You are on page 1of 13

Biomedicine & Pharmacotherapy 170 (2024) 115958

Contents lists available at ScienceDirect

Biomedicine & Pharmacotherapy


journal homepage: www.elsevier.com/locate/biopha

Review

Targeting IL-12 family cytokines: A potential strategy for type 1 and type 2
diabetes mellitus
Jiayu Luo 1, Tingting Ning 1, Xing Li , Tao Jiang , Shenglong Tan , Dandan Ma *
Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong Province, China

A R T I C L E I N F O A B S T R A C T

Keywords: Diabetes is a common metabolic disease characterized by an imbalance in blood glucose levels. The pathogenesis
Diabetes mellitus of diabetes involves the essential role of cytokines, particularly the IL-12 family cytokines. These cytokines,
IL-12 family cytokines which have a similar structure, play multiple roles in regulating the immune response. Recent studies have
Therapeutic targets
emphasized the importance of IL-12 family cytokines in the development of both type 1 and type 2 diabetes
Pathogenesis
mellitus. As a result, they hold promise as potential therapeutic targets for the treatment of these conditions. This
Inflammation
Clinical application review focuses on the potential of targeting IL-12 family cytokines for diabetes therapy based on their roles in the
pathogenesis of both types of diabetes. We have summarized various therapies that target IL-12 family cytokines,
including drug therapy, combination therapy, cell therapy, gene therapy, cytokine engineering therapy, and gut
microbiota modulation. By analyzing the advantages and disadvantages of these therapies, we have evaluated
their feasibility for clinical application and proposed possible solutions to overcome any challenges. In conclu­
sion, targeting IL-12 family cytokines for diabetes therapy provides updated insights into their potential benefits,
such as controlling inflammation, preserving islet β cells, reversing the onset of diabetes, and impeding the
development of diabetic complications.

1. Introduction of IL-12 in both T1DM and T2DM. However, the involvement of other
members of the IL-12 family, such as IL-23, IL-27, and IL-35, in the
Diabetes mellitus is a chronic metabolic disease characterized pri­ pathogenesis of diabetes has not been fully reviewed. Unlike other
marily by hyperglycemia [1]. The prevalence of diabetes is expected to interleukin families, the IL-12 family of cytokines share common chains
reach 537 million worldwide in 2021, as reported by the International and receptors, suggesting that their functions may overlap. These unique
Diabetes Association [1]. The two most common types of diabetes are structures and compositions give the IL-12 family cytokines dual roles in
type 1 diabetes mellitus (T1DM) and type 2 diabetes mellitus (T2DM). promoting both pro- and anti-inflammation in diabetes. Therefore, it is
Despite considerable research, the mechanisms underlying T1DM and intriguing to investigate the functions and interactions of IL-12 family
T2DM are not yet fully understood. Various factors, including immune cytokines in diabetes, considering their compositions and structures.
disorders, contribute to the development of diabetes. Cytokines have Currently, the treatment of T1DM and T2DM primarily focuses on
been extensively studied and are known to play a significant role in the anti-hyperglycemic effects, such as insulin and sulfonylurea. However,
pathogenesis of both T1DM and T2DM. this approach often leads to uncontrolled inflammation in diabetes and
In previous discussions, there has been extensive research on the role subsequent complications, including delayed wound healing,

Abbreviations: T1DM, Type 1 diabetes mellitus; T2DM, Type 2 diabetes mellitus; CTL, cytotoxic T lymphocytes; DCs, dendritic cells; PBMCs, peripheral blood
mononuclear cells; EBI3, Epstein–Barr virus-induced gene 3; IRF-1, interferon regulatory factor 1; Th0, naive CD4 + T helper cells; Th1, mature CD4+T helper cells;
ILCs, innate lymphoid cells; TFH, follicular helper T cells; mDCs, myeloid dendritic cells; NF-kB, Nuclear factor kappa-light-chain-enhancer of activated B cells;
NAFLD, non-alcoholic fatty liver disease; TLR, Toll-Like Receptors; FDA, Food and Drug Administration; JAK-STAT, Janus kinase-signal transducers and activators of
transcription; INGAP, insulin-resistant protein; LSF-LA, a combination of Lisofylline and Linoleic-acid self-assembled into micelles; AIRE, autoimmune Regulators;
CAR-T, Chimeric antigen receptors Tregs; MHC, major histocompatibility complex; Bregs, regulatory B cells; Tregs, regulatory T cells; IL-23R, IL-23 receptors;
Synthekines, synthesize cytokines.
* Corresponding author.
E-mail address: mdd@smu.edu.cn (D. Ma).
1
These authors contributed equally to this work.

https://doi.org/10.1016/j.biopha.2023.115958
Received 31 August 2023; Received in revised form 20 November 2023; Accepted 27 November 2023
Available online 7 December 2023
0753-3322/© 2023 The Authors. Published by Elsevier Masson SAS. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/by-nc-nd/4.0/).
J. Luo et al. Biomedicine & Pharmacotherapy 170 (2024) 115958

retinopathy, neuropathy, and atherosclerosis. It has become increas­ 1.2.1. IL-12 family cytokines may serve as predictive indicators and reflect
ingly apparent that targeting cytokines involved in T1DM and T2DM is the severity of T1DM and T2DM
crucial in controlling inflammation. Unfortunately, the clinical appli­ IL-12 family cytokines appear to serve as warning signals in the body,
cation of cytokine targeting is limited due to low specificity. Therefore, potentially providing insights into the onset of both T1DM and T2DM.
it is necessary to assess the therapeutic potential and obstacles associ­ For instance, a decrease in circulating levels of IL-27 has been suggested
ated with targeting IL-12 family cytokines for diabetes therapy. as a potential predictor for T2DM. It is established that insulin resistance
In this review, we aim to explore the significant correlation between precedes the development of T2DM, and lower levels of circulating IL-27
IL-12 family cytokines and diabetes, focusing on their implications in the have been found in individuals with insulin resistance, indicating that
development of T1DM and T2DM. Additionally, we provide a compre­ IL-27 may serve as a predictive marker for T2DM [13]. This association
hensive overview of the current and potential therapeutic approaches may be attributed to the disrupted regulation of adipocyte thermogen­
targeting IL-12 family cytokines. These include drug therapy, gene esis by IL-27, which can result in adipose accumulation and the subse­
therapy, cell therapy, combination therapy, cytokine engineering ther­ quent development of T2DM [13].
apy, and modulation of gut microbiota. By examining the advantages Contrarily, elevated levels of circulating IL-27 have been linked to an
and drawbacks of each therapy, we identify the existing barriers to the increased risk of T1DM. A Two-Sample Mendelian Randomization Study
application of cytokines and propose potential solutions for future demonstrated that higher circulating protein levels of EBI3 (a subunit of
research. Overall, our objective is to offer valuable insights into the field IL-27) are associated with an enhanced risk of T1DM [14]. Furthermore,
of diabetes therapy. consistent evidence shows that T1DM patients exhibit significantly
higher levels of circulating IL-27 [15]. Mutation of IL-27 gene in T1DM
1.1. Immune disorders contribute to T1DM and T2DM contributes to immune disorders. Expression quantitative trait loci
(eQTL) analysis has revealed that mutations in the IL-27 gene
Type 1 diabetes mellitus (T1DM) is primarily an autoimmune disease (rs181206) result in increased expression of interferon regulatory factor
characterized by the involvement of CD4+ and CD8+ T cells, which play 1 (IRF-1) in CD4+T cells, promoting the production of IFN-γ in T1DM
a role in its pathogenesis [2]. In T1DM, the immune system mistakenly patients [16]. In summary, the levels of circulating IL-27 exhibit oppo­
identifies islet cells as foreign entities, leading to their attack by the site trends during the early stages of T1DM and T2DM, suggesting its
immune system [2]. This process is further exacerbated by the contin­ potential as a diagnostic marker for both conditions.
uous release of pro-inflammatory cytokines [3]. The complex cause of Circulating levels of IL-35 also hold predictive potential in the
immune disorders in T1DM may be closely associated with individual context of T1DM. It has been directly observed that T1DM patients with
susceptibility, with individuals carrying genetic variations in tumor remaining C-peptide exhibit higher levels of IL-35 [17]. The presence of
necrosis factor-alpha (TNF-α) being at a higher risk [4]. C-peptide is indicative of better functioning islet β cells, which suggests
Type 2 diabetes mellitus (T2DM) is characterized by insulin resis­ that the severity of T1DM could be reflected by measuring circulating
tance and reduced insulin levels [5]. Although the exact mechanisms levels of IL-35. Therefore, circulating IL-35 can serve as a valuable
underlying T2DM are still unclear, obesity is widely recognized as a marker for assessing the progression and severity of T1DM.
major risk factor. Obesity contributes to chronic low-grade inflamma­ Unlike IL-27 and IL-35, the levels of IL-23 in the circulation do not
tion by increasing the levels of pro-inflammatory cytokines such as show significant variations in individuals with T1DM or T2DM [18].
TNF-α and IL-6, while reducing the levels of protective factors like However, IL-23 plays a role in local inflammation of the islets. In both
adiponectin [6,7]. This inflammation-induced insulin resistance ulti­ T1DM and T2DM patients, there is an increased production of IL-23 by
mately leads to the development of T2DM [8]. To prevent the onset of peripheral blood mononuclear cells (PBMCs) surrounding the islets
T2DM, it is crucial to disrupt the association between obesity and insulin compared to healthy individuals [19,20]. Furthermore, an upregulation
resistance through anti-inflammatory therapies. of the IL-23 subunit, IL-23p19 (IL-23A), was observed in pancreatic is­
Both T1DM and T2DM are influenced by an excessive inflammatory lets from individuals with short-duration T1DM [21]. Mutations in the
response. This inflammatory response causes the activation of islet cells IL-23 gene have also been linked to the development of both T1DM and
in the pancreas through a mechanism involving the accumulation of pro- T2DM [22,23]. However, there is a debate on whether circulating levels
inflammatory cytokines, ultimately leading to their caspase-dependent of IL-23 also change. A study reported elevated levels of circulating
activation or even apoptosis [9]. Specifically, IL-12 family cytokines IL-23 in individuals with T2DM aged over 51 years old [18]. This finding
play a crucial role in exacerbating immune disorders in both T1DM and suggests that circulating levels of IL-23 may be associated with the
T2DM. progression of diabetes. This is consistent with the findings of Eiris et al.,
who reported that genetic variations in IL-23 were linked to the severity
1.2. Changes of IL-12 family cytokines in T1DM and T2DM of T2DM [23]. Although it is not recommended to use circulating IL-23
levels as an early-stage diagnostic tool for T1DM and T2DM, it could
IL-12 family cytokines, which include IL-12, IL-27, IL-23, IL-35, and serve as a reference for the progression of T2DM.
IL-39, play crucial roles in immunoregulation [10,11]. These cytokines It is well-known that levels of IL-12 are increased in patients with
share structural similarities and have overlapping functions. They are all both T1DM and T2DM [24,25]. Recent studies have focused on the
heterodimeric cytokines composed of two subunits. IL-12 and IL-23 relationship between circulating levels of IL-12 and diabetic complica­
share the same subunit, IL-12p40, while IL-27 and IL-35 share EBI3. tions, particularly cardiovascular diseases. For instance, in individuals
Besides, IL-35 and IL-12 share IL-12p35 as a subunit. These cytokines with T2DM, higher serum levels of IL-12 have been associated with
bind to specific receptors and activate JAK-STAT signaling pathways reduced heart rate variability [26]. Similarly, T1DM patients with car­
[12]. In terms of function, IL-12, IL-23, and IL-27 primarily promote diovascular autonomic neuropathy exhibit significantly elevated circu­
inflammation in both T1DM and T2DM. On the other hand, IL-35 and lating levels of IL-12 compared to healthy individuals [27]. Recent
IL-27 enable to counteract inflammation. It is important to note that research has suggested that varying levels of IL-12 can exacerbate
each member of the IL-12 family has distinct characteristics and con­ glucose variability, increasing the risk of diabetes complications [28].
tributes to various aspects of T1DM and T2DM. The role of IL-39 in these Other members of the IL-12 family of cytokines are also closely
conditions remains to be explored in future research. Therefore, the associated with diabetes complications. Elevated levels of IL-23 were
remainder of this discussion will focus on IL-12, IL-27, IL-23, and IL-35. found to be accompanied by impaired heart rate variability [26].
Additionally, a multiplex proteomics study highlighted the close asso­
ciation between IL-27p28 protein levels and major adverse cardiovas­
cular events in T2DM [29].

