ArticlePDF Available

Integrative Functional Genomics Identifies Systemic Lupus Erythematosus Causal Genetic Variant in the IRF5 Risk Locus

Wiley
Arthritis & Rheumatology
Authors:

Abstract and Figures

Objective IRF5 plays a crucial role in the development of lupus. Genome‐wide association studies have identified several systemic lupus erythematosus (SLE) risk single‐nucleotide polymorphisms (SNPs) enriched in the IRF5 locus. However, no comprehensive genome editing–based functional analysis exists to establish a direct link between these variants and altered IRF5 expression, particularly for enhancer variants. This study was undertaken to dissect the regulatory function and mechanisms of SLE IRF5 enhancer risk variants and to explore the utilization of clustered regularly interspaced short palindromic repeat interference (CRISPRi) to regulate the expression of disease risk gene to intervene in the disease. Methods Epigenomic profiles and expression quantitative trait locus analysis were applied to prioritize putative functional variants in the IRF5 locus. CRISPR‐mediated deletion, activation, and interference were performed to investigate the genetic function of rs4728142. Allele‐specific chromatin immunoprecipitation–quantitative polymerase chain reaction and allele‐specific formaldehyde‐assisted isolation of regulatory element–quantitative polymerase chain reaction were used to decipher the mechanism of alleles differentially regulating IRF5 expression. The CRISPRi approach was used to evaluate the intervention effect in monocytes from SLE patients. Results SLE risk SNP rs4728142 was located in an enhancer region, indicating a disease‐related regulatory function, and risk allele rs4728142‐A was closely associated with increased IRF5 expression. We demonstrated that an rs4728142‐containing region could act as an enhancer to regulate the expression of IRF5. Moreover, rs4728142 affected the binding affinity of zinc finger and BTB domain–containing protein 3 (ZBTB3), a transcription factor involved in regulation. Furthermore, in monocytes from SLE patients, CRISPR‐based interference with the regulation of this enhancer attenuated the production of disease‐associated cytokines. Conclusion These results demonstrate that the rs4728142‐A allele increases the SLE risk by affecting ZBTB3 binding, chromatin status, and regulating IRF5 expression, establishing a biologic link between genetic variation and lupus pathogenesis.
The genomic region containing rs4728142 is a functional enhancer that regulates the expression of IRF5. A, Epigenomic analysis of the rs4728142‐containing region in different immune cell subsets. B, Flow chart for generating wild‐type (WT) clones and knockout (KO) clones using CRISPR/Cas9 technology. C, Genotype of WT clones and KO clones at the rs4728142 site. D, RT‐qPCR analysis of IRF5 expression in U937 WT and KO clones (n = 3 biologic sample replicates). E, ChIP‐qPCR analysis of the enrichment of H3K27ac in the rs4728142‐containing region. F, ChIP‐seq and the assay for transposase‐accessible chromatin using sequencing (ATAC‐seq) analysis of the enrichment of H3K27ac and chromatin accessibility of rs4728142‐containing region in U937 cells. G, Experimental design to activate or inhibit IRF5 expression by targeting the rs4728142‐containing region using the CRISPR activation (CRISPRa) system or the CRISPR interference (CRISPRi) system. H, Components of CRISPRa and CRISPRi. I and J, RT‐qPCR analysis of the expression of IRF5 in CRISPRa U937 cells and CRISPRi U937 cells (n = 3 biologic sample replicates). Bars show the mean ± SEM. * = P < 0.05; ** = P < 0.01; *** = P < 0.001, by Student's unpaired 2‐tailed t‐test. See Supplementary Figure 3 (https://onlinelibrary.wiley.com/doi/10.1002/art.42390). sgRNA = targeting rs4728142 enhancer single‐guide RNA; GFP = green fluorescent protein; FACS = fluorescence‐activated cell sorting; sgNC = negative control sgRNA (see Figure 1 for other definitions). Color figure can be viewed in the online issue, which is available at http://onlinelibrary.wiley.com/doi/10.1002/art.42390/abstract.
… 
SLE risk single‐nucleotide polymorphism rs4728142 affects zinc finger and BTB domain–containing protein 3 (ZBTB3) binding and chromatin state to regulate gene expression. A and B, Expression QTL analysis revealing genotype‐dependent expression of IRF5 for rs4728142 in different immune cell subpopulations (ImmuNexUT project) and whole blood (Genotype‐Tissue Expression project). Data are shown as box plots. Each box represents the range of values. Lines inside the boxes represent the mean. C, Flow chart for creating clones containing different rs4728142 alleles. D, RT‐qPCR analysis showing increased IRF5 expression in G/A clones compared to G/G clones (n = 7 biologic sample replicates). E, The ZBTB3 family DNA binding motif in the context of the DNA sequence surrounding rs4728142. Tall nucleotides above the x‐axis indicate preferred DNA bases. Bases below the x‐axis are disfavored. F, Transcription factor ZBTB3 preference to bind to the A risk allele at rs4728142 as determined by AS‐ChIP‐qPCR in rs4728142 heterozygous U937 cell clone (n = 3 biologic sample replicates). G, The rs4728142‐A risk allele showing higher chromatin accessibility than the nonrisk allele G, as detected by AS‐FAIRE‐qPCR (n = 3 biologic sample replicates). H and I, RT‐qPCR showing the expression of ZBTB3 and IRF5 after ZBTB3 knockdown (n = 3 biologic sample replicates). Bars show the mean ± SEM. * = P < 0.05; ** = P < 0.01; *** = P < 0.001, by Student's unpaired 2‐tailed t‐test. See Supplementary Figure 4 (https://onlinelibrary.wiley.com/doi/10.1002/art.42390). shNC = negative control short hairpin; shZBTB3 = short hairpin ZBTB3 (see Figure 1 for other definitions). Color figure can be viewed in the online issue, which is available at http://onlinelibrary.wiley.com/doi/10.1002/art.42390/abstract.
… 
This content is subject to copyright. Terms and conditions apply.
Integrative Functional Genomics Identies Systemic Lupus
Erythematosus Causal Genetic Variant in the IRF5 Risk Locus
Guojun Hou,
1
Tian Zhou,
2
Ning Xu,
2
Zhihua Yin,
3
Xinyi Zhu,
2
Yutong Zhang,
2
Yange Cui,
4
Jianyang Ma,
2
Yuanjia Tang,
2
Zhaorui Cheng,
2
Yiwei Shen,
2
Yashuo Chen,
3
Ling-Hua Zou,
3
Yong-fei Wang,
5
Zihang Yin,
6
Ya Guo,
6
Huihua Ding,
2
Zhizhong Ye,
3
and Nan Shen
7
Objective. IRF5 plays a crucial role in the development of lupus. Genome-wide association studies have identied
several systemic lupus erythematosus (SLE) risk single-nucleotide polymorphisms (SNPs) enriched in the IRF5 locus.
However, no comprehensive genome editingbased functional analysis exists to establish a direct link between these
variants and altered IRF5 expression, particularly for enhancer variants. This study was undertaken to dissect the reg-
ulatory function and mechanisms of SLE IRF5 enhancer risk variants and to explore the utilization of clustered regularly
interspaced short palindromic repeat interference (CRISPRi) to regulate the expression of disease risk gene to
intervene in the disease.
Methods. Epigenomic proles and expression quantitative trait locus analysis were applied to prioritize putative
functional variants in the IRF5 locus. CRISPR-mediated deletion, activation, and interference were performed to inves-
tigate the genetic function of rs4728142. Allele-specic chromatin immunoprecipitationquantitative polymerase chain
reaction and allele-specic formaldehyde-assisted isolation of regulatory elementquantitative polymerase chain reac-
tion were used to decipher the mechanism of alleles differentially regulating IRF5 expression. The CRISPRi approach
was used to evaluate the intervention effect in monocytes from SLE patients.
Results. SLE risk SNP rs4728142 was located in an enhancer region, indicating a disease-related regulatory func-
tion, and risk allele rs4728142-A was closely associated with increased IRF5 expression. We demonstrated that an
rs4728142-containing region could act as an enhancer to regulate the expression of IRF5. Moreover, rs4728142
affected the binding afnity of zinc nger and BTB domaincontaining protein 3 (ZBTB3), a transcription factor involved
in regulation. Furthermore, in monocytes from SLE patients, CRISPR-based interference with the regulation of this
enhancer attenuated the production of disease-associated cytokines.
Conclusion. These results demonstrate that the rs4728142-A allele increases the SLE risk by affecting ZBTB3
binding, chromatin status, and regulating IRF5 expression, establishing a biologic link between genetic variation and
lupus pathogenesis.
INTRODUCTION
Systemic lupus erythematosus (SLE) is a complex autoim-
mune disease, which is characterized by the breakdown of
tolerance to self antigens, with the presentation of autoantibody
production, inammation, and organ damage (1). Although the
pathogenesis of SLE is not well characterized, the genetic factors
denitely contribute to disease etiology (24). Genome-wide
Supported by the National Natural Science Foundation of China (grants
31930037 and 32141004), the Shanghai Science and Technology Innovation
Plan (grant 21Y31900200), and the Sanming Project of Medicine in Shenzhen
(grant SZSM201602087).
Drs. Hou, Zhou, Xu, and Zhihua Yin contributed equally to this work.
1
Guojun Hou, PhD: Shanghai Institute of Rheumatology, Renji Hospital,
Shanghai Jiao Tong University School of Medicine, Shanghai, China, Shenzhen
Futian Hospital for Rheumatic Diseases, Shenzhen, China, and State Key Lab-
oratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji
Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China;
2
Tian Zhou, PhD, Ning Xu, MSc, Xinyi Zhu, BS, Yutong Zhang, BS, Jianyang
Ma, MSc, Yuanjia Tang, PhD, Zhaorui Cheng, BS, Yiwei Shen, BS, Huihua Ding,
MD: Shanghai Institute of Rheumatology, Renji Hospital, Shanghai Jiao Tong
University School of Medicine, Shanghai, China;
3
Zhihua Yin, PhD, Yashuo
Chen, MD, Ling-Hua Zou, MD, Zhizhong Ye, MD: Shenzhen Futian Hospital
for Rheumatic Diseases, and Joint Research Laboratory for Rheumatology of
Shenzhen University Health Science Center and Shenzhen Futian Hospital
for Rheumatic Diseases, Shenzhen, China;
4
Yange Cui, PhD: Shanghai Insti-
tute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Uni-
versity of Chinese Academy of Sciences, Chinese Academy of Sciences,
Shanghai, China;
5
Yong-fei Wang, PhD: School of Life and Health Sciences,
School of Medicine, and Warshel Institute for Computational Biology, The Chi-
nese University of Hong Kong, Shenzhen, Guangdong, China;
6
Zihang Yin,
PhD, Ya Guo, PhD: Sheng Yushou Center of Cell Biology and Immunology,
Joint International Research Laboratory of Metabolic & Developmental Sci-
ences, School of Life Sciences and Biotechnology, Shanghai Jiao Tong Univer-
sity, Shanghai, China;
7
Nan Shen, MD, PhD: Shanghai Institute of
Rheumatology, Renji Hospital, Shanghai Jiao Tong University School of
574
Arthritis & Rheumatology
Vol. 75, No. 4, April 2023, pp 574585
DOI 10.1002/art.42390
© 2022 American College of Rheumatology
association studies (GWAS) have identied >100 genetic variants
associated with SLE susceptibility (4,5). Most of these SNPs are
located in the noncoding regions of the genome, particularly in
enhancers (6,7), suggesting the important role of enhancers in
SLE development.
