Introduction

Traumatic spinal cord injury (SCI) leads to neurological deficits below the level of injury. In the United States alone there were approximately 282,000 people living with SCI in 2016, and an additional 17,000 new SCI cases occur every year [1]. To date, there has been no effective treatment available for SCI patients [2].

Phospholipids are the main components of a neural cell bi-layer membrane. They not only constitute the backbone of a neural membrane, but also provide the membrane with suitable physical properties, fluidity, and ion permeability, which are required for the proper function of integral membrane proteins, receptors, and ion channels [3,4,5]. Increasing evidence points to the roles of phospholipids as signaling molecules, as participants and coordinators of responses to physiological regulation, and as danger signals [6,7,8]. Although phospholipid alterations have been implicated in SCI [9,10,11,12], their profiles and specific roles in mediating damage remain elusive.

Cardiolipin (CL) is a class of structurally unique dimeric phospholipids that is exclusively present in the inner mitochondrial membrane (IMM) where it is required for optimal mitochondrial function [13, 14]. In addition to its role in maintaining membrane potential and architecture, CL is known to provide essential structural and functional support to several proteins involved in mitochondrial bioenergetics [13,14,15]. Recently, CL is emerging as an important player in the control of the mitochondrial phase of apoptosis [13, 14, 16]. CL alteration (peroxidation and/or loss of CL content) has been implicated in mitochondrial dysfunction and cell death in multiple tissues in a variety of pathological conditions including ischemia, hypothyroidism, aging, heart failure, and traumatic brain injury [13, 17,18,19]. However, limited information is available concerning its role and underlying mechanism following a trauma, such as a SCI. In this study, we employed lipidomics to investigate CL alterations following SCI, and explored the molecular mechanisms of its alterations. We used both in vitro and in vivo experiments to determine whether CL alteration would contribute to the pathogenesis of cell death and functional impairments following SCI.

Results

An increase in biomarkers of apoptosis indicates cardiolipin alteration after SCI

CL is a class of mitochondrial phospholipids which are known to be intimately linked to the mitochondrial bioenergetic machinery [13, 14]. In recent years, CL has been increasingly recognized as playing a key role in mitochondria-mediated apoptosis [13, 14, 16]. CL loss causes the release of cytochrome c and Smac/DIABLO, as well as an increase in caspase-3 activity during the induction of apoptosis [16, 20, 21]. Simultaneous measurements of cytochrome c release, Smac/DIABLO release, and caspase-3 activity can be used to assess the extent of cardiolipin loss. Western blot analysis revealed that both releases of cytochrome c (Fig. 1A) and Smac/DIABLO (Fig. 1B) markedly increased and peaked at 3 h post-SCI compared to the control. Although both releases declined after 1 day post-injury, they remained at significantly elevated levels up to 7 days post-injury (p < 0.05) (Fig. 1A, B). Expression of active caspase-3 was also markedly increased at 1 day post-injury, and reached its peak at 3 and 7 days post-injury (Fig. 1C, Supplementary Fig. 1). These findings suggest that SCI may induce CL alteration.

Fig. 1: Cardiolipin (CL) alteration in the injured spinal cord following spinal cord injury (SCI).
figure 1