2
J. Luo et al. Biomedicine & Pharmacotherapy 170 (2024) 115958

IL-12 family cytokines have the potential to not only predict and which in turn promoted the development of T1DM [39]. However, the
reflect the severity of diabetes but also serve as suitable indicators for mechanism behind the activation of IL-27 on CD8+ T cells remains un­
estimating the risk of diabetes complications. clear. In contrast, Fujimoto et al. presented evidence supporting the role
of IL-27 in suppressing the infiltration of CD8+ T cells induced by
2. Roles of IL-12 family cytokines in the pathogenesis of T1DM streptozocin [40]. Above all, effect of IL-12 family cytokines on CD4+T
and T2DM and CD8+T cells are mainly discussed in the pathogenesis of T1DM.
Notably, similar progress might be observed in the pathogenesis of
2.1. Pro-inflammatory role of IL-12 family cytokines T2DM. This is due to the recent Mendelian randomization study that the
presence of T-lymphocyte including CD4+CD8dim and CD8+T cells
2.1.1. Direct effects of IL-12 family cytokines on islet β cells enhanced the risk of T2DM [41].
The cytokines belonging to the IL-12 family have been found to As the key regulators of immune responses, Regulatory T cells
worsen inflammation by directly triggering the apoptosis of islet β cells. (Tregs), known as FOXP3+T cells, play a vital role. There is a quanti­
This was evidenced by the activation of the Caspase-3 apoptosis marker tative decrease in circulating Tregs in T1DM patients compared to
and the induction of dysfunction in mouse INS-1 cells (insulinoma cells healthy individuals [42]. This decrease is attributed to the accumulation
releasing insulin) when exogenous IL-12 was introduced [30]. More­ of IL-17, which skews the balance between Tregs and Th17 cells towards
over, Radwan et al. demonstrated that inhibiting IL-12 and CHOP (an the latter [42]. The disruption of this balance may be facilitated by the
endoplasmic reticulum stress marker) significantly enhanced the crucial activator of Th17, IL-23. Furthermore, the functional phenotype
glucose test tolerance in mice with type 2 diabetes. Since endoplasmic of Tregs is altered in the context of inflammation. They start producing
reticulum stress is associated with apoptosis in islet cells, it implies that more IFN-γ and IL-17, possibly due to the cooperative action of IL-12 and
IL-12 may activate this stress response to promote apoptosis [31]. IL-18, which activated natural killing cells to compete with Tregs for
However, there is conflicting evidence suggesting that IL-12-induced IL-2, thereby promoting phenotype alteration [38]. The decrease in
endoplasmic reticulum stress does not directly contribute to β-cell circulating levels of IL-35 is also unfavorable for maintaining the
destruction [32]. Therefore, IL-12 may not be the primary mediator in anti-inflammatory role of Tregs [43].
the process of β-cell destruction. Both CD4+T cells and CD8+T cells require major histocompatibility
Although sharing the subunit with IL-12, neither IL-23 nor IL-35 was complex (MHC) to be activated. However, Innate lymphoid cells (ILCs),
reported directly destroy islet β cells. One possibility is that islet β cells a distinct group of cells independent of specific antigen presentation,
are more sensitive to IL-12 for the upregulation of STAT4 gene in T1DM also play a crucial role in the development of insulin resistance. This
individuals [33]. However, IL-12 family cytokines gene has not signifi­ process is closely linked to the activity of IL-12 family cytokines. For
cantly altered in T2DM according to the islet gene map, suggesting an example, when stimulated with IL-12, adipose resident ILC1s produce
indirect role on islet β cells [33]. increased levels of IFN-γ, which in turn leads to the polarization of M1-
type macrophages and the development of insulin resistance [44]. To
2.1.2. Indirect effects of IL-12 family cytokines on islet β cells elucidate whether other members of the IL-12 family also promote
The activation of IL-12 family cytokines leads to an abnormal im­ pro-inflammatory responses in ILC1s, it would be beneficial to analyze
mune response that exacerbates the burden of inflammation in the the expression of surface receptors on these cells. In the context of
pancreatic islets. This process is influenced by the dysregulation of CD4+ inflammation, IL-23 emerges as a significant mediator for ILC3s [45].
T cells, CD8+ T cells, regulatory T cells (Tregs), and other cell types ILC3s contribute to adipose inflammation in the obese state by
(Fig. 1). enhancing IL-17 signaling [46]. Therefore, it is plausible that IL-23
Once antigen-presenting cells (APCs) receive signals from damaged modulates ILC3s to exacerbate inflammation associated with adipose
islet cells, they transmit the signal to naive CD4+T cells. IL-12 family accumulation through the enhancement of IL-17 signaling.
cytokines play a crucial role in determining the density of CD4+T cells.
In 2002, it was discovered that IL-12 activates JAK2-STAT4, promoting 2.2. Anti-inflammatory role of IL-12 family cytokines
the differentiation of naive CD4+T cells (Th0) into IFN-γ producing
CD4+T cells (Th1) [34]. This activation of Th1 cells by IL-12 leads to a Five members of the IL-12 family of cytokines play a role in the
positive feedback loop, as the accumulation of IFN-γ stimulates APCs to immune response. Among these, IL-27 and IL-35 have been shown to
produce more IL-12. Concretely, in the inflammatory context, IL-1β, have anti-inflammatory effects (Fig. 1). At the initiation of the immune
TNF-α,and IFN-γ collectively enhance the expression of IL-12 as pre­ response, IL-27 and IL-35 interact to disrupt antigen presentation. IL-27
sented in Weaver et al. research [35]. This perpetuates the ongoing islet inhibits the activity of myeloid dendritic cells (mDCs) through signaling
inflammation seen in T1DM and T2DM. Although IL-23 shares the same pathways dependent on STAT-1 and STAT-3 [47]. Studies have shown
subunit IL-12p40 with IL-12, it does not facilitate the differentiation of that IL-27 released by monocytes from individuals with T1DM helps
Th0 cells due to the lack of IL-23R expression on Th0 cells initially [36]. alleviate the disease by reducing the production of pro-inflammatory
However, with the help of IL-6, IL-23R is expressed, allowing IL-23 to cytokines IL-17 [48]. On the other hand, IL-35 promotes polarization
play a role in the differentiation and proliferation of Th17 cells [36]. of macrophages towards anti-inflammatory M2 types with higher Arg1
Additionally, Ciecko and colleagues found that complete knockout of expression in spleen of NOD mice[49]. Both IL-27 and IL-35 are involved
the IL-27 gene in NOD mice provided complete protection against T1DM in preventing inflammation in pancreatic islets and targeting various
by dampening the function of Th1 cells in the spleen, pancreatic lymph effector T cells, including Th1, natural killer (NK), and Th17 cells [40,
nodes, and pancreatic islets [37]. 43,48,50]. Additionally, IL-35, one of the most powerful negative
In addition to CD4+ T cells, IL-12 family cytokines have also been immunoregulatory factors in the IL-12 family, enhances the ability of
implicated in the exacerbation of CD8+ T cell toxicity in both T1DM and Tregs and B regulatory cells (Bregs) to produce more anti-inflammatory
T2DM progression. Dean et al., in their study, observed that the com­ cytokines, such as IL-35 and IL-10 [43,51].
bination of IL-12 and IL-18 led to increased expression of granzyme B in As shown, IL-27 and IL-35 have distinct roles in regulating the im­
CD8+ T cells [38]. The knockdown of the IL-27 or IL-27 receptor gene in mune response, with IL-27 primarily suppressing the activity of mDCs,
mice resulted in a weaker ability of autoreactive CD8+ T cells to pro­ while IL-35 promotes the polarization of macrophages towards an anti-
liferate and activate [37]. More recently, the pro-inflammatory role of inflammatory phenotype. Together, these cytokines work to prevent
IL-27 on CD8+ T cells has been further substantiated. CD8+ T cells islet inflammation and exert immunoregulatory effects through their
lacking IL-27 receptors exhibited a reduced capacity to differentiate into interactions with various immune cell subsets.
terminal populations, namely, CD44highTCF1-CXCR6+ CD8+ T cells, As diabetes progresses, the anti-inflammatory functions of IL-27 and

3
J. Luo et al. Biomedicine & Pharmacotherapy 170 (2024) 115958

Fig. 1. The roles of IL-12 family in the pathogenesis of diabetes mellitus.

IL-35 are diminished, possibly due to the inhibition of their main sources induces the polarization of macrophages into a distinct subpopulation
under inflammatory conditions [52]. The failure of activated Tregs, called M(IL-23), which differs from both M1 and M2 types. These M
which are the main producers of IL-35, to effectively suppress inflam­ (IL-23) cells selectively express IL-17A and IFN-γ. The accumulation of
mation can be partially attributed to systemic inflammation. Addition­ IFN-γ, mediated by IL-23, further promotes the M1-type polarization of
ally, the roles of IL-27 may vary depending on their cellular sources. macrophages [57].
IL-27 released by activated dendritic cells (DCs) promotes inflamma­ IL-27 is known to play a role in promoting wound healing at the early
tion [37], while that released by monocytes inhibits it [48]. This stages of the process [58]. It is produced by dermal CD301b+ DCs and
discrepancy implies the future modulation of IL-27 for diabetes therapy. acts by activating epidermal proliferation. This activation is achieved by
increasing the expression of re-epithelialization markers, namely KRT6
and Ki67 [58]. However, in the context of diabetes, it is unclear whether
2.3. IL-12 family cytokines in T1DM and T2DM complications IL-27 retains its reparative capabilities. Therefore, further investigations
are needed to determine the effect of IL-27 on wound healing in diabetic
2.3.1. IL-12 family cytokines in delayed wound healing individuals.
Inflammation management and angiogenesis play crucial roles in Although there is no direct evidence about IL-35 regulation of dia­
promoting wound healing [53]. However, patients with T2DM experi­ betic wound healing, indirect evidence implies that IL-35 may be
ence impaired wound healing due to the failure of pro-inflammatory beneficial to wound healing. Patients with diabetes suffer impaired
macrophages in the bone marrow to convert into M2 Tregs, which contribute to delayed wound healing [59]. Activated Tregs
anti-inflammatory macrophages, primarily because of increased are important sources of IL-35. Impaired Tregs might lead to decreased
expression of IL-12 [54] (Fig. 2). Excessive IL-12 levels contribute to an IL-35 and ongoing inflammation. Single-cell sequencing of a diabetic
escalated inflammatory response and inhibit collagen deposition [54]. wound is needed to clarify the relationship between IL-35 and diabetic
Furthermore, the accumulation of IL-12 in the wound hampers angio­ wound healing.
genesis by triggering oxidative stress, promoting local inflammation,
and suppressing key angiogenic activities such as serine/threonine ki­ 2.3.2. IL-12 family cytokines in diabetic retinopathy
nase Akt activity, endothelial nitric oxide synthase (eNOS) activity, and Th17 cells play a pathogenetic role in diabetic retinopathy. IL-12
vascular endothelial growth factor (VEGF) signaling [55]. Conse­ family cytokines mainly modulate Th17 activity and indirectly partici­
quently, the elevated levels of IL-12 in T2DM patients may have detri­ pate in diabetic retinopathy.
mental effects on the wound healing process. When diabetic retinopathy (DR) develops, IL-23 triggers the release
IL-23, sharing the subunit IL-12p40 with IL-12, has been shown to of IL-17 by Th17 cells in the retina (Fig. 2). This interaction between IL-
delay wound healing by promoting the polarization of macrophages 23 and IL-17 contributes to the destruction of the internal blood-retinal
towards the M1 type [56]. However, it is important to note that IL-23 barrier and the tight retinal pigment epithelium [60]. However, IL-35
may not directly stimulate macrophage M1 polarization. Instead, it

4
J. Luo et al. Biomedicine & Pharmacotherapy 170 (2024) 115958

Fig. 2. IL-12 family cytokines participate in the development of diabetes complications.

inhibits the production of IL-17 by suppressing the RORα and RPRγt arteriosclerosis by indirectly inhibiting macrophage activation, pri­
pathways in Th17 cells, thereby alleviating proliferative diabetic reti­ marily through enhancing Tregs function [69]. The role of IL-27 in
nopathy [61]. The impact of IL-27 on DR is still unclear, but some diabetic arteriosclerosis remains contradictory. On one hand, IL-27 has
research suggests that it may have a negative regulatory effect on Th17 been found to improve arteriosclerosis in obese mice by inhibiting M1
function [48,62,63]. Houssen et al. found that patients with DR have macrophages [70]. On the other hand, increased levels of IL-27 have
increased levels of IL-27 in their aqueous humor as a compensatory been associated with an elevated risk of adverse cardiovascular events in
response to inflammation [64]. This suggests that IL-27 could poten­ patients with both type 1 and type 2 diabetes [29,71]. Recent research
tially protect against DR by suppressing Th17 cells. Interestingly, while tends to support a protective role for IL-27 in arteriosclerosis, as re­
IL-23 and IL-12 share the same subunit IL-12p40, the levels of IL-12 in combinant IL-27 has been shown to prevent the initiation of athero­
the aqueous humor have no correlation with DR [65]. This implies that sclerosis [72]. It is worth noting that M1 macrophages can also
IL-23-mediated DR may primarily depend on the signaling pathway upregulate the expression of IL-12, IL-23, and IL-27 [73]. Therefore, this
activated by the other subunit, IL-23p19. Xu et al. supported this hy­ positive feedback loop involving macrophages and cytokines could
pothesis by demonstrating that local administration of an anti-IL23Rp19 contribute to the development of complications in diabetes.
antibody in the vitreous cavity of STZ-treated rats improved the
blood-retinal barriers [66]. Therefore, targeting anti-IL-23p19 or its 2.3.4. IL-12 family cytokines in diabetic neuropathy
receptor may hold promise for DR therapy. IL-35 has been found to alleviate the neuropathic pain associated
with diabetes by increasing the proportion of anti-inflammatory M2 type
2.3.3. IL-12 family cytokines in diabetic arteriosclerosis macrophages in the central nervous system, namely microglia (Fig. 2)
Macrophages play a crucial role in the development of diabetic [57]. On the other hand, IL-23 has been shown to exacerbate cognitive
arteriosclerosis. The pro-inflammatory macrophages, promoted by both decline in individuals with diabetes by promoting the production of
IL-12 and IL-23, contribute to the worsening of arteriosclerosis [67,68]. IL-17 [74,75]. Additionally, reducing the levels of circulating IL-12
However, IL-35 acts as a protective factor against diabetic through the use of Resveratrol also contributes to the relief of

