ArticlePDF Available

Undenatured type II collagen protects against collagen-induced arthritis by restoring gut-joint homeostasis and immunity

Authors:

Abstract and Figures

Oral administration of harmless antigens can induce suppression of reactive immune responses, a process that capitalises on the ability of the gastrointestinal tract to tolerate exposure to food and commensal microbiome without triggering inflammatory responses. Repeating exposure to type II collagen induces oral tolerance and inhibits induction of arthritis, a chronic inflammatory joint condition. Although some mechanisms underlying oral tolerance are described, how dysregulation of gut immune networks impacts on inflammation of distant tissues like the joints is unclear. We used undenatured type II collagen in a prophylactic regime -7.33 mg/kg three times/week- to describe the mechanisms associated with protective oral immune-therapy (OIT) in gut and joint during experimental Collagen-Induced Arthritis (CIA). OIT reduced disease incidence to 50%, with reduced expression of IL-17 and IL-22 in the joints of asymptomatic mice. Moreover, whilst the gut tissue of arthritic mice shows substantial damage and activation of tissue-specific immune networks, oral administration of undenatured type II collagen protects against gut pathology in all mice, symptomatic and asymptomatic, rewiring IL-17/IL-22 networks. Furthermore, gut fucosylation and microbiome composition were also modulated. These results corroborate the relevance of the gut-joint axis in arthritis, showing novel regulatory mechanisms linked to therapeutic OIT in joint disease.
Disease scores and histological analysis in response to oral immunotherapy (OIT) in CIA mice a Disease scores (left panel) and incidence (right panel) were evaluated at the indicated time points for naïve (grey), CIA (red) and CIA-OIT (blue). OIT consisted of administration of undenatured type II collagen by oral gavage 3 times a week, starting 2 weeks before induction of arthritis. Cumulative disease scores were given for each limb, 0 = no disease and 4 = highest score. Each dot represents the mean of individual mice and error bars show SEM (n = 15 for CIA, n = 16 for OIT groups, n = 9 for naïve mice, data from 2 independent experiments). Statistical significance was determined by one-way ANOVA; *p < 0.05, ***p < 0.001 between CIA and naïve, #p < 0.05, ##p < 0.01 comparing CIA and OIT groups. Disease incidence was calculated by dividing the number of cases by the total number of mice in the group at the indicated times. b Paw width (left panel) and weight change (percentage of initial weight, right panel) were evaluated at the indicated time points. Each dot represents the mean of individual mice, error bars show SEM (n = 15 for CIA, n = 7 for OIT asymptomatic, n = 9 for OIT symptomatic, n = 9 for naïve mice, data from 2 independent experiments). Statistical significance was determined by one-way ANOVA; *p < 0.05, ***p < 0.01,***p < 0.001, ns = non-significant. c Hind paws were collected at the end of the experiment (day 34) for each mouse, sectioned, and stained with haematoxylin and eosin for histological analysis. Images show one representative mouse (Disease score = median for each group). Superimposed dotted lines show bone limits; scale bar = 500 μm. d Cell infiltration, pannus formation, cartilage and bone damage were quantified for individual mice in hematoxilin/eosin-stained sections. Dot plots show histological scores (from 0 to 4, 0 = healthy tissue, 4 = highest possible score). Individual dots represent individual paws (n = 32 for CIA, n = 8 for OIT asymptomatic, and n = 24 for OIT symptomatic, data collected from 2 independent experiments). Bars show the mean value for each group. Statistical significance was determined by the Kruskal–Wallis test; **p < 0.01, ***p < 0.001. The percentage of mice with no sign of pathology (green) for the indicated parameter is shown in vertical column graphs.
… 
OIT protection is associated with reduced upregulation of inflammatory IL-17 and IL-22 in the joint and draining lymph nodes Naïve, CIA and CIA-OIT mice were culled at day 33 when tissue was collected for further analysis. a, b Correlation between numbers of IL-17 and IL-22 positive lymph node cells and clinical scores in T cells (a) and B cells (b) isolated from draining lymph nodes (DLNs). Every dot represents values for individual mice from all groups. Data are presented as mean ± SEM, r: Pearson’s coefficient. c Expression of IL-17 and IL-22 (red) was evaluated in the joint tissue by immunofluorescence in naïve, CIA and OIT asymptomatic mice. DAPI (Blue) was used to stain nuclei as counterstaining. Superimposed dotted lines show bone tissue and areas of cell infiltration are indicated by white arrows. Scale bars: 500 μm. Graphs show the quantification of the mean intensity of individual mice. d IL-17 concentration was evaluated by ELISA in the supernatants of draining lymph node cells upon PMA (50 ng/ml)/Ionomycin (500 ng/ml) stimulation for 12 h. Data show naïve, CIA and OIT (symptomatic and asymptomatic). Each dot represents cells from one individual mouse. Error bars show mean ± SEM; *p < 0.05, **p < 0.01 analysed by one-way ANOVA from one experimental model. e Relative frequency and total cell number of IL-17+ and IL-22+ DLN cells were evaluated by flow cytometry. Data show mean ± SEM; each dot represents individual mice from two independent experimental models; *p < 0.05, analysed by one-way ANOVA. f Cell frequency and total cell numbers of IL-17+ and IL-22+ CD4 T cells, CD8 T cells and B cells in DLNs, represented at the corners of radar charts: Naïve (grey), CIA (red) and OIT asymptomatic (orange) and OIT symptomatic (blue); data were normalised to maximum expression in each group. Significance on the raw data among groups was evaluated by ordinary one-way ANOVA, where *p < 0.05, **p < 0.01 [CIA vs naïve]; $p < 0.05 [naïve vs OIT symptomatic]; ▲p < 0.05 [naïve vs OIT asymptomatic]; ⁺p < 0.05 [CIA vs OIT asymptomatic]; §p < 0.05 [CIA vs OIT asymptomatic]; #p < 0.05 [OIT asymptomatic vs OIT symptomatic].
… 
OIT protects against gastrointestinal damage associated with inflammatory arthritis in both symptomatic and asymptomatic cases Naïve, CIA and OIT asymptomatic mice and OIT symptomatic mice were culled at day 33 when gut tissue and total mesenteric lymph nodes (MLNs) were collected. a Isolated gastrointestinal tract from a naïve mouse showing the four anatomical areas used for further study. b Duodenum, jejunum, ileum, and colon were fixed, and tissue sections were subjected to hematoxylin and eosin staining. The length of villi and crypts was measured using Image J software, and the ratio of villi/crypt was quantified. Each dot represents the villi/crypt ratio for individual mice, and data were collated from two independent experiments. Statistical significance was evaluated by ordinary one-way ANOVA, where *p < 0.05, **p < 0.01 and ***p < 0.001. c Colon tissue sections were stained with PAS to detect changes in the mucus layer and associated pathology. Scale bars = 500 μm. The depicted mice are representative of individual mice whose disease scores fall within the median value for each group. d MLNs were collected to generate single-cell suspensions, and a total number of cells was counted. Cell number and percentage of B cells, total T cells, CD4 and CD8 T cells were evaluated by Flow Cytometry. Each dot represents one individual mouse and error bars show mean ± SEM. Mice were pooled from 2 independent experiments. Statistical significance was determined using Ordinary one-way ANOVA. Significance is indicated by asterisks, *p < 0.05, **p < 0.01.
… 
This content is subject to copyright. Terms and conditions apply.
communications biology Article
https://doi.org/10.1038/s42003-024-06476-z
Undenatured type II collagen protects
against collagen-induced arthritis by
restoring gut-joint homeostasis and
immunity
Check for updates
Piaopiao Pan1,YilinWang
2, Mukanthu H. Nyirenda3,5, Zainulabedin Saiyed4, Elnaz Karimian Azari4,
Amy Sunderman4, Simon Milling3,MargaretM.Harnett
3& Miguel Pineda 1
Oral administration of harmless antigens can induce suppression of reactive immune responses, a
process that capitalises on the ability of the gastrointestinal tract to tolerate exposure to food and
commensal microbiome without triggering inammatory responses. Repeating exposure to type II
collagen induces oral tolerance and inhibits induction of arthritis, a chronic inammatory joint
condition. Although some mechanisms underlying oral tolerance are described, how dysregulation of
gut immune networks impacts on inammation of distant tissues like the joints is unclear. We used
undenatured type II collagen in a prophylactic regime -7.33 mg/kg three times/week- to describe the
mechanisms associated with protective oral immune-therapy (OIT) in gut and joint during experimental
Collagen-Induced Arthritis (CIA). OIT reduced disease incidence to 50%, with reduced expression of
IL-17 and IL-22 in the joints of asymptomatic mice. Moreover, whilst the gut tissue of arthritic mice
shows substantial damage and activation of tissue-specic immune networks, oral administration of
undenatured type II collagen protects against gut pathology in all mice, symptomatic and
asymptomatic, rewiring IL-17/IL-22 networks. Furthermore, gut fucosylation and microbiome
composition were also modulated. These results corroborate the relevance of the gut-joint axis in
arthritis, showing novel regulatory mechanisms linked to therapeutic OIT in joint disease.
Rheumatoid arthritis (RA) is a chronic, autoimmune inammatory con-
dition affecting primarily the synovial joints, although inammation can
affect other tissues like the skin, eye, heart, lung, kidney and gastrointestinal
systems1,2. Despite advancements in immunosuppressive treatments,
approximately 2030% of patients still do not respond to them, and a clear
increase of incidence rates in the global ageing population is described3.
Thus, new therapeutic interventions are needed, but the aetiology of RA is
not yet completely understood, and most disease triggers are still uni-
dentied. Although genetic factors are involved, it is known that environ-
mental effects can play a key role in disease initiation and progression. For
example, smoking can induce post-translational modications leading to
major histocompatibility complex (MHC) presentation of modied self-
proteins to T cells. As a result, autoimmune responses are initiated,
including the production of self-reactive antibodies against immunoglo-
bulin G (Rheumatoid Factor), antibodies to citrullinated protein antigens
(ACPAs)4,5or the cartilage component collagen type II (C-II)6. Interestingly,
and contrary to the heterogenous pool of antigens recognised by ACPAs or
Rheumatoid Factor, type II collagen, being a single molecule, constitutes a
good target for clinical interventions based on oral tolerance mechanisms.
Experimentally, responses against collagen can be studied in vivo using the
collagen-induced arthritis (CIA) model7, in which intradermal injections of
type II collagen and Freuds adjuvant break tolerance and initiate inam-
matory responses in the joint. Data generated with the CIA model have
shown that repeated oral collagen administration stimulates systemic
1Centre for the Cellular Microenvironment, School of Molecular Biology, University of Glasgow, Glasgow, UK. 2Department of Bacteriology and Immunology,
Beijing Chest Hospital, Capital Medical University/Beijing Tuberculosis & Thoracic Tumor Research Institute, Beijing, China. 3Institute of Infection and Immunity,
University of Glasgow, Glasgow, UK. 4Research and Development, Lonza Greenwood LLC, North Emerald Road, Greenwood, SC, USA.
5
Present address: Institute
of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK.
e-mail: miguel.pineda@glasgow.ac.uk
Communications Biology | (2024) 7:804 1
1234567890():,;
1234567890():,;
Content courtesy of Springer Nature, terms of use apply. Rights reserved
tolerance810. Various underpinning mechanisms have been described
including expansion of tolerogenic dendritic cells in the gut-associated
lymphoid tissue (GALT), with subsequent induction of antigen-specic
Tregs, IL-10 and TGFβresponses9,11,12 and suppression of inammatory IL-
17 pathways13. These positive results in pre-clinical studies encouraged
further development of collagen-based formulations. In this study, we used
undenatured type II collagen, a form of collagen isolated from the cartilage
of chicken sternum that preserves the physiological structure of collagen
bres14. Unlike the production of denatured collagen molecules, the unde-
natured type II collagen manufacturing process maintains the intact gly-
cosylation and protein tertiary structure15 that is required to shape the
proteins characteristic triple helix16. The preservation of this structure is key
because it determines the three-dimensional epitopes presented to immune
cells in the Peyers Patches of the gut17. Furthermore, by resisting the
hydrolytic action of human gastric uid15, the preservation of triple helix
domains allows a more efcient presentation of epitopes and subsequent
regulatory responses. Supporting these ndings, some studies indicated that
undenatured type II collagen reduces pain and joint stiffness in osteoar-
thritis, both in experimental models and patients1820. Mechanistically, oral
collagen administration promotes regulatory T cell function in the gut tis-
sue, increasing IL-4, IL-10 and TGFβlevels20, whilst down-regulating
inammatory cytokines (IL-2, TNF, IL-6 and IL-1) and metabolites, like β-
hydroxybutyrate2123. Thus, undenatured type II collagen has also shown
efcacy in ameliorating pain in joint disease15,althoughtheprecise
immunological basis of protection remains unclear. Perhaps surprisingly,
less attention has been given to the structural aspects of the gastrointestinal
regions where the described effector mechanisms take place. Recent
research indicates that maintenance of the local gut tissue architecture and
microenvironmental cues are critical for correct immunological dis-
crimination and homoeostatic resolution of inammation. Loss of mucosal
barrier function in the gut has been implicated in the aetiology of arthritis24,25
suggesting that restoration of the gut barrier integrity could be exploited
therapeutically. Moreover, it has also been postulated that RA could initiate
at mucosal sites before transitioning to more distant synovial joints26.Thus,
a better understanding of the microenvironmental changes in gut mucosal
sites in health and disease will allow us to understand and ultimately utilise,
induction of oral tolerance.