AC Biomarkers of apoptosis following SCI. Time course of cytochrome c release (A), Smac/DIABLO release (B), and active caspase-3 expression (C) following SCI. *P < 0.05, **P < 0.01 vs sham (One-way ANOVA, Dunnett post-test, n = 4 rats/group). Data represent mean ± s.e.m. Cyt. c cytochrome c, Smac/D. Smac/DIABLO, A. casp.-3 active casepase-3. DI Lipidomic analysis after SCI. Lipid extracts of spinal cord from sham, 3 h, and 24 h SCI rats were prepared by using the methyl-tert-butyl ether (MTBE) extraction. D, E Expanded negative-ion ESI mass spectra of rat spinal cord lipid extracts obtained using a Q Exactive™ Hybrid Quadrupole-Orbitrap mass spectrometer. The asterisks indicate the identified CL plus-one isotopologues, which were characteristic of the doubly charged CL molecular species and were used to quantify individual CL molecular species. Each spectrum is displayed after being normalized to the intensity of internal standard 1’, 3’-bis [1, 2-dimyristoyl-sn-glycero-3-phospho]-sn-glycerol (tetra14:0 CL, [CL-2H + 1]2- = 619.91791) ion. The x-axis is mass-to-charge ratio (m/z). A significant decrease in CL was detected at 3 and 24 h post-injury (DF) while a corresponding increase in lyso-CL and ratio of lyso-CL/CL was observed at 24 h post-injury (G, H). 4-HNE, a marker of lipid peroxidation, was significantly increased at 3 and 24 h after SCI (I). Data represent mean ± s.e.m. *P < 0.05, **P < 0.01 (One-way ANOVA, Tukey’s multiple comparisons test, n = 4 rats/groups). CL cardiolipin, Lyso-CL lyso-cardiolipin, 4-HNE 4-hydroxy Nonenal.

Lipidomic analysis shows cardiolipin alterations in the spinal cord after SCI

To further determine CL alteration after SCI, molecular species of CL were characterized by electrospray ionization mass spectrometry (ESI-MS) using the negative-ion mode as previously described [22]. Representative ESI-MS spectra of lipids in the spinal cord with and without injury are presented in Fig. 1D, E. Our lipidomic analysis showed that CL was significantly reduced (Fig. 1D–F) and 4-hydroxy nonenal (4-HNE), a lipid peroxidation marker, significantly increased at 3 and 24 h after SCI in a rat contusive SCI (Fig. 1I). Furthermore, lyso-CL (Fig. 1G) and the ratio of lyso-CL/CL (Fig. 1H) increased at 24 h after SCI. These results suggest that CL may undergo peroxidation at 3 h, and both peroxidation and loss at 24 h after SCI.

Lipidomic analysis identifies changes of molecular species and acyl composition of cardiolipin after SCI

The profile of CL molecular species was observed through a broad mass range (Fig. 1D, E and Fig. 2A). Over 50 distinct CL molecular species were readily identified which included polyunsaturated fatty acids such as arachidonic acid (C20:4) (AA), docosahexaenoic acid (C22:6) (DHA), and linoleic acid (C18:2) (LA) that are highly susceptible to peroxidation (Fig. 2B, C). The 18:1 fatty acid is predominantly composed of CL and represents 49.8% of CL compositions (Fig. 2B, C). Major other species of acyls are 20:4 (AA, 20.0%), 22:6 (DHA, 12.3%) and 18:2 (LA, 8.1%) (Fig. 2B, C). Quantitative assessments revealed a significant reduction of CL molecular ions with m/z (mass-to-charge ratio) 714.00, 727.00, 728.01, 736.99, 738.00, 739.00, 750.00, 751.00, 759.98, 760.99, 762.00, 763.00, 774.00, 796.99 as doubly-charged ions after SCI (Fig. 2A). About 21.4% of CL molecular species were significantly reduced at 3 h after SCI and 50% of CL molecular species was significantly reduced at 24 h after SCI. Acyls of CL such as 18:1, 20:4, 22:6 18:2, and 20:3 fatty acids were significantly reduced after SCI (Fig. 2B, C).

Fig. 2: Quantification of molecular species and acyl composition of cardiolipin (CL) in the injured spinal cord at 3 and 24 h after SCI.
figure 2

A Quantification of CL molecular species at 3 and 24 h after SCI. Over 50 distinct CL molecular species were readily identified. Of them, 50% of species of CL were significantly reduced after SCI. *P < 0.05, **P < 0.01 vs. sham (Two-way ANOVA, Tukey’s multiple comparisons test, n = 4 rats/groups). Data represent mean ± s.e.m. The x-axis is mass-to-charge ratio (m/z). B, C Quantification of acyl composition in CL species at 3 and 24 h after SCI. 18:1 fatty acid was predominantly composed of CL and was 49.86% of CL compositions. The following enrichment of acyl was 20:4 (AA, 18.99%), 22:6 (DHA, 12.29%) and 18:2 (LA, 8.08%). After SCI, all decreased CL acyls were unsaturated fatty acids; of them, 50% were poly-unsaturated fatty acids. *P < 0.05, **P < 0.01 vs. sham (Two-way ANOVA, Tukey’s multiple comparisons test, n = 4 rats/groups). Data represent mean ± s.e.m.

