ArticlePDF AvailableLiterature Review

The non-coding RNA interactome in joint health and disease

Authors:

Abstract

Non-coding RNAs have distinct regulatory roles in the pathogenesis of joint diseases including osteoarthritis (OA) and rheumatoid arthritis (RA). As the amount of high-throughput profiling studies and mechanistic investigations of microRNAs, long non-coding RNAs and circular RNAs in joint tissues and biofluids has increased, data have emerged that suggest complex interactions among non-coding RNAs that are often overlooked as critical regulators of gene expression. Identifying these non-coding RNAs and their interactions is useful for understanding both joint health and disease. Non-coding RNAs regulate signalling pathways and biological processes that are important for normal joint development but, when dysregulated, can contribute to disease. The specific expression profiles of non-coding RNAs in various disease states support their roles as promising candidate biomarkers, mediators of pathogenic mechanisms and potential therapeutic targets. This Review synthesizes literature published in the past 2 years on the role of non-coding RNAs in OA and RA with a focus on inflammation, cell death, cell proliferation and extracellular matrix dysregulation. Research to date makes it apparent that ‘non-coding’ does not mean ‘non-essential’ and that non-coding RNAs are important parts of a complex interactome that underlies OA and RA.
0123456789();:
Non-coding RNAs constitute 99% of total cellular RNA
content and, alongside DNA methylation and histone
modification, represent one of three major epigenetic
mechanisms that contribute to health and disease1.
Although non-coding RNAs are encoded in DNA and
transcribed to RNA, they are not translated to protein;
however, this does not negate their important role in
regulating cellular processes. The precise mechanism of
action is dependent on the class of non-coding RNA —
short non-coding RNAs (such as microRNAs (miRNAs)),
long non-coding RNAs (lncRNAs) or circular RNAs
(circRNAs) — although all types ultimately function
to regulate the expression of specific gene targets1. As
such, non-coding RNAs are essential for establishing
and maintaining homeostatic balance in biological sys-
tems, including regulating the signalling pathways and
biological processes that govern joint development2.
Deregulation of this balance contributes to the patho-
genesis of joint diseases such as osteoarthritis (OA) and
rheumatoid arthritis (RA)3,4.
Non-coding RNAs are found in almost all joint tissues
and biofluids across different species, demonstrating
their biological importance5. In joint disease, non-coding
RNAs have been explored as potential biomarkers,
mediators of pathogenesis and therapeutic targets.
Adding to the complexity of these epigenetic regula-
tors, the different classes of non-coding RNAs can pro-
vide redundancy by targeting the same genes, work in
concert by targeting the same pathways and directly
interact to regulate gene expression6. Although this
putative ‘interactome’ of non-coding RNAs has yet
to be comprehensively explored in joint health and
disease, its elucidation is improving with the use of tech-
nologies for high-throughput profiling and integrative
computational analysis.
To demonstrate that ‘non-coding’ does not mean
‘non-essential’, in this Review we discuss literature pub-
lished in the past 2 years on non-coding RNAs in OA
and RA. We first describe the classes of non-coding
RNAs and their mechanisms of action, followed by the
role of non-coding RNAs in osteogenesis and chondro-
genesis, two vital biological processes in joint develop-
ment. We next review non-coding RNAs in OA and RA
joint tissues and biofluids, and their roles in inflam-
mation, cell death, cell proliferation and extracellular
matrix (ECM) dysregulation. Finally, we discuss the
therapeutic potential of non-coding RNAs in OA and
RA and the deep-dive efforts that will be required in
the future to unravel the complex interactions among
non-coding RNAs.
The non-coding RNA interactome
in joint health and disease
ShabanaA.Ali
1,2 ✉ , MandyJ.Peffers3, MichelleJ.Ormseth4,5, IgorJurisica6,7 and
MohitKapoor
6,8 ✉
Abstract | Non-coding RNAs have distinct regulatory roles in the pathogenesis of joint diseases
including osteoarthritis (OA) and rheumatoid arthritis (RA). As the amount of high-throughput
profiling studies and mechanistic investigations of microRNAs, long non-coding RNAs and
circular RNAs in joint tissues and biofluids has increased, data have emerged that suggest
complex interactions among non-coding RNAs that are often overlooked as critical regulators
of gene expression. Identifying these non-coding RNAs and their interactions is useful for
understanding both joint health and disease. Non-coding RNAs regulate signalling pathways
and biological processes that are important for normal joint development but, when
dysregulated, can contribute to disease. The specific expression profiles of non-coding
RNAs in various disease states support their roles as promising candidate biomarkers,
mediators of pathogenic mechanisms and potential therapeutic targets. This Review
synthesizes literature published in the past 2 years on the role of non-coding RNAs in
OA and RA with a focus on inflammation, cell death, cell proliferation and extracellular
matrix dysregulation. Research to date makes it apparent that ‘non-coding’ does not mean
‘non-essential’ and that non-coding RNAs are important parts of a complex interactome
that underlies OA and RA.
e-mail: sali14@hfhs.org;
mohit.kapoor@uhnresearch.ca
https://doi.org/10.1038/
s41584-021-00687-y
REVIEWS
Nature reviews
|
Rheumatology
0123456789();:
Classes of non-coding RNAs
Non-coding RNAs are classified on the basis of their
biogenesis, length and mechanism of action (FIG.1a).
Following transcription, non-coding RNAs are pro-
cessed to form short, long or circular non-coding RNAs
with unique secondary and tertiary structures. The first
class are short non-coding RNAs, which are fewer than
200 nucleotides in length. This class includes miRNAs,
small nucleolar RNAs (snoRNAs), small nuclear RNAs,
Piwi-interacting RNAs (piRNAs), small interfering
RNAs (siRNAs), transfer RNAs (tRNAs), tRNA-derived
fragments (tRFs) and Y RNA fragments (YRFs)5. The
most frequently studied of the short non-coding RNAs
are miRNAs. miRNA biogenesis begins with a primary
miRNA transcript from the intron, exon or intergenic
region of the host gene, followed by processing within
the nucleus to produce a precursor miRNA. After
export to the cytoplasm, the precursor miRNA under-
goes cleavage to produce the mature miRNA. Mature
miRNAs are single stranded, 18–24 nucleotides in length
and function to inhibit target gene expression through
mRNA degradation or repression of translation4 (FIG.1b).
The second class of non-coding RNAs are lncRNAs,
which are greater than 200 nucleotides in length. Similar to
short non-coding RNAs, lncRNAs function to modulate
mRNA stability and translation in the cytoplasm through
multiple mechanisms, including the post-translational
modification of target molecules7 (FIG.1c). Circular forms
of lncRNAs, or circRNAs, comprise 1–5 introns or exons
and form a covalently closed loop structure that func-
tions as an miRNA sponge, protein sponge or scaffold
for translation5,6 (FIG.1d). miRNA sponging depends
on the presence of miRNA response elements within
lncRNAs and circRNAs that can specifically bind and
sequester miRNAs, thereby blocking their activity. This
sponging is a type of competing endogenous RNA activity
andisa mechanism through which the different classes
of non-coding RNAs can directly interact. circRNAs pri-
marily function as competing endogenous RNAs, binding
to miRNA response elements and reducing the quantity
of miRNAs available to target mRNA, thereby promoting
mRNA stability or protein expression6. In this Review, we
focus on miRNAs, lncRNAs and circRNAs, as these types
of non-coding RNA have been explored the most in OA
and RA to date3,8.
Non-coding RNAs in joint development
Healthy joint development is dependent on precise
regulation of the signalling pathways that govern oste-
ogenesis and chondrogenesis, among other processes,
and if these become dysregulated, joint pathologies
can result. miRNAs, lncRNAs, circRNAs and even
piRNAs are differentially expressed during the early
stages of osteogenic and chondrogenic differentiation
in human bone marrow-derived mesenchymal stro-
mal cells and/or bone marrow-derived mesenchymal
stem cells (BMSCs), suggesting that non-coding RNAs
might affect these processes9,10. Non-coding RNAs can
also regulate important signalling pathways, including
the Wnt–β-catenin and Hedgehog signalling pathways,
which are essential for tissue induction, patterning,
growth and morphogenesis11. For example, overexpres-
sion of miR-378 in transgenic mice results in abnormal
bone formation and quality, as well as compromised
osteogenic differentiation in both mouse and human
BMSCs12. Interestingly, miR-378 targets two Wnt fam-
ily members, Wnt6 and Wnt10a, thereby attenuating
Wnt–β-catenin signalling12. These results suggest that
miRNAs might be upstream regulators of certain devel-
opmental signalling pathways, which has implications
for bone health.
Chondrogenesis is essential for endochondral and
intramembranous ossification and for tissue homeo-
stasis, and is also subject to regulation by non-coding
RNAs. The Indian Hedgehog signalling pathway is
well known to regulate chondrogenesis during normal
development13. The gene encoding Indian Hedgehog
contains two putative sites at which miR-1 can bind and
inhibit its activity, resulting in increased expression of
type II collagen and aggrecan and decreased expression
of type X collagen and matrix metalloproteinase 13 in
mouse thorax chondrocytes14. These results suggest
that miR-1 induces an anabolic effect in chondrocytes
through inhibition of the Indian Hedgehog pathway,
which is consistent with previous findings that aber-
rant activation of the Indian Hedgehog pathway can
have catabolic effects on cartilage15. Furthermore, the
transcription factor SOX9, which is critical for mes-
enchymal condensation prior to chondrogenesis, is
targeted by miR-30a to inhibit chondrogenic differenti-
ation in human BMSCs16, again demonstrating a direct
Key points
•Anincreasingbodyofliteratureonnon-codingRNAsinjointhealthanddiseasehas
revealedimportantregulatoryfunctionsthatindicatethat‘non-coding’doesnot
equateto‘non-essential’.
•Non-codingRNAs,includingmicroRNAs,longnon-codingRNAsandcircularRNAs,
candirectlyinteractandhaveco-regulatoryfunctions.
•Inosteoarthritisandrheumatoidarthritis,non-codingRNAsareimportantcontributors
topathogenesisandserveaspotentialbiomarkersandtherapeutictargets.
•Withtheemergenceofdatafromhigh-throughputstudies,detailedreportingand
accurateannotationofresultsarerequiredtointegrateindividualstudiesandenable
interrogationofthenon-codingRNAinteractome.
•Anexpandedunderstandingofthenon-codingRNAinteractomecouldreveal
essentialregulatorymechanismsandnoveltherapeuticopportunitiesfor
osteoarthritis,rheumatoidarthritisandotherrelatedjointdiseases.
Author addresses
1BoneandJointCenter,DepartmentofOrthopaedicSurgery,HenryFordHealthSystem,
Detroit,MI,USA.
2CenterforMolecularMedicineandGenetics,SchoolofMedicine,WayneState
University,Detroit,MI,USA.
3DepartmentofMusculoskeletalBiology,InstituteofLifeCourseandMedicalSciences,
UniversityofLiverpool,Liverpool,UK.
4DepartmentofResearchandDevelopment,VeteransAffairsMedicalCenter,Nashville,
TN,USA.
5DepartmentofMedicine,VanderbiltUniversityMedicalCenter,Nashville,TN,USA.
6OsteoarthritisResearchProgram,DivisionofOrthopaedics,SchroederArthritisInstitute,
KrembilResearchInstitute,UniversityHealthNetwork,Toronto,Ontario,Canada.
7DataScienceDiscoveryCentreforChronicDiseases,KrembilResearchInstitute,
UniversityHealthNetwork,Toronto,Ontario,Canada.
8DepartmentofSurgeryandDepartmentofLaboratoryMedicineandPathobiology,
UniversityofToronto,Toronto,Ontario,Canada.
www.nature.com/nrrheum
Reviews
0123456789();:
regulatory role for miRNAs in established mechanisms
that govern chondrogenesis.
Looking at interactions between classes of non-
coding RNAs in osteogenesis and chondrogenesis, evi-
dence exists of competing endogenous RNA activity.
The lncRNA LINC00707 sponges miR-145 in human
BMSCs and increases the expression of lipoprotein
receptor-related protein 5, a co-receptor for Wnt pro-
teins, thereby promoting osteogenic differentiation17.
Similarly, the lncRNA ADAMTS9-AS2 sponges miR-
942-5p in human BMSCs and increases expression of the
transcription factor SCRG1, thereby promoting chon-
drogenic differentiation18. circRNAs have also emerged
as novel orchestrators of signalling pathways that gov-
ern osteogenesis19. Relevant to development, an axis
has been identified whereby circRNA_0079201 sponges
miR-140-3p in human chondrocytes and increases
expression of the transcription factor SMAD2, thereby
suppressing cell proliferation, hypertrophy and endo-
chondral ossification20. Taken together, these examples
illustrate an important role for non-coding RNAs in gov-
erning signalling pathways and biological processes in
joint development that have implications for joint health
and disease.
Non-coding RNAs in OA and RA
Expression in joint tissues
Strong evidence exists to support cell-specific and
tissue-specific expression patterns of non-coding RNAs
in OA and RA21,22. Two studies used microarrays to
compare cartilage from patients with OA and healthy
individuals and identified 58 and 70 differentially
expressed miRNAs, respectively23,24. Beyond miRNAs,
a diverse range of non-coding RNAs have been identified
miRNA IncRNA
Immune cells
Synovial cells
Chondrocytes
Bone cells
Adipocytes
Chromatin
Cell
DNA RNA
Short non-coding RNAs
(<200 nt)
Long non-coding RNAs
(>200 nt)
snRNA, snoRNA, piRNA,
siRNA, tRNA, tRF & YRF
Blood
Synovial
fluid
Urine
Extracellular
vesicles
pre-miRNA miRNA
IncRNA circRNA
mRNA cleavage
Protein scaffolding
Translational
repression
a
b c d
Transcriptional regulation
circRNA
Synovial joint
Translational regulation
miRNA sponging
miRNA sponging
Translational
scaffolding
Protein sponging
Fig. 1 | Biogenesis and function of microRNAs, long non-coding RNAs and circular RNAs. Within the synovial joint,
several cell types can be a source of non-coding RNAs that are transcribed from DNA (part a). Non-coding RNAs can
function within the producing cell or in a target cell, and are secreted into biofluids as free molecules or within extracellular
vesicles. Potential functions for microRNAs (miRNAs) include mRNA cleavage and translational repression (part b); for long
non-coding RNAs (lncRNAs) include transcriptional regulation, translational regulation, protein scaffolding and miRNA
sponging (part c); and for circular RNAs (circRNAs) include miRNA sponging, protein sponging and translational scaffolding
(part d). nt, nucleotides; piRNA, Piwi-interacting RNA; siRNA, small interfering RNA; snRNA, small nuclear RNA; snoRNA,
small nucleolar RNA; tRF, tRNA-derived fragment; tRNA, transfer RNA; YRF, Y RNA fragment.
Nature reviews
|
Rheumatology
Reviews
0123456789();:
in primary human OA chondrocytes and cartilage,
including lncRNA MFI2-AS1, lncRNA LOXL1-AS1,
tRF-3003a and U3 snoRNA2528. In primary human
synoviocytes and synovial tissue in OA and RA, reports
have focused on lncRNAs such as MALAT1, NEAT1
and PVT1 (REFS2931). MALAT1 expression crosses tis-
sue types, being increased in both the synovium29 and
subchondral bone32 of patients with OA compared
with healthy individuals. Similarly, NEAT1 expression
crosses both tissue types and diseases, being increased
in OA cartilage30 and in RA synovium33. These examples
demonstrate a non-coding RNA expression pattern that
is not only tissue specific, but potentially also disease
specific, and support the need for further studies focused
on profiling non-coding RNAs in other tissues and cells
implicated in OA and RA, including bone34,35, adipose
tissue36, meniscus37 and macrophages38,39. Furthermore,
this profiling should take into account unique patient
endotypes40,41 and apply appropriate inclusion and exclu-
sion criteria during the selection of participants to facil-
itate the interpretation of findings and integration with
other studies.
Sequencing is the gold standard approach for iden-
tifying tissue-specific non-coding RNAs, their targets
and their interactions. In a 2019 study that compared
lesioned with preserved cartilage from patients with
knee or hip OA, RNA sequencing was used to identify
142 miRNAs and 2,387 mRNAs that were prioritized
into a regulatory network comprising 62 miRNAs that
targeted 238 mRNAs42, which showed joint-specific
expression patterns. Similarly, 1,068 mRNAs, 21miRNAs
and 395miRNA–mRNA pairs were identified in syn-
ovial tissue from patients with knee OA using RNA
sequencing43. Given the large number of candidate
non-coding RNAs identified through sequencing,
a deeper dive into the biological relevance of prioritized
candidates is required through validation studies. In syn-
ovium, canonical correlation analysis of RNA sequenc-
ing and small RNA sequencing data has been used to
identify miRNA–mRNA co-expression patterns44.
