Вы находитесь на странице: 1из 9

Autoinflammatory disorders

Ann Rheum Dis: first published as 10.1136/annrheumdis-2018-214656 on 27 February 2019. Downloaded from http://ard.bmj.com/ on 27 February 2019 by guest. Protected by copyright.
Translational science

mTOR inhibition by metformin impacts monosodium


urate crystal–induced inflammation and cell death in
gout: a prelude to a new add-on therapy?
Nadia Vazirpanah,‍ ‍ 1 Andrea Ottria,1 Maarten van der Linden,1
Catharina G K Wichers,1 Mark Schuiveling,1 Ellen van Lochem,2
Amanda Phipps-Green,3 Tony Merriman,3 Maili Zimmermann,1 Matthijs Jansen,4
Timothy R D J Radstake,1,5 Jasper C A Broen1,5

Handling editor Josef S Abstract


Smolen Key messages
Objective  Gout is the most common inflammatory
►► Additional material is
arthritis worldwide, and patients experience a heavy
What is already known about this subject?
published online only. To view burden of cardiovascular and metabolic diseases. The
►► Increased mechanistic target of rapamycin
please visit the journal online inflammation is caused by the deposition of monosodium
(http://d​ x.​doi.o​ rg/​10.​1136/​ (mTOR) signalling has recently been observed
urate (MSU) crystals in tissues, especially in the joints,
annrheumdis-​2018-​214656). in monocytes after encountering uric acid–
triggering immune cells to mount an inflammatory
containing medium.
1
Department of Rheumatology reaction. Recently, it was shown that MSU crystals can
►► Gout is mediated by monosodium urate crystals
and Clinical Immunology, induce mechanistic target of rapamycin (mTOR) signalling
consisting of precipitated uric acid in joints.
Laboratory of Translational in monocytes encountering these crystals in vitro. The
Immunology, University Medical mTOR pathway is strongly implicated in cardiovascular
Center Utrecht, Utrecht, The What does this study add?
Netherlands and metabolic disease. We hypothesised that inhibiting
►► We show that monocytes encountering MSU
2
Medical Microbiology and this pathway in gout might be a novel avenue of
crystals go into pyroptosis and provoke an
Immunology, Rijnstate Hospital, treatment in these patients, targeting both inflammation
Arnhem, The Netherlands mTOR-mediated proinflammatory environment.
3
and comorbidities.
Department of Biochemistry,
Methods   We used a translational approach starting
University of Otago, Dunedin, How might this impact on clinical practice or
New Zealand from ex vivo to in vitro and back to in vivo.
future developments?
4
Department of Immunology, Results  We show that ex vivo immune cells from
►► Both pyroptosis and inflammation are reduced
Rijnstate Hospital, Arnhem, The patients with gout exhibit higher expression of the mTOR
Netherlands with mTOR inhibitors metformin and rapamycin,
pathway, which we can mimic in vitro by stimulating
5
Department of Rheumatology which leads to a lower gout flare rate in clinical
and Clinical Immunology, healthy immune cells (B lymphocytes, monocytes, T
practice.
University Medical Center, lymphocytes) with MSU crystals. Monocytes are the most
Utrech, The Netherlands prominent mTOR expressers. By using live imaging, we
demonstrate that monocytes, on encountering MSU
Correspondence to crystals, initiate cell death and release a wide array of diseases and cancer.4–7 Recently, it has become
Dr Nadia Vazirpanah, proinflammatory cytokines. By inhibiting mTOR signalling apparent that an important driver of inflamma-
Universitair Medisch Centrum
with metformin or rapamycin, a reduction of cell death tion in gout is interleukin-1 beta (IL-1β)–mediated
Utrecht, Utrecht 3584EA, The
Netherlands; and release of inflammatory mediators was observed. NLRP3-inflammasome activation.8–10 This process
​nadiavazirpanah@​gmail.​com Consistent with this, we show that patients with gout is initiated by autophagy of MSU crystals in macro-
who are treated with the mTOR inhibitor metformin have phages, and the same effect is observed when stimu-
Received 27 October 2018 lating peripheral blood mononuclear cells (PBMCs)
a lower frequency of gout attacks.
Revised 29 January 2019
Accepted 6 February 2019 Conclusions  We propose mTOR inhibition as a novel or monocytes in vitro with MSU crystals.10–12 In
therapeutic target of interest in gout treatment. addition, interleukin 8 (IL-8) levels seem to be
constitutionally increased in the circulation of
patients with gout with concomitant cardiovascular
disease and diabetes.13
Introduction A recent study showed that stimulating mono-
Gout is the most common inflammatory arthritis cytes with MSU crystals in vitro leads to a higher
© Author(s) (or their affecting approximately 4% of the population in expression of mechanistic target of rapamycin
employer(s)) 2019. No Europe and the USA. The inflammation is caused by (mammalian target of Rapamycin) (mTOR) and
commercial re-use. See rights the deposition of monosodium urate (MSU) crystals increased IL-1β.10 The mTOR signalling pathway
and permissions. Published
by BMJ. in the joints, which predominantly occurs in hyper- partially regulates IL-8 production and IL-1β and
uricemia (0.42 mmol/L serum urate). The level of therefore might be of interest as a target in inhibiting
To cite: Vazirpanah N, comorbidity in gout patients is high; 74% have the chronic inflammation in patients with gout.14 15
Ottria A, van der
Linden M, et al.
hypertension, 71% have chronic kidney disease The mTOR pathway is well conserved in eukary-
Ann Rheum Dis Epub ahead and more than 10% suffer from either a myocardial otes, and its signalling is tightly entwined with
of print: [please include Day infarction, heart failure or a major stroke.1–3 Gout regulation of lymphocyte proliferation, immune-
Month Year]. doi:10.1136/ is associated with senescence and with increased cell activation, autophagy, and lipid and glucose
annrheumdis-2018-214656 mortality due to cardiovascular and infectious metabolism. As a consequence of its central role in
Vazirpanah N, et al. Ann Rheum Dis 2019;0:1–9. doi:10.1136/annrheumdis-2018-214656    1
Autoinflammatory disorders