In this study, we use the murine model of CIA to further characterise (i)
immune mechanisms activated upon administration of prophylactic doses
ofundenaturedtypeIIcollageninmice,and(ii)functionallyrelevant
structural changes of the gut mucosal sites associated with undenatured type
II collagen protection against CIA. Using this system as a model of oral
immune therapy (OIT), we show that mice treated with undenatured type II
collagen present a signicant reduction in disease incidence, which is
associated with the regulation of IL-17 and IL-22 cytokines in the joint and
rewiring of cellular and cytokine networks in the gut. Specically, OIT was
associated with the protection of gut villi and crypt structure and the
maintenance of local levels of fucosylation. In describing the local gut
responses by which OIT acts to suppress inammatory responses, these
ndings increase our fundamental understanding of the role(s) of the gut in
autoimmunity and will support the development of alternative areas for
clinical intervention and prevention in RA.
Results
Undenatured type II collagen protects against experimental
arthritis
We chose the CIA model to study the effects of oral administration of
undenatured collagen in arthritis progression. We dened three experi-
mental groups: (i) non-treated animals (Naïve), (ii) mice undergoing
experimental arthritis (CIA) and (iii) mice undergoing arthritis with pro-
phylactic undenatured type II collagen oral immunotherapy (OIT). OIT
started 2 weeks prior to CIA induction, and oral gavage of undenatured type
II collagen was applied 3 times per week. All mice in the CIA group
developed symptoms. Monitoring of disease progression showed that OIT
suppressed joint inammation as evidenced by the signicant reduction in
disease scores and incidence (Fig. 1a), showing that ~50% of the mice did not
show any clinical symptoms. To evaluate whether OIT impacted pre-
dominantly on disease incidence, or it was inuencing both incidence and
severity of disease, we divided OIT mice into symptomatic and asympto-
matic groups to measure paw swelling and relative weight change (Fig. 1b).
OIT symptomatic mice were not signicantly different to CIA controls, in
terms of paw swelling and weight loss, whilst OIT asymptomatic mice
showed similar values to healthy naïve controls. Values for individual mice
are represented in Supplementary Fig. 1.
Corroborating the clinical results, histopathological analysis of CIA
joints showed high numbers of inltrating immune cells, causing bone
damage and severe loss of cartilage that was absent in those OIT mice that
were asymptomatic (Fig. 1c). Quantication of these disease indicators
showed that asymptomatic OIT mice were completely protected against
cartilage and bone damage in all cases, although they presented low levels of
cell inltration and pannus formation in some cases (Fig. 1d). Symptomatic
OIT mice showed higher percentages of mice with no pathological signs for
cell inltration, cartilage damage and bone damage, although the results
were not statistically signicant and overall, scores were similar to those
observed in the CIA control group (Fig. 1d).
OIT inhibits systemic inammatory and cellular pathways
Disease scores and histology demonstrated the therapeutic effect of unde-
natured type II collagen, protecting around 50% of the CIA mice from
developing symptomatic arthritis. To gain insight into the protective
mechanisms, we rst evaluated the effect on pathogenic humoral responses
as determined by the levels of IgG1 and IgG2a anti-CII antibodies27.As
expected, all mice undergoing CIA had increased levels of anti-collagen
antibodies in serum, with OIT mice exhibiting slightly lower anti-CII IgG1
but similar IgG2a levels compared to CIA (Fig. 2a). Nevertheless, both
symptomatic and asymptomatic mice generated high levels of anti-CII
antibodies, suggesting that the inhibition of humoral responses was not a
pivotal factor in preventing arthritis development in our model.
Hence, we next assessed cellular immunity, working under the
hypothesis that cellular immune responses were rewired in protected OIT
mice. First, we counted the total number of cells in joint-draining lymph
nodes (DLN), i.e., axillary, brachial and popliteal. As expected, CIA mice had
asignicantly higher number of cells compared to naïve, whereas the
numbers in naïve and asymptomatic OIT were not signicantly different
and symptomatic OIT mice resembled CIA controls (Fig. 2b). Further
analysis of distinct immune cell populations by ow cytometry showed that
this trend was conserved in B and T cells, including CD4+and CD8+subsets,
as all these lymphocyte groups in asymptomatic OIT were not signicantly
different from the levels found in naïve mice (Fig. 2c).
Since regulatory T cells (Tregs) have been extensively implicated in the
establishment of tolerance, we evaluated the levels of CD3+CD25+FoxP3+
Tregs in the DLNs of asymptomatic OIT mice compared to those of the
naïve and CIA groups (Supplementary Fig. 2a). We also evaluated CD39
and CD73 expression as functional indicators of their effector suppressive
capacity, since these markers are regardedasimmunologicalswitchesthat
shift ATP-driven pro-inammatory immune cell activity towards an anti-
inammatory state mediated by adenosine28 (Supplementary Fig. 2b, c). We
did not observe any expansion of Tregs in asymptomatic OIT mice at the full
day (day 33), suggesting that (systemic) Treg-mediated mechanisms were
not responsible for the observed undenatured type II collage n protection
against CIA during this late stage. Although we cannot rule out that Tregs
play a role in protection during pre-clinical disease stages, we hypothesised
that protection in CIA-OIT mice was associated with a reduction in pro-
inammatory cytokines. Specically, we investigated the expression of IL-
17, a highly pathogenic factor in CIA29,andIL-22,acytokinewereportedto
promote the development of joint disease3033.Toconrm the pathogenic
role of IL-17 and IL-22 in the CIA joint, we rst collated mice from all groups
to directly compare their clinical scores with IL-17 and IL-22 expression in
CD4+T cells (Fig. 3a) and B cells (Fig. 3b), analysed by ow cytometry.
Results indicate that levels of IL-17+and IL-22+CD4 T cells signicantly
https://doi.org/10.1038/s42003-024-06476-z Article
Communications Biology | (2024) 7:804 2
Content courtesy of Springer Nature, terms of use apply. Rights reserved
correlate with disease severity, regardless of treatment. The impact of B cell-
derived IL-17 and IL-22 production on pathology was diminished com-
pared to that of CD4+T cells, corroborating the leading role of Th17 cells in
progressing joint inammation. To corroborate these ndings, we next
analysed IL-17 and IL-22 expression in the joint tissue of CIA and
asymptomatic OIT mice by immunouorescence (Fig. 3c). In line with our
previous results, non-inamed tissue from naive mice exhibited minimal
cytokine expression, whereas effective OIT mitigated the heightened
expression observed in CIA, with a signicant reduction in IL-22 (Fig. 3c).
OIT also reduced production of IL-17 by DLN cells from OIT mice,
including both symptomatic and asymptomatic animals, compared with
their CIA counterparts upon TPA/Ionomycin stimulation in vitro (Fig. 3d).
https://doi.org/10.1038/s42003-024-06476-z Article
Communications Biology | (2024) 7:804 3
Content courtesy of Springer Nature, terms of use apply. Rights reserved
Fig. 1 | Disease scores and histological analysis in response to oral immu-
notherapy (OIT) in CIA mice. a Disease scores (left panel) and incidence (right
panel) were evaluated at the indicated time points for naïve (grey), CIA (red) and
CIA-OIT (blue). OIT consisted of administration of undenatured type II collagen by
oral gavage 3 times a week, starting 2 weeks before induction of arthritis. Cumulative
disease scores were given for each limb, 0 =no disease and 4 = highest score. Each
dot represents the mean of individual mice and error bars show SEM (n= 15 for CIA,
n= 16 for OIT groups, n= 9 for naïve mice, data from 2 independent experiments).
Statistical signicance was determined by one-way ANOVA; *p< 0.05, ***p< 0.001
between CIA and naïve, #p< 0.05, ##p< 0.01 comparing CIA and OIT groups. Dis-
ease incidence was calculated by dividing the number of cases by the total number of
mice in the group at the indicated times. bPaw width (left panel) and weight change
(percentage of initial weight, right panel) were evaluated at the indicated time points.
Each dot represents the mean of individual mice, error bars show SEM (n= 15 for
CIA, n= 7 for OIT asymptomatic, n= 9 for OIT symptomatic, n= 9 for naïve mice,
data from 2 independent experiments). Statistical signicance was determined by
one-way ANOVA; *p< 0.05, ***p< 0.01,***p< 0.001, ns = non-signicant. cHind
paws were collected at the end of the experiment (day 34) for each mouse, sectioned,
and stained with haematoxylin and eosin for histological analysis. Images show one
representative mouse (Disease score = median for each group). Superimposed
dotted lines show bone limits; scale bar = 500 μm. dCell inltration, pannus for-
mation, cartilage and bone damage were quantied for individual mice in hema-
toxilin/eosin-stained sections. Dot plots show histological scores (from 0 to 4,
0 = healthy tissue, 4 = highest possible score). Individual dots represent individual
paws (n= 32 for CIA, n= 8 for OIT asymptomatic, and n= 24 for OIT symptomatic,
data collected from 2 independent experiments). Bars show the mean value for each
group. Statistical signicance was determined by the KruskalWallis test; **p< 0.01,
***p< 0.001. The percentage of mice with no sign of pathology (green) for the
indicated parameter is shown in vertical column graphs.
Fig. 2 | Cellular and humoral responses in response to oral immunotherapy (OIT)
in the joint. a Anti type II collagen antibodies, IgG1 and IgG2 isotypes, were eval-
uated by ELISA in serum from naïve control mice (n= 8), CIA (n= 7, mean of
disease score 4.86) and OIT mice (n= 7 for OIT symptomatic, mean of disease
scores = 7; n= 4 for OIT asymptomatic) at day 33 after induction of arthritis. Values
represent Optical Density (OD) at 450 nm. Each dot represents the mean values of
individual mice analysed in technical triplicates. bTotal cell numbers isolated from
draining lymph nodes (DLNs) collected from all naïve, CIA, asymptomatic and
symptomatic OIT mice. cTotal number of B cells, total T cells, CD4 and CD8 T cells
in DLNs from (b) were evaluated by Flow Cytometry. Each dot in accolumn bars
represents values of individual mice collated from 2 independent experiments. Error
bars show mean ± SEM. Statistical signicance was determined using ordinary one-
way ANOVA. Signicance is indicated by asterisks, *p< 0.05, **p< 0.01
and ***p< 0.001.
https://doi.org/10.1038/s42003-024-06476-z Article
Communications Biology | (2024) 7:804 4
Content courtesy of Springer Nature, terms of use apply. Rights reserved
Following this, we conducted ow cytometry (Supplementary Fig. 3 for
gating strategy) to investigate intracellular IL-17 and IL-22 expression in all
DLN cells (Fig. 3e), categorising OIT mice into symptomatic and asymp-
tomatic groups as before. Mice in theOITgroupthatremainedasympto-
matic exhibited markedly reduced expression levels of both IL-17 and IL-22
producers. Symptomatic mice displayed immune proles that resembled
those of arthritic mice, although they still showed a reduction, albeit not
signicant, in IL-17 producers. We further analysed which IL-17- and IL-
22-producing DLNs were modulated in response to OIT. CD4+and CD8+
T cells, and CD19+B cells data (cell frequency and numbers) were collated
and normalised in radar charts for visualisation (Fig. 3f, raw data shown in
Supplementary Fig. 4). This revealed that whilst IL-17+and IL-22+CD4 and
https://doi.org/10.1038/s42003-024-06476-z Article
Communications Biology | (2024) 7:804 5
Content courtesy of Springer Nature, terms of use apply. Rights reserved
CD8 T cells were signicantly reduced, B cell-dependent production of IL-
17 and, to a lesser extent, IL-22 were less affected in asymptomatic OIT
relative to CIA mice. Moreover, an intriguing response was observed in
asymptomatic OIT mice, which exhibited a signicantly higher frequency of
IL-17+B cells (Fig. 3f, Supplementary Fig. 4).
Oral administration of undenatured type II collagen protects
against damage to gut tissue in CIA
The establishment of oral tolerance by feeding of antigens is a direct con-
sequence of specic immune responses triggered in the gastrointestinal tract
and gut-associated lymphoid tissue (GALT), which can lead to loss of
mucosal barrier function in RA, suggesting that gut tissue architecture can
modulate gut immunity and host-microbiome interactions25. Our previous
work showed that gut pathology precedes and perpetuates chronic systemic
inammation driving autoimmunity and joint damage in CIA24. Therefore,
we evaluated the integrity of the gastrointestinal tract (duodenum, jejunum,
ileum and colon) in our experimental OIT model (Fig. 4a). CIA mice
showed substantial damage in all gut areas, such as disruption of the epi-
thelial layer, cell inltration and thickening of the muscular layer (Fig. 4b).
We quantied the ratio of villi to crypt length in the gut to compare the
health and function of the intestinal mucosa. CIA mice exhibited a
diminished villi-to-crypt ratio, reminiscent of ndings in inammatory gut
conditions. This alteration was rectied in the duodenum of asymptomatic
OIT mice, with a similar trend in the jejunum and ileum. This effect was not
observed in the colon. Interestingly, symptomatic OIT mice, characterised
by joint pathology, also exhibited an absence of gut damage or pathology.
Indeed, symptomatic OIT tended to exhibit even higher villi-to-crypt ratios
than healthy mice, particularly in the distal areas of the small intestine such
as the ileum but not in the colon (Fig. 4b). Given the persistently reduced
villi-to-crypt ratio observed in the colon of all OIT mice, we conducted
Periodic Acid-Schiff (PAS) staining to visualise further tissue damage
associated to redistribution or changes in glycogens and mucins. PAS
staining revealed attachment/effacement lesions in the colon of arthritic
mice (Fig. 4c), in line with previous reports24,25. However, such lesions were
absent in asymptomatic OIT mice but not in those showing joint symp-
toms (Fig. 4c).
Finally, to assess how the safeguarding of gut tissue in asymptomatic
OIT mice translated into gut immune responses, we examined the total
number of cells in mesenteric lymph nodes (MLNs), as well as those of B
cells and CD4+and CD8+T cells. We did not observe any signicant
difference in the number of DLN cells amongst these experimental groups.