Specifically, five miRNAs and four genes were predicted
to be associated with pain in knee OA, suggesting their
potential utility as biomarkers.
Although obtaining tissue samples by biopsy might
be considered too invasive for use in biomarker detec-
tion, evidence from RA suggests that the amount of
non-coding RNAs in the circulation might differ from
that found in tissues. For example, the amount of miR-22
is increased in plasma from patients with RA compared
with that from healthy individuals and is associated with
disease activity in RA45,46, but is decreased in synovial
tissue from patients with RA compared with synovium
from healthy individuals47. It is unclear whether this
tissue-specific difference in miR-22 expression is due
to sponging (as is the case for miR-145-5p, which is
sponged by the lncRNA PVT1 in RA synovium to pro-
duce lower concentrations in the synovium than in the
serum31) or other mechanisms of non-coding RNA regu-
lation. Nevertheless, these data suggest that, in addition
to tissue-specific expression patterns, biofluid-specific
patterns of non-coding RNA expression must also be
considered.
Expression in biofluids
Non-coding RNAs can be secreted by cells either as
free RNA molecules or encapsulated into extracellu-
lar vesicles such as exosomes and can be identified in
biofluids including blood, urine and synovial fluid36,48,49
(FIG.1). Given their association with disease activity,
non-coding RNAs are thought to represent excellent
candidate biomarkers50. Non-coding RNA classes are
broadly altered in plasma from patients with RA, in
which sets of miRNAs and tRFs are enriched and sets of
YRFs are depleted compared with healthy individuals46.
Such non-coding RNA class shifts might be caused by
broad changes in RNA processing mechanisms, such as
the upregulation of Dicer and Drosha in RA peripheral
blood mononuclear cells (PBMCs)51, which are a major
source of non-coding RNAs in plasma. Surprisingly,
non-coding RNAs of microbial origin have also been
detected in human plasma; the abundance of microbial
small RNAs and specific microbial tRFs were inversely
associated with disease activity in two separate cohorts of
patients with RA and also predicted response to therapy,
suggesting that they might be useful as biomarkers52.
Non-coding RNA profiling in OA biofluids has
focused on miRNAs in the circulation because sam-
ples are accessible by minimally invasive blood draw
(TABLE1). Approaches used include real-time PCR53,54,
real-time PCR miRNA arrays55, miRNA microarrays35,38
and, most recently, miRNA sequencing of serum56,
plasma57 and plasma-isolated extracellular vesicles58.
miRNA sequencing is of particular interest as it ena-
bles the discovery of novel miRNAs that are potentially
unique to a disease stage or phenotype57,59. Fewer reports
have described miRNA profiles in urine or synovial
fluid than in blood60,61. Real-time PCR miRNA arrays
were used to interrogate synovial fluid samples taken
before and 6 months after high tibial osteotomy in six
patients with knee OA at Kellgren–Lawrence grade II
or III60. Three miRNAs were identified as being differ-
entially expressed at the two time points and, following
validation by real-time PCR in 22 additional patients,
increased miR-30c-5p was found to correlate with
reduced postoperative pain60. Looking beyond miRNAs,
lncRNAs and circRNAs are also dysregulated in OA
biofluids62. For example, the expression of lncRNAs
CAIF, LUADT1 and SNHG9 are decreased in OA syno-
vial fluid6365, whereas CTBP1-AS2, MCM3AP-AS1 and
CASC2 are increased6668, although the utility of these
lncRNAs as biomarkers requires further research.
To date, no consistent profile of non-coding RNAs
has been identified and validated in biofluids across
OA or RA studies. Among the challenges faced by
researchers are differences across studies in the joints
characterized, the profiling platforms used, the bioflu-
ids profiled and how the patient groups are defined, all
of which make it difficult to directly compare findings
(TABLE1). Going forward, panels of non-coding RNAs
(potentially from multiple classes) could prove to be
more reliable as biomarkers than an individual entity or
class owing to the variable expression and interactions
of individual non-coding RNAs. In addition to their
roles as biomarkers, non-coding RNAs in biofluids
might also function as systemic regulators of disease in
www.nature.com/nrrheum
Reviews
0123456789();:
distal joints. Two studies from 2020 found alterations in
the concentrations of circulating miRNAs that directly
target single-nucleotide polymorphisms in CXCR4 and
ADAMTS5 loci69,70, both of which are related to OA risk.
These results point to non-coding RNAs as circulating
epigenetic factors that regulate risk loci in arthritis as an
exciting new avenue for future research.
Role in pathogenesis
As important regulators of gene expression, non-coding
RNAs can be expected to have pleiotropic effects in poly-
genic diseases such as OA and RA. Data suggest that
non-coding RNAs can have both beneficial (such as
maintaining tissue homeostasis) and detrimental (such
as inducing tissue destruction) effects on the joints4,22.
In fact, miRNAs regulate a diverse range of cellular
processes (including inflammation7174, apoptosis7578,
ECM dysregulation79,80, chondrocyte differentiation81,
oxidative stress82 and autophagy8385), signalling path-
ways (including transforming growth factor-β86,87,
fibroblast growth factor (FGF)88, Wnt–β-catenin8991 and
Hedgehog92) and mediators (including the transcription
factors FOXM1, SOX5, SOX6 and SOX9, and oestro-
gen receptor-α9398) that are relevant for OA and RA.
Similarly, lncRNAs (TABLE2) and circRNAs (TABLE3)
can have multi-target regulatory effects on cell pheno-
type and tissue homeostasis and have the potential to
mediate pathogenic mechanisms in OA and RA6,62,99.
Table 1 | Circulating microRNAs with potential for use as biomarkers in OA and RA
Disease Platform Biofluid Number of
patients
Number of controls
(type)
Differentially expressed
miRNAs
Ref.
Knee OA Real-time PCR Plasma 150 150 (healthy individuals,
traumatic amputation or
meniscus injury)
Reduced in OA: miR-200c-3p,
miR-100-5p and miR-1826
53
Knee OA Real-time PCR Serum 10 10 (trauma) Reduced in OA: let-7e 54
Hip OA Real-time PCR Serum 28 2 (femoral neck fracture) Increased in OA: miR-146a-5p 169
RA Real-time PCR Plasma 125 30 (healthy individuals) Reduced in RA: miR-155 170
RA Real-time PCR Serum 20 20 (healthy individuals) Increased in RA: miR-138 171
RA Real-time PCR Blood 90 30 (healthy individuals) Increased in RA: miR-155,
miR-150, miR-146a, miR-146b,
miR-125a-5p and miR-223
172
RA Real-time PCR Serum 18 76 (SLE, SSc or MCTD) Increased in RA: miR-145 and
miR-181a
173
Knee OA Real-time PCR array Serum 114 (high pain relief
1 year post-TKR)
22 (low pain relief 1 year
post TKR)
Increased in low pain
relief group: miR-146a-5p,
miR-145-5p and miR-130b-3p
55
Knee OA Microarray Blood 55 (healthy individuals) Decreased in OA: miR-582-5p
and miR-424-5p
35
RA Microarray Blood 5 (early RA) 5 (healthy individuals),
5 (CPP+ healthy individuals)
Increased in RA: miR-361-5p 174
RA Microarray Serum 9 (divided into
3 pools)
15 (healthy individuals;
divided into 5 pools)
Increased in RA: miR-187-5p,
miR-4532 and miR-4516;
decreased in RA: miR-125a-3p,
miR-575, miR-191-3p,
miR-6865-3p, miR-197-3p,
miR-6886-3p, miR-1237-3p
and miR-4436b-5p
175
RA Microarray Serum exosomes 22 (in clinical
remission)
20 (not in clinical
remission)
Increased in clinical remission
group: miR-1915-3p and
miR-6511-5p
176
Knee OA Next-generation
sequencing
Serum 10 10 (healthy individuals) Increased in OA: miR-146a-5p
and miR-186-5p
56
Knee OA Next-generation
sequencing
Plasma 41 (early-stage OA) 50 (late-stage OA) Increased in early-stage OA:
miR-335-3p, miR-199a-5p,
miR-671-3p, miR-1260b,
miR-191-3p, miR-335-5p
and miR-543
57
Knee, hip or
hand OA
Next-generation
sequencing
Plasma extracellular
vesicles
23 23 (healthy individuals) None 58
RA Next-generation
sequencing
Plasma 167 91 (healthy individuals) Increased in RA: miR-22-3p,
miR-24-3p, miR-96-5p,
miR-134-5p, miR-140-3p
and miR-627-5p
45
Includes articles published between 2019 and 2021. CPP, cyclic citrullinated peptide antibody; MCTD, mixed connective tissue disease; miRNA, microRNA;
OA, osteoarthritis; RA, rheumatoid arthritis; SLE, systemic lupus erythematosus; SSc, systemic sclerosis; TKR, total knee replacement.
Nature reviews
|
Rheumatology
Reviews
0123456789();:
For example, lncRNAs can function through regulation
of histone methylation100, targeting single-nucleotide
polymorphisms101 and miRNA sponging30,102 to regu-
late cellular processes as diverse as apoptosis103,104, cell
proliferation103 and ECM degradation102,105,106. In the fol-
lowing sections, we curate literature published in thepast
2 years on non-coding RNAs in inflammation, cell
death, cell proliferation and ECM dysregulation in OA
and RA. Overall, although further research is required
to elucidate the interrelated effects of non-coding RNAs
on the pathogenesis of OA and RA, existing evidence
suggests that there could be merit to therapeutically
targeting non-coding RNAs in these diseases.
Inflammation. A variety of signalling molecules,
including non-coding RNAs, can induce and regulate
joint inflammation. Cytokines such as IL-1β, TNF and
IL-6 are often used as markers to gauge inflammatory
responses in chondrocytes and fibroblast-like syn-
oviocytes (FLSs). Amounts of these three cytokines
were reduced in mouse primary chondrocytes by an
increase in miR-410-3p107, and in supernatant from
lipopolysaccharide-treated human chondrocytes by
a decrease in miR-20a108; both outcomes were medi-
ated by nuclear factor-κB (NF-κB) signalling. NF-κB
can regulate the expression of miRNAs, but miRNAs
can also regulate the expression of NF-κB; for exam-
ple, an increase in miR‐382‐3p leads to a decrease in
phosphorylated NF-κB in IL‐1β‐stimulated human
OA chondrocytes109. Furthermore, miR-140-5p can
reduce human chondrocyte senescence110 and can work
synergistically with miR-146a to reduce NF-κΒ phos-
phorylation and the production of pro-inflammatory
cytokines in OA chondrocytes111. These studies suggest
that miRNAs regulate inflammatory responses through
mechanisms that include canonical signalling path-
ways (such as NF-κB) and cytokines (such as IL-1β,
IL-6 and TNF) in OA, and similar results have been
reported in RA47. lncRNAs are also important media-
tors of inflammation in human OA chondrocytes. The
lncRNAs PACER, CILinc01 and CILinc02 all show
rapid and transient induction in response to IL-1β and
other pro-inflammatory stimuli, indicating important
regulatory roles112.
In RA, non-coding RNAs in circulating immune
cells, synovial immune cells and FLSs contribute to
excess inflammation. T helper 17 (TH17) cells that
produce cytokines such as IL-17 and IL-22 stimulate
inflammatory responses from FLSs and macrophages in
RA to further promote synovial inflammation113.
In RA PBMCs, the lncRNA NEAT1 (which is present in
increased amounts compared with healthy individuals)
Table 2 | Long non-coding RNAs of mechanistic importance in OA and RA
lncRNA Change in expression Mechanism Effects Ref.
CTBP1-AS2 Increased in OA synovial fluid Increased methylation of
miR-130a
Promotes proliferation in OA chondrocytes 66
XIST Increased in OA cartilage Binds TIMP3 promoter and
accelerates methylation
Increases collagen degradation in OA chondrocytes 106
LINC01534 Increased in OA cartilage Sponges miR-140-5p Promotes ECM degradation (decreases aggrecan, type II
collagen and increases MMP3, MMP9 and MMP13) and
increases pro-inflammatory mediators (NO, PGE2, IL-6, IL-8
and TNF) in IL-1β-treated chondrocytes
138
H19 Increased in OA cartilage Sponges miR-140-5p Increases apoptosis, reduces cell proliferation, increases ECM
degradation (increases MMP1 and MMP13 and decreases type II
collagen) and increases ECM calcification in chondrocytes
137
Increased in RA FLSs and
synovium
Sponges miR-103a, which
negatively regulates IL15
and DKK1
Increases inflammation and joint destruction in mice with CAIA 177
NEAT1 Increased in OA cartilage Sponges miR-377-3p, which
negatively regulates ITGA6
Reduces cell proliferation and increases apoptosis, ECM
degradation and inflammation in IL-1β-treated chondrocytes
30
Increased in RA FLSs and
synovium
Sponges miR-410-3p, which
negatively regulates YY1
Increases cell proliferation and TNF and MMP9 expression and
decreases apoptosis in RA FLSs
33
Increased in RA PBMC
exosomes
Sponges miR-23a, which
negatively regulates the
MDM2–SIRT6 axis
Increases FLS proliferation and inflammation 135
Increased in RA PBMCs and
TH17 cells
Reduces ubiquitylation
of STAT3
Increases TH17 cell differentiation and disease severity in mice
with CIA
114
PINT Decreased in RA FLSs and
synovium
Sponges miR-155-5p, which
negatively regulates SOCS1
Increases cell proliferation, invasion and pro-inflammatory
cytokine production in RA FLSs
178
PVT1 Increased in RA synovium Sponges miR-145-5p Increases cell proliferation and pro-inflammatory cytokine
production and decreases apoptosis in RA FLSs
31
Increased in RA synovium Sponges miR-543, which
negatively regulates SCUBE2
Increases cell proliferation and IL-1β expression and decreases
apoptosis in RA FLSs
179
Includes articles published between 2019 and 2021. CAIA, collagen antibody-induced arthritis; CIA, collagen-induced arthritis; ECM, extracellular matrix;
FLS, fibroblast-like synoviocyte; lncRNA, long non-coding RNA; MDM2, E3 ubiquitin-protein ligase MDM2; MMP, matrix metalloproteinase; NO, nitric oxide;
OA, osteoarthritis; PBMC, peripheral blood mononuclear cell; PGE2, prostaglandin E2; RA, rheumatoid arthritis; SIRT6, sirtuin 6; STAT3, signal transducer and
activator of transcription 3; TH17 cell, T helper 17 cell.
www.nature.com/nrrheum
Reviews
0123456789();:
targets signal transducer and activator of transcription 3,
causing decreased ubiquitylation-mediated degradation
and leading to an increase in TH17 cell differentiation114.
Similarly, a lack of the miRNA let-7g-5p in patients with
RA promotes the differentiation of naive CD4+ Tcells
into TH17 cells, whereas the treatment of mice with
collagen-induced arthritis (CIA) with let-7g-5p mim-
ics decreases the number of TH17 cells in the blood and
spleen, leading to reduced synovial hyperplasia, pannus
formation and cartilage destruction115. Macrophages
with a pro-inflammatory phenotype (M1-like) are
also enriched in active RA synovium116. An increase in
miR-155 in monocytes from patients with RA impairs
monocyte differentiation into an inflammation-resolving
phenotype (M2-like)117 and, in RA synovial tissue and
fluid, an increase in miR-221-3p leads to decreased
IL-10 production (via direct targeting of Janus kinase 3)
in M2-like macrophages and acts synergistically with
miR-155-5p to increase the production of IL-12 (which
is specific to M1-like macrophages)118. Given the role
of inflammation in OA and RA, understanding the
contribution of non-coding RNAs to its underlying
mechanisms could provide new insights.
Cell death and cell proliferation. Abnormal cell death
and cell proliferation in joint tissues create hallmark
features of OA (such as cartilage degeneration) and RA
(such as synovial hyperplasia). Studies have reported
the effects of a variety of miRNAs on chondrocyte
apoptosis. For example, increased expression of miR-
33b-3p, miR‐9‐5p or miR‐27a decreased chondro-
cyte apoptosis119121, whereas increased expression of
miR-486-5p, miR-363-3p or miR-455-3p increased
chondrocyte apoptosis75,122,123. The mechanisms through
which unique miRNAs affect cell death and cell prolif-
eration can often converge onto a single pathway, such
as the phosphoinositide 3-kinase (PI3K)–AKT sig-
nalling pathway124131. Beneficial effects produced by
miRNAs through regulation of the PI3K–AKT pathway
include a reduction in apoptosis and cartilage degen-
eration caused by an increase in miR-455-3p124, the
promotion of chondrocyte proliferation and reduced
apoptosis caused by a decrease in miR-34a125 and a
reduction in chondrocyte apoptosis and inflammation
caused by an increase in miR-128-3p126. Conversely,
increased amounts of miR‐155, miR-1236 or miR-103 all
promote chondrocyte apoptosis by targeting PI3K127129.
