ArticlePDF Available

Nucleic acid-containing amyloid fibrils potently induce type I interferon and stimulate systemic autoimmunity

Authors:

Abstract

The immunopathophysiologic development of systemic autoimmunity involves numerous factors through complex mechanisms that are not fully understood. In systemic lupus erythematosus, type I IFN (IFN-I) produced by plasmacytoid dendritic cells (pDCs) critically promotes the autoimmunity through its pleiotropic effects on immune cells. However, the host-derived factors that enable abnormal IFN-I production and initial immune tolerance breakdown are largely unknown. Previously, we found that amyloid precursor proteins form amyloid fibrils in the presence of nucleic acids. Here we report that nucleic acid-containing amyloid fibrils can potently activate pDCs and enable IFN-I production in response to self-DNA, self-RNA, and dead cell debris. pDCs can take up DNA-containing amyloid fibrils, which are retained in the early endosomes to activate TLR9, leading to high IFNα/β production. In mice treated with DNA-containing amyloid fibrils, a rapid IFN response correlated with pDC infiltration and activation. Immunization of nonautoimmune mice with DNA-containing amyloid fibrils induced antinuclear serology against a panel of self-antigens. The mice exhibited positive proteinuria and deposited antibodies in their kidneys. Intriguingly, pDC depletion obstructed IFN-I response and selectively abolished autoantibody generation. Our study reveals an innate immune function of nucleic acid-containing amyloid fibrils and provides a potential link between compromised protein homeostasis and autoimmunity via a pDC-IFN axis.
Nucleic acid-containing amyloid brils potently induce
type I interferon and stimulate systemic autoimmunity
Jeremy Di Domizio
a
, Stephanie Dorta-Estremera
a
, Mihai Gagea
b
, Dipyaman Ganguly
c
, Stephan Meller
c
, Ping Li
d
,
Bihong Zhao
e
, Filemon K. Tan
f
, Liqi Bi
d
, Michel Gilliet
c
, and Wei Cao
a,1
Departments of
a
Immunology and
b
Veterinary Medicine and Surgery, University of Texas MD Anderson Cancer Center, Houston, TX 77030;
c
Department of
Dermatology, University Hospital Lausanne, CH-1011 Lausanne, Switzerland;
d
Section of Rheumatology, University of Jilin University, Changchun 130033,
China;
e
Department of Pathology and Laboratory Medicine and
f
Division of Rheumatology, University of Texas Medical School, Houston, TX 77030
Edited* by Michael B. A. Oldstone, The Scripps Research Institute, La Jolla, CA, and approved July 24, 2012 (received for review April 25, 2012)
The immunopathophysiologic development of systemic autoimmu-
nity involves numerous factors through complex mechanisms that are
not fully understood. In systemic lupus erythematosus, type I IFN (IFN-
I) produced by plasmacytoid dendritic cells (pDCs) critically promotes
the autoimmunity through its pleiotropic effects on immune cells.
However, the host-derived factors that enable abnormal IFN-I pro-
duction and initial immune tolerance breakdown are largely un-
known. Previously, we found that amyloid precursor proteins form
amyloid brils in the presence of nucleic acids. Here we report that
nucleic acid-containing amyloid brils can potently activate pDCs and
enable IFN-I production in response to self-DNA, self-RNA, and dead
cell debris. pDCs can take up DNA-containing amyloid brils, which
are retained in the early endosomes to activate TLR9, leading to high
IFNα/βproduction. In mice treated with DNA-containing amyloid
brils, a rapid IFN response correlated with pDC inltration and acti-
vation. Immunization of nonautoimmune mice with DNA-containing
amyloid brils induced antinuclear serology against a panel of self-
antigens. The mice exhibited positive proteinuria and deposited anti-
bodies in their kidneys. Intriguingly, pDC depletion obstructed IFN-I
response and selectively abolished autoantibody generation. Our
study reveals an innate immune function of nucleic acid-containing
amyloid brils and provides a potential link between compromised
protein homeostasis and autoimmunity via a pDC-IFN axis.
autoimmune disease
|
innate immune response
|
disease model
The precise etiology of systemic lupus erythematosus (SLE), a
heterogeneous autoimmune disease with multiple organ in-
volvement, is unclear. SLE manifests with characteristic antinuclear
antibodies (ANA), including those directed against DNA, ribonu-
cleoprotein complex (RNP), and nucleosomes (1, 2). These auto-
antibodies can form immune complexes (ICs), which are deposited
within the kidneys and blood vessels, and contribute critically to the
pathogenesis of such diseases as lupus nephritis and vasculitis. A
signicant number of patients with SLE have inadequate clearing
of apoptotic cell remnants, which include complex antigens con-
taining nucleic acids. The accumulation of these autoantigens
permits the eventualdevelopment of ANA. However, because self-
nucleic acids and apoptotic cell debris are poorly immunogenic, the
mechanism behind the initial breakdown of immune tolerance
leading to systemic autoimmunity remains enigmatic.
Patients with SLE show increased levels of IFN-I in the serum
and expression of IFN-inducible genes in both peripheral blood
cells and affected kidneys, frequently correlating with disease ares
(35). Administration of IFNαto patients with malignant or viral
disease occasionally induces a lupus-like syndrome (5). In auto-
immune-prone mice, exogenous IFNαcan accelerate autoantibody
production and glomerulonephritis, whereas IFNAR deciency
signicantly ameliorates the disease (68). Functionally, IFN-I
potently differentiates monocytes, matures dendritic cells (DCs),
promotes B-cell differentiation and antibody production, modu-
lates survival, proliferation, and differentiation of T cells, and
primes neutrophils for death by NETosis (914). Therefore, IFN-I
acts as a central effector molecule to promote autoimmunity.
Amyloid brils are stable insoluble aggregates of misfolded
protein products with extensive β-sheet structures (15). Multiple
aberrant polypeptides are implicated in more than 20 human
pathologies (16). Amyloid and related misfolded protein spe-
cies critically affect neuronal functions in the central nervous
system (CNS) and participate in inammatory responses in both
CNS and peripheral organs (15, 17, 18). Previously, we charac-
terized how misfolded amyloid precursor proteins form amyloid
brils in the presence of DNA, RNA, and glycosaminoglycans (19).
The brous aggregates containing nonproteinaceous cofactors
displayed the biophysical and biochemical features of amyloids
obtained in vitro and from patients, the latter of which are well
known to harbor signicant amounts of nucleic acids and/or gly-
cosaminoglycans (20, 21).
Plasmacytoid dendritic cells (pDCs) are a unique innate im-
mune cell population that produces high amounts of IFN-I
(IFNα,-β,-ω, and -τ) upon sensing RNA or DNA by endosomal
TLR7 and TLR9, respectively (22, 23). ICs of autoantibodies to
chromatin and RNPs from SLE patients trigger the production
of IFN-I via activation of pDCs, a process that is mediated by the
Fcɣreceptor (24, 25). DNA-containing neutrophil extracellular
traps (NETs), production of which is accelerated by IFN-I and
autoantibodies in the SLE serum, also induce IFN-I production
by pDCs (13, 14). In another autoimmune condition, psoriasis,
complex of antimicrobial peptide LL-37 and self-nucleic acids
stimulates pDCs to secrete IFN-I (26). Given these ndings on
proteinnucleic acid complexes, we examined whether nucleic
acid-containing amyloid brils can activate pDCs to induce IFN-
I and its immunological effects in vivo. Our results suggest that
nucleic acid-containing amyloid brils can function as a potent
IFN-I inducer both in vitro and in vivo. Intriguingly, a healthy
rodent host, in response to these complexes, developed systemic
autoimmunity with features mimicking SLE.
Results
DNA-Containing Amyloid Fibrils Induce Strong IFNα/βProduction by
pDCs. Two prototypic amyloidogenic peptides, prion fragment and
amyloid βpeptide 142 (Aβ), bind directly to DNA (19, 21). To
test whether DNA-containing prion or Aβbrils can activate
pDCs, we complexed them with oligonucleotide CpG B. CpG B
engages TLR9 in the late endosome and induces pDC to produce
abundant TNFαand IL-6, but little IFN-I (22). Interestingly with
CpG B, both peptides stimulated pDCs to produce elevated levels
of IFNαin a dose-dependent manner, with little effect on other
cytokines (Fig. S1), suggesting that DNA-containing amyloid
brils may selectively enhance IFN-I response by pDCs.
The natural amyloidogenic peptides, such as Aβ, undergo
spontaneous intermolecular rearrangement in solution to gen-
erate miscellaneous misfolding species (27). However, in fact,
Author contributions: J.D.D. and W.C. designed research; J.D.D., S.D.-E., and W.C. per-
formed research; D.G., S.M., P.L., and F.T. contributed new reagents/analytic tools;
M. Gagea, B.Z., L.B., and M. Gilliet analyzed data; and J.D.D. and W.C. wrote the paper.
The authors declare no conict of interest.