5
J. Luo et al. Biomedicine & Pharmacotherapy 170 (2024) 115958

neuroinflammation [76]. regulatory cytokines, have shown promising potential in modulating


Intriguingly, the pathogenesis of diabetic complications does not inflammation. Therefore, this discussion aims to provide an overview of
occur independently and may be mutually influenced. The involvement current and potential treatment options for T1DM and T2DM, focusing
of IL-12 family cytokines plays a crucial role in mediating these com­ on targeting the IL-12 family of cytokines. These treatment options
plications. For instance, the development of diabetic psoriasis and dia­ include drug therapy, combination therapy, gene therapy, cell therapy,
betic atherosclerosis is closely linked to the IL-23/IL-17 pathway (Fig. 2) cytokine engineering therapy, and modulation of gut microbiota.
[77]. This suggests the potential for interrupting the progression of other
diabetic complications by timely modulation of circulating IL-12 family
3.1. Drug therapy
cytokine levels. With this perspective in mind, exploring the relationship
between diabetic retinopathy (DR) and neuropathic pain, both of which
Inhibition of IL-12 family cytokines of synthesis or signaling pathway
are influenced by IL-35, becomes particularly intriguing. Additionally, it
allows drug therapy to suppress inflammation of islets (Fig. 3).
would be worth investigating whether increased levels of IL-27 in pa­
In many cases, targeting the main sources of cytokines has proven
tients with DR could potentially enhance the risks of atherosclerosis in
effective in reducing their levels. For example, by inducing autophagy in
the future.
liver macrophages, which are the primary source of IL-23, Empagliflozin
has been shown to improve non-alcoholic fatty liver disease (NAFLD)
2.4. Interplays of IL-12 family cytokines associated with T2DM [82]. Sitagliptin, on the other hand, reduces
IL-23 production by decreasing circulating Th17 cells [19]. It is
The pathogenesis of diabetes is not solely driven by IL-12 family important to note that targeting the source cells does not involve their
cytokines but also by their interactions with other factors. For instance, direct destruction, but rather disruption of the synthetic signaling
IL-27 can activate Th cells through a STAT1-dependent pathway, lead­ pathway. One approach to achieve this is by modifying Toll-like re­
ing to the upregulation of T-box expression and IL-12p35 receptor ceptors (TLRs). For instance, Lactobacillus plantarum OLL2712 en­
expression. This enhances T cells’ sensitivity to IL-12 signals [78]. While hances the activity of TLR-2 and reduces IL-12 and IL-23 production
IL-23 shares the IL-12p40 subunit with IL-12, it actually suppresses the from bone marrow-derived dendritic cells [83]. Interestingly, TLR-2
effects of IL-12 on Th1 responses [79]. Although IL-27 can suppress the inhibitors have been shown to reduce IL-27 production and alleviate
function of Th17, it has little effect on committed Th17, which is islet inflammation [84]. This discrepancy may be due to the involve­
induced by IL-23 [62]. Furthermore, IL-35 competes with IL-12 for the ment of different types of TLR-2-expressing cells, suggesting that sys­
receptor IL-12Rβ2 and partially inhibits the function of IL-12 [80]. temic drug administration could potentially disrupt normal immune
Maintaining a balance in the immune response depends on the coordi­ responses. Additionally, other components of the signaling pathway
nation of members of the IL-12 family. Therefore, further exploration of may hold therapeutic value. Quercetin, for example, inhibits β-catenin
the interactions between the cytokines in the IL-12 family is necessary to in bone marrow-derived DCs, leading to a decrease in IL-12 [85].
better understand the pathogenesis of diabetes. However, it is worth noting that monocyte-derived DCs rely on β-catenin
to decrease IL-12 and limit inflammation [86]. Thus, the effect of sta­
3. Targeting IL-12 family cytokines for DM therapy bilizing or inhibiting β-catenin may depend on the types of DCs. Another
approach involves blocking the production of IL-12 subunits, IL-12p35
T1DM is currently managed through insulin therapy or even islet and IL-12p40, using Apilimod (STA-5326), which inhibits the migra­
transplantation during the latent stage [5]. On the other hand, the tion of C-Rel (a protein involved in the NF-κB signaling pathway) from
treatment approach for T2DM involves a combination of diet therapy, the cytoplasm to the nucleus [87]. It is recommended that future
exercise, and medication. The primary objective of drug therapy is to research investigate the effects of different pathways targeted by these
increase insulin levels and improve insulin sensitivity. Insulin therapy drugs on relieving inflammation in the islets.
still remains the preferred method for managing T1DM [5]. However, Another potential approach is to inhibit the signaling pathway of IL-
long-term administration of insulin can result in complications such as 12 family cytokines. Currently, there are two methods available for
disrupted postprandial glucose absorption and an increased risk of blocking these pathways. Since IL-12 family cytokines share subunits,
obesity and cardiovascular disease [81]. drugs that bind to these subunits can hinder the activation of their
Insulin therapy and traditional drug therapy do not directly address respective receptors on immune cells. Ustekinumab serves as one
the inflammation of islets in both type 1 and type 2 diabetes. However, example of such a drugs.
timely restriction of inflammation can be beneficial in preserving the Ustekinumab, an anti-p40 monoclonal antibody approved by the
islet β cells. The IL-12 family cytokines, which are important immune- FDA for treating psoriasis, has also shown effectiveness in the treatment

Fig. 3. Present and potential therapies for targeting IL-12 family cytokines.

6
J. Luo et al. Biomedicine & Pharmacotherapy 170 (2024) 115958

Table 1
Clinical trials of drugs for targeting IL-12 family cytokines.
Drug Target Clinical trial name patients phase Identifier www.clini
caltrials. gov/

Baricitinib IL-12/ A Phase 2, Randomized, Placebo Controlled Study Investigating the 91 participants(10–30 years old) II NCT04774224
IL-23 Efficacy of Baricitinib in New Onset Type 1 Diabetes Mellitus within 100 days of diagnosis
Ustekinumab IL-12/ Phase I/II Study of Ustekinumab in Patients With New-onset Type 1 20 adults(18–35 years old)within I/II NCT02117765
IL-23 Diabetes 100 days of diagnosis
Ustekinumab IL-12/ Clinical Phase II/III Trial of Ustekinumab to Treat Type 1 Diabetes 66 adults (18–35 years old) with II/III NCT03941132
IL-23 recent-onset T1D
Lisofylline IL-12 Lisofylline as Continuous Subcutaneous and Intravenous 1 Participant (18–35 years old) with Terminated NCT01603121
Administration in Subjects With Type 1 Diabetes Mellitus T1DM
Empagliflozin IL-23 Empagliflozin add-on to Insulin in Type 1 Diabetes Mellitus Over 28 75 participants aged 18–35 years II NCT01969747
Days with T1DM
Sitagliptin IL-23 Effectiveness of Sitagliptin for the Treatment of Newly Diagnosed 50 Patients with newly diagnosed IV NCT04495881
Type 2 Diabetes T2DM

of T1DM (Table 1). Its mechanism of action involves interrupting the it unsuitable for T1DM patients with infectious diseases [101]. In the
downstream signaling pathway of IL-12 and IL-23 when combined with future, it is proposed to identify more specific targets for Sorafenib and
IL-12p40. This disruption leads to a reduction in the levels of IFN-γ and minimize its limitations.
IL-6 from Th1 cells, as well as IL-17 from Th17 cells. Animal experiments There are several obstacles that hinder the application of drug
have demonstrated the feasibility of prescribing Ustekinumab for the therapy in the treatment of T1DM. One of the main challenges is the
treatment of diabetes [88]. Furthermore, in a phase I clinical trial uncertainty surrounding the timing of drug administration. Both animal
(NCT02117765), Ustekinumab was found to improve inflammation in study and clinical trial recommended that Ustekinumab should be
islets of patients with T1DM when administered subcutaneously [89]. As administered early in the course of T1DM. For instance, in NOD mice,
a result, the systematic application of Ustekinumab can be considered the injection of an IL-12p40 antagonist did not significantly delay the
relatively safe. Other drugs, such as Mirikizumab [90] and Guselkumab onset of diabetes at 8 weeks, while the opposite effect was observed at 6
[91], which antagonize IL-23p19, may also hold potential for diabetes weeks [102]. Consistent with it, a phase I clinical trial (NCT02117765)
treatment. reported a significant decrease in the number of inflammatory cells in
In recent years, targeting the Janus kinase-signal transducers and C-peptide responders, suggesting that the drug may exhibit higher
activators of transcription (JAK-STAT) pathway as an alternative sensitivity to islet β cells when administered in the early stages of T1DM
approach to inhibiting the IL-12 family has gained attention [12]. This is [89]. Also, these findings highlight the complexity of determining the
because IL-12 family cytokines have a common characteristic of acti­ optimal timing for drug therapy in T1DM.
vating the JAK-STAT pathways. One JAK2 inhibitor, Baricitinib, has Additionally, there is a lack of consensus regarding the appropriate
been shown to inhibit Th1 differentiation through IL-12 stimulation and dosage of certain drugs, such as Baricitinib, Lisofylline, and Sorafenib, as
Th17 differentiation through IL-23 stimulation (Table 1) [92]. they are dose-dependent medications. In the case of Baricitinib, a daily
Currently, Baricitinib is being tested in a clinical trial for newly-onset dosage of 4 mg is recommended for the treatment of diabetes [93]. It has
T1DM patients (NCT04774224) [93]. Additionally, its therapeutic po­ been reported that Lisofylline at concentrations ranging from 20 to
tential for the treatment of T2DM should be explored, as it has the ability 100 mM is effective in combating islet inflammation in vitro [103]. For
to alleviate insulin resistance by reducing T-cell infiltration around the inhibition of islet inflammation in diabetes patients, particularly in
skeletal muscles and adipose tissue [94]. However, the role of other JAK adults with latent autoimmune diabetes, a dosage of 10 mg/kg/day of
inhibitors such as Tofacitinib, Filgotinib, and Upadacitinib in the Sorafenib is proposed [101]. However, further clinical evidence is
anti-inflammatory response during the pathogenesis of T1DM or T2DM required to establish the safe and effective dosages for these drugs.
remains unclear, as there is a lack of evidence from rodent studies. Finally, the use of certain medications has been found to have sig­
The inhibition of phosphorylation of STAT is also being considered in nificant side effects that can cause concerns in various applications. For
the treatment of diabetes. Lisofylline, an anti-inflammatory agent, was example, the administration of Baricitinib systematically has been
first reported to be effective for Type 1 Diabetes Mellitus (T1DM) ther­ observed to weaken the immune response. Similarly, taking Empagli­
apy in 2002 (Table 1) [95]. Lisofylline works by suppressing the phos­ flozin orally has been associated with inducing hypotension [104].
phorylation of STAT4, thereby inhibiting the IL-12 signaling pathway Consequently, exploring alternative administration methods for these
[95]. What makes Lisofylline particularly attractive for diabetes therapy drugs may offer a potential solution to mitigate such side effects.
is not only its ability to improve islet inflammation [96] but also its
potential to prevent the onset of diabetes and improve complications 3.2. Combination therapy
[97,98]. It is possible that patients who are treated with Lisofylline at an
early stage of diabetes may be able to maintain glucose tolerance. Combination therapy options have been explored to further enhance
However, the clinical trial did not yield favorable results. The current the anti-inflammatory properties of IL-12 family cytokines (Fig. 3).
clinical trial (NCT06103121) was terminated due to the participant’s Various bioactive factors have been proposed for combination with
illness after treatment with Lisofylline. This might be attributed to the these drugs. One potential strategy is to combine Ustekinumab with IL-2,
low specificity of Lisofylline [99]. Another drug that targets the sup­ Treg cells, or specific adjuvants that facilitate antigen-based toleriza­
pression of STAT4 phosphorylation is Sorafenib (Table 1). Sorafenib, an tion, aiming to preserve islet β cells [90]. Another approach involves
FDA-approved antineoplastic agent, also has the potential to reduce coadministration of Lisofylline with insulin-resistant protein (INGAP),
inflammation [100]. Although clinical trials of Sorafenib for the treat­ which has shown improved efficacy in enhancing glucose tolerance
ment of diabetes have not been conducted, its therapeutic potential has [105]. IL-35, when combined with other immune factors, has demon­
been evaluated. Sorafenib indirectly blocks the JAK2-STAT4 signaling strated promising effects in the treatment of autoimmune diseases such
pathway of IL-12, leading to a significant decrease in Th1 cells around as asthma and colitis [106,107]. Therefore, combining IL-35 with other
islets [101]. Due to its higher specificity and lower dosage requirements, immune factors may enhance its anti-inflammatory effect on islet cells.
Sorafenib is an attractive option for diabetes therapy. However, it is Of particular interest is IL-10, which can serve as a suitable partner for
important to note that Sorafenib’s immunosuppressive properties make IL-35. On the one hand, IL-10 exhibits immunosuppressive properties.