Table 3 | Circular RNAs of mechanistic importance in OA and RA
circRNA Change in expression Mechanism Effects Ref.
circ_0136474 Increased in OA cartilage Sponges miR-127-5p, which
negatively regulates MMP13
Suppresses cell proliferation and increases
apoptosis in OA chondrocytes
180
circ_0009119 Decreased in OA cartilage Sponges miR-26a, which
negatively regulates PTEN
Protects OA chondrocytes from IL-1β-induced
apoptosis
181
circ_0001722 (circCDK14) Decreased in OA cartilage Sponges miR-125a-5p, which
negatively regulates SMAD2
Regulates ECM metabolism (decreases MMP3
and MMP13; increases SOX9 and type II
collagen), inhibits apoptosis and promotes cell
proliferation in chondrocytes
182
circ_0023404 (circRNF121) Increased in OA cartilage LEF1 increases circRNF121
expression, which sponges
miR-665, which negatively
regulates MYD88
Regulates degradation of ECM (increases
MMP13 and ADAMTS5; decreases type II
collagen and aggrecan), apoptosis and cell
proliferation in chondrocytes
163
circ_0000284 (circHIPK3) Increased in OA cartilage Sponges miR-124, which
negatively regulates SOX8
Inhibits apoptosis in chondrocytes 183
circVCAN Increased in OA cartilage Inhibits activation of NF-κB
signalling pathway
Increases cell proliferation and decreases
apoptosis in OA chondrocytes
184
circ_0008956 (circUBE2G1) Increased in OA cartilage Sponges miR-373, which
negatively regulates HIF1A
circUBE2G1 inhibition reduces the effects of
LPS in OA chondrocyte viability and apoptosis
185
circPSM3 Increased in OA cartilage Sponges miRNA-296-5p Inhibits cell proliferation and differentiation in
OA chondrocytes
186
circCDR1as Increased in OA cartilage Sponges miR-641, which
negatively regulates FGF2
Regulates ECM metabolism (increases MMP13
and IL-6; decreases type II collagen)
187
circTMBIM6 Increased in OA cartilage Sponges miR-27a, which
negatively regulates MMP13
Regulates ECM degradation (increases
MMP13)
188
circ_0000448 (circGCN1L1) Increased in OA TMJ
synovium
Sponges miR-330-3p, which
negatively regulates TNF
Increases chondrocyte apoptosis and ECM
metabolism (increases MMP3, MMP13 and
ADAMTS4; decreases type II collagen),
and increases synoviocyte hyperplasia and
inflammation
189
circ_0088036 Increased in RA FLSs Sponges miR-140-3p, which
negatively regulates SIRT1
Increases FLS proliferation and migration 190
circ_09505 Increased in RA PBMCs Sponges miR-6089, which
negatively regulates AKT1
Increases macrophage proliferation and
cell-cycle progression
191
Includes articles published between 2019 and 2021. ADAMTS, a disintegrin and metalloproteinase with thrombospondin motifs; circRNA, circular RNA; ECM,
extracellular matrix; FLS, fibroblast-like synoviocyte; LEF1, lymphoid enhancer-binding factor 1; LPS, lipopolysaccharide; MMP, matrix metalloproteinase; NF-κB, nuclear
factor-κB; OA, osteoarthritis; PBMC, peripheral blood mononuclear cell; RA, rheumatoid arthritis; SOX9, transcription factor SOX9; TMJ, temporomandibular joint.
Nature reviews
|
Rheumatology
Reviews
0123456789();:
Other pathways, such as NF-κB132, can also mediate the
effect of non-coding RNAs on chondrocyte apoptosis.
In addition, lncRNAs and circRNAs can interact with
miRNAs to regulate cell death and cell proliferation.
For example, lncRNA CTBP1-AS2 is upregulated
in OA synovial fluid and regulates the expression of
miR-130a through methylation in OA chondrocytes,
but not in healthy chondrocytes, to promote cell
proliferation66. New evidence to support the impor-
tance of non-coding RNAs in regulating cell death and
proliferation is also continuing to emerge133 (TABLE3).
In RA, expression of miR-483-3p, which is thought to
be oncogenic in several human cancers, is increased; in
FLSs, this miRNA directly targets IGF1 mRNA (which
encodes insulin-like growth factor 1; IGF1) to impair
apoptosis and induce tumour-like proliferation134.
Expression of the lncRNA NEAT1 is also increased in
human RA synovial tissue33 and PBMCs114. Delivery of
NEAT1 to RA FLSs via plasma exosomes isolated from
humans and mice caused sponging of miR-23a and
miR-410-3p and thereby increased the expression of
their targets (including E3 ubiquitin-protein ligase
MDM2 and transcriptional repressor protein YY1), lead-
ing to a decrease in apoptosis and an increase in FLS
proliferation and inflammation33,135. Furthermore, the
reduction of NEAT1 via siRNA can reduce the severity
of CIA in mice114. These results illustrate the possibility of
targeting non-coding RNAs to modulate cell death and
cell proliferation.
Extracellular matrix dysregulation. miRNAs, lncRNAs,
circRNAs and even snoRNAs have been implicated in
ECM dysregulation in joint tissues such as cartilage, syn-
ovium and bone. IL-1β is widely used to induce cellular
responses that mimic pathological conditions including
inflammation109, apoptosis127 and cartilage degradation97.
Researchers often use IL-1β to stimulate a response in
chondrocytes invitro that is subsequently rescued or
exacerbated by manipulating a non-coding RNA. In
cultured human chondrocytes, increased miR-377-3p
expression reversed IL-1β-induced upregulation of
inflammatory markers, cartilage degradation markers
and chondrocyte apoptosis30. In a different experimen-
tal model, human chondrocytes transfected with an
miR-613 agonist for 48 hours prior to administration of
IL-1β had reduced markers of inflammation, apoptosis
and cartilage degradation compared with IL-1β-treated
cells136. A tentative role for the NEAT1–miR-377-
3p–ITGA6 axis has been described in IL-1β-treated
chondrocytes, in which NEAT1 might function as
an miR-377-3p sponge, thereby upregulating ITGA6
expression to affect inflammatory responses, apoptosis
and ECM degradation30. Among other notable lncR-
NAs, XIST increases collagen degradation in OA chon-
drocytes via increasing TIMP3 promoter methylation
through the recruitment of a DNA methyltransferase106.
Furthermore, in OA cartilage, expression of the lncRNAs
H19 and LINC01534 is increased and ECM degrada-
tion is promoted through their individual binding to
miR-140-5p137,138, an miRNA that is well characterized in
OA for its cartilage matrix remodelling effects139. Roles
for circRNAs (TABLE3) and snoRNAs28,140,141 have also been
described in ECM metabolism. For example, impaired
expression of U3 snoRNA, SNORD26 or SNORD96A
alters the protein translation capacity of chondrocytes,
chondrocyte differentiation, pro-inflammatory pathways
and the expression of markers of OA28,141.
Notably, ECM dysregulation can actually facilitate
intercellular communication by making otherwise dense
cartilage and bone matrices more permeable to extra-
cellular vesicles that carry non-coding RNAs. Evidence
from RA suggests that both chondrocytes and osteo-
blasts can respond to miRNAs carried by FLS-derived
exosomes. In one study, exosomes from FLSs carrying
miR-106b induced chondrocyte apoptosis and reduced
proliferation by directly targeting PDK4 mRNA142.
In another study, exosomes from FLSs carrying
miR-486-5p were phagocytosed by osteoblasts and pro-
moted their differentiation and the expression of ECM
markers such as type I collagen143. Given that inflamed
and hyperplastic synovium is a common feature of RA,
exosomes secreted by FLSs could serve as messengers to
induce damage in surrounding joint tissues.
Therapeutic potential
Non-coding RNAs represent promising therapeu-
tic targets in OA and RA because their activity can be
modulated via small molecules and biological delivery
systems (such as exosomes144146) to reduce features
of disease, and even pain, in experimental models of
arthritis147,148. The cargo of these delivery systems can
include non-coding RNA mimics (known as agomirs),
non-coding RNA inhibitors (known as antagomirs) and
other molecules, such as transcription factors. For exam-
ple, in mouse cells invitro, the lncRNA MM2P promotes
macrophage polarization towards an M2-like pheno-
type and stimulates the release of exosomes containing
SOX9 mRNA and protein from these cells, which can
induce the production of ECM components in cultured
chondrocytes149. As these exosomes contain a functional
transcription factor (SOX9) that is known to promote
chondrocyte anabolism, they represent a potential
therapeutic strategy to restore cartilage homeostasis.
An important consideration in harnessing the
potential of non-coding RNAs as possible therapeu-
tics is the route of administration, whether systemic or
intra-articular, so that the therapy can reach the intended
target tissues. Systemic administration of non-coding
RNAs can be achieved by intravenous injection. Using
this method, an miR-365 agomir decreased disease
severity in mice with CIA, potentially through downreg-
ulation of IGF1 (REF.78). Similarly, following intravenous
injection, the concentration of an miR-26a mimic was
increased in articular cartilage and could reduce disease
severity in rats with CIA through the downregulation of
connective tissue growth factor (CTGF)150. Intravenous
injection of exosomes has also been explored.
BMSC-derived exosomes enriched with miR-320a,
which directly downregulates C-X-C chemokine
ligand 9, could decrease disease severity in mice with
CIA following intravenous injection151. Furthermore,
BMSC-secreted exosomes enriched with miR-192-5p
could be found in the synovial tissue of rats with CIA
after intravenous injection, and could decrease disease
www.nature.com/nrrheum
Reviews
0123456789();:
severity, potentially through downregulation of the
signalling molecule RAC2 (REF.152). These data suggest
that non-coding RNAs were able to traffic to the joints
in rats and mice with CIA, where they reduced syno-
vial inflammation, cartilage damage and bone erosion.
However, because a single non-coding RNA can have
multiple gene targets, local modulation (such as direct
injection of miRNA agomirs and antagomirs into the
joint) is also being explored to avoid unwanted systemic
effects. In mice with CIA, intra-articular delivery of
miR-146a-5p agomir decreased disease activity, synovial
hyperplasia, the invasiveness of the pannus and cartilage
erosion, potentially through downregulation of CTGF153.
Similarly in rats with CIA, intra-articular delivery of
miR-141-3p agomir improved disease outcomes via
direct binding of the transcription factor FOXC1, which
functions as an oncogene to promote tumour and RA
FLS proliferation154. Intra-articular delivery of exosomes
is also possible, and exosomes can even be engineered
to target cells of interest. For example, the fusion of
chondrocyte-affinity peptide to lysosome-associated
membrane glycoprotein 2b molecules on the surface
of exosomes promoted the trafficking and fusion of the
exosomes to chondrocytes, the efficient delivery of
miR-140 and the mitigation of disease progression in a
rat surgical model of OA155.
Alternative strategies for non-coding RNA modula-
tion continue to emerge. For example, transplantation of
cartilage pellets derived from human BMSCs that over-
express beneficial miRNAs (such as miR-27b) inhibited
hypertrophic chondrocyte differentiation during carti-
lage defect repair156. Similarly, intra-articular injection
of human umbilical cord-derived mesenchymal stem
cells transfected to overexpress miR-140 had protec-
tive effects in a rat model of OA157. A biodegradable
delivery system for an miR-365 antagomir based on
non-pathogenic yeast cell wall particles has been devel-
oped that could resist degradation in the gastrointes-
tinal system following oral administration, and which
reduced features of disease in a mouse surgical model
of OA102. Furthermore, cationic liposomes (lipoplexes)
have been used for the intra-articular administration of
miR-17-5p to reduce synovial immune cell infiltration,
inflammation and bone erosion in mice with CIA72.
As an alternative means of suppressing miRNA function,
tough decoy RNAs have been developed, wherein vectors
expressing miRNA target sites bind and reduce specific
miRNA activity in cells; a tough decoy for miR-195-5p
reduced its activity and the occurrence of hypertrophy
in cultured chondrocytes158. Moreover, miRNA agomirs
and antagomirs can be directly modified to improve
their therapeutic properties. To improve specific bind-
ing, locking the conformation of antagomirs (known
as locked nucleic acids) is effective, and intra-articular
delivery of locked nucleic acid antagomirs for
miR-181a-5p and miR-34a-5p could reduce disease
severity in experimental models of OA159,160. To improve
stability and delivery, non-coding RNAs can be conjugated
to other molecules such as atelocollagen; intra-articular
administration of an miR-9a-5p agomir–atelocollagen
complex could effectively reduce disease severity in
rats with CIA161. Additional delivery mechanisms and
considerations for achieving clinical translation of anti-
sense oligonucleotide-based therapies for OA have been
reviewed in detail elsewhere104,162.
Studying the interactions among non-coding
RNAs, including regulators and effectors of circRNA
miRNA–mRNA axes, could also reveal new avenues
for targeted treatment. One molecular mechanism pro-
posed as a prospective therapeutic target for OA is the
circRNF121–miR-665–MYD88 axis, which is regulated
by the transcription factor LEF1 (REF.163). In human chon-
drocytes, LEF1 increases the expression of circRNF121,
which functions as a sponge for miR-665, thereby
indirectly targeting MYD88. As such, modulation of
miR-665 and circRNF121 could alter MYD88 expression
to promote chondrocyte apoptosis, proliferation and
ECM degradation, both invitro in human chondrocytes
and invivo in a rat model of OA. Furthermore, this axis
was shown to activate the NF-κB signalling pathway163.
Although the data suggest that miR-665 could be tar-
geted to mitigate the detrimental effects of circRNF121,
it is evident that the upstream regulator (LEF1), circRNA
(circRNF121), miRNA (miR-665), gene target (MYD88)
and downstream pathway (NF-кB) could all be potential
targets. These data illustrate the importance of consid-
ering the non-coding RNA interactome for therapeutic
targeting, as one or more of these factors might need to
be modulated to improve disease outcomes (BOX1).
On the basis of the current literature, outstanding
questions remain to be answered before targeting of
non-coding RNAs can be translated into a therapeutic
strategy to improve patient care. First, the appropriate
target must be identified, whether it is the non-coding
RNA, its upstream regulator or the downstream medi-
ator. Second, the appropriate tissue or tissues must be
identified for targeting, as non-coding RNAs are known
to have tissue-specific effects. Third, the appropriate
Box 1 | The non-coding RNA interactome in gene expression regulation
Theregulationofgeneexpressionistightlycontrolled.Non-codingRNAshave
importantrolesinthisprocess,operatingthroughdirectmechanisms(suchas
degradationofgenetranscripts)andindirectmechanisms(suchasinhibitionof
othernon-codingRNAs).Together,thesemechanismscomprisethenon-codingRNA
interactome,whichcanbethoughtofasthecompletesetofthemolecularinteractions
ofnon-codingRNAs.Emergingliteraturesuggeststhatinteractionsamongnon-coding
RNAentitiesandclassesarecommonandhaveconsiderableimplicationsforjoint
diseases.High-throughputprofilingisausefulapproachforbeginningtounravelthe
non-codingRNAinteractome.Forexample,researchershaveusedthreepublicly
availablemicroarraydatasetsforsynoviumfrompatientswithrheumatoidarthritis
(RA)todemonstratepotentialdirectregulationofinterconnectedgenetargetsby
specificlongnon-codingRNAs(lncRNAs),whereinthelncRNAsNEAT1andFAM30A
werepredictedtointeractwithmajorRAhubgenes192.Toexploreinteractionsacross
non-codingRNAclasses,circularRNA(circRNA)–microRNA(miRNA)networkshave
beenconstructedforsynovium193,cartilage194andchondrocytes133,195frompatientswith
osteoarthritis(OA).Forexample,researchershaveusedRNAsequencingtoidentify
OA-relatedcircRNAsincartilage,followedbybioinformaticsanalysestodiscover
166,394circRNA–miRNA–mRNAaxes194.LncRNA–miRNAnetworkshavealsobeen
exploredinOA.Inhumankneecartilage,publiclyavailableRNAsequencingdatahave
beenminedtoidentifydifferentiallyexpressedlncRNAsandmRNAsthatcontribute
toanintegratednetworkofcompetingendogenousRNAs,including10lncRNAs,
69miRNAsand72mRNAs196.Theseindividualprofilingstudiesareanimportantfirst
steptowardsunderstandingthenon-codingRNAinteractome,butneedtobefollowed
byeffortstointegratefindingsacrossstudiessothatcandidatesforfurthervalidation
andpotentialtherapeutictargetingcanbeprioritized.
