ALL Metrics
-
Views
-
Downloads
Get PDF
Get XML
Cite
Export
Track
Review

Advances in the understanding of mitochondrial DNA as a pathogenic factor in inflammatory diseases

[version 1; peer review: 3 approved]
PUBLISHED 20 Feb 2017
Author details Author details
OPEN PEER REVIEW
REVIEWER STATUS

Abstract

Mitochondrial DNA (mtDNA) has many similarities with bacterial DNA because of their shared common ancestry. Increasing evidence demonstrates mtDNA to be a potent danger signal that is recognised by the innate immune system and can directly modulate the inflammatory response. In humans, elevated circulating mtDNA is found in conditions with significant tissue injury such as trauma and sepsis and increasingly in chronic organ-specific and systemic illnesses such as steatohepatitis and systemic lupus erythematosus. In this review, we examine our current understanding of mtDNA-mediated inflammation and how the mechanisms regulating mitochondrial homeostasis and mtDNA release represent exciting and previously under-recognised important factors in many human inflammatory diseases, offering many new translational opportunities.

Keywords

mitochondrial DNA, mtDNA, mtDNA-mediated inflammation, inflammatory diseases

Introduction

Mitochondria are intracellular double-membrane-bound organelles (“cellular powerhouses”) with many essential physiological roles in energy production, programmed cell death, calcium homeostasis, and the synthesis of lipids, amino acids, and haem. In addition, they are involved in antibacterial, antiviral, and stress responses to hypoxia and tissue injury1,2. Mitochondria are evolutionarily derived from energy-producing alpha-bacteria, engulfed by archezoan cells approximately 2 billion years ago leading to a symbiotic relationship that forms the basis of the eukaryotic cells3. The mitochondria share several features with bacteria, including the double-membrane structure, a circular genome that replicates independently of nuclear DNA, and the synthesis of N-formylated proteins4. As the innate immune system recognises conserved bacterial molecules, mitochondrial constituents are similarly immunogenic, acting as damage-associated molecular patterns (DAMPs) when released into the cytosol and extracellular environment, triggering innate immune responses, and promoting inflammation5. In this review, we focus particularly on the role of mitochondrial DNA (mtDNA) as a specific inflammatory factor, the mechanisms behind its abnormal release, and its effects on downstream inflammatory pathways in human inflammatory diseases.

Elevated circulating mtDNA in human diseases

Freely circulating mtDNA can be detected, and over 60 studies have quantified mtDNA by quantitative polymerase chain reaction (PCR) in plasma and serum in human diseases (Table 1). In general, they are increased in conditions with acute tissue injury such as trauma, acute myocardial infarction, and sepsis, implicating major cellular stress and uncontrolled cell death as key factors in the release of mtDNA (Figure 1). In cancer, where its role as “liquid biopsies” is a topic of considerable interest, the pattern is less clear, and relatively lower circulating levels are found in some cancers6.

Table 1. Circulating mitochondrial DNA in human disease.

