Abstract
Immunometabolism is a dynamic process involving the interplay of metabolism and immune response in health and diseases. Increasing evidence suggests that impaired immunometabolism contributes to infectious and inflammatory diseases. In particular, the mitochondrial enzyme aconitate decarboxylase 1 (ACOD1, best known as immunoresponsive gene 1 [IRG1]) is upregulated under various inflammatory conditions and serves as a pivotal regulator of immunometabolism involved in itaconate production, macrophage polarization, inflammasome activation, and oxidative stress. Consequently, the activation of the ACOD1 pathway is implicated in regulating the pathogenic process of sepsis and septic shock, which are part of a clinical syndrome of life-threatening organ failure caused by a dysregulated host response to pathogen infection. In this review, we discuss the latest research advances in ACOD1 expression and function, with particular attention to how the ACOD1-itaconate pathway affects infection and sterile inflammation diseases. These new insights may give us a deeper understanding of the role of immunometabolism in innate immunity.
Keywords: Aconitate decarboxylase 1 (ACOD1), Sepsis, Inflammation, Metabolism, Disease
Introduction
The innate immune system is comprised of different components, including various immune cells (e.g., macrophages, monocytes, dendritic cells [DCs], and neutrophils), which can rapidly recognize invading pathogens through pattern recognition receptors (PRRs). The innate immune response is not only the first line of defense against invading pathogens, it also triggers an inflammatory response through the production of various immune mediators, especially cytokines.[1], [2], [3] A cytokine storm is a pathological hallmark of various critical illnesses caused by sepsis and septic shock.[4], [5], [6], [7] For example, the current coronavirus disease 2019 (COVID-19), caused by the virus designated as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is associated with an excessive activation of innate immune response and a cytokine storm.[8,9]. Therefore, understanding the mechanism and modulation of inflammatory responses during infection is important for the prevention and treatment of critical diseases.
Under inflammatory conditions, immune cell activation is characterized by significant metabolic changes, including altered cell-intrinsic metabolite levels, increased aerobic glycolysis, a remodeled tricarboxylic acid (TCA) cycle, and impaired mitochondria respiration.[10], [11], [12], [13], [14], [15] These metabolic reprogramming processes are mediated by the upregulation of specific enzymes, which usually have low expression or activity under normal conditions.[10,16] The production of metabolites in the process of metabolic reprogramming can affect the function of immune cells through different mechanisms, such as signal transduction, protein activity modification, and gene expression regulation.[17], [18], [19] Excessive cytokine production further aggravates the reprogramming of the metabolic pathways of immune cells. The study of immunometabolism, namely the interaction between metabolism and immune response, has recently been greatly expanded, leading to new therapeutic targets for translational medicine.
Aconitate decarboxylase 1 (ACOD1, also known as immunoresponsive gene 1 [IRG1]) is a key regulator of immunometabolism during infection and inflammation.[20], [21], [22] It was first discovered in 1992 as a lipopolysaccharide (LPS)-inducible gene in murine macrophages.[23] Later, metabolomics profiling analyses revealed that ACOD1 is responsible for itaconate production in macrophages during an inflammatory response[24] [Figure 1]. Accumulated evidence in different disease models shows that ACOD1-dependent itaconate production can either promote or inhibit inflammation. In this review, we not only explain the expression and function of ACOD1 in innate immunity but also discuss the potential of manipulating ACOD1 as a treatment for infectious and inflammatory diseases.
Figure 1.
Metabolism of itaconate. Multiple stimuli, such as live pathogens, PAMPs, and DAMPs, trigger the activation of innate immune cells and induce the expression of ACOD1 in mitochondria. Activated immune cells (such as monocytes, macrophages, and DCs) undergo metabolic reprogramming and alter the TCA cycle to produce high levels of itaconic acid through ACOD1. ACOD1: Aconitate decarboxylase 1; DAMPs: Damage-associated molecular patterns; DCs: Dendritic cells; PAMPs: Pathogen-associated molecular patterns; SDH: Succinate dehydrogenase; TCA: Tricarboxylic acid.
The Structure and Localization of ACOD1
Human ACOD1 contains 481 amino acids and is highly conserved across species, including in chimpanzees, rats, mice, cattle, dogs, chickens, zebrafish, Xenopus species (frogs), and mussels.[25], [26], [27], [28] Crystal structure analysis showed that human ACOD1 is a homodimer, and eight active site residues (Asp93, Thr97, His103, His159, Lys207, Lys272, His277, and Tyr318) are required for its catalytic activity.[26] In Bacillus subtilis, cis-aconitic acid decarboxylase activity requires His102 (a residue of His103 in humans) instead of His277 and Tyr318, indicating that certain protein structures of ACOD1 are not conserved.[29] Cell-staining studies have shown the co-localization of ectopically overexpressed ACOD1 and MitoTracker, revealing that ACOD1 is a mitochondrial protein.[21,30] However, the mitochondrial localization signal or mitochondrial targeting sequence of ACOD1 remains unknown.
The Expression and Upregulation of ACOD1
The expression of ACOD1 is cell- and tissue-specific. Under normal conditions, the expression of ACOD1 is very low. Under stress conditions, especially inflammatory stimulation, the expression of ACOD1 is upregulated by macrophages, monocytes, and DCs in the innate immunity system. Pathogen-associated molecular patterns (PAMPs), such as LPS, lipoteichoic acid, poly I:C, and CpG-DNA, are structural components or products of microorganisms, which can strongly upregulate ACOD1 expression by combining different PRRs in macrophages. In addition, live pathogens (e.g., bacteria, viruses, fungi, protozoa, spirochetes, and Chlamydia), cytokines (e.g., interferon beta 1 [IFNB1], interferon-gamma [IFNG], tumor necrosis factor [TNF], and interleukin-1β [IL1B]), and small molecule drugs (e.g., cycloheximide, carbon monoxide releasing molecule-2, cobalt protoporphyrin IX, or chemical inducer of heme oxygenase-1 [HMOX1]) can stimulate the expression of ACOD1 in immune cells in a context-dependent manner.[22,31,32] These findings make inducible ACOD1 a biomarker of an activated innate immune response.
In addition to immune cells, the expression of ACOD1 is also upregulated in the tissue under infection, such as nervous tissue infected by West Nile virus (WNV) or Zika virus (ZIKV),[33] lung tissues infected by influenza A virus (IAV)[34] or respiratory syncytial virus (RSV) or Chlamydia pneumoniae,[35,36] splenic tissues infected by Leishmania donovani,[37] and pouch membrane tissues exposed to monosodium urate (MSU).[38] Overall, these findings highlight that the upregulation of ACOD1 may be a universal genetic event in infection and inflammation.
Although the precise mechanism of ACOD1 upregulation is poorly understood, the activation of several immune-related transcription factors contributes to ACOD1 expression.[39,40] The upregulated ACOD1 also acts as a feedback mechanism to regulate the activation of transcription factors. For example, lipid A induces Acod1 mRNA upregulation by activating two transcription factors, namely nuclear factor kappa B subunit 1 (NFKB1) and interferon regulatory factor 3 (IRF3),[41] while the increased expression of ACOD1 inhibits NFKB1 and IRF3, leading to LPS tolerance.[42] This negative feedback mechanism between ACOD1 and NFKB1 can be mediated by the deubiquitinase TNF alpha-induced protein 3 (TNFAIP3, also known as A20), which inhibits NFKB1 activation and subsequent ACOD1 expression in myeloid cells in response to LPS, TNF, or carbon monoxide (CO).[31,[42], [43], [44] Increased ACOD1 expression limits NFKB1 activation by sustaining the expression of TNFAIP3. In addition, ACOD1-mediated itaconate production leads to the expression of activating transcription factor 3 (ATF3), thereby inhibiting the translation of the NFKB inhibitor zeta (NFKBIZ) and subsequent interleukin-6 (IL6) expression.[45] Unlike NFKB, the transcriptional factor signal transducer and activator of transcription 1 (STAT1), STAT3, or CCAAT enhancer-binding protein beta (CEBPB) contributes to ACOD1 upregulation in activated macrophages caused by infection with Mycobacterium tuberculosis (Mtb) or Salmonella,[30,46] but it is unclear whether ACOD1 affects the activity of these transcriptional factors. In conclusion, the mechanism of inducible ACOD1 upregulation involves multiple transcription factors, which produce complex regulatory feedback for regulating the immune response.
The Immunological Activities of ACOD1
ACOD1 is identified as the enzyme that catalyzes the production of itaconate through the decarboxylation of cis-aconitate[24] [Figure 1]. ACOD1-dependent itaconate production is increased in activated macrophages in vitro and in vivo, which contributes to a broad range of immunological activities.[24,47,48] In addition to limiting inflammation, the activation of the ACOD1 pathway also promotes an inflammation response. Moreover, the reciprocal regulation between ACOD1 and other inflammatory mediators has been found to shape the immune response. Along with the feedback effect between ACOD1 and pro-inflammatory transcription factors (e.g., NFKB1) discussed earlier, we will now review the dual role of ACOD1 in immune regulation.
