ArticlePDF AvailableLiterature Review

Abstract and Figures

Cytosolic nucleic acid sensors have a critical role in detecting endogenous nucleic acids to initiate innate immune responses during microbial infections and/or cell death. Several seminal studies over the past decade have delineated the conserved mechanism of cytosolic DNA sensor cyclic GMP-AMP synthase (cGAS) and the downstream signaling adapter stimulator of interferon genes (STING) in mediating innate immune signaling pathways as a host defense mechanism. Besides the predominant role in microbial infections and inflammatory diseases, there is an increased attention on alternative functional responses of cGAS-STING-mediated signaling. Here we review the complexity of interactions between the cGAS-STING signaling and cell death pathways. A better understanding of molecular mechanisms of this interplay is important with regard to the development of new therapeutics targeting cGAS-STING signaling in cancer, infectious, and chronic inflammatory diseases.
Content may be subject to copyright.
Cell Death & Differentiation
https://doi.org/10.1038/s41418-020-00624-8
REVIEW ARTICLE
Crosstalk between cGASSTING signaling and cell death
Ambika M. V. Murthy 1Nirmal Robinson 1Sharad Kumar 1
Received: 2 July 2020 / Accepted: 7 September 2020
© The Author(s), under exclusive licence to ADMC Associazione Differenziamento e Morte Cellulare 2020
Abstract
Cytosolic nucleic acid sensors have a critical role in detecting endogenous nucleic acids to initiate innate immune responses
during microbial infections and/or cell death. Several seminal studies over the past decade have delineated the conserved
mechanism of cytosolic DNA sensor cyclic GMP-AMP synthase (cGAS) and the downstream signaling adapter stimulator of
interferon genes (STING) in mediating innate immune signaling pathways as a host defense mechanism. Besides the
predominant role in microbial infections and inammatory diseases, there is an increased attention on alternative functional
responses of cGASSTING-mediated signaling. Here we review the complexity of interactions between the cGASSTING
signaling and cell death pathways. A better understanding of molecular mechanisms of this interplay is important with regard to
the development of new therapeutics targeting cGASSTING signaling in cancer, infectious, and chronic inammatory diseases.
Facts
Cytosolic DNA stimulates various signaling pathways
that often lead to different modes of cell death.
The cGASSTING pathway recognizes cytosolic DNA
to initiate type I interferon production.
Autophagy has a dual role in cGASSTING signaling,
by initiating inammatory responses and by targeting
STING for degradation.
Open questions
What dictates the redundancy in cell death pathways
triggered by the cGASSTING?
What are the mechanisms by which the cGASSTING
induces autophagy-dependent cell death?
Can STING-mediated cell death be targeted for treating
cancer and other diseases?
Introduction
Cell death is indispensable for the development of tissues
and organs, and for eliminating infected, damaged, or
transformed cells to maintain organismal homeostasis. Not
that long-ago cell death was broadly categorized into just
two types, apoptosis (immunologically silent) and necrosis
(accidental or inammatory cell death) [1,2]. Subsequent
studies uncovered various other cell death modalities
including (but not limited to) programmed necrosis, pyr-
optosis, ferroptosis, and autophagy-dependent cell death
(ADCD) [3]. In the last two decades, the ability of DNA to
initiate different types of cell death as a host protective
responsehaveemergedasakeyfeatureofinnateimmu-
nity in mammalian cells. Under normal circumstances
DNA is strictly conned to the nucleus or the mitochon-
drion of a eukaryotic cell. Therefore, the presence of self-
DNA in unusual sites such as cytoplasm or endosomes
and/or the presence of exogeneous DNA from microbial
challenge is considered as a threat. To counteract these
danger signals, cells are equipped with various DNA
sensors, such as toll-like receptor 9 (TLR9), absent
in melanoma 2 (AIM2), cyclic GMP-AMP synthase
(cGAS), RNA polymerase III, DNA-dependent activa-
tors of IRFs, interferon-inducible protein 16, DDX41,
LSm14A [4]. Of these, only TLR9, AIM2, and cGAS are
Edited by G. Melino
*Ambika M. V. Murthy
ambika.mosalevenkateshmurthy@unisa.edu.au
*Nirmal Robinson
nirmal.robinson@unisa.edu.au
*Sharad Kumar
sharad.kumar@unisa.edu.au
1Centre for Cancer Biology, University of South Australia, GPO
Box 2471, Adelaide, SA 5001, Australia
1234567890();,:
1234567890();,:
well characterized, whereas the rest of them are yet to be
validated by genetic approaches [5].
TLR9 was the rst DNA-sensing receptor identied in
immune cells such as dendritic cells (DCs), B cells and
macrophages [6]. Under basal conditions, TLR9 is retained
in the endoplasmic reticulum (ER) [7,8]. However, detec-
tion of unmethylated cytosine-guanine (CpG)-rich motifs
that are relatively abundant in bacteria and DNA viruses,
occurs in early endosomes that activate NF-κB via MyD88
and type I interferon (IFN) production to initiate a cascade
of innate and adaptive immune responses [6,9,10]. Yet,
mitochondrial DNA (mtDNA) can also activate TLR9
marked by p38 mitogen-activated protein kinase activation
in human PMNs and immortalized podocytes [11,12]. This
is unsurprising as mitochondria evolved from bacteria that
are endosymbionts in mammalian cells and therefore,
mtDNA contains non-methylated CpG motifs [13,14].
Another sensor for cytosolic DNA is AIM2 that can bind
to DNA directly in a sequence-independent manner via C-
terminal HIN-200 domain, where an N-terminal pyrin domain
interacts with apoptosis-associated speck-like protein con-
taining a caspase activation and recruitment domain (ASC) to
initiate inammasome formation [15]. AIM2 requires at least
80 bp of dsDNA to initiate inammasome activation [16]
resulting in IL-1βand IL-18 release accompanied by
inammatory cell death termed pyroptosis (discussed below)
to offer protection against cancer and infections caused by
DNA viruses, bacteria, and fungal pathogens [15].
In recent years, cytosolic sensing of mislocalized DNA
by cGAS has emerged as a key event in cellular responses
to pathogen invasion, DNA damage, and mitochondrial
stress. cGAS recognizes DNA of various sizes in a
sequence-independent manner, with human cGAS able to
sense DNA sequences as short as 45 bp [17], however,
larger DNA fragments result in better immune response
[18]. cGAS-mediated stimulator of interferon genes
(STING) activation drives IFN responses and also activates
NF-κB (discussed in detail below). Historically, the nucleus
has been regarded as immune-privileged, however, this
paradigm was overturned by nuclear localization of cGAS
and its association with centromeric DNA in DCs [19], and
also the identication of heterogeneous nuclear ribonu-
cleoprotein A2B1 that can bind viral DNA in the nucleus
during herpes simplex virus-1 infections to drive IFN
responses [20]. Notably, nuclear localized cGAS is 200-fold
less efcient to nuclear DNA than extranuclear cGAS to
cytosolic DNA to induce IFN responses [19], which could
be due to suppression of cyclic guanosine monophosphate-
adenosine monophosphate (cGAMP) production by
nucleosomes [21]. In addition, positioning of cGAS to
plasma membrane could not be discounted [22]. This raises
the question if localization of the cGAS affects intensity of
the IFN responses. However, another persistent question is
why does any cell require multiple DNA sensors? One
possibility is that these sensors act as a safety checkpoints to
ensure any danger signal in the form of cytoplasmic DNA is
not missed in different cellular contexts. The other possi-
bility is that if one DNA-sensing pathway becomes non-
functional and/or inhibited by microbial or danger signals,
the other DNA sensors could act as a backup. Although,
mammalian cells are equipped with an array of DNA sen-
sors, cGASSTING-mediated IFN response is gaining
increasing attention due its critical role in inammatory and
infectious diseases, and cancer [23,24]. This is possibly due
to widespread STING expression in various cell-types and
the ability of cGASSTING to regulate different pro-
grammed cell death pathways. In this review we discuss our
current understanding of the interplay between the
cGASSTING signaling and different modes of cell death
such as apoptosis, necroptosis, and pyroptosis. Finally, we
summarize the new work linking the cGASSTING path-
way to autophagy and ADCD.
The cGASSTING signaling pathway
cGAS recognizes viral, bacterial, protozoal, mitochondrial,
and self-DNA from tumor or dead cells in the cytosol to
activate a cascade of signaling pathways leading to inam-
mation [2528](Fig.1). Binding of DNA to the cGAS indu-
ces conformational changes to cGAS, which in turn catalyzes
the formation of 2,3-cGAMP, a cyclic di-nucleotide (CDN)
with a unique phosphodiester linkage that uses adenosine
triphosphate (ATP) and guanosine triphosphate (GTP) [29].
DNA binding to cGAS induces liquidlike droplets that act as
a microreactor, enriching enzyme, and reactants to enhance
cGAMP production, as demonstrated by cGAS-DNA puncta
in both human broblast cell line and mouse embryonic
broblasts (MEF) [30]. The generated cGAMP acts as a
secondary messenger and activates STING (also known as
transmembrane protein 173 (TMEM173), N-terminal
methionineprolinetyrosineserine plasma membrane trans-
panner (MPYS), mediator of interferon regulatory factor 3
(IRF3) activation, and endoplasmic reticulum IFN stimulator)
[31]. Notably, cGAMP can transactivate STING in neigh-
boring cells [32] with the aid of gap junction proteins such as
connexins to amplify inammatory responses [33]. In addi-
tion, bacterial CDNs such as cGAMP, c-di-AMP and c-di-
GMP can also activate STING directly [3436].
In resting state, STING is contained in the ER as a
transmembrane homodimer by a Ca2+sensing transmem-
brane protein, stromal interaction molecule 1 (STIM1) [37].
Upon activation, STING undergoes a conformational
change and translocates to the perinuclear region via an
unknown mechanism, where it oligomerizes and recruit
tank-binding kinase 1 (TBK1) [38]. Oligomerized STING
A. M. V. Murthy et al.
offers a platform for trans-TBK1 autophosphorylation, and
in turn gets phosphorylated by TBK1 as delineated by
structural studies using human TBK1 complex with
cGAMP and full-length chicken STING [38]. STING can
interact with both TBK1 and the transcription factor IRF3,
therefore specifying IRF3 for phosphorylation by TBK1
[39,40]. Phosphorylated IRF3 dimerizes and translocates
into the nucleus to induce the expression of inammatory
genes including IFN and IFN-stimulated genes (ISG). IFNs
are indispensable for antitumor immune responses in DCs
and CD8+T cells, and mediate adaptive immunity [41,42].
In addition, STINGTBK1 association also phosphorylates
IκB kinase, leading to noncanonical activation of NF-κB
that regulates the transcription of inammatory cytokines,
including IL-6 and tumor necrosis factor (TNF) [25,26].
However, TBK1 and IκB kinase are dispensable for
STING-induced NF-κB activation in mouse myeloid cells
both in vivo and in vitro [43]. Notably, STING can be
degraded by autophagy (discussed later in this review) to
tightly regulate the cGASSTING pathway, as aberrant
activation of this pathway causes chronic inammatory
diseases such as systemic lupus erythematosus (SLE) [28].
However, IFN-independent functions of STING in antiviral
responses and T cell death to control immune evasion
cannot be discounted [44].