Disease
category
DiseaseBlood
fraction
FindingReference(s)
TraumaTraumaPlasmaHigh mtDNA levels in trauma compared with HCs and correlated
with injury severity
7
TraumaPlasmaHigh mtDNA levels in trauma8,9
Trauma with MODSPlasmaHigher levels of mtDNA had higher relative risk for mortality
Higher levels of mtDNA in those with SIRS/MODS compared with
those without
10
Trauma and severe sepsisPlasmamtDNA higher in patients with trauma compared with HCs on day
1
mtDNA correlates with injury severity scores in trauma patients
mtDNA higher on day 1 in non-survivors compared with survivors
11
Post-traumatic SIRSPlasmamtDNA is an independent predictor for post-traumatic SIRS12
TraumaPlasmamtDNA higher in trauma patients with correlation with injury
severity
7
Trauma (femur
fracture)
PlasmamtDNA higher in trauma patients than HCs 13
TraumaPlasmamtDNA higher in trauma patients compared with HCs at two time
points (pre-hospital and day 1)
14
TraumaPlasmamtDNA higher in trauma patients than HCs
mtDNA higher in non-survivors compared with survivors
15
SepsisSevere sepsisPlasmamtDNA higher in patients with severe sepsis compared with HCs
No significant difference in mtDNA between non-survivors and
survivors in severe sepsis
11
Severe sepsis in the EDPlasmamtDNA higher on admission in severe septic patients than in HCs
mtDNA is higher in non-survivors than in survivors, increases
initially and gradually decreases after antimicrobial therapy, and is
an independent predictor of fatality
16
SepsisPlasmamtDNA higher in septic patients compared with HCs 17
Septic shockPlasmamtDNA higher in patients with septic shock 18
Adult community-
acquired bacterial
meningitis
PlasmamtDNA levels were higher in patients with aseptic or bacterial
meningitis compared with HCs
mtDNA levels fall during course of admission
High mtDNA levels associated with poorer outcome in adult
community-acquired bacterial meningitis
19
Infectious SIRSPlasmamtDNA higher in septic patients compared with HCs 20
Paediatric sepsisPlasmamtDNA higher in septic patients compared with critically ill non-
septic and HC patients
21
Severe sepsis in
the ED
PlasmaNo significant difference in mtDNA between sepsis and HC
cohorts
22
Critically ill
patients
ICU patientsPlasmaIncreased mtDNA levels associated with medical ICU mortality 23
Critically ill patients
(in the ICU)
PlasmaPatients with highest quartile of mtDNA in plasma had higher risk
of dying When stratified by TLR9 expression, only patients with high
expression of TLR9 had an association with mortality and mtDNA
level
24
Out-of-hospital
cardiac arrest
PlasmaSignificantly higher levels in non-survivors than in survivors 56a
Liver failureAcetaminophen-
induced acute liver
failure
SerummtDNA higher in acetaminophen-induced acute liver failure
patients compared with HCs
mtDNA higher in non-survivors compared with survivors
25
Acetaminophen-
induced acute liver
injury
PlasmamtDNA higher in patients with acetaminophen overdose with
abnormal liver function tests compared with HCs and those with
acetaminophen overdose but normal liver function tests
27
Fulminant liver
failure
SerumHigher during acute liver injury 26
Heart diseaseAMIPlasmaSignificantly higher mtDNA in ST elevation myocardial infarction
patients than in stable angina pectoris patients (reducing rapidly
to similar levels 3 days after PCI)
28
AMIPlasmaSignificantly higher levels in AMI patients compared with HCs
Levels dropped to normal immediately after PCI
29
AMIPlasmaSignificantly higher levels in acute AMI patients compared with
HCs on admission
30
T2DM with CADPlasmaSignificantly elevated levels in T2DM compared with HCs
Higher levels in those with diabetes mellitus and CAD compared
with those without CAD
mtDNA levels correlated with C-reactive protein in patients with
CAD
31
T2DM with CADPlasmaSignificantly higher levels in CAD patients with T2DM 32
Heart failurePlasmaHigher levels of mtDNA in heart failure patients compared with
age- and sex-matched HCs; no association with disease severity
110
StrokeAcute ischaemic
stroke
PlasmamtDNA levels higher in acute cerebral infarction than in HCs
No significant difference in mtDNA between good versus poor
outcome cohorts
33
Subarachnoid
haemorrhage
PlasmaNo significant difference in mtDNA between subarachnoid
haemorrahge and HC groups
Overall plasma mtDNA not a good marker of prognosis
34
Intracerebral
haemorrhage
PlasmaNo significant difference in mtDNA between intracerebral
haemorrhage and HC groups
No correlation between mtDNA and disease severity
35
MalignancyBreast cancerPlasmaReduced levels of mtDNA in benign or malignant breast cancer
compared with HCs
111
Ovarian cancerPlasma and
serum
Plasma: significantly higher levels of mtDNA in ovarian cancer
group compared with HCs and ovarian benign tumour group
Serum: no significant difference between groups above
112
Testicular germ cell
cancer
SerummtDNA levels were significantly higher in patients with testicular
cancer than in HCs, although it did not correlate with any
clinicopathological variable of disease status
113
Urological
malignancies
SerummtDNA were significantly higher in “urological malignancies”
(bladder cell, renal cell, and prostate cancer)
114
Prostate cancerSerummtDNA could not distinguish between benign prostatic
hypertrophy and prostate cancer
Patients with early biochemical recurrence after radical
prostatectomy have higher mtDNA levels
115
Ewing’s sarcomaSerummtDNA significantly lower in patients with Ewing’s sarcoma
compared with HCs
116
Lung cancerSerummtDNA significantly higher in lung cancer patients compared
with those with benign lung diseases and healthy individuals and
closely associated with tumour, lymph node, metastasis (TNM)
stage
117
Advanced prostate
cancer
PlasmamtDNA levels are elevated in advanced prostate cancer patients
and are associated with decreased survival
118
Adenocarcinoma of
the lung in patients
receiving erlotinib
PlasmaRise in mtDNA levels in patients with partial response; drop in
mtDNA levels in those with progressive disease or no response
No correlation with progression-free survival
119
Exposure to
carcinogenic
halo-alkane-based
pesticides
SerumExposure to these carcinogens was significantly associated with
elevated serum levels of circulating mtDNA (case control study)
120
Renal cell
carcinoma
PlasmaHigher levels in metastatic compared with non-metastatic patients
and controls
121
HIVHIVPlasmaHigher levels in acute HIV infection, late presenters compared with
long-term non-progressors and HCs
Also correlated with viral load
122
Lipodystrophy
in HIV patients
treated with highly
active anti-retroviral therapy
PlasmaSignificantly higher levels in HIV-infected versus non-infected
individuals
Significantly higher levels in those with lipodystrophy compared
with those without lipodystrophy at month 24
123
HIVPlasmaNo significant association between HIV disease status and mtDNA 124
Inflammatory
autoimmune
conditions
Rheumatoid arthritisPlasmaHigher percentage of detectable levels in rheumatoid arthritis
patients compared with controls
37b
Granulomatosis with
polyangiitis
SerumSignificantly higher levels in granulomatosis with polyangiitis
patients compared with controls
38
Age and
exercise
AgePlasmamtDNA levels increased gradually after the fifth decade of life 125
AgePlasmaNo association with age but mtDNA associated with HLA-DR 126
Aging and “frailty”PlasmaAging: no difference in mtDNA between younger and older
subjects
Frailty: mtDNA copy number directly correlated with frailty score
127
ExercisePlasmaReduced mtDNA in response to exercise 128
Male volleyball
players
PlasmaLower levels in professional volleyball players compared with
healthy non-athlete controls
129
MiscellaneousCorrosive injury
(gastrointestinal
ingestion)
PlasmaSignificantly higher mtDNA in mortality group versus survival
group at presentation and after
12 hours
130
Pulmonary
embolism
PlasmaPredictor of 15-day mortality 131c
AutismSerumSignificantly higher mtDNA in young autistic children compared
with HCs
132
HaemodialysisPlasmaSignificantly higher levels in maintenance haemodialysis patients
compared with HCs
133
End-stage renal
failure in Han
population
PlasmaEnd-stage renal failure patients had higher mtDNA copy number 134
Bipolar disorderSerumNo difference between bipolar disorder and HC groups
Higher levels in bipolar disorder group compared with sepsis
135
Low levels of
ionising radiation
SerumHigher levels in interventional cardiologists exposed to low levels
of ionising radiation compared with controls
136
Friedreich’s ataxiaPlasmaSignificantly reduced mtDNA in Friedreich’s ataxia patients
compared with HCs
137
Non-haemolytic
transfusion reaction
Platelet
concentrates
Higher mtDNA copy number in non-haemolytic transfusion
reaction platelet concentrate versus normal platelet concentrate
138

