Download Mitochondria in Lung Biology and Pathology: More than - AJP-Lung

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts

Cell culture wikipedia , lookup

Mitosis wikipedia , lookup

Cell encapsulation wikipedia , lookup

Cell cycle wikipedia , lookup

Signal transduction wikipedia , lookup

Cellular differentiation wikipedia , lookup

Amitosis wikipedia , lookup

DNA damage theory of aging wikipedia , lookup

Organ-on-a-chip wikipedia , lookup

List of types of proteins wikipedia , lookup

JADE1 wikipedia , lookup

Transcript
Articles in PresS. Am J Physiol Lung Cell Mol Physiol (April 18, 2014). doi:10.1152/ajplung.00073.2014
1
2
L-00073-2014 Revised (clean)
3
Mitochondria in Lung Biology and Pathology:
4
More than just a powerhouse
5
6
7
8
9
Paul T. Schumacker1, Mark N. Gillespie2†,
10
Kiichi Nakahira3, Augustine M.K. Choi3, Elliott D. Crouser4
11
Claude A. Piantadosi5, and Jahar Bhattacharya6
12
13
14
1
Northwestern University Feinberg School of Medicine, Department of
15
Pediatrics, Chicago, IL, 2University of South Alabama College of Medicine,
16
Department of Pharmacology, Mobile, AL, 3Weill Cornell Medical College,
17
Department of Medicine, New York, NY, 4The Ohio State University College
18
of Medicine, Department of Internal Medicine, Columbus, OH,
19
University School of Medicine, Department of Medicine, Durham, NC, and
20
6
21
Cellular Biophysics, New York, NY
Duke
Columbia University Medical Center, Department of Physiology and
22
23
24
25
26
27
28
29
30
5
†
Correspondence:
e-mail:
telephone:
Mark N. Gillespie, Ph.D
Department of Pharmacology
College of Medicine
University of South Alabama
Mobile, AL 36688
[email protected]
(251) 460-6497
Copyright © 2014 by the American Physiological Society.
31
32
ABSTRACT
33
An explosion of new information about mitochondria reveals that their importance
34
extends well beyond their time honored function as the “powerhouse of the cell.” In this
35
Perspective, we summarize new evidence showing that mitochondria are at the center
36
of a reactive oxygen species (ROS)-dependent pathway governing the response to
37
hypoxia and to mitochondrial quality control. The potential role of the mitochondrial
38
genome as a sentinel molecule governing cytotoxic responses of lung cells to ROS
39
stress also is highlighted. Additional attention is devoted to the fate of damaged mtDNA
40
relative to its involvement as a Damage Associated Molecular Pattern driving adverse
41
lung and systemic cell responses in severe illness or trauma.
42
strategies for replenishing normal populations of mitochondria after damage, either
43
through promotion of mitochondrial biogenesis or via mitochondrial transfer, are
44
discussed.
45
46
47
48
49
50
51
52
53
54
55
56
57
58
59
60
2
Finally, emerging
61
62
63
INTRODUCTION
“The powerhouse of the cell.”
All of the contributors to the 2013 ATS
64
symposium forming the basis of this Perspective Article, and perhaps many of the
65
attendees, too, recalled this as their first introduction to mitochondria and often their first
66
memorable phrase in biology. Though still true, it now seems almost quaint given the
67
explosion of information about mitochondrial functions unrelated to energy metabolism
68
that has occurred over the past two decades or so. Our symposium was motivated
69
partly by this “explosion” and partly by the equally rapid accumulation of evidence that
70
mitochondria likely play crucial roles in lung disease with significant potential to serve as
71
targets in pulmonary and critical care medicine.
72
This Perspective article does not seek to comprehensively review all
73
developments in the mitochondrial field that bear on lung disease. Rather, it highlights
74
evolving concepts of how mitochondria contribute to normal lung health, and how
75
mitochondria could be targeted for specific intervention in lung disease. Having
76
registered this, we present concise summaries pertaining to mitochondria, reactive
77
oxygen species (ROS) and oxygen sensing in the lung, the role of the mitochondrial
78
genome as a sentinel molecule governing lung cell survival in response to oxidant
79
stress, the nature of mtDNA Damage Associated Molecular Patterns (DAMPs)
80
originating from the lung and their potential use and importance as biomarkers and
81
proinflammatory alarm signals, respectively. Finally, we explore the intriguing notion
82
that therapeutic strategies aimed at reconstituting normal mitochondrial population and
83
function may comprise therapeutic approaches in acute lung injury.
3
84
85
Mitochondrial oxygen sensing
Mitochondria consume O2 at cytochrome c oxidase, the terminal complex in the
86
electron transport chain (ETC).
Electron flux along the ETC is linked to proton
87
translocation across the inner mitochondrial membrane, resulting in the generation of an
88
electrochemical gradient that is used by the ATP synthase to generate ATP during
89
oxidative phosphorylation. As mitochondria convert O2 to water the intracellular O2
90
tension decreases, thereby generating a gradient for O2 diffusion into the cell from the
91
extracellular space. Mitochondria are the principal site of oxygen consumption in the
92
cell, so their oxygen tension is lower than any other organelle. This characteristic has
93
led investigators to ask whether mitochondria function as oxygen sensors in the cell
94
(64). A conceptually simple idea is that decreases in cellular O2 levels (tissue hypoxia)
95
would limit mitochondrial oxygen consumption, leading to redox changes as the ETC
96
complexes became saturated with electrons due to the loss of cytochrome c oxidase
97
activity (56).
98
systems that feed electrons into the ETC, such as the Krebs cycle.
99
conceptual problem with this idea arose from experimental studies showing that
100
mitochondria do not become limited by oxygen availability until the cell is nearly anoxic
101
(12, 95). Specifically, how could the mitochondria respond to physiological levels of
102
hypoxia if their ability to signal via redox alterations was not activated until all of the
103
oxygen was gone? For many years this paradox represented a conceptual barrier to
104
the idea that mitochondria might be involved in oxygen sensing. Some investigators
105
suggested that specialized oxygen sensors such as the glomus cells of the carotid body
106
might circumvent this problem by expressing special isoforms of cytochrome c oxidase
These redox changes would then be reflected in the redox status of
4
However, a
107
with low apparent affinity for O2 (59). In those cells, electron transport and oxygen
108
consumption would become O2 supply-limited at milder levels of hypoxia, allowing the
109
mitochondria to detect subtle decreases in cellular oxygenation (58).
110
experimental evidence for the existence of such a low-affinity oxidase has been
111
reported. Moreover, mammalian cells with high-affinity forms of cytochrome c oxidase
112
are still capable of detecting and responding to hypoxia.
However, no
113
A second hypothesized model of oxygen sensing was promoted by Michelakis,
114
Archer and Weir, who sought to explain how pulmonary artery smooth muscle cells
115
detect hypoxia and trigger acute hypoxic pulmonary vasoconstriction (4). Their model
116
proposed the existence of an intracellular oxidase that would generate more ROS at
117
high O2 levels and less under hypoxic conditions. According to that model, ROS would
118
then activate voltage-dependent potassium (Kv) channels in the plasma membrane.
119
Conversely, the lesser oxidation of these channels during hypoxia would promote
120
channel closure, causing membrane depolarization that would lead to the entry of
121
extracellular Ca2+ through L-type calcium channels. However, when they tested the
122
effect of gp91-(phox) deletion on this response, using mice with genetic deletion of
123
NOX2, they found that the cellular response to hypoxia was still present (3). They
124
subsequently considered that mitochondria were the source of these ROS signals,
125
based on studies using various mitochondrial inhibitors (57). However, a conceptual
126
problem with that model has been the consistent failure of diverse chemical antioxidants
127
or the expression of enzymatic antioxidant systems to activate hypoxic responses in a
128
normoxic cell.
5
129
A third model of mitochondrial oxygen sensing (FIGURE 1) arose from initial
130
studies showing that intracellular ROS levels actually increase during hypoxia (13, 26).
131
Parallel studies using chemical mitochondrial inhibitors implicated complex III as the
132
likely source of ROS generation.
133
because it seemed counter-intuitive, and was based on the use of non-specific
134
fluorescent probes to detect intracellular ROS levels and pharmacological inhibitors to
135
block the ETC (34, 90).
136
inhibitors at various concentrations reported conflicting responses. To address this,
137
Waypa et al. utilized a novel genetically encoded fluorescent protein sensor to detect
138
intracellular ROS levels. The initial sensor, HSP-FRET, exhibited ratiometric behavior
139
that obviated an important limitation of the fluorescent chemical probes (91).
140
Specifically, ratiometric sensors provide a signal that is independent of the light intensity
141
used to excite the probe.
142
pulmonary artery smooth muscle cells confirmed the previous observation that ROS
143
levels increase in the cytosol during hypoxia (91).
144
during hypoxia was shown to be required for the increase in cytosolic Ca2+ activation in
145
pulmonary artery smooth muscle cells. Further advances in this hypoxia-induced ROS
146
signaling model came with the introduction of roGFP, a redox-sensitive mutant of green
147
fluorescent protein (24, 35). This ratiometric sensor provides a specific measure of
148
protein thiol oxidation/reduction status in the cell. Two important features contributed to
149
the importance of this advance. First, its oxidation is reversible in the cell allowing it to
150
be calibrated, analogous to the behavior of ratiometric Ca2+ sensors.
151
expression can be targeted to specific intracellular compartments, allowing assessment
This paradoxical response was initially criticized
Importantly, different laboratory groups using the same
Importantly, studies using HSP-FRET expressed in
6
Moreover, the increase in ROS
Second, its
152
of redox at the subcellular level.
Using roGFP expressed in cytosol, mitochondrial
153
intermembrane space and in the mitochondrial matrix, Waypa et al. found that
154
significant differences in basal oxidant status exist across these compartments in
155
PASMC, with the cytosol existing in a more reduced state, the mitochondrial matrix in a
156
highly oxidized state, and the intermembrane space at an intermediate level (92).
157
During acute hypoxia, oxidation status in the intermembrane space and the cytosol
158
increased, whereas oxidation in the mitochondrial matrix decreased. These findings
159
were consistent with a model in which the release of superoxide from complex III to the
160
intermembrane space increases during hypoxia, allowing the ROS to potentially exit to
161
the cytosol and initiate redox signaling involved in triggering the release of intracellular
162
calcium stores (94). By contrast, hypoxia caused an attenuation of oxidant stress in
163
mitochondrial matrix.
164
compartments could reflect a distinct operation of two independent mechanisms
165
controlling ROS generation during hypoxia in the two domains.
166
possibility that ROS alters the direction of ROS secretion from the membrane – toward
167
the matrix during normoxia and toward the intermembrane space during hypoxia – has
168
not been ruled out.