Phylogenetic analysis has revealed that both cGAS and
STING homologs are present in mammals and the origin of
this pathway has been traced back to a unicellular eukaryote
Fig. 1 Activation of the
cGASSTING signaling
pathway to drive innate
immune responses. a cGAS is
activated by mislocalized DNA
in the cytosol by various sources
such as self-DNA from damaged
mitochondrion or micronucleus,
or exogeneous DNA derived
from virus, dead cell, and
bacterium. Activated cGAS uses
ATP and GTP as substrates to
catalyze the formation of
cGAMP, which acts as a second
messenger to activate STING.
bCyclic dinucleotides secreted
from bacteria can also activate
STING directly. cActivated
STING dimerizes and
translocates from the ER to the
perinuclear region via Golgi to
recruit and promote TBK1-
IRF3-IκB assembly, which
subsequently drives activation of
IRF3 and NF-κB to produce IFN
and inammatory cytokines
(TNF, IL-6), respectively.
Crosstalk between cGASSTING signaling and cell death
Monosiga brevicollis, with the core CDN-binding function
of STING showing evolutionary conservation over 600
million years [45]. Interestingly, STING or cGAS alone is
unable to drive IFN-βexpression effectively as shown fol-
lowing Ectromelia Virus infections in HEK293T cells [46].
This is further supported by the fact that these two proteins
have coevolved, such that different species will either
contain homologs of both cGAS and STING or have lost
both, highlighting their crucial requirement to cooperatively
mediate the inammatory responses [45].
Interactions between the cGASSTING
pathway and apoptosis
Apoptosis is one of the most conserved and best studied cell
death pathways in terms of mechanism and regulation for its
vital role in development and homeostasis [47,48]. In brief,
apoptosis is mediated by caspases, a family of cysteine
proteases [49]. There are three major pathways that signal
caspase activation and apoptosis in mammalian cells, often
termed as intrinsic, extrinsic (or mitochondrial pathway),
and granzyme-mediated cell death pathway [50]. The
extrinsic pathway is initiated by binding of death ligands
such as TNF, FASL, TRAIL, to TNF superfamily receptors
such as TNF receptor 1 (TNFR1), FAS, death receptor 4/5,
respectively, to recruit cytoplasmic adapter proteins such as
Fas-associated death domain or TNF-associated death
domain. These adapters in-turn recruit and activate caspase-
8. Caspase-8 is the central protease in death receptor
induced apoptosis, and once activated, it mediates caspase-3
activation, either directly or through (BH3 Interacting
Domain Death Agonist) BID cleavage and engagement of
the mitochondrial pathway [51]. The intrinsic pathway is
centered on B-cell lymphoma-2 (BCL-2) family proteins
that comprises of both antiapoptotic and proapoptotic
members [47,52]. Cellular stress or danger signals can
either activate proapoptotic proteins such as BCL-2 asso-
ciated X protein (BAX) and BCL-2 homologous antagonist/
killer (BAK) or inhibit antiapoptotic proteins (BCL-2, BCL-
x, BCL-w, and MCL-1) through the activation of several
BH3 only domain family members (BIM, BID). This results
in mitochondrial outer membrane permeabilization, result-
ing in cytochrome c release, that binds to an adapter
apoptotic protease activating factor-1 (APAF1) to initiate
the formation of the apoptosome, a complex required for
recruitment and activation of caspase-9. Activated caspase-
9 cleaves and activates caspase-3 and -7, which targets a
number of cellular substrates to initiate apoptotic cell death
[53]. Last, in the granzyme pathway cytotoxic T lympho-
cytes kill target cells via secreted granular proteases and
perforins [54,55]. Despite the specic requirements of each
of these pathways, intrinsic, extrinsic, and granzyme B
converge on execution pathway (caspase-3 and -7 activa-
tion) of apoptosis [50]. It is however worth noting that
cytotoxic T lymphocytes can also induce non-apoptotic
types of death in target cells [56].
Apoptosis is generally regarded as silent cell death, where
apoptotic bodies are rapidly phagocytosed by neighboring
cells or macrophages to prevent the activation of inamma-
tory pathways. Therefore, not surprisingly apoptotic caspase
activation has inhibitory effect on cGASSTING inamma-
tory pathways. For example, both in vitro and in vivo ISG is
constitutively expressed in caspase-9-decient, and caspase-3/
7 double knockout MEFs. Furthermore, STING pathway is
constitutively activated in caspase-9 decient cells [57].
Finally, mtDNA released from Bak/Bax activation is shown
to be the endogenous ligand that triggers the cGASSTING
pathway activation, with caspases regulating the inammatory
responses [57]. These ndings are further corroborated by an
independent study that conrmed the role of mtDNA in
initiating cGASSTING-mediated IFN responses and active
caspase-9 inhibiting these inammatory responses in hema-
topoietic cells both in vivo and in vitro [58]. The mechanism
of apoptotic caspases downregulating inammatory responses
was uncovered in a recent study, where active caspase-3
cleaved cGAS, and IRF3 in THP-1 cells during viral infec-
tions to prevent excessive IFN production [59]. Moreover,
defects in clearing apoptotic cells led to the formation of
apoptosis-derived membrane vesicles (ADMVs) that have
been associated with autoimmune diseases such as SLE [60].
ADMVs isolated from the sera of SLE patients contained
dsDNA that triggered cGASSTING signaling pathway
leading to ISG expression in THP-1 cells [61]. Based on
these studies it can be argued that apoptosis prevents
cGASSTING-mediated inammatory responses (Fig. 2). By
contrast, emerging evidence suggests that ER stress associated
with STING activation can trigger apoptosis. For instance,
during mycobacterial infections, STING activation causes ER
stress leading to BAX activation and cytochrome c release
resulting in apoptosis in RAW.247 cells [62]. In addition, ER
stress-mediated STING activation in mouse hepatocytes
results in apoptosis in an early alcoholic liver disease model
[63]. It is possible that STING regulates calcium homeostasis
and ER stress, as STING gain-of-function mutant disrupts
calcium homeostasis causing ER stress that primes T cell
apoptosis [64]. Also, STING pathway activation by the
STING agonist 10-carboxymethyl-9-acridanone (CMA) trig-
gers apoptosis in primary and lymphoblastic leukemia murine
T cells by coordinated action of IRF3 and p53 but not in
murine DCs [65]. Similarly, the STING agonist cGAMP
initiates ER-stress-mediated apoptosis in normal and malig-
nant B cells, but not in MEF, B16 melanoma, Hepa 16
hepatoma, LL/2 Lewis lung cancer cell lines and wild-type
T cells from mice [66]. Murine-specic small molecule
STING agonists CMA and DMXAA also failed to trigger
A. M. V. Murthy et al.
apoptosis in MEF, B16 melanoma, Hepa 16 hepatoma, LL/2
Lewis lung cancer cell lines [66]. Inability of these small
molecules to activate human STING is attributed to dynamic
structural differences between mouse and human STING [67].
These ndings also suggest that STING-mediated apoptosis is
cell- and/or context-dependent, which could be due to varia-
tion in endogenous STING expression amongst different cell
types as observed in some studies [65,66], or cell-specic
kinetics of STING regulation (phosphorylation or degrada-
tion). Further mechanistic insights on how STING regulates
calcium homeostasis, whether directly or indirectly by STIM1
or via other calcium signaling pathways, are required from the
therapeutic viewpoint of inhibiting ER stress to prevent
chronic STING activation in autoimmune diseases [64].
Interplay between the cGASSTING signaling
and necroptosis
Necroptosis is a regulated form of necrosis, mediated by
receptor interacting protein kinases activation and its sub-
strate mixed lineage kinase like (MLKL) that leads to
Fig. 2 Interplay between the
cGASSTING signaling
pathway and apoptosis. During
apoptosis signaling the
proapoptotic proteins such as
BAK/BAX cause mitochondrial
membrane permeabilisation and
release of cytochrome c and
mitochondrial DNA.
Mitochondrial DNA can trigger
cGASSTINGTBK1 pathway
to drive IFN-mediated immune
responses. Whereas cytosolic
cytochrome c drives the
assembly of the Apaf1-
caspaspe-9 apoptosome that
subsequently activate the
initiator caspases (e.g. caspase-3
and caspase-7) to mediate
apoptosis. Caspase-3 negatively
regulates cGASSTING
pathway resulting in cleavage of
IRF3 and STING. On the other
hand, STING activation causes
ER-stress that ultimately results
in apoptosis by via BAK/BAX
activation and cytochrome c
release. Red arrows indicate
negative regulation.
Crosstalk between cGASSTING signaling and cell death
plasma membrane disruption [6870]. When apoptosis is
inhibited by genetic defects, viral or bacterial infections, or
experimentally by caspase inhibitors, necroptosis is pro-
moted as a mode of regulated cell death that releases
DAMPS to initiate innate immune responses [71]. Impor-
tantly, IFN signaling is indispensable to trigger necroptosis
and IFN-αreceptor 1 (IFNAR1) decient mice are resistant
to necroptosis during Salmonella Typhimurium infections
[72]. Also, in IFNAR1 decient bone marrow-derived
macrophages (BMMs) necroptosis is inhibited, despite of
caspase inhibition and TLR induction by stimulation with
LPS, polyinosinic-polycytidilic acid, TNFα,orIFN-β[73].
Therefore, it is logical to deduce that the cGASSTING
pathway could play a predominant role in triggering
necroptosis (Fig. 3). Along the same lines, cGASSTING-
dependent TNF and IFN signaling triggers necroptosis in
response to cytosolic DNA, when apoptotic caspases are
inhibited in BMMs [74]. In addition, mtDNA activates
the STING pathway that subsequently enhances RIPK3/
MLKL expression to trigger necroptosis, when caspases are
inhibited in HT29 colon cancer cell line and MEFs [75].
Furthermore, constitutive IFN levels mediated by
cGASSTING activation maintain MLKL at a threshold
level to predispose BMMs to necroptosis [76]. Yet, over-
expression of MLKL in BMMs from Ifnar/mice does not
sensitize cells to necroptosis [76,77]. This suggests the
presence of additional IFN-inducible necroptotic pathway
components, that are required for necroptosis, yet to iden-
tied. However, during Salmonella Typhimurium infec-
tions, IFN-mediated RIPK3 activation impairs antioxidative
responses that could sensitize BMMs to necroptosis [78].
Considering that loss of apoptosis is a hallmark of cancer
[79], it is rational to trigger secondary cell death pathway
such as necroptosis that is mechanistically distinct.
Based on some studies (as discussed above) indicating
cGASSTING signaling enhances expression of necroptotic
apparatus to trigger robust cell death upon specic stimuli
[75,76], it is reasonable to consider using STING agonists
to trigger necroptosis in cancer cells. However, the use of
the STING agonist DMXAA combined with caspase inhi-
bition in vivo increases TNF and IFN levels subsequently
leading to necroptosis, but this resulted in increased mor-
tality due to septic shock [74]. Although, DMXAA is a
failure in clinics due to its inability to effectively activate
human STING, small molecule STING agonist amido-
benzimidazole (ABZI) has been reported to bind and acti-
vate both human and mouse STING [80,81]. Therefore,
such STING agonists in clinical trials will need to be
assessed for their roles in triggering necroptosis without
extensive secondary effects. This conclusion is reinforced
by the observation that necroptotic rather than apoptotic cell
death is efcient in DC-mediated cross-priming of CD8+
T cells to achieve antitumor immunity [82]. Despite the
clinical interest in use of STING agonists as anticancer
drug, surprisingly little is known about the role of necrop-
tosis during STING activation, however, this is clearly an
area of ongoing investigations. On the other hand, it would
be of interest to examine if STING antagonists can be used
Fig. 3 Initiation of necroptosis by cGASSTING signaling. Acti-
vation of the cGASSTING pathway by mitochondrial DNA results in
production of IFN and TNF. Binding of IFN to IFNAR1 and TNF to
TNFR1 can results in RIPK1RIPK3 activation, when caspase-8 is
inhibited. RIPK1RIPK3 activates MLKL to execute necroptosis.