Cardiolipin alteration induces neuronal death

To explore whether SCI-induced CL alteration (peroxidation and loss) was sufficient to induce cell death, we first investigated the effect of CL peroxidation on cell death of spinal cord neurons. We found that exposure of primary neurons to oxidized cardiolipin resulted in dose-dependent cell death (Fig. 3A, B). Second, we examined the effects of H2O2, a ROS inducer, and rotenone, an endogenous mitochondrial ROS inducer on CL peroxidation, mitochondrial function, and cell death in cultured spinal cord neurons. CL content was detected with acridine orange 10-nonyl bromide (NAO, Invitrogen), which binds to CL with high affinity. The results showed that rotenone and H2O2 induced CL loss (Fig. 3C, F), mitochondrial dysfunction (Fig. 3D, G), and neuronal death (Fig. 3E, H). Importantly, such effects were reversed by XJB-5-131 (XJB), a novel mitochondria-targeted electron and reactive oxygen species (ROS) scavenger that reduces CL degradation and cell death [18, 23], suggesting that CL oxidation induced neuronal death. Last, we investigated the effect of reduced CL on spinal cord neuronal death by knocking down CL synthase, a key enzyme of de novo CL biosynthesis, with cardiolipin synthase 1 (CLS1) siRNA in vitro. The results showed that CLS1 siRNA reduced CL content (Fig. 3I) and increased apoptotic cells positively stained with activated caspase-3/7 in their nuclei (bright green, Fig. 3J–P). These findings suggest that CL alteration could induce neuronal death.

Fig. 3: Effects of cardiolipin (CL) alteration on neuronal death.
figure 3

A, B Cultured spinal cord neurons were treated with the designated concentrations of oxidized CL (OXCL) and non-oxidized CL for 24 h. Neuronal cell death was measured by LDH, a stable cytoplasmic enzyme that is present in all cells but only released when the plasma membrane is damaged. MTT assay was used to determine the cell viability and mitochondrial activity, since tetrazolium is reduced to formazan by mitochondrial dehydrogenase activity. The oxidized CL induced spinal cord neuronal death (A) as measured by LDH release and mitochondrial dysfunction (B) as measured by MTT in a dose-dependent manner. **P < 0.01 (Two-way ANOVA, Tukey’s multiple comparisons test). Data represent the mean ± s.e.m. from 3 independent experiments. CH The spinal neuronal cultures were exposed to rotenone (125 nM) or H2O2 (50 μM) either in the absence or presence of 10 μM XJB-5-131 (XJB) for 24 h. XJB was added 30 min before rotenone or H2O2 administration, and the culture medium was removed for LDH at 24 h after oxidative stress. XJB-5-131 reversed rotenone or H2O2-induced CL loss (C, F), mitochondrial dysfunction (D, G), and neuronal death (E, H). *P < 0.05, **P < 0.01 (One-way ANOVA, Tukey post hoc test, n = 6/group). Data represent the mean ± s.e.m. from 3 independent experiments. IP Effects of CL synthase (CLS) siRNA on primary spinal neuronal death in vitro. Cultured spinal cord neurons were transfected with CLS1 siRNA (r) for 24 h. I CL content was assayed using acridine orange 10-nonyl bromide (NAO, Invitrogen). Knocking down CL synthase, the key enzyme of de novo CL biosynthesis, with CLS1 siRNA significantly decreased CL. JO Activated caspase-3/7 was examined using CellEvent™ Caspase-3/7 Green Detection Reagent (Invitrogen). Apoptotic cells with activated caspase-3/7 showed bright green nuclei (arrows). Bar = 100 μm. P Bar graph showed that CLS1 siRNA significantly induced neuronal apoptosis, evidenced by increased number of activated caspase-3/7 cells. **P < 0.01 (Student t-test, n = 6/group). Data represent the mean ± s.e.m. from 3 independent experiments. QS Effects of exogenous CL on mitochondrial dysfunction and spinal neuronal death in the scratch injury model in vitro. Cultured spinal cord neurons were pre-incubated with CL liposomes for 30 min before scratch injury. Q Scratch injury-induced neuronal death was significantly reversed by exogenous CL in a dose-dependent manner. R, S Exogenous CL (100 µM) also significantly reversed scratch injury-induced CL loss (R) and decreased mitochondrial membrane potential (MMP) (S). Veh, vehicle; *P < 0.05, **P < 0.01 (One-way ANOVA, Tukey’s multiple comparisons test, n = 6/group). Data represent the mean ± s.e.m. from 3 independent experiments.