*This Direct Submission article had a prearranged editor.
1
To whom correspondence should be addressed. E-mail: wcao@mdanderson.org.
This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10.
1073/pnas.1206923109/-/DCSupplemental.
www.pnas.org/cgi/doi/10.1073/pnas.1206923109 PNAS Early Edition
|
1of6
IMMUNOLOGY
most polypeptides can adopt a native structure or form amyloid
brils by transitioning through a precursor state (16, 28). We have
characterized stabilized amyloid precursors with misfolded struc-
tures from various native proteins (19). Human serum albumin
(HSA), an abundant protein with native globular structure without
apparent immune stimulatory function, was chosen as a model to
investigate the innate immune functions of dened amyloid brils.
For that, four distinct structural variants of HSA were prepared:
native (HSA), amyloid precursor (AP-HSA), amyloid (A-HSA),
and fully denatured (D-HSA). Both native and AP-HSA are sol-
uble, whereas A-HSA and D-HSA are insoluble precipitates. A-
HSA readily bound to amyloid-specic dye Congo Red, indicating
the presence of β-sheet rich amyloid brils (Fig. 1A). AP-HSA, the
amyloid precursor species, which interacted mildly with Congo
Red (19), formed an insoluble precipitate upon mixing with CpG
B, which exhibited signicantly enhanced Congo Red uorescence
(Fig. 1A). This result is consistent with our previous observation
that amyloid precursors convert to amyloid brils upon binding to
DNA in a sequence-independent manner (19).
After overnight culture, AP-HSA complexed with CpG B in-
duced human primary pDCs to secrete signicant levels of IFNα
and slightly increased TNFαand IL-6 (Fig. 1B). This result was
veried by FACS staining on intracellular IFNαassociated with
pDCs and detection of increased IFN-I gene products after
stimulation by AP-HSA complexed with CpG B (Fig. 1 Cand D).
Despite the strong IFN-I stimulation, AP-HSA did not affect pDC
maturation by CpG B (Fig. 1Dand Fig. S2). In contrast, neither A-
HSA nor D-HSA, two other misfolded variants, affected IFN-I
production (Fig. 1 Band C). Because pDCs account for <1% of
the mononuclear cells in the blood (23), we investigated whether
CpG-containing HSA amyloid brils could selectively activate
pDCs amid other leukocytes. AP-HSA in the presence of CpG B
stimulated prominent and selective IFNαsecretion from PBMCs,
which depended on the function of pDCs as IFN production was
abrogated by selective pDC depletion (Fig. 1E).
AP-HSA readily binds to genomic DNA isolated from salmon
sperm, which resulted in the generation of Congo Red positive
complex that displayed apple-green birefringence under polar-
ized light, a denitive indication of amyloid formation (Fig. 1F).
pDCs secreted signicant levels of IFNαafter stimulation by
HSA amyloid containing salmon sperm DNA (Fig. 1F, Lower)
and similarly by amyloid containing human genomic DNA (Fig.
S3). Therefore, our data indicate that DNA-containing amyloid
brils potently induce IFN-I by activating human pDCs.
Amyloid Fibrils Containing DNA Are Required for IFN-I Induction.
Under our experimental condition, the stabilized amyloid precursor
forms amyloid brils by complexing with DNA (19); however, DNA
and AP-HSAmay trigger separate signaling pathways in pDCs that
synergistically heighten the IFN-I response. To examine this pos-
sibility, we cultured pDCs with DNA and AP-HSA sequentially:
pDCs were cultured with DNA for 2 h, the cells were washed, and
then AP-HSA was added; in a second test, the cells were cultured
rst with AP-HSA then incubated with DNA. pDCs produced
IFNαonly in the presence of both DNA and AP-HSA, a condition
favoring the generation of DNA-containing amyloid brils (Fig.
S4A). Furthermore, uorescent staining with the amyloid-specic
dye thioavin S revealed the presence of amyloid aggregates inside
the pDC cells after exposure to AP-HSA complexed with DNA, but
not to the native HSA-DNA mixture (Fig. S4B).
In addition to nucleic acids, amyloid precursor proteins bind to
other polyanionic cofactors, such as heparan sulfate glycosamino-
glycan, and form amyloid brils (19). In contrast to DNA-con-
taining amyloid brils, AP-HSA mixed with heparin failed to
activate pDCs to induce IFN-I (Fig. S4C). Moreover, heparin
inhibited the production of IFNαby PBMCs in response to CpG-
containing HSA amyloid brils (Fig. S4D), which is consistent with
its ability to compete with the formation of DNA-containing HSA
amyloid brils (19). Therefore, among the different types of amy-
loid brils, only the nucleic acid-containing aggregates can potently
induce IFN-I. A small compound polyphenol(-)-epigallocatechin
A
IFNαIL-6TNFα (pg /ml)(pg/ml)(pg/ml)
CpG B
B
C IL-6 CD86 CD80
6hr 20hr 6hr 20hr 6hr 20hr
Relative Expression
AP-HSA
HSA
IFN 1 IFN 4 IFN
6hr 20hr 6hr 20hr 6hr 20hr
D
E
AP-HSA AP-HSA + DNA
F
med
HSA
AP-HSA
A-HSA
D-HSA
IFN-α
TNF-α
No CpG B
Med
HSA
AP-HSA
A-HSA
D-HSA
6
23
54
40
42
42
1
4
2
24
1
1
CpG B
Fig. 1. DNA-containing amyloid induces prominent IFN-I production by pDCs. (A) Detection of β-sheetrich structures in the HSA structural variants in the
absence or presence of CpG B with Congo Red. The intensity of uorescent emission at 646 nm was plotted (mean ±SD from three independent experiments).
*P<0.05. (Band C) Cytokine production by human pDCs stimulated with 1 μMCpGBwith5μg/mL HSA variants measured by ELISA (B), or by intracellular
FACS staining (C). Data shown in Bare the mean ±SEM from a representative donor (n>10). Numbers in Cindicate the percentage of population (rep-
resentative of four donors). (D) Gene expression by pDCs after stimulation determined by quantitative PCR analysis (levels are relative to that of a resting
PBMC sample). (E) Cytokine production by human PBMC cultured with HSA variants (10 μg/mL) with or without CpG B. Data shown are the mean ±SEM from
a representative donor (n=4). (F) Birefringence analysis of AP-HSA in without or with salmon sperm DNA (Upper). IFNαsecreted from pDCs cultured with HSA
or AP-HSA (5 μg/mL) and salmon sperm DNA (Lower) (mean ±SD).
2of6
|
www.pnas.org/cgi/doi/10.1073/pnas.1206923109 Di Domizio et al.
gallate (EGCG) selectively binds to amyloid precursors and
interferes with the amyloid formation (29). Preincubation of AP-
HSA with EGCG signicantly reduced levels of IFNαproduced by
PBMCs stimulated by CpG-containing HSA amyloid brils (Fig.
S4E). These results illustrate the functional importance of bril
formation for DNA-containing amyloid to induce IFN-I.
DNA-Containing Amyloid Uptake by pDCs Activates TLR9 in Early
Endosomes. To understand how IFN-I production is selectively
enhanced by DNA-containing amyloid brils, we examined the
uptake of genomic DNAs by pDCs. In contrast to native HSA,
AP-HSA signicantly increased the amount of DNA associated
with pDCs in a dose-dependent manner (Fig. 2A). The uptake of
DNA-containing amyloid seems to be cell type independent,
because effective internalization of DNA-containing amyloid
brils was also observed by Jurkat cells (Fig. S5).
Because TLR signaling from different endosomal compart-
ments leads to distinct cytokine response by pDCs (22), we then
examined the subcellular localization of AP-HSA complexed with
DNA in pDCs by costaining the cells with the early endosomal
marker EEA1 (Fig. 2B) and the late endosome/lysosome marker
LAMP1 (Fig. S6). After 4 h in culture, uorescent AP-HSA and
DNA were detected inside pDCs, where they remained tightly
bound, as illustrated by the complete colocalization of their sig-
nals (Fig. 2B). Interestingly, the DNAamyloid complex showed
signicant colocalization with EEA1 but not with LAMP1, sug-
gesting the exclusive early endosome localization of the amyloid
brils. Similarly, both Aβand prion peptides facilitate CpG B
to early ensosome (Fig. S7), which is likely responsible for the
enhanced IFN-I response (Fig. S1).
