IMR Press / JIN / Volume 19 / Issue 4 / DOI: 10.31083/j.jin.2020.04.255
Open Access Original Research
Cryogen spray cooling mitigates inflammation and injury-induced CISD2 decline in rat spinal cord hemisection model
Show Less
1 Department of Exercise and Health Promotion, College of Kinesiology and Health, Chinese Culture University, 111, Taipei, Taiwan
2 Department of Plastic Surgery, Taipei Medical University Hospital, Taipei Medical University, 110, Taipei, Taiwan
3 Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, 110, Taipei, Taiwan
4 Graduate Institute of Biomedical Optomechatronics, College of Biomedical Engineering, Taipei Medical University, 110, Taipei, Taiwan
5 Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital, Chang Gung University, 333, Taipei, Taiwan
6 Division of Neurosurgery, Department of Surgery, Kuang Tien General Hospital, 433, Taichung, Taiwan
7 Department of Biotechnology and Animal Science, College of Bioresources, National Ilan University, 260, Yilan, Taiwan
8 Department of Biotechnology, College of Medical and Health Care, Hung Kuang University, 433, Taichung, Taiwan
9 Department of Health Business Administration, College of Medical and Health Care, Hung Kuang University, 433, Taichung, Taiwan
*Correspondence: neurosurgery2005@yahoo.com.tw (Muh-Shi Lin)
J. Integr. Neurosci. 2020, 19(4), 619–628; https://doi.org/10.31083/j.jin.2020.04.255
Submitted: 24 August 2020 | Revised: 4 December 2020 | Accepted: 4 December 2020 | Published: 30 December 2020
Copyright: © 2020 Kung et al. Published by IMR Press.
This is an open access article under the CC BY 4.0 license ( https://creativecommons.org/licenses/by/4.0/).
Abstract

Therapeutic strategies for traumatic spinal cord injury generally involve rectifying concomitant destruction to the spinal cord from inflammation, mitochondrial dysfunction, and eventual neuronal apoptosis. Elevating the expression of spinal cord injury-attenuated CDGSH iron-sulfur domain-2 has been shown to mitigate the pathologies above. In the current work, hypothermia was induced via continuous cryogen spray cooling in a rat spinal cord hemisection model. Spinal cord injury was shown to elevate the mRNA expression of proinflammatory mediators, including NF κ B, iNOS, TNF- α , and regulated upon activation, normal T-cell expressed and secreted as well as lower CDGSH iron-sulfur domain-2 expression. Cryogen spray cooling treatment was shown to attenuate inflammatory reactions and elevate CDGSH iron-sulfur domain-2 expression. Immunohistochemical analysis of the glial fibrillary acidic protein, caspase-3 and NeuN in spinal cord injured rats that underwent cryogen spray cooling treatment revealed notable reductions in injury-induced astrocytic activation, apoptosis, neuronal loss, and decline in CDGSH iron-sulfur domain-2 expression. These results demonstrate the CDGSH iron-sulfur domain-2 preserving effects of cryogen spray cooling, which could contribute to the prevention of astrocytic activation, astrocyte-mediated neuroinflammation, apoptosis, and neuron loss.

Keywords
Cryogen spray cooling
hypothermia
CISD2
inflammatory response
astrocyte activation
apoptosis
neuronal loss
spinal cord injury
1. Introduction

The incidence of acute spinal cord injury (SCI) in the general population ranges from 15-40 cases per million worldwide. The pathophysiology of acute SCI initially involves significant insult to the spinal cord, resulting in immediate structural damage, including cell membrane rupture, myelin and axon damage, and microvascular destruction. Secondary injuries include profound proinflammatory responses, excitotoxicity, hyperoxidation, mitochondrial dysfunction, and eventual apoptotic cell death (Houle and Tessler, 2003; Bethea and Dietrich, 2002). Several molecular mechanisms have been implicated in SCI’s pathogenesis, including hypoxia, ischemia, lipid peroxidation, free radical production, neutral protease activation, prostaglandin production, and programmed cell death (Bethea and Dietrich, 2002; Hung et al., 2007; Tator and Fehlings, 1991). Therapeutic strategies targeting secondary damage focus on reducing neuronal loss.