Exogenous cardiolipin protects spinal neurons from mechanically-induced cell death in vitro

To further explore the role of CL alteration in SCI, we first investigated whether exogenous CL reduces mechanical injury-induced neuronal death in an in vitro SCI model of mechanical scratch injury in rat primary spinal cord neurons. This model is useful for investigating the effect of mechanical damage on neurons, such as what happened in a SCI, and for testing the neuroprotective effects of certain agents on mechanically injured neurons [24]. Our results showed that pre-treatment with CL liposomes reduced scratch-induced neuronal death in a dose-dependent manner (Fig. 3Q). At 24 h after the scratch injury, mitochondria were isolated from cultured spinal cord neurons for CL and mitochondrial function analysis. Our results showed that scratch injury induced CL loss (Fig. 3R) and reduced mitochondrial membrane potential (MMP) (Δψm) (Fig. 3S) in cultured spinal cord neurons. Importantly, such effects were significantly reversed by the administration of 100 µM exogenous CL (Fig. 3R, S).

Oxidative stress induces mitochondrial cytosolic phospholipase A2 (cPLA2) activation, which mediates cardiolipin loss, leading to mitochondrial dysfunction and neuronal death after SCI

CL, a mitochondrial phospholipid, is a substrate of phospholipase A2 (PLA2) [13, 25, 26]. In physiological conditions, PLA2 hydrolysis is preferentially limited to oxidized acyl chains, including those of oxidized CL, and this is helpful for the overall membrane physiology within mitochondria [27]. However, when PLA2 is overactivated, such as in pathological conditions, the enzymatic activities are therefore elevated and even non-oxidized CL are subject to degradation by the enhanced hydrolysis. cPLA2 is the most important PLA2 isoform [9, 28]. To explore whether cPLA2 was involved in SCI-induced CL loss, we first isolated the mitochondrial fraction from the injured spinal cord in a rat contusive SCI model using a Focus Subcell fraction kit. Western blot analysis showed that cPLA2 expression and activation were increased in the mitochondrial fraction after SCI and peaked at 3 days (Fig. 4A). An immuno-electron microscopic image showed the localization of phosphorylated cPLA2 (p-cPLA2)-immunoreactivity in both outer and inner mitochondrial membranes (Fig. 4B). These findings suggest that mitochondrial cPLA2 may play a role in mediating CL loss. Our previous report showed that a ROS and radical inducer H2O2 induced cPLA2 activation in cultured spinal cord neurons [29]. We also found that microinjections of H2O2 at two doses (15 and 30 μg) into the normal rat spinal cord at T9 resulted in a marked increase in the expression of p-cPLA2, an activated form of cPLA2, in a dose-dependent manner (Fig. 4I). We further found that SCI-induced mitochondrial cPLA2 activation was effectively reversed by the novel ROS and radical scavenger, XJB-5-131 (Fig. 4J–M). These findings suggest that mitochondrial oxidative stress can trigger cPLA2 activation. To further confirm the role of cPLA2 in CL loss after SCI, we determined whether blocking cPLA2 reduced CL loss. Our in vitro experiments showed cPLA2 activation by a cPLA2 activator ceramide-1-phosphate (C-1-P) induced CL loss (Fig. 5A), which was substantially reversed by AACOCF3, a cPLA2 inhibitor (Fig. 5A). Remarkably, blocking cPLA2 pharmacologically with AACOCF3 at 30 min postinjury in rats or genetically deleting cPLA2 in mice significantly reduced CL loss after SCI (Fig. 4C, Fig. 5L). These findings suggest that mitochondrial cPLA2 may play a role in mediating CL loss. To further assess whether activation of cPLA2 mediates CL alteration-induced cell death after SCI, we determined whether blocking cPLA2 activation with the cPLA2 inhibitor AACOCF3 would reduce injury-induced CL loss, resulting in improved mitochondrial function and reduced neuronal death after a contusive SCI in adult rats. Our results showed that cPLA2 activation by both cPLA2 direct and indirect activators C-1-P and A23187, respectively, induced mitochondrial dysfunction and neuronal death (Fig. 5C–K). Such C-1-P- or A23187-induced mitochondrial dysfunction and neuronal death were significantly reversed by AACOCF3, a cPLA2 inhibitor (Fig. 5K). Importantly, pre-treatment with CL liposomes reduced C-1-P-induced neuronal death in a dose-dependent manner (Fig. 5B), suggesting that CL loss mediates cPLA2 activation-induced neuronal death. Most importantly, both pharmacological blockade in rats and genetic deletion of cPLA2 in mice significantly reduced mitochondria dysfunction (Figs. 4D, 5M), cytochrome c release (Figs. 4E, 5N), Smac/DIABLO release (Fig. 4F), active caspase-3 expression (Figs. 4G, 5O), and cleaved PARP expression (Fig. 4H) after SCI. These findings strongly suggest that cPLA2 mediates CL alteration-induced cell death and that such cell death is mediated through a mitochondrial apoptotic cascade.

