ArticlePDF AvailableLiterature Review

SALSA: A Regulator of the early Steps of Complement Activation on Mucosal Surfaces

Authors:

Abstract and Figures

Complement is present mainly in blood. However, following mechanical damage or inflammation, serous exudates enter the mucosal surfaces. Here the complement proteins interact with other endogenous molecules to keep microbes from entering the parenteral tissues. One of the mucosal proteins known to interact with the early complement components of both the classical and the lectin pathway, is the salivary scavenger and agglutinin (SALSA). SALSA is also known as DMBT1 (deleted in malignant brain tumors 1) and gp340. It is found both attached to the epithelium and secreted into the surrounding fluids of most mucosal surfaces. SALSA has been shown to bind directly to C1q, mannose binding lectin (MBL) and the ficolins. Through these interactions SALSA regulates activation of the complement system. In addition, SALSA interacts with surfactant proteins A and D, secretory IgA and lactoferrin. Ulcerative colitis and Crohn’s disease are examples of diseases, where complement activation in mucosal tissues may occur. This review describes the latest advances in our understanding of how the early complement components interact with the SALSA molecule. Furthermore, we discuss how these interactions may affect disease propagation on mucosal surfaces in immunological and inflammatory diseases.
Function and structure of SALSA at the mucosal surfaces. (A) At the mucosal surfaces, the SALSA protein is mainly found associated with the epithelium and secreted into the surrounding fluids. The known features and functions of SALSA are presented in four panels (I–IV). (I) SALSA is present on the epithelial cell surface and deposited in the extracellular matrix, where it is involved in maintaining epithelial homeostasis. (II) Fluid-phase SALSA binds a broad array of microbes. It has been shown to agglutinate viruses, as well as both Gram-positive and Gram-negative bacteria thus preventing them from invading the parenteral spaces. (III) SALSA interacts with other endogenous molecules present at the mucosal surfaces, such as surfactant proteins SpA and SpD as well as IgA. It is believed that these molecules cooperate in antimicrobial defense. (IV) In the case of epithelial damage, cells and molecules from the tissue become mixed with the luminal contents. In this context, SALSA may bind the complement sensor molecules C1q, MBL, and the ficolins, thereby SALSA could initiate complement activation against distinct microbes or participate in the clearance of injured tissue components. (B) In its molecular structure, SALSA contains a stretch of 13 scavenger receptor cysteine-rich (SRCR) domains separated by SRCR interspersed domains. These are followed by two C1r/C1s, urchin embryonic growth factor and bone morphogenetic protein-1 (CUB) domains surrounding the 14th SRCR domain. Finally, a zona pellucida (ZP) domain is found at the most C-terminal end of the protein.
… 
Content may be subject to copyright.
March 2016 | Volume 7 | Article 851
MINI REVIEW
published: 08 March 2016
doi: 10.3389/mmu.2016.00085
Frontiers in Immunology | www.frontiersin.org
Edited by:
Michael C. Carroll,
Boston Children’s Hospital and
Harvard Medical School, USA
Reviewed by:
Masahide Tone,
Cedars-Sinai Medical Center, USA
Matthew Cook,
Canberra Hospital and Australian
National University, Australia
*Correspondence:
Seppo Meri
seppo.meri@helsinki.
Specialty section:
This article was submitted to
Immunological Tolerance,
a section of the journal
Frontiers in Immunology
Received: 02November2015
Accepted: 22February2016
Published: 08March2016
Citation:
ReichhardtMP and MeriS (2016)
SALSA: A Regulator of the Early
Steps of Complement Activation on
Mucosal Surfaces.
Front. Immunol. 7:85.
doi: 10.3389/mmu.2016.00085
SALSA: A Regulator of the Early
Steps of Complement Activation on
Mucosal Surfaces
Martin Parnov Reichhardt and Seppo Meri*
Immunobiology Research Program, Research Programs Unit, Department of Bacteriology and Immunology, Haartman
Institute, University of Helsinki, Helsinki, Finland
Complement is present mainly in blood. However, following mechanical damage or
inammation, serous exudates enter the mucosal surfaces. Here, the complement
proteins interact with other endogenous molecules to keep microbes from entering
the parenteral tissues. One of the mucosal proteins known to interact with the early
complement components of both the classical and the lectin pathway is the salivary
scavenger and agglutinin (SALSA). SALSA is also known as deleted in malignant brain
tumors 1 and gp340. It is found both attached to the epithelium and secreted into the
surrounding uids of most mucosal surfaces. SALSA has been shown to bind directly to
C1q, mannose-binding lectin, and the colins. Through these interactions SALSA regu-
lates activation of the complement system. In addition, SALSA interacts with surfactant
proteins A and D, secretory IgA, and lactoferrin. Ulcerative colitis and Crohn’s disease are
examples of diseases, where complement activation in mucosal tissues may occur. This
review describes the latest advances in our understanding of how the early complement
components interact with the SALSA molecule. Furthermore, we discuss how these
interactions may affect disease propagation on mucosal surfaces in immunological and
inammatory diseases.
Keywords: gp340, DMBT1, Crohn’s disease, colins, MBL, C1q, ulcerative colitis, IBD
INTRODUCTION
Activation of the complement system is strongly involved in generating inammation, combatting
microbial infections, and participating in clearance of non-viable tissue. Although complement is
present mainly in blood, it is also found in serous exudates on mucosal surfaces, such as in the oral
cavity or the airways (1, 2). is is particularly seen aer mechanical, infectious, or immune damage,
e.g., in periodontal disease or SLE (3). When serous exudates enter the mucosal surfaces, innate
immune proteins interact with mucosal surface proteins. Together, these molecules participate in
clearance and defense against invading microorganisms. Although bleeding at the mucosal surfaces
is observed daily, even in healthy individuals, the role of the complement system in this environment
has so far been studied very little. Of particular interest would be the need to suppress complement-
mediated inammation, while still mediating the antimicrobial defense functions.
TABLE 1 | Endogenous and microbial ligands of SALSA.
Endogenous
ligand
Suggested functional relevance
C1q Complement regulation (4)
MBL Complement regulation (6)
Ficolins Complement regulation (6)
SpD Microbial agglutination (9)
SpA Microbial agglutination (16)
IgA Microbial agglutination (8)
Lactoferrin Bacterial binding (20)
DNA Inammation (21)
Heparan sulfate Inammation (21)
Trefoil factors Tissue homeostasis (17)
MUC5B Microbial agglutination (22)
Fibrin Not known (19)
Fibrinogen Not known (19)
Erythrocytes Aggregation (19)
Platelets Aggregation (19)
Microbe Specic strains
Streptococcus S. pyogenes, S. agalactiae, S. pneumonia, S. mutans, S. mitis,
S. oralis, S. salivarius, S. gordonii, S. crista, S. parasanguinis,
S. vestibularis, S. intermedius, S. anginosus, S. suis (7, 2325)
Lactobacillus L. rhamnosus, L. casei, L. reuteri, L. lactis (26)
Other bacteria Staphylococcus aureus, Bidobacterium, Actinomyces,
Salmonella enterica serovar Typhimurium, Helicobacter pylori,
Haemophilus inuenzae, Klebsiella oxytoca (2328)
Viruses HIV, IAV (29, 30)
The listed ligands have been found to bind either human SALSA, the murine-ortholog
of SALSA, or the recombinantly expressed bacterial-binding peptide, SRCRP2.
March 2016 | Volume 7 | Article 852
Reichhardt and Meri
SALSA and Complement Activation
Frontiers in Immunology | www.frontiersin.org
Salivary Scavenger and Agglutinin
One of the molecules at the mucosal surfaces that interact with the
early complement components is a protein that we named salivary
scavenger and agglutinin (SALSA) (47). SALSA, also known as
gp340, “deleted in malignant brain tumors 1” (DMBT1), and sali-
vary agglutinin (SAG), was rst described as a 300–400kDa strep-
tococcus agglutinating agent from saliva (810). Subsequently,
SALSA has been suggested to function in epithelial homeostasis,
innate immunity, inammation, and tumor suppression (1113).
Many of these functions are mediated through interactions with
endogenous ligands. SALSA has been shown to bind the comple-
ment components C1q, mannose-binding lectin (MBL), and the
colins (4, 6). Furthermore, SALSA has been found to interact
with surfactant proteins A and D (SpA and SpD, respectively),
secretory IgA, lactoferrin, brin/brinogen, trefoil factors, and
mucin-5B (Tab le 1 ) (9, 1419). e multiple binding partners
suggest that SALSA plays a central role in regulating inamma-
tion and immune responses on mucosal surfaces.
SALSA in Antimicrobial Defense
Salivary scavenger and agglutinin is expressed at most mucosal
surfaces, including the lungs, oral cavity, gastrointestinal tract,
and vagina (3135). It has been found both attached to the epi-
thelium and secreted into the lining uids, such as saliva, tear
uid, and respiratory mucosal secretions (8, 9, 14, 36). Recent
studies detected SALSA in the amniotic uid and in the intestines
of neonates (37). SALSA was estimated to constitute up to 10% of
the total protein amount in meconium and in the saliva of young
children (<3years), making it one of the most abundant proteins
in these environments (37, 38).