Moreover, IFN upregulates RIPK3 and MLKL expression. It would be
interesting to understand if STING agonists can initiate necroptosis to
limit tumor growth when apoptotic caspases are usually inhibited in
cancer cells. Green arrow indicate upregulation.
A. M. V. Murthy et al.
to treat septic shock caused by bacterial infections. This will
require further understanding of the mechanism and role of
necroptosis in inducing septic shock in the context of bac-
terial infections.
Crosstalk between the cGASSTING pathway
and pyroptosis
Pyroptosis is an inammatory form of cell death initiated by
inammasomes, eventually leading to gasdermin-D (or E)
cleavage to generate the N-terminal domain, which forms a
pore in the cell membrane [8385]. In brief, inammasomes
are large multiprotein complexes comprised of germline-
encoded pattern recognition receptors of the Nod-like receptor
(NLR) family (NLR family pyrin domain-containing 3,
NLRP3; NLR family CARD domain-containing protein 4,
NLRC4; and AIM2), the adapter ASC and procaspase-1. In
response to pathogenic or physiological perturbation in the
cytosol, procaspase-1 undergoes inammasome-assisted
proximity-induced autoproteolysis resulting in active
caspase-1 [86]. Active caspase-1 converts pro-IL-1βand pro-
IL-18 into its mature form and also cleaves gasdermin-D to
initiate pyroptosis [87]. A considerable body of literature
highlights the critical role of the AIM2 inammasome in the
host defense mechanism, and AIM2 decient mice are highly
susceptible to infections with Francisella tularensis,Myco-
bacterium tuberculosis,Staphylococcus aureus, and display
higher mortality and bacterial burden in comparison to wild-
type mice [8890]. Nonetheless, the role of the
cGASSTING in these scenarios are yet to be examined.
A key question is whether both cGAS and AIM2 func-
tion concurrently or act sequentially to initiate immune
responses? To address this, a study demonstrated that
cGASSTING-IFN1 pathway activation amplied AIM2
protein levels to induce robust innate immune responses
during murine cytomegaloviral infections in BMMs [91].
Similarly, cGAS-mediated IFN responses increase caspase-
1 and caspase-11 expression, which in turn increased IL-1β
release and pyroptotic cell death during Chlamydia tra-
chomatis infections [92]. Therefore, robust activation of
both canonical and noncanonical inammasomes are
dependent on early IFN priming of inammasome compo-
nents. With these observations it can be proposed that
AIM2 activation follows STING signaling, and the activa-
tion of these signaling pathways amplify the host defense
response (Fig. 4). However, cGASSTING-NLRP3 acti-
vation replaces the AIM2 function in human myeloid cells
in response to DNA, as STING activation induces lysoso-
mal cell death leading to K+efux, which in turn activates
the NLRP3 inammasome [93]. This nding raises the
question as to whether STING is more sensitive to mis-
localized DNA than AIM2? It is possible that
cGASSTING can detect lower levels of DNA in the
cytosol than AIM2. Another possible explanation is that
there is still an unknown factor that could determine which
DNA sensor to be activated depending on the levels of
DNA in the cytosol, that would correspond to the degree of
the danger signal or localization of DNA.
Conversely, to prevent overactivation of inammatory
responses, the innate immune system maintains a balance
between different DNA sensors expression and/or activa-
tion. For instance, gasdermin-D activated by the AIM2
inammasome inhibits IFN-βresponses to cytoplasmic
DNA via K+efux in BMM [94]. In addition, AIM2
decient BMMs and bone marrow-derived DCs display
elevated IFN-βlevels in response to cytoplasmic DNA-
mediated cGASSTING activation [95]. Moreover, to pre-
vent deleterious effects of inammatory responses not only
the DNA-sensing AIM2 inammasomes, but also other
components of inammasomes can negatively regulate the
cGASSTING pathway. For example, the NLRC3 interacts
directly with STING to prevent its translocation into the
perinuclear region and binding to TBK1, that ultimately
hinders IFN responses and NF-κB activation [96]. Also,
caspase-1 limits cytosolic DNA-mediated cGASSTING
activation by directly cleaving cGAS during canonical
inammasome activation [97]. Similarly, during LPS-
induced noncanonical inammasome activation caspase-4,
-5 (human), and -11 (mouse) can cleave human and mouse
cGAS, respectively [97]. Furthermore, NLRP4 can recruit
the ubiquitin ligase DTX4 to TBK1 for ubiquitination,
which leads to TBK1 degradation and thereby regulation of
DNA-mediated IFN responses [98]. By contrast, IFN
inhibited NLRP3 inammasome activation in STAT1-
dependent manner, thereby preventing caspase-1 depen-
dent IL-1βrelease [99]. Collectively, these studies highlight
the sophisticated interplay between cGASSTING and
inammasome components to prevent hyper-inammation.
cGASSTING and autophagy
The evolutionarily conserved catabolic process of autop-
hagy is essential for cellular health and organismal home-
ostasis [100,101]. Autophagy involves a complex
machinery that targets damaged or long-lived organelles
and/or proteins packaged in double membrane vesicles
termed autophagosome, to lysosomes for degradation and/
or recycling. Based on the mode of cargo delivery to
lysosomes, autophagy can be broadly classied into mac-
roautophagy (cytoplasmic contents are delivered to lyso-
some by autophagosomes), microautophagy (direct
lysosomal membrane invagination), and chaperone-
mediated (direct translocation across lysosomal mem-
brane), which is a non-conserved process only observed in
Crosstalk between cGASSTING signaling and cell death
mammalian cells [102]. Macroautophagy, henceforth
referred to as autophagy, involves several steps: initiation,
nucleation or phagophore formation, elongation of phago-
phore membrane to form autophagosome, fusion of autop-
hagosome with the lysosomes, and degradation of
autophagosome contents. Each of these steps involves a
series of autophagy-related gene (Atg) products. Briey,
autophagy initiation is assisted by the Atg1/ULK1 serine-
threonine complex comprising ULK1, ATG13, ATG101,
and FAK-family interacting protein (FLIP2000) [103]. The
class III phosphatidylinositol 3-kinase (PI3K) complex,
consisting of ATG15, vacuolar protein sorting (VPS) 15,
VPS 34, and Beclin1-related proteins mediate nucleation by
generating phosphatidylinositol 3-phosphate (PI3P) [104].
ATG9 vesicles offer a portion of the autophagosomal
membrane, whilst WIPI proteins and its interacting partners
(ATG2a or ATG2b), and two transmembrane ER proteins,
vacuole membrane protein 1 and TMEM41 B are necessary
to form isolation membrane [105108]. Two unique
ubiquitin-like protein conjugation systems, ATG12-ATG5-
ATG16L complex and ubiquitin-like microtubule-asso-
ciated protein 1 light chain 3 (MAP1LC3B/ATG8) are
conjugated to a lipid molecule phosphatidylethanolamine to
aid in elongation [109]. LC3 is synthesized in its precursor
form as pro-LC3, which is cleaved by ATG4 to LC3-I
[110,111]. ATG3 and ATG7 convert LC3-I to LC3-II,
which aids in elongating autophagosome membrane
[112114]. While the autophagosome closure remains
Fig. 4 Interplay between the
cGASSTING signaling
pathway and pyroptosis. A
model that proposes DNA
concentration could determine
type of DNA sensor and level of
innate immune responses
required to maintain
homeostasis. During low
concentration of DNA in the
cytosol, cGASSTING could be
activated to drive IFN-mediated
immune responses. Whereas,
when high concentrations of
DNA accumulate in the cytosol,
the AIM2 inammasome could
be activated to intensify the
innate immune responses by IL-
1βrelease, and pyroptosis
mediated by active caspase-1
and gasdermin cleavage.
Therefore, IFN release from
cGASSTING activation primes
AIM2 inammasome
components as marked by
increased expression of AIM2,
procaspase-1/11/4/5.
Conversely, to prevent
hyperactivation of the immune
responses, K+efux mediated
by gasdermin-d cleavage
inhibits cGAS, and active
caspase-1/11/4/5 can cleave
cGAS directly. Red arrows
indicate negative regulation, and
green arrows indicate positive
regulation.
A. M. V. Murthy et al.
poorly understood [115], autophagosomes fusion with the
lysosomes requires specic SNARE proteins, small GTPa-
ses and their effectors [116] to initiate degradation of cargo
by a series of lysosomal enzymes. Although, autophagy is
regarded as an unselective process, selective autophagy is
mediated by ubiquitin-binding proteins (p62, autophagy
receptor optineurin, and NDP52), or proteins with trans-
membrane domains such as Nix (NIP3-like protein X as
mitophagy receptor) [117,118].
In recent years growing evidence has indicated interac-
tions between autophagy machinery and the cGASSTING
pathway. Over 10 years ago Akira et al. [119] identied that
ATG9a and LC3-II co-localize with STING in vesicles after
dsDNA stimulation in MEFs. In addition, deletion of
ATG9a increases STINGTBK1 assembly upon sensing
dsDNA, suggesting the possible roles of autophagy proteins
in STING regulation [119]. Subsequent studies demon-
strated interactions between autophagy and cGASSTING
using both genetic and biochemical approaches [120122].
It is possible that STING activation can initiate autophagy
and follow the key steps of autophagy pathway (discussed
above), or STINGTBK1 can be translocated directly to
autophagosome via an unknown mechanism independent of
p62 (Fig. 5). As an example, cGAS-Beclin-1 interaction
initiates autophagy by releasing the autophagy negative
regulator, Rubicon to allow PI3K complex activation upon
dsDNA recognition during HSV infections [123]. Further-
more, STING-induced autophagy requires WIPI2 and
ATG5 proteins [122], ATG3, p62 and increase in LC3-II
conversion followed by degradation of STING via con-
ventional autophagy [121]. On the other hand, STING
interacts directly with LC3 leading to degradation of STING
itself and pTBK1, highlighting unconventional means of
autophagy activation [120]. Undoubtedly, there is a high
degree of complexity associated with re-localization of
STING with different autophagy proteins. It should be
noted that most of the STING-autophagy studies used V-
ATPase inhibitors such as Balomycin or concanamycin A,
or chloroquine to inhibit autophagy pathway that could
affect other signaling pathways. Also, studies involving
genetic manipulation of autophagy should account for the
non-autophagic role of autophagy proteins in cell survival,
maintenance, and death [124].
During Mycobacterium tuberculosis infection bacterial
DNA-mediated STINGTBK1 activation targets a sub-
population of bacteria to autophagosomes in BMMs [125]
that is likely to attenuate virulence and pathogenicity in
murine lungs [126]. However, during Mycobacterium tuber-
culosis infections, NF-κB-induced expression of the DNA
damage-regulated autophagy modulator DRAM1, which then
localized with p62 and STING in zebrash and human
macrophages initiating autophagic defense responses [127]. It
is possible that autophagy prevents sustained STING
phosphorylation but not NF-κB activation [128]. Although,
the NF-κB activation is dependent on MyD88, the role of
STING in NF-κB activation cannot be overlooked [127].