Fig. 4: Oxidative stress induces cPLA2 activation and blockade of cytosolic phospholipase A2 (cPLA2) with AACOCF3 reduced cardiolipin (CL) loss, mitochondrial dysfunction, and apoptosis after spinal cord injury (SCI).
figure 4

A Time course of mitochondrial cPLA2 expression and activation after SCI. Data represent mean ± s.e.m., *P < 0.05, **P < 0.01 vs sham (Two-way ANOVA, Tukey’s multiple comparisons test, n = 6 rats/group). B An electron microscopic image shows phosphorylated cPLA2 (p-cPLA2) immunopositive peroxidase reaction products in a neuronal dendrite in the rat spinal ventral horn. p-cPLA2 immunoreactivity (IR) was observed to localize in mitochondria (*) including both the outer (yellow arrow) and inner (red arrows) mitochondrial membranes, and on the cell membrane (green arrow) at 3 days after SCI. Bar = 0.25 μm. C cPLA2 activation-induced CL loss was reversed by AACOCF3, a cPLA2 inhibitor. D SCI induced a decrease in the ratio of red/green in the MMP (Δψm) assay, which was reversed by AACOCF3. (EH) AACOCF3, a cPLA2 inhibitor, reversed SCI-induced cytochrome c release (E), Smac/DIABLO release (F), active caspase-3 expression (G), and cleaved PAPRP expression (H). Data represent mean ± s.e.m. *P < 0.05, **P < 0.01 (One-way ANOVA, Tukey’s multiple comparisons test, n = 6 rats/group). Cyt. c cytochrome c, Smac/D. Smac/DIABLO, A. casp-3 active caspase-3. IM Oxidative stress induces cPLA2 activation after SCI. I Immunohistochemistry shows a marked increase in p-cPLA2-expression, a marker of cPLA2 activation, at the injury epicenter and 3 mm rostral to it after a single injection of H2O2 into the T9 spinal cord. Right column images show that H2O2-induced p-cPLA2 expression was substantially reversed by mepacrine (5 mg/kg), a PLA2 inhibitor. The expression of p-cPLA2 was found in neurons (arrows), swelling axons (open arrows), and glial cells (arrowheads). Bar, 100 μm. IL Administration of XJB (10 mg/kg, i.p.) at 30 min post-injury significantly reduced SCI-induced mitochondrial cPLA2 activation. J Representative images of phosphorylated cPLA2 (p-cPLA2), cPLA2, and VDAC expression. K Compiled results in a bar graph for the ratio of p-cPLA2/cPLA2 expression. L Compiled results in a bar graph for the ratio of p-cPLA2/VDAC expression. M Compiled results in a bar graph for the ratio of cPLA2/VDAC expression. *P < 0.05, **P < 0.01 (One-way ANOVA, Tukey’s multiple comparisons test, n = 6). Data represent the mean ± s.e.m.