This table lists studies reporting mitochondrial DNA (mtDNA) analysed by polymerase chain reaction (PCR) on serum or plasma—that is, circulating as a damage-associated molecular pattern (DAMP)—in human diseases. aLetter. bPCR rather than quantitative PCR used. cEarlier study in 2010 not included. AMI, acute myocardial infarction; CAD, coronary artery disease; ED, emergency department; HC, healthy control; HIV, human immunodeficiency virus; HLA-DR, human leukocyte antigen–antigen D related; ICU, intensive care unit; MODS, multiple organ dysfunction syndrome; PCI, percutaneous coronary intervention; SIRS, systemic inflammatory response syndrome; T2DM, type 2 diabetes mellitus; TLR9, Toll-like receptor 9.

403a6cba-11b0-4a07-ab52-b7d877634176_figure1.gif

Figure 1. The contribution of mitochondrial DNA to disease pathogenesis.

Medical conditions are in italics. Where and how mitochondria are released are indicated in red. Box in dotted line frames mitochondrial DNA (mtDNA) sensor target. cGAS, cyclic GMP-AMP synthetase; IFN, interferon; IL, interleukin; MAPK, mitogen-activated protein kinase; MMP, matrix metalloproteinase; mtROS, mitochondria-derived reactive oxygen species; NET, neutrophil extracellular trap; NFκB, nuclear factor kappa B; pDC, plasmacytoid dendritic cell; SIRS, systemic inflammatory response syndrome; SLE, systemic lupus erythematosus; STING, stimulator of interferon genes; TLR9, Toll-like receptor 9.

Systemic inflammatory response syndrome

Systemic inflammatory response syndrome (SIRS) is a serious condition associated with high mortality, and affected individuals display progressive signs or symptoms of systemic upset reflecting widespread inflammation, often involving multiple organ dysfunction and failure (for example, lungs, kidneys, and brain). SIRS is often a result of major sepsis but also commonly occurs in the context of injury such as trauma. An early study by Lam et al. found that individuals admitted for blunt traumatic injury had increased plasma nuclear DNA and mtDNA levels7. Subsequently, Hauser et al. made the seminal observation that it is the freely circulating mtDNA following traumatic injury which possesses the distinct ability to trigger and drive the clinical manifestation of SIRS8. Several studies have confirmed the observation of elevated plasma mtDNA in trauma and SIRS915. A number of studies have found correlations with injury severity in trauma7,11 and higher mtDNA in non-survivors compared with survivors11,15. Furthermore, Gu et al. found that elevated plasma mtDNA was an independent predictor of SIRS in trauma patients12. In sepsis, elevated levels of circulating mtDNA have also been found in multiple studies11,1621. De Caro et al. found higher mtDNA in the plasma of critically ill paediatric patients who were septic compared with similarly unwell but non-septic patients21. The one negative study in sepsis may be explained by numerous factors, including a relatively well patient cohort, only one “spot” measurement being taken at presentation, and the potentially confounding factor of cellular content/debris22. Studies of patients in the intensive care setting have found that higher mtDNA levels are associated with poorer outcomes23,24.

Acute single-organ injury: liver, heart, and brain

High levels of mtDNA are present in the serum and plasma of patients with acute injury to a variety of single organs. Acetaminophen overdose induces massive hepatocyte necrosis and in severe cases can lead to multi-organ failure and remains one of the commonest indications for liver transplantation. In fulminant liver failure secondary to acetaminophen overdose, mtDNA in the serum was found to be 30 to 40 times higher than normal, and non-survivors had higher levels than survivors25; a separate study of drug-induced acute liver failure found serum mtDNA levels to be 10,000-fold higher26. Serum mtDNA of acetaminophen overdose patients with derangement in the liver enzyme alanine aminotransferase (a marker of hepatocyte damage) is significantly higher than that of overdose patients who had normal liver enzymes27, suggesting that the extent of mtDNA release into the circulation depends on the extent of hepatocyte necrosis. Similarly, extensive cardiomyocyte necrosis is found in acute myocardial infarction, which is also associated with elevated mtDNA in multiple studies2830 and falls after angioplasty or coronary stent insertion to restore blood flow to the damaged myocardium28,29. Patients with diabetes mellitus and coronary artery disease have higher mtDNA levels than those with diabetes but without coronary artery disease31,32. mtDNA is also higher in acute cerebral ischaemia, caused by a reduction in cerebral blood flow by embolus or local thrombosis, and plasma levels gradually drop over time after the initial tissue injury33. Interestingly, studies by the same group relating to plasma mtDNA in subarachnoid haemorrhage and spontaneous intracerebral haemorrhage found no significant difference compared with healthy controls, although both were small studies34,35. Higher mtDNA is found in the cerebrospinal fluid of patients with subarachnoid haemorrhage34 and traumatic brain injury36 and is associated with worse clinical outcomes. Overall, in these conditions, significant mtDNA release following massive tissue or cellular injury is evident and likely contributes to the uncontrolled inflammatory response25.

