You are on page 1of 9

Inflammation Research (2021) 70:379–387

https://doi.org/10.1007/s00011-021-01447-0 Inflammation Research


REVIEW

Sepsis‑induced myocardial dysfunction: the role of mitochondrial


dysfunction
Hang Yang1 · Zhaocai Zhang2

Received: 18 August 2020 / Revised: 18 February 2021 / Accepted: 22 February 2021 / Published online: 8 March 2021
© The Author(s), under exclusive licence to Springer Nature Switzerland AG part of Springer Nature 2021

Abstract
Introduction Sepsis-induced myocardial dysfunction (SIMD) is a condition manifested by an intrinsic myocardial systolic
and diastolic dysfunction during sepsis, which is associated with worse clinical outcomes and a higher mortality.
Materials and methods Several pathophysiological mechanisms including mitochondrial dysfunction, abnormal body
immune reaction, metabolic reprogramming, excessive production of reactive oxygen species (ROS), and disorder of calcium
regulation have been involved in SIMD. Mitophagy has potential role in protecting myocardial cells in sepsis, especially in
survivors.
Conclusion In the current review, we focus on the role of mitochondrial dysfunction and other mitochondria-related mecha-
nisms including immunologic imbalance, energetic reprogramming, mitophagy, and pyroptosis in the mechanisms of SIMD.

Keywords Sepsis-induced myocardial dysfunction · Mitochondrial dysfunction · Lactate · Immunosuppression

Introduction [6], high-mobility group box 1 (HMBG1) [7], and activated


complement components [8]. In addition, the role of pro-
SIMD is defined as a reversible myocardial dysfunction inflammatory cytokines, decreased coronary perfusion, and
characterized by typical ventricular dilation, decreased altered microcirculation in the presence of severe hypoten-
ventricular contractility, and/or reduced response to vol- sion during sepsis can also lead to myocardial hypoxia and
ume resuscitation. SIMD is a major complication of sepsis dysfunction [9, 10]. In this article, the potential role of mito-
and has a negative impact on patients’ survivals [1, 2]. Evi- chondria in the mechanism of SIMD will be reviewed.
dence suggests several mechanisms and signal pathways.
Mitochondrial dysfunction has become a serious concern,
including oxidative phosphorylation, production of ROS, Mitochondrial dysfunction
reprogramming metabolism of energy [3], and mitophagy
[4]. And body’s immune reaction changes may also play a Changed ultrastructure and structural protein
potential role in SIMD. Previous research has also shown of mitochondria
other factors are associated with SIMD including altered
epigenetic modification [5], myocardial depressant factors, Mitochondrial permeability transition pore (mPTP) is pro-
pathogen-associated molecular patterns (PAMPs), host- posed as a high-conductance channel of the almost imper-
produced danger-associated molecular patterns (DAMPs) meable inner mitochondrial membrane (IMM) [11]. Physi-
ologically, it serves as a rapid mitochondrial efflux channel
for ROS and ­Ca2+ via a transient opening [12]. Its abnormal
* Zhaocai Zhang opening can increase the IMM permeability and change the
2313003@zju.edu.cn
membrane potential of the mitochondria, leading to a reduc-
1
Department of Gastroenterology, West China Hospital, tion of the electrochemical transmembrane gradient, uncou-
Sichuan University, No. 37, Guo Xue Xiang, Wu Hou pling of oxidative phosphorylation, increasing production of
District, Chengdu 610041, China ROS, and decreasing production of ATP [13]. Furthermore,
2
Department of Critical Care Medicine, Second Affiliated the c-ring of the F1F0-ATP synthase has been demonstrated
Hospital, School of Medicine, Zhejiang University, Jiefang as a component of mPTP and might directly influence ATP
Road No. 88, Hangzhou 310000, Zhejiang province, China

13
Vol.:(0123456789)
380 H. Yang, Z. Zhang

synthase [14]. In addition, mPTP is tightly associated with opening of mPTP is properly functioning, when myocardial
cell apoptosis [15]. When outer mitochondrial membrane cells suffer from deteriorating sepsis (Fig. 1).
(OMM) ruptures, N-formyl peptides, ATP, and cytochrome
C are released, and cellular ATP is depleted after the irre- Regulation and dysregulation of mitochondrial
versible opening of mPTP [16, 17], which can be initiated by calcium in sepsis
calcium retention, a central stimulator. Besides the calcium
concentration, pH, ROS, nitric oxide (NO) production in The dysregulation of mitochondrial calcium was observed
excess, peroxynitrite formed from NO can affect the func- in the case of sepsis. In myocardial cells, the homeostasis
tion of mPTP [18]. An mPTP opening was observed in the of calcium is both cytoplasmic and mitochondrial. Mito-
swelling mitochondria [16]. Multiple mPTP inhibitors have chondrial ­Ca2+ channels in IMM include mitochondrial
demonstrated efficacy in preclinical disease models [19]. ­Ca2+ uniporter (MCU), mitochondrial ryanodine recep-
However, although mPTP is involved in cell damage and tor (mRyR), mPTP, mitochondrial Na+/Ca2+ exchanger
death, SIMD lacks cell necrosis or apoptosis probably asso- (mNCX) [20], and H ­ +/Ca 2+ exchanger (mHCX) [13].
ciated with the severity of sepsis. This also indicates that the MCU is dependent on the IMM potential, associated with

