Skip to main content
Cellular and Molecular Immunology logoLink to Cellular and Molecular Immunology
. 2020 Jun 29;17(8):822–833. doi: 10.1038/s41423-020-0489-5

ACOD1 in immunometabolism and disease

Runliu Wu 1, Feng Chen 1, Nian Wang 1, Daolin Tang 1,, Rui Kang 1,
PMCID: PMC7395145  PMID: 32601305

Abstract

Immunometabolism plays a fundamental role in health and diseases and involves multiple genes and signals. Aconitate decarboxylase 1 (ACOD1; also known as IRG1) is emerging as a regulator of immunometabolism in inflammation and infection. Upregulation of ACOD1 expression occurs in activated immune cells (e.g., macrophages and monocytes) in response to pathogen infection (e.g., bacteria and viruses), pathogen-associated molecular pattern molecules (e.g., LPS), cytokines (e.g., TNF and IFNs), and damage-associated molecular patterns (e.g., monosodium urate). Mechanistically, several immune receptors (e.g., TLRs and IFNAR), adapter proteins (e.g., MYD88), ubiquitin ligases (e.g., A20), and transcription factors (e.g., NF-κB, IRFs, and STATs) form complex signal transduction networks to control ACOD1 expression in a context-dependent manner. Functionally, ACOD1 mediates itaconate production, oxidative stress, and antigen processing and plays dual roles in immunity and diseases. On the one hand, activation of the ACOD1 pathway may limit pathogen infection and promote embryo implantation. On the other hand, abnormal ACOD1 expression can lead to tumor progression, neurodegenerative disease, and immune paralysis. Further understanding of the function and regulation of ACOD1 is important for the application of ACOD1-based therapeutic strategies in disease.

Key words: ACOD1, immunometabolism, disease

Subject terms: Gene regulation in immune cells, Signal transduction

Introduction

Pathogens are microorganisms, including viruses, bacteria, fungi, protozoa, and worms, that can cause various human diseases. Potential hosts have two lines of defense against microbial infections. The first line of defense is innate immunity (also known as natural immunity), which rapidly destroys invading pathogens, mainly through phagocytosis mediated by myeloid cells (e.g., neutrophils, monocytes, macrophages, and dendritic cells [DCs]).1,2 The second line of defense is adaptive immunity (also known as acquired immunity), which relies on cytotoxic T cells or antigen-antibody responses mediated by lymphocytes (e.g., T cells and B cells)3. After successfully preventing an infection, the host will have an immune memory of the invading pathogen. If the same pathogen reappears, the host can fight the infection more effectively. In contrast, a dysfunctional immune system may cause inflammation and autoimmune diseases.

Immunometabolism describes changes in the interactions between immune and metabolic pathways during stress, which is a complex dynamic process involving many regulators or signals.4,5 Among them, aconitate decarboxylase 1 (ACOD1, also known as immunoresponsive gene 1 [IRG1]), has attracted much attention as a multifunctional regulator of inflammation and infection.6,7 ACOD1 expression is markedly upregulated by invading pathogens (e.g., Escherichia coli [E. coli], Mycobacterium tuberculosis [Mtb], and Zika virus [ZIKV]) and their derivatives (e.g., lipopolysaccharide [LPS] and CpG DNA).812 In addition, cytokines (e.g., interferon beta [IFNB] and tumor necrosis factor [TNF]), hormones (e.g., progesterone and estradiol), and certain compounds (e.g., cycloheximide [CHX]) also trigger ACOD1 expression under some conditions.8,13,14 Abnormal ACOD1 expression is associated with inflammatory responses, antibacterial processes, tumorigenesis, neurodegenerative changes, and embryo implantation.1519 Consequently, an impaired ACOD1 pathway affects metabolism-related innate immune signaling, leading to a variety of diseases.2022 In this review, we highlight the key features of the ACOD1 protein in immunometabolism and discuss its potential pathological roles in diseases. This knowledge can provide a new perspective for the development of effective treatments for ACOD1-related diseases.

Discovery of ACOD1

As stated above, the innate immune response is the first barrier against microorganisms, activating signaling pathways to trigger the secretion of cytokines or the expression of genes involved in immune responses.2 This process relies on host pattern recognition receptors (PRRs) to recognize pathogen-associated molecular patterns (PAMPs), which are the components of microorganisms.23,24 LPS is a typical PAMP in the outer membrane of gram-negative bacteria and has been widely used to study the mechanisms of innate immunity. In 1995, the Acod1 gene was first cloned as a 2.3-kb cDNA from a mouse macrophage cell line (RAW264.7 cells) stimulated with LPS (50 ng/ml).8 This original study showed that Acod1 transcripts could be detected as early as 1.5 h after stimulation, and the maximum level was observed at 4–6 h after LPS stimulation, with a 25-fold increase in induction.8 The human ACOD1 gene is located on chromosome 13q22.3 and is highly conserved among genes from other species, such as chimpanzees, dogs, cattle, mice, rats, chickens, zebrafish, Xenopus, and Aspergillus.16,2527 ACOD1 is thought to be an enzyme that is active in itaconate metabolism and is mainly found in mitochondria.14,16,28 Recently, the crystal structure was analyzed, and eight active-site residues (Asp93, Thr97, His103, His159, Lys207, Lys272, His277, and Tyr318) were found to be critical for human ACOD1 function, whereas mutations in these residues could lead to a substantial loss of ACOD1 enzymatic activity and decreased itaconate production.27 However, specific inhibitors targeting these ACOD1 sites have not yet been identified.

Induction of ACOD1

Infection with live pathogens can rapidly induce ACOD1 expression in vitro and in vivo (Table 1).16 For example, Acod1 mRNA expression is induced by Mtb infection in murine bone marrow-derived macrophages (BMDMs), with an approximately fivefold increase at 8 h after infection.9 At 48 h after whole-brain infeciton with ZIKV by intracranial administration, tissue homogenates were analyzed, and the expression of Acod1 mRNA was upregulated 16-fold compared to that of the control group.11 These findings suggest that the upregulation of ACOD1 may be important even in the response to bacterial and viral infections.

Table 1.

Pathogens that induce ACOD1 expression

Pathogens Host Burden Induction time Fold change Function Animal model Ref
Bacteria
 Staphylococcus aureus Blood from healthy human donors 1 × 106/ml 4 h 4 Sepsis biomarker NA 12
 Salmonella enterica serovar Typhimurium NA 600 CFU 10–50 Antimicrobial activity Zebrafish 16
 Salmonella enterica serovar Enteritidis

Cecum

Spleen

107 CFU

107 CFU

4 dpi

4 dpi

222

96.62

NA

NA

S. Enteritidis-infected chickens 102104
 Escherichia coli Blood from healthy human donors 4 ×103/ml 4 h 4 Sepsis biomarker NA 12
 Mycobacterium avium subspecies paratuberculosis

RAW264.7 cells

J774A.1 cells

BMDMs

NA 2–6 h 4 NA NA 40
 Mycobacterium smegmatis RAW264.7 cells NA 8 h NA NA NA 40
 Mycobacterium tuberculosis WT, Tlr2−/−, Tlr4−/−, Myd88−/−, Tirap−/−, and Trif−/− MOI 5 24 h >4 NA WT, Tlr2−/−/Tlr4−/−, and Myd88-/ C57BL/6 mice 9
BMDMs 600 CFU 21 dpi >3
Lung
 Borrelia burgdorferi spirochete J774A.1 cells NA 24 h 196.02 NA NA 90
 Burkholderia pseudomallei LPS mutant strain RAW264.7 cells MOI 2 4 h NA NA NA 48
 Legionella pneumophila

Lung

BMDMs

1 × 106/40 µl in vivo 8 h

100

400

Antimicrobial activity L. pneumophila-infected C57BL/6 J mice 28
 Chlamydia pneumoniae

Lung

ANA-1 cells

2.5 ×106 IFU

MOI 10

3 dpi

8 h

20.1

9.1

NA C. pneumoniae-infected C57BL/6 mice 46
 Listeria monocytogenes Spleen 0.1 × LD50 48 h 500 Mitochondrial localization L. monocytogenes-infected C57BL/6 mice 14
Virus
 Respiratory syncytial virus

A549 cells

Lung

MOI 1

5 ×106 PFU/100 µl in vivo

36 h 6 Promote ROS production and immune lung injury RSV-infected BALB/c mice 93
 Zika virus

Primary neurons

Brain

MOI 0.1 in vitro 24 h >8 Antimicrobial activity ZIKV-infected C57BL/6NJ and C57BL/6 J mice 11
103 PFU/50 µl in vivo >16
 Marek’s disease virus Spleen 103 PFU/200 µl 2–4 dpi 14 SNPs promote MDV susceptibility MDV-infected chicks 89
 Influenza A virus Lung 2 ×103 FFU 48 h 8 NA IAV-infected DBA/2 J mice 105
 West Nile virus

Brain

Primary neurons

102 PFU/50 µl in vivo 8 dpi 4 Antimicrobial activity WNV-infected C57BL/6NJ mice 11
MOI 0.001 24 h >4
Vesicular stomatitis virus