Chronic inflammatory and immune-mediated diseases

The role for mtDNA in immune-mediated inflammatory diseases, unlike conditions relating to injury, is now also emerging. In rheumatoid arthritis, a chronic relapsing autoimmune condition affecting the joints, mtDNA was present in the plasma and synovial fluid of most patients but undetectable in healthy controls37. Similarly, higher plasma mtDNA is found in granulomatosis with polyangiitis, an autoimmune disease whose features include necrotising granulomatous inflammation and vasculitis38. Systemic lupus erythematosus (SLE) is a multi-organ autoimmune disease with hallmarks including excessive type I interferon (IFN) and antibodies against nucleic acids. Caielli et al. explored the potential pathogenic importance of oxidised mtDNA in SLE39. They showed that there is a defect in mitochondrial clearance that leads to abnormal extrusion of oxidised mtDNA, which triggers a subsequent interferogenic response. Elevated anti-mtDNA antibodies were found in a separate study of SLE, particularly in lupus nephritis, where levels correlated with the lupus nephritis activity index better than anti-double-stranded DNA (anti-dsDNA) antibody levels did40. In a further study of SLE, neutrophil extracellular traps (NETs) released from the inflammatory subset of low-density granulocyte were highly enriched in mtDNA compared with NETs from healthy control neutrophils41. NETs are networks of extracellular fibres that are primarily composed of DNA and that are strikingly expelled following a form of neutrophil cell death (NETosis) with an aim to control pathogens; however, this study demonstrates that mtDNA-enriched NETs are pro-inflammatory in nature. Similar findings are reported in chronic granulomatous disease in this study. Higher levels of mtDNA have been found in the chronic inflammatory states of HIV (although not in all studies), end-stage renal failure, and diabetes mellitus (Table 1). In obese individuals with steatohepatitis, mitochondria enclosed in microparticles can also be detected in plasma42. These findings suggest that mtDNA, otherwise a “self-signal”, may be an active component in the aberrant immune or inflammatory response in chronic diseases and in autoimmunity.

mtDNA contributes to inflammatory response

mtDNA was first directly implicated as a key factor in the development of inflammatory pathology over a decade ago when intra-articular injection of oxidised mtDNA, but not nuclear DNA, triggered inflammatory arthritis in mice43. There are now numerous studies using in vivo injection of mtDNA to provoke local or systemic inflammation or both9,4446. Moreover, there are now several in vivo studies to show that genetic deletion or pharmacologic interference of these pathways reduces the inflammatory effect of mtDNA (as will be discussed in the next section). Hence, it is clear that mtDNA release is not an epiphenomenon but directly contributes to the genesis of inflammation (Figure 1). Current evidence shows that mtDNA-mediated inflammation is predominantly driven by the Toll-like receptor 9 (TLR9), inflammasome, and, more recently, stimulator of interferon genes (STING) pathways.

Toll-like receptor 9

TLR9 is located in the endoplasmic reticulum (ER) of various immune cells and translocates to the endosome upon sensing of hypomethylated DNA with CpG motifs, such as bacterial DNA47,48. Given its high frequency of unmethylated CpG dinucleotide repeats, it is postulated that mtDNA mediates inflammation dependent on the TLR9 pathway and potentially exerts a similar effect as on bacterial CpG. TLR9 recognises a variety of types of oligodeoxynucleotides (ODNs); for example, class A ODNs preferentially activate plasmacytoid dendritic cells whilst class B CpG ODNs activate B cells49. Some of our understanding of how mtDNA may interact with TLR9 is extrapolated from work with class A ODNs, although they do not necessarily have the same effect. After activation of TLR9 by CpG DNA, inflammatory cytokine induction and Th1 immune responses occur50 and TLR9 is necessary in CpG DNA-driven responses51. TLR9 ligands can preferentially activate downstream pathways, including pro-inflammatory nuclear factor kappa B (NFκB), nucleotide-bindingdomain and leucine-rich repeat (NLR) pyrin domain containing 3 (NLRP3) inflammasomes, and interferon regulatory factor (IRF)-dependent type 1 IFN, which can upregulate IL-1 receptor antagonist52,53.

Most tissue injury models show better outcomes when the tlr9 gene is deleted. Wei et al. recently observed that tlr9−/− mice have improved survival outcome in a necrotic lung model of cationic nanocarrier-induced necrosis and mtDNA release in vivo54. Furthermore, the pulmonary inflammation seen after injection of mtDNA was significantly reduced in tlr9−/− and MyD88−/− mice, underlining the importance of the TLR9–MyD88 pathway54. Intravenous injection of mitochondrial debris with substantial amounts of mtDNA into mice induced a systemic inflammatory response in wild-type mice that was significantly attenuated in tlr9−/− mice45. Tlr9−/− mice also have better survival compared with wild-type counterparts in severe renal ischaemia reperfusion injury with associated decreased circulating mtDNA55. A similar protective effect is seen in tlr9−/− mice with acute acetaminophen overdose with observed lower serum mtDNA and an absence of lung inflammation in contrast to the findings of wild-type mice26. Nevertheless, the reduction in mtDNA in tlr9−/− mice is intriguing and could be explained by the reduced inflammation with lower resultant cellular necrosis. Alternatively, it is possible that TLR9 is somehow involved in mtDNA release into the extracellular circulation. In a recent study using a murine model of non-alcoholic steatohepatitis (NASH), mtDNA from NASH hepatocytes resulted in greater activation of TLR9 than did mtDNA from control livers42. This suggests that mtDNA that is selectively modified during pathologic disease processes can augment the ensuing inflammatory response. Similarly, the level of TLR9 expression (due to various factors) appears to be important. In those with high mtDNA levels, higher TLR9 expression is associated with increased mortality in the intensive care unit (ICU), as discussed earlier56.