Ann Rheum Dis: first published as 10.1136/annrheumdis-2018-214656 on 27 February 2019. Downloaded from http://ard.bmj.com/ on 27 February 2019 by guest. Protected by copyright.
tested using the Mann-Whitney U test (non-parametrical contin-
Table 1  Baseline characteristics of patients with gout and healthy
uous values) and Fisher’s exact test (categorical values) and
participants
(p<0.05). The data are presented as mean±SD.
Healthy participants
Gout (n=89) (n=89)
Male N (%) 72 (81.5) 77 (85.60)
Cell isolation and culture
Using lithium heparin tubes, peripheral blood of patients and
Age 62.66±13.44 47.84±17.87
healthy participants was collected. Total PBMCs were isolated
Colchicine (yes) N (%) 44 (48.9) –
using Ficoll (Ficoll-Paque Plus; GE Healthcare).
NSAID (yes) N (%) 14 (16.20) –
Monocytes (CD14+/CD16−) were isolated from total PBMCs
Allopurinol (yes) N (%) (mean 200 mg/ 76 (84.40) –
of healthy participants (online supplementary table 1) through
day)
a monocyte isolation microbead kit (lot no. 5170817557) by
Corticosteroids (yes) N (%) 40 (44.40) –
AutoMACS apparatus (Miltenyi) according to the manufacturer
Metformin (yes) N (%) 23 (25.84) –
guidelines. After 30 min of resting in RPMI-1640 (Gibco RPMI
Diabetes (type 2) (yes) N (%) 19 (21.35) –
1640 Glutamax medium enriched with 10% fetal bovine serum
Stroke (yes/no) N (%) 5 (5.6) – [FBS] and 1% penicillin/streptavidin; Sigma-Aldrich), 0.5×106
Myocardial infarction (non-fatal) (yes) 14 (15.6) – monocytes per condition were either kept unstimulated or stim-
N (%)
ulated with 0.1 mg/mL of MSU crystals (5 mg, catalogue no.
Heart failure (yes) N (%) 12 (13.30) –
tlrl-msu; InvivoGen) suspended in sterile phosphate buffered
Angina pectoris (yes) N (%) 12 (13.30) – saline buffer (lot no. RNBG2264; Sigma-Aldrich Life Science),
Creatinine level (µmol/L) 95.59 (±31.32) – MSU in combination with 10 nM rapamycin (catalogue no.
Body mass index (kg/m2) (mean±SD) 29.95 (±6.12) 25.79 (±4.18) S1039, batch no. S103911 [sirolimus]) and MSU in combina-
Smoking (yes) N (%) 12 (13.30) 1 (1.24) tion with sterile metformin (1 g, catalogue no. tlrl-metf; Invi-
Serum urate (mmol/L) 0.50 (±0.12) – voGen) suspended in RPMI medium (as described above) at a
Total no of flares per year (mean±SD) 4.41 (±5.17) – final concentration of 38.71 µM (1 g, catalogue no. tlrl-metf;
Presence of tophi (yes) N (%) 40 (45) – InvivoGen). The study design was optimised and the incuba-
Systolic blood pressure mean (mm Hg) 142.65 (±17.37) – tion times were applied according to the readout of the experi-
(SD) ment. To exclude bacterial endotoxin contamination within the
Diastolic blood pressure mean (mm Hg) 85.72 (±10.19) – MSU crystal preparation that might cause activation of the cells
(SD) during incubations, a Limulus Amebocyte Lysate (LAL) assay
NSAID, non-steroidal anti-inflammatory drug. (LAL Chromogenic Endotoxin Quantitation Kit, catalogue no.
88282; ThermoFisher Scientific) was performed following the
cellular signalling, increased mTOR signalling has been impli- manufacturers’ procedure. The quantified endotoxin level (EU/
cated in multiple diseases and is a common causative pathway in mL) was below the detection limit which excludes any endotoxin
vascular disease, inflammation, obesity, progressive renal disease contamination in MSU crystals.
and diabetes.16–18 These comorbidities are a heavy concomitant
disease burden in gout, for which contemporary urate-lowering Gene expression analysis
treatments have not been effective. The most potent clinically RNA was isolated from total PBMCs of patients with gout and
approved drug that inhibits mTOR is rapamycin, which is used healthy individuals (catalogue no./ID: 80204, Qiagen All-prep
as an immunosuppressant agent in transplant patients and as a RNA purification) according to the manufacturer guidelines.
coating for coronary stents.19 In addition, a number of reports Subsequently, cDNA was created using the Biorad iScript kit.
have been published on using rapamycin as an add-on therapy in Quantitative PCR (qPCR) was performed on a Quantstudio
rheumatoid arthritis, systemic lupus erythematosus and Sjögren’s QPCR apparatus, with Taqman Beadchip technology (Applied
disease.20–22 A less well-known, weak inhibitor of mTOR but Biosystems) under conditions as specified by the manufacturer.
more widely used is metformin. Metformin inhibits mTOR As housekeeping genes, GUSB and GAPDH were included to
signalling indirectly through AMPK activation and has been normalise expression. The following genes were included in
shown to reduce IL-8 production and might be able to reduce the analyses: protein kinase B (AkT1), DEP domain-containing
inflammasome activation.23 24 In addition, metformin has been mTOR-interacting protein (DEPTOR), glyceraldehyde 3-phos-
shown to reduce the risk for cardiovascular disease and diabetes phate dehydrogenase (GAPDH), beta-glucuronidase (GUSB),
development in clinical trials and might have a beneficiary effect interleukin 10 (IL-10), interleukin 6 (IL-6), mammalian target
on these concomitant diseases in gout.25 26 of rapamycin (mTOR), nuclear factor-kappa-B p105 subunit
In the current translational study, we were interested whether (NFκB1), phosphatase and tensin homolog (PTEN), rapa-
we could find evidence for increased mTOR signalling in mycin-insensitive companion of mammalian target of rapa-
patients with gout, to pinpoint the immune cells mostly involved mycin (RICTOR) and regulatory-associated protein of mTOR
and to test whether mTOR inhibition might be an approach to (RAPTOR). These specific genes were chosen due to their
reduce MSU crystal–induced inflammation in vitro and in vivo involvement in mTOR complex.
in patients with gout. The expression level of mTOR genes was determined using
synthesised cDNA (Biorad iScript kit) from RNA that was
Patients and methods extracted from 0.5×106 of total PBMCs, T lymphocytes (CD3+/
Demographics of patients and healthy participants CD56−), B lymphocytes (CD19+), monocytes (CD14+/CD16−)
We included 89 Dutch patients with intercritical gout and 89 and classical, intermediate and non-classical monocytes. The cells
healthy participants (table 1 and online supplementary table 1). were lysed after 6 and 24 hours and consecutively cDNA was
The significance of the association between the two classified generated. Taqman single-gene qPCR assays were performed on a
subgroups of patients with gout and healthy participants was Quantstudio apparatus (Applied Biosystems). The Housekeeping
2 Vazirpanah N, et al. Ann Rheum Dis 2019;0:1–9. doi:10.1136/annrheumdis-2018-214656
Autoinflammatory disorders