Fig. 1  Summary of the possible mechanisms involved in SIMD, mitochondrial calcium uniporter; MPC: mitochondrial pyruvate car-
including dysregulation of calcium, dysfunction of mitochondria, rier; mPTP: mitochondrial permeability transition pore; mTOR:
reprogramming of the energy metabolism, overproduction of ROS, mechanistic target of rapamycin; N ­ AD+: nicotinamide adenine dinu-
opening of mPTP, and mitophagy. During sepsis, energy metabo- cleotide; PAMPs/DAMPs: pathogen/damage-associated molecular
lism is switched to aerobic glycolysis, the same as the pathway of patterns; PARP: poly (ADP-ribose) polymerase; PDC: pyruvate dehy-
hypoxia that has been suggested as a strong inductor of mitophagy. drogenase complex; PDKs: pyruvate dehydrogenase kinases; PI3K:
AMPK: AMP-activated protein kinase; cAMP: 3′-5′-cyclic adeno- phosphatidylinositol 3-kinase; RIP3: receptor‑interacting serine/thre-
sine monophosphate; CAT: catalase; C1: carrier 1; C2: carrier 2; onine kinase 3; RyR: ryanodine receptors; SERCA: sarcoendoplasmic
Cyt C: cytochrome c; ECC: excitation–contraction coupling; HIF- reticulum Ca2+ ATPase; SOD: superoxide dismutase; TAC: tricarbo-
1a: hypoxia-inducible factor-1a; GPCR: G protein-coupled receptor; xylic acid cycle; TLR: Toll-like receptor; TNF: tumor necrosis factor;
GPx: glutathione peroxidase; LTCC: L-type calcium channels; MCU: VDAC: voltage-dependent anion channel

13
Sepsis‑induced myocardial dysfunction: the role of mitochondrial dysfunction 381

the matrix calcium and extramitochondrial ­Ca2+ concen- mtDNA [30], and opening of mPTP [31]. It can also cause
tration. It contributes to oxidative phosphorylation and decreasing oxygen consumption and ATP production [32],
activates the dehydrogenase of the TCA cycle to increase decreasing mitochondrial membrane potential and activity,
the production of NADH for electron transport chain and decreasing expression of respiratory complexes [33].
(ETC) and ATP generation [21]. Voltage-dependent anion Evidence suggested the suppression of mtROS protected car-
channel (VDAC), a part of mPTP, provides a pathway for diac mitochondria, attenuated inflammation, and improved
­Ca2+ and metabolite transportation across OMM [22]. In heart function in the animal models of sepsis. The electron
addition, approximately 20% of the mitochondrial surface leakage depends on the ATP demand [34]. Some models
has been found to be close to the endoplasmic reticulum showed that mitochondria-targeted ROS scavengers such
in mammalian cells generating high C ­ a2+ microdomains, as mitochondrial Q was cardioprotective [35]. The anaes-
termed as mitochondria-associated membranes (MAMs) thetic agent propofol manifested its cardioprotective effect
[23]. When the calcium-induced calcium release (CICR) by scavenging free radical [36]. Clinical therapeutic effects
is activated by autonomic nervous system, a considerable of some typical antioxidants such as vitamin C, vitamin E,
amount of calcium is released from ER via RyR, and cal- and SOD were conflicting. Vitamin B and C combined with
cium flows into mitochondria through VDAC in OMM corticosteroid in sepsis has been suggested as a promising
[24]. Thereafter, sarcoplasmic reticulum ­C a 2+ ATPase therapy but currently lacks robust evidence to support its
(SERCA) reabsorbs calcium to prevent cytoplastic over- widespread use [37]. NOX inhibitors were also indicated to
load, which is a determinant of calcium homeostasis. selectively attenuate disease-related ROS formation without
Tumor suppressor p53 localized in ER can interact with altering physiological signaling ROS [38]. The myocardial
SERCA and exert its pro-apoptotic function by regulating cells switch their metabolism to aerobic glycolysis leading
the transporting calcium into mitochondria [25]. An anti- to the reduction of the ATP generation [3]. It is reasonable to
apoptotic member of the Bcl-2 family has been suggested hypothesize that glycolysis increases and oxidative phospho-
to interact with VDAC and protect cells from death by rylation decreases, contributing to decreasing production of
limiting the transfer of C­ a2+ signals to mitochondria [26, mtROS and reduce the damage from mtROS to mitochondria
27]. Although calcium overload has been widely shown in and cells [39]. Glycolysis can be initiated by hypoxia dur-
septic models which can be via C ­ a2+ channels mentioned ing sepsis. Theoretically, TCA cycle will decrease and the
above, little necrosis has been observed in survivors, production of mtROS will correspondingly decrease. Fur-
compared with autophagy which are mediated by mito- ther in this way, damage from mtROS to mitochondria will
chondria. In mice, autophagy could play a protective role be alleviated, especially ETC which is prone to mtROS. It
against pressure-overload-induced mitochondrial dysfunc- may also indicate self-protection of mitochondria is initi-
tion and heart failure via regulating Drp1 [28]. Upregu- ated when exposed to sepsis, as continuous impairment of
lation of autophagy via activation of SIRT1 could pro- mitochondria including ETC can induce NLRP3 inflamma-
tected against SIMD [29]. Therefore, calcium overload and some [40]. On the other hand, there is a tightly association
necrosis may occur via ­Ca2+ channels mentioned above, between mitochondrial energy metabolism and mitochon-
but not be the first and appropriate choice in survivors, as drial dynamics. As it was known that, LPS-induced M1
the balance of apoptosis via apoptotic member interacting macrophages could reprogram their energetic metabolism
with ­Ca2+ channels would be kept and autophagy would to glycolysis during infection to afford the energy need to
be activated to protect mitochondria and myocardial cells kill pathogens and survive, similar to cancer cells [41]. The
(Fig. 1). metabolic reprogramming in mitochondria is also associ-
ated with altered mitochondrial morphology [42]. Mitochon-
Production and over‑production of mitochondrial drial dynamics can control T cell fate through metabolic
ROS programming. Mitochondrial fission will facilitate glycolysis
and mitophagy which eliminate impaired fragmented mito-
Sepsis can cause cardiac mitochondrial damage by increas- chondria via fission [43] (Fig. 1).
ing oxidative stress and decreasing antioxidant defense,
causing mitochondrial ROS (mtROS) overproduction, fur-
ther mitochondrial functional deficiency, and structural rup- Mitophagy and its potential role
ture due to direct oxidation. The scavenging of mtROS can in protecting myocardial cells in sepsis
be achieved via enzymatic and non-enzymatic antioxidants,
such as glutathione peroxidase (GPx), catalase (CAT), and Mitophagy is an important method to remove damaged
superoxide dismutase (SOD). The accumulation of ROS can mitochondria by isolating dysfunctional mitochondria from
lead to more electron leakage after damaging ETC, activa- healthy ones by double-membrane vesicles called autophago-
tion of poly ADP-ribose polymerase (PARP) after damaging somes and subsequently delivering autophagosomes to

