open access

Ahead of Print
Original article
Submitted: 2023-05-28
Accepted: 2023-08-21
Published online: 2023-09-07
Get Citation

Oxidative stress mediates hippocampal neuronal apoptosis through ROS/JNK/P53 pathway in rats with PTSD triggered by high-voltage electrical burn

Ying Lv12, Xuegang Zhao3, Rui Zhang3, Zhaopeng He1, Yanfeng Xu1, Lihong Tu1, Lei Jiang3, Shunjiang Xu3, Qingfu Zhang1
·
Pubmed: 37691512
Affiliations
  1. Department of Burns and Plastic Surgery, the First Hospital of Hebei Medical University, Shijiazhuang, China
  2. Department of Cardiac Surgery, the First Hospital of Hebei Medical University, Shijiazhuang, China
  3. Hebei Key Laboratory of Brain Science and Psychiatric-Psychologic Disease, Shijiazhuang, China

open access

Ahead of Print
ORIGINAL ARTICLES
Submitted: 2023-05-28
Accepted: 2023-08-21
Published online: 2023-09-07

Abstract

Background: The pathogenesis of post-traumatic stress disorder (PTSD) triggered by high-voltage electrical burn (HVEB) remains unclear and the oxidative stress plays a role in this process. The purpose of this study is to investigate the underlying mechanism of oxidative stress mediates hippocampal neuronal apoptosis in rats with PTSD triggered by HVEB.

Materials and methods: The PTSD rat model was developed by stimulating with high voltage electricity and screened using behavioral performance including Morris water maze (MWM), elevated plus-maze (EPM) and open-field test (OFT). The reactive oxygen species (ROS) generation was measured by DHE fluorescence staining or flow cytometry. Western blotting assay was used to detect the proteins of p-JNK, JNK, P53, PUMA, Bcl-2 and Bax in hippocampal tissue or HT22 cells treated with electrical stimulation.

Results: The serum MDA and 8-OHdG levels were increased (P < 0.001), while the activities of SOD and CAT were decreased (P < 0.001) significantly in patients with HVEB. Behavioral test results showed that high-voltage electric stimulation induced the PTSD-like symptoms and the ROS-JNK-P53 pathway was involved in the neuronal apoptosis in rats with PTSD induced by HVEB. In vitro experiments further confirmed the electrical stimulation induced neuronal apoptosis through ROS/JNK/P53 signaling pathway and the antioxidant NAC could rescued the ROS generation, activation of JNK/P53 proteins and improved the cell apoptosis rate in HT22 cells. Finally, the JNK inhibitor SP600125 could significantly inhibited the percentage of HT22 cell apoptosis induced by electrical stimulation (P < 0.001).

Conclusions: These results indicated that oxidative stress mediates hippocampal neuronal apoptosis through ROS/JNK/P53 pathway in rats with PTSD triggered by HVEB.

Abstract

Background: The pathogenesis of post-traumatic stress disorder (PTSD) triggered by high-voltage electrical burn (HVEB) remains unclear and the oxidative stress plays a role in this process. The purpose of this study is to investigate the underlying mechanism of oxidative stress mediates hippocampal neuronal apoptosis in rats with PTSD triggered by HVEB.

Materials and methods: The PTSD rat model was developed by stimulating with high voltage electricity and screened using behavioral performance including Morris water maze (MWM), elevated plus-maze (EPM) and open-field test (OFT). The reactive oxygen species (ROS) generation was measured by DHE fluorescence staining or flow cytometry. Western blotting assay was used to detect the proteins of p-JNK, JNK, P53, PUMA, Bcl-2 and Bax in hippocampal tissue or HT22 cells treated with electrical stimulation.

Results: The serum MDA and 8-OHdG levels were increased (P < 0.001), while the activities of SOD and CAT were decreased (P < 0.001) significantly in patients with HVEB. Behavioral test results showed that high-voltage electric stimulation induced the PTSD-like symptoms and the ROS-JNK-P53 pathway was involved in the neuronal apoptosis in rats with PTSD induced by HVEB. In vitro experiments further confirmed the electrical stimulation induced neuronal apoptosis through ROS/JNK/P53 signaling pathway and the antioxidant NAC could rescued the ROS generation, activation of JNK/P53 proteins and improved the cell apoptosis rate in HT22 cells. Finally, the JNK inhibitor SP600125 could significantly inhibited the percentage of HT22 cell apoptosis induced by electrical stimulation (P < 0.001).