Although p62 was demonstrated to be crucial for trans-
location of STING to lysosome-associated compartments
for STING degradation, it should be noted that p62
knockout MEFs and THP-1 cells show a delay rather than
abrogation of STING degradation in comparison to wild-
type cells [121]. This suggests that ubiquitination by other
ubiquitin ligases such as RNF5 and TRIM30 can also target
STING for proteasomal degradation [129,130]. Additional
studies are required to understand the role of ubiquitin
ligases in autophagy-dependent STING activation and reg-
ulation. Collectively, STING activation initiates autophagy
followed by its own degradation. However, cell-type, con-
centration of cytoplasmic DNA and the strength of
inammatory responses may dictate how and whether
autophagy components mediate STING pathway signaling.
Importantly, it is possible that STING-mediated autophagy
can be a potential way to clear infections effectively, and
further studies are warranted to understand the role of
STING agonists to treat bacterial infections.
Some studies have reported that rather than STING, it is
cGAS that interacts with autophagy components, thereby
increasing the complexity of interactions between the
cGASSTING pathway and autophagy. In keeping with this
concept, cGAS/mice display increased susceptibility to
Mycobacterium tuberculosis than their wild-type or STING-
decient counterparts [131]. In addition, cGAS-induced
autophagy protects hepatocytes that lack STING [132]from
hypoxia-induced ischemia-reperfusion injury, independent of
Beclin-1 [133]. Overall, it could be envisaged that cGAS can
activate STING-dependent and -independent autophagy as
host protective response depending on the intensity of the
threat posed, and the degree of inammatory responses
required to clear danger signals. One possibility is that
STING, but not its activation is required for basal autophagy
as STING-dependent autophagy occurs independent of TBK1
activation and IFN production [122]. In contrast,
cGASSTING pathway activation leading to inammatory
responses as a host defense mechanism is required to coun-
teract a severe threat. Alternatively, there could be an
unknown sensor that licenses STING-dependent versus
STING-independent autophagy in a cell-type or cytosolic
DNA concentration context [134].
ADCD following cGASSTING activation
As autophagy removes damaged organelles and protects
cells under nutrient-limiting conditions, it is primarily a
cytoprotective process. However, autophagy often accom-
panies cell death and has been shown to be a driver of cell
Crosstalk between cGASSTING signaling and cell death
death in specic contexts [135]. ADCD is dened by The
Nomenclature Committee of Cell Death as a form of
regulated cell death that mechanistically depends on the
autophagy components[3]. To be termed as ADCD,
genetic or chemical inhibition of autophagy should prevent
cell death [102,136]. Also, more than one autophagy pro-
teins must be involved in triggering cell death [135].
To date, the most robust genetic evidence for ADCD
comes from studies involving the degradation of larval
midgut during larval-pupal transition in Drosophila
[137,138]. Genetic studies have demonstrated that larval
midgut degradation can occur in the complete absence of
the apoptosis machinery in Drosophila but has an essential
requirement for autophagy induction [131,134,135]. Stu-
dies in mammalian and other model systems have also
identied instances of ADCD in specic developmental or
pathophysiological context [135]. For example, autosis is a
specic form of ADCD that involves the Na+/K+-ATPase
[139]. In another instance, the removal of interdigital web
cells during embryonic development is further delayed in
Atg5/Bax/Bak triple decient mice, compared to Bax/Bak
double knockouts, suggesting a role for autophagy in
removing interdigital web in mammals [140]. Notably,
invertebrates lack C-terminal tail of STING that recruits
Fig. 5 Complexity of
interactions between
autophagy machinery and the
cGASSTING pathway.
Autophagy process involves ve
key steps, (1) initiation, (2)
nucleation, (3) elongation, (4)
autophagosome formation, and
(5) degradation, where each step
is regulated by specic ATG
proteins as highlighted, or
explicit proteins such as ULK1,
Beclin-1, P13P, LC3
conjugation system as
mentioned. cGASSTING
activation can initiate autophagy
and follow the ve key steps
triggering its own degradation,
however the requirement of
specic autophagy components
at each step is ambiguous. In
addition, cGASSTING
activation mediated by
cGASSTINGTBK1IRF3
assembly complex can
translocate directly to
autophagosomes via an
unknown mechanism. Also,
cGAS can interact directly with
Beclin-1 to initiate autophagy
independent and/or absence of
STING. Furthermore, ULK1 can
phosphorylate STING and
prevent its activation.
A. M. V. Murthy et al.
TBK1 and IRF3 and can still mediate ADCD [141]. How-
ever, recent studies link self-DNA stimulated cGAS
STING to excessive autophagy leading to ADCD
[139,140]. One study demonstrated that cell crisis due to
telomeric damage results in release of DNA fragments into
the cytoplasm, that trigger autophagy as observed by
increased in LC3-II or decrease in p62 [142]. Inhibition of
autophagy or STING pathway components prevented cell
crisis resulting in unstable genome and chromosomal
aberrations, suggesting that this pathway is involved in
deleting damaged and potentially premalignant cells [142].
In other recent study the infection by Burkholderia pseu-
domallei, that causes host cell fusion, was shown to activate
cGASSTING activation, independent of bacterial ligands
[140]. Infection induced cell fusion lead to aberrant mitosis,
formation of micronuclei, which colocalised with cGAS,
followed by the activation of cGASSTING pathway and
ADCD [140]. These two studies demonstrate that cell crisis
or bacterial-mediated cell fusion results in cGASSTING-
mediated autophagy and that subsequent ADCD limits
accumulation of damaged cells [142,143]. Thus, these
studies suggest that one of the functions of cGASSTING-
dependent ADCD is to prevent tumorigenesis. However,
autophagy is known to have dual role in cancer [144]
and therefore, further mechanistic studies are required to
understand context-dependent ADCD. Although, excessive
autophagy can clearly trigger cell death, further work is
warranted to establish if different molecular factors are
involved in developmental versus stress-induced ADCD.
Conclusion and perspectives
Activation of the cGASSTING pathway is a major
responder to viral and bacterial infections. However, this
pathway can be activated by self-DNA and can act as a
double-edged sword, where it can be benecial or detri-
mental to the host depending on the context of its activation
(self-DNA, infection, and chronic inammation), cell-type,
cell death pathways activated, and the magnitude and
duration of activation. Structural studies of STING have
shown that a closed conformational change in the lidloop
of STING upon binding to CDN is necessary for its acti-
vation. However, studies with the newly identied ABZI
STING agonist demonstrated that the activation can occur
in an open conformation. Identication of such novel small
molecules also gives a perspective that structures other than
CDN with distinct physiochemical and biological properties
could bypass cGAS to activate STING and provides more
options for the clinical development of STING agonists.
Considering that multiple cell death pathways can be trig-
gered following cGASSTING signaling, a better under-
standing of the regulatory mechanism distinct to cell-types,
context and/or stimulus-dependency will require further
investigations. Moreover, the role of STING in the co-
existence of various cell death pathways such as the newly
described PANopoptosis [145] and switch between cell
death pathways remain to be studied. Further knowledge
will help answer questions such as what dictates the
redundancy in cell death pathways triggered by the
cGASSTING. Any direct role of cGASSTING signaling
in ADCD and the mechanisms of ADCD also require fur-
ther investigations. More importantly, answers to these
questions will eventually open new avenues to target
STING-mediated cell death pathways for treating cancer
and infectious diseases.
Acknowledgements The work in our laboratories is supported by the
National Health & Medical Research Council (NHMRC) project
grants (1144500, 1156601), a NHMRC Senior Principal Research
Fellowship (1103006) to SK, and the University of South Australia
internal research support to SK and NR.
Author contributions AMVM drafted, edited, and revised the manu-
script text and prepared gures, NR provided intellectual input and
edited the manuscript, SK conceptualized, edited, and revised the
manuscript, and contributed intellectual input.
Compliance with ethical standards
Conict of interest The authors declare that they have no conict of
interest.
Publishers note Springer Nature remains neutral with regard to
jurisdictional claims in published maps and institutional afliations.
References
1. Sun EW, Shi YF. Apoptosis: the quiet death silences the immune
system. Pharm Ther. 2001;92:13545.
2. Kerr JF, Wyllie AH, Currie AR. Apoptosis: a basic biological
phenomenon with wide-ranging implications in tissue kinetics.
Br J Cancer. 1972;26:23957.
3. Galluzzi L, Vitale I, Aaronson SA, Abrams JM, Adam D,
Agostinis P, et al. Molecular mechanisms of cell death: recom-
mendations of the Nomenclature Committee on Cell Death 2018.
Cell Death Differ. 2018;25:486541.
4. Unterholzner L. The interferon response to intracellular DNA:
why so many receptors? Immunobiology. 2013;218:131221.
5. Vance RE. Cytosolic DNA sensing: the eld narrows. Immunity.
2016;45:2278.
6. Kumagai Y, Takeuchi O, Akira S. TLR9 as a key receptor for the
recognition of DNA. Adv Drug Deliv Rev. 2008;60:
795804.
7. Latz E, Schoenemeyer A, Visintin A, Fitzgerald KA, Monks BG,
Knetter CF, et al. TLR9 signals after translocating from the ER to
CpG DNA in the lysosome. Nat Immunol. 2004;5:1908.
8. Barton GM, Kagan JC, Medzhitov R. Intracellular localization of
Toll-like receptor 9 prevents recognition of self DNA but facil-
itates access to viral DNA. Nat Immunol. 2006;7:4956.
Crosstalk between cGASSTING signaling and cell death
9. Marongiu L, Gornati L, Artuso I, Zanoni I, Granucci F. Below
the surface: the inner lives of TLR4 and TLR9. J Leukoc Biol.
2019;106:14760.
10. Ewald SE, Lee BL, Lau L, Wickliffe KE, Shi GP, Chapman HA,
et al. The ectodomain of Toll-like receptor 9 is cleaved to gen-
erate a functional receptor. Nature 2008;456:65862.
11. Bao W, Xia H, Liang Y, Ye Y, Lu Y, Xu X, et al. Toll-like
receptor 9 can be activated by endogenous mitochondrial DNA
to induce podocyte apoptosis. Sci Rep. 2016;6:22579.
12. Zhang Q, Raoof M, Chen Y, Sumi Y, Sursal T, Junger W, et al.
Circulating mitochondrial DAMPs cause inammatory responses
to injury. Nature. 2010;464:1047.
13. Gray MW. Mitochondrial evolution. Cold Spring Harb Perspect
Biol. 2012;4:a011403.
14. Liu B, Du Q, Chen L, Fu G, Li S, Fu L, et al. CpG methylation
patterns of human mitochondrial DNA. Sci Rep. 2016;6:23421.
15. Man SM, Karki R, Kanneganti TD. AIM2 inammasome in
infection, cancer, and autoimmunity: role in DNA sensing,
inammation, and innate immunity. Eur J Immunol.
2016;46:26980.
16. Jin T, Perry A, Jiang J, Smith P, Curry JA, Unterholzner L, et al.
Structures of the HIN domain:DNA complexes reveal ligand
binding and activation mechanisms of the AIM2 inammasome
and IFI16 receptor. Immunity. 2012;36:56171.
17. Zhou W, Whiteley AT, de Oliveira Mann CC, Morehouse BR,
Nowak RP, Fischer ES, et al. Structure of the human cGAS-
DNA complex reveals enhanced control of immune surveillance.
Cell. 2018;174:30011 e11.
18. Li X, Shu C, Yi G, Chaton CT, Shelton CL, Diao J, et al. Cyclic
GMP-AMP synthase is activated by double-stranded DNA-
induced oligomerization. Immunity. 2013;39:101931.