Ann Rheum Dis: first published as 10.1136/annrheumdis-2018-214656 on 27 February 2019. Downloaded from http://ard.bmj.com/ on 27 February 2019 by guest. Protected by copyright.
GUSB and GAPDH Genes (HKG) were included to normalise Monocyte markers
the gene expression. Cytokine measurements by Luminex
Cytokines were quantified using a multiplex Luminex assay.
Fluorescence-activated cell sorting (FACS) quantification and Quantification of the cytokines was done using an in-house
developed and further validated (ISO9001 certified) multi-
analysis
plex immunoassay (Laboratory of Translational Immunology,
Healthy participants’ PBMCs were assessed by FACS (FACSAria_
University Medical Center Utrecht) based on Luminex tech-
III; BD Biosciences) (online supplementary table 2). Cellular
nology (xMAP; Luminex, Austin, Texas, USA). Each sample was
markers that were included in FACS quantifications were CD3+
a supernatant of 0.5×106 monocytes per condition that were
(AF700, mouse anti-human, Clone UCHT1 [isotype IgG2a], 1:50
left either untreated, incubated with MSU crystals, MSU crys-
dilution, catalogue no. 300424; Biolegend)/CD56− (PE-CF594,
tals and rapamycin and MSU crystals and metformin during 6
mouse anti-human, Clone B159 [isotype IgG1], 1:25 dilu-
and 24 hours. The monocytes were centrifuged (300g, 8 min)
tion, catalogue no. 562328; BD) for T lymphocytes, CD19+
and the supernatant was collected and kept in −80°C until
(PECy7, mouse anti-human, Clone LT19 [isotype IgG1], 1:40
measured. The cytokine panel included interleukin 1 receptor
dilution, catalogue no. 130-091-247; Miltenyi) for B lympho-
alpha (IL-1Rα), interleukin 1 (IL-1α), interleukin 1 beta (IL-1β),
cytes, CD14+ (BV785, mouse anti-human, Clone M5E2 [isotype
interleukin 6 (IL-6), interleukin 8 (IL-8), interleukin 10 (IL-10),
IgG2a], 1:100 dilution, catalogue no. 301840; Biolegend)/
interleukin 18 (IL-18), tumour necrosis factor alpha (TNF-α),
CD16+(FCγRII) (APC, mouse anti-human, Clone ebio-CB16
interferon gamma (IFN-γ), monocyte chemotactic protein-1
[isotype IgG1], 1:20 dilution, catalogue no. 17-0168-42; eBio-
(MCP-1), macrophage inflammatory protein (MIP-1) and inter-
science) monocytes and CD3−/CD56+ NK cells. The three cell
feron gamma-induced protein 10 (IP-10). A Biorad FlexMAP3D
subsets within the main group of monocytes were differentiated
(Biorad Laboratories, Hercules, California, USA) in combina-
by gating the cells from the CD14+/CD16+ gate according to the
tion with xPONENT software V.4.2 (Luminex) was included
brightness of CD14++ (classical), CD14+CD16+ (intermediate)
to perform the acquisition. To analyse the data, five-parametric
and CD16++ (non-classical).
curve fitting using Bio-Plex Manager software V.6.1.1 (biorad)
The percentage of activation markers of classical, interme-
was assessed.
diate and non-classical monocytes’ subsets were quantified
after gating the CD14+/CD16+ monocytes, by measuring the
expressed CD163+ (APC, mouse anti-human, eBioGHI/61 Statistical analysis
[isotype IgG1], 1:20 dilution, catalogue no. 17-1639-42; eBio- Statistical analyses were performed using IBM SPSS Statistics
science) and CD86+ (BV605, mouse anti-human IT2.2 [isotype V.23 and GraphPad Prism V.6 (GraphPad Software, San Diego,
IgG2b], 1:70 dilution, catalogue no. 2127150; Sony Biotech- California, USA). Microscopic live imaging captures were
nology) percentage on the surface of the cells. Isolation and analysed using ImageJ 1.51 hour program (Java V.1.8.0_111;
stimulation (6 and 24 hours) of the cell subsets were performed National Institutes of Health, USA). Where appropriate, testing
as described above. The cells were subsequently acquired using for significant differences in categorical groups was performed
flow cytometry (FACSAria_III; BD Biosciences). using Student’s t-test (p<0.05).
Intracellular FACS was applied to assess the activation level
of intracellular mTOR pathway at the protein level after stimu- Results
lating monocytes for 15 min according to the abovementioned Genes of mTOR pathway have a higher relative expression in
protocol. Monocytes were first stained extracellularly for the patients with gout compared with healthy controls
abovementioned cell marker panel to distinguish classical, Exploiting a custom Taqman gene expression array, we investi-
non-classical and intermediate monocytes. After being fixed gated the expression of genes involved in the mTOR pathway
and permeabilised, monocytes were stained for phosphorylated (mTOR, Rictor, Raptor, Deptor, AKT1 and PTEN) in ex vivo
S6 (pS6) with human anti-pS6 antibody (anti-S6 pS240-FITC PBMCs from 89 crystal-proven patients with gout and 89
human, monoclonal recombinant IgG1, 1:5 dilution; Miltenyi healthy controls (table 1). A higher expression of the genes
biotec). The pS6 level was quantified and represented as the involved in the mTOR complex was observed in patients with
mean fluorescence intensity in monocytes. gout (p<0.0001). The expression of PTEN, an mTOR inhib-
itor, was lower in patients (p<0.0001). Taken together, these
results demonstrate an upregulation of various genes involved in
Live imaging technique
mTOR signalling in gout (figure 1A).
The microscopic live imaging technique was used to visualise
the monocytes over time. Medium rested monocytes (2×105/
condition) were administered to the medium (RPMI 1640 [10% Stimulation of PBMCs from healthy subjects with MSU
FBS, 1% penicillin–streptavidin]) containing Hoechst 33342 (20 crystals leads to increased mTOR gene expression in vitro
µM) for 30 min at 37°C. The cells were then washed and stim- To investigate if the increased expression of mTOR genes in
ulated according to the previously described stimuli/inhibitors patients with gout could be caused by contact with MSU crys-
in RPMI 1640 (without phenol red) (10% FBS, 1% penicillin– tals in these patients, we cultured PBMCs from healthy subjects
streptavidin) containing 4 nM Sytox Green (Life Technologies) with MSU crystals in vitro for 24 hours and quantified mTOR
and plated in precoated wells of a 96-well plate (clear bottom) expression. We observed an increase of mTOR expression in the
(Ibidi). Monocytes were recorded on the Pathway 855 bioim- PBMCs challenged with MSU crystals (p=0.0007) (figure 1B).
aging system (BD Biosciences) with a ×20 objective during a
period of 5 hours at 5% CO2 at 37°C. Using an Orca high-reso- MSU crystal stimulation induces mTOR gene expression in
lution CCD camera and four fields of view, every 13 min, a set immune-cell subsets in vitro
of two images (Exc/Em: 350/461 nm [Hoechst] and 504/523 nm The gene expression level of mTOR on (in vitro) MSU crystal
[Sytox Green]) was captured. AttoVision software (V.1.7/855) stimulation was measured in T and B lymphocytes and total
controlled the system. monocytes of 10 healthy participants immediately after isolation
Vazirpanah N, et al. Ann Rheum Dis 2019;0:1–9. doi:10.1136/annrheumdis-2018-214656 3
Autoinflammatory disorders

Ann Rheum Dis: first published as 10.1136/annrheumdis-2018-214656 on 27 February 2019. Downloaded from http://ard.bmj.com/ on 27 February 2019 by guest. Protected by copyright.
A) 20
B)
**** 1.5
****
* **** ****
****
to mean array signal
Relative expression

****

mTOR gene expression


10 ****

relative to HKG
1

0 0.5

-10 0

mTOR Rictor Rptor Deptor AKT1 Pten Blank Blank PBMCs


(0hrs) (24hrs) +MSU

C) D)
8 * 100 Blank
Blank
mTOR gene expression

MSU
MSU 80
* ***
6

Cell percentage
relative to HKG

*** *** 60
4
**** 40
** ***
***
2
20
* ****

0 0

T B Monocytes T B Monocytes T B NK Monocytes T B NK Monocytes


lymphocytes lymphocytes lymphocytes lymphocytes lymphocytes lymphocytes cells lymphocytes lymphocytes cells

