ArticlePDF AvailableLiterature Review

How the Innate Immune DNA Sensing cGAS–STING Pathway Is Involved in Autophagy

Authors:

Abstract and Figures

The cGAS–STING pathway is a key component of the innate immune system and exerts crucial roles in the detection of cytosolic DNA and invading pathogens. Accumulating evidence suggests that the intrinsic cGAS–STING pathway not only facilitates the production of type I interferons (IFN-I) and inflammatory responses but also triggers autophagy. Autophagy is a homeostatic process that exerts multiple effects on innate immunity. However, systematic evidence linking the cGAS–STING pathway and autophagy is still lacking. Therefore, one goal of this review is to summarize the known mechanisms of autophagy induced by the cGAS–STING pathway and their consequences. The cGAS–STING pathway can trigger canonical autophagy through liquid-phase separation of the cGAS–DNA complex, interaction of cGAS and Beclin-1, and STING-triggered ER stress–mTOR signaling. Furthermore, both cGAS and STING can induce non-canonical autophagy via LC3-interacting regions and binding with LC3. Subsequently, autophagy induced by the cGAS–STING pathway plays crucial roles in balancing innate immune responses, maintaining intracellular environmental homeostasis, alleviating liver injury, and limiting tumor growth and transformation.
Content may be subject to copyright.
International Journal of
Molecular Sciences
Review
How the Innate Immune DNA Sensing cGAS–STING Pathway
Is Involved in Autophagy
Wanglong Zheng 1,2,3,4,† , Nengwen Xia 1,2,3,4,† , Jiajia Zhang 1,2,3,4, Nanhua Chen 1,2,3,4 ,
François Meurens 5,6 , Zongping Liu 1,2,3,4,* and Jianzhong Zhu 1,2,3,4,*


Citation: Zheng, W.; Xia, N.; Zhang,
J.; Chen, N.; Meurens, F.; Liu, Z.; Zhu,
J. How the Innate Immune DNA
Sensing cGAS–STING Pathway Is
Involved in Autophagy. Int. J. Mol.
Sci. 2021,22, 13232. https://doi.org/
10.3390/ijms222413232
Academic Editor: James L. Stafford
Received: 28 October 2021
Accepted: 7 December 2021
Published: 8 December 2021
Publisher’s Note: MDPI stays neutral
with regard to jurisdictional claims in
published maps and institutional affil-
iations.
Copyright: © 2021 by the authors.
Licensee MDPI, Basel, Switzerland.
This article is an open access article
distributed under the terms and
conditions of the Creative Commons
Attribution (CC BY) license (https://
creativecommons.org/licenses/by/
4.0/).
1Comparative Medicine Research Institute, Yangzhou University, Yangzhou 225009, China;
wanglongzheng@yzu.edu.cn (W.Z.); 13089071533@163.com (N.X.); zjj1608291303@163.com (J.Z.);
hnchen@yzu.edu.cn (N.C.)
2College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
3Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou 225009, China
4Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and
Zoonoses, Yangzhou 225009, China
5BIOEPAR, INRAE, Oniris, 44307 Nantes, France; francois.meurens@inra.fr
6Department of Veterinary Microbiology and Immunology, Western College of Veterinary Medicine,
University of Saskatchewan, Saskatoon, SK S7N 5E2, Canada
*Correspondence: liuzongping@yzu.edu.cn (Z.L.); jzzhu@yzu.edu.cn (J.Z.)
These authors contributed equally to this work.
Abstract:
The cGAS–STING pathway is a key component of the innate immune system and exerts
crucial roles in the detection of cytosolic DNA and invading pathogens. Accumulating evidence
suggests that the intrinsic cGAS–STING pathway not only facilitates the production of type I interfer-
ons (IFN-I) and inflammatory responses but also triggers autophagy. Autophagy is a homeostatic
process that exerts multiple effects on innate immunity. However, systematic evidence linking
the cGAS–STING pathway and autophagy is still lacking. Therefore, one goal of this review is to
summarize the known mechanisms of autophagy induced by the cGAS–STING pathway and their
consequences. The cGAS–STING pathway can trigger canonical autophagy through liquid-phase
separation of the cGAS–DNA complex, interaction of cGAS and Beclin-1, and STING-triggered ER
stress–mTOR signaling. Furthermore, both cGAS and STING can induce non-canonical autophagy
via LC3-interacting regions and binding with LC3. Subsequently, autophagy induced by the cGAS–
STING pathway plays crucial roles in balancing innate immune responses, maintaining intracellular
environmental homeostasis, alleviating liver injury, and limiting tumor growth and transformation.
Keywords: cGAS; STING; autophagy; innate immunity; IFN; DNA sensing
1. Introduction
Pattern recognition receptors (PRRs) serve as innate immune sensors of danger sig-
nals, including pathogen-associated molecular patterns (PAMPs) and danger-associated
molecular patterns (DAMPs), and activate the cellular stress response [
1
]. DNA, as PAMPs
or DAMPs, can be sensed by PRRs to alert cells about the presence of microbial pathogens
as well as of damaged or malignant cells and trigger innate immune responses [
2
]. The
cGAS–STING pathway, comprising the cyclic GMP–AMP synthase (cGAS) and the stimula-
tor of interferon genes (STING), was discovered as an important DNA-sensing machinery
in innate immunity and pathogenic defense [
3
5
]. The cGAS–STING axis can be activated
by both non-self DNA and self DNA including microbial DNA, released mitochondrial
DNA, extra nuclear chromatin, cytosolic micronuclei, and aberrant chromosomal DNA
(Figure 1). Upon binding DNA, cGAS produces cyclic GMP–AMP (2
0
3
0
-cGAMP) that binds
to and activates the adaptor protein STING [
6
,
7
]. Activated STING translocates from the
endoplasmic reticulum (ER) to the Golgi apparatus via the endoplasmic reticulum–Golgi
intermediate compartment (ERGIC) [
8
]. In the meantime, activated STING proteins form
Int. J. Mol. Sci. 2021,22, 13232. https://doi.org/10.3390/ijms222413232 https://www.mdpi.com/journal/ijms
Int. J. Mol. Sci. 2021,22, 13232 2 of 14
large oligomers and recruit the downstream TANK-binding kinase 1 (TBK1) via the TBK1
binding motif (TBM) at the C-terminal tail (CTT). The CTT-bound TBK1 phosphorylates
the serine in the STING motif pLxIS (Ser365 in mouse and Ser366 in humans) at the CTT
domain, which provides a binding site for interferon regulatory factor 3 (IRF3) [
9
]. IRF3,
upon phosphorylation by the nearby TBK1, forms a dimer and translocates to the nucleus,
inducing type I IFNs (IFN-I) and other cytokines. After activation by STING, TBK1 and
its homolog I
κ
B kinase epsilon (IKK
ε
) activate the transcription factor nuclear factor
κ
B
(NF-κB) through the IKK complex [10].
Recently, several studies reported that the intrinsic cGAS–STING pathway not only
facilitates IFN-I and inflammatory production, but also activates other cellular processes
such as autophagy [
1
,
11
]. The cGAS–STING pathway could induce autophagy in a variety
of species, including mammals, Drosophila, and sea anemone (Nematostella vectensis).
The cGAS–STING pathway was shown to be responsible for the activation of autophagy
in human cells with extra chromosomes, such as in cells from individuals with Down
syndrome or other aneuploidy-associated pathologies [
12
]. Studies in herpes simplex
virus type1 (HSV-1)-infected murine embryonic fibroblasts showed that HSV-1 infection
significantly increased LC3 punctum formation and autophagic flux through the cGAS–
STING pathway [
13
]. A research in Zika virus-infected Drosophila (Drosophila melanogaster)
brain indicated that Zika virus induced LC3 lipidation through the STING pathway, which
induced antiviral autophagy to limit Zika virus in the Drosophila brain [
14
]. It also
suggested that autophagy is an ancient and highly conserved function of the cGAS–STING
pathway, which predates the emergence of the type-I interferon pathway in vertebrates [
8
].
STING from the sea anemone (Nematostella vectensis) induces autophagy but not interferon
in response to stimulation by cGAMP, which suggests that autophagy is a primordial
function of the cGAS–STING pathway [8].
Several studies have provided evidence showing that autophagy could be induced
by cGAS directly without any involvement of STING during DNA virus infections [
15
,
16
].
However, other studies have revealed that autophagy could be activated by STING after
stimulation by cGAMP or poly(dA:dT) [
13
,
17
]. Besides, many studies reported that the
cGAS–STING pathway induces a classical and canonical pathway-associated autophagy,
whereas other researches indicated that the cGAS–STING pathway induces a non-canonical
pathway-associated autophagy [
13
,
18
]. Given the complex mechanisms used by the cGAS–
STING pathway to activate autophagy, several crucial and meaningful questions naturally
arise: How does the cGAS–STING pathway regulate autophagy? What kind of autophagy
is induced by the cGAS–STING pathway? Why does the cGAS–STING pathway trigger
autophagy? What are the effects of cGAS–STING-induced autophagy? Thus, the aim of
this review is to summarize the available mechanisms and the consequences of cGAS–
STING-activated autophagy.
Int. J. Mol. Sci. 2021,22, 13232 3 of 14
Figure 1.
Overview of the cGAS–STING signaling pathway. The cytosolic DNA receptor cGAS could be activated by
both non-self DNA and self DNA including microbial DNA, released mitochondrial DNA, cytosolic micronuclei, and
aberrant chromosomal DNA. Mn
2+
is also able to activate cGAS, leading to an unconventional catalytic synthesis of
2
0
3
0
-cGAMP [
19
]. The second messenger 2
0
3
0
-cGAMP and other cyclic dinucleotides (CDNs) could cross the cell membrane
through gap junction intercellular communication (GJIC) [
20
]. Then, 2
0
3
0
-cGAMP binds to STING dimers localized at the
ER membrane, which leads to profound conformational changes that trigger STING oligomerization and liberation from
anchoring factors. STING also can be activated by cGAMP from bacterial and GJIC sources, which establishes a role for
STING as an independent pattern recognition receptor. Next, STING translocates from the ER to the Golgi apparatus through
ERGIC. During transportation, STING recruits TBK1, thus promoting TBK1 autophosphorylation, and the phosphorylation
of STING at the C-terminal tail, which provides a binding site for IRF3. The cGAS–STING signaling pathway not only
induces type I IFN and inflammatory signaling responses but also activates other cellular processes including autophagy
and apoptosis.
2. Canonical and Non-Canonical Autophagy
Autophagy is an evolutionally conserved, highly regulated catabolic process that not
only degrades and recycles long-lived proteins, but also eliminates dysfunctional or broken
organelles including mitochondria, peroxisomes, and ribosomes [
21
]. Currently, three
main types of autophagy are recognized, including macroautophagy, microautophagy
and chaperone-mediated autophagy, according to the different ways by which the cellu-
lar content is incorporated into lysosomes. Among these, macroautophagy is the most
prevalent and best studied type, and is usually referred to as canonical autophagy. It
is characterized by the delivery of cytoplasmic cargo to the lysosomes, which depends
Int. J. Mol. Sci. 2021,22, 13232 4 of 14
on the hierarchically ordered activity of autophagy-related (Atg) proteins recruited at
the phagophore assembly site to form an autophagosome and ultimately fuse with the
lysosomal compartment [
22
]. In contrast to canonical autophagy, recent findings sug-
gested that macroautophagy can also occur in the absence of some key autophagy proteins
through the unconventional biogenesis of autophagosomes. This type has been defined as
non-canonical autophagy [23].