19. Gentili M, Lahaye X, Nadalin F, Nader GPF, Puig Lombardi E,
Herve S, et al. The N-terminal domain of cGAS determines pre-
ferential association with centromeric DNA and innate immune
activation in the nucleus. Cell Rep. 2019;26:237793 e13.
20. Wang L, Wen M, Cao X. Nuclear hnRNPA2B1 initiates and
amplies the innate immune response to DNA viruses. Science.
2019;365:6454.
21. Zierhut C, Yamaguchi N, Paredes M, Luo JD, Carroll T, Funa-
biki H. The cytoplasmic DNA sensor cGAS promotes mitotic
cell death. Cell. 2019;178:30215 e23.
22. Barnett KC, Coronas-Serna JM, Zhou W, Ernandes MJ, Cao A,
Kranzusch PJ, et al. Phosphoinositide interactions position cGAS
at the plasma membrane to ensure efcient distinction between
self- and viral DNA. Cell. 2019;176:143246 e11.
23. Barber GN. STING: infection, inammation and cancer. Nat Rev
Immunol. 2015;15:76070.
24. Motwani M, Pesiridis S, Fitzgerald KA. DNA sensing by the
cGASSTING pathway in health and disease. Nat Rev Genet.
2019;20:65774.
25. Ishikawa H, Ma Z, Barber GN. STING regulates intracellular
DNA-mediated, type I interferon-dependent innate immunity.
Nature. 2009;461:78892.
26. Ishikawa H, Barber GN. STING is an endoplasmic reticulum
adaptor that facilitates innate immune signalling. Nature.
2008;455:6748.
27. Gall A, Treuting P, Elkon KB, Loo YM, Gale M Jr., Barber GN,
et al. Autoimmunity initiates in nonhematopoietic cells and
progresses via lymphocytes in an interferon-dependent auto-
immune disease. Immunity. 2012;36:12031.
28. Zhou R, Xie X, Li X, Qin Z, Wei C, Liu J, et al. The triggers of
the cGASSTING pathway and the connection with inamma-
tory and autoimmune diseases. Infect Genet Evol.
2020;77:104094.
29. Gao P, Ascano M, Wu Y, Barchet W, Gaffney BL, Zillinger T,
et al. Cyclic [G(2,5)pA(3,5)p] is the metazoan second
messenger produced by DNA-activated cyclic GMP-AMP syn-
thase. Cell. 2013;153:1094107.
30. Du M, Chen ZJ. DNA-induced liquid phase condensation of
cGAS activates innate immune signaling. Science.
2018;361:7049.
31. Ablasser A, Goldeck M, Cavlar T, Deimling T, Witte G, Rohl I,
et al. cGAS produces a 2-5-linked cyclic dinucleotide second
messenger that activates STING. Nature. 2013;498:3804.
32. Chen Q, Boire A, Jin X, Valiente M, Er EE, Lopez-Soto A, et al.
Carcinoma-astrocyte gap junctions promote brain metastasis by
cGAMP transfer. Nature. 2016;533:4938.
33. Pepin G, De Nardo D, Rootes CL, Ullah TR, Al-Asmari SS,
Balka KR, et al. Connexin-dependent transfer of cGAMP to
phagocytes modulates antiviral responses. mBio. 2020;11:
e0318719.
34. Burdette DL, Monroe KM, Sotelo-Troha K, Iwig JS, Eckert B,
Hyodo M, et al. STING is a direct innate immune sensor of
cyclic di-GMP. Nature. 2011;478:5158.
35. Woodward JJ, Iavarone AT, Portnoy DA. c-di-AMP secreted by
intracellular Listeria monocytogenes activates a host type I
interferon response. Science. 2010;328:17035.
36. Danilchanka O, Mekalanos JJ. Cyclic dinucleotides and the
innate immune response. Cell. 2013;154:96270.
37. Srikanth S, Woo JS, Wu B, El-Sherbiny YM, Leung J, Chupradit
K, 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;20:15262.
38. Zhang C, Shang G, Gui X, Zhang X, Bai XC, Chen ZJ. Struc-
tural basis of STING binding with and phosphorylation by
TBK1. Nature. 2019;567:3948.
39. Tanaka Y, Chen ZJ. STING species IRF3 phosphorylation by
TBK1 in the cytosolic DNA signaling pathway. Sci Signal.
2012;5:ra20.
40. Liu S, Cai X, Wu J, Cong Q, Chen X, Li T, et al. Phosphor-
ylation of innate immune adaptor proteins MAVS, STING, and
TRIF induces IRF3 activation. Science. 2015;347:aaa2630.
41. Diamond MS, Kinder M, Matsushita H, Mashayekhi M, Dunn
GP, Archambault JM, et al. Type I interferon is selectively
required by dendritic cells for immune rejection of tumors. J Exp
Med. 2011;208:19892003.
42. Fuertes MB, Kacha AK, Kline J, Woo SR, Kranz DM, Murphy
KM, et al. Host type I IFN signals are required for antitumor
CD8+T cell responses through CD8{alpha}+dendritic cells. J
Exp Med. 2011;208:200516.
43. Balka KR, Louis C, Saunders TL, Smith AM, Calleja DJ,
DSilva DB, et al. TBK1 and IKKepsilon act redundantly to
mediate STING-induced NF-kappaB responses in myeloid cells.
Cell Rep. 2020;31:107492.
44. Wu J, Dobbs N, Yang K, Yan N. Interferon-independent activ-
ities of mammalian STING mediate antiviral response and tumor
immune evasion. Immunity. 2020;53:11526 e5.
45. Wu X, Wu FH, Wang X, Wang L, Siedow JN, Zhang W, et al.
Molecular evolutionary and structural analysis of the cytosolic
DNA sensor cGAS and STING. Nucleic Acids Res
2014;42:824357.
46. Cheng WY, He XB, Jia HJ, Chen GH, Jin QW, Long ZL, et al.
The cGASSTING signaling pathway is required for the innate
immune response against ectromelia virus. Front Immunol.
2018;9:1297.
47. Singh R, Letai A, Sarosiek K. Regulation of apoptosis in health
and disease: the balancing act of BCL-2 family proteins. Nat Rev
Mol Cell Biol. 2019;20:17593.
48. Strasser A, OConnor L, Dixit VM. Apoptosis signaling. Annu
Rev Biochem. 2000;69:21745.
49. Shalini S, Dorstyn L, Dawar S, Kumar S. Old, new and emerging
functions of caspases. Cell Death Differ. 2015;22:52639.
A. M. V. Murthy et al.
50. Elmore S. Apoptosis: a review of programmed cell death. Tox-
icol Pathol. 2007;35:495516.
51. Strasser A, Vaux DL. Viewing BCL2 and cell death control from
an evolutionary perspective. Cell Death Differ. 2018;25:1320.
52. Reed JC, Jurgensmeier JM, Matsuyama S. Bcl-2 family proteins
and mitochondria. Biochim Biophys Acta. 1998;1366:12737.
53. Porter AG. Flipping the safety catch of procaspase-3. Nat Chem
Biol. 2006;2:50910.
54. Voskoboinik I, Smyth MJ, Trapani JA. Perforin-mediated target-
cell death and immune homeostasis. Nat Rev Immunol.
2006;6:94052.
55. Martinvalet D, Zhu P, Lieberman J. Granzyme A induces
caspase-independent mitochondrial damage, a required rst step
for apoptosis. Immunity. 2005;22:35570.
56. Lieberman J. Granzyme A activates another way to die. Immunol
Rev. 2010;235:93104.
57. Rongvaux A, Jackson R, Harman CC, Li T, West AP, de Zoete
MR, et al. Apoptotic caspases prevent the induction of type I
interferons by mitochondrial DNA. Cell. 2014;159:156377.
58. White MJ, McArthur K, Metcalf D, Lane RM, Cambier JC,
Herold MJ, et al. Apoptotic caspases suppress mtDNA-induced
STING-mediated type I IFN production. Cell.
2014;159:154962.
59. Ning X, Wang Y, Jing M, Sha M, Lv M, Gao P, et al. Apoptotic
caspases suppress type I interferon production via the cleavage of
cGAS, MAVS, and IRF3. Mol Cell. 2019;74:1931 e7.
60. Fehr EM, Spoerl S, Heyder P, Herrmann M, Bekeredjian-Ding I,
Blank N, et al. Apoptotic-cell-derived membrane vesicles induce
an alternative maturation of human dendritic cells which is dis-
turbed in SLE. J Autoimmun. 2013;40:8695.
61. Kato Y, Park J, Takamatsu H, Konaka H, Aoki W, Aburaya S,
et al. Apoptosis-derived membrane vesicles drive the
cGASSTING pathway and enhance type I IFN production in
systemic lupus erythematosus. Ann Rheum Dis. 2018;77:150715.
62. Cui Y, Zhao D, Sreevatsan S, Liu C, Yang W, Song Z, et al.
Mycobacterium bovis induces endoplasmic reticulum stress
mediated-apoptosis by activating IRF3 in a murine macrophage
cell line. Front Cell Infect Microbiol. 2016;6:182.
63. Petrasek J, Iracheta-Vellve A, Csak T, Satishchandran A, Kodys
K, Kurt-Jones EA, et al. STING-IRF3 pathway links endo-
plasmic reticulum stress with hepatocyte apoptosis in early
alcoholic liver disease. Proc Natl Acad Sci USA.
2013;110:165449.
64. Wu J, Chen YJ, Dobbs N, Sakai T, Liou J, Miner JJ, et al.
STING-mediated disruption of calcium homeostasis chronically
activates ER stress and primes T cell death. J Exp Med.
2019;216:86783.
65. Gulen MF, Koch U, Haag SM, Schuler F, Apetoh L, Villunger
A, et al. Signalling strength determines proapoptotic functions of
STING. Nat Commun. 2017;8:427.
66. Tang CH, Zundell JA, Ranatunga S, Lin C, Nefedova Y, Del
Valle JR, et al. Agonist-mediated activation of STING induces
apoptosis in malignant B cells. Cancer Res. 2016;76:213752.
67. Shih AY, Damm-Ganamet KL, Mirzadegan T. Dynamic struc-
tural differences between human and mouse STING lead to
differing sensitivity to DMXAA. Biophys J. 2018;114:329.
68. Pasparakis M, Vandenabeele P. Necroptosis and its role in
inammation. Nature 2015;517:31120.
69. Cai Z, Jitkaew S, Zhao J, Chiang HC, Choksi S, Liu J, et al.
Plasma membrane translocation of trimerized MLKL protein is
required for TNF-induced necroptosis. Nat Cell Biol.
2014;16:5565.
70. Chen X, Li W, Ren J, Huang D, He WT, Song Y, et al.
Translocation of mixed lineage kinase domain-like protein to
plasma membrane leads to necrotic cell death. Cell Res.
2014;24:10521.
71. Cho YS. The role of necroptosis in the treatment of diseases.
BMB Rep. 2018;51:21924.
72. Robinson N, McComb S, Mulligan R, Dudani R, Krishnan L,
Sad S. Type I interferon induces necroptosis in macrophages
during infection with Salmonella enterica serovar Typhimurium.
Nat Immunol. 2012;13:95462.
73. McComb S, Cessford E, Alturki NA, Joseph J, Shutinoski B,
Startek JB, et al. Type-I interferon signaling through ISGF3
complex is required for sustained Rip3 activation and necroptosis
in macrophages. Proc Natl Acad Sci USA. 2014;111:E320613.
74. Brault M, Olsen TM, Martinez J, Stetson DB, Oberst A. Intra-
cellular nucleic acid sensing triggers necroptosis through syner-
gistic type I IFN and TNF signaling. J Immunol.
