• Title/Summary/Keyword: mitochondrial potential

Search Result 575, Processing Time 0.038 seconds

Multi-walled Carbon Nanotubes Affect the Morphology and Membrane Potential of Mitochondria in HeLa Cell

  • Lee, Wonwoo;Cho, Hyo Min;Oh, Chung Seok;Kim, Eun Hae;Sun, Woong
    • Applied Microscopy
    • /
    • v.44 no.2
    • /
    • pp.68-73
    • /
    • 2014
  • With wide use of nano-materials, it is increasingly important to address their potential toxicity to mammalian cells. However, toxic effects of these materials have been mainly assessed by the cell survival assays. Considering that mitochondrial morphology and quality are highly sensitive to the condition of the cells, and the impairment of mitochondrial function greatly affect the survival of cells, here we tested the impact of multi-walled carbon nanotubes (MWNT) on the survival, mitochondrial morphology, and their membrane potential in HeLa cells. Interestingly, although MWNT did not induce cell death until 24 hours as assessed by pyknotic cell assay, mitochondrial length was elongated and the mitochondrial membrane potential was significantly reduced by exposure of HeLa cells to MWNT. These results suggest that MWNT exposure is potentially harmful to the cell, and the mechanism how MWNT alters mitochondrial quality should be further explored to assess the safety of MWNT use.

The coordinated regulation of mitochondrial structure and function by Drp1 for mitochondrial quality surveillance

  • Cho, Hyo Min;Sun, Woong
    • BMB Reports
    • /
    • v.52 no.2
    • /
    • pp.109-110
    • /
    • 2019
  • Mitochondrial morphology is known to be continuously changing via fusion and fission, but it is unclear what the biological importance of this energy-consuming process is and how it develops. Several data have suggested that mitochondrial fission executed by Drp1 is necessary to select out a damaged spot from the interconnected mitochondrial network, but the precise mechanism for the recognition and isolation of a damaged sub-mitochondrial region during mitochondrial fission is yet unclear. Recently, Cho et al. found that the mitochondrial membrane potential (MMP) is transiently reduced by the physical interaction of Drp1 and mitochondrial Zinc transporter, Zip1, at the fission site prior to the typical mitochondrial division, and we found that this event is essential for a mitochondrial quality surveillance. In this review, Cho et al. discuss the role of a mitochondrial fission in the mitochondrial quality surveillance system.

Effect of Dioxin on the Change of Mitochondrial Inner Membrane Potential and the Induction of ROS (다이옥신이 미토콘드리아 내막의 전위차 변화 및 ROS 생성에 미치는 영향)

  • Cho, Il-Young;Sheen, Yhun-Yhong
    • Environmental Analysis Health and Toxicology
    • /
    • v.24 no.1
    • /
    • pp.33-41
    • /
    • 2009
  • Among the toxicants in the environment dioxin-like compounds, including TCDD(2,3,7,8-Tetrachlorodibenzo-p-Dioxin), are well known as carcinogen and teratogen. TCDD the most toxic of these compounds, may result in a wide variety of adverse health effects in humans and environment, including carconogenesis, hepatotoxicity, teratogenesis, and immunotoxicity. Also TCDD increases superoxide, peroxide radicals and induces oxidative stress that leads to breakage of DNA single-strand and mitochondrial dysfunction. Recently, there have been reports that persistent organic pollutants(POPs) may be causing metabolic disease through mitochondrial toxicity. In order to examine if dioxin brings about toxicity on mitochondria directly, we measured the change of the mitochondrial membrane potential after exposure to TCDD using JC-1 dye. After short time exposure of dioxin, mitochondrial depolarization was observed but it recovered to the control level immediately. This TCDD effect on mitochondrial membrane potential was not correlated either to the production of reactive oxygen species(ROS) or extracellular $Ca^{2+}$ by TCDD. Less than 2 hours exposure of TCDD did not show any change in ROS production but 0.25 nM TCDD for 48 hours or 0.5 nM TCDD for 12 hours exposure did increase in ROS production. Under these conditions of ROS production by TCDD, no changes in the mitochondrial membrane potential by TCDD was observed.