PMs

Lung

Liver

Spleen

MOI 0.1

5 × 107 PFU/g

5 × 107 PFU/g

5 × 107 PFU/g

8 h

24 h

24 h

24 h

>250

>100

>200

>400

Promote viral replication VSV-infected C57BL/6 J mice 22
 Herpes simplex virus 1

PMs

Lung

Liver

Spleen

MOI 1

5 × 107 PFU/g

5 × 107 PFU/g

5 × 107 PFU/g

8 h

24 h

24 h

24 h

300

20

10

20

Promote viral replication HSV-1–infected C57BL/6 J mice 22
Parasite
 Toxoplasma gondii

Spleen

Lung

20 cysts

12 dpi

12 dpi

2

10

Mitochondrial localization T. gondii-infected C57BL/6 mice 14
 Leishmania donovani

Splenic macrophages

Spleen

5 Parasites per macrophage

1 ×106

28 dpi

>10

>100

NA L. donovani-infected hamsters 32

Infection-mediated ACOD1 upregulation is related to the activation of Toll-like receptors (TLRs), a class of proteins that play a key role in innate immunity (Table 2). Many studies indicate that TLR4 is required for LPS (10 ng/ml to 10 µg/ml)- or lipid A (1 µg/ml)-induced ACOD1 expression in primary immune cells (e.g., human peripheral blood mononuclear cells [PBMCs], human PBMC-derived macrophages and DCs, murine peritoneal macrophages [PMs], murine BMDMs and murine bone marrow-derived dendritic cells) and cell lines (e.g., RAW264.7 cells, immortalized BMDMs, and THP1 [a human peripheral blood monocyte from acute monocytic leukemia] cells). In addition, TLR2 is responsible for lipoteichoic acid (LTA; 1 µg/ml)- or botulinum neurotoxin type A (BoNT/A; 1 nM or 5 nM)-induced Acod1 mRNA expression in ANA-1 cells, BMDMs, and RAW264.7 cells, whereas TLR9 facilitates CpG-DNA (1 µM)-induced Acod1 mRNA expression in BMDMs.14,29 The effects of poly I:C, a TLR3 ligand, on ACOD1 expression are context-dependent. A high concentration (40 µg/ml, 24 h) of poly I:C induces Acod1 mRNA expression in BMDMs,30 but a low concentration (1 µg/ml, 16 h) of poly I:C does not change ACOD1 expression in ANA-1 cells or BMDMs.14 As expected, TLR3 is required for high-concentration poly I:C-induced ACOD1 expression in macrophages. Although the administration of the TLR5 agonist KMRC011 in irradiated mice increases the expression of Acod1 mRNA in the small intestine,31 it has not been proven whether KMRC011 directly induces ACOD1 expression in cultured cells.31 It is also unclear whether other TLR members (e.g., TLR1, TLR6, TLR7, TLR8, TLR10, and TLR11) regulate ACOD1 expression in response to their corresponding ligands.

Table 2.

Reagents that induce ACOD1 expression

Compound Target Host cell Induction time Concentration Fold change Function Animal Model Ref
Infection
 LPS TLR4 RAW264.7 cells

4–6 h

6–9 h

6 h

50 ng/ml

10 ng/ml (055:B5)

10 ng/ml or 5 µg/ml

25

>400

NA

NA

NA

NA

NA

8, 50, 40, 53
ANA-1 cells 16 h 100 ng/ml (055:B5) 50 NA NA 14
J774A.1 cells 6 h 5 µg/ml NA NA NA 40
BMDMs

6 h

6 h

16 h

24 h

5 µg/ml

10 ng/ml

100 ng/ml (055:B5)

100 ng/ml

NA

NA

500

>50

NA

Promote itaconate production

NA

Promote itaconate production

NA

NA

NA

NA

40, 7, 14, 30
A20−/− BMDMs

1 h

6 h

10 µg/ml (0111:B4)

3

1.44

NA NA 53
WT, Myd88−/−, Ifnar−/−, and Stat−/− murine DCs 4 h 100 ng/ml (055:B5) NA NA NA 10
Tlr4-overexpressing ovine blood macrophages 1 h 1 µg/ml 3 Anti-inflammatory activity NA 45
Human monocyte-derived macrophages 3 h 100 ng/ml (0111:B4) 125 Promote itaconate production LPS-infected C57BL/6 mice 41
Human PBMC-derived monocytes, macrophages, and DCs 6–9 h 10 ng/ml (055:B5) >400 NA NA 50
Human PBMC-derived monocytes 4 h 10 ng/ml (055:B5) 25 NA NA 15
 Lipid A TLR4 WT and Myd88−/− PMs

4 h

8 h

1 µg/ml NA NA NA 44
 IFNB IFNAR

ANA-1 cells

BMDMs

16 h

16 h

24 h

10 ng/ml

10 ng/ml

1000 U/ml

9

3

>4

NA

NA

Promote itaconate production

NA 14, 30
 IFNG IFNGR ANA-1 cells 16 h 10 ng/ml 33.91 NA NA 14
Human PBMC-derived monocytes, macrophages, and DCs 6–9 h 10 ng/ml >400 NA NA 50
 TNF TNFR ANA-1 cells 16 h 10 ng/ml 5.89 NA NA 14
Human PBMC-derived monocytes, macrophages, and DCs 6–9 h 50 ng/ml >400 NA NA 50
A20−/− BMDMs

1 h

6 h

1000 U/ml

2.45

2.27

NA NA 53
 LTA TLR2

ANA-1 cells

BMDMs

16 h 1 µg/ml

9

50

NA NA 14
 BoNT/A TLR2 RAW264.7 cells

4 h

2 h

1 nM

5 nM

28.04

319.27

NA NA 29
 Poly I:C TLR3 BMDMs 24 h 40 µg/ml >50 NA WT and Ifnar1−/− C57BL/6 mice 30
 CpG1668 DNA TLR9 ANA-1 cells 16 h 1 µM 60 NA NA 14
 CpG DNA TLR9 WT, Ifnar−/−, and Stat−/− murine DCs 4 h 0.1 µM NA NA NA 10
 KMRC011 TLR5 Mouse small intestine Day 5 post TBI 0.2 mg/kg >2 NA Total-body irradiation (TBI) C57BL6/N mice
 IL1B IL1R

ANA-1

BMDMs

16 h 10 ng/ml

10

3

NA NA 14
Pregnancy
 Pregnancy Uteri Day 4 pregnancy NA  > 75 Facilitate embryo implantation CD1 female mice 13
 Progesterone PR Uteri Day 4 pregnancy

40 mg/kg

1 mg

Additional 50 ng estradiol

>100

>50

>75

Facilitate embryo implantation Ovariectomized CD1 female mice 13, 18
 LIF Uteri 12 h

5 µg

50 ng/ml

3.7

40

Facilitate embryo implantation WT and Lif−/− MF1 female mice 33, 34
Other
 CHX RAW264.7 cells 4 h 50 µM NA NA NA 8
 CoPP RAW264.7 cells 8–16 h 10 µM 8

Regulate A20 expression

Anti-inflammatory activity

LPS-infected C57BL6 mice 38
 Hemin RAW264.7 cells 8–16 h 10 µM 6.5

Regulate A20 expression

Anti-inflammatory activity

LPS-infected C57BL6 mice 38
 CORM-2 RAW264.7 cells 8–16 h 20 µM 4.5

Regulate A20 expression

Anti-inflammatory activity

LPS-infected C57BL6 mice 38
 Blast overpressure BV-2 cells Pulse duration of 12 ms 3.7 NA NA 39
 MSU Air pouch membrane 9 h 2 mg/ml 19.8 NA BALB/c mice 37

In addition to TLR ligands, certain inflammatory cytokines are strong inducers of ACOD1 expression (Table 2). Acod1 mRNA is highly upregulated in ANA-1 cells following treatment with IFNB (10 ng/ml), IFNG (10 ng/ml), and TNF (10 ng/ml or 50 ng/ml).14,28,32 IFNB (1000 U/ml) enhances LPS-induced Acod1 mRNA expression in BMDMs, and this process is blocked by the depletion of the type I interferon receptor (Ifnar), indicating that IFNAR is required for IFNB/LPS-induced ACOD1 expression.30 Similar to IFNB, interleukin-1β (IL1B; 10 ng/ml) also enhances LPS-induced Acod1 mRNA expression.14 Thus, there is a synergistic effect between PAMPs (e.g., LPS) and cytokines (e.g., IFNB and IL1B) that mediates the production of ACOD1 during inflammation.

In addition to infection, the upregulation of ACOD1 has also been observed during embryo implantation, which is a highly organized process that involves interactions between a receptive uterus and a competent blastocyst. In the early stage of pregnancy, progesterone (P4),13,18 estradiol (E2),13 and leukemia inhibitory factor (LIF)3335 play vital roles in embryo implantation. Microarray analysis18,36 and northern blot analysis13 identified Acod1 as a P4 response gene in the uterine epithelium. Although the mechanism of costimulation is unclear, P4-induced Acod1 mRNA expression can be further enhanced by administering E2 in luminal epithelial cells.13 Intrauterine administration of recombinant LIF also induces a 3.7-fold increase in Acod1 mRNA expression.33 Therefore, the upregulation of ACOD1 is implicated in P4-, E2-, and LIF-mediated implantation responses.