On the mucosal surfaces, SALSA has been shown to regulate
the local immune system. On one hand, it scavenges invading
microorganisms, whereas, on the other hand, it interacts with the
mucosal epithelium to improve the integrity of this physical bar-
rier (Figure1A) (13, 39). SALSA binds a broad range of microbes,
including viruses and bacteria (Table1). Studies have shown that
SALSA in the oral and intestinal mucosal secretions is sucient
to suppress infection by agglutinating microorganisms and keep-
ing them from infecting the tissue. is has been observed for
Salmonella enterica, HIV-1, and inuenza A-virus (IAV) (27, 29,
30, 40). ese studies suggested that SALSA simply functioned by
agglutinating the microbes. However, the role of SALSA appears
to be more complex than that. SALSA binds, e.g., to epithelial and
tooth surfaces in addition to being secreted into the uid phase
(23). e epithelium-attached localization of a protein with a solely
bacteria-agglutinating function would not appear to be benecial
for the human host. is paradox has been made clear by studies
showing that SALSA, in some cases, may actually be exploited
by the invading microbes. A study of dental caries showed that
certain variants of the SALSA protein correlated positively with
Streptococcus mutans adhesion to SALSA-coated hydroxyapatite
surfaces and the development of dental caries. Other SALSA
variants displayed the opposite correlation (41). In the case of
HIV-1 infection, the salivary uid SALSA protein was found to
interfere with oral transmission. However, SALSA expressed on
the vaginal epithelium had an enhancing eect on the infectivity
of the virus (35). ese ndings suggest that some microbes have
evolved mechanisms to utilize SALSA to infect the human body.
Isoforms of the SALSA Protein
As indicated above, various variants of SALSA may interact dier-
ently with microbes. Indeed, dierent SALSA isoforms have been
identied on various mucosal surfaces. ese have been shown to
vary both in protein sequence and in the glycosylation patterns
(23, 36, 37). e gene for SALSA (in chromosome 10q26.13)
encodes 13 highly conserved scavenger receptor cysteine-rich
(SRCR) domains. ese 109-amino acid-long motifs are found
as “pearls on a string” separated by SRCR interspersed domains
(SIDs) (Figure1B). e stretch of 13 SRCR domains is followed
by 2 C1r/C1s, urchin embryonic growth factor and bone morpho-
genetic protein-1 (CUB) domains encompassing the 14th SRCR
domain. Finally, a zona pellucida (ZP) domain is found at the
most C-terminal end (31, 42). e repetitive sequence of SRCR
domains may facilitate alternative splicing (43). Indeed, mRNA
transcripts encoding between 8 and 13 of the N-terminal SRCR
domains have been observed, all in all revealing up to 7 distinct
alleles (31, 42, 44). It has been estimated that SALSA contains
25–45% (w/w) of carbohydrate (8, 31). SALSA contains all the
major sugar components. However, dierences have been found
to correlate to the secretor [Se(+/)] status (±expression of the
α1-2fucosyl-transferase). e blood group antigens, ABO, and
Lewis antigens b and y (Leb and Ley) were found on SALSA from
Se(+) individuals. In contrast, SALSA from Se() individuals did
not contain ABO, Leb nor Ley antigens. Instead, Lewis antigens a
SRCR domain
CUB domain ZP domain
SRCR interspersed domain
A
B
IgA and IgG
C1q/MBL/ficolins
SALSA
Epithelial damage
Healthy epithelium
SpA/SpD
Lume
n
Tissue
Microbe
IIIIII IV
Epithelial cell
FIGURE 1 | Function and structure of SALSA at the mucosal surfaces. (A) At the mucosal surfaces, the SALSA protein is mainly found associated with the
epithelium and secreted into the surrounding uids. The known features and functions of SALSA are presented in four panels (I–IV). (I) SALSA is present on the
epithelial cell surface and deposited in the extracellular matrix, where it is involved in maintaining epithelial homeostasis. (II) Fluid-phase SALSA binds a broad array
of microbes. It has been shown to agglutinate viruses, as well as both Gram-positive and Gram-negative bacteria thus preventing them from invading the parenteral
spaces. (III) SALSA interacts with other endogenous molecules present at the mucosal surfaces, such as surfactant proteins SpA and SpD as well as IgA. It is
believed that these molecules cooperate in antimicrobial defense. (IV) In the case of epithelial damage, cells and molecules from the tissue become mixed with the
luminal contents. In this context, SALSA may bind the complement sensor molecules C1q, MBL, and the colins, thereby SALSA could initiate complement
activation against distinct microbes or participate in the clearance of injured tissue components. (B) In its molecular structure, SALSA contains a stretch of 13
scavenger receptor cysteine-rich (SRCR) domains separated by SRCR interspersed domains. These are followed by two C1r/C1s, urchin embryonic growth factor
and bone morphogenetic protein-1 (CUB) domains surrounding the 14th SRCR domain. Finally, a zona pellucida (ZP) domain is found at the most C-terminal end of
the protein.
March 2016 | Volume 7 | Article 853
Reichhardt and Meri
SALSA and Complement Activation
Frontiers in Immunology | www.frontiersin.org
and x (Lea and Lex) were present (45, 46). us, dierent forms
of the SALSA protein exist. ey are produced both by variations
in the protein chain and in the extent and nature of glycosyla-
tion. e SALSA protein composition varies not only between
individuals but also in dierent body compartments within the
same individual (23, 36, 37).
SALSA AND COMPLEMENT
Interactions of C1q, MBL, and Ficolins
with SALSA
C1q, MBL, and colins all form bouquet-like structures, where
each subunit contains a collagen-like domain (stalk) and a carboxy-
terminal globular domain (the “ower”) (47, 48). C1q binds speci-
cally to surface-attached IgG and IgM. However, other endogenous
non-immunoglobulin ligands have been found, including SALSA
(4, 6). Also, MBL, M-colin, H-colin, and L-colin were found
to bind to SALSA (6). All interactions between SALSA and the
complement molecules were calcium dependent.
C1q was shown to bind SALSA through the globular domain in
a region close to the immunoglobulin-binding site (49). Similarly,
it appears that MBL utilizes the globular carbohydrate recognition
domain (CRD) for the interaction with SALSA. Due to the heavy
glycosylation of SALSA, sugar structures may function as a target
for the CRD of MBL (8, 31). When the binding of MBL was tested
to SALSA puried from the saliva of a single donor up to 60%
inhibition of the SALSA–MBL interaction was observed when
5 mM fucose was added to the uid phase (7). MBL binds to
the Leb antigen, a fucose-containing oligosaccharide (7). A clear
dierence was observed in the binding of MBL to SALSA from
secretors vs. non-secretors (7). is correlates to the nding that
only SALSA from Se(+) individuals contains the Leb antigen (45,
46). is strongly suggests that MBL binds via the CRD to the Leb
antigen of SALSA.
Complement Activation and Regulation by
SALSA
It has been shown that the binding of C1q to SALSA is sucient to
initiate activation of the classical complement activation pathway
(4, 6). In addition, SALSA was shown to inuence the activation
of the lectin pathway through interactions with MBL and the co-
lins (5, 6). e overall outcome of SALSA-mediated complement
regulation varies with the specic location of SALSA (6). SALSA
coated onto a microtiter plate surface activated complement as
March 2016 | Volume 7 | Article 854
Reichhardt and Meri
SALSA and Complement Activation
Frontiers in Immunology | www.frontiersin.org
measured by deposition of C4b and C3b aer incubation with
normal human serum (NHS). Using MBL-decient serum,
approximately 30% of the total complement activation was lost
(6). e residual activation is likely mediated by C1q and possibly
also by the colins (4, 6). In contrast to complement activation
observed by surface-bound SALSA, SALSA in the uid phase
caused a dose-dependent inhibition of the lectin pathway (6). No
such eect was observed on the classical pathway, which may be
due to the almost 100 times higher concentrations of C1q vs. MBL.
SALSA was able to interfere with the binding of the MBL–MASP2
complex to surface-coated mannan. Candida albicans is a known
target for MBL-mediated complement activation. When NHS was
mixed with increasing concentrations of SALSA and incubated
with C. albicans a dose-dependent inhibition of the deposition of
both C4b and C3b was observed on the Candida surface (6). e
dual eects of SALSA on the complement system appear con-
tradictory at rst glance. On one hand, by binding to MBL and
colins in the uid phase, SALSA can prevent their binding to
targets. On the other hand, when bound to a surface, SALSA can
direct complement activation against appropriate targets, such as
microbes. Overall, it appears that SALSA is a mucosal rst line
recognition molecule that can distinguish between targets to be
cleared vs. structures to be tolerated.
Increased SALSA expression may alone and in concert with,
e.g., C1q and MBL, lead to increased microbial clearance. In addi-
tion to the interactions with the complement proteins, SALSA
can also mediate its anti-bacterial and inammation regulating
functions through interactions with IgA, SpA, and SpD (8, 9, 16).
e functional outcome of these interactions is a cooperative
eect on the microbial agglutination (Figure1A) (50, 51). SALSA,
SpA, and SpD have a dual eect against IAV: viral agglutination
and inammatory modulation (52). e binding of the SALSA
ligand SpD to IAV has been shown to induce a strong respiratory
burst response in neutrophils invitro. is response was reduced
by the addition of SALSA (51). It has been suggested that this
allows a regulated response by the neutrophils, with an increased
uptake of IAV but without an excessive and potentially harmful
burst response (13). A similar feature is observed in the case of
C-reactive protein and the other pentraxins. ey target C1q to
apoptotic and necrotic tissue, while simultaneously recruiting
factor H to limit the complement activation (53, 54). is process
is relevant during the removal of apoptotic debris at the mucosal
surfaces, as well (55). e dierential outcome of the interaction
of SALSA with complement may represent a similar balanced
eector mechanism against invading microbes.