Nonetheless, both asymptomatic and symptomatic OIT mice exhibited an
expansion of B cells in mesenteric lymph nodes (MLNs), not only in
comparison to the CIA group but also in relation to the naive controls, albeit
statistical signicance was attained only for the symptomatic group
(Fig. 4d). In contrast, the numbers of CD4+and CD8+TcellsinMLNs
remained unaltered in both cases.
Overall, our results indicate that OIT modulates gut immunity and
triggers protective mechanisms to preserve gut integrity, even in those mice
who ultimately develop joint pathology. Hence, our next objective was to
conduct a comprehensive characterisation of the local immunological
pathways responsible for protection. We, therefore, worked with asymp-
tomatic OIT mice, a cohort demonstrating neither joint nor gut pathology.
We used cells from MLNs and cells isolated directly from the gut, following
enzymatic tissue digestion (Gating strategy shown in Supplementary Fig. 5).
In line with our experiments in the joint tissue (Fig. 3), we investigated IL-17
expression by ow cytometry in the mesenteric lymph nodes (MLNs) and
ileum and colon tissue (Fig. 5), to assess correlations between gut cellular
networks with distant responses within the joint. Contrary to the DLN data
(Fig. 3), the levels of IL-17 producers in total MLN cells (Fig. 5a), ileum
(Fig. 5b) or colon (Fig. 5c) tissue were not signicantly elevated in CIA mice.
Perhaps surprisingly, there was a trend towards reduced levels of IL-17+cells
in the ileum, although there tended to be higher levels of these cells in the
colon of CIA mice, inverse patterns that were reversed in asymptomatic OIT
mice (Fig. 5b, c). However, analysis of specic cell types revealed a distinct
rewiring of the IL-17-producing networks associated with healthy animals,
in both the CIA and asymptomatic OIT groups, in each of the MLNs
(Fig. 5d), ileum (Fig. 5e) and colon (Fig. 5f) tissue. Overall, a broad analysis
indicates that in healthy conditions, IL-17 is generally produced by innate
cells (NKT, ILC3 and γδ T cells), whilst in CIA this predominantly switches
to adaptive CD4/CD8 cell responses in MLNs, and expansion of more
specialised cell types in the gut tissue, with OIT showing a mixed pattern
distinct to both naive and CIA networks (Raw data from radar charts are
shown in Supplementary Fig. 6). Intriguingly, OIT protection tended to
correlate with increased levels of IL-17+producers in the ileum, although the
results did not reach statistical signicance. Moreover, their production of
IL-17 showed a general increase in the OIT group when assessing the mean
uorescence intensity in all cell types.
We conducted a similar approach to evaluate IL-22 expression in
MLNs and gut tissue (Fig. 6), bearing in mind that IL-22 exerts key mucosal
healing mechanisms, promoting epithelial regeneration and integrity,
mucus production and synthesis of antimicrobial peptides3436.Wedidnot
observe any signicant differences among groups in total IL-22+MLNs
(Fig. 6a), or in the cells isolated from the ileum (Fig. 6b) or colon (Fig. 6c).
Nevertheless, and in line with the previous results shown for IL-17, ow
cytometric analysis of the distinct populations of producers suggests that IL-
22 networks are actively rewired in asymptomatic OIT mice compared to
CIA and naïve controls. Thus, production of IL-22 in naïve MLNs comes
mostly from B cells and innate ILC3 and NKT cells whilst in CIA mice it
extends to CD4+T cells, NKTs and γδ T cells, the latter being the only
enhanced populations in asymptomatic OIT (Fig. 6d). Analysis of IL-22
mean uorescence intensity revealed a generalised increase in IL-22 pro-
ductioninMLNsduringCIA,whereasthiswasreducedbothintheileum
(Fig. 6e) and colon (Fig. 6f), thereby corroborating the protective role of
local IL-22 in the gut tissue. In fact, and in contrast to the IL-22 results in
MLNs, CIA mice did not increase IL-22 production, whilst all innate
populations located in the gastrointestinal tract increased the expression
of IL-22 in asymptomatic OIT mice, as measured by mean uorescence
intensity (MFI) (Fig. 6e, f) (Raw data from radar charts are shown in
Supplementary Fig. 7). Although we cannot unequivocally dene intestinal
Fig. 3 | OIT protection is associated with reduced upregulation of inammatory
IL-17 and IL-22 in the joint and draining lymph nodes. Naïve, CIA and CIA-OIT
mice were culled at day 33 when tissue was collected for further analysis.
a,bCorrelation between numbers of IL-17 and IL-22 positive lymph node cells and
clinical scores in T cells (a) and B cells (b) isolated from draining lymph nodes
(DLNs). Every dot represents values for individual mice from all groups. Data are
presented as mean ± SEM, r: Pearsons coefcient. cExpression of IL-17 and IL-22
(red) was evaluated in the joint tissue by immunouorescence in naïve, CIA and OIT
asymptomatic mice. DAPI (Blue) was used to stain nuclei as counterstaining.
Superimposed dotted lines show bone tissue and areas of cell inltration are indi-
cated by white arrows. Scale bars: 500 μm. Graphs show the quantication of the
mean intensity of individual mice. dIL-17 concentration was evaluated by ELISA in
the supernatants of draining lymph node cells upon PMA (50 ng/ml)/Ionomycin
(500 ng/ml) stimulation for 12 h. Data show naïve, CIA and OIT (symptomatic and
asymptomatic). Each dot represents cells from one individual mouse. Error bars
show mean ± SEM; *p< 0.05, **p< 0.01 analysed by one-way ANOVA from one
experimental model. eRelative frequency and total cell number of IL-17+and IL-
22+DLN cells were evaluated by ow cytometry. Data show mean ± SEM; each dot
represents individual mice from two independent experimental models; *p< 0.05,
analysed by one-way ANOVA. fCell frequency and total cell numbers of IL-17+and
IL-22+CD4 T cells, CD8 T cells and B cells in DLNs, represented at the corners of
radar charts: Naïve (grey), CIA (red) and OIT asymptomatic (orange) and OIT
symptomatic (blue); data were normalised to maximum expression in each group.
Signicance on the raw data among groups was evaluated by ordinary one-way
ANOVA, where *p< 0.05, **p< 0.01 [CIA vs naïve]; $p< 0.05 [naïve vs OIT
symptomatic]; p< 0.05 [naïve vs OIT asymptomatic]; +p< 0.05 [CIA vs OIT
asymptomatic]; §p< 0.05 [CIA vs OIT asymptomatic]; #p< 0.05 [OIT asymptoma tic
vs OIT symptomatic].
https://doi.org/10.1038/s42003-024-06476-z Article
Communications Biology | (2024) 7:804 6
Content courtesy of Springer Nature, terms of use apply. Rights reserved
IL-22 as a protective or pathogenic factor, our results highlight that differ-
ential rewiring of the IL-22+cell network in the gut is associated with
inammatory or homoeostatic conditions, with a general expansion of
IL-22+innate populations and signicant increase in IL-22 production by
NKT cells in asymptomatic OIT mice. Finally, IL-22 staining in gut tissue
provided further support for the protective role of local gut IL-22 expression
in OIT mice, as there was a signicant increase in IL-22 secretion/deposition
inthetissueepitheliuminthecolonof asymptomatic OIT mice compared to
that CIA mice (Fig. 6g, h). Interestingly, increased staining of IL-22 was also
observed in symptomatic OIT mice (Fig. 6g, h), perhaps explaining why the
gut of symptomatic OIT mice also exhibited protected tissue integrity
(Fig. 4b, c).
https://doi.org/10.1038/s42003-024-06476-z Article
Communications Biology | (2024) 7:804 7
Content courtesy of Springer Nature, terms of use apply. Rights reserved
Gut fucosylation and microbiome composition are reshaped in
OIT and arthritic mice
Since our results suggest that intestinal IL-22 networks correlate with
effective OIT, we next investigated potential protective mechanisms trig-
gered by this cytokine in asymptomatic OIT mice. In health, IL-22 not only
supports homeostatic expression of mucins in epithelial cells37,butalsotheir
fucosylation stage38. Fucosylation is a specic type of post-translational
glycosylation, that prevents infections and enhances the integrity of the
epithelial gut layer39,40. Therefore, because asymptomatic OIT mice pre-
served gut tissue integrity and had enhanced gut IL-22 expression, we rst
hypothesised that such protection was associated with increased epithelial
fucosylation, which would, in turn, prevent gut damage during inamma-
tion. To investigate this, we stained the tissue with two lectins that recognise
fucosylated glycans, Ulex european Agglutinin (UEA) and Auleria aureate
lectin (AAL), that bind to terminal and core fucosylation respectively.
Consistent with the protective role proposed for terminal fucosylation, UEA
binding (recognising terminal alpha[1,2] linked fucose residues) was sig-
nicantly reduced in the ileum of CIA mice, while asymptomatic OIT mice
more resembled the prole of healthy tissue, as they were not signicantly
different to the Naïve group (Fig. 7a). By contrast, no signicant difference
was seen in the colon across the groups in this model (Fig. 7a). Moreover,
neither the ileum nor the colon, showed signicant differences in core
fucosylation as detected by AAL binding (Fig. 7b). Next, we evaluated
mRNA expression of fucosyltransferases (FUTs) in whole gut tissue,
enzymes responsible for fucosylated glycan biosynthesis. In the ileum, only
expression of FUT8 mRNA was signicantly different inasymptomatic OIT
mice (Fig. 7c), whereas no signicant changes were seen in the colon
(Fig. 7d). These results in FUT expression do not explain the observed
maintenance of fucosylation in OIT mice compared to the reduced levels in
CIA mice (Fig. 7a), suggesting that other factors may be involved. This is not
completely unexpected since gut epithelial fucosylation is strongly depen-
dent on environmental factors, such as microbiome composition, which is
also regulated by cytokine-dependent mechanisms41. Dysregulation of some
microorganisms in the gut has been linked to RA pathogenesis and mor-
bidity in multiple studies, perhaps through subsequent variation of intestinal
metabolites that promote inammation in the target tissue42,43. Therefore, an
alternative hypothesis is that the rewiring of IL-17/IL22 immune networks
upon undenatured type II collagen administration modies the composi-
tion of the gut microbiome, or vice versa, which in turn, can affect mucin
fucosylation consolidating dysregulation of the microbiome to perpetuate
systemic inammatory response. To provide support for this hypothesis, we
conducted 16 S amplicon sequencing to investigate the microbial diversity
in the ileum (Fig. 8)andcolon(Fig.9), using faecal samples from naïve, CIA
and OIT mice. As before, OIT mice were separated into symptomatic
(disease scores 5 ± 0.6, n= 3) and asymptomatic mice. We also separated
CIA mice into established disease with high scores (9.3 ± 2.8, n=3), and
mice with more recently initiated joint inammation and lower disease
scores (3.6 ± 0.22, n= 3), to identify changes in microbial content led by OIT
and not a reduced inammatory environment. To understand the microbial
community diversity in the samples (Within-community) we looked at α-
diversity indexes Dominance, Simpson, Observed_otus, Shannon and
Chao1 (Fig. 8a). The ileum microbiome of OIT asymptomatic group
showed signicant differences in the Dominance (higher) and Simpson
index (lower), suggesting a microbial community that is characterised by
lower diversity, skewed abundance distribution, and less evenness across
different taxa. Differences in β-diversity visualised by the UniFrac distance
suggest that the microbial communities in the OIT asymptomatic mice have
distinct compositions (Fig. 8b). The composition and relative abundance of
the ileum microbiota at the phylum level were examined (Fig. 8c). Firmi-
cutes was the dominant phylum in all groups, followed by Bacteroidetes
(Bacteroidota). CIA mice at earlier joint disease stages (CIA low) exhibited a
vefold reduction in the proportion of the Bacteroidota, a loss that was
absent in both symptomatic and asymptomatic OIT mice. Perhaps unex-
pectedly, CIA mice exhibiting more established, high-score arthritis, rather
than the more recently developed low-score arthritic mice, displayed
microbiota proles and indexes closer to those of naïve mice. This is likely
related to the appearance of self-resolving mechanisms described in the CIA
model that can occur around two weeks after joint disease onset such as TGF
production44, which can affect the microbiome composition, particularly the
Bacteroidetes45, thereby suggesting that CIA mice with lower scores due to
them being at earlier stages of disease may provide a better reference for
identication of pathogenic microbiome changes. Compared to sympto-
matic mice, the asymptomatic OIT group increased the diversity of bacteria
in the colon, expanding the Campylobacteria and Proteobacteria phyla.
Moreover, the analysis of the top 35 genera in abundance (Fig. 8d) revealed
that whilst the Lachnospiraceae,Mucispirillium,andAnaerotruncus showed
a consistent reduction in the ileum of asymptomatic OIT mice, Mur-
ibaculaceae species were increased (Fig. 8e).
A similar analysis was carried out on the colon faecal material, speci-
cally showing alpha diversity indexes (Fig. 9a), beta diversity by UniFrac
distance (Fig. 9b), and relative abundances at the phyla (Fig. 9c) and genera
(Fig. 9d, e) levels. Contrary to the ileum, the colon microbiome of OIT
asymptomatic mice presents a lower dominance index and a higher
Simpson index compared to symptomatic OIT and arthritic mice with lower
disease scores (Fig. 9a), suggesting a more diverse and even population that
is not strongly dominated by a few specic taxa in asymptomatic OIT.
Consistent with previous reports, analysis at the phyla level reveals higher
diversity in the colon than in the ileum in naïve mice. Firmicutes remain the
most abundant phylum, but there is an increased proportion of Bacter-
oidetes and Deferribacteriota. OIT asymptomatic mice showed a more
diverse prole overall but exhibited a reduction in the Bacteoridetes and
Patesciabacteria phyla relative to the CIA low-score arthritic mice (Fig. 9c).
