• Title/Summary/Keyword: mitochondrial injury

Search Result 105, Processing Time 0.023 seconds

Cardioprotection via mitochondrial transplantation supports fatty acid metabolism in ischemia-reperfusion injured rat heart

  • Jehee Jang;Ki-Woon Kang;Young-Won Kim;Seohyun Jeong;Jaeyoon Park;Jihoon Park;Jisung Moon;Junghyun Jang;Seohyeon Kim;Sunghun Kim;Sungjoo Cho;Yurim Lee;Hyoung Kyu Kim;Jin Han;Eun-A Ko;Sung-Cherl Jung;Jung-Ha Kim;Jae-Hong Ko
    • The Korean Journal of Physiology and Pharmacology
    • /
    • v.28 no.3
    • /
    • pp.209-217
    • /
    • 2024
  • In addition to cellular damage, ischemia-reperfusion (IR) injury induces substantial damage to the mitochondria and endoplasmic reticulum. In this study, we sought to determine whether impaired mitochondrial function owing to IR could be restored by transplanting mitochondria into the heart under ex vivo IR states. Additionally, we aimed to provide preliminary results to inform therapeutic options for ischemic heart disease (IHD). Healthy mitochondria isolated from autologous gluteus maximus muscle were transplanted into the hearts of Sprague-Dawley rats damaged by IR using the Langendorff system, and the heart rate and oxygen consumption capacity of the mitochondria were measured to confirm whether heart function was restored. In addition, relative expression levels were measured to identify the genes related to IR injury. Mitochondrial oxygen consumption capacity was found to be lower in the IR group than in the group that underwent mitochondrial transplantation after IR injury (p < 0.05), and the control group showed a tendency toward increased oxygen consumption capacity compared with the IR group. Among the genes related to fatty acid metabolism, Cpt1b (p < 0.05) and Fads1 (p < 0.01) showed significant expression in the following order: IR group, IR + transplantation group, and control group. These results suggest that mitochondrial transplantation protects the heart from IR damage and may be feasible as a therapeutic option for IHD.

Regional Differences in Mitochondrial Anti-oxidant State during Ischemic Preconditioning in Rat Heart

  • Thu, Vu Thi;Cuong, Dang Van;Kim, Na-Ri;Youm, Jae-Boum;Warda, Mohamad;Park, Won-Sun;Ko, Jae-Hong;Kim, Eui-Yong;Han, Jin
    • The Korean Journal of Physiology and Pharmacology
    • /
    • v.11 no.2
    • /
    • pp.57-64
    • /
    • 2007
  • Ischemic preconditioning (IPC) is known to protect the heart against ischemia/reperfusion (IR)-induced injuries, and regional differences in the mitochondrial antioxidant state during IR or IPC may promote the death or survival of viable and infarcted cardiac tissues under oxidative stress. To date, however, the interplay between the mitochondrial antioxidant enzyme system and the level of reactive oxygen species (ROS) in the body has not yet been resolved. In the present study, we examined the effects of IR- and IPC-induced oxidative stresses on mitochondrial function in viable and infarcted cardiac tissues. Our results showed that the mitochondria from viable areas in the IR-induced group were swollen and fused, whereas those in the infarcted area were heavily damaged. IPC protected the mitochondria, thus reducing cardiac injury. We also found that the activity of the mitochondrial antioxidant enzyme system, which includes manganese superoxide dismutase (Mn-SOD), was enhanced in the viable areas compared to the infarcted areas in proportion with decreasing levels of ROS and mitochondrial DNA (mtDNA) damage. These changes were also present between the IPC and IR groups. Regional differences in Mn-SOD expression were shown to be related to a reduction in mtDNA damage as well as to the release of mitochondrial cytochrome c (Cyt c). To the best of our knowledge, this might be the first study to explore the regional mitochondrial changes during IPC. The present findings are expected to help elucidate the molecular mechanism involved in IPC and helpful in the development of new clinical strategies against ischemic heart disease.