SALSA AND COMPLEMENT IN
INFLAMMATORY BOWEL DISEASE
Intestines are one of the primary sites, where an imbalance
between activation and control of immune responses leads to
disease. Inammatory bowel disease (IBD) encompasses two
chronic relapsing and remitting inammatory conditions of the
gastrointestinal tract. ese are known as ulcerative colitis (UC)
and Crohns disease (CD). Together, they aect up to 1:250 in
the adult population (56). In children, the incidence of IBD is on
the rise. Disease onset is during childhood or adolescence for up
to 25% of the patients; although the mortality of the disease has
been declining, it has a major impact on the development of these
young individuals (57). e fundamental causes of the diseases
are still obscure.
Several associations have been found between complement
components and IBD. Specically for the gut mucosa, the devel-
opment of CD has been associated with an altered expression of
components of the lectin pathway. e frequency of the MBL2 gene
allele, which results in MBL deciency, was signicantly elevated
in pediatric patients with CD compared to healthy controls or
adults with Sjögren’s syndrome (58, 59). Deciencies in classical
and alternative pathway components are rarer. Some patients
decient in C1 inhibitor, which is commonly associated with
hereditary angioedema, were found to develop non-infectious
enteritits and IBD (6062). To further highlight an involvement
of the classical and lectin pathways of complement, we recently
observed an association of pediatric IBD to an MHC haplotype
that involves a deciency of two C4 genes (HLA-A03; HLA-B07;
one C4A gene; one C4B gene; HLA-DRB115) (63).
A study of lectin pathway components during CD treatment
found a dramatic impact on M-colin and MASP-3 levels in
patients responding to anti-TNF-α therapy (64). However, how
the complement components specically aect the local inam-
matory environment of the gut is not clear yet. e above described
interactions with the SALSA molecule present a potential way
for complement to aect a balanced mucosal immunological
response. Current models of CD pathogenesis include an altered
response to the local microbiota, and an increased SALSA expres-
sion has been linked to several of these responses (65). Studies
have shown that SALSA can be strongly induced by various
immunological stimuli (66, 67). e increased levels of SALSA in
the intestinal epithelium of patients with IBD and in the ethmoid
sinusoidal mucosa of patients with chronic sinusitis suggest that
SALSA expression is part of the mucosal inammatory response
(6668). Furthermore, a study of preterm infants revealed that an
increase in the pulmonary SALSA levels was part of the mucosal
response to neonatal infection (69).
Salivary scavenger and agglutinin expression by the intesti-
nal epithelium is induced by NOD2 and TLR4 activation (27).
However, the outcome of an induced SALSA expression during
IBD may not necessarily lead to enhanced clearance only. Rather,
the interaction of SALSA with several endogenous molecules may
be part of an ecient but limited immunological response. Failure
in these processes could propagate an unbalanced and overactive
local immune response in IBD. It has been shown that the previ-
ously described SALSA isoforms inuence both the interaction
with microbes and the endogenous ligands, such as IgA, C1q,
and MBL (37). Interestingly, the specic bacterial-binding ability
of SALSA has been found to depend not only on the isoform of
the protein but also on the location of the protein. Fluid-phase
SALSA can bind and aggregate some streptococcal strains, while
SALSA coated to a hydroxyapatite surface does not (23). us,
the association with the mucosal epithelium or the secretion
into the lining uids may further aect the local immunological
environment dierently. Finally, the described interaction of
SALSA and trefoil factors, being important in maintaining the
March 2016 | Volume 7 | Article 855
Reichhardt and Meri
SALSA and Complement Activation
Frontiers in Immunology | www.frontiersin.org
mucosal epithelial barrier, has also been suggested to play a role
in IBD (70, 71).
Salivary scavenger and agglutinin may be part of the normal
immunological response of the mucosal epithelium during infec-
tion. Individual variations in the expression of SALSA isoforms
alternate the ability of SALSA to interact with endogenous
ligands, to invade microbes, and perhaps even to induce a limited
burst response in neutrophils. It is therefore not surprising that a
specic SALSA isoform, lacking the ve most N-terminal SRCR
domains, has been associated with CD (65, 67).
We speculate that the individual variations in the SALSA inter-
actions are key in understanding how this molecule could play
a role in shiing the immunological balance toward increased
inammation at the mucosal surfaces, with detrimental eects
for IBD patients.
FUTURE PERSPECTIVES
At the mucosal surfaces, a very tight immunological response
to infection and inammation is essential. e SALSA molecule
is central player interacting with a multitude of endogenous
molecules, invading microbes and the epithelial barrier. Due to
the tightly linked interactions, a balanced function of the SALSA
molecule is key in avoiding an overactive immune response.
e interplay between the various SALSA isoforms, colins,
MBL, and C1q with modied tissue components, carbohydrates,
acetylated molecules, and microbes on mucosal surfaces provides
an interesting area for future research that may open a new under-
standing of mechanisms underlying the development of mucosal
immunological disorders.
AUTHOR CONTRIBUTIONS
Both authors contributed to the design and writing of this
review.
FUNDING
is work was supported by the Helsinki Biomedical Graduate
Program, the Jenni and Antti Wihuri foundation, Helsinki
University Central Hospital funds (EVO), the Sigrid Jusélius
Foundation, Signe and Ane Gyllenberg Foundation, Magnus
Ehrnrooth Foundation, Helsinki University Funds, the
Stockmann Foundation, and the Academy of Finland.
REFERENCES
1. Boackle RJ. e interaction of salivary secretions with the human complement
system–a model for the study of host defense systems on inamed mucosal
surfaces. Crit Rev Oral Biol Med (1991) 2:355–67.
2. Persson CG, Erjefalt I, Alkner U, Baumgarten C, Grei L, Gustafsson B, etal.
Plasma exudation as a rst line respiratory mucosal defence. Clin Exp Allergy
(1991) 21:17–24. doi:10.1111/j.1365-2222.1991.tb00799.x
3. Courts FJ, Boackle RJ, Fudenberg HH, Silverman MS. Detection of functional
complement components in gingival crevicular uid from humans with
periodontal diseases. J Dent Res (1977) 56:327–31. doi:10.1177/0022034577
0560032001
4. Boackle RJ, Connor MH, Vesely J. High molecular weight non-immunoglobu-
lin salivary agglutinins (NIA) bind C1Q globular heads and have the potential
to activate the rst complement component. Mol Immunol (1993) 30:309–19.
doi:10.1016/0161-5890(93)90059-K
5. Leito JT, Ligtenberg AJ, van Houdt M, van den Berg TK, Wouters D. e
bacteria binding glycoprotein salivary agglutinin (SAG/gp340) activates com-
plement via the lectin pathway. Mol Immunol (2011) 49:185–90. doi:10.1016/j.
molimm.2011.08.010
6. Reichhardt MP, Loimaranta V, iel S, Finne J, Meri S, Jarva H. e salivary
scavenger and agglutinin binds MBL and regulates the lectin pathway of com-
plement in solution and on surfaces. Front Immunol (2012) 3:205. doi:10.3389/
mmu.2012.00205
7. Gunput ST, Ligtenberg AJ, Terlouw B, Brouwer M, Veerman EC, Wouters D.
Complement activation by salivary agglutinin is secretor status dependent.
Biol Chem (2015) 396:35–43. doi:10.1515/hsz-2014-0200
8. Ericson T, Rundegren J. Characterization of a salivary agglutinin reacting with
a serotype c strain of Streptococcus mutans. Eur J Biochem (1983) 133:255–61.
doi:10.1111/j.1432-1033.1983.tb07456.x
9. Holmskov U, Lawson P, Teisner B, Tornoe I, Willis AC, Morgan C, et al.
Isolation and characterization of a new member of the scavenger receptor
superfamily, glycoprotein-340 (gp-340), as a lung surfactant protein-D bind-
ing molecule. J Biol Chem (1997) 272:13743–9. doi:10.1074/jbc.272.21.13743
10. Mollenhauer J, Wiemann S, Scheurlen W, Korn B, Hayashi Y, Wilgenbus
KK, etal. DMBT1, a new member of the SRCR superfamily, on chromosome
10q25.3-26.1 is deleted in malignant brain tumours. Nat Genet (1997) 17:32–9.
doi:10.1038/ng0997-32
11. Kang W, Reid KB. DMBT1, a regulator of mucosal homeostasis through the
linking of mucosal defense and regeneration? FEBS Lett (2003) 540:21–5.
doi:10.1016/S0014-5793(03)00217-5
12. Ligtenberg AJ, Veerman EC, Nieuw Amerongen AV, Mollenhauer J. Salivary
agglutinin/glycoprotein-340/DMBT1: a single molecule with variable compo-
sition and with dierent functions in infection, inammation and cancer. Biol
Chem (2007) 388:1275–89. doi:10.1515/BC.2007.158
13. Madsen J, Mollenhauer J, Holmskov U. Review: gp-340/DMBT1
in mucosal innate immunity. Innate Immun (2010) 16:160–7.
doi:10.1177/1753425910368447
14. ornton DJ, Davies JR, Kirkham S, Gautrey A, Khan N, Richardson PS, etal.