Asiatic Acid Protects Dopaminergic Neurons from Neuroinflammation by Suppressing Mitochondrial ROS Production

  • Chen, Dong;Zhang, Xiao-Ya;Sun, Jing;Cong, Qi-Jie;Chen, Wei-Xiong;Ahsan, Hafiz Muhammad;Gao, Jing;Qian, Jin-Jun
    • Biomolecules & Therapeutics
    • /
    • v.27 no.5
    • /
    • pp.442-449
    • /
    • 2019
  • This study sought to evaluate the effects of Asiatic acid in LPS-induced BV2 microglia cells and 1-methyl-4-phenyl-pyridine ($MPP^+$)-induced SH-SY5Y cells, to investigate the potential anti-inflammatory mechanisms of Asiatic acid in Parkinson's disease (PD). SH-SY5Y cells were induced using $MPP^+$ to establish as an in vitro model of PD, so that the effects of Asiatic acid on dopaminergic neurons could be examined. The NLRP3 inflammasome was activated in BV2 microglia cells to explore potential mechanisms for the neuroprotective effects of Asiatic acid. We showed that Asiatic acid reduced intracellular production of mitochondrial reactive oxygen species and altered the mitochondrial membrane potential to regulate mitochondrial dysfunction, and suppressed the NLRP3 inflammasome in microglia cells. We additionally found that treatment with Asiatic acid directly improved SH-SY5Y cell viability and mitochondrial dysfunction induced by $MPP^+$. These data demonstrate that Asiatic acid both inhibits the activation of the NLRP3 inflammasome by downregulating mitochondrial reactive oxygen species directly to protect dopaminergic neurons from, and improves mitochondrial dysfunction in SH-SY5Y cells, which were established as a model of Parkinson's disease. Our finding reveals that Asiatic acid protects dopaminergic neurons from neuroinflammation by suppressing NLRP3 inflammasome activation in microglia cells as well as protecting dopaminergic neurons directly. This suggests a promising clinical use of Asiatic acid for PD therapy.

Mitochondrial Transplantation Ameliorates the Development and Progression of Osteoarthritis

  • A Ram Lee;Jin Seok Woo;Seon-Yeong Lee;Hyun Sik Na;Keun-Hyung Cho;Yeon Su Lee;Jeong Su Lee;Seon Ae Kim;Sung-Hwan Park;Seok Jung Kim;Mi-La Cho
    • IMMUNE NETWORK
    • /
    • v.22 no.2
    • /
    • pp.14.1-14.17
    • /
    • 2022
  • Osteoarthritis (OA) is a common degenerative joint disease characterized by breakdown of joint cartilage. Mitochondrial dysfunction of the chondrocyte is a risk factor for OA progression. We examined the therapeutic potential of mitochondrial transplantation for OA. Mitochondria were injected into the knee joint of monosodium iodoacetate-induced OA rats. Chondrocytes from OA rats or patients with OA were cultured to examine mitochondrial function in cellular pathophysiology. Pain, cartilage destruction, and bone loss were improved in mitochondrial transplanted-OA rats. The transcript levels of IL-1β, TNF-α, matrix metallopeptidase 13, and MCP-1 in cartilage were markedly decreased by mitochondrial transplantation. Mitochondrial function, as indicated by membrane potential and oxygen consumption rate, in chondrocytes from OA rats was improved by mitochondrial transplantation. Likewise, the mitochondrial function of chondrocytes from OA patients was improved by coculture with mitochondria. Furthermore, inflammatory cell death was significantly decreased by coculture with mitochondria. Mitochondrial transplantation ameliorated OA progression, which is caused by mitochondrial dysfunction. These results suggest the therapeutic potential of mitochondrial transplantation for OA.