Canonical autophagy is initiated by various cellular stresses and can be roughly di-
vided into five steps: (1) initiation, (2) nucleation, (3) expansion, (4) fusion, (5) degradation.
It is controlled by Atg family proteins, kinases, and lipid metabolism (
Figure 2
). The initia-
tion step is triggered by inhibiting mechanistic target of rapamycin kinase (mTOR) or acti-
vating AMP-activated protein kinase (AMPK), leading to the assembly and activation of the
ULK1 complex including ULK1, Atg13, Atg101, and FIP200 [
24
26
]. The step of nucleation
requires the ULK1 complex to phosphorylate the class III PI3KC3 complex which consists
of the tumor suppressor Beclin-1, Beclin-1-regulated autophagy protein 1 (AMBRA1), the
lipid kinase VPS34, the scaffolding protein VPS15, and the autophagy-specific subunit
Atg14 [
27
]. The ATG12–ATG5–ATG16L1 complex and LC3–phosphatidylethanolamine
(PE) are necessary for promoting the expansion of the phagophore membrane [
28
]. Af-
ter autophagosome formation, autophagosomes undergo a stepwise maturation process
including fusion events with multivesicular endosomes and lysosomes. Fusion with
lysosomes is necessary for the complete degradation of the segregated cytoplasm [29].
Non-canonical autophagy is also a key cellular pathway in immunity, characterized
by the conjugation of LC3 to endolysosomal membranes such as phagosomes and en-
dosomes [
30
]. Non-canonical autophagy is associated with endocytic and engulfment
processes, including LC3-associated endocytosis (LANDO) and LC3-associated phagocy-
tosis (LAP), which have essential functions in the clearance of pathogens and cytokines
and in responses to lysosome biogenesis. During the process of LANDO and LAP, LC3
coats the surface of endosomes and phagosomes, respectively, regulating the trafficking
and lysosomal clearance of engulfed extracellular materials [
31
]. The canonical and non-
canonical autophagy pathways share overlapping machineries, but there are essential
differences (Figure 2). First of all, canonical autophagy is initiated by the formation of
a double-membrane phagophore known as the isolation membrane, which engulfs the
cytosolic cargo. Compared to canonical autophagy, non-canonical autophagy is character-
ized by a single-membrane phagosome [
32
,
33
]. Furthermore, the non-canonical pathway
is independent of the upstream autophagy regulators mTOR and ULK1 initiation com-
plex; nevertheless, it needs the class III PI3KC3 complex and members of the protein
lipid conjugation complex [
34
]. Additionally, the WD (tryptophan–aspartic acid) repeat-
containing C-terminal domain of ATG16L is essential for LC3 recruitment to endolysosomal
membranes during non-canonical forms of autophagy but is dispensable for canonical
autophagy [35].
Int. J. Mol. Sci. 2021,22, 13232 5 of 14
Figure 2.
Schematic presentation of canonical and non-canonical autophagy. (
A
) The process of canonical autophagy.
Canonical autophagy can be roughly divided into five steps: (1) initiation, (2) nucleation, (3) expansion, (4) fusion,
(5) degradation and is mediated by Atg family proteins, kinases, and lipid metabolism. (
B
) The process of LAP-like non-
canonical autophagy. LAP-like non-canonical autophagy is initiated by autophagy receptors through their LIR interacting
with LC3. LC3 is recruited to single-membrane phagosomes. LAP-like non-canonical autophagy is independent of the
upstream autophagy regulators mTOR and ULK1 initiation complex, while it needs the class III PI3KC3 complex and
members of the protein lipid conjugation complex.
3. Molecular Mechanisms of cGAS–STING Pathway-Induced Autophagy
3.1. cGAS Can Directly Interact with Beclin-1 to Induce Autophagy
Liang and collaborators indicated that cytoplasmic DNA could trigger autophagy
and recruit LC3-positive autophagic vesicles by promoting cGAS to interaction with
Beclin-1 [15]
. cGAS contains an amino (N)-terminal regulatory domain (RD, a.a. 1–160), a
central nucleotidyl transferase (NTase) domain (a.a. 161–330), and a carboxyl (C)-terminal
domain (CTD, a.a. 331–522) [
36
]. The central region of Beclin-1 contains a Bcl-2 binding
domain (BD, a.a. 112–159) for cellular and viral Bcl-2 interactions, a coiled-coil domain
(CCD, a.a.
142–270
) for Atg14L and UVRAG interaction, and an evolutionarily conserved
domain (ECD, a.a. 245–450) for PI3KC3 (Vps34) interaction [
15
,
37
]. It was found that the
Int. J. Mol. Sci. 2021,22, 13232 6 of 14
central NTase domain of cGAS and the central CCD of Beclin-1 are responsible for their
interactions [15].
The interaction between cGAS and Beclin-1 increases the level of autophagy by com-
peting with the binding of Rubicon to Beclin-1 (Figure 3). Beclin-1 is a core component
of the Beclin-1–PI3KC3 complex, a lipid–kinase complex involved in autophagosome nu-
cleation [
38
]. The activity of the Beclin-1–PI3KC3 complex is tightly regulated by many
factors, especially different binding partners [
39
]. Atg14L and Rubicon associate with
the Beclin-1–PI3KC3 complex; Atg14L positively regulates autophagy at various steps,
whereas Rubicon negatively regulates both autophagy and endocytosis at the membrane
fusion step by suppressing PI3KC3 lipid kinase activity [
15
,
40
]. It has been shown that
Rubicon is stably associated with Beclin-1 complexes in 293T cells. Upon stimulation by
dsDNA, Rubicon progressively dissociated from the Beclin-1–PI3KC3 complex, while the
association between Atg14L and the Beclin-1–PI3KC3 complex was not affected [
15
]. Since
both cGAS and Rubicon competitively bind to the first two CCDs of Beclin-1, increasing
the cGAS–Beclin-1 interaction could cause the decrease of the Rubicon–Beclin-1 interac-
tion [
15
]. Taken together, the interaction of cGAS with Beclin-1 leads to the dissociation of
the negative autophagy factor Rubicon from the Beclin-1 complex, initiating the activation
of PI3KC3 kinase and thereby increasing autophagy.
3.2. cGAS Can Directly Interact with LC3 to Induce Autophagy
The abundance of proteins and cellular organelles is usually controlled by selective
autophagy, a process mediated by autophagy receptors that simultaneously bind the car-
goes and LC3 proteins on phagophores’ membranes. Studies have shown that cGAS is a
receptor that induces selective autophagy by interacting with LC3 [
18
]. LC3-interacting
regions (LIRs) are short linear motifs within selective autophagy receptors, which can
bind to LC3 and induce autophagy [
41
]. cGAS was found to contain a total of five LIRs,
two of which are conserved in different species [
18
]. The LIR motif of cGAS mediates the
interaction with LC3, and deletion of LIR355–360 or mutation of the evolutionarily con-
served phenylalanine 357 and valine 360 impairs the interaction of cGAS with endogenous
LC3 [
18
]. Additionally, it was indicated that Atg14 and Atg7 are involved in the process
of autophagy induced by cGAS [
18
]. Atg14, a subunit of the PI3K complex, localizes to
isolation phagophores and is essential for autophagosome formation during the process
of canonical autophagy [
42
]. ATG-7 is an E1 ubiquitin-activating enzyme required for
the activation of the ATG12–ATG5–ATG16L1 complex, which is crucial for phagophore
elongation and autophagosome biogenesis during canonical autophagy. These studies
have suggested that cGAS could trigger non-canonical autophagy by interacting with LC3,
and this process shares machineries with canonical autophagy, such as the PI3K complex
and members of the protein lipid conjugation complex (Figure 3).
3.3. The cGAS–dsDNA Polymer Can Form a Liquid-Phase Condensate, Which Could Be an
Initiator of Autophagy
An accumulating amount of DNA in the cytoplasm could activate cGAS and drive
the formation of liquid-like droplets, which allows cGAS to detect the presence of DNA in
the cytoplasm above a certain threshold and trigger a switch-like response [
43
,
44
]. When
cGAS is activated by DNA ligands, it assembles into a dimer, in which the DNA strands are
sandwiched between two cGAS protomers [
3
]. Each cGAS protomer contains two principal
DNA binding sites, named A and B. In the dimer, two dsDNA strands are bound with
two cGAS, while one dsDNA binds to site A of one cGAS protomer, and the other dsDNA
strand binds to site B of the other cGAS protomer [
3
]. The formation of the initial 2:2
cGAS–DNA complex prearranges the two DNA duplexes in a roughly parallel fashion, and
a recent study has indicated that multiple cGAS dimers can form a ladder-like assembly
with long DNA ligands [
45
]. DNA binding to cGAS induces a robust phase transition to
liquid-like droplets, which function as microreactors in which the enzyme and reactants
are concentrated to drastically enhance the production of 2
0
3
0
-cGAMP [
43
]. Several studies
suggested that liquid-like droplets of cGAS could provide a mechanism that prevents cGAS
Int. J. Mol. Sci. 2021,22, 13232 7 of 14
activation by a low amount of genomic or mitochondrial DNA that might be present in
the cytoplasm at different cell cycle stages [
46
]. Because cGAS cannot distinguish foreign
DNA from self-DNA, it relies on liquid-phase separation to measure the concentrations of
cGAS and DNA in the cytoplasm and mount a switch-like immune response only when
the concentrations of these molecules rise above a certain threshold [46].
Increasing evidence suggests that the liquid-like condensate is a main regulator of
both bulk and selective autophagy [
47
]. Liquid-like protein droplets undergo internal
rearrangement and can recruit other molecules, with a potential to host biochemical re-
actions. Therefore, it is possible that thanks to its liquid-like properties, the condensates
could provide a platform for recruiting autophagy-related molecules such as LC3, p62, and
Atg proteins that drive autophagosome formation [48,49]. In this pathway, the liquid-like
properties of condensates gradually form larger aggregates, are labelled by ubiquitin,
and eventually become autophagosomes and are degraded when they fuse with lyso-
somes [
48
,
50
]. Further, it was suggested that liquid-like condensates play important roles
in multiple steps of autophagy, mediating the assembly of autophagosome formation
sites, acting as a modulators of TORC1-mediated autophagy regulation, and triaging pro-
tein cargos for degradation [
47
].Thus, autophagy could be initiated by the Liquid-Phase
Condensate formed by The cGAS-dsDNA Polymer (Figure 3).