Fig. 5: Cytosolic phospholipase A2 (cPLA2) activation induces cardiolipin (CL) loss, leading to mitochondrial dysfunction and neuronal death.
figure 5

A cPLA2 inhibitor AACOCF3 revised cPLA2 activation-induced CL loss. **P < 0.01 (One-way ANOVA, Tukey’s multiple comparisons test, n = 4–6). Data represent the mean ± s.e.m. from 3 independent culture experiments. B CL reduced cPLA2 activation-induced neuronal death. CL liposomes were added 30 min before C-1-P (2 µM) treatment. **P < 0.01; ns no significance. (One-way ANOVA, Tukey’s multiple comparisons test, n = 6). Data represent the mean ± s.e.m. from 3 independent culture experiments. CH C-1-P induced mitochondrial membrane potential (MMP, Δψm) change measured with the cationic dye JC-1 in cultured spinal neurons. Vehicle-treated neurons showed strong J-aggregation (red). After C-1-P treatment, the majority of neurons showed green staining due to low Δψm. Bar, 50 µm. I Bar graph shows C-1-P induced a significant decrease in the ratio of red/green, indicating that activation of cPLA2 induced mitochondrial dysfunction (**P < 0.01, Student t test, n = 3). J Cultured spinal cord neurons were treated with the designated concentrations of C-1-P or A23187 for 24 h. MTT assay revealed that both cPLA2 activators, C-1-P and A23187, induced mitochondrial dysfunction and neuronal death in a dose-dependent manner. **P < 0.01, ##P < 0.01 versus the vehicle control (One-way ANOVA, Tukey’s multiple comparisons test, n = 6–8). Data represent the mean ± s.e.m. from 3 independent experiments. K Importantly, mitochondrial dysfunction and neuronal death induced by C-1-P (2 µM) or A23187 (5 µM) were significantly reversed by AACOCF3 (15 µM), a cPLA2 inhibitor. AACOCF3 was added 30 min before C-1-P or A23187 administration, and the culture cells were examined for MTT at 24 h after the activator treatment. **P < 0.01 versus the vehicle control, ##P < 0.01 versus the C-1-P or A23187 group (Two-way ANOVA, Tukey’s multiple comparisons test, n = 7–8. Data represent the mean ± s.e.m. from 3 independent experiments. LO cPLA2 ablation protected against CL loss (L), mitochondrial dysfunction (M), and apoptosis (NO) induced by SCI. L NAO-labeled cardiolipin expression at 1 day after SCI. M A ratio of red/green, indicating mitochondrial function. N Cytochrome c release at 1 day after SCI. O Expression of active caspase-3 at 1 day after SCI. Data represent mean ± s.e.m. *P < 0.05, **P < 0.01 vs sham; ##P < 0.01 vs WT (Two-way ANOVA, Tukey’s multiple comparisons test, n = 6 mice/group).

XJB-5-131 attenuates SCI-induced cardiolipin peroxidation, mitochondria dysfunction, and apoptotic neuronal death after SCI

Mitochondrial oxidative stress not only induced CL peroxidation, but also stimulated cPLA2 activation. XJB-5-131 is a novel mitochondria-targeted ROS, electron, and radical scavenger which can cross the BBB and prevent the peroxidation of CL [18, 30,31,Full size image

Female animals are routinely used in SCI studies because female animals allow for easier manual expression of bladders after SCI, less urinary tract infection, and less mortality [34,35,36,37]. In addition, we and others have shown that no significant differences were detected in histological and behavioral outcomes between male and female animals after SCI [38,39,40]. Therefore, female animals were used in this study.

