Cytosolic DNA is recognized by the innate immune system as a potential threat. During apoptotic cell death, mitochondrial DNA (mtDNA) release activates the DNA sensor cyclic GMP–AMP synthase (cGAS) to promote a pro-inflammatory type I interferon response. Inflammation following mtDNA release during apoptotic cell death can be exploited to engage anti-tumor immunity and represents a potential avenue for cancer therapy. Additionally, various studies have described leakage of mtDNA, independent of cell death, with different underlying cues such as pathogenic infections, changes in mtDNA packaging, mtDNA stress or reduced mitochondrial clearance. The interferon response in these scenarios can be beneficial but also potentially disadvantageous, as suggested by a variety of disease phenotypes. In this review, we discuss mechanisms underlying mtDNA release governed by cell death pathways and summarize release mechanisms independent of cell death. We further highlight the similarities and differences in mtDNA release pathways, outlining gaps in our knowledge and questions for further research. Together, a deeper understanding of how and when mtDNA is released may enable the development of drugs to specifically target or inhibit mtDNA release in different disease settings.

Mitochondria are distinct, highly dynamic cellular organelles with myriad functions including, metabolism, energy production, cell death and immune signaling [1,2]. Many of these functions require the double-membrane system compartmentalizing the mitochondria into intermembrane space (IMS) and matrix, separated by an outer and inner mitochondrial membrane (OMM and IMM). Alongside double-membrane compartmentalization, the origin of mitochondria as an archaebacterial endosymbiont also imparts the organelle with its own double stranded, circular genome able to replicate, in the matrix, independently of the nuclear genome [3]. Mitochondria are essential for mitochondrial (intrinsic) apoptosis. Particularly during extrinsic apoptosis [4–8] and intracellular death complexes [9–12] mitochondria can amplify the initial signal leading to cell death. During pyroptosis mitochondria can supply a ligand for inflammasome activation (Figure 1). However, the role of mitochondria in other types of cell death such as necroptosis and ferroptosis is less clear (Figure 1) (see review [13]).

Role of mitochondria in cell death.

Figure 1.
Role of mitochondria in cell death.

Apoptosis: Mitochondrial apoptosis is held in check by the balance of pro- and anti-apoptotic proteins. An apoptotic trigger promotes BAX BAK oligomerization and lipid pore formation in the mitochondrial outer membrane — mitochondrial outer membrane permeabilisation (MOMP), apoptotosome formation (cyto c, APAF1 and caspase-9) in the cytosol and activation of executioner caspases-3 and -7 enforcing cell death. The intermembrane space protein SMAC binds to and inhibits XIAP thereby relieving the brake on caspase-9 and -3 activity. Activation of caspase-8 results in caspase-3 activation and in type II cells, as well as after activation of the ripoptosome and necrosome; apoptotic cell death requires caspase-8 mediated cleavage of BID, mitochondrial outer membrane permeabilization and mitochondrial apoptosis. Pyroptosis: mtDNA and mtROS can act as ligands for the inflammasome receptor NLRP3. Activation of the inflammasome platform (receptor, ASC, caspase-1), allows autoproteolytic cleavage and activation of caspase-1, thus promoting cleavage of GSDMD. The N-terminal domain of GSDMD (GSDMDNT) inserts into the plasma membrane — promoting pyroptotic cell death. GSDMDNT also forms pores in the mitochondrial membrane. While caspase-3 cleaves GSDMD in its N-terminus (amino acid D87 mouse) deactivating the N-terminal fragment, caspase-8 cleavage of GSDMD results in an active GSDMDNT. Activation of caspase-3 can additionally cleave GSDME yielding an active N-terminal fragment promoting pore formation in the mitochondrial and plasma membrane. Necroptosis: Mitochondria are not required for the execution of necroptosis, however, mtROS can promote RIP1 kinase phosphorylation and thus promote the initial steps of necroptosis. Ferroptosis: Mitochondria impact ferroptosis in at least two ways; firstly, promoting ferroptosis due to mtROS oxidizing mitochondrial lipids and secondly dampening ferroptosis by chelating iron (Abbreviations: MOMP, mitochondrial outer membrane permabilization; APAF1, apoptotic protease-activating factor1; cyto c, cytochrome c; SMAC, second mitochondria-derived activator of caspases; XIAP, X-linked inhibitor of apoptosis protein; BID, BH3-interacting domain death agonist; mtDNA, mitochondrial DNA; mtROS, mitochondrial reactive oxygen species; NLRP3, NACHT, LRR and PYD domains-containing protein 3; ASC, apoptosis-associated speck-like containing a card; GSDMD, gasdermin-D; RIP1, receptor-interacting protein 1).

Figure 1.
Role of mitochondria in cell death.

Apoptosis: Mitochondrial apoptosis is held in check by the balance of pro- and anti-apoptotic proteins. An apoptotic trigger promotes BAX BAK oligomerization and lipid pore formation in the mitochondrial outer membrane — mitochondrial outer membrane permeabilisation (MOMP), apoptotosome formation (cyto c, APAF1 and caspase-9) in the cytosol and activation of executioner caspases-3 and -7 enforcing cell death. The intermembrane space protein SMAC binds to and inhibits XIAP thereby relieving the brake on caspase-9 and -3 activity. Activation of caspase-8 results in caspase-3 activation and in type II cells, as well as after activation of the ripoptosome and necrosome; apoptotic cell death requires caspase-8 mediated cleavage of BID, mitochondrial outer membrane permeabilization and mitochondrial apoptosis. Pyroptosis: mtDNA and mtROS can act as ligands for the inflammasome receptor NLRP3. Activation of the inflammasome platform (receptor, ASC, caspase-1), allows autoproteolytic cleavage and activation of caspase-1, thus promoting cleavage of GSDMD. The N-terminal domain of GSDMD (GSDMDNT) inserts into the plasma membrane — promoting pyroptotic cell death. GSDMDNT also forms pores in the mitochondrial membrane. While caspase-3 cleaves GSDMD in its N-terminus (amino acid D87 mouse) deactivating the N-terminal fragment, caspase-8 cleavage of GSDMD results in an active GSDMDNT. Activation of caspase-3 can additionally cleave GSDME yielding an active N-terminal fragment promoting pore formation in the mitochondrial and plasma membrane. Necroptosis: Mitochondria are not required for the execution of necroptosis, however, mtROS can promote RIP1 kinase phosphorylation and thus promote the initial steps of necroptosis. Ferroptosis: Mitochondria impact ferroptosis in at least two ways; firstly, promoting ferroptosis due to mtROS oxidizing mitochondrial lipids and secondly dampening ferroptosis by chelating iron (Abbreviations: MOMP, mitochondrial outer membrane permabilization; APAF1, apoptotic protease-activating factor1; cyto c, cytochrome c; SMAC, second mitochondria-derived activator of caspases; XIAP, X-linked inhibitor of apoptosis protein; BID, BH3-interacting domain death agonist; mtDNA, mitochondrial DNA; mtROS, mitochondrial reactive oxygen species; NLRP3, NACHT, LRR and PYD domains-containing protein 3; ASC, apoptosis-associated speck-like containing a card; GSDMD, gasdermin-D; RIP1, receptor-interacting protein 1).

Close modal

In this review, we focus on signaling pathways specifically induced by the presence of mitochondrial DNA (mtDNA) in the cytosol as a consequence of mitochondrial damage. In addition, attention will be centered on cell death pathways promoting mtDNA release and how this release is mediated.

Eukaryotic cells contain a highly elaborate, organellar system ensuring cellular homeostasis. DNA resides in two distinct organelles, nuclei and mitochondria. The presence of DNA in other compartments indicates one of two things: damaged or misfunctioning nucleus and/or mitochondria or the presence of foreign DNA. DNA is detected by DNA sensors in the cytosol or in endocytic vesicles (Toll-like receptor 9 — TLR9) to alert the cell's immune system to misplaced or foreign DNA. The activation of the DNA sensor will govern, in turn, the activation of the signaling pathway and immune response. Two cytosolic pattern recognition receptors and one DNA sensor have specifically been associated with mtDNA detection, the absent in melanoma 2 (AIM2), the NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) and cyclic GMP–AMP synthase (cGAS).

mtDNA activates cGAS-STING signaling pathway

cGAS is a DNA-binding protein localized to the cytosol as well as the nucleus [14,15]. While nuclear cGAS activity is tightly held in check, to avoid an unwanted inflammatory response [16–24], binding of dsDNA in the cytosol frees the catalytic site of cGAS, allowing its activation. Active cGAS then synthesizes 2′,3′ GMP–AMP (cGAMP) using ATP and GTP to act as a second messenger activating the endoplasmic reticulum (ER) membrane adaptor STING upon binding [25–28]. The active dimeric STING relocates from the ER to the Golgi compartment, recruits TBK1 which subsequently phosphorylates STING at its C-terminal tail (CTT). Phosphorylated STING acts as a platform for the recruitment and phosphorylation of interferon regulatory factor 3 (IRF3) by TBK1 [29]. Phosphorylated IRF3 (pIRF3) dimerizes and translocates to the nucleus to drive type I interferon transcription (Figure 2). In this review, we specifically highlight the signaling pathways mediated by cytosolic mtDNA promoting cGAS-STING activation resulting in IRF3 but it is important to note that activation of cGAS has also been implicated in the induction of other transcriptional responses mediated by NF-κB [28,30], mitogen-activated protein kinases (MAPK) and STAT factors [31]. Activation of these signaling pathways ensures the transcription of a wide variety of proteins including pro-inflammatory cytokines and type I interferons.

Signaling pathways activated upon mitochondrial DNA recognition in the cytosol.

Figure 2.
Signaling pathways activated upon mitochondrial DNA recognition in the cytosol.

(a) Cytosolic mtDNA binds and activates cGAS inducing the production of the second messenger cGAMP and dimerization of STING. STING recruits TBK1 to phosphorylate STING, itself as well as the transcription factor IRF3, thus enabling transcription of pro-inflammatory target genes via NF-κB and IRF3 in the nucleus. (b) MtDNA activates the NLRP3 and potentially AIM2 inflammasome receptor which act as a platform for caspase-1 activation and induction of pyroptotic cell death via GSDMD pore formation in the plasma membrane. (c) TLR9 recognizes mtDNA in the endosomal compartment and relays the signaling to the adaptor protein MyD88-IRAK1/IRAK4-TRAF6 and IRF7, TAK1-IκKBα or TAK1-MAPK1 which in turn promotes pro-inflammatory gene expression via activation of the transcription factor IRF7, NF-κB or AP-1 (Abbreviations: TLR9, Toll-like receptor 9; mtDNA, mitochondrial DNA; MyD88, myeloid differentiation primary response protein; IRAK, interleukin-1 receptor-associated kinase; TRAF6, TNF receptor-associated factor 6; MAPK, mitogen-activated protein kinase 1; IRF7, interferon regulatory factor 7; cGAMP, cyclic GMP–AMP; cGAS, cyclic GMP–AMP synthase; STING, stimulator of interferon genes protein; IRF3, Inteferon regulatory factor 3; AIM2, absent in melanoma 2; NLRP3, NACHT, LRR and PYD domains-containing protein 3).