Enhancers are the specic DNA sequences that bind tran-
scription factors to increase the transcription of target genes.
Enhancers could shape the expression pattern of genes to con-
trol cell identity and cell fate. An increasing number of diseases
are linked to enhancer dysfunction (6,8,9). Identication of the
disease-associated enhancers and deciphering the underlying
mechanism involved in the disease will help us understand the
development of SLE pathogenesis. Notably, the disease func-
tional variants could pinpoint the disease-associated enhancers
(1013). However, few studies have identied the functional
enhancer variants of SLE, let alone deciphered the mechanisms.
Interferon regulatory factor 5 (IRF5) is a transcription factor
that plays a critical role in mediating innate and adaptive immunity.
Many genetic studies have revealed a strong association between
IRF5 and SLE (1418). The use of IRF5-knockout mice directly
demonstrated the critical role of IRF5 in the pathogenesis of SLE
(19,20). Moreover, IRF5 is abnormally expressed and activated
in SLE patients and affects various pathogenic pathways by regu-
lating the production of type I interferons (IFNs), proinammatory
cytokines, autoantibodies, etc. (2124). Genetic variants were
considered as an important factor that affects the expression of
IRF5. For example, rs2004640 is located in the 50untranslated
region (UTR) of IRF5 and creates an alternative splice site to regu-
late the expression of IRF5 different isoforms (14). Also,
rs10954213 is located in the 30UTR of IRF5 and alters a polyade-
nylation signal, inuencing IRF5 expression (16,25).
In addition, a 5-bp CGGGG indel located at the 50UTR of
IRF5 creates an Sp-1 binding site involved in the regulation of
IRF5 expression (17). However, the genetic variants in the
enhancers of IRF5 are rarely studied, and the mechanism of
these variants acting in SLE remains elusive. To ll this gap, we
focused our study on the enhancer variants at the IRF5 locus.
By integrating genetic studies, epigenomic analysis, and clus-
tered regularly interspaced short palindromic repeat (CRISPR)
editing, we found that rs4728142 is an SLE-associated causal
genetic variant, and the genomic region containing rs4728142
regulates the expression of IRF5 as a functional enhancer.
Moreover, the rs4728142 alleles differentially bind with zinc n-
ger and BTB domaincontaining protein 3 (ZBTB3) and alter
the chromatin state to ne-tune IRF5 expression involved in dis-
ease pathogenesis.
SUBJECTS AND METHODS
Study design. The overall study design is shown in
Figure 1. First, we integrated public genetic data, epigenomic
data, and expression quantitative trait locus (eQTL) data to iden-
tify the candidate variants. Next, we performed functional stud-
ies such as CRISPR activation assay, CRISPR interference
(CRISPRi) assay, CRISPR-mediated deletion, allele-specic
chromatin immunoprecipitationquantitative polymerase chain
reaction (AS-ChIP-qPCR), and allele-specic formaldehyde-
assisted isolation of regulatory elementquantitative polymerase
chain reaction (AS-FAIRE-qPCR) to validate whether rs4728142
is a functional variant.
Cell culture. U937 and HEK 293T cells were purchased
from the Cell Bank of Shanghai Institutes for Biological Sciences.
U937 cells were cultured with RPMI 1640 (no. 22400105; Gibco)
together with 10% fetal bovine serum (FBS) (no. 10099141,
Gibco), and HEK 293T cells were cultured with Dulbeccos mod-
ied Eagles medium (no. 11039047; Gibco) together with 10%
FBS. Cells were all cultured at 37Cin5%CO
2
.
RNA extraction and reverse transcriptase
quantitative PCR (RT-qPCR) analysis. Total RNA was isolated
using Trizol reagent (no. 15596026; Invitrogen). Complementary
DNA (cDNA) was synthesized using PrimeScript RT reagent Kit
(no. RR037A; Takara) using 500 ng of RNA per cDNA reaction.
RT-qPCR reactions were performed using TB Green Premix Ex
Taq reagent (no. RR420A) according to the protocol of the manu-
facturer (Takara). Results were analyzed using the comparative
Ct method, and the GAPDH messenger RNA level was chosen
as the normalization. The primers used in the present study are
listed in Supplementary Table 1 (available on the Arthritis &
Rheumatology website at https://onlinelibrary.wiley.com/doi/
10.1002/art.42390).
Lentivirus production. Lentiviral particles were produced
in HEK 293T cells using the pMD2.G (no. 12259; Addgene) and
psPAX2 (no. 12260; Addgene) packaging plasmids. Briey,
5×10
5
cells were seeded into the wells of 6-well plates. After
incubation overnight, cells were transfected with 1 μg of a plasmid
expressing single-guide RNA (sgRNA) or short hairpin RNA
(shRNA), 250 ng of pMD2.G, and 750 ng of psPAX2, using 3 μlof
Lipofectamine 2000 (no. 11668019; Thermo Fisher). After transfec-
tion for 6 hours, the media was changed. After transfection for
72 hours, the virus supernatant was collected and centrifuged at
Medicine, Shanghai, China, Shenzhen Futian Hospital for Rheumatic Diseases,
Shenzhen, China, State Key Laboratory of Oncogenes and Related Genes,
Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University
School of Medicine, Shanghai, China, Center for Autoimmune Genomics and
Etiology, Division of Human Genetics, Cincinnati Childrens Hospital Medical
Center, Cincinnati, Ohio, and Department of Pediatrics, University of Cincin-
nati, Cincinnati, Ohio.
Author disclosures are available at https://onlinelibrary.wiley.com/action/
downloadSupplement?doi=10.1002%2Fart.42390&le=art42390-sup-0001-
Disclosureform.pdf.
Address correspondence via email to Nan Shen, MD, PhD, at
nanshensibs@gmail.com.
Submitted for publication July 11, 2022; accepted in revised form October
4, 2022.
SLE-ASSOCIATED RISK VARIANT REGULATES IRF5 EXPRESSION 575
23265205, 2023, 4, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/art.42390 by Affiliated Hospitals, Wiley Online Library on [31/03/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
4C for 10 minutes. The supernatant was aliquoted and stored
at 80C.
Short hairpin RNA knockdown assay. Single-guide
RNA expression vector pKLV-U6-gRNA-PGKpuro2ABFP plas-
mid (no. 50946; Addgene) was used to express shRNA. The plas-
mid was cut with restriction enzyme BbsI (no. R0539L; NEB) and
BamHI (no. R0136L; NEB), and then the large fragment was gel-
puried (no. DC301-01; Vazyme). Oligos were synthesized in
Tsingke (Shanghai), annealed, and subsequently cloned into the
digested plasmid. Short hairpin RNA lentiviral particles were pro-
duced as mentioned above. The virus particles were added into
2×10
5
cells, and centrifuged at 1,000gfor 90 minutes to trans-
duce the cells. After infection for 24 hours, the media was
changed and cells were cultured for another 72 hours with 1 μg/ml
puromycin (ant-pr-1; InvivoGen). Then, cells were collected and
RNA was extracted to detect target gene expression. The oligos
used in the present study are listed in Supplementary Table 2
(https://onlinelibrary.wiley.com/doi/10.1002/art.42390).
Genome editing in cell line. To delete the fragment har-
boring rs4728142 or introduce rs4728142 different alleles,
sgRNAs targeting the rs4728142 site were designed using online
CRISPR design tool CHOPCHOP. Single-guide RNA oligos were
annealed and subcloned into PX458 (no. 48138; Addgene) vec-
tor, which can express a chimeric guide RNA, a human codon-
optimized Cas9, and a green uorescent protein (GFP). Plasmids
were transformed into Tstbl3 chemically competent Escherichia
coli (no. TSC-C06; Tsingke) and grown, and plasmid DNA was
extracted and puried. Next, 6 μg of CRISPR plasmid and 30 μg
of a 121-bp single-stranded oligodeoxynucleotide donor tem-
plate (Sango Biotech) were electroporated into 1.5 × 10
6
cells
using the 100-μl Neon Transfection System (Thermo Fisher),
under conditions of 1,400V, 10 msec, and 3 pulses. After trans-
fection for 12 hours, 1 μMSCR-7 (no. SML1546; Sigma-Aldrich)
was added to enhance homology-directed repair (HDR) ef-
ciency. After transfection for 3 days, single cells with high GFP
uorescence was sorted into 96-well plates using ow cytometry
(FACS Aris II; BD Biosciences). After 14 days of cell growth, the
genomic DNA of individual clone was isolated using a TransDirect
Animal Tissue PCR Kit (no. AD201-02; Transgen Biotech), the
sequence containing rs4728142 was amplied, and the genotype
was identied by Sanger sequencing. Clones with different
rs4728142 alleles or rs4728142-containing region knockout
clones were chosen for further study. The oligos used in the pres-
ent study are listed in Supplementary Tables 3 and 4 (https://
onlinelibrary.wiley.com/doi/10.1002/art.42390).
CRISPRi assays. Cells stably expressingKruppel-associated
box (KRAB)/dCas9 were generated using the pHR-SFFV-KRAB-
dCas9-P2A-mCherry plasmid (no. 60954; Addgene). Briey,
lentiviral particles were generated in HEK 293T cells using the
pMD2.G (no. 12259; Addgene) and psPAX2 (no. 12260; Addgene)
packaging plasmids. U937 cells were transduced by the lentivi-
ruses for 24 hours, the medium was changed, and cells were
cultured for another 48 hours. Next, cells with a strong mCherry
signal were sorted by uorescence-activated cell sorting
(FACS). Single-guide RNAs targeting the rs4728142-containing
region were designed by CHOPCHOP. Oligos were synthesized
in Tsingke and subcloned into pKLV-U6-gRNA(BbsI)-
PGKpuro2ABFP plasmid (no. 50946; Addgene). Single-guide
RNA lentiviral particles were produced and transduced
KRAB/dCas9/mCherry U937 cells as mentioned above, and
cells were further treated with 1 μg/ml puromycin for 3 days.
Subsequently, cells were collected and RNA was extracted to
test target gene expression. The oligos used in the present study
Genome wide association studies
0
5
15
10
-log
10
(P)
GG GA AA
Expression
Tissue and cell-type
eQTL analysis
cell-type
Epigenetic modication
H3K27ac H3K4me1 DHS
SLE patients
Monocytes isolation
Transfect KRAB-dCas9
interference system
Inhibit IRF5 expression
IL-6 IFN-β
Proinammatory cytokines, Type I IFNs
HDR
Knock out Activation Interference
IRF5 mRNA expression
Chromatin accessebility
Transcription factor binding
Single base regulatory mechanism
RT-qPCR
AS-FAIRE-qPCR
AS-ChIP-qPCR
rs4728142 G/A
Enhancer verication by CRISPR
Figure 1. Study design and experimental steps. eQTL = expression quantitative trait locus; CRISPR = clustered regularly interspaced short
palindromic repeat; HDR = homology-directed repair; RT-qPCR = reversetranscriptase quantitative polymerase chain reaction; AS-FAIRE-
qPCR = allele-specic formaldehyde-assisted isolation of regulatory elementqPCR; AS-ChIP-qPCR = allele-specic chromatin
immunoprecipitationqPCR; SLE = systemic lupus erythematosus; KRAB = Kruppel-associated box; IL-6 = interleukin 6; IFNβ= interferon-β.