TLR9 recognizes the unmethylated CpG motifs present in the
2deoxyribose backbone of natural DNA (22, 23). In addition to
TLR9, other cellular DNA sensors can participate in IFN-I re-
sponse by recognizing biochemical features of DNA irrespective
of unmethylated CpGs (30). To investigate the specic role of
TLR9, we enzymatically methylated two DNA species, i.e.,
plasmid DNA and human genomic DNA, and prepared amyloid
brils by complexing them with AP-HSA. After methylation,
plasmid DNA obtained from a bacterial source became resistant
to digestion by the restriction enzyme BstU1, which recognizes
unmethylated CGCG sequences, whereas human DNA that
contains low-frequency unmethylated CpGs showed enhanced
BstU1 resistance (Fig. 2C, Left). When added to pDCs, amyloid
containing methylated DNA completely lost its ability to induce
IFN-I, demonstrating the requirement of unmethylated CpGs to
trigger IFN production (Fig. 2C, Right). Therefore, our data
collectively reveal that DNA as part of the complex amyloid is
effectively taken up by pDCs and delivered to early endosomes
to potently trigger TLR9-mediated IFN-I production.
Nucleic Acid-Containing Amyloid Fibrils Mediate IFN-I Response to
Self-RNA and Dead Cell Debris. Similar to their interaction with
DNA, AP-HSA mixed with total cellular RNA produced in-
soluble high molecular aggregate with enhanced Congo Red
emission, bril formation (19), and retarded migration during
electrophoresis (Fig. 3A). Within this complex, RNA became
resistant to RNase digestion, indicating a protective effect of
the amyloid structure to the complexed nucleic acids. When
added to pDCs, the RNA-containing amyloid brils induced
signicant levels of IFNα(Fig. 3B).
Upon death, cells release their cellular components and nu-
clear antigens. Preincubation of AP-HSA, but not other forms of
HSA, with the lysates of necrotic cells stimulated puried pDCs
to secrete IFNα(Fig. 3C). Consistently, IFNαproduction was
detected in PBMCs stimulated by AP-HSA complexed with the
cell debris (Fig. 3D). Such activation was sensitive to the pre-
treatment of the lysates with DNase and RNase, suggesting that
DNA- and RNA-containing amyloid brils formed in the mixture
are likely responsible for triggering IFN-I secretion.
Because IFN-inducing ICs implicated in SLE rely on the func-
tion of FcγR to mediate their cellular entry (5, 22), we investigated
whether DNA-containing amyloid brils also use surface FcγRIIa
(CD32) on pDCs. Although blocking CD32 signicantly reduced
the amount of internalized DNA and secreted IFNαinduced
by SLE serum, it had no effect on either the uptake of DNA-
containing amyloid brils or the amount of IFNαsecreted by
pDCs after amyloid stimulation (Fig. 3 Eand F). Overall, our
results demonstrate that amyloid brils containing self-DNA,
self-RNA, and dead cell debris can directly activate pDCs to
produce IFN-I.
DNA-Containing Amyloid Fibrils Induce Inltration of pDCs and IFNα/β
Production in Vivo. To examine the function of nucleic acid-con-
taining amyloid brils in an in vivo tissue environment, we injected
mixtures of native HSA or AP-HSA together with endotoxin-free
bacterial DNA into the peritoneal cavities of mice. pDCs inl-
trated to the site where DNA-containing amyloid brils were in-
oculated and retained locally for days afterward (Fig. 4A). In
contrast, few pDCs were found in the peritoneal cavities of mice
injected with DNA and/or HSA. An elevated number of DCs and
macrophages were also detected, but no signicant difference
found between the groups receiving HSA/DNA or AP-HSA/DNA.
Hence, pDCs selectively inltrate in vivo in response to DNA-
containing amyloid brils. We next analyzed the gene expression
by the peritoneal exudate cells 24 h after injection. Strikingly, in-
oculation of DNAAP-HSA complex triggered the transcription of
multiple IFN-I subtypes and a group of IFN inducible genes at
levels signicantly higher than other treatments (Fig. 4B).
To examine the functional involvement, we depleted pDCs by
injecting mAb 120G8, an antibody recognizing the mouse pDC-
specic receptor BST2 (31), i.p. 24 h before amyloid inoculation.
AP-HSAHSA
A
DNAAP-HSA EEA1
DNA
EEA1
EEA1
AP-HSA AP-HSA
DNA EEA1
DNAAP-HSA
B
plasmid genomic DNA
-+
methylaon -+
IFNα (pg/ml)
BstU1
digeson
-+
methylaon -+
plasmid genomic DNA
AP-HSA + + + +
C
Fig. 2. DNA-containing amyloid brils are endocytosed into pDCs to activate
TLR9. (A) Uptake of DNA by pDCs after incubation with HSA or AP-HSA and
Alexa 647-labeled salmon sperm DNA. (B) Confocal analysis of pDCs con-
taining biotinylated AP-HSA (green) and A647-labeled DNA (red). Cells were
also stained for the early endosome marker EEA1 (blue). (CLeft) Plasmid
DNA and human genomic DNA, unmodied or treated with CpG methyl-
transferase, were digested by BstU1. Shown is the separation of DNA fragments
together with a DNA ladder (center lane). (CRight)IFNαsecretion by human
pDCs stimulated by AP-HSA complexed with unmodied or methylated DNA.
Shown are representative results from at least three different donors.
Di Domizio et al. PNAS Early Edition
|
3of6
IMMUNOLOGY
Preinjection of 120G8, but not isotype-matched control, reduced
the inltrating pDCs by more than 90% in the peritoneal cavity
after DNAAP-HSA complex inoculation (Fig. S8). As a result,
the transcription of not only IFN-I genes but also IFN-inducible
genes induced by amyloid brils was drastically decreased (Fig.
4C). Note that although masked in the heat map, the levels of
irf7 and isg15, two prominent IFN-inducible genes, were reduced
after pDC depletion (Fig. S9). Interestingly, the up-regulated
expression of several chemokines, such as CCL5 and CXCL911,
was unaffected by pDC depletion, suggesting an inammatory
reaction that does not rely on the function of pDCs. Further
analysis revealed an elevated transcription of IL-1βtriggered by
the DNA-containing amyloid brils, which was likewise inde-
pendent of pDCs (Fig. 4C). Overall, our in vivo analysis conrms
the in vitro human study and suggests that pDCs acutely sense and
respond to nucleic acid-containing amyloid in tissues.
necSN
AP-HSA
DNAse I
RNAse H
-
-
-
-
-
+
-
-
-
-
+
-
-
+
-
+
+
-
+
-
+
+
-
-
+
+
+
-
+
+
+
-
-
-
+
+
-
-
+
+
+
+
-
-
-
HSA
+
-
+
+
-
+
-
+
-
+
+
-
+
+
+
+
-
+
-
-
RNase A - + - + - + - + - +
buffer HSA AP-HSA A-HSA D-HSA
total RNA
AB
IFNα
α
(pg/ml)
med
HSA
AP-HSA
A-HSA
D-HSA
necrotic cell supernatan
t
C
DEF
Fig. 3. Amyloid brils containing self-nucleic acids trigger IFN-I production by pDCs. (A) Gel shift analysis of human total RNA mixed with HSA structural
variants in the absence or presence of RNase A. (Band C) Cytokine secreted by pDCs stimulated with HSA proteins in the presence of human total RNA
(B; mean ±SD, four donors) or supernatants of necrotic Jurkat cells (C; mean ±SEM, representative of at least three donors). (D) IFNαsecretion by PBMCs in
response to necrotic Jurkat supernatants in the presence of HSA or AP-HSA (n=11). In some, necrotic supernatants were treated with enzyme before mixing
with HSA. Pvalues were determined by a two-way ANOVA test. **P<0.01. (E) Blocking of CD32 on uptake of DNA by pDCs in the presence of HSA proteins or
sera from healthy donor or SLE patient. Values of mean uorescent intensity (MFI) are shown. (F) IFNαproduced by pDCs in the presence of anti-CD32
blocking antibody. (Eand F) Data are presented as mean ±SD (n=4).
DNA
PBS PBS HSA AP-HSA
ifna1
ifna4
ifna6
ifnb
mx1
mx2
isg15
isg20
irf7
ifit1
ifi202
cxcl9
cxcl10
cxcl11
Type I IFN
genes
IFN-
response
genes
-1 1.5 10+
B
DNA + HSA DNA + AP-HSA
rat IgG 120G8 rat IgG 120G8
ifna1
ifna4
ifna6
ifnb
mx1
mx2
irf7
isg15
isg20
ifit1
ifi202
ccl5
cxcl9
cxcl10
cxcl11
il-1a
il-1b
il-18
C
A
120G8
B220
PBS DNA DNA + HSA DNA + AP-HSA
24 hrs
93 502 600 4121
Fig. 4. DNA-containing amyloid induces pDC-mediated IFN-I response in vivo. (A) Number of inltrating antigen presenting cells in the peritoneum of mice.