Conclusions: These results indicated that oxidative stress mediates hippocampal neuronal apoptosis through ROS/JNK/P53 pathway in rats with PTSD triggered by HVEB.

Get Citation

Keywords

high voltage electric burn, P53 protein, oxidative stress, brain injury, PTSD

About this article
Title

Oxidative stress mediates hippocampal neuronal apoptosis through ROS/JNK/P53 pathway in rats with PTSD triggered by high-voltage electrical burn

Journal

Folia Morphologica

Issue

Ahead of Print

Article type

Original article

Published online

2023-09-07

Page views

383

Article views/downloads

364

DOI

10.5603/fm.95727

Pubmed

37691512

Keywords

high voltage electric burn
P53 protein
oxidative stress
brain injury
PTSD

Authors

Ying Lv
Xuegang Zhao
Rui Zhang
Zhaopeng He
Yanfeng Xu
Lihong Tu
Lei Jiang
Shunjiang Xu
Qingfu Zhang

References (52)
  1. Barbano AC, van der Mei WF, Bryant RA, et al. Clinical implications of the proposed ICD-11 PTSD diagnostic criteria. Psychol Med. 2019; 49(3): 483–490.
  2. Beyfuss K, Hood DA. A systematic review of p53 regulation of oxidative stress in skeletal muscle. Redox Rep. 2018; 23(1): 100–117.
  3. Boeke EA, Moscarello JM, LeDoux JE, et al. Active avoidance: neural mechanisms and attenuation of pavlovian conditioned responding. J Neurosci. 2017; 37(18): 4808–4818.
  4. Clausen AN, Francisco AJ, Thelen J, et al. PTSD and cognitive symptoms relate to inhibition-related prefrontal activation and functional connectivity. Depress Anxiety. 2017; 34(5): 427–436.
  5. Daskal Y, Beicker A, Dudkiewicz M, et al. High voltage electric injury: mechanism of injury, clinical features and initial evaluation. Harefuah. 2019; 158(1): 65–69.
  6. DePamphilis MA, Cauley RP, Sadeq F, et al. Reconstruction of the upper extremity high-voltage electrical injury: a pediatric burn hospital's 13-year experience. J Burn Care Res. 2022; 43(3): 696–703.
  7. Di J, Tang J, Qian H, et al. p53 upregulates PLCε-IP3-Ca pathway and inhibits autophagy through its target gene Rap2B. Oncotarget. 2017; 8(39): 64657–64669.
  8. Garufi A, Pistritto G, Baldari S, et al. p53-dependent PUMA to DRAM antagonistic interplay as a key molecular switch in cell-fate decision in normal/high glucose conditions. J Exp Clin Cancer Res. 2017; 36(1): 126.
  9. Gentges J, Schieche C, Nusbaum J, et al. Electrical injuries in the emergency department: an evidence-based review. Emerg Med Pract. 2018; 20(11): 1–20.
  10. Piotrowski A, Fillet AM, Perez P, et al. Outcome of occupational electrical injuries among french electric company workers: a retrospective report of 311 cases, 1996–2005. Burns. 2014; 40(3): 480–488.
  11. Hasin D, Hatzenbuehler ML, Keyes K, et al. Substance use disorders: diagnostic and statistical manual of mental disorders, fourth edition (DSM-IV) and international classification of diseases, tenth edition (ICD-10). Addiction. 2006; 101 Suppl 1: 59–75.
  12. Hyland P, Vallières F, Cloitre M, et al. Trauma, PTSD, and complex PTSD in the Republic of Ireland: prevalence, service use, comorbidity, and risk factors. Soc Psychiatry Psychiatr Epidemiol. 2021; 56(4): 649–658.
  13. Imbriani P, Martella G, Bonsi P, et al. Oxidative stress and synaptic dysfunction in rodent models of Parkinson's disease. Neurobiol Dis. 2022; 173: 105851.
  14. Jeong CH, Joo SH. Downregulation of reactive oxygen species in apoptosis. J Cancer Prev. 2016; 21(1): 13–20.
  15. Jiang Le, Kon N, Li T, et al. Ferroptosis as a p53-mediated activity during tumour suppression. Nature. 2015; 520(7545): 57–62.
  16. Jiso A, Demuth P, Bachowsky M, et al. Natural merosesquiterpenes activate the DNA damage response via DNA strand break formation and trigger apoptotic cell death in p53-Wild-type and mutant colorectal cancer. Cancers (Basel). 2021; 13(13).