HOU ET AL576
23265205, 2023, 4, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/art.42390 by Affiliated Hospitals, Wiley Online Library on [31/03/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
are listed in Supplementary Table 5 (https://onlinelibrary.wiley.
com/doi/10.1002/art.42390).
Inhibition by CRISPRi of the expression of proin-
ammatory cytokines and type I IFNs in U937 cells. For
this assay, we rst generated U937 cells stably expressing Toll-
like receptor 7 (TLR-7) and KRAB/dCas9. The lentivirus particles
of pLV-TLR7-GFPSpark plasmid (no. HG11204-ACGLN; Sino-
Biological, lnc.) were packaged and transduced into the U937/
KRAB/dCas9 cells. The cells with strong GFP signal were sorted
by FACS to generate cells stably expressing TLR-7. Single-guide
RNA lentiviral particles targeting the rs4728142 site were pro-
duced and transduced the cells, as mentioned above, and cells
were further treated with 1 μg/ml puromycin for 3 days. Then,
cells were stimulated with 1 μg/ml R837 (TLR-7 agonist tlrl-imqs;
InvivoGen) for 24 hours to induce the expression of proinamma-
tory cytokines and type I IFNs. The cells were collected and RNA
was extracted to test target gene expression. The oligos used in
the present study are listed in Supplementary Table 1 (https://
onlinelibrary.wiley.com/doi/10.1002/art.42390).
CRISPR activation (CRISPRa) assay. Cells stably
expressing the CRISPRa system were generated using the
lenti-dCas-VP64-Blast plasmid (no. 61425; Addgene) and
lenti-MS2-P65-HSF1-Hygro plasmid (no. 61426; Addgene), as
mentioned above. Cells were transduced by the lentivirus and
treated with 10 μg/ml blasticidin (no. ant-bl-1; InvivoGen) and
300 μg/ml hygromcin B (no. 60224ES03; Yeasen) for 1 week to iden-
tify the cells stably expressing CRISPRa. Single-guide RNAs targeting
the rs4728142-containing region were subcloned into lenti-sgRNA
(MS2)-zeo backbone plasmid (no. 61427; Addgene). Single-guide
RNA lentivirus particles were produced, and the cells (stably express-
ing dCas/VP64 and MS2-P65-HSF1) were transduced as mentioned
above. Next, the cells were treated with 400 μg/ml Zeocin
(no. R25001; Thermo Fisher) for 72 hours. After that, the cells were
collected and RNA was extracted to test target gene expression.
The oligos used in the present study are listed in Supplementary
Table 5 (https://onlinelibrary.wiley.com/doi/10.1002/art.42390).
AS-ChIP-qPCR. ChIP assay was carried out using a Simple-
ChIP Plus Enzymatic Chromatin IP Kit (no. 9005; Cell Signaling
Technology). Briey, 1 × 10
7
cells were crosslinked with 1%formal-
dehyde solution for 10 minutes at room temperature and quenched
with 0.125Mglycine. Cells were lysed in buffer A and treated
with 1 μl of micrococcal nuclease for 30 minutes at 37Ctodigest
DNA. After digestion, pellet nuclei were resuspended in ChIP
buffer and subsequently sonicated with a Bioruptor sonicator
(Diagenode) at high power for 5 cycles for 30 seconds, with
30 seconds between cycles. After removing insoluble debris, the
supernatant was separately incubated with specicantibodies
against rabbit IgG ZBTB3 (no. ab106536; Abcam), H3K27ac
(no. ab177178; Abcam), and Protein G Magnetic Beads at 4C
overnight. Beads were washed and DNA was reverse-crosslinked
and puried to detect the enrichment. Allele-specic qPCR primers
were designed to specically amplify the rs4728142 region with a
G or A allele in theDNA samples from the ChIP experiment. Results
were analyzed in a manner similar to normal qPCR. The oligos used
in the present study are listed in Supplementary Table 4 (https://
onlinelibrary.wiley.com/doi/10.1002/art.42390).
AS-FAIRE-qPCR. The FAIRE DNA sample was prepared
using the aliquot from the crosslinked and sonicated ChIP chro-
matin. The chromatin lysate was treated with phenol/chloroform/
isoamyl alcohol and then with chloroform/isoamyl alcohol. Then
the aqueous phase was precipitated with ethanol and dissolved
in 10 mMTris HCL. After that, samples were reverse-crosslinked,
puried, and analyzed using AS-qPCR primers. Results were
analyzed in a manner similar to normal qPCR. The oligos used in
the present study are listed in Supplementary Table 4 (https://
onlinelibrary.wiley.com/doi/10.1002/art.42390).
SLE patients. Patients were diagnosed based on the 1997
update of the American College of Rheumatology revised SLE cri-
teria (26) in the rheumatology department at Shanghai Renji Hos-
pital. The clinical characteristics of the SLE patients are displayed
in Supplementary Table 6 (https://onlinelibrary.wiley.com/doi/10.
1002/art.42390). All patients provided written informed consent
according to the internal review and ethics boards of Renji Hospi-
tal and Shanghai Jiao Tong University.
CRISPRi in SLE monocytes. Blood samples from SLE
patients were collected with 10 ml vacuum sterile tube, and
peripheral blood mononuclear cells (PBMCs) were isolated by
Ficoll gradient centrifugation. After isolation, PBMCs were incu-
bated with CD14 microbeads (no. 130-050-201; Miltenyi Biotec)
to isolate CD14+ monocytes. Monocytes (3 × 10
5
) were trans-
fected with 1 μg KRAB/dCas9 plasmid and 1 μg sgRNA plasmid
using the Neon Transfection System under the following condi-
tions: 1,600V, 10 msec, and 3 pulses. After transfection for
24 hours, the cells were collected and RNA was extracted to
detect target gene expression.
Statistical analysis. All statistical analyses were per-
formed using GraphPad Prism 8 software and calculated using a
Students paired or unpaired 2-tailed t-test as indicated in the g-
ure legends, unless otherwise mentioned. Pvalues less than 0.05
are considered signicant.
Data availability statement. The data from GSE155555
(7) were used to analyze the assay for transposase-accessible
chromatin using sequencing (ATAC-seq) and H3K27ac signal of
U937 cells. All data relevant to the study are included in the article
or uploaded as supplementary information.
SLE-ASSOCIATED RISK VARIANT REGULATES IRF5 EXPRESSION 577
23265205, 2023, 4, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/art.42390 by Affiliated Hospitals, Wiley Online Library on [31/03/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
RESULTS
Signicant association of rs4728142 with SLE and
with interferon regulatory factor 5 (IRF5) expression in
a tissue-dependent manner. IRF5 is a key transcription factor
involved in multiple steps of SLE pathogenesis (22). In SLE
patients, IRF5 is abnormally expressed and activated. This dys-
function is associated with genetic variants (14,16,17,21). Several
studies have shown that genetic variants, such as rs2004640
(14), rs10954213 (16,25), and 5-bp CGGGG indel (17), could reg-
ulate the expression of IRF5. However, these causal variants are
located in UTR region or promoter region (Figure 2A), and the func-
tional variants in enhancers are poorly studied. Given the impor-
tance of enhancers in controlling disease-critical gene expression,
we decided to focus our study on the enhancer variants of IRF5.
To identify the SLE causal variants in the enhancers of IRF5,we
rst collected all SLE risk variants at IRF5 locus (GWAS catalog var-
iants and the variants that have been reported to affect IRF5 expres-
sion in SLE patients) (2,5,14,16,17,2735). Next, we used the
SCREEN system (https://screen.encodeproject.org/), a tool for
searching candidate cis-regulatory elements derived from Encyclo-
pedia of DNA Elements (ENCODE) data, to analyze the enhancer
characters of these single-nucleotide polymorphisms (SNPs) (36).
Finally, we chose noncoding SNPs located in the enhancers as
our candidates (Figure 2B). After analysis, we identied 12 SLE risk
variants at this locus. Among them, 6 SNPs were intergenic vari-
ants, 3 SNPs were UTR variants, 2 SNPs affected the splicing of
IRF5, and 1 SNP was an intron variant (Figure 2C,Supplementary
Figures 1AH, and Supplementary Table 7, https://onlinelibrary.
wiley.com/doi/10.1002/art.42390). Notably, only intergenic SNP
rs4728142 is located in the distal enhancer-like element
(Supplementary Figure 1AH). rs4728142 is located in the
upstream region of IRF5 (Figure 2A); a genetic study (18)has
identied rs4728142-A as a risk allele that is associated with an
increased risk for SLE development at the IRF5 locus, and there
is no linkage disequilibrium with the reported functional variants
(rs2004640, rs10954213, and 5-bp CGGGG indel).
Meanwhile, a phenome-wide association study (PheWAS),
which is a powerful approach to comprehensively evaluate associ-
ations between genetic variants and different phenotypes, revealed
that rs4728142 has the strongest association with the SLE pheno-
type (n = 4,756 GWAS) (https://atlas.ctglab.nl/)(37,38) (Figure 2D
and Supplementary Table 8, https://onlinelibrary.wiley.com/doi/
10.1002/art.42390). In addition, eQTL data from the Genotype-
Tissue Expression (GTEx) project suggest that rs4728142 is
strongly associated with the expression of IRF5 (Supplementary
Figure 2 and Supplementary Table 9) and the effect of
rs4728142 different alleles on IRF5 expression is tissue-
Ex2 Ex3 Ex4 Ex6 Ex7 Ex8 Ex9Ex1a Ex1b Ex1c
rs2004640
Exon 1b
splicing site
rs10954213
Alter the
polyadenylation site
5 bp CGGGG
indel
Alters
SP1 binding
rs4728142
A
Activities Aging Body Structures Cardiovascular Cell Cognitive Connective Tissue Dermatological Endocrine Environment
Environmental Gastrointestinal Hematological Immunological Infection Metabolic Mortality Neoplasms Neurological Nutritional
Ophthalmolo
g
ical Psychiatric Reproduction Respiratory Skeletal Social Interactions
-log10 P-value
Systemic Lupus Erythematosus
Systemic Lupus Erythematosus
Estimated glomerular filtration rate
Estimated glomerular filtration rate
Primary biliary cirrhosis
Rheumatoid Arthritis
Mouth/teeth dental problems: Mouth ulcers
D
GWAS
SLE risk SNPs at IRF5 locus
12 SNPs
Analysis of the Enhancer characters
of SNP by SCREEN system
of ENCODE
SNP in the Enhancer region
as candidate
B
6
3
21
Intergenic region
UTR
Splicing site
Intron
region
C
IRF5
Figure 2. Identication of rs4728142 as a candidate enhancer variant at IRF5 locus. A, Locations of rs4728142, CGGGG indel, rs2004640, and
rs10954213 relative to IRF5.B, Flow chart identifying the enhancer variants at IRF5 locus. C, Pie chart showing the distribution of functional anno-
tation of systemic lupus erythematosus (SLE) risk variants at the IRF5 locus. D, Manhattan plot of phenome-wide association studies showing the
signicance of association between single-nucleotide polymorphism (SNP) rs4728142 and different phenotypes, with phenotypes ordered within
each phenotype category by Pvalue. SLE was the most signicant phenotypic association with rs4728142. Along the x-axis, different phenotypes
are color-coded. The y-axis indicates level of signicance. SNPs with Pvalues >0.05 are not shown in the plot. There are 287 data points displayed
in the plot (Bonferroni corrected P= 1.74 × 10
4
). The association results were retrieved from the genome-wide association study (GWAS) ATLAS
(ref. 36). See Supplementary Figures 1 and 2 (https://onlinelibrary.wiley.com/doi/10.1002/art.42390). SCREEN = Search Candidate cis-
Regulatory Elements by ENCODE; ENCODE = Encyclopedia of DNA Elements; UTR = untranslated region. Color gure can be viewed in the online
issue, which is available at http://onlinelibrary.wiley.com/doi/10.1002/art.42390/abstract.