Amino acid CDGSH iron sulfur domain 2 (CISD2) plays an important role in calcium metabolism (Shen et al., 2017) as well as anti-apoptotic (Chang et al., 2010) and anti-inflammatory activities (Lin, 2020; Kung et al., 2020). CISD2 helps preserve the mitochondrial membrane’s integrity and thereby prevents mitochondrial malfunction and ultimate cell death. CISD2 depletion has been linked to mitochondrial breakdown and dysfunction accompanied by autophagic cell death in CISD2 knockout mice (Chen et al., 2009). At the endoplasmic reticulum, CISD2 has been shown to attenuate calcium-mediated excitotoxicity through the binding of CISD2 to BCL2 and the inositol 1, 4, 5-triphosphate receptor (Chang et al., 2012). CISD2 has also been shown to enhance the combination of BCL2 and BECN1, which regulates cellular autophagy/apoptosis (Kang et al., 2011). CISD2 deficiency has been linked to an increase in iNOS and a reduction in BCL2 levels in SH-SY5Y cells challenged with lipopolysaccharide (LPS) (Lin et al., 2017). CISD2 deficiency has also been linked to augmented nuclear translocation of the NF κ B (nuclear factor κ B) p65 subunit and a shift of the M2 phenotype toward M1 microglia-associated inflammation in EOC microglia (Lin, 2020). Its reduction has even been linked to a decrease in PPAR- β expression in SH-SY5Y cells, indicating enhanced degradation of I κ B (an NF- κ B inhibitor) and consequent NF κ B activation (Kung et al., 2020). In mice, CISD2 expression levels have been shown to decline as a function of aging (Lin et al., 2019) or spinal cord hemisection injury (Lin et al., 2017). In neural cell models involving CISD2 knockdown, CISD2 deficiency has been shown to augment inflammatory responses and mitochondrial dysfunction, as indicated by low DeltaPsi (m) levels, high reactive oxygen species (ROS) levels, and increased cellular apoptosis (Lin et al., 2019). Therapeutic strategies aimed at elevating CISD2 levels through curcumin administration (Lin et al., 2017, 2019) or wild bitter melon (Kung et al., 2020) have also been shown to mitigate the pathologies above.

Therapeutic hypothermia aimed at halting tumor progression was first reported in 1945 (Kwon et al., 2008). Over the last half-century, therapeutic hypothermia has been evaluated for its neuroprotective effects on the brain and spinal cord in situations such as ischemic or traumatic brain injury (TBI) (Bernard and Buist, 2003; Lo Jr et al., 2009; Hansebout et al., 1984; Yu et al., 2000). Therapeutic hypothermia can be managed systemically or locally. Systemic treatment involves the cooling of external surfaces or internally via the endovascular system. Local hypothermia avoids the side effects of systemic hypothermia while allowing a more pronounced reduction in temperature at the injury site (Jordan and Carhuapoma, 2007). Researchers have demonstrated therapeutic hypothermia’s efficacy in attenuating secondary injury by reducing neuroinflammation and cellular apoptosis in ischemic brain injury (Ohmura et al., 2005). Therapeutic hypothermia has been shown to preserve damaged tissue and promote impaired locomotor function in experiments on SCI rats and SCI patients undergoing systemic hypothermia (Lo Jr et al., 2009; Yu et al., 2000) or local spinal cord cooling (Hansebout et al., 1984).

In 1994, Nelson et al. (1995) introduced cryogen spray cooling (CSC) to induce local hypothermia during the spatially selective photocoagulation of biological tissue. This method involves spraying cryogen on the skin’s surface for a short duration, such that the effects of cooling remain localized in the epidermis and the temperature of deeper blood vessels remains unaffected (Waldorf et al., 1997; Nelson et al., 1996). As mentioned above, neural injuries can lead to CISD2 downregulation, resulting in exaggerated inflammatory reactions. In the current work, we sought to verify the neuroprotective (anti-inflammatory and CISD2-preserving) effects of therapeutic hypothermia induced through the continuous delivery of CSC in a rat spinal cord hemisection model.

2. Materials and methods
2.1 Hemisection of the spinal cord in rats

Male Sprague-Dawley (SD) rats (Academia Sinica, Taipei, Taiwan) weighing 280-330 g were maintained at a density of two animals per cage for at least 5 days after they arrived at our laboratory. The rats were given ad libitum access to food and water while housed within a room with a 12 : 12 h light-dark cycle. All rats were treated as per the guidelines stipulated by the Experimental Animal Laboratory and approved by the Animal Care and Use Committee of Chang Gung University (IACUC Approval No.: CGU14-052).

The surgical procedure used in the lateral hemisection of the spinal cord in rats can be found in our previous studies (Lin et al., 2011, 2012). Briefly, isoflurane was delivered to the rats as general anesthesia, before the animals were fastened within a stereotaxic apparatus (David Kopf Instruments, Tujunga, CA). An adjustable wire knife was used to create a spinal cord lesion on the left side. Control animals were subjected to a sham operation (laminectomy without hemisection). Posterior decompression was achieved via laminectomy at the eleventh thoracic vertebrae using a fine diamond drill. The wire knife guide was positioned vertically adjacent to the lateral surface at the spinal cord’s lower thoracic level. This permitted analysis of the healthy cephalad and caudal segments of the spinal cord. The knife was turned medially and then extended 1.5 mm, at which point the guide was lifted by 4.0 mm to hemi-transect the spinal cord. Iridectomy scissors were then used to cut to ascertain the thoroughness of the hemisection. The wound was closed in layers.

2.2 Cryogen spray cooling for SCI animals

The rats were divided into three groups: (1) Vehicle-treated sham-operated control (sham; laminectomy only) (n = 6); (2) Vehicle-treated SCI group (SCI) (n = 6); (3) SCI rats post-treated with CSC treatment (SCI + CSC) (n = 6).

The cryogen is 1, 1, 1, 2-tetrafluoroethane, an FDA-approved replacement for Freon, biodegradable, non-toxic, and incombustible. Following spinal cord hemisection, cryogen spurts were immediately sprayed onto the injured spinal cord through an electrically powered nozzle from a distance of 3 cm above the dural surface. Coolant was delivered continuously in spurts of 20 milliseconds at intervals of 5 spurts per minute for 10 minutes. CSC treatment lowered the surface temperature in the treatment region to -10 C. The surface temperature returned to normal within roughly 500 milliseconds after CSC treatment (Anvari et al., 1996; Smithies et al., 1998). Note that spraying was precisely regulated using a programmable digital delay generator. A schematic diagram showing the SCI model and CSC treatment are presented in Fig. 1A.