Figure 2.
Signaling pathways activated upon mitochondrial DNA recognition in the cytosol.

(a) Cytosolic mtDNA binds and activates cGAS inducing the production of the second messenger cGAMP and dimerization of STING. STING recruits TBK1 to phosphorylate STING, itself as well as the transcription factor IRF3, thus enabling transcription of pro-inflammatory target genes via NF-κB and IRF3 in the nucleus. (b) MtDNA activates the NLRP3 and potentially AIM2 inflammasome receptor which act as a platform for caspase-1 activation and induction of pyroptotic cell death via GSDMD pore formation in the plasma membrane. (c) TLR9 recognizes mtDNA in the endosomal compartment and relays the signaling to the adaptor protein MyD88-IRAK1/IRAK4-TRAF6 and IRF7, TAK1-IκKBα or TAK1-MAPK1 which in turn promotes pro-inflammatory gene expression via activation of the transcription factor IRF7, NF-κB or AP-1 (Abbreviations: TLR9, Toll-like receptor 9; mtDNA, mitochondrial DNA; MyD88, myeloid differentiation primary response protein; IRAK, interleukin-1 receptor-associated kinase; TRAF6, TNF receptor-associated factor 6; MAPK, mitogen-activated protein kinase 1; IRF7, interferon regulatory factor 7; cGAMP, cyclic GMP–AMP; cGAS, cyclic GMP–AMP synthase; STING, stimulator of interferon genes protein; IRF3, Inteferon regulatory factor 3; AIM2, absent in melanoma 2; NLRP3, NACHT, LRR and PYD domains-containing protein 3).

Close modal

mtDNA can activate the NLRP3 inflammasome

Activation of the cytosolic pattern recognition receptor NLRP3 leading to inflammasome activation and pyroptosis has been linked to cytosolic mtDNA [32–34]. How the NLRP3 receptor is activated remains unclear, often — but not always — it is linked to sensing potassium efflux [35]. Interestingly, mitochondrial ROS (mtROS) [36,37] as well as leakage of oxidized mtDNA was suggested as an activator of the NLRP3 inflammasome [32–34] (Figure 2). The release of mtROS/mtDNA has been proposed to depend on VDAC (VDAC1 and 2) [36] and the mitochondrial permeability transition pore (mPTP), an unselective (and poorly defined) Ca2+ regulated pore in the IMM [32,34,38–40]. How mtROS and mtDNA are detected by the NLRP3 inflammasome remains ambiguous as no DNA recognition/binding domains in the NLRP3 receptor have been identified. One possibility for receptor activation may relate to the leucine-rich repeat domain (LRR) which was proposed to recognize RNA : DNA hybrids during enterohemorrhagic Escherichia coli infections [41]. Of note, the bona fide DNA sensor AIM2 can possibly also recognize cytosolic mtDNA [42].

mtDNA can activate TLR9

In contrast with to cytosolic DNA, the presence of mtDNA in endosomes is detected by a different receptor, TLR9, predominantly expressed in B-cells, plasma cells, Paneth cells and microglial cells [43]. TLR9 drives expression of type I interferons and pro-inflammatory cytokines upon activation of the myeloid differentiation primary response 88 protein (MyD88) which signals through interferon regulatory factor 7 (IRF7), MAPK or NF-kB signaling pathways [44,45]. TLR9 detects specifically unmethylated cytidine-phosphate-guanosine (CpG) dinucleotides, DNA motifs that are frequently found in bacterial DNA (Figure 2) [43,46,47]. The mtDNA can stem various cell types including neutrophils or hepatocytes. Neutrophils release mtDNA due to accumulated oxidized nucleoids or during neutrophil extracellular trap (NET) formation [48–50]. Whereas the mtDNA released by hepatocytes is important during nonalcoholic steatohepatitis (NASH), where the mtDNA is taken up by macrophages inducing inflammation via TLR9 signaling [50–53].

Cell death is important during development, for homeostasis as well as for the clearance of pathogens and destruction of transformed cells. Mitochondria are implicated in many functions ensuring cellular maintenance and survival; thus it is perhaps not surprising that mitochondria and cell death are tightly linked. Interestingly, the activation of two contrasting cell death pathways apoptosis and pyroptosis can result in the release of mtDNA.

mtDNA release during apoptosis

Apoptosis is divided into extrinsic and intrinsic apoptotic cell death depending on the origin of cell death ligand. Activation of apoptosis from the cell exterior — extrinsic apoptosis — is induced upon binding of cognate ligands to death receptors [4–7], as well as the assembly of intracellular death complexes such as complex IIa, IIb [9,10], ripoptosome or necrosome [11,12] may relay (via tBID) or even require MOMP to drive cell death [8] (Figure 1). Mitochondrial apoptosis is established as an immunologically silent type of cell death [54]. Under homeostasis, apoptosis is held in check by a balance of anti- and pro-apoptotic proteins of the BCL-2 protein family [55]. Upon an apoptotic stimulus such as DNA damage, ER stress, hypoxia or metabolic stress, BH3-only proteins bind pro-survival BCL-2 proteins and pro-apoptotic BAX and BAK proteins thereby activating the latter [56–60]. This enables cytosolic BAX to translocate and embed into the OMM, where BAX and BAK oligomerize to form pores, causing MOMP and release of soluble IMS proteins such as cyto c [61–64]. The presence of cyto c in the cytosol allows the formation of the apoptosome, a protein complex consisting of APAF1, cyto c, inactive caspase-9, enabling activation of caspase-9 and driving apoptotic cell death via caspase-3/-7 activation [13].

Interestingly, while caspase activity is important during development for cell clearance and cellular organization e.g. in the brain [65,66], APAF1 and caspase-9, -3 and -7 only play a subordinate role compared with BAX/BAK in cellular homeostasis. In particular, APAF1 and caspase-9 deficient cells provide a short-term resistance to apoptotic stimuli, while BAX/BAK deficiency grants full protection [56,67–70]. Similarly, BAX and BAK play a superior role in the clearance of mature blood cell in the hematopoietic stem cell compartment in vivo compared with APAF1-, caspase-9 or caspase-3 [71]. These results imply that there are other, caspase-independent, ways for cells to die following MOMP — collectively referred to as caspase-independent cell death (CICD). In contrast with canonical apoptosis, lack of caspases was observed to trigger a type I interferon response prior to death [72–74]. Two independent studies investigated this phenomenon and found that MOMP-induced death in the absence of caspases cause the accumulation of mtDNA in the cytosol activating the cGAS-STING DNA sensing pathway [72,73]. Two separate studies reinforced the previous findings that CICD is pro-inflammatory by visualizing the gradual widening of BAX/BAK macropores allowing the extrusion of the mitochondrial inner membrane, its permeabilization and the release of mtDNA into the cytosol ultimately resulting in transcription of type I interferons (Figure 3) [75,76]. These studies substantiate that apoptosis, a silent type of cell death, can be pro-inflammatory under caspase-deficient conditions. Since the discovery of mitochondrial-driven CICD, additional studies have described a type I interferon response during apoptosis [77,78].

Overview of how mtDNA is released during cell death.

Figure 3.
Overview of how mtDNA is released during cell death.

Apoptosis: Extrinsic or intrinsic apoptosis signals result in BAX/BAK pore formation activation of the apoptotic pathway, extrusion of the inner mitochondrial membrane and release of mtDNA into the cytosol. DNA is recognized by cGAS and the cGAS-STING pathway is activated resulting in IRF3 activation and a type I IFN response. This gene expression is counteracted by caspase-3 cleaving and inactivating cGAS and IRF3. Pyroptosis: Activation of gasdermins (GSDMD, GSDME, Gasdermin A3) promotes pore formation in the mitochondria inducing mtDNA release and activation of the cGAS-STING signaling cascade resulting in a type I interferon response. However, the type I IFN reponse is prohibited by caspase-1 and -3 by inactivating cleavage of cGAS and IRF3 (Abbreviations: cGAS, cyclic GMP–AMP synthase; STING, stimulator of interferon genes protein; IRF3, inteferon regulatory factor 3; ISG, interferon stimulatory genes; GSDMD, gasdermin-D; GSDME, gasdermin-E; mtDNA, mitochondrial DNA).

Figure 3.
Overview of how mtDNA is released during cell death.

Apoptosis: Extrinsic or intrinsic apoptosis signals result in BAX/BAK pore formation activation of the apoptotic pathway, extrusion of the inner mitochondrial membrane and release of mtDNA into the cytosol. DNA is recognized by cGAS and the cGAS-STING pathway is activated resulting in IRF3 activation and a type I IFN response. This gene expression is counteracted by caspase-3 cleaving and inactivating cGAS and IRF3. Pyroptosis: Activation of gasdermins (GSDMD, GSDME, Gasdermin A3) promotes pore formation in the mitochondria inducing mtDNA release and activation of the cGAS-STING signaling cascade resulting in a type I interferon response. However, the type I IFN reponse is prohibited by caspase-1 and -3 by inactivating cleavage of cGAS and IRF3 (Abbreviations: cGAS, cyclic GMP–AMP synthase; STING, stimulator of interferon genes protein; IRF3, inteferon regulatory factor 3; ISG, interferon stimulatory genes; GSDMD, gasdermin-D; GSDME, gasdermin-E; mtDNA, mitochondrial DNA).

Close modal

Two consecutive studies have advanced the in vitro discovery of CICD by deliberately inducing inflammation using CICD in the tumor microenvironment, thus turning a ‘cold’ tumor ‘hot’ [74,79]. Specifically, activating CICD in a colorectal cancer mouse model prompted a strong anti-tumorigenic immune response driven by infiltration of macrophages and T cells ultimately resulting in regression of the tumor [74]. Subsequent to this, a combination of radiation and the pan-caspase-inhibitor emricasan as a cancer treatment in mice showed synergistic therapeutical effects in cancer regression caused by the inflammatory response of mtDNA released during CICD [79].

The finding that apoptosis can induce inflammation, raises the question of how and why an interferon response is lacking under caspase proficiency, especially since mtDNA is released into the cytosol independent of caspase activity [75] Indeed, caspase-3/7 were shown to cleave essential substrates of the cGAS-STING pathway namely cGAS and IRF3 inactivating these substrates [80]. Additionally, caspases have also been shown to prohibit de novo protein translation [81,82]. These findings suggest that caspases harbor a ‘non-cell death function’ important for the regulation of interferon induction by limiting signaling through cGAS-STING and IRF3, as well as protein translation, ultimately prohibiting type I interferon expression (Figure 3) [72,83].

mtDNA release during pyroptosis

In contrast with canonical apoptosis, pyroptotic cell death is a lytic, highly pro-inflammatory type of cell death. Pyroptosis entails activation of the canonical or non-canonical inflammasome signaling cascade; for canonical signaling this comprises the formation of a multiprotein complex, inflammasome complex, required to activate caspase-1 by autoproteolytic cleavage [84,85]. Activation of the caspase causes cleavage of the cell death substrate gasdermin-D into a N- and a C-terminal domain [86,87]. The N-terminal domain (GSDMDNT) translocates to the plasma membrane where it oligomerizes to form large permeability pores promoting cellular lysis [88–91].