HOU ET AL578
23265205, 2023, 4, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/art.42390 by Affiliated Hospitals, Wiley Online Library on [31/03/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
dependent (Supplementary Figure 2). More importantly, epige-
nomic analysis in different immune cell subsets shows that the
rs4728142-containing region is enriched with the signals of
H3K27ac modication, H3K4me1 modication, and DNase I hyper-
sensitivity sites occupancy (http://www.roadmapepigenomics.org/)
(Figure 3A), suggesting that this region is an active enhancer-like ele-
ment. Based on these observations, we focused our study on
rs4728142.
The rs4728142-containing region is a functional
regulatory element modulating IRF5 expression.
Expression QTL data indicate an association between
rs4728412 and IRF5, and epigenomic analysis shows that the
rs4728142-containing region is an active enhancer-like element.
To directly evaluate the regulatory function and target genes of
this region, we attempted to delete the enhancer fragment con-
taining rs4728142 using CRISPR/Cas9-mediated knockout.
Since the rs4728142-containing region has the strongest
enhancer marker signal in monocytes (Figure 3A) and the expres-
sion of IRF5 is highest in monocytes (Supplementary Figure 3A,
https://onlinelibrary.wiley.com/doi/10.1002/art.42390), we per-
formed this assay in U937 monocyte cells.
We designed a sgRNA targeting the rs4728142 site and
transfected the sgRNA/Cas9 expression vector into the U937 cell
with the Neon Transfection System. Then, single cells were sorted
by FACS, cultured, and the genotype was identied. After screen-
ing, we successfully identied rs4728142 knockout clones
(Figures 3B and C). Compared to rs4728142 wild-type clones,
deletion of the rs4728142-containing region decreased the
expression of IRF5 (Figure 3D), which is consistent with strong
signals of H3K27ac and chromatin accessibility of this region in
U937 cells (Figures 3E and F). Moreover, the expression of other
nearby genes such as TNPO3, TPI1P2, and ATP6V1F was not
affected by the deletion of this region (Supplementary
Figures 3BD, https://onlinelibrary.wiley.com/doi/10.1002/art.
42390). Further, we used CRISPRa or CRISPRi to conrm the
regulatory function of this region (Figures 3G and H). As shown
in Figures 3I and J, the CRISPRa or CRISPRi assay successfully
up-regulated or down-regulated the expression of IRF5, respec-
tively. Taken together, these results demonstrate that the
rs4728142-containing region functions as a regulatory element
regulating the expression of IRF5.
IRF5 expression differentially affected by rs4728142
alleles. After verifying the regulatory function of the
rs4728142-containing region, we sought to explore whether the
genotype of rs4728142 could affect the IRF5 expression. Regulo-
meDB (https://regulomedb.org/regulome-search)(39), a data-
base that annotates SNPs with known or predicted regulatory
elements in the noncoding regions of the genome, ranked
rs4728142 as a potential function variant with a high score of 1f
(Supplementary Figure 4A, https://onlinelibrary.wiley.com/doi/
10.1002/art.42390). Meanwhile, data from the GTEx project
(https://gtexportal.org/home/)(40) and the ImmuNexUT project
(https://www.immunexut.org/)(41) suggest that rs4728142 is
strongly associated with the expression of IRF5, and the effect of
different rs4728142 alleles on IRF5 expression is tissue- or cell-
typedependent, where the risk allele rs4728142-A is associated
with a higher expression of IRF5 relative to the nonrisk
allele rs4728142-G (Figures 4A and Band Supplementary
Figures 4BG).
To establish the direct link between rs4728142 alleles and
the expression of IRF5, we performed CRISPR-mediated HDR
at the rs4728142 site to introduce different alleles of rs4728142
in U937 cells (Figure 4C). After screening 200 clones, we nally
identied 7 clones with the homozygous G allele and 7 clones with
the heterogenous GA allele. We then detected IRF5 expression in
these clones. As shown in Figure 4D, rs4728142 risk allele
A demonstrated higher IRF5 expression than the rs4728142
nonrisk allele G, which is consistent with the public eQTL results.
Collectively, these data suggest that rs4728142 is a functional
variant modulating the expression of IRF5 in a genotype-
dependent manner in monocytes.
Altered transcription factor ZBTB3 binding and
chromatin status via rs4728142 alleles to regulate IRF5
expression. Functional SNPs usually ne-tune the expression
of the target gene by mediating the critical transcription factors
binding (42). To identify possible transcription factors that bind
to rs4728142, we rst used bioinformatic tools such as JASPAR
(43), the Catalog of Inferred Sequence Binding Preferences (44)
database, and the Human Transcription Factors catalog (45)to
predict the candidate transcription factors. We found that the
binding motif of ZBTB3 overlaps with the rs4728142 sequence
(Figure 4E). Notably, the ZBTB3 binding motif has a higher binding
afnity with the rs4728142-A risk allele relative to that of the
rs4728142-G nonrisk allele (Figure 4E). Further, we carried out
AS-ChIP-qPCR to conrm this genotype-dependent binding. As
expected, we observed that ZBTB3 prefers to bind to risk allele
A (Figure 4F). Consistent with this observation, AS-FAIRE-qPCR
data suggest that the rs4728142-A risk allele enriched more
FAIRE signal than nonrisk allele G (Figure 4G), indicating that
chromatin harboring risk allele A has higher chromatin accessibil-
ity than nonrisk allele G. In addition, knockdown of ZBTB3
reduced the expression of IRF5 in the U937 monocytes
(Figures 4H and I). These data reveal that the rs4728142 allele
specic bind to ZBTB3 differentially regulates the expression
of IRF5.
KRAB/dCas9 system targeting the rs4728142 locus
reduces the production of proinammatory cytokines
and type I IFNs by regulating the expression of IRF5.The
abnormal expression and activation of IRF5 induce the expression
of proinammatory cytokines and type I IFNs, thus contributing to
SLE-ASSOCIATED RISK VARIANT REGULATES IRF5 EXPRESSION 579
23265205, 2023, 4, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/art.42390 by Affiliated Hospitals, Wiley Online Library on [31/03/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
B
Scale
chr7:
5 kb hg19
128
,
570
,
000 128
,
571
,
000 128
,
572
,
000 128
,
573
,
000 128
,
574
,
000 128
,
575
,
000 128
,
576
,
000 128
,
577
,
000 128
,
578
,
000
IRF5
DNaseI
50 _
1
H3K4me1
100 _
0
H3K27ac
25 _
0
DNaseI
175 _
1
H3K4me1
175 _
0
H3K27ac
175 _
0
DNaseI
50 _
1
H3K4me1
100 _
0
H3K27ac
50
0
CD3+T cellsCD14+Monocytes
CD19+B cells
rs4728142
A
Neon transfection system
sgRNA-Cas9
expression vector Single GFP+ cell sorting by FACS
2 weeks culture
TTCCAGGTCACACCCCAAAAAGCTCTGAGCCGGTGTTAGTAAGAAATGGGGAGG
TTCCAGGTCACA----------------------------------------------------------AAGAAATGGGGAGG
rs4728142
sgRNA
WT
KO
C
WT KO
0.0
0.5
1.0
1.5
**
IRF5 relative expression
D
050100150200
IgG
H3K27ac
H3K27ac ChIP-qPCR in rs4728142 region
Fold enrichment
***
E
F
rs4728142
0-56
0-35
0-51
0-47
U-937 H3K27ac ChIP-seq 1
U-937 H3K27ac ChIP-seq 2
U-937 ATAC-seq 1
U-937 ATAC-seq 2
inhibitor
dCas9 protein
genome sgRNA
CRISPRi
rs4728142
IRF5
activator
dCas9 protein
genome sgRNA
CRISPRa
rs4728142
IRF5
G
HJ
CRISPRa or CRISPRi U-937 cells
lentivirus infection
(sgRNA targeting rs4728142
or non-targeting region)
selection for 3 days
CRISPRa or CRISPRi expressed
and sgRNA expressed U-937 cells RT-qPCR detection the
expression of IRF5
I
sgNC sgRNA1sgRNA2
0
1
2
3
IRF5 relative expression
**
**
CRISPRa
sgNC sgRNA1sgRNA2
0.0
0.5
1.0
1.5
IRF5 relative expression
*
**
CRISPRi
Figure 3. The genomic region containing rs4728142 is a functional enhancer that regulates the expression of IRF5.A, Epigenomic analysis of
the rs4728142-containing region in different immune cell subsets. B, Flow chart for generating wild-type (WT) clones and knockout (KO) clones
using CRISPR/Cas9 technology. C, Genotype of WT clones and KO clones at the rs4728142 site. D, RT-qPCR analysis of IRF5 expression in
U937 WT and KO clones (n = 3 biologic sample replicates). E, ChIP-qPCR analysis of the enrichment of H3K27ac in the rs4728142-containing
region. F, ChIP-seq and the assay for transposase-accessible chromatin using sequencing (ATAC-seq) analysis of the enrichment of H3K27ac
and chromatin accessibility of rs4728142-containing region in U937 cells. G, Experimental design to activate or inhibit IRF5 expression by target-
ing the rs4728142-containing region using the CRISPR activation (CRISPRa) system or the CRISPR interference (CRISPRi) system. H, Compo-
nents of CRISPRa and CRISPRi. Iand J, RT-qPCR analysis of the expression of IRF5 in CRISPRa U937 cells and CRISPRi U937 cells (n = 3
biologic sample replicates). Bars show the mean ± SEM. * = P< 0.05; ** = P< 0.01; *** = P< 0.001, by Students unpaired 2-tailed t-test. See
Supplementary Figure 3 (https://onlinelibrary.wiley.com/doi/10.1002/art.42390). sgRNA = targeting rs4728142 enhancer single-guide RNA;
GFP = green uorescent protein; FACS = uorescence-activated cell sorting; sgNC = negative control sgRNA (see Figure 1for other denitions).
Color gure can be viewed in the online issue, which is available at http://onlinelibrary.wiley.com/doi/10.1002/art.42390/abstract.
