Vous êtes sur la page 1sur 10

Cytokine and Growth Factor Reviews 44 (2018) 18–27

Contents lists available at ScienceDirect

Cytokine and Growth Factor Reviews


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

Potential roles of IL-1 subfamily members in glycolysis in disease T


a,1 a,1 a,1 a b c
Qi Tan , Qi Huang , Yan Ling Ma , KaiMin Mao , GuangHai Yang , Ping Luo ,
GuanZhou Maa, PeiYuan Meib, Yang Jina,

a
Department of Respiratory and Critical Care Medicine, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of
Science and Technology, Wuhan, 430022, China
b
Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
c
Center for Translational Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China

ARTICLE INFO ABSTRACT

Keywords: The interleukin-(IL)-1 subfamily consists of IL-1α, IL-1β, IL-1 receptor antagonist IL-1Ra and IL-33. These cy-
IL-1α tokines are the main members of the IL-1 family and have been widely recognized as having significant roles in
IL-1β pro-inflammatory and immunomodulatory actions. Mounting evidence has revealed that these cytokines also
IL-1Ra play key roles in the regulation of glycolysis, which is an important metabolic pathway in most organisms that
IL-33
provides energy. Dysregulation of glycolysis is associated with various diseases, including type 2 diabetes,
Glycolysis
rheumatoid arthritis (RA) and cancer. We reviewed studies addressing the important roles of IL-1 subfamily
Metabolism
cytokines, with particular focus on their ability to regulate glycolysis in disease states. In this review, we
summarize the potential roles of IL-1 subfamily members in glycolysis in disease states and address the un-
derlying mechanisms. Furthermore, we discuss the potential of these cytokines as therapeutic targets in clinical
applications to provide insight into possible therapeutic strategies for treatment, especially for cancers.

1. Introduction major regulators, playing active roles in pro-inflammatory and im-


munomodulatory actions and include mainly the IL-1 subfamily cyto-
Interleukin (IL) was first described in studies on fever pathogenesis kines, which were the first IL-1 cytokines discovered and have been
by Menkin and Beeson in 1943–1948, and it functions as a major reg- widely studied since [6]. Recent research has revealed that IL-1 sub-
ulator of responses to infections or sterile insults [1]. Subsequent stu- family cytokines are involved in the pathogenesis of several diseases,
dies on the origin and function of this regulator led to the name “in- including type 2 diabetes [7], rheumatoid arthritis (RA) [8] and cancer
terleukin,” which was used to describe the soluble factors secreted by [9], through directly or indirectly targeting the process of glycolysis,
macrophages and lymphocytes. Thus, IL-1 was used to describe the which is an important metabolic pathway for providing energy in most
macrophage product [2]. Subsequent research has revealed that IL-1 is organisms [10]. This phenomenon was first reported by Taylor and
a product not only of macrophages but also of neutrophils, dendritic coworkers in 1988; in their study, IL-1α and IL-1β were implicated in
cells and NK cells [3]. At present, the IL-1 family consists of 12 cyto- the increased glycolysis of rheumatoid synovial cells in RA [8]. Sub-
kines (IL-1α, IL-1β, IL-1Ra, IL-33, IL-18, IL-18BP, IL-37, IL-36α, IL-36β, sequently, these authors obtained consistent results with human dermal
IL-36γ, IL-36Ra and IL-38). These cytokines have similar gene struc- fibroblasts (HDF) from familial adenopolyposis (FAP) of the colon and
tures and exert their biological properties via binding to specific severe sepsis states in vitro, which suggested that the changes in gly-
membrane receptors such as intracellular Toll/IL-1 receptor (TIR) do- colysis induced by these two cytokines were the molecular mechanisms
mains or extracellular immunoglobulin-like binding domains on target underlying the disease, a possibility warranting further study [11,12].
cells [4]. Furthermore, according to the lengths of their precursors, Since those studies, many experiments have been conducted to in-
these cytokines can be classified into three subfamilies [1]: the IL-1 vestigate the roles of the IL-1 subfamily in glycolysis. In this review, we
subfamily (IL-1α, IL-1β, IL-1Ra, IL-33) [2], the IL-18 subfamily (IL-18, summarize the major research on the regulation of glycolysis by IL-1
IL-18BP, IL-37) and [3] the IL-36 subfamily (IL-36α, IL-36β, IL-36γ, IL- subfamily members and discuss the underlying mechanisms that result
36Ra, IL-38) [4,5]. Members of the IL-1 family are well recognized as in disease.

Corresponding author.

E-mail addresses: m201775412@hust.edu.cn (Q. Tan), huangqi66@126.com (Q. Huang), mayanling811@hust.edu.cn (Y.L. Ma), 444931158@qq.com (K. Mao),
2004XH0838@hust.edu.cn (G. Yang), 985066021@qq.com (P. Luo), 837324889@qq.com (G. Ma), M13260630812@163.com (P. Mei), whuhjy@126.com (Y. Jin).
1
Qi Tan, Qi Huang and YanLing Ma contributed equally to this work.

https://doi.org/10.1016/j.cytogfr.2018.11.001
Received 4 November 2018; Received in revised form 8 November 2018; Accepted 12 November 2018
Available online 16 November 2018
1359-6101/ © 2018 The Authors. Published by Elsevier Ltd. This is an open access article under the CC BY-NC-ND license
(http://creativecommons.org/licenses/BY-NC-ND/4.0/).
Q. Tan et al. Cytokine and Growth Factor Reviews 44 (2018) 18–27

