You are on page 1of 7

Immunological Medicine

ISSN: (Print) 2578-5826 (Online) Journal homepage: https://www.tandfonline.com/loi/timm20

Basic mechanism of immune system activation by


mitochondria

Yukiko Iwasaki, Yusuke Takeshima & Keishi Fujio

To cite this article: Yukiko Iwasaki, Yusuke Takeshima & Keishi Fujio (2020) Basic mechanism
of immune system activation by mitochondria, Immunological Medicine, 43:4, 142-147, DOI:
10.1080/25785826.2020.1756609

To link to this article: https://doi.org/10.1080/25785826.2020.1756609

© 2020 The Author(s). Published by Informa


UK Limited, trading as Taylor & Francis
Group on behalf of the Japanese Society of
Clinical Immunology.

Published online: 12 May 2020.

Submit your article to this journal

Article views: 5887

View related articles

View Crossmark data

Citing articles: 14 View citing articles

Full Terms & Conditions of access and use can be found at


https://www.tandfonline.com/action/journalInformation?journalCode=timm20
IMMUNOLOGICAL MEDICINE
2020, VOL. 43, NO. 4, 142–147
https://doi.org/10.1080/25785826.2020.1756609

REVIEW ARTICLE

Basic mechanism of immune system activation by mitochondria


Yukiko Iwasaki, Yusuke Takeshima and Keishi Fujio
Department of Allergy and Rheumatology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan

ABSTRACT ARTICLE HISTORY


Almost 160 years after the discovery of mitochondria, they are known for their production of Received 19 March 2020
energy and are called “the powerhouse of the cell”. Recently, immune-metabolism has been Accepted 8 April 2020
revealed as a key factor controlling immune cell proliferation and differentiation. Resting
KEYWORDS
lymphocytes generate energy through oxidative phosphorylation and fatty acid oxidation,
Mitochondrial dysfunction;
whereas activated lymphocytes rapidly shift to glycolysis. Oxidative phosphorylation mtDNA; OXPHOS; mtROS;
(OXPHOS) as well as mitochondrial reactive oxygen species (mtROS) generated through the autoimmune diseases
electron transport chain (ETC) are involved in many immune cell functions. Moreover, mito-
chondria are dynamic organelles that can provide immunogenic molecules, such as mito-
chondrial DNA (mtDNA) resulting in innate immune system activation. Here, we describe the
role of mitochondria in immune system regulation, highlighting metabolism-dependent and
other immunogenic aspects.

1. Introduction There is accumulating evidence that mitochon-


dria are a hub of the immune system [6]. In this
In the Warburg effect, cancer cells shift from oxida-
tive phosphorylation towards aerobic glycolysis. review, we discuss the importance of mitochondrial
Immune-metabolism refers to the processes control- functions in autoimmune diseases from the point of
ling immune cell differentiation, proliferation and view of immunogenicity and immune-metabolism.
function. In clinical settings, insights into metabolic
regulation of autoimmune diseases have been accu- 2. Mitochondria as activators of the
mulating. Inhibition of mammalian target of rapamy- immune system
cin (mTOR), a key protein kinase regulating cellular
Mitochondria are the source of damage-associated
metabolism and cell growth, was reported to be
molecular patterns (DAMPs). Immunogenicity of
effective in the treatment of systemic lupus erythema-
mitochondria can be partly explained by the fact
tosus [1]. Mitochondria are the main energy producer
of cells through the process of oxidative phosphoryl- that they originated through endosymbiosis accom-
ation (OXPHOS). They also function as biosynthetic panied by gene transfer from the endosymbiont
centers and balance redox signaling [2]. In a genome- to the host. The hypomethylated CpG motif of
wide association study (GWAS), several single nucleo- mitochondrial DNA (mtDNA) is thought to be a
tide polymorphisms (SNPs) related to mitochondria remnant of engulfed bacteria and they are immuno-
were associated with systemic lupus erythematosus, genic. In addition to mtDNA, mitochondria produce
suggesting the importance of mitochondrial function several DAMPs, such as ATP, succinate, cardiolipin,
in SLE pathogenesis [3]. Mitochondrial dysfunctions, N-formyl peptides and transcriptional factor A
including increased production of reactive oxygen (TFAM). In concordance with impaired mitochon-
species (ROS) and decreased respiration were drial quality, these molecules are recognized by pat-
reported in untreated skeletal muscles of dermato- tern recognition receptors (PRRs) and activate the
myositis (DM) patients [4]. Interferon (IFN) b, a immune system. Nine kinds of anti-mitochondrial
type I IFN, induced ROS in human myotubes and it antibodies (AMA), M1 to M9, have been character-
reportedly contributed to mitochondrial dysfunction. ized. AMA-M2, -M4, -M8, and -M9 classes were
In Sjӧgren’s syndrome (SS), a relation between mito- described in primary biliary cholangitis (PBC) and
chondrial dysfunction and oxidative stress was postu- AMA-M1, -M3 and -M5 classes were detected in
lated in its pathogenesis [5]. SLE patients. The molecular target of M1 is known