Activation of pyroptosis affects mitochondria in different ways: reduction in mitochondrial membrane potential, dysregulating intracellular ion homeostasis, impediment of mitophagy and even permeabilization of mitochondrial membranes [92–95]. As discussed before, mtDNA can act upstream of NLRP3 and possibly AIM2 inflammasome receptor — as a ligand, but inflammasome activation can also cause mtDNA release. It has been shown that the GSDMDNT interacts with lipid species, including cardiolipin, only found in mitochondrial and bacterial membranes, and forms conduits in various lipidic liposomes [88–91]. Besides GSDMD, gasdermin A3 and gasdermin-E (GSDME) have been proposed to target mitochondrial membranes [96,97]. GSDME is cleaved and activated by the apoptotic caspase-3, and pore formation by GSDME promotes cyto c efflux [98]. In two recent reports, these findings were extended to show that gasdermin pores (upon activation of the NLRP3 and pyrin inflammasome) can induce mtDNA release directly and independent of cellular lysis (Figure 3) [99,100]. Interestingly, the osmoprotectant glycine was shown to inhibit the release of larger proteins (>25 kDa) as well as mtDNA from GSDM pores in the plasma membrane [101,102], but did not impact release of mtDNA into the cytosol [99].

Notably, similar to apoptotic caspases, the pro-inflammatory caspase-1 can cleave cGAS thereby inhibiting the cGAS-STING pathway (Figure 3) [103]. Moreover, potassium efflux via GSDMD pores was sufficient to impede cGAS-mediated type I interferon activation [95]. These studies suggest that although mtDNA can be released during inflammasome activation, caspase activity as well as changes in cytosolic ion concentrations during pyroptotic cell death may limit the expression of type I interferons.

Besides cell death-dependent release of mtDNA many other cell death-independent scenarios have been proposed to also result in mtDNA leakage and cGAS-STING pathway activation. These possibilities are summarized in Table 1 and Figure 4.

Overview of factors inducing mtDNA release.

Figure 4.
Overview of factors inducing mtDNA release.

mtDNA release has been reported in cell death during apoptosis and pyroptosis but also independent of cell death pathways. The non-cell death cues can be categorized in exterior signals, mitochondrial stress, mtDNA stress, inefficient mitochondrial clearance and pathogenic infections. Factors in gray induce a type I IFN response possibly mediating by mtDNA release.

Figure 4.
Overview of factors inducing mtDNA release.

mtDNA release has been reported in cell death during apoptosis and pyroptosis but also independent of cell death pathways. The non-cell death cues can be categorized in exterior signals, mitochondrial stress, mtDNA stress, inefficient mitochondrial clearance and pathogenic infections. Factors in gray induce a type I IFN response possibly mediating by mtDNA release.

Close modal
Table 1.
Cell death independent mtDNA release
Mitochondrial featuresCytosolic DNARef.
Mitochondrial stress and dysfunction 
Fatty acid binding protein (FABP5) inhibition in T cells Distorted cristae structure, altered lipid metabolism, lowered OXPHOS 
  • Cytosolic DNA

  • cGAS-STING activation

  • Type I IFN signature

 
[104
Endog−/− Accumulation of ROS in MEF cells 
  • Cytosolic DNA

  • cGAS-STING activation

  • Type I IFN signature

 
[105
Tfam+/− Alteration of mtDNA packaging — bigger nucleoids 
  • Cytosolic DNA

  • cGAS-STING activation

  • Type I IFN signature

 
[106
Ymel−/− Imbalance of mitochondrial pyrimidine pool 
  • Cytosolic DNA

  • cGAS-STING activation

  • Type I IFN signature

 
[107
ATAD3−/− Heterozygous mutations in human ATAD3, THP-1 ATAD3−/− 
  • Cytosolic DNA

  • cGAS-STING activation

  • Type I IFN signature

 
[108
CLPP−/−, Clpp−/− Mutation in the protease results in altered nucleoid size and induction of type I interferons 
  • Cytosolic DNA

  • cGAS-STING activation

  • Type I IFN signature

 
[109
Samm50−/− Inner membrane remodeling induced mtDNA release 
  • Cytosolic DNA

  • cGAS-STING activation

  • Type I IFN signature

 
[110
Mic19−/−
Mtx2−/−
Dnajc−/− 
Cristae disorganization induced mtDNA release 
  • Cytosolic DNA

  • cGAS-STING activation

  • Type I IFN signature

 
[111
Reduced and inefficient clearance of mitochondria 
Pink1−/−/Prkn−/− Deficiency for PINK or PRK in mice during exhaustive exercise 
  • cGAS-STING activation

  • Type I IFN signature

 
[112
TNF-α Down-regulation of PINK1 expression, decreased autophagy Cytosolic DNA [113,114
IRGM1−/− Accumulation of defective mitochondria — higher mtROS, lower respiratory capacity, more depolarized mitochondria Indirect effect [115
Infections 
HSV-I/II — herpes simplex I/II Degradation of TFAM and mtDNA by viral nucelase Type I IFN signature [106
SARS-COV2 — severe acute respiratory syndrome coronavirus 2 Virus induced mtDNA release in endothelial cells 
  • Cytosolic DNA

  • cGAS-STING activation

  • Type I IFN signature

 
[116
DENV — dengue virus NSB2 a DENV protease prohibits cGAS detection of mtDNA 
  • Cytosolic DNA

  • cGAS-STING activation

  • Type I IFN signature

 
[117,118
IAV — influenza A virus
ECMV — encephalomyocarditis virus 
Viroporins Cytosolic DNA [119
Extracellular cues inducing mtDNA release 
IL-1β In myeloid, fibroblast and epithelial cells 
  • Cytosolic DNA

  • cGAS-STING activation

  • Type I IFN signature

 
[120
Intracellular cues inducing mtDNA release 
TDP-43 Cytoplasmic accumulation of the nuclear DNA/RNA binding protein TDP-43 induces mtDNA release 
  • Cytosolic DNA

  • cGAS-STING activation

  • Type I IFN signature

 
[121
Mitochondrial featuresCytosolic DNARef.
Mitochondrial stress and dysfunction 
Fatty acid binding protein (FABP5) inhibition in T cells Distorted cristae structure, altered lipid metabolism, lowered OXPHOS 
  • Cytosolic DNA

  • cGAS-STING activation

  • Type I IFN signature

 
[104
Endog−/− Accumulation of ROS in MEF cells 
  • Cytosolic DNA

  • cGAS-STING activation

  • Type I IFN signature

 
[105
Tfam+/− Alteration of mtDNA packaging — bigger nucleoids 
  • Cytosolic DNA

  • cGAS-STING activation

  • Type I IFN signature

 
[106
Ymel−/− Imbalance of mitochondrial pyrimidine pool 
  • Cytosolic DNA

  • cGAS-STING activation

  • Type I IFN signature

 
[107
ATAD3−/− Heterozygous mutations in human ATAD3, THP-1 ATAD3−/− 
  • Cytosolic DNA

  • cGAS-STING activation

  • Type I IFN signature

 
[108
CLPP−/−, Clpp−/− Mutation in the protease results in altered nucleoid size and induction of type I interferons 
  • Cytosolic DNA

  • cGAS-STING activation

  • Type I IFN signature

 
[109
Samm50−/− Inner membrane remodeling induced mtDNA release 
  • Cytosolic DNA

  • cGAS-STING activation

  • Type I IFN signature

 
[110
Mic19−/−
Mtx2−/−
Dnajc−/− 
Cristae disorganization induced mtDNA release 
  • Cytosolic DNA

  • cGAS-STING activation

  • Type I IFN signature

 
[111
Reduced and inefficient clearance of mitochondria 
Pink1−/−/Prkn−/− Deficiency for PINK or PRK in mice during exhaustive exercise 
  • cGAS-STING activation

  • Type I IFN signature

 
[112
TNF-α Down-regulation of PINK1 expression, decreased autophagy Cytosolic DNA [113,114
IRGM1−/− Accumulation of defective mitochondria — higher mtROS, lower respiratory capacity, more depolarized mitochondria Indirect effect [115
Infections 
HSV-I/II — herpes simplex I/II Degradation of TFAM and mtDNA by viral nucelase Type I IFN signature [106
SARS-COV2 — severe acute respiratory syndrome coronavirus 2 Virus induced mtDNA release in endothelial cells 
  • Cytosolic DNA

  • cGAS-STING activation

  • Type I IFN signature

 
[116
DENV — dengue virus NSB2 a DENV protease prohibits cGAS detection of mtDNA 
  • Cytosolic DNA

  • cGAS-STING activation

  • Type I IFN signature

 
[117,118
IAV — influenza A virus
ECMV — encephalomyocarditis virus 
Viroporins Cytosolic DNA [119
Extracellular cues inducing mtDNA release 
IL-1β In myeloid, fibroblast and epithelial cells 
  • Cytosolic DNA

  • cGAS-STING activation

  • Type I IFN signature

 
[120
Intracellular cues inducing mtDNA release 
TDP-43 Cytoplasmic accumulation of the nuclear DNA/RNA binding protein TDP-43 induces mtDNA release 
  • Cytosolic DNA

  • cGAS-STING activation

  • Type I IFN signature

 
[121

Many studies have documented escape of mtDNA into the cytosol, but how is a large, negatively charged DNA nucleoid that resides in the mitochondrial matrix detected by a cytosolic DNA sensor cGAS? Does the DNA pass through two lipidic membranes into the cytosol? Are membranes permeabilized to facilitate its release?

BAX/BAK macropores

During apoptosis, the release of mtDNA into the cytosol is dependent on pro-apoptotic proteins BAX and BAK, thus suggesting the requirement of BAX/BAK pores to perforate the OMM. BAX/BAK form dynamic pores in the OMM upon oligomerization [122–126]. The inner membrane then protrudes through the BAX/BAK macropore and must thus be large enough to allow the passage of mtDNA [75,76]. Considering that cGAS could not be detected inside the protruding inner membrane [75,76]; breaching of the inner membrane likely facilitates mtDNA detection by cGAS. The exact mechanism of mitochondrial inner membrane permeabilization remains unclear. However, several possibilities are excluded; for example, mitochondrial dynamics (fusion and fission) does not impact mtDNA release [76]. In addition, rupture of the inner membrane due to the prolonged opening of the mPTP and ensuing IMM swelling could also be excluded on the basis that lack of mPTP regulator cyclophilin D did not reduce mtDNA release [76]. Nevertheless, the data does not exclude that permeabilization of the IMM is a passive event. New data however indicate shedding of large portions of the outer membrane prompted by Toxoplasma gondii infection was not sufficient to breach the IMM [127]. This may indicate that permeabilization of the IMM requires more than disruption of the OMM.