Metabolic rewiring
Succinate dehydrogenase (SDH) is the enzyme involved in the citric acid cycle and electron transport chain.[49] Itaconate acts as an endogenous inhibitor of SDH [Figure 2]. Due to the structural similarity of itaconate to succinate, itaconate competitively limits the activity of SDH during inflammation.[50] For example, ACOD1-dependent itaconate production can impair SDH activity, reduce succinate oxidation, and promote succinate accumulation in LPS-activated macrophages, finally resulting in a decrease in the oxygen consumption rate and inhibiting the production of mitochondrial reactive oxygen species (mROS).[48,51,52] In contrast, the loss of itaconate production in Acod1-deficient macrophages decreases the level of succinate, leading to increased expression of hypoxia-inducible factor 1 subunit alpha (HIF1A), IL1B, and other inflammatory cytokines in response to LPS.[51] ACOD1 and itaconate also restrict ZIKV replication in mouse neurons by inhibiting SDH activity, supporting that itaconate exerts an anti-inflammatory effect in a succinate-dependent manner.[33]
Figure 2.
Role of ACOD1-dependent itaconate production in signal transduction. ACOD1-mediated itaconate production impairs SDH activity, decreases succinate oxidation, and promotes succinate accumulation, thereby suppressing the inflammatory processes mediated by succinate, such as ROS production, HIF1A activity, and IL1B expression. Itaconate alkylates the Cyss on KEPA1 or GAPDH, leading to increased NFE2L2 activation or inhibited glycolysis, thereby limiting the expression of inflammatory genes (e.g., IL1B, IL6, TNF, and NOS2) or ROS production. In addition, itaconate-mediated ATF3 expression inhibits the NFKBIZ pathway. ACOD1: Aconitate decarboxylase 1; ATF3: Activating transcription factor 3; Cys: Cysteine residue; GAPDH: Glycolytic enzyme glyceraldehyde 3-phosphate dehydrogenase; HIF1A: Hypoxia-inducible factor 1 subunit alpha; IL6: Interleukin 6; IL1B: Interleukin-1β; KEAP1: Kelch-like ECH-associated protein 1; NFE2L2: Nuclear factor erythroid 2-like 2; NFKBIZ: NFKB inhibitor zeta; NOS2: Nitric oxide synthase 2; SDH: Succinate dehydrogenase; ROS: Reactive oxygen species; TNF: Tumor necrosis factor.
Of note, ACOD1-dependent itaconate production drives the immune paralysis of human monocytes, and β-glucan (a fungal cell wall component) reverses this effect by reducing ACOD1 expression and restoring SDH expression.[53] Accumulated succinate caused by the ACOD1-itaconate pathway favors the metabolic adaptation of bacteria Pseudomonas aeruginosa in tissue-resident macrophages and epithelial cells from infected mice.[54] These findings indicate that an appropriate level of itaconate is important for initiating an immune response.
Glyceraldehyde 3-phosphate dehydrogenase (GAPDH) is an enzyme involved in breaking down glucose to obtain energy through glycolysis.[55] The 4-octyl itaconate (4-OI)-induced alkylation of cysteine residue (Cys) 22 on the protein GAPDH inhibits its enzymatic activity, thereby downregulating aerobic glycolysis and subsequent production of IL1B, NOS2, and TNF in activated macrophages.[56] The anti-inflammatory effect of itaconate can be counteracted by over-expressing wild-type GAPDH, rather than cys-22 mutant GAPDH.[56] Moreover, the activity of GAPDH-mediated glycolysis is enhanced in Acod1-deficient macrophages following LPS exposure.[56] These findings indicate that itaconate inhibits glycolysis through the alkylation of GAPDH.
Itaconate also regulates other metabolic rewiring pathways, including prenylation, mitochondria substrate-level phosphorylation, and the glyoxylate shunt. Prenylation, a modification of proteins by isoprenoid lipids, aids viruses in anchoring to cell membranes.[57] ACOD1 induced by the vesicular stomatitis virus (VSV) and itaconate upregulation enhance the synthesis of geranylgeranyl diphosphate and the prenylation processes that follow, facilitating the entry and replication of viruses in macrophages.[58] The inhibitory effect on viral infection caused by Acod1 deficiency can be abrogated by the addition of 4-OI or dimethyl itaconate (DMI) in murine bone marrow-derived macrophages (BMDMs).[58] Mitochondrial substrate-level phosphorylation is a process by which adenosine triphosphate (ATP) is generated in the absence of oxidative phosphorylation.[59] The inhibition of Acod1 by siRNA reverses LPS-induced mitochondrial substrate-level phosphorylation impairment, showing an elevated oxygen consumption rate. Macrophages treated with itaconate or overexpressed with Acod1 lose the capacity of mitochondrial substrate-level phosphorylation to mount an immune defense, although this observation requires more evidence from in vivo research.[60] Despite affecting the metabolic pathway in host immune cells, itaconate also inhibits the activity of isocitrate lyase, a critical enzyme of glyoxylate shunt in Mtb, thereby exhibiting an antimicrobial effect by limiting bacterial growth.[24] Overall, the contribution of itaconate to inflammation-related metabolic rewiring is context-dependent.
Oxidative stress
Reactive oxygen species (ROS) plays a dual role in inflammation regulation and pathogen clearance.[61,62] LPS or IFNB1 triggers ACOD1-dependent ROS production and activates STAT1 and STAT3, thereby promoting the expression of transporter 1 ATP binding cassette subfamily B (TAP1) and proteasome 20S subunit beta 9 (PSMB9), which are transporters involved in antigen processing.[63] Similarly, increased ACOD1 in LPS-treated or LPS-tolerized macrophages promotes ROS-mediated TNFAIP3 transcription.[42,43] As a negative regulator of NFKB1 signaling, the upregulation of TNFAIP3 induced by ACOD1 inhibits the activation of NFKB1 and IRF3, thereby reducing the release of IL1B, IL6, TNF, and IFNB1.[42,43] In Acod1-overexpressing macrophages, ROS scavengers abrogate the upregulation of TAP1, PSMB9, and TNFAIP3, indicating that ROS plays an indispensable role in ACOD1-driven inflammatory gene expression[42,63] [Figure 3]. Consistent with the view that ACOD1-mediated ROS production promotes inflammation, ACOD1 favors ROS-dependent lung inflammation and lung tissue damage during RSV infection.[35] Nonetheless, whether ACOD1-mediated ROS production depends on itaconate production remains to be investigated.
Figure 3.
Role of ACOD1-dependent ROS production in signal transduction. ACOD1-dependent ROS production activates STAT1 and STAT3, thereby increasing the expression of TAP1 and PSMB9, which are transporter proteins for promoting the presentation of MHC-I on the cell surface. ACOD1-dependent ROS production also promotes the expression of TNFAIP3 by increasing the methylation of H3K4. TNFAIP3 acts as a negative regulator of NFKB1 signaling to inhibit NFKB1 and IRF3 activation, subsequently reducing the release of IL1B, IL6, TNF, and IFNB1. Increased TNFAIP3 inhibits ACOD1 transcription. ACOD1: Aconitate decarboxylase 1; ATP: Adenosine triphosphate; H3K4me3: H3K4 methylation; IFNB1: Interferon beta 1; IL6: Interleukin 6; IL1B: Interleukin-1β; IRF3: Interferon regulatory factor 3; MHC-I: Class I major histocompatibility complex; NFKB1: Nuclear factor-kappa B subunit 1; PSMB9: Proteasome 20S subunit beta 9; ROS: Reactive oxygen species; STAT1: Signal transducer and activator of transcription 1; STAT3: Signal transducer and activator of transcription 3; TAP1: Transporter 1 ATP binding cassette subfamily B; TNF: Tumor necrosis factor; TNFAIP3: TNF alpha-induced protein 3.
Immune cells also use the production of ROS to fight pathogens. When exposed to LPS, elevated ACOD1 enhances the production of ROS in the mitochondria by promoting the utilization of fatty acids during oxidative phosphorylation.[30] The bactericidal activity of ROS is reduced after the depletion of Acod1 in a zebrafish infection model.[30] Thus, ACOD1-mediated ROS production plays dual roles in modulating the immune response.
