• 제목/요약/키워드: Mitochondrial fragmentation

검색결과 142건 처리시간 0.03초

Melatonin Attenuates Mitochondrial Damage in Aristolochic Acid-Induced Acute Kidney Injury

  • Jian Sun;Jinjin Pan;Qinlong Liu;Jizhong Cheng;Qing Tang;Yuke Ji;Ke Cheng;Rui wang;Liang Liu;Dingyou Wang;Na Wu;Xu Zheng;Junxia Li;Xueyan Zhang;Zhilong Zhu;Yanchun Ding;Feng Zheng;Jia Li;Ying Zhang;Yuhui Yuan
    • Biomolecules & Therapeutics
    • /
    • 제31권1호
    • /
    • pp.97-107
    • /
    • 2023
  • Aristolochic acid (AA), extracted from Aristolochiaceae plants, plays an essential role in traditional herbal medicines and is used for different diseases. However, AA has been found to be nephrotoxic and is known to cause aristolochic acid nephropathy (AAN). AA-induced acute kidney injury (AKI) is a syndrome in AAN with a high morbidity that manifests mitochondrial damage as a key part of its pathological progression. Melatonin primarily serves as a mitochondria-targeted antioxidant. However, its mitochondrial protective role in AA-induced AKI is barely reported. In this study, mice were administrated 2.5 mg/kg AA to induce AKI. Melatonin reduced the increase in Upro and Scr and attenuated the necrosis and atrophy of renal proximal tubules in mice exposed to AA. Melatonin suppressed ROS generation, MDA levels and iNOS expression and increased SOD activities in vivo and in vitro. Intriguingly, the in vivo study revealed that melatonin decreased mitochondrial fragmentation in renal proximal tubular cells and increased ATP levels in kidney tissues in response to AA. In vitro, melatonin restored the mitochondrial membrane potential (MMP) in NRK-52E and HK-2 cells and led to an elevation in ATP levels. Confocal immunofluorescence data showed that puncta containing Mito-tracker and GFP-LC3A/B were reduced, thereby impeding the mitophagy of tubular epithelial cells. Furthermore, melatonin decreased LC3A/B-II expression and increased p62 expression. The apoptosis of tubular epithelial cells induced by AA was decreased. Therefore, our findings revealed that melatonin could prevent AA-induced AKI by attenuating mitochondrial damage, which may provide a potential therapeutic method for renal AA toxicity.

칠제향부환(七製香附丸)이 자궁근종세포의 성장억제와 세포자멸사에 미치는 영향 (Induced apoptosis in human Uterine Leiomyoma Cells by treatment with Chiljehyangbu-hwan)

  • 김석중;백승희;김은하;김동철
    • 대한한방부인과학회지
    • /
    • 제20권2호
    • /
    • pp.25-42
    • /
    • 2007
  • Purpose : Uterine leiomyoma (fibroids) are benign smooth muscle tumors originating from the myometrium. These benign neoplasms of monoclonal origin are typically diagnosed during the reproductive years, occurring only after puberty and tending to regress after menopause. In the present study we used Chiljehyangbu-hwan to determine its growth inhibitory effect and apoptosis in human uterine leiomyoma cells. Methods : Primary cultured human uterine leiomyoma cells were treated with Chiljehyangbu-hwan. Cell viability analysis was analyzed by MTS assay and FACS was performed to ascertain the effects Chiljehyangbu-hwan. DNA fragmentation analysis and casapase-3 activity test were done. Expression of apoptosis related proteins were evaluated by Western blot analysis. Results : Cell viability was significantly influenced by Chiljehyangbu-hwan treatment in a dose-dependent manner in leiomyoma cells compare to normal myometrial cells. FACS showed that Chiljehyangbu-hwan induced Sub G1 arrest. DNA fragmentation assay was carried out and apoptosis was detected. Activation of caspase-3, down-regulation of Bcl-2, with concomitant increased expression in Bid and Bax were observed. Chiljehyangbu-hwan treatment of uterine leiomyoma cells resulted in a concentration-dependent cell death induced via the mitochondrial pathway.