6 hrs stimulation 24 hrs stimulation 6 hrs stimulation 24 hrs stimulation

Figure 1  (A) Gene expression of mTOR pathway–related genes in patients with gout (N=89) and healthy participants (N=89) (filled dots and empty
dots, respectively). (B) Expression of mTOR after stimulating peripheral blood mononuclear cells (PBMCs) of healthy participants with monosodium
urate (MSU) crystals in vitro (N=28). (C) Gene expression level of mTOR on MSU stimulation after 6 and 24 hours of stimulation as compared with
basal level of the gene at T=0 in immune cell subsets. The gene expression level of mTOR is increased in B lymphocytes (p=0.0006) and monocytes
(p=0.024) after 6 hours of stimulation. After 24 hours of stimulation, there was a significant induction of mTOR gene expression in T lymphocytes
(p=0.0001), B lymphocytes (p=0.0008) and monocytes (p<0.0001) as compared with the T=0 conditions. (D) After 6 hours of stimulation with MSU
crystals, there was a reduction in the proportion of monocytes within the total PBMCs cultured (p=0.0002) within the MSU-challenged condition
compared with the control. Reciprocally, there was an increase in the proportion of T lymphocytes (p=0.012). In line with this, the proportion of
monocytes in the PBMCs that had been incubated for 24 hours showed a further decrease in the proportion of monocytes (p=0.001). Accordingly, an
increment of the proportion of T lymphocytes (p=0.0004) and NK cells (p<0.0001) was observed.

of the cells (T=0) and after stimulating the cells for 6 and 24 lower in patients with gout as compared with healthy partici-
hours. After 6 hours of stimulation, MSU crystals induced mTOR pants (p=0.007). The percentage of classical (p=0.01) and inter-
gene expression in B lymphocytes (p=0.0006) and monocytes mediate (p=0.03) monocytes were lower in patients with gout.
(p=0.024) but not in T lymphocytes (p=0.085). After 24 hours, There was a similar trend in non-classical monocytes (p=0.05)
there was an induction of mTOR gene expression in T lympho- (online supplementary figure 2). The mean percentages (±SD)
cytes (p=0.0001), B lymphocytes (p=0.0008) and monocytes of the immune-cell subset of patients and healthy participants
(p<0.0001) as compared with the T=0 conditions (figure 1C). are presented in online supplementary table 2.

Encountering MSU crystals in vitro substantiates a reduction


of monocytes in PBMCs Monocytes actively engage MSU crystals and undergo cell
In order to study the effect of MSU crystal stimulation on death after contact
immune-cell subsets in more detail, PBMCs were challenged To better gauge the reaction of monocytes towards MSU crystals,
with MSU crystals for 6 and 24 hours (figure 1D). After 6 hours we performed live imaging of CD14+ monocytes encountering
of stimulation with MSU crystals, there was a significant reduc- MSU crystals. We used two dyes, namely, Sytox Green (green
tion in the proportion of monocytes within the total PBMCs colour that visualises dead cells) and Hoechst (blue colour that
cultured (p=0.0002). Reciprocally, there was an increase in the visualises live cells), to quantify the number of monocytes dying
proportion of T lymphocytes (p=0.012). Consistent with this, on encountering MSU crystals. During 7 hours of imaging, we
the proportion of monocytes in the PBMCs that had been incu- observed an active movement of monocytes towards MSU crys-
bated for 24 hours showed a further decrease in the propor- tals. A large proportion of these monocytes undergo cell death
tion of monocytes (p=0.001). Accordingly, an increase of the on encountering these crystals. The full movies are made avail-
proportion (as ratio of the total) of T lymphocytes (p=0.0004) able on the website of the journal (online supplementary movies
and NK cells (p<0.0001) was observed (figure 1D). In order S1). In figure 2A, we show representative snapshots made every
to investigate the immune-cell subsets that might be responsible hour. After 7 hours, 61% of the monocytes cultured in medium
for mTOR activation and subsequently the inflammatory reac- only were still alive, whereas only 35% of the monocytes stim-
tion in patients with gout, we evaluated the ratio of the subsets. ulated with MSU crystals survived (p<0.0001) (figure 2B). The
In PBMCs of patients with gout and healthy participants, the imaging experiment was repeated eight times with analogous
ratio of the T (p=0.22) and B (p=0.01) lymphocytes and NK outcomes. The results of the similar assessments and analysis on
(p=0.01) cells was higher in patients with gout as compared classical, non-classical and intermediate monocytes are presented
with healthy participants. The total monocytes, however, were in online supplementary figure 1.
4 Vazirpanah N, et al. Ann Rheum Dis 2019;0:1–9. doi:10.1136/annrheumdis-2018-214656
Autoinflammatory disorders

Ann Rheum Dis: first published as 10.1136/annrheumdis-2018-214656 on 27 February 2019. Downloaded from http://ard.bmj.com/ on 27 February 2019 by guest. Protected by copyright.
Figure 2  (A) Captures made from monocytes, collected from one healthy participant that were kept unstimulated, stimulated with monosodium
urate (MSU) crystals, MSU crystals with rapamycin and MSU crystals with metformin at the time 0 to 7 hours are demonstrated (blue is live cell, green
is dead cell). (B) Captures made every 13 min from the same cells were analysed and plotted against the time represented in hours. Treating the
monocytes with rapamycin (p<0.0001) and metformin (p<0.0001) on MSU stimulation induces cell survival as compared with MSU crystal stimulation
alone. (C) In vitro, 6 hours of MSU stimulation of peripheral blood mononuclear cells showed a reduction in percentage of classical (p=0.0008),
non-classical (p=0.02) and intermediate (p=0.04) cells. After 24 hours of stimulation, classical (p<0.0001) and intermediate (p=0.001) monocytes
were significantly reduced. After crystal stimulation, while non-classical are unchanged (p=0.34) (D), in monocytes from healthy participants (N=10),
metformin gave a significant reduction in phosphorylation of S6 protein after 15 min of stimulation with MSU crystals.

