• Title/Summary/Keyword: Mitochondria membrane potential

Search Result 161, Processing Time 0.024 seconds

Mitochondria Control Protein as a Novel Therapeutic Target for Metabolic Syndrome

  • KimPak, Young-Mi
    • Proceedings of the Korean Society of Applied Pharmacology
    • /
    • 2008.04a
    • /
    • pp.23-30
    • /
    • 2008
  • Mitochondria biogenesis requires a coordination of two genomes, nuclear DNA (nDNA) and mitochondrial DNA (mtDNA). Disruption of mitochondria function leads to a loss of mitochondrial membrane potential and ATP generating capacity and consequently results in chronic degenerative diseases including insulin resistance, metabolic syndrome and neurodegenerative diseases. Although PPAR-${\gamma}$ coactivator-$1{\alpha}$ (PGC-$1{\alpha}$) was discovered as a central regulator of mitochondria biogenesis and a transcriptional co-activator of nuclear respiratory factor (NRF) and mitochondrial transcription factor A (Tfam), the expressions of PGC-$1{\alpha}$, NRF and Tfam were not significantly altered in tissues showing abnormal mitochondria functions. This observation suggests that there should be another regulator(s) for mitochondria function. Here, we demonstrate microRNAs (miRNAs) can modulate mitochondria function. Overexpression of microRNA dissipated mitochondrial membrane potential and increased ROS production in vitro and in vivo. It will be discussed the target of microRNA and its role in metabolic syndrome.

  • PDF

Anticancer Activity of Bispidinone Derivative by Induction of Apoptosis

  • Lee, Man Gi;Kwon, Ryong
    • Biomedical Science Letters
    • /
    • v.26 no.4
    • /
    • pp.336-343
    • /
    • 2020
  • The present study was carried out to investigate the possibility that bispidinone derivative makes anticancer drug availability to human cervical carcinoma cell. The B8 has the lowest IC50 value among B8, B9 and B10 which are bispidinone analogue with bromide. According to cytotoxic test through WST-8 assay, B8 shows the most magnificent cytotoxicity effectiveness with 76 μM of IC50 value. In human cervical carcinoma cell treated with B8, it noticeably controlled cellular multiplication by increase of concentration and time. Furthermore, morphological changes like cellular shrink, disruption and nuclear condensation, feature of apoptosis, are observed. Annexin V-FITC/PI double staining assay test proved that B8 can cause apoptosis. Moreover, after treatment with 76 μM of B8, flow cytometry analysis shows that increase of active oxygen species are induced and membrane potential in mitochondria is decreased. Manifestation of Bcl-2 family and caspase cascades protein provides evidence that B8 induces apoptosis through mitochondria and caspase-related pathway. Taken together, we suggested that B8 reduced membrane potential in mitochondria and induce apoptosis through the pathway depended on mitochondria and caspase.

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.

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.

Iron Toxicity to Peritoneal Macrophage Due to Alteration of Mitochondria by NO

  • Yoon, Ji-Yeon;Kim, Jin-Sun;Lee, Heum-Sook;Lee, Kyo-Young;Cheon, Choong-Ill;Lee, Myeong-Sok;Park, Jong-Hoon;Song, Eun-Sook
    • Animal cells and systems
    • /
    • v.8 no.2
    • /
    • pp.97-103
    • /
    • 2004
  • The cytotoxic effect of iron was examined in peritoneal macrophage to determine contributing factors by iron injection to rat. Viability was reduced by 24% by the iron-overload and by 30% by short-term iron addition. Total iron was increased by 45% in the iron-overloaded with remarkable elevation (9 to 14 fold) in the presence of $FeSO_4$. Free calcium was also increased by 19% in control and 44% in iron-overloaded group due to additional $FeSO_4$ NO and MDA were increased by 40% and 136%, respectively, with significant reduction (37%) of NAD(P)H. RCR and cytochrome c oxidase activity were lowered approximately by 10% with reduction of mitochondrial membrane potential. Addition of iron was frequently associated with altered distribution of mitochondria of high membrane potential in the iron-overloaded macrophage. These results suggest altered mitochondria with high NO and low NAD(P)H due to iron.