Fig. 1.

CSC reduced injury-induced inflammation following spinal cord hemisection in rats. (A) Schematic illustration of the CSC technique applied to tissue adjacent to the spinal cord hemisection under the following conditions: (i) Sham controls; (ii) SCI rats undergoing SCI; (iii) SCI rats undergoing SCI followed by CSC (SCI + CSC); mRNA expression levels of (B) NF κ B P105, (C) TNF- α , (D) iNOS, and (E) RANTES. Vertical bars indicate the mean ( ± SEM) mRNA expression (n = 3 in each group). *, P < 0.05 and ***, P < 0.001 indicate the level of statistical significance.

At 24 h post-treatment, all of the animals in all groups underwent deep anesthesia via isoflurane. The animals were then sacrificed, and the spinal cord tissue was removed to investigate mRNA expression (n = 3 in each group) and perform immunohistochemical analysis (n = 3 in each group).

2.3 Reverse transcription-polymerase chain reaction (RT-PCR) and real-time quantitative reverse-transcription polymerase chain reaction (qRT-PCR)

Total RNA was prepared through the direct lysis of animal tissue using TRIzol reagent (Invitrogen, Carlsbad, CA, USA). Then, reverse transcribing the mRNA into cDNA using oligo-dT, and SuperScript II reverse transcriptase (Invitrogen, Carlsbad, CA, USA). Quantitative RT-PCR was performed using the ABI StepOne sequence detector system (Applied Biosystems, Foster City, CA, USA) in accordance with the SYBR Green methodology, normalized to the expression of a housekeeping gene (GAPDH), as previously described (Lee et al., 2015). The primer sets and product size of each cDNA of interest were as follows: rat CISD2 (Gene ID: 295457, accession number: NM_001191608, 5’- CCCTTCCTTGGTGTACTTGCA -3’ and 5’- TTTCGTTCACCACTTTGGGATT -3’, 129 bp); NF κ B P105 (Gene ID: 81736, accession number: NM_001276711, 5’- GGGCCTGCAAAGGTTATCGT -3’ and 5’- TGCCGTCCTCACAGTGCTT -3’, 128 bp); TNF- α (Gene ID: 24835, accession number: NM_012675, 5’- GACGTGGAACTGGCAGAAGAG -3’ and 5’- AGGAATGAGAAGAGGCTGAGACA -3’, 147 bp); iNOS (Gene ID: 24599, accession number: NM_012611, 5’- GTCTTTGACGCTCGGAACTGT -3’ and 5’- CGACCTGATGTTGCCATTGT -3’, 101 bp); RANTES (Regulated on Activation, Normal T Cell Expressed and Secreted) (Gene ID: 81780, accession number: NM_031116, 5’- AACCCCCTACTCCCACTCATTC -3’ and 5’- GGCCATAGGAGAGGACACAGTT -3’, 106 bp); and GAPDH (Gene ID: 24383, accession number: NM_017008, 5’- CGTGTTCCTACCCCCAATGT -3’ and 5’- CCTGCTTCACCACCTTCTTGAT -3’, 103 bp).

2.4 Immunohistochemical staining

On the second day after SCI, the animals underwent deep anesthesia via isoflurane before being sacrificed. They were then thoroughly perfused with phosphate-buffered saline (PBS), followed by cold 4% paraformaldehyde in 0.15 M sodium phosphate buffer. The spinal cords were removed immediately, fixed at 4 C for 8 h, and cryoprotected in 15% and 30% sucrose for 3 days. Tissue from the spinal cords was frozen (-80 C) before analysis. Sections (5 μ m in thickness) were cut from the center of the spinal cord hemisection on a frozen stage using a sliding microtome. The sections were then washed in PBS and incubated in 3% normal goat serum with 0.3% Triton X-100 in PBS for 1 h. The free-floating sections were incubated at 4 C with the following antibodies: Anti-GFAP (Catalog: 16825-1-AP, Proteintech, Chicago, USA); Anti-caspase-3 (Catalog: 19677-1-AP, Proteintech, Chicago, IL, USA); Anti-NeuN (clone A60, Catalog: MAB377, Merk, Taipei City, Taiwan); Anti-CISD2 (Catalog: 13318-1-AP, Proteintech, Chicago, IL, USA). Immunoreactivity was visualized using the Vectastain Elite ABC Peroxidase method (Vector Laboratories, Burlingame, CA, USA) with diaminobenzidine (DAB) as the chromagen. The morphological and quantitative analysis focused on injured grey and white matter adjacent to the spinal cord hemisection (epicenter), which is indicated as the region of interest (ROI) within the dashed box in Fig. 3A. Cell counting was performed at a magnification of 400 × on every sixth section stained using the antibodies mentioned above. Only cells presenting visible indications of staining were counted. All data are presented as mean ± SEM of three consecutive measurements.