Proportions of CD14++ (classical), CD14+CD16+ (intermediate) we investigated whether mTOR inhibition, which promotes
and CD16++ (non-classical) monocytes within the total PBMCs autophagy and decreases inflammatory responses and response
are all decreased after encountering MSU crystals to apoptotic cells, would have a dampening effect on monocyte
After 6 hours of stimulating PBMCs with MSU crystals, we quan- death and MSU crystal–induced inflammation. First, we eval-
tified the number of monocytes by flow cytometry and further uated whether the observed increased mTOR gene expression
differentiated the monocytes from the CD14+/CD16+ gate was reflected in the protein level. We measured the phosphor-
according to the brightness of CD14++ (classical), CD14+CD16+ ylation of S6 ribosomal protein (S6) at serine 240/244, which
(intermediate) and CD16++ (non-classical) monocytes. is downstream from mTOR activation and therefore commonly
After culturing PBMCs for 6 hours with MSU crystals, we used as readout of mTOR activation. After resting, monocytes
observed a significant reduction in proportion of classical were stimulated for 15 min with MSU crystals and MSU crys-
monocytes (p=0.0008), non-classical monocytes (p=0.02) tals with metformin. As presented in figure 2D, metformin
and intermediate monocytes (p=0.04) within the total PBMC caused a decrease of the pS6 mean fluorescence intensity in total
number. The PBMCs that were incubated for 24 hours showed (p=0.013), classical (p=0.015) and non-classical (p=0.040)
a significant reduction in classical (p<0.0001) and intermediate monocytes within 15 min.
(p=0.001) monocytes, while the reduction of non-classical To investigate temporal stability of the inhibitory effect of
monocytes (p=0.34) was not significant (figure 2C). The mean metformin in monocytes, we performed titration assays where
percentages (±SD) of the immune-cell subsets of patients and we quantified the expression level of mTOR gene in monocytes
healthy participants are presented in online supplementary table (N=5) after 3, 6, 9 and 12 hours of incubation in the presence
2. of MSU crystals. After 3 hours of metformin stimulation in
MSU crystal–challenged monocytes, we observed a significant
MTOR inhibition by rapamycin or metformin reduces MSU decrease in mTOR gene expression as compared with MSU
crystal–induced monocyte death crystal–challenged monocytes (p=0.0007). This inhibitory effect
Since we observed an increased rate of cell death and an increased of metformin was stable after 6 hours (p=0.008) (figure 3D).
expression of mTOR in monocytes encountering MSU crystals, The inhibitory effect of metformin in MSU crystal–challenged
Vazirpanah N, et al. Ann Rheum Dis 2019;0:1–9. doi:10.1136/annrheumdis-2018-214656 5
Autoinflammatory disorders

Ann Rheum Dis: first published as 10.1136/annrheumdis-2018-214656 on 27 February 2019. Downloaded from http://ard.bmj.com/ on 27 February 2019 by guest. Protected by copyright.
Figure 3  (A, B) Differential cytokine expression in monocytes stimulated with monosodium urate (MSU) crystals with and without mTOR inhibition
by metformin or rapamycin. The cytokines that were significantly differently secreted from monocytes treated with MSU crystals and rapamycin as
compared with MSU only after respectively 6 and 24 hours. (C) Heatmap representing the changes in monocytes’ cytokine expression on stimulation
with MSU crystals only, MSU crystals with rapamycin, and MSU crystals with metformin. (D) Inhibitory effect over time of metformin on mTOR gene
expression (normalised for housekeeping gene (HKG) expression) in MSU-challenged monocytes. In monocytes of healthy participants (N=5), blank
condition contains unstimulated monocytes that have the same incubation time as the stimulated conditions. Only the significant differences are
indicated. (E) Patients with gout treated with a combination of allopurinol and metformin have significantly less recurrent flares as compared with
patients treated only with allopurinol (p=0.010).

monocytes was until 9 hours of stimulation (p=0.19) and medium only, with MSU crystals, with MSU crystals and
reached its minimum after 12 hours of stimulation (p=0.27). In metformin, or with MSU crystals and rapamycin. In addition,
the same monocytes and the same setting, NFkB gene expression we cultured monocytes with rapamycin and metformin without
was quantified. After 3 (p=0.027) and 9 (p=0.026) hours of MSU crystals. These conditions were all evaluated alongside live
metformin stimulation, there was a significant inhibition of NFkB imaging, at the same time, in which monocytes from a healthy
(online supplementary figure 3A). Interestingly, metformin had participant were cultured in every condition mentioned and
an inhibitory effect on IL-1β in monocytes after 3 (p=0.023), analysed. This experiment was performed six times. When we
9 (p=0.024) and 12 (p=0.041) hours. Similarly, after 6 hours quantified the proportion of cell death by ImageJ, comparing
(p=0.063), there was a trend of inhibitory effect of metformin ‘live cell’ dye within each snapshot (time between each snapshot
on monocytes (online supplementary figure 3B). T=13 min), we observed that monocytes co-cultured with MSU
Since we were now able to inhibit mTOR in vitro with crystals and rapamycin (cells alive 56%) or metformin (cells
metformin and rapamycin, we co-cultured monocytes with alive 59%) had a significantly lower death rate as compared
6 Vazirpanah N, et al. Ann Rheum Dis 2019;0:1–9. doi:10.1136/annrheumdis-2018-214656
Autoinflammatory disorders