At the genus level, the abundance of the top 35 genera present changed
between asymptomatic OIT mice and the rest of the groups (Fig. 9d).
Clostridia_vadinBB60 were reduced, whereas others like Colidextribacter,
Roseburia, Rikenella,andRuminococcaceae were increased, relative to the
CIA-low score group (Fig. 9e). Critically, the distinct proles of healthy
(Naïve) and OIT asymptomatic mice suggest that rather than simply pre-
venting CIA-induced changes in the microbial species, OIT rewires the
interactive network between the gut immunoregulatory pathways and
commensal bacteria to a phenotype that protects against arthritis. Such a
network is likely to be bidirectional, with bacterial metabolites further
adjusting host immunity. To gain some insight into the potential biological
activities of microbial communities without directly measuring gene
expression or protein function, we conducted some metagenomic function
prediction based on the 16 S sequencing using the bioinformatic tool
PICRUSt246. This analysis indicated differences among naïve, CIA and OIT
groups, particularly in the ileum of OIT asymptomatic mice
Fig. 4 | OIT protects against gastrointestinal damage associated with inam-
matory arthritis in both symptomatic and asymptomatic cases. Naïve, CIA and
OIT asymptomatic mice and OIT symptomatic mice were culled at day 33 when gut
tissue and total mesenteric lymph nodes (MLNs) were collected. aIsolated gastro-
intestinal tract from a naïve mouse showing the four anatomical areas used for
further study. bDuodenum, jejunum, ileum, and colon were xed, and tissue sec-
tions were subjected to hematoxylin and eosin staining. The length of villi and crypts
was measured using Image J software, and the ratio of villi/crypt was quantied. Each
dot represents the villi/crypt ratio for individual mice, and data were collated from
two independent experiments. Statistical signicance was evaluated by ordinary
one-way ANOVA, where *p< 0.05, **p< 0.01 and ***p< 0.001. cColon tissue
sections were stained with PAS to detect changes in the mucus layer and associated
pathology. Scale bars = 500 μm. The depicted mice are representative of individual
mice whose disease scores fall within the median value for each group. dMLNs were
collected to generate single-cell suspensions, and a total number of cells was counted.
Cell number and percentage of B cells, total T cells, CD4 and CD8 T cells were
evaluated by Flow Cytometry. Each dot represents one individual mouse and error
bars show mean ± SEM. Mice were pooled from 2 independent experiments. Sta-
tistical signicance was determined using Ordinary one-way ANOVA. Signicance
is indicated by asterisks, *p< 0.05, **p< 0.01.
https://doi.org/10.1038/s42003-024-06476-z Article
Communications Biology | (2024) 7:804 8
Content courtesy of Springer Nature, terms of use apply. Rights reserved
(Supplementary Fig. 8a), but also in the colon (Supplementary Fig. 8b).
Although further experimental work is required to demonstrate this, this
data suggest that effective OIT could impact of the functional host-
microbiome crosstalk during chronic arthritis.
Discussion
Oral tolerance mechanisms have been the subject of investigation since the
beginning of the 20th century, but translation of these ndings into a clinical
setting has not always been successful, indicating that key pathways are still
Fig. 5 | Expression of IL-17 in mesenteric lymph nodes gastrointestinal tract.
Naïve, CIA and OIT asymptomatic mice were culled at day 33, when mesenteric
lymph nodes (MLNs), ileum and colon samples were collected. Single-cell suspen-
sions were obtained from MLNs and digested gut tissue, and IL-17 expression was
subsequently evaluated by ow cytometry in total isolated cells (ac) and specic cell
populations, including CD4 T cells, CD8 T cells, group 3 innate lymphoid cells
(ILC3), γδ T cells and NK cells (df). acPercentage and number of total IL-17+
cells in MLNs (a), ileum samples (b) and colon (c). Each dot represents values of
individual mice; bars show mean values for each group ±SEM; Naïve n= 10. CIA
n= 15, asymptomatic OIT n= 7. Statistical signicance was determined using
ordinary one-way ANOVA, *p< 0.05. dfRelative cell frequency and mean uor-
escence intensity (MFI) of IL-17 in the indicated cell populations in MLNs (d), single
cells isolated from the ileum (e) and colon (f). Each corner of the radar charts
represents the indicated normalised parameter for naïve (grey), CIA (red) and
asymptomatic OIT (orange) mice. Data were normalised to maximum expression in
each group; naïve n= 5, CIA n= 5, asymptomatic OIT n= 4. Statistical signicance
was determined using raw data and ordinary one-way ANOVA. *p< 0.05,
**p< 0.01 in CIA versus Naïve; p< 0.05 [naïve vs OIT asymptom atic].
https://doi.org/10.1038/s42003-024-06476-z Article
Communications Biology | (2024) 7:804 9
Content courtesy of Springer Nature, terms of use apply. Rights reserved
unknown. Our results show that a high percentage of animals (~55%) that
received OIT were fully protected against experimental arthritis, with no
symptoms of disease, supporting the previously described protective role of
undenatured type II collagen in inammatory joint disease15,22,4749.Our
analysis of asymptomatic and symptomatic OIT mice, in comparison with
the CIA group, therefore, contributes to our understanding of the
mechanisms triggered by OIT to achieve a therapeutic effect. Asymptomatic
OIT mice are strongly protected against cartilage and bone damage, which
may be a direct consequence of the lower IL-17 and IL-22 levels in the joint.
We still observed some cell inltration in these mice, however, suggesting
that this was not sufcient to initiate any inammatory response or reected
recruitment of inammation resolving/suppressing cells. By contrast,
https://doi.org/10.1038/s42003-024-06476-z Article
Communications Biology | (2024) 7:804 10
Content courtesy of Springer Nature, terms of use apply. Rights reserved
symptomatic OIT animals exhibited immune cell inltration and clinical
joint scores similar to those of CIA mice, suggesting that OIT impacted
disease incidence rather than disease severity. Nevertheless, both sympto-
matic and asymptomatic mice showed protection against gut damage,
indicating that all OIT mice had undergone some local immunoregulation
regardless of whether they developed clinical joint symptoms or not. Such
gut local protection could be a consequence of the elevated IL-22 gut levels
observed in both groups. IL-22 promotes the protection of the gut tissue,
primarily through the maintenance of mucosal homoeostasis and activation
of anti-microbial responses50. Thus collectively, the high IL-22 in the joint
and gut of symptomatic OIT mice may reect pro-inammatory effects in
the joint, but homeostatic mechanisms in the gut. IL-22, therefore, could
play a central role in OIT, a factor that has been involved not only in joint
inammation and epithelial repair, as discussed, but also in the regulation of
fucosylation of mucins and modulation of the gut microbiome, factors that
are also modulated in our experimental model. However, we still do not
know the factors that ultimately drove protection in the asymptomatic OIT
mice. Environmental factors could be affecting the functional outcomes of
IL-22 in some mice, and further studies should address these to harness all
the therapeutic potential of undenatured type II collagen.
Following the comparative analysis of symptomatic and asymptomatic
OIT mice, we next focused on the investigation of the protective mechan-
isms activated in mice with no clinical symptoms. This demonstrated that
the protective effect of undenatured type II collagen occurs, at least in part,
through the protection of targeted organs distal to the joint such as the gut,
where it acts broadly to rewire cytokine networks and regulate the gut
dysbiosis observed in arthritic mice. Interestingly, asymptomatic OIT mice
displayed a distinct ileum and colon microbiome rather than a preservation
of the bacteria found in healthy animals, indicating an active modulation of
the bacterial community and potentially, the subsequent metabolite pro-
duction to effect protection. Interestingly, therefore, Roseburia species were
expanded in asymptomatic OIT, a genus which produces immunor-
egulatory Short Chain Fatty Acids (SFCA) and inhibits Th17 but increases
IL-22 responses in the gut5154.Theseactionsoverlapwithourinvivo
ndings in the intestine of asymptomatic OIT mice, providing support to
the hypothesis that rewiring of functional microbiome-cytokine networks is
boosted in protected OIT. Such networks will likely include more players,
increasing the level of complexity and bidirectional regulation. For example,
Ruminococcaceae, another genus increased in the colon of asymptomatic
OIT mice, produces SFCA which has been directly implicated in activating
the Wnt/β-catenin pathway responsible for gut epithelium regeneration55.
Likewise, modulation of local gut immune system populations in OIT may
exert some selective pressure on the microbiome composition, to facilitate
the dominance of protective genera. For example, the expansion of ILC3
cells observed in asymptomatic OIT mice could contribute to this by
modulating the microbiota composition via epithelial fucosylation40,41.
Additional experiments are needed to provide further support to these data,
but our ndings provide additional evidence for the role of a pathogenic
gutjoint axis in arthritis and describe new networks that may underpin the
induction of anti-inammatory responses upon exposure to fed antigens
such as undenatured type II collagen.
Amelioration of joint inammation and bone damage during protec-
tive OIT was also associated with the downregulation of pro-inammatory
IL-17 in the joint and draining lymph nodes. Inammatory factors pro-
duced systemically and in the joint, such as IL-17, could induce damage in
the gut tissue to exacerbate disease pathology, but a breach of tolerance at
mucosal surfaces is thought to be an initial event in RA that can occur many
yearsbeforediseaseonset.Whichfactoristhecauseoreffectisnotknown,
although both scenarios appear to happen in specic clinical cases. More-
over, we and others have shown that microbiome dysbiosis and gut
pathology precede systemic inammation, autoimmunity and joint disease
in the CIA model24,56. In any case, gut dysbiosis and metabolite production
seem to be a driving factor to ultimately lead to disease progression in mouse
models and human studies57,58.
Some well-dened tolerogenic mechanisms did not appear to be cri-
tical to protection in OIT mice, including the reduced production of anti-
collagen antibodies or induction of regulatory T cells (Treg) in the gut and
draining lymph nodes, although it has previously been reported that type II
collagen increased levels of Treg and TGFβ, in addition to the reduction of
IL-1759. This lack of effect on Tregs can be a consequence of the antigen dose
used in our study since Treg activity is stimulated by lower doses60.Thedose
of 7.33 mg/kg was chosen because our preliminary studies suggested this to
be more efcient under our experimental conditions, perhaps because of the
strong inhibitory effect of IL-17-dependent pathways which are the main
drivers of CIA. A dose of 2 mg/kg was efcient in a recent study of
experimental osteoarthritis, both in young and older animals49. Our study
focused on the late stages of the disease, and we cannot rule out that Tregs
are involved prior to the induction of IL-17-mediated pathways. Likewise,
we did not evaluate the numbers of regulatory B cells (Bregs), but there is
experimental evidence showing that aberrant regulation of this cell type
modulates arthritis progression via gut-dependent mechanisms24,25.Infact,
we observed a signicant increase in the number of B cells in the gut in
response to undenatured collagen treatment but no effects in the production
of anti-collagen antibodies, perhaps indicating a role for Bregs, an area we
plan to address in follow-up studies. For example, since Tregs are generated
in the gut during the induction phase of disease61, additional studies,
including pre-clinical disease stages are required to provide a full under-
standing of undenatured type II collagen-dependent regulatory responses.
The impact of oral administration of undenatured type II collagen on B cells
in MLNs prior to the initiation of disease might also be needed to fully
understand its protective mechanisms, although due to the lack of lineage-
specicmarkers,theseexperimentswillbemorechallengingtoconduct.
Reduction of IL-17 can be directly related to bone protection, as it
increases RANKL and osteoclastogenesis62. Interestingly, IL-22 expression
in draining lymph nodes and joints was increased in CIA mice and
decreased in the asymptomatic OIT group (both IL-17+/IL-22+CD4 T cell
levels show positive correlations with increasing articular score). This is
contrary to the observations in the gut, where IL-22 was generally up-
regulated in OIT mice and provides further support for the dual role
described for IL-22 during arthritis31,63, perhaps related to the ability of this
cytokine to promote gut epithelial cell regeneration50.Thus,IL-22canbea
critical contributor to both disease pathology and protective responses, as
the newly dened gutjoint axis theory64 proposes that the gut mucosa is the
rst site of disease initiation. Consistent with this, CIA mice displayed
signicant gut pathology and structural damage that was absent in OIT
mice. IL-17 acts to regulate mucosal host defence against many invading
Fig. 6 | Expression of IL-22 in mesenteric lymph nodes and gastrointestinal tract.
Naïve, CIA and OIT asymptomatic mice were culled at day 33, when mesenteric
lymph nodes (MLNs), ileum and colon samples were collected. Single-cell suspen-
sions were obtained from MLNs and digested gut tissue, and IL-22 expression was
subsequently evaluated by ow cytometry in total isolated cells (ac) and specic cell
populations, including CD4 T cells, CD8 T cells, B cells, group 3 innate lymphoid
cells (ILC3), γδ T cells, NK cells and NKT cells (df). acPercentage and number of
total IL-22+cells in MLNs (a), ileum samples (b) and colon (c). Naïve n= 10. CIA
n= 15, asymptomatic OIT n= 7. Each dot represents values for individual mice; bars
show mean values for each group ± SEM. dfRelative cell frequency and mean
uorescence intensity (MFI) of IL-22 in the indicated cell populations in MLNs (d),
single cells isolated from the ileum (e) and colon (f). Each corner of the radar charts
represents the indicated normalised parameter for naïve (grey), CIA (red) and
asymptomatic OIT (orange) mice. Data were normalised to maximum expression in
each group; naïve n= 5, CIA n= 5, asymptomatic OIT n=4.gIleum and colon
sections were stained with anti-IL-22 antibodies and specic secondary antibodies
(Red) and DAPI (Blue) as counterstaining . Scale bars = 500 μm. Pixel intensity for
IL-22 staining was quantied using ImageJ, each dot shows the mean value of 10
different areas for each individual mouse. Error bars show standard error (SEM).