Gasdermin pores

Activation of gasdermin A3, D and E during pyroptosis have been suggested to trigger mtDNA release. Since the N-terminal domain of gasdermin forms large pores in membranes and has been shown to release soluble cytosolic proteins during pyroptosis [101,102,128], it was perhaps natural to assume that mtDNA can be released via gasdermin pores. The gasdermin pore however would have to perforate both mitochondrial membranes, IMM and OMM or allow extrusion of the inner membrane when the outer membrane is perforated. The outer diameter of the GSDM pore was measured to 28/40 nm (GSDMA3/GSDMD) and the inner diameter between 10 and 18 nm [88–91,129]. Although gasdermin pores clearly interact with cardiolipin and other lipids, the measured pore size is too small to transfer full-sized (80 × 100 nm) nucleoids. Thus, two possibilities for mtDNA release are probable, release as a result of collateral damage or a direct release of mtDNA fragments over the pore.

Mitochondrial permeability transition pores and VDAC oligomers

An early report proposed the release of mtDNA fragments via mPTP [130], which was described to have a minimal diameter of 2.8 nm [130–132]. A more recent report shows mtDNA release from Endog−/− cells being independent of apoptosis. Instead, Tfam+/− and ENDOG-deficient cells release mtDNA fragments into the IMS upon Ca2+ and ROS-induced mPTP opening [105]. In a second step the mtDNA fragments are released upon VDAC1 (possibly VDAC3) oligomerization — not part of the mPTP — from the outer mitochondrial membrane [105]. Since the release of mtDNA observed in cells deficient for YME1L or TDP-43 invasion into mitochondria, is independent of BAX/BAK pores but requires VDAC oligomerization it is likely also released via mPTP and VDAC oligomers [107,121]. Molecules smaller than 1.5 nm have been suggested to permeate the mPTP pore suggesting nucleoids are too big, but mtDNA fragments (DNA molecules in solution 0.33 nm [133]) can pass through the IMM into the IMS [134]. According to the NMR structure of human VDAC1, the pore has an approximate 3.4 nm diameter, and would thus be large enough to allow passage of mtDNA fragments (PDB 6TIQ) [135]. It was further suggested that mtDNA interacts with the VDAC1 to stabilize the pore; however, what signals promote dissociation to allow release of mtDNA through the pore remains ambiguous. Interestingly, double-stranded mtDNA (dsmtDNA) breaks are immediately cleared rather than repaired [136,137]. Thus, it is puzzling, that fragments of dsmtDNA are not degraded but instead released through mPTP and VDAC1 pores. Possibly this is suggestive of a protective mechanism to inhibit mtDNA degradation.

The detection and reaction of the cell to mtDNA in the cytosol is multifold. It is dependent on cell type and availability of receptor, but also on the way mtDNA is released (apoptotic conditions, etc.). This complicates the study of how mtDNA is released and activates cGAS-STING signaling. Many questions such as how and if mtDNA is fragmented before release, how mtDNA exits the IMM, how cGAS gets access to mtDNA and recognizes mtDNA, and whether the characteristics of mtDNA play a role in its recognition (circularity, CpG methylation, proteins and packaging of mtDNA) remain to be answered. It is also unclear how much mtDNA is required for the activation of cGAS, whether longer DNA stretches induce a more potent inflammatory response as suggested previously [138], and whether mtDNA leakage can occur in absence of mitochondrial rupture. In particular, can limited mitochondrial permeabilization — minority MOMP cause DNA damage and genome instability in absence of cell death [139], but can it also be a source of inflammation as suggested for a variety of pathogenic infections [140]? Furthermore, it is unclear whether mtDNA recognition by cGAS can be masked and potentially regulated, as reported for nuclear cGAS interacting with histones, thus limiting its activation by the nuclear genome [16,22–24]. Does mitochondrial TFAM adopt a similar role? Future research should address some of these questions, as elucidation and characterization of mtDNA is of importance to further our understanding in the role of mtDNA in health and disease as well as exploit the pathway pharmacologically.

Inflammation induced by the release of mtDNA is double-sided and can be beneficial as well as detrimental for the cell and organism. For instance, the release of mtDNA by BAX/BAK pores, in absence of caspases, induces pro-inflammatory cGAS-STING and NF-κB signaling which is important to repress tumorigenesis and induce tumor clearance. In addition, inhibition of the autophagic machinery fueling mtDNA release can potentially have synergistic therapeutical effects in combination with radiation in breast cancer cells [74,77]. Treatment of cancers using a combination therapy of apoptosis inducers and caspase inhibitors could thus improve therapeutic outcome. Activation of cGAS-STING pathway and expression of type I interferons has also shown to be beneficial in various virus infections to counteract viral proliferation and limiting viral spread [141–143].

Mitochondrial DNA release can also be unfavorable, for example promoting disease progression in systemic lupus erythematosus (SLE), amyotrophic lateral sclerosis (ALS), rheumatoid arthritis (RA) and others [105,114,121]. It is seen that inhibition of VDAC pores, thereby inhibiting mtDNA release, reduces disease severity in a mouse model of SLE [105]. Moreover, the hallmark protein TDP-43 in ALS was shown to induce mtDNA release via mPTP, linking mtDNA release to the ALS disease phenotype [121]. Blocking of mtDNA release in this setting would thus ameliorate the disease phenotype.

Understanding and elucidating the mechanism of mtDNA is of high importance pharmacologically. Hopefully, it will be employed to develop future treatment for cancer in respect to limiting cancer growth. But also used to develop agents hindering mtDNA release to limit progression in diseases phenotypes associated to mtDNA release.

  • Importance in the field. Escape of mtDNA into the cytosol is associated with the up-regulation of an inflammatory, type I interferon-driven response. Understanding the cues and factors inducing leakage of mtDNA into the cytosol in cell death and beyond is of importance to purposely promote (e.g. in cancer, infections) or hinder (mtDNA-associated disease phenotypes) mtDNA release pharmacologically.

  • Summary of the current thinking. mtDNA is released upon MOMP during apoptosis in caspase-deficient conditions. During apoptosis the cytosolic mtDNA activates the DNA sensor cGAS inducing a NFκB and type I interferon response, thus promoting inflammation. Besides apoptosis, cues such as mitochondrial, stress, mtDNA stress, pathogenic infections and others have been found to induce mtDNA release by distinct mechanisms causing inflammation via activation of the DNA sensors (cGAS) and receptors (NLRP3, AIM2 or TLR9).

  • A comment on future directions. To date, mtDNA release has been observed in a variety of systems, circumstances and reasons resulting in inflammatory gene expression. However, mechanistical insight into how mtDNA reaches the cytosol as well as how recognition is regulated is still ambiguous and will require further efforts. Furthermore, the impact of mtDNA-driven inflammation in health and disease is of high importance for pharmacological intervention and will require in-depth knowledge of the pathway.

The authors declare that there are no competing interests associated with the manuscript.

The laboratory is supported by Cancer Research UK Programme Foundation Award [A20145; S.W.G.T.] and R.H. is supported by a fellowship from the Swiss National Science Foundation.

Open access for this article was enabled by the participation of University of Glasgow in an all-inclusive Read & Publish agreement with Portland Press and the Biochemical Society under a transformative agreement with JISC.

R.H., J.L. and S.W.G.T. wrote the manuscript.

We apologized to colleagues we did not cited due to limited space.