Neutrophils have received the most attention in studies on mtDNA–TLR9 signalling in several different inflammatory settings. Zhang et al. found that mtDNA activates neutrophil p38 mitogen-activated protein kinase (MAPK) through TLR9 with release of matrix metalloproteinase 8 (MMP8) and MMP98,9, a finding confirmed in a study in which phosphorylated p38 and MMP9 increased after mtDNA treatment of neutrophils57. A separate study reported similar findings where pre-treatment with TLR9 inhibitor ODN2088 inhibited the activation of p38 MAPK and release of MMP854. Gu et al. also found that intratracheal administration of mtDNA provokes lung inflammation through TLR9–p38 MAPK58. Hip fracture in rats resulted in mtDNA release into the circulation as well as higher TLR9 and NFκB p65 activation and subsequent lung injury46. The role of other MAPKs such as extracellular signal-regulated kinases (ERKs) and c-Jun N-terminal kinases (JNKs) remains unclear and, to our knowledge, unexamined in this context. These data suggest a pathway where mtDNA activates neutrophils through TLR9 binding and activation of the MAPK pathway with subsequent MMP8 and MMP9 release (Figure 1).

When mtDNA is considered vis-à-vis the site and location of TLR9 receptor, mtDNA must be either displaced from whole mitochondria and moved into the cytosol or, when extracellular, internalised by some mechanism(s) to act on endosomal TLR9. The endosomal location of TLR9 is most likely a mechanism to avoid unwanted activation59. It is unclear how extracellular mtDNA is internalised, but possibilities include endocytosis, transmembrane diffusion, phagocytosis, and receptor-mediated endocytosis60. Transmembrane diffusion is unlikely because of the highly (negatively) charged nature of DNA, which makes it difficult to pass through the cellular membrane. A recent study found that monocyte-derived macrophages can take up whole mitochondria released from necroptosis, suggesting that phagocytosis could be a relevant mechanism61. The macrophage has a clear role in resolving inflammation by clearing up cellular debris and apoptotic bodies by phagocytosis. When mitochondria are not cleared during non-apoptotic cell death, the macrophage may phagocytose cellular corpses with mtDNA still abundantly present. Typically, apoptotic corpses can suppress the transcription of pro-inflammatory cytokine genes, promote the secretion of anti-inflammatory cytokines by phagocytes, and cause antigen-presenting cells to present dead cell antigen in a manner that promotes immunological tolerance (reviewed by Zitvogel et al.62). It will be of interest to consider the fate of mtDNA when macrophages or dendritic cells phagocytose cellular corpses with mtDNA. Does this clear the mtDNA or does it regulate subsequent functions (for example, immune responsiveness) in these cell types? This has yet to be studied in detail. It is also possible that binding to additional cofactors facilitates the internalisation into immune cells, and, in this instance, high-mobility group box 1 (HMGB1) and receptor for advanced glycation end products (RAGE) have been implicated63. In this study, HMGB1–CpG (class A) complexes resulted in TLR9/RAGE association and recruitment of MyD88 in B cells63. Here, RAGE was visualised as associating with the DNA and was internalised with some sequestered in endosome-like structures. However, this possible mechanism requires further investigation. It has also been proposed that activation of autoreactive B cells by CpG DNA occurs after B-cell receptor engagement, leading to the delivery of CpG DNA to endosomal TLR964.

Although nucleic acid-sensing TLRs on immune cells are found mainly within cells, cell surface expression has also been described. Via flow cytometry, TLR9 has been detected on the surface of resting B lymphocytes65,66 and peripheral blood mononuclear cells67,68. One functional ex vivo study found primary human and mouse TLR9 surface expression in neutrophils, which are upregulated by a variety of stimuli, including TLR9 agonists69. However, it remains unclear whether TLR9 is able to signal from the cell surface. In other cell types, TLR9 is also expressed on the cell surface. For example, TLR9 is expressed on both the apical and the basolateral membranes of intestinal epithelial cells, although NFκB is activated only via basolateral stimulation of CpG ligands70,71. This is relevant at the gut mucosal interface, as this limits the extent of TLR9 activation at the apical surface, which is in contact with a luminal milieu rich with bacterial DNA. Hence, compromised intestinal barrier integrity and translocation of bacterial CpG from the lumen during gut pathology will lead to basolateral stimulation in this context. Whether mtDNA has a different propensity compared with bacterial CpG to trigger TLR9 depending on epithelial site has not been studied.

The inflammasome

The inflammasomes are targets of mtDNA leading to cleavage and activation of caspase-1 and downstream maturation of interleukin-1β (IL-1β) and IL-1872. Here, it is the cytosolic release of mtDNA that exerts the dominant effect on inflammasome activation. Of the several inflammasomes described, the NLRP3 inflammasome is the best characterised in this regard. Nakahira et al. showed that depletion of mtDNA reduced IL-1β secretion in macrophages following treatment with known inflammasome triggers lipopolysaccharide (LPS) and ATP73. Of interest, mitochondria-derived reactive oxygen species (mtROS) is a further key mediator in this process. Pharmacologic induction of mtROS correlates with higher secretion of active IL-1β in an NLRP3- and caspase-1-dependent manner, and treatment with mtROS scavengers suppresses this effect74. The requirement for mtROS in NLRP3 activation has been confirmed by other studies73,7577 and may be explained by its oxidising effects on mtDNA. Shimada et al. showed that it is the oxidised form of mtDNA that confers the inflammatogenic potential to mtDNA75. mtROS enhances not only the oxidative process but also the cytosolic translocation of oxidised mtDNA that then binds directly to NLRP375. Non-oxidised mtDNA is insufficient to activate the NLRP3 inflammasome, although it may stimulate IL-1β production via other inflammasomes such as the absent in melanoma 2 (AIM2)78. Interestingly, genetic deletion of NLRP3 and caspase-1 results in less mtDNA release73,77. This suggests a positive-feedback loop, in which activation of the NLRP3 inflammasome by oxidised mtDNA further promotes mtDNA release. The overwhelming or persisting (or both) ROS production by inflammatory cells, for example, is known to damage macromolecules (DNA as well as RNA, lipids, carbohydrates, and proteins) of the surrounding cells. Activated neutrophils produce large amounts of ROS as part of their essential role in host defense79. Hence, this is a likely major contributory factor to mtDNA damage once the inflammatory process is triggered.