Rhizoma Scirpi induced Apoptosis in Human Cervical Carcinoma HeLa Cells (삼릉(三稜)이 자궁경부암세포(子宮頸部癌細胞)(HeLa cell)의 Apoptosis에 미치는 영향(影響))

  • Hong, Ki-Cheul;Kim, Joo-Yeon;Kong, Bok-Cheul;Choi, Chang-Min;Yoo, Sim-Keun
    • The Journal of Korean Obstetrics and Gynecology
    • /
    • v.18 no.4
    • /
    • pp.10-23
    • /
    • 2005
  • Purpose : This study is to examine the ability of Rhizoma Scirpi (RS) to induce HeLa cell viability. Methods : We culture HeLa cell which is human metrocarcinoma cell in D-MEM included 10% fetal bovine serum(Hyclone Laboratories) below $37^{\circ}C$, 5% CO2. Then we observed apoptosis of log phage cell which is changed cultivation liquid 24 Hours periodically. Results : 1. RS induces mitochondria membrane potential collapse. 2. P38 MAPK is involved in RS-induced death in HeLa cells. 3. P38 MAPK is involved in RS-induced apoptosis in HeLa cells. 4. P38 MAPK reguates RS-induced caspase-3, -8 and -9 activation in HeLa cells. 5. The inhibition of caspase regulates RS-induced cell death in HeLa cells. 6. RS induces mitochondria membrane potential collapse in HeLa cells. 7. P38 MPK is involved in the regulation of Bcl-2 and Bfu in HeLa cells.8. RS regulates the expression of Bcl-2 and Bax in HeLa cells. 9. SR induces p38 MAPK activation in HeLa cells. Conclusion : RS induces apoptosis in HeLa cells via p38 MAPK activation.

  • PDF

Effects of Harmaline and Harmalol on Dopamine Quinone-induced Brain Mitochondrial Dysfunction

  • Han, Eun-Sook;Lee, Chung-Soo
    • Biomolecules & Therapeutics
    • /
    • v.10 no.3
    • /
    • pp.152-158
    • /
    • 2002
  • The present study elucidated the effect of $\beta$-carbolines (harmaline and harmalol) on brain mitochondlial dysfunction caused by the tyrosinase-induced oxidation of dopamine. Harmaline, harmalol and antioxidant enzymes (SOD and catalase) attenuated the dopamine-induced alteration of membrane potential, cytochrome c release and thiol oxidation in mitochondria. In contrast, antioxidant enzymes failed to reverse mitochondrial dysfunction induced by dopmnine plus tyrosinase. $\beta$-Carbolines decreased the damaging effect of dopamine plus tyrosinase against mitochondria, except no effect of harmalol on thiol oxidation. Antioxidant enzymes decreased the melanin formation from dopamine in the reaction mixture containing mitochondria but did not reduce the formation of dopamine quinone caused by tyrosinase. Both harmalol and harmaline inhibited the formation of reactive quinone and melanin. Harmalol being more effective for quinone formation and vise versa. The results indicate that compared to MAO-induced dopamine oxidation, the toxic effect of dopamine in the presence of tyrosinase against mitochondria may be accomplished by the dopamine quinone and toxic substances other than reactive oxygen species. $\beta$-Carbolines may decrease the dopamine plus tyrosinase-induced brain mitochondrial dysfunction by inhibition of the formation of reactive quinone and the change in membrane permeability.