Glycolysis, which was first clearly described by Meyerhof et al in the associated with inflammatory factor production and immune regulation
1940s, is usually defined as the conversion of glucose to lactate with the [23]. Accordingly, a major property of IL-1α is the mediation of the
synthesis of adenosine triphosphate (ATP) under anaerobic conditions early phases of sterile inflammation and the T helper (Th) 17 response.
[10,13]. Although glycolysis is a low-efficiency process that produces 2 In addition, IL-1α has been demonstrated to play a role in the nucleus
ATP molecules from 1 molecule of glucose, it can synthesize ATP ra- by stimulating transcription.
pidly. Accordingly, it plays an essential role in providing energy for
most organisms, especially for metabolically active cells such as cancer 2.2. IL-1β
cells [14]. Under aerobic conditions, glycolysis is necessary for the
survival and growth of cancer cells. Accumulating evidence suggests IL-1β (IL-1F2) is another member of IL-1 subfamily. It has biological
that cancer cells prefer to synthesize ATP through glycolysis even when properties similar to those of IL-1α due to its binding to the same re-
there is a sufficient oxygen supply. This phenomenon was first de- ceptor, IL-1R1. IL-1β can also activate NF-κB and AP-1 through the
scribed by Otto Warburg in the 1920s; thus, “aerobic glycolysis” is also IRAK pathway and activates the PI3K pathway leading to NF-κB acti-
known as “the Warburg effect” [15]. In general, glycolysis plays pivotal vation. Accumulating evidence indicates that the PI3K/protein kinase B
roles in energy production and cell proliferation. It has been reported (Akt)/mammalian target of rapamycin (mTOR) signaling pathway,
that the dysregulation of glycolysis is responsible for various human which is recognized as a regulatory pathway in glycolysis (described
diseases, such as type 2 diabetes, RA and cancer. Evidence that this below), is involved in IL-1β-dependent signal transduction [24]. How-
dysregulation can result in disease comprises the following [1]: De- ever, unlike IL-1α, IL-1β is only expressed by a small number of cell
creased glucose utilization in pancreatic islets leads to type 2 diabetes types including monocytes, macrophages and dendritic cells. In addi-
mellitus [7,2]. Increased levels of lactate, which is the product of gly- tion, the IL-1β precursor is not active, and its synthesis requires the
colysis in the rheumatoid joint, are associated with RA [8,3]. Lactate induction of transcription factor NF-κB after the stimulation of innate
acts as the most important energy source for cancer cells, and inhibiting immune cells, such as macrophages and dendritic cells. The synthesis of
glycolysis can inhibit tumor growth [9]. Understanding the mechanisms IL-1β precursor can also be regulated by tumor necrosis factor-alpha
of glycolysis dysregulation can lead to the development of novel ther- (TNFα), IL-18 or itself [25]. However, to increase the potency of the
apeutic strategies for treating disease. Several studies have identified final product, the IL-1β precursor has to be cleaved via proteolytic
correlations between the levels of regulating factors, including those of processing by caspase-1. The activation of caspase-1 is dependent on a
the IL-1 subfamily, and the dysregulation of glycolysis. Therefore, the complex called the inflammasome, which is associated with cyto-
present review focuses in particular on the roles of IL-1 subfamily plasmic pattern recognition receptor signaling [26]. Another distinction
members in glycolysis and their potentials as therapeutic targets for between IL-1α and IL-1β is in their ability to bind IL-R2; IL-R2 binds to
treating associated diseases. IL-1β with high affinity. However, IL-R2 is unable to initiate signaling
[27]. Major biological functions of IL-1β include the mediation of in-
2. Biological features of cytokines in the IL-1 subfamily flammation and the regulation of the immune response via the activa-
tion of Th1 and Th17 cells.
2.1. IL-1α
2.3. IL-1Ra
IL-1α (IL-1F1) is one of most studied members of the IL-1 subfamily
due to its multiple effects and the fact that it was the first member IL-1Ra, also designated as IL-1LF3, functions as a natural antagonist
discovered [16]. IL-1α is constitutively expressed in many types of cells for both IL-1α and IL-1β and is produced by monocytes and macro-
under normal conditions, especially in epithelial cells, and its precursor phages. All three cytokines share the same receptors, i.e., IL-1R1 and IL-
is synthesized and stored in the cytoplasm. The conversion of pro-IL-1α R2. Although IL-1Ra binds to specific receptors with almost the same
to a mature 17-kDa protein requires a Ca2+-activated protease, calpain affinity as IL-1α and IL-1β, such binding does not initiate signal
[17]. However, both forms of IL-1α are fully active and are released transduction [17]. Accordingly, the function of IL-1Ra is to inhibit the
from intracellular to extracellular space under necrosis or pyroptosis. As activity of IL-1α or IL-1β via competing with them for receptor binding
a result, IL-1α in the extracellular space is an indicator of cell stress due sites. The biological features of IL-1Ra have important medical appli-
to infection, injury or hypoxia. Accordingly, IL-1α is used as a damage- cations; inhibition by IL-1Ra is utilized clinically to monitor treatment
associated molecular pattern molecule for danger signals involved in responses of RA and type 2 diabetes mellitus [28,29]. In addition, there
inflammatory responses [18,19]. Following the release of IL-1α, its is ongoing research to treat pancreatic cancer using IL-1Ra by targeting
function is exerted primarily through IL-1 receptor type 1 (IL-1R1). For IL-1α [30].
signaling to occur, the activation of signal transduction of IL-1/IL-1RI
complexes is necessary, which requires the IL-1 receptor accessory 2.4. IL-33
protein (IL-1RAcP). IL-1RAcP functions as a coreceptor and is recruited
when IL-1α binds to IL-1R1. Interestingly, IL-1RAcP is also necessary IL-33 is a newly identified cytokine in the IL-1 subfamily and is
for IL-1β and IL-33 (see below) [20]. IL-1α binding causes the assem- mainly expressed in normal endothelial stromal, epithelial and en-
bling of signaling modules, which are formed by IL-1, IL-1RI, IL-1RAcP, dothelial cells. IL-33 acts as an alarm, such as when IL-1α is released
myeloid differentiation primary response gene 88 (MYD88) and IL-1 upon cell injury or tissue damage. Generally, IL-33 is converted into
receptor–activated protein kinase (IRAK)-4. Through the phosphoryla- mature bioactive forms by neutrophil elastase and cathepsin G and
tion of IRAK-4, IRAK-1 and IRAK-2 are activated; this activation is as- calpain [31]. Mature IL-33 initiates its signaling pathway through the
sociated with the recruitment of tumor necrosis factor–associated factor formation of the IL-33/ST2L/IL1-RAcP complex, which is mainly ex-
6 (TRAF6). TRAF6 activates two pathways. One pathway signals pressed in Th2 cells. Mechanically, IL-33 mediates its immune response
through mitogen-activated protein kinase (MAPK) to trigger the acti- primarily via polarized Th2 cells [32]. The signal transduction is similar
vation of P38, c-Jun N-terminal kinases (JNKs), extracellular signal- to that triggered by IL-1α. The IL-33/ST2L/IL1-RAcP complex also
regulated kinases (ERKs) and activator protein 1 (AP-1), and the other causes the recruitment of MyD88 and further induces IRAK1/4, which
pathway leads to the activation of nuclear factor κB (NF-κB) tran- in turn activates TRAF6 and consequently drives the production of NF-
scription factors [21,22]. IL-1α also signals through mitogen-activated κB. As a result, IL-33 has been described as a nuclear factor with
phosphatidylinositol-3-phosphate (PI3K), which functions as a med- transcriptional regulatory properties that interacts with the activation
iator of glycolysis (see below), to activate NF-κB and thereby induce of NF-κB as well as the P53 submit [33]. IL-33 is a “dual cytokine” that
gene expression, including the expression of IL-6 and IL-8, which is exerts its regulatory and transcriptional effects by serving as a regular

19
Q. Tan et al. Cytokine and Growth Factor Reviews 44 (2018) 18–27

Table 1
Biological features of cytokines in the IL-1 subfamily and targeted agents.
IL-1 subfamily Family name Specific receptor Coreceptor Activity Targeted agents
(s)

IL-1α IL-1F1 IL-1RI;IL-1R2 IL-1RAcP Alarmin;Pro-inflammation; Th17 cell response;Nuclear Anakinra;Rilonacept;MABp1


transcription
IL-1β IL-1F2 IL-1RI;IL-1R2 IL-1RAcP Th17 cell response;Pro-inflammation; Anakinra;Rilonacept;Canakinumab
IL-1Ra IL-1F3 IL-1RI None Anti-inflammation None
IL-33 IL-1F4 ST2 IL-1RAcP Alarmin; Pro-inflammation; Th2 response;Nuclear transcription ANB020;CNTO7160

Fig. 1. Signaling pathways of the IL-1 subfamily and currently available inhibitors.

cytokine or a nuclear transcription factor. transporter (GLUT)1, hexokinase (HK)2, phosphofructokinase (PFK)
The biological features of the IL-1 subfamily members are sum- and enolase 1 (ENO1) as well as lactate dehydrogenase A (LDHA)
marized in Table 1, and their signaling pathways are described in Fig. 1. [34–37]. The latter pathway is activated by the ligand-bound receptor
(Table 2) tyrosine kinases (RTKs) through Akt and directly activates c-Myc
transcription factor and hypoxia inducible factor-1α (HIF-1α) to reg-
3. A review of glycolysis ulate GLUT1 and several glycolytic enzymes, including HK2, PFK, al-
dolase A (ALDOA) and LDHA [38–40]. The enzymes of this pathway
Glycolysis is a determined sequence of enzyme-catalyzed reactions also play important roles in the regulation of glycolysis. The regulation
that result in the conversion of glucose to lactate with the synthesis of of glycolysis by the external signaling pathways and glycolytic enzymes
ATP. ATP plays an essential role in providing energy for most organ- is summarized in Fig. 2.
isms, especially under anaerobic conditions. In nearly all organisms,
glycolysis occurs in the cytosol of cells and can be divided into two
steps. The first step is the Embden–Meyerhof–Parnas pathway, whereby 4. The roles of the IL-1 subfamily in regulating glycolysis
glucose is converted to pyruvate, and the last step is the production of
lactate (Fig. 2) [10]. The flux through the glycolytic pathway is regu- As described above, glycolysis is the most important cellular energy
lated directly or indirectly by external signaling pathways and the en- pathway, and its dysregulation is responsible for disease progression.
zymes of glycolysis. The Ras and PI3K signaling pathways are two The IL-1 subfamily has been implicated in the regulation of glycolysis
common pathways in the regulation of glycolysis. The former activates by influencing glucose uptake or glycolytic enzyme activation.
MAPK and PI3K to trigger the transcription factor c-Myc, which directly
induces the expression of numerous glycolytic genes such as glucose

20
Q. Tan et al. Cytokine and Growth Factor Reviews 44 (2018) 18–27

Table 2
The potential roles of IL-1 subfamily members in glycolysis in disease.
Cytokine Disease model Cell type/Tissue Mechanism Effect on Reference
glycolysis

IL-1α RA Human rheumatoid synovial cell Fru(2,6)P2 ↑ ↑ [8]


