM-1 amelior-
cornuside on experimental sepsis. Planta Med. 2009;
ate collagen-induced arthritis. J Cell Mol Med. 2017;
75(06):614–619. doi:10.1055/s-0029-1185383.
21(10):2524–2534. doi:10.1111/jcmm.13173.
99. Wang HM, Gao JH, Lu JL. Pravastatin improves ath-
87. Tornai D, Furi I, Shen ZT, Sigalov AB, Coban S,
erosclerosis in mice with hyperlipidemia by inhibit-
Szabo G. Inhibition of triggering receptor expressed
ing TREM-1/DAP12. Eur Rev Med Pharmacol Sci.
on myeloid cells 1 ameliorates inflammation and
macrophage and neutrophil activation in alcoholic. 2018;22(15):4995–5003. doi:10.26355/eurrev_201808_
Hepatol Commun. 2019;3(1):99–115. doi:10.1002/ 15640.
hep4.1269. 100. Dai M, Chen Y, Mei X. Pravastatin sodium attenu-
88. Gibot S, Jolly L, Lemarie J, Carrasco K, Derive M, ated TREM-1-mediated inflammation in human per-
Boufenzer A. Triggering Receptor Expressed on ipheral blood mononuclear cells. Biochem Biophys
Myeloid Cells-1 Inhibitor Targeted to Endothelium Res Commun. 2019;508(1):225–229. doi:10.1016/j.
Decreases Cell Activation. Front Immunol. 2019;10: bbrc.2018.11.098.
2314doi:10.3389/fimmu.2019.02314. 101. Kumar PS. From focal sepsis to periodontal medi-
89. Hosoda H, Nakamura K, Hu Z, et al. Antimicrobial cine: a century of exploring the role of the oral
cathelicidin peptide LL37 induces NET formation microbiome in systemic disease. J Physiol. 2017;
and suppresses the inflammatory response in a 595(2):465–476. doi:10.1113/JP272427.
mouse septic model. Mol Med Rep. 2017;16(4): 102. Parent M, Clarot I, Gibot S, et al. One-week in vivo
5618–5626. doi:10.3892/mmr.2017.7267. sustained release of a peptide formulated into in situ
90. Sun G-Y, Guan C-X, Zhou Y, et al. Vasoactive intes- forming implants. Int J Pharm. 2017;521(1-2):
tinal peptide re-balances TREM-1/TREM-2 ratio in 357–360. doi:10.1016/j.ijpharm.2017.02.046.
acute lung injury. Regul Pept. 2011;167(1):56–64. doi: 103. Cuvier V, Lorch U, Witte S, et al. A first-in-man
10.1016/j.regpep.2010.11.008. safety and pharmacokinetics study of nangibotide, a
91. Mihailidou I, Pelekanou A, Pistiki A, et al. new modulator of innate immune response through
Dexamethasone Down-Regulates Expression of TREM-1 receptor inhibition. Br J Clin Pharmacol.
Triggering Receptor Expressed on Myeloid Cells-1: 2018;84(10):2270–2279. doi:10.1111/bcp.13668.
Evidence for a TNFa-Related Effect. Front Public 104. Gibot S, Kolopp-Sarda M-N, Bene M-C, et al. A sol-
Health. 2013;1:50doi:10.3389/fpubh.2013.00050. uble form of the triggering receptor expressed on
92. Jeon K, Chung CR, Yang JH, Park CM, Suh GY. myeloid cells-1 modulates the inflammatory response
Predictors of response to corticosteroid treatment in in murine sepsis. J Exp Med. 2004b;200(11):
patients with early acute respiratory distress syn- 1419–1426. doi:10.1084/jem.20040708.
drome: results of a pilot study. Yonsei Med J. 2015; 105. Gibot S, Massin F, Alauzet C, et al. Effects of the
56(1):287–291. doi:10.3349/ymj.2015.56.1.287. TREM-1 pathway modulation during mesenteric
INTERNATIONAL REVIEWS OF IMMUNOLOGY 15

ischemia-reperfusion in rats. Crit Care Med. 2008; 113. Delgado M, Ganea D. Vasoactive intestinal peptide:
36(2):504–510. doi:10.1097/01.CCM.0B013E318161FAF3. a neuropeptide with pleiotropic immune functions.
106. Genua M, Rutella S, Correale C, Danese S. The trig- Amino Acids. 2013;45(1):25–39. doi:10.1007/s00726-
gering receptor expressed on myeloid cells (TREM) 011-1184-8.
in inflammatory bowel disease pathogenesis. J Transl 114. Ramamoorthy S, Cidlowski JA. Corticosteroids:
Med. 2014;12(1):293. doi:10.1186/s12967-014-0293-z. Mechanisms of Action in Health and Disease Rheum
107. Mashima R. Physiological roles of miR-155.
Dis. Clin North Am. 2016;42(1):15–31. vii doi:10.
Immunology. 2015;145(3):323–333. doi:10.1111/imm.
1016/j.rdc.2015.08.002.
12468.
115. Auphan N, DiDonato JA, Rosette C, Helmberg A,
108. Ho C-C, Liao W-Y, Wang C-Y, et al. TREM-1
expression in tumor-associated macrophages and Karin M. Immunosuppression by glucocorticoids:
clinical outcome in lung cancer. Am J Respir Crit inhibition of NF-kappa B activity through induction
Care Med. 2008;177(7):763–770. doi:10.1164/rccm. of I kappa B synthesis. Science. 1995;270(5234):
200704-641OC. 286–290. doi:10.1126/science.270.5234.286.
109. Lin Y, Xu J, Lan H. Tumor-associated macrophages 116. Hoyt JC, Robbins RA. Macrolide antibiotics and pul-
in tumor metastasis: biological roles and clinical monary inflammation. FEMS Microbiol Lett. 2001;
therapeutic applications. J Hematol Oncol. 2019; 205(1):1–7. doi:10.1111/j.1574-6968.2001.tb10917.x.
12(1):76. doi:10.1186/s13045-019-0760-3. 117. Wales D, Woodhead M. The anti-inflammatory
110. Fabisiak A, Murawska N, Fichna J. LL-37: effects of macrolides. Thorax. 1999;54(Supplement
Cathelicidin-related antimicrobial peptide with pleio- 2):S58–S62. doi:10.1136/thx.54.2008.S58.
tropic activity. Pharmacol Rep. 2016;68(4):802–808. 118. Bedi O, Dhawan V, Sharma PL, Kumar P.
doi:10.1016/j.pharep.2016.03.015. Pleiotropic effects of statins: new therapeutic targets
111. Xhindoli D, Pacor S, Benincasa M, Scocchi M, in drug design. Naunyn-Schmiedeberg’s Arch
Gennaro R, Tossi A. The human cathelicidin LL-
Pharmacol. 2016;389(7):695–712. doi:10.1007/s00210-
37–A pore-forming antibacterial peptide and host-
016-1252-4.
cell modulator. Biochim Biophys Acta. 2016;1858(3):
119. Liu M-W, Liu R, Wu H-Y, et al. Atorvastatin has a
546–566. doi:10.1016/j.bbamem.2015.11.003.
112. Mookherjee N, Brown KL, Bowdish DME, et al. protective effect in a mouse model of bronchial
Modulation of the TLR-mediated inflammatory asthma through regulating tissue transglutaminase
response by the endogenous human host defense and triggering receptor expressed on myeloid cells-1
peptide LL-37. J Immunol. 2006;176(4):2455–2464. expression. Exp Ther Med. 2017;14(2):917–930. doi:
doi:10.4049/jimmunol.176.4.2455. 10.3892/etm.2017.4576.

You might also like