Statistical signicance was determined using raw data and ordinary one-way
ANOVA, where *p< 0.05, **p< 0.01. hDetailed images of colon sections stained for
IL-22, scale bars = 100 μm and 20 μm for zoomed areas.
https://doi.org/10.1038/s42003-024-06476-z Article
Communications Biology | (2024) 7:804 11
Content courtesy of Springer Nature, terms of use apply. Rights reserved
Fig. 7 | Effect of protective OIT on fucosylation and fucosyltransferase expression
in the gut tissue during arthritis. a,bIleum and colon sections from naïve, CIA and
asymptomatic OIT mice were stained with UEA (a) and AAL (b) biotinylated lectins
and uorescence streptavidin (yellow) to detect terminal and core fucosylation
respectively. DAPI (Blue) was used as counterstaining. Scale bars: 500 μm. Images
show one representative example of each group. Graphs show the mean pixel
intensity for lectin staining in individual mice; each dot shows the mean value from
10 different analysed areas, quantied using Image J software. c,dRelative
expression of fucosyltransferases mRNA was evaluated by RT-PCR in the ileum (c)
and colon (d) samples, including FUT1, FUT2, FUT4, FUT7, FUT8 and FUT9.
Expression is shown as relative to actin expression. Statistical signicance was
evaluated by one-way ANOVA where *p< 0.05. Each dot represents values of
individual mice where data are collected from two independent experiments. Error
bars represent standard error (SEM).
https://doi.org/10.1038/s42003-024-06476-z Article
Communications Biology | (2024) 7:804 12
Content courtesy of Springer Nature, terms of use apply. Rights reserved
pathogensinthegut,butunresolved IL-17-associated inammation may
cause gut pathology65. In addition, IL-22 has been proposed to play both
pathologic and protective roles in gut integrity and homeostasis. Thus, to
identify pathological mechanisms, we examined the cells producing IL-17
and IL-22 in MLNs, ileum and colon as sites of Th17/22 induction and
tolerance. Interestingly, whilst our results show upregulation of IL-17 in the
gut tissue from CIA but not asymptomatic OIT mice, the most striking
changes appear to reect OIT-induced switches in the cellular networks of
IL-17+and IL-22+cells in the gut, with these cytokines being predominantly
produced by innate effector cells like NKT and ILCs, rather than the CD4+
helper and γδ T cells driving systemic autoimmunity in CIA29.IL-22pro-
duction was increased in all cell types isolated from the ileum in response to
https://doi.org/10.1038/s42003-024-06476-z Article
Communications Biology | (2024) 7:804 13
Content courtesy of Springer Nature, terms of use apply. Rights reserved
undenatured type II collagen administration, and this reected an expan-
sion of IL-22+γδT cells, NKT cells and CD4 T cells. Overall, our data shows
that the protective role of gut IL-17/IL-22 likely depends on the relative
contribution coming from innate and adaptive immune cells, although we
have not investigated all the molecular mechanisms underlying these pro-
tective cytokine and cellular networks.
Future studies plan to focus on the ability of undenatured type II
collagen to harness protective IL-22 actions in the mucosal epithelium50.IL-
22 promotes epithelial fucosylation, which is crucial to maintaining barrier
integrity and immunity66. Indeed, perhaps linking the gut-joint axis, fucose
can play a protective role in both local gut and systemic inammation67.
Interestingly, OIT restored the reduced gut fucosylation observed in CIA
mice. We did not identify the mechanism(s) controlling such tissue fuco-
sylation, but since this was not associated with changes in the expression of
fucosyltransferases, environmental factors could be controlling local fucose
content. For example, our results indicate that OIT induces microbiome
changes that could be responsible for the modulation of fucosylation status
as well as some of the therapeutic anti-inammatory effects. In line with this,
rewiring of immune-glycan-microbiome networks could affect the pro-
duction of protective metabolites such as short-chain fatty acids with known
anti-inammatory effects in models of autoimmunity68,69.Additionally,
specic microbial species may play an integral role in the induction of the
protective glycan-dependent mechanisms associated with OIT. For exam-
ple, segmentedlamentous bacteria (SFB) is a c ommongut resident that can
induce differentiation of Th17+CD4 T cells in arthritis70,71, but also induce
IL-22 production in the small intestine lamina propria to modulate fuco-
sylation of the ileum72,73. Thus, understanding the gut microenvironment,
including cytokine networks, regulation of microbiome composition, and
mechanisms linking both sides, such as epithelial glycosylation, may ll
some of the gaps in the eld, opening new avenues for translational
opportunities, which consider human disease heterogeneity. For example,
several clinical subgroups have been described in RA based on the presence
of specic autoantibodies and/or specic immune responses; where for
instance, the myeloid RA phenotype is driven by TNF, contrary to lymphoid
RA type that is IL-17/IL-1-mediated, and these RA phenotypes correlate
with response to treatments74. Thus, it is likely that specicgroupsof
patients (with distinct pathogenic signatures) respond differently to unde-
natured type II collagen therapies, and the search for predictive biomarkers
should be considered in future clinical trials. Additionally, patients
responses may differ in early or late disease stages, since anti-collagen
antibodies show a higher concentration during early disease stages, and it
has been associated with specic disease phenotypes75,76.
Collectively, our study demonstrates the importance of differential IL-
17- and IL-22-producing cell networks in the joint-gut inammatory axis
and the therapeutic potential of undenatured type II collagen. Regulation of
cytokine networks correlates with protection against arthritis, indicating
that factors affecting them may determine effective OIT. For translational
purposes, understanding the impact of exogenous factors in regulating these
immune networks can offer a way to enhance the clinical value of oral
immunomodulation, and to identify patient groups which may benetfrom
these interventions. In this regard, we have identied molecular and cellular
parameters in the gastrointestinal tract correlating with low inammatory
conditions, including regulation of cytokines, cellular networks, tissue gly-
cosylation and microbial proles. These ndings could offer new oppor-
tunities to treat arthritis and other chronic inammatory disorders affected
by changes in mucosal tissues.
Methods
Collagen-induced arthritis and undenatured type II collagen
treatment
Male DBA/1 mice were purchased at 810 weeks of age (Envigo; Bicester,
UK) and housed and maintained in the Central Research Facility of the
University of Glasgow. We have complied with all relevant ethical reg-
ulations for animal use. All experiments were approved by and conducted
in accordance with the Animal Welfare and Ethical Review Board of the
University of Glasgow, UK Home Ofce Regulations and Licences PPL
P8C60C865, PIL I62988261, PIL I675F0C46. To induce arthritis, mice
were immunised with type II chicken collagen emulsion (1 mg/mL; in
complete Freunds adjuvant [CFA] i.d.) and then administered type II
chicken collagen in PBS i.p. at 21 days after the rst injection. Mice were
divided into three experimental groups, naïve, CIA, and mice receiving
undenatured collagen. Undenatured type II collagen (7.33 mg/kg) was
given to the animals by oral gavage three times a week, starting two weeks
before induction of arthritis. The dose was selected based on pilot studies,
suggesting that 7.33 mg/kg was the most protective under our lab
experimental conditions. A concentrate of undenatured type II collagen
was obtained from Lonza Greenwood LLC (UC-II®undenatured type II
collagen). Monitoring of mouse health, clinical scores and weight were
evaluated every 2 days. Clinical scoring of each limb was used to assess
disease progression, where each paw received a score ranging from 0 (no
inammation) to 4 (highly inamed/loss of functionality) as described
previously29. An aggregated Clinical score of 10 for an individual mouse
was considered an experimental endpoint, with any such mice imme-
diately euthanized.
Hematoxylin and eosin (H&E) staining
Gut tissue and paws from individual mice were collected for histology. Gut
tissue was processed according to the Swiss roll technique, including duo-
denum, jejunum, ileum and colon. Gut tissue and paws were xed in 4%
paraformaldehyde for up to 24 h, then transferred into 70% ethanol for
storage. Following dehydration, tissue was embedded in parafnblocksand
sectioned on a standard microtome at 7 μm thickness using a microtome
(Leica RM2125). For joint blocks, bone decalcication, parafn sectioning
and H&E staining were completed at Histology Research Service, School of
Veterinary Medicine, University of Glasgow. For gut tissue parafnblocks,
tissue processor Lecia Asap300 was used. Sections were heated for 35 min at
60 °C, then followed by H&E staining, which was performed on all tissues
for identication of pathological changes. For analysis of gut sections, the
length of gut villi and crypt depth were determined using Image J software to
calculate villi/crypt length ratios. Pathology of the joint tissue (cell inltra-
tion, pannus formation, bone damage and cartilage damage) was assessed by
visual evaluation according to a score system ranging from 0 (no effect) to 4
(high pathology). The nal score for each mouse paw is representative of two
hind paws. No cut off was used to exclude any joints in the analysis.
Fig. 8 | Analysis of the microbial composition in the ileum. Faecal matter was
taken from the ileum of naïve mice (n= 4), severe arthritis (CIA high, n= 3, disease
scores 11, 10 and 7), mild arthritis (CIA low, n= 3; disease scores 3, 4 and 4),
symptomatic OIT mice (OIT high, n= 3; disease scores 5, 6 and 4) and asympto-
matic OIT mice (OIT low, n= 3) at day 33 when DNA was isolated and subjected to
16 S ribosomal RNA (rRNA) amplication and sequencing. aAlpha Diversity
analysis, including observed_ otus, shannon, simpson, chao1, dominance and pie-
lou_e indices. Each dot represents individual mice; graphs show the mean ± SEM.
KruskalWallis test was used to analyse whether the differences in species diversity
between groups were signicant, *p< 0.05. bBeta diversity indices heatmap of
unweighted unifrac distance Matrix. The size and colour of the circle in the square
represent the differences in coefcient between the two samples. The larger the circle
is, the darker the corresponding colour is, indicating that the differences between the
two samples are greater. cRelative abundance of the indicated phyla. Each column
shows data from individual mice. Data are also presented as pie charts, presenting
proportion values for each group as means. dClustering of Species Abundance. The
top 35 genera in abundance were clustered from the species and sample levels
according to their abundance information in each sample. Heatmap in grey scale
shows the mean value of all mice in the group for Zvalue of taxonomic relative
abundance after standardisation. The coloured heatmap represents the values for
individual mice: the x-axis represents the sample name, and the y-axis represents the
function annotation. The cluster tree on the left side is the species cluster tree.
eRelativeabundance of the indicated genera. Data show mean ± SEM and dots
represent individual mice.
https://doi.org/10.1038/s42003-024-06476-z Article
Communications Biology | (2024) 7:804 14
Content courtesy of Springer Nature, terms of use apply. Rights reserved
Assessment was conducted blindly, by two independent researchers. Images
were acquired using an EVOS microscope.
Periodic acid-Schiff (PAS) staining
Parafn sections were dewaxed using the same method used for H&E
staining and rehydration. The slides were stained with Periodic acid for
5 min and rinsed with water before staining with Schiffs reagent for
15 min. After washing with tap water for 5 min, the sections were
counterstained with haematoxylin for 1 min. Sections were then rinsed
with water and dehydrated using a series of ethanol and xylene, mounted
with DPX and coverslip. Images were acquired on an EVOS brighteld
microscope.
https://doi.org/10.1038/s42003-024-06476-z Article
Communications Biology | (2024) 7:804 15
Content courtesy of Springer Nature, terms of use apply. Rights reserved
Immunouorescence
Tissue sections (7 μm thickness) were dewaxed in xylene followed by
100% EtOH, 90% EtOH, 70% EtOH, 50% EtOH, and 30% EtOH for
3 min x2, respectively. Sections were then put in antigen retrieval buffer
(Citrate buffer, Ph: 5.0) for 20 min at 95 °C31. To block non-specic
antibody binding, sections were incubated with Carbo-Free Blocking
Solution (Vector labs) for 30 min. The slides were washed in PBS-T (PBS
0.05% Tween20), and Streptavidin/Biotin blocking was performed
following kit instructions (Vector laboratories, SP-2002). Sections
were incubated with biotinylated lectins (Vector laboratories, 1:200) or
primary antibodies in PBS and incubated overnight at 4 °C. Sections
were washed with PBS-T at least 3 times. Streptavidin-Alexa 647 or
secondary antibodies (1:400) were applied in PBS at room temperature
for 60 min. Slides were rinsed with PBS and mounted with mounting
media containing DAPI. Images were acquired with an EVOS micro-
scope and confocal microscopy and were later analysed in Image J
software.
Serum cytokine and antibody ELISAs
Single-cell suspensions of lymph nodes (LN) were obtained, and cells were
then cultured (106/ml) and stimulated with TPA (50 ng/ml)/Ionomycin
(500 ng/ml) for 12 h when supernatants were collected for detection of
secreted cytokines. Interleukin-17 (IL-17) was measured by ELISA
according to the manufacturers instructions (Invitrogen). For determina-
tion of anti-collagen type II-specic IgG1 and IgG2a antibodies in serum,
high-binding 96-well ELISA plates were coated with chicken collagen (5 μg/
ml) overnight at 4 °C before washing and blocking with PBS 5% BSA. Serum
from individual animals was diluted (1:100) and incubated with HRP-
conjugated goat anti-mouse IgG1 or IgG2a (1:10,000) in PBS 10% FBS prior
to developing with TMB. Samples were read in a microplate reader (Sunrise)
at an optical density of 450 nm.
Flow cytometry
LN cells were suspended in FACS buffer (1% FBS; 0.5 mM EDTA, in PBS).