7
J. Luo et al. Biomedicine & Pharmacotherapy 170 (2024) 115958

On the other hand, IL-35 enhances the protective role of Tregs, given with the IL-35 gene (IL-35-MSCs) have been successful in inhibiting
sufficient IL-10 levels in the islets [43]. Additionally, considering the CD4+T cell proliferation and increasing the proportion of Tregs [116]. In
context of inflammation, the application of IL-27 in combination with animal models of autoimmune hepatitis and ulcerative colitis,
anti-inflammatory cytokines like IL-10 and TGF-β appears to be a IL-35-MSCs have demonstrated efficacy, indicating their potential for
reasonable approach. the treatment of Type 1 Diabetes Mellitus (T1DM) [116]. Another
In addition to incorporating assistant cytokines, an alternative avenue for cell modification under consideration is the modification of
approach to address drug shortages could involve the use of modified DCs to tolerant phenotypes. Spleen DC cells transfected with the
materials. Lisofylline, due to its high hydrophilicity and rapid metabolic single-chain IL-35Ig fusion gene have shown promising effects in
interconversion, poses challenges in maintaining its therapeutic effect reducing pro-inflammatory immune cells surrounding islet β cells [117].
over an extended period. However, when combined with linoleic acid to Similarly, modifying bone marrow-derived DCs to overexpress autoim­
form self-assembled micelles (LSF-LA), Lisofylline exhibits a longer half- mune regulators (AIRE) can decrease IL-27 production, which impedes
life and improved glucose tolerance compared to the pure formulation the secretion of islet-specific antibodies [118]. However, it is important
[99,108]. Recent studies have demonstrated that transforming LSF-LA to carefully select the type of DCs for cell modification, as increased
micelles into nanoparticulate tablets can further enhance the drug’s expression of IL-27 from myeloid DCs can induce the tolerant phenotype
half-life and bioavailability [109]. of DCs [47]. Adoptive cellular immunotherapy involves extracting
Combination therapy faces several challenges that must be overcome anti-inflammatory immune cells surrounding the islet and proliferating
for its successful implementation. Firstly, the utilization of bioactive them in vitro. These proliferated cells, along with additional factors such
factors is hindered by their low concentrations and short lifespan, as IL-2, IL-10, and IL-35, are then implanted into the body to combat
making it difficult to effectively employ them in treatment. Moreover, inflammation. Chimeric antigen receptor Tregs (CAR-T) therapy is an
estimating the cost associated with modifying the delivery system for example of adoptive cellular immunotherapy [119]. CAR-T has advan­
these bioactive factors is essential, as it directly impacts the clinical tages in enhancing specificity towards islets independently of MHC and
feasibility of such therapy [110]. Finally, the scarcity of clinical trials increasing the ratio of Tregs/Teffs [119]. However, a single infusion
poses a significant obstacle in establishing the safety and efficacy of only delays the development of T1DM instead of preventing it entirely
combination therapy. [120]. To address this, the addition of a low dose of IL-2 is proposed in
CAR-T therapy to enhance the function of Tregs [121]. Likewise,
3.3. Gene therapy IL-35-producing regulatory B cells (Bregs) also play a crucial role in
maintaining immune balance. Bregs directly induce the intolerance of
Gene therapy involves vaccines by modifying viral capsid proteins DCs, indirectly inhibiting the activation of pathogenic CD8+T cells
and combining them with transgenic systems (Fig. 3). By engineering [122]. Further research is needed to determine whether it is possible to
capsid proteins loaded with cytokine genes, gene therapy allows for design CAR-B cells with additional IL-35 to improve diabetes.
enhanced tropism for islet β cells without triggering an antigen-antibody
response in the body [111]. Besides, gene therapy avoids administrating 3.5. Cytokines engineering therapy
the drug continuously. Hence, notable advantages of gene therapy are
the improved stability and specificity of cytokines. For example, when To optimize efficiency, it is advantageous to manipulate cytokines
an islet β cell-targeted adeno-associated virus vector with IL-35 trans­ and their receptors (Fig. 3). Cytokine engineering holds great promise as
gene (AAV8mIP-IL-35) is injected into NOD mice, only CD4+ and CD8+ it enables to modify the signaling pathways of IL-23 by modulating IL-23
T cells in the islets are reduced, resulting in decreased levels of receptors (IL-23R). When cytokines bind to receptors, it triggers a
pro-inflammatory factors like IFN-γ and TNF-α [50]. This demonstrates conformational rearrangement, which in turn activates downstream
the crucial role of adeno-associated virus vectors in maintaining the signaling pathways. An interesting finding reported that the deletion of
stability of exogenous IL-35. Moreover, the use of the IL-35 transgene the extended stalk regions of IL-23R prevented IL-23-mediated confor­
with an insulin promoter enhances the specificity of the islets. mational rearrangement while maintaining the activation status in cells
Although gene therapy involving the other members of the IL-12 [123]. This technique, known as receptor interface engineering, inspired
family of cytokines has not been previously explored for diabetes us to explore the possibility of specifically controlling signaling and
treatment, it holds great potential. One possible approach is the use of restraining the activity of target cells by modifying the receptor. If
adeno-associated virus vectors with soluble IL-23 receptors, which has successfully applied to diabetes patients, this could protect pancreatic
been proved effective to protect against autoimmune encephalomyelitis islets from inflammation without adversely affecting the immune
through inhibiting IL-23/Th17 signaling pathway [112]. Similarly, an system.
adeno-associated virus vector containing IL-27 is successful to halt the Recently, researchers have developed Supercytokines to address the
onset of autoimmune encephalomyelitis and colitis, with the common limitations of natural cytokines. One approach to fabricating Super­
characteristic of inhibiting Th17 activity [113,114]. Another promising cytokines is the synthesis of cytokines called Synthekines, which are not
method of gene therapy involves modifying the transcript of IL-27. For found in nature. These Synthekines are created by fusing two subunits
instance, by targeting the IL-27 gene c-terminus with IL-27pL, the from different cytokines to activate distinct signaling cascades [124].
effectiveness of glucocorticoids in improving the treatment of rheuma­ Promising results have been obtained in animal experiments investi­
toid arthritis was enhanced [115]. This is attributed to the peptide’s gating the therapeutic potential of Synthekines for diabetes treatment.
specific binding to the interleukin 6 receptor type 1 (IL-6Rα), which is Specifically, a fusion of two subunits from the IL-12 family cytokines,
also involved in the inflammatory response observed in diabetic islets IL-12p40 and IL-12p28, called scIL-Y, demonstrated effectiveness in
[90]. Altogether, gene therapy of IL-12 family cytokines are applicable preventing the onset of diabetes in NOD mice [125]. Overall, further
for harmonizing the immune disorders, which is hopeful for diabetes research could explore the fusion of subunits from other IL-12 family
therapy. cytokines, such as IL-12p28 and IL-12p35, to expand the range of ap­
plications for Supercytokines [125].
3.4. Cell therapy
3.6. Gut microbiota modulation
Cell therapy refers to the enhancement of immunoregulation through
either modifying cells or adopting cellular immunotherapy (Fig. 3). One Recent research has highlighted the strong correlation between gut
approach to cell modification involves transfecting cells with modified microbiota disorder and the development of both T1DM and T2DM
cytokine genes. For example, mesenchymal stem cells (MSCs) modified [126,127]. One significant link between gut microbiota and diabetes is

8
J. Luo et al. Biomedicine & Pharmacotherapy 170 (2024) 115958

the role of IL-12 family cytokines, which serve as crucial intermediaries maintaining stability. This will pave the way for improved treatment of
(Fig. 3). In NOD mice lacking gut microbiota, higher levels of IL-12 were T1DM and T2DM. Further research are needed to overcome the existing
observed, accompanied by elevated autoantibody titers [128]. More­ challenges and advance the field of targeting IL-12 family cytokine for
over, the dysbiosis of gut microbiota contributes to the disruption of the diabetes therapy.
intestinal barrier through the release of IL-12 family cytokines.
Maintaining the integrity of the intestinal barrier is crucial in Funding
combating systemic inflammation and reducing the risk of obesity-
related conditions. One way to achieve this is by reducing IL-23 and This work was supported by National Natural Science Foundation of
increasing IL-27 levels, as these cytokines contribute to intestinal barrier China [Grant/Award Numbers: 81970930].
integrity [129,130]. For instance, the probiotic Clostridium Faecali­
bacterium prausnitzii has been found to stimulate IL-27 secretion by CRediT authorship contribution statement
dendritic cells and promote IL-10 production by regulatory T cells,
effectively enhancing the body’s anti-inflammatory response. This Jiayu Luo: Conceptualization, Investigation, Writing – original
finding has potential implications for reducing inflammation in the islets draft. Tingting Ning: Conceptualization, Data curation. Xing Li: Formal
[131]. analysis, Resources. Tao Jiang: Formal analysis. Shenglong Tan:
Surprisingly, even if the intestinal barrier is disrupted, the body has Visualization. Dandan Ma: Funding acquisition, Project administration,
another mechanism to resist obesity. Intestinal bacteria produce a Supervision, Writing – review & editing.
metabolite that induces intestinal B cells to secrete IL-35, especially
under high-fat diet conditions. This secretion of IL-35 helps suppress
Declaration of generative AI and AI-assisted technologies in the
inflammation and prevents the development of type 2 diabetes [132].
writing process
When targeting IL-12 family cytokines through manipulation of the gut
microbiota, it is crucial to consider the order of treatment [133]. In
During the preparation of this work, the author(s) used Wordtune in
healthy individuals, maintaining intestinal homeostasis relies on the
order to improve language. After using this tool, the author(s) reviewed
early involvement of CD4+ T cells and the later maintenance of innate
and edited the content as needed and take(s) full responsibility for the
lymphoid cells (ILCs). Therefore, it is suggested that in the initial stages,
content of the publication.
establishing a gut microbiota profile related to IL-12 can enhance the
role of CD4+ T cells. Subsequently, establishing a gut microbiota profile
related to IL-27 and IL-35 can further enhance ILC stability and function. Declaration of Competing Interest