Nuclear factor erythroid 2-like 2 (NFE2L2, also known as NRF2) is the master transcription factor that activates antioxidant genes to prevent oxidative damage during inflammation.[64] Under normal circumstances, NFE2L2 is mainly degraded by a proteasome pathway mediated by kelch-like ECH-associated protein 1 (KEAP1). In contrast, cell-permeable itaconate derivatives 4-OI and DMI can inhibit the activity of KEAP1 by alkylating the cysteine residues (cys-151, cys-257, cys-288, cys-273, and cys-297) on KEAP1, thereby increasing the stability of the NFE2L2 protein. This process does not rely on itaconate-mediated SDH inhibition.[65] Consequently, ACOD1-dependent itaconate production and subsequent activation of NFE2L2 limit the production of ROS and inflammatory cytokines in LPS models[45,64,65] and in sterile inflammation models, such as those for acute kidney injury, liver ischemia–reperfusion injury, and abdominal aortic aneurysms.[66], [67], [68] However, it is possible that the activation of the NFE2L2 pathway is due to the stress response caused by ACOD1-mediated ROS production.
Macrophage polarization
Depending on the stimuli, macrophages are extremely plastic cells that can have two distinct functional phenotypes via classical M1 activation or alternative M2 activation.[69] The M1-like phenotype is characterized by an increased pro-inflammatory state, while M2-type macrophages have an anti-inflammatory phenotype.[69] The dysregulated state of macrophage M1–M2 polarization leads to inflammatory conditions or diseases by changing the production profile of pro-inflammatory or anti-inflammatory cytokines.[70] Metabolic reprogramming, including itaconate production, plays a significant role in the modulation of macrophage polarization. One study showed that under normal and hypoxic conditions, the overexpression of Acod1 in macrophages leads to the upregulation of the M1 marker nitric oxide synthase 2 (NOS2),[71] highlighting a molecular link between hypoxia and inflammation mediated by ACOD1. In contrast, the suppression of Acod1 in mouse macrophages reduces itaconate production, resulting in an increased expression of the M2 marker arginase 1 (ARG1).[72] These findings demonstrate the potential role of the activation of the ACOD1–itaconate axis in maintaining inflammation by M1 polarization.
Inflammasome inhibition
Inflammasomes are intracellular multiprotein complexes that respond to various PAMPs and damage-associated molecular patterns (DAMPs).[73,74] Activated inflammasomes trigger the cleavage of pro-caspase-1 (pro-CASP1) or pro-caspase-11 (pro-CASP11) into active CASP1 or CASP11, thereby leading to maturation and secretion of interleukin 1 cytokines (e.g., IL1B and interleukin-18 [IL18]), cleaved gasdermin D (GSDMD)-mediated pyroptosis, or the release of coagulation factor and DAMPs (e.g., high mobility group box 1 [HMGB1] and sequestosome 1 [SQSTM1]).[75], [76], [77], [78], [79], [80], [81], [82], [83], [84], [85] The NOD-like receptor (NLR) family pyrin domain containing 3 (NLRP3) inflammasome is one of the best-characterized inflammasomes, consisting of its sensor NLRP3, the adaptor apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC), and the effector zymogen (pro-CASP1).[86] The activation of NLRP3 inflammasomes requires a priming signal (e.g., toll-like receptor ligands) and the subsequent second signal (e.g., ATP, nigericin, and MSU).[87] DMI inhibits the expression of LPS-induced genes (e.g., Il1b, Il18, Casp1, and Asc) that are associated with inflammasome activation and function in BMDMs,[51] indicating that DMI inhibits inflammasome activation at the priming phase. Consistently, 4-OI shares similar properties with DMI that blocks pro-IL1B induction and IL1B secretion, resulting in an immunosuppressive phenotype of macrophages. In contrast, natural itaconate suppresses IL1B release, rather than pro-IL1B induction, suggesting that natural itaconate-mediated inhibition of inflammasome function is due to a defective second signal.[52] These different inhibitory effects between modified and unmodified itaconate may attribute to the stronger electrophilic properties of itaconate derivatives.[52]
Mechanistically, itaconate inhibits the activation of NLRP3 inflammasomes by modifying the Cys-of NLRP3 or GSDMD, and this process of protein posttranslational modification is called itaconation. Cys 548 on NLRP3 is itaconated by 4-OI, thereby blocking the interaction between NLRP3 and NIMA-related kinase 7 (NEK7), a key step in the activation process.[88] In the late stage of macrophage activation, itaconate modifies the Cys 77 on GSDMD, which reduces the sustained CASP1 activation and GSDMD cleavage, and decreases IL1B and pyroptosis.[89] Conversely, macrophages from Acod1-deficient mice exhibit depleted itaconate and increased NLRP3 inflammasome activation, while increased ACOD1 expression in HEK293T cells has the opposite effect.[88,89] Thus, both endogenous and exogenous itaconate regulates the activation of NLRP3 inflammasomes [Figure 4]. Overall, these findings establish an anti-inflammatory role for ACOD1 in blocking inflammasome activation, mainly through itaconation.
Figure 4.
Role of ACOD1 in inflammasomes. In response to signal 1 and signal 2, the sensor NLRP3, the partner NEK7, the adaptor ASC, and the effector CASP1 assemble activated NLLRP3 inflammasomes, which trigger the maturation and secretion of cytokines (e.g., IL1B and IL18) or cleaved GSDMD-mediated pyroptosis. ACOD1 induced by signal 1 promotes the production of itaconate, which modifies Cys548 on NLRP3 and Cys77 on GSDMD. This process is called itaconation. Itaconated NLRP3 fails to interact with NEK7 in response to signal 2, whereas itaconated GSDMD loses the ability to trigger pyroptosis. ACOD1: Aconitate decarboxylase 1; ASC: Apoptosis-associated speck-like protein containing a caspase recruitment domain; CASP1: Caspase 1; Cys: Cysteine residue; GSDMD: Gasdermin D; IL18: Interleukin 18; IL1B: Interleukin-1β; NEK7: NIMA-related kinase 7; NLRP3: NLR family pyrin domain containing 3.
ACOD1 in Infectious and Inflammatory Diseases
ACOD1 plays a significant role at the intersection of the host's inflammatory response to infectious and sterile threats. Infectious diseases are disorders caused by microorganisms (especially bacteria and viruses), while sterile inflammatory occurs in the absence of microorganisms. Typical sterile inflammatory diseases include peripheral arterial diseases, ischemia–reperfusion injury diseases, and neuroinflammatory diseases. These inflammatory reactions contribute to the progress of pathophysiology. In this section, we discuss the complex role of ACOD1 in immune-related pathological conditions and diseases [Figure 5].
Figure 5.
ACOD1 in immune-related diseases. A: Itaconate protects mice against experimental sepsis by reducing inflammatory cytokine production and release (e.g., IL1B, TNF, and IL6) in the early stage. The upregulation of ACOD1 also leads to immune paralysis at the late stage. B: ACOD1-induced itaconate and ROS are delivered to pathogen-containing vacuoles, thereby contributing to bacterial killing during infection. C: ACOD1-induced itaconate production triggers bacteria or virus-infected cells to undergo metabolic reprogramming, thereby favoring the survival of pathogens. D: ACOD1-related immune response in disease. ACOD1 plays a context-dependent role in inflammatory diseases. Generally, excessive inflammation leads to tissue injury and multiple diseases, whereas appropriate inflammation is beneficial for the recovery from diseases. ACOD1: Aconitate decarboxylase 1; IL6: Interleukin 6; IL1B: Interleukin-1β; ROS: Reactive oxygen species; TNF: Tumor necrosis factor.
ACOD1 in infectious diseases
Sepsis is a severe clinical syndrome characterized by a dysregulated host response to infection (e.g., from bacteria and viruses), which can lead to hypotension, tissue damage, immunocoagulation, multiple organ failure, and even death.[90], [91], [92], [93] Preclinical studies using animal models of acute endotoxemia confirmed the protective effect of ACOD1 in limiting lethal inflammation in sepsis.[24] The administration of 4-OI prolongs survival, reduces inflammatory cytokine release (e.g., IL1B, TNF, and IL6), and reduces lactate production in mice exposed to lethal endotoxemia.[56,65] In mouse models, the treatment of endotoxemia with CO and HMOX1 inducers reduces the production of TNF in liver tissue and serum by inducing the expression of ACOD1.[31] Clinically, although there is acute inflammation in the early stage of sepsis, the immunosuppression in the late stage is the main cause of patient death. Thus, ACOD1-mediated immune paralysis may lead to life-threatening secondary infections during sepsis.[42,53] In contrast, β-glucan treatment inhibits LPS-induced ACOD1 expression and restores the immune activity of macrophages[42,53] which provides a potential strategy to restore immune responsiveness in patients with sepsis.
In addition to the endotoxemia model, the role of ACOD1 in sepsis has also been studied in bacterial or viral infection models in vitro and in vivo. ACOD1 mainly exhibits bactericidal effects during acute bacterial infections. For example, Mtb infection, during the etiology of tuberculosis, promotes the expression of ACOD1 and subsequent itaconate production in mouse macrophages and DCs.[46] The loss of Acod1 in macrophages increases the intracellular replication of Mtb.[24,94,95] Consistently, Acod1-deficient mice are more susceptible to Mtb infection, leading to increased mycobacterial burden and lethal inflammation in the lungs.[94,95] This phenotype is reversed by itaconate,[94] indicating that ACOD1-mediated itaconate production can prevent Mtb infection.