The opposing redox responses to hypoxia in these two
Alternatively, the
169
In either case, studies were needed to more critically test the hypothesis that the
170
mitochondrial electron transport chain is the source of ROS signaling during hypoxia.
171
Accordingly, Waypa et al. developed a genetic model by generating a mouse to permit
172
conditional deletion of the Rieske iron-sulfur protein (RISP) (93). This nuclear encoded
173
protein is critical for the function of complex III, where it accepts an electron from
174
ubiquinol, the carrier that moves electron into complex III from complexes I and II (34).
7
175
ROS generation from complex III can occur after RISP accepts an electron from
176
ubiquinol, generating the intermediate free radical, ubisemiquinone.
177
electron on ubisemiquinone is normally transferred to the b-cytochromes in the
178
complex, but can potentially be transferred instead to O2, generating superoxide. If
179
RISP is deleted, ubisemiquinone, and thus superoxide, cannot be generated at complex
180
III (33, 54). Accordingly, that group studied PASMC isolated from the conditional RISP
181
mice and used Cre recombinase expression to delete the RISP gene. After loss of
182
RISP was confirmed, subsequent roGFP studies demonstrated that the hypoxia-
183
induced changes in redox status in the cytosol, intermembrane space and the
184
mitochondrial matrix were abolished.
185
functional complex III was required for the changes in redox observed in different
186
subcellular compartments during hypoxia (93). In parallel studies, it was found that the
187
transient increase in cytosolic Ca2+ in PASMC during acute hypoxia was abrogated in
188
cells where RISP had been deleted (93).
189
The remaining
These studies therefore demonstrated that a
A technical limitation of studies using cultured PASMC is the possibility that the
190
cells will undergo a phenotypic shift in culture.
191
cannot provide an assessment of the role of those responses in the intact lung.
192
Accordingly, that group conducted in vivo studies in which RISP was deleted from
193
smooth muscle cells by breeding the conditional RISP mice with a transgenic line
194
expressing a smooth muscle-specific, tamoxifen-inducible Cre (96). The resulting mice
195
were administered tamoxifen to activate nuclear localization of the Cre in smooth
196
muscle cells, and subsequent deletion of RISP from smooth muscle was confirmed.
197
The acute pulmonary vasoconstriction response was then compared in the knockout
8
Moreover, studies in cultured cells
198
and control mice, by measuring the right ventricular systolic pressure (RVSP) response
199
to acute alveolar hypoxia. Control mice exhibited a significant increase in RVSP during
200
5% O2 ventilation, which was significantly attenuated in the RISP-deficient mice (93).
201
These findings indicate that a functional complex III is required for the acute
202
vasopressor response to hypoxia in the lung.
203
Does the same mitochondrial ROS signal trigger other functional responses to
204
hypoxia? Mungai et al. tested the hypothesis that hypoxia activates the cellular energy
205
sensor, AMP-dependent kinase (AMPK), through an ROS-dependent mechanism. In
206
PASMC, they found that ROS generation during moderate hypoxia led to AMPK
207
activation, even in the absence of any increase in AMP or ADP concentrations (60).
208
Those findings suggest that mitochondrial ROS signals activate AMPK as an early
209
response to hypoxia, allowing it to activate protective mechanisms before the cell
210
progresses into a bioenergetic crisis where AMP and ADP concentrations are
211
increased.
212
Do specific antioxidants block these responses? To test this, Sabharwal et al.
213
reasoned that the expression of an enzymatic scavenger of H2O2 exclusively in the
214
mitochondrial intermembrane space (IMS) should abolish functional responses to
215
hypoxia by preventing ROS released from complex III to reach the cytosol (79).
216
Peroxiredoxin-5 (PRDX5) is an atypical 2-cysteine-containing peroxidase that is
217
normally expressed in multiple subcellular compartments including the mitochondrial
218
matrix (45). This enzyme functions as a monomer, whereas other H2O2 scavengers
219
only function as multimers. They targeted its expression to the intermembrane space
220
using the N-terminal sequence that directs the SMAC/Diablo protein to that
9
221
compartment (66), and confirmed correct localization using immunogold electron
222
microscopy (79).
223
PASMC, it attenuated the increase in ROS in the IMS and the cytosol during hypoxia.
224
Importantly, the PRDX5-expressing cells also showed a significant attenuation of the
225
acute hypoxia-induced activation of cytosolic Ca2+.
226
independent demonstration that ROS release from the mitochondrial inner membrane to
227
the IMS and subsequently to the cytosol is critical for triggering acute responses to
228
hypoxia in PASMC.
When PRDX5 was expressed in the intermembrane space of
These findings provide an
229
Previous studies by this group had shown that the stabilization of Hypoxia-
230
Inducible Factor-1alpha (HIF-1) is triggered by hypoxia-induced release of ROS from
231
mitochondria. In PASMC, hypoxia also triggers stabilization of HIF-
232
prolyl hydroxylase, which targets the protein for degradation by hydroxylating conserved
233
proline residues during normoxia (38, 41).
234
PRDX5 in the IMS, hypoxia-induced HIF-1 stabilization was inhibited in a dose-
235
dependent manner (79).
In PASMC expressing graded levels of
236
Collectively, these findings indicate that mitochondria function as more than
237
simple factories supplying ATP to the cell. Through the regulated release of ROS to the
238
cytosol, these organelles have the capacity to orchestrate a diverse group of adaptive
239
responses to hypoxia. In that regard, the moniker of “cellular oxygen sensors” clearly
240
applies to these organelles.
241
242
10
243
Sentinel functions of mitochondrial DNA (mtDNA) in pulmonary
244
vascular cells
245
The nuclear and mitochondrial genomes differ in certain fundamental respects.
246
For example, in comparison to the nuclear genome, the mitochondrial genome is tiny.
247
Even if all the mtDNA molecules in cells with high mitochondrial densities are totaled, it
248
is still a small fraction of the size of the nuclear genome. Following from this, mtDNA
249
encodes for only a handful of proteins compared to the many thousands encoded by
250
nuclear DNA. Second, the structure and regulation of the mitochondrial genome is
251
comparatively simple. Unlike nuclear DNA, in which the state of compaction, its
252
interactions with multiple transcription factors and co-activators, and epigenetic
253
determinants are all exquisitely regulated processes culminating in precise control of
254
gene expression, the mitochondrial genome is expressed as a single unit and its
255
transcription is controlled by a handful of mtDNA binding proteins, particularly
256
Mitochondrial Transcription Factor A (TFAM, (10, 44). Instead of compaction within a
257
nucleosome context, a single molecule of mtDNA is associated with about 30 core
258
proteins in a structure known as the nucleoid. Most if not all of the enzymes needed for
259
mtDNA replication and mtRNA expression appear to be components of the nucleoid (9).
260
Another difference between mitochondrial and nuclear DNA particularly relevant
261
to the present discussion is that mtDNA is about 50-fold more sensitive to oxidative
262
damage (7, 16, 32, 97). Reasons for this heightened sensitivity are not entirely known,
263
but have been attributed to a comparatively simple process of mtDNA repair
264
(mitochondria seem to contain only base excision repair while the nucleus has multiple,
11
265
overlapping pathways (8), and the prospect that the open structure of mtDNA is more
266
accessible to DNA-damaging ROS. In light of the forgoing comments that mitochondrial
267
ROS generation plays a central role in hypoxic signal transduction, the question can be
268
raised as to whether mtDNA is damaged by oxidation in hypoxic environments. While
269
DNA integrity across the entire mitochondrial genome in hypoxic cells has not been
270
examined, a large expanse of the mtDNA coding region - including the “common
271
deletion sequence” known to be sensitive to oxidant stress (25) - fails to display
272
oxidative base modifications in hypoxia (31). Perhaps this is because in hypoxia, the
273
matrix, where mtDNA resides, becomes more reduced in hypoxia as ROS are vectored
274
into the intermembrane space and cytosol (92).
275
In marked contrast to the lack of effect of hypoxia-induced mitochondrial ROS
276
generation on mtDNA integrity, when cells or intact lung tissue are challenged with
277
exogenous ROS the nuclear genome is largely spared of lesions while mtDNA is
278
extensively damaged (7, 16, 32, 97). One question that emerges from this interesting
279
observation pertains to the biological value in having a mitochondrial genome sensitive
280
to damage by exogenous oxidants while the nuclear genome is relatively resistant
281
potential answer, discussed subsequently, is that mtDNA functions as a “fuse” or a
282
“sentinel molecule” such that before an externally applied oxidant stress rises to a level
283
that threatens the nuclear genome with mutation, oxidative mtDNA damage triggers
284
both death of the affected cell and promotes the dissemination signals to “alarm”
285
neighboring and itinerant cells.
A
286
Multiple lines of evidence support the idea that mtDNA serves as a molecular
287
sentinel dictating cell survival in response to oxidant stress. As just noted, in virtually all
12
288
cell types studied thus far mtDNA is highly sensitive to ROS-induced damage (7, 32,
289
97).
290
mediated cell death, with the propensity for cytotoxicity inversely related to the efficiency
291
of mtDNA repair (32, 37). Genetic modulation of the first and rate-limiting step in mtDNA
292
repair, mediated by Ogg1 (19), a DNA glycosylase that detects and excises oxidatively
293
damaged purines – coordinately regulates ROS- and reactive nitrogen species-induced
294
mtDNA damage and cell death in all cultured cell populations so far examined (14, 21,
295
47, 67, 72, 73). Particularly relevant here are studies using mitochondrially-targeted
296
Ogg1 constructs, which eliminate the possibility that the biological consequences of
297
Ogg1 over-expression are related to effects on nuclear DNA repair.
298
pulmonary artery endothelial cells show that over-expression of mitochondrially-targeted
299
Ogg1 reduces sensitivities to both oxidative mtDNA damage and cytotoxicity evoked by
300
exogenous ROS stressors (21, 77).
301
siRNA, while not sensitizing nuclear DNA to oxidative damage, leads to persistence of
302
ROS-induced damage to the mitochondrial genome, increased apoptosis and
303
cytotoxicity (78).
There is also a conspicuous association between mtDNA damage and ROS-
Studies in
Conversely, reduction in total cell Ogg1 using
304
While these observations suggest that mtDNA integrity is a determinant of ROS-
305
induced cytotoxicity, many questions remain about the specific pathway(s) by which
306
oxidative mtDNA damage dictates lung cell survival and function. The link between
307
ROS-induced mtDNA damage and cell death traditionally has been ascribed to the idea
308
that persistent damage leads to diminished mtDNA transcription, defective ETC
309
function, and resulting formation of a regenerative cycle of excessive pro-apoptotic ROS
310
production and increasing mtDNA damage that ultimately culminates in mitochondrially-
13
311
driven cell death (11, 39, 75). However, this traditional model has never been
312
thoroughly explored, and more importantly, emerging evidence suggests that other
313
pathways are equally plausible. Protective actions of mtDNA repair enzymes against
314
ROS-induced lung cell death and dysfunction might not be linked to mtDNA repair, but
315
rather, to their interaction with oxidatively damaged mtDNA or mtDNA damage-related
316
proteins.