Intracellular pH is a Critical Element in Apoptosis Triggered by GM-CSF Deprivation in TF1 Cells

  • Yoon, Suk Ran;Choi, In Pyo
    • IMMUNE NETWORK
    • /
    • v.3 no.4
    • /
    • pp.268-275
    • /
    • 2003
  • Background: Hemopoietic cells require the constant presence of growth factors for survival in vitro and in vivo. Caspases have been known as central executors of apoptotic cell death. We have, therefore, investigated the pathways that regulate caspase activity and apoptosis using the $CD34^+$ cell line, TF-1 which requires GM-CSF for survival. Methods: Apoptosis was measured by annexin V staining and mitochondrial membrane potential was measured by DiOC6 labelling. Intracellular pH was measured using pH sensitive fluorochrome, BCECF or SNARF-1, followed by flow cytometry analysis. Caspase activation was analyzed by PARP cleavage using anti-PARP antibody. Results: Removal of GM-CSF induceed PARP cleavage, a hallmark of caspase activity, concomitant with pHi acidification and a drop in mitochondrial potential. Treatment with ZVAD, a competitive inhibitor of caspases, partially rescued cell death without affecting pHi acidification and the reduction of mitochondrial potential, suggesting that both these events act upstream of caspases. Overexpression of Bcl-2 prevented cell death induced by GM-CSF deprivation as well as pHi acidification and the reduction in mitochondrial membrane potential. In parental cells maintained with GM-CSF, EIPA, a competitive inhibitor of $Na^+/H^+$ antiporter induced apoptosis, accompanied by a drastic reduction in mitochondrial potential. In contrast, EIPA induced apoptosis in Bcl-2 transfectants without causing mitochondrial membrane depolarization. Conclusion: Taken together, our results suggest that the regulation of $H^+$fluxes, either through a mitochondriondependent or independent pathway, is central to caspase activation and apoptosis.

Hypermethylation-mediated silencing of NDRG4 promotes pancreatic ductal adenocarcinoma by regulating mitochondrial function

  • Shi, Hao-Hong;Liu, Hai-E;Luo, Xing-Jing
    • BMB Reports
    • /
    • v.53 no.12
    • /
    • pp.658-663
    • /
    • 2020
  • The N-myc downstream regulated gene (NDRG) family members are dysregulated in several tumors. Functionally, NDRGs play an important role in the malignant progression of cancer cells. However, little is known about the potential implications of NDRG4 in pancreatic ductal adenocarcinoma (PDAC). The aim of the current study was to elucidate the expression pattern of NDRG4 in PDAC and evaluate its potential cellular biological effects. Here, we firstly report that epigenetic-mediated silencing of NDRG4 promotes PDAC by regulating mitochondrial function. Data mining demonstrated that NDRG4 was significantly down-regulated in PDAC tissues and cells. PDAC patients with low NDRG4 expression showed poor prognosis. Epigenetic regulation by DNA methylation was closely associated with NDRG4 down-regulation. NDRG4 overexpression dramatically suppressed PDAC cell growth and metastasis. Further functional analysis demonstrated that up-regulated NDRG4 in SW1990 and Canpan1 cells resulted in attenuated mitochondrial function, including reduced ATP production, decreased mitochondrial membrane potential, and increased fragmented mitochondria. However, opposite results were obtained for HPNE cells with NDRG4 knockdown. These results indicate that hypermethylation-driven silencing of NDRG4 can promote PDAC by regulating mitochondrial function and that NDRG4 could be as a potential biomarker for PDAC patients.

Up-regulation of Cyelin A-Cdk2 activity is associated with depolarization of mitochondrial membrane potential during apoptosis of human hepatoma SK-HEP1 cells induced by treatment with panaxadiol