Small molecular compounds also have the ability to induce ACOD1 expression in immune cells (Table 2). As a protein synthesis inhibitor, CHX (50 µM) alone induces Acod1 mRNA expression in RAW264.7 cells at 4 h, suggesting that an interaction between protein synthesis and gene transcription may determine ACOD1 expression.8 Unlike cytokines, CHX fails to enhance LPS-induced Acod1 mRNA expression in RAW264.7 cells. Certain damage-associated molecular patterns, such as monosodium urate (MSU) or uric acid, can induce Acod1 expression in murine air pouch membranes,37 indicating a potential pathological role of ACOD1 in gout. Other reagents or stressors, including the CO inducer CO-releasing molecule (CORM-2),38 HO-1 inducer hemin or cobalt protoporphyrin IX (CoPP),38 and even stimulated explosive overpressure treatments, have been shown to induce ACOD1 expression.39 These findings further highlight ACOD1 as a stress-related inducible protein associated with inflammation, infection, and tissue damage.

Although ACOD1 expression can be elevated in most activated immune cells, the extent and peak time of the increase in ACOD1 will vary under different circumstances. The dynamic changes in Acod1 mRNA were first observed in LPS-stimulated macrophages, such as RAW264.7 cells, J774A.1 cells, and BMDMs. Acod1 mRNA can usually be detected after 2 h and reaches maximum expression levels ~6 h after LPS (10 ng/ml to 5 µg/ml) treatment.6,8,40 Unlike mouse macrophages, different kinetics of ACOD1 mRNA are observed in LPS-stimulated human PBMCs, and the expression levels peak is at 18 h.25 PAMPs, such as LPS, often induce higher ACOD1 expression than infection by live bacteria.40 Compared to individual stimulation in ANA-1 cells and BMDMs, certain cytokines (e.g., TNF and IFNG) produce a synergistic effect on Acod1 mRNA induction in the absence of LPS.14 The degree of ACOD1 induction during live pathogen infection also varies from species to species. Details are listed in Table 1.

Several amino acid metabolic pathways may affect ACOD1 expression in response to inflammatory stimuli. Branched-chain aminotransferase 1 (BCAT1) has been identified as a positive regulator of ACOD1 expression in activated macrophages.41 The loss of BCAT1 inhibits LPS-induced ACOD1 expression and reduces oxygen consumption and glycolysis, thereby preventing the infiltration and activation of macrophages.41 Since BCAT1 is the enzyme responsible for reversible transamination of some amino acids, the mechanism by which BCAT1 regulates ACOD1 may be related to amino acid metabolism. Indoleamine 2,3-dioxygenase 1 (IDO1) is a rate-limiting metabolic enzyme that converts the essential amino acid tryptophan into kynurenines. The expression of Acod1 mRNA is upregulated in primary microglial cells from Ido1−/− mice compared to wild-type (WT) mice,42 suggesting that tryptophan metabolism has a potential role in regulating ACOD1 expression. However, whether the altered ACOD1 levels in turn regulate the metabolism of these amino acids is still unclear.43

Notably, ACOD1 is generally considered to be an inducible gene, and the baseline expression of ACOD1 is usually very low under normal conditions. However, ACOD1 can be detected in unstimulated human fetal PBMCs but not in human fetal tissue samples.25 It is unclear whether this difference is due to experimental detection methods or physiological stress.

Modulation of ACOD1

TLRs

TLRs are the most important PRRs in host immune cells for recognizing PAMPs. As discussed above, TLR2, TLR3, TLR4, TLR5, and TLR9 have been shown to mediate ACOD1 expression in response to LPS, LTA, BoNT/A, CpG-DNA, poly I:C, or KMRC011 (Fig. 1).8,10,14,2931 Depletion of Tlr4 or Tlr2 blocks ACOD1 expression in activated immune cells,29,44 whereas overexpression of Tlr4 in ovine macrophages promotes Acod1 mRNA expression.45 While TLR3- or TLR5-mediated Acod1 upregulation is observed under certain conditions, detailed signal transduction has not been reported. As a key adapter molecule, myeloid differentiation primary response 88 (MYD88), is recruited to the cytoplasmic portion of the TLR and triggers a downstream signal cascade.2 CpG DNA-induced Acod1 mRNA expression is diminished in Myd88−/− DCs, suggesting that TLR9-mediated ACOD1 upregulation is MYD88-dependent.10 Of note, Acod1 is one of the most upregulated genes in C. pneumoniae = Chlamydia pneumoniae infection, but this upregulation is not observed in Myd88−/− macrophages, suggesting that MYD88 is essential in mediating ACOD1 expression.46 Interestingly, MYD88 seems to not always be important for LPS-induced Acod1 mRNA expression in murine macrophages and DCs,10,44 indicating a context-dependent role of MYD88 in the regulation of LPS-induced ACOD1 expression. In addition, Mtb infection-mediated Acod1 mRNA expression in BMDMs is independent of TLR2 and TLR4, as well as their downstream adapter proteins, such as MYD88, TIR domain-containing adapter protein (TIRAP), and toll-like receptor adaptor molecule 1.9 Possible signaling pathways that induce ACOD1 expression in MYD88-deficient cells may involve the activation of interferon regulatory factor 3 (IRF3),44 nuclear factor-κB (NF-κB),44 IFNAR,9 and signal transducer and activator of transcription 1 (STAT1).9 Thus, TLR-signaling pathway-mediated ACOD1 induction is intricate and largely depends on the type of host cell and stimulation.

Fig. 1.

Fig. 1

Expression of ACOD1 in infection and immunity. ac Many inflammatory stimuli (e.g., PAMPs, IFNs, TNF, and IL1B) can induce ACOD1 expression by activating receptors (e.g., TLRs, IFNAR, TNFR, and IL1R) and transcription factors (such as NF-κB, IRFs, and STATs) in a context-dependent manner

IRFs

The IRF family, which consists of nine members in mammals, plays an important role in the regulation of immune responses by producing type I IFN and interferon-stimulated genes (ISGs).47 ACOD1 is generally regarded as an ISG in activated immune cells (Fig. 1). Pathogen infection (e.g., by Legionella pneumophila [L. pneumophila]28 and B. pseudomallei = Burkholderia pseudomallei48) leads to the production of type I and type II IFN and the subsequent expression of hundreds of ISGs, including ACOD1. In contrast, the loss of Ifnar limits inducible Acod1 mRNA expression in macrophages during Mtb9 or L. pneumophila infection28 or LPS stimulation.30 However, LPS or CpG DNA induces Acod1 mRNA expression in DCs in an IFNB-independent manner,10 although TLR4 and TLR9 are required for Acod1 mRNA upregulation in DCs.10 Inhibition of IRF9 expression by miR-93 reduces ACOD1 induction in a model of peripheral artery disease (Fig. 1a),49 whereas IRF3 is required for IFNB-induced Acod1 mRNA expression in ovine macrophages.45 In addition, a bioinformatics analysis suggested that IRF1 may regulate ACOD1 expression, but experimental evidence is still lacking.50 In summary, IFN induces ACOD1 expression mainly in an IFN-dependent manner.

A20

A20, also known as TNFα-inducible protein 3, is a cytokine-inducible protein that controls inflammatory responses by negatively regulating NF-κB signaling.51,52 Some studies have showed that LPS-, CORM-2- or hemin-mediated Acod1 mRNA upregulation is required for the subsequent upregulation of A20 in various types of macrophages.20,38 Consistently, overexpression of ACOD1 in LPS-tolerized RAW264.7 cells restores LPS-induced A20 expression.20 Genetic silencing of A20 eliminates ACOD1-dependent inhibition of proinflammatory cytokine (e.g., TNF, IL6, and IFNB) production, suggesting that A20 functions as a downstream signal of the ACOD1 pathway.20,38 Another proteomics study showed that ACOD1 expression is upregulated in A20−/− BMDMs in the absence or presence of LPS or TNF compared to that of WT cells, suggesting that A20 functions as a negative regulator of ACOD1 expression in macrophages.53 Thus, there may be a feedback modulation loop between A20 and ACOD1 expression, but this mechanism requires further study.

PKC

The protein kinase C (PKC) family of protein kinases regulates the activity of other proteins by phosphorylating the hydroxyl groups on the serine/threonine side chains of these target proteins.54 PKC has been identified as a positive regulator of LPS-induced ACOD1 expression through the phosphorylation of unknown substrates (Fig. 1a).8 Phorbol myristate acetate, an activator of the PKC pathway, can further augment LPS-induced Acod1 mRNA expression in macrophages. Consistently, pharmacological inhibition of PKC using H7 blocks LPS-induced Acod1 mRNA expression in RAW264.7 cells.8 In addition, genistein abolishes LPS-induced Acod1 mRNA expression by preventing tyrosine phosphorylation and PKC δ activation.8,55 Unlike PKC, the protein kinase A (PKA) family of protein kinases has anti-inflammatory effects that rely on cellular levels of cyclic AMP (cAMP).56 In contrast, the PKA agonist cAMP has no effect on inducible Acod1 mRNA expression.8 Taken together, these findings indicate that PKC but not PKA is required for LPS-induced ACOD1 expression in macrophages.

STATs

Seven mammalian STAT family members have been identified (STAT1, STAT2, STAT3, STAT4, STAT5A, STAT5B, and STAT6), which play multiple roles in orchestrating a cytokine-induced immune response during infection and cell death.57 Among them, STAT3 and STAT1 play important roles in regulating ACOD1 expression in different situations (Fig. 1b). Activation of the STAT3 and glucocorticoid receptor (GR) signaling pathways can synergistically induce Acod1 mRNA expression in macrophage lineage cells from a zebrafish model.16 In addition to STATs, the expression of ACOD1 by GR signaling requires the activation of CCAAT enhancer-binding protein beta, a transcription factor that contains a basic leucine zipper domain (Fig. 1b). In addition, the lack of Stat1 limits Mtb infection-induced Acod1 mRNA expression in BMDMs.9 Therefore, STAT is also required for ACOD1 induction under certain conditions.