Shown at Top are proles of pDCs within CD11c
+
MHC-II
+
CD11b
population 24 h after i.p. injection. Quantication of kinetic inltration is shown at Bottom
(mean ±SD, four mice per time point). (B) Peritoneal gene expression presented as a heat map. A PBS-treated animal was used as a reference. (C) Gene
expression by peritoneal cells 24 h after i.p. injection of HSA proteins with DNA in mice received pretreatment of antibodies (Band C) Shown are results from
one experiment of at least two independent experiments with similar results.
4of6
|
www.pnas.org/cgi/doi/10.1073/pnas.1206923109 Di Domizio et al.
Immunization of DNA-Containing Amyloid Fibrils Induces Autoantibodies
and Proteinuria in Healthy Mice. To evaluate the long-term immune
response by a healthy host to nucleic acid-containing amyloid
brils, we immunized wild-type female BALB/c mice with such
amyloid in comparison with PBS, DNA, and DNA mixed with
native protein. The mice rst received i.p. injection in the presence
of complete Freunds adjuvant (CFA), followed at 2-wk intervals
by two boost i.p. injections with IFA. No difference was detected in
the total IgM and IgG levels between the experimental groups
(Fig. S10). However, the sera from the mice that received DNA-
containing HSA amyloid, but not from the other groups, displayed
strong ANA staining onHep-2 cells (Fig. 5A). Clearly visible in the
nuclei of Hep-2 cells, the staining was also positive in the cyto-
plasm, implying a broad reactivity toward cellular antigens.
To elucidate the specicity of the ANA, we examined sera re-
activity to several well-known autoantigens implicated in SLE. In-
terestingly, mice that received DNA-containing HSA amyloid
developed signicant antibody responses against single-stranded
DNA (ssDNA), total RNA, Sm/RNP complex, and histone over
a period of several months (Fig. 5B). Further analysis demonstrated
that IgG1 and, to a lesser extent, IgG2a were the major Ig isotypes
within the anti-ssDNA response (Fig. S11). Consistent with the
negative ANA, control animals and mice immunized with DNA
and HSA showed no sign of autoantibody. The autoantibodies in-
duced by DNA-HSA amyloid are independent of the Abs against
HSA, because depletion of HSA-specic Ig had no impact on the
serum reactivity to the autoantigens (Fig. S12). At the time when
the mice were terminated 16 wk after immunization, none of sera
reacted with double-strand DNA. Because lupus nephritis is a ma-
jor organ-specic pathology associated with SLE, we analyzed the
renal function of the immunized mice and detected proteinuria in
the group that received DNA-containing HSA amyloid (Fig. 5C).
Furthermore, the deposition of IgG was found in the glomeruli of
the kidneys from these mice (Fig. 5D).
Because pDC depletion resulted in diminished acute IFN-I
response (Fig. 4C), we next investigated the role of pDCs in
antibody development. Strikingly, 120G8 preinjection largely
abolished the ANA response and severely affected the generation
of specic autoantibodies induced by DNA-containing amyloid
(Fig. 5 Eand F). However, it did not affect the titer of anti-HSA
antibody or the proteinuria in the immunized mice (Fig. S13).
This nding suggests that pDC-IFN axis strongly inuence the
immune reactions leading to autoantibody development. Overall,
these data collectively demonstrate that exposure of nucleic acid-
containing amyloid brils to a nonautoimmune host can result in
the development of lupus-like systemic autoimmunity.
Discussion
By forming brous aggregates with amyloid precursor proteins,
self-nucleic acids are protected from nucleases in the environ-
ment, effectively taken up by pDCs, and then transported to the
endocytic compartment. A unique membrane trafcking pathway
with characteristics of endolysosomes are essential for TLR7/9
signaling and IFN production in pDCs (22, 32, 33). The nucleic
acid-containing amyloid is retained in the early endosomes of
pDCs, where the prolonged TLR9 activation can promote MyD88
signaling and subsequent IRF7 activation, which initiates the
transcription of all IFN-I subtypes. This mechanism is analogous
to other potent IFN-I inducers, i.e., type A CpG oligonucleotide
and LL-37 complexed with nucleic acids (22, 26). It is unclear
whether any specic pDC surface receptor mediates this process.
Amyloid βbrils effectively attach to cells by interacting with
a wide array of surface receptors and directly with the phospho-
lipid bilayer (34). Interestingly, multiple amyloidgenic peptides
PBS DNA
DNA + HSA DNA + AP-HSA
Hep-2 cells
A
D
DNA + HSA DNA + AP-HSA
rat IgG
injected
120G8
injected
Immunization
E
BF
C
PBS DNA
DNA + HSA DNA + AP-HSA
Fig. 5. BALB/c mice develop lupus-like autoimmunity after immunization with DNA-containing amyloid. (A) ANA reactivity from sera of mice 13 wk after
immunization. (B) Levels of autoantibodies in the sera of the immunized mice determined by ELISA analysis (mean ±SD, ve mice per group). (C) Levels of
albumin in the urine of immunized mice (mean ±SD, ve mice per group). (D) Detection of IgG in the kidneys from immunized mice by staining with A488-
labeled anti-mouse IgG. (Aand D) Shown are results with a representative mouse (n=5). (AD) Shown are results from one experiment of two independent
experiments with similar results (812 mice per group in combination). ANA response (E) and levels of antigen-specic autoantibodies (F) from mice that
received Ab pretreatment. Hep-2 cells were stained with a representative serum 9 wk after immunization (E). The box and whiskers plots of the data dis-
tribution of four mice per group are shown in F.Pvalues were determined by a two-way ANOVA test.
Di Domizio et al. PNAS Early Edition
|
5of6
IMMUNOLOGY
enhance HIV attachment and entry into cells (35). Therefore,
nucleic acid-containing amyloid brils are unusually effective to
deliver nucleic acids to elicit IFN-I production by pDCs.
Despite the recognized importance of IFN-I in many autoim-
mune diseases, its role in the initiation phase of autoimmunity has
not been fully dened. In fact, only a minor fraction of patients
treated with IFNαdevelop ANA, and even a smaller fraction
manifest with SLE (5). Distinct from human SLE, mice that develop
spontaneous lupus do not exhibit signicant upregulation of IFN-I.
Excessive IFN-I exposure exacerbates disease only in certain lupus-
prone strains but has no effect in nonautoimmune mice (6), implying
that IFN-I requires certain genetic susceptibility or perhaps activa-
tion of additional pathway(s) to break immune tolerance. Here, we
demonstrate the capacity of nucleic acid-containing amyloid to in-
duce early IFN-I production upstream in a cascade of immune
responses, which eventually lead to the autoantibody generation.
Aberrant IFN-I production by pDCs has been implicated in
several human autoimmune disorders (4, 23). In SLE patients, the
numbers of circulating pDCs are reduced, whereas increased pDC
presence has been observed in the inamed tissues (5, 22). Besides
secreting IFN-I, TLR-activated pDCs promote the generation of
plasma cells and antibody responses via IFN and IL-6 in vitro (10).
However, how pDCs participate in systemic autoimmunity in vivo
remains obscure. Here, we show the inltration of pDCs shortly
after inoculation of nucleic acid-containing amyloid. Depletion of
pDCs not only abolished the IFN-I induction, but also severely and
selectively diminished the development of autoantibodies against
nuclear antigens. Therefore, IFN-producing pDCs play an essential
role in initiating systemic autoimmunity.
Amyloid brils are a product of failed protein homeostasis be-
cause of germ-line mutation, erroneous transcription/translation,
physical damage, or abnormal posttranslational processing (15).
Because human amylomeconstitutes approximately 15% of
all coding polypeptides in the genome, many selfproteins have
the potential to form amyloid (28). Amyloid depositions are fre-
quently heterogeneous containing nonproteinaceous cofactors
(15, 20, 21). Our results suggest that only the type of amyloid-
containing nucleic acids is capable of inducing IFN-I through
activating nucleic acid-sensing TLRs. Interestingly, protein
misfolding products display another innate immune function: both
brillar Aβand amyloid precursor of islet amyloid polypeptide
potently activate NALP3 inammasome and induce IL-1βmatu-
ration (17, 18). We also observed that DNA-containing amyloid
brils induced peritoneal inammation in a pDC- and IFN-
independent manner, likely due to inammasome activation
and IL-1βinduction. Therefore, the protein misfolding products
can activate multiple innate immune pathways in vivo.
By immunizing nucleic acid-containing amyloid brils, we have,
in effect, created an inducible experimental lupus model. Previous
attempts to immunize nonautoimmune mice with self-antigens,
such as DNA, apoptotic cells, or puried nucleosome, only re-
sulted in limited or transient autoantibody generation (3638).
Tetramethylpentadecane (TMPD) induces an array of auto-
antibodies and glomerulonephritis in BALB/c mice (39). In-
terestingly, prolonged oral administration of TMPD reportedly
leads to amyloidosis (40). Our model of experimental lupus
uniquely centers on the activation of pDC-IFN axis. It would be
important to investigate the critical cellular players and pathways
that lead to systemic autoimmunity.