Xylene Induces Oxidative Stress and Mitochondria Damage in Isolated Human Lymphocytes

  • Salimi, Ahmad;Talatappe, Behnaz Shoja;Pourahmad, Jalal
    • Toxicological Research
    • /
    • v.33 no.3
    • /
    • pp.233-238
    • /
    • 2017
  • Xylene is a cyclic hydrocarbon and an environmental pollutant. It is also used in medical technology, paints, dyes, polishes and in many industries as a solvent; therefore, an understanding of the interaction between xylene and human lymphocytes is of significant interest. Biochemical assessment was used to demonstrate that exposure of lymphocytes to xylene induces cytotoxicity (at 6 hr), generates intracellular reactive oxygen species, collapse of mitochondrial membrane potential, lysosomal injury, lipid peroxidation and depletion of glutathione (at 3 hr). The findings show that xylene triggers oxidative stress and organelle damage in lymphocytes. The results of our study suggest that the use of antioxidant, mitochondrial and lysosomal protective agents can be helpful for individuals subject to chronic exposure to xylene.

NecroX-5 protects mitochondrial oxidative phosphorylation capacity and preserves PGC1α expression levels during hypoxia/reoxygenation injury

  • Vu, Thi Thu;Kim, Hyoung Kyu;Le, Thanh Long;Nyamaa, Bayalagmaa;Song, In-Sung;To, Thanh Thuy;Nguyen, Quang Huy;Marquez, Jubert;Kim, Soon Ha;Kim, Nari;Ko, Kyung Soo;Rhee, Byoung Doo;Han, Jin
    • The Korean Journal of Physiology and Pharmacology
    • /
    • v.20 no.2
    • /
    • pp.201-211
    • /
    • 2016
  • Although the antioxidant and cardioprotective effects of NecroX-5 on various in vitro and in vivo models have been demonstrated, the action of this compound on the mitochondrial oxidative phosphorylation system remains unclear. Here we verify the role of NecroX-5 in protecting mitochondrial oxidative phosphorylation capacity during hypoxia-reoxygenation (HR). Necrox-5 treatment ($10{\mu}M$) and non-treatment were employed on isolated rat hearts during hypoxia/reoxygenation treatment using an ex vivo Langendorff system. Proteomic analysis was performed using liquid chromatography-mass spectrometry (LC-MS) and non-labeling peptide count protein quantification. Real-time PCR, western blot, citrate synthases and mitochondrial complex activity assays were then performed to assess heart function. Treatment with NecroX-5 during hypoxia significantly preserved electron transport chain proteins involved in oxidative phosphorylation and metabolic functions. NecroX-5 also improved mitochondrial complex I, II, and V function. Additionally, markedly higher peroxisome proliferator-activated receptor-gamma coactivator-$1{\alpha}$ ($PGC1{\alpha}$) expression levels were observed in NecroX-5-treated rat hearts. These novel results provide convincing evidence for the role of NecroX-5 in protecting mitochondrial oxidative phosphorylation capacity and in preserving $PGC1{\alpha}$ during cardiac HR injuries.

Effect of Polygoni Multiflori Ramulus extract against arachidonic acid and iron-induced oxidative stress in HepG2 cell and CCl4-induced liver injury in mice (야교등의 항산화 및 간보호효과)

  • Jeon, Chang Kwon;Jung, Ji Yun;Park, Chung A;Jee, Seon Young;Kim, Sang Chan
    • Herbal Formula Science
    • /
    • v.25 no.2
    • /
    • pp.155-166
    • /
    • 2017
  • Objectives : Polygoni Multiflori Ramulus has been widely used as a traditional medicinal herb for the treatment of insomnia, limb pain and itch. The extract of Polygoni Multiflori Ramulus (PMRE) is known to have a modulatory effect of many inflammatory responses. This study was performed to investigate the hepatoprotective effect of PMRE against arachidonic acid (AA) + iron-induced oxidative stress on HepG2 cell and carbon tetrachloride ($CCl_4$)-induced liver injury on mice. Methods : The effects of PMRE on cell viability was assessed by MTT assay. And flow cytometric analysis was performed to estimate the effects on mitochondrial function. To investigate its underlying mechanism, apoptosis-related proteins were analysed by using immunoblot analysis. In addition, ICR mouse were administrated (po) with the PMRE (30, 100 mg/kg) for 3 days and then, injected (ip) with $CCl_4$ (0.5 ml/kg body weight) to induce acute liver damage. The level of pro-caspase-3 was measured. Results : Treatment of PMRE increased relative cell viability, prevented a cleavage of poly (ADP ribose) polymerase and pro-caspase-3, and also reduced mitochondrial membrane permeability against AA + iron-induced oxidative stress. In addition, PMRE treatment decreased liver injury induced by $CCl_4$, as evidenced by increases in pro-caspase-3 level. Conclusions : These results demonstrate that PMRE has an ability to anti-oxidant and hepatoprotective effect against AA + iron-induced oxidative stress and $CCl_4$-induced liver injury.