4. Conclusion We declare that we have no financial and personal relationships with


other people or organizations that can inappropriately influence our
Diabetes is the chronic disease largely associated with immune dis­ work, there is no professional or other personal interest of any nature or
orders. Here, we highlight the strong relationship between IL-12 family kind in any product, service and/ or company that could be constructed
cytokines and diabetes by analyzing the changing rules of IL-12 family as influencing the position presented in, or the review of, the manuscript
cytokines. Further, we investigate the roles of IL-12 family cytokines in entitled.
T1DM and T2DM. The imbalance of pro- and anti-inflammation in islet β
cells is attributed to the disordered regulation of IL-12 family cytokines. Acknowledgments
We provide the new insights to investigate the functions of IL-12 family
cytokines in diabetes and complications from the view of compositions. This work was supported by the National Natural Science Foundation
Novelly, IL-12 family cytokines serve as the bridges between metabolism of China Grants (No. 81970930). We thank the Southern Medical Uni­
and immune disorders by modulating ILCs. As for the role of anti- versity of China for language support. Figures in the review were created
inflammation, the inhibitory effects of IL-27 and IL-35 reflects on with Biorender.com.
almost every step of innate response, including antigen-presentation,
effector cells activation, and inhibitory cytokines releasing. Also, IL-12
References
family cytokines play a crucial role in diabetes complications
including delayed wound healing, retinopathy, arteriosclerosis, and [1] Home, Resources, L. with diabetes, Acknowledgement, FAQs, Contact, P. Policy,
neuropathy. It implies that targeting IL-12 family cytokines not only has IDF Diabetes Atlas | Tenth Edition, (n.d.). https://diabetesatlas.org/ (accessed
the advantages of relieving islet inflammation, but also improves dia­ August 21, 2022).
[2] P. Pirot, A.K. Cardozo, D.L. Eizirik, Mediators and mechanisms of pancreatic beta-
betes complications. Hence, targeting IL-12 family cytokines possess cell death in type 1 diabetes, Arq. Bras. Endocrinol. Metab. 52 (2008) 156–165,
tremendous potentials for diabetes therapy. https://doi.org/10.1590/s0004-27302008000200003.
We introduce present and potentials strategies, including drug [3] I. Zentsova, Z. Parackova, J. Kayserova, L. Palova-Jelinkova, P. Vrabcova,
N. Volfova, Z. Sumnik, S. Pruhova, L. Petruzelkova, A. Sediva, Monocytes
therapy, combination therapy, gene therapy, cell therapy, cytokines contribute to DNA sensing through the TBK1 signaling pathway in type 1 diabetes
engineering therapy, and gut microbiota modulation. Drug therapy re­ patients, J. Autoimmun. 105 (2019), 102294, https://doi.org/10.1016/j.
lieves diabetes through hindering the synthesis and disrupting the jaut.2019.06.005.
[4] Y.R. Timasheva, Z.R. Balkhiyarova, T.R. Nasibullin, D.Sh Avzaletdinova, T.
signaling pathways. However, issues like medication safety (e.g., timing, V. Morugova, O.E. Mustafina, I. Prokopenko, Multilocus associations of
dosage, and toxicity) require further investigation. Combination therapy inflammatory genes with the risk of type 1 diabetes, Gene 707 (2019) 1–8,
complements the effect of cytokine targeting by supplementing bioac­ https://doi.org/10.1016/j.gene.2019.04.085.
[5] S.Y. Tan, J.L. Mei Wong, Y.J. Sim, S.S. Wong, S.A. Mohamed Elhassan, S.H. Tan,
tive factors and modifying delivery systems. Unfortunately, the efficacy G.P. Ling Lim, N.W. Rong Tay, N.C. Annan, S.K. Bhattamisra, M. Candasamy,
of bioactive factors and the costs associated with optimizing delivery Type 1 and 2 diabetes mellitus: a review on current treatment approach and gene
systems hinder widespread application. Gene therapy raises the speci­ therapy as potential intervention, Diabetes Metab. Syndr.: Clin. Res. Rev. 13
(2019) 364–372, https://doi.org/10.1016/j.dsx.2018.10.008.
ficity and stability of cytokines. But related research was only conducted
[6] K. Esposito, D. Giugliano, The metabolic syndrome and inflammation: association
in animals. Cell therapy, cytokines engineering therapy and gut micro­ or causation? Nutr., Metab. Cardiovasc. Dis. 14 (2004) 228–232, https://doi.org/
biota modulation have an attractive effect on enhanced specificity and 10.1016/S0939-4753(04)80048-6.
controlling inflammation, becoming momentum for further study. It is [7] P.A. Kern, G.B. Di Gregorio, T. Lu, N. Rassouli, G. Ranganathan, Adiponectin
expression from human adipose tissue: relation to obesity, insulin resistance, and
crucial to explore various strategies for targeting IL-12 family cytokines tumor necrosis factor-alpha expression, Diabetes 52 (2003) 1779–1785, https://
that are safe, effective, accessible, cost-effective, and capable of doi.org/10.2337/diabetes.52.7.1779.

9
J. Luo et al. Biomedicine & Pharmacotherapy 170 (2024) 115958

[8] W. Milano, F. Carizzone, M. Foia, M. Marchese, M. Milano, B. Saetta, A. Capasso, prediction of major cardiovascular events in type 2 diabetes, Diabetologia 61
Obesity and its multiple clinical implications between inflammatory states and (2018) 1748–1757, https://doi.org/10.1007/s00125-018-4641-z.
gut microbiotic alterations, Diseases 11 (2022) 7, https://doi.org/10.3390/ [30] D.A. Taylor-Fishwick, J.R. Weaver, W. Grzesik, S. Chakrabarti, S. Green-Mitchell,
diseases11010007. Y. Imai, N. Kuhn, J.L. Nadler, Production and function of IL-12 in islets and beta
[9] L.A. Berchtold, M. Prause, J. Størling, T. Mandrup-Poulsen, Cytokines and cells, Diabetologia 56 (2013) 126–135, https://doi.org/10.1007/s00125-012-
pancreatic β-cell apoptosis, in: Advances in Clinical Chemistry, Elsevier, 2016, 2732-9.
pp. 99–158, https://doi.org/10.1016/bs.acc.2016.02.001. [31] E. Radwan, S. Belmadani, K. Matrougui, Disrupting interleukin 12 improves
[10] E.D. Tait Wojno, C.A. Hunter, J.S. Stumhofer, The immunobiology of the microvascular endothelial function in type 2 diabetes through ER stress CHOP
interleukin-12 family: room for discovery, Immunity 50 (2019) 851–870, https:// and oxidative stress mechanisms, Diabetes Metab. Syndr. Obes. 15 (2022)
doi.org/10.1016/j.immuni.2019.03.011. 2633–2642, https://doi.org/10.2147/DMSO.S369488.
[11] Z. Lu, K. Xu, X. Wang, Y. Li, M. Li, Interleukin 39: a new member of interleukin 12 [32] T. Satoh, N. Abiru, M. Kobayashi, H. Zhou, K. Nakamura, G. Kuriya,
family, Cent. Eur. J. Immunol. 45 (2020) 214–217, https://doi.org/10.5114/ H. Nakamura, Y. Nagayama, E. Kawasaki, H. Yamasaki, L. Yu, G.S. Eisenbarth,
ceji.2020.97911. E. Araki, M. Mori, S. Oyadomari, K. Eguchi, CHOP deletion does not impact the
[12] D.A.A. Vignali, V.K. Kuchroo, IL-12 family cytokines: immunological playmakers, development of diabetes but suppresses the early production of insulin
Nat. Immunol. 13 (2012) 722–728, https://doi.org/10.1038/ni.2366. autoantibody in the NOD mouse, Apoptosis 16 (2011) 438–448, https://doi.org/
[13] Q. Wang, D. Li, G. Cao, Q. Shi, J. Zhu, M. Zhang, H. Cheng, Q. Wen, H. Xu, L. Zhu, 10.1007/s10495-011-0576-2.
H. Zhang, R.J. Perry, O. Spadaro, Y. Yang, S. He, Y. Chen, B. Wang, G. Li, Z. Liu, [33] R.M. Elgamal, P. Kudtarkar, R.L. Melton, H.M. Mummey, P. Benaglio, M.-
C. Yang, X. Wu, L. Zhou, Q. Zhou, Z. Ju, H. Lu, Y. Xin, X. Yang, C. Wang, Y. Liu, G. L. Okino, K.J. Gaulton, An integrated map of cell type–specific gene expression in
I. Shulman, V.D. Dixit, L. Lu, H. Yang, R.A. Flavell, Z. Yin, IL-27 signalling pancreatic islets, Diabetes 72 (2023) 1719–1728, https://doi.org/10.2337/db23-
promotes adipocyte thermogenesis and energy expenditure, Nature 600 (2021) 0130.
314–318, https://doi.org/10.1038/s41586-021-04127-5. [34] A. Morinobu, M. Gadina, W. Strober, R. Visconti, A. Fornace, C. Montagna, G.
[14] N. Yazdanpanah, M. Yazdanpanah, Y. Wang, V. Forgetta, M. Pollak, M. Feldman, R. Nishikomori, J.J. O’Shea, STAT4 serine phosphorylation is critical
C. Polychronakos, J.B. Richards, D. Manousaki, Clinically relevant circulating for IL-12-induced IFN-γ production but not for cell proliferation, Proc. Natl. Acad.
protein biomarkers for type 1 diabetes: evidence from a two-sample mendelian Sci. USA 99 (2002) 12281–12286, https://doi.org/10.1073/pnas.182618999.
randomization study, Diabetes Care 45 (2022) 169–177, https://doi.org/ [35] J.R. Weaver, J.L. Nadler, D.A. Taylor-Fishwick, Interleukin-12 (IL-12)/STAT4
10.2337/dc21-1049. axis is an important element for β-cell dysfunction induced by inflammatory