Ann Rheum Dis: first published as 10.1136/annrheumdis-2018-214656 on 27 February 2019. Downloaded from http://ard.bmj.com/ on 27 February 2019 by guest. Protected by copyright.
with monocytes stimulated with MSU crystals only after 7 hours as compared with patients who did not receive any colchicine
(cells alive 35%) (both p<0.0001). The rate of cell death in the treatment (p=0.0412). An additional analysis demonstrated no
monocytes treated with mTOR inhibitors and MSU crystals significant difference in mTOR expression level in patients who
was similar to that of monocytes cultured without MSU crys- were treated/not treated with non-steroidal anti-inflammatory
tals (61% alive). In figure 2A, we show representative snapshots drugs (p=0.8139). Assessing the association between mTOR
made every hour of monocyte cell culture at T=0 to 7 hours, gene expression level and uric acid level (by Pearson correlation)
and figure 2B shows the number of alive cells per condition over showed a significant positive correlation (p=0.014).
time. We did not observe any differences when cells were stim-
ulated with metformin and rapamycin only mTOR inhibition
by metformin or rapamycin reduces proinflammatory cytokine Discussion
release by monocytes on encountering MSU crystals in vitro The main conclusion of this study is that PBMCs from patients
To assess whether mTOR inhibition leads to less cytokine with gout have a signature of increased mTOR signalling as
production on monocyte exposure to MSU crystals in vitro, we compared with healthy participants. By performing in vitro
quantified the release of IL-1Rα, IL-1α, IL-1β, IL-6, IL-8, IL-10, experiments, we showed that MSU crystals provoke upregu-
IL-18, TNF-α, IFN-γ, MCP-1, MIP-1 and IP-10 by Luminex lation of mTOR pathways gene expression, IL-1β, IL-6, IL-8,
in monocytes from 11 donors. Monocytes were cultured with IL-18 release and cell death in monocytes. We were able to
MSU crystals. We compared cytokine levels between MSU-cul- inhibit these phenomena by adding mTOR inhibitors rapamycin
tured monocytes with MSU crystals alone or co-cultured with and metformin. When we analysed the effect of metformin
metformin or rapamycin, which are both mTOR inhibitors. on gout flares in a retrospective analysis of patients with gout
The monocytes co-cultured with MSU crystals and rapamycin with diabetes stratified according to metformin treatment, we
showed a reduction in levels of IL-1β (p=0.02), IL-6 (p=0.02), observed a significantly lower gout attack frequency as compared
IL-8 (p=0.0017), IL-10 (p=0.024), IL-18 (p=0.0009), IFN-γ with patients not treated with metformin.
(p=0.001), MCP-1 (p=0.01), MIP-1 (p=0.0026) and IP-10 An interesting finding of our study is the active engagement of
(p=0.029). In the presence of metformin, a reduction in monocytes towards MSU crystals, which induces a form of acute
levels of IL-1β (p=0.02), IL-6 (p=0.31), IL-8 (p=0.01), IL-10 cell death. It is well known that there is an overlap in apoptosis
(p=0.0051), IL-18 (p=0.045), TNF-α (p=0.042), MCP-1 and necrosis in vivo when immune cells encounter strong danger
(p=0.006), MIP-1 (p=0.04) and IP-10 (p=0.046) was observed signals.27 It is established that necrosis leads to NACHT, LRR
when compared with the monocytes cultured with MSU crystals and PYD domains–containing protein 3 (NLRP3) activation and
alone. The reduced cytokine level after 24 hours of incubation increased IL-1β production, an important feature of gout and also
with MSU crystals and rapamycin was IL-1α (p=0.027), IL-8 observed in our study. Interestingly, mTOR activation enhances
(p=0.02), IL-10 (p=0.005), MCP-1 (p=0.0032) and MIP-1 the process of necrosis.28 To apply this to gout and our study, it is
(p=0.005). Reduced cytokine levels in monocytes incubated conceivable that necrosis of monocytes when encountering MSU
with metformin and crystal stimulation after 24 hours was IL-1α crystals leads to activation of the inflammasome pathway and
(p=0.032), IL-1β (p=0.04), IL-6 (p=0.034), IL-10 (p=0.015), release of proinflammatory cytokines, as we demonstrate. The
TNF-α (p=0.0024), MCP-1 (p=0.008) and MIP-1 (p=0.04). high expression of mTOR within monocytes further facilitates
The quantified values are represented on a logarithmic scale the pro-necrotic state within patients with gout. When mTOR
(figure 3A). Colour heatmaps represent the effect of stimuli and is inhibited, there is a lower tendency towards cell death and
inhibitors on the cells (figure 3B). There was no difference in consequently less inflammasome activity and inflammation, as
cytokine secretion by the cells when stimulated with metformin we display in our study as well. Hence, the very start of the gout
and rapamycin only. attack might lie in the encounter of monocytes with MSU crys-
tals and seems to be modulated by mTOR.
Our findings are in line with a recent study also showing that
Metformin treatment associates with low flare frequency in stimulation of monocytes with MSU crystals enhances mTOR
patients with gout activation.10 Very little research has been performed within the
To scrutinise whether mTOR inhibition through metformin in field of mTOR inhibition and gout; however, most available
patients with gout leads to a lower frequency of gout flares, data concern the effects of metformin treatment in gout disease
we performed a retrospective cohort analyses in 23 Caucasian activity. A large retrospective case–control study (N=7536)
patients with gout and metformin use in comparison with 19 in patients with diabetes showed that the use of metformin
patients with gout and diabetes without using metformin. As decreases the ORs for developing gout compared with patients
diabetic comedication, insulin use was allowed. Patients were not using metformin.29 The authors, however, mainly focused
selected from the Dutch cohort (table 1) and Caucasian patients on the finding that poorly controlled diabetes as defined by
with gout from New Zealand (online supplementary table 3). HbA1c levels is correlated with a decreased incidence of gout.
Our analysis demonstrates that patients with gout who were Two small-scale studies conducted in Russia (N=30 and N=26)
treated with a combination of metformin and allopurinol in patients without diabetes with gout showed that metformin
have a significantly lower attack frequency as compared with reduces the frequency of gout attacks, lowers uric acid and led
patients who were treated with allopurinol alone (p=0.010) to normo-uricemia in 11 patients.30 31 Of interest is the observa-
(figure 3E). In our small retrospective cohort, we recorded a tion that metformin is able to interfere directly with the purine
mean flare frequency of 2.04 (95% CI 1.29 to 2.38) flares per pathway, which might be the mode of action for the lowering
year in the allopurinol with metformin group versus the 4.00 of uric acid levels; the latter, however, has not yet been clearly
(95% CI 2.57 to 5.43) flares in the allopurinol-only group. proven.32 33
Finally, our results demonstrate that patients with gout have a The evidence for an anti-inflammatory effect of metformin has
significantly higher mTOR gene expression level. We observed been mounting over the past years. It is known that metformin
that patients with gout who received colchicine treatment do activates AMPK (5′ adenosine monophosphate-activated
have a significantly lower level of mTOR gene expression level protein kinase) to inhibit NF-κB via the PI3K (phosphoinositide
Vazirpanah N, et al. Ann Rheum Dis 2019;0:1–9. doi:10.1136/annrheumdis-2018-214656 7
Autoinflammatory disorders

