ArticlePDF Available

Ficolin-3 Deficiency Is Associated with Disease and an Increased Risk of Systemic Lupus Erythematosus

Authors:
  • Aarhus University & Aarhus University Hospital

Abstract and Figures

Purpose Ficolin-3 deficiency is caused by a mutation (+1637delC) in the FCN3 gene. It is a rare condition and has been associated with both infection and autoimmune disease including systemic lupus erythematosus (SLE). Here we investigated if ficolin-3 deficiency is more frequent in patients than in controls and tried to identify a common phenotype among ficolin-3 deficient individuals. Since a significant part of patients identified with ficolin-3 deficiency was diagnosed with SLE, we explored whether the heterozygous state of the FCN3+1637delC variant represents a risk factor in the development of SLE. Further, we examined other possible causes of ficolin-3 deficiency when the FCN3+1637delC is not present. Methods A systematic literature search for studies measuring ficolin-3 was carried out. We examined 362 SLE patients and 596 controls for the presence of the variant FCN3+1637delC. We established assays for measurements of ficolin-3 and of auto-antibodies against ficolin-3. We sequenced the coding and non-coding regions of the FCN3 gene in an SLE patient with ficolin-3 deficiency not carrying the +1637delC. Results Ficolin-3 deficiency leads to an 8-time increased odds of having a disease (p < 0.05). Three out of nine patients with deficiency had SLE. The heterozygous state of the deficiency variant is not associated with increased risk of developing SLE (p = 0.18). Conclusion By systematically reviewing the literature for the described cases of ficolin-3 deficiency, an autoimmune phenotype is emerging. Thirty-three percent of the ficolin-3 deficient patients had SLE. Heterozygosity for the FCN3 gene deletion causing the deficiency does not seem to be associated with the development of SLE.
Content may be subject to copyright.
ORIGINAL ARTICLE
Ficolin-3 Deficiency Is Associated with Disease and an Increased Risk
of Systemic Lupus Erythematosus
Anne Troldborg
1,2,3
&Rudi Steffensen
4
&Marten Trendelenburg
5
&Thomas Hauser
6
&Kasper G. Winther
2
&
Annette G. Hansen
2
&Kristian Stengaard-Pedersen
3
&Anne Voss
7
&Steffen Thiel
2
Received: 5 March 2019 / Accepted: 7 April 2019
#Springer Science+Business Media, LLC, part of Springer Nature 2019
Abstract
Purpose Ficolin-3 deficiency is caused by a mutation (+1637delC)intheFCN3 gene. It is a rare condition and has been
associated with both infection and autoimmune disease including systemic lupus erythematosus (SLE). Here we investigated
if ficolin-3 deficiency is more frequent in patients than in controls and tried to identify a common phenotype among ficolin-3
deficient individuals. Since a significant part of patients identified with ficolin-3 deficiency was diagnosed with SLE, we
explored whether the heterozygous state of the FCN3+1637delC variant represents a risk factor in the development of SLE.
Further, we examined other possible causes of ficolin-3 deficiency when the FCN3+1637delC is not present.
Methods A systematic literature search for studies measuring ficolin-3 was carried out. We examined 362 SLE patients and 596
controls for the presence of the variant FCN3+1637delC. We established assays for measurements of ficolin-3 and of auto-
antibodies against ficolin-3. We sequenced the coding and non-coding regions of the FCN3 gene in an SLE patient with ficolin-3
deficiency not carrying the +1637delC.
Results Ficolin-3 deficiency leads to an 8-time increased odds of having a disease (p< 0.05). Three out of nine patients with deficiency
had SLE. The heterozygous state of the deficiency variant is not associated with increased risk of developing SLE (p=0.18).
Conclusion By systematically reviewing the literature for the described cases of ficolin-3 deficiency, an autoimmune phenotype
is emerging. Thirty-three percent of the ficolin-3 deficient patients had SLE. Heterozygosity for the FCN3 gene deletion causing
the deficiency does not seem to be associated with the development of SLE.
Keywords SLE .ficolin-3 deficiency .complement .complement deficiency .autoimmunity
Introduction
The plasma protein ficolin-3 (also called H-ficolin and origi-
nally named Hakata antigen) was discovered in 1990 as an
autoantigen when antibodies against the protein were identi-
fied in patients with systemic lupus erythematosus (SLE) [1].
Ficolin-3 is a soluble pattern recognition molecule (PRM) of
the innate immune system. It binds to acetylated molecules,
e.g., as found in acetylated carbohydrate structures or on pro-
teins via its fibrinogen-like recognition domain [2]. The fitting
patterns recognized by ficolin-3 may be present on apoptotic
and necrotic cells, and this gives ficolin-3 the possibility of
acting as an initiator of scavenging actions [3]. When bound to
a surface, ficolin-3 can activate the complementsystem via the
lectin pathway (LP), giving rise to both anti-microbial defense
and homeostatic balance. To enable this activation, ficolin-3
relies on activation of attached enzymes, i.e., so-called
Mannose-binding lectin (MBL)-associated serine proteases
Electronic supplementary material The online version of this article
(https://doi.org/10.1007/s10875-019-00627-2) contains supplementary
material, which is available to authorized users.
*Anne Troldborg
annetrol@rm.dk
1
Department of Rheumatology, Aarhus University Hospital, Palle
Juul-Jensens Boulevard 99, 8200 Aarhus N, Denmark
2
Department of Biomedicine, Aarhus University, Aarhus, Denmark
3
Department of Clinical Medicine, Aarhus University,
Aarhus, Denmark
4
Department of Clinical Immunology, Aalborg University Hospital,
Aalborg, Denmark
5
Division of Internal Medicine and Clinical Immunology lab,
Department of Biomedicine, University Hospital Basel, University of
Basel, Basel, Switzerland
6
IZZ Immunologie-Zentrum Zürich, Zürich, Switzerland
7
Department of Rheumatology, Odense University Hospital,
Odense, Denmark
Journal of Clinical Immunology
https://doi.org/10.1007/s10875-019-00627-2
(MASPs) [4]. Ficolin-3 is the most abundant of the five PRMs
of the LP (as compared with ficolin-2, ficolin-1, MBL, and
collectin-LK).
In our previous investigations, we have consistently ob-
served high levels of ficolin-3 in patients with SLE [5,6].
This is in line with investigations by other research groups
[1,7,8]. However, when examining a large SLE cohort of
424 patients, we also identified two ficolin-3 deficient indi-
viduals [6]. One of the patients carried the only known
mutation-causing ficolin-3 deficiency; the other patient
(patient-X) did not.
Ficolin-3 is encoded by the FCN3 gene. A functional gene
is only present in primates whereas, e.g., the rodent FCN3
orthologue is a pseudogene [9,10]. Among the known poly-
morphisms of FCN3, only one leads to complete deficiency in
variant homozygotes. A frameshift mutation causing ficolin-3
deficiency is located on exon 5 of the FCN3 gene
(+1637delC, rs532781899) [11]. It has a gene frequency of
0.01, and thus, homozygosity is expected in 1 out of 10,000
individuals [12,13]. Before our findings in a cohort of SLE
patients [6], only males with ficolin-3 deficiency had been
identified [14].
The PRMs of the LP provide defense against a large array
of pathogens and are primarily mediated by recognition of
highly conserved pathogen-associated molecular patterns,
i.e., repetitive sugar arrays on the surface of microorganisms
that are rare or non-existent on mammalian cells [15].
Deficiencies of the PRMs are therefore hypothesized to be
associated with serious infections by several research groups
[16,17]. Ficolin-3 deficiency has only been described in a
limited number of humans, and the alleged infectious pheno-
type is controversial [14]. Secondary ficolin-3 deficiency
caused by autoantibodies against the protein has been de-
scribed in SLE patients and has been suggested as a biomarker
for disease activity and glomerulonephritis [1,18,19].
SLE is a chronic autoimmune disease. The disease course
is unpredictable and characterized by periods of remission
followed by periods of ongoing disease activity (flares).
Defects in the clearance of apoptotic cells leading to the re-
lease of intracellular autoantigens resulting in the induction of
autoantibody production have been proposed as a key patho-
genic mechanism of SLE [20,21]. In agreement with this,
proteins that are implicated in the stringently orchestrated
clearance of dying cells are likely to play a role in host pro-
tection against SLE.