[15] K. Alnek, K. Kisand, K. Heilman, A. Peet, K. Varik, R. Uibo, Increased blood levels cytokines, PLOS One 10 (2015), e0142735, https://doi.org/10.1371/journal.
of growth factors, proinflammatory cytokines, and Th17 cytokines in patients pone.0142735.
with newly diagnosed type 1 diabetes, PLOS One 10 (2015), e0142976, https:// [36] K. Ghoreschi, A. Laurence, X.-P. Yang, C.M. Tato, M.J. McGeachy, J.E. Konkel, H.
doi.org/10.1371/journal.pone.0142976. L. Ramos, L. Wei, T.S. Davidson, N. Bouladoux, J.R. Grainger, Q. Chen, Y. Kanno,
[16] S. Kasela, K. Kisand, L. Tserel, E. Kaleviste, A. Remm, K. Fischer, T. Esko, H.- W.T. Watford, H.-W. Sun, G. Eberl, E.M. Shevach, Y. Belkaid, D.J. Cua, W. Chen,
J. Westra, B.P. Fairfax, S. Makino, J.C. Knight, L. Franke, A. Metspalu, J.J. O’Shea, Generation of pathogenic T(H)17 cells in the absence of TGF-β
P. Peterson, L. Milani, Pathogenic implications for autoimmune mechanisms signalling, Nature 467 (2010) 967–971, https://doi.org/10.1038/nature09447.
derived by comparative eQTL analysis of CD4+ versus CD8+ T cells, PLOS Genet. [37] A.E. Ciecko, B. Foda, J.Y. Barr, S. Ramanathan, M.A. Atkinson, D.V. Serreze, A.
13 (2017), e1006643, https://doi.org/10.1371/journal.pgen.1006643. M. Geurts, S.M. Lieberman, Y.-G. Chen, Interleukin-27 is essential for type 1
[17] D. Espes, K. Singh, S. Sandler, P.-O. Carlsson, Increased interleukin-35 levels in diabetes development and sjögren syndrome-like inflammation, Cell Rep. 29
patients with type 1 diabetes with remaining C-peptide, Diabetes Care 40 (2017) (2019) 3073–3086.e5, https://doi.org/10.1016/j.celrep.2019.11.010.
1090–1095, https://doi.org/10.2337/dc16-2121. [38] J.W. Dean, L.D. Peters, C.A. Fuhrman, H.R. Seay, A.L. Posgai, S.E. Stimpson, M.
[18] A. Roohi, M. Tabrizi, F. Abbasi, A. Ataie-Jafari, B. Nikbin, B. Larijani, M. Qorbani, A. Brusko, D.J. Perry, W.-I. Yeh, B.N. Newby, M.J. Haller, A.B. Muir, M.
A. Meysamie, H. Asgarian-Omran, B. Nikmanesh, A. Bajouri, N. Shafiey, A. Atkinson, C.E. Mathews, T.M. Brusko, Innate inflammation drives NK cell
A. Maleki, Serum IL-17, IL-23, and TGF-β levels in type 1 and Type 2 diabetic activation to impair treg activity, J. Autoimmun. 108 (2020), 102417, https://
patients and age-matched healthy controls, Biomed. Res Int 2014 (2014), doi.org/10.1016/j.jaut.2020.102417.
718946, https://doi.org/10.1155/2014/718946. [39] A.E. Ciecko, D.M. Schauder, B. Foda, G. Petrova, M.Y. Kasmani, R. Burns, C.-
[19] M. Rezaeepoor, M. Hoseini-Aghdam, V. Sheikh, M.M. Eftekharian, M. Behzad, W. Lin, W.R. Drobyski, W. Cui, Y.-G. Chen, Self-renewing islet TCF1+ CD8 T cells
Evaluation of interleukin-23 and JAKs/STATs/SOCSs/ROR-γt expression in type 2 undergo IL-27-controlled differentiation to become TCF1- terminal effectors
diabetes mellitus patients treated with or without sitagliptin, J. Interferon during the progression of type 1 diabetes, J. Immunol. 207 (2021) 1990–2004,
Cytokine Res 40 (2020) 515–523, https://doi.org/10.1089/jir.2020.0113. https://doi.org/10.4049/jimmunol.2100362.
[20] L. Qian, H. Shi, M. Ding, Comparative analysis of gene expression profiles in [40] H. Fujimoto, T. Hirase, Y. Miyazaki, H. Hara, N. Ide-Iwata, A. Nishimoto-Hazuku,
children with type 1 diabetes mellitus, Mol. Med. Rep. (2019), https://doi.org/ C.J.M. Saris, H. Yoshida, K. Node, IL-27 inhibits hyperglycemia and pancreatic
10.3892/mmr.2019.10099. islet inflammation induced by streptozotocin in mice, Am. J. Pathol. 179 (2011)
[21] T.L. Mastracci, J.-V. Turatsinze, B.K. Book, I.A. Restrepo, M.J. Pugia, E.A. Wiebke, 2327–2336, https://doi.org/10.1016/j.ajpath.2011.08.001.
M.D. Pescovitz, D.L. Eizirik, R.G. Mirmira, Distinct gene expression pathways in [41] J. Li, Q. Niu, A. Wu, Y. Zhang, L. Hong, H. Wang, Causal relationship between
islets from individuals with short- and long-duration type 1 diabetes, Diabetes circulating immune cells and the risk of type 2 diabetes: a Mendelian
Obes. Metab. 20 (2018) 1859–1867, https://doi.org/10.1111/dom.13298. randomization study, Front Endocrinol. (Lausanne) 14 (2023), 1210415, https://
[22] V.S. Costa, A.S. Santos, R.T. Fukui, T.C.C. Mattana, S.R. Matioli, M.E.R. Silva, doi.org/10.3389/fendo.2023.1210415.
Protective effect of interleukin-23A (IL23A) haplotype variants on type 1A [42] M. Ryba-Stanisławowska, M. Skrzypkowska, J. Myśliwska, M. Myśliwiec, The
diabetes mellitus in a Brazilian population, Cytokine 62 (2013) 327–333, https:// serum IL-6 profile and treg/Th17 peripheral cell populations in patients with type
doi.org/10.1016/j.cyto.2013.03.015. 1 diabetes, Mediat. Inflamm. 2013 (2013) 1–7, https://doi.org/10.1155/2013/
[23] N. Eirís, L. González-Lara, J. Santos-Juanes, R. Queiro, E. Coto, P. Coto-Segura, 205284.
Genetic variation at IL12B, IL23R and IL23A is associated with psoriasis severity, [43] K. Singh, E. Kadesjö, J. Lindroos, M. Hjort, M. Lundberg, D. Espes, P.-O. Carlsson,
psoriatic arthritis and type 2 diabetes mellitus, J. Dermatol. Sci. 75 (2014) S. Sandler, L. Thorvaldson, Interleukin-35 administration counteracts established
167–172, https://doi.org/10.1016/j.jdermsci.2014.05.010. murine type 1 diabetes – possible involvement of regulatory T cells, Sci. Rep. 5
[24] M. Ryba-Stanisławowska, K. Rybarczyk-Kapturska, M. Myśliwiec, J. Myśliwska, (2015), 12633, https://doi.org/10.1038/srep12633.
Elevated levels of serum IL-12 and IL-18 are associated with lower frequencies of [44] T.E. O’Sullivan, M. Rapp, X. Fan, O.-E. Weizman, P. Bhardwaj, N.M. Adams,
CD4+CD25highFOXP3+ regulatory T cells in young patients with type 1 T. Walzer, A.J. Dannenberg, J.C. Sun, Adipose-resident group 1 innate lymphoid
diabetes, Inflammation 37 (2014) 1513–1520, https://doi.org/10.1007/s10753- cells promote obesity-associated insulin resistance, Immunity 45 (2016) 428–441,
014-9878-1. https://doi.org/10.1016/j.immuni.2016.06.016.
[25] M. Mishra, H. Kumar, S. Bajpai, R.K. Singh, K. Tripathi, Level of serum IL-12 and [45] A. Geremia, C.V. Arancibia-Cárcamo, M.P.P. Fleming, N. Rust, B. Singh, N.
its correlation with endothelial dysfunction, insulin resistance, proinflammatory J. Mortensen, S.P.L. Travis, F. Powrie, IL-23-responsive innate lymphoid cells are
cytokines and lipid profile in newly diagnosed type 2 diabetes, Diabetes Res. Clin. increased in inflammatory bowel disease, J. Exp. Med. 208 (2011) 1127–1133,
Pract. 94 (2011) 255–261, https://doi.org/10.1016/j.diabres.2011.07.037. https://doi.org/10.1084/jem.20101712.
[26] A.M. Wegeberg, T. Okdahl, S. Riahi, N. Ejskjaer, F. Pociot, J. Størling, B. Brock, [46] R. Castillo-González, A. Valle-Noguera, M.J. Gomez-Sánchez, P. Xia, A. Cruz-
C. Brock, Elevated levels of interleukin-12/23p40 may serve as a potential Adalia, Innate lymphoid cells type 3 in cancer, Front. Immunol. 13 (2022),
indicator of dysfunctional heart rate variability in type 2 diabetes, Cardiovasc 1033252, https://doi.org/10.3389/fimmu.2022.1033252.
Diabetol. 21 (2022), 5, https://doi.org/10.1186/s12933-021-01437-w. [47] Z. Parackova, P. Vrabcova, I. Zentsova, J. Kayserova, I. Richtrova, L. Sojka,
[27] A.-M.L. Wegeberg, T. Okdahl, T. Fløyel, C. Brock, N. Ejskjaer, S. Riahi, F. Pociot, K. Stechova, Z. Sumnik, A. Sediva, Enhanced STAT3 phosphorylation and PD-L1
J. Størling, B. Brock, Circulating inflammatory markers are inversely associated expression in myeloid dendritic cells indicate impaired IL-27Ralpha signaling in
with heart rate variability measures in type 1 diabetes, Mediat. Inflamm. 2020 type 1 diabetes, Sci. Rep. 10 (2020), 493, https://doi.org/10.1038/s41598-020-
(2020), 3590389, https://doi.org/10.1155/2020/3590389. 57507-8.
[28] V.V. Klimontov, K.R. Mavlianova, N.B. Orlov, J.F. Semenova, A.I. Korbut, Serum [48] M. Frangieh, A. McHenry, R. Phillips, C. Ye, A. Bernier, L. Laffel, W. Elyaman, E.
cytokines and growth factors in subjects with type 1 diabetes: associations with M. Bradshaw, IL-27: an endogenous constitutive repressor of human monocytes,
time in ranges and glucose variability, Biomedicines 11 (2023) 2843, https://doi. Clin. Immunol. 217 (2020), 108498, https://doi.org/10.1016/j.
org/10.3390/biomedicines11102843. clim.2020.108498.
[29] C. Nowak, A.C. Carlsson, C.J. Östgren, F.H. Nyström, M. Alam, T. Feldreich, [49] Z. Luo, C. Soläng, R. Larsson, K. Singh, Interleukin-35 prevents the elevation of
J. Sundström, J.-J. Carrero, J. Leppert, P. Hedberg, E. Henriksen, A.C. Cordeiro, the M1/M2 ratio of macrophages in experimental type 1 diabetes, Int. J. Mol. Sci.
V. Giedraitis, L. Lind, E. Ingelsson, T. Fall, J. Ärnlöv, Multiplex proteomics for 23 (2022) 7970, https://doi.org/10.3390/ijms23147970.