Niclosamide induces mitochondria fragmentation and promotes both apoptotic and autophagic cell death

  • Park, So-Jung;Shin, Ji-Hyun;Kang, Hee;Hwang, Jung-Jin;Cho, Dong-Hyung
    • BMB Reports
    • /
    • v.44 no.8
    • /
    • pp.517-522
    • /
    • 2011
  • Mitochondrial dynamics not only involves mitochondrial morphology but also mitochondrial biogenesis, mitochondrial distribution, and cell death. To identify specific regulators to mitochondria dynamics, we screened a chemical library and identified niclosamide as a potent inducer of mitochondria fission. Niclosamide promoted mitochondrial fragmentation but this was blocked by down-regulation of Drp1. Niclosamide treatment resulted in the disruption of mitochondria membrane potential and reduction of ATP levels. Moreover, niclosamide led to apoptotic cell death by caspase-3 activation. Interestingly, niclosamide also increased autophagic activity. Inhibition of autophagy suppressed niclosamide-induced cell death. Therefore, our findings suggest that niclosamide induces mitochondria fragmentation and may contribute to apoptotic and autophagic cell death.

Modulation of Mitochondrial Membrane Potential and ROS Generation by Nicotinamide in a Manner Independent of SIRT1 and Mitophagy

  • Song, Seon Beom;Jang, So-Young;Kang, Hyun Tae;Wei, Bie;Jeoun, Un-woo;Yoon, Gye Soon;Hwang, Eun Seong
    • Molecules and Cells
    • /
    • v.40 no.7
    • /
    • pp.503-514
    • /
    • 2017
  • Nicotinamide (NAM) plays essential roles in physiology through facilitating $NAD^+$ redox homeostasis. Importantly, at high doses, it protects cells under oxidative stresses, and has shown therapeutic effectiveness in a variety of disease conditions. In our previous studies, NAM lowered reactive oxygen species (ROS) levels and extended cellular life span in primary human cells. In the treated cells, levels of $NAD^+/NADH$ and SIRT1 activity increased, while mitochondrial content decreased through autophagy activation. The remaining mitochondria were marked with low superoxide levels and high membrane potentials (${\Delta}_{{\Psi}m}$); we posited that the treatment of NAM induced an activation of mitophagy that is selective for depolarized mitochondria, which produce high levels of ROS. However, evidence for the selective mitophagy that is mediated by SIRT1 has never been provided. This study sought to explain the mechanisms by which NAM lowers ROS levels and increases ${\Delta}_{{\Psi}m}$. Our results showed that NAM and SIRT1 activation exert quite different effects on mitochondrial physiology. Furthermore, the changes in ROS and ${\Delta}_{{\Psi}m}$ were not found to be mediated through autophagy or SIRT activation. Rather, NAM suppressed superoxide generation via a direct reduction of electron transport, and increased ${\Delta}_{{\Psi}m}$ via suppression of mitochondrial permeability transition pore formation. Our results dissected the effects of cellular $NAD^+$ redox modulation, and emphasized the importance of the $NAD^+/NADH$ ratio in the mitochondria as well as the cytosol in maintaining mitochondrial quality.

Bufalin Induces Mitochondrial Pathway-Mediated Apoptosis in Lung Adenocarcinoma Cells

  • Ding, Da-Wei;Zhang, Yong-Hong;Huang, Xin-En;An, Qing;Zhang, Xun
    • Asian Pacific Journal of Cancer Prevention
    • /
    • v.15 no.23
    • /
    • pp.10495-10500
    • /
    • 2015
  • Background: To evaluate the effects of bufalin in A549 human lung adenocarcinoma epithelial cells in vitro and assess the underlying mechanisms. Materials and Methods: Human A549 non-small cell lung cancer (NSCLC) cells were treated with various concentrations of bufalin. Cell proliferation was measured by CCK-8 assay, apoptotic cell percentage was calculated by flow cytometry and morphological change was observed by inverted phase contrast microscopy/transmission electron microscopy. In addition, the membrane potential of mitochondria was detected by JC-1 fluorescence microscopy assay, and the related protein expression of cytochrome C and caspase-3 was analyzed by Western blotting. Results: Bufalin could inhibit the proliferation of A549 cells via induction of apoptosis, with the evidence of characteristic morphological changes in the nucleus and mitochondria. Furthermore, bufalin decreased the mitochondrial membrane potential with up-regulation of cytochrome C in the cytosol, and activation of caspase-3. Conclusions: Bufalin inhibits the proliferation of A549 cells and triggers mitochondria-dependent apoptosis, pointing to therapeutic application for NSCLC.