AIM2

absent in melanoma 2

ALS

amyotrophic lateral sclerosis

cGAS

cyclic GMP–AMP synthase

CICD

caspase-independent cell death

CpG

cytidine-phosphate-guanosine

ER

endoplasmic reticulum

IMM

inner mitochondrial membrane

IMS

intermembrane space

IRF3

interferon regulatory factor 3

IRF7

interferon regulatory factor 7

LRR

leucine-rich repeat

MAPK

mitogen-activated protein kinases

mPTP

mitochondrial permeability transition pore

mtDNA

mitochondrial DNA

OMM

outer mitochondrial membrane

SLE

systemic lupus erythematosus

1
Pfanner
,
N.
,
Warscheid
,
B.
and
Wiedemann
,
N.
(
2018
)
Mitochondrial proteins: from biogenesis
to functional networks
.
Nat. Rev. Mol. Cell Biol.
20
,
267
284
2
Kummer
,
E.
and
Ban
,
N.
(
2021
)
Mechanisms and regulation of protein synthesis in mitochondria
.
Nat. Rev. Mol. Cell Biol.
22
,
307
325
3
Bonekamp
,
N.A.
and
Larsson
,
N.G.
(
2018
)
Snapshot: mitochondrial nucleoid
.
Cell
172
,
388
388.e1
4
Micheau
,
O.
and
Tschopp
,
J.
(
2003
)
Induction of TNF receptor I-mediated apoptosis via two sequential signaling complexes
.
Cell
114
,
181
190
5
Hsu
,
H.
,
Shu
,
H.B.
,
Pan
,
M.G.
and
Goeddel D
,
V.
(
1996
)
TRADD-TRAF2 and TRADD-FADD interactions define two distinct TNF receptor 1 signal transduction pathways
.
Cell
84
,
299
308
6
Haas
,
T.L.
,
Emmerich
,
C.H.
,
Gerlach
,
B.
,
Schmukle
,
A.C.
,
Cordier
,
S.M.
,
Rieser
,
E.
et al (
2009
)
Recruitment of the linear ubiquitin chain assembly complex stabilizes the TNF-R1 signaling complex and is required for TNF-mediated gene induction
.
Mol. Cell
36
,
831
844
7
Vince
,
J.E.
,
Pantaki
,
D.
,
Feltham
,
R.
,
Mace
,
P.D.
,
Cordier
,
S.M.
,
Schmukle
,
A.C.
et al (
2009
)
TRAF2 must bind to cellular inhibitors of apoptosis for tumor necrosis factor (TNF) to efficiently activate NF-κB and to prevent TNF-induced apoptosis
.
J. Biol. Chem.
284
,
35906
35915
8
Jost
,
P.J.
,
Grabow
,
S.
,
Gray
,
D.
,
McKenzie
,
M.D.
,
Nachbur
,
U.
,
Huang
,
D.C.S.
et al (
2009
)
XIAP discriminates between type I and type II FAS-induced apoptosis
.
Nature
460
,
1035
1039
9
Wang
,
L.
,
Du
,
F.
and
Wang
,
X.
(
2008
)
TNF-α Induces two distinct caspase-8 activation pathways
.
Cell
133
,
693
703
10
Petersen
,
S.L.
,
Wang
,
L.
,
Yalcin-Chin
,
A.
,
Li
,
L.
,
Peyton
,
M.
,
Minna
,
J.
et al (
2007
)
Autocrine TNFα signaling renders human cancer cells susceptible to Smac-mimetic-induced apoptosis
.
Cancer Cell
12
,
445
456
11
Tenev
,
T.
,
Bianchi
,
K.
,
Darding
,
M.
,
Broemer
,
M.
,
Langlais
,
C.
,
Wallberg
,
F.
et al (
2011
)
The ripoptosome, a signaling platform that assembles in response to genotoxic stress and loss of IAPs
.
Mol. Cell
43
,
432
448
12
Feoktistova
,
M.
,
Geserick
,
P.
,
Kellert
,
B.
,
Dimitrova
,
D.P.
,
Langlais
,
C.
,
Hupe
,
M.
et al (
2011
)
CIAPs block ripoptosome formation, a RIP1/Caspase-8 containing intracellular cell death complex differentially regulated by cFLIP isoforms
.
Mol. Cell
43
,
449
463
13
Bock
,
F.J.
and
Tait
,
S.W.G.
(
2019
)
Mitochondria as multifaceted regulators of cell death
.
Nat. Rev. Mol. Cell Biol.
21
,
85
100
14
Sun
,
L.
,
Wu
,
J.
,
Du
,
F.
,
Chen
,
X.
and
Chen
,
Z.J.
(
2013
)
Cyclic GMP-AMP synthase is a cytosolic DNA sensor that activates the type I interferon pathway
.
Science
339
,
786
791
15
Orzalli
,
M.H.
,
Broekema
,
N.M.
,
Diner
,
B.A.
,
Hancks
,
D.C.
,
Elde
,
N.C.
,
Cristea
,
I.M.
et al (
2015
)
CGAS-mediated stabilization of IFI16 promotes innate signaling during herpes simplex virus infection
.
Proc. Natl Acad. Sci. U.S.A.
112
,
E1773
E1781
16
Volkman
,
H.E.
,
Cambier
,
S.
,
Gray
,
E.E.
and
Stetson
,
D.B.
(
2019
)
Tight nuclear tethering of cGAS is essential for preventing autoreactivity
.
eLife
8
,
e47491
17
Zierhut
,
C.
,
Yamaguchi
,
N.
,
Paredes
,
M.
,
Luo
,
J.D.
,
Carroll
,
T.
and
Funabiki
,
H.
(
2019
)
The cytoplasmic DNA sensor cGAS promotes mitotic cell death
.
Cell
178
,
302
315.e23
18
Lahaye
,
X.
,
Gentili
,
M.
,
Silvin
,
A.
,
Conrad
,
C.
,
Picard
,
L.
,
Jouve
,
M.
et al (
2018
)
NONO detects the nuclear HIV capsid to promote cGAS-mediated innate immune activation
.
Cell
175
,
488
501.e22
19
Boyer
,
J.A.
,
Spangler
,
C.J.
,
Strauss
,
J.D.
,
Cesmat
,
A.P.
,
Liu
,
P.
,
McGinty
,
R.K.
et al (
2020
)
Structural basis of nucleosome-dependent cGAS inhibition
.
Science
370
,
450
454
20
Cao
,
D.
,
Han
,
X.
,
Fan
,
X.
,
Xu
,
R.M.
and
Zhang
,
X.
(
2020
)
Structural basis for nucleosome-mediated inhibition of cGAS activity
.
Cell Res.
30
,
1088
1097
21
Kujirai
,
T.
,
Zierhut
,
C.
,
Takizawa
,
Y.
,
Kim
,
R.
,
Negishi
,
L.
,
Uruma
,
N.
et al (
2020
)
Structural basis for the inhibition of cGAS by nucleosomes
.
Science
370
.
455
458
22
Michalski
,
S.
,
de Oliveira Mann
,
C.C.
,
Stafford
,
C.A.
,
Witte
,
G.
,
Bartho
,
J.
,
Lammens
,
K.
et al (
2020
)
Structural basis for sequestration and autoinhibition of cGAS by chromatin
.
Nature
587
,
678
682
23
Pathare
,
G.R.
,
Decout
,
A.
,
Glück
,
S.
,
Cavadini
,
S.
,
Makasheva
,
K.
,
Hovius
,
R.
et al (
2020
)
Structural mechanism of cGAS inhibition by the nucleosome
.
Nature
587
,
668
672
24
Zhao
,
B.
,
Xu
,
P.
,
Rowlett
,
C.M.
,
Jing
,
T.
,
Shinde
,
O.
,
Lei
,
Y.
et al (
2020
)
The molecular basis of tight nuclear tethering and inactivation of cGAS
.
Nature
587
,
673
677
25
Wu
,
J.
,
Sun
,
L.
,
Chen
,
X.
,
Du
,
F.
,
Shi
,
H.
,
Chen
,
C.
et al (
2013
)
Cyclic GMP-AMP is an endogenous second messenger in innate immune signaling by cytosolic DNA
.
Science
339
,
826
830
26
Ablasser
,
A.
,
Goldeck
,
M.
,
Cavlar
,
T.
,
Deimling
,
T.
,
Witte
,
G.
,
Röhl
,
I.
et al (
2013
)
cGAS produces a 2′-5′-linked cyclic dinucleotide second messenger that activates STING
.
Nature
498
,
380
384
27
Burdette
,
D.L.
,
Monroe
,
K.M.
,
Sotelo-Troha
,
K.
,
Iwig
,
J.S.
,
Eckert
,
B.
,
Hyodo
,
M.
et al (
2011
)
STING is a direct innate immune sensor of cyclic di-GMP
.
Nature
478
,
515
518
28
Ishikawa
,
H.
and
Barber
,
G.N.
(
2008
)
STING is an endoplasmic reticulum adaptor that facilitates innate immune signalling
.
Nature
455
,
674
678
29
Liu
,
S.
,
Cai
,
X.
,
Wu
,
J.
,
Cong
,
Q.
,
Chen
,
X.
,
Li
,
T.
et al (
2015
)
Phosphorylation of innate immune adaptor proteins MAVS, STING, and TRIF induces IRF3 activation
.
Science
347
,
aaa2630
30
McWhirter
,
S.M.
,
Barbalat
,
R.
,
Monroe
,
K.M.
,
Fontana
,
M.F.
,
Hyodo
,
M.
,
Joncker
,
N.T.
et al (
2009
)
A host type I interferon response is induced by cytosolic sensing of the bacterial second messenger cyclic-di-GMP
.
J. Exp. Med.
206
,
1899
1911
31
Ablasser
,
A.
and
Chen
,
Z.J.
(
2019
)
CGAS in action: expanding roles in immunity and inflammation
.
Science
363
,
eaat8657
32
Nakahira
,
K.
,
Haspel
,
J.A.
,
Rathinam
,
V.A.K.
,
Lee
,
S.J.
,
Dolinay
,
T.
,
Lam
,
H.C.
et al (
2010
)
Autophagy proteins regulate innate immune responses by inhibiting the release of mitochondrial DNA mediated by the NALP3 inflammasome
.
Nat. Immunol.
12
,
222
230
33
Shimada
,
K.
,
Crother
,
T.R.
,
Karlin
,
J.
,
Dagvadorj
,
J.
,
Chiba
,
N.
,
Chen
,
S.
et al (
2012
)
Oxidized mitochondrial DNA activates the NLRP3 inflammasome during apoptosis
.
Immunity
36
,
401
414
34
Zhong
,
Z.
,
Liang
,
S.
,
Sanchez-Lopez
,
E.
,
He
,
F.
,
Shalapour
,
S.
,
Lin
,
X.J.
et al (
2018
)
New mitochondrial DNA synthesis enables NLRP3 inflammasome activation
.
Nature
560
,
198
203
35
Mangan
,
M.S.J.J.
,
Olhava
,
E.J.
,
Roush
,
W.R.
,
Seidel
,
H.M.
,
Glick
,
G.D.
and
Latz
,
E.
(
2018
)
Targeting the NLRP3 inflammasome in inflammatory diseases
.
Nat. Rev. Drug Discov.
17
,
588
606
36
Zhou
,
R.
,
Yazdi
,
A.S.
,
Menu
,
P.
and
Tschopp
,
J.
(
2010
)
A role for mitochondria in NLRP3 inflammasome activation
.
Nature
469
,
221
225
37
Heid
,
M.E.
,
Keyel
,
P.A.
,
Kamga
,
C.
,
Shiva
,
S.
,
Watkins
,
S.C.
and
Salter
,
R.D.
(
2013
)