The objectives of the current study were to investigate if
ficolin-3 deficiency would be more common in patients than
in controls by reviewing published literature on ficolin-3 and
identifying the number of patients and controls that had been
investigated. We aimed to identify a common phenotype
among ficolin-3 deficient individuals. Further, since a signif-
icant part of patients identified with ficolin-3 deficiency were
diagnosed with SLE, we explored whether the heterozygous
state (C/del) of rs532781899 would represent a risk factor in
the development of SLE. Lastly, we examined an SLE patient,
who is ficolin-3 deficient, but not carrying the +1637delC
mutation for other potential causes leading to ficolin-3 defi-
ciency. Further, we describe the first female patient with
ficolin-3 deficiency.
Methods
Study Populations
SLE patients followed at the out-patient clinic of the
Department of Rheumatology,
Aarhus University Hospital, Denmark (n=169), the
Rheumatology out-patient clinic of Odense University
Hospital, Denmark (n= 203), and SLE patients from the rheu-
matology out-patient clinic of Basel, Switzerland, (n= 52)
have previously been described [6]. The Danish cohort is a
cross-sectional cohort and represents patients in full remission
as well as patients with active disease. All patients fulfilled the
1997 revised ACR classification criteria for SLE [22].
Controls (n= 596) used in the study were included with in-
formed consent at the blood bank at Aalborg University
Hospital, Denmark.
Patient-X
The patient is a male SLE patient (from our Danish SLE cohort)
diagnosed at the age of 27 with classical SLE symptoms (ar-
thritis, malar rash, lupus nephritis, positive anti-dsDNA, anti-
nuclear antibodies, low complement). In the 8-year course of
disease, he has had three severe nephritis flares. He currently
has ongoing proteinuria of more than 5 g/day and is treated with
prednisolone (10 mg/day), mycophenolate-mofetil (3 g/day),
and hydroxychloroquine (400 mg/day). The patient was admit-
ted with biopsy-verified vasculitis of the colon in 2015 treated
with high dose intravenous prednisolone. He has not had doc-
umented recurrent infectious episodes.
First Female Patient with Ficolin-3 Deficiency
The patient is a Swiss SLE patient (from our own SLE cohort)
diagnosed at the age of 18 with classical SLE symptoms (ar-
thritis, pleuritis, malar rash, lupus nephritis, positive anti-
dsDNA, anti-nuclear antibodies, low complement, lupus
headache, Coombs-positive anemia, lymphopenia, high anti-
C1q). One year after diagnosis, she presented with meningo-
coccal meningitis (type non-B/C) in cerebrospinal fluid, and
in peripheral blood (culture), consecutively, she developed
septic shock with septic cardiomyopathy, acute renal failure,
disseminated intravascular coagulation, and respiratory fail-
ure. At the time, she was under mild immunosuppression with
JClinImmunol
prednisone (10 mg/day) and azathioprine (50 mg/day). She
was successfully treated with ceftriaxone. Four years after
her SLE diagnosis, she presented with severe pneumonia
(without identification of a pathogen), and 5 years after diag-
nosis with acute Escherichia coli cystitis with pyelonephritis.
In the same year, she had a renal flare and was started on
mycophenolate-mofetil. The mother of this patient was also
diagnosed with SLE and was found to be heterozygous for the
FCN3 gene deletion [6].
Literature Review Search
Articles were identified by systematic literature searches in
MEDLINE, Embase, Web of science, and Scopus covering
the period from 1980 to January 1, 2018. The following search
terms were used and adjusted based on the search engine used:
BH-ficolin^,BFicolin 3^,BFCN3 gene^,BFicolin^,
BComplement Pathway, Mannose-Binding Lectin^.
Inclusion criteria were clinical studies on humans where
ficolin-3 concentrations had been measured in a patient group
and/or control group, or studies where genetic testing for
ficolin-3 deficiency had been performed.
Exclusion criteria were studies in which methods were not
described sufficiently, data were not available for evaluation,
studies in which neither ficolin-3 plasma or serum concentra-
tions nor genetic data were available, or review articles not
containing original data (Supplementary Fig. 1outlines the
screening and selection process of the literature search).
After removing duplicates, the remaining articles were
screened by title and abstract based on the exclusion criteria,
which left 64 articles for full-text assessment. Sixty-four arti-
cles were screened independently and evaluated for inclusion
and exclusion criteria. Ten of these articles were excluded due
to missing information on measured plasma concentrations. In
the remaining 54 articles, the number of patients and controls
examined for ficolin-3 were then added up. Results from the
current article and the article in which our two ficolin-3 defi-
cient SLE patients were identified [6] were added to the group
of articles ending up with a total of 56 articles.
PCR Analysis for FCN3+1637delC Mutation
DNA from SLE patients and controls were extracted from
peripheral blood mononuclear cells (PBMCs) using the
Maxwell16 Blood DNA kit on the Maxwell16 Instrument
(Promega).
To cover the frameshift mutation FCN3+1637delC
(rs532781899) [12], a fragment of 366 bp of FCN3 exon 5
was amplified by PCR using the following primers: 5-
ggccaagatcctccccaca-3and 5-tctggtgggttctggctcc-3.PCR
amplifications were carried out in 50 μl volumes containing
~ 50 ng genomic DNA, 0.5 mM of each primer, 1× PCR
buffer II, 2.5 mM MgCl2, 0.2 mM dNTP, and 0.75 units of
AmpliTaq DNA polymerase (Invitrogen Life Technologies).
The PCR reactions were performed at; 5 m94 °C, 35 cycles
(30 s94 °C, 30 s62 °C, 30 s72 °C), 5 m72 °C.
After a cleaning step (FlashGel Recovery System, Lonza,
Inc.), the fragment was sequenced in both directions using the
ABI BigDye cycle sequencing terminator kit, V 1.1 (Applied
Biosystems). PCR amplifications were performed in 20 μl
volumes at: 1 m96 °C, 25 cycles (10 s96 °C, 5 s50 °C,
4 m60 °C). PCR products were purified (BigDye
XTerminator, Applied Biosystems) and sequences analyzed
(ABI Prism 3500 Genetic Analyzer, Applied Biosystems).
CLC main workbench software was used for alignment of
resulting DNA sequence.
FCN3 Gene Sequencing
A total of 6510 bases of the FCN3 gene (covering all 8 exons
of the gene) were sequenced covering 1350 base pairs (bp)
upstream of exon 1, exon 1, intron 1, exon 2, intron 2, exon 3,
intron 3, exon 4, intron 4, exon 5, and 300 bp upstream of
intron 5, 482 bp downstream of exon 6, exon 6, intron 6, exon
7, and 271 bp upstream of intron 7, 308 bp downstream of
exon 8, besides 1485 bp upstream of the 3UTR part. The
sequence was aligned to the NG_016279 sequence from the
NCBI nucleotide database. The sequencing was performed as
a resequencing service at Eurofins Genomics GmbH Freiburg,
Germany.
Ficolin-3 Assay
Microtitre plates (Nunc, #437958 or # 43791) were incubated
with acetylated bovine serum albumin (Ac-BSA) (Sigma
#B2518) 10 μg/ml coating buffer (0.1 M sodium bicarbonate,
pH 9.6 with 0.09% (w/v) sodium azide, and Ampliqon
#AMPQ44048.1000) and incubated overnight at room tem-
perature (RT). Residual binding sites were blocked by incu-
bation for 1 hour (h) with 200 μl of 1 mg human serum albu-
min (HSA)/ml TBS (10 mM Tris, 145 mM NaCl, pH 7.4) per
well, and plates were washed three times with TBS/TW (TBS
with 0.05% Tween 20). Standard, quality controls, and sam-
ples were then added to the plate in duplicates. The standard
curve was obtained by diluting the standard (pool of human
plasma with a known concentration of 20 μg/ml ficolin-3) 1/
10 and further seven times twofold dilutions with TBS/Tween/
Ca
2+
/HSA (TBS/TW, 5 mM CaCl
2
, and 1 mg/ml HSA). The
concentration of ficolin-3 in the standard is based on compar-
isons of the signal obtained in dilutions of standard with that
obtained in dilutions of purified plasma ficolin-3. Samples and
quality controls were diluted 1/50 in TBS/Tween/Ca
2+
/HSA.