3.4. STING Can Induce Autophagy by Interacting with LC3
It was found that STING is an autophagy receptor that can directly interact with LC3
via its LIR motifs to mediate autophagy [
13
]. Co-IP experiments and pulldown analyses
have shown that STING and LC3 could physically interact with each other [
13
]. The iLIR
web server was used to analyze the STING protein sequence, and it was revealed that
STING contains seven putative LIRs (a.a. 40–52, 76–88, 100–112, 161–173, 180–192, 193–205,
and 239–251) [
13
]. It has been demonstrated that three LIRs (a.a. 161–173, 193–205, and
239–251) are involved in the interactions between STING and LC3 [
13
]. Further studies
have indicated that the process of STING-induced autophagy requires the activation of
STING by phosphorylation [
8
]. Activated STING could leave the ER, moving to the ERGIC
and Golgi in a process dependent on the COP-II complex and ARF GTPases [51]. Vesicles
budding from the ER and ERGIC could serve as membrane sources for LC3 lipidation and
autophagosome biogenesis [
52
]. Thus, it is possible that autophagy induced by STING re-
quires the membrane fraction derived from ER or ERGIC as the substrate for LC3 lipidation.
It was shown that treatment with Brefeldin A (BFA), a specific inhibitor of protein traffick-
ing, specifically inhibited protein trafficking from ER to ERGIC by targeting ARF GTPases.
Under this condition, STING-stimulated cells lost the ability to stimulate LC3 lipidation [
8
].
Therefore, the process of budding from ER is required for STING-induced autophagy.
It was reported that autophagy induced by STING is independent of upstream au-
tophagy regulators such as Beclin-1, ULK1, and Atg9a, but dependent on downstream
autophagy regulators such as Atg5 and Atg16L1 [
13
,
17
]. It was also shown that knockout
of Beclin-1, ULK1, or Atg9a failed to block poly(dA:dT) and STING-induced LC3-II conver-
sion and punctum formation. However, knockdown of Atg5 could block STING-induced
LC3 punctum formation and autophagic flux in HeLa cells [
13
]. Additionally, STING
activation induced LC3 lipidation onto single-membrane perinuclear vesicles, mediated
by ATG16L1 via its WD40 domain, bypassing the requirement of the canonical upstream
autophagy machinery [
17
]. These studies suggest that STING initiates non-canonical
autophagy by interacting with LC3 (Figure 3).
3.5. STING Triggers Autophagy through the ER Stress–mTOR Pathway
Wu and collaborators indicated that STING was involved in the activation of ER stress
and unfolded protein response (UPR) [
53
]. The accumulation of unfolded or misfolded
proteins in the ER could result in ER stress, which triggers the UPR to alleviate it and restore
ER homeostasis [
54
]. The process of UPR is relayed through three ER transmembrane
sensors, i.e., IRE1
α
, PERK, and ATF6, and these UPR sensors are typically bound to
Int. J. Mol. Sci. 2021,22, 13232 8 of 14
the key ER chaperone GRP78 to prevent downstream signaling [
55
]. When unfolded
protein stress reaches a critical threshold, GRP78 can dissociate from these sensors thus
allowing downstream signaling activation that regulates transcription and translation
and restores ER homeostasis [
56
]. It was shown that co-transfecting cGAS and STING
into HEK293T cells to transiently activate STING signaling could upregulate UPR gene
expression including the levels of Activating transcription factor 3 (ATF3) and Growth
arrest and DNA damage-inducible protein 34 (GADD34) mRNAs, which are markers of
ER stress and UPR [
53
]. It has been indicated that live bacteria could elicit an ER stress
response through c-di-AMP to activate the STING pathway [
57
]. Further, STING resides in
the ER where it maintains ER calcium homeostasis through its interaction with the Ca
2+
sensor stromal interaction molecule 1 (STIM1) [
58
]. Additionally, it was shown that STING
is a crucial regulator of ER stress, as the protein expression levels of p-PERK, p-IRE-1
α
, and
p-eIF2
α
were markedly stimulated by aortic banding surgery; however, these results were
markedly restrained in STING-KO mice [59].
It has been observed that STING mediated ER stress and the UPR through a novel
motif, named “the UPR motif” [
53
]. Structural and functional analyses demonstrated
that the UPR and IFN are mediated through distinct domains of STING. The helix (a.a
322–343) of STING, specifically, residues R331 and R334, is critically required for STING-
mediated UPR. Deletion of the helix from full-length STING (
322–343) could abrogate
STING-mediated GADD34 induction [53].
STING triggers autophagy through the ER stress–mTOR pathway. Autophagy is neg-
atively controlled by the mTOR signaling pathway [
60
]. mTOR is a downstream effector of
the PI3K/AKT pathway and forms two distinct multiprotein complexes, namely, mTORC1
and mTORC2 [
61
]. mTORC1 is a key inhibitor of autophagy activated by diverse stimuli
such as growth factors, nutrients, energy, and stress signals to control cell growth, prolifer-
ation, and survival; however, the function of mTORC2 in autophagy is controversial [
62
].
It was shown that ER stress induces autophagy through a negative regulation of the AKT
mTOR pathway [
63
]. The PERK–CHOP pathway activated by ER stress could mediate
the induction of TRB3, which can directly bind to AKT and suppress the AKT–mTOR
signaling pathway. Additionally, one study indicated that bacterial c-di-AMP activated
STING and triggered ER stress, leading to mTOR inactivation and subsequent autophagy
in macrophages [
57
]. These studies suggest that STING could trigger autophagy through
the ER Stress–mTOR pathway (Figure 3).
3.6. Role of TBK1 and IRF3 in cGAS–STING Pathway-Induced Autophagy
Accumulating evidence indicates that TBK1 and IRF3 are dispensable for STING-
induced formation of the autophagosome [
9
]. The CTT of STING (a.a 341–379) is required
and essential for recruiting TBK1 and IRF3. It was shown that deletion of the CTT from
STING (
341–379) could impair the downstream phosphorylation of TBK1 and IRF3 but
did not affect LC3 lipidation and the interaction between STING and LC3 [
9
]. The S365A
mutation of STING is defective in phosphorylation, thereby disrupting the recruitment of
IRF3 while retaining the ability to recruit TBK1. A STING mouse strain with a mutation at
position 365 (S365A) showed a disruption in IRF3 binding and the resulting absence of type
I interferon induction but not of autophagy [
9
]. The mutation L373A of STING prevents
the recruitment of TBK1 and the activation of both IRF3 and NF-
κ
B [
64
]. STING knock-in
mice carrying the mutation L373A of STING present normal LC3 lipidation. Additionally,
RNA interference (RNAi) to knock down the STING downstream kinase TBK1 and the
transcription factor IRF3 had no obvious effects on STING-induced autophagy. Neither
TBK1 nor IRF3 deficiency blocked the increase of LC3-II levels and LC3 punctum formation
induced by poly(dA:dT) stimulation [64].
TBK1 is not involved in the formation of autophagosomes but is implicated in their
maturation and degradation. Prabakaran and collaborators indicated that STING was not
degraded by autophagy in TBK1-deficient cells [
65
]. TBK-1 knockdown did not affect the
formation of autophagosomes but suppressed their maturation [
65
]. It was noticed that
Int. J. Mol. Sci. 2021,22, 13232 9 of 14
TBK-1 is a key regulator of immunological autophagy and is responsible for the maturation
of autophagosomes [
65
]. TBK1 mediates maturation and degradation of autophagosomes
through the following mechanisms. Firstly, it was reported that TBK-1 could phosphorylate
the autophagic adaptor p62 on Serine 403, which increased the affinity of p62 for ubiquitin
chains [
66
]. After STING stimulation, p62 and phospho-TBK1 showed significant co-
localization, and phospho-TBK1 was recruited to p62 in stimulated cells [
65
]. Secondly, it
has been shown that in addition to p62, TBK1 could bind to another autophagy receptor,
i.e., NDP52 to form the NDP52–ULK–TBK1 complex, which is essential for the growth of
WIPI2-positive phagophores and LC3-positive autophagosomes to drive xenophagy [
67
].
Thirdly, TBK1 could regulate microtubule dynamics in mitosis and the cytoplasmic levels
of dynein. The maturation of autophagosomes transport to lysosome-rich areas occurs
through microtubules and is dependent on the action of the motor protein dynein [
68
]. It
has been shown that the loss of TBK1 could impair microtubule transport and then the
maturation of autophagosomes into autophagolysosomes [69].
Figure 3.
Schematic mechanism of cGAS–STING pathway-mediated autophagy.
1
cGAS could directly interact with
Beclin-1 to induce autophagy.
2
cGAS could directly interact with LC3 to induce autophagy.
3
cGAS–dsDNA polymers
can form a liquid-phase condensate, which could be an initiator of autophagy.
4
STING could induce autophagy through its
interactions with LC3.
5
STING triggers autophagy through the ER stress–mTOR pathway.
6
Autophagy induced by the
cGAS–STING pathway is crucial for the modulation of innate immune responses, to maintain intracellular environmental
homeostasis, to protect against liver injuries, and to limit cellular transformation and tumor growth.
4. Functions of Autophagy Induced by the cGAS–STING Pathway
Autophagy induced by the cGAS–STING pathway is crucial for preventing excessive
immune response and persistent immune stimulation. Autophagy induced by cGAS–
STING could modulate the innate immune responses through the degradation of cGAS
and STING and the elimination of DNA and pathogens from the cytosol. cGAS-induced
autophagy through its interaction with LC3 and Beclin-1 not only suppresses the synthesis
of 2
0
3
0
-cGAMP to halt type I IFN production, but also enhances autophagy-mediated degra-
dation of cGAS and DNA to prevent excessive cGAS activation and persistent immune
stimulation [
15
,
70
]. STING can be degraded by autophagy through TBK1-mediated p62
Int. J. Mol. Sci. 2021,22, 13232 10 of 14
phosphorylation to drive STING ubiquitination and autophagic degradation that could
attenuate the innate immune signaling [
65
,
70
]. Autophagy is thus able to control infections,
as it functions as an intracellular defense mechanism capturing cytosol-invading pathogens
and driving them to lysosomal degradation [
71
]. During Mycobacterium tuberculosis or
HSV-1 infections, cytosolic pathogenic DNA triggers cGAS–STING pathway-mediated
autophagy to eliminate the pathogens [72].
Further, autophagy induced by the cGAS–STING pathway is crucial for maintaining
intracellular environmental homeostasis [
73
]. The cGAS–STING pathway induced au-
tophagy through the ER stress pathway, which could in turn alleviate ER stress and restore
homeostasis via isolating and sequestering it [
74
]. Moretti and colleagues revealed that after
sensing c-di-AMP derived from Gram-positive bacteria, STING triggered ER stress, mTOR
inactivation, and ER-phagy and eventually curtailed cell death [
57
]. It was also suggested
that autophagy induced by the cGAS–STING pathway plays a crucial role in micronuclei
homeostasis [
18
,
75
]. cGAS reduced the abundance of micronuclei through the induction
of autophagy, cleaning micronuclei and preventing chromosomal instability [
18
]. cGAS
is a crucial regulator of selective autophagy to clear the micronuclei caused by genotoxic
stress [
18
]. Knockdown of cGAS along with induction of mitotic arrest in HeLa and U2OS
cancer cells clearly resulted in an increase of micronuclei formation and chromosome mis-
segregation [
75
]. Amar and his collaborators suggested that STING-mediated autophagy is
protective against H
2
O
2
-induced cell death [
73
]. STING-mediated autophagy serves as a
protective mechanism to remove damaged cells and promotes a protective environment in
MEFSV40 cells after treatment with H2O2[73].