Materials and Methods
Reagents. HSA structural variant proteins were prepared essentially as de-
scribed (19). Wi ld-ty pe BALB /cByJ m ice were obtained from the Jackson
Laboratory. Additional methods and detailed information can be found
in SI Materials and Methods.
ACKNOWLEDGMENTS. We thank Cametria Thompson, Ming Zhuo, and Ran
Zhang for technical assistance, and Drs. Yong-Jun Liu, Stephanie Watowich,
and Shao-Cong Sun for valuable suggestions. This research is supported by
the University of Texas MD Anderson Cancer Center Institutional Research
Grant (to W.C.), National Institutes of Health (NIH) Grant AI074809 (to W.C.),
and NIH MD Andersons Cancer Center Support Grant CA016672.
1. Cho JH, Gregersen PK (2011) Genomics and the multifactorial nature of human au-
toimmune disease. N Engl J Med 365:16121623.
2. Hahn BH (1998) Antibodies to DNA. N Engl J Med 338:13591368.
3. Theolopoulos AN, Baccala R, Beutler B, Kono DH (2005) Type I interferons (α/β)in
immunity and autoimmunity. Annu Rev Immunol 23:307336.
4. Banchereau J, Pascual V (2006) Type I interferon in systemic lupus erythematosus and
other autoimmune diseases. Immunity 25:383392.
5. Rönnblom L, Pascual V (2008) The innate immune system in SLE: Type I interferons
and dendritic cells. Lupus 17:394399.
6. Mathian A, Weinberg A, Gallegos M, Banchereau J, Koutouzov S (2005) IFN-αinduces
early lethal lupus in preautoimmune but not in BALB/c mice. J Immunol 174:24992506.
7. Santiago-Raber M-L, et al. (2003) Type-I interferon receptor deciency reduces lupus-
like disease in NZB mice. J Exp Med 197:777788.
8. Agrawal H, et al. (2009) Deciency of type I IFN receptor in lupus-prone New Zealand
mixed 2328 mice decreases dendritic cell numbers and activation and protects from
disease. J Immunol 183:60216029.
9. Blanco P, Palucka AK, Gill M, Pascual V, Banchereau J (2001) Induction of dendritic cell
differentiation by IFN-alpha in systemic lupus erythematosus. Science 294:15401543.
10. Jego G, et al. (2003) Plasmacytoid dendritic cells induce plasma cell differentiation
through type I interferon and interleukin 6. Immunity 19:225234.
11. Le Bon A, et al. (2001) Type i interferons potently enhance humoral immunity and can
promote isotype switching by stimulating dendritic cells in vivo. Immunity 14:461470.
12. Tough DF (2012) Modulation of T-cell function by type I interferon. Immunol Cell Biol
90:492497.
13. Garcia-Romo GS, et al. (2011) Netting neutrophils are major inducers of type I IFN
production in pediatric systemic lupus erythematosus. Sci Transl Med 3:73ra20.
14. Lande R, et al. (2011) Neutrophils activate plasmacytoid dendritic cells by releasing self-
DNA-peptide complexes in systemic lupus erythematosus. Sci Transl Med 3:73ra19.
15. Sideras K, Gertz MA (2009) Amyloidosis. Adv Clin Chem 47:144.
16. Schnabel J (2010) Protein folding: The dark side of proteins. Nature 464:828829.
17. Halle A, et al. (2008) The NALP3 inammasome is involved in the innate immune
response to amyloid-β.Nat Immunol 9:857865.
18. Masters SL, et al. (2010) Activation of the NLRP3 inammasome by islet amyloid polypeptide
provides a mechanism for enhanced IL-1βin type 2 diabetes. Nat Immunol 11:897904.
19. Di Domizio J , et al. (2012) Binding with nucleic acids or glycosaminoglycans converts
soluble protein oligomers to amyloid. J Biol Chem 287:736747.
20. Zhang X, Li J-P, Lijuan Z (2010) Heparan sulfate proteoglycans in amyloidosis. Prog
Mol Biol Transl Sci 93:309334.
21. Jiménez JS (2010) Protein-DNA interaction at the origin of neurological diseases:
A hypothesis. J Alzheimers Dis 22:375391.
22. Gilliet M, Cao W, Liu Y-J (2008) Plasmacytoid dendritic cells: Sensing nucleic acids in
viral infection and autoimmune diseases. Nat Rev Immunol 8:594606.
23. Colonna M, Trinchieri G, Liu YJ (2004) Plasmacytoid dendritic cells in immunity. Nat
Immunol 5:12191226.
24. Vollmer J, et al. (2005) Immune stimulation mediated by autoantigen binding sites
within small nuclear RNAs i nvolves Toll-like recep tors 7 and 8. J ExpMed 202 :15751585.
25. Means TK, et al. (2005) Human lupus autoantibody-DNA complexes activate DCs
through cooperation of CD32 and TLR9. J Clin Invest 115:407417.
26. Lande R, et al. (2007) Plasmacytoid dendritic cells sense self-DNA coupled with anti-
microbial peptide. Nature 449:564569.
27. Stefani M (2010) Biochemical and biophysical features of both oligomer/bril and cell
membrane in amyloid cytotoxicity. FEBS J 277:46024613.
28. Goldschmidt L, Teng PK, Riek R, Eisenberg D (2010) Identifying the amylome, proteins
capable of forming amyloid-like brils. Proc Natl Acad Sci USA 107:34873492.
29. Ehrnhoefer DE, et al. (2008) EGCG redirects amyloidogenic polypeptides into un-
structured, off-pathway oligomers. Nat Struct Mol Biol 15:558566.
30. Kumar H, Kawai T, Akira S (2011) Pathogen recognition by the innate immune system.
Int Rev Immunol 30:1634.
31. Asselin-Paturel C, Brizard G,Pin J-J, Brière F, Trinchieri G (2003) Mouse strain differences
in plasmacytoid dendritic cell frequency and function revealed by a novel monoclonal
antibody. J Immunol 171:64666477.
32. Blasius AL, et al. (2010) Slc15a4, AP-3, and Hermansky-Pudlak syndrome proteins are
required for Toll-like receptor signaling in plasmacytoid dendritic cells. Proc Natl Acad
Sci USA 107:1997319978.
33. Sasai M, Linehan MM, Iwasaki A (2010) Bifurcation of Toll-like receptor 9 signaling by
adaptor protein 3. Science 329:15301534.
34. Williams TL, Serpell LC (2011) Membrane and surface interactions of AlzheimersAβ
peptideinsights into the mechanism of cytotoxicity. FEBS J 278:39053917.
35. Roan NR, et al. (2011) Peptides released by physiological cleavage of semen coagulum
proteins form amyloids that enhance HIV infection. Cell Host Microbe 10:541550.
36. Pisetsky DS (1996) Immune activation by bacterial DNA: A new genetic code. Immu-
nity 5:303310.
37. Mevorach D, Zhou JL, Song X, Elkon KB (1998) Systemic exposure to irradiated apo-
ptotic cells induces autoantibody production. J Exp Med 188:387392.
38. Urbonaviciute V, et al. (2008) Induction of inammatory and immune responses by HMGB1-
nucleosome complexes: Implications for the pathogenesis of SLE. JExpMed205:30073018.
39. Reeves WH, Lee PY, Weinstein JS, Satoh M, Lu L (2009) Induction of autoimmunity by
pristane and other naturally occurring hydrocarbons. Trends Immunol 30:455464.
40. Ho FC, Fu KH (1987) A new model of AA-amyloidosis induced by oral pristane in BALB/c
mice. BrJExpPathol68:413420.
6of6
|
www.pnas.org/cgi/doi/10.1073/pnas.1206923109 Di Domizio et al.
... The lines of demarcation between amyloids and AMPs have blurred in the last 10 years; however, it is now clear that AMPs can be autoantigens. AMPs and amyloids can adopt similar structures and biophysical properties, and both can self-assemble with immune ligands like DNA to amplify immune responses [28,82,83]. Although AMPs and bacterial amyloids have been implicated in the pathogenesis of autoimmune diseases like SLE, psoriasis, rheumatoid arthritis [5,[9][10][11][12][13], recent work has shown that many amyloids possess antimicrobial activity, suggesting a potential role in host defense [84]. ...
... Importantly, even in the protease-rich environment of the gut where only fragments of TcdA exist, the TcdA transduction domain alone can organize DNA into complexes capable of strong multivalent TLR9 activation. Consistent with these results, Di Domizio et al. showed that artificially formed amyloid-DNA complexes administered systematically promote systematic autoimmunity, autoantibody production, and lupus-like syndrome in mice through TLR9 signaling in plasmacytoid dendritic cells [83,90]. In sum, amyloids and AMPs can both organize and chaperone immune ligands into supramolecular structures with optimized geometries that promote multivalent binding to toll-like receptors and thereby amplify immune activation. ...