IL-1α RA Human rheumatoid synovial cell Fru(2,6)P2↑, Glucose utilization ↑ ↑ [42]
IL-1α Severe sepsis HDF Glucose uptake ↑ ↑ [12]
IL-1α Glomerular Injury Murine mesangial cell HK2 ↑ ↑ [61]
IL-1α Hyperlactatemia Rat skeletal muscle cells Lactate ↑ ↑ [72]
IL-1α Severe scalding Rat skeletal muscle cells PFK ↑ ↑ [66]
IL-1α Fasted catheterized rat model Rat skeletal muscle Glucose uptake ↑ ↑ [49]
IL-1α / HDF from FAP of colon and normal state Glucose uptake ↑; Fru(2,6)P2 ↑ ↑ [11]
IL-1α / HuGi and PSF Glucose uptake ↑ ↑ [43]
IL-1α / Rat adipose cells No effect on glucose uptake None [53]
IL-1α / Porcine Sertoli cells LDHA expression ↑ ↑ [69]
IL-1α / Rat hepatocytes Lactate ↑ ↑ [71]
IL-1β Type 2 diabetes mellitus Rat pancreatic islets Glucokinase mRNA content ↓ ↓ [7]
IL-1β Type 2 diabetes mellitus HIT-T 15β-cells Glucokinase activity ↓ ↓ [65]
IL-1β Type 2 diabetes mellitus 3T3-L1 adipocytes GLUT4 translocation ↓; ↓ [60]
Glucose uptake ↓
IL-1β Type 2 diabetes mellitus Mouse pancreatic islets cells Glucokinase activity ↓ ↓ [59]
IL-1β RA Human rheumatoid synovial cell Fru(2,6)P2 ↑ ↑ [8]
IL-1β RA;osteoarthritis Human synovial cells from patient with osteoarthritis and Glucose uptake ↑ ↑ [41]
rheumatoid arthritis
IL-1β Osteoarthritis Rat articular chondrocytes GLUT1 mRNA ↑ ↑ [55]
IL-1β Osteoarthritis Human chondrocytes Glucose uptake ↑ ↑ [45]
IL-1β Glomerular Injury Murine mesangial cell HK2 ↑ ↑ [61]
IL-1β Allergic asthma Human airway epithelial cells; mouse tracheal epithelial LDHA ↑ ↑ [70]
cells
IL-1β Peritoneum injury Rat peritoneal mesothelial cells GLUT1 mRNA ↑ ↑ [54]
IL-1β Tendon injuries inTPCs from injured Achilles tendons from mouse HK2 mRNA ↑ ↑ [62]
IL-1β OSCC CAFs from patients with OSCC HK2 ↑ ↑ [9]
IL-1β Glioblastoma Glioblastoma cell lines HK2 mRNA ↑ ↑ [63]
IL-1β Hypoglycemia Spleen, lung, heart Glucose uptake ↑ ↑ [52]
IL-1β Rat model with intravenous Lung, spleen, liver, skin Glucose uptake ↑ ↑ [50,51]
injection
IL-1β / Rat ovarian cells Glucose uptake ↑ ↑ [47]
IL-1β / HuGi and PSF Glucose uptake ↑ ↑ [43]
IL-1β / Rat Sertoli cells Glucose transport ↑; LDHA ↑ ↑ [57]
IL-1β / Mouse astrocytes Glucose uptake ↑ ↑ [44]
IL-1β / Immature rat ovarian cells GLUT3 translocation ↑ ↑ [56]
IL-1β / Human peritoneal mesothelial cells Glucose uptake ↑ ↑ [46]
IL-1β / L6-GLUT4myc cells GLUT4 translocation ↑; Glucose ↑ [58]
uptake ↑
IL-1Ra GVHD Rat Th17 cells HK2 and GLUT1 ↓ ↓ [73]
IL-1Ra OSCC CAFs HK2 ↓ ↓ [9]
IL-1Ra Glomerular injury Murine mesangial cells HK2 ↓ ↓ [61]
IL-33 NSCLC NSCLC cells isolated from surgical tissues GLUT1 ↑; Glucose uptake ↑ ↑ [48]

4.1. The role of the IL-1 subfamily in the regulation of glycolysis through model increased glucose uptake in the lung, spleen, liver and skin,
targeting glucose uptake whereas that of IL-1α enhanced glucose uptake in skeletal muscle,
diaphragm and heart [49–51,2]. Metzger et al performed an experiment
Increasing evidence suggests that IL-1 subfamily members mediate on mice bearing IL-1β-secreting tumors to investigate the chronic effect
glycolysis by targeting glucose uptake into cells, which occurs during of IL-1β on glucose uptake and discovered that IL-1β was associated
the early stage of glycolysis. Findings from in vitro studies that are with increased glucose uptake through the peripheral tissues, including
consistent with this view include the following: [1] Both IL-1α and IL- the spleen, lung and heart [52]. These studies suggest a positive role of
1β have been observed to stimulate glucose uptake in rheumatoid sy- the IL-1 subfamily in regulating glucose uptake.
novial cells [41,42,2]. Bird et al. reported that IL-1α and IL-1β sig- Several studies of the associations between the IL-1 subfamily and
nificantly induced glucose uptake in human gingival fibroblasts (HuGi) glucose uptake have been conducted in recent years. Further studies are
and porcine synoviocytes (PSF) by facilitating the maximal transport needed to address the following issues: [1] One study found that IL-1α
velocity of the glucose transporter (Vmax) [43,3]. Administration of IL- did not trigger a change in glucose uptake in rat adipose cells, sug-
1α was found to be positively correlated with the improvement of gesting that cell sensitivity to IL-1 subfamily members may vary among
glucose uptake in the HDF of patients with FAP of the colon [11,4]. IL- different cell types [53,2]. IL-1β has been found to have positive and
1β has been identified as a promoter of glucose uptake in different cell negative roles on glucose uptake, which seem to be associated with cell
types, including astrocytic brain cells [44], human chondrocytes [45], type. In addition, IL-1β was found to enhance glucose uptake in rat
human peritoneal mesothelial cells and rat ovarian cells [46,47,5]. IL- peritoneal mesothelial cells and rat articular chondrocytes by in-
33 enhances non-small cell lung cancer (NSCLC) cell outgrowth and creasing GLUT1 mRNA [54,55]. In immature rat ovarian cells, in-
metastasis in vitro through upregulating membrane GLUT1 on NSCLC creased glucose uptake was induced by IL-1β via GLUT3 production
cells via the IL-33/ST2 pathway, leading to enhanced glycolysis [48]. [56]. Riera et al confirmed that IL-1β activates the PI3K/PKB pathway
Furthermore, animal studies have confirmed the actions of the IL-1 to upregulate glucose transport in Sertoli cells [57]. In a line of skeletal
subfamily in targeting glucose uptake. The main findings include the muscle cells (L6-GLUT4myc cells), IL-1β facilitated glucose uptake by
following [1]: The intracerebroventricular injection of IL-1β in a rat increasing GLUT4 translocation via the activation of AMPK [58].

21
Q. Tan et al. Cytokine and Growth Factor Reviews 44 (2018) 18–27

Fig. 2. The Ras pathway and the PI3K-AKT-mTOR pathway in the regulation of glycolysis.

Nevertheless, IL-1β acts as a strong inhibitor of glucose uptake in some HK2 promoter [63]. Recently, increased IL-1β derived from oral squa-
tissues, such as liver and pancreatic islets, via the downregulation of mous cell carcinoma (OSCC) in the tumor microenvironment (TME) and
GLUT2 expression [52,59]. Interestingly, a report revealed that IL-1β a correlation between IL-1β secretion and the glycolysis of cancer-as-
inhibited insulin-induced glucose uptake via decreasing GLUT4 trans- sociated fibroblasts (CAFs) in vitro was described by Wu et al [9]. These
location in 3T3-L1 adipocytes, which conflicts with the findings in results suggest that IL-1β secreted by OSCC cells increases HK2 ex-
skeletal muscle cells (described above) [60]. These data suggest that IL- pression in CAFs. As a result, the lactate produced by CAFs serves as
1β may have different roles in glucose uptake depending on cell type. 'fuel' for the proliferation of cancer cells. However, IL-1β appears to be a
Further studies are needed to identify the mechanisms underlying IL- functional mediator between CAFs and cancer cells. Interestingly,
1β's effects on glucose uptake. macrophages are the primary manufacturers of IL-1β, and one hallmark
of cancer is aerobic glycolysis (the Warburg effect) [64]. Thus, it re-
4.2. The role of the IL-1 subfamily in the regulation of glycolysis via mains to be determined whether tumor-associated macrophages (TAM),
glycolytic enzyme activation which are considered to be central players in the TME, can enhance the
glycolysis of cancer cells through the production of IL-1β or IL-1β by
IL-1 subfamily cytokines have been found to influence the activities cancer cells, which would increase the activity and positive role of
of glycolytic enzymes, including HK, Glucokinase, PFK and LDHA, and TAM. This area is a major topic of interest to our research group. The
thereby lead to glycolysis changes. regulatory associations between TAM and cancer cells require further
study.