HOU ET AL580
23265205, 2023, 4, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/art.42390 by Affiliated Hospitals, Wiley Online Library on [31/03/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
GA
0
2
4
6
8
AS-FAIRE-qPCR
*
Fold enrichment
GG
GA
0.0
0.5
1.0
1.5
2.0
**
IRF5 relative expression
G
EF
H
I
genome sgRNA
Double strands break
ssODN
rs4728142 GGrs4728142 AA
rs4728142
D
C
GCCGGTG
GCCAGTG
rs4728142-A
rs4728142-G
ZBTB3 motif
IRF5
(225) (326) (119)
GG GA AA
-3.0
-2.0
-1.0
0.0
1.0
2.0
3.0
Norm. Expression
Whole Blood
rs4728142
p value: 3.42e-78
B
shNC shZBTB3
0.0
0.5
1.0
1.5
ZBTB3 relative expression
***
shNC shZBTB3
0.0
0.5
1.0
1.5
IRF5 relative expression
**
GAGA
0
5
10
15 AS-ChIP-qPCR
Fold enrichment
IgG ZBTB3
A
Figure 4. SLE risk single-nucleotide polymorphism rs4728142 affects zinc nger and BTB domaincontaining protein 3 (ZBTB3) binding and
chromatin state to regulate gene expression. Aand B, Expression QTL analysis revealing genotype-dependent expression of IRF5 for
rs4728142 in different immune cell subpopulations (ImmuNexUT project) and whole blood (Genotype-Tissue Expression project). Data are shown
as box plots. Each box represents the range of values. Lines inside the boxes represent the mean. C, Flow chart for creating clones containing dif-
ferent rs4728142 alleles. D, RT-qPCR analysis showing increased IRF5 expression in G/A clones compared to G/G clones (n = 7 biologic sample
replicates). E, The ZBTB3 family DNA binding motif in the context of the DNA sequence surrounding rs4728142. Tall nucleotides above the x-axis
indicate preferred DNA bases. Bases below the x-axis are disfavored. F, Transcription factor ZBTB3 preference to bind to the A risk allele at
rs4728142 as determined by AS-ChIP-qPCR in rs4728142 heterozygous U937 cell clone (n = 3 biologic sample replicates). G, The
rs4728142-A risk allele showing higher chromatin accessibility than the nonrisk allele G, as detected by AS-FAIRE-qPCR (n = 3 biologic sample
replicates). Hand I, RT-qPCR showing the expression of ZBTB3 and IRF5 after ZBTB3 knockdown (n = 3 biologic sample replicates). Bars show
the mean ± SEM. * = P< 0.05; ** = P< 0.01; *** = P< 0.001, by Students unpaired 2-tailed t-test. See Supplementary Figure 4 (https://onlineli
brary.wiley.com/doi/10.1002/art.42390). shNC = negative control short hairpin; shZBTB3 = short hairpin ZBTB3 (see Figure 1for other deni-
tions). Color gure can be viewed in the online issue, which is available at http://onlinelibrary.wiley.com/doi/10.1002/art.42390/abstract.
SLE-ASSOCIATED RISK VARIANT REGULATES IRF5 EXPRESSION 581
23265205, 2023, 4, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/art.42390 by Affiliated Hospitals, Wiley Online Library on [31/03/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
SLE pathogenesis. Previous studies have demonstrated that IRF5
can act as a therapeutic target for SLE intervention (23,24) and
that inhibition of IRF5 could strongly down-regulate the expres-
sion of proinammatory cytokine and type I IFNs, thereby improv-
ing the symptoms of SLE. Since the rs4728142-containing region
acts as an enhancer modulating the expression of IRF5 in mono-
cytes, we speculated that KRAB/dCas9 that targets the
rs4728142-containing region could decrease proinammatory
cytokine and type I IFN production by reducing the expression of
IRF5. To test this hypothesis, we rst delivered the KRAB/dCas9
system (targeting the rs4728142-containing region) to the U937
cells. Subsequently, the cells were stimulated with R837, a
TLR-7 agonist that can lead to lupus-like autoimmunity (46), and
cytokine expression induced by R837 was detected.
As expected, the expression of proinammatory cyto-
kines and type I IFNs, such as IL6 and IFN
β
,wassignicantly
reduced (Figure 5AC). Next, we attempted to apply the
KRAB/dCas9 system to SLE monocytes to test the disease
intervention effect. We recruited SLE patients and isolated
the CD14+ monocytes. Next, the KRAB/dCas9 system was
transduced into these cells using the Neon Transfection
System (Figure 5D). We found that IRF5 expression was
effectively inhibited (Figure 5E), and IL6 and IFN
β
expression
were also reduced (Figures 5F and G). Taken together,
SLE patients’ monocytes SLE patients’ monocytesSLE patients’ monocytes
ABC
E
FG
D
sgNC sg1
0.0
0.5
1.0
1.5 IRF5
Relative expression
**
sgNC sg1
0.0
0.5
1.0
1.5 IL6
Relative expression
**
sgNC sg1
0.0
0.5
1.0
1.5
IFNβ
Relative expression
**
rs4728142
Monocytes
High IRF5 expression
High IL6
High IFNβ
Monocytes
Decreased IRF5 expression
Decreased IL6
Decreased IFNβ
Target rs4728142-containing region by CRISPRi
dCas9 KRAB
IRF5
sgRNA
SLE patients
sgNC sgRNA
0.0
0.5
1.0
1.5
IRF5
Relative expression
sgNC sgRNA
0.0
0.5
1.0
1.5
IL6
Relative expression
sgNC sgRNA
0.0
0.5
1.0
1.5
IFNβ
Relative expression
✱✱
Figure 5. Targeting the rs4728142-containing region inhibits production of proinammatory cytokines and type I IFNs by regulating IRF5 expres-
sion. AC, KRAB/dCas9 system targeting rs4728142 site inhibits the expression of IRF5,IL6, and IFN
β
induced by R837 in U937 cells (n = 3 bio-
logic sample replicates). D, Experimental design to inhibit the production of proinammatory cytokines and type I IFNs by the CRISPR interference
(CRISPRi) system in SLE patients is shown. EG, CRISPRi therapy decreases the production of proinammatory cytokines and type I IFNs in SLE
monocytes by inhibiting IRF5 expression through targeting the rs4728142 site (n = 5 biologic sample replicates). Bars show the mean ± SEM.
*=P< 0.05; ** = P< 0.01, by Students unpaired 2-tailed t-test (AC) and by Students paired 2-tailed t-test (EG). sgNC = negative control sin-
gle-guide RNA; sgRNA = targeting rs4728142 enhancer single-guide RNA (see Figure 1for other denitions). Color gure can be viewed in the
online issue, which is available at http://onlinelibrary.wiley.com/doi/10.1002/art.42390/abstract.
HOU ET AL582
23265205, 2023, 4, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/art.42390 by Affiliated Hospitals, Wiley Online Library on [31/03/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
these ndings suggest that the rs4728142-containing region
could be a prospective target to regulate the expression
of IRF5 and modulate SLE etiopathogenesisrelevant gene
expression.
DISCUSSION
GWAS have identied hundreds of genetic variants that are
associated with SLE; however, dening the causal variant and
understanding the functional mechanism of genetic variants
underlying the pathologic process is a challenge, particularly for
the variants in the noncoding genome (5,47,48). In this study,
we identied that enhancer variant rs4728142 is a functionally
causal variant of SLE, by integrating genetic studies, epigenomic
analysis, and CRISPR editing. We investigated the functional
mechanism of rs4728142 that mediates SLE risk and found that
it may regulate IRF5 expression involving SLE pathogenesis
(Figure 6).
Genetic variants are widely involved in the regulation of
gene expression, and many genetic studies have indicated
a strong association between rs4718142 and IRF5 (17,18).
Through analysis of the epigenetic mark around the
rs4728142-containing region, we have demonstrated that the
rs4728142-harboring region has a potential regulatory role.
Using CRISPR/Cas9-mediated deletion, CRISPRa, and CRIS-
PRi, we identied that the rs4728142-containing region func-
tions as an enhancer regulating the expression of IRF5 in
monocytes. Notably, eQTL data and epigenomic analysis
revealed that the rs4728142-containing region also has poten-
tial regulatory functions in B cells and T cells. However, whether
this region could regulate IRF5 expression in these 2 cell types
still needs to be studied.
Expression QTL studies suggest an association between
rs4728142 alleles and IRF5 expression. Using CRISPR/
Cas9-mediated HDR, we constructed cell clones harboring differ-
ent rs4728142 alleles and conrmed the allele-specic regulation
of IRF5 expression. Furthermore, our ndings revealed that tran-
scription factor ZBTB3 preferentially binds to the rs4728142 risk
allele, which might alter the chromatin state and thus lead to
genotype-dependent expression of IRF5, which is consistent with
a previous study that used electrophoretic mobility shift assays
(18). Our ndings conrm the link between enhancer variant
rs4728142 and IRF5 for the rst time and provide a plausible
mechanism by which rs4728142 modulates IRF5 expression.
SLE is accompanied by dysregulated production of proin-
ammatory cytokine and type I IFNs (49,50), and IRF5 plays a crit-
ical role in mediating the production of proinammatory cytokines
and type I IFNs (22). Here, we directed the CRISPRi system to the
rs4728142-harboring region, effectively down-regulating expres-
sion of proinammatory cytokines and type I IFNs by inhibiting
IRF5 expression in SLE monocytes, which indicates that targeting
the rs4872142 enhancer region might attenuate SLE develop-
ment. Our study provides new insights into the therapy of human
disease by targeting the enhancers of disease-critical genes. In
summary, we have provided evidence that the SLE-associated
enhancer risk variant regulates IRF5 expression and proves useful
in the treatment of SLE by targeting noncoding sequences.
ACKNOWLEDGMENT
We thank the participants in this study.
AUTHOR CONTRIBUTIONS
All authors were involved in drafting the article or revising it critically
for important intellectual content, and all authors approved the nal ver-
sion to be published. Dr. Shen had full access to all of the data in the
study and takes responsibility for the integrity of the data and the accu-
racy of the data analysis.
Study conception and design. Hou, Ye, N. Shen.
Acquisition of data. Hou, Zhou, Xu, Zhihua Yin, Zhu, Zhang, Cui, Ma,
Tang, Cheng, Y. Shen, Chen, Zou, Wang, Zihang Yin, Guo, Ding, Ye,
N. Shen.
Analysis and interpretation of data. Hou, Zhou, Xu, Zhihua Yin, Ye,
N. Shen.
GCC
G
GTG
ZBTB3
GCCAGTG
ZBTB3
IRF
5
IRF
5
3.7 kb 3.7 kb
high IRF5
high IL-6
high IFNβ
low IRF5
low IL-6
low IFNβ
rs4728142 A/G
SLE risk allele SLE protective allele
Figure 6. The proposed model for rs4728142 alleles mediates SLE risk. The rs4728142-A risk allele has a high binding afnity with transcription
factor zinc nger and BTB domaincontaining protein 3 (ZBTB3), resulting in the high expression of IRF5 and increasing SLE risk. In contrast, the
rs4728142-G nonrisk allele has a low binding afnity with ZBTB3, leading to the reduced expression of IRF5 and decreasing SLE risk. See Figure 1
for other denitions. Color gure can be viewed in the online issue, which is available at http://onlinelibrary.wiley.com/doi/10.1002/art.42390/abstract.
SLE-ASSOCIATED RISK VARIANT REGULATES IRF5 EXPRESSION 583
23265205, 2023, 4, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/art.42390 by Affiliated Hospitals, Wiley Online Library on [31/03/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
REFERENCES
1. Tsokos GC, Lo MS, Costa Reis P, et al. New insights into the immuno-
pathogenesis of systemic lupus erythematosus [review]. Nat Rev
Rheumatol 2016;12:71630.
2. Bentham J, Morris DL, Graham DS, et al. Genetic association analy-
ses implicate aberrant regulation of innate and adaptive immunity
genes in the pathogenesis of systemic lupus erythematosus. Nat
Genet 2015;47:145764.
3. Ramos PS, Shedlock AM, Langefeld CD. Genetics of autoimmune
diseases: insights from population genetics. J Hum Genet 2015;60:
65764.
4. Wang YF, Zhang Y, Lin Z, et al. Identication of 38 novel loci for sys-
temic lupus erythematosus and genetic heterogeneity between
ancestral groups. Nat Commun 2021;12:772.