Other

Posttranscriptional mechanisms are also implicated in the regulation of ACOD1 gene expression, including events that may cause Acod1 mRNA degradation or prevent translation initiation.58 For example, Acod1 mRNA in murine macrophages is increasingly stabilized by LPS treatment but not by infection with Mycobacterium avium subspecies paratuberculosis.40 In addition, a miRNA is defined as a 21–25 nucleotide single-stranded RNA that regulates mRNA translation or stability.59 MiR-378 was found to directly target and downregulate ACOD1 expression in glioma cells (Fig. 1b).60 Taken together, these findings indicate that Acod1 mRNA expression is regulated at multiple levels. It remains unknown whether protein degradation is involved in the modulation of ACOD1 levels in activated immune cells.

Functions of ACOD1

Immune cells undergo extensive metabolic reprogramming under inflammatory conditions. Activated macrophages usually switch from oxidative phosphorylation (OXPHOS) to glycolysis, along with increasing glucose consumption or reactive oxygen species (ROS) production for host defense.61,62 This metabolic reprogramming requires the expression of certain metabolic enzymes during macrophage activation. ACOD1 participates in immune responses through multiple pathways, including mediating the production of itaconate (an immune metabolite) and mitochondrial reactive oxygen species (mROS). In addition to metabolic functions, nonmetabolic functions are also important for ACOD1-mediated immune regulation. In the following sections, we discuss the main functions of ACOD1 in infection and immunity.

Itaconate metabolism

ACOD1 is a member of the bacterial 2-citrate citrate dehydratase family of proteins, whose enzymes are encoded by prpD.63 A metabolomics analysis of LPS-activated macrophages identified ACOD1 as an enzyme that catalyzes itaconate production through cis-itaconate decarboxylation.6,7 Increased itaconate production and secretion are observed in LPS- and IFNG-activated macrophages, whereas Acod1 depletion can prevent these processes.64 Moreover, ACOD1-mediated itaconate production has been shown to inhibit LPS-induced IL6, IL12, IL18, IL1B, and type I IFN production in BMDMs.21,30 Itaconate regulates the immune response through multiple mechanisms (Fig. 2a),64,65 as described below.

Fig. 2.

Fig. 2

Function of ACOD1 in infection and immunity. a ACOD1-mediated itaconate production leads to NFE2L2 activation, SDH inhibition, isocitrate lyase inhibition, and prenylation induction. b ACOD1-mediated ROS production contributes to antigen processing, pathogen killing, and A20 modulation. c ACOD1 mediates the utilization of fatty acids in OXPHOS to produce mROS for bacterial killing

Cysteine alkylation

Nuclear factor erythroid 2-like 2 (NFE2L2, also known as NRF2) is a master transcription factor in the response to oxidative stress and inflammation. NFE2L2 expression is reduced under normal conditions, which is due to protein degradation mediated by Kelch-like ECH-associated protein 1 (KEAP1), a component of the cullin 3-based E3 ubiquitin ligase complex.66 Itaconate is an anti-inflammatory metabolite that can inactivate KEAP1 by alkylating certain cysteine residues (cys-151, -257, -288, -273 and -297), resulting in NFE2L2 accumulation and activation.30,67 In contrast, this process is not observed in LPS-activated Acod1−/− macrophages.67 NFE2L2 activation promotes the expression of heme oxygenase 1 and the production of glutathione, thereby enhancing the anti-inflammatory and antioxidant capacity of immune cells.68 Indeed, itaconate limits the production of LPS-induced cytokines (e.g., IL1B and TNF) and IRGs in an NFE2L2-dependent manner.30 Consequently, itaconate-mediated activation of the NFE2L2 pathway prevents endotoxemia and liver ischemia-reperfusion injury in mice.30,69 Whether ACOD1 overexpression promotes NFE2L2 activation requires further characterization.

Itaconate can also inactivate glyceraldehyde 3-phosphate dehydrogenase (GAPDH) by directly alkylating the cysteine residue at position 22 (cys-22), thereby reducing IL1B, iNOS, and TNF production by limiting aerobic glycolysis.70 In contrast, the loss of Acod1 in macrophages promotes LPS-induced inflammation in part by enhancing GAPDH-dependent glycolysis. Overexpression of cys-22 mutant GAPDH results in a failure to reverse the inhibitory effect of itaconate.71 Taken together, these findings indicate that itaconate inhibits inflammation in part by cysteine alkylation of inflammatory or metabolic regulators.

SDH inhibition

Succinate dehydrogenase (SDH), also known as succinate:quinone oxidoreductase, is a key component of the citrate cycle and electron transport chain, contributing to mROS production and inflammatory conditions.72 SDH inactivation leads to the accumulation of succinic acid, thereby promoting hypoxia-inducible factor 1 subunit alpha-mediated IL1B production and the inflammatory response by inhibiting prolyl hydroxylase enzymes.73 Itaconate directly inhibits the enzymatic activity of SDH in vivo and in vitro by competitively blocking the active site of SDH.21,74 Both ACOD1 activation and itaconate production have been shown to increase succinate accumulation following LPS treatment.7,21 In contrast, the production of itaconate and succinate is blocked in Acod1−/− BMDMs after LPS and IFNG stimulation7,21 In addition to bacterial infections, ACOD1-mediated itaconate inhibits SDH activity in ZIKV-infected neurons in vivo and in vitro.11 Itaconate-mediated inhibition of the oxygen consumption rate occurs only in the presence of succinic acid and rotenone, and this process is abolished in Acod1-deficient macrophages,7,21,75 further highlighting that itaconate controls inflammation in a succinate-dependent manner. Notably, the excessive inhibition of SDH by ACOD1 and itaconate may cause immune tolerance.15 In contrast, the recovery of SDH expression by β-glucan can offset the immune paralysis caused by ACOD1 and itaconate, which in turn contributes to the complete tricarboxylic acid (TCA) cycle and enhanced immune response.15 Therefore, proper itaconate levels are necessary for immune regulation.

Isocitrate lyase inhibition

The glyoxylate shunt is a two-step metabolic pathway that serves as an alternative to the TCA cycle and generates energy for bacterial growth and survival.76 Apart from host cells, itaconate can be used as an antimicrobial metabolite by inhibiting the activity of isocitrate lyase, a key enzyme of the glyoxylate shunt.6

Prenylation induction

Prenylation is a site-specific lipid modification of proteins that plays a key role in the viral life cycle.77,78 During vesicular stomatitis virus (VSV) infection, ACOD1 promotes viral entry and overgrowth by producing itaconate, which can increase the expression of enzymes related to geranylgeranyl diphosphate (GGPP) synthesis and promote GGPP-mediated protein prenylation, rather than in an IFN-dependent manner. VSV replication is inhibited when itaconate production is downregulated.22,79 These findings indicate dual roles of itaconate in innate immunity, in a context-dependent manner.

Oxidative stress

ROS play a wide range of roles in immune defense, antibacterial effects, and signal transduction.80 In addition to directly killing pathogens by causing oxidative damage, ROS can also promote the expression of genes involved in immune regulation.81 ACOD1-mediated ROS production has been observed in LPS-untolerized and LPS-tolerized macrophages,20,38 which promotes the upregulation of A20 expression (Fig. 2b). ACOD1-mediated A20 expression may form a negative feedback loop to regulate NFκB signals, thereby limiting the production of proinflammatory cytokines38 or causing immune tolerance.20 Furthermore, ACOD1-mediated ROS production contributes to STAT1/3 pathway activation and subsequent cytokine production (Fig. 2b).82 These findings indicate dual roles of ACOD1-mediated oxidative stress in shaping the immune response in a downstream signal-dependent manner.

As a byproduct of OXPHOS, mROS aid in macrophage-mediated bactericidal clearance.83 Indeed, the induction of ACOD1 promotes the utilization of fatty acids in OXPHOS, thereby increasing mROS production and subsequently enhancing the antibacterial activity of macrophages (Fig. 2c).16 In contrast, the loss of Acod1 limits the production of mROS and inhibits bacterial killing capacity.16 It is worth noting that mROS-independent signals are also involved in ACOD1-mediated bactericidal clearance. For example, ACOD1 restricts the growth of L. pneumophila in alveolar macrophages through a TLR-dependent but not mROS-dependent mechanism.28 Given that ROS are an important cause of cell death, it is unclear whether ACOD1 regulates the death of host cells, especially by pyroptosis, which is a type of inflammatory cell death in immune cells.84

Antigen processing

T-cell recognition by antigen-presenting cells depends on the expression of a series of peptides that bind to the major histocompatibility complex class I (MHC I) and class II (MHC II) molecules.85 In particular, MHC I can recognize and bind viral proteins, present these proteins to cytotoxic T lymphocytes, and cause apoptosis in virally infected cells.86 It has been shown that ACOD1 upregulation is the bridge linking innate immunity and adaptive immunity. LPS- or IFNB-induced ACOD1 expression can enhance MHC I function by promoting the expression of transporter associated with antigen processing 1 (TAP1) and proteasome subunit beta 9 (PSMB9), which are transporter proteins associated with antigen processing (Fig. 2b).82,87 Furthermore, ACOD1-induced activation of MHC I exerts its antiviral effects through antigen presentation, although the exact mechanism is unknown.82 Recently, eight missense polymorphisms have been identified that are closely related to the active center of human ACOD1. Six of these polymorphisms are rare and may have no enzymatic activity.27 These findings also suggest that ACOD1 may affect susceptibility to certain diseases, such as hepatitis B virus infection in humans and Marek’s disease virus (MDV) infection in chickens, through its own nucleotide mutations.88,89 Single nucleotide polymorphism (SNP) analysis confirmed that two SNPs of the ACOD1 gene (rs17470171 and rs17385627) enhance the expression of ACOD1, which may activate the immune response to hepatitis B vaccination.88 Two other SNPs of ACOD1 were found at the transcriptional binding site, which is related to sensitivity to Marek’s disease (MD), while homozygous genotypes increase resistance to MD.89 The genetic variation of ACOD1 in other diseases merits further investigation.