4.2.1. The IL-1 subfamily and HK activity


HK catalyzes the conversion of glucose to G-6-P when glucose enters 4.2.2. The role of the IL-1 subfamily in glucokinase activity
the cell. In vitro studies have been conducted to verify the role of IL-1α Glucokinase is another enzyme that catalyzes the conversion of
or IL-1β in HK activity. Taneja et al. reported that IL-1α and IL-1β are glucose to G-6-P; however, it specific to hepatocytes and β cells. Studies
two common and important activators of HK activity and that both of pancreatic islets and HIT-T15 β-cells have shown that IL-1β is a
cytokines increase HK activity through the IL-1 receptor-, Ras- and strong inhibitor of glucokinase expression. The inhibition of glucoki-
classic MAPK pathways [61]. In vitro experiments with injured tendon- nase reduces glycolysis, leading to decreased insulin release, which is
derived progenitor cells from mouse Achilles tendons revealed a cor- considered a pathogenic factor of type 2 diabetes mellitus [7,59,65]. To
relation between IL-1β overexpression and the expression of HK2 [62]. investigate the role of IL-1β in regulating glucokinase activity, in-
In another study, IL-1β enhanced HK2 expression and appeared to act vestigators measured islet glucokinase mRNA through polymerase
as a transcription factor that induces chromatin reorganization at the chain reaction (PCR) and the concentration of nitric oxide (NO), which

22
Q. Tan et al. Cytokine and Growth Factor Reviews 44 (2018) 18–27

is a positive regulator in the dysfunction of pancreatic β-cells. They IL-1Ra significantly suppressed the IL-1β-induced effects on the ex-
found that IL-1β provoked a delayed increase in the production of NO pression of glycolysis-related genes in Th17 cells, slowing the progress
but reduced the islet content of glucokinase mRNA. This effects were of the disease [73]. Furthermore, IL-1Ra was found to decrease the
attenuated due to the activity of N-monomethyl-L-arginine (NMA), an development of CAFs, the central players in TME (described above), in
inhibitor of NO synthase, suggesting that IL-1β induces a decrease in samples of patients with OSCC.
the synthesis of islet glucokinase protein by an NO-dependent me- However, IL-1β plays a key role in TME and induces the develop-
chanism [7]. The different roles of IL-1β in the conversion of glucose to ment of CAFs. This latter effect was neutralized by IL-1Ra in a previous
G-6-P depend on the cell type involved. study, suggesting that IL-1α and IL-1β have key functions in the pro-
gression of disease and that IL-1α, IL-1β and IL-1Ra may also play roles
4.2.3. The IL-1 subfamily affects PFK activity in the targeted agent [9].
In catalyzing the rate-limiting step of the conversion of fructose-6-
phosphate (F6P) to fructose-1,6-bisphosphate (F1,6P2), PFK functions 5. The dysregulation of glycolysis caused by the IL-1 subfamily in
as a strong regulator of flux through glycolysis. An experimental mouse disease
model of severe scalding revealed that IL-1α can enhance PFK activity
in skeletal muscles and lower pyruvate dehydrogenase (PDH) activity, As described in the previous section, IL-1 subfamily-induced effects
which is associated with glucose oxidation [66]. In addition, PFK can be on glycolysis have been linked to the dysregulation of glycolysis, which
stimulated by Fru(2,6)P2. Accordingly, an increase in Fru(2,6)P2 has contributes to the development and progression of several glycolysis-
been proposed to be part of the mechanism by which IL-1α induces PFK related diseases. Evidence that the dysregulation of glycolysis was in-
in rheumatoid synovial cells [8,42,67]. Taylor used HDF from a normal duced by the IL-1 subfamily in disease is described below.
colon and FAP of the colon and confirmed that Fru(2,6)P2 was actively
regulated by IL-1α(11). These findings suggest that the role of IL-1α in 5.1. Type 2 diabetes
the regulation of glycolysis involves both direct and indirect targeting
of PFK. Type 2 diabetes is characterized by diminished insulin secretion and
insulin resistance, which result from decreased glycolysis in islet β-cells
4.2.4. The IL-1 subfamily plays an active role in LDHA expression and insulin-sensitive tissues such as adipose tissue. IL-1β has been im-
Lactate is the end product of glycolysis, and its accumulation is plicated in the occurrence and development of type 2 diabetes in as-
among the most sensitive parameters for detecting improvements in sociation with the downregulation of glucokinase expression in β-cells
glycolysis. LDHA is a rate-limiting enzyme in glycolysis and has es- and decreased glucose uptake in adipocytes, as described above. These
sential roles in the conversion of pyruvate to lactate [68]. Correlations observations suggest the potential of the inhibition of IL-1β activity as a
between IL-1 subfamily members and LDHA have also been demon- new targeted therapeutic strategy in type 2 diabetes patients
strated in vitro studies. Researchers have revealed that IL-1α stimulates [7,59,60,65]. In a randomized, placebo-controlled, double-blind Phase
lactate production in porcine Sertoli cells by upregulating LDHA ex- I/II clinical trial, patients with type 2 diabetes received a novel vaccine
pression [69]. In vitro and animal studies using an allergic asthma against IL-1β and showed improved glycemia as a result. Furthermore,
model to investigate the expression of LDHA induced by IL-1α or IL-1β the vaccine was suggested to be safe for use in clinical practice [74].
revealed active roles of these cytokines in the regulation of LDHA in
human airway epithelial cells and mouse tracheal epithelial cells [70]. 5.2. Rheumatoid arthritis
Riera et al. investigated the mechanism of IL-1β-triggered glycolysis
increase by utilizing Sertoli cells and found that IL-1β enhanced LDHA Increased glycolysis is associated with increased accumulation of
mRNA levels via the ERK1/2 pathway [57]. lactate in joints. This accumulation inhibits the synthesis of matrix
Collectively, the above-described studies reveal that IL-1α, IL-1β macromolecules, such as proteoglycans, in synovial cells, leading to
and IL-33 influence glycolysis via different mechanisms. Additional joint degeneration; this process has been described as a mechanism of
studies have found correlations between IL-1 subfamily members and RA. Several lines of evidence described above suggest that both IL-1α
glycolysis for which the underlying mechanisms remain unknown: [1] and IL-1β play active roles in glycolysis and its inhibition in synovial
Vaartjes et al. reported that IL-1α increased lactate accumulation in rat cells. IL-1 blockers such as anakinra, which is a recombinant human IL-
hepatocytes in vitro and depressed glycogen synthesis [71,2]. Admin- 1 receptor antagonist (IL-1Ra, described below) and has been FDA-
istration of IL-1β induced hyperlactatemia in a rat model and enhanced approved for RA, offer potential strategies for treatment [8,41,42,67].
lactate content in rat skeletal muscle while decreasing the activity of Another study involving synovium and fibroblast-like synoviocytes
PDH [72]. These studies suggest that cytokines in the IL-1 subfamily from patients with RA demonstrated the efficacy of 2-deoxy-D-glucose
have important roles in glycolysis not only in targeting glucose uptake (2-DG), a glycolysis inhibitor, in the treatment of RA [75]. Collectively,
or glycolytic enzyme activation but also in influencing alternative these findings suggest that the increased glycolysis in synovial cells
pathways of glucose metabolism. This possibility requires further in- caused by IL-1α and IL-1β plays a pivotal role in the pathogenesis of
vestigation. The mechanisms by which these cytokines influence gly- RA.
colysis are varied and seem to be dependent on cell type. Regardless,
the abovementioned studies support to the view that IL-1 subfamily 5.3. Cancer
cytokines and the process of glycolysis are correlated.
Cancer cells use glycolysis as the main pathway for obtaining en-
4.2.5. The potential role of IL-1Ra as antagonist in glycolysis regulation ergy. Even under a sufficient O2 supply, the tumor cells seem to prefer
As one of the main members of the IL-1 subfamily and the natural to rely on glycolysis for the production of ATP as fuel. Therefore, sig-
antagonist of both IL-1α and IL-1β, IL-1Ra dampens IL-1α- or IL-1β- nificant metabolic characteristics of cancer tissue have been found to be
enhanced glycolysis via competing with these cytokines for receptor associated with high glycolysis rates [15]. Recent research has revealed
binding sites. Consistent with this observation, a study demonstrated that lactate, a product of the glycolysis pathway, is the most important
that IL-1Ra could prevent IL-1α or IL-1β stimulation of HK activity energy resource for cancer cells. These findings suggest that high
[61]. In a study of Graf-versus-host disease (GVHD), Th17 cells were aerobic glycolysis is a feature of cancer cells and can be used as a target
found to be the central players in GVHD, and IL-1β was found to be in cancer therapy [76]. IL-1β and IL-33 were described as common
involved in the development of Th17 cells through inducing the ex- promoters in the regulation of glycolysis in studies of OSCC and NSCLC
pression of glycolysis-related genes, such as HK2 and GLUT1. However, [9,48]. These observations support the concept that an IL-1 subfamily