The Effects of Difumarate Salt S-15176 after Spinal Cord Injury in Rats

  • Erdogan, Hakan;Tuncdemir, Matem;Kelten, Bilal;Akdemir, Osman;Karaoglan, Alper;Tasdemiroglu, Erol
    • Journal of Korean Neurosurgical Society
    • /
    • v.57 no.6
    • /
    • pp.445-454
    • /
    • 2015
  • Objective : In the present study we analyzed neuroprotective and antiapoptotic effect of the difumarate salt S-15176, as an anti-ischemic, an antioxidant and a stabilizer of mitochondrial membrane in secondary damage following spinal cord injury (SCI) in a rat model. Methods : Three groups were performed with 30 Wistar rats; control (1), trauma (2), and a trauma+S-15176 (10 mg/kg i.p., dimethyl sulfoxide) treatment (3). SCI was performed at the thoracic level using the weight-drop technique. Spinal cord tissues were collected following intracardiac perfusion in 3rd and 7th days of posttrauma. Hematoxylin and eosin staining for histopatology, terminal deoxynucleotidyl transferase dUTP nick end labeling assay for apoptotic cells and immunohistochemistry for proapoptotic cytochrome-c, Bax and caspase 9 were performed to all groups. Functional recovery test were applied to each group in 3rd and 7th days following SCI. Results : In trauma group, edematous regions, diffuse hemorrhage, necrosis, leukocyte infiltration and severe degeneration in motor neurons were observed prominently in gray matter. The number of apoptotic cells was significantly higher (p<0.05) than control group. In the S-15176-treated groups, apoptotic cell number in 3rd and 7th days (p<0.001), also cytochrome-c (p<0.001), Bax (p<0.001) and caspase 9 immunoreactive cells (p<0.001) were significantly decreased in number compared to trauma groups. Hemorrhage and edema in the focal areas were also noticed in gray matter of treatment groups. Results of the locomotor test were significantly increased in treatment group (p<0.05) when compared to trauma groups. Conclusion : We suggest that difumarate salt S-15176 prevents mitochondrial pathways of apoptosis and protects spinal cord from secondary injury and helps to preserve motor function following SCI in rats.

Enhanced Production of Adenosine Triphosphate by Pharmacological Activation of Adenosine Monophosphate-Activated Protein Kinase Ameliorates Acetaminophen-Induced Liver Injury

  • Hwang, Jung Hwan;Kim, Yong-Hoon;Noh, Jung-Ran;Choi, Dong-Hee;Kim, Kyoung-Shim;Lee, Chul-Ho
    • Molecules and Cells
    • /
    • v.38 no.10
    • /
    • pp.843-850
    • /
    • 2015
  • The1hepatic cell death induced by acetaminophen (APAP) is closely related to cellular adenosine triphosphate (ATP) depletion, which is mainly caused by mitochondrial dysfunction. Adenosine monophosphate (AMP)-activated protein kinase (AMPK) is a key sensor of low energy status. AMPK regulates metabolic homeostasis by stimulating catabolic metabolism and suppressing anabolic pathways to increase cellular energy levels. We found that the decrease in active phosphorylation of AMPK in response to APAP correlates with decreased ATP levels, in vivo. Therefore, we hypothesized that the enhanced production of ATP via AMPK stimulation can lead to amelioration of APAP-induced liver failure. A769662, an allosteric activator of AMPK, produced a strong synergistic effect on AMPK Thr172 phosphorylation with APAP in primary hepatocytes and liver tissue. Interestingly, activation of AMPK by A769662 ameliorated the APAP-induced hepatotoxicity in C57BL/6N mice treated with APAP at a dose of 400 mg/kg intraperitoneally. However, mice treated with APAP alone developed massive centrilobular necrosis, and APAP increased their serum alanine aminotransferase and aspartate aminotransferase levels. Furthermore, A769662 administration prevented the loss of intracellular ATP without interfering with the APAP-mediated reduction of mitochondrial dysfunction. In contrast, inhibition of glycolysis by 2-deoxy-glucose eliminated the beneficial effects of A769662 on APAP-mediated liver injury. In conclusion, A769662 can effectively protect mice against APAP-induced liver injury through ATP synthesis by anaerobic glycolysis. Furthermore, stimulation of AMPK may have potential therapeutic application for APAP overdose.