  17. Kasana RA, Baba PU, Wani AH. Pattern of high voltage electrical injuries in the Kashmir valley: a 10-year single centre experience. Ann Burns Fire Disasters. 2016; 29(4): 259–263.
  18. Keshavarzi A, Rahgozar Z, Mortazavi M, et al. Epidemiology, geographical distribution, and outcome analysis of patients with electrical burns referred to Shiraz Burn Center, Shiraz, Iran during 2008-2019. World J Plast Surg. 2022; 11(2): 102–109.
  19. Kinarivala N, Patel R, Boustany RM, et al. Discovery of aromatic carbamates that confer neuroprotective activity by enhancing autophagy and inducing the anti-apoptotic protein b-cell lymphoma 2 (Bcl-2). J Med Chem. 2017; 60(23): 9739–9756.
  20. La Colla A, Vasconsuelo A, Milanesi L, et al. 17β-Estradiol protects skeletal myoblasts from apoptosis through p53, Bcl-2, and foxo families. J Cell Biochem. 2017; 118(1): 104–115.
  21. Liberzon I, Krstov M, Young EA. Stress-restress: effects on ACTH and fast feedback. Psychoneuroendocrinology. 1997; 22(6): 443–453.
  22. Liu X, Wang Li, Cai J, et al. N-acetylcysteine alleviates H2O2-induced damage via regulating the redox status of intracellular antioxidants in H9c2 cells. Int J Mol Med. 2019; 43(1): 199–208.
  23. Logue MW, van Rooij SJH, Dennis EL, et al. Smaller hippocampal volume in posttraumatic stress disorder: a multisite ENIGMA-PGC study: subcortical volumetry results from posttraumatic stress disorder consortia. Biol Psychiatry. 2018; 83(3): 244–253.
  24. Lu Z, Miao Y, Muhammad I, et al. Colistin-induced autophagy and apoptosis involves the JNK-Bcl2-Bax signaling pathway and JNK-p53-ROS positive feedback loop in PC-12 cells. Chem Biol Interact. 2017; 277: 62–73.
  25. Maercker A, Cloitre M, Bachem R, et al. Complex post-traumatic stress disorder. Lancet. 2022; 400(10345): 60–72.
  26. Merians AN, Spiller T, Harpaz-Rotem I, et al. Post-traumatic stress disorder. Med Clin North Am. 2023; 107(1): 85–99.
  27. Mobayen M, Sadeghi M. Prevalence and related factors of electrical burns in patients referred to iranian medical centers: a systematic review and meta-analysis. World J Plast Surg. 2022; 11(1): 3–11.
  28. Nielsen KJ, Carstensen O, Kærgaard A, et al. Neurological symptoms and disorders following electrical injury: a register-based matched cohort study. PLoS One. 2022; 17(3): e0264857.
  29. Peruzzolo TL, Pinto JV, Roza TH, et al. Inflammatory and oxidative stress markers in post-traumatic stress disorder: a systematic review and meta-analysis. Mol Psychiatry. 2022; 27(8): 3150–3163.
  30. Piotrowski A, Fillet AM, Perez P, et al. Outcome of occupational electrical injuries among French electric company workers: a retrospective report of 311 cases, 1996-2005. Burns. 2014; 40(3): 480–488.
  31. Radulovic N, Mason SA, Rehou S, et al. Acute and long-term clinical, neuropsychological and return-to-work sequelae following electrical injury: a retrospective cohort study. BMJ Open. 2019; 9(5): e025990.
  32. Ressler KJ, Berretta S, Bolshakov VY, et al. Post-traumatic stress disorder: clinical and translational neuroscience from cells to circuits. Nat Rev Neurol. 2022; 18(5): 273–288.
  33. Ritov G, Boltyansky B, Richter-Levin G. A novel approach to PTSD modeling in rats reveals alternating patterns of limbic activity in different types of stress reaction. Mol Psychiatry. 2016; 21(5): 630–641.
  34. Rosenberg M, Mehta N, Rosenberg L, et al. Immediate and long-term psychological problems for survivors of severe pediatric electrical injury. Burns. 2015; 41(8): 1823–1830.
  35. Roy K, Wu Y, Meitzler JL, et al. NADPH oxidases and cancer. Clin Sci (Lond). 2015; 128(12): 863–875.