23
Q. Tan et al. Cytokine and Growth Factor Reviews 44 (2018) 18–27

blockade as a potential therapeutic target may be beneficial for cancer in Atopic Dermatitis) and represent the first proof-of-concept data re-
patients. Regarding NSCLC, a current phase III trial is seeking patients garding anti-IL-33 activity. Research to confirm and expand its clinical
to evaluate the safety and efficacy of Canakinumab (a neutralizing applications is in progress. For example, a phase II clinical trial is un-
monoclonal anti-IL-1β antibody; described below) (ClinicalTrials.gov derway to assess the safety and efficacy of ANB020 in adult patients
Identifier: NCT03447769). with severe eosinophilic asthma (ClinicalTrials.gov Identifier:
NCT03469934).
5.4. Other glycolysis-related diseases The findings described above showed that these targeted agents
have been widely acknowledged and have applications in clinical
Correlations of the IL-1 subfamily with glycolysis dysfunction have practice. The targeted agents are summarized in Table 1.
been reported for some glycolysis-related diseases, including osteoar-
thritis and asthma, in a limited number of studies, However, more at- 7. Conclusions and future directions
tention should be paid to the possibility that the IL-1 subfamily has
important roles in the pathogenesis of theses disease for the following In summary, the cytokines of the IL-1 subfamily act as promoters in
reasons: [1] Lactate inhibits the synthesis of matrix macromolecules in regulating glycolysis. However, some reports have revealed negative
chondrocytes and is believed to be a mechanism of osteoarthritis roles of these cytokines in glycolysis in disease. The specific roles of
[41,2]. Increases in lactate in human nasal epithelial cells from asth- these cytokines in regulating glycolysis might depend on cell type and
matics relative to those from healthy subjects have been observed, the type of cytokine.
suggesting the importance of dysregulated glycolysis in allergies [41]. The correlations between the IL-1 subfamily and the dysfunction of
Studies (described above) have demonstrated that IL-1β, as a positive glycolysis in disease states should not be neglected. Our review pro-
regulator, is important in augmenting glycolysis in human chon- vides the first summary of the potential roles of IL-1 subfamily members
drocytes and human nasal epithelial cells. Currently, a phase I/II trial in in glycolysis in disease. Previous studies have focused mainly the re-
patients with asthma is underway to determine the effect of anakinra on lationships between inflammatory tissue injury induced by these cyto-
asthma (ClinicalTrials.gov Identifier: NCT03513458). kines and the pathogenesis of disease. However, IL-1 subfamily-induced
changes in glycolysis in disease states are gradually being identified and
6. Therapies available to inhibit the actions of the IL-1 subfamily becoming recognized. Given the findings summarized in the present
review, it is essential that the following aspects be addressed in future
As described above, studies have identified correlations between the studies: [1] It is clear that inflammatory factors, such as the cytokines of
IL-1 subfamily and the dysregulation of glycolysis, which seem to play IL-1 subfamily, can influence glycolysis. Positron emission tomography
an important role in the pathogenesis of disease. The use of IL-Ra has (PET) detects the high aerobic glycolysis of malignant tumors [81],
been shown to partially correct glycolysis dysfunction due to its an- with standardized uptake (SUVmax) measured by PET serving as an
tagonistic effects. On the other hand, IL-1 subfamily blockers may re- index of metabolism and reflecting the rate of glycolysis in tissues.
present promising adjuvant therapies. Here, we summarize current Therefore, whether SUVmax is positively correlated with IL-1α, IL-1β
targeted agents that antagonize the actions of these cytokines (see and IL-33 in peripheral blood warrants investigation [2]. Patients suf-
Fig. 2). Currently, therapeutic strategies include the following: [1] fering from glycolysis-related diseases such as type 2 diabetes or RA
anakinra, the recombinant form of hIL-1Ra [2]; soluble decoy receptors, have been shown to benefit from anti-IL-1 agents [77]. Since aerobic
including rilonacept; and [3] neutralizing monoclonal anti-IL-1β anti- glycolysis is the main pathway by which cancer cells obtain energy,
bodies, such as canakinumab. Anakinra, rilonacept and canakinumab whether IL-1 subfamily blockers can improve prognosis in cancer pa-
have been widely studied and FDA-approved for inflammatory diseases tients, e.g., by prolonging survival time and relieving cancer-associated
such as gout and type 2 diabetes. Their effects on inflammatory diseases symptoms such as anorexia, fatigue and pain, should be investigated.
and the associated mechanisms are reviewed in previous work [77]. In The blocking of the IL-1 subfamily has been performed in clinical trials
addition, a monoclonal antibody directed against IL-1α (MABp1) and of cancer treatment and has been reported effective for patients with
the monoclonal anti-IL-33R antibody (CNTO7160) as well as a neu- advanced colorectal cancer [80]. However, few studies have evaluated
tralizing anti-IL-33 (ANB020) are being investigated in clinical trials. whether IL-1 subfamily blockers improve prognosis in lung-cancer pa-
MABp1, a neutralizing monoclonal anti-IL-1α antibody, appears to tients, which is a future research direction of our group. Furthermore,
be effective in inhibiting the progression of type 2 diabetes mellitus and inflammatory factors, especially the cytokines of the IL-1 subfamily, in
has been found to be effective in the treatment of moderate to severe TME warrant investigation and will be a focus of our future studies.
hidradenitis suppurativa [78,79]. Furthermore, in a phase III trial of
patients with metastatic colorectal cancer, MABp1 treatment led to Conflict of interests
improvements in the prognosis of patients ineligible for standard che-
motherapy, including the relief of cancer-associated symptoms [80]. The authors declare that they have no competing interests.
MABp1 has been shown to have a satisfactory safety profile. These
observations indicate that MABp1 may be another promising agent for Acknowledgements
targeting IL-1α; further studies are needed to confirm this possibility
and develop clinical applications. Currently, the efficacy of MABp1 in This work was greatly supported by the National Natural Science
treating patients with atopic dermatitis is being tested in a phase II Foundation of China (Nos. 81572942 and No. 81770096), and the
study (ClinicalTrials.gov Identifier: NCT03496974). Hubei province natural science foundation of China (No. 2017CFB260).
CNTO-7160, a monoclonal antibody to IL-33R, inhibits IL-33 by
preventing it from binding to its receptor (ST2) and was designed to References
treat patients with severe asthma. Presently, this antibody is in a phase I
clinical trial (ClinicalTrials.gov Identifier: NCT02345928). No data [1] V. Menkin, Chemical basis of fever, Science 100 (2598) (1944) 337–338.
have yet been reported. [2] C.J. March, B. Mosley, A. Larsen, D.P. Cerretti, G. Braedt, V. Price, et al., Cloning,
sequence and expression of two distinct human interleukin-1 complementary DNAs,
ANB020 is another potent IL-33-targeted agent that directly neu- Nature. 315 (6021) (1985) 641.
tralizes IL-33. It has been licensed and has passed a phase II clinical trial [3] C.A. Dinarello, Biology of interleukin 1, FAESB J. Off. Publ. Feder. Am. Soc. Exp.
for the treatment of moderate-to-severe atopic dermatitis. Data are Biol. 2 (2) (1988) 108–115.
[4] J. Sims, D. Smith, H. Blumberg, M. Nold, A. Mantovani, A. Rubartelli, et al., IL-1
available from AnaptysBio reports (AnaptysBio Reports Positive Family Nomenclature, Queen’s University of Belfast, 1987.
Topline Proof-of-Concept Data from Phase 2a Clinical Trial of ANB020