13
382 H. Yang, Z. Zhang

lysosomes. It protects body cells against septic stress [44], scope. Further research on the GPx4 expressed in a myocar-
which can be strongly activated by mitochondrial depolari- dial cell is required, especially focusing on the correlation
zation before damage triggers necrosis [45]. The canonical between mitophagy and expression of GPx4 (Fig. 1).
signal pathways are PINK1/Parkin and DJ-1 pathway. ROS
induced mitochondrial damage might be a necessary activa-
tor to mitophagy [46]. In the pathway, PINK1 kinase accu- Immunosuppression and pyroptosis in SIMD
mulates on the mitochondria and phosphorylates mitofusin 2
(MFN2). Parkin E3 ligase binds phosphorylated MFN2 and Sepsis is a result of uncontrolled primary and secondary
localizes to the mitochondria [4]. The ubiquitination of sev- infection under the condition of abnormal immunity [56].
eral proteins on mitochondrial surface and various adapter It will further result in tissue damage, cellular compromise,
proteins, including MFN1, MFN2, optic atrophy protein 1 and molecular dysregulation, further initiating organ dys-
(OPA1), and VDAC, are associated with the mitophagy, ulti- function and multiorgan failure [57]. In SIMD survivors,
mately facilitating the elimination of the mitochondrial con- necrosis of myocardial cells is little, which indicates the
tent [47, 48]. Moreover, VDAC was shown to be necessary damage from host immune cells and pathogens is not inten-
for the PINK1/Parkin-directed mitophagy of damaged mito- sive enough to induce necrosis, compared with not massive
chondria [49]. LPS injection significantly downregulated apoptosis. Therefore, there would be a balance between host
MFN1, MFN2, and OPA1, which are primary regulators of immunity and pathogens during SIMD as immune cells can
mitochondrial fusion. However, excessive mitochondrial fis- still not clear the pathogens. Immunosuppression involving
sion and mitophagy can compromise the metabolic capacity innate immune and adaptive immunity has always been seen
of a cell [50]. During sepsis, mitochondrial dysfunction is in sepsis because of lymphocyte apoptosis via caspases 8/9
characterized with calcium dysregulation, overproduction and finally caspase 3 by both mitochondrial-mediated and
of ROS, opening of mPTP, and a final loss in mitochondrial receptor-mediated pathways [58, 59]. A decreased produc-
membrane potential, which induces cell death if progressive. tion of inflammatory cytokines has also been demonstrated
But if mitophagy occurs in myocardial cells instead of the such as decreased TNF, IL‑1α, IL‑6, and IL‑12 [60], inter-
above processes, myocardial cells will avoid massive death, feron γ (IFN γ) [61], and macrophage colony-stimulating
benefiting cell survival. factor (GM-CSF). Protein expressed on the lymphocyte
On the other hand, the opening of mPTP is pivotal to the also change. And these changes include the downregula-
fate of mitochondria and cells. And from the content men- tion of CD127 and Bcl-2, and the upregulation of inhibitory
tioned above, it is not the first choice of survival cells. There- receptors [62]. Sepsis can also induce a subset of neutrophils
fore, the reason to keep its functional opening is of interest. with suppressive properties, as these neutrophils produce
Cytochrome C can be released after mPTP opening to induce large amounts of the immunosuppressive cytokine IL-10
apoptosis. The cardiolipin located in IMM is required for the during sepsis. The most dominant function of macrophages
dissociation of cytochrome C after its oxidation, as activated is the phagocytosis of both pathogens and apoptotic cells.
calpain 1 cleaves Bid, inducing cytochrome C release [51]. Macrophages’ clearance of innate immune cells can also be
Therefore, oxidation resistance is critical for cell survival. weakened by an increasing production of IL-10 from neutro-
mGPx4, is a kind of GPx4 transported to mitochondria after phils [63] and pyroptosis, a type of programmed cell death,
synthesis in cell nucleus. It can suppress the peroxidation of via caspases 11 after detecting the LPS invasion in cytosol
cardiolipin in mitochondria and inhibit the dissociation of [64]. Pyroptosic cell can be finally uptake by the neutrophil
cytochrome C from IMM in apoptosis [52]. Furthermore, in response to phagocytosis, which might be able to resist
mGPx4 catalyzes the decomposition of hydrogen perox- pyroptosis in response to some inflammasome activators
ide or organic hydroperoxides using the reduced form of [65]. During sepsis, neutrophil apoptosis is delayed in con-
glutathione as an electron donor [53]. Meanwhile, mGPx4 trast to the lymphocytes undergoing accelerated apoptosis
can inactivate Adenine nucleotide translocator (ANT), a [66], which indicates that (1) neutrophils may last longer
component of mPTP, to inhibit the opening of mPTP and in sepsis than lymphocytes, (2) the clearance function of
the opening of VDAC [54]. If the expression of GPx4 is lymphocytes is actually impaired, and (3) increasing pyrop-
upregulated when a myocardial cell is exposed to sepsis, tosis exists. In addition, Gpx4 expressed by cells from the
the pathway of cell death including apoptosis and necrosis myeloid lineage plays a major role in attenuating lipid perox-
is suppressed, and then, the mitophagy is chosen to begin. idation, inflammasome activation, and pyroptotic cell death
However, cardiac mGPx4 decreases after sepsis, and the in the context of sepsis. The depletion of Gpx4 results in
activity of GPx4 falls to 70% after 12–24 h [55]. As mGPx4 increased septic lethality [67]. GPx4 may be a requisite gate-
is reductive and protective, the more reasonable explana- way to pyroptosis at least via caspases 11 [53]. Therefore,
tion for the “contradiction” is that the expression of mGPx4 pyroptosis will take more account of the programmed cell
is not always increasing, and also has its compensational death of innate immune cells. In this way, a balance can be