Additionally, it was found that autophagy induced by the cGAS–STING pathway
exerts a protective role in liver cancer, limiting transformation and tumor growth [
16
].
cGAS-mediated autophagy protects the liver from ischemia/reperfusion (I/R) injury, which
is independent of STING. Deletion of cGAS suppressed hypoxia-induced autophagy in
hepatocytes and thus led to apoptotic cell death during liver I/R [
16
]. Autophagy is
a therapeutic strategy in cancer, through the modulation of both therapeutic resistance
and the death of cancer cells [
76
]. STING activation induced by host DNA damages can
trigger autophagy-dependent cell death to remove cancer cells or other stressed cells [
25
].
Guendalina and colleagues suggested that oncolytic viruses can successfully activate
antitumor immunity via the activation of a STING-dependent antiviral cascade in cancer
cells [
77
].The nucleoside analog zalcitabine (an antiviral drug) induces mitochondrial
damage and the release of mtDNA into the cytosol, resulting in the activation of the cGAS–
STING pathway, which in turn induces autophagy-dependent ferroptosis and suppresses
pancreatic tumor growth in mice [
78
]. The release of DNA from the nucleus to the cytosol
caused by nuclear cathepsin B (CTSB)-mediated genomic DNA damage activates STING-
dependent autophagy and leads to ferroptotic cell death in human pancreatic cancer
cells [79].
5. Conclusions and Future Perspective
The cGAS–STING pathway triggers autophagy through both canonical and non-
canonical pathways. It triggers canonical autophagy through the interactions of cGAS and
Beclin-1, the liquid-phase separation of the cGAS–DNA complex, and STING-triggered
ER stress–mTOR signaling. It also triggers non-canonical autophagy through the binding
of cGAS and/or STING to LC3. Autophagy induced by the cGAS–STING pathway is
crucial for balancing innate immune responses, maintaining intracellular environmental
homeostasis, preventing liver injury, and limiting tumor growth and transformation.
It was reported that the process of budding from ER is required for STING-induced
autophagy; however, whether STING transportation from ERGIC to Golgi is involved in
the process of STING-induced autophagy is unknown. Further, it was suggested that IRF3
is not involved in the formation of autophagosome induced by the cGAS–STING pathway
but is essential for DNA-stimulated degradation of STING by autophagy [
65
]. However,
the molecular mechanisms activated by IRF3 in STING degradation are not resolved. Thus,
Int. J. Mol. Sci. 2021,22, 13232 11 of 14
it is recommended that studies should be designed to explore these aspects. Additionally,
evidence has shown that the cGAS–STING pathway not only activates autophagy but also
induces apoptosis [
80
]. Apoptosis can play a key role in the innate response to pathogenic
infection [
81
]. It is recommended that the relationships between apoptosis, autophagy, and
IFN responses induced by the cGAS–STING pathway draw more attention.
Author Contributions:
J.Z. (Jianzhong Zhu), Z.L. and W.Z. proposed the idea and wrote the
manuscript; N.X. and J.Z. (Jiajia Zhang) contributed to reference collection and figures; N.C. and F.M.
read the manuscript and provided advice. All authors have read and agreed to the published version
of the manuscript.
Funding:
The work was partly supported by the National Key Research and Development Program
of China (2017YFD0502301), Jiangsu provincial key R & D plan (BE2020398), National Natural Science
Foundation of China (32172867; 31872450), the Priority Academic Program Development of Jiangsu
Higher Education Institutions (PAPD), and China Postdoctoral Science Foundation (2019M661952).
Institutional Review Board Statement: Not applicable.
Informed Consent Statement: Not applicable.
Data Availability Statement: Not applicable.
Acknowledgments:
We would like to thank Qi Shao for her polishing the language. Special thanks
belong to Wei Wu for her support to draw figures.
Conflicts of Interest: The authors declare no conflict of interest.
References
1.
Wan, D.; Jiang, W.; Hao, J. Research Advances in How the cGAS-STING Pathway Controls the Cellular Inflammatory Response.
Front. Immunol. 2020,11, 615. [CrossRef]
2.
Santoni, G.; Cardinali, C.; Morelli, M.B.; Santoni, M.; Nabissi, M.; Amantini, C. Danger- and pathogen-associated molecular
patterns recognition by pattern-recognition receptors and ion channels of the transient receptor potential family triggers the
inflammasome activation in immune cells and sensory neurons. J. Neuroinflamm. 2015,12, 21. [CrossRef]
3.
Hopfner, K.P.; Hornung, V. Molecular mechanisms and cellular functions of cGAS-STING signalling. Nat. Reviews. Mol. Cell Biol.
2020,21, 501–521. [CrossRef]
4.
Storozynsky, Q.; Hitt, M.M. The Impact of Radiation-Induced DNA Damage on cGAS-STING-Mediated Immune Responses to
Cancer. Int. J. Mol. Sci. 2020,21, 8877. [CrossRef] [PubMed]
5.
Li, Q.; Liu, C.; Yue, R.; El-Ashram, S.; Wang, J.; He, X.; Zhao, D.; Zhou, X.; Xu, L. cGAS/STING/TBK1/IRF3 Signaling Pathway
Activates BMDCs Maturation Following Mycobacterium bovis Infection. Int. J. Mol. Sci. 2019,20, 895. [CrossRef]
6.
Ma, R.; Ortiz Serrano, T.P.; Davis, J.; Prigge, A.D.; Ridge, K.M. The cGAS-STING pathway: The role of self-DNA sensing in
inflammatory lung disease. FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol. 2020,34, 13156–13170. [CrossRef]
7.
Rolf, J.; Siedentop, R.; Lutz, S.; Rosenthal, K. Screening and Identification of Novel cGAS Homologues Using a Combination of In
Vitro and In Vivo Protein Synthesis. Int. J. Mol. Sci. 2020,21, 105. [CrossRef]
8.
Gui, X.; Yang, H.; Li, T.; Tan, X.; Shi, P.; Li, M.; Du, F.; Chen, Z.J. Autophagy induction via STING trafficking is a primordial
function of the cGAS pathway. Nature 2019,567, 262–266. [CrossRef] [PubMed]
9.
Yum, S.; Li, M.; Fang, Y.; Chen, Z.J. TBK1 recruitment to STING activates both IRF3 and NF-kappaB that mediate immune defense
against tumors and viral infections. Proc. Natl. Acad. Sci. USA 2021,118, e2100225118. [CrossRef] [PubMed]
10.
Balka, K.R.; Louis, C.; Saunders, T.L.; Smith, A.M.; Calleja, D.J.; D’Silva, D.B.; Moghaddas, F.; Tailler, M.; Lawlor, K.E.; Zhan, Y.;
et al. TBK1 and IKKepsilon Act Redundantly to Mediate STING-Induced NF-kappaB Responses in Myeloid Cells. Cell Rep.
2020
,
31, 107492. [CrossRef]
11.
Liu, Y.; Gordesky-Gold, B.; Leney-Greene, M.; Weinbren, N.L.; Tudor, M.; Cherry, S. Inflammation-Induced, STING-Dependent
Autophagy Restricts Zika Virus Infection in the Drosophila Brain. Cell Host Microbe 2018,24, 57–68.e3. [CrossRef]
12.
Krivega, M.; Stiefel, C.M.; Karbassi, S.; Andersen, L.L.; Chunduri, N.K.; Donnelly, N.; Pichlmair, A.; Storchova, Z. Genotoxic
stress in constitutive trisomies induces autophagy and the innate immune response via the cGAS-STING pathway. Commun. Biol.
2021,4, 831. [CrossRef]
13.
Liu, D.; Wu, H.; Wang, C.; Li, Y.; Tian, H.; Siraj, S.; Sehgal, S.A.; Wang, X.; Wang, J.; Shang, Y.; et al. STING directly activates
autophagy to tune the innate immune response. Cell Death Differ. 2019,26, 1735–1749. [CrossRef]
14.
Delorme-Axford, E.; Klionsky, D.J. Inflammatory-dependent Sting activation induces antiviral autophagy to limit zika virus in
the Drosophila brain. Autophagy 2019,15, 1–3. [CrossRef] [PubMed]
Int. J. Mol. Sci. 2021,22, 13232 12 of 14
15.
Liang, Q.; Seo, G.J.; Choi, Y.J.; Kwak, M.J.; Ge, J.; Rodgers, M.A.; Shi, M.; Leslie, B.J.; Hopfner, K.P.; Ha, T.; et al. Crosstalk between
the cGAS DNA sensor and Beclin-1 autophagy protein shapes innate antimicrobial immune responses. Cell Host Microbe
2014
,15,
228–238. [CrossRef] [PubMed]
16.
Lei, Z.; Deng, M.; Yi, Z.; Sun, Q.; Shapiro, R.A.; Xu, H.; Li, T.; Loughran, P.A.; Griepentrog, J.E.; Huang, H.; et al. cGAS-mediated
autophagy protects the liver from ischemia-reperfusion injury independently of STING. Am. J. Physiol. Gastrointest. Liver Physiol.
2018,314, G655–G667. [CrossRef]
17.
Fischer, T.D.; Wang, C.; Padman, B.S.; Lazarou, M.; Youle, R.J. STING induces LC3B lipidation onto single-membrane vesicles via
the V-ATPase and ATG16L1-WD40 domain. J. Cell Biol. 2020,219, e202009128. [CrossRef] [PubMed]
18.
Zhao, M.; Wang, F.; Wu, J.; Cheng, Y.; Cao, Y.; Wu, X.; Ma, M.; Tang, F.; Liu, Z.; Liu, H.; et al. CGAS is a micronucleophagy
receptor for the clearance of micronuclei. Autophagy 2021, 1–17. [CrossRef]
19.
Zhang, R.; Wang, C.; Guan, Y.; Wei, X.; Sha, M.; Yi, M.; Jing, M.; Lv, M.; Guo, W.; Xu, J.; et al. Manganese salts function as potent
adjuvants. Cell. Mol. Immunol. 2021,18, 1222–1234. [CrossRef] [PubMed]
20.
Jindal, S.; Chockalingam, S.; Ghosh, S.S.; Packirisamy, G. Connexin and gap junctions: Perspectives from biology to nanotechnol-
ogy based therapeutics. Transl. Res. J. Lab. Clin. Med. 2021,235, 144–167. [CrossRef] [PubMed]
21.
Jacquin, E.; Leclerc-Mercier, S.; Judon, C.; Blanchard, E.; Fraitag, S.; Florey, O. Pharmacological modulators of autophagy activate
a parallel noncanonical pathway driving unconventional LC3 lipidation. Autophagy 2017,13, 854–867. [CrossRef] [PubMed]
22.