Pathological role of ACOD1

Infectious diseases

In response to infection, host cells trigger the activation of immune pathways, which leads to the production of inflammatory mediators (e.g., cytokines, chemokines, and interferons) and maintains a balance between pro- and anti-inflammatory states. In this context, many studies have identified ACOD1 as one of the most upregulated genes under various conditions associated with infection (Table 1). For example, gene expression profiling revealed that infection with live B. burgdorferi can cause a 196.2-fold induction of Acod1 expression in J774A.1 macrophages,90 and stimulation with IFNG and TNF leads to a 73-fold upregulation of Acod1 expression in ANA-1 macrophages.14 These findings suggest that ACOD1 may function as an important alarm signal in the immune response to infection.

Several studies have shown that Acod1 prevents infection in mice. Compared with WT mice, Acod1 global knockout mice generally have poorer survival, inflammatory cytokine bursts, uncontrolled neutrophil recruitment, and immune-mediated tissue damage.11,28,91 Acod1 global knockout mice also exhibit higher mortality and viral burden in the brain than WT mice when infected with ZIKV and West Nile virus (WNV).11 Moreover, myeloid cell-specific Acod1 knockout mice are sensitive to Mtb infection and have an increased Mtb burden in the lungs.91 In vitro studies in Acod1-deficient BMDMs show enhanced replication of Lactobacillus halophilus.28 In primary neurons, Acod1 knockdown results in increased ZIKV and WNV replication,11,92 while Acod1 overexpression reduces the production of multiple viruses, including WNV, St. Louis encephalitis virus (SLEV), and mouse hepatitis virus (MHV).92 These in vitro and in vivo studies support the anti-infective effects of ACOD1 under multiple conditions.

However, ACOD1 does not always play a protective role in a complex inflammatory environment and sometimes leads to increased pathogen burden and tissue damage. Acod1 mRNA is highly upregulated in splenic macrophages of V. Leishmania-infected mice, which aids parasite growth and survival.32 ACOD1 may also contribute to Borrelia-induced immunopathology, as ACOD1 expression can be induced by Borrelia burgdorferi infection and suppressed by the anti-inflammatory cytokine IL10.90 In addition, ACOD1 expression contributes to mouse respiratory syncytial virus (RSV) infection, thereby promoting the production of proinflammatory cytokines and ROS, leading to extensive lung damage.93 Moreover, ACOD1-mediated itaconate production promotes the replication of VSV, while Acod1−/− mice show a reduction in inflammatory cell infiltration in the lung during VSV infection.22 The increase in ACOD1 and itaconate production in macrophages leads to the induction of M1-like polarization, leading to impaired angiogenesis, arteriogenesis, and perfusion in peripheral artery disease.49 Furthermore, the immunosuppressive effects of ACOD1 on endotoxin-resistant macrophages and monocytes may lead to immune paralysis and life-threatening secondary infection.15,20 Collectively, these studies suggest that ACOD1 plays dual roles in infection and may elicit different immune responses.

Cancer

Although many studies have focused on the role of ACOD1 in immune cells, especially macrophages, some studies have noted that dysregulated ACOD1 expression may promote tumorigenesis by modulating antitumor immunity. Although considerable progress has been made in surgery and conventional chemotherapy, ovarian cancer continues to have a high mortality rate in women. In the early and advanced stages of ovarian cancer, strong expression of ACOD1 was observed in malignant cells, which may limit the antitumor effect of infiltrating CD8+ T cells and IgY cell-containing cells in the tumor microenvironment (TME) through unknown mechanisms.94 Tumor-associated macrophages (TAMs) are key cells that produce an immunosuppressive TME. A recent study showed that ACOD1-mediated itaconate production is an immunometabolic component of crosstalk between tumors and TAMs within the TME.95 Cancer cells can increase ACOD1 expression in TAMs, leading to itaconate production, oxidative stress, and subsequent protumor macrophage polarization in the TME, ultimately promoting tumor progression in melanoma and ovarian cancer.95 ACOD1 plays an oncogene-like role in gliomas and can promote the growth and invasion of cancer cells, resulting in a poor prognosis.17,60 In contrast, intrauterine bisphenol A exposure inhibits Acod1 mRNA expression, which may promote breast cancer cell growth in mice.96 These findings also suggest that ACOD1 plays dual roles in tumor biology, but the direct role of ACOD1 in cancer cells remains to be fully elucidated.

Neurological disorders

Microglia are the most abundant macrophages in the central nervous system (CNS) and play a dynamic role in maintaining immune homeostasis in the brain. Activation of microglia from a priming state to a destructive state contributes to the neuropathy of CNS diseases.97 Recent studies have reported that upregulated ACOD1 is involved in the initiation and activation of microglia. Mice primed and reinfected with the intracranial parasite Toxoplasma gondii (T. gondii) had high gene expression of Acod1 and Ccr9 in microglia, thereby increasing microglial resistance to apoptosis and subsequent neurotoxicity.19 In contrast, inhibiting Acod1 in parasite-infected mice reduces parasitic DNA levels in brain tissue and shifts microglia into a protective phenotype.19 The production of Acod1-deficient microglia is triggered in lymphocytic choriomeningitis virus (LCMV)-infected mice, which promotes neuronal regeneration. This finding suggests that increased ACOD1 expression can be considered a marker of neurotoxic microglia. Elevated Acod1 mRNA is also observed in microglia in high-fat diet mice or mice infected with neurotrophic pathogens (e.g., T. gondii, HIV vaccine, recombinant A3A capsid protein of Theiler’s murine encephalomyelitis virus, and β-amyloid protein fragments). This change in ACOD1 expression is associated with neurotoxic microglial activation and chronic neuroinflammation, which causes progressive neuronal loss and ultimately leads to cognitive decline and neurodegenerative diseases, such as Alzheimer’s disease and dementia.19,98 Although it is not clear how ACOD1 specifically regulates the immune function of microglia, ACOD1 may exert its enzymatic effects by altering microglial metabolism. The precise contributions of ACOD1 in other tissue-resident macrophages remain unclear.

Embryo implantation

Embryo implantation takes place in the first trimester of pregnancy, and this stage is related to the production of a large number of inflammatory molecules by epithelial cells and immune cells.99 This proinflammatory condition is regulated by ovarian steroid hormones, estradiol, and progesterone, which help and protect early pregnancy. Regulation of the expression of certain inflammatory molecules is essential for obtaining and developing uterine receptivity, which is one of the key factors for successful implantation. ACOD1 has been shown to be related to implantation. Acod1 mRNA expression in the luminal epithelium has been shown to occur in a specific manner at the implantation stage, increase on day 3 after pregnancy, peak at day 4, and abruptly decrease at day 5.18 In mice, day 4 after pregnancy is the time at which the blastocyst disappears from the recipient endometrium.100 The administration of progesterone for 24 h in ovariectomized mice can induce expression of Acod1 mRNA. This increase in Acod1 expression can be reversed by pharmacologically (e.g., with PR486) or genetically inhibiting the progesterone receptor (PR).18,36 A similar inhibition of Acod1 expression is observed when mice are treated with PKC inhibitors during implantation, while E2 and P4 synergistically and independently increase PKC activity on day 4 of pregnancy.13 These findings indicate that the PR pathway is essential for P-responsive ACOD1 expression in the uterine epithelium by regulating PKC activity. Triggered by estrogen, the level of the endometrial cytokine LIF, which is also a well-known modulator of successful implantation, increases and peaks on day 4.101 In pseudopregnant Lif-deficient mice, an intrauterine Lif injection promotes Acod1 expression in the uterus through Stat3 activation.33,34 Therefore, the injection of anti-Lif antibodies or Stat3 peptide inhibitors into the uterine cavity of pregnant mice on day 3 prevents the induction of Acod1 and subsequent embryo implantation.34,35 In addition, the protective role of Acod1 in the implantation process has been further demonstrated by using Acod1-related antisense oligodeoxynucleotides on the lumen on day 2.18 These findings highlight the need for ACOD1 in embryo implantation, despite the complex molecular network of the implantation process.

Conclusions and perspectives

ACOD1 has emerged as an important modulator with different functions in innate and adaptive immunity. Activation of the ACOD1 pathway may limit pathogen infection and promote embryo implantation. Moreover, abnormal ACOD1 expression can cause tumor progression, neurodegeneration, and immune paralysis. Although the dual roles of ACOD1 in immunity are mainly related to ACOD1-mediated itaconate metabolism, oxidative stress, and antigen processing, more research is needed to clarify the roles and regulatory mechanisms of ACOD1 in both immune and nonimmune cells. In addition to the broad role of ACOD1 in innate immunity, ACOD1 may regulate adaptive immunity by promoting MHC I expression82. However, how to express ACOD1 in specialized antigen-presenting cells to regulate adaptive immunity in diseases requires further investigation.