13
Sepsis‑induced myocardial dysfunction: the role of mitochondrial dysfunction 383

achieved. Although immune cells are suppressed, pathogens (Flt3L) used to stimulate the expansion and differentiation
can still be swallowed in their cytoplasm which can further of NK cells and dendritic cells (DC) in infectious mice, and
decrease the damage from both immunity and pathogens. IFN γ used to restore energy metabolism in tolerant mono-
This “balanced” status of survivors can be ended with the cytes of mice [70]. And in clinical, the administration of
improvement of host immunity. And continuous increase of GM-CSF therapy was showed to facilitate the reversal of
pyroptosis means the host immunity is still in suppressed immunoparalysis in patients with multiple organ dysfunc-
and pathogens’ virulence is not attenuated, indicating poor tion syndrome (MODS) [71]. As SIMD is characteristic
outcomes. It was also demonstrated that increased LPS- with insufficient function without obvious necrosis of myo-
induced pyroptosis and the inhibition of autophagy by highly cardial cells, it will improve after sepsis is over. Therefore,
expressed ZFAS1 could aggravate the progression of SIMD enhancing body immunity as mentioned examples including
in the in vivo and in vitro model [68] (Fig. 2). enhancing the function of associated cytokines, complement,
immune cells and their energy metabolism will contribute
to SIMD directly and indirectly. As further explanation, for
Enhancing body immunity benefits SIMD the heart function under the condition of immunosuppres-
sion, clearing necrotic or apoptotic myocardial cells will
Some basic research related to the enhancement of immu- also further weaken the function of the innate immune cells
nity has been carried out such as C5a inhibitor used to [72], which is not beneficial for the recovery [73]. The func-
treat sepsis in mice [69], Fms-like tyrosine kinase 3 ligand tion of immune cells will be impaired and further apoptosis

Fig. 2  Body immune reaction


is activated by both patho-
gens and their metabolites via
PAMPs and DAMPs in sepsis.
Immunosuppression involving
innate and adaptive immunity
has always been seen in sepsis
including lymphocyte apoptosis.
Pyroptosis can also be activated
after detecting the LPS invasion
in cytosol. GPx4: glutathione
peroxidase 4; NF-kB: nuclear
factor kB; PRRs: pattern recog-
nition receptors

13
384 H. Yang, Z. Zhang

will be induced by the swallowed pathogen which cannot [81]. Physiologically, oxidative phosphorylation is the
be cleared, if the mitochondria are reduced to an extent via first choice, as ATP is the basis for cells. Even under the
mitophagy. Therefore, enhancing the function of immunity depression of various signal pathways mainly and primar-
and treating the primary infection can decrease the apopto- ily caused by PAMPs and DAMPs, the epigenetic regulat-
sis of immune cells and the proportion of pyroptosis. It can ing gene expression can have a more positive effect on
further improve the clearance of pathogens by host immune a healthy cell than on a suppressed cell in sepsis, which
cells and weaken the suppressing signal from the inflamma- can try to generate more ATP. In contrast, the signal of
tory environment, further recovering myocardial cells after mitophagy may be the dominant regulating signal in a
the fading of related stimulus of sepsis. suppressed cell, which may also process the apoptosis or
necrosis when the damage is caused by severe inflamma-
tion (Fig. 1).
Metabolism reprogramming is pivotal
in SIMD

In SIMD, metabolism reprogramming would occur in both Conclusions


immune cells and myocardial cells. Under the homeostatic
condition, immune cells rely on oxidative phosphorylation SIMD, as a common complication in septic patients, is asso-
as the energy sources of ATP production. It will be shifted to ciated with increased mortality. Changes in any part of CICR
aerobic glycolysis during sepsis, which is known as Warburg and excitation–contraction coupling (ECC) influence the
effect [3]. It was suggested that the glycolysis of macrophage heart function upon exposure to inflammatory environment
facilitated a macrophage proinflammatory phenotype and and damage from inflammatory mediators and cytokines
induced IL-1β production [3, 74]. Moreover, aerobic glyco- after the initiation of an immune reaction on pathogens.
lysis could provide ATP for the immune response immedi- Mitophagy may be the compressive choice of a myocardial
ately [75]. Aerobic glycolysis exists during critical illness in cell when facing sepsis, and is even chosen in the case of
both immune cells and the other cells [76]. And if fission, death through necrosis or apoptosis or swallowing by mac-
mitophagy, and aerobic glycolysis occur in myocardial cells, rophages and degeneration. Sepsis is a result of uncontrolled
it will reduce the damage from mtROS and keep myocar- primary and secondary infections under the condition of
dial cells from massive apoptosis, which provide myocardial abnormal immunity. Immunosuppression has always been
cells more opportunities to recover after sepsis. However, observed in the case of sepsis, featured by a significant apop-
the inhibition of aerobic glycolysis by 2-DG significantly tosis of immune cells and a comparative increase in regula-
improves the survival outcome in bacterial sepsis [77]. It tory T cell and IL-10. Enhancing the body’s immunity may
is because lactate significantly increases after the shift of be more efficient to prevent or improve SIMD and control
the energy metabolism to glycolysis during sepsis. And the sepsis, besides the treatment of the primary infection. How-
immunosuppressive effects of the tumor-derived lactate have ever, the research on the correlation between mitophagy and
been shown on a variety of cell types in the surrounding expression of GPx4, the recognition of the transitional or
microenvironment [78], and lactate has been suggested to cut-off point of SIMD and immunosuppression to achieve
induce M2-like macrophage polarization through an HIF- early prevention, and the selection of the biomarkers that can
1a-dependent mechanism [79]. According to the content be used to indicate the possibility of SIMD and immunosup-
mentioned above, if the inhibition of aerobic glycolysis is pression need further research.
applied during immunosuppression, it has the similar role
with the enhancement of body immunity, as it can alleviate
the immunosuppressive effects of lactate by decreasing its Authors’ contributions Hang Yang contributed to design and draft the
manuscript. And Zhaocai Zhang contributed to design and review the
production. manuscript.
Another question is whether SIMD is mainly caused
by inflammatory environment because of both slightly Funding This study was funded by National Natural Science Founda-
reversible damage and the epigenetic regulation of gene tion of China (Nos. 81772110, 82072202).
expression. It has been demonstrated that sirtuin 1 and sir-
tuin 6 can epigenetically shift substrate selection in septic Declarations
mouse or human monocytes from the glucose fueling of
the immune resistance to the fatty acid fueling of immune Conflict of interest Author Hang Yang declares that she has no conflict
of interest. Author Zhaocai Zhang declares that he has no conflict of
tolerance [80], and mitochondrial sirtuin 3 expression interest.
could be increased by sirtuin 1 to support mitochondrial
catabolic energetics and activate the antioxidant pathways