Mitochondrial reactive oxygen species induces NLRP3-dependent lysosomal damage and inflammasome activation
.
J. Immunol.
191
,
5230
5238
38
Xian
,
H.
,
Watari
,
K.
,
Sanchez-Lopez
,
E.
,
Offenberger
,
J.
,
Onyuru
,
J.
,
Sampath
,
H.
et al (
2022
)
Oxidized DNA fragments exit mitochondria via mPTP- and VDAC-dependent channels to activate NLRP3 inflammasome and interferon signaling
.
Immunity
55
,
1370
1385.e8
39
Nguyen
,
T.T.
,
Stevens M
,
V.
,
Kohr
,
M.
,
Steenbergen
,
C.
,
Sack
,
M.N.
and
Murphy
,
E.
(
2011
)
Cysteine 203 of cyclophilin D is critical for cyclophilin D activation of the mitochondrial permeability transition pore
.
J. Biol. Chem.
286
,
40184
40192
40
Du
,
H.
,
Guo
,
L.
,
Fang
,
F.
,
Chen
,
D.
,
Sosunov A
,
A.
,
McKhann G
,
M.
et al (
2008
)
Cyclophilin D deficiency attenuates mitochondrial and neuronal perturbation and ameliorates learning and memory in Alzheimer's disease
.
Nat. Med.
14
,
1097
1105
41
Vanaja S
,
K.
,
Rathinam
,
V.A.K.
,
Atianand
,
M.K.
,
Kalantari
,
P.
,
Skehan
,
B.
,
Fitzgerald
,
K.A.
et al)
Bacterial RNA:DNA hybrids are activators of the NLRP3 inflammasome
.
Proc. Natl Acad. Sci. U.S.A.
111
,
7765
7770
42
Dang
,
E.V.
,
McDonald
,
J.G.
,
Russell
,
D.W.
,
Cyster
,
J.G.
(
2017
)
Oxysterol restraint of cholesterol synthesis prevents AIM2 inflammasome activation
.
Cell
171
,
1057
1071.e11
43
Hemmi
,
H.
,
Takeuchi
,
O.
,
Kawai
,
T.
,
Kaisho
,
T.
,
Sato
,
S.
,
Sanjo
,
H.
et al (
2000
)
A Toll-like receptor recognizes bacterial DNA
.
Nature
408
,
740
745
44
Paludan
,
S.R.
,
Reinert
,
L.S.
and
Hornung
,
V.
(
2019
)
DNA-stimulated cell death: implications for host defence, inflammatory diseases and cancer
.
Nat. Rev. Immunol.
19
,
141
153
45
Broz
,
P.
and
Monack
,
D.M.
(
2013
)
Newly described pattern recognition receptors team up against intracellular pathogens
.
Nat. Rev. Immunol.
13
,
551
565
46
Bauer
,
S.
,
Kirschning
,
C.J.
,
Häcker
,
H.
,
Redecke
,
V.
,
Hausmann
,
S.
,
Akira
,
S.
et al (
2001
)
Human TLR9 confers responsiveness to bacterial DNA via species-specific CpG motif recognition
.
Proc Natl Acad. Sci. U.S.A.
98
,
9237
9242
47
Takeshita
,
F.
,
Leifer
,
C.A.
,
Gursel
,
I.
,
Ishii
,
K.J.
,
Takeshita
,
S.
,
Gursel
,
M.
et al (
2001
)
Cutting edge: role of toll-like receptor 9 in CpG DNA-induced activation of human cells
.
J. Immunol.
167
,
3555
3558
48
Yousefi
,
S.
,
Mihalache
,
C.
,
Kozlowski
,
E.
,
Schmid
,
I.
and
Simon
,
H.U.
(
2009
)
Viable neutrophils release mitochondrial DNA to form neutrophil extracellular traps
.
Cell Death Differ.
16
,
1438
1444
49
Lood
,
C.
,
Blanco
,
L.P.
,
Purmalek
,
M.M.
,
Carmona-Rivera
,
C.
,
de Ravin
,
S.S.
,
Smith
,
C.K.
et al (
2015
)
Neutrophil extracellular traps enriched in oxidized mitochondrial DNA are interferogenic and contribute to lupus-like disease
.
Nat. Med.
22
,
146
153
50
Caielli
,
S.
,
Athale
,
S.
,
Domic
,
B.
,
Murat
,
E.
,
Chandra
,
M.
,
Banchereau
,
R.
et al (
2016
)
Oxidized mitochondrial nucleoids released by neutrophils drive type I interferon production in human lupus
.
J. Exp. Med.
213
,
697
713
51
Pazmandi
,
K.
,
Agod
,
Z.
,
Kumar B
,
V.
,
Szabo
,
A.
,
Fekete
,
T.
,
Sogor
,
V.
et al (
2014
)
Oxidative modification enhances the immunostimulatory effects of extracellular mitochondrial DNA on plasmacytoid dendritic cells
.
Free Radic. Biol. Med.
77
,
281
290
52
McIlroy
,
D.J.
,
Jarnicki
,
A.G.
,
Au
,
G.G.
,
Lott
,
N.
,
Smith
,
D.W.
,
Hansbro
,
P.M.
et al (
2014
)
Mitochondrial DNA neutrophil extracellular traps are formed after trauma and subsequent surgery
.
J. Crit. Care
29
,
1133.e1
1133.e5
53
Garcia-Martinez
,
I.
,
Santoro
,
N.
,
Chen
,
Y.
,
Hoque
,
R.
,
Ouyang
,
X.
,
Caprio
,
S.
et al (
2016
)
Hepatocyte mitochondrial DNA drives nonalcoholic steatohepatitis by activation of TLR9
.
J. Clin. Investig.
126
,
859
864
54
Kerr
,
J.F.R.
,
Wyllie
,
A.H.
and
Curriet
,
A.R.
(
1972
)
APOPTOSIS: a basic biological phenomenon with wide-ranging implications in tissue kinetics
.
Br. J. Cancer
26
,
239
257
55
Campbell
,
K.J.
and
Tait
,
S.W.G.
(
2018
)
Targeting BCL-2 regulated apoptosis in cancer
.
Open Biol.
8
,
180002
56
Wei
,
M.C.
,
Zong
,
W.X.
,
Cheng
,
E.H.Y.
,
Lindsten
,
T.
,
Panoutsakopoulou
,
V.
,
Ross
,
A.J.
et al (
2001
)
Proapoptotic BAX and BAK: a requisite gateway to mitochondrial dysfunction and death
.
Science
292
,
727
730
57
Letai
,
A.
,
Bassik
,
M.C.
,
Walensky
,
L.D.
,
Sorcinelli
,
M.D.
,
Weiler
,
S.
and
Korsmeyer
,
S.J.
(
2002
)
Distinct BH3 domains either sensitize or activate mitochondrial apoptosis, serving as prototype cancer therapeutics
.
Cancer Cell
2
,
183
192
58
Czabotar
,
P.E.
,
Westphal
,
D.
,
Dewson
,
G.
,
Ma
,
S.
,
Hockings
,
C.
,
Fairlie
,
W.D.
et al (
2013
)
Bax crystal structures reveal how BH3 domains activate Bax and nucleate its oligomerization to induce apoptosis
.
Cell
152
,
519
531
59
Moldoveanu
,
T.
,
Grace
,
C.R.
,
Llambi
,
F.
,
Nourse
,
A.
,
Fitzgerald
,
P.
,
Gehring
,
K.
et al (
2013
)
BID-induced structural changes in BAK promote apoptosis
.
Nat. Struct. Mol. Biol.
20
,
589
60
Leshchiner
,
E.S.
,
Braun
,
C.R.
,
Bird
,
G.H.
and
Walensky
,
L.D.
(
2013
)
Direct activation of full-length proapoptotic BAK
.
Proc. Natl Acad. Sci. U.S.A.
110
,
E986
E995
61
Dewson
,
G.
,
Kratina
,
T.
,
Czabotar
,
P.
,
Day
,
C.L.
,
Adams
,
J.M.
and
Kluck
,
R.M.
(
2009
)
Bak activation for apoptosis involves oligomerization of dimers via their α6 helices
.
Mol. Cell
36
,
696
703
62
Dewson
,
G.
,
Ma
,
S.
,
Frederick
,
P.
,
Hockings
,
C.
,
Tan
,
I.
,
Kratina
,
T.
et al (
2012
)
Bax dimerizes via a symmetric BH3:groove interface during apoptosis
.
Cell Death Differ.
19
,
661
670
63
Bleicken
,
S.
,
Classen
,
M.
,
Padmavathi
,
P.V.L.
,
Ishikawa
,
T.
,
Zeth
,
K.
,
Steinhoff
,
H.J.
et al (
2010
)
Molecular details of Bax activation, oligomerization, and membrane insertion
.
J. Biol. Chem.
285
,
6636
6647
64
Subburaj
,
Y.
,
Cosentino
,
K.
,
Axmann
,
M.
,
Pedrueza-Villalmanzo
,
E.
,
Hermann
,
E.
,
Bleicken
,
S.
et al (
2015
)
Bax monomers form dimer units in the membrane that further self-assemble into multiple oligomeric species
.
Nat. Commun.
6
,
1
11
65
Kulda
,
K.
,
Zheng
,
T.S.
,
Na
,
S.
,
Kuan
,
C.Y.
,
Yang
,
D.
,
Karasuyama
,
H.
et al (
1996
)
Decreased apoptosis in the brain and premature lethality in CPP32-deficient mice
.
Nature
384
,
368
372
66
Yoshida
,
H.
,
Kong
,
Y.Y.
,
Yoshida
,
R.
,
Elia
,
A.J.
,
Hakem
,
A.
,
Hakem
,
R.
et al (
1998
)
Apaf1 is required for mitochondrial pathways of apoptosis and brain development
.
Cell
94
,
739
750
67
Lindsten
,
T.
,
Ross
,
A.J.
,
King
,
A.
,
Zong
,
W.X.
,
Rathmell
,
J.C.
,
Shiels
,
H.A.
et al (
2000
)
The combined functions of proapoptotic Bcl-2 family members Bak and Bax are essential for normal development of multiple tissues
.
Mol. Cell
6
,
1389
1399
68
Ekert
,
P.G.
,
Read
,
S.H.
,
Silke
,
J.
,
Marsden
,
V.S.
,
Kaufmann
,
H.
,
Hawkins
,
C.J.
et al (
2004
)
Apaf-1 and caspase-9 accelerate apoptosis, but do not determine whether factor-deprived or drug-treated cells die
.
J. Cell Biol.
165
,
835
842
69
Marsden
,
V.S.
,
O'Connor
,
L.
,
O'Reilly
,
L.A.
,
Silke
,
J.
,
Metcalf
,
D.
,
Ekert
,
P.G.
et al (
2002
)
Apoptosis initiated by Bcl-2-regulated caspase activation independently of the cytochrome c/Apaf-1/caspase-9 apoptosome
.
Nature
419
,
634
637
70
Van Delft
,
M.F.
,
Smith
,
D.P.
,
Lahoud
,
M.H.
,
Huang
,
D.C.S.
and
Adams
,
J.M.
(
2010
)
Apoptosis and non-inflammatory phagocytosis can be induced by mitochondrial damage without caspases
.
Cell Death Differ.
17
,
821
832
71
Janzen
,
V.
,
Fleming
,
H.E.
,
Riedt
,
T.
,
Karlsson
,
G.
,
Riese
,
M.J.
,
lo Celso
,
C.
et al (
2008
)
Hematopoietic stem cell responsiveness to exogenous signals is limited by caspase-3
.
Cell Stem Cell
2
,
584
594
72
Rongvaux
,
A.
,
Jackson
,
R.
,
Harman
,
C.C.D.
,
Li
,
T.
,
West
,
A.P.
,
De Zoete
,