Another frequent cause of severe pneumonia in humans is Legionella pneumophila infection. IFNB1 or IFNG limits the intracellular growth of L. pneumophila by inducing the expression of ACOD1 and itaconate production in vitro and in vivo.[37,96] Infection from Salmonella enterica serovar Typhimurium (S. Typhimurium) or Mycobacterium avium (M. avium) is also restricted by the induction of ACOD1 expression or itaconate production.[24,30,40,43,97] In contrast, the deletion of Acod1 in mice or zebrafish larvae leads to enhanced susceptibility to S. Typhimurium or M. avium infection. Mechanistically, enhanced tethering between mitochondria and bacteria-containing vacuoles facilitates the subsequent delivery of itaconate into the vacuoles, leading to reduced replication of S. Typhimurium or M. avium.[40,97] Alernatively, ACOD1-dependent mROS production delivers itaconate to bacteria-containing phagosomes, thereby contributing to bacterial killing during S. Typhimurium infection.[30,98] Furthermore, ACOD1 is upregulated in Staphylococcus aureus- or Escherichia coli-infected human blood cells,[99] Listeria monocytogenes-infected primary murine macrophages and splenic tissue,[21] Burkholderia pseudomallei-infected J774A.1 cells (a murine macrophage cell line),[100] and Brucella melitensis-infected BMDMs,[101] indicating a wide role for ACOD1 in the defense against bacterial infections.
Unlike in bacterial infections, ACOD1 plays a dual role in viral infections. On the one hand, the antiviral effect of ACOD1 has been observed during WNV, ZIKV, and hepatitis B virus (HBV) infections.[33,102,103] Acod1-deficient mice show higher mortality, increased brain viral load, and worsening neurological diseases after ZIKV infection, which is rescued by 4-OI treatment.[33] On the other hand, ACOD1 favors viral infection under certain conditions. For example, RSV infection induces ACOD1 expression and subsequent ROS production in A549 cells (a human alveolar epithelial cell line) and lung tissues of RSV-infected mice, leading to increased inflammatory cell infiltration, oxidative damage, and lung injury in vivo.[35] Similarly, VSV-induced ACOD1 expression and itaconate production promote geranylgeranyl diphosphate synthesis and prenylation, thereby facilitating virus entry and intracellular replication.[58] Consequently, the suppression of ACOD1 impairs viral overgrowth and protects mice from RSV and VSV infection-induced lung injury.[35,58] Further understanding the different mechanisms of ACOD1 in viral infection may help clarify the selectivity of viral immunity.
In addition to the previously mentioned bacterial and viral infections, the immune function of ACOD1 is confirmed in other pathogen infections, including Chlamydia and spirochete and protozoan infections. For example, Chlamydia pneumoniae-induced Acod1 mRNA expression in lung tissue is associated with reduced pulmonary inflammatory infiltration in an MYD88 innate immune signal transduction adaptor (MYD88)-dependent manner.[36] ACOD1 is related to the pro-inflammatory response of Borrelia burgdorferi spirochete infection, the cause of Lyme disease.[20] The upregulation of ACOD1 contributes to the susceptibility of splenic macrophages to the parasite L. donovani,[104] but limits neurotoxicity in brain macrophages (microglia) that is mediated by the parasite Toxoplasma gondii.[105] Moreover, microglia that fail to induce ACOD1 expression during lymphocytic choriomeningitis virus (LCMV) infection develop into a neuroprotective phenotype, supporting the neurotoxic effect of ACOD1.[105] Taken together, these findings confirm a context-dependent role of ACOD1 during pathogen infections.
ACOD1 in sterile inflammatory diseases
In addition to pathogen infections, inflammation can also be triggered by tissue damage and stress, leading to various diseases. The abnormal expression of ACOD1 is also related to the development of sterile inflammatory diseases. Below, we highlight some examples of how ACOD1 is implicated in those diseases.
Peripheral artery disease
Peripheral artery disease is a circulatory problem in which occlusions in arteries to organs cause tissue ischemia. Severe peripheral artery disease often results in tissue necrosis and limb amputation.[106] In peripheral artery disease, macrophages are recruited into the occluded vessels to induce a potent M1 polarization, leading to decreased angiogenesis and arteriogenesis.[107,108] The ACOD1–itaconate pathway mediates M1 polarization, leading to impaired perfusion recovery in experimental peripheral artery disease.[72] The suppression of ACOD1 by miR93 mimic or Acod1 knockout in macrophages induces an M2-like phenotype, which is beneficial for the recovery of ischemic muscle.[72] Studies of these topics are advancing our understanding of the mechanism of ACOD1-mediated macrophage polarization in peripheral artery disease.
Ischemia–reperfusion injury disease
In addition to ischemic injury, re-established blood supply may increase local inflammation and ROS production, leading to secondary injury in ischemic tissue, called ischemia–reperfusion injury.[109] Despite the upregulation of Acod1 being involved in ligands (soluble CD74 and recombinant migration inhibitory factor [MIF]) that induced necroptosis of cardiac myofibroblasts,[110] itaconate exerts an anti-inflammatory effect and reduces the myocardial infarction size in a mice model of ischemia–reperfusion injury.[51] A similar protective effect for ACOD1 and itaconate has been validated in experimental models of ischemia–reperfusion injury of the liver, kidneys, and brain.[32,66,67] It is unclear whether and to what extent different immune cells confer increased itaconate production resulting in decreased ischemia–reperfusion injury. Additional investigations are required to determine the specific effect of ACOD1 in ischemia–reperfusion injury, especially investigations that use genetic models of ACOD1 inhibition in different cells or tissues.
Neuroinflammatory disorders
Activated microglia (macrophages of the brain and spinal cord) mediate dysregulated neuroinflammatory processes, which contributes to the pathogenesis of neurological diseases.[111] In addition to promoting LCMV infection in microglia, the pathological effects of ACOD1 have also been observed in mice exposed to a high-fat diet, leading to chronic neuroinflammation and ultimately cognitive impairment.[71] However, the administration of DMI in mice with experimental autoimmune encephalomyelitis or ischemic stroke suppresses neuroinflammation and ameliorates disease severity,[32,112] indicating that ACOD1 and itaconate may have non-overlapping functions. In addition, α-synuclein increases the expression of ACOD1 in microglia, indicating an inflammatory state in Parkinson's disease.[113] These findings highlight the functional role of ACOD1 in highly specialized macrophages in tissue.
Pulmonary fibrotic diseases
A dysregulated inflammatory response in lung tissue is a crucial factor in pulmonary fibrotic diseases, including cystic fibrosis and idiopathic pulmonary fibrosis,[114,115] but the contribution of ACOD1 in these diseases are different. In the cystic fibrosis airway, ACOD1-dependent itaconate production helps bacteria to undergo metabolic reprogramming, thereby promoting their survival, lung colonization, and persistent infection in the airways.[54,116] Idiopathic pulmonary fibrosis is featured by a deposition of excessive extracellular matrix in the lung parenchyma, and this process is regulated by airway macrophages.[117,118] In a bleomycin-induced pulmonary fibrosis model, the loss of Acod1 results in increased expression of profibrotic genes (e.g., Cebpb, transforming growth factor 2 [Tgf2], and Smad family member 7 [Smad7]) in airway macrophages and persistent fibrosis in mice. The application of itaconate decreases the fibrotic activity of lung fibroblasts and ameliorates the severity of pulmonary fibrosis in vivo.[119] Thus, although itaconate shows therapeutic potential in idiopathic pulmonary fibrosis, researchers should carefully consider its application to avoid unexpected bacterial infection favored by itaconate.
Rheumatoid arthritis
Rheumatoid arthritis is an autoimmune disease that can cause joint pain and damage. ERG240 is an inhibitor of branched-chain amino acid transaminase 1 (BCAT1), which inhibits LPS-induced ACOD1 expression and subsequent itaconic production in macrophages.[120] The administration of ERG240 reduces the severity of rheumatoid arthritis and crescentic glomerulonephritis in murine models.[120] Through microarray analysis, investigators have identified highly upregulated genes, including Acod1, in synovial macrophages in mice with collagen antibody-induced arthritis or in air pouch membranes stimulated by MSU crystals.[38,121] Collectively, these findings indicate the association of ACOD1 with collagen- or crystal-induced inflammation or inflammatory arthropathies (such as rheumatoid arthritis or gout), but additional studies are needed to precisely determine the function of ACOD1 in these conditions.