5. Yin X, Kim K, Suetsugu H, et al. Meta-analysis of 208370 East Asians
identies 113 susceptibility loci for systemic lupus erythematosus.
Ann Rheum Dis 2021;80:63240.
6. Hnisz D, Brian JA, Tong IL, et al. Super-Enhancers in the control of cell
identity and disease. Cell 2013;155:93447.
7. Nasser J, Bergman DT, Fulco CP, et al. Genome-wide enhancer
maps link risk variants to disease genes. Nature 2021;593:23843.
8. Bulger M, Groudine M. Functional and mechanistic diversity of distal
transcription enhancers. Cell 2011;144:32739.
9. Heinz S, Romanoski CE, Benner C, et al. The selection and function of
cell type-specic enhancers [review]. Nat Rev Mol Cell Bio 2015;
16:144.
10. Hou G, Harley IT, Lu X, et al. SLE non-coding genetic risk variant
determines the epigenetic dysfunction of an immune cell specic
enhancer that controls disease-critical microRNA expression. Nat
Commun 2021;12:135.
11. Zhou T, Zhu X, Ye Z, et al. Lupus enhancer risk variant causes dysreg-
ulation of IRF8 through cooperative lncRNA and DNA methylation
machinery. Nat Commun 2022;13:1855.
12. Gupta RM, Hadaya J, Trehan A, et al. A genetic variant associated
with ve vascular diseases is a distal regulator of endothelin-1 gene
expression. Cell 2017;170:52233.
13. Soldner F, Stelzer Y, Shivalila CS, et al. Parkinson-associated risk var-
iant in distal enhancer of alpha-synuclein modulates target gene
expression. Nature 2016;533:959.
14. Graham RR, Kozyrev SV, Baechler EC, et al. A common haplotype of
interferon regulatory factor 5 (IRF5) regulates splicing and expression
and is associated with increased risk of systemic lupus erythemato-
sus. Nat Genet 2006;38:5505.
15. Kozyrev SV, Lewen S, Reddy PM, et al. Structural insertion/deletion
variation in IRF5 is associated with a risk haplotype and denes the
precise IRF5 isoforms expressed in systemic lupus erythematosus.
Arthritis Rheum 2007;56:123441.
16. Graham RR, Kyogoku C, Sigurdsson S, et al. Three functional variants
of IFN regulatory factor 5 (IRF5) dene risk and protective haplotypes
for human lupus. Proc Natl Acad Sci U S A 2007;104:675863.
17. Sigurdsson S, Goring HH, Kristjansdottir G, et al. Comprehensive
evaluation of the genetic variants of interferon regulatory factor
5 (IRF5) reveals a novel 5 bp length polymorphism as strong risk factor
for systemic lupus erythematosus. Hum Mol Genet 2008;17:87281.
18. Kottyan LC, Zoller EE, Bene J, et al. The IRF5-TNPO3 association with
systemic lupus erythematosus has two components that other auto-
immune disorders variably share. Hum Mol Genet 2015;24:58296.
19. Savitsky DA, Yanai H, Tamura T, et al. Contribution of IRF5 in B cells to
the development of murine SLE-like disease through its transcriptional
control of the IgG2a locus. Proc Nat Acad Sci 2010;107:101549.
20. Ban T, Sato GR, Nishiyama A, et al. Lyn kinase suppresses the tran-
scriptional activity of IRF5 in the TLR-MyD88 pathway to restrain the
development of autoimmunity. Immunity 2016;45:31932.
21. Feng D, Stone RC, Eloranta ML, et al. Genetic variants and disease-
associated factors contribute to enhanced interferon regulatory factor
5 expression in blood cells of patients with systemic lupus erythema-
tosus. Arthritis and Rheum 2010;62:56273.
22. Lazzari E, Jefferies CA. IRF5-mediated signaling and implications for
SLE. Clin Immunol 2014;153:34352.
23. Ban T, Kikuchi M, Sato GR, et al. Genetic and chemical inhibition of
IRF5 suppresses pre-existing mouse lupus-like disease. Nat Com-
mun 2021;12:4379.
24. Song S, De S, Nelson V, et al. Inhibition of IRF5 hyperactivation pro-
tects from lupus onset and severity. J Clin Invest 2020;130:670017.
25. Graham DS, Manku H, Wagner S, et al. Association of IRF5 in UK SLE
families identies a variant involved in polyadenylation. Hum Mol
Genet 2007;16:57991.
26. Hochberg MC, for the Diagnostic and Therapeutic Criteria Committee
of the American College of Rheumatology. Updating the American
College of Rheumatology revised criteria for the classication of sys-
temic lupus erythematosus [letter]. Arthritis Rheum 1997;40:1725.
27. MacArthur J, Bowler E, Cerezo M, et al. The new NHGRI-EBI catalog
of published genome-wide association studies (GWAS Catalog).
Nucleic Acids Res 2016;45:D896901.
28. Tangtanatakul P, Thumarat C, Satproedprai N, et al. Meta-analysis of
genome-wide association study identies FBN2 as a novel locus
associated with systemic lupus erythematosus in Thai population.
Arthritis Res Ther 2020;22:185.
29. Yang W, Tang H, Zhang Y, et al. Meta-analysis followed by replication
identies loci in or near CDKN1B, TET3, CD80, DRAM1, and ARID5B
as associated with systemic lupus erythematosus in Asians.
Am J Hum Genet 2013;92:4151.
30. Han JW, Zheng HF, Cui Y, et al. Genome-wide association study in a
Chinese Han population identies nine new susceptibility loci for sys-
temic lupus erythematosus. Nat Genet 2009;41:12347.
31. Alarc
on-Riquelme ME, Ziegler JT, Molineros J, et al. Genome-wide
association study in an Amerindian ancestry population reveals novel
systemic lupus erythematosus risk loci and the role of European
admixture. Arthritis Rheumatol 2016;68:93243.
32. Lessard CJ, Sajuthi S, Zhao J, et al. Identication of a systemic lupus
erythematosus risk locus spanning ATG16L2, FCHSD2, and P2RY2
in Koreans. Arthritis Rheumatol 2016;68:11971209.
33. Langefeld CD, Ainsworth HC, Graham DS, et al. Transancestral map-
ping and genetic load in systemic lupus erythematosus. Nat Commun
2017;8:16021.
34. Morris DL, Sheng Y, Zhang Y, et al. Genome-wide association meta-
analysis in Chinese and European individuals identies ten new loci
associated with systemic lupus erythematosus. Nat Genet 2016;48:
9406.
35. Gateva V, Sandling JK, Hom G, et al. A large-scale replication study
identies TNIP1, PRDM1, JAZF1, UHRF1BP1 and IL10 as risk loci
for systemic lupus erythematosus. Nat Genet 2009;41:122833.
36. Moore JE, Purcaro MJ, Pratt HE, et al. Expanded encyclopaedias of
DNA elements in the human and mouse genomes. Nature 2020;
583:699710.
37. Watanabe K, Stringer S, Frei O, et al. A global overview of pleiotropy
and genetic architecture in complex traits. Nat Genet 2019;51:
133948.
38. Ge T, Chen CY, Neale BM, et al. Phenome-wide heritability analysis of
the UK Biobank. PLoS Genet 2017;13:e1006711.
39. Boyle AP, Hong EL, Hariharan M, et al. Annotation of functional varia-
tion in personal genomes using RegulomeDB. Genome Res 2012;22:
17907.
HOU ET AL584
23265205, 2023, 4, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/art.42390 by Affiliated Hospitals, Wiley Online Library on [31/03/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
40. The GTEx Consortium. The GTEx Consortium atlas of genetic regula-
tory effects across human tissues. Science 2020,369:131830.
41. Ota M, Nagafuchi Y, Hatano H, et al. Dynamic landscape of immune
cell-specic gene regulation in immune-mediated diseases. Cell
2021;184:300621.
42. Abramov S, Boytsov A, Bykova D, et al. Landscape of allele-specic
transcription factor binding in the human genome. Nat Commun
2021;12:2751.
43. Fornes O, Castro-Mondragon JA, Khan A, et al. JASPAR 2020:
update of the open-access database of transcription factor binding
proles. Nucleic Acids Res 2020;48:D8792.
44. Weirauch MT, Yang A, Albu M, et al. Determination and inference of
eukaryotic transcription factor sequence specicity. Cell 2014;158:
143143.
45. Lambert SA, Jolma A, Campitelli LF, et al. The human transcription
factors. Cell 2018;172:65065.
46. Yokogawa M, Takaishi M, Nakajima K, et al. Epicutaneous application
of toll-like receptor 7 agonists leads to systemic autoimmunity in wild-
type mice: a new model of systemic Lupus erythematosus. Arthritis
Rheumatol 2014;66:694706.
47. Chen L, Wang YF, Liu L, et al. Genome-wide assessment of genetic
risk for systemic lupus erythematosus and disease severity. Hum
Mol Genet 2020;29:174556.
48. Sharon E, Chen SA, Khosla NM, et al. Functional genetic variants
revealed by massively parallel precise genome editing. Cell 2018;
175:54457.
49. Umare V, Pradhan V, Nadkar M. Effect of proinammatory cytokines
(IL-6, TNF-α, and IL-1β) on clinical manifestations in Indian SLE
patients. Mediators Inamm 2014;2014:385297.
50. Pacheco Y, Acosta-Ampudia Y, Ramírez-Santana C, et al. Cytokines
and inammatory mediators in systemic lupus erythematosus. Eur
Med J Rheumatol 2018;5:8392.
SLE-ASSOCIATED RISK VARIANT REGULATES IRF5 EXPRESSION 585
23265205, 2023, 4, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/art.42390 by Affiliated Hospitals, Wiley Online Library on [31/03/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
... The findings revealed a significant correlation between the rs4728142 allele and heightened IRF5 expression, highlighting the rs4728142 locus as an enhancer regulating IRF5 expression. Furthermore, CRISPRi interference on this enhancer led to a decrease in the production of disease-related cytokines [12]. Therefore, utilizing CRISPR solely to intervene in the expression of associated genes can serve as a means to regulate the production of pathogenic cytokines in patients and be incorporated into therapeutic research purposes. ...
Article
Autoimmune diseases (ADs) result from the immune system’s misidentification of self-antigens, leading to harmful responses and pathological states. The CRISPR/Cas9 system, a groundbreaking gene-editing technology, offers promising avenues for addressing ADs. Drawing inspiration from prokaryotic immune mechanisms, CRISPR/Cas9 precisely targets and edits foreign DNA, providing unprecedented genome-editing capabilities. Manipulating immune cell genetics or regulating relevant protein expression with CRISPR/Cas9 shows significant therapeutic potential in managing ADs. Recent research and clinical studies emphasize the valuable role of CRISPR/Cas9 in understanding the underlying pathogenic mechanisms of ADs, especially concerning non-viral triggers. Through strategic and targeted approaches, substantial progress is anticipated in comprehending and managing ADs. CRISPR/Cas9 emerges as a safe and efficient tool for tackling a wide range of ADs with diverse etiologies, offering transformative advancements in treatment strategies. This technology holds immense promise in reshaping the landscape of autoimmune disease management, providing hope for improved outcomes and enhanced quality of life for affected individuals.
... The most compelling evidence suggesting the direct involvement of the IFN-I pathway in the development and progression of GD comes from patients diagnosed with multiple sclerosis, hepatitis C, and some types of cancer, all developing glomerular lesions after receiving recombinant IFN therapy [16][17][18][19]. Moreover, patients with inborn errors causing enhanced IFN-I production such as Aicardi-Goutières syndrome [20], STING-associated vasculopathy with onset in infancy (SAVI) [21,22], and systemic lupus erythematosus (SLE) [23] also present with glomerular lesions. ...