Codogno, P.; Mehrpour, M.; Proikas-Cezanne, T. Canonical and non-canonical autophagy: Variations on a common theme of
self-eating? Nat. Rev. Mol. Cell Biol. 2011,13, 7–12. [CrossRef] [PubMed]
23. Juenemann, K.; Reits, E.A. Alternative macroautophagic pathways. Int. J. Cell Biol. 2012,2012, 189794. [CrossRef] [PubMed]
24.
Xie, Y.; Kang, R.; Sun, X.; Zhong, M.; Huang, J.; Klionsky, D.J.; Tang, D. Posttranslational modification of autophagy-related
proteins in macroautophagy. Autophagy 2015,11, 28–45. [CrossRef] [PubMed]
25.
Zhang, R.; Kang, R.; Tang, D. The STING1 network regulates autophagy and cell death. Signal. Transduct. Target. Ther.
2021
,6,
208. [CrossRef] [PubMed]
26.
LeBlond, N.D.; Nunes, J.R.C.; Smith, T.K.T.; O’Dwyer, C.; Robichaud, S.; Gadde, S.; Cote, M.; Kemp, B.E.; Ouimet, M.; Fullerton,
M.D. Foam Cell Induction Activates AMPK But Uncouples Its Regulation of Autophagy and Lysosomal Homeostasis. Int. J. Mol.
Sci. 2020,21, 9033. [CrossRef]
27.
Russell, R.C.; Tian, Y.; Yuan, H.; Park, H.W.; Chang, Y.Y.; Kim, J.; Kim, H.; Neufeld, T.P.; Dillin, A.; Guan, K.L. ULK1 induces
autophagy by phosphorylating Beclin-1 and activating VPS34 lipid kinase. Nat. Cell Biol.
2013
,15, 741–750. [CrossRef] [PubMed]
28.
Wei, Y.; Liu, M.; Li, X.; Liu, J.; Li, H. Origin of the Autophagosome Membrane in Mammals. Biomed. Res. Int.
2018
,2018, 1012789.
[CrossRef]
29.
Eskelinen, E.L.; Saftig, P. Autophagy: A lysosomal degradation pathway with a central role in health and disease. Biochim.
Biophys. Acta 2009,1793, 664–673. [CrossRef]
30.
Romao, S.; Gasser, N.; Becker, A.C.; Guhl, B.; Bajagic, M.; Vanoaica, D.; Ziegler, U.; Roesler, J.; Dengjel, J.; Reichenbach, J.; et al.
Autophagy proteins stabilize pathogen-containing phagosomes for prolonged MHC II antigen processing. J. Cell Biol.
2013
,203,
757–766. [CrossRef] [PubMed]
31.
Leidal, A.M.; Debnath, J. LC3-dependent extracellular vesicle loading and secretion (LDELS). Autophagy
2020
,16, 1162–1163.
[CrossRef]
32. Noda, T.; Yoshimori, T. Molecular basis of canonical and bactericidal autophagy. Int. Immunol. 2009,21, 1199–1204. [CrossRef]
33.
Mehrpour, M.; Esclatine, A.; Beau, I.; Codogno, P. Autophagy in health and disease. 1. Regulation and significance of autophagy:
An overview. Am. J. Physiol. Cell Physiol. 2010,298, C776–C785. [CrossRef]
34.
Sprenkeler, E.G.; Gresnigt, M.S.; van de Veerdonk, F.L. LC3-associated phagocytosis: A crucial mechanism for antifungal host
defence against Aspergillus fumigatus. Cell. Microbiol. 2016,18, 1208–1216. [CrossRef] [PubMed]
35.
Fletcher, K.; Ulferts, R.; Jacquin, E.; Veith, T.; Gammoh, N.; Arasteh, J.M.; Mayer, U.; Carding, S.R.; Wileman, T.; Beale, R.; et al.
The WD40 domain of ATG16L1 is required for its non-canonical role in lipidation of LC3 at single membranes. EMBO J.
2018
,
37, e97840. [CrossRef] [PubMed]
36.
Seo, G.J.; Kim, C.; Shin, W.J.; Sklan, E.H.; Eoh, H.; Jung, J.U. TRIM56-mediated monoubiquitination of cGAS for cytosolic DNA
sensing. Nat. Commun. 2018,9, 613. [CrossRef]
37.
Ashkenazi, A.; Bento, C.F.; Ricketts, T.; Vicinanza, M.; Siddiqi, F.; Pavel, M.; Squitieri, F.; Hardenberg, M.C.; Imarisio, S.; Menzies,
F.M.; et al. Polyglutamine tracts regulate beclin 1-dependent autophagy. Nature 2017,545, 108–111. [CrossRef]
38.
McKnight, N.C.; Zhenyu, Y. Beclin 1, an Essential Component and Master Regulator of PI3K-III in Health and Disease. Curr.
Pathobiol. Rep. 2013,1, 231–238. [CrossRef]
39.
Vega-Rubin-de-Celis, S.; Kinch, L.; Pena-Llopis, S. Regulation of Beclin 1-Mediated Autophagy by Oncogenic Tyrosine Kinases.
Int. J. Mol. Sci. 2020,21, 9210. [CrossRef]
40.
Xu, D.Q.; Wang, Z.; Wang, C.Y.; Zhang, D.Y.; Wan, H.D.; Zhao, Z.L.; Gu, J.; Zhang, Y.X.; Li, Z.G.; Man, K.Y.; et al. PAQR3 controls
autophagy by integrating AMPK signaling to enhance ATG14L-associated PI3K activity. EMBO J.
2016
,35, 496–514. [CrossRef]
[PubMed]
41.
Johansen, T.; Birgisdottir, A.B.; Huber, J.; Kniss, A.; Dotsch, V.; Kirkin, V.; Rogov, V.V. Methods for Studying Interactions Between
Atg8/LC3/GABARAP and LIR-Containing Proteins. Methods Enzymol. 2017,587, 143–169. [CrossRef]
Int. J. Mol. Sci. 2021,22, 13232 13 of 14
42.
Matsunaga, K.; Morita, E.; Saitoh, T.; Akira, S.; Ktistakis, N.T.; Izumi, T.; Noda, T.; Yoshimori, T. Autophagy requires endoplasmic
reticulum targeting of the PI3-kinase complex via Atg14L. J. Cell Biol. 2010,190, 511–521. [CrossRef]
43.
Du, M.; Chen, Z.J. DNA-induced liquid phase condensation of cGAS activates innate immune signaling. Science
2018
,361,
704–709. [CrossRef] [PubMed]
44.
Baba, T.; Yoshida, T.; Tanabe, Y.; Nishimura, T.; Morishita, S.; Gotoh, N.; Hirao, A.; Hanayama, R.; Mukaida, N. Cytoplasmic DNA
accumulation preferentially triggers cell death of myeloid leukemia cells by interacting with intracellular DNA sensing pathway.
Cell Death Dis. 2021,12, 322. [CrossRef] [PubMed]
45.
Andreeva, L.; Hiller, B.; Kostrewa, D.; Lassig, C.; de Oliveira Mann, C.C.; Jan Drexler, D.; Maiser, A.; Gaidt, M.; Leonhardt, H.;
Hornung, V.; et al. cGAS senses long and HMGB/TFAM-bound U-turn DNA by forming protein-DNA ladders. Nature
2017
,549,
394–398. [CrossRef]
46.
Zhang, X.; Bai, X.C.; Chen, Z.J. Structures and Mechanisms in the cGAS-STING Innate Immunity Pathway. Immunity
2020
,53,
43–53. [CrossRef] [PubMed]
47. Noda, N.N.; Wang, Z.; Zhang, H. Liquid-liquid phase separation in autophagy. J. Cell Biol. 2020,219, e202004062. [CrossRef]
48.
Sun, D.; Wu, R.; Li, P.; Yu, L. Phase Separation in Regulation of Aggrephagy. J. Mol. Biol.
2020
,432, 160–169. [CrossRef] [PubMed]
49. Fujioka, Y.; Noda, N.N. Biomolecular condensates in autophagy regulation. Curr. Opin. Cell Biol. 2021,69, 23–29. [CrossRef]
50.
Hyttinen, J.M.; Amadio, M.; Viiri, J.; Pascale, A.; Salminen, A.; Kaarniranta, K. Clearance of misfolded and aggregated proteins by
aggrephagy and implications for aggregation diseases. Ageing Res. Rev. 2014,18, 16–28. [CrossRef]
51.
Dobbs, N.; Burnaevskiy, N.; Chen, D.; Gonugunta, V.K.; Alto, N.M.; Yan, N. STING Activation by Translocation from the ER Is
Associated with Infection and Autoinflammatory Disease. Cell Host Microbe 2015,18, 157–168. [CrossRef]
52.
Ge, L.; Melville, D.; Zhang, M.; Schekman, R. The ER-Golgi intermediate compartment is a key membrane source for the LC3
lipidation step of autophagosome biogenesis. eLife 2013,2, e00947. [CrossRef]
53.
Wu, J.; Chen, Y.J.; Dobbs, N.; Sakai, T.; Liou, J.; Miner, J.J.; Yan, N. STING-mediated disruption of calcium homeostasis chronically
activates ER stress and primes T cell death. J. Exp. Med. 2019,216, 867–883. [CrossRef]
54.
Zhang, L.; Wang, A. Virus-induced ER stress and the unfolded protein response. Front. Plant. Sci.
2012
,3, 293. [CrossRef]
[PubMed]
55.
Karna, K.K.; Choi, N.Y.; Kim, C.Y.; Kim, H.K.; Shin, Y.S.; Park, J.K. Gui-A-Gra Attenuates Testicular Dysfunction in Varicocele-
Induced Rats via Oxidative Stress, ER Stress and Mitochondrial Apoptosis Pathway. Int. J. Mol. Sci.
2020
,21, 9231. [CrossRef]
[PubMed]
56.
Bhardwaj, M.; Leli, N.M.; Koumenis, C.; Amaravadi, R.K. Regulation of autophagy by canonical and non-canonical ER stress
responses. Semin. Cancer Biol. 2020,66, 116–128. [CrossRef]
57.
Moretti, J.; Roy, S.; Bozec, D.; Martinez, J.; Chapman, J.R.; Ueberheide, B.; Lamming, D.W.; Chen, Z.J.; Horng, T.; Yeretssian, G.;
et al. STING Senses Microbial Viability to Orchestrate Stress-Mediated Autophagy of the Endoplasmic Reticulum. Cell
2017
,171,
809–823.e13. [CrossRef] [PubMed]
58.
Srikanth, S.; Woo, J.S.; Wu, B.; El-Sherbiny, Y.M.; Leung, J.; Chupradit, K.; Rice, L.; Seo, G.J.; Calmettes, G.; Ramakrishna, C.; et al.
The Ca
2+
sensor STIM1 regulates the type I interferon response by retaining the signaling adaptor STING at the endoplasmic
reticulum. Nat. Immunol. 2019,20, 152–162. [CrossRef]
59.