2018;200:274856.
75. Chen D, Tong J, Yang L, Wei L, Stolz DB, Yu J, et al. PUMA
amplies necroptosis signaling by activating cytosolic DNA
sensors. Proc Natl Acad Sci USA. 2018;115:39305.
76. Sarhan J, Liu BC, Muendlein HI, Weindel CG, Smirnova I, Tang
AY, et al. Constitutive interferon signaling maintains critical
threshold of MLKL expression to license necroptosis. Cell Death
Differ. 2019;26:33247.
77. Legarda D, Justus SJ, Ang RL, Rikhi N, Li W, Moran TM, et al.
CYLD proteolysis protects macrophages from TNF-mediated
auto-necroptosis induced by LPS and licensed by type I IFN.
Cell Rep. 2016;15:244961.
78. Hos NJ, Ganesan R, Gutierrez S, Hos D, Klimek J, Abdullah Z,
et al. Type I interferon enhances necroptosis of Salmonella
Typhimurium-infected macrophages by impairing antioxidative
stress responses. J Cell Biol. 2017;216:410721.
79. Wong RS. Apoptosis in cancer: from pathogenesis to treatment. J
Exp Clin Cancer Res. 2011;30:87.
80. Ramanjulu JM, Pesiridis GS, Yang J, Concha N, Singhaus R,
Zhang SY, et al. Design of amidobenzimidazole STING receptor
agonists with systemic activity. Nature. 2018;564:43943.
81. Chipurupalli S, Ganesan R, Dhanabal SP, Kumar MS, Robinson
N. Pharmacological STING activation is a potential alternative to
overcome drug-resistance in melanoma. Front Oncol. 2020;10:758.
82. Yatim N, Jusforgues-Saklani H, Orozco S, Schulz O, Barreira da
Silva R, Reis e Sousa C, et al. RIPK1 and NF-kappaB signaling
in dying cells determines cross-priming of CD8(+) T cells.
Science. 2015;350:32834.
83. Voet S, Srinivasan S, LamkanM, van Loo G. Inammasomes
in neuroinammatory and neurodegenerative diseases. EMBO
Mol Med. 2019;11:e10248.
84. Broz P, Dixit VM. Inammasomes: mechanism of assembly,
regulation and signalling. Nat Rev Immunol. 2016;16:40720.
85. Man SM, Karki R, Kanneganti TD. Molecular mechanisms and
functions of pyroptosis, inammatory caspases and inamma-
somes in infectious diseases. Immunol Rev. 2017;277:6175.
86. Guo H, Callaway JB, Ting JP. Inammasomes: mechanism of
action, role in disease, and therapeutics. Nat Med.
2015;21:67787.
87. He WT, Wan H, Hu L, Chen P, Wang X, Huang Z, et al. Gas-
dermin D is an executor of pyroptosis and required for
interleukin-1beta secretion. Cell Res. 2015;25:128598.
88. Fernandes-Alnemri T, Yu JW, Juliana C, Solorzano L, Kang S,
Wu J, et al. The AIM2 inammasome is critical for innate
immunity to Francisella tularensis. Nat Immunol. 2010;11:38593.
89. Saiga H, Kitada S, Shimada Y, Kamiyama N, Okuyama M,
Makino M, et al. Critical role of AIM2 in Mycobacterium
tuberculosis infection. Int Immunol. 2012;24:63744.
90. Hanamsagar R, Aldrich A, Kielian T. Critical role for the AIM2
inammasome during acute CNS bacterial infection. J Neu-
rochem. 2014;129:70411.
91. Swanson KV, Junkins RD, Kurkjian CJ, Holley-Guthrie E,
Pendse AA, El Morabiti R, et al. A noncanonical function of
Crosstalk between cGASSTING signaling and cell death
cGAMP in inammasome priming and activation. J Exp Med.
2017;214:361126.
92. Webster SJ, Brode S, Ellis L, Fitzmaurice TJ, Elder MJ, Gekara
NO, et al. Detection of a microbial metabolite by STING reg-
ulates inammasome activation in response to Chlamydia tra-
chomatis infection. PLoS Pathog. 2017;13:e1006383.
93. Gaidt MM, Ebert TS, Chauhan D, Ramshorn K, Pinci F, Zuber
S, et al. The DNA inammasome in human myeloid cells is
initiated by a STING-cell death program upstream of NLRP3.
Cell. 2017;171:111024 e18.
94. Banerjee I, Behl B, Mendonca M, Shrivastava G, Russo AJ,
Menoret A, et al. Gasdermin D restrains type I interferon
response to cytosolic DNA by disrupting ionic homeostasis.
Immunity. 2018;49:41326 e5.
95. Corrales L, Woo SR, Williams JB, McWhirter SM, Dubensky
TW Jr., Gajewski TF. Antagonism of the STING pathway via
activation of the AIM2 inammasome by intracellular DNA. J
Immunol. 2016;196:31918.
96. Zhang L, Mo J, Swanson KV, Wen H, Petrucelli A, Gregory SM,
et al. NLRC3, a member of the NLR family of proteins, is a
negative regulator of innate immune signaling induced by the
DNA sensor STING. Immunity. 2014;40:32941.
97. Wang Y, Ning X, Gao P, Wu S, Sha M, Lv M, et al. Inam-
masome activation triggers caspase-1-mediated cleavage of
cGAS to regulate responses to DNA virus infection. Immunity.
2017;46:393404.
98. Cui J, Li Y, Zhu L, Liu D, Songyang Z, Wang HY, et al. NLRP4
negatively regulates type I interferon signaling by targeting the
kinase TBK1 for degradation via the ubiquitin ligase DTX4. Nat
Immunol. 2012;13:38795.
99. Guarda G, Braun M, Staehli F, Tardivel A, Mattmann C, Forster
I, et al. Type I interferon inhibits interleukin-1 production and
inammasome activation. Immunity. 2011;34:21323.
100. Clarke AJ, Simon AK. Autophagy in the renewal, differentiation
and homeostasis of immune cells. Nat Rev Immunol.
2019;19:17083.
101. Dikic I, Elazar Z. Mechanism and medical implications of
mammalian autophagy. Nat Rev Mol Cell Biol. 2018;19:34964.
102. Galluzzi L, Baehrecke EH, Ballabio A, Boya P, Bravo-San Pedro
JM, Cecconi F, et al. Molecular denitions of autophagy and
related processes. EMBO J. 2017;36:181136.
103. Zachari M, Ganley IG. The mammalian ULK1 complex and
autophagy initiation. Essays Biochem. 2017;61:58596.
104. Russell RC, Tian Y, Yuan H, Park HW, Chang YY, Kim J, et al.
ULK1 induces autophagy by phosphorylating Beclin-1 and
activating VPS34 lipid kinase. Nat Cell Biol. 2013;15:74150.
105. Mizushima N, Yoshimori T, Ohsumi Y. The role of Atg proteins
in autophagosome formation. Annu Rev Cell Dev Biol.
2011;27:10732.
106. Morishita H, Mizushima N. Diverse cellular roles of autophagy.
Annu Rev Cell Dev Biol. 2019;35:45375.
107. Molejon MI, Ropolo A, Vaccaro MI. VMP1 is a new player in
the regulation of the autophagy-specic phosphatidylinositol 3-
kinase complex activation. Autophagy. 2013;9:9335.
108. Moretti F, Bergman P, Dodgson S, Marcellin D, Claerr I,
Goodwin JM, et al. TMEM41B is a novel regulator of autophagy
and lipid mobilization. EMBO Rep. 2018;19:e45889.
109. Geng J, Klionsky DJ. The Atg8 and Atg12 ubiquitin-like conjuga-
tion systems in macroautophagy. Protein modications: beyond the
usual suspectsreview series. EMBO Rep. 2008;9:85964.
110. Fujita N, Hayashi-Nishino M, Fukumoto H, Omori H, Yama-
moto A, Noda T, et al. An Atg4B mutant hampers the lipidation
of LC3 paralogues and causes defects in autophagosome closure.
Mol Biol Cell. 2008;19:46519.
111. Kabeya Y, Mizushima N, Yamamoto A, Oshitani-Okamoto S,
Ohsumi Y, Yoshimori T. LC3, GABARAP and GATE16
localize to autophagosomal membrane depending on form-II
formation. J Cell Sci. 2004;117:280512.
112. Kabeya Y, Mizushima N, Ueno T, Yamamoto A, Kirisako T,
Noda T, et al. LC3, a mammalian homologue of yeast Apg8p, is
localized in autophagosome membranes after processing. EMBO
J. 2000;19:57208.
113. Tsuboyama K, Koyama-Honda I, Sakamaki Y, Koike M, Mor-
ishita H, Mizushima N. The ATG conjugation systems are
important for degradation of the inner autophagosomal mem-
brane. Science. 2016;354:103641.
114. Ichimura Y, Kirisako T, Takao T, Satomi Y, Shimonishi Y,
Ishihara N, et al. A ubiquitin-like system mediates protein lipi-
dation. Nature. 2000;408:48892.
115. Noda T, Fujita N, Yoshimori T. The late stages of autophagy:
how does the end begin? Cell Death Differ. 2009;16:98490.
116. Nakamura S, Yoshimori T. New insights into autophagosome-
lysosome fusion. J Cell Sci. 2017;130:120916.
117. Behrends C, Fulda S. Receptor proteins in selective autophagy.
Int J Cell Biol. 2012;2012:673290.
118. Johansen T, Lamark T. Selective autophagy mediated by
autophagic adapter proteins. Autophagy. 2011;7:27996.
119. Saitoh T, Fujita N, Hayashi T, Takahara K, Satoh T, Lee H, et al.
Atg9a controls dsDNA-driven dynamic translocation of STING
and the innate immune response. Proc Natl Acad Sci USA.
2009;106:208426.
120. Liu D, Wu H, Wang C, Li Y, Tian H, Siraj S, et al. STING
directly activates autophagy to tune the innate immune response.
Cell Death Differ. 2019;26:173549.
121. Prabakaran T, Bodda C, Krapp C, Zhang BC, Christensen MH,
Sun C, et al. Attenuation of cGASSTING signaling is mediated
by a p62/SQSTM1-dependent autophagy pathway activated by
TBK1. EMBO J. 2018;37:e97858.
122. Gui X, Yang H, Li T, Tan X, Shi P, Li M, et al. Autophagy
induction via STING trafcking is a primordial function of the
cGAS pathway. Nature. 2019;567:2626.
123. Liang Q, Seo GJ, Choi YJ, Kwak MJ, Ge J, Rodgers MA, et al.
Crosstalk between the cGAS DNA sensor and Beclin-1 autop-
hagy protein shapes innate antimicrobial immune responses. Cell
Host Microbe. 2014;15:22838.
124. Subramani S, Malhotra V. Non-autophagic roles of autophagy-
related proteins. EMBO Rep. 2013;14:14351.
125. Watson RO, Manzanillo PS, Cox JS. Extracellular M. tubercu-
losis DNA targets bacteria for autophagy by activating the host
DNA-sensing pathway. Cell 2012;150:80315.
126. Dey B, Dey RJ, Cheung LS, Pokkali S, Guo H, Lee JH, et al. A
bacterial cyclic dinucleotide activates the cytosolic surveillance
pathway and mediates innate resistance to tuberculosis. Nat Med.
2015;21:4016.
127. van der Vaart M, Korbee CJ, Lamers GE, Tengeler AC, Hosseini
R, Haks MC, et al. The DNA damage-regulated autophagy
modulator DRAM1 links mycobacterial recognition via TLR-
MYD88 to autophagic defense [corrected]. Cell Host Microbe.