Article
Full-text available
The field of nephrology has recently directed a considerable amount of attention towards the stimulator of interferon genes (STING) molecule since it appears to be a potent driver of chronic kidney disease (CKD). STING and its activator, the cyclic GMP-AMP synthase (cGAS), along with intracellular RIG-like receptors (RLRs) and toll-like receptors (TLRs), are potent inducers of type I interferon (IFN-I) expression. These cytokines have been long recognized as part of the mechanism used by the innate immune system to battle viral infections; however, their involvement in sterile inflammation remains unclear. Mounting evidence pointing to the involvement of the IFN-I pathway in sterile kidney inflammation provides potential insights into the complex interplay between the innate immune system and damage to the most sensitive segment of the nephron, the glomerulus. The STING pathway is often cited as one cause of renal disease not attributed to viral infections. Instead, this pathway can recognize and signal in response to host-derived nucleic acids, which are also recognized by RLRs and TLRs. It is still unclear, however, whether the development of renal diseases depends on subsequent IFN-I induction or other processes involved. This review aims to explore the main endogenous inducers of IFN-I in glomerular cells, to discuss what effects autocrine and paracrine signaling have on IFN-I induction, and to identify the pathways that are implicated in the development of glomerular damage.
... [11][12][13][14] Among the inborn errors causing enhanced IFN-I production are Aicardi-Goutières syndrome, 15 STING-associated vasculopathy with onset in infancy (SAVI), 16,17 and systemic lupus erythematosus (SLE). 18 These genetic diseases all present with glomerular injury. ...
Preprint
Full-text available
The field of nephrology has recently directed a considerable amount of attention towards the stimulator of interferon genes (STING) molecule since it appears to be a potent driver of chronic kidney disease (CKD). STING and its activator, the cyclic GMP-AMP synthase (cGAS) are among the most relevant inducers that promote the expression of type I interferons (IFN-Is). These cytokines have been long recognized as part of the mechanism used by the innate immune system to battle viral infections; however, their involvement in sterile inflammation remains unclear. Mounting evidence pointing to the involvement of the IFN-I pathway in sterile kidney inflammation provides potential insights into the complex interplay between the innate immune system and damage to the most sensitive segment of the nephron, the glomerulus. The STING pathway is often cited as the cause of renal disease originating from non-infection related caused by the induction of IFN-I expression. However, other receptors have been implicated in this process, mainly by recognizing host-derived nucleic acids. This review explores the main endogenous inducers of IFN-I in glomerular cells, discusses what exposure to the main autocrine and paracrine cytokines has on these cells, and identifies the pathways that are implicated in the development of glomerular damage.
... using LDpop online tool on 1000 Genomes European populations [26]). An enhancer variant rs4728142, affecting IRF5 expression and causal in the association with systemic lupus erythematosus [28], is also in high linkage disequilibrium with some of the SNPS we studied (r²=0.71 both with rs4731532 and rs3807306 in European populations). ...
Article
Full-text available
Inflammation has been associated with renal diseases. The Interferon Regulatory Factor (IRF)-5 is a key transcription factor in the pro-inflammatory polarization of M1-like macrophages. GWAS have reported that the IRF5 locus is associated with autoimmune diseases and with the estimated glomerular filtration rate (eGFR). We study whether allelic variations in IRF5 are associated with the incidence of chronic kidney disease (CKD) in a general population. We genotyped eleven IRF5 SNPs in the French D.E.S.I.R. cohort from the general population (n = 4820). Associations of SNPs with baseline renal parameters were assessed. Data were analyzed for three endpoints during a 9-year follow-up, incidence of:at least stage 3 CKD, the KDIGO criterion “certain drop in eGFR”, and incidence of micro/macro albuminuria. In the cross-sectional analysis, rs10954213 and rs10954214 were associated with eGFR and rs1874328 with urinary albumin/creatinine ratio (ACR). Rs3807306, rs11761199, rs78658945, rs1874328, rs10954213 and rs11770589 were associated with the incidence of stage 3 CKD in multi-adjusted models. Rs4731532, rs3807306, and rs11761199 were associated with the incidence of CKD defined by the KDIGO. Rs4731532, rs3807306, rs11761199 and rs79288514 were associated with the incidence of micro/macro albuminuria. Our results support the hypothesis of the importance of IRF5 mediated macrophage polarization in the etiology of CKD.
... Interferon regulatory factor 5 (IRF5) is involved in a variety of inflammatory signaling pathways and mediates interferon Rare Variants in Systemic Lupus Erythematosus activation and apoptosis through complex transcriptional regulation 85 . Recently, rs4728142 variant was identified as a functionally causal variant of SLE, by integrating genetic studies, epigenomic analysis and CRISPR editing 86 . Expression QTL studies suggest an association between rs4728142 alleles and IRF5 expression involving SLE pathogenesis 85 . ...
Article
Full-text available
The clinical and genotypic characterization of autoimmune diseases, including systemic lupus erythematosus (SLE), has made great strides recently as a result of tremendous advancements in gene sequencing technologies. Systemic lupus erythematosus is a complex multisystem disease characterized by high clinical variability due to abnormalities in both the innate and adaptive immune systems. Several genetic variants as well as environmental and hormonal factors have been identified, but the etiology of lupus is not fully understood yet. The ability of genome-wide association studies to scan thousands of individuals has enabled researchers to associate thousands of common variants to lupus. Common polymorphisms may jointly predispose to lupus, but their individual impact on the disease is minimal. It's becoming progressively more evident that rare mutations have a far higher influence. The role of rare variation in lupus has been the subject of intense research. Several approaches including genotyped-based follow-up of the variants in families, hierarchical screening, and imputation, have been applied to elucidate their functional involvement. Nevertheless, due to their rarity and the absence of standardized methodology, rare variants are still challenging to study. Most lupus patients present a polygenic form of the disease, which is defined by the complex interplay between genetic and environmental factors. Still, certain lupus patients and patients with lupus-like phenotypes might be affected by monogenic lupus, a group of disorders largely caused by individual gene mutation abnormalities. Although monogenic lupus is rare, it has been associated with a sizable number of genes in a range of pathways, mostly resulting in early-onset phenotypes. The study of rare variants causing monogenic lupus has resulted in incredibly useful breakthroughs in our understanding of the function of rare variants in the disease, nonetheless further research is still required.
... Targeting disease-associated enhancers is a novel potential strategy for disease therapy. 6,38 Here, we found that targeting the SLE-associated 32.5-kb enhancer with the CRISPRa system effectively down-regulated the activity of the IFN pathway and the production of inflammatory cytokines, suggesting a promising target for therapy in the future. Deletion of the 32.5-kb enhancer strongly reduced miR-146a expression in U-937 monocytes (n = 6, biologically independent samples). ...
Article
Full-text available
Objective The diminished expression of microRNA‐146a (miR‐146a) in systemic lupus erythematosus (SLE) contributes to the aberrant activation of the interferon pathway. Despite its significance, the underlying mechanism driving this reduced expression remains elusive. Considering the integral role of enhancers in steering gene expression, our study seeks to pinpoint the SLE‐affected enhancers responsible for modulating miR‐146a expression. Additionally, we aim to elucidate the mechanisms by which these enhancers influence the contribution of miR‐146a to the activation of the interferon pathway. Methods Circular chromosome conformation capture sequencing and epigenomic profiles were applied to identify candidate enhancers of miR‐146a. CRISPR activation was performed to screen functional enhancers. Differential analysis of chromatin accessibility was used to identify SLE‐dysregulated enhancers, and the mechanism underlying enhancer dysfunction was investigated by analyzing transcription factor binding. The therapeutic value of a lupus‐related enhancer was further evaluated by targeting it in the peripheral blood mononuclear cells (PBMCs) of patients with SLE through a CRISPR activation approach. Results We identified shared and cell‐specific enhancers of miR‐146a in distinct immune cells. An enhancer 32.5 kb downstream of miR‐146a possesses less accessibility in SLE, and its chromatin openness was negatively correlated with SLE disease activity. Moreover, CCAAT/enhancer binding protein α, a down‐regulated transcription factor in patients with SLE, binds to the 32.5‐kb enhancer and induces the epigenomic change of this locus. Furthermore, CRISPR‐based activation of this enhancer in SLE PBMCs could inhibit the activity of interferon pathway. Conclusion Our work defines a promising target for SLE intervention. We adopted integrative approaches to define cell‐specific and functional enhancers of the SLE critical gene and investigated the mechanism underlying its dysregulation mediated by a lupus‐related enhancer. image
... In ImmuNexUT, on the other hand, rs3757387 in the enhancer region of IRF5 exhibited the strongest eQTL effect in neutrophils, plasmacytoid dendritic cells, CD4 + T cells, and CD8 + T cells (https://www.immunexut.org/). Hou et al. reanalyzed the SLE GWAS results, focusing on enhancers near IRF5, and found that rs4728142 located within an enhancer in linkage disequilibrium with rs3757387 was associated most strongly with SLE [42]. They reported that the SLE-risk genotype of rs4728142 enhanced transcription factor ZBTB3, and deletion of the rs4728142 region in monocytes reduced the expression of IRF5, IL6, and IFNB. ...
Article
Full-text available
Systemic lupus erythematosus (SLE) is caused by a combination of genetic and environmental factors. Recently, analysis of a functional genome database of genetic polymorphisms and transcriptomic data from various immune cell subsets revealed the importance of the oxidative phosphorylation (OXPHOS) pathway in the pathogenesis of SLE. In particular, activation of the OXPHOS pathway is persistent in inactive SLE, and this activation is associated with organ damage. The finding that hydroxychloroquine (HCQ), which improves the prognosis of SLE, targets toll-like receptor (TLR) signaling upstream of OXPHOS suggests the clinical importance of this pathway. IRF5 and SLC15A4, which are regulated by polymorphisms associated with SLE susceptibility, are functionally associated with OXPHOS as well as blood interferon activity and metabolome. Future analyses of OXPHOS-associated disease-susceptibility polymorphisms, gene expression, and protein function may be useful for risk stratification of SLE.
Article
Understanding the influence of cis ‐regulatory elements on gene regulation poses numerous challenges given complexities stemming from variations in transcription factor (TF) binding, chromatin accessibility, structural constraints, and cell‐type differences. This review discusses the role of gene regulatory networks in enhancing understanding of transcriptional regulation and covers construction methods ranging from expression‐based approaches to supervised machine learning. Additionally, key experimental methods, including MPRAs and CRISPR‐Cas9‐based screening, which have significantly contributed to understanding TF binding preferences and cis‐regulatory element functions, are explored. Lastly, the potential of machine learning and artificial intelligence to unravel cis ‐regulatory logic is analyzed. These computational advances have far‐reaching implications for precision medicine, therapeutic target discovery, and the study of genetic variations in health and disease.