CONTACT Yukiko Iwasaki yunyan-todai@umin.ac.jp Department of Allergy and Rheumatology, Graduate School of Medicine, The University of
Tokyo, Tokyo, Japan
ß 2020 The Author(s). Published by Informa UK Limited, trading as Taylor & Francis Group on behalf of the Japanese Society of Clinical Immunology.
This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0/), which permits
unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
IMMUNOLOGICAL MEDICINE 143

to be cardiolipin, which exists within the inner variety of cytoplasmic and nuclear components,
mitochondrial membrane [7]. including DNA and RNA. Large MPs (approxi-
mately 3000 nm) containing mitochondrial mole-
cules (mitoMPs) can be elevated in the blood and
2.1. Nlrp3 inflammasome activation by
are associated with major SLE features in SLE
mitochondrial components
patients. This indicates mitoMPs play an important
The inflammasome, the immune surveillance system role in SLE pathogenesis [13].
of cells, consists of high molecular weight protein
complexes composed of PRR-containing pyrin and a
2.3. Toll-like receptor (TLR) 9 and cGAS-STING
caspase recruitment and activation domain, an
signaling activated by mtDNA
adaptor protein apoptosis-related speck-like protein
(ASC) and the effector protease caspase-1. Assembly Toll-like receptor (TLR) 9 is one of the PRRs that
of the inflammasome complex occurs in response to recognizes unmethylated CpG DNA originating
pathogen- or damage-associated molecular patterns. from bacteria, viruses or mtDNA. mitoMPs trans-
PRRs have several subgroups, such as toll-like recep- ported to endosomes are detected by TLR9 and sub-
tors (TLRs), C-type lectin receptors (CLRs), RIG-I sequently induce the production of proinflammatory
like receptor (RLR), AIM2-like receptor (ALR) and cytokines through MyD88-NF-jB signaling or type I
nucleotide-binding oligomerization domain-like IFNs in plasmacytoid dendritic cells (pDCs). In add-
receptors (NLRs). Among the NLRs, nucleotide ition, mtROS and mtDNA induce the formation of
oligomerization domain-like (NOD-like) receptor neutrophil extracellular traps (NETs), and a TLR9
family pyrin domain 3 (NLRP3) is the most exten- antagonist was reported to prevent NETosis in
sively studied and is a multiprotein platform whose human neutrophils [14]. NETs are also important
activation leads to caspase-1-dependent secretion of for SLE pathogenesis by releasing nuclear antigens
proinflammatory cytokines, such as interleukin-1b outside of cells.
(IL-1b) and IL-18. Mitochondria play a pivotal DNA released to the cytosol is detected by cyclic
role in NLRP3 inflammasome activation. GMP-AMP synthase (cGAS) that is linked to stimu-
Mitochondrion-derived molecules, mitochondrial lator of interferon genes (STING), a DNA sensor in
ROS (mtROS), oxidized mtDNA and cardiolipin, the cytosol. It induces the expression of IFN-b and
contribute to NLRP3 inflammasome activation [8]. IFN-stimulated genes through IFN regulatory factor
Mitochondrial antiviral signaling protein (MAVS), 3 (IRF3) phosphorylation. mtDNA can also induce
which is involved in regulating the production of type I IFN production through the cGAS/STING
type I IFN, associates with NLRP3, thereby recruit- pathway [15] and oxidized mtDNA is associated
ing NLRP3 to mitochondria [9]. Recently, Pim-1, a with SLE pathogenesis through type I IFN induction
serine/threonine kinase, was revealed to regulate the in a cGAS/STING dependent manner [16]. In add-
NLRP3 inflammasome in podocytes of lupus neph- ition, STING-gain-of-function mutations induced
ritis [10]. Moreover, NLRP3 inflammasome activa- IFN-b1 in vitro [17]. The STING mutations discov-
tion plays a crucial role in the pathogenesis of ered in patients with lupus-like syndromes and
rheumatoid arthritis (RA), systemic sclerosis (SSc) autoinflammatory disease are called STING-associ-
and inflammatory bowel diseases (IBD) [11]. In ated vasculopathy with onset in infancy (SAVI).
these autoimmune diseases, mitochondrial compo-
nents might play a crucial role in their pathophysi-
3. Metabolic regulation by mitochondria in
ology through activating NLRP3 inflammasome.
immune cells
Mitochondria have fundamental roles in regulating
2.2. Microparticles containing mitochondrial
metabolic pathways through glucose oxidation as
components detected as foreign antigens
well as the biosynthesis of fatty acids, amino acids
Mitochondrial components such as mtDNA can and hormones. Tight control of mitochondrial func-
move to the cytosol through mitochondrial perme- tions and dynamics is critical for maintaining
ability transition pores (mPTP) and act as intracellu- homeostasis of the immune system. Historically,
lar antigens [12]. It remains unclear how metabolic regulation of immune cells has been
mitochondria-derived components are detected as extensively investigated in CD4 T cells and mono-
foreign antigens. Generally, cells rearrange the cyto- cytes. The regulation of metabolism in other
skeleton and package intracellular components into immune cell subsets is important for their survival,
small vesicles and secrete them into the circulation. proliferation and activation [3,18]. In addition,
These microparticles (MPs) are 100–1000 nm in mtROS predominantly produced by OXPHOS and
diameter, larger than exosomes. MPs carry a wide the electron transport chain (ETC) can regulate
144 Y. IWASAKI ET AL.