317
defective Ogg1 mutant protected against asbestos-induced apoptosis in A549 cells (67).
318
Because the repair-defective enzyme, like the wild-type enzyme, bound aconitase, the
319
possibility was advanced that Ogg1 influences ROS-mediated signaling events
320
downstream of mtDNA damage (80). In a related context, it has recently been shown
321
that Ogg1 in complex with free 8-oxoguanine serves as a guanine exchange factor to
322
activate Ras-mediated signaling (29), but whether this occurs in intact cells with ROS-
323
induced mtDNA damage has not been established.
In this regard, over-expression of a mitochondrially-targeted DNA repair-
324
As a means of exploring involvement of oxidative mtDNA damage in intact lung
325
and animal models of pulmonary disease, Wilson and colleagues developed novel
326
fusion-protein constructs targeting DNA repair glycosylases, either Ogg1 or the bacterial
327
enzyme Endo III, to mitochondria (48). Early work showed that these fusion protein
328
constructs prevented both hydrogen peroxide-induced mtDNA damage and increases in
329
the vascular filtration coefficient in isolated perfused rat lungs (16). The Ogg1 construct
330
also attenuated ventilator-induced lung injury and mortality in mice (36) and hyperoxia-
331
induced dysmorphogenesis in fetal rat lung explants (28).
332
suggest that mitochondrially-targeted Ogg1 inhibits mortality and acute lung injury
333
evoked by intratracheal P. aeruginosa in rats (15).
14
Preliminary studies also
334
These observations on the effectiveness of increasing mtDNA repair in isolated
335
lungs and animal models are a step towards determining if mtDNA integrity is a
336
legitimate pharmacologic target in lung disease. However, there are many unresolved
337
issues. As noted above, some pertain to the specific mechanism whereby increased
338
mtDNA repair protects against cytotoxicity. As an extension of this, whereas protection
339
of mtDNA integrity in cultured cells inhibits ROS-induced cytotoxicity, the involvement of
340
cytotoxicity per se in intact animal models of acute lung injury is complex. Taking this
341
analysis one step further, it is easy to envision how oxidative mtDNA damage could, by
342
different mechanisms, mediate different types of cellular responses in lung disease. For
343
example, ROS-induced cytotoxicity, driven by persistent mtDNA damage, defective
344
mtDNA transcription and attendant bioenergetic insufficiency, could be lethal to some
345
populations of lung cells, while oxidant-induced alterations in mtDNA-protein stability
346
might acutely influence behavior of other lung cell populations secondary to alterations
347
in cytoskeletal integrity (74).
348
mechanism(s) of action and pharmacologic utility of protecting mtDNA integrity in the
349
many pulmonary disorders where acute oxidant stress plays a pathogenic role.
Additional studies will be required to determine the
350
It has been appreciated for some time that ROS contribute to the evolution of
351
ALI/ARDS and other lung diseases, possibly through an adverse effect on mitochondrial
352
function (11). Indeed, about 40 clinical trials have examined the effect of anti-oxidant
353
strategies in acute lung injury and multiple organ system failure, but the therapeutic
354
effects of anti-oxidant strategies have been unimpressive (88). These disappointing
355
outcomes may be attributed in part to the heterogeneous nature and onset of ALI/ARDS
356
that tend to confound design and interpretation of clinical trials. There also remains
15
357
considerable scientific uncertainty about molecular targets of anti-oxidant drug action.
358
For example, non-selective antioxidants may disrupt signaling required for physiologic
359
adaption, such as hypoxia-mediated ventilation-perfusion matching in the injured lung,
360
lung cell survival and recovery. Strategies currently available also may not target the
361
key sentinel molecule(s) triggering adverse cellular effects of ROS.
362
suggested by the foregoing discussion, mtDNA could be such a sentinel molecule.
Perhaps, as
363
364
Mitochondrial DNA DAMP Release and its Cellular Targets in Lung
365
Disease
366
An intriguing question to emerge from the prospect that mtDNA damaged by
367
oxidation in the setting of oxidant lung injury pertains to the fate of the molecule. As has
368
been suggested from studies in other experimental systems (20), if mtDNA damage is
369
severe and not promptly repaired, does the molecule fragment after which it is exported
370
from the organelle, possibly as part of mitophagy? And if oxidatively-damaged mtDNA
371
fragments are exported from mitochondria, what are the next destinations of the
372
fragments and do they play a role in disease pathogenesis? These questions, depicted
373
in FIGURE 2, are discussed below.
374
If unrepaired, damaged mtDNA is rapidly degraded (81). Interestingly, mtDNA so
375
damaged displays a higher abundance of sugar-phosphate backbone lesions and
376
abasic sites relative to oxidized base products, which would be expected to fragment
377
the DNA rather then lead to persistent mutagenic lesions. There is scant evidence
378
concerning the fate of mtDNA fragments formed as a consequence of oxidative
16
379
damage. In a study using isolated mitochondria subjected to an exogenous oxidant
380
stress, Garcia and Chavez reported that mtDNA fragments less than 1000bp were lost
381
from the organelle via the permeability transition pore (27). Studies in macrophages
382
also indicate that the permeability transition pore is required for cytosolic accumulation
383
of mtDNA after immunologic stimulation (61), but whether there is some specificity for
384
fragment size or other attributes is unknown. In cardiomyocytes subjected to
385
hemodymamic stress-induced mitochondrial damage, mtDNA fragments accumulate in
386
autolysosomes where, on the one hand, Dnase II is capable of degrading the fragments
387
to biological insignificance (65).
388
degradation appear capable of autonomously activating endosomal TLR9 (65). In
389
immunologically-stimulated macrophages, mtDNA fragments accumulating in the
390
cytoplasm function as a co-activator in the NALP3-dependent activation of caspase 1
391
(61). Importantly, both TLR9 and NALP3 activation can initiate downstream pro-
392
inflammatory signaling.
On the other hand, mtDNA fragments escaping
393
In severely injured (100) and critically ill human subjects (62) mtDNA
394
accumulates in the circulation where it functions as a DAMP and causes TLR9-
395
dependent activation of the innate immune system. Here, too, the mechanism by which
396
mtDNA fragments exit cells has received little study.
397
interferon-primed
398
accumulation of mtDNA fragments in the extracellular space. The release process –
399
described as “catapult-like” – occurred rapidly and was not dependent on death of the
400
cells (98). Whether a similar process accounts for release of mtDNA from other types of
401
oxidatively- or mechanically-stressed cells is entirely unknown. Indeed, whether mtDNA
eosinophils,
lipopolysaccharide
17
In interleukin-5- or gamma
causes
the
ROS-dependent
402
DAMP release is inextricably linked to autophagy, mitophagy or cell death remains to be
403
established.
404
As noted previously, mtDNA is normally localized in a multi-protein complex
405
within the mitochondrial matrix, referred to as a nucleoid, which contains the essential
406
transcription factor and structural protein TFAM (9, 30, 44). TFAM has a high binding
407
affinity for mtDNA and remains associated when DNA is immunoprecipitated from lysed
408
cells (43). TFAM is a functional and structural homologue of the nuclear DNA binding
409
protein HMGB1, and both TFAM and HMGB1 are potent ligands for RAGE.
410
context promoting inflammation, RAGE ligands are shown to amplify the immune
411
responses to pattern-recognizing Toll-like receptors (43). In contrast to nuclear DNA
412
which is non-immunogenic, mtDNA, like viral and bacterial DNA, is enriched with
413
hypomethylated CpG motifs, which are natural ligands for TLR9, a highly specific
414
intracellular (endosomal) pattern recognizing receptor (50).
415
TFAM and mtDNA, it is not surprising that TFAM remains associated with mtDNA
416
following cell necrosis, and this complex is particularly potent in terms of promoting
417
TLR9-responsive immune cell activation (43).
In the
Given these properties of
418
Whereas many cell types are capable of responding to exogenous and
419
endogenous danger signals, including PMNs, monocytes, and even epithelial cells (50),
420
dendritic cells are specialized for sensing and processing immunogenic antigens to
421
initiate and coordinate complex and sustained immune responses.
422
present in low abundance (<1%) in blood and other tissues, immature dendritic cells,
423
particularly plasmacytoid DCs (pDCs) and immature myeloid DCs (iDCs), are uniquely
424
equipped to initiate and ultimately sustain sterile immune responses to immunogenic
18
Despite being
425
DNA, including viral and bacterial DNA. Immature DCs are unique in their capacity to
426
simultaneously promote pro- and anti-inflammatory immune responses.
427
distinguished from other immune cell lines by the lack of expression of common T-cell,
428
B-cell or myeloid cell surface markers, and are further discriminated by their ability to
429
produce large quantities of type I interferons in response to immunogenic (e.g., viral or
430
bacterial) RNA and DNA, recognized by TLR7/8 and TLR9, respectively.
431
iDCs and pDCs, through the production of TNFa, further promote “bystander” activation
432
of regional macrophages, are induced to mature, and are targeted to various tissues via
433
expression of specific chemokine receptors [e.g., CCR2 targets DCs to lung (53), where
434
they are capable of presenting antigen via MHC class II molecules to promote potent T
435
cell-mediated adaptive and counter-regulatory immune responses (FIGURE 3).
They are
Activated
436
In the context of acute cell damage, immature DCs respond specifically to
437
mitochondrial danger signals; indeed, the TFAM/mtDNA complex has been shown to be
438
sufficient to promote the simultaneous release of TNFa and IFNa from purified pDCs
439
(42, 43) (FIGURE 3A).
440
approximating the complex interaction of immune cells in vivo, TFAM was found to
441
augment the immune response to CpGA DNA (a readily available surrogate for mtDNA).
442
Furthermore, the depletion of pDCs, representing a small fraction of the total splenocyte
443
culture population, dramatically suppressed the innate immune response to TFAM/CpG
444
complex, demonstrating the capacity of pDCs to amplify both proinflammatory and
445
counter-regulatory immune pathways (FIGURE 3B). Together, these data indicate that
446
TFAM remains associated with mtDNA following acute cell damage, that TFAM serves
447
to augment the immune response to mtDNA, and immature DCs are important sensors
Moreover, using a splenocyte culture model closely
19
448
for TFAM/mtDNA complex and important regulators of the sterile immune response to
449
these mitochondrial danger signals.
450
New evidence indicates that DCs, particularly pDC-like macrophages, are of
451
critical importance during the pathogenesis of acute lung injury in the context of sterile
452
inflammation, and mitochondrial danger signals are incriminated as proximal mediators.