2.5 Statistical analyses

Independent two-sample t-tests were used to compare data between the experimental groups. One-way analysis of variance (ANOVA) was used to determine whether there were significant differences between the means of three or more independent (unrelated) groups. In cases where the differences were apparent, multiple comparisons were performed using the Newman-Keuls method. Data are presented as mean ± SEM. All statistical assessments were two-sided and evaluated at a 0.05 level of significance. Statistical analysis was performed using GraphPad Prism software version 5.0 (GraphPad Software, Inc; La Jolla, CA).

3. Results
3.1 CSC treatment attenuated the expression of proinflammatory mediators in mRNA levels at 24 h after SCI

In the current spinal cord hemisection model, rats were subjected to CSC treatment involving short spurts (20 milliseconds) at intervals of 5 spurts per min for 10 min, or untreated with CSC (Fig. 1A). qRT-PCR was used to examine the mRNA expression of proinflammatory mediators at the site of SCI. Compared to the sham control group, animals in the SCI only group presented higher expression levels of NF κ B (P105), TNF- α , iNOS, and RANTES (*P < 0.05, Fig. 1B; *P < 0.05, Fig. 1C; ***P < 0.001, Fig. 1D; and *P < 0.05, Fig. 1E, respectively). Compared to the SCI only group, animals in the SCI + CSC group presented significantly lower mRNA expression levels of NF κ B (P105), TNF- α , iNOS, and RANTES (*P < 0.05, Fig. 1B; *P < 0.05, Fig. 1C; *P < 0.05, Fig. 1D; and *P < 0.05, Fig. 1E, respectively). Overall, these findings indicate that CSC treatment had an anti-inflammatory effect on rats previously subjected to SCI.

3.2 CSC treatment upregulated the expression of CISD2 mRNA at 24 h after SCI

The qRT-PCR analysis revealed that CISD2 expression levels were significantly lower in the SCI only group than in the sham control group (P < 0.05, Fig. 2). CISD2 expression levels were considerably higher in the SCI only group than in the SCI + CSC group (P < 0.05). In a previous paper, we reported on the anti-inflammatory effects of CISD2 (Lin et al., 2017). In the present work, CSC treatment was shown to attenuate the injury-induced pro-inflammatory response (i.e., SCI-induced reduction in CISD2 expression).

Fig. 2.

Spinal cord hemisection produced a corresponding decrease in CISD2 expression. CSC mediated the injury-induced decrease in CISD2 expression. Vertical bars indicate the mean ( ± SEM) mRNA expression (n = 3 in each group). *, P < 0.05 and ***, P < 0.001 indicate the level of statistical significance.

3.3 CSC treatment attenuated GFAP expression following SCI

Astrocytes can be stimulated to release various pro-inflammatory mediators in response to SCI, as indicated by GFAP over-expression. Immunostaining for GFAP (arrow in Fig. 3C) was more robust in the SCI only group than in the sham control group (Fig. 3B) (***P < 0.001, Fig. 3E). Immunostaining for GFAP was lower in the SCI + CSC treatment (indicated by an arrow in Fig. 3D) than in the SCI only group (**P < 0.01, Fig. 3E). These results indicate that CSC therapy mediated the activation of astrocytes and astrocyte-mediated proinflammatory responses following SCI.

Fig. 3.

Representative photographs of GFAP-stained sections of tissue samples were obtained from the spinal cords of rats in the sham control group (B), SCI (C), and SCI + CSC (D) after SCI. (A) Schematic depiction of ROI (labeled as the dashed box) in injured spinal cords for all groups. Staining of GFAP-positive cells was weaker in the sham group (B) than in the SCI group (C). CSC treatment decreased GFAP immunofluorescence. (A-C: magnification 400 × ). (E) Vertical bars indicate the mean ( ± SEM) number of GFAP-stained cells per tissue section in each group (n = 3). *, P < 0.05 and ***, P < 0.001 indicate the level of statistical significance.

3.4 CSC treatment could limit apoptosis in SCI animals

The blue arrows in Fig. 4A-C indicates the immunoreactivity of caspase-3 as a proxy of cellular apoptosis. Following spinal cord hemisection, immunostaining for caspase-3 was significantly higher in the SCI only group than in the sham control group (***P < 0.001, Fig. 4D). Immunostaining for caspase-3 was significantly lower in the SCI + CSC group than in the SCI only group (**P < 0.01, Fig. 4D), indicating that CSC attenuated cellular apoptosis.

Fig. 4.

Representative photographs of caspase-3-stained sections of tissue samples obtained from the spinal cords of rats in the (A) sham control group, (B) SCI only group, and (C) SCI + CSC group (A-C: 400 × ). The blue arrows in A-C indicate caspase-3-positive cells. (D) Vertical bars indicate the mean ( ± SEM) number of caspase-3-stained cells per tissue section in each group (n = 3). **, P < 0.01 and ***, P < 0.001 indicate the level of statistical significance.

3.5 CSC treatment could reduce neuronal damage following spinal cord hemisection

The blue arrows in Fig. 5A present the immunohistochemistry indicative of the neuron-specific marker NeuN in the sham group. As indicated by the blue arrows in Fig. 5B, the morphological integrity was far worse in the SCI only group than in the sham control group, and neuronal loss was also evident (***P < 0.001, Fig. 5D). The blue arrows in Fig. 5C indicate that NeuN staining was more preserved in the SCI + CSC group than in the SCI only group, and neuronal loss was less evident (*P < 0.05, Fig. 5D). Overall, the anti-inflammatory and anti-apoptotic effects of CSC could potentially help to prevent neuronal deficits.