M.R.
et al (
2014
)
Apoptotic caspases prevent the induction of type I interferons by mitochondrial DNA
.
Cell
159
,
1563
1577
73
White
,
M.J.
,
McArthur
,
K.
,
Metcalf
,
D.
,
Lane
,
R.M.
,
Cambier
,
J.C.
,
Herold
,
M.J.
et al (
2014
)
Apoptotic caspases suppress mtDNA-induced STING-mediated type I IFN production
.
Cell
159
,
1549
1562
74
Giampazolias
,
E.
,
Zunino
,
B.
,
Dhayade
,
S.
,
Bock
,
F.
,
Cloix
,
C.
,
Cao
,
K.
et al (
2017
)
Mitochondrial permeabilization engages NF-κB-dependent anti-tumour activity under caspase deficiency
.
Nat. Cell Biol.
19
,
1116
1129
75
McArthur
,
K.
,
Whitehead
,
L.W.
,
Heddleston
,
J.M.
,
Li
,
L.
,
Padman
,
B.S.
,
Oorschot
,
V.
et al (
2018
)
BAK/BAX macropores facilitate mitochondrial herniation and mtDNA efflux during apoptosis
.
Science
359
,
eaao6047
76
Riley
,
J.S.
,
Quarato
,
G.
,
Cloix
,
C.
,
Lopez
,
J.
,
O'Prey
,
J.
,
Pearson
,
M.
et al (
2018
)
Mitochondrial inner membrane permeabilisation enables mtDNA release during apoptosis
.
EMBO J.
37
,
e99238
77
Yamazaki
,
T.
,
Kirchmair
,
A.
,
Sato
,
A.
,
Buqué
,
A.
,
Rybstein
,
M.
,
Petroni
,
G.
et al (
2020
)
Mitochondrial DNA drives abscopal responses to radiation that are inhibited by autophagy
.
Nat. Immunol.
21
,
1160
1171
78
Maekawa
,
H.
,
Inoue
,
T.
,
Hirokawa
,
N.
,
Nangaku
,
M.
and
Inagi
,
R.
(
2019
)
Mitochondrial damage causes inflammation via cGAS-STING signaling in acute kidney injury
.
Cell Rep.
29
,
1261
1273.e6
79
Han
,
C.
,
Liu
,
Z.
,
Zhang
,
Y.
,
Shen
,
A.
,
Dong
,
C.
,
Zhang
,
A.
et al (
2020
)
Tumor cells suppress radiation-induced immunity by hijacking caspase 9 signaling
.
Nat. Immunol.
21
,
546
554
80
Ning
,
X.
,
Wang
,
Y.
,
Jing
,
M.
,
Sha
,
M.
,
Lv
,
M.
,
Gao
,
P.
et al (
2019
)
Apoptotic caspases suppress type I interferon production via the cleavage of cGAS, MAVS, and IRF3
.
Mol. Cell
74
,
19
31
81
Marissen
,
W.E.
and
Lloyd
,
R.E.
(
1998
)
Eukaryotic translation initiation factor 4G is targeted for proteolytic cleavage by caspase 3 during inhibition of translation in apoptotic cells
.
Mol. Cell. Biol.
18
,
7565
7574
82
Saelens
,
X.
,
Kalai
,
M.
and
Vandenabeele
,
P.
(
2001
)
Translation inhibition in apoptosis: caspase-dependent PKR activation and eIF2-α phosphorylation
.
J. Biol. Chem.
276
,
41620
41628
83
White
,
M.J.
,
Mcarthur
,
K.
,
Metcalf
,
D.
,
Lane
,
R.M.
,
Cambier
,
J.C.
,
Herold
,
M.J.
et al (
2014
)
Article apoptotic caspases suppress mtDNA-induced STING-mediated type I IFN production
.
Cell
159
,
1549
1562
84
Broz
,
P.
and
Dixit
,
V.M.
(
2016
)
Inflammasomes: mechanism of assembly, regulation and signalling
.
Nat. Rev. Immunol.
16
,
407
420
85
Broz
,
P.
,
Pelegrín
,
P.
and
Shao
,
F.
(
2019
)
The gasdermins, a protein family executing cell death and inflammation
.
Nat. Rev. Immunol.
20
,
143
157
86
Kayagaki
,
N.
,
Stowe
,
I.B.
,
Lee
,
B.L.
,
O'Rourke
,
K.
,
Anderson
,
K.
,
Warming
,
S.
et al (
2015
)
Caspase-11 cleaves gasdermin D for non-canonical inflammasome signalling
.
Nature
526
,
666
671
87
Shi
,
J.
,
Zhao
,
Y.
,
Wang
,
K.
,
Shi
,
X.
,
Wang
,
Y.
,
Huang
,
H.
et al (
2015
)
Cleavage of GSDMD by inflammatory caspases determines pyroptotic cell death
.
Nature
526
,
660
665
88
Ding
,
J.
,
Wang
,
K.
,
Liu
,
W.
,
She
,
Y.
,
Sun
,
Q.
,
Shi
,
J.
et al (
2016
)
Pore-forming activity and structural autoinhibition of the gasdermin family
.
Nature
535
,
111
116
89
Aglietti
,
R.A.
,
Estevez
,
A.
,
Gupta
,
A.
,
Ramirez
,
M.G.
,
Liu
,
P.S.
,
Kayagaki
,
N.
et al (
2016
)
Gsdmd p30 elicited by caspase-11 during pyroptosis forms pores in membranes
.
Proc. Natl Acad. Sci. U.S.A.
113
,
7858
7863
90
Sborgi
,
L.
,
Rühl
,
S.
,
Mulvihill
,
E.
,
Pipercevic
,
J.
,
Heilig
,
R.
,
Stahlberg
,
H.
et al (
2016
)
GSDMD membrane pore formation constitutes the mechanism of pyroptotic cell death
.
EMBO J.
35
,
e201694696-13
91
Liu
,
X.
,
Zhang
,
Z.
,
Ruan
,
J.
,
Pan
,
Y.
,
Magupalli
,
V.G.
,
Wu
,
H.
et al (
2016
)
Inflammasome-activated gasdermin D causes pyroptosis by forming membrane pores
.
Nature
535
,
153
158
92
de Vasconcelos
,
N.M.
,
Van Opdenbosch
,
N.
,
Van Gorp
,
H.
,
Parthoens
,
E.
and
Lamkanfi
,
M.
(
2018
)
Single-cell analysis of pyroptosis dynamics reveals conserved GSDMD-mediated subcellular events that precede plasma membrane rupture
.
Cell Death Differ.
26
,
146
161
93
Cryns
,
V.L.
,
Byun
,
Y.
,
Rana
,
A.
,
Mellor
,
H.
,
Lustig
,
K.D.
,
Ghanem
,
L.
et al (
1997
)
Specific proteolysis of the kinase protein kinase C-related kinase 2 by caspase-3 during apoptosis. Identification by a novel, small pool expression cloning strategy
.
J. Biol. Chem.
272
,
29449
29453
94
Kahns
,
S.
,
Kalai
,
M.
,
Diness Jakobsen
,
L.
,
Clark
,
B.F.C.
,
Vandenabeele
,
P.
and
Jensen
,
P.H.
(
2003
)
Caspase-1 and caspase-8 cleave and inactivate cellular parkin
.
J. Biol. Chem.
278
,
23376
23380
95
Banerjee
,
I.
,
Behl
,
B.
,
Mendonca
,
M.
,
Shrivastava
,
G.
,
Russo
,
A.J.
,
Menoret
,
A.
et al (
2018
)
Gasdermin D restrains type I interferon response to cytosolic DNA by disrupting ionic homeostasis
.
Immunity
49
,
413
426.e5
96
Lin
,
P.H.
,
Lin
,
H.Y.
,
Kuo
,
C.C.
and
Yang
,
L.T.
(
2015
)
N-terminal functional domain of gasdermin A3 regulates mitochondrial homeostasis via mitochondrial targeting
.
J. Biomed. Sci.
22
,
1
18
97
Rogers
,
C.
,
Erkes
,
D.A.
,
Nardone
,
A.
,
Aplin
,
A.E.
,
Fernandes-Alnemri
,
T.
and
Alnemri
,
E.S.
(
2019
)
Gasdermin pores permeabilize mitochondria to augment caspase-3 activation during apoptosis and inflammasome activation
.
Nat. Commun.
10
,
1689
98
Rogers
,
C.
,
Fernandes-Alnemri
,
T.
,
Mayes
,
L.
,
Alnemri
,
D.
,
Cingolani
,
G.
and
Alnemri
,
E.S.
(
2017
)
Cleavage of DFNA5 by caspase-3 during apoptosis mediates progression to secondary necrotic/pyroptotic cell death
.
Nat. Commun.
8
,
14128
99
de Torre-Minguela
,
C.
,
Gómez
,
A.I.
,
Couillin
,
I.
and
Pelegrín
,
P.
(
2021
)
Gasdermins mediate cellular release of mitochondrial DNA during pyroptosis and apoptosis
.
FASEB J.
35
,
e21757
100
Huang
,
L.S.
,
Hong
,
Z.
,
Wu
,
W.
,
Xiong
,
S.
,
Zhong
,
M.
,
Gao
,
X.
et al (
2020
)
mtDNA activates cGAS signaling and suppresses the YAP-mediated endothelial cell proliferation program to promote inflammatory injury
.
Immunity
52
,
475
486.e5
101
Evavold
,
C.L.
,
Ruan
,
J.
,
Tan
,
Y.
,
Xia
,
S.
,
Wu
,
H.
and
Kagan
,
J.C.
(
2018
)
The pore-forming protein gasdermin D regulates interleukin-1 secretion from living macrophages
.
Immunity
48
,
35
44.e6
102
Heilig
,
R.
,
Dick
,
M.S.
,
Sborgi
,
L.
,
Meunier
,
E.
,
Hiller
,
S.
and
Broz
,
P.
(
2018
)
The gasdermin-D pore acts as a conduit for IL-1β secretion in mice
.
Eur. J. Immunol.
48
,
584
592
103
Wang
,
Y.
,
Ning
,
X.
,
Gao
,
P.
,
Wu
,
S.
,
Sha
,
M.
,
Lv
,
M.
et al (
2017
)
Inflammasome activation triggers caspase-1-mediated cleavage of cGAS to regulate responses to DNA virus infection
.
Immunity
46
,
393
404
104
Field
,
C.S.
,
Baixauli
,
F.
,
Kyle
,
R.L.
,
Puleston
,
D.J.
,
Cameron
,
A.M.
,
Sanin
,
D.E.
et al (
2020
)
Mitochondrial integrity regulated by lipid metabolism is a cell-intrinsic checkpoint for treg suppressive function
.
Cell Metab.
31
,
422
437.e5
105
Kim
,
J.
,
Gupta
,
R.
,
Blanco
,
L.P.
,
Yang
,
S.
,
Shteinfer-Kuzmine
,
A.
,
Wang
,
K.
et al (
2019
)
VDAC oligomers form mitochondrial pores to release mtDNA fragments and promote lupus-like disease
.
Science
366
,
1531
1536
106
West
,
A.P.
,
Khoury-Hanold
,
W.
,
Staron
,
M.
,
Tal
,
M.C.
,
Pineda
,
C.M.
,
Lang
,
S.M.
et al (
2015
)
Mitochondrial DNA stress primes the antiviral innate immune response
.
Nature
520
,
553
557
107
Sprenger
,
H.G.
,
MacVicar
,
T.
,
Bahat
,
A.
,
Fiedler
,
K.U.
,
Hermans
,
S.
,
Ehrentraut
,
D.
et al (
2021
)
Cellular pyrimidine imbalance triggers mitochondrial DNA–dependent innate immunity
.
Nat. Metabol.
3
,
636
650
108
Lepelley
,
A.
,