  • Park, Byoung-Duck;Jin, Ying-Hua;Yim, Hyung-Shin;Lee, Seung-Ki
    • Proceedings of the PSK Conference
    • /
    • 2003.10b
    • /
    • pp.167.1-167.1
    • /
    • 2003
  • Here we show that panaxadiol, a ginseng saponin with a dammarane skeleton, induces acute apoptotic cell death in human hepatoma SK-HEP-1 cells as evidenced by analysis of DNA fragmentation, caspase activation, and changes in cell morphology. The kinetic study showed that panaxadiol-induced apoptosis is associated with depolarization of mitochondrial membrane potential and cytochrome c release. Sequential activations of caspases-depolarization of mitochondrial membrane potential and cytochrome c release. Sequential activations of caspases-9, and -3, or -7, but not of caspase 8 coincide well in a time dependent manner with mitochondrial membrane depolarization and cytochrome c release from mitochondria during apoptosis of SK-HEP-1 cells induced by treatment with panaxadiol. (omitted)

  • PDF

Uncoupling Protein, UCP-4 May Be Involved in Neuronal Defects During Aging and Resistance to Pathogens in Caenorhabditis elegans

  • Cho, Injeong;Hwang, Gyu Jin;Cho, Jeong Hoon
    • Molecules and Cells
    • /
    • v.39 no.9
    • /
    • pp.680-686
    • /
    • 2016
  • Uncoupling proteins (UCPs) are mitochondrial inner membrane proteins that function to dissipate proton motive force and mitochondrial membrane potential. One UCP has been identified in Caenorhabditis elegans (C. elegans), namely UCP-4. In this study, we examined its expression and localization using a GFP marker in C. elegans. ucp-4 was expressed throughout the body from early embryo to aged adult and UCP-4 was localized in the mitochondria. It is known that increased mitochondrial membrane protential leads to a reactive oxygen species (ROS) increase, which is associated with age-related diseases, including neurodegenerative diseases in humans. A ucp-4 mutant showed increased mitochondrial membrane protential in association with increased neuronal defects during aging, and the neurons of ucp-4 overexpressing animals showed decreased neuronal defects during aging. These results suggest that UCP-4 may be involved in neuroprotection during aging via relieving mitochondrial membrane protential. We also investigated the relationship between UCP-4 and innate immunity because increased ROS can affect innate immunity. ucp-4 mutant displayed increased resistance to the pathogen Staphylococcus aureus compared to wild type. The enhanced immunity in the ucp-4 mutant could be related to increased mitochondrial membrane protential, presumably followed by increased ROS. In summary, UCP-4 might have an important role in neuronal aging and innate immune responses through mediating mitochondrial membrane protential.

Adenosine and Purine Nucleosides Prevent the Disruption of Mitochondrial Transmembrane Potential by Peroxynitrite in Rat Primary Astrocytes

  • Choi, Ji-Woong;Yoo, Byung-Kwon;Ryu, Mi-Kyoung;Choi, Min-Sik;Park, Gyu-Hwan;Ko, Kwang-Ho
    • Archives of Pharmacal Research
    • /
    • v.28 no.7
    • /
    • pp.810-815
    • /
    • 2005
  • Previously, we have shown that astrocytes deprived of glucose became highly vulnerable to peroxynitrite, and adenosine and its metabolites attenuated the gliotoxicity via the preservation of cellular ATP level. Here, we found that adenosine and related metabolites prevented the disruption of mitochondrial transmembrane potential (MTP) in glucose-deprived rat primary astrocytes exposed to 3-morpholinosydnonimine (SIN-1), a peroxynitrite releasing agent. Exposure to glucose deprivation and SIN-1(2h) significantly disrupted MTP in astrocytes, and adenosine prevented it in dose-dependent manner with an $EC_{50}\;of\;5.08{\mu}M$. Adenosine also partially prevented the cell death by myxothiazol, a well-known inhibitor of mitochondrial respiration. Blockade of adenosine deamination or intracellular transport with erythro-9-(-hydroxy-3-nonyl)adenosine (EHNA) or S-(4-nitrobenzyl)-6-thioinosine (NBTI), respectively, completely reversed the protective effect of adenosine. Other purine nucleos(t)ides including inosine, guanosine, ATP, ADP, AMP, ITP, and GTP also showed similar protective effects. This study indicates that adenosine and related purine nucleos(t)ides may protect astrocytes from peroxynitrite-induced mitochondrial dysfunction.