Fig. 5.

Representative photographs of NeuN-stained sections of tissue samples obtained from the spinal cords of rats in the (A) sham control group, (B) SCI only group, and (C) SCI + CSC group (A-C: 400 × ). Compared to sham controls, the arrows in (A-C) indicate NeuN immunoreactivity (exhibited neuronal loss in SCI group). CSC treatment preserved neurons after SCI. (D) Vertical bars indicate the mean ( ± SEM) number of NeuN-stained cells per tissue section in each group (n = 3). *, P < 0.05 and ***, P < 0.001 indicate the level of statistical significance.

3.6 CSC treatment upregulated injury-attenuated CISD2 expression following spinal cord hemisection

As in our previous research, we determined in the current study that spinal injury led to a decline in CISD2 expression, as evidenced by a reduction in CISD2 immunoreactivity in the SCI only group (***P < 0.001, Fig. 6D). Far more CISD2-positive cells were observed in the sham group (blue arrows in Fig. 6A) than in injured tissue (showing signs of vacuolation) at the site of the injury in the SCI only group (dashed circle in Fig. 6B). The number of CISD2-positive cells in the same injured tissue in the SCI + CSC group (dashed circles in Fig. 6B and 6C) was far higher than in the SCI only group (*P < 0.05, Fig. 6D). Our results demonstrate the CISD2-preserving beneficial effects of CSC. The eventual anti-inflammatory property could contribute to the prevention of astrocytic activation, astrocyte-mediated neuroinflammation, apoptosis, and neuron loss.

Fig. 6.

SCI-induced a decrease in CISD2 expression in the injured spinal cord, the effect of which was attenuated by CSC. Representative photographs of CISD2-stained sections of tissue samples obtained from the spinal cords of rats in the (A) sham control group, (B) SCI only group, and (C) SCI + CSC group (A-C: 400 × ). (A) The sham group presented a high degree of CISD2 immunoreactivity. (B) CISD2 expression was lower in tissue samples with follicles from the hemisected spinal cord (indicated by a dashed circle). At the injury site of a similar situation in (C) (dashed circle), a densely packed pattern of CISD2 immunoreactivities proved the CISD2-elevating effect of CSC treatment in SCI rats, compared to (B). (D) Vertical bars indicate the mean ( ± SEM) number of CISD2-stained cells per tissue section in each group (n = 3). *, P < 0.05 and ***, P < 0.001 indicate the level of statistical significance.

4. Discussion

SCI can induce the activation of immunity-related cells in the spinal cord’s parenchyma and the activation of astrocytes and microglia (Hung et al., 2005; Davies et al., 1996). Reactivated astrocytes produce inflammatory cytokines and chemokines, which trigger microglial activation (favoring the detrimental M1 over the protective M2 phenotype) (Liu et al., 2011; Luo et al., 2002). Injury-activated astrocytes and microglia release considerable nitric oxide and ROS quantities, which amplify the inflammatory cascade (Li et al., 2007). Profound inflammatory responses can eventually lead to mitochondrial dysfunction and the enhanced production of ROS and nitric oxygen species (NOS) (Lucas et al., 2006; van Horssen et al., 2019), which can, in turn, contribute to neuronal apoptosis (Xu et al., 2006) and potential neurological deficits (Hurlbert, 2006). Thus, the therapeutic targeting of acute SCI can help to mitigate secondary insults to the spinal cord.

Therapeutic hypothermia has been shown to exert neuroprotective effects in the central nervous system (CNS) in response to injuries, such as TBI and SCI (Bernard and Buist, 2003). Animal studies have demonstrated that physiological ATP concentrations can be maintained by decreasing cellular consumption or increasing production through glycolysis, thereby preserving the CNS’s neurological function (Arrica and Bissonnette, 2007). In cases of hypoxic-ischemic brain insults, therapeutic hypothermia has been shown to attenuate secondary damage by decreasing proinflammatory responses, such as neutrophil and microglia stimulation. Therapeutic hypothermia has also been shown to decrease the generation of free radicals (which would otherwise lead to oxidative stress) and extracellular glutamate levels (which would otherwise lead to excitotoxicity) (Hachimi-Idrissi et al., 2004). Finally, therapeutic hypothermia has been shown to mediate secondary insults related to SCI, including the release of cytochrome-3 and the activation of caspase-3 and calpain (Ohmura et al., 2005).

One secondary injury related to SCI involves the hypertrophic formation of astrocytes and increased GFAP expression, which can, in turn, lead to glial scarring and a corresponding decrease in axonal and neuronal regeneration (Morino et al., 2003; Stichel and Müller, 1998). Glia-mediated neuroinflammation following acute SCI can lead to mitochondrial dysfunction, cellular apoptosis, and eventual neuron loss (Scholpa and Schnellmann, 2017). Ameliorating injury-induced astrocyte activation has been shown to help prevent pathogenesis in cases of SCI. In the current study, the injury-induced expression of GFAP, caspase-3, and NeuN indicates pathogenesis, including respective astrocytic proliferation, cellular apoptosis, and neuronal damage. We discovered that CSC-induced hypothermia helped attenuate secondary injuries following SCI, including astrocyte-associated inflammatory response, apoptosis, and neuronal deficit.