13
Sepsis‑induced myocardial dysfunction: the role of mitochondrial dysfunction 385

Ethics approval This article does not contain any studies with human 15. Bernardi P, Rasola A, Forte M, Lippe G. The Mitochondrial per-
participants or animals performed by any of the authors. meability transition pore: channel formation by F-ATP synthase,
integration in signal transduction, and role in pathophysiology.
Physiol Rev. 2015;95(4):1111–55. https​://doi.org/10.1152/physr​
ev.00001​.2015.
References 16. Elrod JW, Molkentin JD. Physiologic functions of cyclophilin
D and the mitochondrial permeability transition pore. Circ J.
2013;77(5):1111–22.
1. Stanzani G, Duchen MR, Singer M. The role of mitochon-
17. Izzo V, Bravo-San Pedro JM, Sica V, Kroemer G, Galluzzi L.
dria in sepsis-induced cardiomyopathy. Biochim Biophys
Mitochondrial permeability transition: new findings and persist-
Acta. 2019;1865(4):759–73. https​: //doi.org/10.1016/j.bbadi​
ing uncertainties. Trends Cell Biol. 2016;26(9):655–67. https​://
s.2018.10.011.
doi.org/10.1016/j.tcb.2016.04.006.
2. Wilhelm J, Hettwer S, Schuermann M, Bagger S, Gerhardt F,
18. Halestrap AP. What is the mitochondrial permeability transi-
Mundt S, et al. Severity of cardiac impairment in the early stage
tion pore? J Mol Cell Cardiol. 2009;46(6):821–31. https​://doi.
of community-acquired sepsis determines worse prognosis. Clin
org/10.1016/j.yjmcc​.2009.02.021.
Res Cardiol. 2013;102(10):735–44. https​: //doi.org/10.1007/
19. Briston T, Selwood DL, Szabadkai G, Duchen MR. Mitochon-
s0039​2-013-0584-z.
drial permeability transition: a molecular lesion with multiple
3. Van Wyngene L, Vandewalle J, Libert C. Reprogramming of
drug targets. Trends Pharmacol Sci. 2019;40(1):50–70. https​://
basic metabolic pathways in microbial sepsis: therapeutic tar-
doi.org/10.1016/j.tips.2018.11.004.
gets at last? EMBO Mol Med. 2018. https​://doi.org/10.15252​/
20. Kim B, Takeuchi A, Koga O, Hikida M, Matsuoka S. Mito-
emmm.20170​8712.
chondria Na(+)-Ca (2+) exchange in cardiomyocytes and lym-
4. Vasquez-Trincado C, Garcia-Carvajal I, Pennanen C, Parra
phocytes. Adv Exp Med Biol. 2013;961:193–201. https​://doi.
V, Hill JA, Rothermel BA, et al. Mitochondrial dynam-
org/10.1007/978-1-4614-4756-6_16.
ics, mitophagy and cardiovascular disease. J Physiol.
21. Luongo TS, Lambert JP, Yuan A, Zhang X, Gross P, Song J,
2016;594(3):509–25. https​://doi.org/10.1113/jp271​301.
et al. The mitochondrial calcium uniporter matches energetic
5. Sack MN. Emerging characterization of the role of SIRT3-
supply with cardiac workload during stress and modulates per-
mediated mitochondrial protein deacetylation in the heart. Am
meability transition. Cell Rep. 2015;12(1):23–34. https​://doi.
J Physiol Heart Circ Physiol. 2011;301(6):H2191–7. https​://doi.
org/10.1016/j.celre​p.2015.06.017.
org/10.1152/ajphe​art.00199​.2011.
22. Szabadkai G, Bianchi K, Varnai P, De Stefani D, Wieckowski
6. Liu YC, Yu MM, Shou ST, Chai YF. Sepsis-induced car-
MR, Cavagna D, et al. Chaperone-mediated coupling of endo-
diomyopathy: mechanisms and treatments. Front Immunol.
plasmic reticulum and mitochondrial Ca2+ channels. J Cell
2017;8:1021. https​://doi.org/10.3389/fimmu​.2017.01021​.
Biol. 2006;175(6):901–11. https​://doi.org/10.1083/jcb.20060​
7. Zhang Z, Deng W, Kang R, Xie M, Billiar T, Wang H, et al.
8073.
Plumbagin protects mice from lethal sepsis by modulat-
23. Giorgi C, Missiroli S, Patergnani S, Duszynski J, Wieckowski
ing immunometabolism upstream of PKM2. Mol Med.
MR, Pinton P. Mitochondria-associated membranes: composi-
2016;22:162–72. https​://doi.org/10.2119/molme​d.2015.00250​.
tion, molecular mechanisms, and physiopathological implica-
8. Kalbitz M, Fattahi F, Grailer JJ, Jajou L, Malan EA, Zetoune
tions. Antioxid Redox Signal. 2015;22(12):995–1019. https​://
FS, et al. Complement-induced activation of the cardiac NLRP3
doi.org/10.1089/ars.2014.6223.
inflammasome in sepsis. FASEB J. 2016;30(12):3997–4006.
24. De Pinto VD, Palmieri F. Transmembrane arrangement of mito-
https​://doi.org/10.1096/fj.20160​0728R​.
chondrial porin or voltage-dependent anion channel (VDAC). J
9. Chagnon F, Bentourkia M, Lecomte R, Lessard M, Lesur O.
Bioenerg Biomembr. 1992;24(1):21–6.
Endotoxin-induced heart dysfunction in rats: assessment of
25. Giorgi C, Bonora M, Sorrentino G, Missiroli S, Poletti F, Suski
myocardial perfusion and permeability and the role of fluid
JM, et al. p53 at the endoplasmic reticulum regulates apop-
resuscitation. Crit Care Med. 2006;34(1):127–33. https​://doi.
tosis in a Ca2+-dependent manner. Proc Natl Acad Sci USA.
org/10.1097/01.ccm.00001​90622​.02222​.df.
2015;112(6):1779–84. https​://doi.org/10.1073/pnas.14107​23112​.
10. Sun LJ, Qiao W, Xiao YJ, Cui L, Wang X, Ren WD. Naringin
26. Monaco G, Decrock E, Arbel N, van Vliet AR, La Rovere RM, De
mitigates myocardial strain and the inflammatory response
Smedt H, et al. The BH4 domain of anti-apoptotic Bcl-XL, but not
in sepsis-induced myocardial dysfunction through regula-
that of the related Bcl-2, limits the voltage-dependent anion chan-
tion of PI3K/AKT/NF-κB pathway. Int Immunopharmacol.
nel 1 (VDAC1)-mediated transfer of pro-apoptotic Ca2+ signals
2019;75:105782. https​://doi.org/10.1016/j.intim​p.2019.10578​2.
to mitochondria. J Biol Chem. 2015;290(14):9150–61. https:​ //doi.
11. Perez MJ, Quintanilla RA. Development or disease: duality
org/10.1074/jbc.M114.62251​4.
of the mitochondrial permeability transition pore. Dev Biol.
27. Arbel N, Ben-Hail D, Shoshan-Barmatz V. Mediation of the
2017;426(1):1–7. https​://doi.org/10.1016/j.ydbio​.2017.04.018.
antiapoptotic activity of Bcl-xL protein upon interaction with
12. Kwong JQ, Molkentin JD. Physiological and pathological roles
VDAC1 protein. J Biol Chem. 2012;287(27):23152–61. https​://
of the mitochondrial permeability transition pore in the heart.
doi.org/10.1074/jbc.M112.34591​8.
Cell Metab. 2015;21(2):206–14. https​: //doi.org/10.1016/j.
28. Shirakabe A, Zhai P, Ikeda Y, Saito T, Maejima Y, Hsu CP, et al.
cmet.2014.12.001.
Drp1-dependent mitochondrial autophagy plays a protective role
13. Hurst S, Hoek J, Sheu SS. Mitochondrial Ca(2+) and regula-
against pressure overload-induced mitochondrial dysfunction and
tion of the permeability transition pore. J Bioenerg Biomembr.
heart failure. Circulation. 2016;133(13):1249–63. https​://doi.
2017;49(1):27–47. https​://doi.org/10.1007/s1086​3-016-9672-x.
org/10.1161/circu​latio​naha.115.02050​2.
14. Jonas EA, Porter GA Jr, Beutner G, Mnatsakanyan N, Ala-
29. Zhang WX, He BM, Wu Y, Qiao JF, Peng ZY. Melatonin protects
vian KN. Cell death disguised: the mitochondrial permeability
against sepsis-induced cardiac dysfunction by regulating apop-
transition pore as the c-subunit of the F(1)F(O) ATP synthase.
tosis and autophagy via activation of SIRT1 in mice. Life Sci.
Pharmacol Res. 2015;99:382–92. https​: //doi.org/10.1016/j.
2019;217:8–15. https​://doi.org/10.1016/j.lfs.2018.11.055.
phrs.2015.04.013.
30. Bai P, Nagy L, Fodor T, Liaudet L, Pacher P. Poly(ADP-ribose)
polymerases as modulators of mitochondrial activity. Trends in