Nature reviews
|
Rheumatology
Reviews
0123456789();:
delivery mechanism must be identified, including the
vehicle (such as exosomes) and route of administration.
For example, intra-articular delivery might offer ben-
efits over systemic administration by providing local
modulation and thereby reducing unwanted off-target
effects. The answers to these questions and others (such
as the best dosage to use) might be patient-specific,
and tailored RNA-based therapeutics might need
to be administered in a phenotype-dependent manner to
achieve precision medicine in OA and RA. The utility of
RNA-based therapeutics has now achieved global recog-
nition through RNA vaccines, which were first described
over two decades ago164, and it is therefore reasonable
to expect bolstered research efforts into RNA-based
therapeutics, which should include non-coding RNAs.
Future directions
Unravelling the complex interactions among non-coding
RNAs is becoming an important goal; however, the com-
prehensive high-throughput profiling of joint tissues
and biofluids that will be necessary to achieve this aim
comes with its own set of challenges (TABLE4). Although
microarrays are useful for profiling a pre-selected sub-
set of known candidate RNAs, this technique is lim-
ited by factors such as the appropriate selection of an
endogenous reference, which can vary by tissue type165.
Increasingly, next-generation sequencing technologies
are being used to achieve unbiased and quantitative
measures of all varieties of non-coding RNAs. For exam-
ple, next-generation sequencing can be used to identify
the direct binding of miRNAs to target genes through
RNA-immunoprecipitation and high-throughput
sequencing (RIP sequencing), as has been described in
human articular chondrocytes166. This approach ena-
bles the validation of predicted gene targets that are
commonly obtained using prediction tools (TABLE4).
Furthermore, applying sequencing technology to fun-
damental processes in model systems has the potential
to uncover important mechanisms that underlie disease.
For example, combinations of RNA sequencing and
small non-coding RNA sequencing have been applied
to explore chondrogenesis and metabolism in human
BMSCs167, inflammatory cytokine responses in mouse
induced pluripotent stem cells168 and cartilage ageing in
horse chondrocytes140.
Among the limitations of unbiased discovery of
non-coding RNAs is that researchers often focus on just
one or two molecules for further investigation. How and
why these molecules are chosen can be unclear, as other
non-coding RNA entities that could have promising
roles in joint pathobiology are often not investigated
further. Notably, very few non-coding RNA studies in
OA and RA include comparisons with other studies or
meta-analyses with other available non-coding RNA
datasets in order to validate, expand and build a com-
prehensive interactome of these important epigenetic
regulators. However, efforts are ongoing around the
world to curate comprehensive databases of published
evidence to help researchers to investigate the complex
interactions between non-coding RNAs, genes and pro-
teins (TABLE4). To this end, it is critical that nomenclature
and annotations for the non-coding RNAs identified in
studies are systematically reported. For example, inves-
tigators are encouraged to report the strands of miRNAs
(3p or 5p) to ensure accurate integration of their data
with other datasets and analyses. Furthermore, report-
ing of the clinical annotation of samples involved in
non-coding RNA studies is required to enable correla-
tion with molecular and clinical phenotypes. Finally, to
improve the quality of basic and translational research
by applying integrative analytical and machine learning
techniques, well-annotated high-throughput data must
be made available in the correct format (for example, the
raw sequencing datasets) in online repositories.
Conclusions
A substantial surge has occurred in the number of pub-
lished articles related to non-coding RNAs in OA and
RA in the past few years, mostly for miRNAs, lncRNAs
and circRNAs, and to a lesser degree for snoRNAs,
Table 4 | Use of bioinformatics and computational biology tools in non-coding RNA research
Challenge Approach Resources
Analysing high-throughput profiling
data for non-coding RNAs in both
health and disease contexts
A search tool can be used to discover novel
non-coding RNA sequences in deep sequencing data
miRDeep2
Ensuring proper and consistent
naming of all non-coding RNA
entities so that data can be
accurately integrated across studies
Several databases are helpful to ensure the correct
use of primary names and identifiers
miRbase for microRNAs; DIANA-lncBase for long
non-coding RNAs; circBase and circAtlas for circular
RNAs; and Hugo Gene Nomenclature for gene names
Elucidating the potential functions of
non-coding RNAs
Multiple tools can be used for target gene prediction,
including for novel microRNA sequences
TargetScan; mirDIP; miRDB; and miRanda
Interpreting predicted target genes Pathway prediction tools can be used to create
functional groups with biological relevance
(such as signalling cascades)
The Gene Ontology Resource for gene enrichment
analysis; pathDIP for integrated pathway enrichment
analysis; and integrated web portals, such as
Enrichr, for access to diverse types of computational
annotation and overrepresentation analysis
Combining non-coding RNA
datasets to promote integrative
computational analyses
Public repositories can be used to access and deposit
high-throughput data
The NCBI Gene Expression Omnibus repository; the
NCBI Sequence Read Archive; the NCBI Database
of Genotypes and Phenotypes; and the EMBL-EBI
European Nucleotide Archive
EMBL-EBI, European Molecular Biology Laboratory European Bioinformatics Institute; NCBI, National Center for Biotechnology Information.
www.nature.com/nrrheum
Reviews
0123456789();:
tRFs and other non-coding RNAs. This increased
research output has been possible because of advances
in next-generation sequencing technology and the
availability of computational and analytical tools for
data mining. Some of the non-coding RNAs that have
been identified using these methods, as well as their
regulatory interactome, could have crucial roles in joint
health and disease, affecting biological processes and
functioning as biomarkers, mediators of pathogenesis
and potential therapeutic targets. Although these dis-
coveries are promising, a concerted effort is required to
validate, integrate and translate findings from current
studies to harness the full potential of non-coding RNAs
in OA and RA.
Published online xx xx xxxx
1. Palazzo, A. F. & Lee, E. S. Non-coding RNA: what
is functional and what is junk? Front. Genet. 6, 2
(2015).
2. Rice, S. J., Beier, F., Young, D. A. & Loughlin, J.
Interplay between genetics and epigenetics in
osteoarthritis. Nat. Rev. Rheumatol. 16, 268–281
(2020).
3. Reynard, L. N. & Barter, M. J. Osteoarthritis year
in review 2019: genetics, genomics and epigenetics.
Osteoarthritis Cartilage 28, 275–284 (2020).
4. Vicente, R., Noel, D., Pers, Y. M., Apparailly, F.
& Jorgensen, C. Deregulation and therapeutic
potential of microRNAs in arthritic diseases.
Nat. Rev. Rheumatol. 12, 211–220 (2016).
5. Zhang, P., Wu, W., Chen, Q. & Chen, M. Non-coding
RNAs and their integrated networks. J. Integr.
Bioinform 16, 20190027 (2019).
6. Ren, S. etal. Circular RNAs: promising molecular
biomarkers of human aging-related diseases via
functioning as an miRNA sponge. Mol. Ther. Methods
Clin. Dev. 18, 215–229 (2020).
7. Chillon, I. & Marcia, M. The molecular structure
of long non-coding RNAs: emerging patterns and
functional implications. Crit. Rev. Biochem. Mol. Biol.
55, 662–690 (2020).
8. Jiang, S., Liu, Y., Xu, B., Zhang, Y. & Yang, M.
Noncoding RNAs: new regulatory code in chondrocyte
apoptosis and autophagy. Wiley Interdiscip. Rev. RNA
11, e1584 (2020).
9. Wang, J. etal. The role of lncRNAs in osteogenic
differentiation of bone marrow mesenchymal stem
cells. Curr. Stem Cell Res. Ther. 15, 243–249 (2020).
10. Della Bella, E. etal. Differential regulation of circRNA,
miRNA, and piRNA during early osteogenic and
chondrogenic differentiation of human mesenchymal
stromal cells. Cells 9, 398 (2020).
11. Inui, M., Martello, G. & Piccolo, S. MicroRNA control
of signal transduction. Nat. Rev. Mol. Cell Biol. 11,
252–263 (2010).
12. Feng, L. etal. MicroRNA-378 suppressed osteogenesis
of MSCs and impaired bone formation via inactivating
Wnt/β-catenin signaling. Mol. Ther. Nucleic Acids 21,
1017–1028 (2020).
13. Ohba, S. Hedgehog signaling in endochondral
ossification. J. Dev. Biol. 4, 20 (2016).
14. Chen, T. etal. MicroRNA-1 promotes cartilage matrix
synthesis and regulates chondrocyte differentiation
via post-transcriptional suppression of Ihh expression.
Mol. Med. Rep. 22, 2404–2414 (2020).
15. Lin, A. C. etal. Modulating hedgehog signaling can
attenuate the severity of osteoarthritis. Nat. Med.
15, 1421–1425 (2009).
16. Zhang, H. etal. MicroRNA-30a regulates chondrogenic
differentiation of human bone marrow-derived
mesenchymal stem cells through targeting Sox9.
Exp. Ther. Med. 18, 4689–4697 (2019).
17. Cai, W. L. etal. LncRNA LINC00707 promotes
osteogenic differentiation of hBMSCs through the
Wnt/β-catenin pathway activated by LINC00707/
miR-145/LRP5 axis. Eur. Rev. Med. Pharmacol. Sci.
24, 18–28 (2020).
18. Huang, M. J. etal. lncRNA ADAMTS9-AS2 controls
human mesenchymal stem cell chondrogenic
differentiation and functions as a ceRNA. Mol. Ther.
Nucleic Acids 18, 533–545 (2019).
19. Huang, X. etal. Prospect of circular RNA in
osteogenesis: a novel orchestrator of signaling
pathways. J. Cell Physiol. 234, 21450–21459
(2019).
20. Liu, X., Yan, C., Deng, X. & Jia, J. Hsa_circularRNA_
0079201 suppresses chondrocyte proliferation
and endochondral ossification by regulating the
microRNA1403p/SMAD2 signaling pathway in
idiopathic short stature. Int. J. Mol. Med. 46,
1993–2006 (2020).
21. Tavallaee, G., Rockel, J. S., Lively, S. & Kapoor, M.
MicroRNAs in synovial pathology associated with
osteoarthritis. Front. Med. 7, 376 (2020).
22. Endisha, H., Rockel, J., Jurisica, I. & Kapoor, M.
The complex landscape of microRNAs in articular
cartilage: biology, pathology, and therapeutic targets.
JCI Insight 3, e121630 (2018).
23. Qiu, W. J., Xu, M. Z., Zhu, X. D. & Ji, Y. H.
MicroRNA-27a alleviates IL-1β-induced inflammatory
response and articular cartilage degradation via TLR4/
NF-κB signaling pathway in articular chondrocytes.
Int. Immunopharmacol. 76, 105839 (2019).
24. Xiang, Y. etal. miR-142-5p as a CXCR4-targeted
microRNA attenuates SDF-1-induced chondrocyte
apoptosis and cartilage degradation via inactivating
MAPK signaling pathway. Biochem. Res. Int. 2020,
4508108 (2020).
25. Luo, X., Wang, J., Wei, X., Wang, S. & Wang, A.
Knockdown of lncRNA MFI2-AS1 inhibits
lipopolysaccharide-induced osteoarthritis progression
by miR-130a-3p/TCF4. Life Sci. 240, 117019 (2020).
26. Chen, K., Fang, H. & Xu, N. LncRNA LOXL1-AS1
is transcriptionally activated by JUND and contributes
to osteoarthritis progression via targeting the
miR-423-5p/KDM5C axis. Life Sci. 258, 118095
(2020).
27. Green, J. A., Ansari, M. Y., Ball, H. C. & Haqqi, T. M.
tRNA-derived fragments (tRFs) regulate post-
transcriptional gene expression via AGO-dependent
mechanism in IL-1β stimulated chondrocytes.
Osteoarthritis Cartilage 28, 1102–1110 (2020).
28. Ripmeester, E. G. J. etal. Impaired chondrocyte
U3 snoRNA expression in osteoarthritis impacts the
chondrocyte protein translation apparatus. Sci. Rep.
10, 13426 (2020).
29. Nanus, D. E. etal. Regulation of the inflammatory
synovial fibroblast phenotype by metastasis-
associated lung adenocarcinoma transcript 1 long
noncoding RNA in obese patients with osteoarthritis.
Arthritis Rheumatol. 72, 609–619 (2020).
30. Tu, Y. etal. MicroRNA-377-3p alleviates IL-1β-caused
chondrocyte apoptosis and cartilage degradation
in osteoarthritis in part by downregulating ITGA6.
Biochem. Biophys. Res. Commun. 523, 46–53
(2020).
31. Tang, J., Yi, S. & Liu, Y. Long non-coding RNA PVT1
can regulate the proliferation and inflammatory
responses of rheumatoid arthritis fibroblast-like
synoviocytes by targeting microRNA-145-5p.
Hum. Cell 33, 1081–1090 (2020).
32. Alnajjar, F. A. etal. The expression and function
of metastases associated lung adenocarcinoma
transcript-1 long non-coding RNA in subchondral
bone and osteoblasts from patients with
osteoarthritis. Cells 10, 786 (2021).
33. Wang, Y. etal. LncRNA NEAT1 targets fibroblast-like
synoviocytes in rheumatoid arthritis via the miR-410-
3p/YY1 Axis. Front. Immunol. 11, 1975 (2020).
34. Sun, J. L. etal. MicroRNA-29b promotes subchondral
bone loss in TMJ osteoarthritis. J. Dent. Res. 99,
1469–1477 (2020).
35. Wang, P. etal. Genome-wide microRNA screening
reveals miR-582-5p as a mesenchymal stem
cell-specific microRNA in subchondral bone of the
human knee joint. J. Cell Physiol. 234, 21877–21888
(2019).
36. Zhao, C. etal. Exosomes from adipose-derived
stem cells promote chondrogenesis and suppress
inflammation by upregulating miR145 and miR221.
Mol. Med. Rep. 21, 1881–1889 (2020).
37. Zhao, J. etal. Identification of lncRNA and mRNA
biomarkers in osteoarthritic degenerative meniscus by
weighted gene coexpression network and competing
endogenous RNA network analysis. Biomed. Res. Int.
2020, 2123787 (2020).
38. Liu, C., Gao, J., Su, G., Xiang, Y. & Wan, L.
MicroRNA-1202 plays a vital role in osteoarthritis via
KCNQ1OT1 has-miR-1202-ETS1 regulatory pathway.
J. Orthop. Surg. Res. 15, 130 (2020).
39. Zhang, P. etal. lncRNA IGHCγ1 acts as a ceRNA
to regulate macrophage inflammation via the
miR-6891-3p/TLR4 axis in osteoarthritis. Mediators
Inflamm. 2020, 9743037 (2020).
40. Soul, J. etal. Stratification of knee osteoarthritis:
two major patient subgroups identified by genome-
wide expression analysis of articular cartilage.
Ann. Rheum. Dis. 77, 423 (2018).
41. Coutinho de Almeida, R. etal. Identification and
characterization of two consistent osteoarthritis
subtypes by transcriptome and clinical data
integration. Rheumatology 60, 1166–1175 (2021).
42. Coutinho de Almeida, R. etal. RNA sequencing
data integration reveals an miRNA interactome
of osteoarthritis cartilage. Ann. Rheum. Dis. 78,
270–277 (2019).
43. Zhou, Y. etal. Identification of differentially expressed
miRNAs and mRNAs in synovial of osteoarthritis via
RNA-sequencing. BMC Med. Genet. 21, 46 (2020).
44. Wang, H. etal. Prediction of microRNA and
gene target in synovium-associated pain of knee
osteoarthritis based on canonical correlation analysis.
Biomed. Res. Int. 2019, 4506876 (2019).
45. Ormseth, M. J. etal. Development and validation
of a microRNA panel to differentiate between
patients with rheumatoid arthritis or systemic
lupus erythematosus and controls. J. Rheumatol. 47,
188–196 (2020).
46. Ormseth, M. J. etal. The endogenous plasma small
RNAome of rheumatoid arthritis. ACR Open. Rheumatol.
2, 97–105 (2020).
47. Zhang, C. etal. miR-22 inhibits synovial fibroblasts
proliferation and proinflammatory cytokine production
in RASF via targeting SIRT1. Gene 724, 144144
(2020).
48. Jin, Z., Ren, J. & Qi, S. Human bone mesenchymal
stem cells-derived exosomes overexpressing
microRNA-26a-5p alleviate osteoarthritis via
down-regulation of PTGS2. Int. Immunopharmacol.
78, 105946 (2020).
49. Xie, F. etal. Role of microRNA, lncRNA, and exosomes
in the progression of osteoarthritis: a review of recent
literature. Orthop. Surg. 12, 708–716 (2020).
50. Alevizos, I. & Illei, G. G. MicroRNAs as biomarkers
in rheumatic diseases. Nat. Rev. Rheumatol. 6,
391–398 (2010).