We found that CSC-induced hypothermia led to the upregulation of CISD2, even though acute SCI tends to downregulate the expression of CISD2. CSC’s anti-inflammatory effects can be attributed to the fact that CSC treatment attenuated the downregulation of CISD2 mRNA and proteins. In previous siRNA knockdown models aimed at decreasing CISD2 expression levels, we observed elevated inflammatory responses in SH-SY5Y neuronal-like cells (Lin et al., 2019) and EOC microglia (Lin, 2020). We also confirmed that CISD2 inhibits inflammation via the NF κ B pathway (Lin, 2020; Kung et al., 2020). NF κ B acts as a regulator of inflammation in mitochondria (Albensi, 2019; Laforge et al., 2016), and NF κ B signaling has been implicated in the link between mitochondrial dysfunction and acute SCI. CSC-driven CISD2 augmentation can help overcome the detrimental effects of NF κ B signaling, including excessive inflammation and potential apoptosis related to mitochondrial dysfunction. The above mechanisms help explain how CSC treatment attenuates or prevents inflammation, astrocytic activation, apoptosis, and neuronal loss in the tissue surrounding spinal cord injuries. Furthermore, this is the first study to report that CSC-induced therapeutic hypothermia regulates CISD2 expression.

The current study has several limitations, which must be considered in the interpretation of our findings. In the current study, we were limited to only three animals in each group. Many inflammation-related inflammatory factors, such as IL-4, were not addressed, and we did not collect samples at other time points. Subsequent in vivo studies will be required to assess CSC treatment’s long-term effects on upstream and downstream inflammatory signaling. Note that we did not perform CISD2 knockdown or knockout; therefore, a causal relationship cannot be determined between CISD2 and injury-induced inflammation following CSC.

Nonetheless, the anti-inflammatory effects of CISD2, including the antagonism of PPAR- β , NF- κ B, and injury-induced inflammatory mediators, have previously been established in CISD2 knockdown cell models (Lin, 2020; Kung et al., 2020). From this, it is safe to surmise that CSC’s effects on CISD2 limit the inflammatory response and reduce the likelihood of apoptosis. Future in vivo investigations must elucidate the mechanism(s) underlying CISD2-associated anti-inflammation and glial activation regulation.

5. Conclusions

The results obtained in this study demonstrate that CSC treatment inhibited astrocyte activation, astrocyte-mediated inflammatory responses, cellular apoptosis, and neuronal loss in rats following spinal cord hemisection. These results demonstrate the CISD2-preserving effects of CSC, which could contribute to the prevention of astrocytic activation, astrocyte-mediated neuroinflammation, apoptosis, and neuron loss.

Author contributions

WMK is responsible for the writing of the original draft and formal analysis. CJC was accountable for the resources, supervision, and investigation. TYC conceptualized the study and participated in the experiments and data analysis. MSL acted as the corresponding author and funded, wrote, reviewed, and edited the final draft.

Ethics approval and consent to participate

All rats were treated as per the Experimental Animal Laboratory guidelines under the approval of the Animal Care and Use Committee of Chang Gung University (IACUC Approval No.: CGU14-052).

Acknowledgment

This work was supported by grants from the Health Bureau, Taipei City Government, Taiwan (10401-62-038).

Conflict of Interest

The authors declare no conflict of interest.