The ACOD1-itaconate axis is becoming a target of disease treatment. In particular, the development of metabolically stable itaconate derivatives may have great potential in the treatment of inflammatory diseases. In contrast, for patients with peritoneal tumors, inhibition of ACOD1 may be beneficial because ACOD1-expressing TAMs mediate tumor growth.95 With the discovery of its crystal structure, technical modifications of the ACOD1 protein to interfere with or improve its activity may be a promising therapeutic strategy. Some agents have been found to inhibit the induction of ACOD1 under certain conditions, such as dexamethasone,8 ethylene glycol tetraacetic acid,8 and β-glucan.15 Further research needs to not only distinguish the overlapping and unique functions of ACOD1 and itaconic acid in immunity but also evaluate the pathological consequences of drugs targeting ACOD1 on disease.

Acknowledgements

We thank Dave Primm (Department of Surgery, University of Texas Southwestern Medical Center) for his critical reading of the manuscript.

Competing interests

The authors declare no competing interests.

Contributor Information

Daolin Tang, Email: daolin.tang@utsouthwestern.edu.

Rui Kang, Email: rui.kang@utsouthwestern.edu.

References

  • 1.Mogensen TH. Pathogen recognition and inflammatory signaling in innate immune defenses. Clin. Microbiol Rev. 2009;22:240–273. doi: 10.1128/CMR.00046-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Akira S, Uematsu S, Takeuchi O. Pathogen recognition and innate immunity. Cell. 2006;124:783–801. doi: 10.1016/j.cell.2006.02.015. [DOI] [PubMed] [Google Scholar]
  • 3.Chaplin DD. Overview of the immune response. J. Allergy Clin. Immunol. 2010;125:S3–S23. doi: 10.1016/j.jaci.2009.12.980. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.O’Neill LA, Kishton RJ, Rathmell J. A guide to immunometabolism for immunologists. Nat. Rev. Immunol. 2016;16:553–565. doi: 10.1038/nri.2016.70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Mathis D, Shoelson SE. Immunometabolism: an emerging frontier. Nat. Rev. Immunol. 2011;11:81. doi: 10.1038/nri2922. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Michelucci A, et al. Immune-responsive gene 1 protein links metabolism to immunity by catalyzing itaconic acid production. Proc. Natl Acad. Sci. USA. 2013;110:7820–7825. doi: 10.1073/pnas.1218599110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Cordes T, et al. Immunoresponsive gene 1 and itaconate inhibit succinate dehydrogenase to modulate intracellular succinate levels. J. Biol. Chem. 2016;291:14274–14284. doi: 10.1074/jbc.M115.685792. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Lee CG, Jenkins NA, Gilbert DJ, Copeland NG, O’Brien WE. Cloning and analysis of gene regulation of a novel LPS-inducible cDNA. Immunogenetics. 1995;41:263–270. doi: 10.1007/BF00172150. [DOI] [PubMed] [Google Scholar]
  • 9.Shi S, et al. Expression of many immunologically important genes in Mycobacterium tuberculosis-infected macrophages is independent of both TLR2 and TLR4 but dependent on IFN-alphabeta receptor and STAT1. J. Immunol. 2005;175:3318–3328. doi: 10.4049/jimmunol.175.5.3318. [DOI] [PubMed] [Google Scholar]
  • 10.Hoshino K, Kaisho T, Iwabe T, Takeuchi O, Akira S. Differential involvement of IFN-beta in Toll-like receptor-stimulated dendritic cell activation. Int Immunol. 2002;14:1225–1231. doi: 10.1093/intimm/dxf089. [DOI] [PubMed] [Google Scholar]
  • 11.Daniels BP, et al. The nucleotide sensor ZBP1 and kinase RIPK3 induce the enzyme IRG1 to promote an antiviral metabolic state in neurons. Immunity. 2019;50:64–76. e64. doi: 10.1016/j.immuni.2018.11.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Dix A, et al. Biomarker-based classification of bacterial and fungal whole-blood infections in a genome-wide expression study. Front. Microbiol. 2015;6:171. doi: 10.3389/fmicb.2015.00171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Chen B, Zhang D, Pollard JW. Progesterone regulation of the mammalian ortholog of methylcitrate dehydratase (immune response gene 1) in the uterine epithelium during implantation through the protein kinase C pathway. Mol. Endocrinol. 2003;17:2340–2354. doi: 10.1210/me.2003-0207. [DOI] [PubMed] [Google Scholar]
  • 14.Degrandi D, Hoffmann R, Beuter-Gunia C, Pfeffer K. The proinflammatory cytokine-induced IRG1 protein associates with mitochondria. J. Interferon Cytokine Res. 2009;29:55–67. doi: 10.1089/jir.2008.0013. [DOI] [PubMed] [Google Scholar]
  • 15.Dominguez-Andres J, et al. The itaconate pathway is a central regulatory node linking innate immune tolerance and trained immunity. Cell Metab. 2019;29:211–220. e215. doi: 10.1016/j.cmet.2018.09.003. [DOI] [PubMed] [Google Scholar]
  • 16.Hall CJ, et al. Immunoresponsive gene 1 augments bactericidal activity of macrophage-lineage cells by regulating beta-oxidation-dependent mitochondrial ROS production. Cell Metab. 2013;18:265–278. doi: 10.1016/j.cmet.2013.06.018. [DOI] [PubMed] [Google Scholar]
  • 17.Pan J, et al. Immune responsive gene 1, a novel oncogene, increases the growth and tumorigenicity of glioma. Oncol. Rep. 2014;32:1957–1966. doi: 10.3892/or.2014.3474. [DOI] [PubMed] [Google Scholar]
  • 18.Cheon YP, Xu X, Bagchi MK, Bagchi IC. Immune-responsive gene 1 is a novel target of progesterone receptor and plays a critical role during implantation in the mouse. Endocrinology. 2003;144:5623–5630. doi: 10.1210/en.2003-0585. [DOI] [PubMed] [Google Scholar]
  • 19.Li H, et al. Different neurotropic pathogens elicit neurotoxic CCR9- or neurosupportive CXCR3-expressing microglia. J. Immunol. 2006;177:3644–3656. doi: 10.4049/jimmunol.177.6.3644. [DOI] [PubMed] [Google Scholar]
  • 20.Li Y, et al. Immune responsive gene 1 (IRG1) promotes endotoxin tolerance by increasing A20 expression in macrophages through reactive oxygen species. J. Biol. Chem. 2013;288:16225–16234. doi: 10.1074/jbc.M113.454538. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Lampropoulou V, et al. Itaconate links inhibition of succinate dehydrogenase with macrophage metabolic remodeling and regulation of inflammation. Cell Metab. 2016;24:158–166. doi: 10.1016/j.cmet.2016.06.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Yin, S. et al. The IRG1-itaconate axis promotes viral replication via metabolic reprogramming and protein prenylation. Cell Metab.19-00910, 54 (2019).
  • 23.Brubaker SW, Bonham KS, Zanoni I, Kagan JC. Innate immune pattern recognition: a cell biological perspective. Annu. Rev. Immunol. 2015;33:257–290. doi: 10.1146/annurev-immunol-032414-112240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Medzhitov R, Janeway C., Jr. Innate immunity. N. Engl. J. Med. 2000;343:338–344. doi: 10.1056/NEJM200008033430506. [DOI] [PubMed] [Google Scholar]
  • 25.Xiao W, et al. Expression profile of human immune-responsive gene 1 and generation and characterization of polyclonal antiserum. Mol. Cell Biochem. 2011;353:177–187. doi: 10.1007/s11010-011-0784-7. [DOI] [PubMed] [Google Scholar]
  • 26.Schmidt G, Richter K. Expression pattern of XIRG, a marker for non-neural ectoderm. Dev. Genes Evol. 2000;210:575–578. doi: 10.1007/s004270000095. [DOI] [PubMed] [Google Scholar]
  • 27.Chen F, et al. Crystal structure of cis-aconitate decarboxylase reveals the impact of naturally occurring human mutations on itaconate synthesis. Proc. Natl Acad. Sci. USA. 2019;116:20644–20654. doi: 10.1073/pnas.1908770116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Naujoks J, et al. IFNs modify the proteome of legionella-containing vacuoles and restrict infection via IRG1-derived itaconic acid. PLoS Pathog. 2016;12:e1005408. doi: 10.1371/journal.ppat.1005408. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Kim YJ, et al. Botulinum neurotoxin type A induces TLR2-mediated inflammatory responses in macrophages. PLoS ONE. 2015;10:e0120840. doi: 10.1371/journal.pone.0120840. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Mills EL, et al. Itaconate is an anti-inflammatory metabolite that activates Nrf2 via alkylation of KEAP1. Nature. 2018;556:113–117. doi: 10.1038/nature25986. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Kim JY, et al. Radioprotective effect of newly synthesized toll-like receptor 5 agonist, KMRC011, in mice exposed to total-body irradiation. J. Radiat. Res. 2019;60:432–441. doi: 10.1093/jrr/rrz024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Kong F, et al. Transcriptional profiling in experimental visceral leishmaniasis reveals a broad splenic inflammatory environment that conditions macrophages toward a disease-promoting phenotype. PLoS Pathog. 2017;13:e1006165. doi: 10.1371/journal.ppat.1006165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Sherwin JR, et al. Identification of genes regulated by leukemia-inhibitory factor in the mouse uterus at the time of implantation. Mol. Endocrinol. 2004;18:2185–2195. doi: 10.1210/me.2004-0110. [DOI] [PubMed] [Google Scholar]