13
386 H. Yang, Z. Zhang

endocrinology and metabolism: TEM. 2015;26(2):75–83. https​:// 47. Chan NC, Salazar AM, Pham AH, Sweredoski MJ, Kolawa NJ,
doi.org/10.1016/j.tem.2014.11.003. Graham RL, et al. Broad activation of the ubiquitin-proteasome
31. Larche J, Lancel S, Hassoun SM, Favory R, Decoster B, Mar- system by Parkin is critical for mitophagy. Hum Mol Genet.
chetti P, et al. Inhibition of mitochondrial permeability transition 2011;20(9):1726–37. https​://doi.org/10.1093/hmg/ddr04​8.
prevents sepsis-induced myocardial dysfunction and mortality. J 48. Chen Y, Dorn GW 2nd. PINK1-phosphorylated mitofusin 2 is
Am Coll Cardiol. 2006;48(2):377–85. https​://doi.org/10.1016/j. a Parkin receptor for culling damaged mitochondria. Science.
jacc.2006.02.069. 2013;340(6131):471–5. https:​ //doi.org/10.1126/scienc​ e.123103​ 1.
32. Lee I, Huttemann M. Energy crisis: the role of oxidative phos- 49. Geisler S, Holmstrom KM, Skujat D, Fiesel FC, Rothfuss OC,
phorylation in acute inflammation and sepsis. Biochem Biophys Kahle PJ, et al. PINK1/Parkin-mediated mitophagy is dependent
Acta. 2014;1842(9):1579–86. https​://doi.org/10.1016/j.bbadi​ on VDAC1 and p62/SQSTM1. Nat Cell Biol. 2010;12(2):119–31.
s.2014.05.031. https​://doi.org/10.1038/ncb20​12.
33. Brand MD, Nicholls DG. Assessing mitochondrial dysfunction in 50. Twig G, Shirihai OS. The interplay between mitochondrial dynam-
cells. Biochem J. 2011;435(2):297–312. https​://doi.org/10.1042/ ics and mitophagy. Antioxid Redox Signal. 2011;14(10):1939–51.
bj201​10162​. https​://doi.org/10.1089/ars.2010.3779.
34. Goncalves RL, Quinlan CL, Perevoshchikova IV, Hey-Mogensen 51. Smith MA, Schnellmann RG. Calpains, mitochondria, and apop-
M, Brand MD. Sites of superoxide and hydrogen peroxide produc- tosis. Cardiovasc Res. 2012;96(1):32–7. https​://doi.org/10.1093/
tion by muscle mitochondria assessed ex vivo under conditions cvr/cvs16​3.
mimicking rest and exercise. J Biol Chem. 2015;290(1):209–27. 52. Nomura K, Imai H, Koumura T, Kobayashi T, Nakagawa Y.
https​://doi.org/10.1074/jbc.M114.61907​2. Mitochondrial phospholipid hydroperoxide glutathione peroxi-
35. Chandran K, Aggarwal D, Migrino RQ, Joseph J, McAllis- dase inhibits the release of cytochrome c from mitochondria by
ter D, Konorev EA, et al. Doxorubicin inactivates myocardial suppressing the peroxidation of cardiolipin in hypoglycaemia-
cytochrome c oxidase in rats: cardioprotection by Mito-Q. Biophys induced apoptosis. Biochem J. 2000;351(Pt 1):183–93. https​://
J. 2009;96(4):1388–98. https:​ //doi.org/10.1016/j.bpj.2008.10.042. doi.org/10.1042/0264-6021:35101​83.
36. Liu XR, Cao L, Li T, Chen LL, Yu YY, Huang WJ, et al. Propofol 53. Zhu H, Santo A, Jia Z, Robert LY. GPx4 in bacterial infection and
attenuates H(2)O(2)-induced oxidative stress and apoptosis via the polymicrobial sepsis: involvement of ferroptosis and pyroptosis.
mitochondria- and ER-medicated pathways in neonatal rat cardio- React Oxyg Species. 2019;7(21):154–60. https:​ //doi.org/10.20455​
myocytes. Apoptosis. 2017;22(5):639–46. https:​ //doi.org/10.1007/ /ros.2019.835.
s1049​5-017-1349-3. 54. Imai H, Nakagawa Y. Biological significance of phospholipid
37. Moskowitz A, Andersen LW, Huang DT, Berg KM, Grossestreuer hydroperoxide glutathione peroxidase (PHGPx, GPx4) in mam-
AV, Marik PE, et al. Ascorbic acid, corticosteroids, and thiamine malian cells. Free Radical Biol Med. 2003;34(2):145–69.
in sepsis: a review of the biologic rationale and the present state 55. Zang Q, Maass DL, Tsai SJ, Horton JW. Cardiac mitochon-
of clinical evaluation. Crit Care. 2018;22(1):283. https​://doi. drial damage and inflammation responses in sepsis. Surg Infect.