Lymphocytes were identied by labelling with antibodies against CD3
(FITC) and CD4 (AF700). CD8+T cells were identied by labelling with
antibodies against CD3 (FITC) and CD8 (PE-Cy5). B cells were identied by
labelling with antibodies against CD19 (BV421). Data were acquired using
an LSR-II Flow Cytometer, and populations were gated based on isotype
controls using FlowJo software. For gut sample preparation, the ileum and
colon were collected into HBSS 10% FBS following removal of all excess fat
and Peyers patches and immediately digested following established pro-
tocol in our group77.Briey, tissue was washed with PBS and cut into small
pieces (around 1 cm), rinsed with warm HBSS and transferred to wash
buffer (HBSS no Ca++,Mg
++ and containing 2 mM EDTA) for 15 min
(37 °C, 220 rpm). Ileum samples were digested with 0.5 mg/mL of Col-
lagenase IV (15 min, 37 °C) and the colon was digested with 0.5 mg/mL of
Collagenase IV containing 24 μg/mL DNase I (20 min, 37 °C) to generate
single cell suspensions prior to resuspending for antibody staining and ow
cytometric analysis.
RT-PCR
Gut tissues were dissolved in Trizol Reagent (1 ml/50100 mg of tissue)
following tissue homogenisation, and the lysates were centrifuged for 5 min
at 12,000 rpm at 4 °C. The supernatants were transferred to a fresh tube, and
chloroform (0.2 ml/mL of Trizol) was added to the samples for 5 minutes of
incubation followed by centrifugation (15 minutes at 12,000gC).RNAin
the aqueous phase was precipitated by adding 100% EtOH. Isolated RNA
was cleaned using the RNeasy Plus Mini kit (Qiagen, Germany) according to
the manufacturers instructions. The High-Capacity cDNA Reverse Tran-
scriptase kit (Applied Biosystems, Life Technology, UK) was used to gen-
erate cDNA. qPCR reactions were conducted using StepOne PlusTM real-
time PCR system (Applied Biosystems, UK) and KiCqStart®qPCR Ready
Mix (Sigma-Aldrich). Taqman probes were used to evaluate gene expres-
sion, including fucosyltransferases (FUT1, FUT2, FUT4, FUT7, FUT8 and
FUT9). Data were normalised to the reference gene β-actin to obtain ΔCT
values.
Microbial diversity sequencing and analysis
Genomic DNA from the ileum and colon faecal matter was puried using
QIAamp DNA Stool Mini Kit (Qiagen, Germany) and stored at 20 °C. All
samples were subjected to paired-end sequencing by Novogene (UK)
Company Limited, according to established protocols. Briey, specic16S
target hypervariable regions (amplicons) were amplied by PCR. Amplied
DNA fragments were end-repaired, and A-tailed. The sequencing adapters
were ligated to the ends of the DNA fragments using a DNA-binding
enzyme, and the DNA fragments were puried using AMpure PB magnetic
beads to construct an SMRTbell library. Finally, the sequencing primer was
annealed to the SMRTbell templates, followed by the binding of the
sequence polymerase to the annealed template. The library was checked
with Qubit for quantication. Quantied libraries were pooled and
sequenced on PacBio Sequel II/IIe systems. For sequencing data processing,
the PacBio BAM le was split according to barcode and ltered to get clean
data, used to generate Amplicon Sequence Variants (ASVs). Denoising,
species annotation and alpha diversity indices (including observed_ otus,
shannon, simpson, chao1, goods_ coverage, dominance and pielou_e
indices), were performed by the DADA2 and Qiime2 software. Sequences
with less than 5 abundance were ltered out to obtain the nal ASVs and
feature table. Then, the Classify-sklearn moduler in QIIME2 software was
used to compare ASVs with the database and to obtain the species anno-
tation of each ASV.
Statistics
All data were analysed using one-way ANOVA with FishersLSDpost-tests
for parametric data or KruskalWallis test and Dunns post-test for non-
parametric data (GraphPad Prism software). ShapiroWilk test was used to
assess normality.
Reporting summary
Further information on research design is available in the Nature Portfolio
Reporting Summary linked to this article.
Fig. 9 | Analysis of the microbial composition in the colon. Faecal matter was taken
from the colon of naïve mice (n= 3), severe arthritis (CIA high, n= 3, disease scores
11, 10 and 7), mild arthritis (CIA low, n= 3; disease scores 3, 4 and 4), symptomatic
OIT mice (OIT high, n= 3; disease scores 5, 6 and 4) and asymptomatic OIT mice
(OIT low, n= 3) at day 33, when DNA was isolated and subjected to 16 S ribosomal
RNA (rRNA) amplication and sequencing. aAlpha Diversity analysis, including
observed_ otus, shannon, simpson, chao1, dominance and pielou_e indices. Each
dot represents one individual mouse; graphs show the mean ± SEM. KruskalWallis
test was used to analyze whether the differences in species diversity between groups
were signicant, *p< 0.05. bBeta diversity indices heatmap of unweighted unifrac
distance Matrix. The size and colour of the circle in the square represent the dif-
ferences in coefcient between the two samples. The larger the circle is, the darker the
corresponding colour is, indicating that the differences between the two samples are
greater. cRelative abundance of the indicated phyla. Each column shows data from
individual mice. Data are also presented as pie charts, presenting proportion values
for each group as means. dClustering of species abundance. The top 35 genera in
abundance were clustered from the species and sample levels according to their
abundance information in each sample. Heatmap in grey scale shows the mean value
of all mice in the group for Zvalue of taxonomic relative abundance after standar-
disation. The coloured heatmap represents the values for individual mice: the x-axis
represents the sample name, and the y-axis represents the function annotation. The
cluster tree on the left side is the species cluster tree. eRelative abundance of the
indicated genera. Data show mean ± SEM and dots represent individual mice.
https://doi.org/10.1038/s42003-024-06476-z Article
Communications Biology | (2024) 7:804 16
Content courtesy of Springer Nature, terms of use apply. Rights reserved
Data availability
Metagenomic data have been deposited in NCBIs Gene Expression
Omnibus SRA under the Bioproject accession code PRJNA1117859. The
source data behind the graphs in the paper can be found in Supplementary
Data 1. All other data are available in the article and Supplementary les or
from the corresponding authors upon reasonable request. Numerical source
data for all graphs in the manuscript can be found in Supplementary
Data 1 File.
Received: 9 June 2023; Accepted: 20 June 2024;
References
1. Figus, F. A., Piga, M., Azzolin, I., McConnell, R. & Iagnocco, A.
Rheumatoid arthritis: extra-articular manifestations and
comorbidities. Autoimmun. Rev. 20, 102776 (2021).
2. Smolen, J. S. et al. Rheumatoid arthritis. Nat. Rev. Dis. Prim. 4,
18001 (2018).
3. Sari, S. et al. Global, regional and national burden of rheumatoid
arthritis 1990-2017: a systematic analysis of the Global Burden of
Disease study 2017. Ann. Rheum. Dis. 78, 14631471 (2019).
4. Holers, V. M. Autoimmunity to citrullinated proteins and the initiation
of rheumatoid arthritis. Curr. Opin. Immunol. 25, 728735 (2013).
5. Muller, S. & Radic, M. Citrullinated autoantigens: from diagnostic
markers to pathogenetic mechanisms. Clin. Rev. Allergy Immunol. 49,
232239 (2015).
6. Cook, A. D., Rowley, M. J., Mackay, I. R., Gough, A. & Emery, P.
Antibodies to type II collagen in early rheumatoid arthritis. Correlation
with disease progression. Arthritis Rheum. 39, 17201727 (1996).
7. Williams, R. O. Collagen-induced arthritis as a model for rheumatoid
arthritis. Methods Mol. Med. 98, 207216 (2004).
8. Thompson, H. S., Harper, N., Bevan, D. J. & Staines, N. A.
Suppression of collagen induced arthritis by oral administration of
type II collagen: changes in immune and arthritic responses mediated
by active peripheral suppression. Autoimmunity 16, 189199 (1993).
9. Kim, W. U. et al. Suppression of collagen-induced arthritis by single
administration of poly(lactic-co-glycolic acid) nanoparticles
entrapping type II collagen: a novel treatment strategy for induction of
oral tolerance. Arthritis Rheum. 46, 11091120 (2002).
10. Ding, C. H. et al. Oral administration of type II collagen suppresses
pro-inammatory mediator production by synoviocytes in rats with
adjuvant arthritis. Clin. Exp. Immunol. 132, 416423 (2003).
11. Kim, G. Y. et al. Oral administration of proteoglycan isolated from
Phellinus linteus in the prevention and treatment of collagen-induced
arthritis in mice. Biol. Pharm. Bull. 26, 823831 (2003).
12. Garcia, G., Komagata, Y., Slavin, A. J., Maron, R. & Weiner, H. L.
Suppression of collagen-induced arthritis by oral or nasal
administration of type II collagen. J. Autoimmun. 13, 315324 (1999).
13. Ju, J. H. et al. Oral administration of type-II collagen suppresses IL-17-
associated RANKL expression of CD4+T cells in collagen-induced
arthritis. Immunol. Lett. 117,1625 (2008).
14. Lugo, J. P. et al. Undenatured type II collagen (UC-II(R)) for joint
support: a randomized, double-blind, placebo-controlled study in
healthy volunteers. J. Int. Soc. Sports Nutr. 10, 48 (2013).
15. Bagchi, D. et al. Effects of orally administered undenatured type II
collagen against arthritic inammatory diseases: a mechanistic
exploration. Int J. Clin. Pharm. Res. 22, 101110 (2002).
16. Bann, J. G., Peyton, D. H. & Bachinger, H. P. Sweet is stable:
glycosylation stabilizes collagen. FEBS Lett. 473, 237240 (2000).
17. Corthay, A. et al. Epitope glycosylation plays a critical role for T cell
recognition of type II collagen in collagen-induced arthritis. Eur. J.
Immunol. 28, 25802590 (1998).
18. Bagi, C. M., Berryman, E. R., Teo, S. & Lane, N. E. Oral administration
of undenatured native chicken type II collagen (UC-II) diminished
deterioration of articular cartilage in a rat model of osteoarthritis (OA).
Osteoarthr. Cartil. 25, 20802090 (2017).
19. Lugo, J. P., Saiyed, Z. M. & Lane, N. E. Efcacy and tolerability of an
undenatured type II collagen supplement in modulating knee
osteoarthritis symptoms: a multicenter randomized, double-blind,
placebo-controlled study. Nutr. J. 15, 14 (2016).
20. Rui, F. et al. Undenatured type II collagen prevents and treats
osteoarthritis and motor function degradation in T2DM patients and
db/db mice. Food Funct. 12, 43734391 (2021).
21. Stabile, M. et al. 1)H-NMR metabolomic prole of healthy and
osteoarthritic canine synovial uid before and after UC-II
supplementation. Sci. Rep 12, 19716 (2002).
22. Orhan, C. et al. Undenaturedtype II collagenameliorates inammatory
responses and articular cartilage damage in the rat model of
osteoarthritis. Front. Vet. Sci. 8, 617789 (2021).
23. Varney, J. L., Fowler, J. W. & Coon, C. N. Undenatured type II collagen
mitigates inammation and cartilage degeneration in healthy
Labrador Retrievers during an exercise regimen. Transl. Anim. Sci. 5,
txab084 (2021).
24. Doonan, J. et al. The parasitic worm product ES-62 normalises the gut
microbiota bone marrow axis in inammatory arthritis. Nat. Commun.
10, 1554 (2019).
25. Matei, D. E. et al. Intestinal barrier dysfunction plays an integral role in
arthritis pathology and can be targeted to ameliorate disease.
Medicines 2, 864883 e869 (2021).
26. Holers, V. M. et al. Rheumatoid arthritis and the mucosal origins
hypothesis: protection turns to destruction. Nat. Rev. Rheumatol. 14,
542557 (2018).
27. Brand, D. D. et al. Autoantibodies to murine type II collagen in
collagen-induced arthritis: a comparison of susceptible and
nonsusceptible strains. J. Immunol. 157, 51785184 (1996).
28. Deaglio, S. et al. Adenosine generation catalyzed by CD39 and CD73
expressed on regulatory T cells mediates immune suppression. J.
Exp. Med. 204, 12571265 (2007).
29. Pineda, M. A. et al. The parasitic helminth product ES-62 suppresses
pathogenesis in collagen-induced arthritis by targeting the
interleukin-17-producing cellular network at multiple sites. Arthritis
Rheum. 64, 31683178 (2012).
30. Harnett, M. M., Harnett, W. & Pineda, M. A. The parasitic worm
product ES-62 up-regulates IL-22 production by gammadelta T cells
in the murine model of Collagen-Induced Arthritis. Inamm. Cell
Signal. https://doi.org/10.14800/ics.308 (2014).
31. Pineda, M. A., Rodgers, D. T., Al-Riyami, L., Harnett, W. & Harnett, M.
M. ES-62 protects against collagen-induced arthritis by resetting
interleukin-22 toward resolution of inammation in the joints. Arthritis
Rheumatol. 66, 14921503 (2014).
32. Kim, K. W. et al. Interleukin-22 promotes osteoclastogenesis in
rheumatoid arthritis through induction of RANKL in human synovial
broblasts. Arthritis Rheum. 64, 10151023 (2012).
33. Geboes, L. et al. Proinammatory role of the Th17 cytokine
interleukin-22 in collagen-induced arthritis in C57BL/6 mice. Arthritis
Rheum. 60, 390395 (2009).
34. Lindemans, C. A. et al. Interleukin-22 promotes intestinal-stem-cell-
mediated epithelial regeneration. Nature 528, 560564 (2015).
35. Zhang, X. et al. Interleukin22 regulates the homeostasis of the
intestinal epithelium during inammation. Int. J. Mol. Med. 43,
16571668 (2019).
36. Patnaude, L. et al. Mechanisms and regulation of IL-22-mediated
intestinal epithelial homeostasis and repair.Life Sci. 271, 119195 (2021).