  • 34.Catalano RD, et al. Inhibition of Stat3 activation in the endometrium prevents implantation: a nonsteroidal approach to contraception. Proc. Natl Acad. Sci. USA. 2005;102:8585–8590. doi: 10.1073/pnas.0502343102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Terakawa J, et al. Embryo implantation is blocked by intraperitoneal injection with anti-LIF antibody in mice. J. Reprod. Dev. 2011;57:700–707. doi: 10.1262/jrd.11-048h. [DOI] [PubMed] [Google Scholar]
  • 36.Cheon YP, et al. A genomic approach to identify novel progesterone receptor regulated pathways in the uterus during implantation. Mol. Endocrinol. 2002;16:2853–2871. doi: 10.1210/me.2002-0270. [DOI] [PubMed] [Google Scholar]
  • 37.Pessler F, et al. Identification of novel monosodium urate crystal regulated mRNAs by transcript profiling of dissected murine air pouch membranes. Arthritis Res. Ther. 2008;10:R64. doi: 10.1186/ar2435. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Jamal Uddin M, et al. IRG1 induced by heme oxygenase-1/carbon monoxide inhibits LPS-mediated sepsis and pro-inflammatory cytokine production. Cell Mol. Immunol. 2016;13:170–179. doi: 10.1038/cmi.2015.02. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Kane MJ, et al. Altered gene expression in cultured microglia in response to simulated blast overpressure: possible role of pulse duration. Neurosci. Lett. 2012;522:47–51. doi: 10.1016/j.neulet.2012.06.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Basler T, Jeckstadt S, Valentin-Weigand P, Goethe R. Mycobacterium paratuberculosis, Mycobacterium smegmatis, and lipopolysaccharide induce different transcriptional and post-transcriptional regulation of the IRG1 gene in murine macrophages. J. Leukoc. Biol. 2006;79:628–638. doi: 10.1189/jlb.0905520. [DOI] [PubMed] [Google Scholar]
  • 41.Papathanassiu AE, et al. BCAT1 controls metabolic reprogramming in activated human macrophages and is associated with inflammatory diseases. Nat. Commun. 2017;8:16040. doi: 10.1038/ncomms16040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Gonzalez-Pena D, et al. Differential transcriptome networks between IDO1-knockout and wild-type mice in brain microglia and macrophages. PLoS ONE. 2016;11:e0157727. doi: 10.1371/journal.pone.0157727. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Munn DH, Mellor AL. Indoleamine 2,3 dioxygenase and metabolic control of immune responses. Trends Immunol. 2013;34:137–143. doi: 10.1016/j.it.2012.10.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Kawai T, et al. Lipopolysaccharide stimulates the MyD88-independent pathway and results in activation of IFN-regulatory factor 3 and the expression of a subset of lipopolysaccharide-inducible genes. J. Immunol. 2001;167:5887–5894. doi: 10.4049/jimmunol.167.10.5887. [DOI] [PubMed] [Google Scholar]
  • 45.Wei S, et al. Overexpression of Toll-like receptor 4 enhances LPS-induced inflammatory response and inhibits Salmonella Typhimurium growth in ovine macrophages. Eur. J. Cell Biol. 2019;98:36–50. doi: 10.1016/j.ejcb.2018.11.004. [DOI] [PubMed] [Google Scholar]
  • 46.Rodriguez N, et al. MyD88-dependent changes in the pulmonary transcriptome after infection with Chlamydia pneumoniae. Physiol. Genom. 2007;30:134–145. doi: 10.1152/physiolgenomics.00011.2007. [DOI] [PubMed] [Google Scholar]
  • 47.Zhao GN, Jiang DS, Li H. Interferon regulatory factors: at the crossroads of immunity, metabolism, and disease. Biochim. Biophys. Acta. 2015;1852:365–378. doi: 10.1016/j.bbadis.2014.04.030. [DOI] [PubMed] [Google Scholar]
  • 48.Tangsudjai S, et al. Involvement of the MyD88-independent pathway in controlling the intracellular fate of Burkholderia pseudomallei infection in the mouse macrophage cell line RAW 264.7. Microbiol. Immunol. 2010;54:282–290. doi: 10.1111/j.1348-0421.2010.00205.x. [DOI] [PubMed] [Google Scholar]
  • 49.Ganta VC, et al. A MicroRNA93-interferon regulatory factor-9-immunoresponsive gene-1-itaconic acid pathway modulates M2-like macrophage polarization to revascularize ischemic muscle. Circulation. 2017;135:2403–2425. doi: 10.1161/CIRCULATIONAHA.116.025490. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Tallam A, et al. Gene regulatory network inference of immunoresponsive gene 1 (IRG1) identifies interferon regulatory factor 1 (IRF1) as its transcriptional regulator in mammalian macrophages. PLoS ONE. 2016;11:e0149050. doi: 10.1371/journal.pone.0149050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Opipari AW, Jr., Boguski MS, Dixit VM. The A20 cDNA induced by tumor necrosis factor alpha encodes a novel type of zinc finger protein. J. Biol. Chem. 1990;265:14705–14708. [PubMed] [Google Scholar]
  • 52.Jaattela M, Mouritzen H, Elling F, Bastholm L. A20 zinc finger protein inhibits TNF and IL-1 signaling. J. Immunol. 1996;156:1166–1173. [PubMed] [Google Scholar]
  • 53.Van Quickelberghe E, et al. Identification of immune-responsive gene 1 (IRG1) as a target of A20. J. Proteome Res. 2018;17:2182–2191. doi: 10.1021/acs.jproteome.8b00139. [DOI] [PubMed] [Google Scholar]
  • 54.Newton AC. Protein kinase C: structure, function, and regulation. J. Biol. Chem. 1995;270:28495–28498. doi: 10.1074/jbc.270.48.28495. [DOI] [PubMed] [Google Scholar]
  • 55.Konishi H, et al. Activation of protein kinase C by tyrosine phosphorylation in response to H2O2. Proc. Natl Acad. Sci. USA. 1997;94:11233–11237. doi: 10.1073/pnas.94.21.11233. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Aronoff DM, Canetti C, Serezani CH, Luo M, Peters-Golden M. Cutting edge: macrophage inhibition by cyclic AMP (cAMP): differential roles of protein kinase A and exchange protein directly activated by cAMP-1. J. Immunol. 2005;174:595–599. doi: 10.4049/jimmunol.174.2.595. [DOI] [PubMed] [Google Scholar]
  • 57.Hanada T, Yoshimura A. Regulation of cytokine signaling and inflammation. Cytokine Growth Factor Rev. 2002;13:413–421. doi: 10.1016/s1359-6101(02)00026-6. [DOI] [PubMed] [Google Scholar]
  • 58.Day DA, Tuite MF. Post-transcriptional gene regulatory mechanisms in eukaryotes: an overview. J. Endocrinol. 1998;157:361–371. doi: 10.1677/joe.0.1570361. [DOI] [PubMed] [Google Scholar]
  • 59.Filipowicz W, Bhattacharyya SN, Sonenberg N. Mechanisms of post-transcriptional regulation by microRNAs: are the answers in sight? Nat. Rev. Genet. 2008;9:102–114. doi: 10.1038/nrg2290. [DOI] [PubMed] [Google Scholar]
  • 60.Shi HZ, Wang D, Sun XN, Sheng L. MicroRNA-378 acts as a prognosis marker and inhibits cell migration, invasion and epithelial-mesenchymal transition in human glioma by targeting IRG1. Eur. Rev. Med Pharmacol. Sci. 2018;22:3837–3846. doi: 10.26355/eurrev_201806_15268. [DOI] [PubMed] [Google Scholar]
  • 61.Kelly B, O’Neill LA. Metabolic reprogramming in macrophages and dendritic cells in innate immunity. Cell Res. 2015;25:771–784. doi: 10.1038/cr.2015.68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Pearce EL, Pearce EJ. Metabolic pathways in immune cell activation and quiescence. Immunity. 2013;38:633–643. doi: 10.1016/j.immuni.2013.04.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Lohkamp B, Bauerle B, Rieger PG, Schneider G. Three-dimensional structure of iminodisuccinate epimerase defines the fold of the MmgE/PrpD protein family. J. Mol. Biol. 2006;362:555–566. doi: 10.1016/j.jmb.2006.07.051. [DOI] [PubMed] [Google Scholar]
  • 64.Strelko CL, et al. Itaconic acid is a mammalian metabolite induced during macrophage activation. J. Am. Chem. Soc. 2011;133:16386–16389. doi: 10.1021/ja2070889. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Shin JH, et al. (1)H NMR-based metabolomic profiling in mice infected with Mycobacterium tuberculosis. J. Proteome Res. 2011;10:2238–2247. doi: 10.1021/pr101054m. [DOI] [PubMed] [Google Scholar]
  • 66.Kobayashi A, et al. Oxidative stress sensor Keap1 functions as an adaptor for Cul3-based E3 ligase to regulate proteasomal degradation of Nrf2. Mol. Cell Biol. 2004;24:7130–7139. doi: 10.1128/MCB.24.16.7130-7139.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Bambouskova M, et al. Electrophilic properties of itaconate and derivatives regulate the IkappaBzeta-ATF3 inflammatory axis. Nature. 2018;556:501–504. doi: 10.1038/s41586-018-0052-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Ahmed SM, Luo L, Namani A, Wang XJ, Tang X. Nrf2 signaling pathway: Pivotal roles in inflammation. Biochim. Biophys. Acta Mol. Basis Dis. 2017;1863:585–597. doi: 10.1016/j.bbadis.2016.11.005. [DOI] [PubMed] [Google Scholar]