After overnight incubation at 4 °C and three times wash in
TBS/Tween/Ca
2+
biotinylated mouse anti-human-ficolin-3,
clone 4H5 (conc. 0.34 μg/ml, Hycult biotechnology
#HM2089b) diluted 1/1000 in TBS/Tween/Ca
2+
were added
JClinImmunol
to each well at 100 μl/well and incubated for 2 h at RT. The
subsequent steps followed our standard procedures for time-
resolved immunofluorometric assay (TRIFMA) as previously
described [6], i.e., incubation with europium-labeled
streptavidin followed by wash and detection of the signal by
time-resolved fluorometry.
Purification of Ficolin-3 from Serum
Ficolin-3 was purified from human serum following a previ-
ously described procedure [23]. Serum was precipitated with
polyethylene glycol (PEG) in two steps each followed by cen-
trifugation, and at 4 °C, the supernatant was passed through a
10 ml HSA-sepharose (HSA coupled to CNBr-activated se-
pharose beads) column (this removes most L-ficolin), and the
effluent was loaded onto a 20 ml-acetylated HSA-sepharose
column to bind ficolin-3. The column was washed with 80 ml
of TBS/tw/Ca
2+
. Ficolin-3 was subsequently eluted from the
beads with 1 M sodium acetate, pH 7.5, 1 ml/fraction.
Fractions with ficolin-3 were pooled and dialyzed against
50 mM sodium acetate, 0.01% Tween-20, and pH 8.3. After
dialysis, the preparation was centrifuged 9000gfor 10 min.
The ficolin-3 in the supernatant was purified further on an ion
exchange column MonoQ 5/50 GL (GE Healthcare), using a
gradient from 50 mM NaCl to 350 mM NaCl over 30 ml. The
purity of the protein was tested by SDS-PAGE protein stain
(Supplementary Fig. 2).
Assay Measuring Auto-antibodies Against Ficolin-3
Microtitre plates (Nunc, Roskilde, Denmark; # 437958 or #
43791) were incubated with purified ficolin-3 at 0.5 μg/ml
phosphate buffered saline (PBS) overnight at RT. Residual
binding sites were blocked with 1 mg of HSA per ml of
TBS. After washing with TBS/TW, 100 μl of test samples
and three quality controls (plasma chosen to reflect a wide
distribution of concentrations) were added to the plate diluted
1/10 in sample buffer (10 mM Tris/base, 145 mM NaCl,
10 mM EDTA, 0.05% Tween-20, 1 mg HSA/ml, heat-
aggregated human immunoglobulin (Ig)G 100 mg/ml,
pH 7.4). All samples were added in duplicates using a 1/10
dilution. A standard curve was constructed from a pool of four
SLE EDTA plasma samples, initially diluted tenfold followed
by serial threefold dilutions. After incubation overnight at
4 °C, the wells were washed with TBS/TWand incubated with
biotin-rabbit-anti-human-kappa/lambda (Dako #A0194 and
A0191) at 1/5000 in TBS/TW for 2 h at RT. After washing
with TBS/TW, the wells were incubated with europium-
labeled streptavidin (Perkin Elmer, USA; #1244-360) 1/1000
in TBS/TW, 25 μM EDTA for 1 h at RT. After washing,
quantification of europium was performed by adding 200 ml
of enhancement solution (Ampliqon laboratory reagents
#Q99800; Ampliqon, Denmark) per well releasing and
encapsulating the bound europium, and the fluorescence was
read as time-resolved fluorometry on a Victor 5 from Perkin
Elmer.
As a control, plates were also coated with human IgG to be
able to adjust for the difference in background signal between
SLE patients and controls. No significant binding was ob-
served for neither SLE patient sera nor for controls, and no
difference in background signal was observed between pa-
tients and controls.
Western Blot
We tested whether ficolin-3 protein from patient-X could be
detected by Western blot and compared with the signals seen
in other SLE patient samples. Serum samples from six SLE
patients with known ficolin-3 serum concentrations (one de-
ficient, two with high concentrations, one with medium con-
centrations and two with low concentration), based on mea-
surements in TRIFMA assays as described above, were added
to SDS PAGE sample buffer (30 mM Tris-HCL, 10% (v/V)
glycerol, 8 M urea, 3% (w/v)SDS,0.1%(W/v) bromophenol
blue, pH 8.9). TBS was added to reach the desired sample
volume (3045 μl). Dithiothreitol (DDT) was added to reach
60 mM in the samples to be reduced, followed by heating and
addition of iodoacetic acid. Proteins were separated using an
18 well 415% gradient gel (Bio-Rad, Criterion TGX gels #
567-1084). Following electrophoresis, the proteins were blot-
ted onto nitrocellulose membranes (Bio-Rad #170-4159). The
membranes were then blocked by incubation for 30 min at RT
in TBS with 0.1% Tween (v/v), washed, and developed with
goat-anti-human-ficolin-3 (RD#F2367) at 0.5 mg/ml) in pri-
mary buffer (TBS, 1 mM EDTA, pH 7.4, with 1 mg HSA
(CSL Behring #109697) and 100 μg human IgG (CSL
Behring #007815) per ml). The membrane was subsequently
washed and incubated with HRP-conjugated rabbit-anti-goat
IgG antibody (DAKO #P0449) diluted 1/3000 in secondary
buffer (TBS/Tween, 100 μg human IgG/ml, 1 mM EDTA,
pH 7.4). After washing, the blot was developed with
SuperSignal West Dura extended duration substrate (Pierce),
and emission recorded by a charge-coupled device camera.
Ethics
The project was performed according to the Helsinki
Declaration. The Danish Data Protection Agency and the
Central Denmark Region Committees on Health Research.
Ethics approved the study conducted in Aarhus (#1-10-72-
214-13). The Southern Denmark Region Committees on
Health Research Ethics approved the inclusion of patients in
Odense (#2010 0015). For inclusion of the Swiss SLE popula-
tion, the Ethical committee of Northwest and Central
Switzerland approved the project (EKNZ.Ref. no. EK 262/06).
JClinImmunol
Table 1 Patients with ficolin-3 deficiency described in the literature
Clinical presentation Age Gender (M/F) Ficolin-3 concentration
μg/ml (plasma or serum)
Homozygosity for the
FCN3+1637delC
Autoantibodies
against ficolin-3
Reference Dominant
phenotype
Recurrent pulmonary infections, brain abscesses,
and recurrent warts on fingers; bronchiectasis
and pulmonary fibrosis
32 M < 0.1 Yes NA [12] Autoimmune
SLE no other clinical data available NA NA < 1.1 No No [7] Autoimmune
Invasive necrotizing enterocolitis and repeated
skin infections with staphylococci; no other
infectious diseases during 4-year follow-up
29 weeks GA M < 0.1 Yes NA [24] Infectious
Fatal necrotizing enterocolitis 28 weeks GA M 0.8 NA NA [24] Infectious
Perinatal S. agalactiae infection; microcephaly,
growth and mental retardation; ADHD; no
severe infections during 8-year follow-up
35 weeks GA M < 0.1 Yes NA [25] Infectious
Membranous nephropathy; nephrotic syndrome;
EBV infection; no other recurrent infections
50 M < 0.1 Yes NA [14] Autoimmune
Congenital heart disease; pneumonia; no severe
infections during follow-up after cardiac
surgery
11 months M < 0.1 Yes NA [14] Infectious
Healthy individual (control person) no clinical
data available
NA NA < 0.1 Yes NA [26]NA
Healthy individual (control person) no clinical
data available
NA NA < 0.1 Yes NA [26]NA
SLE (arthritis, positive ANA, anti-ds-DNA,
glomerulonephritis, low C3 and C4).
Meningococcal sepsis. 2nd severe lung
infections.
18 F < 0.1 Yes NA [6] Autoimmune
SLE (malar rash, arthritis, glomerulonephritis,
ANA positive, anti-ds-DNA positive,
low complement C3 and C4)
27 M 0.3 No No [6] Autoimmune
JClinImmunol
Results
Ficolin-3 Deficiency
Based on our literature search, a total of 10 individuals with
ficolin-3 deficiency are now described in the literature, nine
males and one female. All individuals with described ficolin-3
deficiency where evaluated for disease phenotype, age, gen-
der, ficolin-3 concentration, genetic data, and autoantibody
status against the ficolin-3 protein where this information
was available (Table 1).