51. Wang, S. etal. Expression of Dicer in rheumatoid
arthritis is associated with disease activity and
balances the production of TNF-α. Mol. Med. Rep.
16, 1590–1595 (2017).
52. Ormseth, M. J. etal. Circulating microbial small
RNAs are altered in patients with rheumatoid arthritis.
Ann. Rheum. Dis. 79, 1557–1564 (2020).
53. Lai, Z. & Cao, Y. Plasma miR-200c-3p, miR-100-5p,
and miR-1826 serve as potential diagnostic
biomarkers for knee osteoarthritis: randomized
controlled trials. Medicine 98, e18110 (2019).
54. Feng, L. etal. Circulating microRNA let7e is decreased
in knee osteoarthritis, accompanied by elevated
apoptosis and reduced autophagy. Int. J. Mol. Med.
45, 1464–1476 (2020).
55. Giordano, R. etal. Preoperative serum circulating
microRNAs as potential biomarkers for chronic
postoperative pain after total knee replacement.
Mol. Pain. 16, 1744806920962925 (2020).
56. Rousseau, J. C. etal. Association of circulating
microRNAs with prevalent and incident knee
osteoarthritis in women: the OFELY study. Arthritis
Res. Ther. 22, 2 (2020).
57. Ali, S. A. etal. Sequencing identifies a distinct
signature of circulating microRNAs in early
radiographic knee osteoarthritis. Osteoarthritis
Cartilage 28, 1471–1481 (2020).
58. Aae, T. F. etal. Evaluating plasma extracellular
vesicle microRNAs as possible biomarkers for
osteoarthritis. Osteoarthritis Cartilage Open 1,
100018 (2020).
59. Potla, P., Ali, S. A. & Kapoor, M. A bioinformatics
approach to microRNA-sequencing analysis.
Osteoarthritis Cartilage Open 3, 100131 (2021).
Nature reviews
|
Rheumatology
Reviews
0123456789();:
60. Kwak, Y. H. etal. Significant changes in synovial fluid
microRNAs after high tibial osteotomy in medial
compartmental knee osteoarthritis: identification
of potential prognostic biomarkers. PLoS ONE 15,
e0227596 (2020).
61. Li, Y. H. etal. Identification of synovial fluid microRNA
signature in knee osteoarthritis: differentiating
early- and late-stage knee osteoarthritis.
Osteoarthritis Cartilage 24, 1577–1586 (2016).
62. Ehrlich, G. D. Circular RNAs as diagnostic biomarkers
for osteoarthritis. Genet. Test. Mol. Biomark. 23,
701–702 (2019).
63. Qi, K. etal. Long non-coding RNA (lncRNA) CAIF
is downregulated in osteoarthritis and inhibits
LPS-induced interleukin 6 (IL-6) upregulation by
downregulation of miR-1246. Med. Sci. Monit. 25,
8019–8024 (2019).
64. Ni, S. etal. LncRNA LUADT1 regulates miR-34a/SIRT1
to participate in chondrocyte apoptosis. J. Cell Biochem.
https://doi.org/10.1002/jcb.29637 (2020).
65. Zhang, H., Li, J., Shao, W. & Shen, N. LncRNA
SNHG9 is downregulated in osteoarthritis and inhibits
chondrocyte apoptosis by downregulating miR-34a
through methylation. BMC Musculoskelet. Disord.
21, 511 (2020).
66. Zhang, H., Li, J., Shao, W. & Shen, N. LncRNA
CTBP1-AS2 is upregulated in osteoarthritis and
increases the methylation of miR-130a gene to
inhibit chondrocyte proliferation. Clin. Rheumatol.
39, 3473–3478 (2020).
67. Gao, Y., Zhao, H. & Li, Y. LncRNA MCM3AP-AS1
regulates miR-142-3p/HMGB1 to promote LPS-induced
chondrocyte apoptosis. BMC Musculoskelet. Disord.
20, 605 (2019).
68. Sun, Y., Kang, S., Pei, S., Sang, C. & Huang, Y.
MiR93-5p inhibits chondrocyte apoptosis in
osteoarthritis by targeting lncRNA CASC2.
BMC Musculoskelet. Disord. 21, 26 (2020).
69. Wang, Y., Li, Y., Jia, D., Zheng, J. & Wang, G.
Correlation between single nucleotide polymorphisms
in CXCR4 microRNA binding site and the susceptibility
to knee osteoarthritis in Han Chinese population.
J. Clin. Lab. Anal. 35, e23600 (2021).
70. Weng, K., Luo, M. & Dong, D. Elucidation of the
mechanism by which a ADAMTS5 gene microRNA-
binding site single nucleotide polymorphism affects
the risk of osteoarthritis. Genet. Test. Mol. Biomark.
24, 467–477 (2020).
71. Cao, J., Liu, Z., Zhang, L. & Li, J. miR-940 regulates
the inflammatory response of chondrocytes by
targeting MyD88 in osteoarthritis. Mol. Cell Biochem.
461, 183–193 (2019).
72. Najm, A. etal. MicroRNA-17-5p reduces inflammation
and bone erosions in mice with collagen-induced
arthritis and directly targets the JAK/STAT pathway
in rheumatoid arthritis fibroblast-like synoviocytes.
Arthritis Rheumatol. 72, 2030–2039 (2020).
73. Wu, S., Wang, J., Li, J. & Li, F. microRNA-21
aggravates lipopolysaccharide-induced inflammation
in MH7A cells through targeting SNF5. Inflammation
43, 441–454 (2020).
74. Jiang, F. etal. MicroRNA-421 promotes inflammatory
response of fibroblast-like synoviocytes in rheumatoid
arthritis by targeting SPRY1. Eur. Rev. Med.
Pharmacol. Sci. 23, 8186–8193 (2019).
75. Zhang, M. etal. Identification of microRNA-363-3p
as an essential regulator of chondrocyte apoptosis
in osteoarthritis by targeting NRF1 through
the p53-signaling pathway. Mol. Med. Rep. 21,
1077–1088 (2020).
76. Wang, Y. etal. miR-410-3p regulates proliferation and
apoptosis of fibroblast-like synoviocytes by targeting
YY1 in rheumatoid arthritis. Biomed. Pharmacother.
119 , 109426 (2019).
77. Huang, Y. etal. Up-regulated microRNA-411 or
declined RIPK1 inhibits proliferation and promotes
apoptosis of synoviocytes in rheumatoid arthritis
mice via decreased NF-κB pathway. Cell Cycle 19,
666–683 (2020).
78. Wang, X. etal. Up-regulation of miR-365 promotes the
apoptosis and restrains proliferation of synoviocytes
through downregulation of IGF1 and the inactivation of
the PI3K/AKT/mTOR pathway in mice with rheumatoid
arthritis. Int. Immunopharmacol. 79, 106067 (2020).
79. Zhang, Y. etal. miR-137 suppresses cell growth and
extracellular matrix degradation through regulating
ADAMTS-5 in chondrocytes. Am. J. Transl Res. 11,
7027–7034 (2019).
80. Bai, Y., Chen, K., Zhan, J. & Wu, M. miR-122/SIRT1
axis regulates chondrocyte extracellular matrix
degradation in osteoarthritis. Biosci. Rep. 40,
BSR20191908 (2020).
81. Liu, H. & Luo, J. miR-211-5p contributes to chondrocyte
differentiation by suppressing fibulin-4 expression to
play a role in osteoarthritis. J. Biochem. 166, 495–502
(2019).
82. Cheleschi, S. etal. Hydrostatic pressure regulates
oxidative stress through microRNA in human
osteoarthritic chondrocytes. Int. J. Mol. Sci. 21,
3653 (2020).
83. Li, H. etal. MicroRNA-375 exacerbates knee
osteoarthritis through repressing chondrocyte
autophagy by targeting ATG2B. Aging 12,
7248–7261 (2020).
84. He, B. & Jiang, D. HOTAIR-induced apoptosis is
mediated by sponging miR-130a-3p to repress
chondrocyte autophagy in knee osteoarthritis.
Cell Biol. Int. 44, 524–535 (2020).
85. Tian, F., Wang, J., Zhang, Z. & Yang, J. LncRNA
SNHG7/miR-34a-5p/SYVN1 axis plays a vital
role in proliferation, apoptosis and autophagy
in osteoarthritis. Biol. Res. 53, 9 (2020).
86. Lei, J., Fu, Y., Zhuang, Y., Zhang, K. & Lu, D. LncRNA
SNHG1 alleviates IL-1β-induced osteoarthritis by
inhibiting miR-16-5p-mediated p38 MAPK and NF-κB
signaling pathways. Biosci. Rep. 39, BSR20191523
(2019).
87. An, Y. etal. Down-regulation of microRNA-203a
suppresses IL-1β-induced inflammation and cartilage
degradation in human chondrocytes through Smad3
signaling. Biosci. Rep. 40, BSR20192723 (2020).
88. Li, Y., Nie, J. & Jiang, P. Oleanolic acid mitigates
interleukin-1β-induced chondrocyte dysfunction by
regulating miR-148-3p-modulated FGF2 expression.
J. Gene Med. 22, e3169 (2020).
89. Shi, F. L. & Ren, L. X. Up-regulated miR-374a-3p
relieves lipopolysaccharides induced injury in
CHON-001 cells via regulating Wingless-type MMTV
integration site family member 5B. Mol. Cell Probes
51, 101541 (2020).
90. Wang, J. etal. Forkhead box C1 promotes the
pathology of osteoarthritis by upregulating β-catenin in
synovial fibroblasts. FEBS J. 287, 3065–3087 (2020).
91. Dinesh, P., Kalaiselvan, S., Sujitha, S. & Rasool, M.
MiR-145-5p mitigates dysregulated Wnt1/β-catenin
signaling pathway in rheumatoid arthritis.
Int. Immunopharmacol. 82, 106328 (2020).
92. Shui, X. etal. Identification and functional analysis of
long non-coding RNAs in the synovial membrane
of osteoarthritis patients. Cell Biochem. Funct. 38,
460–471 (2020).
93. Zhu, S., Deng, Y., Gao, H., Huang, K. & Nie, Z.
miR-877-5p alleviates chondrocyte dysfunction
in osteoarthritis models via repressing FOXM1.
J. Gene Med. 22, e3246 (2020).
94. Wei, H. etal. MicroRNA-15a/16/SOX5 axis promotes
migration, invasion and inflammatory response in
rheumatoid arthritis fibroblast-like synoviocytes.
Aging 12, 14376–14390 (2020).
95. Chen, J. & Wu, X. MicroRNA-103 contributes to
osteoarthritis development by targeting Sox6.
Biomed. Pharmacother. 118 , 109186 (2019).
96. Chunlei, H. etal. Down-regulation of mir-138-5p
protects chondrocytes ATDC5 and CHON-001 from
IL-1 β-induced inflammation via up-regulating SOX9.
Curr. Pharm. Des. 25, 4613–4621 (2020).
97. Guo, Y., Tian, L., Du, X. & Deng, Z. MiR-203 regulates
estrogen receptor α and cartilage degradation in
IL-1β-stimulated chondrocytes. J. Bone Min. Metab.
38, 346–356 (2020).
98. Tian, L., Su, Z., Ma, X., Wang, F. & Guo, Y. Inhibition
of miR-203 ameliorates osteoarthritis cartilage
degradation in the postmenopausal rat model:
involvement of estrogen receptor α. Hum. Gene
Ther. Clin. Dev. 30, 160–168 (2019).
99. Zhu, J. etal. lncRNAs: function and mechanism
in cartilage development, degeneration, and
regeneration. Stem Cell Res. Ther. 10, 344 (2019).
100. Yang, Y. etal. A long non-coding RNA, HOTAIR,
promotes cartilage degradation in osteoarthritis
by inhibiting WIF-1 expression and activating Wnt
pathway. BMC Mol. Cell Biol. 21, 53 (2020).
101. Wang, H., Li, J., Cheng, Y. & Yao, J. Association
of long-chain noncoding RNA H19 and MEG3
gene polymorphisms and their interaction with
risk of osteoarthritis in a Chinese Han population.
Genet. Test. Mol. Biomarkers 24, 328–337 (2020).
102. Zhang, L. etal. Yeast cell wall particle mediated
nanotube-RNA delivery system loaded with miR365
antagomir for post-traumatic osteoarthritis therapy
via oral route. Theranostics 10, 8479–8493 (2020).
103. Wang, Q., Wang, W., Zhang, F., Deng, Y. &
Long, Z. NEAT1/miR-181c Regulates osteopontin
(OPN)-mediated synoviocyte proliferation in
osteoarthritis. J. Cell Biochem. 118, 3775–3784
(2017).
104. Nakamura, A., Ali, S. A. & Kapoor, M. Antisense
oligonucleotide-based therapies for the treatment
of osteoarthritis: opportunities and roadblocks.
Bone 138, 115461 (2020).
105. Chen, Y. etal. Long non-coding RNA (lncRNA) small
nucleolar RNA host gene 15 (SNHG15) alleviates
osteoarthritis progression by regulation of
extracellular matrix homeostasis. Med. Sci. Monit.
26, e923868 (2020).
106. Chen, H., Yang, S. & Shao, R. Long non-coding XIST
raises methylation of TIMP-3 promoter to regulate
collagen degradation in osteoarthritic chondrocytes
after tibial plateau fracture. Arthritis Res. Ther. 21,
271 (2019).
107. Pan, H. etal. MicroRNA-410-3p modulates chondrocyte
apoptosis and inflammation by targeting high mobility
group Box 1 (HMGB1) in an osteoarthritis mouse
model. BMC Musculoskelet. Disord. 21, 486 (2020).
108. Zhao, H. & Gong, N. miR-20a regulates inflammatory
in osteoarthritis by targeting the IκBβ and regulates
NK-κB signaling pathway activation. Biochem.
Biophys. Res. Commun. 518, 632–637 (2019).
109. Lei, J., Fu, Y., Zhuang, Y., Zhang, K. & Lu, D. miR-382-3p
suppressed IL-1β induced inflammatory response of
chondrocytes via the TLR4/MyD88/NF-κB signaling
pathway by directly targeting CX43. J. Cell Physiol.
234, 23160–23168 (2019).
110 . Si, H. B. etal. miR-140 attenuates the progression
of early-stage osteoarthritis by retarding chondrocyte
senescence. Mol. Ther. Nucleic Acids 19, 15–30
(2020).
111. Papathanasiou, I., Balis, C., Trachana, V., Mourmoura, E.
& Tsezou, A. The synergistic function of miR-140-5p
and miR-146a on TLR4-mediated cytokine secretion
in osteoarthritic chondrocytes. Biochem. Biophys.
Res. Commun. 522, 783–791 (2020).
112 . Pearson, M. J. etal. Long intergenic noncoding
RNAs mediate the human chondrocyte inflammatory
response and are differentially expressed in
osteoarthritis cartilage. Arthritis Rheumatol. 68,
845–856 (2016).
113 . Yang, P. etal. Th17 cell pathogenicity and plasticity
in rheumatoid arthritis. J. Leukoc. Biol. 106,
1233–1240 (2019).
114 . Shui, X. etal. Knockdown of lncRNA NEAT1 inhibits
Th17/CD4(+) Tcell differentiation through reducing
the STAT3 protein level. J. Cell Physiol. 234,
22477–22484 (2019).
115 . Yang, P. etal. MicroRNA let-7g-5p alleviates murine
collagen-induced arthritis by inhibiting Th17 cell
differentiation. Biochem. Pharmacol. 174, 113822
(2020).
116 . Alivernini, S. etal. Distinct synovial tissue macrophage
subsets regulate inflammation and remission in
rheumatoid arthritis. Nat. Med. 26, 1295–1306
(2020).
117 . Paoletti, A. etal. Monocyte/macrophage
abnormalities specific to rheumatoid arthritis are
linked to miR-155 and are differentially modulated
by different TNF inhibitors. J. Immunol. 203,
1766–1775 (2019).
118 . Quero, L. etal. miR-221-3p drives the shift of
M2-macrophages to a pro-inflammatory function by
suppressing JAK3/STAT3 activation. Front. Immunol.
10, 3087 (2019).
119 . Ma, F., Li, G., Yu, Y., Xu, J. & Wu, X. MiR-33b-3p
promotes chondrocyte proliferation and inhibits
chondrocyte apoptosis and cartilage ECM degradation
by targeting DNMT3A in osteoarthritis. Biochem.
Biophys. Res. Commun. 519, 430–437 (2019).
120. Chen, H., Yang, J. & Tan, Z. Upregulation of
microRNA-9-5p inhibits apoptosis of chondrocytes
through downregulating Tnc in mice with
osteoarthritis following tibial plateau fracture.