Article
Full-text available
Bacteria are microscopic, single-celled organisms known for their ability to adapt to their environment. In response to stressful environmental conditions or in the presence of a contact surface, they commonly form multicellular aggregates called biofilms. Biofilms form on various abiotic or biotic surfaces through a dynamic stepwise process involving adhesion, growth, and extracellular matrix production. Biofilms develop on tissues as well as on implanted devices during infections, providing the bacteria with a mechanism for survival under harsh conditions including targeting by the immune system and antimicrobial therapy. Like pathogenic bacteria, members of the human microbiota can form biofilms. Biofilms formed by enteric bacteria contribute to several human diseases including autoimmune diseases and cancer. However, until recently the interactions of immune cells with biofilms had been mostly uncharacterized. Here, we will discuss how components of the enteric biofilm produced in vivo, specifically amyloid curli and extracellular DNA, could be interacting with the host's immune system causing an unpredicted immune response.
... The subunits of HU and IHF have a highly conserved sequence homology (15). As human immune system is considered to be exposed to biofilm components throughout life and as DNA interaction with protein antigens is reported to induce autoimmune disorders (6,17), we investigated whether DNABII proteins are involved in the induction of autoimmunity. In the present study, we had demonstrated that a conserved peptide derived from DNABII proteins specifically recognized by sera from patients with SLE, especially in those with lupus nephritis (LN). ...
... The formation of aggregated and condensed structures by several proteins incorporated with DNA is documented to trigger the production of type I interferon and is involved the pathogenesis of autoimmune diseases (6,17,25). The antimicrobial peptide, LL37, binds DNA to form a coil-node structure and subsequently mediates pDC activation and drives autoimmunity in psoriasis (27)(28)(29)(30). ...
Article
Full-text available
Systemic lupus erythematosus (SLE) is a systemic autoimmune disease with multiple autoantibody production and often affects the kidneys, known as lupus nephritis. However, the mechanism underlying lupus nephritis development is unclear. Biofilms that protect bacteria from stress are ubiquitous in almost every environment. Here, we identified that a conserved peptide (HU1) derived from DNABII proteins, one of major bacterial biofilm components, was specifically recognized by sera from about 47% patients with SLE. Moreover, the serum anti-HU1 levels showed a significant positive correlation with lupus nephritis occurrence. Presence of antibodies against HU1 in pristane-induced mice aggravated lupus nephritis, although these antibodies also attenuated bacterial biofilm formation. We further identified that antibodies against HU1 cross-recognized protein disulfide isomerase (P4HB) located on the renal cell surface and inhibited the activities of this enzyme. Our findings reveal a novel mechanism underlying the development of lupus nephritis triggered by bacterial biofilms.
... 405 In addition, the damaged host cells released nucleic acids containing Aβ aggregates, which were immunogenic and elicited the secretion of type I interferons (IFNs) by adjacent microglia to accomplish the antiviral response. 406 The produced interferon-γ (IFN-γ) further facilitated Aβ generation to form a positive feedback loop. 407 Similar to anti-viral activity, Aβ peptides also bound to fungal cells and stimulated the phagocytosis of microglia. ...
Article
Full-text available
Amyloid β protein (Aβ) is the main component of neuritic plaques in Alzheimer's disease (AD), and its accumulation has been considered as the molecular driver of Alzheimer's pathogenesis and progression. Aβ has been the prime target for the development of AD therapy. However, the repeated failures of Aβ-targeted clinical trials have cast considerable doubt on the amyloid cascade hypothesis and whether the development of Alzheimer's drug has followed the correct course. However, the recent successes of Aβ targeted trials have assuaged those doubts. In this review, we discussed the evolution of the amyloid cascade hypothesis over the last 30 years and summarized its application in Alzheimer's diagnosis and modification. In particular, we extensively discussed the pitfalls, promises and important unanswered questions regarding the current anti-Aβ therapy, as well as strategies for further study and development of more feasible Aβ-targeted approaches in the optimization of AD prevention and treatment.
... Aβ fibrils with bound nucleic acids (that can originate either from viral particles of damaged host cells) are highly immunogenic and elicit robust type I interferon secretion by adjacent microglia [175,176], which promotes their antiviral response. Activated microglia or other cells infiltrating the infected brain can also produce interferon-γ (IFN-γ) [177,178], which facilitates Aβ production (see above). ...
Article
Full-text available
The amyloid cascade hypothesis, focusing on pathological proteins aggregation, has so far failed to uncover the root cause of Alzheimer’s disease (AD), or to provide an effective therapy. This traditional paradigm essentially explains a mechanism involved in the development of sporadic AD rather than its cause. The failure of an overwhelming majority of clinical studies (99.6%) demonstrates that a breakthrough in therapy would be difficult if not impossible without understanding the etiology of AD. It becomes more and more apparent that the AD pathology might originate from brain infection. In this review, we discuss a potential role of bacteria, viruses, fungi, and eukaryotic parasites as triggers of AD pathology. We show evidence from the current literature that amyloid beta, traditionally viewed as pathological, actually acts as an antimicrobial peptide, protecting the brain against pathogens. However, in case of a prolonged or excessive activation of a senescent immune system, amyloid beta accumulation and aggregation becomes damaging and supports runaway neurodegenerative processes in AD. This is paralleled by the recent study by Alam and colleagues (2022) who showed that alpha-synuclein, the protein accumulating in synucleinopathies, also plays a critical physiological role in immune reactions and inflammation, showing an unforeseen link between the 2 unrelated classes of neurodegenerative disorders. The multiplication of the amyloid precursor protein gene, recently described by Lee and collegues (2018), and possible reactivation of human endogenous retroviruses by pathogens fits well into the same picture. We discuss these new findings from the viewpoint of the infection hypothesis of AD and offer suggestions for future research.
... Amyloidosis promotes progressive cellular activation by IFN-I in the brain Previously, we observed an age-dependent increase of NA-containing plaques in 5XFAD brains (Roy et al., 2020). NAs, when complexed to amyloid, serve as an immunogenic stimulus that elicits an IFN-I response (Di Domizio et al., 2012). We thus detected microglia with active IFN-I signaling, marked by nuclear Stat1, exclusively near NA + amyloid plaques in young 5XFAD brains. ...
Article
The principal signals that drive memory and cognitive impairment in Alzheimer’s disease (AD) remain elusive. Here, we revealed brain-wide cellular reactions to type I interferon (IFN-I), an innate immune cytokine aberrantly elicited by amyloid β plaques, and examined their role in cognition and neuropathology relevant to AD in a murine amyloidosis model. Using a fate-mapping reporter system to track cellular responses to IFN-I, we detected robust, Aβ-pathology-dependent IFN-I activation in microglia and other cell types. Long-term blockade of IFN-I receptor (IFNAR) rescued both memory and synaptic deficits and resulted in reduced microgliosis, inflammation, and neuritic pathology. Microglia-specific Ifnar1 deletion attenuated the loss of post-synaptic terminals by selective engulfment, whereas neural Ifnar1 deletion restored pre-synaptic terminals and decreased plaque accumulation. Overall, IFN-I signaling represents a critical module within the neuroinflammatory network of AD and prompts concerted cellular states that are detrimental to memory and cognition.
... Amyloidosis promotes progressive cellular activation by IFN-I in the brain Previously, we observed an age-dependent increase of NA-containing plaques in 5XFAD brains (Roy et al., 2020). NAs, when complexed to amyloid, serve as an immunogenic stimulus that elicits an IFN-I response (Di Domizio et al., 2012). We thus detected microglia with active IFN-I signaling, marked by nuclear Stat1, exclusively near NA + amyloid plaques in young 5XFAD brains. ...
Article
Background: Despite well-documented maladaptive neuroinflammation in Alzheimer's disease (AD), the principal signal that drives memory and cognitive impairment remains elusive. Type I interferon (IFN) is an innate immune cytokine aberrantly elicited by β amyloid plaques. Here, we seek to understand its role in cognition and neuropathology relevant to AD. Method: We introduced a genetically-encoded IFN-responsive reporter system into 5XFAD mice, a β-amyloid plaque model, to gauge IFN-stimulated cellular response. We administered an antibody that specifically blocks the signaling of IFN receptor into 5XFAD mice and examined their memory performance, neuropathologies, and gene expression. Moreover, we generated and analyzed 5XFAD mice that lack IFN receptor selectively in microglia or neural cells. Result: Here, we reveal an age-dependent, brain-wide, and profound accrual of brain cells responding to IFN signaling activation in 5XFAD mice. Long-term blockade of IFN receptor rescued both memory and synaptic deficits, and also resulted in reduced microgliosis, inflammation, and neuritic pathology. Interestingly, microglia-specific IFN receptor ablation attenuated the loss of post-synaptic terminals, whereas IFN signaling in neural cells contributed to pre-synaptic alteration and plaque accumulation. Intriguingly, IFN pathway activation displayed a strong inverse correlation with cognitive performance, promoting selective synapse engulfment by microglia rather than amyloid plaques. Conclusion: IFN signaling represents a critical module within the neuroinflammatory network of AD and prompts a concerted cellular state that is detrimental to memory and cognition.