Other diseases
In an animal model of psoriasis induced by imiquimod, NFKBIZ-driven skin inflammation was inhibited by DMI and increased by Acod1 depletion.[45,89] In a mouse model of abdominal aortic aneurysm induced by angiotensinogen, 4-OI suppressed the formation of abdominal aortic aneurysms in apolipoprotein E (Apoe)-deficient mice by inhibiting NFE2L2-mediated vascular inflammation, while Acod1 deficiency exerted the opposite effect.[68] In a dextran sodium sulfate (DSS)-induced ulcerative colitis mouse model, DMI inhibited the inflammatory response by decreasing the secretion of cytokines (IL1B and C-C motif chemokine ligand 2 [CCL2]) from intestinal epithelial cells, thereby reducing macrophage infiltration.[122] Researchers have found that 4-OI not only protects against urate-induced peritonitis in vivo by inhibiting NLRP3-dependent inflammasome activation[88] but also suppresses the inflammation response of peripheral blood mononuclear cells (PBMCs) from cryopyrin-associated periodic syndrome patients with NLRP3 mutations.[88,123] These studies support a beneficial effect of ACOD1-mediated itaconate in alleviating excessive inflammation in immunological disorders.
Conclusions and Perspective
Emerging evidence on inducing ACOD1 expression in immunometabolism supports it as a therapeutic approach, especially for infection and inflammatory diseases.[22] To date, the immunoregulatory role of ACOD1 has been revealed mostly in processes of innate immunity, including metabolism reprogramming, oxidative stress, macrophage polarization, inflammasome inhibition, and inflammatory gene regulation. The function of ACOD1 in adaptive immune cells (e.g., T cells and B cells) or adaptive immunity has not been well-defined, although the expression of ACOD1 in macrophages may affect antigen presentation and the function of T cells in infection or antitumor immunity.[124] Another characteristic of ACOD1’s immune function is closely related to its production of itaconate. The anti-inflammatory activity of itaconate makes it a promising therapeutic strategy to limit the pathological consequences of immune-mediated diseases in multiple preclinical models. However, the induction of ACOD1 or the use of itaconate also has detrimental effects, such as promoting virus replication, aggravating tissue damage, inducing immune paralysis, or causing cell death (e.g., ferroptosis).[35,53,125,126] Nevertheless, identifying itaconate-independent functions of ACOD1 may facilitate the development of more precise treatments to overcome the side effects of itaconate.
In addition to ACOD1, oxoglutarate dehydrogenase (OGDH) also contributes to the production of itaconate.[127] Thus, it is important to investigate the signaling, mechanism, and regulation of OGDH-dependent itaconate production in the context of inflammation and immune response. Since ACOD1 is mainly induced and expressed in myeloid cells, OGDH-dependent itaconate production may play a wide range of roles in physiological processes. Given that itaconate derivatives and endogenous itaconate have opposite immunological effects due to their distinct electrophilic properties, an appropriate form of itaconate to select should be considered in future experiments.[52] More importantly, whether itaconate is useful in clinical situations needs further verification.
Funding
This research did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors.
Conflicts of Interest
The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.
Acknowledgment
We thank Dave Primm (Department of Surgery, University of Texas Southwestern Medical Center) for his critical reading of the manuscript.
Managing Editor: Jingling Bao
Footnotes
Given his role as Editorial Board Member, Prof. Daolin Tang had no involvement in the peer-review of this article and has no access to information regarding its peer-review. Prof. Dechang Chen who is the co-editor-in-chief took the responsibility for peer-review progress and made the final decision.
References
- 1.Akira S, Takeda K. Toll-like receptor signalling. Nat Rev Immunol. 2004;4(7):499–511. doi: 10.1038/nri1391. [DOI] [PubMed] [Google Scholar]
- 2.Medzhitov R, Janeway C., Jr. Innate immunity. N Engl J Med. 2000;343(5):338–344. doi: 10.1056/NEJM200008033430506. [DOI] [PubMed] [Google Scholar]
- 3.Akira S, Uematsu S, Takeuchi O. Pathogen recognition and innate immunity. Cell. 2006;124(4):783–801. doi: 10.1016/j.cell.2006.02.015. [DOI] [PubMed] [Google Scholar]
- 4.Bosmann M, Ward PA. The inflammatory response in sepsis. Trends Immunol. 2013;34(3):129–136. doi: 10.1016/j.it.2012.09.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Kany S, Vollrath JT, Relja B. Cytokines in inflammatory disease. Int J Mol Sci. 2019;20(23):6008. doi: 10.3390/ijms20236008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Woo SR, Corrales L, Gajewski TF. Innate immune recognition of cancer. Annu Rev Immunol. 2015;33:445–474. doi: 10.1146/annurev-immunol-032414-112043. [DOI] [PubMed] [Google Scholar]
- 7.Zhang RX, Kang R, Tang DL. STING1 in sepsis: Mechanisms, functions, and implications. Chin J Traumatol. 2021;S1008-1275(21) doi: 10.1016/j.cjtee.2021.07.009. 00121-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Schultze JL, Aschenbrenner AC. COVID-19 and the human innate immune system. Cell. 2021;184(7):1671–1692. doi: 10.1016/j.cell.2021.02.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Tang D, Comish P, Kang R. The hallmarks of COVID-19 disease. PLoS Pathog. 2020;16(5) doi: 10.1371/journal.ppat.1008536. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Kelly B, O'Neill LA. Metabolic reprogramming in macrophages and dendritic cells in innate immunity. Cell Res. 2015;25(7):771–784. doi: 10.1038/cr.2015.68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Pearce EL, Pearce EJ. Metabolic pathways in immune cell activation and quiescence. Immunity. 2013;38(4):633–643. doi: 10.1016/j.immuni.2013.04.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Jha AK, Huang SC, Sergushichev A, Lampropoulou V, Ivanova Y, Loginicheva E, et al. Network integration of parallel metabolic and transcriptional data reveals metabolic modules that regulate macrophage polarization. Immunity. 2015;42(3):419–430. doi: 10.1016/j.immuni.2015.02.005. [DOI] [PubMed] [Google Scholar]
- 13.Kominsky DJ, Campbell EL, Colgan SP. Metabolic shifts in immunity and inflammation. J Immunol. 2010;184(8):4062–4068. doi: 10.4049/jimmunol.0903002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Yang L, Xie M, Yang M, Yu Y, Zhu S, Hou W, et al. PKM2 regulates the Warburg effect and promotes HMGB1 release in sepsis. Nat Commun. 2014;5:4436. doi: 10.1038/ncomms5436. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Xie M, Yu Y, Kang R, Zhu S, Yang L, Zeng L, et al. PKM2-dependent glycolysis promotes NLRP3 and AIM2 inflammasome activation. Nat Commun. 2016;7:13280. doi: 10.1038/ncomms13280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Krawczyk CM, Holowka T, Sun J, Blagih J, Amiel E, DeBerardinis RJ, et al. Toll-like receptor-induced changes in glycolytic metabolism regulate dendritic cell activation. Blood. 2010;115(23):4742–4749. doi: 10.1182/blood-2009-10-249540. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Diskin C, Ryan T, O'Neill L. Modification of proteins by metabolites in immunity. Immunity. 2021;54(1):19–31. doi: 10.1016/j.immuni.2020.09.014. [DOI] [PubMed] [Google Scholar]
- 18.Mills E, O'Neill LA. Succinate: a metabolic signal in inflammation. Trends Cell Biol. 2014;24(5):313–320. doi: 10.1016/j.tcb.2013.11.008. [DOI] [PubMed] [Google Scholar]
- 19.Li R, Zhang P, Wang Y, Tao K. Itaconate: A metabolite regulates inflammation response and oxidative stress. Oxid Med Cell Longev. 2020;2020 doi: 10.1155/2020/5404780. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Gautam A, Dixit S, Philipp MT, Singh SR, Morici LA, Kaushal D, 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(12):4876–4892. doi: 10.1128/IAI.05451-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Degrandi D, Hoffmann R, Beuter-Gunia C, Pfeffer K. The proinflammatory cytokine-induced IRG1 protein associates with mitochondria. J Interferon Cytokine Res. 2009;29(1):55–67. doi: 10.1089/jir.2008.0013. [DOI] [PubMed] [Google Scholar]