Cardiolipin alteration occurs at an early stage after SCI triggering an apoptotic cascade

A decrease in the content of mitochondrial CL is the most frequently reported pathological alteration of the CL profile [13]. In our study, increases of biomarkers of apoptosis (cytochrome c release, Smac/DIABLO release, and active caspase-3 expression) at the early stage of SCI suggested an early CL alteration post-SCI. CL alteration has been reported to cause the release of cytochrome c and Smac/DIABLO and to activate caspase-3 during induction of apoptosis [16, 20, 21]. In our SCI model, the release of cytochrome c and Smac/DIABLO preceded active caspase-3 expression, which was consistent with a previous report that CL oxidation preceded caspase-3 activation after traumatic brain injury [17]. The decrease of CL at 3 and 24 h after SCI, shown by the lipidomic analysis, further confirmed that CL alteration is an early event after SCI. A corresponding increase in lyso-CL at only 24 h post-injury indicated that SCI induced CL degradation at 24 h after SCI. Decreases in CL content are often used as an indicator of CL oxidation because CL oxidation may lead to an overall loss of detectable CL content, either by preferential hydrolysis of peroxidized acyl chains by PLA2, direct decomposition of lipid peroxides, or the formation of CL-protein complexes that would no longer be detected as phospholipids [13]. Lipid peroxidation increased at 3 and 24 h after SCI (Fig. 1F), suggesting that CL peroxidation may occur at 3 and 24 h after SCI. After SCI, 50% of CL species were significantly reduced and the reduced CL species contained mainly polyunsaturated fatty acids that are highly susceptible to peroxidation. Additionally, 4-HNE, a lipid peroxidation marker, also increased after SCI. CL is a likely early target of ROS attack in mitochondria due not only to its high content of unsaturated fatty acids, but also to its location in the IMM near the sites of major ROS production [41]. A change in the oxidative state or content of CL has been proposed to trigger the mitochondrial switch from ATP generation to apoptosis initiation [13, 14, 42]. Thus, these results suggest that SCI induces early CL peroxidation, degradation, and loss, triggering an apoptotic cascade.

Cardiolipin loss is mediated by phospholipase A2

Although CL content was significantly decreased after SCI, the mechanism(s) by which it was decreased remains unclear. Under normal conditions, PLA2 preferentially hydrolyzes oxidized phospholipids including oxidized CL to maintain mitochondrial membrane stability and function [27]. Under pathological conditions, over-activated PLA2 can directly induce CL loss through enhanced hydrolysis and degradation of CL, resulting in mitochondrial dysfunction. This can trigger an apoptotic signaling cascade, eventually leading to cell death and tissue damage. The phospholipases A2 can be classified into three major enzyme families: cytosolic PLA2 (cPLA2), secretory PLA2 (sPLA2), and Ca2+-independent PLA2 (iPLA2) [3, 9, 43]. Mammalian phospholipases, including cPLA2, sPLA2, and iPLA2, are reported to have CL-hydrolyzing activities [25, 26], and several studies suggest that iPLA2 could be a candidate catalyst capable of hydrolyzing CL in mitochondria [44,45,46,47,48,49]. However, which PLA2 subtype is involved in CL hydrolysis after SCI, or its preference for peroxidized CL, remains to be elucidated. Cytosolic PLA2 is the most important PLA2 isoform because it has been implicated as an effector in the receptor-mediated release of AA and exhibits a strong preference for deacylation of AA over other fatty acids [4, 50]. Our in vitro experiments showed that activated cPLA2 induced CL loss, leading to mitochondrial dysfunction and neuronal death. Such effects, however, were substantially reversed by AACOCF3, a cPLA2 inhibitor. Our in vivo experiments further showed that the expression and activity of cPLA2 significantly increased in the mitochondria following SCI. Immuno-EM analysis revealed that activated cPLA2 was expressed in the mitochondria. Remarkably, blocking cPLA2 pharmacologically with AACOCF3 reduced CL loss, and alleviated mitochondrial dysfunction and neural death after SCI. Furthermore, genetic deletion of cPLA2 reduced CL loss, resulting in neuroprotection after SCI. Taken together, these findings suggest that SCI-induced CL loss, at least part, is mediated by cPLA2 activation.