Preprint
Full-text available
Despite well-documented maladaptive neuroinflammation in Alzheimer's disease (AD), the principal signal that drives memory and cognitive impairment remains elusive. Here, we reveal robust, age-dependent cellular reactions to type I interferon (IFN), an innate immune cytokine aberrantly elicited by beta-amyloid plaques, and examine their role in cognition and neuropathology relevant to AD in a murine amyloidosis model. Long-term blockade of IFN receptor rescued both memory and synaptic deficits and resulted in reduced microgliosis, inflammation, and neuritic pathology. Interestingly, microglia-specific IFN receptor ablation attenuated the loss of post-synaptic terminals, whereas IFN signaling in neural cells contributed to pre-synaptic alteration and plaque accumulation. Intriguingly, IFN pathway activation displayed a strong inverse correlation with cognitive performance, promoting selective synapse engulfment by microglia rather than amyloid plaques. Overall, IFN signaling represents a critical module within the neuroinflammatory network of AD and prompts a concerted cellular state that is detrimental to memory and cognition.
Article
Objective: To compare the frequency of rare variants in genes of the pathophysiologically relevant endosomal Toll-like receptor (eTLR) pathway and any quantifiable differences in variant rarity, predicted deleteriousness, or molecular proximity in patients with systemic lupus erythematosus (SLE) and healthy controls. Patients and methods: 65 genes associated with the eTLR pathway were identified by literature search and pathway analysis. Using next generation sequencing techniques, these were compared in two randomised cohorts of patients with SLE (n = 114 and n = 113) with 197 healthy controls. Genetically determined ethnicity was used to normalise minor allele frequencies (MAF) for the identified genetic variants and these were then compared by their frequency: rare (MAF < 0.005), uncommon (MAF 0.005-0.02), and common (MAF >0.02). This was compared to the results for 65 randomly selected genes. Results: Patients with SLE are more likely to carry a rare nonsynonymous variant affecting proteins within the eTLR pathway than healthy controls. Furthermore, individuals with SLE are more likely to have multiple rare variants in this pathway. There were no differences in rarity, Combined Annotation Dependent Depletion (CADD) score, or molecular proximity for rare eTLR pathway variants. Conclusions: Rare non-synonymous variants are enriched in patients with SLE in the eTLR pathway. This supports the hypothesis that SLE arises from several rare variants of relatively large effect rather than many common variants of small effect.
Article
Full-text available
All neurodegenerative diseases feature aggregates, which usually contain disease‐specific diagnostic proteins; non‐protein constituents, however, have rarely been explored. Aggregates from SY5Y‐APPSw neuroblastoma, a cell model of familial Alzheimer's disease, were crosslinked and sequences of linked peptides identified. We constructed a normalized “contactome” comprising 11 subnetworks, centered on 24 high‐connectivity hubs. Remarkably, all 24 are nucleic acid‐binding proteins. This led us to isolate and sequence RNA and DNA from Alzheimer's and control aggregates. RNA fragments were mapped to the human genome by RNA‐seq and DNA by ChIP‐seq. Nearly all aggregate RNA sequences mapped to specific genes, whereas DNA fragments were predominantly intergenic. These nucleic acid mappings are all significantly nonrandom, making an artifactual origin extremely unlikely. RNA (mostly cytoplasmic) exceeded DNA (chiefly nuclear) by twofold to fivefold. RNA fragments recovered from AD tissue were ~1.5‐to 2.5‐fold more abundant than those recovered from control tissue, similar to the increase in protein. Aggregate abundances of specific RNA sequences were strikingly differential between cultured SY5Y‐APPSw glioblastoma cells expressing APOE3 vs. APOE4, consistent with APOE4 competition for E‐box/CLEAR motifs. We identified many G‐quadruplex and viral sequences within RNA and DNA of aggregates, suggesting that sequestration of viral genomes may have driven the evolution of disordered nucleic acid‐binding proteins. After RNA‐interference knockdown of the translational‐procession factor EEF2 to suppress translation in SY5Y‐APPSw cells, the RNA content of aggregates declined by >90%, while reducing protein content by only 30% and altering DNA content by ≤10%. This implies that cotranslational misfolding of nascent proteins may ensnare polysomes into aggregates, accounting for most of their RNA content.
Article
Full-text available
Ample evidence suggests that almost all polypeptides can either adopt a native structure (folded or intrinsically disordered) or form misfolded amyloid fibrils. Soluble protein oligomers exist as an intermediate between these two states, and their cytotoxicity has been implicated in the pathology of multiple human diseases. However, the mechanism by which soluble protein oligomers develop into insoluble amyloid fibrils is not clear, and investigation of this important issue is hindered by the unavailability of stable protein oligomers. Here, we have obtained stabilized protein oligomers generated from common native proteins. These oligomers exert strong cytotoxicity and display a common conformational structure shared with known protein oligomers. They are soluble and remain stable in solution. Intriguingly, the stabilized protein oligomers interact preferentially with both nucleic acids and glycosaminoglycans (GAG), which facilitates their rapid conversion into insoluble amyloid. Concomitantly, binding with nucleic acids or GAG strongly diminished the cytotoxicity of the protein oligomers. EGCG, a small molecule that was previously shown to directly bind to protein oligomers, effectively inhibits the conversion to amyloid. These results indicate that stabilized oligomers of common proteins display characteristics similar to those of disease-associated protein oligomers and represent immediate precursors of less toxic amyloid fibrils. Amyloid conversion is potently expedited by certain physiological factors, such as nucleic acids and GAGs. These findings concur with reports of cofactor involvement with disease-associated amyloid and shed light on potential means to interfere with the pathogenic properties of misfolded proteins.
Article
Full-text available
Alzheimer's disease is the most common form of dementia and its pathological hallmarks include the loss of neurones through cell death, as well as the accumulation of amyloid fibres in the form of extracellular neuritic plaques. Amyloid fibrils are composed of the amyloid-β peptide (Aβ), which is known to assemble to form 'toxic' oligomers that may be central to disease pathology. Aβ is produced by cleavage from the amyloid precursor protein within the transmembrane region, and the cleaved peptide may retain some membrane affinity. It has been shown that Aβ is capable of specifically binding to phospholipid membranes with a relatively high affinity, and that modulation of the composition of the membrane can alter both membrane-amyloid interactions and toxicity. Various biomimetic membrane models have been used (e.g. lipid vesicles in solution and tethered lipid bilayers) to examine the binding and interactions between Aβ and the membrane surfaces, as well as the resulting permeation. Oligomeric Aβ has been observed to bind more avidly to membranes and cause greater permeation than fibrillar Aβ. We review some of the recent advances in studying Aβ-membrane interactions and discuss their implications with respect to understanding the causes of Alzheimer's disease.
Article
Full-text available
Plasmacytoid dendritic cells (pDCs) are important mediators of antiviral immunity through their ability to produce large amounts of type I interferons (IFNs) on viral infection. This function of pDCs is linked to their expression of Toll-like receptor 7 (TLR7) and TLR9, which sense viral nucleic acids within the early endosomes. Exclusion of self nucleic acids from TLR-containing early endosomes normally prevents pDC responses to them. However, in some autoimmune diseases, self nucleic acids can be modified by host factors and gain entrance to pDC endosomes, where they activate TLR signalling. Several pDC receptors negatively regulate type I IFN responses by pDCs during viral infection and for normal homeostasis.
Article
Full-text available
Systemic lupus erythematosus (SLE) is a systemic autoimmune disease characterized by a breakdown of tolerance to nuclear antigens and the development of immune complexes. Genomic approaches have shown that human SLE leukocytes homogeneously express type I interferon (IFN)-induced and neutrophil-related transcripts. Increased production and/or bioavailability of IFN-α and associated alterations in dendritic cell (DC) homeostasis have been linked to lupus pathogenesis. Although neutrophils have long been shown to be associated with lupus, their potential role in disease pathogenesis remains elusive. Here, we show that mature SLE neutrophils are primed in vivo by type I IFN and die upon exposure to SLE-derived anti-ribonucleoprotein antibodies, releasing neutrophil extracellular traps (NETs). SLE NETs contain DNA as well as large amounts of LL37 and HMGB1, neutrophil proteins that facilitate the uptake and recognition of mammalian DNA by plasmacytoid DCs (pDCs). Indeed, SLE NETs activate pDCs to produce high levels of IFN-α in a DNA- and TLR9 (Toll-like receptor 9)-dependent manner. Our results reveal an unsuspected role for neutrophils in SLE pathogenesis and identify a novel link between nucleic acid-recognizing antibodies and type I IFN production in this disease.