453
Immunogenic DNA, such as is present in bacteria and mitochondria, is sufficient to
454
promote ALI in animal models (46, 87), and mtDNA has been specifically linked to the
455
pathogenesis of ALI in setting of sterile inflammation induced experimentally by acid
456
aspiration(17), surgical trauma (100), and acute liver necrosis (55).
457
through activation of TLR9, appear to play a major role in initiating ALI under these
458
conditions. By contrast, recruitment of pDC-like macrophages to the lung is shown to
459
attenuate ALI induced by LPS, particularly in the acute phase of lung injury (49). These
460
pDC-like macrophages express CCR2 and are recruited to the lung by CC chemokine
461
ligand-2 (CCL2), whereupon they facilitate the clearance of damaged cells (52). In
462
addition to enhanced clearance of damaged cells, pDC activation in the lungs results in
463
the release of Type 1 interferons which are capable of suppressing ALI by altering the
464
balance between pro-inflammatory (e.g. IL-1b, IL-6) and anti-inflammatory (e.g., IL-10)
465
cytokines (5). Finally, lung DCs present antigen to and thereby activate regulatory T
466
cells (Tregs), which, in turn, contribute to the resolution of inflammation during the sub-
467
acute phase of ALI(1) (FIGURE 3A).
468
acute damage inflicted by activated PMNs is counterbalanced by pDCs in the context of
469
ALI induced by mitochondrial danger signals.
Neutrophils,
Thus, the current evidence suggests that the
470
20
471
Circulating Mitochondria DNA: A Biomarker of Sepsis and ARDS in
472
the ICU
473
As noted above, accumulating evidence suggests that mitochondrial damage
474
may engender the export mtDNA DAMPs that activate innate immune responses and
475
possibly propagate injury to distant targets. In this regard, while biomarkers can be
476
useful for identifying patients with critically illness or assessing the responses to the
477
therapies for the patients in the intensive care unit (ICU), a small number of biomarkers
478
reflecting the disease severity are commonly measured in clinical practice (e.g. lactate,
479
(71). The question thus arises as to whether mtDNA DAMP may be a useful biomarker.
480
Inflammasomes are multi-protein complexes that activate caspase-1 and
481
downstream immune responses, including the maturation and secretion of pro-
482
inflammatory cytokines (i.e. interleukin (IL)-1β and IL-18) (18). Recent studies show
483
inflammasomes are involved in the pathogenesis of various diseases such as infection,
484
cancer and metabolic diseases (18). Inflammasomes are activated by a wide range of
485
signals of pathogenic and non-pathogenic origins (host cell derived molecules including
486
extracellular ATP, hyaluronan, amyloid-β fibrils and uric acid crystals) (18).
487
To examine the involvement of inflammasome in critical illnesses, Choi and
488
colleagues first performed gene expression profiling analysis of lungs from mice
489
subjected to mechanical ventilation (MV), an experimental animal model of acute lung
490
injury (ALI). Specific changes in expression were identified for caspase-activator
491
domain-10, interleukin-18 receptor-1 (Il18r1), and Il1r2 and interleukin-1 (Il1b) as
492
genes following MV (22, 23).
Since genes representing inflammasome complex
21
493
molecules and the downstream cytokines were regulated in animal models of ALI, it was
494
then determined whether inflammasome family genes are also regulated in human
495
critical illnesses such as acute respiratory distress syndrome (ARDS), a lethal disease
496
in ICU, and sepsis, a major cause of ARDS and a leading cause of death in ICU. Total
497
blood RNA was extracted from patients admitted in medical ICU to determine the global
498
gene expression profile of ICU controls, patients with systemic inflammatory response
499
syndrome (SIRS), sepsis, and sepsis/ARDS. The gene expression profiling analysis
500
revealed that inflammasome-associated genes (e.g. asc and Il1b) were up-regulated in
501
patients with sepsis/ARDS when compared to SIRS (23). In addition, plasma levels of
502
IL-18, a representative inflammasome-mediated pro-inflammatory cytokine, at the time
503
of medical ICU admission were higher with increasing disease severity categories
504
(control, SIRS, sepsis and sepsis/ARDS) and the highest in sepsis/ARDS patients (23).
505
Importantly, patients with elevated IL-18 levels at the time of medical ICU hospitalization
506
had increased mortality (23). In mice, MV enhanced IL-18 levels in the serum and
507
bronchoalveolar lavage fluid (23). Furthermore, IL-18 neutralizing antibody treatment, or
508
genetic deletion of IL-18 or caspase-1, reduced lung injury in response to MV (23).
509
These data suggest critical roles of inflammasome-mediated immune responses in ICU
510
patients.
511
Recent studies suggest mtDNA is a potent inflammasome activator (61) and is
512
released from mitochondria to the cytosol or the extracellular space as a mitochondrial
513
DAMP. In addition, exogenous administration of disrupted mitochondria containing
514
mtDNA induces acute inflammatory tissue injuries (100).
515
whether the levels of circulating mtDNA are associated with disease severity or mortality
22
Therefore, we examined
516
in ICU (62). Circulating cell-free mtDNA level was higher in ICU patients who died within
517
28 days of medical ICU admission as well as in patients with sepsis or ARDS in the
518
ICU. Medical ICU patients with an elevated mtDNA level had an increase in their odds
519
of dying within 28 days of ICU admission, while no evidence for association was noted
520
in non-medical ICU patients (e.g. surgical ICU patients). There was no evidence that the
521
association between mtDNA level and medical ICU mortality was attenuated in models
522
adjusted for clinical covariates, including Acute Physiology and Chronic Health
523
Evaluation (APACHE) II score, sepsis, or ARDS. The inclusion of an elevated mtDNA to
524
a clinical model (including age, gender, race/ethnicity, APACHE II score, and sepsis)
525
improved the net-reclassification of 28-day mortality among medical ICU patients. The
526
improvement in net reclassification remained significant even after additionally adjusting
527
for measures of procalcitonin or lactate, commonly used biomarkers in ICU. Similar
528
relationships between circulating mtDNA and outcome have been noted in patients with
529
severe trauma (82) or patients with sepsis in the emergency room (51).
530
circulating mtDNA could serve as a viable plasma biomarker in medical ICU patients
531
and, may represent an important pathogenic determinant that contributes to a
532
exaggerated systemic inflammatory response observed in patients with sepsis or
533
ARDS.
Thus,
534
535
Mitochondrial biogenesis to the rescue
536
Decades of laboratory and clinical studies have revealed that acute systemic
537
inflammatory syndromes, including sepsis, generate substantial macromolecular
23
538
damage to mitochondria, particularly in the cells of highly metabolically active tissues
539
like the liver, heart, and kidneys (83, 85). Such mitochondrial damage may be
540
indiscernible in vivo because it does not necessarily activate cell death pathways, and
541
some of the same factors that damage mitochondria – e.g., ROS - can also activate the
542
process of mitochondrial biogenesis (86). Recently mitochondrial biogenesis has even
543
been detected even in distal lung cells, including alveolar type II epithelial cells(6).
544
Indeed, all cells that rely on aerobic respiration and oxidative phosphorylation for energy
545
conservation express a robust genetic program of mitochondrial quality control that is
546
designed to maintain a fully functional complement of mitochondria through the
547
conjoined
548
(mitophagy) (89). These closely regulated processes serve a pro-survival capacity by
549
allowing cells to quickly replace metabolically dysfunctional mitochondria with fresh
550
organelles from a pool of undamaged organelles before energy failure (FIGURE 4).
activities
of
mitochondrial
biogenesis
and
mitochondrial
autophagy
551
During inflammation mitochondrial damage is caused by the over-production of
552
reactive oxygen and nitrogen species (ROS/RNS) at multiple sites, leading to the
553
inactivation of critical mitochondrial macromolecules through biochemical oxidation,
554
nitration, and/or S-nitrosation. The critical mitochondrial targets are diverse, including
555
many proteins, lipids, and mtDNA that embody integral mitochondrial processes
556
including protein importation, transcription, protein synthesis, TCA cycle function,
557
electron transport, heme synthesis, and control of the mitochondrial permeability
558
transition pore (69). The upshot of damage from elevated ROS production is the
559
compromised efficiency of oxidative phosphorylation, which may destabilize energy
560
homeostasis, and lead to further inflammation that may perpetuate the mitochondrial
24
561
damage (101). However, the production of mitochondrial ROS also provides signals to
562
activate redox-sensitive transcriptional pathways that regulate mitochondrial quality
563
control and trigger the synthesis of counter-inflammatory mediators, such as IL-10 (70).
564
Moreover, the enhanced induction of enzymatic NO and CO production also promotes
565
the expression of a large group of genes that is essential for mitochondrial biogenesis
566
(63, 84).
567
A number of the transcriptional pathways activated by the NO synthase 2 (NOS2)
568
and heme oxygenase-1 (HO-1) enzymes for the induction of mitochondrial biogenesis
569
during inflammation have been worked out over the last several years. For both
570
enzymes, the endogenous gaseous product (NO or CO) is directly involved in the
571
induction mechanisms through the activation of redox-sensitive pathways. For NOS2,
572
as for the other NOS isoforms, a key step involves the cyclic GMP-dependent
573
phosphorylation of cyclic AMP-responsive element-binding protein 1 (CREB1), which
574
undergoes translocation to the nucleus to activate the gene for PGC-1a, a critical co-
575
activator of mitochondrial biogenesis and oxidative metabolism. Activated PGC-1a
576
protein is a partner for nuclear respiratory factor-1 (NRF-1) and NRF-2 (GABPA); these
577
are both essential transcription factors for mitochondrial biogenesis in most cells. For
578
HO-1, heme catabolism releases CO, which binds to the reduced terminal oxidase of
579
the respiratory chain and increases the mitochondrial H2O2 leak rate. The increased
580
H2O2 production activates several pro-survival kinases, such as Akt, as well as the
581
transcription factor Nfe2l2 (Nrf2)(68). Nrf2, in its gene transactivation function, binds to
582
anti-oxidant response elements (ARE) in the NRF-1 gene promoter, while Akt
583
phosphorylates GSK-3b, which relieves a restriction on the entry of Nrf2 into the
25
584
nucleus.
Akt phosphorylates the NRF-1 protein as well, allowing both transcription
585
factors to gain entrance to the nucleus to induce an assemblage of genes required for
586
mitochondrial biogenesis.