Article
Full-text available
Three-dimensional (3D) genomics is an emerging field of research that investigates the relationship between gene regulatory function and the spatial structure of chromatin. Chromatin folding can be studied using chromosome conformation capture (3C) technology and 3C-based derivative sequencing technologies, including chromosome conformation capture-on-chip (4C), chromosome conformation capture carbon copy (5C), and high-throughput chromosome conformation capture (Hi-C), which allow scientists to capture 3D conformations from a single site to the entire genome. A comprehensive analysis of the relationships between various regulatory components and gene function also requires the integration of multi-omics data such as genomics, transcriptomics, and epigenomics. 3D genome folding is involved in immune cell differentiation, activation, and dysfunction and participates in a wide range of diseases, including autoimmune diseases. We describe hierarchical 3D chromatin organization in this review and conclude with characteristics of C-techniques and multi-omics applications of the 3D genome. In addition, we describe the relationship between 3D genome structure and the differentiation and maturation of immune cells and address how changes in chromosome folding contribute to autoimmune diseases.
Article
Full-text available
Despite strong evidence that human genetic variants affect the expression of many key transcription factors involved in autoimmune diseases, establishing biological links between non-coding risk variants and the gene targets they regulate remains a considerable challenge. Here, we combine genetic, epigenomic, and CRISPR activation approaches to screen for functional variants that regulate IRF8 expression. We demonstrate that the locus containing rs2280381 is a cell-type-specific enhancer for IRF8 that spatially interacts with the IRF8 promoter. Further, rs2280381 mediates IRF8 expression through enhancer RNA AC092723.1, which recruits TET1 to the IRF8 promoter regulating IRF8 expression by affecting methylation levels. The alleles of rs2280381 modulate PU.1 binding and chromatin state to regulate AC092723.1 and IRF8 expression differentially. Our work illustrates an integrative strategy to define functional genetic variants that regulate the expression of critical genes in autoimmune diseases and decipher the mechanisms underlying the dysregulation of IRF8 expression mediated by lupus risk variants. The functional effects of genetic loci associated with autoimmune disease are not well understood. By dissecting an autoimmune disease genetic locus, the authors define an immune cell-type-specific enhancer and the molecular mechanisms underlying the dysregulation of IRF8 expression by lupus risk variants.
Article
Full-text available
The transcription factor IRF5 has been implicated as a therapeutic target for the autoimmune disease systemic lupus erythematosus (SLE). However, IRF5 activation status during the disease course and the effects of IRF5 inhibition after disease onset are unclear. Here, we show that SLE patients in both the active and remission phase have aberrant activation of IRF5 and interferon-stimulated genes. Partial inhibition of IRF5 is superior to full inhibition of type I interferon signaling in suppressing disease in a mouse model of SLE, possibly due to the function of IRF5 in oxidative phosphorylation. We further demonstrate that inhibition of IRF5 via conditional Irf5 deletion and a newly developed small-molecule inhibitor of IRF5 after disease onset suppresses disease progression and is effective for maintenance of remission in mice. These results suggest that IRF5 inhibition might overcome the limitations of current SLE therapies, thus promoting drug discovery research on IRF5 inhibitors.
Article
Full-text available
Sequence variants in gene regulatory regions alter gene expression and contribute to phenotypes of individual cells and the whole organism, including disease susceptibility and progression. Single-nucleotide variants in enhancers or promoters may affect gene transcription by altering transcription factor binding sites. Differential transcription factor binding in heterozygous genomic loci provides a natural source of information on such regulatory variants. We present a novel approach to call the allele-specific transcription factor binding events at single-nucleotide variants in ChIP-Seq data, taking into account the joint contribution of aneuploidy and local copy number variation, that is estimated directly from variant calls. We have conducted a meta-analysis of more than 7 thousand ChIP-Seq experiments and assembled the database of allele-specific binding events listing more than half a million entries at nearly 270 thousand single-nucleotide polymorphisms for several hundred human transcription factors and cell types. These polymorphisms are enriched for associations with phenotypes of medical relevance and often overlap eQTLs, making candidates for causality by linking variants with molecular mechanisms. Specifically, there is a special class of switching sites, where different transcription factors preferably bind alternative alleles, thus revealing allele-specific rewiring of molecular circuitry.
Article
Full-text available
Genome-wide association studies (GWAS) have identified thousands of noncoding loci that are associated with human diseases and complex traits, each of which could reveal insights into the mechanisms of disease1. Many of the underlying causal variants may affect enhancers2,3, but we lack accurate maps of enhancers and their target genes to interpret such variants. We recently developed the activity-by-contact (ABC) model to predict which enhancers regulate which genes and validated the model using CRISPR perturbations in several cell types4. Here we apply this ABC model to create enhancer–gene maps in 131 human cell types and tissues, and use these maps to interpret the functions of GWAS variants. Across 72 diseases and complex traits, ABC links 5,036 GWAS signals to 2,249 unique genes, including a class of 577 genes that appear to influence multiple phenotypes through variants in enhancers that act in different cell types. In inflammatory bowel disease (IBD), causal variants are enriched in predicted enhancers by more than 20-fold in particular cell types such as dendritic cells, and ABC achieves higher precision than other regulatory methods at connecting noncoding variants to target genes. These variant-to-function maps reveal an enhancer that contains an IBD risk variant and that regulates the expression of PPIF to alter the membrane potential of mitochondria in macrophages. Our study reveals principles of genome regulation, identifies genes that affect IBD and provides a resource and generalizable strategy to connect risk variants of common diseases to their molecular and cellular functions. Mapping enhancer regulation across human cell types and tissues illuminates genome function and provides a resource to connect risk variants for common diseases to their molecular and cellular functions.
Article
Full-text available
Systemic lupus erythematosus (SLE), a worldwide autoimmune disease with high heritability, shows differences in prevalence, severity and age of onset among different ancestral groups. Previous genetic studies have focused more on European populations, which appear to be the least affected. Consequently, the genetic variations that underlie the commonalities, differences and treatment options in SLE among ancestral groups have not been well elucidated. To address this, we undertake a genome-wide association study, increasing the sample size of Chinese populations to the level of existing European studies. Thirty-eight novel SLE-associated loci and incomplete sharing of genetic architecture are identified. In addition to the human leukocyte antigen (HLA) region, nine disease loci show clear ancestral differences and implicate antibody production as a potential mechanism for differences in disease manifestation. Polygenic risk scores perform significantly better when trained on ancestry-matched data sets. These analyses help to reveal the genetic basis for disparities in SLE among ancestral groups.
Article
Full-text available
Since most variants that impact polygenic disease phenotypes localize to non-coding genomic regions, understanding the consequences of regulatory element variants will advance understanding of human disease mechanisms. Here, we report that the systemic lupus erythematosus (SLE) risk variant rs2431697 as likely causal for SLE through disruption of a regulatory element, modulating miR-146a expression. Using epigenomic analysis, genome-editing and 3D chromatin structure analysis, we show that rs2431697 tags a cell-type dependent distal enhancer specific for miR-146a that physically interacts with the miR-146a promoter. NF-kB binds the disease protective allele in a sequence-specific manner, increasing expression of this immunoregulatory microRNA. Finally, CRISPR activation-based modulation of this enhancer in the PBMCs of SLE patients attenuates type I interferon pathway activation by increasing miR-146a expression. Our work provides a strategy to define non-coding RNA functional regulatory elements using disease-associated variants and provides mechanistic links between autoimmune disease risk genetic variation and disease etiology.
Article
Full-text available
The Genotype-Tissue Expression (GTEx) project was established to characterize genetic effects on the transcriptome across human tissues and to link these regulatory mechanisms to trait and disease associations. Here, we present analyses of the version 8 data, examining 15,201 RNA-sequencing samples from 49 tissues of 838 postmortem donors. We comprehensively characterize genetic associations for gene expression and splicing in cis and trans, showing that regulatory associations are found for almost all genes, and describe the underlying molecular mechanisms and their contribution to allelic heterogeneity and pleiotropy of complex traits. Leveraging the large diversity of tissues, we provide insights into the tissue specificity of genetic effects and show that cell type composition is a key factor in understanding gene regulatory mechanisms in human tissues. © 2020 American Association for the Advancement of Science. All rights reserved.
Article
Full-text available
Objective Systemic lupus erythematosus (SLE), an autoimmune disorder, has been associated with nearly 100 susceptibility loci. Nevertheless, these loci only partially explain SLE heritability and their putative causal variants are rarely prioritised, which make challenging to elucidate disease biology. To detect new SLE loci and causal variants, we performed the largest genome-wide meta-analysis for SLE in East Asian populations. Methods We newly genotyped 10 029 SLE cases and 180 167 controls and subsequently meta-analysed them jointly with 3348 SLE cases and 14 826 controls from published studies in East Asians. We further applied a Bayesian statistical approach to localise the putative causal variants for SLE associations. Results We identified 113 genetic regions including 46 novel loci at genome-wide significance (p<5×10 ⁻⁸ ). Conditional analysis detected 233 association signals within these loci, which suggest widespread allelic heterogeneity. We detected genome-wide associations at six new missense variants. Bayesian statistical fine-mapping analysis prioritised the putative causal variants to a small set of variants (95% credible set size ≤10) for 28 association signals. We identified 110 putative causal variants with posterior probabilities ≥0.1 for 57 SLE loci, among which we prioritised 10 most likely putative causal variants (posterior probability ≥0.8). Linkage disequilibrium score regression detected genetic correlations for SLE with albumin/globulin ratio (r g =−0.242) and non-albumin protein (r g =0.238). Conclusion This study reiterates the power of large-scale genome-wide meta-analysis for novel genetic discovery. These findings shed light on genetic and biological understandings of SLE.
Article
Full-text available
The transcription factor interferon regulatory factor 5 (IRF5) is a central mediator of innate and adaptive immunity. Genetic variations within IRF5 associate with risk of systemic lupus erythematosus (SLE) and mice lacking Irf5 are protected from lupus onset and severity, but how IRF5 functions in the context of SLE disease progression remains unclear. Using the NZB/W F1 model of murine lupus, we show that murine Irf5 becomes hyper-activated before clinical onset. In SLE patients, IRF5 hyper-activation correlated with dsDNA titers. To test whether IRF5 hyper-activation is a targetable function, we developed novel inhibitors that are cell permeable, non-toxic and selectively bind to the inactive IRF5 monomer. Preclinical treatment of NZB/W F1 mice with inhibitor attenuated lupus pathology by reducing serum ANA, dsDNA titers and the number of circulating plasma cells, which alleviated kidney pathology and improved survival. Clinical treatment of MRL/lpr and pristane-induced mice with inhibitor led to significant reductions in dsDNA levels and improved survival. In ex vivo human studies, the inhibitor blocked SLE serum-induced IRF5 activation in healthy immune cells and reversed basal IRF5 hyper-activation in SLE immune cells. Altogether, this study provides the first in vivo clinical support for treating SLE patients with an IRF5 inhibitor.
Article
Genetic studies have revealed many variant loci that are associated with immune-mediated diseases. To elucidate the disease pathogenesis, it is essential to understand the function of these variants, especially under disease-associated conditions. Here, we performed a large-scale immune cell gene-expression analysis, together with whole-genome sequence analysis. Our dataset consists of 28 distinct immune cell subsets from 337 patients diagnosed with 10 categories of immune-mediated diseases and 79 healthy volunteers. Our dataset captured distinctive gene-expression profiles across immune cell types and diseases. Expression quantitative trait loci (eQTL) analysis revealed dynamic variations of eQTL effects in the context of immunological conditions, as well as cell types. These cell-type-specific and context-dependent eQTLs showed significant enrichment in immune disease-associated genetic variants, and they implicated the disease-relevant cell types, genes, and environment. This atlas deepens our understanding of the immunogenetic functions of disease-associated variants under in vivo disease conditions.