References
[1]
Albensi, B. C. (2019) What is nuclear factor kappa B (NF-kB) doing in and to the mitochondrion? Frontiers in Cell and Developmental Biology 7, 154.
[2]
Anvari, B., Tanenbaum, B. S., Milner, T. E., Tang, K., Liaw, L. H., Kalafus, K., Kimel, S. and Nelson, J. S. (1996) Spatially selective photocoagulation of biological tissues: feasibility study utilizing cryogen spray cooling. Applied Optics 35, 3314-3320.
[3]
Arrica, M. and Bissonnette, B. (2007) Therapeutic hypothermia. Seminars in Cardiothoracic and Vascular Anesthesia 11, 6-15.
[4]
Bernard, S. A. and Buist, M. (2003) Induced hypothermia in critical care medicine: a review. Critical Care Medicine 31, 2041-2051.
[5]
Bethea, J. R. and Dietrich, W. D. (2002) Targeting the host inflammatory response in traumatic spinal cord injury. Current Opinion in Neurology 15, 355-360.
[6]
Chang, N. C., Nguyen, M., Germain, M. and Shore, G. C. (2010) Antagonism of Beclin 1-dependent autophagy by BCL-2 at the endoplasmic reticulum requires NAF-1. The EMBO Journal 29, 606-618.
[7]
Chang, N. C., Nguyen, M. and Shore, G. C. (2012) BCL2-CISD2. Autophagy 8, 856-857.
[8]
Chen, Y. F., Kao, C. H., Chen, Y. T., Wang, C. H., Wu, C. Y., Tsai, C. Y., Liu, F. C., Yang, C. W., Wei, Y. H., Hsu, M. T., Tsai, S. F. and Tsai, T. F. (2009) Cisd2 deficiency drives premature aging and causes mitochondria-mediated defects in mice. Genes & Development 23, 1183-1194.
[9]
Davies, S. J., Field, P. M. and Raisman, G. (1996) Regeneration of cut adult axons fails even in the presence of continuous aligned glial pathways. Experimental Neurology 142, 203-216.
[10]
Hachimi-Idrissi, S., Van, H. A., Michotte, A., Smolders, I., Sarre, S., Ebinger, G., Huyghens, L. and Michotte, Y. (2004) Postischemic mild hypothermia reduces neurotransmitter release and astroglial cell proliferation during reperfusion after asphyxial cardiac arrest in rats. Brain Research 1019, 217-225.
[11]
Hansebout, R. R., Tanner, J. A. and Romero-Sierra, C. (1984) Current status of spinal cord cooling in the treatment of acute spinal cord injury. Spine 9, 508-511.
[12]
Houle, J. D. and Tessler, A. (2003) Repair of chronic spinal cord injury. Experimental Neurology 182, 247-260.
[13]
Hung, K., Tsai, S., Lee, T., Lin, J., Chang, C. and Chiu, W. (2007) Gene transfer of insulin-like growth factor-I providing neuroprotection after spinal cord injury in rats. Journal of Neurosurgery Spine 6, 35-46.
[14]
Hung, K. S., Hwang, S. L., Liang, C. L., Chen, Y. J., Lee, T. H., Liu, J. K., Howng, S. L. and Wang, C. H. (2005) Calpain inhibitor inhibits p35-p25-Cdk5 activation, decreases tau hyperphosphorylation, and improves neurological function after spinal cord hemisection in rats. Journal of Neuropathology and Experimental Neurology 64, 15-26.
[15]
Hurlbert, R. J. (2006) Strategies of medical intervention in the management of acute spinal cord injury. Spine 31, S16-21; discussion S36.
[16]
Jordan, J. D. and Carhuapoma, J. R. (2007) Hypothermia: comparing technology. Journal of the Neurological Sciences 261, 35-38.
[17]
Kang, R., Zeh, H. J., Lotze, M. T. and Tang, D. (2011) The Beclin 1 network regulates autophagy and apoptosis. Cell Death and Differentiation 18, 571-580.
[18]
Kung, W. M., Lin, C. C., Kuo, C. Y., Juin, Y. C., Wu, P. C. and Lin, M. S. (2020) Wild bitter melon exerts anti-inflammatory effects by upregulating injury-attenuated CISD2 expression following spinal cord injury. Behavioural Neurology 2020, 1080521.
[19]
Kwon, B. K., Mann, C., Sohn, H. M., Hilibrand, A. S., Phillips, F. M., Wang, J. C. and Fehlings, M. G. (2008) Hypothermia for spinal cord injury. The Spine Journal 8, 859-874.
[20]
Kwon, B. K., Tetzlaff, W., Grauer, J. N., Beiner, J. and Vaccaro, A. R. (2004) Pathophysiology and pharmacologic treatment of acute spinal cord injury. The Spine Journal 4, 451-464.
[21]
Laforge, M., Rodrigues, V., Silvestre, R., Gautier, C., Weil, R., Corti, O. and Estaquier, J. (2016) NF-κB pathway controls mitochondrial dynamics. Cell Death & Differentiation 23, 89-98.
[22]
Lee, Y. H., Lin, C. H., Hsu, P. C., Sun, Y. Y., Huang, Y. J., Zhuo, J. H., Wang, C. Y., Gan, Y. L., Hung, C. C., Kuan, C. Y. and Shie, F. S. (2015) Aryl hydrocarbon receptor mediates both proinflammatory and anti-inflammatory effects in lipopolysaccharide-activated microglia. Glia 63, 1138-1154.
[23]
Li, S., Wang, L., Berman, M. A., Zhang, Y. and Dorf, M. E. (2007) RNAi screen in mouse astrocytes identifies phosphatases that regulate NF-kappaB signaling. Molecular Cell 24, 497-509.
[24]
Lin, C., Chiang, T., Chen, W., Sun, Y., Lee, Y. and Lin, M. (2017) CISD2 serves a novel role as a suppressor of nitric oxide signalling and curcumin increases CISD2 expression in spinal cord injuries. Injury 46, 2341-2350.
[25]
Lin, C., Chiang, T., Sun, Y. and Lin, M. (2019) Protective effects of CISD2 and influence of curcumin on CISD2 expression in aged animals and inflammatory cell model. Nutrients 11, 700.
[26]