10
J. Luo et al. Biomedicine & Pharmacotherapy 170 (2024) 115958

[50] F. Manzoor, M.C. Johnson, C. Li, R.J. Samulski, B. Wang, R. Tisch, β-cell-specific [71] M. Łukawska-Tatarczuk, E. Franek, L. Czupryniak, I. Joniec-Maciejak, A. Pawlak,
IL-35 therapy suppresses ongoing autoimmune diabetes in NOD mice, Eur. J. E. Wojnar, J. Zieliński, D. Mirowska-Guzel, B. Mrozikiewicz-Rakowska, Sirtuin 1,
Immunol. 47 (2017) 144–154, https://doi.org/10.1002/eji.201646493. visfatin and IL-27 serum levels of type 1 diabetic females in relation to
[51] M.D. Sharma, L. Huang, J.-H. Choi, E.-J. Lee, J.M. Wilson, H. Lemos, F. Pan, B. cardiovascular parameters and autoimmune thyroid disease, Biomolecules 11
R. Blazar, D.M. Pardoll, A.L. Mellor, H. Shi, D.H. Munn, An inherently (2021) 1110, https://doi.org/10.3390/biom11081110.
bifunctional subset of Foxp3+ T helper cells is controlled by the transcription [72] W. Xu, R. Zhu, Z. Zhu, K. Yu, Y. Wang, Y. Ding, J. Yu, H. Tang, Q. Zeng, Y. Zhong,
factor eos, Immunity 38 (2013) 998–1012, https://doi.org/10.1016/j. Interleukin-27 ameliorates atherosclerosis in ApoE-/- mice through regulatory T
immuni.2013.01.013. cell augmentation and dendritic cell tolerance, Mediat. Inflamm. 2022 (2022)
[52] M. Tomaszewicz, A. Ronowska, M. Zieliński, A. Jankowska-Kulawy, 1–18, https://doi.org/10.1155/2022/2054879.
P. Trzonkowski, T regulatory cells metabolism: the influence on functional [73] C. Oyarce, A. Vizcaino-Castro, S. Chen, A. Boerma, T. Daemen, Re-polarization of
properties and treatment potential, Front. Immunol. 14 (2023), 1122063, https:// immunosuppressive macrophages to tumor-cytotoxic macrophages by repurposed
doi.org/10.3389/fimmu.2023.1122063. metabolic drugs, Oncoimmunology 10 (2021), 1898753, https://doi.org/
[53] M. Hao, C. Ding, S. Sun, X. Peng, W. Liu, Chitosan/sodium alginate/velvet antler 10.1080/2162402X.2021.1898753.
blood peptides hydrogel promotes diabetic wound healing via regulating [74] S. Ma, D. Zhong, P. Ma, G. Li, W. Hua, Y. Sun, N. Liu, L. Zhang, W. Zhang,
angiogenesis, inflammatory response and skin flora, J. Inflamm. Res 15 (2022) Exogenous hydrogen sulfide ameliorates diabetes-associated cognitive decline by
4921–4938, https://doi.org/10.2147/JIR.S376692. regulating the mitochondria-mediated apoptotic pathway and IL-23/IL-17
[54] K.A. Gallagher, A. Joshi, W.F. Carson, M. Schaller, R. Allen, S. Mukerjee, expression in db/db mice, Cell Physiol. Biochem. 41 (2017) 1838–1850, https://
N. Kittan, E.L. Feldman, P.K. Henke, C. Hogaboam, C.F. Burant, S.L. Kunkel, doi.org/10.1159/000471932.
Epigenetic changes in bone marrow progenitor cells influence the inflammatory [75] P. Chen, L. Lin, X. Xu, Z. Zhang, W. Cai, Z. Shao, S. Chen, X. Chen, Q. Weng,
phenotype and alter wound healing in type 2 diabetes, Diabetes 64 (2015) Liraglutide improved inflammation via mediating IL-23/Th-17 pathway in obese
1420–1430, https://doi.org/10.2337/db14-0872. diabetic mice with psoriasiform skin, J. Dermatol. Treat. 32 (2021) 745–751,
[55] M. Ali, V. Mali, S. Haddox, S.M. AbdelGhany, S.E.M. El-deek, A. Abulfadl, https://doi.org/10.1080/09546634.2019.1708853.
K. Matrougui, S. Belmadani, Essential role of IL-12 in angiogenesis in type 2 [76] J. Komorowska, M. Wątroba, M. Bednarzak, A.D. Grabowska, D. Szukiewicz, The
diabetes, Am. J. Pathol. 187 (2017) 2590–2601, https://doi.org/10.1016/j. role of glucose concentration and resveratrol in modulating neuroinflammatory
ajpath.2017.07.021. cytokines: insights from an in vitro blood-brain barrier model, Med. Sci. Monit. 29
[56] J. Lee, M.P. Rodero, J. Patel, D. Moi, R. Mazzieri, K. Khosrotehrani, Interleukin- (2023), e941044, https://doi.org/10.12659/MSM.941044.
23 regulates interleukin-17 expression in wounds, and its inhibition accelerates [77] D.M. Schwartz, P. Parel, H. Li, A.V. Sorokin, A.R. Berg, M. Chen, A. Dey, C.
diabetic wound healing through the alteration of macrophage polarization, G. Hong, M. Playford, M. Sylvester, H. Teague, E. Siegel, N.N. Mehta, PET/CT-
FASEB J. 32 (2018) 2086–2094, https://doi.org/10.1096/fj.201700773R. based characterization of 18F-FDG uptake in various tissues reveals novel
[57] Y. Hou, L. Zhu, H. Tian, H.-X. Sun, R. Wang, L. Zhang, Y. Zhao, IL-23-induced potential contributions to coronary artery disease in psoriatic arthritis, Front.
macrophage polarization and its pathological roles in mice with imiquimod- Immunol. 13 (2022), 909760, https://doi.org/10.3389/fimmu.2022.909760.
induced psoriasis, Protein Cell 9 (2018) 1027–1038, https://doi.org/10.1007/ [78] E.D. Tait Wojno, N. Hosken, J.S. Stumhofer, A.C. O’Hara, E. Mauldin, Q. Fang, L.
s13238-018-0505-z. A. Turka, S.D. Levin, C.A. Hunter, A role for IL-27 in limiting T regulatory cell
[58] B. Yang, J. Suwanpradid, R. Sanchez-Lagunes, H.W. Choi, P. Hoang, D. Wang, S. populations, J. I. 187 (2011) 266–273, https://doi.org/10.4049/
N. Abraham, A.S. MacLeod, IL-27 facilitates skin wound healing through jimmunol.1004182.
induction of epidermal proliferation and host defense, J. Invest Dermatol. 137 [79] A.N. Sieve, K.D. Meeks, S. Lee, R.E. Berg, A novel immunoregulatory function for
(2017) 1166–1175, https://doi.org/10.1016/j.jid.2017.01.010. IL-23: inhibition of IL-12 dependent IFN-γ production, Eur. J. Immunol. 40 (2010)
[59] C.M. Hull, M. Peakman, T.I.M. Tree, Regulatory T cell dysfunction in type 1 2236–2247, https://doi.org/10.1002/eji.200939759.
diabetes: what’s broken and how can we fix it? Diabetologia 60 (2017) [80] N.S. Mahfooz, M.R. Merling, T.A. Claeys, J.W. Dowling, A. Forero, R.T. Robinson,
1839–1850, https://doi.org/10.1007/s00125-017-4377-1. Human IL-35 inhibits the bioactivity of IL-12 and its interaction with IL-12Rβ2,
[60] H. Zhang, L. Liang, R. Huang, P. Wu, L. He, Comparison of inflammatory Immunohorizons 7 (2023) 431–441, https://doi.org/10.4049/
cytokines levels in the aqueous humor with diabetic retinopathy, Int. immunohorizons.2300039.
Ophthalmol. 40 (2020) 2763–2769, https://doi.org/10.1007/s10792-020-01463- [81] C.S. Frandsen, T.F. Dejgaard, S. Madsbad, Non-insulin drugs to treat
9. hyperglycaemia in type 1 diabetes mellitus, Lancet Diabetes Endocrinol. 4 (2016)
[61] A. Yan, Y. Zhang, X. Wang, Y. Cui, W. Tan, Interleukin 35 regulates interleukin 17 766–780, https://doi.org/10.1016/S2213-8587(16)00039-5.
expression and T helper 17 in patients with proliferative diabetic retinopathy, [82] Z. Meng, X. Liu, T. Li, T. Fang, Y. Cheng, L. Han, B. Sun, L. Chen, The SGLT2
Bioengineered 13 (2022) 13293–13299, https://doi.org/10.1080/ inhibitor empagliflozin negatively regulates IL-17/IL-23 axis-mediated
21655979.2022.2080367. inflammatory responses in T2DM with NAFLD via the AMPK/mTOR/autophagy
[62] C. Diveu, M.J. McGeachy, K. Boniface, J.S. Stumhofer, M. Sathe, B. Joyce-Shaikh, pathway, Int Immunopharmacol. 94 (2021), 107492, https://doi.org/10.1016/j.
Y. Chen, C.M. Tato, T.K. McClanahan, R. de Waal Malefyt, C.A. Hunter, D.J. Cua, intimp.2021.107492.
R.A. Kastelein, IL-27 blocks RORc expression to inhibit lineage commitment of [83] T. Toshimitsu, S. Ozaki, J. Mochizuki, K. Furuichi, Y. Asami, Effects of
Th17 cells, J. Immunol. 182 (2009) 5748–5756, https://doi.org/10.4049/ lactobacillus plantarum strain OLL2712 culture conditions on the anti-
jimmunol.0801162. inflammatory activities for murine immune cells and obese and type 2 diabetic
[63] S. Wu, R. Ma, Y. Zhong, Z. Chen, H. Zhou, M. Zhou, W. Chong, J. Chen, Deficiency mice, Appl. Environ. Microbiol. 83 (2017), e03001-16, https://doi.org/10.1128/
of IL-27 signaling exacerbates experimental autoimmune uveitis with elevated AEM.03001-16.
uveitogenic Th1 and Th17 responses, Int. J. Mol. Sci. 22 (2021) 7517, https://doi. [84] F. Zamani, S. Almasi, T. Kazemi, R. Jahanban Esfahlan, M.R. Aliparasti, New
org/10.3390/ijms22147517. approaches to the immunotherapy of type 1 diabetes mellitus using interleukin-
[64] M.E. Houssen, M.A.B. El-Hussiny, A. El-Kannishy, D. Sabry, R. El Mahdy, M. 27, Adv. Pharm. Bull. 5 (2015) 599–603, https://doi.org/10.15171/
E. Shaker, Serum and aqueous humor concentrations of interleukin-27 in diabetic apb.2015.081.
retinopathy patients, Int. Ophthalmol. 38 (2018) 1817–1823, https://doi.org/ [85] A.L. Zirnheld, M. Villard, A.M. Harrison, M.M. Kosiewicz, P. Alard, β-catenin
10.1007/s10792-017-0655-7. stabilization in NOD dendritic cells increases IL-12 production and subsequent
[65] J. Wu, Y. Zhong, S. Yue, K. Yang, G. Zhang, L. Chen, L. Liu, Aqueous humor induction of IFN-γ-producing T cells, J. Leukoc. Biol. 106 (2019) 1349–1358,
mediator and cytokine aberrations in diabetic retinopathy and diabetic macular https://doi.org/10.1002/JLB.3A0919-244R.
edema: a systematic review and meta-analysis, Dis. Markers 2019 (2019), [86] W. Azeem, R.M. Bakke, S. Appel, A.M. Øyan, K.-H. Kalland, Dual pro- and anti-
6928524, https://doi.org/10.1155/2019/6928524. inflammatory features of monocyte-derived dendritic cells, Front. Immunol. 11
[66] H. Xu, M. Cai, X. Zhang, Effect of the blockade of the IL-23-Th17-IL-17A pathway (2020), 438, https://doi.org/10.3389/fimmu.2020.00438.
on streptozotocin-induced diabetic retinopathy in rats, Graefes Arch. Clin. Exp. [87] A. Billich, Drug evaluation: apilimod, an oral IL-12/IL-23 inhibitor for the
Ophthalmol. 253 (2015) 1485–1492, https://doi.org/10.1007/s00417-014-2842- treatment of autoimmune diseases and common variable immunodeficiency,
9. IDrugs 10 (2007) 53–59.
[67] P. Davenport, P.G. Tipping, The role of interleukin-4 and interleukin-12 in the [88] K. Fujihira, M. Nagata, H. Moriyama, H. Yasuda, K. Arisawa, M. Nakayama, S.
progression of atherosclerosis in apolipoprotein E-deficient mice, Am. J. Pathol. Maeda, M. Kasuga, K. Okumura, H. Yagita, K. Yokono, Suppression and
163 (2003) 1117–1125, https://doi.org/10.1016/S0002-9440(10)63471-2. acceleration of autoimmune diabetes by neutralization of endogenous
[68] M. Subramanian, E. Thorp, I. Tabas, Identification of a non-growth factor role for interleukin-12 in NOD Mice, 49 (2000) 9.
GM-CSF in advanced atherosclerosis: promotion of macrophage apoptosis and [89] A.K. Marwaha, S. Chow, A.M. Pesenacker, L. Cook, A. Sun, S.A. Long, J.H.
plaque necrosis through IL-23 signaling, Circ. Res 116 (2015), e13-24, https:// M. Yang, K.A. Ward-Hartstonge, E. Williams, C. Domingo-Vila, K. Halani, K.
doi.org/10.1161/CIRCRESAHA.116.304794. M. Harris, T.I.M. Tree, M.K. Levings, T. Elliott, R. Tan, J.P. Dutz, A phase 1b open-
[69] Y. Shao, W.Y. Yang, F. Saaoud, C. Drummer, Y. Sun, K. Xu, Y. Lu, H. Shan, E. label dose-finding study of ustekinumab in young adults with type 1 diabetes,
M. Shevach, X. Jiang, H. Wang, X. Yang, IL-35 promotes CD4+Foxp3+ Tregs and Immunother. Adv. 2 (2021), ltab022, https://doi.org/10.1093/immadv/ltab022.
inhibits atherosclerosis via maintaining CCR5-amplified Treg-suppressive [90] G.T. Nepom, M. Ehlers, T. Mandrup-Poulsen, Anti-cytokine therapies in T1D:
mechanisms, JCI Insight 6 (2021), e152511, https://doi.org/10.1172/jci. concepts and strategies, Clin. Immunol. 149 (2013) 279–285, https://doi.org/
insight.152511. 10.1016/j.clim.2013.02.003.
[70] T. Hirase, H. Hara, Y. Miyazaki, N. Ide, A. Nishimoto-Hazuku, H. Fujimoto, C.J. [91] Z. Yao, C. Hu, Y. Zhu, Z. Xu, B. Randazzo, Y. Wasfi, Y. Chen, A. Sharma, H. Zhou,
M. Saris, H. Yoshida, K. Node, Interleukin 27 inhibits atherosclerosis via Population pharmacokinetic modeling of guselkumab, a human IgG1λ
immunoregulation of macrophages in mice, Am. J. Physiol. -Heart Circ. Physiol. monoclonal antibody targeting IL-23, in patients with moderate to severe plaque
305 (2013) H420–H429, https://doi.org/10.1152/ajpheart.00198.2013. psoriasis, J. Clin. Pharmacol. 58 (2018) 613–627, https://doi.org/10.1002/
jcph.1063.