- 22.Wu R, Chen F, Wang N, Tang D, Kang R. ACOD1 in immunometabolism and disease. Cell Mol Immunol. 2020;17(8):822–833. doi: 10.1038/s41423-020-0489-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.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(5):263–270. doi: 10.1007/BF00172150. [DOI] [PubMed] [Google Scholar]
- 24.Michelucci A, Cordes T, Ghelfi J, Pailot A, Reiling N, Goldmann O, et al. Immune-responsive gene 1 protein links metabolism to immunity by catalyzing itaconic acid production. Proc Natl Acad Sci USA. 2013;110(19):7820–7825. doi: 10.1073/pnas.1218599110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Xiao W, Wang L, Xiao R, Wu M, Tan J, He Y. Expression profile of human immune-responsive gene 1 and generation and characterization of polyclonal antiserum. Mol Cell Biochem. 2011;353(1–2):177–187. doi: 10.1007/s11010-011-0784-7. [DOI] [PubMed] [Google Scholar]
- 26.Chen F, Lukat P, Iqbal AA, Saile K, Kaever V, van den Heuvel J, 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(41):20644–20654. doi: 10.1073/pnas.1908770116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Schmidt G, Richter K. Expression pattern of XIRG, a marker for non-neural ectoderm. Dev Genes Evol. 2000;210(11):575–578. doi: 10.1007/s004270000095. [DOI] [PubMed] [Google Scholar]
- 28.Sendra M, Saco A, Rey-Campos M, Novoa B, Figueras A. Immune-responsive gene 1 (IRG1) and dimethyl itaconate are involved in the mussel immune response. Fish Shellfish Immunol. 2020;106:645–655. doi: 10.1016/j.fsi.2020.07.034. [DOI] [PubMed] [Google Scholar]
- 29.Chun HL, Lee SY, Lee SH, Lee CS, Park HH. Enzymatic reaction mechanism of cis-aconitate decarboxylase based on the crystal structure of IRG1 from Bacillus subtilis. Sci Rep. 2020;10(1):11305. doi: 10.1038/s41598-020-68419-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Hall CJ, Boyle RH, Astin JW, Flores MV, Oehlers SH, Sanderson LE, et al. Immunoresponsive gene 1 augments bactericidal activity of macrophage-lineage cells by regulating β-oxidation-dependent mitochondrial ROS production. Cell Metab. 2013;18(2):265–278. doi: 10.1016/j.cmet.2013.06.018. [DOI] [PubMed] [Google Scholar]
- 31.Jamal Uddin M, Joe Y, Kim SK, Oh Jeong S, Ryter SW, Pae HO, et al. IRG1 induced by heme oxygenase-1/carbon monoxide inhibits LPS-mediated sepsis and pro-inflammatory cytokine production. Cell Mol Immunol. 2016;13(2):170–179. doi: 10.1038/cmi.2015.02. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Kuo PC, Weng WT, Scofield BA, Furnas D, Paraiso HC, Yu IC, et al. Immunoresponsive gene 1 modulates the severity of brain injury in cerebral ischaemia. Brain Commun. 2021;3(3):fcab187. doi: 10.1093/braincomms/fcab187. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Daniels BP, Kofman SB, Smith JR, Norris GT, Snyder AG, Kolb JP, et al. The nucleotide sensor ZBP1 and kinase RIPK3 induce the enzyme IRG1 to promote an antiviral metabolic state in neurons. Immunity. 2019;50(1):64–76. doi: 10.1016/j.immuni.2018.11.017. .e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.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]
- 35.Ren K, Lv Y, Zhuo Y, Chen C, Shi H, Guo L, 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(16):7313–7322. doi: 10.1128/JVI.00563-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Rodríguez N, Mages J, Dietrich H, Wantia N, Wagner H, Lang R, et al. MyD88-dependent changes in the pulmonary transcriptome after infection with Chlamydia pneumoniae. Physiol Genomics. 2007;30(2):134–145. doi: 10.1152/physiolgenomics.00011.2007. [DOI] [PubMed] [Google Scholar]
- 37.Naujoks J, Tabeling C, Dill BD, Hoffmann C, Brown AS, Kunze M, et al. IFNs modify the proteome of Legionella-containing vacuoles and restrict infection via IRG1-derived itaconic acid. PLoS Pathog. 2016;12(2) doi: 10.1371/journal.ppat.1005408. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Pessler F, Mayer CT, Jung SM, Behrens EM, Dai L, Menetski JP, et al. Identification of novel monosodium urate crystal regulated mRNAs by transcript profiling of dissected murine air pouch membranes. Arthritis Res Ther. 2008;10(3):R64. doi: 10.1186/ar2435. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Tallam A, Perumal TM, Antony PM, Jäger C, Fritz JV, Vallar L, 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(2) doi: 10.1371/journal.pone.0149050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Gidon A, Louet C, Røst LM, Bruheim P, Flo TH. The tumor necrosis factor alpha and interleukin 6 auto-paracrine signaling loop controls Mycobacterium avium infection via induction of IRF1/IRG1 in human primary macrophages. mBio. 2021;12(5) doi: 10.1128/mBio.02121-21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Kawai T, Takeuchi O, Fujita T, Inoue J, Mühlradt PF, Sato S, 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(10):5887–5894. doi: 10.4049/jimmunol.167.10.5887. [DOI] [PubMed] [Google Scholar]
- 42.Li Y, Zhang P, Wang C, Han C, Meng J, Liu X, 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(23):16225–16234. doi: 10.1074/jbc.M113.454538. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Zhu X, Guo Y, Liu Z, Yang J, Tang H, Wang Y. Itaconic acid exerts anti-inflammatory and antibacterial effects via promoting pentose phosphate pathway to produce ROS. Sci Rep. 2021;11(1):18173. doi: 10.1038/s41598-021-97352-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Van Quickelberghe E, Martens A, Goeminne L, Clement L, van Loo G, Gevaert K. Identification of immune-responsive gene 1 (IRG1) as a target of A20. J Proteome Res. 2018;17(6):2182–2191. doi: 10.1021/acs.jproteome.8b00139. [DOI] [PubMed] [Google Scholar]
- 45.Bambouskova M, Gorvel L, Lampropoulou V, Sergushichev A, Loginicheva E, Johnson K, et al. Electrophilic properties of itaconate and derivatives regulate the IκBζ-ATF3 inflammatory axis. Nature. 2018;556(7702):501–504. doi: 10.1038/s41586-018-0052-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Shi S, Blumenthal A, Hickey CM, Gandotra S, Levy D, Ehrt S. 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(5):3318–3328. doi: 10.4049/jimmunol.175.5.3318. [DOI] [PubMed] [Google Scholar]
- 47.Strelko CL, Lu W, Dufort FJ, Seyfried TN, Chiles TC, Rabinowitz JD, et al. Itaconic acid is a mammalian metabolite induced during macrophage activation. J Am Chem Soc. 2011;133(41):16386–16389. doi: 10.1021/ja2070889. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Cordes T, Wallace M, Michelucci A, Divakaruni AS, Sapcariu SC, Sousa C, et al. Immunoresponsive gene 1 and itaconate inhibit succinate dehydrogenase to modulate intracellular succinate levels. J Biol Chem. 2016;291(27):14274–14284. doi: 10.1074/jbc.M115.685792. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Rustin P, Munnich A, Rötig A. Succinate dehydrogenase and human diseases: New insights into a well-known enzyme. Eur J Hum Genet. 2002;10(5):289–291. doi: 10.1038/sj.ejhg.5200793. [DOI] [PubMed] [Google Scholar]
- 50.Tannahill GM, Curtis AM, Adamik J, Palsson-McDermott EM, McGettrick AF, Goel G, et al. Succinate is an inflammatory signal that induces IL-1β through HIF-1α. Nature. 2013;496(7444):238–242. doi: 10.1038/nature11986. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Lampropoulou V, Sergushichev A, Bambouskova M, Nair S, Vincent EE, Loginicheva E, et al. Itaconate links inhibition of succinate dehydrogenase with macrophage metabolic remodeling and regulation of inflammation. Cell Metab. 2016;24(1):158–166. doi: 10.1016/j.cmet.2016.06.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Swain A, Bambouskova M, Kim H, Andhey PS, Duncan D, Auclair K, et al. Comparative evaluation of itaconate and its derivatives reveals divergent inflammasome and type I interferon regulation in macrophages. Nat Metab. 2020;2(7):594–602. doi: 10.1038/s42255-020-0210-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Domínguez-Andrés J, Novakovic B, Li Y, Scicluna BP, Gresnigt MS, Arts R, et al. The itaconate pathway is a central regulatory node linking innate immune tolerance and trained immunity. Cell Metab. 2019;29(1):211–220. doi: 10.1016/j.cmet.2018.09.003. .e5. [DOI] [PubMed] [Google Scholar]