Other factors controlling mitochondria-mediated NLRP3 activation are also relevant. For example, defective autophagy increases caspase-1 activation, IL-1β and IL-18 production, and cytosolic mtDNA translocation in LPS- and ATP-primed macrophages76. Pharmacological inhibition of mitophagy/autophagy in human macrophages results in the accumulation of damaged mitochondria, ROS generation and IL-1β secretion74, and increased NLRP3 expression in the presence of LPS80. Hence, defective autophagy leads to inadequate clearance of damaged mitochondria, priming the internal cellular environment for NLRP3 activation. It is noteworthy that, given the diversity of NLRP3 activators, current literature suggests that the precise mechanism of NLRP3 activation is still under debate81. Although the role of the inflammasome is often considered separately from TLR9 here, there is evidence that TLR/NFκB activation is a necessary priming step leading to NLRP3 upregulation and subsequent downstream signalling. NFκB-activating stimulus is required for cells to express pro-IL-1β and NLRP382. Imeada et al. showed that stimulation of TLR9 by DNA fragments during early acetaminophen-induced cell death can lead to the transcriptional activation of the IL-1β gene, resulting in the formation of pro-IL-1β83. Using the acetaminophen hepatotoxicity model, they showed that NLRP3 deletion and related inflammasome components ASC and Caspase-1 were protective against induced liver failure83. A further study, however, did not show any effect of NLRP3 deletion on the outcomes of acetaminophen-induced liver failure84. Hence, in the context of liver necrosis, the role for NLRP3 inflammasome remains controversial.

STING pathway

The role of mtDNA in innate immunity through the STING pathway has also been a focus of recent studies. STING is a cytosolic protein anchored to the ER85. STING can be activated either by direct association with dsDNA or by cyclic dinucleotides, which can be derived from intracellular bacteria or viruses or produced by a DNA sensor, cyclic GMP–AMP (cGAMP) synthetase (cGAS)86. This, in turn, activates IRF3, which ultimately translocates to the nucleus and transcribes type I IFN genes, and also the NFκB pathway85.

Two independent groups recently discovered that the STING-mediated IFN response can also be activated by mtDNA87,88. They first observed that deficiency of apoptotic caspases (3, 7, and 9) resulted in upregulation of type I IFN genes. This response was dependent on Bak/Bax, pro-apoptotic proteins responsible for mitochondrial outer membrane permeabilisation leading to mtDNA release, and the release of cytochrome C, which activates the intrinsic apoptotic pathway. Typically, apoptosis is considered immunologic-silent; for example, it does not trigger an inflammatory response. However, these studies demonstrated that, when caspases (9 and 3/7) responsible for the completion of apoptotic process are inhibited or deleted, cytosolic mtDNA goes on to activate cGAS/STING-mediated type I IFN signalling87,88. Hence, these caspases serve as a “brake” on the mtDNA-inflammatory effect during cell death. mtDNA released during cell death has been previously reported to provide a second signal that cooperates with an additional inflammatory signal (for example, LPS) to activate the NLRP3 inflammasome and induce IL-1β production in murine macrophages75. Further evidence of an mtDNA role in STING-mediated IFN responses comes from West et al.89. Here, partial deficiency of the mtDNA-binding protein mitochondrial transcription factor A (TFAM) was associated with increased concentrations of cytosolic mtDNA and enhanced type I IFN response, which was attenuated by knockdown of components of the STING pathway.

Aberrant mtDNA–STING signalling has also been implicated in human inflammatory diseases, such as SLE. As discussed earlier, Lood et al. showed that treatment of human neutrophils with SLE-abundant ribonucleoprotein immune complexes induces mtROS, mtDNA oxidation, and translocation of the mitochondria to the plasma membrane41. Oxidised mtDNA is then released extracellularly as a component of NETs. Transfection of NET-derived mtDNA results in expression of IFN-β in human peripheral mononuclear cells. Systemic injection of oxidised mtDNA increases IFN-stimulated gene expression in the spleen of wild-type but not STING-deficient mice. Similar to inflammasomes, uncontrolled mtROS production promoting cytosolic mtDNA release is important in STING activation and potentially in the case of autoimmunity. These studies highlight the importance of the innate cellular functions to handle mtDNA release during the initiation of cell death, which ultimately will decide whether the ensuing fate will be that of a silent or inflammatory outcome.

Mechanisms for mtDNA release

Two levels of mtDNA release—cytosolic and then extracellular—are critically important steps (Figure 1). In the former, the mechanism of release of mtDNA from mitochondria relies on the opening of mitochondrial permeability transition (MPT) pores in the inner mitochondrial membrane90. Inhibition of pore opening with cyclosporine A resulted in lower mtDNA in the cytosol after stimulation with LPS and ATP73. Ding et al. showed that the induction of ROS using oxidised low-density lipoprotein (ox-LDL) increased mtDNA leakage into the cytosol in a dose-dependent manner, and this effect was ameliorated with blockade of the ox-LDL receptor or a ROS inhibitor91.