24
Q. Tan et al. Cytokine and Growth Factor Reviews 44 (2018) 18–27

[5] P. Italiani, M.L. Manca, F. Angelotti, D. Melillo, F. Pratesi, I. Puxeddu, et al., IL-1 metabolic switches hexokinase 2 and pyruvate dehydrogenase kinase 1, Mol. Cell.
family cytokines and soluble receptors in systemic lupus erythematosus, Arthritis Biol. 27 (21) (2007) 7381–7393.
Res. Ther. 20 (1) (2018) 27. [37] A. Le, C.R. Cooper, A.M. Gouw, R. Dinavahi, A. Maitra, L.M. Deck, et al., Inhibition
[6] J.E. Sims, S.K. Dower, cDNA expression cloning of the IL-1 receptor, a member of of lactate dehydrogenase A induces oxidative stress and inhibits tumor progression,
the immunoglobulin superfamily, Science 241 (4865) (1988) 585. Proc. Natl. Acad. Sci. U. S. A. 107 (5) (2010) 2037–2042.
[7] Z. Ma, M. Landt, A. Bohrer, S. Ramanadham, D.M. Kipnis, J. Turk, Interleukin-1 [38] N. Robichaud, N. Sonenberg, D. Ruggero, R.J. Schneider, Translational control in
reduces the glycolytic utilization of glucose by pancreatic islets and reduces glu- cancer, Cold Spring Harb. Perspect. Biol. (2018) a032896.
cokinase mRNA content and protein synthesis by a nitric oxide-dependent me- [39] R. Bartrons, J. Caro, Hypoxia, glucose metabolism and the Warburg’s effect, J.
chanism, J. Biol. Chem. 272 (28) (1997) 17827. Bioenerg. Biomembr. 39 (3) (2007) 223–229.
[8] D.J. Taylor, R.J. Whitehead, J.M. Evanson, D. Westmacott, M. Feldmann, [40] J.J. Lum, T. Bui, M. Gruber, J.D. Gordan, R.J. Deberardinis, K.L. Covello, et al., The
H. Bertfield, et al., Effect of recombinant cytokines on glycolysis and fructose 2,6- transcription factor HIF-1α plays a critical role in the growth factor-dependent
bisphosphate in rheumatoid synovial cells in vitro, Biochem. J. 250 (1) (1988) regulation of both aerobic and anaerobic glycolysis, Genes Dev. 21 (9) (2007)
111–115. 1037–1049.
[9] J. Wu, Y. Hong, T. Wu, J. Wang, X. Chen, Z. Wang, et al., Stromal-epithelial lactate [41] A. Hernvann, B. Bourely, V.L. Maire, C. Aussel, C.-J. Menkes, O.G. Ekindjian, Action
shuttle induced by tumor-derived interleukin-1β promotes cell proliferation in oral of anti-inflammatory drugs on interleukin-1β-mediated glucose uptake by syno-
squamous cell carcinoma, Int. J. Mol. Med. 41 (2) (2018) 687–696. viocytes, Eur. J. Pharmacol. 314 (1–2) (1996) 193–196.
[10] J. Peretó, Embden-Meyerhof-Parnas Pathway, Springer Berlin Heidelberg, 2011 [42] D.J. Taylor, M. Feldmann, J.M. Evanson, D.E. Woolley, Comparative and combined
485- p. effects of transforming growth factors alpha and beta, interleukin-1 and interferon-
[11] D.J. Taylor, Interleukin-1 stimulation of fibroblast glycolysis is accompanied by gamma on rheumatoid synovial cell proliferation, glycolysis and prostaglandin E
reduced glucose oxidation in the tricarboxylic acid cycle, Biochem. Soc. Trans. 18 production, Rheumatol. Int. 9 (2) (1989) 65–70.
(5) (1990) 982. [43] T.A. Bird, A. Davies, S.A. Baldwin, J. Saklatvala, Interleukin 1 stimulates hexose
[12] D.J. Taylor, E.B. Faragher, J.M. Evanson, Inflammatory cytokines stimulate glucose transport in fibroblasts by increasing the expression of glucose transporters, J. Biol.
uptake and glycolysis but reduce glucose oxidation in human dermal fibroblasts in Chem. 265 (23) (1990) 13578.
vitro, Circ. Shock 37 (2) (1992) 105. [44] E. Shmueli, O. Ben-Menchem-Zidon, S.B. Park, R. Yirmiya, 92. The role of astrocytic
[13] N. Kresge, R.D. Simoni, R.L. Hill, JBC centennial 1905-2005 100 years of bio- glucose uptake in the effect of the inflammatory cytokine interleukin-1-beta on
chemistry and molecular biology (Reprinted), J. Biol. Chem. 285 (36) (2010). hippocampal-dependent memory, Brain Behav. Immun. 25 (Suppl2) (2011) S205-S.
[14] A.H. Romano, T. Conway, Evolution of carbohydrate metabolic pathways, Res. [45] G.M. Lee, M.E. Tioran, M. Jansen, R.D. Graff, S.S. Kelley, P. Lin, Development of
Microbiol. 147 (6-7) (1996) 448–455. selective tolerance to interleukin-1β by human chondrocytes in vitro *, J. Cell.
[15] O. Warburg, The metabolism of carcinoma cells, J. Cancer Res. 9 (1925) 148–163. Physiol. 192 (1) (2010) 113.
[16] P.E. Auron, A.C. Webb, L.J. Rosenwasser, S.F. Mucci, A. Rich, S.M. Wolff, et al., [46] M. Kruse, A. Mahiout, V. Kliem, P. Kurz, K.M. Koch, R. Brunkhorst, Interleukin-1
Nucleotide sequence of human monocyte interleukin 1 precursor cDNA, Proc. Natl. beta stimulates glucose uptake of human peritoneal mesothelial cells in vitro, Perit.
Acad. Sci. U. S. A. 81 (24) (1984) 7907–7911. Dial. Int. 16 (Suppl 1) (1996) S58.
[17] C.A. Dinarello, Biologic basis for interleukin-1 in disease, Blood. 87 (6) (1996) [47] I. Ben-Shlomo, S. Kol, L.M. Roeder, C.E. Resnick, A. Hurwitz, D.W. Payne, et al.,
2095–2147. Interleukin (IL)-1beta increases glucose uptake and induces glycolysis in aerobically
[18] P. Rider, Y. Carmi, O. Guttman, A. Braiman, I. Cohen, E. Voronov, et al., IL-1α and cultured rat ovarian cells: evidence that IL-1beta may mediate the gonadotropin-
IL-1β recruit different myeloid cells and promote different stages of sterile in- induced midcycle metabolic shift, Endocrinology 138 (7) (1997) 2680.
flammation, J. Immunol. 187 (9) (2011) 4835–4843. [48] C. Wang, Z. Chen, X. Bu, H. Yang, S. Shan, R. Tao, et al., IL-33 signaling fuels
[19] I. Cohen, P. Rider, Y. Carmi, A. Braiman, S. Dotan, M.R. White, et al., Differential outgrowth and metastasis of human lung cancer, Biochem. Biophys. Res. Commun.
release of chromatin-bound IL-1alpha discriminates between necrotic and apoptotic 479 (3) (2016) 461–468.
cell death by the ability to induce sterile inflammation, Proc. Natl. Acad. Sci. U. S. [49] A. Jarrous, C.H. Lang, Central administration of IL-1 alpha increases glucose uptake
A. 107 (6) (2010) 2574–2579. by muscle, Cytokine 7 (1) (1995) 57–63.
[20] C.A. Dinarello, The many worlds of reducing interleukin-1, Arthritis Rheum. 52 (7) [50] C.H. Lang, C. Dobrescu, Interleukin-1 induced increases in glucose utilization are
(2014) 1960–1967. insulin mediated, Life Sci. 45 (22) (1989) 2127–2134.
[21] M. Muzio, J. Ni, P. Feng, V.M. Dixit, IRAK (Pelle) family member IRAK-2 and [51] A.D. Rey, E. Roggero, A. Randolf, C. Mahuad, S. Mccann, V. Rettori, et al., IL-1