Identication of a nonmucin glycoprotein (gp-340) from a puried respiratory
mucin preparation: evidence for an association involving the MUC5B mucin.
Glycobiology (2001) 11:969–77. doi:10.1093/glycob/11.11.969
15. Rundegren J, Arnold RR. Dierentiation and interaction of secretory immu-
noglobulin A and a calcium-dependent parotid agglutinin for several bacterial
strains. Infect Immun (1987) 55:288–92.
16. Tino MJ, Wright JR. Glycoprotein-340 binds surfactant protein-A (SP-A) and
stimulates alveolar macrophage migration in an SP-A-independent manner.
Am J Respir Cell Mol Biol (1999) 20:759–68. doi:10.1165/ajrcmb.20.4.3439
17. im L, Mortz E. Isolation and characterization of putative trefoil peptide
receptors. Regul Pept (2000) 90:61–8. doi:10.1016/S0167-0115(00)00110-5
18. Oho T, Bikker FJ, Nieuw Amerongen AV, Groenink J. A peptide domain of
bovine milk lactoferrin inhibits the interaction between streptococcal surface
protein antigen and a salivary agglutinin peptide domain. Infect Im mun (2004)
72:6181–4. doi:10.1128/IAI.72.10.6181-6184.2004
19. Muller H, Renner M, Helmke BM, End C, Weiss C, Poeschl J, etal. Deleted in
malignant brain tumors 1 is up-regulated in bacterial endocarditis and binds
to components of vegetations. J orac Cardiovasc Surg (2009) 138:725–32.
doi:10.1016/j.jtcvs.2009.05.021
20. Mitoma M, Oho T, Shimazaki Y, Koga T. Inhibitory eect of bovine milk lactofer-
rin on the interaction between a streptococcal surface protein antigen and human
salivary agglutinin. J Biol Chem (2001) 276:18060–5. doi:10.1074/jbc.M101459200
21. End C, Bikker F, Renner M, Bergmann G, Lyer S, Blaich S, etal. DMBT1 func-
tions as pattern-recognition molecule for poly-sulfated and poly-phosphor-
ylated ligands. Eur J Immunol (2009) 39:833–42. doi:10.1002/eji.200838689
22. Wickstrom C, Christersson C, Davies JR, Carlstedt I. Macromolecular
organization of saliva: identication of ‘insoluble’ MUC5B assemblies and
March 2016 | Volume 7 | Article 856
Reichhardt and Meri
SALSA and Complement Activation
Frontiers in Immunology | www.frontiersin.org
non-mucin proteins in the gel phase. Biochem J (2000) 351(Pt 2):421–8.
doi:10.1042/bj3510421
23. Loimaranta V, Jakubovics NS, Hytonen J, Finne J, Jenkinson HF, Stromberg
N. Fluid- or surface-phase human salivary scavenger protein gp340 exposes
dierent bacterial recognition properties. Infect Immun (2005) 73:2245–52.
doi:10.1128/IAI.73.4.2245-2252.2005
24. Prakobphol A, Xu F, Hoang VM, Larsson T, Bergstrom J, Johansson I, etal.
Salivary agglutinin, which binds Streptococcus mutans and Helicobacter pylori,
is the lung scavenger receptor cysteine-rich protein gp-340. J Biol Chem (2000)
275:39860–6. doi:10.1074/jbc.M006928200
25. Madsen J, Tornoe I, Nielsen O, Lausen M, Krebs I, Mollenhauer J, etal.
CRP-ductin, the mouse homologue of gp-340/deleted in malignant brain
tumors 1 (DMBT1), binds Gram-positive and Gram-negative bacteria and
interacts with lung surfactant protein D. Eur J Immunol (2003) 33:2327–36.
doi:10.1002/eji.200323972
26. Haukioja A, Loimaranta V, Tenovuo J. Probiotic bacteria aect the compo-
sition of salivary pellicle and streptococcal adhesion invitro. Oral Microbiol
Immunol (2008) 23:336–43. doi:10.1111/j.1399-302X.2008.00435.x
27. Rosenstiel P, Sina C, End C, Renner M, Lyer S, Till A, etal. Regulation of DMBT1
via NOD2 and TLR4 in intestinal epithelial cells modulates bacterial recognition
and invasion. J Immunol (2007) 178:8203–11. doi:10.4049/jimmunol.178.12.8203
28. Jumblatt MM, Imbert Y, Young WW Jr, Foulks GN, Steele PS, Demuth DR.
Glycoprotein 340 in normal human ocular surface tissues and tear lm. Infect
Immun (2006) 74:4058–63. doi:10.1128/IAI.01951-05
29. Nagashunmugam T, Malamud D, Davis C, Abrams WR, Friedman HM.
Human submandibular saliva inhibits human immunodeciency virus type
1 infection by displacing envelope glycoprotein gp120 from the virus. J Infect
Dis (1998) 178:1635–41. doi:10.1086/314511
30. Hartshorn KL, White MR, Mogues T, Ligtenberg T, Crouch E, Holmskov U.
Lung and salivary scavenger receptor glycoprotein-340 contribute to the host
defense against inuenza A viruses. Am J Physiol Lung Cell Mol Physiol (2003)
285:L1066–76. doi:10.1152/ajplung.00057.2003
31. Holmskov U, Mollenhauer J, Madsen J, Vitved L, Gronlund J, Tornoe I, etal.
Cloning of gp-340, a putative opsonin receptor for lung surfactant protein D.
Proc Natl Acad Sci U S A (1999) 96:10794–9. doi:10.1073/pnas.96.19.10794
32. Mollenhauer J, Herbertz S, Holmskov U, Tolnay M, Krebs I, Merlo A, etal.
DMBT1 encodes a protein involved in the immune defense and in epithelial
dierentiation and is highly unstable in cancer. Cancer Res (2000) 60:1704–10.
33. Mollenhauer J, Herbertz S, Helmke B, Kollender G, Krebs I, Madsen J, etal.
Deleted in malignant brain tumors 1 is a versatile mucin-like molecule likely to
play a dierential role in digestive tract cancer. Cancer Res (2001) 61:8880–6.
34. Kang W, Nielsen O, Fenger C, Madsen J, Hansen S, Tornoe I, et al. e
scavenger receptor, cysteine-rich domain-containing molecule gp-340 is dif-
ferentially regulated in epithelial cell lines by phorbol ester. Clin Exp Immunol
(2002) 130:449–58. doi:10.1046/j.1365-2249.2002.01992.x
35. Stoddard E, Cannon G, Ni H, Kariko K, Capodici J, Malamud D, etal. gp340
expressed on human genital epithelia binds HIV-1 envelope protein and
facilitates viral transmission. J Immunol (2007) 179:3126–32. doi:10.4049/
jimmunol.179.5.3126
36. Schulz BL, Oxley D, Packer NH, Karlsson NG. Identication of two highly
sialylated human tear-uid DMBT1 isoforms: the major high-molecular-mass
glycoproteins in human tears. Biochem J (2002) 366:511–20. doi:10.1042/
bj20011876
37. Reichhardt MP, Jarva H, de Been M, Rodriguez JM, Jimenez Quintana
E, Loimaranta V, et al. e salivary scavenger and agglutinin in early life:
diverse roles in amniotic uid and in the infant intestine. J Immunol (2014)
193:5240–8. doi:10.4049/jimmunol.1401631
38. Sonesson M, Ericson D, Kinnby B, Wickstrom C. Glycoprotein 340 and sialic
acid in minor-gland and whole saliva of children, adolescents, and adults. Eur
J Oral Sci (2011) 119:435–40. doi:10.1111/j.1600-0722.2011.00879.x
39. Ligtenberg AJ, Karlsson NG, Veerman EC. Deleted in malignant brain
tumors-1 protein (DMBT1): a pattern recognition receptor with multiple
binding sites. Int J Mol Sci (2010) 11:5212–33. doi:10.3390/ijms1112521
40. Wu Z, Van Ryk D, Davis C, Abrams WR, Chaiken I, Magnani J, et al.
Salivary agglutinin inhibits HIV type 1 infectivity through interaction
with viral glycoprotein 120. AIDS Res Hum Retroviruses (2003) 19:201–9.
doi:10.1089/088922203763315704
41. Jonasson A, Eriksson C, Jenkinson HF, Kallestal C, Johansson I, Stromberg N.
Innate immunity glycoprotein gp-340 variants may modulate human suscepti-
bility to dental caries. BMC Infect Dis (2007) 7:57. doi:10.1186/1471-2334-7-57
42. Mollenhauer J, Holmskov U, Wiemann S, Krebs I, Herbertz S, Madsen J, etal.
e genomic structure of the DMBT1 gene: evidence for a region with sus-
ceptibility to genomic instability. Oncogene (1999) 18:6233–40. doi:10.1038/
sj.onc.1203071
43. Mollenhauer J, Muller H, Kollender G, Lyer S, Diedrichs L, Helmke B, etal.
e SRCR/SID region of DMBT1 denes a complex multi-allele system
representing the major basis for its variability in cancer. Genes Chromosomes
Cancer (2002) 35:242–55. doi:10.1002/gcc.10115
44. Mollenhauer J, Helmke B, Muller H, Kollender G, Lyer S, Diedrichs L, etal.
Sequential changes of the DMBT1 expression and location in normal lung
tissue and lung carcinomas. Genes Chromosomes Cancer (2002) 35:164–9.