org/10.1186/s1305​4-018-2217-4. 2007;8(1):41–54. https​://doi.org/10.1089/sur.2006.033.
38. Brandes RP, Weissmann N, Schröder K. Nox family NADPH 56. Kumar V. Correction to: Immunometabolism: another road to sep-
oxidases: Molecular mechanisms of activation. Free Radical sis and its therapeutic targeting. Inflammation. 2019;42(3):789.
Biol Med. 2014;76:208–26. https​://doi.org/10.1016/j.freer​adbio​ https​://doi.org/10.1007/s1075​3-019-00970​-x.
med.2014.07.046. 57. Rittirsch D, Flierl MA, Ward PA. Harmful molecular mecha-
39. Li S, Wu H, Han D, Ma S, Fan W, Wang Y, et al. A novel nisms in sepsis. Nat Rev Immunol. 2008;8(10):776–87. https​://
mechanism of mesenchymal stromal cell-mediated protection doi.org/10.1038/nri24​02.
against sepsis: restricting inflammasome activation in mac- 58. Hotchkiss RS, Osmon SB, Chang KC, Wagner TH, Cooper-
rophages by increasing mitophagy and decreasing mitochondrial smith CM, Karl IE. Accelerated lymphocyte death in sepsis
ROS. Oxid Med Cell Longev. 2018;2018:3537609. https​://doi. occurs by both the death receptor and mitochondrial pathways.
org/10.1155/2018/35376​09. J Immunol. 2005;174(8):5110–8. https​://doi.org/10.4049/jimmu​
40. Liu Q, Zhang D, Hu D, Zhou X, Zhou Y. The role of mito- nol.174.8.5110.
chondria in NLRP3 inflammasome activation. Mol Immunol. 59. Chang KC, Unsinger J, Davis CG, Schwulst SJ, Muenzer JT,
2018;103:115–24. https:​ //doi.org/10.1016/j.molimm ​ .2018.09.010. Strasser A, et al. Multiple triggers of cell death in sepsis: death
41. Chang M, Hamilton JA, Scholz GM, Elsegood CL. Glycolytic receptor and mitochondrial-mediated apoptosis. FASEB J.
control of adjuvant-induced macrophage survival: role of PI3K, 2007;21(3):708–19. https​://doi.org/10.1096/fj.06-6805c​om.
MEK1/2, and Bcl-2. J Leukoc Biol. 2009;85(6):947–56. https​:// 60. Cavaillon JM, Adib-Conquy M. Bench-to-bedside review: endo-
doi.org/10.1189/jlb.09085​22. toxin tolerance as a model of leukocyte reprogramming in sepsis.
42. Zuo H, Wan Y. Metabolic reprogramming in mitochondria of Crit Care. 2006;10(5):233. https​://doi.org/10.1186/cc505​5.
myeloid cells. Cells. 2019. https:​ //doi.org/10.3390/cells​90100​05. 61. Delano MJ, Scumpia PO, Weinstein JS, Coco D, Nagaraj S, Kelly-
43. Buck MD, O’Sullivan D, Klein Geltink RI, Curtis JD, Chang Scumpia KM, et al. MyD88-dependent expansion of an imma-
CH, Sanin DE, et al. Mitochondrial dynamics controls T cell fate ture GR-1(+)CD11b(+) population induces T cell suppression
through metabolic programming. Cell. 2016;166(1):63–76. https​ and Th2 polarization in sepsis. J Exp Med. 2007;204(6):1463–74.
://doi.org/10.1016/j.cell.2016.05.035. https​://doi.org/10.1084/jem.20062​602.
44. Youle RJ, Narendra DP. Mechanisms of mitophagy. Nat Rev Mol 62. Boomer JS, To K, Chang KC, Takasu O, Osborne DF, Walton
Cell Biol. 2011;12(1):9–14. https​://doi.org/10.1038/nrm30​28. AH, et al. Immunosuppression in patients who die of sepsis and
45. Kubli DA, Gustafsson AB. Mitochondria and mitophagy: the yin multiple organ failure. JAMA. 2011;306(23):2594–605. https​://
and yang of cell death control. Circ Res. 2012;111(9):1208–21. doi.org/10.1001/jama.2011.1829.
https​://doi.org/10.1161/circr​esaha​.112.26581​9. 63. Hotchkiss RS, Monneret G, Payen D. Sepsis-induced immunosup-
46. Wang Y, Nartiss Y, Steipe B, McQuibban GA, Kim PK. ROS- pression: from cellular dysfunctions to immunotherapy. Nat Rev
induced mitochondrial depolarization initiates PARK2/PARKIN- Immunol. 2013;13(12):862–74. https​://doi.org/10.1038/nri35​52.
dependent mitochondrial degradation by autophagy. Autophagy.
2012;8(10):1462–76. https​://doi.org/10.4161/auto.21211​.