Zhang, Y.; Chen, W.; Wang, Y. STING is an essential regulator of heart inflammation and fibrosis in mice with pathological cardiac
hypertrophy via endoplasmic reticulum (ER) stress. Biomed. Pharmacother. 2020,125, 110022. [CrossRef]
60.
Yurube, T.; Ito, M.; Kakiuchi, Y.; Kuroda, R.; Kakutani, K. Autophagy and mTOR signaling during intervertebral disc aging and
degeneration. JOR Spine 2020,3, e1082. [CrossRef] [PubMed]
61.
Populo, H.; Lopes, J.M.; Soares, P. The mTOR signalling pathway in human cancer. Int. J. Mol. Sci.
2012
,13, 1886–1918. [CrossRef]
62.
Aspernig, H.; Heimbucher, T.; Qi, W.; Gangurde, D.; Curic, S.; Yan, Y.; von Gromoff, E.D.; Baumeister, R.; Thien, A. Mitochondrial
Perturbations Couple mTORC2 to Autophagy in C. elegans. Cell Rep. 2019,29, 1399–1409.e5. [CrossRef] [PubMed]
63.
Qin, L.; Wang, Z.; Tao, L.; Wang, Y. ER stress negatively regulates AKT/TSC/mTOR pathway to enhance autophagy. Autophagy
2010,6, 239–247. [CrossRef]
64.
Cai, H.; Imler, J.L. cGAS-STING: Insight on the evolution of a primordial antiviral signaling cassette. Fac. Rev.
2021
,10, 54.
[CrossRef]
65.
Prabakaran, T.; Bodda, C.; Krapp, C.; Zhang, B.C.; Christensen, M.H.; Sun, C.; Reinert, L.; Cai, Y.; Jensen, S.B.; Skouboe, M.K.;
et al. Attenuation of cGAS-STING signaling is mediated by a p62/SQSTM1-dependent autophagy pathway activated by TBK1.
EMBO J. 2018,37, e97858. [CrossRef]
66. Wang, Y.; Qin, Z.H. Coordination of autophagy with other cellular activities. Acta Pharmacol. Sin. 2013,34, 585–594. [CrossRef]
67. Thurston, T.L.; Boyle, K.B.; Allen, M.; Ravenhill, B.J.; Karpiyevich, M.; Bloor, S.; Kaul, A.; Noad, J.; Foeglein, A.; Matthews, S.A.;
et al. Recruitment of TBK1 to cytosol-invading Salmonella induces WIPI2-dependent antibacterial autophagy. EMBO J.
2016
,35,
1779–1792. [CrossRef] [PubMed]
68.
Hyttinen, J.M.; Niittykoski, M.; Salminen, A.; Kaarniranta, K. Maturation of autophagosomes and endosomes: A key role for
Rab7. Biochim. Biophys. Acta 2013,1833, 503–510. [CrossRef]
69.
Oakes, J.A.; Davies, M.C.; Collins, M.O. TBK1: A new player in ALS linking autophagy and neuroinflammation. Mol. Brain
2017
,
10, 5. [CrossRef]
70.
Wu, M.Y.; Lu, J.H. Autophagy and Macrophage Functions: Inflammatory Response and Phagocytosis. Cells
2019
,9, 70. [CrossRef]
Int. J. Mol. Sci. 2021,22, 13232 14 of 14
71.
Gutierrez, M.G.; Master, S.S.; Singh, S.B.; Taylor, G.A.; Colombo, M.I.; Deretic, V. Autophagy is a defense mechanism inhibiting
BCG and Mycobacterium tuberculosis survival in infected macrophages. Cell 2004,119, 753–766. [CrossRef] [PubMed]
72.
Watson, R.O.; Bell, S.L.; MacDuff, D.A.; Kimmey, J.M.; Diner, E.J.; Olivas, J.; Vance, R.E.; Stallings, C.L.; Virgin, H.W.; Cox, J.S. The
Cytosolic Sensor cGAS Detects Mycobacterium tuberculosis DNA to Induce Type I Interferons and Activate Autophagy. Cell Host
Microbe 2015,17, 811–819. [CrossRef] [PubMed]
73.
Abdullah, A.; Mobilio, F.; Crack, P.J.; Taylor, J.M. STING-Mediated Autophagy Is Protective against H
2
O
2
-Induced Cell Death.
Int. J. Mol. Sci. 2020,21, 7059. [CrossRef]
74. Moretti, J.; Blander, J.M. Detection of a vita-PAMP STINGs cells into reticulophagy. Autophagy 2018,14, 1102–1104. [CrossRef]
75.
Basit, A.; Cho, M.G.; Kim, E.Y.; Kwon, D.; Kang, S.J.; Lee, J.H. The cGAS/STING/TBK1/IRF3 innate immunity pathway maintains
chromosomal stability through regulation of p21 levels. Exp. Mol. Med. 2020,52, 643–657. [CrossRef]
76.
Silva, V.R.; Neves, S.P.; Santos, L.S.; Dias, R.B.; Bezerra, D.P. Challenges and Therapeutic Opportunities of Autophagy in Cancer
Therapy. Cancers 2020,12, 3461. [CrossRef] [PubMed]
77.
Froechlich, G.; Caiazza, C.; Gentile, C.; D’Alise, A.M.; De Lucia, M.; Langone, F.; Leoni, G.; Cotugno, G.; Scisciola, V.; Nicosia, A.;
et al. Integrity of the Antiviral STING-mediated DNA Sensing in Tumor Cells Is Required to Sustain the Immunotherapeutic
Efficacy of Herpes Simplex Oncolytic Virus. Cancers 2020,12, 3407. [CrossRef] [PubMed]
78.
Li, C.; Zhang, Y.; Liu, J.; Kang, R.; Klionsky, D.J.; Tang, D. Mitochondrial DNA stress triggers autophagy-dependent ferroptotic
death. Autophagy 2021,17, 948–960. [CrossRef]
79.
Kuang, F.; Liu, J.; Li, C.; Kang, R.; Tang, D. Cathepsin B is a mediator of organelle-specific initiation of ferroptosis. Biochem.
Biophys. Res. Commun. 2020,533, 1464–1469. [CrossRef]
80.
Guo, Q.; Chen, X.; Chen, J.; Zheng, G.; Xie, C.; Wu, H.; Miao, Z.; Lin, Y.; Wang, X.; Gao, W.; et al. STING promotes senescence,
apoptosis, and extracellular matrix degradation in osteoarthritis via the NF-kappaB signaling pathway. Cell Death Dis.
2021
,
12, 13. [CrossRef]
81.
Everett, H.; McFadden, G. Apoptosis: An innate immune response to virus infection. Trends Microbiol.
1999
,7, 160–165. [CrossRef]
... In addition to IRF3-and NF-κB-mediated gene transcriptions, STING also triggers cell autophagy and apoptosis [12,13]. Recent studies have shown that STING can cause canonical autophagy and non-canonical autophagy [12]. ...
... In addition to IRF3-and NF-κB-mediated gene transcriptions, STING also triggers cell autophagy and apoptosis [12,13]. Recent studies have shown that STING can cause canonical autophagy and non-canonical autophagy [12]. STING was shown to induce endoplasmic reticulum (ER) stress and unfolded protein response (UPR) through a unique UPR motif in the cyclic dinucleotide binding (CBD) domain, which negatively regulates the AKT/TSC/mTOR pathway to enhance canonical autophagy [14]. ...
... In this study, we first demonstrated the IFN-independent antiviral activity of chSTING in both mammalian and chicken cells (Figures 1-3). It is known that STING can not only induce IFN, but also activate NF-κB, autophagy and apoptosis [12,13,32]. First, chSTING pLxVS sub S366A and chSTING ∆CTT have differential NF-κB signaling ( Figure 2C), but the antiviral activity of these two chSTING mutants are similar ( Figure 2F,G). ...
Article
Full-text available
It has been recently recognized that the DNA sensing innate immune cGAS-STING pathway exerts an IFN-independent antiviral function; however, whether and how chicken STING (chSTING) exerts such an IFN-independent antiviral activity is still unknown. Here, we showed that chSTING exerts an antiviral activity in HEK293 cells and chicken cells, independent of IFN production. chSTING was able to trigger cell apoptosis and autophagy independently of IFN, and the apoptosis inhibitors, rather than autophagy inhibitors, could antagonize the antiviral function of chSTING, suggesting the involvement of apoptosis in IFN-independent antiviral function. In addition, chSTING lost its antiviral function in IRF7-knockout chicken macrophages, indicating that IRF7 is not only essential for the production of IFN, but also participates in the other activities of chSTING, such as the apoptosis. Collectively, our results showed that chSTING exerts an antiviral function independent of IFN, likely via apoptosis.
... In addition to IRF3 and NF-κB mediated gene transcriptions, STING also triggers cell autophagy and apoptosis [12,13]. Recent studies have shown that STING can cause canonical autophagy and noncanonical autophagy [12]. ...
... In addition to IRF3 and NF-κB mediated gene transcriptions, STING also triggers cell autophagy and apoptosis [12,13]. Recent studies have shown that STING can cause canonical autophagy and noncanonical autophagy [12]. STING was shown to induce endoplasmic reticulum (ER) stress and unfolded protein response (UPR) through a unique UPR motif in the cyclic dinucleotide binding (CBD) domain, which negatively regulates AKT/TSC/mTOR pathway to enhance canonical autophagy [14]. ...
... Despite that the IFN independent antiviral activity of STING has been appreciated [23,30,31], in this study, we rst demonstrated the IFN-independent antiviral activity of chSTING in both mammalian and chicken cells (Fig. 1-3). It is known that STING can not only induce IFN, but also activate NF-κB, autophagy and apoptosis [12,13,32]. First, the NF-κB signaling of chSTING pLxVS sub S366A and chSTING △CTT increased and decreased respectively, but the antiviral activity of these two chSTING mutants did not change accordingly (Fig. 2C). ...
Preprint
Full-text available
It has been recently recognized that the DNA sensing innate immune cGAS-STING pathway exerts an IFN-independent antiviral function; however, whether and how chicken STING (chSTING) exerts such an IFN-independent antiviral activity is still unknown. Here, we showed that chSTING exerts an antiviral activity in both mammalian cells and chicken cells, either with or without the production of IFN. On the other hand, chSTING was able to trigger cell apoptosis and autophagy independently of IFN, and the apoptosis inhibitors instead of autophagy inhibitors could antagonize the antiviral function of chSTING, suggesting the involvement of apoptosis in the IFN independent antiviral function. In addition, chSTING lost its antiviral function in IRF7 knockout chicken macrophages, indicating that IRF7 is not only essential for the production of IFN, but also participates in the other activities of chSTING, such as the apoptosis. Collectively, our results showed that chSTING exerts the antiviral function independently of IFN, likely via the apoptosis.
... The cGAS-STING pathway has garnered attention for its role in sensing cellular stress and triggering downstream inflammatory responses. Additionally, this pathway can intersect with autophagy [17,18], thereby influencing cellular fate during ischemiareperfusion (IR) injury. Despite these intriguing insights, the precise mechanistic underpinnings and functional consequences of the cGAS-STING pathway in modulating NCOA4-mediated ferritinophagy, iron homeostasis, and oxidative stress within the framework of cerebral IR remain incompletely understood. ...