J. Cell Physiol. 234, 23326–23336 (2019).
121. Liu, W., Zha, Z. & Wang, H. Upregulation of
microRNA-27a inhibits synovial angiogenesis and
chondrocyte apoptosis in knee osteoarthritis rats
through the inhibition of PLK2. J. Cell Physiol. 234,
22972–22984 (2019).
122. Chang, Q., Ji, M., Li, C. & Geng, R. Downregulation
of miR4865p alleviates LPS-induced inflammatory
injury, oxidative stress and apoptosis in chondrogenic
cell ATDC5 by targeting NRF1. Mol. Med. Rep. 22,
2123–2131 (2020).
123. Cheng, F., Hu, H., Sun, K., Yan, F. & Geng, Y.
miR-455-3p enhances chondrocytes apoptosis and
inflammation by targeting COL2A1 in the invitro
osteoarthritis model. Biosci. Biotechnol. Biochem. 84,
695–702 (2020).
www.nature.com/nrrheum
Reviews
0123456789();:
124. Wen, X. etal. MiR-455-3p reduces apoptosis and
alleviates degeneration of chondrocyte through
regulating PI3K/AKT pathway. Life Sci. 253, 117718
(2020).
125. Tao, H., Cheng, L. & Yang, R. Downregulation of
miR-34a promotes proliferation and inhibits apoptosis
of rat osteoarthritic cartilage cells by activating PI3K/Akt
pathway. Clin. Interv. Aging 15, 373–385 (2020).
126. Chen, S. & Li, B. MiR-128-3p post-transcriptionally
inhibits WISP1 to suppress apoptosis and
inflammation in human articular chondrocytes via the
PI3K/AKT/NF-κB signaling pathway. Cell Transpl. 29,
963689720939131 (2020).
127. Fan, Z. etal. MiR-155 promotes interleukin-
1β-induced chondrocyte apoptosis and catabolic
activity by targeting PIK3R1-mediated PI3K/Akt
pathway. J. Cell Mol. Med. 24, 8441–8451 (2020).
128. Wang, W. T. etal. microRNA-1236 promotes
chondrocyte apoptosis in osteoarthritis via direct
suppression of PIK3R3. Life Sci. 253, 117694 (2020).
129. Li, F., Yao, J., Hao, Q. & Duan, Z. miRNA-103
promotes chondrocyte apoptosis by down-regulation
of sphingosine kinase-1 and ameliorates PI3K/AKT
pathway in osteoarthritis. Biosci. Rep. 39,
BSR20191255 (2019).
130. Dong, C. etal. microRNA-mediated GAS1
downregulation promotes the proliferation of synovial
fibroblasts by PI3K-Akt signaling in osteoarthritis.
Exp. Ther. Med. 18, 4273–4286 (2019).
131. Shirdel, E. A., Xie, W., Mak, T. W. & Jurisica, I.
NAViGaTing the micronome — using multiple
microRNA prediction databases to identify signalling
pathway-associated microRNAs. PLoS ONE 6, e17429
(2011).
132. Liu, X. C. etal. MiR-1207-5p/CX3CR1 axis regulates
the progression of osteoarthritis via the modulation
of the activity of NF-κB pathway. Int. J. Rheum. Dis.
23, 1057–1065 (2020).
133. Li, L., Lv, G., Wang, B. & Kuang, L. XIST/miR-376c-5p/
OPN axis modulates the influence of proinflammatory
M1 macrophages on osteoarthritis chondrocyte
apoptosis. J. Cell Physiol. 235, 281–293 (2020).
134. Wang, Y. etal. miR-483-3p promotes cell proliferation
and suppresses apoptosis in rheumatoid arthritis
fibroblast-like synoviocytes by targeting IGF-1.
Biomed. Pharmacother. 130, 110519 (2020).
135. Rao, Y. etal. Delivery of long non-coding RNA NEAT1
by peripheral blood mononuclear cells-derived
exosomes promotes the occurrence of rheumatoid
arthritis via the microRNA-23a/MDM2/SIRT6 axis.
Front. Cell Dev. Biol. 8, 551681 (2020).
136. Xiao, P. etal. MicroRNA-613 alleviates IL-1β-induced
injury in chondrogenic CHON-001 cells by targeting
fibronectin 1. Am. J. Transl Res. 12, 5308–5319
(2020).
137. Yang, B., Xu, L. & Wang, S. Regulation of lncRNA-H19/
miR-140-5p in cartilage matrix degradation and
calcification in osteoarthritis. Ann. Palliat. Med. 9,
1896–1904 (2020).
138. Wei, W. etal. LINC01534 promotes the aberrant
metabolic dysfunction and inflammation in
IL-1β-simulated osteoarthritic chondrocytes
by targeting miR-140-5p. Cartilage https://doi.org/
10.1177/1947603519888787 (2019).
139. Duan, L., Liang, Y., Xu, X., Xiao, Y. & Wang, D.
Recent progress on the role of miR-140 in
cartilage matrix remodelling and its implications
for osteoarthritis treatment. Arthritis Res. Ther. 22,
194 (2020).
140. Balaskas, P. etal. Small non-coding RNAome of ageing
chondrocytes. Int. J. Mol. Sci. 21, 5675 (2020).
141. Peffers, M. J. etal. SnoRNA signatures in cartilage
ageing and osteoarthritis. Sci. Rep. 10, 10641
(2020).
142. Liu, D. etal. Synovial fibroblast-derived exosomal
microRNA-106b suppresses chondrocyte
proliferation and migration in rheumatoid arthritis via
down-regulation of PDK4. J. Mol. Med. 98, 409–423
(2020).
143. Chen, J. etal. Exosomal miRNA-486-5p derived
from rheumatoid arthritis fibroblast-like synoviocytes
induces osteoblast differentiation through the
Tob1/BMP/Smad pathway. Biomater. Sci. 8,
3430–3442 (2020).
144. Esmaeili, A., Hosseini, S. & Baghaban Eslaminejad, M.
Engineered-extracellular vesicles as an optimistic
tool for microRNA delivery for osteoarthritis
treatment. Cell Mol. Life Sci. 78, 79–91 (2020).
145. Asghar, S., Litherland, G. J., Lockhart, J. C.,
Goodyear, C. S. & Crilly, A. Exosomes in intercellular
communication and implications for osteoarthritis.
Rheumatology 59, 57–68 (2020).
146. Wu, X. etal. Extracellular vesicles: potential role in
osteoarthritis regenerative medicine. J. Orthop. Transl
21, 73–80 (2020).
147. Zhang, G., Zhou, Y., Su, M., Yang, X. & Zeng, B.
Inhibition of microRNA-27b-3p relieves osteoarthritis
pain via regulation of KDM4B-dependent DLX5.
Biofactors 46, 788–802 (2020).
148. Hoshikawa, N., Sakai, A., Takai, S. & Suzuki, H.
Targeting extracellular miR-21-TLR7 signaling
provides long-lasting analgesia in osteoarthritis.
Mol. Ther. Nucleic Acids 19, 199–207 (2020).
149. Bai, J. etal. LncRNA MM2P-induced, exosome-
mediated transfer of Sox9 from monocyte-derived
cells modulates primary chondrocytes. Cell Death Dis.
11, 763 (2020).
150. Jiang, L. & Cao, S. Role of microRNA-26a in cartilage
injury and chondrocyte proliferation and apoptosis
in rheumatoid arthritis rats by regulating expression
of CTGF. J. Cell Physiol. 235, 979–992 (2020).
151. Meng, Q. & Qiu, B. Exosomal microRNA-320a derived
from mesenchymal stem cells regulates rheumatoid
arthritis fibroblast-like synoviocyte activation by
suppressing CXCL9 expression. Front. Physiol. 11,
441 (2020).
152. Zheng, J. etal. Bone marrow-derived mesenchymal
stem cells-secreted exosomal microRNA-192-5p
delays inflammatory response in rheumatoid arthritis.
Int. Immunopharmacol. 78, 105985 (2020).
153. Sun, W. etal. Resolvin D1 suppresses pannus
formation via decreasing connective tissue growth
factor caused by upregulation of miRNA-146a-5p
in rheumatoid arthritis. Arthritis Res. Ther. 22, 61
(2020).
154. Wang, J. etal. Identification of a novel microRNA-141-
3p/Forkhead box C1/β-catenin axis associated with
rheumatoid arthritis synovial fibroblast function in vivo
and in vitro. Theranostics 10, 5412–5434 (2020).
155. Liang, Y. etal. Chondrocyte-targeted microRNA
delivery by engineered exosomes toward a cell-free
osteoarthritis therapy. ACS Appl. Mater. Interfaces
12, 36938–36947 (2020).
156. Lv, S. etal. MicroRNA-27b targets CBFB to inhibit
differentiation of human bone marrow mesenchymal
stem cells into hypertrophic chondrocytes. Stem Cell
Res. Ther. 11, 392 (2020).
157. Geng, Y. etal. Intra-articular injection of hUC-MSCs
expressing miR-140-5p induces cartilage self-repairing
in the rat osteoarthritis. J. Bone Min. Metab. 38,
277–288 (2020).
158. Abbasi Pashaki, P., Rahim, F., Habibi Roudkenar, M.,
Razavi-Toosi, S. & Ebrahimi, A. MicroRNA tough
decoy knockdowns miR-195 and represses
hypertrophy in chondrocytes. Appl. Biochem.
Biotechnol. 191, 1056–1071 (2020).
159. Nakamura, A. etal. microRNA-181a-5p antisense
oligonucleotides attenuate osteoarthritis in facet and
knee joints. Ann. Rheum. Dis. 78, 111–121 (2019).
160. Endisha, H. etal. MicroRNA-34a-5p promotes joint
destruction during osteoarthritis. Arthritis Rheumatol.
73, 426–439 (2020).
161. Lee, W. S. etal. MicroRNA-9 ameliorates destructive
arthritis through down-regulation of NF-κB1-RANKL
pathway in fibroblast-like synoviocytes. Clin. Immunol.
212, 108348 (2020).
162. Rai, M. F. etal. Applications of RNA interference in the
treatment of arthritis. Transl Res. 214, 1–16 (2019).
163. Wang, T. etal. LEF1 mediates osteoarthritis
progression through circRNF121/miR-665/MYD88
axis via NF-κB signaling pathway. Cell Death Dis. 11,
598 (2020).
164. Ying, H. etal. Cancer therapy using a self-replicating
RNA vaccine. Nat. Med. 5, 823–827 (1999).
165. Ragni, E. etal. miR-22-5p and miR-29a-5p are
reliable reference genes for analyzing extracellular
vesicle-associated miRNAs in adipose-derived
mesenchymal stem cells and are stable under
inflammatory priming mimicking osteoarthritis
condition. Stem Cell Rev. Rep. 15, 743–754 (2019).
166. Martinez-Sanchez, A., Lazzarano, S., Sharma, E.,
Lockstone, H. & Murphy, C. L. High-throughput
identification of miR-145 targets in human articular
chondrocytes. Life 10, 58 (2020).
167. Lu, J. etal. MiR-520d-5p modulates chondrogenesis
and chondrocyte metabolism through targeting
HDAC1. Aging 12, 18545–18560 (2020).
168. Ross, A. K. etal. The miRNA-mRNA interactome
of murine induced pluripotent stem cell-derived
chondrocytes in response to inflammatory cytokines.
FASEB J. 34, 11546–11561 (2020).
169. Skrzypa, M. etal. miRNA-146a-5p is upregulated
in serum and cartilage samples of patients with
osteoarthritis. Pol. Przegl. Chir. 91, 1–5 (2019).
170. Kolarz, B., Ciesla, M., Dryglewska, M., Rosenthal, A. K.
& Majdan, M. Hypermethylation of the miR-155 gene
in the whole blood and decreased plasma level
of miR-155 in rheumatoid arthritis. PLoS ONE 15,
e0233897 (2020).
171. Shao, L. & Hou, C. miR-138 activates NF-κB signaling
and PGRN to promote rheumatoid arthritis via
regulating HDAC4. Biochem. Biophys. Res. Commun.
519, 166–171 (2019).
172. Rezaeepoor, M. etal. Altered expression of
microRNAs may predict therapeutic response in
rheumatoid arthritis patients. Int. Immunopharmacol.
83, 106404 (2020).
173. Stypinska, B. etal. The serum cell-free microRNA
expression profile in MCTD, SLE, SSc, and RA
patients. J. Clin. Med. 9, 161 (2020).
174. Romo-Garcia, M. F. etal. Identification of putative
miRNA biomarkers in early rheumatoid arthritis
by genome-wide microarray profiling: a pilot study.
Gene 720, 144081 (2019).
175. Guo, D. etal. Study of miRNA interactome in active
rheumatoid arthritis patients reveals key pathogenic
roles of dysbiosis in the infection-immune network.
Rheumatology 60, 1512–1522 (2021).
176. Lim, M. K. etal. Serum exosomal miRNA-1915-3p is
correlated with disease activity of Korean rheumatoid
arthritis. Vivo 34, 2941–2945 (2020).
177. Mu, N. etal. Blockade of discoidin domain receptor 2
as a strategy for reducing inflammation and joint
destruction in rheumatoid arthritis via altered
interleukin-15 and Dkk-1 signaling in fibroblast-like
synoviocytes. Arthritis Rheumatol. 72, 943–956
(2020).
178. Wang, J. & Zhao, Q. LncRNA LINC-PINT increases
SOCS1 expression by sponging miR-155-5p to inhibit
the activation of ERK signaling pathway in rheumatoid
arthritis synovial fibroblasts induced by TNF-α.
Int. Immunopharmacol. 84, 106497 (2020).
179. Wang, J., Kong, X., Hu, H. & Shi, S. Knockdown
of long non-coding RNA PVT1 induces apoptosis of
fibroblast-like synoviocytes through modulating
miR-543-dependent SCUBE2 in rheumatoid arthritis.
J. Orthop. Surg. Res. 15, 142 (2020).
180. Li, Z. etal. Circ_0136474 and MMP-13 suppressed
cell proliferation by competitive binding to
miR-127-5p in osteoarthritis. J. Cell Mol. Med.
23, 6554–6564 (2019).
181. Chen, C., Yin, P., Hu, S., Sun, X. & Li, B. Circular
RNA-9119 protects IL-1β-treated chondrocytes
from apoptosis in an osteoarthritis cell model by
intercepting the microRNA-26a/PTEN axis. Life Sci.
256, 117924 (2020).
182. Shen, P. etal. CircCDK14 protects against
osteoarthritis by sponging miR-125a-5p and
promoting the expression of Smad2. Theranostics
10, 9113–9131 (2020).
183. Wu, Q., Yuan, Z. H., Ma, X. B. & Tang, X. H.
Low expression of CircRNA HIPK3 promotes
osteoarthritis chondrocyte apoptosis by serving as a
sponge of miR-124 to regulate SOX8. Eur. Rev. Med.
Pharmacol. Sci. 24, 7937–7945 (2020).
184. Ma, H. R. etal. CircVCAN regulates the proliferation
and apoptosis of osteoarthritis chondrocyte through
NF-κB signaling pathway. Eur. Rev. Med. Pharmacol.
Sci. 24, 6517–6525 (2020).
185. Chen, G., Liu, T., Yu, B., Wang, B. & Peng, Q.
CircRNA-UBE2G1 regulates LPS-induced
osteoarthritis through miR-373/HIF-1a axis.
Cell Cycle 19, 1696–1705 (2020).
186. Ni, J. L., Dang, X. Q. & Shi, Z. B. CircPSM3
inhibits the proliferation and differentiation of
OA chondrocytes by targeting miRNA-296-5p.
Eur. Rev. Med. Pharmacol. Sci. 24, 3467–3475
(2020).
187. Zhang, W. etal. Circular RNA-CDR1as acts as the
sponge of microRNA-641 to promote osteoarthritis
progression. J. Inflamm. 17, 8 (2020).
188. Bai, Z. M., Kang, M. M., Zhou, X. F. & Wang, D.
CircTMBIM6 promotes osteoarthritis-induced
chondrocyte extracellular matrix degradation via
miR-27a/MMP13 axis. Eur. Rev. Med. Pharmacol. Sci.
24, 7927–7936 (2020).
189. Zhu, H., Hu, Y., Wang, C., Zhang, X. & He, D.
CircGCN1L1 promotes synoviocyte proliferation and
chondrocyte apoptosis by targeting miR-330-3p
and TNF-α in TMJ osteoarthritis. Cell Death Dis. 11,
284 (2020).
190. Zhong, S. etal. Hsa_circ_0088036 promotes
the proliferation and migration of fibroblast-like
synoviocytes by sponging miR-140-3p and
upregulating SIRT 1 expression in rheumatoid
arthritis. Mol. Immunol. 125, 131–139 (2020).