diverse environmental and pathological stimuli. regulator of multiple B cell functions, there is an
Excessive production of ROS is observed in various increased survival rate due to upregulated expres-
disease conditions including autoimmune diseases. sion of Glut1 and hexokinase 2 (Hk2). The latter
phosphorylates glucose to produce glucose-6-phos-
3.1. Activation and functional changes of phate (G6P) in the first step of glucose metabolism
immune cells by regulating metabolism [25]. Moreover, in our own transcriptomic analysis
of immune cell subsets in peripheral blood mono-
Metabolic regulation in monocytes/macrophages has nuclear cells, the OXPHOS signature in memory B
long been investigated. M1 macrophages, inflamma- cells appears to be a key player in SLE pathogenesis
tory macrophages, depend on glycolysis for the pro- (unpublished data). B cells chronically stimulated by
duction of ATP. The induction of nicotinamide B-cell activating factor, a member of the tumor
adenine dinucleotide phosphate (NADPH) synthesis necrosis factor family (BAFF), an important cyto-
is achieved through the pentose phosphate pathway kine for SLE pathogenesis, were metabolically
(PPP). ROS is produced through catabolizing reprogrammed, resulting in upregulated glycolysis
NADPH by NADPH oxidase functions, and it pre- and enhanced antibody production [26]. Moreover,
vents infection. M2 macrophages, which contribute
the survival of long-lived plasma cells relies on
to tissue repair and release the anti-inflammatory
pyruvate imported into mitochondria [27].
cytokine IL-10, reportedly depend on OXPHOS and
In activated dendritic cells, glucose uptake and
fatty acid oxidation to produce energy [19]. 
lactate production are elevated through glycolysis, as
In activated CD4þ T cells, the metabolic shifts
observed in M1 macrophage. However, metabolites
from OXPHOS to aerobic glycolysis, the PPP and
from glycolysis are utilized for fatty acids synthesis
glutaminolysis have been investigated in detail [20].
and ATP production, a process dependent on
Overexpression of glucose transporter type 1
OXPHOS [28]. Plasmacytoid dendritic cells (pDCs),
(GLUT1), the expression of which is upregulated by
the main type I IFN-producing cells, are important
T-cell receptor (TCR) stimulation under hypoxia,
for SLE pathogenesis and are known to be metabol-
causes an autoimmune phenotype in mice
ically reprogrammed by an autocrine type I IFN
[21,22].  Sanroque mice (Roquinsan/san) present a
lupus-like phenotype because of Inducible receptor-dependent pathway. Such autocrine type I
Costimulator (ICOS) overexpression. In this setting, IFN signaling is characterized by increased fatty acid
metformin, an ETC inhibitor, suppresses Th17 cell oxidation (FAO) and OXPHOS for full pDC activa-
differentiation and promotes regulatory T cell tion [29], suggesting that the inhibition of FAO and
(Treg) differentiation. It also prevents germinal cen- OXPHOS might allow the amelioration of type I
ter B cell (GCB) and plasma cell differentiation IFN-related diseases, such as SLE.
through activation of the AMP-activated protein
kinase (AMPK)-mTOR pathway [23]. Tregs are 3.2. Immune system affected by mitochondrial
necessary for the maintenance of immuno-tolerance reactive oxygen species (mtROS)
and homeostasis. The suppressive function of Tregs
in mice was shown to be impaired by the deficiency Mitochondria have a key role in oxygen metabolism
of mitochondrial complex III, 1 of the 5 protein and are the major contributor to ROS formation.
complexes in the inner mitochondrial membrane, ROS are important to many physiological processes,
the main source of ATP in a cell [24]. Mice lacking such as aging, cancer and type 2 diabetes mellitus
complex III in Tregs displayed a loss of T cell-sup- [5,30,31]. mtROS in peripheral blood mononuclear
pression capacity without altering Treg proliferation cells from SLE patients induced mitochondrial anti-
and survival. Increased DNA methylation and the viral signaling (MAVS) oligomerization resulting in
metabolites 2-hydroxyglutarate (2-HG) and succin- the induction of type I IFN [32]. Mitochondrial
ate in these Tregs inhibited the ten-eleven transloca- hyperpolarization and elevated mtROS production
tion (TET) family of DNA demethylases. have been observed in T cells from SLE patients
Mitochondrial complex III in Tregs was critical for [33]. mito TEMPO, the mitochondria-targeted
maintaining immune regulatory gene expression and superoxide dismutase mimetic with superoxide scav-
suppressive function. enging properties, was able to prevent inflamma-
Unlike T lymphocytes, in B cells both OXPHOS some-dependent IL-1b production in human
and aerobic glycolysis are upregulated during cell macrophages, suggesting the importance of mtROS
activation. The importance of metabolic regulation in macrophage activation [34]. In B cells, mtROS
of B cells was suggested by the following observa- promoted class switch recombination and sup-
tions. First, in B cells deficient for TNF receptor- pressed plasma cell differentiation by attenuating
associated factor 3 (TRAF3), which is a negative heme synthesis [35]. mtROS provides signals for
IMMUNOLOGICAL MEDICINE 145