Human umbilical cord mesenchymal stem cell-derived mitochondria (PN-101) attenuate LPS-induced inflammatory responses by inhibiting NFκB signaling pathway

  • Yu, Shin-Hye;Kim, Soomin;Kim, Yujin;Lee, Seo-Eun;Park, Jong Hyeok;Cho, Gayoung;Ha, Jong-Cheon;Jung, Hahnsun;Lim, Sang-Min;Han, Kyuboem;Lee, Hong Kyu;Kang, Young Cheol;Kim, Chun-Hyung
    • BMB Reports
    • /
    • v.55 no.3
    • /
    • pp.136-141
    • /
    • 2022
  • Inflammation is one of the body's natural responses to injury and illness as part of the healing process. However, persistent inflammation can lead to chronic inflammatory diseases and multi-organ failure. Altered mitochondrial function has been implicated in several acute and chronic inflammatory diseases by inducing an abnormal inflammatory response. Therefore, treating inflammatory diseases by recovering mitochondrial function may be a potential therapeutic approach. Recently, mitochondrial transplantation has been proven to be beneficial in hyperinflammatory animal models. However, it is unclear how mitochondrial transplantation attenuates inflammatory responses induced by external stimuli. Here, we isolated mitochondria from umbilical cord-derived mesenchymal stem cells, referred as to PN-101. We found that PN-101 could significantly reduce LPS-induced mortality in mice. In addition, in phorbol 12-myristate 13-acetate (PMA)-treated THP-1 macrophages, PN-101 attenuated LPS-induced increase production of pro-inflammatory cytokines. Furthermore, the anti-inflammatory effect of PN-101 was mediated by blockade of phosphorylation, nuclear translocation, and trans-activity of NFκB. Taken together, our results demonstrate that PN-101 has therapeutic potential to attenuate pathological inflammatory responses.

Effect of suprascapular nerve injury on muscle and regenerated enthesis in a rat rotator cuff tear model

  • Kenichiro Eshima;Hiroki Ohzono;Masafumi Gotoh;Hisao Shimokobe;Koji Tanaka;Hidehiro Nakamura;Tomonoshin Kanazawa;Takahiro Okawa;Naoto Shiba
    • Clinics in Shoulder and Elbow
    • /
    • v.26 no.2
    • /
    • pp.131-139
    • /
    • 2023
  • Background: Massive rotator cuff tears (RCTs) are complicated by muscle atrophy, fibrosis, and intramuscular fatty degeneration, which are associated with postoperative tendon-to-bone healing failure and poor clinical outcomes. We evaluated muscle and enthesis changes in large tears with or without suprascapular nerve (SN) injury in a rat model. Methods: Sixty-two adult Sprague-Dawley rats were divided into SN injury (+) and SN injury (-) groups (n=31 each), comprising tendon (supraspinatus [SSP]/infraspinatus [ISP]) and nerve resection and tendon resection only cases, respectively. Muscle weight measurement, histological evaluation, and biomechanical testing were performed 4, 8, and 12 weeks postoperatively. Ultrastructural analysis with block face imaging was performed 8 weeks postoperatively. Results: SSP/ISP muscles in the SN injury (+) group appeared atrophic, with increased fatty tissue and decreased muscle weight, compared to those in the control and SN injury (-) groups. Immunoreactivity was only positive in the SN injury (+) group. Myofibril arrangement irregularity and mitochondrial swelling severity, along with number of fatty cells, were higher in the SN injury (+) group than in the SN injury (-) group. The bone-tendon junction enthesis was firm in the SN injury (-) group; this was atrophic and thinner in the SN injury (+) group, with decreased cell density and immature fibrocartilage. Mechanically, the tendon-bone insertion was significantly weaker in the SN injury (+) group than in the control and SN injury (+) groups. Conclusions: In clinical settings, SN injury may cause severe fatty changes and inhibition of postoperative tendon healing in large RCTs. Level of evidence: Level Basic research, controlled laboratory study.