- 54.Riquelme SA, Lozano C, Moustafa AM, Liimatta K, Tomlinson KL, Britto C, et al. CFTR-PTEN-dependent mitochondrial metabolic dysfunction promotes Pseudomonas aeruginosa airway infection. Sci Transl Med. 2019;11(499):eaav4634. doi: 10.1126/scitranslmed.aav4634. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Tristan C, Shahani N, Sedlak TW, Sawa A. The diverse functions of GAPDH: Views from different subcellular compartments. Cell Signal. 2011;23(2):317–323. doi: 10.1016/j.cellsig.2010.08.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Liao ST, Han C, Xu DQ, Fu XW, Wang JS, Kong LY. 4-Octyl itaconate inhibits aerobic glycolysis by targeting GAPDH to exert anti-inflammatory effects. Nat Commun. 2019;10(1):5091. doi: 10.1038/s41467-019-13078-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Zhang FL, Casey PJ. Protein prenylation: Molecular mechanisms and functional consequences. Annu Rev Biochem. 1996;65:241–269. doi: 10.1146/annurev.bi.65.070196.001325. [DOI] [PubMed] [Google Scholar]
- 58.Lin J, Ren J, Gao DS, Dai Y, Yu L. The emerging application of itaconate: Promising molecular targets and therapeutic opportunities. Front Chem. 2021;9 doi: 10.3389/fchem.2021.669308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Bui D, Ravasz D, Chinopoulos C. The effect of 2-ketobutyrate on mitochondrial substrate-level phosphorylation. Neurochem Res. 2019;44(10):2301–2306. doi: 10.1007/s11064-019-02759-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Németh B, Doczi J, Csete D, Kacso G, Ravasz D, Adams D, 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(1):286–300. doi: 10.1096/fj.15-279398. [DOI] [PubMed] [Google Scholar]
- 61.Forrester SJ, Kikuchi DS, Hernandes MS, Xu Q, Griendling KK. Reactive oxygen species in metabolic and inflammatory signaling. Circ Res. 2018;122(6):877–902. doi: 10.1161/CIRCRESAHA.117.311401. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Alfadda AA, Sallam RM. Reactive oxygen species in health and disease. J Biomed Biotechnol. 2012;2012 doi: 10.1155/2012/936486. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.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]
- 64.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(2):585–597. doi: 10.1016/j.bbadis.2016.11.005. [DOI] [PubMed] [Google Scholar]
- 65.Mills EL, Ryan DG, Prag HA, Dikovskaya D, Menon D, Zaslona Z, et al. Itaconate is an anti-inflammatory metabolite that activates Nrf2 via alkylation of KEAP1. Nature. 2018;556(7699):113–117. doi: 10.1038/nature25986. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Zhu D, Zhao Y, Luo Y, Qian X, Zhang Z, Jiang G, et al. Irg1-itaconate axis protects against acute kidney injury via activation of Nrf2. Am J Transl Res. 2021;13(3):1155–1169. [PMC free article] [PubMed] [Google Scholar]
- 67.Yi Z, Deng M, Scott MJ, Fu G, Loughran PA, Lei Z, et al. Immune-responsive gene 1/itaconate activates nuclear factor erythroid 2-related factor 2 in hepatocytes to protect against liver ischemia-reperfusion injury. Hepatology. 2020;72(4):1394–1411. doi: 10.1002/hep.31147. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Song H, Xu T, Feng X, Lai Y, Yang Y, Zheng H, et al. Itaconate prevents abdominal aortic aneurysm formation through inhibiting inflammation via activation of Nrf2. EBioMedicine. 2020;57 doi: 10.1016/j.ebiom.2020.102832. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Sica A, Mantovani A. Macrophage plasticity and polarization: In vivo veritas. J Clin Invest. 2012;122(3):787–795. doi: 10.1172/JCI59643. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Murray PJ. Macrophage polarization. Annu Rev Physiol. 2017;79:541–566. doi: 10.1146/annurev-physiol-022516-034339. [DOI] [PubMed] [Google Scholar]
- 71.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]
- 72.Ganta VC, Choi MH, Kutateladze A, Fox TE, Farber CR, Annex BH. A microRNA93-interferon regulatory factor-9-immunoresponsive gene-1-itaconic acid pathway modulates M2-like macrophage polarization to revascularize ischemic muscle. Circulation. 2017;135(24):2403–2425. doi: 10.1161/CIRCULATIONAHA.116.025490. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Tang D, Kang R, Coyne CB, Zeh HJ, Lotze MT. PAMPs and DAMPs: Signal 0s that spur autophagy and immunity. Immunol Rev. 2012;249(1):158–175. doi: 10.1111/j.1600-065X.2012.01146.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Guo H, Callaway JB, Ting JP. Inflammasomes: Mechanism of action, role in disease, and therapeutics. Nat Med. 2015;21(7):677–687. doi: 10.1038/nm.3893. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Lamkanfi M, Dixit VM. Mechanisms and functions of inflammasomes. Cell. 2014;157(5):1013–1022. doi: 10.1016/j.cell.2014.04.007. [DOI] [PubMed] [Google Scholar]
- 76.Wu R, Wang N, Comish PB, Tang D, Kang R. Inflammasome-dependent coagulation activation in sepsis. Front Immunol. 2021;12 doi: 10.3389/fimmu.2021.641750. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Zhang H, Zeng L, Xie M, Liu J, Zhou B, Wu R, et al. TMEM173 drives lethal coagulation in sepsis. Cell Host Microbe. 2020;27(4):556–570. doi: 10.1016/j.chom.2020.02.004. .e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Wu C, Lu W, Zhang Y, Zhang G, Shi X, Hisada Y, et al. Inflammasome activation triggers blood clotting and host death through pyroptosis. Immunity. 2019;50(6):1401–1411. doi: 10.1016/j.immuni.2019.04.003. .e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Yang X, Cheng X, Tang Y, Qiu X, Wang Y, Kang H, et al. Bacterial endotoxin activates the coagulation cascade through gasdermin D-dependent phosphatidylserine exposure. Immunity. 2019;51(6):983–996. doi: 10.1016/j.immuni.2019.11.005. .e6. [DOI] [PubMed] [Google Scholar]
- 80.Zhou B, Liu J, Zeng L, Zhu S, Wang H, Billiar TR, et al. Extracellular SQSTM1 mediates bacterial septic death in mice through insulin receptor signalling. Nat Microbiol. 2020;5(12):1576–1587. doi: 10.1038/s41564-020-00795-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Vande Walle L, Kanneganti TD, Lamkanfi M. HMGB1 release by inflammasomes. Virulence. 2011;2(2):162–165. doi: 10.4161/viru.2.2.15480. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Kang R, Zeng L, Zhu S, Xie Y, Liu J, Wen Q, et al. Lipid peroxidation drives gasdermin D-mediated pyroptosis in lethal polymicrobial sepsis. Cell Host Microbe. 2018;24(1):97–108. doi: 10.1016/j.chom.2018.05.009. .e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Zhang H, Zeng L, Xie M, Liu J, Zhou B, Wu R, et al. TMEM173 drives lethal coagulation in sepsis. Cell Host Microbe. 2020;27(4):556–570. doi: 10.1016/j.chom.2020.02.004. .e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Zou B, Liu J, Klionsky DJ, Tang D, Kang R. Extracellular SQSTM1 as an inflammatory mediator. Autophagy. 2020;16(12):2313–2315. doi: 10.1080/15548627.2020.1843253. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Kang R, Chen R, Zhang Q, Hou W, Wu S, Cao L, et al. HMGB1 in health and disease. Mol Aspects Med. 2014;40:1–116. doi: 10.1016/j.mam.2014.05.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.He Y, Hara H, Núñez G. Mechanism and regulation of NLRP3 inflammasome activation. Trends Biochem Sci. 2016;41(12):1012–1021. doi: 10.1016/j.tibs.2016.09.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Bauernfeind FG, Horvath G, Stutz A, Alnemri ES, MacDonald K, Speert D, et al. Cutting edge: NF-kappaB activating pattern recognition and cytokine receptors license NLRP3 inflammasome activation by regulating NLRP3 expression. J Immunol. 2009;183(2):787–791. doi: 10.4049/jimmunol.0901363. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Hooftman A, Angiari S, Hester S, Corcoran SE, Runtsch MC, Ling C, et al. The immunomodulatory metabolite itaconate modifies NLRP3 and inhibits inflammasome activation. Cell Metab. 2020;32(3):468–478. doi: 10.1016/j.cmet.2020.07.016. .e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Bambouskova M, Potuckova L, Paulenda T, Kerndl M, Mogilenko DA, Lizotte K, et al. Itaconate confers tolerance to late NLRP3 inflammasome activation. Cell Rep. 2021;34(10) doi: 10.1016/j.celrep.2021.108756. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Singer M, Deutschman CS, Seymour CW, Shankar-Hari M, Annane D, Bauer M, et al. The third international consensus definitions for sepsis and septic shock (sepsis-3) JAMA. 2016;315(8):801–810. doi: 10.1001/jama.2016.0287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Wang N, Wu R, Comish PB, Kang R, Tang D. Pharmacological modulation of BET family in sepsis. Front Pharmacol. 2021;12 doi: 10.3389/fphar.2021.642294. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Tang D, Wang H, Billiar TR, Kroemer G, Kang R. Emerging mechanisms of immunocoagulation in sepsis and septic shock. Trends Immunol. 2021;42(6):508–522. doi: 10.1016/j.it.2021.04.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Hotchkiss RS, Moldawer LL, Opal SM, Reinhart K, Turnbull IR, Vincent JL. Sepsis and septic shock. Nat Rev Dis Primers. 2016;2:16045. doi: 10.1038/nrdp.2016.45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Nair S, Huynh JP, Lampropoulou V, Loginicheva E, Esaulova E, Gounder AP, et al. Irg1 expression in myeloid cells prevents immunopathology during M. tuberculosis infection. J Exp Med. 2018;215(4):1035–1045. doi: 10.1084/jem.20180118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.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(3):628–638. doi: 10.1189/jlb.0905520. [DOI] [PubMed] [Google Scholar]