various cell fate/state determinations and functions states as well as mtROS production. Mitochondria
as regulators of the immune system. are also a source of immunogenicity following their
loss of integrity. Mitochondrial dysfunction can acti-
vate NLRP3 inflammasomes in immune cells [36]
4. Concluding remarks
and NLRP3 activation is important for pathogenesis
Mitochondria are critical organelles that regulate the and development of autoimmune diseases (Figure 1
immune system. That is, they regulate signaling and Table 1). Metabolic regulation in each immune
pathways and cell fate in both innate and adaptive cell subset reviewed in this article is summarized in
immune cells through their control of metabolic Table 2.

Figure 1. Mitochondrial regulation on immune system. Figures were prepared using BioRender (http://www.biorender.com)
and modified from Sack [37].

Table 1. Role of mitochondrial components in immune responses and related autoimmune diseases.
Mitochondrial components Reported roles in immune responses Related autoimmune diseases
mtDNA secreted as DAMPs detected by TLR9, Inducers of NETs, NLRP3 SLE, SAVI(STING mutation)
mitoMPs, inflammasome activationa, & Activation of cGAS/
oxidized mtDNA STING pathway.
mtROS NLRP3 inflammasome activation, Inducers of NETs, MAVS SLE (especially T cells), Dermatomyositis,
oligomerization, Macrophage activation, Promotion of class Sjӧgren syndrome
switch recombination and suppression on plasma cell
differentiation.
ATP, succinate, cardiolipin, DAMPs produced by mitochondria.
TFAM
Cardiolipin NLRP3 inflammasome activation.
MAVS Recruite NLRP3 inflammasome to mitochondria.
Pim-1 Important for preserving mitochondrial morphology, related Lupus nephritis
to NLRP3 inflammasome activation.
a
NLRP3 inflammasome activation is also related to RA, SSc, and IBD.

Table 2. Metabolic regulation in immune cell subsets.


Immune cell type Reported metabolic regulation Related experimental reports
CD4þ T cell Shift from OXPHOS to aerobic glycolysis, PPP, and glutaminolysis Overexpression of Glut1 causes an
on activation. autoimmune phenotype in mice.
Th17 cell ETC is important for differentiation. Metformin, ETC inhibitor, suppresses its
differentiation in Sanroque mice.
Treg ETC complex III is critical for regulatory gene expression,
suppressive function.
B cells Both OXPHOS and aerobic glycolysis upregulated on activation. Upregulation of Glut1 and Hk2 contributes to
its survival rate.
GCB and plasma cell ETC is important for their differentiations. Metformin prevents their differentiations
through activation of AMPK/mTOR pathway.
Long-lived plasma cell Survival dependence on pyruvate imported to mitochondria.
M1 macrophage Aerobic glycolysis-dependent energy (ATP) production. NADPH
synthesis through PPP is important for their ROS production.
M2 macrophage OXPHOS and fatty acid oxidation (FAO)-dependent production
of ATP.
Dendritic cell Upregulated aerobic glycolysis on activation. Metabolites
produced through glycolysis are utilized for FAO and
ATP production.
pDC Activation is dependent on increased FAO and OXPHOS induced
by autocrine IFN signaling.
146 Y. IWASAKI ET AL.