doi:10.1002/gcc.10096
45. Ligtenberg AJ, Veerman EC, Nieuw Amerongen AV. A role for Lewis a anti-
gens on salivary agglutinin in binding to Streptococcus mutans. Antonie Van
Leeuwenhoek (2000) 77:21–30. doi:10.1023/A:1002054810170
46. Eriksson C, Frangsmyr L, Danielsson Niemi L, Loimaranta V, Holmskov
U, Bergman T, et al. Variant size- and glycoforms of the scavenger receptor
cysteine-rich protein gp-340 with dierential bacterial aggregation. Glycoconj
J (2007) 24:131–42. doi:10.1007/s10719-006-9020-1
47. Reid KB, Porter RR. Subunit composition and structure of subcomponent C1q
of the rst component of human complement. Biochem J (1976) 155:19–23.
doi:10.1042/bj1550019
48. Sellar GC, Blake DJ, Reid KB. Characterization and organization of the genes
encoding the A-, B- and C-chains of human complement subcomponent C1q.
e complete derived amino acid sequence of human C1q. Biochem J (1991)
274(Pt 2):481–90. doi:10.1042/bj2740481
49. Kojouharova MS, Tsacheva IG, Tchorbadjieva MI, Reid KB, Kishore U.
Localization of ligand-binding sites on human C1q globular head region using
recombinant globular head fragments and single-chain antibodies. Biochim
Biophys Acta (2003) 1652:64–74. doi:10.1016/j.bbapap.2003.08.003
50. Ligtenberg AJ, Bikker FJ, De Blieck-Hogervorst JM, Veerman EC, Nieuw
Amerongen AV. Binding of salivary agglutinin to IgA. Biochem J (2004)
383:159–64. doi:10.1042/BJ20040265
51. White MR, Crouch E, Vesona J, Tacken PJ, Batenburg JJ, Leth-Larsen R, etal.
Respiratory innate immune proteins dierentially modulate the neutrophil
respiratory burst response to inuenza A virus. Am J Physiol Lung Cell Mol
Physiol (2005) 289:L606–16. doi:10.1152/ajplung.00130.2005
52. White MR, Crouch E, van Eijk M, Hartshorn M, Pemberton L, Tornoe I, etal.
Cooperative anti-inuenza activities of respiratory innate immune proteins
and neuraminidase inhibitor. Am J Physiol Lung Cell Mol Physiol (2005)
288:L831–40. doi:10.1152/ajplung.00365.2004
53. Jarva H, Jokiranta TS, Hellwage J, Zipfel PF, Meri S. Regulation of complement
activation by C-reactive protein: targeting the complement inhibitory activity
of factor H by an interaction with short consensus repeat domains 7 and 8-11.
J Immunol (1999) 163:3957–62.
54. Deban L, Jarva H, Lehtinen MJ, Bottazzi B, Bastone A, Doni A, etal. Binding
of the long pentraxin PTX3 to factor H: interacting domains and function
in the regulation of complement activation. J Immunol (2008) 181:8433–40.
doi:10.4049/jimmunol.181.12.8433
55. Fox S, Ryan KA, Berger AH, Petro K, Das S, Crowe SE, etal. e role of C1q
in recognition of apoptotic epithelial cells and inammatory cytokine pro-
duction by phagocytes during Helicobacter pylori infection. J Inamm (Lond)
(2015) 12:51. doi:10.1186/s12950-015-0098-8
56. Stone MA, Mayberry JF, Baker R. Prevalence and management of inammatory
bowel disease: a cross-sectional study from central England. Eur J Gastroenterol
Hepatol (2003) 15:1275–80. doi:10.1097/00042737-200312000-00004
57. Van Limbergen J, Russell RK, Drummond HE, Aldhous MC, Round NK,
Nimmo ER, etal. Denition of phenotypic characteristics of childhood-on-
set inammatory bowel disease. Gastroenterology (2008) 135:1114–22.
doi:10.1053/j.gastro.2008.06.081
58. Bak-Romaniszyn L, Szala A, Sokolowska A, Mierzwa G, Czerwionka-Szaarska
M, Swierzko AS, etal. Mannan-binding lectin deciency in pediatric patients
with inammatory bowel disease. Scand J Gastroenterol (2011) 46:1275–8. do
i:10.3109/00365521.2011.594087
March 2016 | Volume 7 | Article 857
Reichhardt and Meri
SALSA and Complement Activation
Frontiers in Immunology | www.frontiersin.org
59. Aittoniemi J, Pertovaara M, Hulkkonen J, Pasternack A, Hurme M, Laippala
P, et al. e signicance of mannan-binding lectin gene alleles in patients
with primary Sjögren’s syndrome. Scand J Rheumatol (2002) 31:362–5.
doi:10.1080/030097402320817095
60. Slade JD, Luskin AT, Gewurz H, Kra SC, Kirsner JB, Zeitz HJ. Inherited
deciency of second component of complement and HLA haplotype A10,B18
associated with inammatory bowel disease. Ann Intern Med (1978) 88:796–8.
doi:10.7326/0003-4819-88-6-796
61. Freeman HJ. Hereditary angioneurotic edema and familial Crohn’s disease.
Can J Gastroenterol (2000) 14:337–9.
62. Farkas H, Gyeney L, Nemesanszky E, Kaldi G, Kukan F, Masszi I, et al.
Coincidence of hereditary angioedema (HAE) with Crohn’s disease. Immunol
Invest (1999) 28:43–53. doi:10.3109/08820139909022722
63. Kolho KL, Paakkanen R, Lepistö AL, Wennerstom A, Meri S, Lokki ML. Novel
associations between major histocompatibility complex and pediatric-onset
inammatory bowel disease. J Pediatr Gastroenterol Nutr (2015). doi:10.1097/
MPG.0000000000000984
64. Sandahl TD, Kelsen J, Dige A, Dahlerup JF, Agnholt J, Hvas CL, etal. e
lectin pathway of the complement system is downregulated in Crohn’s disease
patients who respond to anti-TNF-alpha therapy. J Crohns Colitis (2014)
8:521–8. doi:10.1016/j.crohns.2013.11.007
65. Diegelmann J, Czamara D, Le Bras E, Zimmermann E, Olszak T, Bedynek A,
etal. Intestinal DMBT1 expression is modulated by Crohn’s disease-associated
IL23R variants and by a DMBT1 variant which inuences binding of the tran-
scription factors CREB1 and ATF-2. PLoS One (2013) 8:e77773. doi:10.1371/
journal.pone.0077773
66. Kim TH, Lee SH, Lee HM, Jung HH, Lee SH, Cho WS, etal. Increased expres-
sion of glycoprotein 340 in the ethmoid sinus mucosa of patients with chronic
sinusitis. Arch Otolaryngol Head Neck Surg (2007) 133:1111–4. doi:10.1001/
archotol.133.11.1111
67. Renner M, Bergmann G, Krebs I, End C, Lyer S, Hilberg F, et al. DMBT1
confers mucosal protection in vivo and a deletion variant is associated
with Crohn’s disease. Gastroenterology (2007) 133:1499–509. doi:10.1053/j.
gastro.2007.08.007
68. Hamm CM, Reimers MA, McCullough CK, Gorbe EB, Lu J, Gu CC, etal.
NOD2 status and human ileal gene expression. Inamm Bowel Dis (2010)
16:1649–57. doi:10.1002/ibd.21208
69. Muller H, End C, Weiss C, Renner M, Bhandiwad A, Helmke BM, et al.
Respiratory deleted in malignant brain tumours 1 (DMBT1) levels increase
during lung maturation and infection. Clin Exp Immunol (2008) 151:123–9.
doi:10.1111/j.1365-2249.2007.03528.x
70. Madsen J, Sorensen GL, Nielsen O, Tornoe I, im L, Fenger C, etal. A vari-
ant form of the human deleted in malignant brain tumor 1 (DMBT1) gene
shows increased expression in inammatory bowel diseases and interacts
with dimeric trefoil factor 3 (TFF3). PLoS One (2013) 8:e64441. doi:10.1371/
journal.pone.0064441
71. Aamann L, Vestergaard EM, Gronbaek H. Trefoil factors in inammatory
bowel disease. World J Gastroenterol (2014) 20:3223–30. doi:10.3748/wjg.v20.
i12.3223
Conict of Interest Statement:e authors declare that the research was con-
ducted in the absence of any commercial or nancial relationships that could be
construed as a potential conict of interest.
Copyright © 2016 Reichhardt and Meri. is is an open-access article distributed
under the terms of the Creative Commons Attribution License (CC BY). e use,
distribution or reproduction in other forums is permitted, provided the original
author(s) or licensor are credited and that the original publication in this journal
is cited, in accordance with accepted academic practice. No use, distribution or
reproduction is permitted which does not comply with these terms.
... SAG was originally identified in the parotid saliva as an agglutinating factor of serotype c S. mutans (Ericson and Rundegren 1983) and comprises about 5 to 10% of total salivary proteins of adults and children, respectively (Sonesson et al. 2011). Also known as gp340, Salivary scavenger and agglutinin (SALSA), and "Deleted in malignant brain tumor 1" (DMBT1) (Reichhardt and Meri 2016), SAG is a complex glycoprotein of about 300 to 400 kDa expressed in most epithelial and glandular cells of mucosal tissues, as well as in the endothelium of the vascular system (Polley et al. 2015;Reichhardt et al. 2017). The SAG encoding gene was named DMBT1 because it was identified in an 80 kDa region of the chromosome 10q25.3-q26.1 homozygously deleted in several malignant brain tumours (Mollenhauer et al. 1997;Mollenhauer et al. 1999). ...