2014;15:75367.
128. Konno H, Konno K, Barber GN. Cyclic dinucleotides trigger
ULK1 (ATG1) phosphorylation of STING to prevent sustained
innate immune signaling. Cell. 2013;155:68898.
129. Zhong B, Zhang L, Lei C, Li Y, Mao AP, Yang Y, et al. The
ubiquitin ligase RNF5 regulates antiviral responses by mediating
degradation of the adaptor protein MITA. Immunity.
2009;30:397407.
130. Wang Y, Lian Q, Yang B, Yan S, Zhou H, He L, et al. TRI-
M30alpha is a negative-feedback regulator of the intracellular
DNA and DNA virus-triggered response by targeting STING.
PLoS Pathog. 2015;11:e1005012.
131. Collins AC, Cai H, Li T, Franco LH, Li XD, Nair VR, et al.
Cyclic GMP-AMP Synthase Is an Innate Immune DNA Sensor
A. M. V. Murthy et al.
for Mycobacterium tuberculosis. Cell Host Microbe. 2015;17:
8208.
132. Thomsen MK, Nandakumar R, Stadler D, Malo A, Valls RM,
Wang F, et al. Lack of immunological DNA sensing in hepatocytes
facilitates hepatitis B virus infection. Hepatology. 2016;64:74659.
133. Lei Z, Deng M, Yi Z, Sun Q, Shapiro RA, Xu H, et al. cGAS-
mediated autophagy protects the liver from ischemia-reperfusion
injury independently of STING. Am J Physiol Gastrointest Liver
Physiol. 2018;314:G655G67.
134. Moretti J, Roy S, Bozec D, Martinez J, Chapman JR, Ueberheide
B, et al. STING senses microbial viability to orchestrate stress-
mediated autophagy of the endoplasmic reticulum. Cell.
2017;171:80923 e13.
135. Denton D, Kumar S. Autophagy-dependent cell death. Cell
Death Differ. 2019;26:60516.
136. Klionsky DJ, Abdelmohsen K, Abe A, Abedin MJ, Abeliovich
H, Acevedo Arozena A. et al. Guidelines for the use and inter-
pretation of assays for monitoring autophagy (3rd edition).
Autophagy. 2016;12:1222.
137. Denton D, Aung-Htut MT, Kumar S. Developmentally pro-
grammed cell death in Drosophila. Biochim Biophys Acta.
2013;1833:3499506.
138. Denton D, Shravage B, Simin R, Mills K, Berry DL, Baehrecke
EH, et al. Autophagy, not apoptosis, is essential for midgut cell
death in Drosophila. Curr Biol 2009;19:17416.
139. Liu Y, Shoji-Kawata S, Sumpter RM Jr., Wei Y, Ginet V,
Zhang L, et al. Autosis is a Na+,K+-ATPase-regulated form
of cell death triggered by autophagy-inducing peptides, starva-
tion, and hypoxia-ischemia. Proc Natl Acad Sci USA.
2013;110:2036471.
140. Arakawa S, Tsujioka M, Yoshida T, Tajima-Sakurai H, Nishida
Y, Matsuoka Y, et al. Role of Atg5-dependent cell death in the
embryonic development of Bax/Bak double-knockout mice. Cell
Death Differ. 2017;24:1598608.
141. Margolis SR, Wilson SC, Vance RE. Evolutionary origins of
cGASSTING signaling. Trends Immunol. 2017;38:73343.
142. Nassour J, Radford R, Correia A, Fuste JM, Schoell B, Jauch A,
et al. Autophagic cell death restricts chromosomal instability
during replicative crisis. Nature. 2019;565:65963.
143. Ku JWK, Chen Y, Lim BJW, Gasser S, Crasta KC, Gan YH.
Bacterial-induced cell fusion is a danger signal triggering cGAS-
STING pathway via micronuclei formation. Proc Natl Acad Sci
USA. 2020;117:1592334.
144. Lin L, Baehrecke EH. Autophagy, cell death, and cancer. Mol
Cell Oncol. 2015;2:e985913.
145. Samir P, Malireddi RKS, Kanneganti TD. The PANoptosome: a
deadly protein complex driving pyroptosis, apoptosis, and
necroptosis (PANoptosis). Front Cell Infect Microbiol.
2020;10:238.
Crosstalk between cGASSTING signaling and cell death
... MnP treatment also upregulated interferon-1 beta (IFN-1β) expression ( Figure S3). It is recognized that cGAS-STING/type I IFN plays a key role in promoting immunogenic cell death (ICD) and apoptosis [29]. ...
... While cGAS-STING signaling inhibitor C176 significantly abrogated cisplatin-induced RGCs cell apoptosis, indicating a crucial role of cGAS-STING in regulating cisplatin caused DNA damage in RGCs. Besides the diverse roles in microbial infections, cGAS-STING signaling was found in multiple pathological processes, such as cell death, tumors, as well as inflammation [32][33][34]. Additionally, mitochondrial DNA damage causes inflammation by activation of cGAS-STING signaling in acute kidney injury [34]. ...
Article
Background: Glaucoma is an optic neurodegenerative disease. Retinal ganglion cells (RGCs) are the fundamental neurons in the trabecular meshwork, and their loss is the main pathological reason for glaucoma. The present study was to investigate mechanisms that regulate RGCs survival. Methods: A mouse model of glaucoma was established by injecting hypertonic saline into the limbal veins. RGCs apoptosis was detected by using flow cytometry. Protein expressions in RGCs in response to DNA damage inducer cisplatin treatment were detected by immunofluorescence and western blot. The expressions of inflammatory cytokines were determined using ELISA and real-time PCR. Results: In the hypertonic saline-injected mice, we found visual function was impaired followed by the increased expression of γH2AX and activation of cGAS-STING signaling. We found that DNA damage inducer cisplatin treatment incurred significant DNA damage, cell apoptosis, and inflammatory response. Mechanistically, cisplatin treatment triggered activation of the cGAS-STING signaling by disrupting mitochondrial function. Suppression of cGAS-STING ameliorated inflammation and protected visual function in glaucoma mice. Conclusions: The data demonstrated that cGAS-STING signaling is activated in the damaged retinal ganglion cells, which is associated with increased inflammatory responses, DNA damage, and mitochondrial dysfunction. Targeting the cGAS-STING signaling pathway represents a potential way to alleviate glaucoma-related visual function.
... Inflammation and autoimmunity play significant roles in the development of diabetes, and hence, targeting the inflammatory response has shown therapeutic benefits (79, 80). The NOD signaling pathway is involved in the inflammation triggered by the cGAS-STING pathway (81,82). Perturbations in the gut microbiota in diabetic patients can, in turn, activate the cGAS-STING-NOD pathway, leading to inflammation (83). ...
Article
Full-text available
Introduction With the increasing prevalence of type 2 diabetes mellitus (T2DM), there is an urgent need to discover effective therapeutic targets for this complex condition. Coding and non-coding RNAs, with traditional biochemical parameters, have shown promise as viable targets for therapy. Machine learning (ML) techniques have emerged as powerful tools for predicting drug responses. Method In this study, we developed an ML-based model to identify the most influential features for drug response in the treatment of type 2 diabetes using three medicinal plant-based drugs (Rosavin, Caffeic acid, and Isorhamnetin), and a probiotics drug (Z-biotic), at different doses. A hundred rats were randomly assigned to ten groups, including a normal group, a streptozotocin-induced diabetic group, and eight treated groups. Serum samples were collected for biochemical analysis, while liver tissues (L) and adipose tissues (A) underwent histopathological examination and molecular biomarker extraction using quantitative PCR. Utilizing five machine learning algorithms, we integrated 32 molecular features and 12 biochemical features to select the most predictive targets for each model and the combined model. Results and discussion Our results indicated that high doses of the selected drugs effectively mitigated liver inflammation, reduced insulin resistance, and improved lipid profiles and renal function biomarkers. The machine learning model identified 13 molecular features, 10 biochemical features, and 20 combined features with an accuracy of 80% and AUC (0.894, 0.93, and 0.896), respectively. This study presents an ML model that accurately identifies effective therapeutic targets implicated in the molecular pathways associated with T2DM pathogenesis.
... Another intracellular pathway involved in sensing and mediating ER stress is the pathway of cGAS (cyclic GMP-AMP synthase) and STING (stimulator of interferon genes). The cGAS-STING pathway is known for its nucleic acid-sensing ability in the cytoplasm, mediated through cGAS (Motwani et al. 2019;Murthy et al. 2020). cGAS in turn activates STING through a second messenger. ...
Article
Full-text available
Previous studies have demonstrated a brain volume decrease linked to long-term starvation in patients with anorexia nervosa (AN). Food intake is critically diminished in this disorder, leading to one of the highest mortality rates within the psychiatric disease spectrum. As reported in animal models, astrocytes seem to be the most affected cell type in AN. In a recently established primary cell culture model, an elevated unfolded protein response (UPR) was observed in long-term glucose semi-starved astrocytes. A well-functioning protein machinery is essential for every cell, and prolonged UPR will lead to cell death. As a nucleic acid stress-sensing pathway with the activator located in the endoplasmic reticulum, the regulation of the cGAS-STING pathway (cyclic GMP-AMP synthase/stimulator of interferon genes) was additionally investigated in the starvation context. In the current study, a glucose semi-starvation protocol of 15 days, during which cells were supplied with 2 mM glucose in the medium, was prolonged with an additional 6-day long recovery period. Our findings showed that increased UPR mRNA expression was reversible after re-establishing the standard glucose concentration of 25 mM. Furthermore, we were able to verify the presence of cGAS and STING in astrocytes with a characteristic presence of cGAS in the astrocyte nucleus during starvation. A correlation between STING and the glial fibrillary acidic protein (GFAP) could be established, hinting at a conditional presence of STING with a specific astrocyte phenotype. Graphical Abstract
... Binding of DNA to the cGAS induces conformational changes to cGAS, which in turn catalyzes the formation of 2′,3′-cGAMP, a cyclic di-nucleotide (CDN) with a unique phosphodiester linkage. The generated cGAMP acts as a secondary messenger and activates STING and downstream proteins to initiate inflammatory response [35]. Finally, the downstream inflammatory indicators of STING, including interleukin-6 (IL-6), tumor necrosis factor alpha (TNF-α), and interleukin-1β (IL-1β) were detected and the trends were consistent with Western blot (WB) results ( Fig. 3H-J). ...
Article
Full-text available
Excessive production of reactive oxygen species (ROS) and inflammation are the key problems that impede diabetic wound healing. In particular, dressings with ROS scavenging capacity play a crucial role in the process of chronic wound healing. Herein, Zr-based large-pore mesoporous metal–organic frameworks (mesoMOFs) were successfully developed for the construction of spatially organized cascade bioreactors. Natural superoxide dismutase (SOD) and an artificial enzyme were spatially organized in these hierarchical mesoMOFs, forming a cascade antioxidant defense system, and presenting efficient intracellular and extracellular ROS scavenging performance. In vivo experiments demonstrated that the SOD@HMUiO-MnTCPP nanoparticles (S@M@H NPs) significantly accelerated diabetic wound healing. Transcriptomic and western blot results further indicated that the nanocomposite could inhibit fibroblast senescence and ferroptosis as well as the stimulator of interferon genes (STING) signaling pathway activation in macrophages mediated by mitochondrial oxidative stress through ROS elimination. Thus, the biomimetic multi-enzyme cascade catalytic system with spatial ordering demonstrated a high potential for diabetic wound healing, where senescence, ferroptosis, and STING signaling pathways may be potential targets. Graphical Abstract Schematic diagram showing the proposed mechanism that S@M@H NPs promoted diabetic wound healing
... These data suggested that the cGAS-STING pathway is activated under mechanical stress in the KS pathological setting. Besides mediating IFN-dependent inflammatory response, cGAS-STING activates different evolutionarily ancient defense programs, including non-canonical autophagy and NLRP3-mediated inflammasome, which leads to different DNA-stimulated programmed cell death (PCD) pathways [49][50][51][52] . Indeed, we found that the executors of PCD pathways BAX/BAD 49,[53][54][55] resulted activated in MLL4 Q4092X MSCs grown on stiff matrix (Fig. 7c and Extended Data Fig. 7i). ...