It is notable that the precise mechanism underly- transduction, and role in pathophysiology. Physiol
ing the cross-talk between metabolic regulation and Rev. 2015;95(4):1111–1155.
ROS production in mitochondria remains unclear. [13] Mobarrez F, Fuzzi E, Gunnarsson I, et al.
Microparticles in the blood of patients with SLE:
Moreover, the approaches to analyzing the mutual size, content of mitochondria and role in circulat-
metabolic regulation among various immune cell ing immune complexes. J. Autoimmun. 2019;102:
subsets remain a challenge for future research. 142–149.
Finally, it will be necessary to investigate the contri- [14] Itagaki K, Kaczmarek E, Lee YT, et al.
butions of mitochondria to immune system regula- Mitochondrial DNA released by trauma induces
neutrophil extracellular traps. PLoS One. 2015;
tion by using purified human immune cell samples
10(3):e0120549.
to fully understand the mechanisms underlying vari- [15] West AP, Shadel GS. Mitochondrial DNA in
ous human autoimmune diseases. innate immune responses and inflammatory path-
ology. Nat Rev Immunol. 2017;17(6):363–375.
[16] Lood C, Blanco LP, Purmalek MM, et al.
Disclosure statement Neutrophil extracellular traps enriched in oxidized
mitochondrial DNA are interferogenic and con-
No potential conflict of interest was reported by
tribute to lupus-like disease. Nat Med. 2016;22:
the author(s).
146–153.
[17] Liu Y, Jesus AA, Marrero B, et al. Activated
References STING in a vascular and pulmonary syndrome. N
Engl J Med. 2014;371(6):507–518.
[1] Lai Z-W, Hanczko R, Bonilla E, et al. N -acetylcys- [18] Lightfoot YL, Blanco LP, Kaplan MJ. Metabolic
teine reduces disease activity by blocking mamma- abnormalities and oxidative stress in lupus. Curr
lian target of rapamycin in T cells from systemic Opin Rheumatol. 2017;29(5):442–449.
lupus erythematosus patients: a randomized, dou- [19] Kelly B, O’Neill LA. Metabolic reprogramming in
ble-blind, placebo-controlled trial. Arthritis macrophages and dendritic cells in innate immun-
Rheum. 2012;64(9):2937–2946. ity. Cell Res. 2015;25(7):771–784.
[2] Spinelli JB, Haigis MC. The multifaceted contribu- [20] Maciolek JA, Alex Pasternak J, Wilson HL.
tions of mitochondria to cellular metabolism. Nat Metabolism of activated T lymphocytes. Curr.
Cell Biol. 2018;20(7):745–754. Opin. Immunol. 2014;27:60–74.
[3] Takeshima Y, Iwasaki Y, Fujio K, et al. [21] Jacobs SR, Herman CE, MacIver NJ, et al. Glucose
Metabolism as a key regulator in the pathogenesis uptake is limiting in T cell activation and requires
of systemic lupus erythematosus. Semin. Arthritis CD28-mediated Akt-dependent and independent
Rheum. 2019;48(6):1142–1145. pathways. J Immunol. 2008;180(7):4476–4486.
[4] Meyer A, Laverny G, Allenbach Y, et al. IFN- [22] Cretenet G, Clerc I, Matias M, et al. Cell surface