In terms of extracellular release, cellular stress and necrosis are primary factors in the non-discriminant liberation of a host of mitochondrial components such as mtDNA, N-formyl peptides, ATP, TFAM, and mitochondrial lipids. These mitochondrial constituents also exert their respective effects, which are wide-ranging, on key inflammatory pathways (extensively reviewed by Nakahira et al.81). Aside from this non-selective release after uncontrolled cell death, several studies have suggested additional mechanisms such as necroptosis (or programmed necrosis)92. Blood transfusion-induced endothelial necroptosis was recently found to increase extracellular mtDNA as a potential mechanism to explain transfusion-related lung injury93. A recent study suggested that, during necroptosis, mitochondria were released before plasma membrane rupture and then phagocytosed by monocyte-derived macrophages or dendritic cells, triggering an inflammatory response as shown by cytokine production and cell maturation, respectively61. Thus, ingestion of intact mitochondria may represent a distinct uptake mechanism following necroptosis. In a separate study, platelets were also found to be a source for free extracellular mitochondria release and then to act as an endogenous substrate for bactericidal secreted phospholipase A2IIA (sPLA2-IIA) leading to mitochondrial membrane hydrolysis, loss of mitochondrial structural integrity, and mtDNA release94. Intriguingly, Xin et al. found lower levels of mtROS production when metformin was added to activated platelets, and this was associated with decreased extracellular mtDNA release95. The authors found lower complex I activity of the platelet mitochondrial respiratory chain and suggested this as a mechanism for the observed suppressed mitochondrial dysfunction.

Whether there is an active element in mtDNA release is an interesting point of consideration. Active cellular transfer of mitochondria from stromal cells to rescue stricken lung alveoli cells in acute lung injury has been demonstrated96. Extracellular vesicles are important modes of intercellular communication and comprise exosomes (endosomal) and microvesicles (plasma membrane-derived) and are directed by exocytosis. Both chromosomal DNA97,98 and mtDNA99,100 have been observed in extracellular vesicles; it has been suggested that the transfer of altered mtDNA between cells may play a role in Alzheimer’s disease and skeletal muscle diseases99,100. As described earlier, in patients with NASH, a greater percentage of mitochondria was found inside extracellular microparticles and a higher percentage of microparticles contained mitochondria compared with lean subjects42. Furthermore, subjects with NASH had a higher level of oxidised mtDNA in microparticles. Further clarification is required on the concentration and significance of mtDNA in extracellular vesicles and whether this has different immunostimulatory effects compared with cell-free or surface-bound mtDNA. As previously mentioned, the pro-inflammatory effects of mtDNA are dependent on its oxidisation75,101. The highly oxidative extracellular milieu at sites of tissue inflammation in patients with chronic inflammatory disease may overwhelm anti-oxidant systems, further potentiating the inflammatory potential of DAMPs such as mtDNA5.

mtDNA degradation and clearance

Several well-described clearance mechanisms limit the pro-inflammatory nature of mtDNA. Autophagy as discussed earlier is important102. Defective autophagy has been implicated in several chronic inflammatory human diseases, including Crohn’s disease103. A proportion of circulating DNA in the bloodstream appears to cross the kidney barrier and be excreted in the urine104. Indeed, mtDNA has been detected in the urine at elevated levels in patients with progressive acute kidney injury105. This may be due to the inflammatory state associated with this condition, the increased clearance with a disturbed kidney barrier, or both. Another possible mechanism of mtDNA clearance is phagocytosis by macrophages in a manner similar to the ingestion of the structurally similar bacterial DNA106. As described earlier, the outcome of phagocytosis of intact mitochondria may be pro- rather than anti-inflammatory; these divergent effects may also be dependent on the phenotype of the phagocytosing cells (for example, inflammatory versus pro-resolution macrophages/monocytes, neutrophils, and red blood cells)61,93.

In general, non-host DNA in the circulation is digested in part by circulating nucleases, and mtDNA may be affected by a similar mechanism107. Intracellularly, DNases found in the autophagolysosome play a vital role in degrading mtDNA102,108. Oka et al. showed that cardiac-specific deletion of DNase II resulted in mtDNA accumulation in cardiomyocytes and the development of heart failure102. In human umbilical vein endothelial cells, lysosomal DNases protect cells against inflammation from mtDNA damage induced by ox-LDL91. Here, small interfering RNA (siRNA) knockdown of DNase II amplifies the mtDNA–TLR9-mediated inflammatory response91. It is unclear whether nucleases have a similar action on mtDNA in the extracellular space or are relevant in the physiological setting, especially when mtDNA is present in microvesicles or housed within intact mitochondria, which protect against DNase II. Intriguingly, DNase pre-treatment abolished renal mitochondrial injury that was observed after injection of mitochondrial debris (including mtDNA) in mice45. However, the precise role of DNase and its effect on the immunostimulatory effects of mtDNA is likely to be more complex, as illustrated by a recent study which showed that DNase II was required for TLR9 activation by bacterial genomic DNA109.

Conclusions: translational opportunities for mtDNA-mediated inflammation

mtDNA contributes to inflammation at multiple levels when tissue or cellular homeostasis is perturbed. Damaged mtDNA released into the cytosol has a functional short-range effect on immediate “alarm” systems such as the inflammasome and NFκB. Uncontrolled release of mtDNA into the circulation in conditions with significant tissue injury generates a more systemic effect whilst de-regulation of local mitochondrial homeostatic mechanisms such as autophagy or mtROS detoxification contributes to organ-specific pathology as observed in the heart and liver. Failure of such mechanisms may also give rise to a more wide-ranging consequence (for example, in autoimmune diseases such as SLE).

Our review shows that mtDNA-mediated inflammation is important and relevant to many human inflammatory diseases. However, this remains an underexplored field and more insights will likely emerge in the near future. The current evidence offers a rich realm of translational opportunities to target mtDNA-mediated inflammation. There are many plausible approaches, which include targeting cytosolic mtDNA release (for example, directly at MPT using cyclosporine or by specific mitochondrial anti-oxidant strategies, such as MitoQ1010 to reduce mtROS), augmenting clearance (for example, using autophagy activators or correcting factors leading to impaired autophagy), diverting the cellular response following mitochondrial damage (for example, induction of pro-apoptotic caspases), and reducing the inflammatory potential of mtDNA (for example, DNases to digest NET-bound mtDNA and reducing oxidation of mtDNA).