11
J. Luo et al. Biomedicine & Pharmacotherapy 170 (2024) 115958

[92] S. Kubo, S. Nakayamada, K. Sakata, Y. Kitanaga, X. Ma, S. Lee, A. Ishii, [114] X. Zhu, Z. Liu, J.-Q. Liu, J. Zhu, J. Zhang, J.P. Davis, J. Chu, J. Yu, J. Zhou, M.-
K. Yamagata, K. Nakano, Y. Tanaka, Janus kinase inhibitor baricitinib modulates S. Li, X.-F. Bai, Systemic delivery of IL-27 by an adeno-associated viral vector
human innate and adaptive immune system, Front Immunol. 9 (2018), 1510, inhibits T cell-mediated colitis and induces multiple inhibitory pathways in T
https://doi.org/10.3389/fimmu.2018.01510. cells, J. Leukoc. Biol. 100 (2016) 403–411, https://doi.org/10.1189/jlb.3A1215-
[93] M. Waibel, H.E. Thomas, J.M. Wentworth, J.J. Couper, R.J. MacIsaac, F. 540R.
J. Cameron, M. So, B. Krishnamurthy, M.C. Doyle, T.W. Kay, Investigating the [115] A.A. Marin, R.E. Decker, S. Kumar, Z. Lamantia, H. Yokota, T. Emrick, M.
efficacy of baricitinib in new onset type 1 diabetes mellitus (BANDIT)-study L. Figueiredo, Enhancing prednisone-based arthritis therapy with targeted IL-27
protocol for a phase 2, randomized, placebo controlled trial, Trials 23 (2022), gene delivery, Bioengineering 9 (2022) 248, https://doi.org/10.3390/
433, https://doi.org/10.1186/s13063-022-06356-z. bioengineering9060248.
[94] I.M. Khan, X.Y. Perrard, G. Brunner, H. Lui, L.M. Sparks, S.R. Smith, X. Wang, Z.- [116] H. Guo, B. Li, W. Wang, N. Zhao, H. Gao, Mesenchymal stem cells overexpressing
Z. Shi, D.E. Lewis, H. Wu, C.M. Ballantyne, Intermuscular and perimuscular fat IL-35: a novel immunosuppressive strategy and therapeutic target for inducing
expansion in obesity correlates with skeletal muscle T cell and macrophage transplant tolerance, Stem Cell Res. Ther. 9 (2018), 254, https://doi.org/
infiltration and insulin resistance, Int J. Obes. 39 (2015) 1607–1618, https://doi. 10.1186/s13287-018-0988-9.
org/10.1038/ijo.2015.104. [117] G. Mondanelli, C. Volpi, R. Bianchi, M. Allegrucci, V.N. Talesa, U. Grohmann, M.
[95] Z.-D. Yang, M. Chen, R. Wu, M. McDuffie, J.-L. Nadler, The anti-inflammatory L. Belladonna, Islet antigen-pulsed dendritic cells expressing ectopic IL-35Ig
compound lisofylline prevents Type I diabetes in non-obese diabetic mice, protect nonobese diabetic mice from autoimmune diabetes, Cytokine 75 (2015)
Diabetologia 45 (2002) 1307–1314, https://doi.org/10.1007/s00125-002-0901- 380–388, https://doi.org/10.1016/j.cyto.2015.05.002.
y. [118] X. Zou, S. Wang, Y. Zhang, X. Wang, R. Zhang, W. Yang, Y. Li, AIRE-
[96] J.S. Striffler, J.L. Nadler, Lisofylline, a novel anti-inflammatory agent, enhances overexpressing BMDCs suppress TFH cells through ICOSL to prevent and
glucose-stimulated insulin secretion in vivo and in vitro: studies in prediabetic attenuate autoimmune diabetes in NOD mice, Int. Immunopharmacol. 99 (2021),
and normal rats, Metabolism 53 (2004) 290–296, https://doi.org/10.1016/j. 107979, https://doi.org/10.1016/j.intimp.2021.107979.
metabol.2003.10.008. [119] M. Tenspolde, K. Zimmermann, L.C. Weber, M. Hapke, M. Lieber, J. Dywicki,
[97] Z. Yang, M. Chen, L.B. Fialkow, J.D. Ellett, R. Wu, J.L. Nadler, Inhibition of A. Frenzel, M. Hust, M. Galla, L.E. Buitrago-Molina, M.P. Manns, E. Jaeckel,
STAT4 activation by lisofylline is associated with the protection of autoimmune M. Hardtke-Wolenski, Regulatory T cells engineered with a novel insulin-specific
diabetes, Ann. N. Y. Acad. Sci. 1005 (2003) 409–411, https://doi.org/10.1196/ chimeric antigen receptor as a candidate immunotherapy for type 1 diabetes,
annals.1288.069. J. Autoimmun. 103 (2019), 102289, https://doi.org/10.1016/j.jaut.2019.05.017.
[98] M. Ali, M.H. Bakr, L.A. Abdelzaher, S.A. Sayed, V. Mali, A.A. Desai, E. Radwan, [120] L. Zhang, T. Sosinowski, A.R. Cox, J.R. Cepeda, N.S. Sekhar, S.M. Hartig, D. Miao,
Lisofylline mitigates cardiac inflammation in a mouse model of obesity through L. Yu, M. Pietropaolo, H.W. Davidson, Chimeric antigen receptor (CAR) T cells
improving insulin secretion and activating cardiac AMPK signaling pathway, targeting a pathogenic MHC class II:peptide complex modulate the progression of
Cytokine 138 (2021), 155398, https://doi.org/10.1016/j.cyto.2020.155398. autoimmune diabetes, J. Autoimmun. 96 (2019) 50–58, https://doi.org/10.1016/
[99] K.S. Italiya, S. Mazumdar, S. Sharma, D. Chitkara, R.I. Mahato, A. Mittal, Self- j.jaut.2018.08.004.
assembling lisofylline-fatty acid conjugate for effective treatment of diabetes [121] H. Yu, R. Paiva, R.A. Flavell, Harnessing the power of regulatory T-cells to control
mellitus, Nanomedicine 15 (2019) 175–187, https://doi.org/10.1016/j. autoimmune diabetes: overview and perspective, Immunology 153 (2018)
nano.2018.09.014. 161–170, https://doi.org/10.1111/imm.12867.
[100] S. Wilhelm, C. Carter, M. Lynch, T. Lowinger, J. Dumas, R.A. Smith, B. Schwartz, [122] J. Boldison, L.C. Da Rosa, J. Davies, L. Wen, F.S. Wong, Dendritic cells license
R. Simantov, S. Kelley, Discovery and development of sorafenib: a multikinase regulatory B cells to produce IL-10 and mediate suppression of antigen-specific
inhibitor for treating cancer, Nat. Rev. Drug Discov. 5 (2006) 835–844, https:// CD8 T cells, Cell Mol. Immunol. 17 (2020) 843–855, https://doi.org/10.1038/
doi.org/10.1038/nrd2130. s41423-019-0324-z.
[101] Q. Zeng, J. Song, D. Wang, X. Sun, Y. Xiao, H. Zhang, Y. Xiao, Z. Zhou, T. Deng, [123] T.M. Hummel, T. Ackfeld, M. Schönberg, G. Ciupka, F. Schulz, A. Oberdoerster,
Identification of Sorafenib as a Treatment for Type 1 Diabetes, Front. Immunol. J. Grötzinger, J. Scheller, D.M. Floss, Synthetic deletion of the interleukin 23
13 (2022), 740805, https://doi.org/10.3389/fimmu.2022.740805. receptor (IL-23R) stalk region led to autonomous IL-23R homodimerization and
[102] A.K. Marwaha, S. Tan, J.P. Dutz, Targeting the IL-17/IFN-γ axis as a potential new activation, Mol. Cell Biol. 37 (2017), e00014-17, https://doi.org/10.1128/
clinical therapy for type 1 diabetes, Clin. Immunol. 154 (2014) 84–89, https:// MCB.00014-17.
doi.org/10.1016/j.clim.2014.06.006. [124] I. Moraga, J.B. Spangler, J.L. Mendoza, M. Gakovic, T.S. Wehrman, P. Krutzik, K.
[103] Z. Yang, M. Chen, J.L. Nadler, Lisofylline: a potential lead for the treatment of C. Garcia, Synthekines are surrogate cytokine and growth factor agonists that
diabetes, Biochem. Pharmacol. 69 (2005) 1–5, https://doi.org/10.1016/j. compel signaling through non-natural receptor dimers, eLife 6 (2017), e22882,
bcp.2004.08.012. https://doi.org/10.7554/eLife.22882.
[104] J. Forycka, J. Hajdys, J. Krzemińska, P. Wilczopolski, M. Wronka, E. Młynarska, [125] R.R. Flores, E. Kim, L. Zhou, C. Yang, J. Zhao, A. Gambotto, P.D. Robbins, IL-Y, a
J. Rysz, B. Franczyk, New insights into the use of empagliflozin-a comprehensive synthetic member of the IL-12 cytokine family, suppresses the development of
review, Biomedicines 10 (2022) 3294, https://doi.org/10.3390/ type 1 diabetes in NOD mice, Eur. J. Immunol. 45 (2015) 3114–3125, https://doi.
biomedicines10123294. org/10.1002/eji.201445403.
[105] S.A. Tersey, J.D. Carter, L. Rosenberg, D.A. Taylor-Fishwick, R.G. Mirmira, J. [126] K. Forslund, F. Hildebrand, T. Nielsen, G. Falony, E. Le Chatelier, S. Sunagawa,
L. Nadler, Amelioration of type 1 diabetes following treatment of non-obese E. Prifti, S. Vieira-Silva, V. Gudmundsdottir, H.K. Pedersen, M. Arumugam,
diabetic mice with INGAP and lisofylline, J. Diabetes Mellit. 2 (2012) 251–257, K. Kristiansen, A.Y. Voigt, H. Vestergaard, R. Hercog, P.I. Costea, J.R. Kultima,
https://doi.org/10.4236/jdm.2012.22040. J. Li, T. Jørgensen, F. Levenez, J. Dore, MetaH.I.T. consortium, H.B. Nielsen,
[106] W. Peng, L. Wang, H. Zhang, Z. Zhang, X. Chen, Effects of recombinant IL-35-BCG S. Brunak, J. Raes, T. Hansen, J. Wang, S.D. Ehrlich, P. Bork, O. Pedersen,
on Treg/Th17 cell imbalance and inflammatory response in asthmatic newborn Disentangling type 2 diabetes and metformin treatment signatures in the human
mice induced by RSV, Inflammation 44 (2021) 2476–2485, https://doi.org/ gut microbiota, Nature 528 (2015) 262–266, https://doi.org/10.1038/
10.1007/s10753-021-01517-9. nature15766.
[107] C. Tan, S. Tan, H. Zhang, M. Zhang, H. Fan, Z. Nan, X. Liu, W. Wang, L. Zhang, [127] P. de Groot, T. Nikolic, S. Pellegrini, V. Sordi, S. Imangaliyev, E. Rampanelli,
S. Deng, D. Zuo, Q. Tang, Enhanced migration and immunoregulatory capacity of N. Hanssen, I. Attaye, G. Bakker, G. Duinkerken, A. Joosten, A. Prodan, E. Levin,
BMSCs mediated by overexpression of CXCR4 and IL-35, Mol. Immunol. 150 H. Levels, B.P. van Loon, A. van Bon, C. Brouwer, S. van Dam, S. Simsek, D. van
(2022) 1–8, https://doi.org/10.1016/j.molimm.2022.07.005. Raalte, F. Stam, V. Gerdes, R. Hoogma, M. Diekman, M. Gerding, C. Rustemeijer,
[108] K.S. Italiya, D. Chitkara, Self-assembling lisofylline-fatty acid conjugate for B. de Bakker, J. Hoekstra, A. Zwinderman, J. Bergman, F. Holleman, L. Piemonti,
effective treatment of diabetes mellitus, (n.d.) 43. W.D. Vos, B. Roep, M. Nieuwdorp, Faecal microbiota transplantation halts
[109] K.S. Italiya, A.K. Singh, D. Chitkara, A. Mittal, Nanoparticulate tablet dosage form progression of human new-onset type 1 diabetes in a randomised controlled trial,
of lisofylline-linoleic acid conjugate for type 1 diabetes: in situ single-pass Gut 70 (2021) 92–105, https://doi.org/10.1136/gutjnl-2020-322630.
intestinal perfusion (SPIP) studies and pharmacokinetics in rat, AAPS [128] T.U. Greiner, T. Hyötyläinen, M. Knip, F. Bäckhed, M. Orešič, The gut microbiota
PharmSciTech 22 (2021), 114, https://doi.org/10.1208/s12249-021-01980-5. modulates glycaemic control and serum metabolite profiles in non-obese diabetic
[110] M.Y. Donath, C.A. Dinarello, T. Mandrup-Poulsen, Targeting innate immune mice, PLOS One 9 (2014), e110359, https://doi.org/10.1371/journal.
mediators in type 1 and type 2 diabetes, Nat. Rev. Immunol. 19 (2019) 734–746, pone.0110359.
https://doi.org/10.1038/s41577-019-0213-9. [129] M. Lo Conte, M. Antonini Cencicchio, M. Ulaszewska, A. Nobili, I. Cosorich,
[111] M. Bettini, A.H. Castellaw, G.P. Lennon, A.R. Burton, D.A.A. Vignali, Prevention R. Ferrarese, L. Massimino, A. Andolfo, F. Ungaro, N. Mancini, M. Falcone, A diet
of autoimmune diabetes by ectopic pancreatic β-cell expression of interleukin-35, enriched in omega-3 PUFA and inulin prevents type 1 diabetes by restoring gut
Diabetes 61 (2012) 1519–1526, https://doi.org/10.2337/db11-0784. barrier integrity and immune homeostasis in NOD mice, Front. Immunol. 13
[112] M. Miralles, H. Eixarch, M. Tejero, C. Costa, K. Hirota, A.R. Castaño, M. Puig, (2022), 1089987, https://doi.org/10.3389/fimmu.2022.1089987.
G. Stockinger, X. Montalban, A. Bosch, C. Espejo, M. Chillon, Clinical and [130] S. Lavoie, E. Chun, S. Bae, C.A. Brennan, C.A. Gallini Comeau, J.K. Lang,
histopathological amelioration of experimental autoimmune encephalomyelitis M. Michaud, H.R. Hoveyda, G.L. Fraser, M.H. Fuller, B.T. Layden, J.N. Glickman,
by AAV vectors expressing a soluble interleukin-23 receptor, Neurotherapeutics W.S. Garrett, Expression of free fatty acid receptor 2 by dendritic cells prevents
14 (2017) 1095–1106, https://doi.org/10.1007/s13311-017-0545-8. their expression of interleukin 27 and is required for maintenance of mucosal
[113] J. Zhu, J.-Q. Liu, Z. Liu, L. Wu, M. Shi, J. Zhang, J.P. Davis, X.-F. Bai, Interleukin- barrier and immune response against colorectal tumors in mice, Gastroenterology
27 gene therapy prevents the development of autoimmune encephalomyelitis but 158 (2020) 1359–1372.e9, https://doi.org/10.1053/j.gastro.2019.12.027.
fails to attenuate established inflammation due to the expansion of CD11b+Gr-1+ [131] J. Alameddine, E. Godefroy, L. Papargyris, G. Sarrabayrouse, J. Tabiasco,
myeloid cells, Front. Immunol. 9 (2018), 873, https://doi.org/10.3389/ C. Bridonneau, K. Yazdanbakhsh, H. Sokol, F. Altare, F. Jotereau,
fimmu.2018.00873. Faecalibacterium prausnitzii skews human DC to prime IL10-producing t cells

12
J. Luo et al. Biomedicine & Pharmacotherapy 170 (2024) 115958

through TLR2/6/JNK signaling and IL-10, IL-27, CD39, and IDO-1 induction, [133] K. Mao, A.P. Baptista, S. Tamoutounour, L. Zhuang, N. Bouladoux, A.J. Martins,
Front. Immunol. 10 (2019), 143, https://doi.org/10.3389/fimmu.2019.00143. Y. Huang, M.Y. Gerner, Y. Belkaid, R.N. Germain, Innate and adaptive
[132] X. Su, M. Zhang, H. Qi, Y. Gao, Y. Yang, H. Yun, Q. Zhang, X. Yang, Y. Zhang, lymphocytes sequentially shape the gut microbiota and lipid metabolism, Nature
J. He, Y. Fan, Y. Wang, P. Guo, C. Zhang, R. Yang, Gut microbiota-derived 554 (2018) 255–259, https://doi.org/10.1038/nature25437.
metabolite 3-idoleacetic acid together with LPS induces IL-35+ B cell generation,
Microbiome 10 (2022), 13, https://doi.org/10.1186/s40168-021-01205-8.

13

You might also like