Lin, M. (2020) CISD2 Attenuates inflammation and regulates microglia polarization in EOC microglial cells-as a potential therapeutic target for neurodegenerative dementia. Frontiers in Aging Neuroscience 12, 260.
[27]
Lin, M., Lee, Y., Chiu, W. and Hung, K. (2011) Curcumin provides neuroprotection after spinal cord injury. Journal of Surgical Research 166, 280-289.
[28]
Lin, M., Sun, Y., Chiu, W., Hung, C., Chang, C., Shie, F., Tsai, S., Lin, J., Hung, K. and Lee, Y. (2012) Curcumin attenuates the expression and secretion of RANTES after spinal cord injury in vivo and lipopolysaccharide-induced astrocyte reactivation in vitro. Journal of Neurotrauma 28, 1259-1269.
[29]
Liu, W., Tang, Y. and Feng, J. (2011) Cross talk between activation of microglia and astrocytes in pathological conditions in the central nervous system. Life Sciences 89, 141-146.
[30]
Lo Jr, T. P., Cho, K. S., Garg, M. S., Lynch, M. P., Marcillo, A. E., Koivisto, D. L., Stagg, M., Abril, R. M., Patel, S., Dietrich, W. D. and Pearse, D. D. (2009) Systemic hypothermia improves histological and functional outcome after cervical spinal cord contusion in rats. The Journal of Comparative Neurology 514, 433-448.
[31]
Lucas, S., Rothwell, N. J. and Gibson, R. M. (2006) The role of inflammation in CNS injury and disease. British Journal of Pharmacology 147 Suppl 1, S232-S240.
[32]
Luo, Y., Berman, M. A., Zhai, Q., Fischer, F. R., Abromson-Leeman, S. R., Zhang, Y., Kuziel, W. A., Gerard, C. and Dorf, M. E. (2002) RANTES stimulates inflammatory cascades and receptor modulation in murine astrocytes. Glia 39, 19-30.
[33]
Morino, T., Ogata, T., Horiuchi, H., Takeba, J., Okumura, H., Miyazaki, T. and Yamamoto, H. (2003) Delayed neuronal damage related to microglia proliferation after mild spinal cord compression injury. Neuroscience Research 46, 309-318.
[34]
Nelson, J. S., Milner, T. E., Anvari, B., Tanenbaum, B. S., Kimel, S., Svaasand, L. O. and Jacques, S. L. (1995) Dynamic epidermal cooling during pulsed laser treatment of port-wine stain. A new methodology with preliminary clinical evaluation. Archives of Dermatology 131, 695-700.
[35]
Nelson, J. S., Milner, T. E., Anvari, B., Tanenbaum, B. S., Svaasand, L. O. and Kimel, S. (1996) Dynamic epidermal cooling in conjunction with laser-induced photothermolysis of port wine stain blood vessels. Lasers in Surgery and Medicine 19, 224-229.
[36]
Ohmura, A., Nakajima, W., Ishida, A., Yasuoka, N., Kawamura, M., Miura, S. and Takada, G. (2005) Prolonged hypothermia protects neonatal rat brain against hypoxic-ischemia by reducing both apoptosis and necrosis. Brain and Development 27, 517-526.
[37]
Rossignol, S., Schwab, M., Schwartz, M. and Fehlings, M. G. (2007) Spinal cord injury: time to move? The Journal of Neuroscience 27, 11782-11792.
[38]
Scholpa, N. E. and Schnellmann, R. G. (2017) Mitochondrial-based therapeutics for the treatment of spinal cord injury: mitochondrial biogenesis as a potential pharmacological target. Journal of Pharmacology and Experimental Therapeutics 363, 303-313.
[39]
Shen, Z. Q., Chen, Y. F., Chen, J. R., Jou, Y. S., Wu, P. C., Kao, C. H., Wang, C. H., Huang, Y. L., Chen, C. F., Huang, T. S., Shyu, Y. C., Tsai, S. F., Kao, L. S. and Tsai, T. F. (2017) CISD2 Haploinsufficiency disrupts calcium homeostasis, causes nonalcoholic fatty liver disease, and promotes hepatocellular carcinoma. Cell Reports 21, 2198-2211.
[40]
Smithies, D. J., Milner, T. E., Tanenbaum, B. S., Goodman, D. M. and Nelson, J. S. (1998) Accuracy of subsurface temperature distributions computed from pulsed photothermal radiometry. Physics in Medicine & Biology 43, 2453-2463.
[41]
Stichel, C. C. and Müller, H. W. (1998) The CNS lesion scar: new vistas on an old regeneration barrier. Cell and Tissue Research 294, 1-9.
[42]
Tator, C. H. and Fehlings, M. G. (1991) Review of the secondary injury theory of acute spinal cord trauma with emphasis on vascular mechanisms. Journal of Neurosurgery 75, 15-26.
[43]
van Horssen, J., van Schaik, P. and Witte, M. (2019) Inflammation and mitochondrial dysfunction: a vicious circle in neurodegenerative disorders? Neuroscience Letters 710, 132931.
[44]
Waldorf, H. A., Alster, T. S., McMillan, K., Kauvar, A. N. B., Geronemus, R. G. and Nelson, S. J. (1997) Effect of dynamic cooling on 585-nm pulsed dye laser treatment of port-wine stain birthmarks. Dermatologic Surgery 23, 657-662.
[45]
Xu, Z., Wang, B. R., Wang, X., Kuang, F., Duan, X. L., Jiao, X. Y. and Ju, G. (2006) ERK1/2 and p38 mitogen-activated protein kinase mediate iNOS-induced spinal neuron degeneration after acute traumatic spinal cord injury. Life Science 79, 1895-1905.
[46]
Yu, C. G., Jimenez, O., Marcillo, A. E., Weider, B., Bangerter, K., Dietrich, W. D., Castro, S. and Yezierski, R. P. (2000) Beneficial effects of modest systemic hypothermia on locomotor function and histopathological damage following contusion-induced spinal cord injury in rats. Journal of Neurosurgery 93, 85-93.
Share
Back to top