  • 69.Yi Z, et al. IRG1/Itaconate activates Nrf2 in hepatocytes to protect against liver ischemia-reperfusion injury. Hepatology. 2020 doi: 10.1002/hep.31147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Kornberg MD, et al. Dimethyl fumarate targets GAPDH and aerobic glycolysis to modulate immunity. Science. 2018;360:449–453. doi: 10.1126/science.aan4665. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Liao ST, et al. 4-Octyl itaconate inhibits aerobic glycolysis by targeting GAPDH to exert anti-inflammatory effects. Nat. Commun. 2019;10:5091. doi: 10.1038/s41467-019-13078-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Mills EL, et al. Succinate dehydrogenase supports metabolic repurposing of mitochondria to drive inflammatory macrophages. Cell. 2016;167:457–470. e413. doi: 10.1016/j.cell.2016.08.064. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Tannahill GM, et al. Succinate is an inflammatory signal that induces IL-1beta through HIF-1alpha. Nature. 2013;496:238–242. doi: 10.1038/nature11986. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Ackermann WW, Potter VR. Enzyme inhibition in relation to chemotherapy. Proc. Soc. Exp. Biol. Med. 1949;72:1–9. doi: 10.3181/00379727-72-17313. [DOI] [PubMed] [Google Scholar]
  • 75.Nemeth B, et al. Abolition of mitochondrial substrate-level phosphorylation by itaconic acid produced by LPS-induced Irg1 expression in cells of murine macrophage lineage. FASEB J. 2016;30:286–300. doi: 10.1096/fj.15-279398. [DOI] [PubMed] [Google Scholar]
  • 76.Ahn S, Jung J, Jang IA, Madsen EL, Park W. Role of glyoxylate shunt in oxidative stress response. J. Biol. Chem. 2016;291:11928–11938. doi: 10.1074/jbc.M115.708149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Palsuledesai CC, Distefano MD. Protein prenylation: enzymes, therapeutics, and biotechnology applications. ACS Chem. Biol. 2015;10:51–62. doi: 10.1021/cb500791f. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Einav S, Glenn JS. Prenylation inhibitors: a novel class of antiviral agents. J. Antimicrob. Chemother. 2003;52:883–886. doi: 10.1093/jac/dkg490. [DOI] [PubMed] [Google Scholar]
  • 79.Liu Y, et al. N (6)-methyladenosine RNA modification-mediated cellular metabolism rewiring inhibits viral replication. Science. 2019;365:1171–1176. doi: 10.1126/science.aax4468. [DOI] [PubMed] [Google Scholar]
  • 80.Alfadda AA, Sallam RM. Reactive oxygen species in health and disease. J. Biomed. Biotechnol. 2012;2012:936486. doi: 10.1155/2012/936486. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Paiva CN, Bozza MT. Are reactive oxygen species always detrimental to pathogens? Antioxid. Redox Signal. 2014;20:1000–1037. doi: 10.1089/ars.2013.5447. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Liu X, Wu XP, Zhu XL, Li T, Liu Y. IRG1 increases MHC class I level in macrophages through STAT-TAP1 axis depending on NADPH oxidase mediated reactive oxygen species. Int. Immunopharmacol. 2017;48:76–83. doi: 10.1016/j.intimp.2017.04.012. [DOI] [PubMed] [Google Scholar]
  • 83.West AP, et al. TLR signalling augments macrophage bactericidal activity through mitochondrial ROS. Nature. 2011;472:476–480. doi: 10.1038/nature09973. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Bergsbaken T, Fink SL, Cookson BT. Pyroptosis: host cell death and inflammation. Nat. Rev. Microbiol. 2009;7:99–109. doi: 10.1038/nrmicro2070. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Rossjohn J, et al. T cell antigen receptor recognition of antigen-presenting molecules. Annu. Rev. Immunol. 2015;33:169–200. doi: 10.1146/annurev-immunol-032414-112334. [DOI] [PubMed] [Google Scholar]
  • 86.Hewitt EW. The MHC class I antigen presentation pathway: strategies for viral immune evasion. Immunology. 2003;110:163–169. doi: 10.1046/j.1365-2567.2003.01738.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Ackerman AL, Cresswell P. Cellular mechanisms governing cross-presentation of exogenous antigens. Nat. Immunol. 2004;5:678–684. doi: 10.1038/ni1082. [DOI] [PubMed] [Google Scholar]
  • 88.Liu X, et al. Polymorphisms in IRG1 gene associated with immune responses to hepatitis B vaccination in a Chinese Han population and function to restrain the HBV life cycle. J. Med. Virol. 2017;89:1215–1223. doi: 10.1002/jmv.24756. [DOI] [PubMed] [Google Scholar]
  • 89.Smith J, et al. Systems analysis of immune responses in Marek’s disease virus-infected chickens identifies a gene involved in susceptibility and highlights a possible novel pathogenicity mechanism. J. Virol. 2011;85:11146–11158. doi: 10.1128/JVI.05499-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Gautam A, et al. Interleukin-10 alters effector functions of multiple genes induced by Borrelia burgdorferi in macrophages to regulate Lyme disease inflammation. Infect. Immun. 2011;79:4876–4892. doi: 10.1128/IAI.05451-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Nair S, et al. Irg1 expression in myeloid cells prevents immunopathology during M. tuberculosis infection. J. Exp. Med. 2018;215:1035–1045. doi: 10.1084/jem.20180118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Cho H, et al. Differential innate immune response programs in neuronal subtypes determine susceptibility to infection in the brain by positive-stranded RNA viruses. Nat. Med. 2013;19:458–464. doi: 10.1038/nm.3108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Ren K, et al. Suppression of IRG-1 reduces inflammatory cell infiltration and lung injury in respiratory syncytial virus infection by reducing production of reactive oxygen species. J. Virol. 2016;90:7313–7322. doi: 10.1128/JVI.00563-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.McNeal S, et al. Association of immunosuppression with DR6 expression during the development and progression of spontaneous ovarian cancer in Laying Hen model. J. Immunol. Res. 2016;2016:6729379. doi: 10.1155/2016/6729379. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Weiss JM, et al. Itaconic acid mediates crosstalk between macrophage metabolism and peritoneal tumors. J. Clin. Investig. 2018;128:3794–3805. doi: 10.1172/JCI99169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Fischer C, et al. Bisphenol A (BPA) exposure in utero leads to immunoregulatory cytokine dysregulation in the mouse mammary gland: a potential mechanism programming breast cancer risk. Horm. Cancer. 2016;7:241–251. doi: 10.1007/s12672-016-0254-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Perry VH, Teeling J. Microglia and macrophages of the central nervous system: the contribution of microglia priming and systemic inflammation to chronic neurodegeneration. Semin Immunopathol. 2013;35:601–612. doi: 10.1007/s00281-013-0382-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Duffy CM, Hofmeister JJ, Nixon JP, Butterick TA. High fat diet increases cognitive decline and neuroinflammation in a model of orexin loss. Neurobiol. Learn Mem. 2019;157:41–47. doi: 10.1016/j.nlm.2018.11.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Mor G, Cardenas I, Abrahams V, Guller S. Inflammation and pregnancy: the role of the immune system at the implantation site. Ann. N. Y. Acad. Sci. 2011;1221:80–87. doi: 10.1111/j.1749-6632.2010.05938.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Paria BC, Huet-Hudson YM, Dey SK. Blastocyst’s state of activity determines the “window” of implantation in the receptive mouse uterus. Proc. Natl Acad. Sci. USA. 1993;90:10159–10162. doi: 10.1073/pnas.90.21.10159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Salleh N, Giribabu N. Leukemia inhibitory factor: roles in embryo implantation and in nonhormonal contraception. ScientificWorldJournal. 2014;2014:201514. doi: 10.1155/2014/201514. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Matulova M, et al. Characterization of chicken spleen transcriptome after infection with Salmonella enterica serovar enteritidis. PLoS ONE. 2012;7:e48101. doi: 10.1371/journal.pone.0048101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Matulova M, et al. Chicken innate immune response to oral infection with Salmonella enterica serovar enteritidis. Vet. Res. 2013;44:37. doi: 10.1186/1297-9716-44-37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Rychlik I, Elsheimer-Matulova M, Kyrova K. Gene expression in the chicken caecum in response to infections with non-typhoid Salmonella. Vet. Res. 2014;45:119. doi: 10.1186/s13567-014-0119-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Preusse M, Tantawy MA, Klawonn F, Schughart K, Pessler F. Infection- and procedure-dependent effects on pulmonary gene expression in the early phase of influenza A virus infection in mice. BMC Microbiol. 2013;13:293. doi: 10.1186/1471-2180-13-293. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Cellular and Molecular Immunology are provided here courtesy of Nature Publishing Group

RESOURCES