587
At counterpoise to mitochondrial biogenesis is the removal of senescent or
588
damaged mitochondria that no longer perform oxidative phosphorylation or are
589
triggering oxidative damage to nearby cellular structures. Such damaged mitochondria
590
also propagate certain danger signals, for example, inflammasome activation (101). The
591
deconstruction of obsolescent mitochondria by mitophagy, a form of selective
592
macroautophagy, entails the envelopment of the mitochondrial cargo within double-
593
membrane vesicles or autophagosomes, which fuse with lysosomes to degrade the
594
cargo. The rate of mitophagy is governed by the extent of mitochondrial damage, but
595
also by the cell’s energy requirement and its capacity to meet this mitochondrial
596
disposal function (99). Thus, from the cellular perspective, optimal stress-stabilizing
597
strategies match energy requirements with the rates of mitophagy and mitochondrial
598
biogenesis in order to ensure energy sufficiency and mitochondrial quality control.
599
In the mammalian lung, mitochondrial energy provision is an important function of
600
many of the roughly 40 cell types present there. In the parenchyma for instance,
601
alveolar type II cells are enriched in mitochondria, which provide ATP to support
602
surfactant synthesis, secretion, and recycling. These cells also initiate mitochondrial
603
biogenesis during acute lung injury, pneumonia, hyperoxia, and other stresses through
604
induction of the HO-1/CO and NOS2 systems. These alveolar type II cell mitochondria
605
also proliferate to support not only surfactant metabolism, but multiple other energy-
606
dependent functions in the transition to type I epithelium, such as those involved in
26
607
maintaining alveolar barrier integrity. In fact, mitochondrial proliferation precedes the
608
proliferation of type II cells as well as their differentiation into type I epithelium. Under
609
many of these same conditions, as discussed elsewhere in this Perspective, autophagy
610
programs, including mitophagy, are activated in order to eliminate damaged proteins
611
and other damaged organelles from the cell.
612
In the future, our understanding of the basic intracellular mechanisms of
613
mitochondrial quality control in the lung will be brought into line with newly emerging
614
roles for mitochondrial depletion, immune cell scavenging of exported mitochondrial
615
components, and adoptive mitochondrial rescue-by-transfer during acute and chronic
616
lung injury. The importance of these intricate extra- and intercellular processes will
617
hinge on the relative effectiveness of mitochondrial biogenesis and mitophagy under
618
pathogenic conditions of varying type, intensity, and severity that are currently being
619
investigated in different experimental settings.
620
621
Mitochondrial transfer to the rescue
622
The foregoing discussion raises the prospect that mitochondrial oxygen sensing,
623
maintenance of mtDNA integrity to prevent mtDNA fragmentation and release in the
624
form of mtDNA DAMPs, and mitochondrial biogenesis may be important determinants of
625
outcome in sepsis.
626
severe illness is to prevent mitochondrial dysfunction (11).
627
decreased ATP production could causes failure of various aspects of cell function that in
628
combination could lead to multi-organ failure in ALI/ARDS. The cause of mitochondrial
A related and desirable goal of mitochondrial-directed therapy in
27
Clearly, the attendant
629
failure is not well understood, but one possibility is that NFκB activation, occurring
630
secondary to TLR4 ligation by bacterial endotoxin, causes transcriptional up regulation
631
of inducible nitric oxide synthase. The increased nitric oxide thus produced reacts with
632
mitochondrial superoxide to cause peroxynitration, hence dysfunction of mitochondrial
633
electron transport proteins (69). Although other mechanisms may also apply, the end
634
result is failure of cellular bioenergetics as well-documented by the reduction of tissue
635
ATP levels. The therapeutic objective therefore, would be to replenish dysfunctional
636
mitochondria of host cells with fresh mitochondria.
637
The question is how does one replenish mitochondria? One possibility is to give
638
exogenous mitochondria with the expectation that cells will take up the mitochondria by
639
endocytosis and thereby repair the ATP deficit. However the caveat here is that free
640
mitochondria in the extracellular space might act as DAMPs (damage-associated
641
molecular patterns) that activate pattern recognition receptors that exacerbate tissue
642
inflammation and injury (100).
643
The other option is to enable intercellular mitochondrial transfer, a process that
644
has been reported in cultured cells (76). Islam et al. now show that the process also
645
occurs in vivo and that mitochondrial transfer from exogenously administered BMSCs
646
(bone-marrow-derived stromal cells) to the alveolar epithelium protects against
647
lipopolysaccharide (LPS)-induced ALI (40). A more recent report by Ahmad et al. also
648
affirms that mitochondrial transfer from BMSCs to the airway epithelium occurs in vivo
649
under inflammatory conditions (2).
650
mitochondrial transfer abrogated airway inflammation and protected against bronchial
651
injury caused by mitochondrial dysfunction. These findings attest to the feasibility of
In this paper, the authors show that the
28
652
mitochondrial transfer between specific cell types in vivo, indicating that it might be
653
applicable as a therapeutic tool in that mitochondrial transfer might correct the deficit of
654
mitochondrial function in affected cells through donation of fresh mitochondria from
655
BMSCs.
656
Islam et al. noted two important features regarding mitochondrial transfer. First,
657
the transfer did not occur when BMSCs were instilled in control lungs not exposed to
658
LPS, indicating that pathologic stress to the alveolar epithelium was essential. Second,
659
stabilization of BMSCs on the alveolar epithelium was also essential. The stabilization
660
occurred through formation of gap junctional channels (GJCs) between the BMSCs and
661
the alveolar epithelium. Knockdown of connexin 43, a GJC protein, blocked the BMSC
662
stabilization and also diminished the survival benefit accruing from BMSC instillation.
663
These findings indicate that for mitochondrial transfer to occur from a donor to a
664
recipient cell, a pre-conditioning of the interacting cell surfaces is critical.
665
In the study by Islam et al. the mechanism of mitochondrial transfer in alveoli
666
occurred through a sequence in which BMSCs released mitochondria-containing
667
nanotubes and microvesicles, and then the alveolar epithelium engulfed the
668
microvesicles, internalizing the mitochondria (FIGURE 5).
669
Miro1, a mitochondrial Rho-GTPase, is critical for this process since BMSCs lacking
670
Miro1 fail to transfer mitochondria to the airway epithelium. The mechanisms that
671
regulate microvesicle engulfment and release of the internalized mitochondria in the
672
alveolar cytosol are not clear.
673
internalized mitochondria were functional, since progressive increases of ATP levels
674
were detectable in the mitochondria-recipient epithelium. Islam et al showed that
Ahmad et al. show that
However, in Islam and coworkers found that the
29
675
endotoxin-induced mouse mortality decreased markedly following instillation of BMSCs
676
carrying normal mitochondria, but not when the BMSC mitochondria were made
677
dysfunctional by knockdown of the electron-transporting Riske iron-sulfur protein that
678
determines mitochondrial ATP production. Although mitochondrial transfer has been
679
shown in vitro and in feral dogs (62), the reports by Islam et al. and Ahmad et al indicate
680
that the process occurs in vivo to promote tissue repair in organ systems undergoing
681
pathologic stress. The applicability of mitochondrial transfer as a viable therapeutic
682
modality in inflammatory lung disease requires further consideration.
683
684
685
Acknowledgements
686
The authors’ research described in this Perspective was supported by grants from the National
687
Institutes of Health (R01 HL35440 and R01 HL122062 to P.T.S., R01 HL058234 and R0 1
688
HL113614 to M.N.G., R01 HL055330 and P01 HL114501 to A.M.K.C., R01 AI62740 and R01
689
AI083912 to E.D.C. P01 HL108801 and R01 AI095424 C.A.P., and RO1 HL36024
and RO1
690
HL57556 to J.B.)
691
30
692
References
693
694
695
696
697
698
699
700
701
702
703
704
705
706
707
708
709
710
711
712
713
714
715
716
717
718
719
720
721
722
723
724
725
726
727
728
729
730
731
732
733
734
735
736
1.
Aggarwal NR, D'Alessio FR, Tsushima K, Sidhaye VK, Cheadle C, Grigoryev
DN, Barnes KC, and King LS. Regulatory T cell-mediated resolution of lung injury:
identification of potential target genes via expression profiling. Physiological genomics
41: 109-119, 2010.
2.
Ahmad T, Mukherjee S, Pattnaik B, Kumar M, Singh S, Kumar M, Rehman R,
Tiwari BK, Jha KA, Barhanpurkar AP, Wani MR, Roy SS, Mabalirajan U, Ghosh B,
and Agrawal A. Miro1 regulates intercellular mitochondrial transport & enhances
mesenchymal stem cell rescue efficacy. The EMBO journal 2014.
3.
Archer SL, Reeve HL, Michelakis E, Puttagunta L, Waite R, Nelson DP,
Dinauer MC, and Weir EK. O2 sensing is preserved in mice lacking the gp91 phox
subunit of NADPH oxidase. Proceedings of the National Academy of Sciences of the
United States of America 96: 7944-7949, 1999.
4.
Archer SL, Souil E, Dinh-Xuan AT, Schremmer B, Mercier JC, El Yaagoubi
A, Nguyen-Huu L, Reeve HL, and Hampl V. Molecular identification of the role of
voltage-gated K+ channels, Kv1.5 and Kv2.1, in hypoxic pulmonary vasoconstriction
and control of resting membrane potential in rat pulmonary artery myocytes. The
Journal of clinical investigation 101: 2319-2330, 1998.
5.
Arimori Y, Nakamura R, Yamada H, Shibata K, Maeda N, Kase T, and
Yoshikai Y. Type I interferon limits influenza virus-induced acute lung injury by
regulation of excessive inflammation in mice. Antiviral research 99: 230-237, 2013.
6.
Athale J, Ulrich A, Chou Macgarvey N, Bartz RR, Welty-Wolf KE, Suliman
HB, and Piantadosi CA. Nrf2 promotes alveolar mitochondrial biogenesis and
resolution of lung injury in Staphylococcus aureus pneumonia in mice. Free radical
biology & medicine 53: 1584-1594, 2012.
7.
Ballinger SW, Patterson C, Yan CN, Doan R, Burow DL, Young CG, Yakes
FM, Van Houten B, Ballinger CA, Freeman BA, and Runge MS. Hydrogen peroxideand peroxynitrite-induced mitochondrial DNA damage and dysfunction in vascular
endothelial and smooth muscle cells. Circulation research 86: 960-966, 2000.
8.
Bogenhagen DF. Repair of mtDNA in vertebrates. American journal of human
genetics 64: 1276-1281, 1999.
9.
Bogenhagen DF, Rousseau D, and Burke S. The layered structure of human
mitochondrial DNA nucleoids. The Journal of biological chemistry 283: 3665-3675,
2008.
10.
Bonawitz ND, Clayton DA, and Shadel GS. Initiation and beyond: multiple
functions of the human mitochondrial transcription machinery. Molecular cell 24: 813825, 2006.
11.
Brealey D, Brand M, Hargreaves I, Heales S, Land J, Smolenski R, Davies
NA, Cooper CE, and Singer M. Association between mitochondrial dysfunction and
severity and outcome of septic shock. Lancet 360: 219-223, 2002.
12.