Based on all published data on ficolin-3, we observed that a
total of 9294 patients and 8227 controls had been evaluated
for either ficolin-3 concentration and/or ficolin-3 deficiency
by genetic testing (Supplementary Table 1). These numbers
are based on the addition of the total number of patients and
the total number of controls reported in the included articles of
the literature search. Among these two groups, nine patients
and only one control were deficient for the FCN3del1637,
yielding an odds ratio for having a disease phenotype when
being ficolin-3 deficient of 8.0 (95% CI 1.0163.0, p<0.05)
(Table 2).
FCN3 Mutation in SLE
Since three of the nine identified patients with ficolin-3 deficien-
cy were SLE patients, we examined (in our own cohorts) whether
being a heterozygous carrier of the FCN3 + del1637C would be
a risk factor for the development of SLE. SLE patients (n=362)
and controls (n= 596) were assessed for the mutation. In SLE
patients, 2.8% carried the mutation, and in controls, this was true
for 1.5%. This difference was not significant (p= 0.18) (Fig. 1).
Investigations of the Cause of Ficolin-3 Deficiency
in Patient-X
Sixteen randomly chosen SLE patients from our SLE cohort
with different serum concentrations of ficolin-3 and our ficolin-
3 deficient patient without the known mutation-causing ficolin-
3 deficiency (patient-X), and 18 randomly chosen controls were
examined for auto-antibodies against ficolin-3. No significant
difference was observed between controls and SLE patients
(Fig. 2a),andnocorrelationwasobserved between ficolin-3
plasma concentration and concentration of auto-antibodies
against ficolin-3 (Fig. 2b).Onepatienthadveryhighlevelsof
antibodies against ficolin-3; however, the patient had normal
plasma levels of ficolin-3 (38 μg/ml). Patient-X did not have
higher concentrations of autoantibodies against ficolin-3 than
the controls (open circle Fig. 2a).
To examine whether ficolin-3 protein was present in
patient-X in a form not detectable in our immunoassay, we
investigated patient serum by Western blotting. Ficolin-3 is
normally detected at 34 kDa under reducing conditions
(Fig. 3a) and as several high oligomeric bands above
250 kDa under non-reducing conditions (Fig. 3b). No
Fig. 1 Number of carriers of the
heterozygous mutation FCN3+
1637delC in Danish SLE patients
and controls. In the SLE patients,
2.8% (10 / 362) were heterozy-
gous carriers of the FCN3+
1637delC, the same was true for
1.5% (9/596) of the controls. P
value reflects χ
2
for the difference
between the two groups
Table 2 Cumulated number of patients and controls in the published
literature where ficolin-3 has been measured based on a systematic liter-
ature review. AnOR of havingdisease when being ficolin-3 deficient was
calculated based on the number of ficolin-3 individuals in each group
(patients and controls)
Ficolin-3 measured Patients (p) Controls (c) Deficient (p/c)
Total 9.294 8.227 9/1
Odds for disease when being ficolin-3 deficient: OR 8.0 (95% CI 1.01
63.0, p<0.05)
JClinImmunol
ficolin-3 was detectablein patient-X (lane 1 Fig. 3a (reducing)
and lane 1 Fig. 3b (non-reducing)).
For patient-X, genetic sequencing of all exons [18]and
flanking sequences of the FCN3 gene was carried out to detect
potential new mutations to account for the observed deficien-
cy. No such mutations were discovered. Only a heterozygous
mutation located upstream of exon1 in the promoter region
was observed (Supplementary Fig. 3c).
Fig. 3 Test for ficolin-3 using Western blotting of samples from the SLE
deficient patient not carrying FCN3 1637delC (patient-X). Proteins were
separated on a 415% gel and blotted, and the blot developed with goat-
anti-ficolin-3. aFicolin-3 is detected at 34 k Da under reducing conditions
(lanes 2, 3, 46). No ficolin-3 was present in patient-X (lane 1). bFicolin-
3 is detected as several oligomeric bands above 250 kDa under non-
reducing conditions. No ficolin-3 was present in patient-X (lane 1, a +
b). Lanes 23and57 on each blot represents serum from SLE patients
with high (lane 23), medium (lane 5), and low [6,7] concentrations in
serum of ficolin-3
Fig. 2 Auto-antibodies against ficolin-3. aAuto-antibodies against
ficolin-3 in SLE patients and controls. Open circle represents patient-X
with ficolin-3 deficiency not carrying the known mutation causing
ficolin-3 deficiency (FCN3 + 1637delC). Auto-antibodies against
ficolin-3 did not explain the observed deficiency. bCorrelation between
ficolin-3 plasma concentrations and auto-antibodies against ficolin-3
JClinImmunol
Discussion
By systematically reviewing the literature, we identified 10
individuals with ficolin-3 deficiency. Eight carried the known
FCN3del1637 mutation. Three of the ten described cases had
a diagnosis of SLE. The heterozygous state of the
FCN3del1637 mutation, which leads to a 50 % reduction in
ficolin-3 plasma concentration was not associated with the
development of SLE.
Our immediate hypothesis was, that the discovery of
ficolin-3 deficiency in diseased individuals, could be due to
diseased populations being studied more rigorously than con-
trols. However, when evaluating the literature, we found that
approximately the same number of diseased individuals and
healthy individual hasbeen investigated since the discovery of
ficolin-3. Based on the currently available literature, this im-
plies that the deficient state is a risk factor for disease and not
just a selection bias. The clinical consequences of ficolin-3
deficiency are still unclear and definite conclusions cannot
be drawn based on 10 individuals. However, a pattern of in-
fection in the young cases and an autoimmune phenotype in
the adult cases, does seem to be emerging.
Michalski et al. reported, that heterozygosity for the FCN3
gene deletion does not seem to have major clinical importance
in neonates [25]. This also appears to be the case for SLE, i.e.,
we did not find a significantly higher number of heterozygous
carriers among SLE patients. In concurrence with previous re-
ports, ficolin-3 concentrations in the heterozygous patients were
on average half of the expected normal concentration [6,12].
In patient-X, we were not able to detect a genetic explana-
tion for the observed deficiency. We did find a heterozygous
mutation upstream of exon1. It is, however, unlikely to be the
explanation of the deficiency in patient-X, since the mutation
was heterozygous. Autoantibodies against ficolin-3 could be
an explanation for a pseudo deficient state. This phenomenon
is known from complement C1q, where autoantibodies
against the protein are strongly associated with protein levels
and disease activity [27,28]. Autoantibodies against ficolin-3
were recently reported in as much as 37% of the SLE popu-
lation investigated [18]. We did, however, not observe higher
levels in patient-X, and although only a small number of pa-
tients and controls were examined in the present study (SLE
n= 16, controls n= 16), we did not (on average) see any
difference in antibodies against ficolin-3 between SLE pa-
tients and controls, nor did we see a correlation between
ficolin-3 levels and antibodies against ficolin-3. The deficien-
cy could be caused by epigenetic regulation. Epigenetic regu-
lation of the FCN3 gene has been described in relation to high
ficolin-3 levels in leprosy [29]. We did, however, not have
liver tissue available from patient-X, which would be neces-
sary to investigate this hypothesis further.
In the original studies on antibodies against ficolin-3 by
Inaba et al, 283 SLE patients and 398 patients with other
autoimmune diseases were examined [1]. In the SLE cohort,
15 patients with unmeasurable amounts officolin-3 were iden-
tified, 3 of who were also positive for antibodies against
ficolin-3. In the group of other autoimmune diseases, two
patients were identified as ficolin-3 deficient without detect-
able antibodies against ficolin-3, one with chronic glomerulo-
nephritis and one with primary biliary cirrhosis. The study
was carried out prior to the purification of ficolin-3, and the
authors proposed the deficient state to be caused by autoanti-
bodies against ficolin-3 although they did not detect antibod-
ies against ficolin-3 in most of the patients.