b-induced reactive oxygen species and mitochon- Glut1 levels distinguish human CD4 and CD8 T
drial damage contribute to muscle impairment and lymphocyte subsets with distinct effector functions.
inflammation maintenance in dermatomyositis. Sci Rep. 2016;6:24129.
Acta Neuropathol. 2017;134(4):655–666. [23] Lee S-Y, Moon S-J, Kim E-K, et al. Metformin
[5] Pagano G, Castello G, Pallard o FV. Sjøgren’s syn- suppresses systemic autoimmunity in Roquin san/
drome-associated oxidative stress and mitochon- san mice through inhibiting B cell differentiation
drial dysfunction: Prospects for chemoprevention into plasma cells via regulation of AMPK/mTOR/
trials. Free Radic. Res. 2013;47(2):71–73. STAT3. JI. 2017;198(7):2661–2670.
[6] Breda C. N d S, Davanzo GG, Basso PJ, et al. [24] Weinberg SE, Singer BD, Steinert EM, et al.
Mitochondria as central hub of the immune sys- Mitochondrial complex III is essential for suppres-
tem. Redox Biol. 2019;26:101255. sive function of regulatory T cells. Nature. 2019;
[7] Becker Y, et al. Anti-mitochondrial autoantibodies 565(7740):495–499.
in systemic lupus erythematosus and their associ- [25] Liu YH, et al. Quantitative PPARc expression
ation with disease manifestations. Sci Rep. 2019;9: affects the balance between tolerance and immun-
1–16. ity. Sci. Rep. 2016;6:1–13.
[8] Liu Q, Zhang D, Hu D, et al. The role of mito- [26] Caro-Maldonado A, Wang R, Nichols AG, et al.
chondria in NLRP3 inflammasome activation. Mol Metabolic reprogramming is required for antibody
Immunol. 2018;103:115–124. production that is suppressed in anergic but exag-
[9] Park S, Juliana C, Hong S, et al. The mitochon- gerated in chronically BAFF-exposed B cells. JI.
drial antiviral protein MAVS associates with 2014;192(8):3626–3636.
NLRP3 and regulates its inflammasome activity. JI. [27] Lam WY, Becker AM, Kennerly KM, et al.
2013;191(8):4358–4366. Mitochondrial pyruvate import promotes long-
[10] Fu R, Xia Y, Li M, et al. Pim-1 as a therapeutic term survival of antibody-secreting plasma cells.
target in lupus nephritis. Arthritis Rheumatol. Immunity. 2016;45(1):60–73.
2019;71(8):1308–1318. [28] Everts B, Amiel E, Huang SC, et al. TLR-driven
[11] Shen H-H, Yang Y-X, Meng X, et al. NLRP3: a early glycolytic reprogramming via the kinases
promising therapeutic target for autoimmune dis- TBK1-IKKe supports the anabolic demands of
eases. Autoimmun Rev. 2018;17(7):694–702. dendritic cell activation. Nat Immunol. 2014;15:
[12] Bernardi P, Rasola A, Forte M, et al. The mito- 323–332.
chondrial permeability transition pore: channel [29] Wu D, Sanin DE, Everts B, et al. Type 1 interfer-
formation by F-ATP synthase, integration in signal ons induce changes in core metabolism that are
IMMUNOLOGICAL MEDICINE 147