Chandel N, Budinger GR, Kemp RA, and Schumacker PT. Inhibition of
cytochrome-c oxidase activity during prolonged hypoxia. The American journal of
physiology 268: L918-925, 1995.
31
737
738
739
740
741
742
743
744
745
746
747
748
749
750
751
752
753
754
755
756
757
758
759
760
761
762
763
764
765
766
767
768
769
770
771
772
773
774
775
776
777
778
779
780
13.
Chandel NS, Maltepe E, Goldwasser E, Mathieu CE, Simon MC, and
Schumacker PT. Mitochondrial reactive oxygen species trigger hypoxia-induced
transcription. Proceedings of the National Academy of Sciences of the United States of
America 95: 11715-11720, 1998.
14.
Chatterjee A, Mambo E, Zhang Y, Deweese T, and Sidransky D. Targeting of
mutant hogg1 in mammalian mitochondria and nucleus: effect on cellular survival upon
oxidative stress. BMC cancer 6: 235, 2006.
15.
Chouteau J, Gorodnya O, Ruchko M, Obiako B, Wilson G, and Gillespie M.
Novel Fusion Protein Constructs Targeting DNA Repair Enzymes To Mitochondria
Protect Against Pseudomonas Aeruginosa-Induced Acute Lung Injury In Intact Rats. Am
J Resp Crit Care Med 279: A3763, 2011.
16.
Chouteau JM, Obiako B, Gorodnya OM, Pastukh VM, Ruchko MV, Wright
AJ, Wilson GL, and Gillespie MN. Mitochondrial DNA integrity may be a determinant
of endothelial barrier properties in oxidant-challenged rat lungs. American journal of
physiology Lung cellular and molecular physiology 301: L892-898, 2011.
17.
Davidson BA, Vethanayagam RR, Grimm MJ, Mullan BA, Raghavendran K,
Blackwell TS, Freeman ML, Ayyasamy V, Singh KK, Sporn MB, Itagaki K, Hauser
CJ, Knight PR, and Segal BH. NADPH oxidase and Nrf2 regulate gastric aspirationinduced inflammation and acute lung injury. Journal of immunology 190: 1714-1724,
2013.
18.
Davis BK, Wen H, and Ting JP. The inflammasome NLRs in immunity,
inflammation, and associated diseases. Annual review of immunology 29: 707-735,
2011.
19.
de Souza-Pinto NC, Eide L, Hogue BA, Thybo T, Stevnsner T, Seeberg E,
Klungland A, and Bohr VA. Repair of 8-oxodeoxyguanosine lesions in mitochondrial
dna depends on the oxoguanine dna glycosylase (OGG1) gene and 8-oxoguanine
accumulates in the mitochondrial dna of OGG1-defective mice. Cancer research 61:
5378-5381, 2001.
20.
Ding Z, Liu S, Wang X, Khaidakov M, Dai Y, and Mehta JL. Oxidant stress in
mitochondrial DNA damage, autophagy and inflammation in atherosclerosis. Scientific
reports 3: 1077, 2013.
21.
Dobson AW, Grishko V, LeDoux SP, Kelley MR, Wilson GL, and Gillespie
MN. Enhanced mtDNA repair capacity protects pulmonary artery endothelial cells from
oxidant-mediated death. American journal of physiology Lung cellular and molecular
physiology 283: L205-210, 2002.
22.
Dolinay T, Kaminski N, Felgendreher M, Kim HP, Reynolds P, Watkins SC,
Karp D, Uhlig S, and Choi AM. Gene expression profiling of target genes in ventilatorinduced lung injury. Physiological genomics 26: 68-75, 2006.
23.
Dolinay T, Kim YS, Howrylak J, Hunninghake GM, An CH, Fredenburgh L,
Massaro AF, Rogers A, Gazourian L, Nakahira K, Haspel JA, Landazury R,
Eppanapally S, Christie JD, Meyer NJ, Ware LB, Christiani DC, Ryter SW, Baron
RM, and Choi AM. Inflammasome-regulated cytokines are critical mediators of acute
lung injury. American journal of respiratory and critical care medicine 185: 1225-1234,
2012.
32
781
782
783
784
785
786
787
788
789
790
791
792
793
794
795
796
797
798
799
800
801
802
803
804
805
806
807
808
809
810
811
812
813
814
815
816
817
818
819
820
821
822
823
824
825
826
24.
Dooley CT, Dore TM, Hanson GT, Jackson WC, Remington SJ, and Tsien
RY. Imaging dynamic redox changes in mammalian cells with green fluorescent protein
indicators. The Journal of biological chemistry 279: 22284-22293, 2004.
25.
Driggers WJ, Holmquist GP, LeDoux SP, and Wilson GL. Mapping
frequencies of endogenous oxidative damage and the kinetic response to oxidative
stress in a region of rat mtDNA. Nucleic acids research 25: 4362-4369, 1997.
26.
Duranteau J, Chandel NS, Kulisz A, Shao Z, and Schumacker PT.
Intracellular signaling by reactive oxygen species during hypoxia in cardiomyocytes.
The Journal of biological chemistry 273: 11619-11624, 1998.
27.
Garcia N, and Chavez E. Mitochondrial DNA fragments released through the
permeability transition pore correspond to specific gene size. Life sciences 81: 11601166, 2007.
28.
Gebb SA, Decoux A, Waggoner A, Wilson GL, and Gillespie MN.
Mitochondrial DNA damage mediates hyperoxic dysmorphogenesis in rat fetal lung
explants. Neonatology 103: 91-97, 2013.
29.
German P, Szaniszlo P, Hajas G, Radak Z, Bacsi A, Hazra TK, Hegde ML, Ba
X, and Boldogh I. Activation of cellular signaling by 8-oxoguanine DNA glycosylase-1initiated DNA base excision repair. DNA repair 12: 856-863, 2013.
30.
Gleyzer N, Vercauteren K, and Scarpulla RC. Control of mitochondrial
transcription specificity factors (TFB1M and TFB2M) by nuclear respiratory factors
(NRF-1 and NRF-2) and PGC-1 family coactivators. Molecular and cellular biology 25:
1354-1366, 2005.
31.
Grishko V, Solomon M, Breit JF, Killilea DW, Ledoux SP, Wilson GL, and
Gillespie MN. Hypoxia promotes oxidative base modifications in the pulmonary artery
endothelial cell VEGF gene. FASEB journal : official publication of the Federation of
American Societies for Experimental Biology 15: 1267-1269, 2001.
32.
Grishko V, Solomon M, Wilson GL, LeDoux SP, and Gillespie MN. Oxygen
radical-induced mitochondrial DNA damage and repair in pulmonary vascular
endothelial cell phenotypes. American journal of physiology Lung cellular and molecular
physiology 280: L1300-1308, 2001.
33.
Guzy RD, Hoyos B, Robin E, Chen H, Liu L, Mansfield KD, Simon MC,
Hammerling U, and Schumacker PT. Mitochondrial complex III is required for hypoxiainduced ROS production and cellular oxygen sensing. Cell metabolism 1: 401-408,
2005.
34.
Guzy RD, and Schumacker PT. Oxygen sensing by mitochondria at complex III:
the paradox of increased reactive oxygen species during hypoxia. Experimental
physiology 91: 807-819, 2006.
35.
Hanson GT, Aggeler R, Oglesbee D, Cannon M, Capaldi RA, Tsien RY, and
Remington SJ. Investigating mitochondrial redox potential with redox-sensitive green
fluorescent protein indicators. The Journal of biological chemistry 279: 13044-13053,
2004.
36.
Hashizume M, Mouner M, Chouteau JM, Gorodnya OM, Ruchko MV, Potter
BJ, Wilson GL, Gillespie MN, and Parker JC. Mitochondrial-targeted DNA repair
enzyme 8-oxoguanine DNA glycosylase 1 protects against ventilator-induced lung injury
in intact mice. American journal of physiology Lung cellular and molecular physiology
304: L287-297, 2013.
33
827
828
829
830
831
832
833
834
835
836
837
838
839
840
841
842
843
844
845
846
847
848
849
850
851
852
853
854
855
856
857
858
859
860
861
862
863
864
865
866
867
868
869
870
871
37.
Hollensworth SB, Shen C, Sim JE, Spitz DR, Wilson GL, and LeDoux SP.
Glial cell type-specific responses to menadione-induced oxidative stress. Free radical
biology & medicine 28: 1161-1174, 2000.
38.
Hon WC, Wilson MI, Harlos K, Claridge TD, Schofield CJ, Pugh CW, Maxwell
PH, Ratcliffe PJ, Stuart DI, and Jones EY. Structural basis for the recognition of
hydroxyproline in HIF-1 alpha by pVHL. Nature 417: 975-978, 2002.
39.
Ide T, Tsutsui H, Hayashidani S, Kang D, Suematsu N, Nakamura K, Utsumi
H, Hamasaki N, and Takeshita A. Mitochondrial DNA damage and dysfunction
associated with oxidative stress in failing hearts after myocardial infarction. Circulation
research 88: 529-535, 2001.
40.
Islam MN, Das SR, Emin MT, Wei M, Sun L, Westphalen K, Rowlands DJ,
Quadri SK, Bhattacharya S, and Bhattacharya J. Mitochondrial transfer from bonemarrow-derived stromal cells to pulmonary alveoli protects against acute lung injury.
Nature medicine 18: 759-765, 2012.
41.
Ivan M, Kondo K, Yang H, Kim W, Valiando J, Ohh M, Salic A, Asara JM,
Lane WS, and Kaelin WG, Jr. HIFalpha targeted for VHL-mediated destruction by
proline hydroxylation: implications for O2 sensing. Science 292: 464-468, 2001.
42.
Julian MW, Shao G, Bao S, Knoell DL, Papenfuss TL, VanGundy ZC, and
Crouser ED. Mitochondrial transcription factor A serves as a danger signal by
augmenting plasmacytoid dendritic cell responses to DNA. Journal of immunology 189:
433-443, 2012.
43.
Julian MW, Shao G, Vangundy ZC, Papenfuss TL, and Crouser ED.
Mitochondrial transcription factor A, an endogenous danger signal, promotes TNFalpha
release via RAGE- and TLR9-responsive plasmacytoid dendritic cells. PloS one 8:
e72354, 2013.
44.
Kang D, and Hamasaki N. Mitochondrial transcription factor A in the
maintenance of mitochondrial DNA: overview of its multiple roles. Annals of the New
York Academy of Sciences 1042: 101-108, 2005.
45.
Knoops B, Goemaere J, Van der Eecken V, and Declercq JP. Peroxiredoxin
5: structure, mechanism, and function of the mammalian atypical 2-Cys peroxiredoxin.
Antioxidants & redox signaling 15: 817-829, 2011.
46.