Based on the DNA deposits gathered at the Broad Institute
[13], 26 people homozygous for the FCN3 del1637 mutation
have been detected resulting in an allele frequency of 0.017.
Clinical data on these individuals are not available.
The question still remains, whether ficolin-3 deficiency is
the cause of disease or acts as a disease modifier. In relation
to infections, it has been proposed that the crucial role of
ficolin-3 may be to control normal (commensal) flora that
can cause opportunistic infections rather than to protect from
obligatory pathogens [14]. With regard to autoimmunity, it
has been demonstrated, that ficolin-3 binds to apoptotic cells
and activates the complement system [3,30]. It is well
established, that clearance defects are strongly associated
with autoimmune diseases [20], and it is striking to see that
among the diseased adults with ficolin-3 deficiency SLE,
glomerulonephritis, and lung fibrosis are the clinical
presentations.
We speculate that ficolin-3 deficiency, like C1q deficiency,
could increase the risk of serious infections especially in early
childhood; whereas, the deficient state in adulthood is a po-
tential risk factor for autoimmune diseases like SLE, in which
clearance defects seem to play an important role.
Acknowledgements We thank the Swiss Systemic Lupus Erythematosus
Cohort Study (SSCS) for collaboration in the project. We are grateful to
all patients participating in the project.
Authors Contribution AT, ST, and MT designed the study. AT performed
the literature search. AT, AH, and KG performed the laboratory experi-
ments. RS performed the genetic analysis. AT was in charge of collecting
blood samples and handling the blood samples after they were drawn; AT,
AV, and KS handled patient inclusion and clinical assessments. ST devel-
oped the assays used in the project and supervised laboratory procedures.
AT, ST, and KS wrote the manuscript, and all authors participated in the
editing of the article.
Funding Information The authors would like to acknowledge the Danish
Rheumatism association, Aase and Ejnar Danielsens fond, Lundbeck
Foundation, and the Danish National Research Foundation for financial
support.
Compliance with Ethical Standards
Conflict of Interest The authors declare that they have no conflict of
interest.
JClinImmunol
References
1. Inaba S, Okochi K, Yae Y, Niklasson F, de Verder CH. Serological
studies of an SLE-associated antigen-antibody system discovered
as a precipitation reaction in agarose gel: the HAKATA antigen-
antibody system. Fukuoka Igaku Zasshi. 1990;81:28491.
2. Gout E, Garlatti V, Smith DF, Lacroix M, Dumestre-Pérard C,
Lunardi T, et al. Carbohydrate recognition properties of human
ficolins: glycan array screening reveals the sialic acid binding spec-
ificity of M-ficolin. J Biol Chem. 2010;285:661222.
3. Honoré C, Hummelshoj T, Hansen BE, Madsen HO, Eggleton P,
Garred P. The innate immune component ficolin 3 (Hakata antigen)
mediates the clearance of late apoptotic cells. Arthritis Rheum.
2007;56:1598607.
4. Matsushita M, Kuraya M, Hamasaki N, Tsujimura M, Shiraki H,
Fujita T. Activation of the lectin complement pathway by H-Ficolin
(Hakata antigen). J Immunol. 2002;168:35026.
5. Troldborg A, Thiel S, Jensen L, Hansen S, Laska MJ, Deleuran B,
et al. Collectin liver 1 and collectin kidney 1 and other complement-
associated pattern recognition molecules in systemic lupus erythe-
matosus. Clin Exp Immunol. 2015;182:1328.
6. Troldborg A, Thiel S, Trendelenburg M, Friebus-Kardash J,
Nehring J, Steffensen R, et al. The lectin pathway of complement
activation in patients with systemic lupus erythematosus. J
Rheumatol. 2018;45:113644.
7. Andersen T, Munthe-Fog L, Garred P, Jacobsen S. Serum levels of
ficolin-3 (Hakata antigen) in patients with systemic lupus erythe-
matosus. J Rheumatol. 2009;36:7579.
8. Hein E, Nielsen LA, Nielsen CT, Munthe-Fog L, Skjoedt M-O,
Jacobsen S, et al. Ficolins and the lectin pathway of complement
in patients with systemic lupus erythematosus. Mol Immunol.
2015;63:20914.
9. Endo Y, Liu Y, Kanno K, Takahashi M, Matsushita M, Fujita T.
Identification of the mouse H-ficolin gene as a pseudogene and
orthology between mouse ficolins A/B and human L/M-ficolins.
Genomics. 2004;84:73744.
10. Hummelshøj T, Nissen J, Munthe-Fog L, Koch C, Bertelsen M,
Garred P. Allelic lineages of the ficolin genes (FCNs) are passed
from ancestral to descendant primates. PLoS One. 2011;6:28187.
11. Munthe-Fog L, Hummelshøj T, Ma YJ, Hansen BE, Koch C,
Madsen HO, et al. Characterization of a polymorphism in the cod-
ing sequence of FCN3 resulting in a ficolin-3 (Hakata antigen)
deficiency state. Mol Immunol. 2008;45:26606.
12. Munthe-Fog L, Hummelshøj T, Honoré C, Madsen HO, Permin H,
Garred P. Immunodeficiency associated with FCN3 mutation and
ficolin-3 deficiency. N Engl J Med. 2009;360:263744.
13. ExAC Browser [Internet]. [cited 2019 Jan 8]. Available from: http://
exac.broadinstitute.org/variant/1-27699670-AG-A. Accessed 8
Jan 2019.
14. Michalski M, Świerzko AS, Pągowska-Klimek I, Niemir ZI,
Mazerant K, Domżalska-Popadiuk I, et al. Primary ficolin-3
deficiencyis it associated with increased susceptibility to infec-
tions? Immunobiology. 2015;220:7113.
15. Kjaer TR, Thiel S, Andersen GR. Toward a structure-based com-
prehension of the lectin pathway of complement. Mol Immunol.
2013;56:22231.
16. De Pascale G, Cutuli SL, Pennisi MA, Antonelli M. The role of
mannose-binding lectin in severe sepsis and septic shock. Mediat
Inflamm. 2013;2013:625803.
17. Sprong T, Mollnes TE, Neeleman C, Swinkels D, Netea MG, Van
Der Meer JWM, et al. Mannose-binding lectin is a critical factor in
systemic complement activation during meningococcal septic
shock. Clin Infect Dis. 2009;49(9):13806.
18. Plawecki M, Lheritier E, Clavarino G, Jourde-Chiche N, Ouili S,
Paul S, et al. Association between the presence of autoantibodies
targeting Ficolin-3 and active nephritis in patients with systemic
lupus erythematosus. PLoS One. 2016;11:e0160879.
19. Yoshizawa S, Nagasawa K, Yae Y, Niho Y, Okochi K. A thermo-
labile beta 2-macroglycoprotein (TMG) and the antibody against
TMG in patients with systemic lupus erythematosus. Clin Chim
Acta. 1997;264:21925.
20. Mahajan A, Herrmann M, Muñoz LE. Clearance deficiency and
cell death pathways: a model for the pathogenesis of SLE. Front
Immunol. 2016;7:35.
21. Muñoz LE, Lauber K, Schiller M, Manfredi AA, Herrmann M. The
role of defective clearance of apoptotic cells in systemic autoimmu-
nity. Nat Rev Rheumatol. 2010;6:2809.
22. Hochberg MC. Updating the American College of Rheumatology
revised criteria for the classification of systemic lupus erythemato-
sus. Arthritis Rheum. 1997;40:1725.
23. Zacho RM, Jensen L, Terp R, Jensenius JC, Thiel S. Studies of the
pattern recognition molecule H-ficolin: specificity and purification.
J Biol Chem. 2012;287:807181.
24. Schlapbach LJ, Thiel S, Kessier U, Ammann RA, Aebi C,
Jensenius JC. Congenital H-ficolin deficiency in premature infants
with severe necrotisingenterocolitis. Gut. 2011;60:14389.
25. Michalski M, Szala A, St. Swierzko A, Lukasiewicz J,
Maciejewska A, Kilpatrick DC, et al. H-ficolin (ficolin-3) concen-
trations and FCN3 gene polymorphism in neonates.
Immunobiology. 2012;217:7307.