13
Sepsis‑induced myocardial dysfunction: the role of mitochondrial dysfunction 387

64. Jorgensen I, Rayamajhi M, Miao EA. Programmed cell death as a 74. Vander Heiden MG, Cantley LC, Thompson CB. Understanding
defence against infection. Nat Rev Immunol. 2017;17(3):151–64. the Warburg effect: the metabolic requirements of cell prolifera-
https​://doi.org/10.1038/nri.2016.147. tion. Science. 2009;324(5930):1029–33. https​://doi.org/10.1126/
65. Chen KW, Gross CJ, Sotomayor FV, Stacey KJ, Tschopp J, Sweet scien​ce.11608​09.
MJ, et al. The neutrophil NLRC4 inflammasome selectively pro- 75. Bauer DE, Harris MH, Plas DR, Lum JJ, Hammerman PS,
motes IL-1beta maturation without pyroptosis during acute Sal- Rathmell JC, et al. Cytokine stimulation of aerobic glycolysis
monella challenge. Cell reports. 2014;8(2):570–82. https​://doi. in hematopoietic cells exceeds proliferative demand. FASEB J.
org/10.1016/j.celre​p.2014.06.028. 2004;18(11):1303–5. https​://doi.org/10.1096/fj.03-1001f​j e.
66. Drifte G, Dunn-Siegrist I, Tissieres P, Pugin J. Innate immune 76. Donnelly RP, Finlay DK. Glucose, glycolysis and lympho-
functions of immature neutrophils in patients with sepsis and cyte responses. Mol Immunol. 2015;68(2):513–9. https​://doi.
severe systemic inflammatory response syndrome. Crit Care Med. org/10.1016/j.molim​m.2015.07.034.
2013;41(3):820–32. https​://doi.org/10.1097/CCM.0b013​e3182​ 77. Wang A, Huen SC, Luan HH, Yu S, Zhang C, Gallezot JD, et al.
74647​d. Opposing effects of fasting metabolism on tissue tolerance in
67. Kang R, Zeng L, Zhu S, Xie Y, Liu J, Wen Q, et al. Lipid Peroxi- bacterial and viral inflammation. Cell. 2016;166(6):1512-25.e12.
dation drives gasdermin D-mediated pyroptosis in lethal polymi- https​://doi.org/10.1016/j.cell.2016.07.026.
crobial sepsis. Cell Host Microbe. 2018;24(1):97-108.e4. https​:// 78. Choi SY, Collins CC, Gout PW, Wang Y. Cancer-generated lac-
doi.org/10.1016/j.chom.2018.05.009. tic acid: a regulatory, immunosuppressive metabolite? J Pathol.
68. Liu JJ, Li Y, Yang MS, Chen R, Cen CQ. SP1-induced ZFAS1 2013;230(4):350–5. https​://doi.org/10.1002/path.4218.
aggravates sepsis-induced cardiac dysfunction via miR-590-3p/ 79. Colegio OR, Chu NQ, Szabo AL, Chu T, Rhebergen AM, Jairam
NLRP3-mediated autophagy and pyroptosis. Arch Biochem Bio- V, et al. Functional polarization of tumour-associated macrophages
phys. 2020;695:108611. https:​ //doi.org/10.1016/j.abb.2020.10861​ by tumour-derived lactic acid. Nature. 2014;513(7519):559–63.
1. https​://doi.org/10.1038/natur​e1349​0.
69. Melis JP, Strumane K, Ruuls SR, Beurskens FJ, Schuurman J, 80. Liu TF, Vachharajani VT, Yoza BK, McCall CE. NAD+-depend-
Parren PW. Complement in therapy and disease: Regulating ent sirtuin 1 and 6 proteins coordinate a switch from glucose to
the complement system with antibody-based therapeutics. Mol fatty acid oxidation during the acute inflammatory response. J
Immunol. 2015;67(2):117–30. https​://doi.org/10.1016/j.molim​ Biol Chem. 2012;287(31):25758–69. https​://doi.org/10.1074/jbc.
m.2015.01.028. M112.36234​3.
70. Cheng SC, Scicluna BP, Arts RJ, Gresnigt MS, Lachmandas E, 81. Liu TF, Vachharajani V, Millet P, Bharadwaj MS, Molina AJ,
Giamarellos-Bourboulis EJ, et al. Broad defects in the energy McCall CE. Sequential actions of SIRT1-RELB-SIRT3 coordi-
metabolism of leukocytes underlie immunoparalysis in sepsis. Nat nate nuclear-mitochondrial communication during immunometa-
Immunol. 2016;17(4):406–13. https​://doi.org/10.1038/ni.3398. bolic adaptation to acute inflammation and sepsis. J Biol Chem.
71. Hall MW, Knatz NL, Vetterly C, Tomarello S, Wewers MD, Volk 2015;290(1):396–408. https​://doi.org/10.1074/jbc.M114.56634​9.
HD, et al. Immunoparalysis and nosocomial infection in children
with multiple organ dysfunction syndrome. Intensive Care Med. Publisher’s Note Springer Nature remains neutral with regard to
2011;37(3):525–32. https​://doi.org/10.1007/s0013​4-010-2088-x. jurisdictional claims in published maps and institutional affiliations.
72. Voll RE, Herrmann M, Roth EA, Stach C, Kalden JR, Girkon-
taite I. Immunosuppressive effects of apoptotic cells. Nature.
1997;390(6658):350–1. https​://doi.org/10.1038/37022​.
73. Zhang J. Autophagy and mitophagy in cellular damage control.
Redox biology. 2013;1(1):19–23. https​://doi.org/10.1016/j.redox​
.2012.11.008.

13

You might also like