  • PDF

The Nuclear Orphan Receptor NR4A1 is Involved in the Apoptotic Pathway Induced by LPS and Simvastatin in RAW 264.7 Macrophages

  • Kim, Yong Chan;Song, Seok Bean;Lee, Sang Kyu;Park, Sang Min;Kim, Young Sang
    • IMMUNE NETWORK
    • /
    • 제14권2호
    • /
    • pp.116-122
    • /
    • 2014
  • Macrophage death plays a role in several physiological and inflammatory pathologies such as sepsis and arthritis. In our previous work, we showed that simvastatin triggers cell death in LPS-activated RAW 264.7 mouse macrophage cells through both caspase-dependent and independent apoptotic pathways. Here, we show that the nuclear orphan receptor NR4A1 is involved in a caspase-independent apoptotic process induced by LPS and simvastatin. Simvastatin-induced NR4A1 expression in RAW 264.7 macrophages and ectopic expression of a dominant-negative mutant form of NR4A1 effectively suppressed both DNA fragmentation and the disruption of mitochondrial membrane potential (MMP) during LPS- and simvastatin-induced apoptosis. Furthermore, apoptosis was accompanied by Bcl-2-associated X protein (Bax) translocation to the mitochondria. Our findings suggest that NR4A1 expression and mitochondrial translocation of Bax are related to simvastatin-induced apoptosis in LPS-activated RAW 264.7 macrophages.

Perphenazine and trifluoperazine induce mitochondria-mediated cell death in SH-SY5Y cells

  • Hong, Seok-Heon;Lee, Min-Yeong;Shin, Ki-Soon;Kang, Shin-Jung
    • Animal cells and systems
    • /
    • 제16권1호
    • /
    • pp.20-26
    • /
    • 2012
  • Drug-induced parkinsonism has been associated with an increased risk for Parkinson's disease. Antipsychotic drugs have long been known to cause parkinsonian symptoms. However, it remains unclear whether antipsychotics can directly damage the nigrostriatal pathway. In the present study, we investigated the toxicity mechanism of two typical antipsychotics, perphenazine and trifluoperazine, in a human dopaminergic cell line, SH-SY5Y. Perphenazine and trifluoperazine induced mitochondrial damage as evidenced by fragmentation of mitochondria, activation of Bax, cytochrome c release and a decrease in cellular ATP level. In addition, activation of caspase-3 and apoptotic nuclei were observed following the drug treatment. However, pan-caspase inhibitor did not suppress the cell death induced by the antipsychotics, suggesting that the initiated apoptosis was possibly shifted to necrosis upon caspase inhibition. Damaged mitochondria may have induced oxidative stress since the drug-induced cell death was partially suppressed by an antioxidant. Taken together, our results suggest that perphenazine and trifluoperazine can induce apoptotic cell death in a dopaminergic cell line via mitochondrial damage accompanied by oxidative stress.

Shikonin Exerts Cytotoxic Effects in Human Colon Cancers by Inducing Apoptotic Cell Death via the Endoplasmic Reticulum and Mitochondria-Mediated Pathways

  • Han, Xia;Kang, Kyoung Ah;Piao, Mei Jing;Zhen, Ao Xuan;Hyun, Yu Jae;Kim, Hyun Min;Ryu, Yea Seong;Hyun, Jin Won
    • Biomolecules & Therapeutics
    • /
    • 제27권1호
    • /
    • pp.41-47
    • /
    • 2019
  • The apoptotic effects of shikonin (5,8-dihydroxy-2-[(1R)-1-hydroxy-4-methylpent-3-enyl]naphthalene-1,4-dione) on the human colon cancer cell line SNU-407 were investigated in this study. Shikonin showed dose-dependent cytotoxic activity against SNU-407 cells, with an estimated $IC_{50}$ value of $3{\mu}M$ after 48 h of treatment. Shikonin induced apoptosis, as evidenced by apoptotic body formation, sub-G_1$ phase cells, and DNA fragmentation. Shikonin induced apoptotic cell death by activating mitogen-activated protein kinase family members, and the apoptotic process was mediated by the activation of endoplasmic reticulum (ER) stress, leading to activation of the $PERK/elF2{\alpha}/CHOP$ apoptotic pathway, and mitochondrial $Ca^{2+}$ accumulation. Shikonin increased mitochondrial membrane depolarization and altered the levels of apoptosis-related proteins, with a decrease in B cell lymphoma (Bcl)-2 and an increase in Bcl-2-associated X protein, and subsequently, increased expression of cleaved forms of caspase-9 and -3. Taken together, we suggest that these mechanisms, including MAPK signaling and the ER- and mitochondria-mediated pathways, may underlie shikonin-induced apoptosis related to its anticancer effect.