26. Metzger ML, Michelfelder I, Goldacker S, Melkaoui K, Litzman J,
Guzman D, et al. Low ficolin-2 levels in common variable immu-
nodeficiency patients with bronchiectasis. Clin Exp Immunol.
2015;179(2):25664.
27. Bock M, Heijnen I, Trendelenburg M. Anti- C1q antibodies as a
follow-up marker in SLE patients. PLoS One. 2015;10:e0123572.
28. Thanei S, Vanhecke D, Trendelenburg M. Anti-C1q autoantibodies
from systemic lupus erythematosus patients activate the comple-
ment system via both the classical and lectin pathways. Clin
Immunol. 2015;160:1807.
29. Andrade FA, Beltrame MH, Bini VB, Gonçalves LB, Boldt ABW,
de Messias-Reason IJ. Association of a new FCN3 haplotype with
high ficolin-3 levels in leprosy. Johnson C, editor. PLoS One.
2017;11:e0005409.
30. Kuraya M, Ming Z, Liu X, Matsushita M,Fujita T. Specific binding
of L-ficolin and H-ficolin to apoptotic cells leads to complement
activation. Immunobiology. 2005;209:68997.
PublishersNote Springer Nature remains neutral with regard to juris-
dictional claims in published maps and institutional affiliations.
JClinImmunol
... A rare frameshift variant in the FCN3 gene that encodes ficolin-3 in humans (rs532781899, chr1:27699670_AG_A) causes complete ficolin-3 deficiency in the homozygous state [10]. A recent publication summarized the known cases of ficolin-3 deficiency and suggested an enrichment of severe infections and SLE in deficient individuals [11], similar to the phenotype observed in classical pathway deficiencies. ...
... Instead, we observed four SLE patients with an apparent depletion of ficolin-3 in serum, a phenomenon illustrated by the disappearance of circulating ficolin-3 over time in longitudinal samples. In support of these findings, two of the three ficolin-3 deficient SLE patients described in the literature have no confirmed coding mutations in the FCN3 gene [11,25]. Serum from one of the four deficient patients in our study displayed strong autoreactivity against ficolin-3, implying autoantibodies as a possible cause of the deficient state. ...
... AP serves as an amplification loop during complement activation of any origin pathway and, therefore, is a well-suited target for potent inhibition of unwarranted complement activation. Targeting only the alternative pathway amplification would also preserve the roles of the CP and LP in maintaining tissue homeostasis by promoting a controlled removal of metabolic debris and apoptotic cells without inducing local inflammation [44,45]. ...
Article
Complement activation is a hallmark of systemic lupus erythematosus (SLE) and can proceed through the classical (CP), lectin (LP), or alternative pathway (AP). When managing SLE patients, pathway-specific complement activation is rarely monitored as clinical assays are unavailable. In this study, we aim to differentiate between CP- or LP-mediated complement activation in SLE patients by quantifying pathway-specific protein complexes, namely C1s/C1-inhibitor (C1- INH) (CP-specific activation) and MASP-1/C1-INH (LP-specific activation). Levels for both complexes were assessed in 156 SLE patients and 50 controls using two newly developed ELISAs. We investigated whether pathway-specific complement activation was associated with disease activity and lupus nephritis (LN). Disease activity stratification was performed using SLEDAI scores assessed at inclusion. C1s/C1-INH concentrations were significantly increased in active SLE patients (SLEDAI ≥6) when compared to SLE patients with low disease activity (SLEDAI <6, p<0.01) and correlated with SLEDAI score (r=0.29, p<0.01). In active LN, MASP-1/C1-INH plasma concentrations were significantly increased compared to non-active LN (p=0.02). No differences in MASP-1/C1-INH plasma concentrations were observed between active SLE patients and patients with low disease activity (p=0.11), nor did we observe a significant correlation with disease activity (r=0.12, p=0.15). Our data suggest that the CP and the LP are activated in SLE. The CP is activated in active SLE disease, whereas activation of the LP might be more specific to disease manifestations like LN. Our results warrant further research into specific complement pathway activation in SLE patients to potentially improve specific targeted and tailored treatment approaches.
... For the rs4494157 variant, it has been associated with an increased susceptibility to rheumatic fever with or without rheumatic heart disease (allele A = OR: 2.93, p value: 0.0002 and OR = 2:23, p value = 0.008, respectively) [25], and the +1637delC variant, involving a heterozygous deletion state in the gene and consequent ficolin-3 deficiency, was associated with an increased risk of developing the disease (OR: 8.0; 95% CI 1.01-63.0; p value < 0.05%) [26]. ...
Article
Full-text available
Genetic association studies in rheumatoid arthritis conducted in various populations have yielded heterogeneous results. The present systematic review was conducted to synthesize the results of the studies in order to establish the impact of polymorphisms in the ficolin-coding genes FCN1, FCN2, and FCN3 on the susceptibility to develop rheumatoid arthritis. A systematic literature review was performed using the following keywords “gene (FCN1/FCN2/FCN3)”, “Polymorphism/Genetic Variant”, and “rheumatoid arthritis” in different databases until January 2022. Authors assessed articles by title/abstract and then assessed by full text for data extraction. The risk of bias was assessed using the Newcastle-Ottawa scale. Data synthesis was performed qualitatively and quantitatively. A total of 1519 articles were eligible for inclusion in this review, 3 were identified as relevant for the quantitative synthesis with 670 patients and 1019 controls. For the FCN1 gene, an association was found in the dominant and recessive genetic models of the variants rs2989727 (genotype TT = OR: 0.577, 95% CI: 0.430-0.769) and rs1071583 (genotype GG = OR: 1.537, 95% CI: 1.153-2.049, p = 0.0032 ) with the development of rheumatoid arthritis as a protective or susceptibility factor. FCN2 and FCN3 genes did not show association with disease development. The FCN1 gene variants rs2989727 and rs1071583 are associated with the risk of developing rheumatoid arthritis in populations from Brazil and Belgium, but not in FCN2 and FCN3 gene variants.
... The specifically downregulated protein in AQP-4 IgG positive samples were apolipoprotein L-1, ficolin-3, complement component c1 r subcomponent, complement factor H related protein 2, zinc finger CCH type antiviral protein 1and adiponectin A with functions of immune and inflammation response and lipid transport. Ficolin 3 deficiency is associated with increased risk of systemic lupus erythematosus (SLE), an autoimmune disorder (21). ...
Article
Full-text available
Neuromyelitis optica spectrum disorders (NMOSD) is a demyelinating autoimmune disease affecting the central nervous system causing inflammatory lesions in the optic nerves, spinal cord and other vital areas of CNS. The clinical manifestations include acute transverse myelitis with paraplegia and optic neuritis with impaired vision. In the present study we focussed on comparative expression of serum proteins between NMOSD variants and control. The study has identified a plethora of novel and unexplored acute phase proteins involved in lipid transport and myelination in perspective of NMOSD. The serum proteins obtained after albumin globulin depletion were subjected to LC-MS/MS. The commonly altered proteins in all NMOSD variants with respect to control were smaug homolog protein and serum amyloid A. Truncated breast and ovarian cancer susceptibility protein and cystic fibrosis transmembrane conductance regulator protein were specifically upregulated and can act as potential biomarkers for neuromyelitis optica with autoantibody negativity to Aquaporin - 4 (AQP-4) and myelin oligodendrocyte (MOG) immunoglobulins. In addition, the uniquely downregulated proteins such as antithrombin III and histidine rich glycoprotein in NMO/MOG autoantibody negative samples can be accounted for its dysregulated fibrinolysis associated with NMO. The differentially expressed proteins were involved in cholesterol transport, synaptic vesicle mediated transport, neurotransmission and immune regulation which are closely associated with myelin formation and protection. Supplementary information: The online version contains supplementary material available at 10.1007/s12291-021-01004-w.
... For the three ficolins, only ficolin-3 deficiency, which is caused by a rare frameshift mutation in the FCN3 gene, has been described in humans with variable clinical manifestations mainly related to a tendency for increased infection, autoimmunity, and neurologic complications (Munthe-Fog et al., 2009;Schlapbach et al., 2011;Michalski et al., 2012;Troldborg et al., 2019;Babaha et al., 2020;Dadfar et al., 2020). However, polymorphisms in the FCN1 and FCN2 genes associated with different serum levels of ficolin-1 and ficolin-2, respectively and their functions have been associated with different infectious and autoimmune conditions, but no clear consensus exists about these associations (Garred et al., 2016). ...