... Moreover, there is evidence that the individual patterns or SAG isoform expression are linked to increased risk of microbiome-immune mediated diseases, e.g. Crohn's disease (Reichhardt and Meri 2016), suggesting a role for SAG in individual susceptibilities to diseases resulting from dysbiosis. Fluid-phase SAG can sequester complement activating proteins (MBL and C1q), inhibiting complement activation, whereas surface adsorbed SAG can recruit complement initiating proteins inducing C3b deposition and/or MAC formation (Reichhardt et al. 2012). ...
... Particular SAG isotypes seem to be associated with increased caries increment and with S. mutans adhesion (Johansson and Hansson 2016). Although most bacterial species characterized regarding binding to SAG are streptococci of the oral cavity and nasopharynx (Reichhardt and Meri 2016), information on the effects of these interactions in strain persistence in the oral cavity and other host sites as well as in virulence is largely unknown. ...
Article
In this review, we address the interplay between the complement system and host microbiomes in health and disease, focussing on oral bacteria known to contribute to homeostasis or to promote dysbiosis associated with dental caries and periodontal diseases. Host proteins modulating complement activities in the oral environment and expression profiles of complement proteins in oral tissues were described. In addition, we highlight a sub-set of bacterial proteins involved in complement evasion and/or dysregulation previously characterized in pathogenic species (or strains), but further conserved among prototypical commensal species of the oral microbiome. Potential roles of these proteins in host-microbiome homeostasis and in the emergence of commensal strain lineages with increased virulence were also addressed. Finally, we provide examples of how commensal bacteria might exploit the complement system in competitive or cooperative interactions within the complex microbial communities of oral biofilms. These issues highlight the need for studies investigating the effects of the complement system on bacterial behaviour and competitiveness during their complex interactions within oral and extra-oral host sites.
... It is interesting that Sapt1 seems to target the components of the human innate immune system which normally are involved in the recognition and elimination of Candida. Soluble complement proteins are present mainly in the blood, but also found in serous exudates on mucosal surfaces (Reichhardt and Meri, 2016). This is particularly noticeable under pathological conditions when there is mechanical damage or infection such as periodontitis (Cekici et al., 2013). ...
Article
Full-text available
Candida tropicalis is an opportunistic fungal pathogen and is one of the most frequently isolated non-albicans species. It can cause localised as well as invasive systemic infections particularly in immunocompromised patients. Increased resistance to common anti-fungal drugs is an emerging problem. In order to establish disseminated infections, Candida has evolved several strategies to escape the host immune system. A detailed understanding of how C. tropicalis escapes the host immune attack is needed as it can help develop novel anti-fungal therapies. Secreted aspartyl proteinases (Saps) of C. albicans have been shown to be determinants of virulence and immune evasion. However, the immune evasion properties of C. tropicalis Saps have been poorly characterised. This study investigates the molecular immune evasion properties of C. tropicalis secreted aspartic protease 1 (Sapt1). Sapt1 was recombinantly produced using a Kluyveromyces lactis yeast expression system. A range of complement proteins and immunogloublins were screened to test if Sapt1 had any proteolytic activity. Sapt1 efficiently cleaved human mannose-binding lectin (MBL) and collectin-11, which are the initiating molecules of the lectin pathway of the complement system, but not L-ficolin. In addition, Sapt1 cleaved DC-SIGN, the receptor on antigen presenting dendritic cells. Proteolysis was prominent in acidic condition (pH 5.2), a characteristic of aspartyl protease. No proteolytic activity was detected against complement proteins C1q, C3, C3b, IgG and IgA. In view of the ability of Sapt1 to cleave MBL and collectin-11, we found that Sapt1 could prevent activation of the complement lectin pathway. RT-qPCR analysis using three different C. tropicalis clinical isolates (oral, blood and peritoneal dialysis fluid) revealed relatively higher levels of mRNA expression of Sapt1 gene when compared to a reference strain; Sapt1 protein was found to be secreted by all the tested strains. Lectin pathway and its initiating components are crucial to provide front line defence against Candida infections. For the first time, we have shown that a Candida protease can proteolytically degrade the key initiating components of lectin pathway and inhibit activation. Findings from this study highlight the importance of exploring Sapt1 as a potential therapeutic target. We conclude that C. tropicalis secretes Sapt1 to target the complement lectin pathway, a key pattern recognition and clearance mechanism, for its survival and pathogenesis.
... The small number of viable bacteria observed in our study may be partially explained by fetal bacteriostatic substances. Both the mucosal surface of the intestine as well as the amniotic fluid contain lactoferrin and salivary scavenger and agglutinin (SALSA), which are able to suppress bacterial growth and viability (Reichhardt and Meri, 2016;Lisowska-Myjak et al., 2019). In human meconium, SALSA amounts to 10% of all proteins, highlighting its potential role in antimicrobial defense (Reichhardt et al., 2014). ...
Article
Full-text available
The development of a healthy intestinal immune system requires early microbial exposure. However, it remains unclear whether microbial exposure already begins at the prenatal stage. Analysis of such low microbial biomass environments are challenging due to contamination issues. The aims of the current study were to assess the bacterial load and characterize the bacterial composition of the amniotic fluid and meconium of full-term calves, leading to a better knowledge of prenatal bacterial seeding of the fetal intestine. Amniotic fluid and rectal meconium samples were collected during and immediately after elective cesarean section, performed in 25 Belgian Blue cow-calf couples. The samples were analyzed by qPCR, bacterial culture using GAM agar and 16S rRNA gene amplicon sequencing. To minimize the effects of contaminants, we included multiple technical controls and stringently filtered the 16S rRNA gene sequencing data to exclude putative contaminant sequences. The meconium samples contained a significantly higher amount of bacterial DNA than the negative controls and 5 of 24 samples contained culturable bacteria. In the amniotic fluid, the amount of bacterial DNA was not significantly different from the negative controls and all samples were culture negative. Bacterial sequences were identified in both sample types and were primarily of phyla Proteobacteria, Firmicutes, Bacteroidetes, and Actinobacteria, with some individual variation. We conclude that most calves encounter in utero maternal-fetal transmission of bacterial DNA, but the amount of bacterial DNA is low and viable bacteria are rare.
... Saliva is a key component of the host defense against infection in the mouth and full of immune materials. Salivary scavenger and agglutinin are responsible for innate immunity in the oral cavity [4]. Secretory immunoglobulin A (SIgA), which plays a critical role in mucosal immunity, is secreted from the salivary gland [5]. ...
Article
Full-text available
Influenza virus and severe acute respiratory syndrome coronavirus (SARS-CoV-2) have caused respiratory diseases worldwide. Coronavirus disease 2019 (COVID-19) is now a global health concern requiring emergent measures. These viruses enter the human body through the oral cavity and infect respiratory cells. Since the oral cavity has a complex microbiota, influence of oral bacteria on respiratory virus infection is considered. Saliva has immune molecules which work as the front line in the biophylactic mechanism and has considerable influence on the incidence and progression of respiratory viral infection. Salivary scavenger molecules, such as gp340 and sialic acid, have been reported to exert anti-influenza virus activity. Salivary secretory immunoglobulin A (SIgA) has potential to acquire immunity against these viruses. Biological features of the oral cavity are thought to affect viral infection in respiratory organs in various ways. In this review, we reviewed the literature addressing the impact of oral conditions on respiratory infectious diseases caused by viruses.
... Originally described as a bacterial agglutinating protein in saliva, subsequent studies showed that DMBT1 can serve as a tumor suppressor that is down-regulated in multiple cancers (Braidotti et al., 2004;Du et al., 2011;Imai et al., 2005). DMBT1 is also known as lung glycoprotein-340 (gp340), salivary agglutinin (SAG), salivary scavenger and agglutinin (Reichhardt and Meri, 2016), muclin (De Lisle et al., 2008), or hensin (Polley et al., 2015). In breast cancer, DMBT1 is a susceptibility gene; expression is decreased in mouse strains susceptible to breast cancer but not in strains with a low incidence of breast cancer (Blackburn et al., 2007). ...
Article
Full-text available
Recurrent and new tumors, attributed in part to lateral invasion, are frequent in squamous cell carcinomas and lead to poor survival. We identified a mechanism by which cancer subverts adjacent histologically normal epithelium to enable small clusters of cancer cells to burrow undetected under adjacent histologically normal epithelium. We show that suppression of DMBT1 within cancer promotes aggressive invasion and metastasis in vivo and is associated with metastasis in patients. Cancer cells via TGFβ1 and TNFα also suppress DMBT1 in adjacent histologically normal epithelium, thereby subverting it to promote invasion of a small population of tumor cells. The sufficiency of DMBT1 in this process is demonstrated by significantly higher satellite tumor nests in Dmbt1−/− compared with wild-type mice. Moreover, in patients, invasion of small tumor nests under adjacent histologically normal epithelium is associated with increased risk for recurrence and shorter disease-free survival. This study demonstrates a crucial role of adjacent histologically normal epithelium in invasion and its important role in the tumor microenvironment and opens new possibilities for therapeutic strategies that reduce tumor recurrence.