Nature reviews
|
Rheumatology
Reviews
0123456789();:
191. Yang, J. etal. CircRNA_09505 aggravates
inflammation and joint damage in collagen-induced
arthritis mice via miR-6089/AKT1/NF-κB axis.
Cell Death Dis. 11, 833 (2020).
192. Li, X. etal. Promising targets and drugs in rheumatoid
arthritis: a module-based and cumulatively scoring
approach. Bone Jt. Res. 9, 501–514 (2020).
193. Xiang, S., Li, Z., Bian, Y. & Weng, X. RNA sequencing
reveals the circular RNA expression profiles of
osteoarthritic synovium. J. Cell Biochem. 120,
18031–18040 (2019).
194. Xiao, K. etal. Circular RNA expression profile of knee
condyle in osteoarthritis by illumina HiSeq platform.
J. Cell Biochem. 120, 17500–17511 (2019).
195. Wang, Y. etal. Screening for differentially expressed
circular RNAs in the cartilage of osteoarthritis patients
for their diagnostic value. Genet. Test. Mol. Biomark.
23, 706–716 (2019).
196. Chen, H. & Chen, L. An integrated analysis of the
competing endogenous RNA network and co-expression
network revealed seven hub long non-coding RNAs
in osteoarthritis. Bone Jt. Res. 9, 90–98 (2020).
Acknowledgements
The work of M.J.P. is supported by a Wellcome Trust
Intermediate Clinical Fellowship (107471/Z/15/Z) and by the
Medical Research Council and Versus Arthritis as part of
the Medical Research Council Versus Arthritis Centre for
Integrated Research into Musculoskeletal Ageing
(MR/R502182/1). The work of M.J.O. is supported by the
Veterans Health Administration CDA (IK2CX001269) and by
an Arthritis Foundation Delivering on Discovery grant. I.J. is
also at the Departments of Medical Biophysics and Computer
Science, University of Toronto, Toronto, Ontario, Canada and
the Institute of Neuroimmunology, Slovak Academy of
Sciences, Bratislava, Slovakia. The work of I.J. was funded in
part by the Ontario Research Fund (no. 34876 and GL2-01-
030), the Natural Sciences Research Council (NSERC
no. 203475), Genome Canada (DIG2 no. 14408), the Canada
Foundation for Innovation (CFI no. 29272, no. 225404,
no. 33536) and the Canada Research Chair Program (CRC
no. 203373 and no. 225404). Additional support is provided
by the Schroeder Arthritis Institute via the Toronto General and
Western Hospital Foundation, University Health Network.
The work of M.K. is supported by the Canadian Institute
of Health Research Operating grant (no. 156299) and the
Tier 1 Canada Research Chair Award (no. 950-232237).
Author contributions
The authors contributed equally to all aspects of the article.
Competing interests
S.A.A. and M.K. declare that they have filed a US Provisional
Patent Application no. 63/033,463 titled “Circulating
MicroRNAs in Knee Osteoarthritis and Uses Thereof”. The
other authors declare no competing interests.
Disclaimer
None of the funders had a role in study design, data collection
and analysis, decision to publish or preparation of the
manuscript.
Peer review information
Nature Reviews Rheumatology thanks S. Jones and the other,
anonymous, reviewer(s) for their contribution to the peer
review of this work.
Publisher’s note
Springer Nature remains neutral with regard to jurisdictional
claims in published maps and institutional affiliations.
Review criteria
A literature search was performed in PubMed for articles
published in the past 2 years using combinations of the fol-
lowing key words: “osteoarthritis”, “rheumatoid arthritis”,
“microRNA”, “long non-coding RNA”, “circular RNA”, “small
nucleolar RNAs” and “transfer RNAs”. Some highly relevant
papers outside the search criteria were also included.
RELATED LINKS
circAtlas: http://159.226.67.237:8080/new/index.php
circBase: http://www.circbase.org/
DIANA-lncBase: https://diana.e-ce.uth.gr/lncbasev3
EMBL-EBI European Nucleotide Archive: https://www.ebi.
ac.uk/ena/browser/home
Enrichr: https://maayanlab.cloud/Enrichr/
Gene Ontology Resource: http://geneontology.org
Hugo Gene Nomenclature: https://www.genenames.org/
miRanda: https://bioweb.pasteur.fr/packages/
pack@miRanda@3.3a
miRbase: http://www.mirbase.org/
miRDB: http://mirdb.org/
miRDeep2: https://github.com/rajewsky-lab/mirdeep2
mirDIP: http://ophid.utoronto.ca/mirDIP/
NCBI Database of Genotypes and Phenotypes:
https://www.ncbi.nlm.nih.gov/gap/
NCBI Gene Expression Omnibus repository:
https://www.ncbi.nlm.nih.gov/gds
NCBI Sequence Read Archive: https://www.ncbi.nlm.nih.gov/sra
pathDIP: http://ophid.utoronto.ca/pathDIP/
TargetScan: http://www.targetscan.org/vert_72/
© Springer Nature Limited 2021
www.nature.com/nrrheum
Reviews
... Non-coding RNAs, such as microRNAs (miRNAs), have been considered crucial regulators of the gene expression over the past decades. 15,16 MiRNA has approximately 20 nucleotides, and it can inhibit protein translation by competitively binding to mRNA, without affecting the gene transcription process. 15,16 Past studies have reported that miRNAs are involved in the IVDD process by binding to the 3′-UTR of IVDD-related target genes, followed by mediation of the inflammatory response, NPC death, and ECM metabolism. ...
... 15,16 MiRNA has approximately 20 nucleotides, and it can inhibit protein translation by competitively binding to mRNA, without affecting the gene transcription process. 15,16 Past studies have reported that miRNAs are involved in the IVDD process by binding to the 3′-UTR of IVDD-related target genes, followed by mediation of the inflammatory response, NPC death, and ECM metabolism. [16][17][18][19] Our previous study demonstrated that miR-874-3p is remarkably downregulated in IVDD and negatively correlated with the severity of IVDD. ...
... 15,16 Past studies have reported that miRNAs are involved in the IVDD process by binding to the 3′-UTR of IVDD-related target genes, followed by mediation of the inflammatory response, NPC death, and ECM metabolism. [16][17][18][19] Our previous study demonstrated that miR-874-3p is remarkably downregulated in IVDD and negatively correlated with the severity of IVDD. 20 Nevertheless, the biological effects and regulatory mechanisms of miR-874-3p in IVDD remains unclear. ...
Article
Full-text available
Intervertebral disc degeneration (IVDD) severely affects the work and the quality of life of people. We previously demonstrated that silencing activation transcription factor 3 (ATF3) blocked the IVDD pathological process by regulating nucleus pulposus cell (NPC) ferroptosis, apoptosis, inflammation, and extracellular matrix (ECM) metabolism. Nevertheless, whether miR‐874‐3p mediated the IVDD pathological process by targeting ATF3 remains unclear. We performed single‐cell RNA sequencing (scRNA‐seq) and bioinformatics analysis to identify ATF3 as a key ferroptosis gene in IVDD. Then, Western blotting, flow cytometry, ELISA, and animal experiments were performed to validate the roles and regulatory mechanisms of miR‐874‐3p/ATF3 signalling axis in IVDD. ATF3 was highly expressed in IVDD patients and multiple cell types of IVDD rat, as revealed by scRNA‐seq and bioinformatics analysis. GO analysis unveiled the involvement of ATF3 in regulating cell apoptosis and ECM metabolism. Furthermore, we verified that miR‐874‐3p might protect against IVDD by inhibiting NPC ferroptosis, apoptosis, ECM degradation, and inflammatory response by targeting ATF3. In vivo experiments displayed the protective effect of miR‐874‐3p/ATF3 axis on IVDD. These findings propose the potential of miR‐874‐3p and ATF3 as biomarkers of IVDD and suggest that targeting the miR‐874‐3p/ATF3 axis may be a therapeutic target for IVDD.
... miRNAs are a class of non-coding RNAs ranging from 18 to 24 nucleotides in length, which exert inhibitory effects on the expression of target genes through interactions with mRNA. Studies have demonstrated that miRNAs play a crucial role as a pathogenic factor in OA (Ali et al., 2021). For instance, miR-199b-5p was found to contribute to the osteogenic differentiation of bone marrow stromal cells (Zhao et al., 2016) another miR-140 showed cartilage-specific expression, and its expression was significantly reduced in OA cartilage (Yamashita et al., 2012;Miyaki et al., 2009). ...
... Exosomes are rich in miRNAs, and various cells can secrete exosomes to target cells under physiological and pathological conditions, which function as delivery vehicles for miRNAs (Kalluri and LeBleu, 2020;Sun et al., 2019). There is evidence suggesting that miRNAs can participate in various cellular processes (inflammation, cell viability, ECM dysregulation) and signaling pathways (Hedgehog signaling, PI3K-AKT signaling) relevant to OA (15, Ali et al., 2021;Jiang et al., 2023). Our differential miRNA enrichment analysis also strongly supports the correlation with these findings. ...
Article
Full-text available
Osteoarthritis (OA) is a degenerative disease with a high prevalence in the elderly population, but our understanding of its mechanisms remains incomplete. Analysis of serum exosomal small RNA sequencing data from clinical patients and gene expression data from OA patient serum and cartilage obtained from the GEO database revealed a common dysregulated miRNA, miR-199b-5p. In vitro cell experiments demonstrated that miR-199b-5p inhibits chondrocyte vitality and promotes extracellular matrix degradation. Conversely, inhibition of miR-199b-5p under inflammatory conditions exhibited protective effects against damage. Local viral injection of miR-199b-5p into mice induced a decrease in pain threshold and OA-like changes. In an OA model, inhibition of miR-199b-5p alleviated the pathological progression of OA. Furthermore, bioinformatics analysis and experimental validation identified Gcnt2 and Fzd6 as potential target genes of MiR-199b-5p . Thus, these results indicated that MiR-199b-5p / Gcnt2 and Fzd6 axis might be a novel therapeutic target for the treatment of OA.
... Previous studies have also suggested that these non-coding transcription processes regulate key gene expression under physiological and pathological conditions. 4,5) More importantly, ncRNAs regulate the pathological process of AMI, mainly including long ncRNAs (lncRNAs), microRNAs (microRNAs), and circular RNAs (circRNAs). 6,7) In addition, lncRNA, microRNA, and cir-cRNA interact with each other, making the functions of these ncRNAs more complex and comprehensive. ...
Article
This study aimed to explore the expression of long non-coding RNA (lncRNA) nuclear paraspeckle assembly transcript 1 (NEAT1) in patients with acute myocardial infarction (AMI) and its inflammatory regulation mechanism through miR-211/interleukin 10 (IL-10) axis. A total of 75 participants were enrolled in this study: 25 healthy people in the control group, 25 patients with stable angina pectoris (SAP) in the SAP group, and 25 patients with AMI in the AMI group. Real-time qPCR was used to detect mRNA expression levels of NEAT1, miR-211, and IL-10. The interaction between miR-211, NEAT1, and IL-10 was confirmed by dual-luciferase reporter assay, and protein expression was detected using western blot. High expression of NEAT1 in peripheral blood mononuclear cells (PBMCs) of patients with AMI was negatively related to serum creatine kinase-MB (CK-MB), cardiac troponin I (cTnI), tumor necrosis factor-α (TNF-α), IL-6, and IL-1β and was positively correlated with left ventricular ejection fraction (LVEF). In THP-1 cells, miR-211 was confirmed to target and inhibit IL-10 expression. NEAT1 knockdown and miR-211-mimic markedly decreased IL-10 protein levels, whereas anti-miR-211 markedly increased IL-10 protein levels. Importantly, miR-211 level was negatively related to NEAT1 and IL-10 levels, whereas IL-10 level was positively related to the level of NEAT1 expression in PBMCs of patients with AMI. LncRNA NEAT1 was highly expressed in PBMCs of patients with AMI, and NEAT1 suppressed inflammation via miR-211/IL-10 axis in PBMCs of patients with AMI.
... Furthermore, sEVs boast low immunogenicity and possess the ability to traverse the blood-brain barrier, making them ideal non-cellular carriers of molecular information [15]. miRNAs, a class of short non-coding RNAs, have emerged as prominent regulators of gene expression at the posttranscriptional level [16,17]. The inclusion of miRNAs in sEVs confers protection against enzymatic degradation in bodily fluids, enabling them to reach target cells and exert robust biological regulatory effects [18]. ...
Article
Full-text available
Background Rheumatoid arthritis (RA) is a chronic autoimmune disorder characterized by synovial inflammation, causing substantial disability and reducing life quality. While macrophages are widely appreciated as a master regulator in the inflammatory response of RA, the precise mechanisms underlying the regulation of proliferation and inflammation in RA-derived fibroblast-like synoviocytes (RA-FLS) remain elusive. Here, we provide extensive evidence to demonstrate that macrophage contributes to RA microenvironment remodeling by extracellular vesicles (sEVs) and downstream miR-100-5p/ mammalian target of rapamycin (mTOR) axis. Results We showed that bone marrow derived macrophage (BMDM) derived-sEVs (BMDM-sEVs) from collagen-induced arthritis (CIA) mice (cBMDM-sEVs) exhibited a notable increase in abundance compared with BMDM-sEVs from normal mice (nBMDM-sEVs). cBMDM-sEVs induced significant RA-FLS proliferation and potent inflammatory responses. Mechanistically, decreased levels of miR-100-5p were detected in cBMDM-sEVs compared with nBMDM-sEVs. miR-100-5p overexpression ameliorated RA-FLS proliferation and inflammation by targeting the mTOR pathway. Partial attenuation of the inflammatory effects induced by cBMDM-sEVs on RA-FLS was achieved through the introduction of an overexpression of miR-100-5p. Conclusions Our work reveals the critical role of macrophages in exacerbating RA by facilitating the transfer of miR-100-5p-deficient sEVs to RA-FLS, and sheds light on novel disease mechanisms and provides potential therapeutic targets for RA interventions. Graphical abstract
Chapter
Rheumatic diseases are much more common among women than men, suggesting that the X chromosome and sex hormones are actively involved in the sex difference of the diseases. In particular, X chromosome inactivation is one of the key determinants of sex difference in rheumatic diseases, causing a different gene-dose effect between the two sexes. Another important mechanism is microRNAs (miRNAs), 10% of which are residing in the X chromosome, while a few are found in Y. These small noncoding RNAs regulate the translation of a specific gene and many of X-chromosome-residing miRNAs are related to immune genes. The third factor contributing to the sex difference in rheumatic diseases is sex hormones. In general, testosterones negatively regulate immune response, while estrogens reinforce it. In addition, estrogens can activate or inhibit miRNA related to immune functions, further contributing to the sex difference of rheumatic diseases. This chapter will present how these mechanisms work in rheumatic diseases by taking examples of systemic lupus erythematosus and rheumatoid arthritis.
Article
Osteoclasts are the cells primarily responsible for inflammation‐induced bone loss, as is particularly seen in rheumatoid arthritis. Increasing evidence suggests that osteoclasts formed under homeostatic versus inflammatory conditions may differ in phenotype. While microRNA‐29‐3p family members (miR‐29a‐3p, miR‐29b‐3p, miR‐29c‐3p) promote the function of RANKL‐induced osteoclasts, the role of miR‐29‐3p during inflammatory TNF‐α‐induced osteoclastogenesis is unknown. We used bulk RNA‐seq, histology, qRT‐PCR, reporter assays, and western blot analysis to examine bone marrow monocytic cell cultures and tissue from male mice in which the function of miR‐29‐3p family members was decreased by expression of a miR‐29‐3p tough decoy (TuD) competitive inhibitor in the myeloid lineage (LysM‐cre). We found that RANKL‐treated monocytic cells expressing the miR‐29‐3p TuD developed a hypercytokinemia/proinflammatory gene expression profile in vitro, which is associated with macrophages. These data support the concept that miR‐29‐3p suppresses macrophage lineage commitment and may have anti‐inflammatory effects. In correlation, when miR‐29‐3p activity was decreased, TNF‐α‐induced osteoclast formation was accentuated in an in vivo model of localized osteolysis and in a cell‐autonomous manner in vitro. Further, miR‐29‐3p targets mouse TNF receptor 1 (TNFR1/ Tnfrsf1a ), an evolutionarily conserved regulatory mechanism, which likely contributes to the increased TNF‐α signaling sensitivity observed in the miR‐29‐3p decoy cells. Whereas our previous studies demonstrated that the miR‐29‐3p family promotes RANKL‐induced bone resorption, the present work shows that miR‐29‐3p dampens TNF‐α‐induced osteoclastogenesis, indicating that miR‐29‐3p has pleiotropic effects in bone homeostasis and inflammatory osteolysis. Our data supports the concept that the knockdown of miR‐29‐3p activity could prime myeloid cells to respond to an inflammatory challenge and potentially shift lineage commitment toward macrophage, making the miR‐29‐3p family a potential therapeutic target for modulating inflammatory response.