37. Sugimoto, K. et al. IL-22 ameliorates intestinal inammation in a
mouse model of ulcerative colitis. J. Clin. Invest. 118, 534544 (2008).
38. Pham, T. A. et al. Epithelial IL-22RA1-mediated fucosylation promotes
intestinal colonization resistance to an opportunistic pathogen. Cell
Host Microbe 16, 504516 (2014).
https://doi.org/10.1038/s42003-024-06476-z Article
Communications Biology | (2024) 7:804 17
Content courtesy of Springer Nature, terms of use apply. Rights reserved
39. Magalhaes, A. et al. Fut2-null mice display an altered glycosylation
prole and impaired BabA-mediated Helicobacter pylori adhesion to
gastric mucosa. Glycobiology 19, 15251536 (2009).
40. Goto, Y. et al. Innate lymphoid cells regulate intestinal epithelial cell
glycosylation. Science 345, 1254009 (2014).
41. Nagao-Kitamoto, H. et al. Interleukin-22-mediated host glycosylation
prevents Clostridioides difcile infection by modulating the metabolic
activity of the gut microbiota. Nat. Med. 26, 608617 (2020).
42. Wang, Y. et al. Gut dysbiosis in rheumatic diseases: a systematic
review and meta-analysis of 92 observational studies. EBioMedicine
80, 104055 (2022).
43. Xu, X. et al. The bridge of the gut-joint axis: gut microbial metabolites
in rheumatoid arthritis. Front. Immunol. 13, 1007610 (2022).
44. Marinova-Mutafchieva, L., Gabay, C., Funa, K. & Williams, R. O.
Remission of collagen-induced arthritis is associated with high levels
of transforming growth factor-beta expression in the joint. Clin. Exp.
Immunol. 146, 287293 (2006).
45. Wang, Z. et al. Mice with dysfunctional TGF-beta signaling develop
altered intestinal microbiome and colorectal cancer resistant to 5FU.
Biochim. Biophys. Acta Mol. Basis Dis. 1867, 166179 (2021).
46. Douglas, G. M. et al. PICRUSt2 for prediction of metagenome
functions. Nat. Biotechnol. 38, 685688 (2020).
47. Schon, C. et al. UC-II undenatured type II collagen for knee joint
exibility: a multicenter, randomized, double-blind, placebo-
controlled clinical study. J. Integr. Complement. Med. 28, 540548
(2022).
48. Lerman, R. H., Chang, J. L., Konda, V., Desai, A. & Montalto, M. B.
Nutritional approach for relief of joint discomfort: a 12-week, open-
case series and illustrative case report. Integr. Med. 14,5261 (2015).
49. Sahin, E. et al. The effect of oral administration of undenatured type II
collagen on monosodium iodoacetate-induced osteoarthritis in
young and old rats. Sci. Rep. 13, 6499 (2023).
50. Pickert, G. et al. STAT3 links IL-22 signaling in intestinal epithelial cells
to mucosal wound healing. J. Exp. Med. 206, 14651472 (2009).
51. Duncan, S. H., Hold, G. L., Barcenilla, A., Stewart, C. S. & Flint, H. J.
Roseburia intestinalis sp. nov., a novel saccharolytic, butyrate-
producing bacterium from human faeces. Int. J. Syst. Evol. Microbiol.
52, 16151620 (2002).
52. Hold, G. L., Schwiertz, A., Aminov, R. I., Blaut, M. & Flint, H. J.
Oligonucleotide probes that detect quantitatively signicant groups of
butyrate-producing bacteria in human feces. Appl. Environ. Microbiol.
69, 43204324 (2003).
53. Zhu, C. et al. Roseburia intestinalis inhibits interleukin17 excretion
and promotes regulatory T cells differentiation in colitis. Mol. Med.
Rep. 17, 75677574 (2018).
54. Hoffmann, T. W. et al. Microorganisms linked to inammatory bowel
disease-associated dysbiosis differentially impact host physiology in
gnotobiotic mice. ISME J. 10, 460477 (2016).
55. Xie, J. et al. Short-chain fatty acids produced by ruminococcaceae
mediate alpha-linolenic acid promote intestinal stem cells
proliferation. Mol. Nutr. Food Res. 66, e2100408 (2022).
56. Jubair, W. K. et al. Modulation of inammatory arthritis in mice by gut
microbiota through mucosal inammation and autoantibody
generation. Arthritis Rheumatol. 70, 12201233 (2018).
57. He, J. et al. Intestinal butyrate-metabolizing species contribute to
autoantibody production and bone erosion in rheumatoid arthritis.
Sci. Adv. 8, eabm1511 (2022).
58. Takahashi, D. et al. Microbiota-derived butyrate limits the
autoimmune response by promoting the differentiation of follicular
regulatory T cells. EBioMedicine 58, 102913 (2020).
59. Tong, T. et al. Chicken type II collagen induced immune balance of
main subtype of helper T cells in mesenteric lymph node lymphocytes
in rats with collagen-induced arthritis. Inamm. Res. 59, 369377
(2010).
60. Weiner, H. L., da Cunha, A. P., Quintana, F. & Wu, H. Oral tolerance.
Immunol. Rev. 241, 241259 (2011).
61. Cosovanu, C. & Neumann, C. The many functions of Foxp3(+)
regulatory T cells in the intestine. Front. Immunol. 11, 600973 (2020).
62. Kotake, S. et al. IL-17 in synovial uids from patients with rheumatoid
arthritis is a potent stimulator of osteoclastogenesis. J. Clin. Invest.
103, 13451352 (1999).
63. Justa, S., Zhou, X. & Sarkar, S. Endogenous IL-22 plays a dual role in
arthritis: regulation of established arthritis via IFN-gamma responses.
PLoS ONE 9, e93279 (2014).
64. Zaiss, M. M., Joyce Wu, H. J., Mauro, D., Schett, G. & Ciccia, F.
The gut-joint axis in rheumatoid arthritis. Nat. Rev. Rheumatol. 17,
224237 (2021).
65. OConnor, W. Jr., Zenewicz, L. A. & Flavell, R. A. The dual nature of
T(H)17 cells: shifting the focus to function. Nat. Immunol. 11, 471476
(2010).
66. Zheng, Y. et al. Interleukin-22 mediates early host defense against
attaching and effacing bacterial pathogens. Nat. Med. 14, 282289
(2008).
67. Pickard, J. M. & Chervonsky, A. V. Intestinal fucose as a mediator of
host-microbe symbiosis. J. Immunol. 194, 55885593 (2015).
68. Furusawa, Y. et al. Commensal microbe-derived butyrate induces
the differentiation of colonic regulatory T cells. Nature 504, 446450
(2013).
69. Haghikia, A. et al. Dietary fatty acids directly impact central nervous
system autoimmunity via the small intestine. Immunity 43, 817829
(2015).
70. Teng, F. et al. Gut microbiota drive autoimmune arthritis by promoting
differentiation and migration of Peyers patch T follicular helper cells.
Immunity 44, 875888 (2016).
71. Wu, H. J. et al. Gut-residing segmented lamentous bacteria drive
autoimmune arthritis via T helper 17 cells. Immunity 32, 815827
(2010).
72. Umesaki, Y., Okada, Y., Matsumoto, S., Imaoka, A. & Setoyama, H.
Segmented lamentous bacteria are indigenous intestinal bacteria
that activate intraepithelial lymphocytes and induce MHC class II
molecules and fucosyl asialo GM1 glycolipids on the small intestinal
epithelial cells in the ex-germ-free mouse. Microbiol. Immunol. 39,
555562 (1995).
73. Ivanov, I. I. et al. Induction of intestinal Th17 cells by segmented
lamentous bacteria. Cell 139, 485498 (2009).
74. Dennis, G. Jr. et al. Synovial phenotypes in rheumatoid arthritis
correlate with response to biologic therapeutics. Arthritis Res. Ther.
16, R90 (2014).
75. Pereira, R. S. et al. Disappearing collagen antibodies in rheumatoid
arthritis. Lancet 2, 501502 (1985).
76. Manivel, V. A. et al. Anticollagen type II antibodies are associated with
an acute onset rheumatoid arthritis phenotype and prognosticate
lower degree of inammation during 5 years follow-up. Ann. Rheum.
Dis. 76, 15291536 (2017).
77. Webster, H. C., Andrusaite, A. T., Shergold, A. L., Milling, S. W. F. &
Perona-Wright, G. Isolation and functional characterisation of lamina
propria leukocytes from helminth-infected, murine small intestine. J.
Immunol. Methods 477, 112702 (2020).
Acknowledgements
This work was funded by an Industrial partnership PhD between the
Universityof Glasgow and Lonzaand a Versus Arthritis Career Development
Fellowship Award grant 21221.
Author contributions
P.P., M.M.H., S.M. and M.P. conceivedand designedthe study. P.P., Y.W.,
M.H.N. and M.P. performed the lab work. P.P., Z.S., E.K.A., A.S., S.M.,
M.M.H. andM.P. contributed to data interpretation and manuscript revision.
https://doi.org/10.1038/s42003-024-06476-z Article
Communications Biology | (2024) 7:804 18
Content courtesy of Springer Nature, terms of use apply. Rights reserved
P.P., S.M., M.M.H and M.P. wrote the paper. All authors read and approved
the submitted version.
Competing interests
Z.S., E.K.A.and A.S. are employeesof Lonza GreenwoodLLC., Greenwood,
SC, USA. The remaining authors declare no competing interests.
Additional information
Supplementary information The online version contains
supplementary material available at
https://doi.org/10.1038/s42003-024-06476-z.
Correspondence and requests for materials should be addressed to
Miguel Pineda.
Peer review information Communications Biology thanksZhihua Liu, Julie
Gibbs and the other, anonymous, reviewer for their contribution to the peer
review of this work. Primary Handling Editors: Martina Rauner and Joao
Valente. A peer review le is available.
Reprints and permissions information is available at
http://www.nature.com/reprints
Publishers note Springer Nature remains neutral with regard to
jurisdictional claims in published maps and institutional afliations.
Open Access This article is licensed under a Creative Commons
Attribution 4.0 International License, which permits use, sharing,
adaptation, distribution and reproduction in any medium or format, as long
as you give appropriate credit to the original author(s) and the source,
provide a link to the Creative Commons licence, and indicate if changes
were made. The images or other third party material in this article are
included in the articles Creative Commons licence, unless indicated
otherwise in a credit line to the material. If material is not included in the
articles Creative Commons licence and your intended use is not permitted
by statutory regulation or exceeds the permitted use, you will need to
obtain permission directly from the copyright holder. To view a copy of this
licence, visit http://creativecommons.org/licenses/by/4.0/.
© The Author(s) 2024
https://doi.org/10.1038/s42003-024-06476-z Article
Communications Biology | (2024) 7:804 19
Content courtesy of Springer Nature, terms of use apply. Rights reserved
1.
2.
3.
4.
5.
6.
Terms and Conditions
Springer Nature journal content, brought to you courtesy of Springer Nature Customer Service Center GmbH (“Springer Nature”).
Springer Nature supports a reasonable amount of sharing of research papers by authors, subscribers and authorised users (“Users”), for small-
scale personal, non-commercial use provided that all copyright, trade and service marks and other proprietary notices are maintained. By
accessing, sharing, receiving or otherwise using the Springer Nature journal content you agree to these terms of use (“Terms”). For these
purposes, Springer Nature considers academic use (by researchers and students) to be non-commercial.
These Terms are supplementary and will apply in addition to any applicable website terms and conditions, a relevant site licence or a personal
subscription. These Terms will prevail over any conflict or ambiguity with regards to the relevant terms, a site licence or a personal subscription
(to the extent of the conflict or ambiguity only). For Creative Commons-licensed articles, the terms of the Creative Commons license used will
apply.
We collect and use personal data to provide access to the Springer Nature journal content. We may also use these personal data internally within
ResearchGate and Springer Nature and as agreed share it, in an anonymised way, for purposes of tracking, analysis and reporting. We will not
otherwise disclose your personal data outside the ResearchGate or the Springer Nature group of companies unless we have your permission as
detailed in the Privacy Policy.
While Users may use the Springer Nature journal content for small scale, personal non-commercial use, it is important to note that Users may
not:
use such content for the purpose of providing other users with access on a regular or large scale basis or as a means to circumvent access
control;
use such content where to do so would be considered a criminal or statutory offence in any jurisdiction, or gives rise to civil liability, or is
otherwise unlawful;
falsely or misleadingly imply or suggest endorsement, approval , sponsorship, or association unless explicitly agreed to by Springer Nature in
writing;
use bots or other automated methods to access the content or redirect messages
override any security feature or exclusionary protocol; or
share the content in order to create substitute for Springer Nature products or services or a systematic database of Springer Nature journal
content.
In line with the restriction against commercial use, Springer Nature does not permit the creation of a product or service that creates revenue,
royalties, rent or income from our content or its inclusion as part of a paid for service or for other commercial gain. Springer Nature journal
content cannot be used for inter-library loans and librarians may not upload Springer Nature journal content on a large scale into their, or any
other, institutional repository.
These terms of use are reviewed regularly and may be amended at any time. Springer Nature is not obligated to publish any information or
content on this website and may remove it or features or functionality at our sole discretion, at any time with or without notice. Springer Nature
may revoke this licence to you at any time and remove access to any copies of the Springer Nature journal content which have been saved.
To the fullest extent permitted by law, Springer Nature makes no warranties, representations or guarantees to Users, either express or implied
with respect to the Springer nature journal content and all parties disclaim and waive any implied warranties or warranties imposed by law,
including merchantability or fitness for any particular purpose.
Please note that these rights do not automatically extend to content, data or other material published by Springer Nature that may be licensed
from third parties.