Article
Full-text available
Ischemic stroke presents a global health challenge, necessitating an in-depth comprehension of its pathophysiology and therapeutic strategies. While reperfusion therapy salvages brain tissue, it also triggers detrimental cerebral ischemia-reperfusion injury (CIRI). In our investigation, we observed the activation of nuclear receptor coactivator 4 (NCOA4)-mediated ferritinophagy in an oxygen-glucose deprivation/reoxygenation (OGD/R) model using HT22 cells (P < 0.05). This activation contributed to oxidative stress (P < 0.05), enhanced autophagy (P < 0.05) and cell death (P < 0.05) during CIRI. Silencing NCOA4 effectively mitigated OGD/R-induced damage (P < 0.05). These findings suggested that targeting NCOA4-mediated ferritinophagy held promise for preventing and treating CIRI. Subsequently, we substantiated the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway effectively regulated the NCOA4-mediated ferritinophagy, by applying the cGAS inhibitor RU.521 and performing NCOA4 overexpression (P < 0.05). Suppressing the cGAS-STING pathway efficiently curtailed ferritinophagy (P < 0.05), oxidative stress (P < 0.05), and cell damage (P < 0.05) of CIRI, while NCOA4 overexpression could alleviate this effect (P < 0.05). Finally, we elucidated the specific molecular mechanism underlying the protective effect of the iron chelator deferoxamine (DFO) on CIRI. Our findings revealed that DFO alleviated hypoxia-reoxygenation injury in HT22 cells through inhibiting NCOA4-mediated ferritinophagy and reducing ferrous ion levels (P < 0.05). However, the protective effects of DFO were counteracted by cGAS overexpression (P < 0.05). In summary, our results indicated that the activation of the cGAS-STING pathway intensified cerebral damage during CIRI by inducing NCOA4-mediated ferritinophagy. Administering the iron chelator DFO effectively attenuated NCOA4-induced ferritinophagy, thereby alleviating CIRI. Nevertheless, the role of the cGAS-STING pathway in CIRI regulation likely involves intricate mechanisms, necessitating further validation in subsequent investigations.
... VRK-IN-1 is a potent and selective small-molecule inhibitor for VRK1 [25]. We next sought to explore whether and self-DNA [27]. Numerous studies show that overexpression of each component of the cGAS-STING signaling can upregulate the cGAS-STING-IFN-I signaling and subsequently induce IFNB expression [9]. ...
Article
Full-text available
Background Type I interferons (IFNs) are an essential class of cytokines with antitumor, antiviral and immunoregulatory effects. However, over-productive the type I IFNs are tightly associated with autoimmune disorders. Thus, the induction of type I interferons is precisely regulated to maintain immune hemostasis. This study aimed to identify a novel regulator of type I interferon signaling. Methods and results Primary BMDMs, isolated from mice, and human cell lines (HEK293 cells, Hela cells) and murine cell line (MEF cells) were cultured to generate in vitro models. After knockdown VRK1, real-time PCR and dual-luciferase reporter assay were performed to determine the expression level of the type I IFNs and ISGs following HTDNA and Poly (dA:dT) stimulation. Additionally, cells were treated with the VRK1 inhibitor, and the impact of VRK1 inhibition was detected. Upon HTDNA and Poly (dA:dT) stimulation, knockdown of VRK1 attenuated the induction of the type I IFNs and ISGs. Consistently, VRK-IN-1, a potent and selective VRK1 inhibitor, significantly suppressed the induction of the type I IFNs and ISGs in human and murine cell lines. Further, VRK-IN-1 inhibited induction of the type I IFNs in mouse primary BMDMs. Intriguingly, VRK1 potentiated the cGAS-STING- IFN-I axis response at STING level. Conclusions Our study reveals a novel function of VRK1 in regulating the production of type I IFNs. VRK-IN-1 might be a potential lead compound for suppressing aberrant type I IFNs in autoimmune disorders.
... Although the cGAS-STING signaling pathway plays a very important role in the treatment of innate immune and inflammatory diseases, inappropriate activation of this pathway can also trigger autoimmune and inflammatory diseases such as familial frostbite lupus erythematosus, neuroinflammatory and neurodegenerative diseases, and Parkinson's disease (Zheng et al. 2021). Available studies have shown that the cGAS-STING pathway triggers an autoinflammatory response in threeprime repair exonuclease 1 (Trex1) -deficient mice, with lethality occurring at around week eight (Maltbaek et al. 2022). ...
Article
Full-text available
Background The cGAS-STING signaling pathway is an essential section of the natural immune system. In recent years, an increasing number of studies have shown a strong link between abnormal activation of the cGAS-STING signaling pathway, a natural immune pathway mediated by the nucleic acid receptor cGAS, and the development and progression of autoimmune diseases. Therefore, it is important to identify an effective compound to specifically downregulate this pathway for disease. Methods The effect of Glabridin (Glab) was investigated in BMDMs and Peripheral blood mononuclear cell (PBMC) by establishing an in vitro model of cGAS-STING signaling pathway activation. An activation model stimulated by DMXAA was also established in mice to study the effect of Glab. On the other hand, we investigated the possible mechanism of action of Glab and the effect of Glab on Trex1-deficient mice. Results In this research, we report that Glab, a major component of licorice, specifically inhibits the cGAS-STING signaling pathway by inhibiting the level of type I interferon and inflammatory cytokines (IL-6 and TNF-α). In addition, Glab has a therapeutic effect on innate immune diseases caused by abnormal cytoplasmic DNA in Trex1-deficient mice. Mechanistically, Glab can specifically inhibit the interaction of STING with IRF3. Conclusion Glab is a specific inhibitor of the cGAS-STING signaling pathway and may be used in the clinical therapy of cGAS-STING pathway-mediated autoimmune diseases.
Article
STING1 (stimulator of interferon response cGAMP interactor 1) is the quintessential protein in the CGAS-STING1 signaling pathway, crucial for the induction of type I IFN (interferon) production and eliciting innate immunity. Nevertheless, the overactivation or sustained activation of STING1 has been closely associated with the onset of autoimmune disorders. Notably, the majority of these disorders manifest as an upregulated expression of type I interferons and IFN-stimulated genes (ISGs). Hence, strict regulation of STING1 activity is paramount to preserve immune homeostasis. Here, we reported that CSNK1A1/CK1α, a serine/threonine protein kinase, was essential to prevent the overactivation of STING1-mediated type I IFN signaling through autophagic degradation of STING1. Mechanistically, CSNK1A1 interacted with STING1 upon the CGAS-STING1 pathway activation and promoted STING1 autophagic degradation by enhancing the phosphorylation of SQSTM1/p62 at serine 351 (serine 349 in human), which was critical for SQSTM1-mediated STING1 autophagic degradation. Consistently, SSTC3, a selective CSNK1A1 agonist, significantly attenuated the response of the CGAS-STING1 signaling by promoting STING1 autophagic degradation. Importantly, pharmacological activation of CSNK1A1 using SSTC3 markedly repressed the systemic autoinflammatory responses in the trex1-/- mouse autoimmune disease model and effectively suppressed the production of IFNs and ISGs in the PBMCs of SLE patients. Taken together, our study reveals a novel regulatory role of CSNK1A1 in the autophagic degradation of STING1 to maintain immune homeostasis. Manipulating CSNK1A1 through SSTC3 might be a potential therapeutic strategy for alleviating STING1-mediated aberrant type I IFNs in autoimmune diseases.Abbreviations: BMDMs: bone marrow-derived macrophages; cGAMP: cyclic GMP-AMP; CGAS: cyclic GMP-AMP synthase; HTDNA: herring testes DNA; IFIT1: interferon induced protein with tetratricopeptide repeats 1; IFNA4: interferon alpha 4; IFNB: interferon beta; IRF3: interferon regulatory factor 3; ISD: interferon stimulatory DNA; ISGs: IFN-stimulated genes; MEFs: mouse embryonic fibroblasts; PBMCs: peripheral blood mononuclear cells; RSAD2: radical S-adenosyl methionine domain containing 2; SLE: systemic lupus erythematosus; STING1: stimulator of interferon response cGAMP interactor 1; TBK1: TANK binding kinase 1.
Article
Full-text available
As an essential micronutrient, manganese plays an important role in the physiological process and immune process. In recent decades, cGAS-STING pathway, which can congenitally recognize exogenous and endogenous DNA for activation, has been widely reported to play critical roles in the innate immunity against some important diseases, such as infections and tumor. Manganese ion (Mn²⁺) has been recently proved to specifically bind with cGAS and activate cGAS-STING pathway as a potential cGAS agonist, however, is significantly restricted by the low stability of Mn²⁺ for further medical application. As one of the most stable forms of manganese, manganese dioxide (MnO2) nanomaterials have been reported to show multiple promising functions, such as drug delivery, anti-tumor and anti-infection activities. More importantly, MnO2 nanomaterials are also found to be a potential candidate as cGAS agonist by transforming into Mn²⁺, which indicates their potential for cGAS-STING regulations in different diseased conditions. In this review, we introduced the methods for the preparation of MnO2 nanomaterials as well as their biological activities. Moreover, we emphatically introduced the cGAS-STING pathway and discussed the detailed mechanisms of MnO2 nanomaterials for cGAS activation by converting into Mn²⁺. And we also discussed the application of MnO2 nanomaterials for disease treatment by regulating cGAS-STING pathway, which might benefit the future development of novel cGAS-STING targeted treatments based on MnO2 nanoplatforms.
Article
Full-text available
Autophagy plays an important role in host antiviral defense. The avian leukosis virus subgroup J (ALV-J) has been shown to inhibit autophagy while promoting viral replication. The underlying autophagic mechanisms, however, are unknown. Cholesterol 25-hydroxylase (CH25H) is a conserved interferon-stimulated gene, which converts cholesterol to a soluble antiviral factor, 25-hydroxycholesterol (25HC). In this study, we further investigated the autophagic mechanism of CH25H resistance to ALV-J in chicken embryonic fibroblast cell lines (DF1). Our results found that overexpression of CH25H and treatment with 25HC promoted the autophagic markers microtubule-associated protein 1 light chain 3 II (LC3II) and autophagy-related gene 5(ATG5), while decreased autophagy substrate p62/SQSTM1 (p62) expression in ALV-J infection DF-1 cells. Induction of cellular autophagy also reduces the levels of ALV-J gp85 and p27. ALV-J infection, on the other hand, suppresses autophagic marker protein LC3II expression. These findings suggest that CH25H-induced autophagy is a host defense mechanism that aids in ALV-J replication inhibition. In particular, CH25H interacts with CHMP4B and inhibits ALV-J infection in DF-1 cells by promoting autophagy, revealing a novel mechanism by which CH25H inhibits ALV-J infection. Although the underlying mechanisms are not completely understood, CH25H and 25HC are the first to show inhibiting ALV-J infection via autophagy.