Della Mina
,
E.
,
van Nieuwenhove
,
E.
,
Waumans
,
L.
,
Fraitag
,
S.
,
Rice
,
G.I.
et al (
2021
)
Enhanced cGAS-STING–dependent interferon signaling associated with mutations in ATAD3A
.
J. Exp. Med.
218
,
e20201560
109
Torres-Odio
,
S.
,
Lei
,
Y.
,
Gispert
,
S.
,
Maletzko
,
A.
,
Key
,
J.
,
Menissy
,
S.S.
et al (
2021
)
Loss of mitochondrial protease CLPP activates type I IFN responses through the mitochondrial DNA–cGAS–STING signaling axis
.
J. Immunol.
206
,
1890
1900
110
Chen
,
L.
,
Dong
,
J.
,
Liao
,
S.
,
Wang
,
S.
,
Wu
,
Z.
,
Zuo
,
M.
et al (
2022
)
Loss of Sam50 in hepatocytes induces cardiolipin-dependent mitochondrial membrane remodeling to trigger mtDNA release and liver injury
.
Hepatology
76
,
1389
1408
111
He
,
B.
,
Yu
,
H.
,
Liu
,
S.
,
Wan
,
H.
,
Fu
,
S.
,
Liu
,
S.
et al (
2022
)
Mitochondrial cristae architecture protects against mtDNA release and inflammation
.
Cell Rep.
41
,
111774
112
Sliter
,
D.A.
,
Martinez
,
J.
,
Hao
,
L.
,
Chen
,
X.
,
Sun
,
N.
,
Fischer
,
T.D.
et al (
2018
)
Parkin and PINK1 mitigate STING-induced inflammation
.
Nature
561
,
258
262
113
Bueno
,
M.
,
Zank
,
D.
,
Buendia-Roldán
,
I.
,
Fiedler
,
K.
,
Mays
,
B.G.
,
Alvarez
,
D.
et al (
2019
)
PINK1 attenuates mtDNA release in alveolar epithelial cells and TLR9 mediated profibrotic responses
.
PLoS ONE
14
,
1
20
114
Willemsen
,
J.
,
Neuhoff
,
M.T.
,
Hoyler
,
T.
,
Noir
,
E.
,
Tessier
,
C.
,
Sarret
,
S.
et al (
2021
)
TNF leads to mtDNA release and cGAS/STING-dependent interferon responses that support inflammatory arthritis
.
Cell Rep.
37
,
109977
115
Rai
,
P.
,
Janardhan
,
K.S.
,
Meacham
,
J.
,
Madenspacher
,
J.H.
,
Lin
,
W.C.
,
Karmaus
,
P.W.F.
et al (
2021
)
IRGM1 links mitochondrial quality control to autoimmunity
.
Nat. Immunol.
22
,
312
321
116
Domizio
,
J.D.
,
Gulen
,
M.F.
,
Saidoune
,
F.
,
Thacker
,
V.V.
,
Yatim
,
A.
,
Sharma
,
K.
et al)
The cGAS-STING pathway drives type I IFN immunopathology in COVID-19
.
Nature
603
,
145
151
117
Sun
,
B.
,
Sundström
,
K.B.
,
Chew
,
J.J.
,
Bist
,
P.
,
Gan
,
E.S.
,
Tan
,
H.C.
et al (
2017
)
Dengue virus activates cGAS through the release of mitochondrial DNA
.
Sci. Rep.
7
,
3594
118
Aguirre
,
S.
,
Luthra
,
P.
,
Sanchez-Aparicio
,
M.T.
,
Maestre
,
A.M.
,
Patel
,
J.
,
Lamothe
,
F.
et al (
2017
)
Dengue virus NS2B protein targets cGAS for degradation and prevents mitochondrial DNA sensing during infection
.
Nat. Microbiol.
2
,
17037
119
Moriyama
,
M.
,
Koshiba
,
T.
and
Ichinohe
,
T.
(
2019
)
Influenza A virus M2 protein triggers mitochondrial DNA-mediated antiviral immune responses
.
Nat. Commun.
10
,
1
14
120
Aarreberg
,
L.D.
,
Esser-Nobis
,
K.
,
Driscoll
,
C.
,
Shuvarikov
,
A.
,
Roby
,
J.A.
and
Gale
,
M.
(
2019
)
Interleukin-1β induces mtDNA release to activate innate immune signaling via cGAS-STING
.
Mol. Cell
74
,
801
815.e6
121
Yu
,
C.H.
,
Davidson
,
S.
,
Harapas
,
C.R.
,
Hilton
,
J.B.
,
Mlodzianoski
,
M.J.
,
Laohamonthonkul
,
P.
et al (
2020
)
TDP-43 Triggers mitochondrial DNA release via mPTP to activate cGAS/STING in ALS
.
Cell
183
,
636
649.e18
122
Uren
,
R.T.
,
Iyer
,
S.
and
Kluck
,
R.M.
(
2017
)
Pore formation by dimeric Bak and Bax: an unusual pore?
Phil. Trans. R. Soc. B
372
,
20160218
123
Bleicken
,
S.
,
Landeta
,
O.
,
Landajuela
,
A.
,
Basañez
,
G.
and
García-Sáez
,
A.J.
(
2013
)
Proapoptotic bax and bak proteins form stable protein-permeable pores of tunable size
.
J. Biol. Chem.
288
,
33241
33252
124
Salvador-Gallego
,
R.
,
Mund
,
M.
,
Cosentino
,
K.
,
Schneider
,
J.
,
Unsay
,
J.
,
Schraermeyer
,
U.
et al (
2016
)
Bax assembly into rings and arcs in apoptotic mitochondria is linked to membrane pores
.
EMBO J.
35
,
389
401
125
Große
,
L.
,
Wurm
,
C.A.
,
Brüser
,
C.
,
Neumann
,
D.
,
Jans
,
D.C.
and
Jakobs
,
S.
(
2016
)
Bax assembles into large ring-like structures remodeling the mitochondrial outer membrane in apoptosis
.
EMBO J.
35
,
402
413
126
Cosentino
,
K.
,
Hertlein
,
V.
,
Jenner
,
A.
,
Dellmann
,
T.
,
Gojkovic
,
M.
,
Peña-Blanco
,
A.
et al (
2022
)
The interplay between BAX and BAK tunes apoptotic pore growth to control mitochondrial-DNA-mediated inflammation
.
Mol. Cell
82
,
933
949.e9
127
Li
,
X.
,
Straub
,
J.
,
Medeiros
,
T.C.
,
Mehra
,
C.
,
den Brave
,
F.
,
Peker
,
E.
et al (
2022
)
Mitochondria shed their outer membrane in response to infection-induced stress
.
Science
375
,
eabi4343
128
Monteleone
,
M.
,
Stanley
,
A.C.
,
Chen
,
K.W.
,
Sweet
,
M.J.
,
Stow
,
J.L.
,
Correspondence
,
K.S.
et al (
2018
)
Interleukin-1β maturation triggers its relocation to the plasma membrane for gasdermin-D-dependent and -independent secretion
.
Cell Rep.
24
,
1425
1433
129
Ruan
,
J.
,
Xia
,
S.
,
Liu
,
X.
,
Lieberman
,
J.
and
Wu
,
H.
(
2018
)
Cryo-EM structure of the gasdermin A3 membrane pore
.
Nature
557
,
62
67
130
Patrushev
,
M.
,
Kasymov
,
V.
,
Patrusheva
,
V.
,
Ushakova
,
T.
,
Gogvadze
,
V.
and
Gaziev
,
A.
(
2004
)
Mitochondrial permeability transition triggers the release of mtDNA fragments
.
Cell. Mol. Life Sci.
61
,
3100
3103
131
Massari
,
S.
and
Azzone
,
G.F.
(
1972
)
The equivalent pore radius of intact and damaged mitochondria and the mechanism of active shrinkage
.
Biochim. Biophys. Acta
283
,
23
29
132
Bernardis
,
P.
,
Vassanelli
,
S.
,
Veronese
,
P.
,
Colonna
,
R.
,
Szabo
,
I.
and
Zoratti
,
M.
(
1992
)
Modulation of the mitochondrial permeability transition pore. Effect of protons and divalent cations
.
J. Biol. Chem.
267
,
2934
2939
133
Mandelkern
,
M.
,
Elias
,
J.G.
,
Eden
,
D.
and
Crothers
,
D.M.
(
1981
)
The dimensions of DNA in solution
.
J. Mol. Biol.
152
,
153
161
134
Karch
,
J.
and
Molkentin
,
J.D.
(
2014
)
Identifying the components of the elusive mitochondrial permeability transition pore
.
Proc. Natl Acad. Sci. U.S.A.
111
,
10396
10397
135
,
R.
,
Amodeo
,
G.F.
,
Murlidaran
,
S.
,
Winterhalter
,
M.
,
Brannigan
,
G.
and
Correspondence
,
S.H.
(
2020
)
The structural basis for low conductance in the membrane protein VDAC upon β-NADH binding and voltage gating
.
Struct. Fold. Des.
28
,
206
214.e4
136
Srivastava
,
S.
and
Moraes
,
C.T.
(
2001
)
Manipulating mitochondrial DNA heteroplasmy by a mitochondrially targeted restriction endonuclease
.
Hum. Mol. Genet.
10
,
3093
3099
137
Tanaka
,
M.
,
Borgeld
,
H.J.
,
Zhang
,
J.
,
Muramatsu
,
S.I.
,
Gong
,
J.S.
,
Yoneda
,
M.
et al (
2002
)
Gene therapy for mitochondrial disease by delivering restriction endonuclease SmaI into mitochondria
.
J. Biomed. Sci.
9
,
534
541
138
Andreeva
,
L.
,
Hiller
,
B.
,
Kostrewa
,
D.
,
Lässig
,
C.
,
De Oliveira Mann
,
C.C.
,
Jan Drexler
,
D.
et al (
2017
)
cGAS senses long and HMGB/TFAM-bound U-turn DNA by forming protein–DNA ladders
.
Nature
549
,
394
398
139
Ichim
,
G.
,
Lopez
,
J.
,
Ahmed
,
S.U.
,
Muthalagu
,
N.
,
Giampazolias
,
E.
,
Delgado
,
M.E.
et al (
2015
)
Limited mitochondrial permeabilization causes DNA damage and genomic instability in the absence of cell death
.
Mol. Cell
57
,
860
872
140
Brokatzky
,
D.
,
Kretz
,
O.
and
Häcker
,
G.
(
2020
)
Apoptosis functions in defense against infection of mammalian cells with environmental chlamydiae
.
Infect. Immun.
88
,
e00851-19
141
Miura
,
T.
,
Goto
,
M.
,
Hosoya
,
N.
,
Odawara
,
T.
,
Kitamura
,
Y.
,
Nakamura
,
T.
et al (
2003
)
Depletion of mitochondrial DNA in HIV-1-infected patients and its amelioration by antiretroviral therapy
.
J. Med. Virol.
70
,
497
505
142
McNab
,
F.
,
Mayer-Barber
,
K.
,
Sher
,
A.
,
Wack
,
A.
and
O'Garra
,
A.
(
2015
)
Type I interferons in infectious disease
.
Nat. Rev. Immunol.
15
,
87
103
143
Schoggins
,
J.W.
(
2019
)
Interferon-stimulated genes: what do they all do?
Annu. Rev. Virol.
14
,
567
584
This is an open access article published by Portland Press Limited on behalf of the Biochemical Society and distributed under the Creative Commons Attribution License 4.0 (CC BY).