... Both the mucosal surface of the intestine as well as the amniotic fluid contain bacteriostatic substances, such as lactoferrin and salivary scavenger and agglutinin (SALSA), which are able to suppress bacterial growth and viability (Reichhardt and Meri, 2016;Lisowska-Myjak et al., 2019). In human meconium, SALSA amounts to 10 % of all proteins, highlighting its potential role in antimicrobial defense (Reichhardt et al., 2014). ...
Preprint
Full-text available
The fetal gut microbiome is being investigated for its putative effect on fetal development and maturation of the immune system. In this environment the low amounts of bacteria and bacterial DNA makes them highly challenging to accurately isolate and identify, and sensitive to contamination issues. The aims of the current study were to assess the bacterial load and characterize the bacterial composition of the amniotic fluid and meconium of full-term calves, leading to a better knowledge of prenatal bacterial seeding of the fetal intestine. Amniotic fluid and rectal meconium samples were collected during and immediately after elective caesarean section, performed in 25 Belgian Blue cow-calf couples. The samples were analyzed by qPCR, bacterial culture using GAM agar and 16S rRNA gene amplicon sequencing. To minimize the effects of contaminants, we included numerous technical controls and stringently filtered the 16S rRNA gene sequencing data to exclude putative contaminant sequences. The meconium samples contained a significantly higher amount of bacterial DNA than the negative controls and 5 of the samples contained culturable bacteria. In the amniotic fluid, the amount of bacterial DNA was not significantly different from the negative controls and no colonies were obtained .
Article
Full-text available
Discovering loci under balancing selection in humans can identify loci with alleles that affect response to the environment and disease. Genome variation data have identified the 5’ region of the DMBT1 gene as undergoing balancing selection in humans. DMBT1 encodes the pattern-recognition glycoprotein DMBT1, also known as SALSA, gp340 or salivary agglutinin. DMBT1 binds to a variety of pathogens through a tandemly-arranged scavenger receptor cysteine-rich (SRCR) domain, with the number of domains polymorphic in humans. We show that the signal of balancing selection is driven by one haplotype usually carrying a shorter SRCR repeat, and another usually carrying a longer SRCR repeat. DMBT1 encoded by a shorter SRCR repeat allele does not bind a cariogenic and invasive Streptococcus mutans strain, in contrast to the long SRCR allele which shows binding. Our results suggest that balancing selection at DMBT1 is due to host-microbe interactions of encoded SRCR tandem repeat alleles.
Article
Contact lenses are biomaterials worn on the eye to correct refractive errors. Bacterial adhesion and colonization of these lenses results in adverse events such as microbial keratitis. The adsorption of tear proteins to contact lens materials enhances bacterial adhesion. Glycoprotein 340 (Gp340), a tear component, is known to promote microbial colonization in the oral cavity, however, it has not been investigated in any contact lens-related adverse event. Therefore, this study examined the adsorption of Gp340 and its recombinantly expressed scavenger receptor cysteine rich ( i SRCR 1 Gp340 ) domain on two common contact lens materials, etafilcon A and lotrafilcon B, and the concomitant effects on the adherence of clinical isolates of microbial keratitis causative agents, Pseudomonas aeruginosa (PA6206, PA6294), and Staphylococcus aureus (SA38, USA300). Across all strains and materials, i SRCR 1 Gp340 enhanced adherence of bacteria in a dose-dependent manner. However, i SRCR 1 Gp340 did not modulate lysozyme’s and lactoferrin’s effects on bacterial adhesion to the contact lens. The Gp340 binding surface protein SraP significantly enhanced USA300 binding to i SRCR 1 Gp340 -coated lenses. In addition, i SRCR 1 Gp340 -coated surfaces had significantly diminished biofilms with the SraP mutant (ΔSraP ), and with the Sortase A mutant (ΔSrtA ), there was a further reduction in biofilms, indicating the likely involvement of additional surface proteins. Finally, the binding affinities between i SRCR 1 Gp340 and SraP were determined using surface plasmon resonance (SPR), where the complete SraP binding region displayed nanomolar affinity, whereas its smaller fragments adhered with micromolar affinities. This study concludes that Gp340 and its SRCR domains play an important role in bacterial adhesion to the contact lens.
Article
The first months of life represent a crucial time period for an infant. Alongside establishing the early microbiome, the mucosal immunological homeostasis is being developed. Both processes may be perturbed in prematurely born infants. The glycoprotein SALSA plays a role in mucosal inflammation and microbial clearance. It is one of the most abundant molecules on the intestinal mucosal surfaces in early life. SALSA binds to many types of microbes and host defence molecules like IgA, C1q and collectin molecules. We here describe the development in faecal SALSA levels during the first three months of life. During these 90 days, the median SALSA level in full‐term babies decreased from 1100 μg/mL (range 49‐17 000 μg/mL) to 450 μg/mL (range 33‐1000 μg/mL). Lower levels of SALSA were observed in prematurely born infants in the same time period. Our novel observation thus indicates an impact of prematurity on an important component of the infant intestinal immune system. Changes in SALSA in early life may have an effect on the early establishment of the human microbiome.
Article
Full-text available
Background: Gastric epithelial cells (GECs) undergo apoptosis during H. pylori infection and phagocytes within the mucosa engulf these cells. The recognition and clearance of apoptotic cells is a multifactorial process, enhanced by the presence of various bridging molecules and opsonins which are abundant in serum. However, it is not clear how recognition or clearance may differ in the context of H. pylori infection induced apoptosis. In addition, efferocytosis of sterile apoptotic cells is known to confer anti-inflammatory properties in the engulfing phagocyte, however it is unknown if this is maintained when phagocytes encounter H. pylori-infected cells. Thus, the ability of macrophages to bind and engulf gastric epithelial cells rendered apoptotic by H. pylori infection and the association of these interactions to the modulation of phagocyte inflammatory responses was investigated in the absence and presence of serum with a particular focus on the role of serum protein C1q. Methods: Control (uninfected) or H. pylori-infected AGS cells were co-cultured with THP-1 macrophages in the presence or absence of serum or serum free conditions + C1q protein (40-80 μg/mL). Binding of AGS cells to THP-1 macrophages was assessed by microscopy and cytokine (IL-6 and TNF-α) release from LPS stimulated THP-1 macrophages was quantified by ELISA. Results: We show that macrophages bound preferentially to cells undergoing apoptosis subsequent to infection with H. pylori. Binding of apoptotic AGS to THP-1 macrophages was significantly inhibited when studied in the absence of serum and reconstitution of serum-free medium with purified human C1q restored binding of macrophages to apoptotic cells. Co-culture of sterile apoptotic and H. pylori-infected AGS cells both attenuated LPS-stimulated cytokine production by THP-1 macrophages. Further, direct treatment of THP-1 macrophages with C1q attenuated LPS stimulated TNF-α production. Conclusions: These studies suggest that C1q opsonizes GECs rendered apoptotic by H. pylori. No differences existed in the ability of infected or sterile apoptotic cells to attenuate macrophage cytokine production, however, there may be a direct role for C1q in modulating macrophage inflammatory cytokine production to infectious stimuli.
Article
Full-text available
The salivary scavenger and agglutinin (SALSA), also known as gp340 and dmbt1, is an antimicrobial and inflammation-regulating molecule located at the mucosal surfaces. The present study revealed that SALSA was present in the amniotic fluid (AF) and exceptionally enriched in both meconium and feces of infants. Based on immunological and mass spectrometric analysis, SALSA was estimated to constitute up to 4-10% of the total protein amount in meconium, making it one of the most abundant proteins. SALSA proteins in the AF and intestinal samples were polymorphic and exhibited varying polypeptide compositions. In particular, a different abundance of peptides corresponding to functionally important structures was found in the AF and intestinal SALSA. The AF form of SALSA had a more intact structure and contained peptides from the zona pellucida domain, which is involved in cell differentiation and oligomerization. In contrast, the intestinal SALSA was more enriched with the scavenger receptor cysteine-rich domains. The AF, but not the meconium SALSA, bound to Streptococcus pyogenes, S. agalactiae, S. gordonii, and Escherichia coli. Furthermore, differential binding was observed also to known endogenous ligands C1q, mannose-binding lectin, and secretory IgA. Our results have thus identified mucosal body compartments, where SALSA is particularly abundant, and suggest that SALSA exhibits varying functions in the different mucosal locations. The high levels of SALSA in AF and the infant intestine suggest a robust and important function for SALSA during the fetal development and in the mucosal innate immune defense of infants.
Article
Full-text available
Inflammatory bowel disease (IBD), which comprises ulcerative colitis and Crohn's disease, is characterized by inflammation of the gastrointestinal tract. The trefoil factors 1, 2, and 3 (TFF1-3) are a family of peptides that play important roles in the protection and repair of epithelial surfaces, including the gastrointestinal tract. TFFs may be involved in IBD pathogenesis and are a potential treatment option. In the present review, we describe the TFF family and their potential role in IBD by summarizing the current knowledge of their expression, possible function and pharmacological role in IBD.