Article
Full-text available
Gain of even a single chromosome leads to changes in human cell physiology and uniform perturbations of specific cellular processes, including downregulation of DNA replication pathway, upregulation of autophagy and lysosomal degradation, and constitutive activation of the type I interferon response. Little is known about the molecular mechanisms underlying these changes. We show that the constitutive nuclear localization of TFEB, a transcription factor that activates the expression of autophagy and lysosomal genes, is characteristic of human trisomic cells. Constitutive nuclear localization of TFEB in trisomic cells is independent of mTORC1 signaling, but depends on the cGAS-STING activation. Trisomic cells accumulate cytoplasmic dsDNA, which activates the cGAS-STING signaling cascade, thereby triggering nuclear accumulation of the transcription factor IRF3 and, consequently, upregulation of interferon-stimulated genes. cGAS depletion interferes with TFEB-dependent upregulation of autophagy in model trisomic cells. Importantly, activation of both the innate immune response and autophagy occurs also in primary trisomic embryonic fibroblasts, independent of the identity of the additional chromosome. Our research identifies the cGAS-STING pathway as an upstream regulator responsible for activation of autophagy and inflammatory response in human cells with extra chromosomes, such as in Down syndrome or other aneuploidy-associated pathologies.
Article
Full-text available
Stimulator of interferon genes (STING) functions in the cytosolic DNA-sensing pathway of innate immunity in mammals. It is activated upon binding the cyclic dinucleotide 2'3'-cGAMP, a second messenger produced by the enzyme cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS), which acts as the receptor for DNA in this pathway, and triggers the expression of interferons and other viral stress-induced genes. The ancient origin of STING in the evolution of animals had been noted, but its primitive function was speculative. We review here recent advances in the remarkable history of cGAS-STING signaling, which establish that cGAS is a member of the family of cGAS/DncV-like nucleotidyltransferases (CD-NTases). In bacteria, CD-NTases synthesize a wide range of cyclic oligonucleotide second messengers in response to bacteriophage infections, which in turn activate a variety of effector proteins to abort phage infection. Among these effectors, some are related to STING, revealing an ancestral function for the cGAS-STING cassette in antiviral host defense. Study of STING signaling in invertebrate animals is consistent with an early acquisition in the history of metazoans of CD-NTase- and STING-encoding genes to counter the universal threat of viruses. In particular, STING-dependent immunity appears to play a previously unsuspected important role in some insects. These discoveries open up interesting perspectives for the use of model organisms to decipher emerging aspects of cGAS-STING biology in mammals, such as the activation of interferon-independent responses or the function and regulation of cGAS in the nucleus.
Article
Full-text available
Cell death and immune response are at the core of life. In past decades, the endoplasmic reticulum (ER) protein STING1 (also known as STING or TMEM173) was found to play a fundamental role in the production of type I interferons (IFNs) and pro-inflammatory cytokines in response to DNA derived from invading microbial pathogens or damaged hosts by activating multiple transcription factors. In addition to this well-known function in infection, inflammation, and immunity, emerging evidence suggests that the STING1-dependent signaling network is implicated in health and disease by regulating autophagic degradation or various cell death modalities (e.g., apoptosis, necroptosis, pyroptosis, ferroptosis, mitotic cell death, and immunogenic cell death [ICD]). Here, we outline the latest advances in our understanding of the regulating mechanisms and signaling pathways of STING1 in autophagy and cell death, which may shed light on new targets for therapeutic interventions.
Article
Full-text available
Significance The cGAS-STING pathway is important for immune defense against infection and cancer. STING activation triggers multiple signaling cascades leading to activation of IRF3, NF-κB, and autophagy. By generating mice harboring mutations of STING that specifically inactivate different signaling cascades, we found that ablation of IRF3 activation, which is essential for the induction of type I interferons, was not sufficient to abolish the immune defense against virus infection and cancer in mouse models. Rather, impairing the ability of STING to recruit TBK1, which is important for activating both IRF3 and NF-κB, abolished the immune defense functions of STING. These results demonstrate that the recruitment of TBK1 to STING has functions that are broader than activating IRF3 and inducing type I interferons.
Article
Full-text available
Accumulating evidence indicates the presence of cytoplasmic DNAs in various types of malignant cells, and its involvement in anti-cancer drug- or radiotherapy-mediated DNA damage response and replication stress. However, the pathophysiological roles of cytoplasmic DNAs in leukemias remain largely unknown. We observed that during hematopoietic stem cell transplantation (HSCT) in mouse myeloid leukemia models, double-stranded (ds)DNAs were constitutively secreted in the form of extracellular vesicles (EVs) from myeloid leukemia cells and were transferred to the donor cells to dampen their hematopoietic capabilities. Subsequent analysis of cytoplasmic DNA dynamics in leukemia cells revealed that autophagy regulated cytoplasmic dsDNA accumulation and subsequent redistribution into EVs. Moreover, accumulated cytoplasmic dsDNAs activated STING pathway, thereby reducing leukemia cell viability through reactive oxygen species (ROS) generation. Pharmaceutical inhibition of autophagosome formation induced cytoplasmic DNA accumulation, eventually triggering cytoplasmic DNA sensing pathways to exert cytotoxicity, preferentially in leukemia cells. Thus, manipulation of cytoplasmic dsDNA dynamics can be a novel and potent therapeutic strategy for myeloid leukemias.
Article
Full-text available
Aluminum-containing adjuvants have been used for nearly 100 years to enhance immune responses in billions of doses of vaccines. To date, only a few adjuvants have been approved for use in humans, among which aluminum-containing adjuvants are the only ones widely used. However, the medical need for potent and safe adjuvants is currently continuously increasing, especially those triggering cellular immune responses for cytotoxic T lymphocyte activation, which are urgently needed for the development of efficient virus and cancer vaccines. Manganese is an essential micronutrient required for diverse biological activities, but its functions in immunity remain undefined. We previously reported that Mn ²⁺ is important in the host defense against cytosolic dsDNA by facilitating cGAS-STING activation and that Mn ²⁺ alone directly activates cGAS independent of dsDNA, leading to an unconventional catalytic synthesis of 2′3′-cGAMP. Herein, we found that Mn ²⁺ strongly promoted immune responses by facilitating antigen uptake, presentation, and germinal center formation via both cGAS-STING and NLRP3 activation. Accordingly, a colloidal manganese salt (Mn jelly, MnJ) was formulated to act not only as an immune potentiator but also as a delivery system to stimulate humoral and cellular immune responses, inducing antibody production and CD4 ⁺ /CD8 ⁺ T-cell proliferation and activation by either intramuscular or intranasal immunization. When administered intranasally, MnJ also worked as a mucosal adjuvant, inducing high levels of secretory IgA. MnJ showed good adjuvant effects for all tested antigens, including T cell-dependent and T cell-independent antigens, such as bacterial capsular polysaccharides, thus indicating that it is a promising adjuvant candidate.
Article
Full-text available
Micronuclei are constantly considered as a marker of genome instability and very recently found to be a trigger of innate immune responses. An increased frequency of micronuclei is associated with many diseases, but the mechanism underlying the regulation of micronuclei homeostasis remains largely unknown. Here, we report that CGAS (cyclic GMP-AMP synthase), a known regulator of DNA sensing and DNA repair, reduces the abundance of micronuclei under genotoxic stress in an autophagy-dependent manner. CGAS accumulates in the autophagic machinery and directly interacts with MAP1LC3B/LC3B in a manner dependent upon its MAP1LC3-interacting region (LIR). Importantly, the interaction is essential for MAP1LC3 recruitment to micronuclei and subsequent clearance of micronuclei via autophagy (micronucleophagy) in response to genotoxic stress. Moreover, in contrast to its DNA sensing function to activate micronuclei-driven inflammation, CGAS-mediated micronucleophagy blunts the production of cyclic GMP-AMP (cGAMP) induced by genotoxic stress. We therefore conclude that CGAS is a receptor for the selective autophagic clearance of micronuclei and uncovered an unprecedented role of CGAS in micronuclei homeostasis to dampen innate immune surveillance.
Article
Full-text available
Autophagy is an intracellular degradation system that contributes to cellular homeostasis. Autophagosome formation is a landmark event in autophagy, which sequesters and delivers cytoplasmic components to the lysosome for degradation. Based on selectivity, autophagy can be classified into bulk and selective autophagy, which are mechanistically distinct from each other, especially in the requirement of cargos for autophagosome formation. Recent studies revealed that liquid-like biomolecular condensates, which are formed through liquid–liquid phase separation, regulate the autophagosome formation of both bulk and selective autophagy. Here, we focus on recent findings on the involvement of biomolecular condensates in autophagy regulation and discuss their significance.
Article
Full-text available
Damaged deoxyribonucleic acid (DNA) is a primary pathologic factor for osteoarthritis (OA); however, the mechanism by which DNA damage drives OA is unclear. Previous research demonstrated that the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) participates in DNA damage response. As a result, the current study aimed at exploring the role STING, which is the major effector in the cGAS-STING signaling casacde, in OA progress in vitro, as well as in vivo. In this study, the expression of STING was evaluated in the human and mouse OA tissues, and in chondrocytes exposed to interleukin-1 beta (IL-1β). The influences of STING on the metabolism of the extracellular matrix (ECM), apoptosis, and senescence, were assessed in STING overexpressing and knocking-down chondrocytes. Moreover, the NF-κB-signaling casacde and its role in the regulatory effects of STING on ECM metabolism, apoptosis, and senescence were explored. The STING knockdown lentivirus was intra-articularly injected to evaluate its therapeutic impact on OA in mice in vivo. The results showed that the expression of STING was remarkably elevated in the human and mouse OA tissues and in chondrocytes exposed to IL-1β. Overexpression of STING promoted the expression of MMP13, as well as ADAMTS5, but suppressed the expression of Aggrecan, as well as Collagen II; it also enhanced apoptosis and senescence in chondrocytes exposed to and those untreated with IL-1β. The mechanistic study showed that STING activated NF-κB signaling cascade, whereas the blockage of NF-κB signaling attenuated STING-induced apoptosis and senescence, and ameliorated STING-induced ECM metabolism imbalance. In in vivo study, it was demonstrated that STING knockdown alleviated destabilization of the medial meniscus-induced OA development in mice. In conclusion, STING promotes OA by activating the NF-κB signaling cascade, whereas suppression of STING may provide a novel approach for OA therapy.
Article
The concept of gap junctions and their role in intercellular communication has been known for around 50 years. Considerable progress has been made in understanding the fundamental biology of connexins in mediating gap junction intercellular communication (GJIC) and their role in various cellular processes including pathological conditions. However, this understanding has not led to development of advanced therapeutics utilizing GJIC. Inadequacies in strategies that target specific connexin protein in the affected tissue, with minimal or no collateral damage, are the primary reason for the lack of development of efficient therapeutic models. Herein, nanotechnology has a role to play, giving plenty of scope to circumvent these problems and develop more efficient connexin based therapeutics. In this review, we discuss briefly the role of connexins and gap junctions in various physiological and pathological processes, with special emphasis on cancer. We further discuss the application of nanotechnology and tissue engineering in developing treatments for various connexin based disorders.