MyD88 as proximal mediators of IL-1 signaling, Science 278 (5343) (1997) resets glucose homeostasis at central levels, Proc. Natl. Acad. Sci. U. S. A. 103 (43)
1612–1615. (2006) 16039–16044.
[22] L. Sanz, M.T. Diaz-Meco, H. Nakano, J. Moscat, The atypical PKC‐interacting pro- [52] S. Metzger, S. Nusair, D. Planer, V. Barash, O. Pappo, J. Shilyansky, et al., Inhibition
tein p62 channels NF‐κB activation by the IL‐1–TRAF6 pathway, EMBO J. 19 (7) of hepatic gluconeogenesis and enhanced glucose uptake contribute to the devel-
(2014) 1576–1586. opment of hypoglycemia in mice bearing interleukin-1beta- secreting tumor,
[23] E. Hoffmann, A. Thiefes, D. Buhrow, O. Dittrich-Breiholz, H. Schneider, K. Resch, Endocrinology 145 (11) (2004) 5150.
et al., MEK1-dependent delayed expression of Fos-related antigen-1 counteracts c- [53] A. Garcia-Welsh, J.S. Schneiderman, D.L. Baly, Interleukin-1 stimulates glucose
Fos and p65 NF-kappaB-mediated interleukin-8 transcription in response to cyto- transport in rat adipose cells. Evidence for receptor discrimination between IL-1
kines or growth factors, J. Biol. Chem. 280 (10) (2005) 9706–9718. beta and IL-1 alpha, FEBS Lett. 269 (2) (1990) 421–424.
[24] C. Wang, L. Zeng, T. Zhang, J. Liu, W. Wang, Tenuigenin prevents IL-1β-induced [54] H. Ding, R. Min, Effects of IL-1β on glucose transporter 1 expression in cultured rat
inflammation in human osteoarthritis chondrocytes by suppressing PI3K/AKT/NF- peritoneal mesothelial cells, Chin. J. Immunol. (2011).
κB signaling pathway, Inflammation 39 (2) (2016) 1–6. [55] H.S. Lee, Y.P. Chung, S.L. Su, G.S. Huang, Regulation of glucose transporter 1 by IL-
[25] A.J. Puren, G. Fantuzzi, Y. Gu, M.S. Su, C.A. Dinarello, Interleukin-18 (IFNgamma- 1β stimulation in rat articular chondrocytes, J. Med. Sci. 29 (3) (2009) 125–130.
inducing factor) induces IL-8 and IL-1beta via TNFalpha production from non- [56] S. Kol, I. Ben-Shlomo, K. Ruutiainen, M. Ando, T.M. Davies-Hill, R.M. Rohan, et al.,
CD14+ human blood mononuclear cells, J. Clin. Invest. 101 (3) (1998) 711–721. The midcycle increase in ovarian glucose uptake is associated with enhanced ex-
[26] M. Sahoo, I. Ceballosolvera, L.D. Barrio, F. Re, Role of the inflammasome, IL-1β, pression of glucose transporter 3. Possible role for interleukin-1, a putative inter-
and IL-18 in bacterial infections, Sci. World J. 11 (2011) 2037–2050. mediary in the ovulatory process, J. Clin. Invest. 99 (9) (1997) 2274.
[27] D. Boraschi, P. Italiani, S. Weil, M.U. Martin, The family of the interleukin-1 re- [57] M.F. Riera, M.N. Galardo, E.H. Pellizzari, S.B. Meroni, S.B. Cigorraga, Participation
ceptors, Immunol. Rev. 281 (1) (2018) 197–232. of phosphatidyl inositol 3-kinase/protein kinase B and ERK1/2 pathways in inter-
[28] U. Fiocco, M. Vezzù, L. Cozzi, S. Todesco, IL-1Ra (recombinant human IL-1 receptor leukin-1beta stimulation of lactate production in Sertoli cells, Reproduction 133 (4)
antagonist) in the treatment of rheumatoid arthritis: the efficacy, Reumatismo 56 (2007) 763–773.
(1s) (2011) 62. [58] A. Takaguri, S. Inoue, T. Kubo, K. Satoh, AMPK activation by prolonged stimulation
[29] M.S. Akash, Q. Shen, K. Rehman, S. Chen, Interleukin-1 receptor antagonist: a new with interleukin‐1β contributes to the promotion of GLUT4 translocation in skeletal
therapy for type 2 diabetes mellitus, J. Pharm. Sci. 101 (5) (2012) 1647–1658. muscle cells, Cell Biol. Int. 40 (11) (2016) 1204–1211.
[30] S. Lu, S. Zheng, W. Wang, Inhibitor of pancreatic cancer by RHIL1RA-letter, Clin. [59] C. Park, J.R. Kim, J.K. Shim, B.S. Kang, Y.G. Park, K.S. Nam, et al., Inhibitory effects
Cancer Res. 23 (12) (2017) 3223. of Streptozotocin, tumor necrosis Factor-α, and Interleukin-1β on glucokinase ac-
[31] E. Lefrançais, S. Roga, V. Gautier, A. Gonzalezdeperedo, B. Monsarrat, J.P. Girard, tivity in pancreatic islets and gene expression of GLUT2 and glucokinase, Arch.
et al., IL-33 is processed into mature bioactive forms by neutrophil elastase and Biochem. Biophys. 362 (2) (1999) 217–224.
cathepsin G, Proc. Natl. Acad. Sci. U. S. A. 109 (5) (2012) 1673–1678. [60] T.G. Jennifer Jager, Mireille Cormont, Yannick Le Marchand-Brustel, Jean-
[32] A.G. Besnard, D. Togbe, N. Guillou, F. Erard, V. Quesniaux, B. Ryffel, IL-33-acti- François Tanti, Interleukin-1beta-induced insulin resistance in adipocytes through
vated dendritic cells are critical for allergic airway inflammation, Eur. J. Immunol. down-regulation of insulin receptor substrate-1 expression, Endocrinology 148 (1)
41 (6) (2011) 1675–1686. (2007) 241–251.
[33] W.P. Arend, G. Palmer, C. Gabay, IL-1, IL-18, and IL-33 families of cytokines, [61] N. Taneja, P.E. Coy, I. Lee, J.M. Bryson, R.B. Robey, Proinflammatory interleukin-1
Immunol. Rev. 223 (1) (2010) 20–38. cytokines increase mesangial cell hexokinase activity and hexokinase II isoform
[34] R.C. Osthus, H. Shim, S. Kim, Q. Li, R. Reddy, M. Mukherjee, et al., Deregulation of abundance, Am. J. Physiol. Cell Physiol. 287 (2) (2004) C548.
glucose transporter 1 and glycolytic gene expression by c-Myc, J. Biol. Chem. 275 [62] K. Zhang, S. Asai, B. Yu, Enomotoiwamoto M. IL-1β irreversibly inhibits tenogenic
(29) (2000) 21797–21800. differentiation and alters metabolism in injured tendon-derived progenitor cells in
[35] H. Shim, C. Dolde, B.C. Lewis, C.S. Wu, G. Dang, R.A. Jungmann, et al., c-Myc vitro, Biochem. Biophys. Res. Commun. 463 (4) (2015) 667–672.
transactivation of LDH-A: implications for tumor metabolism and growth, Proc. [63] P. Gupta, T. Sheikh, E. Sen, SIRT6 regulated nucleosomal occupancy affects
Natl. Acad. Sci. U. S. A. 94 (13) (1997) 6658–6663. Hexokinase 2 expression, Exp. Cell Res. 357 (1) (2017) 98.
[36] J.W. Kim, P. Gao, Y.C. Liu, G.L. Semenza, C.V. Dang, Hypoxia-inducible factor 1 and [64] B. Pazár, H.K. Ea, S. Narayan, L. Kolly, N. Bagnoud, V. Chobaz, et al., Basic calcium
dysregulated c-Myc cooperatively induce vascular endothelial growth factor and phosphate crystals induce monocyte/macrophage IL-1β secretion through the