Knuefermann P, Baumgarten G, Koch A, Schwederski M, Velten M,
Ehrentraut H, Mersmann J, Meyer R, Hoeft A, Zacharowski K, and Grohe C. CpG
oligonucleotide activates Toll-like receptor 9 and causes lung inflammation in vivo.
Respiratory research 8: 72, 2007.
47.
Koczor CA, Shokolenko IN, Boyd AK, Balk SP, Wilson GL, and Ledoux SP.
Mitochondrial DNA damage initiates a cell cycle arrest by a Chk2-associated
mechanism in mammalian cells. The Journal of biological chemistry 284: 36191-36201,
2009.
48.
Koczor CA, Snyder JW, Shokolenko IN, Dobson AW, Wilson GL, and
Ledoux SP. Targeting repair proteins to the mitochondria of mammalian cells through
stable transfection, transient transfection, viral transduction, and TAT-mediated protein
transduction. Methods in molecular biology 554: 233-249, 2009.
34
872
873
874
875
876
877
878
879
880
881
882
883
884
885
886
887
888
889
890
891
892
893
894
895
896
897
898
899
900
901
902
903
904
905
906
907
908
909
910
911
912
913
914
915
916
49.
Kojima K, Arikawa T, Saita N, Goto E, Tsumura S, Tanaka R, Masunaga A,
Niki T, Oomizu S, Hirashima M, and Kohrogi H. Galectin-9 attenuates acute lung
injury by expanding CD14- plasmacytoid dendritic cell-like macrophages. American
journal of respiratory and critical care medicine 184: 328-339, 2011.
50.
Krysko DV, Agostinis P, Krysko O, Garg AD, Bachert C, Lambrecht BN, and
Vandenabeele P. Emerging role of damage-associated molecular patterns derived from
mitochondria in inflammation. Trends in immunology 32: 157-164, 2011.
51.
Kung CT, Hsiao SY, Tsai TC, Su CM, Chang WN, Huang CR, Wang HC, Lin
WC, Chang HW, Lin YJ, Cheng BC, Su BY, Tsai NW, and Lu CH. Plasma nuclear
and mitochondrial DNA levels as predictors of outcome in severe sepsis patients in the
emergency room. Journal of translational medicine 10: 130, 2012.
52.
Liang J, Jung Y, Tighe RM, Xie T, Liu N, Leonard M, Gunn MD, Jiang D, and
Noble PW. A macrophage subpopulation recruited by CC chemokine ligand-2 clears
apoptotic cells in noninfectious lung injury. American journal of physiology Lung cellular
and molecular physiology 302: L933-940, 2012.
53.
Lin KL, Suzuki Y, Nakano H, Ramsburg E, and Gunn MD. CCR2+ monocytederived dendritic cells and exudate macrophages produce influenza-induced pulmonary
immune pathology and mortality. Journal of immunology 180: 2562-2572, 2008.
54.
Mansfield KD, Guzy RD, Pan Y, Young RM, Cash TP, Schumacker PT, and
Simon MC. Mitochondrial dysfunction resulting from loss of cytochrome c impairs
cellular oxygen sensing and hypoxic HIF-alpha activation. Cell metabolism 1: 393-399,
2005.
55.
Marques PE, Amaral SS, Pires DA, Nogueira LL, Soriani FM, Lima BH,
Lopes GA, Russo RC, Avila TV, Melgaco JG, Oliveira AG, Pinto MA, Lima CX, De
Paula AM, Cara DC, Leite MF, Teixeira MM, and Menezes GB. Chemokines and
mitochondrial products activate neutrophils to amplify organ injury during mouse acute
liver failure. Hepatology 56: 1971-1982, 2012.
56.
Michelakis ED, Archer SL, and Weir EK. Acute hypoxic pulmonary
vasoconstriction: a model of oxygen sensing. Physiological research / Academia
Scientiarum Bohemoslovaca 44: 361-367, 1995.
57.
Michelakis ED, Rebeyka I, Wu X, Nsair A, Thebaud B, Hashimoto K, Dyck
JR, Haromy A, Harry G, Barr A, and Archer SL. O2 sensing in the human ductus
arteriosus: regulation of voltage-gated K+ channels in smooth muscle cells by a
mitochondrial redox sensor. Circulation research 91: 478-486, 2002.
58.
Mills E, and Jobsis FF. Mitochondrial respiratory chain of carotid body and
chemoreceptor response to changes in oxygen tension. Journal of neurophysiology 35:
405-428, 1972.
59.
Mills E, and Jobsis FF. Simultaneous measurement of cytochrome a3 reduction
and chemoreceptor afferent activity in the carotid body. Nature 225: 1147-1149, 1970.
60.
Mungai PT, Waypa GB, Jairaman A, Prakriya M, Dokic D, Ball MK, and
Schumacker PT. Hypoxia triggers AMPK activation through reactive oxygen speciesmediated activation of calcium release-activated calcium channels. Molecular and
cellular biology 31: 3531-3545, 2011.
35
917
918
919
920
921
922
923
924
925
926
927
928
929
930
931
932
933
934
935
936
937
938
939
940
941
942
943
944
945
946
947
948
949
950
951
952
953
954
955
956
957
958
959
960
961
61.
Nakahira K, Haspel JA, Rathinam VA, Lee SJ, Dolinay T, Lam HC, Englert
JA, Rabinovitch M, Cernadas M, Kim HP, Fitzgerald KA, Ryter SW, and Choi AM.
Autophagy proteins regulate innate immune responses by inhibiting the release of
mitochondrial DNA mediated by the NALP3 inflammasome. Nature immunology 12:
222-230, 2011.
62.
Nakahira K, Kyung SY, Rogers AJ, Gazourian L, Youn S, Massaro AF,
Quintana C, Osorio JC, Wang Z, Zhao Y, Lawler LA, Christie JD, Meyer NJ, Mc
Causland FR, Waikar SS, Waxman AB, Chung RT, Bueno R, Rosas IO,
Fredenburgh LE, Baron RM, Christiani DC, Hunninghake GM, and Choi AM.
Circulating mitochondrial DNA in patients in the ICU as a marker of mortality: derivation
and validation. PLoS medicine 10: e1001577; discussion e1001577, 2013.
63.
Nisoli E, Clementi E, Paolucci C, Cozzi V, Tonello C, Sciorati C, Bracale R,
Valerio A, Francolini M, Moncada S, and Carruba MO. Mitochondrial biogenesis in
mammals: the role of endogenous nitric oxide. Science 299: 896-899, 2003.
64.
Nuutinen EM, Nishiki K, Erecinska M, and Wilson DF. Role of mitochondrial
oxidative phosphorylation in regulation of coronary blood flow. The American journal of
physiology 243: H159-169, 1982.
65.
Oka T, Hikoso S, Yamaguchi O, Taneike M, Takeda T, Tamai T, Oyabu J,
Murakawa T, Nakayama H, Nishida K, Akira S, Yamamoto A, Komuro I, and Otsu
K. Mitochondrial DNA that escapes from autophagy causes inflammation and heart
failure. Nature 485: 251-255, 2012.
66.
Ozawa T, Natori Y, Sako Y, Kuroiwa H, Kuroiwa T, and Umezawa Y. A
minimal peptide sequence that targets fluorescent and functional proteins into the
mitochondrial intermembrane space. ACS chemical biology 2: 176-186, 2007.
67.
Panduri V, Liu G, Surapureddi S, Kondapalli J, Soberanes S, de SouzaPinto NC, Bohr VA, Budinger GR, Schumacker PT, Weitzman SA, and Kamp DW.
Role of mitochondrial hOGG1 and aconitase in oxidant-induced lung epithelial cell
apoptosis. Free radical biology & medicine 47: 750-759, 2009.
68.
Piantadosi CA, Carraway MS, Babiker A, and Suliman HB. Heme oxygenase1 regulates cardiac mitochondrial biogenesis via Nrf2-mediated transcriptional control of
nuclear respiratory factor-1. Circulation research 103: 1232-1240, 2008.
69.
Piantadosi CA, and Suliman HB. Redox regulation of mitochondrial biogenesis.
Free radical biology & medicine 53: 2043-2053, 2012.
70.
Piantadosi CA, Withers CM, Bartz RR, MacGarvey NC, Fu P, Sweeney TE,
Welty-Wolf KE, and Suliman HB. Heme oxygenase-1 couples activation of
mitochondrial biogenesis to anti-inflammatory cytokine expression. The Journal of
biological chemistry 286: 16374-16385, 2011.
71.
Pierrakos C, and Vincent JL. Sepsis biomarkers: a review. Critical care 14:
R15, 2010.
72.
Rachek LI, Grishko VI, Ledoux SP, and Wilson GL. Role of nitric oxideinduced mtDNA damage in mitochondrial dysfunction and apoptosis. Free radical
biology & medicine 40: 754-762, 2006.
73.
Rachek LI, Grishko VI, Musiyenko SI, Kelley MR, LeDoux SP, and Wilson
GL. Conditional targeting of the DNA repair enzyme hOGG1 into mitochondria. The
Journal of biological chemistry 277: 44932-44937, 2002.
36
962
963
964
965
966
967
968
969
970
971
972
973
974
975
976
977
978
979
980
981
982
983
984
985
986
987
988
989
990
991
992
993
994
995
996
997
998
999
1000
1001
1002
1003
1004
1005
1006
1007
74.
Reyes A, He J, Mao CC, Bailey LJ, Di Re M, Sembongi H, Kazak L, Dzionek
K, Holmes JB, Cluett TJ, Harbour ME, Fearnley IM, Crouch RJ, Conti MA,
Adelstein RS, Walker JE, and Holt IJ. Actin and myosin contribute to mammalian
mitochondrial DNA maintenance. Nucleic acids research 39: 5098-5108, 2011.
75.
Ricci C, Pastukh V, Leonard J, Turrens J, Wilson G, Schaffer D, and
Schaffer SW. Mitochondrial DNA damage triggers mitochondrial-superoxide generation
and apoptosis. American journal of physiology Cell physiology 294: C413-422, 2008.
76.
Rogers RS, and Bhattacharya J. When cells become organelle donors.
Physiology 28: 414-422, 2013.
77.
Ruchko M, Gorodnya O, LeDoux SP, Alexeyev MF, Al-Mehdi AB, and
Gillespie MN. Mitochondrial DNA damage triggers mitochondrial dysfunction and
apoptosis in oxidant-challenged lung endothelial cells. American journal of physiology
Lung cellular and molecular physiology 288: L530-535, 2005.
78.
Ruchko MV, Gorodnya OM, Zuleta A, Pastukh VM, and Gillespie MN. The
DNA glycosylase Ogg1 defends against oxidant-induced mtDNA damage and apoptosis
in pulmonary artery endothelial cells. Free radical biology & medicine 50: 1107-1113,
2011.
79.