critical for immune function. Immunity. 2016; and cytoplasmic alkalinization characterize altered
44(6):1325–1336. IL-10 signaling in patients with systemic lupus
[30] Reuter S, Gupta SC, Chaturvedi MM, et al. erythematosus. J Immunol. 2002;169(2):1092–1101.
Oxidative stress, inflammation, and cancer: how [34] Traba J, Kwarteng-Siaw M, Okoli TC, et al.
are they linked? Free Radic. Biol. Med. 2010; Fasting and refeeding differentially regulate NLRP3
49(11):1603–1616. inflammasome activation in human subjects. J Clin
[31] Elmarakby AA, Sullivan JC. Relationship between Invest. 2015;125(12):4592–4600.
oxidative stress and inflammatory cytokines in dia- [35] Jang K-J, Mano H, Aoki K, et al. Mitochondrial
betic nephropathy. Cardiovasc. Ther. 2012;30(1): function provides instructive signals for activation-
49–59. induced B-cell fates. Nat Commun. 2015;6:6750.
[32] Buskiewicz IA, Montgomery T, Yasewicz EC, et al. [36] Sadatomi D, Nakashioya K, Mamiya S, et al.
Reactive oxygen species induce virus-independent Mitochondrial function is required for extracellular
MAVS oligomerization in systemic lupus erythe- ATP-induced NLRP3 inflammasome activation. J.
matosus. Sci Signal. 2016;9(456):ra115–ra115. Biochem. 2017;161:503–512.
[33] Gergely P, Niland B, Gonchoroff N, et al. [37] Sack MN. Mitochondrial fidelity and metabolic
Persistent mitochondrial hyperpolarization, agility control immune cell fate and function. J
increased reactive oxygen intermediate production, Clin Invest. 2018;128:3651–3661.

You might also like