- 96.Price JV, Russo D, Ji DX, Chavez RA, DiPeso L, Lee AY, et al. IRG1 and inducible nitric oxide synthase act redundantly with other interferon-gamma-induced factors to restrict intracellular replication of Legionella pneumophila. mBio. 2019;10(6) doi: 10.1128/mBio.02629-19. e02629–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Chen M, Sun H, Boot M, Shao L, Chang SJ, Wang W, et al. Itaconate is an effector of a Rab GTPase cell-autonomous host defense pathway against Salmonella. Science. 2020;369(6502):450–455. doi: 10.1126/science.aaz1333. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.West AP, Brodsky IE, Rahner C, Woo DK, Erdjument-Bromage H, Tempst P, et al. TLR signalling augments macrophage bactericidal activity through mitochondrial ROS. Nature. 2011;472(7344):476–480. doi: 10.1038/nature09973. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Dix A, Hünniger K, Weber M, Guthke R, Kurzai O, Linde J. 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]
- 100.Tangsudjai S, Pudla M, Limposuwan K, Woods DE, Sirisinha S, Utaisincharoen P. 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(5):282–290. doi: 10.1111/j.1348-0421.2010.00205.x. [DOI] [PubMed] [Google Scholar]
- 101.Lacey CA, Ponzilacqua-Silva B, Chambers CA, Dadelahi AS, Skyberg JA. MyD88-dependent glucose restriction and itaconate production control Brucella infection. Infect Immun. 2021;89(10) doi: 10.1128/IAI.00156-21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Cho H, Proll SC, Szretter KJ, Katze MG, Gale M, Jr, Diamond MS. Differential innate immune response programs in neuronal subtypes determine susceptibility to infection in the brain by positive-stranded RNA viruses. Nat Med. 2013;19(4):458–464. doi: 10.1038/nm.3108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Liu X, Zhang L, Wu XP, Zhu XL, Pan LP, Li T, 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(7):1215–1223. doi: 10.1002/jmv.24756. [DOI] [PubMed] [Google Scholar]
- 104.Kong F, Saldarriaga OA, Spratt H, Osorio EY, Travi BL, Luxon BA, 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(1) doi: 10.1371/journal.ppat.1006165. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Li H, Gang Z, Yuling H, Luokun X, Jie X, Hao L, et al. Different neurotropic pathogens elicit neurotoxic CCR9- or neurosupportive CXCR3-expressing microglia. J Immunol. 2006;177(6):3644–3656. doi: 10.4049/jimmunol.177.6.3644. [DOI] [PubMed] [Google Scholar]
- 106.Hardman RL, Jazaeri O, Yi J, Smith M, Gupta R. Overview of classification systems in peripheral artery disease. Semin Intervent Radiol. 2014;31(4):378–388. doi: 10.1055/s-0034-1393976. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Seaman SA, Cao Y, Campbell CA, Peirce SM. Macrophage recruitment and polarization during collateral vessel remodeling in murine adipose tissue. Microcirculation. 2016;23(1):75–87. doi: 10.1111/micc.12261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Mantovani A, Biswas SK, Galdiero MR, Sica A, Locati M. Macrophage plasticity and polarization in tissue repair and remodelling. J Pathol. 2013;229(2):176–185. doi: 10.1002/path.4133. [DOI] [PubMed] [Google Scholar]
- 109.Wu MY, Yiang GT, Liao WT, Tsai AP, Cheng YL, Cheng PW, et al. Current mechanistic concepts in ischemia and reperfusion injury. Cell Physiol Biochem. 2018;46(4):1650–1667. doi: 10.1159/000489241. [DOI] [PubMed] [Google Scholar]
- 110.Soppert J, Kraemer S, Beckers C, Averdunk L, Möllmann J, Denecke B, et al. Soluble CD74 reroutes MIF/CXCR4/AKT-mediated survival of cardiac myofibroblasts to necroptosis. J Am Heart Assoc. 2018;7(17) doi: 10.1161/JAHA.118.009384. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.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(5):601–612. doi: 10.1007/s00281-013-0382-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Kuo PC, Weng WT, Scofield BA, Paraiso HC, Brown DA, Wang PY, et al. Dimethyl itaconate, an itaconate derivative, exhibits immunomodulatory effects on neuroinflammation in experimental autoimmune encephalomyelitis. J Neuroinflammat. 2020;17(1):138. doi: 10.1186/s12974-020-01768-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Sarkar S, Dammer EB, Malovic E, Olsen AL, Raza SA, Gao T, et al. Molecular signatures of neuroinflammation induced by αsynuclein aggregates in microglial cells. Front Immunol. 2020;11:33. doi: 10.3389/fimmu.2020.00033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Cantin AM, Hartl D, Konstan MW, Chmiel JF. Inflammation in cystic fibrosis lung disease: Pathogenesis and therapy. J Cyst Fibros. 2015;14(4):419–430. doi: 10.1016/j.jcf.2015.03.003. [DOI] [PubMed] [Google Scholar]
- 115.Bringardner BD, Baran CP, Eubank TD, Marsh CB. The role of inflammation in the pathogenesis of idiopathic pulmonary fibrosis. Antioxid Redox Signal. 2008;10(2):287–301. doi: 10.1089/ars.2007.1897. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Tomlinson KL, Lung T, Dach F, Annavajhala MK, Gabryszewski SJ, Groves RA, et al. Staphylococcus aureus induces an itaconate-dominated immunometabolic response that drives biofilm formation. Nat Commun. 2021;12(1):1399. doi: 10.1038/s41467-021-21718-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Martinez FJ, Collard HR, Pardo A, Raghu G, Richeldi L, Selman M, et al. Idiopathic pulmonary fibrosis. Nat Rev Dis Primers. 2017;3:17074. doi: 10.1038/nrdp.2017.74. [DOI] [PubMed] [Google Scholar]
- 118.Wynn TA, Vannella KM. Macrophages in tissue repair, regeneration, and fibrosis. Immunity. 2016;44(3):450–462. doi: 10.1016/j.immuni.2016.02.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Ogger PP, Albers GJ, Hewitt RJ, O'Sullivan BJ, Powell JE, et al. Itaconate controls the severity of pulmonary fibrosis. Sci Immunol. 2020;5(52):eabc1884. doi: 10.1126/sciimmunol.abc1884. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Papathanassiu AE, Ko JH, Imprialou M, Bagnati M, Srivastava PK, Vu HA, 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]
- 121.Saeki N, Imai Y. Reprogramming of synovial macrophage metabolism by synovial fibroblasts under inflammatory conditions. Cell Commun Signal. 2020;18(1):188. doi: 10.1186/s12964-020-00678-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Wang Q, Li XL, Mei Y, Ye JC, Fan W, Cheng GH, et al. The anti-inflammatory drug dimethyl itaconate protects against colitis-associated colorectal cancer. J Mol Med (Berl) 2020;98(10):1457–1466. doi: 10.1007/s00109-020-01963-2. [DOI] [PubMed] [Google Scholar]
- 123.Hoffman HM, Mueller JL, Broide DH, Wanderer AA, Kolodner RD. Mutation of a new gene encoding a putative pyrin-like protein causes familial cold autoinflammatory syndrome and Muckle-Wells syndrome. Nat Genet. 2001;29(3):301–305. doi: 10.1038/ng756. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Liu CP, Zhang X, Tan QL, Xu WX, Zhou CY, Luo M, et al. NF-κB pathways are involved in M1 polarization of RAW 264.7 macrophage by polyporus polysaccharide in the tumor microenvironment. PLoS One. 2017;12(11) doi: 10.1371/journal.pone.0188317. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Qu C, Dai E, Lai T, Cao G, Liu J, Kang R, et al. Itaconic acid induces ferroptosis by activating ferritinophagy. Biochem Biophys Res Commun. 2021;583:56–62. doi: 10.1016/j.bbrc.2021.10.054. [DOI] [PubMed] [Google Scholar]
- 126.Chen X, Li J, Kang R, Klionsky DJ, Tang D. Ferroptosis: Machinery and regulation. Autophagy. 2021;17(9):2054–2081. doi: 10.1080/15548627.2020.1810918. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Liu Y, You Y, Lu Z, Yang J, Li P, Liu L, et al. N (6)-methyladenosine RNA modification-mediated cellular metabolism rewiring inhibits viral replication. Science. 2019;365(6458):1171–1176. doi: 10.1126/science.aax4468. [DOI] [PubMed] [Google Scholar]