Preprint
Cells and tissue integrity is constantly challenged by the necessity to adapt and respond to mechanical loads. Among the cellular components, the nucleus possesses mechano-sensing and mechanotransduction capabilities, yet the molecular mechanisms involved remains poorly defined. We postulated that the mechanical properties of the chromatin and its compartmentalization into condensates contribute to the nuclear adaptation to external forces, while preserving its integrity. By interrogating the effects of MLL4 loss-of-function in Kabuki Syndrome, we found that the balancing of transcriptional and Polycomb condensates tunes the nuclear responsiveness to external mechanical forces. We showed that MLL4 acts as a chromatin mechano-sensor by clustering into condensates through its Prion-like domain, and its response was regulated by the chromatin context. Furthermore, the machano-sensing activity of MLL4 condensates is instrumental to withstand the physical challenges that nuclei experience during cell confinement and migration by preserving their integrity. In Kabuki Syndrome persistent rupture of nuclear envelope triggers cGAS-STING activation, which leads to programmed cell death. Ultimately, these results demonstrate the critical role chromatin compartments play in mechano-responses and how they impact pathological conditions by stimulating cGAS-STING signaling.
Article
Full-text available
Programmed cell death is regulated by evolutionarily conserved pathways that play critical roles in development and the immune response. A newly recognized pathway for proinflammatory programmed cell death called PANoptosis is controlled by a recently identified cytoplasmic multimeric protein complex named the PANoptosome. The PANoptosome can engage, in parallel, three key modes of programmed cell death—pyroptosis, apoptosis, and necroptosis. The PANoptosome components have been implicated in a wide array of human diseases including autoinflammatory diseases, neurodegenerative diseases, cancer, microbial infections, and metabolic diseases. Here, we review putative components of the PANoptosome and present a phylogenetic analysis of their molecular domains and interaction motifs that support complex assembly. We also discuss genetic data that suggest PANoptosis is coordinated by scaffolding and catalytic functions of the complex components and propose mechanistic models for PANoptosome assembly. Overall, this review presents potential mechanisms governing PANoptosis based on evolutionary analysis of the PANoptosome components.
Article
Full-text available
Significance Burkholderia pseudomallei is a bacterial pathogen that causes melioidosis, an infectious disease in the tropics with high morbidity and mortality. It has a unique property among bacteria: to fuse infected host cells. We found that our immune system detects bacterial- or chemical-induced host cell fusion as a danger signal. Abnormal cell fusion leads to genomic instability and formation of micronuclei. This triggers the host to activate a signaling pathway leading to a form of cell death known as autophagic death, which likely serves to limit abnormal cellular transformation.
Article
Full-text available
Melanoma is the most aggressive type of skin cancer and resistance to the conventional chemotherapy is the major cause for its poor prognosis. Metabolic perturbations leading to increased production of reactive oxygen species activate NRF2-dependent anti-oxidative responses to survive oxidative stress. This protective function of NRF2 is the primary cause for therapy resistance in cancer as anti-cancer agents such as BRAF inhibitors also induce NRF2-dependent antioxidative response. We had reported that type I interferons produced upon activation of STING, abrogates NRF2 function. Therefore, we investigated if STING agonists such as the newly developed dimeric aminobenzimidazole (diABZI) could sensitize melanoma cells to the clinically used BRAF inhibitors. Our results reveal that pharmacological activation of STING by diABZI, down regulates NRF2-dependent anti-oxidative responses and potentiates cell-death in melanoma cells when used in combination with BRAF inhibitors.
Article
Full-text available
Stimulator of Interferon Genes (STING) is a critical component of host innate immune defense but can contribute to chronic autoimmune or autoinflammatory disease. Once activated, the cyclic guanosine monophosphate (GMP)-adenosine monophosphate (AMP) (cGAMP) synthase (cGAS)-STING pathway induces both type I interferon (IFN) expression and nuclear factor-κB (NF-κB)-mediated cytokine production. Currently, these two signaling arms are thought to be mediated by a single upstream kinase, TANK-binding kinase 1 (TBK1). Here, using genetic and pharmacological approaches, we show that TBK1 alone is dispensable for STING-induced NF-κB responses in human and mouse immune cells, as well as in vivo. We further demonstrate that TBK1 acts redundantly with IκB kinase ε (IKKε) to drive NF-κB upon STING activation. Interestingly, we show that activation of IFN regulatory factor 3 (IRF3) is highly dependent on TBK1 kinase activity, whereas NF-κB is significantly less sensitive to TBK1/IKKε kinase inhibition. Our work redefines signaling events downstream of cGAS-STING. Our findings further suggest that cGAS-STING will need to be targeted directly to effectively ameliorate the inflammation underpinning disorders associated with STING hyperactivity.
Article
Full-text available
Activation of cyclic GMP-AMP (cGAMP) synthase (cGAS) plays a critical role in antiviral responses to many DNA viruses. Sensing of cytosolic DNA by cGAS results in synthesis of the endogenous second messenger cGAMP that activates stimulator of interferon genes (STING) in infected cells. Critically, cGAMP can also propagate antiviral responses to uninfected cells through intercellular transfer, although the modalities of this transfer between epithelial and immune cells remain poorly defined. We demonstrate here that cGAMP-producing epithelial cells can transactivate STING in cocultured macrophages through direct cGAMP transfer. cGAMP transfer was reliant upon connexin expression by epithelial cells and pharmacological inhibition of connexins blunted STING -dependent transactivation of the macrophage compartment. Macrophage transactivation by cGAMP contributed to a positive-feedback loop amplifying antiviral responses, significantly protecting uninfected epithelial cells against viral infection. Collectively, our findings constitute the first direct evidence of a connexin-dependent cGAMP transfer to macrophages by epithelial cells, to amplify antiviral responses. IMPORTANCE Recent studies suggest that extracellular cGAMP can be taken up by macrophages to engage STING through several mechanisms. Our work demonstrates that connexin-dependent communication between epithelial cells and macrophages plays a significant role in the amplification of antiviral responses mediated by cGAMP and suggests that pharmacological strategies aimed at modulating connexins may have therapeutic applications to control antiviral responses in humans.
Article
Full-text available
Cyclic GMP-AMP synthase (cGAS) is a cytosolic nucleic acid sensor that can bind to dsDNA. It maintains an autoinhibited state in the absence of cytosolic dsDNA, while when activated, it in turn activates its adaptor protein STING, ultimately triggering a cascade that produces inflammatory cytokines and type I interferons (IFNs). With further research, additional types of nucleic acids have been found to be activators of the cGAS-STING pathway. The cGAS-STING pathway can provide protection or resistance against infections; however, improper or overactivation might cause severe inflammatory pathologies, including autoimmunity. This article systematically reviews the latest research progress on the axis, including categorical pathway triggers, the connection with autoimmune disease and drug therapy progress.
Article
Type I interferon (IFN) response is commonly recognized as the main signaling activity of STING. Here, we generate the Sting1S365A/S365A mutant mouse that precisely ablates IFN-dependent activities while preserving IFN-independent activities of STING. StingS365A/S365A mice protect against HSV-1 infection, despite lacking the STING-mediated IFN response. This challenges the prevailing view and suggests that STING controls HSV-1 infection through IFN-independent activities. Transcriptomic analysis reveals widespread IFN-independent activities of STING in macrophages and T cells, and STING activities in T cells are predominantly IFN independent. In mouse tumor models, T cells in the tumor experience substantial cell death that is in part mediated by IFN-independent activities of STING. We found that the tumor induces STING-mediated cell death in T cells to evade immune control. Our data demonstrate that mammalian STING possesses widespread IFN-independent activities that are important for restricting HSV-1 infection, tumor immune evasion and likely also adaptive immunity.
Article
The detection of pathogens through nucleic acid sensors is a defining principle of innate immunity. RNA-sensing and DNA-sensing receptors sample subcellular compartments for foreign nucleic acids and, upon recognition, trigger immune signalling pathways for host defence. Over the past decade, our understanding of how the recognition of nucleic acids is coupled to immune gene expression has advanced considerably, particularly for the DNA-sensing receptor cyclic GMP–AMP synthase (cGAS) and its downstream signalling effector stimulator of interferon genes (STING), as well as the molecular components and regulation of this pathway. Moreover, the ability of self-DNA to engage cGAS has emerged as an important mechanism fuelling the development of inflammation and implicating the cGAS–STING pathway in human inflammatory diseases and cancer. This detailed mechanistic and biological understanding is paving the way for the development and clinical application of pharmacological agonists and antagonists in the treatment of chronic inflammation and cancer. The cGAS–STING pathway forms a major DNA-sensing mechanism in mammalian cells. This Review summarizes the current knowledge of cGAS–STING signalling and regulation, highlighting emerging evidence of its role in monogenic inflammatory diseases and cancer and new therapeutic opportunities.
Article
A nuclear sensor of viral DNA? A signaling pathway in eukaryotes known as cGAS–STING recognizes the presence of cytosolic DNA, which alerts the immune system to viral infection or cellular damage. However, the majority of DNA viruses direct their genomic DNA into nuclei, suggesting that nuclear-specific sensing is also needed. L. Wang et al. find that during herpes simplex virus–1 infection, heterogeneous nuclear ribonucleoprotein A2B1 forms a complex with viral DNA, homodimerizes, and is demethylated. These events result in translocation of the complex to the cytosol and activation of the immune system through type I interferon signaling. Additionally, the complex promotes N ⁶ -methyladenosine modification and translocation of cGAS–STING–related mRNAs after DNA virus infection, further amplifying the immune response. Science , this issue p. eaav0758
Article
Pathogenic and other cytoplasmic DNAs activate the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway to induce inflammation via transcriptional activation by IRF3 and nuclear factor κB (NF-κB), but the functional consequences of exposing cGAS to chromosomes upon mitotic nuclear envelope breakdown are unknown. Here, we show that nucleosomes competitively inhibit DNA-dependent cGAS activation and that the cGAS-STING pathway is not effectively activated during normal mitosis. However, during mitotic arrest, low level cGAS-dependent IRF3 phosphorylation slowly accumulates without triggering inflammation. Phosphorylated IRF3, independently of its DNA-binding domain, stimulates apoptosis through alleviating Bcl-xL-dependent suppression of mitochondrial outer membrane permeabilization. We propose that slow accumulation of phosphorylated IRF3, normally not sufficient for inducing inflammation, can trigger transcription-independent induction of apoptosis upon mitotic aberrations. Accordingly, expression of cGAS and IRF3 in cancer cells makes mouse xenograft tumors responsive to the anti-mitotic agent Taxol. The Cancer Genome Atlas (TCGA) datasets for non-small cell lung cancer patients also suggest an effect of cGAS expression on taxane response.