Article
Full-text available
Systemic lupus erythematosus (SLE) is a severe and incurable autoimmune disease characterized by chronic activation of plasmacytoid dendritic cells (pDCs) and production of autoantibodies against nuclear self-antigens by hyperreactive B cells. Neutrophils are also implicated in disease pathogenesis; however, the mechanisms involved are unknown. Here, we identified in the sera of SLE patients immunogenic complexes composed of neutrophil-derived antimicrobial peptides and self-DNA. These complexes were produced by activated neutrophils in the form of web-like structures known as neutrophil extracellular traps (NETs) and efficiently triggered innate pDC activation via Toll-like receptor 9 (TLR9). SLE patients were found to develop autoantibodies to both the self-DNA and antimicrobial peptides in NETs, indicating that these complexes could also serve as autoantigens to trigger B cell activation. Circulating neutrophils from SLE patients released more NETs than those from healthy donors; this was further stimulated by the antimicrobial autoantibodies, suggesting a mechanism for the chronic release of immunogenic complexes in SLE. Our data establish a link between neutrophils, pDC activation, and autoimmunity in SLE, providing new potential targets for the treatment of this devastating disease.
Article
Amyloidosis is a heterogeneous group of diseases in which an otherwise normal protein, with or without proteolytic cleavage, forms insoluble amyloid fibrils. These, in turn, deposit in various organs and cause dysfunction. A wide range of diseases are associated with amyloidosis such as Alzheimer's disease, multiple myeloma, other plasma cell disorders, and chronic inflammation, either as a cause, or result, of amyloid production. This heterogeneity in cause and presentation leads to an incomplete understanding of the pathophysiology of amyloid disease. As such, study of this complicated disease process presents significant challenges. The purpose of this review article is to introduce the biochemistry of amyloidosis including ultrastructure analysis, models of monomer aggregation, the importance of the amyloid microenvironment, and the mechanisms of organ dysfunction, including the role of "toxic intermediates." Pathophysiologic analysis of amyloidosis will focus on diagnostic tools as well as the classification of the various forms of amyloidosis. Finally, treatment of amyloidosis will be reviewed including traditional and established modalities. We will also introduce new and novel treatment options as they relate to the basic pathophysiology of this complex and heterogeneous disorder.
Article
Production of type I interferon (IFN-α/β) is a common cellular response to virus infection. IFN-α/β has a dual role in combating infection, triggering innate antiviral mechanisms and stimulating the generation of an adaptive immune response. This review focuses on the effects of IFN-α/β on one particular immune cell type, the T cell, and the impact of IFN-α/β-mediated signalling in T cells on the immune response. The critical role of T-cell responsiveness to IFN-α/β for the generation of productive T-cell responses after infections with certain viruses in vivo is discussed in the context of in vitro experiments investigating the mechanisms by which IFN-α/β modifies T-cell function. These studies reveal complex effects of IFN-α/β on T cells, with the consequences of exposure to IFN-α/β depending on the context of other signals received by the T cell.
Article
Semen serves as a vehicle for HIV and promotes sexual transmission of the virus, which accounts for the majority of new HIV cases. The major component of semen is the coagulum, a viscous structure composed predominantly of spermatozoa and semenogelin proteins. Due to the activity of the semen protease PSA, the coagulum is liquefied and semenogelins are cleaved into smaller fragments. Here, we report that a subset of these semenogelin fragments form amyloid fibrils that greatly enhance HIV infection. Like SEVI, another amyloid fibril previously identified in semen, the semenogelin fibrils exhibit a cationic surface and enhance HIV virion attachment and entry. Whereas semen samples from healthy individuals greatly enhance HIV infection, semenogelin-deficient semen samples from patients with ejaculatory duct obstruction are completely deficient in enhancing activity. Semen thus harbors distinct amyloidogenic peptides derived from different precursor proteins that commonly enhance HIV infection and likely contribute to HIV transmission.
Article
S. Gazal, K. Sacre, Y. Allanore, M. Teruel, A. H. Goodall, , S. Tohma, L. Alfredsson, Y. Okada, G. Xie, A. Constantin, A. Balsa, A. Kawasaki, P. Nicaise, C. Amos, L. Rodriguez-Rodriguez, G. Chiocchia, C. Boileau, J. Zhang, O. Vittecoq, T. Barnetche, M. A. G. Gay, H. Furukawa, A. Cantagrel, X. Le Loet, T. Sumida, M. Hurtado-Nedelec, C. Richez, S. Chollet-Martin, T. Schaeverbeke, B. Combe, L. Khoryati, B. Coustet, J. El-Benna, K. Siminovitch, R. Plenge, L. Padyukov, J. Martin, N. Tsuchiya, P. Dieude. (2015) Identification of secreted phosphoprotein 1 gene as a new rheumatoid arthritis susceptibility gene. Annals of the Rheumatic Diseases 74, e19-e19 CrossRef L.-M. Diaz-Gallo, C. P. Simeon, J. C. Broen, N. Ortego-Centeno, L. Beretta, M. C. Vonk, P. E. Carreira, S. Vargas, J. A. Roman-Ivorra, M. A. Gonzalez-Gay, C. Tolosa, F. J. Lopez-Longo, G. Espinosa, E. F. Vicente, R. Hesselstrand, G. Riemekasten, T. Witte, J. H. W. Distler, A. E. Voskuyl, A. J. Schuerwegh, P. G. Shiels, A. Nordin, L. Padyukov, A.-M. Hoffmann-Vold, R. Scorza, C. Lunardi, P. Airo, J. M. van Laar, N. Hunzelmann, B. S. Gathof, A. Kreuter, A. Herrick, J. Worthington, C. P. Denton, X. Zhou, F. C. Arnett, C. Fonseca, B. P. Koeleman, S. Assasi, T. R. D. J. Radstake, M. D. Mayes, J. Martin, , J. L. Callejas, R. Rios, N. Navarrete, R. G. Portales, M. T. Camps, A. Fernandez-Nebro, M. F. Gonzalez-Escribano, J. Sanchez-Roman, F. J. Garcia-Hernandez, M. J. Castillo, M. A. Aguirre, I. Gomez-Gracia, B. Fernandez-Gutierrez, L. Rodriguez-Rodriguez, J. L. Andreu, P. G. de la Pena, L. Martinez, M. A. Robles, N. Oreiro, S. de Reumatologia, V. Fonollosa, A. Pros, M. R. Carballeira, F. J. Narvaez, B. Diaz, L. Trapiella, M. Gallego, M. del Carmen Freire, I. Vaqueiro, M. V. Egurbide, L. Saez-Comet, F. Diaz, V. Hernandez, E. Beltran. (2013) Implication of IL-2/IL-21 region in systemic sclerosis genetic susceptibility. Annals of the Rheumatic Diseases 72, 1233-1238 CrossRef Lara Bossini-Castillo, Carmen P Simeon, Lorenzo Beretta, Jasper Broen, Madelon C Vonk, José Callejas, Patricia Carreira, Luis Rodríguez-Rodríguez, Rosa García-Portales, Miguel A González-Gay, Ivan Castellví, María Camps, Carlos Tolosa, Esther Vicente-Rabaneda, María Egurbide, , Annemie J Schuerwegh, Roger Hesselstrand, Claudio Lunardi, Jacob M van Laar, Paul Shiels, Ariane Herrick, Jane Worthington, Christopher Denton, Timothy RDJ Radstake, Carmen Fonseca, Javier Martin. (2012) KCNA5 gene is not confirmed as a systemic sclerosis-related pulmonary arterial hypertension genetic susceptibility factor. Arthritis Research & Therapy 14, R273 CrossRef
Article
Microbial infection initiates complex interactions between the pathogen and the host. Pathogens express several signature molecules, known as pathogen-associated molecular patterns (PAMPs), which are essential for survival and pathogenicity. PAMPs are sensed by evolutionarily conserved, germline-encoded host sensors known as pathogen recognition receptors (PRRs). Recognition of PAMPs by PRRs rapidly triggers an array of anti-microbial immune responses through the induction of various inflammatory cytokines, chemokines and type I interferons. These responses also initiate the development of pathogen-specific, long-lasting adaptive immunity through B and T lymphocytes. Several families of PRRs, including Toll-like receptors (TLRs), RIG-I-like receptors (RLRs), NOD-like receptors (NLRs), and DNA receptors (cytosolic sensors for DNA), are known to play a crucial role in host defense. In this review, we comprehensively review the recent progress in the field of PAMP recognition by PRRs and the signaling pathways activated by PRRs.