25
Q. Tan et al. Cytokine and Growth Factor Reviews 44 (2018) 18–27

NLRP3 inflammasome in vitro, J. Immunol. 186 (4) (2011) 2495–2502. Qi Huang obtained her PhD in Union Hospital, Tongji
[65] M. Beggs, G. Beresford, J. Clarke, R. Mertz, J. Espinal, P. Hammonds, Interleukin‐1β Medical College, Huazhong University of Science and
inhibits glucokinase activity in clonal HIT‐T15 β‐cells, FEBS Lett. 267 (2) (1990) Technology in 2016. She has been devoted to research the
217–220. glucose metabolism in lung cancer, and the mechanism of
[66] X. Liao, Y. Dong, Regulative effects of IL-1 on key enzyme for glucose catabolism in action of IL-17 in cancer metabolism.
skeletal muscle after severe scalding in rats, Acta Acad. Med. Militaris Tertiae 21
(1999).
[67] D.J. Taylor, J.M. Evanson, D.E. Woolley, Contrasting effects of the protein kinase C
inhibitor, staurosporine, on cytokine and phorbol ester stimulation of fructose 2,6-
bisphosphate and prostaglandin E production by fibroblasts in vitro. Comparative
studies using interleukin-1 alpha, tumour necro, Biochem. J. 269 (3) (1990) 573.
[68] A. Schurr, R.S. Payne, Lactate, not pyruvate, is neuronal aerobic glycolysis end
product: an in vitro electrophysiological study, Neuroscience 147 (3) (2007)
613–619.
[69] D. Nehar, C. Mauduit, F. Boussouar, M. Benahmed, Interleukin 1alpha stimulates
Yan ling Ma received a master's degree from the Union
lactate dehydrogenase A expression and lactate production in cultured porcine
Hospital of Tongji Medical College of Huazhong University
sertoli cells, Biol. Reprod. 59 (6) (1998) 1425–1432.
of Science and Technology in 2015. Her work focuses on
[70] X. Qian, R. Aboushousha, Chia S.B. Van dWC, E. Amiel, R.W. Schneider, et al.,
the role of mechanism of action of inflammatory cytokines
Interleukin-1/inhibitory kappa B kinase epsilon-induced glycolysis augment epi-
in the metabolism of EGFR mutation-positive non-small cell
thelial effector function and promote allergic airways disease, J. Allergy Clin.
lung cancer cells.
Immunol. 142 (2) (2017).
[71] W.J. Vaartjes, C.G. de Haas, M. Houweling, Acute effects of interleukin 1 alpha and
6 on intermediary metabolism in freshly isolated rat hepatocytes, Biochem.
Biophys. Res. Commun. 169 (2) (1990) 623–628.
[72] T.C. Vary, P. O’Neill, R.N. Cooney, M. Shumate, Chronic infusion of interleukin 1
induces hyperlactatemia and altered regulation of lactate metabolism in skeletal
muscle, JPEN J. Parenter. Enteral Nutr. 23 (4) (1999) 213.
[73] P. Min-Jung, L.S. Hoon, L. Sung-Hee, L. Eun-Jung, K. Eun-Kyung, C.J. Young, et al.,
IL-1 receptor blockade alleviates graft-versus-host disease through downregulation
of an Interleukin-1β-dependent glycolytic pathway in Th17 cells, Mediators Kai min Mao started MD studies in 2018 focusing on lung
Inflamm. 2015 (2) (2015) 1–12 2015-12-20. related diseases, especially lung cancer. She is studying at
[74] C. Cavelti-Weder, K. Timper, E. Seelig, C. Keller, M. Osranek, U. Lässing, et al., the Department of Respiratory, Union Hospital, Tongji
Development of an Interleukin-1β vaccine in patients with type 2 diabetes, Mol. Medical College, Huazhong University of Science and
Ther. J. Am. Soc. Gene Ther. 24 (5) (2015) 1003–1012. Technology. Her current topic is to study early diagnosis of
[75] R. Garcia-Carbonell, A.S. Divakaruni, A. Lodi, I. Vicente-Suarez, A. Saha, pulmonary nodules by metabolomics, and the mechanism
H. Cheroutre, et al., Critical role of glucose metabolism in rheumatoid arthritis fi- of RORa in LPS-induced chronic bronchitis.
broblast-like synoviocytes, Arthritis Rheumatol. 68 (7) (2016) 1614–1626.
[76] H. Xie, J.I. Hanai, J.G. Ren, L. Kats, K. Burgess, P. Bhargava, et al., Targeting lactate
dehydrogenase-A inhibits tumorigenesis and tumor progression in mouse models of
lung cancer and impacts tumor-initiating cells, Cell Metab. 19 (5) (2014) 795–809.
[77] C.A. Dinarello, An expanding role for interleukin-1 blockade from gout to cancer,
Mol. Med. 20 (Suppl 1) (2014) S43.
[78] K. Timper, E. Seelig, D.A. Tsakiris, M.Y. Donath, Safety, pharmacokinetics, and
preliminary efficacy of a specific anti-IL-1alpha therapeutic antibody (MABp1) in
patients with type 2 diabetes mellitus, J. Diabetes Complicat. 29 (7) (2015) Guang hai Yang obtained his Master of Medicine in Union
955–960. Hospital, Tongji Medical College, Huazhong University of
[79] T. Kanni, M. Argyropoulou, T. Spyridopoulos, A. Pistiki, M. Stecher, C.A. Dinarello, Science and Technology in 2004.Now he is a doctor of
et al., MABp1 targeting interleukin-1Alpha for moderate to severe hidradenitis Department of Thoracic Surgery, Union Hospital, Tongji
suppurativa not eligible for adalimumab: a randomized study, J. Invest. Dermatol. Medical College, Huazhong University of Science and
138 (4) (2018). Technology.His research interest includes the biological
[80] T. Hickish, T. Andre, L. Wyrwicz, M. Saunders, T. Sarosiek, J. Kocsis, et al., MABp1 targeting therapy for the treatment of lung cancer.
as a novel antibody treatment for advanced colorectal cancer: a randomised,
double-blind, placebo-controlled, phase 3 study, Lancet Oncol. 18 (2) (2017) 192.
[81] J.J. Fox, S.C. Gavane, E. Blancautran, S. Nehmeh, M. Gönen, B. Beattie, et al.,
Positron emission Tomography/Computed tomography-based assessments of an-
drogen receptor expression and glycolytic activity as a prognostic biomarker for
metastatic castration-resistant prostate cancer, JAMA Oncol. (2017) e173588.

Qi Tan started his postgraduate work at the Department of Ping Luo obtained her PhD title in the Dalian Institute of
Respiratory, Union Hospital, Tongji Medical College, Chemical Physics, Chinese Academy of Sciences in 2017,
Huazhong University of Science and Technology in working at the Center for Translational Medicine, Union
2017.His research interests in the role and mechanism of Hospital, Tongji Medical College, Huazhong University of
action of inflammatory cytokines in cancer metabolism. Science and Technology since 2017. Her work mainly fo-
cuses on the methods development of liquid chromato-
graphy-mass spectrometry-based metabolomics and appli-
cations in diseases study. Main clinical and research interest
includes investigation of metabolic disorders in different
diseases and discovery of novel biomarkers for diseases
diagnosis and prognosis through application of transla-
tional medicine technology.

26
Q. Tan et al. Cytokine and Growth Factor Reviews 44 (2018) 18–27

Guan zhou Ma started MD studies at Union Hospital, Professor Yang Jin obtained his PhD in Union Hospital,
Tongji Medical College, Huazhong University of Science Tongji Medical College, Huazhong University of Science
and Technology in 2018. His research is centered on the and Technology in 2003.He is a doctor of the Department of
potential relationship between a series of environmental Respiratory, Union Hospital, Tongji Medical College,
exposures and respiratory disease. Huazhong University of Science and Technology.His re-
search focuses on cancer metabolism and the role of gly-
colysis in lung cancer cells.He is head of scientific research
office in Union Hospital, He is also a member of the Chinese
Thoracic Society.

PeiYuan Mei started his postgraduate work at Department


of Cardiothoracic Surgery, Union Hospital, Tongji Medical
College, Huazhong University of Science and Technology in
2017. His research focuses on the relationship between in-
flammatory cytokines from IL-1 family and glucose meta-
bolism of lung cancer cells.

27

Vous aimerez peut-être aussi