Article
Full-text available
The complement system was discovered at the end of the 19th century as a heat-labile plasma component that “complemented” the antibodies in killing microbes, hence the name “complement.” Complement is also part of the innate immune system, protecting the host by recognition of pathogen-associated molecular patterns. However, complement is multifunctional far beyond infectious defense. It contributes to organ development, such as sculpting neuron synapses, promoting tissue regeneration and repair, and rapidly engaging and synergizing with a number of processes, including hemostasis leading to thromboinflammation. Complement is a double-edged sword. Although it usually protects the host, it may cause tissue damage when dysregulated or overactivated, such as in the systemic inflammatory reaction seen in trauma and sepsis and severe coronavirus disease 2019 (COVID-19). Damage-associated molecular patterns generated during ischemia-reperfusion injuries (myocardial infarction, stroke, and transplant dysfunction) and in chronic neurologic and rheumatic disease activate complement, thereby increasing damaging inflammation. Despite the long list of diseases with potential for ameliorating complement modulation, only a few rare diseases are approved for clinical treatment targeting complement. Those currently being efficiently treated include paroxysmal nocturnal hemoglobinuria, atypical hemolytic-uremic syndrome, myasthenia gravis, and neuromyelitis optica spectrum disorders. Rare diseases, unfortunately, preclude robust clinical trials. The increasing evidence for complement as a pathogenetic driver in many more common diseases suggests an opportunity for future complement therapy, which, however, requires robust clinical trials; one ongoing example is COVID-19 disease. The current review aims to discuss complement in disease pathogenesis and discuss future pharmacological strategies to treat these diseases with complement-targeted therapies. Significance Statement The complement system is the host’s defense friend by protecting it from invading pathogens, promoting tissue repair, and maintaining homeostasis. Complement is a double-edged sword, since when dysregulated or overactivated it becomes the host’s enemy, leading to tissue damage, organ failure, and, in worst case, death. A number of acute and chronic diseases are candidates for pharmacological treatment to avoid complement-dependent damage, ranging from the well established treatment for rare diseases to possible future treatment of large patient groups like the pandemic coronavirus disease 2019.
Article
Purpose of review This review aims to provide an overview of the genes and molecular pathways involved in monogenic lupus, the implications for genome diagnosis, and the potential therapies targeting these molecular mechanisms. Recent findings To date, more than 30 genes have been identified as contributors to monogenic lupus. These genes are primarily related to complement deficiency, activation of the type I interferon (IFN) pathway, disruption of B-cell and T-cell tolerance and metabolic pathways, which reveal the multifaceted nature of systemic lupus erythematosus (SLE) pathogenesis. Summary In-depth study of the causes of monogenic lupus can provide valuable insights into of pathogenic mechanisms of SLE, facilitate the identification of effective biomarkers, and aid in developing therapeutic strategies.
Article
Full-text available
Idiopathic pulmonary fibrosis (IPF) is a chronic progressive lung disease with reduced quality of life and earlier mortality, but its pathogenesis and key genes are still unclear. In this investigation, bioinformatics was used to deeply analyze the pathogenesis of IPF and related key genes, so as to investigate the potential molecular pathogenesis of IPF and provide guidance for clinical treatment. Next-generation sequencing dataset GSE213001 was obtained from Gene Expression Omnibus (GEO), and the differentially expressed genes (DEGs) were identified between IPF and normal control group. The DEGs between IPF and normal control group were screened with the DESeq2 package of R language. The Gene Ontology (GO) and REACTOME pathway enrichment analyses of the DEGs were performed. Using the g:Profiler, the function and pathway enrichment analyses of DEGs were performed. Then, a protein–protein interaction (PPI) network was constructed via the Integrated Interactions Database (IID) database. Cytoscape with Network Analyzer was used to identify the hub genes. miRNet and NetworkAnalyst databaseswereused to construct the targeted microRNAs (miRNAs), transcription factors (TFs), and small drug molecules. Finally, receiver operating characteristic (ROC) curve analysis was used to validate the hub genes. A total of 958 DEGs were screened out in this study, including 479 up regulated genes and 479 down regulated genes. Most of the DEGs were significantly enriched in response to stimulus, GPCR ligand binding, microtubule-based process, and defective GALNT3 causes HFTC. In combination with the results of the PPI network, miRNA-hub gene regulatory network and TF-hub gene regulatory network, hub genes including LRRK2, BMI1, EBP, MNDA, KBTBD7, KRT15, OTX1, TEKT4, SPAG8, and EFHC2 were selected. Cyclothiazide and rotigotinethe are predicted small drug molecules for IPF treatment. Our findings will contribute to identification of potential biomarkers and novel strategies for the treatment of IPF, and provide a novel strategy for clinical therapy.
Article
Knowledge related to the biology of inborn errors of immunity (IEI) and associated laboratory testing methods continue to expand at a tremendous rate. Despite this, many patients with IEI suffer for prolonged periods of time prior to identification of their underlying condition, thereby delaying appropriate care. Understanding that test selection and optimal evaluation for patients with recurrent infections or unusual patterns of inflammation can be unclear, we present a document which distills relevant clinical features of immunological disease due to IEI and related appropriate and available test options. This document is intended to serve the practicing clinical immunologist and, in turn, patients by describing best available test options for initial and expanded immunological evaluations across the disease spectrum. Our goal is to demystify the process of evaluating patients with suspected immune dysfunction and to enable more rapid and accurate diagnosis of such individuals.
Article
Full-text available
Background The aim of our study was to estimate the association of ficolin‐1 (FCN1) gene (rs10120023, rs1071583) and ficolin‐3 (FCN3) gene (rs3813800, rs10794501) polymorphisms and pulmonary tuberculosis (PTB) susceptibility, as well as their several clinical features, in a Chinese population. Methods This study included a cohort of 489 PTB patients and 489 healthy controls, and the four SNPs were genotyped by improved multiple ligase detection reaction (iMLDR). Results We found that there were no significant differences regarding the allele and genotype frequencies of FCN1 rs10120023, rs1071583 and FCN3 rs3813800, rs10794501 between PTB patients and healthy controls (all p > 0.05). The association of three main haplotypes (CC, CT, and TC) in FCN1 and three main haplotypes (CT, GA, and GT) in FCN3 with PTB susceptibility was also analyzed, and no significant association was detected (all p > 0.05). In FCN1, the rs1071583 TT genotype was significantly associated with the occurrence of drug resistance in PTB patients (p = 0.040). In addition, the GG genotype and G allele frequencies of rs3813800 in FCN3 gene were significantly higher in PTB patients with pulmonary infection (p = 0.027, p = 0.020, respectively). Conclusions FCN1 and FCN3 genetic variation were not contributed to the pathogenesis of PTB in Chinese. While rs1071583 and rs3813800 variant might associate with several clinical characteristics of PTB.
Article
Ficolins are pattern-recognition molecules (PRMs) that could form complexes with mannose-binding lectin-associated serine proteases (MASPs) to trigger complement activation via the lectin pathway, thereby mediating a series of immune responses including opsonization, phagocytosis and cytokine production. In the past few decades, accumulating evidence have suggested that ficolins play a major role in the onset and development of several autoimmune diseases (ADs), including systemic lupus erythematosus (SLE), rheumatoid arthritis (RA), systemic sclerosis (SSc), Type 1 diabetes (T1D), inflammatory bowel disease (IBD), etc. In this review, we synthesized previous literatures and recent advances to elucidate the immunological regulations of ficolins and discuss the potential diagnostic ability of ficolins in ADs, as well as giving an insight into the future therapeutic options for ficolins in ADs.