  36. Salehi SH, Sadat Azad Y, Bagheri T, et al. Epidemiology of occupational electrical injuries. J Burn Care Res. 2022; 43(2): 399–402.
  37. Schrader C, Ross AA. A review of PTSD and current treatment strategies. Mo Med. 2021; 118(6): 546–551.
  38. Seetharaman S, Fleshner M, Park CR, et al. Influence of daily social stimulation on behavioral and physiological outcomes in an animal model of PTSD. Brain Behav. 2016; 6(5): e00458.
  39. Shahid F, Gondal MF, Us Sabah N, et al. Psychosocial impact of electrical burn in children: a follow-up study conducted at a tertiary care hospital. Cureus. 2022; 14(12): e32816.
  40. Shi T, van Soest DMK, Polderman PE, et al. DNA damage and oxidant stress activate p53 through differential upstream signaling pathways. Free Radic Biol Med. 2021; 172: 298–311.
  41. Shih JG, Shahrokhi S, Jeschke MG. Review of adult electrical burn injury outcomes worldwide: an analysis of low-voltage vs high-voltage electrical injury. J Burn Care Res. 2017; 38(1): e293–e298.
  42. Simabuco FM, Morale MG, Pavan ICB, et al. p53 and metabolism: from mechanism to therapeutics. Oncotarget. 2018; 9(34): 23780–23823.
  43. Souza RR, Noble LJ, McIntyre CK. Using the single prolonged stress model to examine the pathophysiology of PTSD. Front Pharmacol. 2017; 8: 615.
  44. Steckley D, Karajgikar M, Dale LB, et al. Puma is a dominant regulator of oxidative stress induced Bax activation and neuronal apoptosis. J Neurosci. 2007; 27(47): 12989–12999.
  45. Suha T, Asli M, Aynur S, et al. Effects of N-acetylcysteine and ethyl pyruvate on ischemia-reperfusion injury in experimental electrical burn model. Am J Emerg Med. 2016; 34(7): 1217–1224.
  46. Sun Y, Qu Y, Zhu J. The relationship between inflammation and post-traumatic stress disorder. Front Psychiatry. 2021; 12: 707543.
  47. Suroto H, Asriel A, De Vega B, et al. Early and late apoptosis protein expression (Bcl-2, BAX and p53) in traumatic brachial plexus injury. J Musculoskelet Neuronal Interact. 2021; 21(4): 528–532.
  48. Tamayo-Gómez A, Velásquez-Suárez J, Páramo-Duque L, et al. Epidemiology and factors associated with acute stress disorder in burned patients: a case-control study. Burns. 2022; 48(4): 995–1003.
  49. Tamura RE, Hunger A, Fernandes DC, et al. Induction of oxidants distinguishes susceptibility of prostate carcinoma cell lines to p53 gene transfer mediated by an improved adenoviral vector. Hum Gene Ther. 2017; 28(8): 639–653.
  50. Wang P, Zhang SD, Jiao J, et al. ROS -mediated p53 activation by juglone enhances apoptosis and autophagy in vivo and in vitro. Toxicol Appl Pharmacol. 2019; 379: 114647.
  51. Wei R, Zhang R, Li H, et al. MiR-29 Targets PUMA to Suppress Oxygen and Glucose Deprivation/ Reperfusion (OGD/R)-induced Cell Death in Hippocampal Neurons. Curr Neurovasc Res. 2018; 15(1): 47–54.
  52. Yang H, Xie Y, Yang D, et al. Oxidative stress-induced apoptosis in granulosa cells involves JNK, p53 and Puma. Oncotarget. 2017; 8(15): 25310–25322.

Regulations

Important: This website uses cookies. More >>

The cookies allow us to identify your computer and find out details about your last visit. They remembering whether you've visited the site before, so that you remain logged in - or to help us work out how many new website visitors we get each month. Most internet browsers accept cookies automatically, but you can change the settings of your browser to erase cookies or prevent automatic acceptance if you prefer.

By VM Media Group sp. z o.o., Grupa Via Medica, Świętokrzyska 73, 80–180 Gdańsk, Poland

tel.: +48 58 320 94 94, faks: +48 58 320 94 60, e-mail: viamedica@viamedica.pl