Given that mtDNA can be measured and used as a biomarker, it offers a unique opportunity to stratify and identify individuals who may benefit from specific therapeutic targeting of downstream inflammation pathways (for example, TLR9, NLRP3, or STING pathways). As discussed earlier, there are numerous studies in sepsis, trauma, and acute single-organ injury that have already demonstrated that individuals with high mtDNA levels and TLR9 expressions have worse prognosis. Therefore, there are clear groups in which stratification is useful. However, a number of challenges exist to its implementation as a biomarker, such as the variation in which mtDNA is measured (for example, serum versus plasma, mtDNA-specific PCR primers) and reported in the literature. Standardisation of these protocols, including the identification of “normal” and “abnormal” ranges, will be important prior to clinical use. Furthermore, many studies have failed to include clinically relevant predictive statistics; further studies reporting such statistics in a variety of inflammatory conditions are required.

In conclusion, multiple lines of data show that innate responses to mtDNA, which is similar to and evolutionarily derived from bacteria, are hard-wired into our biology and drive the development inflammation with pathologic consequences in many diseases.

Comments on this article Comments (0)

Version 1
VERSION 1 PUBLISHED 20 Feb 2017
Comment
Author details Author details
Competing interests
Grant information
Copyright
Download
 
Export To
metrics
Views Downloads
F1000Research - -
PubMed Central
Data from PMC are received and updated monthly.
- -
Citations
CITE
how to cite this article
Boyapati RK, Tamborska A, Dorward DA and Ho GT. Advances in the understanding of mitochondrial DNA as a pathogenic factor in inflammatory diseases [version 1; peer review: 3 approved] F1000Research 2017, 6(F1000 Faculty Rev):169 (https://doi.org/10.12688/f1000research.10397.1)
NOTE: it is important to ensure the information in square brackets after the title is included in all citations of this article.
track
receive updates on this article
Track an article to receive email alerts on any updates to this article.

Open Peer Review

Current Reviewer Status: ?
Key to Reviewer Statuses VIEW
ApprovedThe paper is scientifically sound in its current form and only minor, if any, improvements are suggested
Approved with reservations A number of small changes, sometimes more significant revisions are required to address specific details and improve the papers academic merit.
Not approvedFundamental flaws in the paper seriously undermine the findings and conclusions
Version 1
VERSION 1
PUBLISHED 20 Feb 2017
Views
0
Cite
Reviewer Report 20 Feb 2017
Augustine M.K. Choi, Division of Pulmonary and Critical Care Medicine, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, NY, USA 
Kiichi Nakahira, Department of Medicine, Weill Cornell Medicine, New York, NY, USA 
Approved
VIEWS 0
We confirm that we have read this submission and believe that we have an ... Continue reading
CITE
CITE
HOW TO CITE THIS REPORT
Choi AMK and Nakahira K. Reviewer Report For: Advances in the understanding of mitochondrial DNA as a pathogenic factor in inflammatory diseases [version 1; peer review: 3 approved]. F1000Research 2017, 6(F1000 Faculty Rev):169 (https://doi.org/10.5256/f1000research.11203.r19532)
NOTE: it is important to ensure the information in square brackets after the title is included in all citations of this article.
Views
0
Cite
Reviewer Report 20 Feb 2017
Mitchell R. McGill, Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, 63110, USA 
Approved
VIEWS 0
I confirm that I have read this submission and believe that I have an ... Continue reading
CITE
CITE
HOW TO CITE THIS REPORT
McGill MR. Reviewer Report For: Advances in the understanding of mitochondrial DNA as a pathogenic factor in inflammatory diseases [version 1; peer review: 3 approved]. F1000Research 2017, 6(F1000 Faculty Rev):169 (https://doi.org/10.5256/f1000research.11203.r19531)
NOTE: it is important to ensure the information in square brackets after the title is included in all citations of this article.
Views
0
Cite
Reviewer Report 20 Feb 2017
Antonio Ferrante, Women and Children's Hospital Campus, University of Adelaide, Adelaide, Australia 
Approved
VIEWS 0
I confirm that I have read this submission and believe that I have an ... Continue reading
CITE
CITE
HOW TO CITE THIS REPORT
Ferrante A. Reviewer Report For: Advances in the understanding of mitochondrial DNA as a pathogenic factor in inflammatory diseases [version 1; peer review: 3 approved]. F1000Research 2017, 6(F1000 Faculty Rev):169 (https://doi.org/10.5256/f1000research.11203.r20366)
NOTE: it is important to ensure the information in square brackets after the title is included in all citations of this article.

Comments on this article Comments (0)

Version 1
VERSION 1 PUBLISHED 20 Feb 2017
Comment
Alongside their report, reviewers assign a status to the article:
Approved - the paper is scientifically sound in its current form and only minor, if any, improvements are suggested
Approved with reservations - A number of small changes, sometimes more significant revisions are required to address specific details and improve the papers academic merit.
Not approved - fundamental flaws in the paper seriously undermine the findings and conclusions
Sign In
If you've forgotten your password, please enter your email address below and we'll send you instructions on how to reset your password.

The email address should be the one you originally registered with F1000.

Email address not valid, please try again

You registered with F1000 via Google, so we cannot reset your password.

To sign in, please click here.

If you still need help with your Google account password, please click here.

You registered with F1000 via Facebook, so we cannot reset your password.

To sign in, please click here.

If you still need help with your Facebook account password, please click here.

Code not correct, please try again
Email us for further assistance.
Server error, please try again.