Sabharwal SS, Waypa GB, Marks JD, and Schumacker PT. Peroxiredoxin-5
targeted to the mitochondrial intermembrane space attenuates hypoxia-induced reactive
oxygen species signalling. The Biochemical journal 456: 337-346, 2013.
80.
Shadel GS. Mitochondrial DNA, aconitase 'wraps' it up. Trends in biochemical
sciences 30: 294-296, 2005.
81.
Shokolenko I, Venediktova N, Bochkareva A, Wilson GL, and Alexeyev MF.
Oxidative stress induces degradation of mitochondrial DNA. Nucleic acids research 37:
2539-2548, 2009.
82.
Simmons JD, Lee YL, Mulekar S, Kuck JL, Brevard SB, Gonzalez RP,
Gillespie MN, and Richards WO. Elevated levels of plasma mitochondrial DNA
DAMPs are linked to clinical outcome in severely injured human subjects. Annals of
surgery 258: 591-596; discussion 596-598, 2013.
83.
Suliman HB, Carraway MS, and Piantadosi CA. Postlipopolysaccharide
oxidative damage of mitochondrial DNA. American journal of respiratory and critical
care medicine 167: 570-579, 2003.
84.
Suliman HB, Carraway MS, Tatro LG, and Piantadosi CA. A new activating
role for CO in cardiac mitochondrial biogenesis. Journal of cell science 120: 299-308,
2007.
85.
Suliman HB, Welty-Wolf KE, Carraway M, Tatro L, and Piantadosi CA.
Lipopolysaccharide induces oxidative cardiac mitochondrial damage and biogenesis.
Cardiovascular research 64: 279-288, 2004.
86.
Suliman HB, Welty-Wolf KE, Carraway MS, Schwartz DA, Hollingsworth JW,
and Piantadosi CA. Toll-like receptor 4 mediates mitochondrial DNA damage and
biogenic responses after heat-inactivated E. coli. FASEB journal : official publication of
the Federation of American Societies for Experimental Biology 19: 1531-1533, 2005.
87.
Tasaka S, Kamata H, Miyamoto K, Nakano Y, Shinoda H, Kimizuka Y,
Fujiwara H, Hasegawa N, Fujishima S, Miyasho T, and Ishizaka A. Intratracheal
synthetic CpG oligodeoxynucleotide causes acute lung injury with systemic
inflammatory response. Respiratory research 10: 84, 2009.
37
1008
1009
1010
1011
1012
1013
1014
1015
1016
1017
1018
1019
1020
1021
1022
1023
1024
1025
1026
1027
1028
1029
1030
1031
1032
1033
1034
1035
1036
1037
1038
1039
1040
1041
1042
1043
1044
1045
1046
1047
1048
1049
1050
88.
Tsushima K, King LS, Aggarwal NR, De Gorordo A, D'Alessio FR, and Kubo
K. Acute lung injury review. Internal medicine 48: 621-630, 2009.
89.
Wang K, and Klionsky DJ. Mitochondria removal by autophagy. Autophagy 7:
297-300, 2011.
90.
Waypa GB, Chandel NS, and Schumacker PT. Model for hypoxic pulmonary
vasoconstriction involving mitochondrial oxygen sensing. Circulation research 88: 12591266, 2001.
91.
Waypa GB, Guzy R, Mungai PT, Mack MM, Marks JD, Roe MW, and
Schumacker PT. Increases in mitochondrial reactive oxygen species trigger hypoxiainduced calcium responses in pulmonary artery smooth muscle cells. Circulation
research 99: 970-978, 2006.
92.
Waypa GB, Marks JD, Guzy R, Mungai PT, Schriewer J, Dokic D, and
Schumacker PT. Hypoxia triggers subcellular compartmental redox signaling in
vascular smooth muscle cells. Circulation research 106: 526-535, 2010.
93.
Waypa GB, Marks JD, Guzy RD, Mungai PT, Schriewer JM, Dokic D, Ball
MK, and Schumacker PT. Superoxide generated at mitochondrial complex III triggers
acute responses to hypoxia in the pulmonary circulation. American journal of respiratory
and critical care medicine 187: 424-432, 2013.
94.
Waypa GB, and Schumacker PT. Hypoxia-induced changes in pulmonary and
systemic vascular resistance: where is the O2 sensor? Respiratory physiology &
neurobiology 174: 201-211, 2010.
95.
Wilson DF, and Erecinska M. Effect of oxygen concentration on cellular
metabolism. Chest 88: 229S-232S, 1985.
96.
Wirth A, Benyo Z, Lukasova M, Leutgeb B, Wettschureck N, Gorbey S, Orsy
P, Horvath B, Maser-Gluth C, Greiner E, Lemmer B, Schutz G, Gutkind JS, and
Offermanns S. G12-G13-LARG-mediated signaling in vascular smooth muscle is
required for salt-induced hypertension. Nature medicine 14: 64-68, 2008.
97.
Yakes FM, and Van Houten B. Mitochondrial DNA damage is more extensive
and persists longer than nuclear DNA damage in human cells following oxidative stress.
Proceedings of the National Academy of Sciences of the United States of America 94:
514-519, 1997.
98.
Yousefi S, Gold JA, Andina N, Lee JJ, Kelly AM, Kozlowski E, Schmid I,
Straumann A, Reichenbach J, Gleich GJ, and Simon HU. Catapult-like release of
mitochondrial DNA by eosinophils contributes to antibacterial defense. Nature medicine
14: 949-953, 2008.
99.
Zhang J, and Ney PA. Role of BNIP3 and NIX in cell death, autophagy, and
mitophagy. Cell death and differentiation 16: 939-946, 2009.
100. Zhang Q, Raoof M, Chen Y, Sumi Y, Sursal T, Junger W, Brohi K, Itagaki K,
and Hauser CJ. Circulating mitochondrial DAMPs cause inflammatory responses to
injury. Nature 464: 104-107, 2010.
101. Zhou R, Yazdi AS, Menu P, and Tschopp J. A role for mitochondria in NLRP3
inflammasome activation. Nature 469: 221-225, 2011.
1051
1052
38
1053
Figure Legends
1054
FIGURE 1:
1055
regulated generation of reactive oxygen species (ROS) that are released to the
1056
cytosol through the intermembrane space. IMS-PRDX5: Peroxiredoxin-5 targeted to
1057
the intermembrane space. PHD: HIF Prolyl Hydroxylase. III: Mitochondrial complex III.
1058
FIGURE 2: Fate and biological actions of the oxidatively damaged mitochondrial
1059
genome that warrant characterization of mtDNA as a “sentinel molecule”. See
1060
text for details. Abbreviations: DAMPs, Damage Associated Molecular Patterns; IMS,
1061
intermembrane space; PTP, permeability transition pore
1062
FIGURE 3: Plasmacytoid dendritic cells (pDCs) play a central role in orchestrating
1063
both pro- and anti-inflammatory responses.
1064
mitochondrial DNA
1065
contaminating bacteria is sensed by pDCs to promote simultaneous release of TNFα
1066
and IFNα, which promotes the simultaneous activation of proinflammatory and
1067
regulatory immune cells. Thus, pDCs are poised to orchestrate a localized, and self-
1068
limited inflammtory response. Presumably, excessive pDCs activation (e.g., massive
1069
cell injury) could promote systemic pro- and anti-inflammatory immune responses.
1070
Panel B: In splenocyte cultures representing complex immune cell interactions typically
1071
occurring in vivo release of IFNα and TNFα in response to immunogenic (CpGA) DNA is
1072
enhanced by TFAM, and depletion of pDCs [by magnetic MicroBead selection (Miltenyi
1073
Biotec, Auburn, CA)], representing <3% of the total cell populationfrom the splenocyte
1074
cultures, is shown to greatly suppress the immune response. Thus, pDCs are important
1075
“amplifiers” of the immune response to TFAM and DNA. (*p < 0.01, compared to
Conceptual model of oxygen sensing by mitochondria through the
(mtDNA) and TFAM
39
Panel A: During acute tissue injury
released from
damaged cells and
1076
corresponding CpGA DNA treatment alone, †p < 0.01; relative to matching treatment in
1077
the High pDC group, and ‡p < 0.01, compared to corresponding CpGA DNA treatment
1078
alone and the matching treatment in the High pDC group.) PMN = neutrophil; MФ =
1079
macrophage; T-eff = effector CD4+ T cell; T-reg = regulatory T cell
1080
FIGURE 4:
1081
maintenance of mitochondrial homeostasis. The diagram illustrates the simplified
1082
relationship between mitochondrial biogenesis and selective mitochondrial autophagy or
1083
mitophagy. The cycle is accelerated by oxidative stress and is monitored and regulated
1084
by redox sensors including the physiological gases.
1085
FIGURE 5: Sequence of events leading to mitochondrial transfer from alveolus-
1086
attached BMSCs to the alveolar epithelium. The sketch of the distal airway (LEFT)
1087
shows the arrival of airway instilled BMSCs (MSC) in alveoli composed of alveolar type
1088
1 and type 2 cells (AT2 and AT2, respectively) and alveolar macrophages (AM).
1089
Subsequent processes in the alveolus (RIGHT) are: (1) BMSCs adhere to the alveolar
1090
epithelium through Cx43 interactions; (2) Gap junctions form between BMSCs and the
1091
epithelium; (3) Calcium levels increase in the BMSCs; and (4) BMSCs release
1092
mitochondria-containing microvesicles which enter the epithelium and release their
1093
mitochondria in the cytosol.
The integrated process of mitochondrial quality control in the
1094
1095
1096
1097
40
1098
1099
41
FIGURE 1
FIGURE 2
ROS
D-loop
Bioenergetic
defect, cell death
and dysfunction
mtDNA
Damageinduced
mtDNA
Fragmentation
into DAMPs
Mitochondrial
Matrix
IMS
PTP
Cytoplasm
Cellautogenous
TLR9 activation
NALP3
Inflammasome
Activation
Endosome
Plasma Membrane
?
Extracellular Space
Local Inflammation,
TLR9-dependent Immune System Activation,
and Injury Propagation
FIGURE 3
A.
TFAM
RAGE
mtDNA
IFNα
T
reg
T-reg
TLR9
Cell Damage
+/Contamination
Tissue Repair
Antimicrobial
Defense
pDC
TNFα
MФ
T-eff
Local
Inflammation
Negative Feedback
*
6000
5000
No Treatment
CpGA DNA - IFNα
TFAM + CpGA - IFNα
CpGA DNA - TNFα
TFAM + CpGA - TNFα
*
4000
‡
3000
†
2000
†
1000
‡
pD
w
Lo
pD
Cs
Cs
0
Hi
gh
(p g /m l)
B.
S u p e rn a ta n t C yto k in e C o n c e n tra tio n
PMN
FIGURE 4
FIGURE 5
(1)
(2)
AM
MSC
(3)
Cx43
AT2
AT1
(4)