Article
Full-text available
Background Rheumatoid arthritis (RA) is a chronic inflammatory disease that leads to progressive joint damage. Circular RNA (circRNA) can regulate the inflammatory response of fibroblast-like synoviocytes (FLSs) in RA, influencing the disease progression. Paeoniflorin (PF) is the main active ingredient extracted from Paeonia lactiflora and is known for its anti-inflammatory effect. This study aims to explore the potential mechanisms by which hsa_circ_009012 and PF regulate the inflammatory response in RA. Methods RNA expression of hsa_circ_009012, has-microRNA-1286 (miR-1286), toll-like receptor 4 (TLR4), NOD-like receptor thermal protein domain associated protein 3 (NLRP3) was assessed by real-time quantitative polymerase chain reaction (RT-qPCR) or western blotting (WB). Cell inflammation markers (TNF-α, IL-1β, IL-6) were assessed by RT-qPCR and immunofluorescence (IF). Counting Kit-8 (CCK-8) assay, flow cytometry, and transwell assay were utilized to test cell viability, cell cycle distribution, and migration. Results Hsa_circ_009012 was highly expressed in RA-FLS. Hsa_circ_009012 over-expression facilitated the inflammation in RA-FLS and was closely associated with the miR-1286/TLR4 axis. Paeoniflorin inhibited inflammation and the expression of hsa_circ_009012 and TLR4, while upregulating the expression of miR-1286 in RA-FLS. Moreover, the upregulation of hsa_circ_009012 reversed the repressive effect of paeoniflorin on RA-FLS progression. Conclusion Paeoniflorin inhibits the inflammation of RA-FLS via mediating the hsa_circ_009012/miR-1286/TLR4/NLRP3 axis.
Article
Full-text available
Cardiovascular diseases pose a serious threat to human health. The onset of cardiovascular diseases involves the comprehensive effects of multiple genes and environmental factors, and multiple signaling pathways are involved in regulating the occurrence and development of cardiovascular diseases. The Hippo pathway is a highly conserved signaling pathway involved in the regulation of cell proliferation, apoptosis, and differentiation. Recently, it has been widely studied in the fields of cardiovascular disease, cancer, and cell regeneration. Non-coding RNA (ncRNAs), which are important small molecules for the regulation of gene expression in cells, can directly target genes and have diverse regulatory functions. Recent studies have found that ncRNAs interact with Hippo pathway components to regulate myocardial fibrosis, cardiomyocyte proliferation, apoptosis, and hypertrophy and play an important role in cardiovascular disease. In this review, we describe the mode of action of ncRNAs in regulating the Hippo pathway, provide new ideas for further research, and identify molecules involved in the mechanism of action of ncRNAs and the Hippo pathway as potential therapeutic targets, with the aim of finding new modes of action for the treatment and prevention of cardiovascular diseases.
Article
Full-text available
Ultraviolet (UV) is divided into UVA (long-wave, 320-400 nm), UVB (middle-wave, 280-320 nm) and UVC (short-wave, 100-280 nm) based on wavelength. UV radiation (UVR) from sunlight (UVA + UVB) is a major cause of skin photodamage including skin inflammation, aging and pigmentation. Accidental exposure to UVC burns the skin and induces skin cancer. In addition to the skin, UV radiation can also impair visual function. Non-coding RNAs (ncRNAs) are a class of functional RNAs that do not have coding activity but can control cellular processes at the post-transcriptional level, including microRNA (miRNA), long non-coding RNA (lncRNA) and circulatory RNA (circRNA). Through a review of the literature, it was determined that UVR can affect the expression of various ncRNAs, and that this regulation may be wavelength specific. Functionally, ncRNAs participate in the regulation of photodamage through various pathways and play pathogenic or protective regulatory roles. In addition, ncRNAs that are upregulated or downregulated by UVR can serve as biomarkers for UV-induced diseases, aiding in diagnosis and prognosis assessment. Therapeutic strategies targeting ncRNAs, including the use of natural drugs and their extracts, have shown protective effects against UV-induced photodamage. In the present review, an extensive summarization of previous studies was performed and the role and mechanism of ncRNAs in UV-induced radiation effects was reviewed to aid in the diagnosis and treatment of UV-related diseases.
Article
Full-text available
Metastasis Associated Lung Adenocarcinoma Transcript-1 (MALAT1) is implicated in regulating the inflammatory response and in the pathology of several chronic inflammatory diseases, including osteoarthritis (OA). The purpose of this study was to examine the relationship between OA subchondral bone expression of MALAT1 with parameters of joint health and biomarkers of joint inflammation, and to determine its functional role in human OA osteoblasts. Subchondral bone and blood were collected from hip and knee OA patients (n = 17) and bone only from neck of femur fracture patients (n = 6) undergoing joint replacement surgery. Cytokines were determined by multiplex assays and ELISA, and gene expression by qPCR. MALAT1 loss of function was performed in OA patient osteoblasts using locked nucleic acids. The osteoblast transcriptome was analysed by RNASeq and pathway analysis. Bone expression of MALAT1 positively correlated to serum DKK1 and galectin-1 concentrations, and in OA patient osteoblasts was induced in response to IL-1β stimulation. Osteoblasts depleted of MALAT1 exhibited differential expression (>1.5 fold change) of 155 genes, including PTGS2. Both basal and IL-1β-mediated PGE2 secretion was greater in MALAT1 depleted osteoblasts. The induction of MALAT1 in human OA osteoblasts upon inflammatory challenge and its modulation of PGE2 production suggests that MALAT1 may play a role in regulating inflammation in OA subchondral bone.
Article
Full-text available
The rapid expansion of Next Generation Sequencing (NGS) data availability has made exploration of appropriate bioinformatics analysis pipelines a timely issue. Since there are multiple tools and combinations thereof to analyze any dataset, there can be uncertainty in how to best perform an analysis in a robust and reproducible manner. This is especially true for newer omics applications, such as miRNomics, or microRNA sequencing (miRNA-seq). As compared to transcriptomics, there have been far fewer miRNA-seq studies performed to date, and those that are reported seldom provide detailed description of the bioinformatics analysis, including aspects such as Unique Molecular Identifiers (UMIs). In this article, we attempt to fill the gap and help researchers understand their miRNA-seq data and its analysis. This article will specifically discuss a customizable miRNA bioinformatics pipeline, that was developed using miRNA-seq datasets generated from human OA plasma samples. We describe quality assessment of raw sequencing data files, reference-based alignment, counts generation for miRNA expression levels, and novel miRNA discovery. This report is expected to improve clarity and reproducibility of the bioinformatics portion of miRNA-seq analysis, applicable across any sample type, to promote sharing of detailed protocols across the NGS field.
Article
Full-text available
circular (circ)RNAs are an important group of non-coding RNAs involved in different pathological and physiological functions, such as longitudinal bone growth. However, the effects of an increase or decrease in circRNA expression on idiopathic short stature (ISS) remain largely unknown. The present study compared the circRNA expression patterns of patients with ISS and healthy individuals to identify differentially expressed circRNAs involved in the regulation of ISS pathogenesis and their target microRNAs (miR). Microarray analysis revealed that 145 circRNAs were differentially expressed in patients with ISS, including 83 up- and 62 downregulated circRNAs. Reverse transcription-quantitative PCR confirmed that hsa_circRNA_0079201 was increased in patients with ISS compared with that in the normal individuals, whilst hsa_circRNA_0079201 overexpression in human chondrocytes was shown to significantly suppress their proliferation, hypertrophy and endochondral ossification abilities. Luciferase reporter assays identified that circRNA_0079201 acted as an miR-140-3p sponge. In situ hybridization confirmed the co‑localization of circRNA_0079201 and miR‑140‑3p in the human chondrocyte and neonatal femur growth plate of C57 mice, while rescue experiments demonstrated that miR-140-3p overexpression reversed the inhibition of human chondrocyte proliferation, hypertrophy and endochondral ossification, caused by circRNA_0079201 overexpression. Bioinformatics analysis and luciferase reporter assays revealed that SMAd2 was a potential target gene of miR-140-3p. Furthermore, overexpressing circRNA_0079201 in human chondrocytes suppressed miR-140-3p and increased SMAd2
Article
Full-text available
Background Chronic postoperative pain affects approximately 20% of patients with knee osteoarthritis after total knee replacement. Circulating microRNAs can be found in serum and might act as biomarkers in a variety of diseases. The current study aimed to investigate the preoperative expression of circulating microRNAs as potential predictive biomarkers for the development of chronic postoperative pain in the year following total knee replacement. Methods Serum samples, collected preoperatively from 136 knee osteoarthritis patients, were analyzed for 21 circulatory microRNAs. Pain intensity was assessed using a visual analog scale before and one year after total knee replacement. Patients were divided into a low-pain relief group (pain relief percentage <30%) and a high-pain relief group (pain relief percentage >30%) based on their pain relief one year after total knee replacement, and differences in microRNAs expression were analyzed between the two groups. Results We found that three microRNAs were preoperatively dysregulated in serum in the low-pain relief group compared with the high-pain relief group. MicroRNAs hsa-miR-146a-5p, -145-5p, and -130 b-3p exhibited fold changes of 1.50, 1.55, and 1.61, respectively, between the groups (all P values < 0.05). Hsa-miR-146a-5p and preoperative pain intensity correlated positively with postoperative pain relief (respectively, R = 0.300, P = 0.006; R = 0.500, P < 0.001). Discussion This study showed that patients with a low postoperative pain relief present a dysregulation of circulating microRNAs. Altered circulatory microRNAs expression correlated with postoperative pain relief, indicating that microRNAs can serve as predictive biomarkers of pain outcome after surgery and hence may foster new strategies for preventing chronic postoperative pain after total knee replacement (TKR).
Article
Full-text available
Objective MicroRNA‐34a‐5p (miR‐34a‐5p) expression is elevated in the synovial fluid of patients with late‐stage knee osteoarthritis (OA); however, its exact role and therapeutic potential in OA remain to be fully elucidated. This study was undertaken to examine the role of miR‐34a‐5p in OA pathogenesis. Methods Expression of miR‐34a‐5p was determined in joint tissues and human plasma (n = 71). Experiments using miR‐34a‐5p mimic or antisense oligonucleotide (ASO) treatment were performed in human OA chondrocytes, fibroblast‐like synoviocytes (FLS) (n = 7–9), and mouse OA models, including destabilization of the medial meniscus (DMM; n = 22) and the accelerated, more severe model of mice fed a high‐fat diet and subjected to DMM (n = 11). Wild‐type (WT) mice (n = 9) and miR‐34a–knockout (KO) mice (n = 11) were subjected to DMM. Results were expressed as the mean ± SEM and analyzed by t‐test or analysis of variance, with appropriate post hoc tests. P values less than 0.05 were considered significant. RNA sequencing was performed on WT and KO mouse chondrocytes. Results Expression of miR‐34a‐5p was significantly increased in the plasma, cartilage, and synovium of patients with late‐stage OA and in the cartilage and synovium of mice subjected to DMM. Plasma miR‐34a‐5p expression was significantly increased in obese patients with late‐stage OA, and in the plasma and knee joints of mice fed a high‐fat diet. In human OA chondrocytes and FLS, miR‐34a‐5p mimic increased key OA pathology markers, while miR‐34a‐5p ASO improved cellular gene expression. Intraarticular miR‐34a‐5p mimic injection induced an OA‐like phenotype. Conversely, miR‐34a‐5p ASO injection imparted cartilage‐protective effects in the DMM and high‐fat diet/DMM models. The miR‐34a–KO mice exhibited protection against DMM‐induced cartilage damage. RNA sequencing of WT and KO chondrocytes revealed a putative miR‐34a‐5p signaling network. Conclusion Our findings provide comprehensive evidence of the role and therapeutic potential of miR‐34a‐5p in OA.
Article
Full-text available
A number of circular RNAs (circRNAs) have been implicated in rheumatoid arthritis (RA) pathogenesis; however, little is known about their function and hidden molecular mechanism in immune and inflammation regulation. We investigated the role and the underlying mechanism of circRNA_09505 in RA in this study. Real-time PCR and fluorescence in situ hybridization (FISH) are adopted to estimate the quantitative expression and localization of circRNA_09505 in macrophages. The altering effect of circRNA_09505 on inflammation is investigated in vitro and in vivo by use of macrophage cell models and collagen-induced arthritis (CIA) mice. Luciferase reporter assay and RNA-binding protein immunoprecipitation (RIP) are used to confirm the circRNA_09505/miR-6089 ceRNA network predicted by bioinformatics analysis. Compared with controls, the expression of circRNA_09505 is upregulated in peripheral blood mononuclear cells (PBMCs) from patients with RA. The proliferation and cell cycle are significantly promoted when circRNA_09505 is upregulated in macrophages, whereas knockdown of circRNA_09505 inhibits macrophage proliferation and cell- cycle progression. Besides, circRNA_09505 can act as a miRNA sponge for miR-6089 in macrophages, and promote the production of TNF-α, IL-6, and IL-12 through ceRNA mechanism. Moreover, AKT1 is a direct target of miR-6089. CircRNA_09505 can promote AKT1 expression by acting as a miR-6089 sponge via IκBα/NF-κB signaling pathway in macrophages. Most interestingly, knockdown of circRNA_09505 significantly alleviates arthritis and inflammation in vivo in CIA mice. These data support the hypothesis that circRNA_09505 can function as a miR-6089 sponge and regulate inflammation via miR-6089/AKT1/NF-κB axis in CIA mice.
Article
Full-text available
Long non-coding RNAs (lncRNAs) are recently-discovered transcripts that regulate vital cellular processes and are crucially connected to diseases. Despite their unprecedented molecular complexity, it is emerging that lncRNAs possess distinct structural motifs. Remarkably, the 3D shape and topology of full-length, native lncRNAs have been visualized for the first time in the last year. These studies reveal that lncRNA structures dictate lncRNA functions. Here, we review experimentally determined lncRNA structures and emphasize that lncRNA structural characterization requires synergistic integration of computational, biochemical and biophysical approaches. Based on these emerging paradigms, we discuss how to overcome the challenges posed by the complex molecular architecture of lncRNAs, with the goal of obtaining a detailed understanding of lncRNA functions and molecular mechanisms in the future.
Article
This paper presents both inaccuracies and mistakes. Therefore, the article "CircVCAN regulates the proliferation and apoptosis of osteoarthritis chondrocyte through NF-κB signaling pathway, by H.-R. Ma, W.-B. Mu, K.-Y. Zhang, H.-K. Zhou, R.-D. Jiang, L. Cao, published in Eur Rev Med Pharmacol Sci 2020; 24 (12): 6517-6525-DOI: 10.26355/eurrev_202006_21635-PMID: 32633338" has been withdrawn. The Publisher apologizes for any inconvenience this may cause. https://www.europeanreview.org/article/21635.
Article
Background: Osteoarthritis (OA) is an aging-related chronic degenerative joint disease. A number of miRNAs have been found to be involved in the development of OA, but the role of miR-613 in OA remains unclear. Thus, this study aimed to investigate the role of miR-613 during the progression of OA. Methods: CHON-001 cells were transfected with miR-613 agonist for 48 h, and then exposed to 10 ng/mL IL-1β for 24 h. Cell viability, cell proliferation and cell apoptosis in CHON-001 cells were assessed by CCK-8, immunofluorescence, and flow cytometry assays, respectively. In addition, the dual luciferase reporter system assay was used to determine the interaction of miR-613 and fibronectin 1 in CHON-001 cells. Results: The level of miR-613 was significantly decreased in IL-1β-treated CHON-001 cells. Overexpression of miR-613 markedly inhibited IL-1β-induced apoptosis in CHON-001 cells. In addition, upregulation of miR-613 obviously alleviated IL-1β-induced inflammatory response and cartilage matrix degradation in CHON-001 cells. Meanwhile, fibronectin 1 was identified as a direct binding target of miR-613 in CHON-001 cells. Overexpression of miR-613 alleviated IL-1β-induced injury in CHON-001 cells via downregulating the expression of fibronectin 1. Furthermore, overexpression of miR-613 alleviated cartilage degradation, and reduced OARSI scores and subchondral bone thickness in a mouse model of OA. Conclusion: Our data indicated that overexpression of miR-613 could inhibit IL-1β-induced injury in CHON-001 cells via decreasing the level fibronectin 1 in vitro, and alleviate the symptoms of OA in vivo. Therefore, miR-613 might be a potential therapeutic option for the treatment of OA.