Article
Full-text available
DMBT is an antibacterial pattern recognition and scavenger receptor. In this study, we analyzed the role of DMBT1 single nucleotide polymorphisms (SNPs) regarding inflammatory bowel disease (IBD) susceptibility and examined their functional impact on transcription factor binding and downstream gene expression. Seven SNPs in the DMBT1 gene region were analyzed in 2073 individuals including 818 Crohn's disease (CD) patients and 972 healthy controls in two independent case-control panels. Comprehensive epistasis analyses for the known CD susceptibility genes NOD2, IL23R and IL27 were performed. The influence of IL23R variants on DMBT1 expression was analyzed. Functional analysis included siRNA transfection, quantitative PCR, western blot, electrophoretic mobility shift and luciferase assays. IL-22 induces DMBT1 protein expression in intestinal epithelial cells dependent on STAT3, ATF-2 and CREB1. IL-22 expression-modulating, CD risk-associated IL23R variants influence DMBT1 expression in CD patients and DMBT1 levels are increased in the inflamed intestinal mucosa of CD patients. Several DMBT1 SNPs were associated with CD susceptibility. SNP rs2981804 was most strongly associated with CD in the combined panel (p = 3.0×10(-7), OR 1.42; 95% CI 1.24-1.63). All haplotype groups tested showed highly significant associations with CD (including omnibus P-values as low as 6.1×10(-18)). The most strongly CD risk-associated, non-coding DMBT1 SNP rs2981804 modifies the DNA binding sites for the transcription factors CREB1 and ATF-2 and the respective genomic region comprising rs2981804 is able to act as a transcriptional regulator in vitro. Intestinal DMBT1 expression is decreased in CD patients carrying the rs2981804 CD risk allele. We identified novel associations of DMBT1 variants with CD susceptibility and discovered a novel functional role of rs2981804 in regulating DMBT1 expression. Our data suggest an important role of DMBT1 in CD pathogenesis.
Article
Full-text available
The protein deleted in malignant brain tumors (DMBT1) and the trefoil factor (TFF) proteins have all been proposed to have roles in epithelial cell growth and cell differentiation and shown to be up regulated in inflammatory bowel diseases. A panel of monoclonal antibodies was raised against human DMBT1(gp340). Analysis of lung washings and colon tissue extracts by Western blotting in the unreduced state, two antibodies (Hyb213-1 and Hyb213-6) reacted with a double band of 290 kDa in lung lavage. Hyb213-6, in addition, reacted against a double band of 270 kDa in colon extract while Hyb213-1 showed no reaction. Hyb213-6 showed strong cytoplasmic staining in epithelial cells of both the small and large intestine whereas no staining was seen with Hyb213-1. The number of DMBT1(gp340) positive epithelial cells, stained with Hyb213-6, was significantly up regulated in inflammatory colon tissue sections from patients with ulcerative colitis (p<0.0001) and Crohn's disease (p = 0.006) compared to normal colon tissue. Immunohistochemical analysis of trefoil factor TFF1, 2 and 3 showed that TFF1 and 3 localized to goblet cells in both normal colon tissue and in tissue from patients with ulcerative colitis or Crohn's disease. No staining for TFF2 was seen in goblet cells in normal colon tissue whereas the majority of tissue sections in ulcerative colitis and Crohn's disease showed sparse and scattered TFF2 positive goblet cells. DMBT1 and TFF proteins did therefore not co-localize in the same cells but localized in adjacent cells in the colon. The interaction between DMBT1(gp340) and trefoil TFFs proteins was investigated using an ELISA assay. DMBT1(gp340) bound to solid-phase bound recombinant dimeric TFF3 in a calcium dependent manner (p<0.0001) but did not bind to recombinant forms of monomeric TFF3, TFF2 or glycosylated TFF2. This implies a role for DMBT1 and TFF3 together in inflammatory bowel disease.
Article
Purpose: Major Histocompatibility Complex (MHC) genes have been widely studied in adult inflammatory bowel disease (IBD), but data on MHC genes are scarce in pediatric IBD. This study focused on MHC association of genes with pediatric-onset IBD and its different phenotypes. Methods: Blood samples of 103 patients with pediatric IBD (Crohn's disease [CD] or ulcerative colitis [UC]) were collected at Children's Hospital, University of Helsinki, Finland. HLA-A, -B, -DRB1 alleles and complement C4A and C4B gene copy numbers were determined and constructed into haplotypes by a Bayesian algorithm (PHASE). A general population cohort (n = 149) served as a control. HLA-alleles and C4 deficiency frequencies were compared between patients and controls with Chi-squared and Fisher's exact test with Bonferroni correction (pcorr). Results: One MHC haplotype HLA-A03;HLA-B07;one C4A gene;one C4B gene;HLA-DRB115 was more common in CD and UC than in controls (7/61, 11.5%, 6/42, 14.3% and 1/149, 0.7% respectively, OR = 19.19, 95%CI = 2.31-159.57, pcorr = 0.004 for CD vs. controls and OR = 24.67, 95%CI = 2.88-211.36, pcorr = 0.002 for UC vs. controls). Two MHC markers were associated with clinical characteristics. HLA-DRB101 was more common in patients with milder disease course, i.e. no need for anti-TNF-α medication (18/32, 56.2% vs. 19/71, 26.8% without and with anti-TNF-α medication, respectively, OR = 0.28, 95%CI = 0.12-0.68, pcorr = 0.032). C4B deficiency (<2 C4B genes) was associated with complicated recovery after surgery (12/16, 75.0% vs. 4/16, 25.0%, respectively, OR = 9.00, 95%CI = 1.82-44.59, pcorr = 0.025). Conclusions: One MHC haplotype is strongly linked with pediatric-onset IBD, whereas the need for immunomodulatory therapy, and surgery outcome associate with other distinct MHC gene markers.
Article
Objectives: To estimate the prevalence of inflammatory bowel disease (IBD) from the information in general practitioners’ records and to describe patient management, including the prescribing of 5-aminosalicylates and adherance to treatment in ulcerative colitis, and frequency of advice given concerning cessation of smoking. Methods: Fifteen general practices were recruited through the Trent Focus Collaborative Research Network, UK, to take part in a cross-sectional study. They identified confirmed cases of IBD and used a pro-forma to collect data for collation and analysis. Results: Searches identified 344 IBD cases from a combined list of 86 801 patients, suggesting a prevalence of 396 per 100 000 (95% confidence interval, 356–440). Practices considered 32% of patients to be under the sole care of general practitioners; only 59% had been seen in secondary care during the previous year and the numbers of outpatient and general practitioner consultations were similar. Smoking cessation advice was documented for similar numbers of smokers with Crohn's disease and ulcerative colitis. Excluding patients who had undergone surgery, only 65% of patients with ulcerative colitis had been prescribed a 5-aminosalicylate in the previous 6 months and good treatment adherence was suggested in only 42% of ulcerative colitis patients taking a 5-aminosalicylate. Prescribing of aminosalicylates was more common in patients under specialist or shared care than those under general practitioner care only; this remained significant in a regression model also including extent of disease (P < 0.0001). Conclusions: General practitioners play an important role in caring for patients with IBD and may need relevant education and support. Aminosalicylates appear to be under-used in patients with ulcerative colitis.
Article
Abstract After mucosal damage or gingival inflammation, complement proteins leak into the oral cavity and mix with salivary proteins such as salivary agglutinin (SAG/gp-340/DMBT1). This protein is encoded by the gene Deleted in Malignant Brain Tumors 1 (DMBT1), and it aggregates bacteria, viruses and fungi, and activates the lectin pathway of the complement system. In the lectin pathway, carbohydrate structures on pathogens or altered self cells are recognized. SAG is highly glycosylated, partly on the basis of the donor's blood group status. Whereas secretors express Lewis b, Lewis y, and antigens from the ABO-blood group system on SAG, non-secretors do not. Through MBL binding and C4 deposition assays, we aimed to identify the chemical structures on SAG which are responsible for complement activation. The complement-activating properties of SAG were completely abolished by oxidation of its carbohydrate moiety. SAG-mediated activation of complement was also inhibited in the presence of saccharides such as fucose and Lewis b carbohydrates, and also after pretreatment with the fucose-binding lectin, Anguilla anguilla agglutinin. Complement activation was significantly (p<0.01) higher in secretors than in non-secretors. Our results suggest that fucose-rich oligosaccharide sidechains, such as Lewis b antigens, are involved in the activation of complement by SAG.
Article
The lectin pathway of the complement system is initiated through the recognition of pathogens or altered self-structures by mannan-binding lectin (MBL) or ficolins and subsequent activation of MBL-associated serine proteases (MASPs). Altered ficolin levels may contribute to a dysregulated immune response in Crohn's disease (CD). A complete analysis of the lectin pathway has not been performed in patients with CD. We hypothesised that the lectin pathway proteins exacerbate inflammation in CD. We assessed the lectin pathway proteins in 43 patients with active CD & 350 blood donors by measuring the serum levels of MBL; M-, H-, and L-ficolin; MASP-2; MASP-3; and MAp44. In patients with CD, the blood samples were obtained during induction treatment with infliximab or adalimumab. Of 43 patients with CD, 32 (74%) were classified as responders. We observed a nearly 50% decrease in median M-ficolin levels between day 0 and weeks 1/7 in the responders (p<0.001), whereas there was no decrease in the non-responders. MASP-2 levels decreased from baseline to week 1 in both the responders (37%, p<0.0001) and the non-responders (29%, p=0.02). In the responders only, the level of the inhibitory serine protease MASP-3 increased by 26% from baseline to week 1 (p<0.001) and remained high at week 7. Our findings indicated that M-ficolin, MASP-2, and MASP-3 may act in concert to reduce the activity of the lectin pathway, in patients with CD who respond to biological therapy.