Ann Rheum Dis: first published as 10.1136/annrheumdis-2018-214656 on 27 February 2019. Downloaded from http://ard.bmj.com/ on 27 February 2019 by guest. Protected by copyright.
3-kinase)–Akt1 pathway and reduces the production of NO mTOR signalling; a study by Harris et al showed that mTOR
(nitric oxide), prostaglandin E2 and proinflammatory cytokines inhibition with rapamycin in macrophages leads to degradation
(IL-1β, IL8, IL-6 and TNF-α) in monocytes and macrophages.34 35 of pro-IL-1β, subsequently reducing NLRP3 inflammasome acti-
One study that included over 4000 patients with pre-diabetes vation.43 A similar observation was made in an in vitro model
showed a significant reduction of CRP levels when treated with for sepsis.44 Another important study showed that MSU priming
metformin as compared with placebo after 12 months.36 More- in monocytes leads to mTOR activation in concert with IL-1β
over, in monocyte-derived macrophages, metformin seemed expression.12 We believe that therapeutic targeting of the inflam-
to interfere directly with the inflammasome, orchestrating an masome directly and indirectly by inhibiting mTOR in patients
inhibition of IL-1β maturation in patients with type 2 diabetes with gout might have a symbiotic effect in reducing the inflam-
treated with metformin.37 Patients with gout are typified by matory response mediated through IL-1β.
inflammasome induction and high circulating IL-8 levels and Somewhat counterintuitive, we observed an increase in IL-10
metformin is likely to be a suitable treatment for these patients levels when the monocytes were stimulated with MSU crystals.
since it is an effective inflammasome and IL-8 suppressor. Although the role of IL-10 in the biology of monocytes has been
Metformin is the first-choice drug for treating type 2 diabetes; under debate, this might be attributed to the high apoptosis rate
it is effective in reducing the hyperglycaemic state and decreases in these cells.45
insulin resistance. Less obvious but well proven is the fact that As described above, metformin has many potential benefi-
metformin reduces the cardiovascular risk in patients with cial effects on the disease course in gout. It has properties that
diabetes. The UK Prospective Diabetes Study (N=5500) demon- inhibit inflammation through the mTOR and NLRP3 path-
strated a substantial beneficial effect of metformin therapy on ways, it decreases cardiovascular risk and it potentially might
cardiovascular disease outcomes, with a 36% relative risk reduc- be able to decrease gout flares (online supplementary figure
tion in all-cause mortality and a 39% relative risk reduction 4). The currently available drugs are well able to target one of
in myocardial infarction.38 The exact mechanism of action by these domains, (eg, allopurinol/colchicine in uric acid lowering,
which metformin protects the vasculature is not known, but it canakinumab for inflammasome targeting); however, none of
is thought to be a combination of improving lipid metabolism, them are able to target all three domains. Up until now, it is not
AMPK induction and reduction of reactive oxygen species. Of clear if any of the currently used drugs reduce cardiovascular and
interest for the gout population, which is at high risk to have or metabolic risk.
develop diabetes, metformin reduced the incidence of diabetes A large body of evidence shows that metformin reduces
in high-risk groups.39 cardiovascular risk and increases insulin sensitivity, reducing the
Our study has strengths and weaknesses; the strength of our burden of diabetes. Hence, taking also into account the favour-
study lies in the fact that we started from ex vivo patient material able drug profile and our observations, we advocate to investi-
and observations, which we translated in an in vitro model and gate metformin as an add-on therapy for patients with gout in a
validated retrospectively in an in vivo observation. This chain of prospective study to clarify whether metformin is able to reduce
experimental settings makes our findings more robust to transla- the burden of gout flares and comorbidities.
tion to the clinical setting. Our experiments were performed in
Acknowledgements  JCAB is supported by a VENI Award from the Netherlands
parallel on the same apparatus and analysed by the same algo- Organization for Scientific Research (N.W.O. project no. 91614041). Multiplex
rithms to avoid mistakes or bias by measurement or observer. Luminex immunoassays were in-house developed by W De Jager, H te Velthuis, B
Another strength is that the observations were made on both J Prakken, W Kuis and G T Rijkers, validated and performed at the Multiplex core
gene expression and protein level with various techniques. All facility of the Laboratory for Translational Immunology (LTI) of the University Medical
patients included in the ex vivo study had crystal-proven gout, Center Utrecht.
which is the gold standard of diagnosis. Moreover, the concen- Contributors  All authors approved the final version after being involved
trations of metformin and rapamycin used in our experimental in drafting and revising the article for important intellectual content. As the
corresponding author, NV had full access to the data and takes responsibility for
settings were derived from real-life plasma concentrations of the accuracy of the performed analysis and the integrity of the data. NV, TRDJR and
these drugs in patients being treated with these drugs in clin- JCAB were involved in design of the study. Execution, analysis and writing of the
ical practice. This makes the results more relevant to clinical manuscript was performed by NV. AO and MvdL respectively contributed in FACS
use. A weakness of our study is the small cohort in which we and Live Imaging of this study. CGKW was involved in performing gene arrays. MS
performed a retrospective analysis on the effects of metformin and MZ thought along on rapamycin and metformin stimulations. EvL and MJ were
involved in inclusion of Dutch patients with gout and TM participated by including
on the frequency of gout attacks. Although highly informative patients with gout from New Zealand.
in the light of our study, these results need to be confirmed in
Funding  This study was funded by Nederlandse Organisatie voor Wetenschappelijk
a larger prospective study to make way for use of metformin in Onderzoek (91614041).
gout clinical practice. In our retrospective study, we did not have
Competing interests  None declared.
longitudinal data on glucose status, kidney function, treatment
adherence and dose escalation; therefore, these results should be Patient consent for publication  Obtained.
regarded with caution for direct extrapolation to clinical prac- Ethics approval  This study was performed according to the guidelines of
tice without further prospective and preferably randomised clin- the Declaration of Helsinki and study meets the approval of ethical and review
committees of the the Rijnstate hospital (Nijmegen, the Netherlands), University
ical trials.
Medical Center of Utrecht in the Netherlands, VieCuri Hospital of Venlo in the
Our data add to the understanding of the inflammatory Netherlands and University of Otago in Dunedin, New Zealand.
reaction that occurs when monocytes encounter MSU crystals.
Provenance and peer review  Not commissioned; externally peer reviewed.
Previous studies have strongly implicated inflammasome acti-
vation in the pathogenesis of gout as well; the role of inflam- Data sharing statement  Not applicable.
masome activation in gout is well described and witnessed by
the effect that inflammasome-modulating drugs such as canaki- References
1 Vazirpanah N, Kienhorst LBE, Van Lochem E, et al. Patients with gout have
numab and anakinra have in gout.40–42 It is therefore very inter- short telomeres compared with healthy participants: association of telomere
esting to discuss how these pathways might entwine. Previous length with flare frequency and cardiovascular disease in gout. Ann Rheum Dis
studies have shown that IL-1β secretion is partially regulated by 2017;76:1313–9.