Article
Full-text available
Leprosy is a chronic inflammatory disease caused by Mycobacterium leprae that mainly affects the skin and peripheral nervous system, leading to a high disability rate and social stigma. Previous studies have shown a contribution of genes encoding products of the lectin pathway of complement in the modulation of the susceptibility to leprosy; however, the ficolin-3/FCN3 gene impact on leprosy is currently unknown. The aim of the present study was to investigate if FCN3 polymorphisms (rs532781899: g.1637delC, rs28362807: g.3524_3532insTATTTGGCC and rs4494157: g.4473C>A) and ficolin-3 serum levels play a role in the susceptibility to leprosy. We genotyped up to 190 leprosy patients (being 114 (60%) lepromatous), and up to 245 controls with sequence-specific PCR. We also measured protein levels using ELISA in 61 leprosy and 73 controls. FCN3 polymorphisms were not associated with disease, but ficolin-3 levels were higher in patients with FCN3 *2B1 (CinsA) haplotype (p = 0.032). Median concentration of ficolin-3 was higher in leprosy per se (26034 ng/mL, p = 0.005) and lepromatous patients (28295 ng/mL, p = 0.016) than controls (18231 ng/mL). In addition, high ficolin-3 levels (>33362 ng/mL) were more common in leprosy per se (34.4%) and in lepromatous patients (35.5%) than controls (19.2%; p = 0.045 and p = 0.047, respectively). Our results lead us to suggest that polymorphisms in the FCN3 gene cooperate to increase ficolin-3 concentration and that it might contribute to leprosy susceptibility by favoring M. leprae infection.
Article
Full-text available
Systemic lupus erythematosus (SLE) is a chronic autoimmune disease characterized by the production of multiple autoantibodies. Antibodies against Ficolin-3 were previously identified in the sera of some SLE patients, but their prevalence and significance have not been yet investigated. The aims of this study were to determine the prevalence of anti-ficolin-3 antibodies among SLE patients and to investigate their potential as diagnostic and/or prognostic biomarkers in SLE. In this retrospective study, sera from SLE patients (n = 165) were selected from a preexisting declared biological collection. Samples from healthy controls (n = 48) were matched with SLE sera. Disease activity was determined according to the SLEDAI score. Anti-ficolin-3, anti-dsDNA and anti-C1q antibodies levels were measured in sera by ELISA. First, a highly significant difference was found in the anti-ficolin-3 levels between SLE patients and healthy subjects. Anti-ficolin-3 antibodies were detected as positive in 56 of 165 (34%) SLE patients. The titer of anti-ficolin-3 antibodies was correlated with the SLEDAI score (r = 0.38, p<0.0001). The presence of anti-ficolin-3 antibodies was associated with anti-C1q and anti-dsDNA antibodies. Regarding associations with clinical manifestations, the presence of active lupus nephritis was significantly associated with the presence of anti-ficolin-3 antibodies (p≤0.001). This association with renal involvement was higher with anti-ficolin-3 or anti-C1q antibodies than with other auto-antibodies. Interestingly, the combination of anti-ficolin-3 and anti-C1q antibodies demonstrated higher specificity than any other serological biomarker. These results suggest that anti-ficolin-3 antibodies could be useful for the diagnosis of active nephritis in SLE patients.
Article
Full-text available
In cross-sectional studies autoantibodies against complement C1q (anti-C1q) were found to be highly associated with active lupus nephritis. The aim of this retrospective study was to determine the value of anti-C1q as follow-up marker of disease activity and renal involvement in patients with systemic lupus erythematosus (SLE). Fifty-two patients with SLE and a minimum of three anti-C1q measurements during follow-up were analyzed. Anti-C1q levels correlated with global disease activity scores. In subgroup analyses, patients without renal involvement did not show a significant correlation between anti-C1q levels and disease activity. In contrast, in patients with renal involvement, anti-C1q levels correlated well with global disease activity. In addition, a positive correlation with the urine protein-to-creatinine ratio and anti-dsDNA antibody levels as well as a negative correlation with complement levels was observed. Anti-C1q antibodies were found to strongly correlate with parameters of SLE disease activity during follow-up, in particular with regard to renal involvement.
Article
Full-text available
Alterations of cell death pathways, including apoptosis and the neutrophil specific kind of death called NETosis, can represent a potential source of autoantigens. Defects in the clearance of apoptotic cells may be responsible for the initiation of systemic autoimmunity in several chronic inflammatory diseases, including systemic lupus erythematosus (SLE). Autoantigens are released mainly from secondary necrotic cells because of a defective clearance of apoptotic cells or an inefficient degradation of DNA-containing neutrophil extracellular traps (NETs). These modified autoantigens are presented by follicular dendritic cells to autoreactive B cells in germinal centers of secondary lymphoid organs. This results in the loss of self-tolerance and production of autoantibodies, a unifying feature of SLE. Immune complexes (IC) are formed from autoantibodies bound to uncleared cellular debris in blood or tissues. Clearance of IC by blood phagocytes, macrophages, and dendritic cells leads to proinflammatory cytokine secretion. In particular, plasmacytoid dendritic cells produce high amounts of interferon-α upon IC uptake, thereby contributing to the interferon signature of patients with SLE. The clearance of antinuclear IC via Fc-gamma receptors is considered a central event in amplifying inflammatory immune responses in SLE. Along with this, the accumulation of cell remnants represents an initiating event of the etiology, while the subsequent generation of autoantibodies against nuclear antigens (including NETs) results in the perpetuation of inflammation and tissue damage in patients with SLE. Here, we discuss the implications of defective clearance of apoptotic cells and NETs in the development of clinical manifestations in SLE.
Article
Full-text available
The objective was to explore the involvement of collectin liver 1 (CL-L1) and collectin kidney 1 (CL-K1) and other pattern recognition molecules (PRMs) of the lectin pathway of the complement system in a cross-sectional cohort of Systemic Lupus Erythematosus (SLE) patients. Concentrations in plasma of CL-L1, CL-K1, mannan-binding lectin (MBL), M-ficolin, H-ficolin and L-ficolin were determined in 58 patients with SLE and 65 healthy controls using time resolved immuno-flourometric assays. The SLE patients' demographic, diagnostic, clinical, and biochemical data, and collection of plasma samples were done prospectively during 4 months. CL-L1, CL-K1 and M-ficolin plasma concentrations were lower in SLE patients than healthy controls (p values <0.001, 0.033 and p<0.001, respectively). H-ficolin concentration was higher in SLE patients (p<0.0001). CL-L1 and CL-K1 plasma concentrations in the individuals correlated in both patients and controls. Patients with low complement component 3 (C3) demonstrated a negative correlation between C3 and CL-L1 and CL-K1 (p=0.022 and 0.031 respectively). Patients positive for anti-dsDNA-antibodies had lower levels of MBL in plasma than patients negative for anti-dsDNA-antibodies (p=0.02). In a cross-sectional cohort of SLE patients, we found differences in the plasma concentrations of CL-L1, CL-K1, M-ficolin and H-ficolin compared to a group of healthy controls. Alterations in plasma concentrations of the PRMs of the lectin pathway in SLE patients and associations to key elements of the disease support the hypothesis that the lectin pathway plays a role in the pathogenesis of SLE. This article is protected by copyright. All rights reserved. © 2015 British Society for Immunology.
Article
Objective: The pathogenesis of systemic lupus erythematosus (SLE) involves complement activation. Activation of complement through the classical pathway (CP) is well established. However, complement activation through pattern recognition not only happens through the CP, but also through the lectin pathway (LP). We investigated the hypothesis that the LP is activated in SLE and involved in the pathogenesis of the disease. Methods: Using immunoassays developed in-house, we measured concentrations of LP proteins in a cohort of 372 patients with SLE and 170 controls. We estimated complement activation measuring total C3, and investigated whether LP protein concentrations were associated with complement activation and disease activity. Protein changes and disease activity over time were assessed in a cohort of 52 patients with SLE followed with repeated samples over a 5-year period. Results: Concentrations of LP proteins in SLE were altered compared with controls. The differences observed in LP proteins associated with complement activation were reflected by a decrease in total C3. The pattern recognition molecules (M-ficolin, CL-L1, and CL-K1), the serine protease (MASP-3), and the associated protein (MAp19) displayed a negative correlation with disease activity. Changes in MASP-2 concentrations over time correlated significantly with increased disease activity. Association between active proteinuria and serum concentration was observed for MASP-3 and MAp19. Conclusion: In patients with SLE, we measured specific changes in LP proteins that are associated with complement activation and disease activity, indicating that the LP is activated in patients with SLE. These novel findings substantiate the involvement of the LP in SLE.