If you would like to use or distribute our Springer Nature journal content to a wider audience or on a regular basis or in any other manner not
expressly permitted by these Terms, please contact Springer Nature at
onlineservice@springernature.com
Article
Full-text available
Correction for 'Undenatured type II collagen prevents and treats osteoarthritis and motor function degradation in T2DM patients and db/db mice' by Fan Rui et al., Food Funct., 2021, 12, 4373-4391, https://doi.org/10.1039/D0FO03011B.
Article
Full-text available
We investigated whether different doses of undenatured type II collagen (undenatured collagen, UC-II) help improve monosodium iodoacetate (MIA)-induced (osteoarthritis) OA in young and old rats. A total of 70 rats were divided into five groups: (1) control; (2) MIA (a single intra-articular injection of MIA); (3)–(5) MIA+ Undenatured Collagen with various oral doses (0.66, 1.33, and 2 mg/kg). The results showed that all doses of undenatured collagen in both age groups reduced knee diameter, while the two higher doses (1.33 mg/kg and 2 mg/kg) reduced the Mankin score and increased most gait measurements as early as day 14 compared to the MIA rats. However, the 2 mg/kg dose showed the best efficacy in improving Mankin score and gait measurements by 28 days post-OA induction. In young but not old rats, all doses of undenatured collagen reduced the Kellgren-Lawrence score compared to the MIA group. Undenatured collagen reduced the levels of most inflammatory and cartilage breakdown markers in serum and knee joint cartilage in both age groups. In conclusion, this data suggests that while all doses of undenatured collagen supplementation may ameliorate MIA-induced OA symptoms, the higher doses showed faster improvement in gait measurements and were more efficacious for overall joint health in rats.
Article
Full-text available
The aim of the study was to compare the metabolomic synovial fluid (SF) profile of dogs affected by spontaneous osteoarthritis (OA) and supplemented with undenatured type II collagen (UC-II), with that of healthy control dogs. Client-owned dogs were enrolled in the study and randomized in two different groups, based on the presence/absence of OA (OA group and OA-free group). All dogs were clinically evaluated and underwent SF sampling for ¹H-Nuclear Magnetic Resonance spectroscopy (¹H-NMR) analysis at time of presentation. All dogs included in OA group were supplemented with UC-II orally administered for 30 days. After this period, they were reassessed (OA-T30). The differences in the ¹H-NMR metabolic SFs profiles between groups (OA-free, OA-T0 and OA-T30) were studied. The multivariate statistical analysis performed on SFs under different conditions (OA-T0 vs OA-T30 SFs; OA-T0 vs OA-free SFs and OA-T30 vs OA-free SFs) gave models with excellent goodness of fit and predictive parameters, revealed by a marked separation between groups. β-Hydroxybutyrate was identified as a characteristic compound of osteoarthritic joints, showing the important role of fat metabolism during OA. The absence of β-hydroxybutyrate after UC-II supplementation suggests the supplement’s effectiveness in rebalancing the metabolism inside the joint. The unexpectedly high level of lactate in the OA-free group suggests that lactate could not be considered a good marker for OA. These results prove that ¹H-NMR-based metabolomic analysis is a valid tool to study and monitor OA and that UC-II improves clinical symptoms and the SF metabolic profile in OA dogs.
Article
Full-text available
Rheumatoid arthritis (RA) is an autoimmune disease characterized by joint destruction, synovitis, and pannus formation. Gut microbiota dysbiosis may exert direct pathogenic effects on gut homeostasis. It may trigger the host’s innate immune system and activate the “gut–joint axis”, which exacerbates the RA. However, although the importance of the gut microbiota in the development and progression of RA is widely recognized, the mechanisms regulating the interactions between the gut microbiota and the host immune system remain incompletely defined. In this review, we discuss the role of gut microbiota-derived biological mediators, such as short-chain fatty acids, bile acids, and tryptophan metabolites, in maintaining intestinal barrier integrity, immune balance and bone destruction in RA patients as the bridge of the gut–joint axis.
Article
Full-text available
Background Emerging evidence suggests that dysbiosis in gut microbiota may contribute to the occurrence or development of several rheumatic diseases. Since gut microbiota dysbiosis is potentially modifiable, it has been postulated to be a promising preventive or therapeutic target for rheumatic diseases. However, the current understanding on the potential associations between gut microbiota and rheumatic diseases is still inadequate. Therefore, we aimed to synthesise the accumulating evidence for the relation of gut microbiota to rheumatic diseases. Methods The PubMed, Embase and Cochrane Library were searched from inception to March 11, 2022 to include observational studies evaluating the associations between gut microbiota and rheumatic diseases. Standardised mean difference (SMD) of α-diversity indices between rheumatic diseases and controls were estimated using random-effects model. β-diversity indices and relative abundance of gut microbes were summarised qualitatively. Findings Of the included 92 studies (11,998 participants), 68 provided data for α-diversity. Taken together as a whole, decreases in α-diversity indices were consistently found in rheumatic diseases (observed species: SMD = −0.36, [95%CI = −0.63, −0.09]; Chao1: SMD = −0.57, [95%CI = −0.88, −0.26]; Shannon index: SMD = −0.33, [95%CI = −0.48, −0.17]; Simpson index: SMD = −0.32, [95%CI = −0.49, −0.14]). However, when specific rheumatic diseases were examined, decreases were only observed in rheumatoid arthritis (observed species: SMD = −0.51, [95%CI = −0.78, −0.24]; Shannon index: SMD = −0.31, [95%CI = −0.49, −0.13]; Simpson index: SMD = −0.31, [95%CI = −0.54, −0.08]), systemic lupus erythematosus (Chao1: SMD = −1.60, [95%CI = −2.54, −0.66]; Shannon index: SMD = −0.63, [95%CI = −1.08, −0.18]), gout (Simpson index: SMD = −0.64, [95%CI = −1.07, −0.22]) and fibromyalgia (Simpson index: SMD = −0.28, [95%CI = −0.44, −0.11]), whereas an increase was observed in systemic sclerosis (Shannon index: SMD = 1.25, [95%CI = 0.09, 2.41]). Differences with statistical significance in β-diversity were consistently reported in ankylosing spondylitis and IgG4-related diseases. Although little evidence of disease specificity of gut microbes was found, shared alterations of the depletion of anti-inflammatory butyrate-producing microbe (i.e., Faecalibacterium) and the enrichment of pro-inflammatory microbe (i.e., Streptococcus) were observed in rheumatoid arthritis, Sjögren's syndrome and systemic lupus erythematosus. Interpretation Gut microbiota dysbiosis was associated with rheumatic diseases, principally with potentially non-specific, shared alterations of microbes. Funding National Natural Science Foundation of China (81930071, 81902265, 82072502 and U21A20352).
Article
Full-text available
Objective Joint-related stress models have been used in the past to induce a standardized load on physical structures, allowing researchers to observe changes in perceived stress on joints as accurately as possible in healthy individuals. Previous studies support the efficacy of UC-II® undenatured type II collagen (“undenatured collagen”) supplementation in maintaining joint health. The purpose of this study was to assess the effect of undenatured collagen on knee flexibility in healthy subjects who experience activity-related joint discomfort (ArJD). Methods This randomized, double-blind, placebo (PLA)-controlled study was conducted in healthy subjects with ArJD who had no history of osteoarthritis, or joint diseases. Ninety-six (n = 96, 20–55 years old) subjects who reported joint discomfort while performing a standardized single-leg-step-down test were randomized to receive either PLA (n = 48) or 40 mg of undenatured collagen (n = 48) supplementation daily for 24 weeks. Range of motion (ROM) flexion and extension were measured using a digital goniometer. Results At the end of the study, a statistically significant increase in knee ROM flexion was observed in the undenatured collagen group versus the PLA group (3.23° vs. 0.21°; p = 0.025). In addition, an increase in knee ROM extension by 2.21° was observed over time in the undenatured collagen group (p = 0.0061), while the PLA group showed a nonsignificant increase by 1.27° (p > 0.05). Subgroup analysis by age showed a significant increase in knee ROM flexion in subjects >35 years old in the undenatured collagen supplemented group compared with PLA (6.79° vs. 0.30°; p = 0.0092). Conclusion Overall, these results suggest that daily supplementation of 40 mg of undenatured collagen improved knee joint ROM flexibility and extensibility in healthy subjects with ArJD.
Article
Full-text available
Emerging data show a rise in colorectal cancer (CRC) incidence in young men and women that is often chemoresistant. One potential risk factor is an alteration in the microbiome. Here, we investigated the role of TGF-β signaling on the intestinal microbiome and the efficacy of chemotherapy for CRC induced by azoxymethane and dextran sodium sulfate in mice. We used two genotypes of TGF-β-signaling-deficient mice (Smad4+/– and Smad4+/–/Sptbn1+/–), which developed CRC with similar phenotypes and had similar alterations in the intestinal microbiome. Using these mice, we evaluated the intestinal microbiome and determined the effect of dysfunctional TGF-β signaling on the response to the chemotherapeutic agent 5-Fluoro-uracil (5FU) after induction of CRC. Using shotgun metagenomic sequencing, we determined gut microbiota composition in mice with CRC and found reduced amounts of beneficial species of Bacteroides and Parabacteroides in the mutants compared to the WT mice. Furthermore, the mutant mice with CRC were resistant to 5FU. Whereas the abundances of E. boltae, B.dorei, Lachnoclostridium sp., and Mordavella sp. were significantly reduced in mice with CRC, these species only recovered to basal amounts after 5FU treatment in WT mice, suggesting that the alterations in the intestinal microbiome resulting from compromised TGF-β signaling impaired the response to 5FU. These findings could have implications for inhibiting the TGF-β pathway in the treatment of CRC or other cancers.
Article
Full-text available
Background: Evidence suggests an important role for gut-microbiota dysbiosis in the development of rheumatoid arthritis (RA). The link between changes in gut bacteria and the development of joint inflammation is missing. Here, we address whether there are changes to the gut environment and how they contribute to arthritis pathogenesis. Methods: We analyzed changes in markers of gut permeability, damage, and inflammation in peripheral blood and serum of RA patients. Serum, intestines, and lymphoid organs isolated from K/BxN mice with spontaneous arthritis or from wild-type, genetically modified interleukin (IL)-10R-/- or claudin-8 -/- mice with induced arthritis were analyzed by immunofluorescence/histology, ELISA, and flow cytometry. Findings: RA patients display increased levels of serum markers of gut permeability and damage and cellular gut-homing markers, both parameters positively correlating with disease severity. Arthritic mice display increased gut permeability from early stages of disease, as well as bacterial translocation, inflammatory gut damage, increases in interferon γ (IFNγ) + and decreases in IL-10 + intestinal-infiltrating leukocyte frequency, and reduced intestinal epithelial IL-10R expression. Mechanistically, both arthritogenic bacteria and leukocytes are required to disrupt gut-barrier integrity. We show that exposing intestinal organoids to IFNγ reduces IL-10R expression by epithelial cells and that mice lacking epithelial IL-10R display increased intestinal permeability and exacerbated arthritis. Claudin-8 -/- mice with constitutively increased gut permeability also develop worse joint disease. Treatment of mice with AT-1001, a molecule that prevents development of gut permeability, ameliorates arthritis. Conclusions: We suggest that breakdown of gut-barrier integrity contributes to arthritis development and propose restoration of gut-barrier homeostasis as a new therapeutic approach for RA. Funding: Funded by Versus Arthritis (21140 and 21257) and UKRI/MRC (MR/T000910/1).
Article
The imbalance between pathogenic and beneficial species of the intestinal microbiome and metabolism in rheumatoid arthritis (RA) remains unclarified. Here, using shotgun-based metagenome sequencing for a treatment-naïve patient cohort and a “quasi-paired cohort” method, we observed a deficiency of butyrate-producing species and an overwhelming number of butyrate consumers in RA patients. These outcomes mainly occurred in patients with positive ACPA, with a mean AUC of 0.94. This panel was also validated in established RA with an AUC of 0.986 in those with joint deformity. In addition, we showed that butyrate promoted T regs , while suppressing T convs and osteoclasts, due to potentiation of the reduction in HDAC expression and down-regulation of proinflammatory cytokine genes. Dietary butyrate supplementation conferred anti-inflammatory benefits in a mouse model by rebalancing T FH cells and T regs , as well as reducing antibody production. These findings reveal the critical role of butyrate-metabolizing species and suggest the potential of butyrate-based therapies for RA patients.
Article
Scope: The proliferation and differentiation of intestinal stem cells (ISCs) are the basis of intestinal renewal and regeneration, and gut microbiota plays an important role in it. Dietary nutrition has the effect of regulating the activity of ISCs, however, the regulation effect of α-linolenic acid (ALA) has seldom been reported. Methods and results: After intervening mice with different doses of ALA for 30 days, we found that ALA (0.5 g/kg) promoted small intestinal and villus growth by activating the Wnt/β-catenin signaling pathway to stimulate the proliferation of ISCs. Furthermore, ALA administration increased the abundance of the Ruminococcaceae and Prevotellaceae, and promoted the produce of short-chain fatty acids (SCFAs). Subsequent fecal transplantation and antibiotic experiments demonstrated that ALA on the proliferation of ISCs are gut microbiota dependent, among them the functional microorganism may be derived from Ruminococcaceae. Administration of isobutyrate showed a similar effect to ALA in terms of promoting ISCs proliferation. Furthermore, ALA mitigated 5-fluorouracil-induced intestinal mucosal damage by promoting ISCs proliferation. Conclusion: These results indicate that SCFAs produced by Ruminococcaceae mediates ALA promote ISCs proliferation by activating Wnt/β-catenin signaling pathway, and suggest the possibility of ALA as a prebiotic agent for the prevention and treatment of intestinal mucositis. This article is protected by copyright. All rights reserved