8 Vazirpanah N, et al. Ann Rheum Dis 2019;0:1–9. doi:10.1136/annrheumdis-2018-214656


Autoinflammatory disorders

Ann Rheum Dis: first published as 10.1136/annrheumdis-2018-214656 on 27 February 2019. Downloaded from http://ard.bmj.com/ on 27 February 2019 by guest. Protected by copyright.
2 Dalbeth N, Haskard DO. Mechanisms of inflammation in gout. Rheumatology activation in the adipose tissue of diabetic mice. Mol Cell Endocrinol
2005;44:1090–6. 2016;434:36–47.
3 Bardin T, Richette P. Impact of comorbidities on gout and hyperuricaemia: an update 24 Rotondi M, Coperchini F, Pignatti P, et al. Metformin reverts the secretion of CXCL8
on prevalence and treatment options. BMC Med 2017;15. induced by TNF-α in primary cultures of human thyroid cells: an additional indirect
4 Kuo C-F, See L-C, Luo S-F, et al. Gout: an independent risk factor for all-cause and anti-tumor effect of the drug. J Clin Endocrinol Metab 2015;100:E427–E432.
cardiovascular mortality. Rheumatology 2010;49:141–6. 25 Maruthur NM, Tseng E, Hutfless S, et al. Diabetes medications as monotherapy or
5 Kuo C-F, Yu K-H, See L-C, et al. Elevated risk of mortality among gout patients: metformin-based combination therapy for type 2 diabetes: a systematic review and
a comparison with the national population in Taiwan. Joint Bone Spine meta-analysis. Ann Intern Med 2016;164:740–51.
2011;78:577–80. 26 Mazzotti A, Caletti MT, Marchignoli F, et al. Which treatment for type 2 diabetes
6 Krishnan Eet al. Long-term cardiovascular mortality among middle-aged men with associated with non-alcoholic fatty liver disease? Dig Liver Dis 2017;49:235–40.
gout. Arch Intern Med 2008;168:1104–10. 27 Nikoletopoulou V, Markaki M, Palikaras K, et al. Crosstalk between apoptosis, necrosis
7 Perez-Ruiz F, Martínez-Indart L, Carmona L, et al. Tophaceous gout and high level of and autophagy. Biochimica et Biophysica Acta (BBA) - Molecular Cell Research
hyperuricaemia are both associated with increased risk of mortality in patients with 2013;1833:3448–59.
gout. Ann Rheum Dis 2014;73:177–82. 28 Wu Y-T, Tan H-L, Huang Q, et al. Activation of the PI3K-AKT-mTOR signaling
8 Crişan TO, Cleophas MCP, Novakovic B, Cris TO, et al. Uric acid priming in human pathway promotes necrotic cell death via suppression of autophagy. Autophagy
monocytes is driven by the AKT-PRAS40 autophagy pathway. Proc Natl Acad Sci U S A 2009;5:824–34.
2017;114:5485–90. 29 Bruderer SG, Bodmer M, Jick SS, et al. Poorly controlled type 2 diabetes mellitus is
9 Martinon F, Pétrilli V, Mayor A, et al. Gout-associated uric acid crystals activate the associated with a decreased risk of incident gout: a population-based case–control
NALP3 inflammasome. Nature 2006;440:237–41. study. Ann Rheum Dis 2015;74:1651–8.
10 Chung Y-H, Kim D-H, Lee W-W. Monosodium urate crystal-induced pro-interleukin-1β 30 Barskova VG, Eliseev MS, Nasonov EL, et al. [Use of metformin (siofor) in patients with
production is post-transcriptionally regulated via the p38 signaling pathway in human gout and insulin resistance (pilot 6-month results)]. Ter Arkh 2005;77:44–9.
monocytes. Sci Rep 2016;6:1–15. 31 Barskova VG, Eliseev MS, Kudaeva FM, et al. Effect of metformin on the clinical course
11 Mylona EE, Mouktaroudi M, Crisan TO, et al. Enhanced interleukin-1β production of of gout and insulin resistance]. Klin Med 2009;87:41–6.
PBMCs from patients with gout after stimulation with Toll-like receptor-2 ligands and 32 Scotland S, Saland E, Skuli N, et al. Mitochondrial energetic and Akt status mediate
urate crystals. Arthritis Res Ther 2012;14. metabolic effects and apoptosis of metformin in human leukemic cells. Leukemia
12 Crişan TO, Cleophas MCP, Novakovic B, et al. Uric acid priming in human monocytes 2013;27:2129–38.
is driven by the AKT-PRAS40 autophagy pathway. Proc Natl Acad Sci U S A 33 Schuiveling M, Vazirpanah N, Radstake TRDJ, et al. Metformin, a new era for an old
2017;114:5485–90. drug in the treatment of immune mediated disease? CDT 2018;19:945–59.
13 Kienhorst LBE, van Lochem E, Kievit W, et al. Gout is a chronic inflammatory 34 Vasamsetti SB, Karnewar S, Kanugula AK, et al. Metformin inhibits monocyte-to-
disease in which high levels of interleukin-8 (CXCL8), myeloid-related protein 8/ macrophage differentiation via AMPK-mediated inhibition of STAT3 activation:
myeloid-related protein 14 complex, and an altered proteome are associated potential role in atherosclerosis. Diabetes 2015;64:2028–41.
with diabetes mellitus and cardiovascular disease. Arthritis Rheumatol 35 Hattori Y, Suzuki K, Hattori S, et al. Metformin inhibits cytokine-induced nuclear factor
2015;67:3303–13. κB activation via AMP-activated protein kinase activation in vascular endothelial cells.
14 HYH L, Chang KT, Hung CC, et al. Effects of the mTOR inhibitor rapamycin on Hypertension 2006;47:1183–8.
monocyte-secreted chemokines. BMC Immunol 2014;15. 36 Haffner S, Temprosa M, Crandall J, et al. Intensive lifestyle intervention or metformin
15 Castranova V, Asgharian B, Sayre P, et al. Inhalation exposure to carbon nanotubes on. 54, 2005.
(CnT) and carbon nanofibers (CNF): methodology and dosimetry. J Toxicol Environ 37 Lee H-M, Kim J-J, Kim HJ, et al. Upregulated NLRP3 inflammasome activation in
Health B Crit Rev 2016;18:121–212. patients with type 2 diabetes. Diabetes 2013;62:194–204.
16 De Vita V, Melnik BC. Activation of mechanistic target of rapamycin complex 1: the 38 U.K. Prospective Diabetes Study Group. U.K. Prospective Diabetes Study 16:
common link between rheumatoid arthritis and diabetes mellitus. Rheumatology overview of 6 years’ therapy of type II diabetes: a progressive disease. Diabetes
2018;149:1–3. 1995;44:1249–58.
17 Saxton RA, Sabatini DM. mTOR signaling in growth, metabolism, and disease. Cell 39 Knowler WC, Barrett-Connor E, Fowler SE, et al. Reduction in the incidence of type 2
2017;168:960–76. diabetes with lifestyle intervention or metformin. N Engl J Med 2002;346:393–403.
18 Johnson SC, Rabinovitch PS, Kaeberlein M. mTOR is a key modulator of ageing and 40 Joosten LAB, Netea MG, Mylona E, et al. Engagement of fatty acids with Toll-like
age-related disease. Nature 2013;493:338–45. receptor 2 drives interleukin-1β production via the ASC/caspase 1 pathway in
19 Zivelonghi C, van Kuijk JP, Nijenhuis V, et al. First report of the use of long-tapered monosodium urate monohydrate crystal-induced gouty arthritis. Arthritis Rheum
sirolimus-eluting coronary stent for the treatment of chronic total occlusions with the 2010;62:3237–48.
hybrid algorithm. Catheter Cardiovasc Interv 2018;92:E299–E307. 41 Cleophas MCP, Crişan TO, Lemmers H, et al. Suppression of monosodium urate
20 Shao P, Ma L, Ren Y, et al. Modulation of the immune response in rheumatoid arthritis crystal-induced cytokine production by butyrate is mediated by the inhibition of class I
with strategically released rapamycin. Mol Med Rep 2017;16:5257–62. histone deacetylases. Ann Rheum Dis 2016;75:593–600.
21 Koganesawa M, Yamamoto S, Kaneko R, et al. Utility of the attached sample blood for 42 So AK, Martinon F. Inflammation in gout: mechanisms and therapeutic targets. Nat
quality control of long-term cryopreserved umbilical cord blood for hematopoietic cell Rev Rheumatol 2017;13:639–47.
transplantation. J Showa Med Assoc 2016;76:199–206. 43 Harris J, Hartman M, Roche C, et al. Autophagy controls IL-1beta secretion by
22 Kato H, Perl A. Blockade of Treg cell differentiation and function by the targeting pro-IL-1beta for degradation. J Biol Chem 2011;286:9587–97.
interleukin-21–mechanistic target of rapamycin axis via suppression of autophagy in 44 Giegerich AK, Kuchler L, Sha LK, et al. Autophagy-dependent PELI3 degradation
patients with systemic lupus erythematosus. Arthritis Rheumatol 2018;70:427–38. inhibits proinflammatory IL1B expression. Autophagy 2014;10:1937–52.
23 Li A, Zhang S, Li J, et al. Metformin and resveratrol inhibit Drp1-mediated 45 Bzowska M, Guzik K, Barczyk K, et al. Increased IL-10 production during spontaneous
mitochondrial fission and prevent ER stress-associated NLRP3 inflammasome apoptosis of monocytes. Eur J Immunol 2002;32:2011–20.

Vazirpanah N, et al. Ann Rheum Dis 2019;0:1–9. doi:10.1136/annrheumdis-2018-214656 9

Вам также может понравиться