Cardiolipin alteration results in mitochondrial dysfunction and neuronal death

CL is a mitochondrial-specific phospholipid localized in the IMM where it is required for optimal mitochondrial function [13]. It has been reported that CL is the only phospholipid in mitochondria that undergoes early oxidation after injury [17] or during apoptosis [16]. The oxidation is catalyzed by a CL-specific peroxidase activity of CL-bound cytochrome c [16]. Furthermore, oxidized CL is required for the release of proapoptotic factors [16]. Our results showed that decreasing CL by knocking down the CL synthase (CLS1) induced neuronal apoptosis, and pre-treatment with CL liposomes reduced scratch-induced mitochondrial dysfunction and neuronal death, suggesting that CL alteration may mediate mitochondrial dysfunction and neuronal death after SCI. Our in vivo experiments further showed that pharmacological blocking or genetic deletion of cPLA2 in mice attenuated CL loss, leading to improvement of mitochondrial function and neuronal survival. One mechanism of CL alteration-induced neuronal death could be its interaction with cytochrome c. CL electrostatically anchors cytochrome c to the IMM and plays an important regulatory role in cytochrome c release, which triggers the downstream events in apoptosis [13, 16, 30]. CL peroxidation and/or loss weakens its interaction with cytochrome c, which is then released and escapes to the cytosol via the mitochondria transition pore formed by the proapoptotic Bcl-2 family proteins, triggering apoptosis [13, 16, 30].

Pharmacological inhibition of CL alteration results in neuroprotection and functional recovery after SCI

A significant finding of this study is that pharmacological inhibition of CL alteration with XJB-5-131 reduced mitochondrial dysfunction, neuronal death, and tissue damage, and promoted functional recovery in adult rats after SCI. Our results showed that XJB-5-131 not only inhibited CL peroxidation after SCI, but also reduced CL loss by inhibition of cPLA2 activation after SCI. Several studies show that CL content can be preserved when ROS is reduced in ischemic heart and skeletal muscle [51,52,53,54]. Apoptosis has been considered as a key mechanism of cell death following SCI [55,56,57]. Notably, administration of XJB-5-131 at 30 min post-injury significantly reduced SCI-induced biomarkers of apoptosis including cytochrome c release, Smac/DIABLO release, and active caspase-3 expression. Importantly, our post-treatment results showed a long beneficial effect on anatomical and functional improvements. In agreement with our observation, XJB-5-131 inhibited traumatic brain injury (TBI)-induced CL alteration, had potent neuroprotective activity in vitro, reduced cortical lesion volume, and ameliorated behavioral deficits after TBI [18]. XJB-5-131 also reduced oxidative DNA damage, improved mitochondrial function, enhanced neuronal survival, and suppressed motor decline in a mouse model of Huntington’s Disease [58]. Our results suggest that CL alteration contributes to the pathogenesis of SCI and that targeting the CL alteration could be a promising therapeutic strategy for intervention after SCI.

In conclusion, we demonstrated that SCI induced significant CL alteration (peroxidation and loss) at an early stage of SCI, and that such alterations induced mitochondrial dysfunction and neuronal death, ultimately, leading to tissue damage and functional deficits. Notably, the mitochondrial-target ROS scavenger XJB-5-131 treatment reduced CL peroxidation and loss, leading to decreases of mitochondrial dysfunction and neuronal apoptosis after SCI. Remarkably, pharmacologic inhibition of CL alteration with XJB-5-131 reduced tissue damage and ameliorated behavioral deficits after SCI in rats. These findings suggest that CL alteration could be a novel mechanism that mediates injury-induced neuronal death, and therefore represents a potential therapeutic target for ameliorating secondary SCI.