Protective effect of platelet-rich plasma against cold ischemia-induced apoptosis of canine adipose-derived mesenchymal stem cells

  • Suji Shin;Sung-Eon Kim;Seong-Won An;Seong-Mok Jeong;Young-Sam Kwon
    • 대한수의학회지
    • /
    • 제64권1호
    • /
    • pp.2.1-2.8
    • /
    • 2024
  • This study was performed to assess the antiapoptotic effect of canine platelet-rich plasma (PRP) treated on the canine adipose-derived mesenchymal stem cells (cMSCs) under cold ischemic conditions. The effect of preventing apoptosis of cMSCs was evaluated in the apoptotic condition induced by cold ischemic injury in vitro. To determine the progression of apoptosis, the changes in cell nucleus were observed using 4',6-diamidino-2-phenylindole (DAPI) fluorescence staining. In addition, we examined the mitochondrial membrane potential (MMP) and caspase-3 activity. When the cold hypoxic injury was applied to cMSCs, the apoptotic change was observed by DAPI staining, mitochondrial staining for MMP, and caspase-3 assay. PRP significantly decreased the number of apoptotic cells. Nuclear shrinkage and fragmentation of apoptotic cells in control groups were observed by DAPI staining. The MMP was recovered by the treatment of PRP. In addition, when the luminescence intensity was measured for caspase-3 activity, the value was significantly higher in the PRP treated groups than the control groups. The results of this study showed that the PRP may have a beneficial effect on apoptosis induced by cold ischemic injury.

부정항암탕(扶正抗癌湯)의 사람 췌장암 세포주 PANC-1에 대한 항종양(抗腫瘍) 효과(效果) (Anti-cancer Effects of Bujeonghangamtang on Human Pancreatic Cancer Cell Line PANC-1)

  • 김훈;원진희;문구
    • 대한한의학방제학회지
    • /
    • 제15권1호
    • /
    • pp.213-228
    • /
    • 2007
  • Objectives : The purpose of this report was to investigate the chemotherapeutic effect of Bujeonghangamtang against cancer cells. Materials and Methods : Various cancer cell lines including PANC-1, C6 glioma, SH-SY5Y, HepG2, and MCF-7 cells, were used. Apoptosis was determined by DAPI nuclei staining and flow cytometry in PANC-1 cells treated with 1 mg/ml Bujeonghangamtang for 48 hr. Expression of cell cycle arrest mediators including, cdc2p34 and cyclin B1 proteins were measured by Western blot analysis. Mitochondrial membrane potential was measured by fluorescence staining with JC-1, rhodamine 123. Result : Bujeonghangamtang induced the apoptosis of PANC-1, which was characterized as nucleic acid and genomic DNA fragmentation, chromatin condensation, and sub-G0/G1 fraction of cell cycle increase. but not C6 glioma, SH-SY5Y, HepG2, and MCF-7 cells. PANC-1 cells were markedly sensitive to Bujeonghangamtang. Treatment with Bujeonghangamtang resulted in the decreased expression of cdc2p34 and cyclin B1. Treatment with Bujeonghangamtang also increased the ROS production and induced mitochondrial dysfunction. Conclusion : Bujeonghangamtang exerted cytotoxicity against human Pancreatic cancer cells via cell cycle arrest-mediated apoptotic signaling including ROS production and mitochondrial dysfunction. Our data suggest that Bujeonghangamtang may be an important modulator of chemosensitivity of cancer cells against anticancer chemotherapeutic agents.

  • PDF

DNA Bis-intercalating Agent, Echinomycin-induced Apoptosis via Bcl-2 Dependence Pathway in Human Colon Cancer Cells

  • Park, Ju-Youn;Ryang, Yong-Suk;Kim, Jong-Bae;Chang, Jae-Ho;Cho, Hyeon-Cheol;Kim, Soo-Ki
    • Molecular & Cellular Toxicology
    • /
    • 제4권2호
    • /
    • pp.144-149
    • /
    • 2008
  • Despite versatile activity (cancericidal, antimicrobial, hypoxia inducible factor (HIF) inhibition, immune deactivation of DNA bis-intercalation agent, echinomycin, its specific mechanism has been elusive. Of these novel mechanisms, we reported that using human colon cancer cells (HT-29), apoptotic machinery induced by echinomycin might be dependent of caspase-3 pathway. Despite a partial enlightenment of prototypic signal path triggered by echinomycin, the role of Bcl-2 in this signaling pathway is unclear. To address this issue, we explored whether or not echinomycin would overcome the anti-apoptotic impact of Bcl-2 in HT-29 cells by the controlled Bcl-2 overexpression. Prior to this proof, we confirmed that echinomycin induces mitochondrial depolarization, then triggering the mitochondrial pathway of apoptosis with an involvement of upstream cas-pases-3. Transiently transfection with inactive Bax-DNA failed to prevent echinomycin-induced apoptosis in HT-29 cells. To dissect the role of Bcl-2 in echinomycin-induced apoptosis, HT-29 cells were transiently transfected with Bcl-2 DNA for overexpression and then treated with echinomycin for 24h. Combined analyses of DNA fragmentation and flow cytometric analysis clearly verified that echinomycin-induced apoptosis was drastically attenuated by Bcl-2 overexpression, whereas a control vector rarely affected echinomycin-induced apoptosis. Collectively, these data verify that Bcl-2 regulates echinomycin-induced apoptosis in HT-29 cells. To my knowledge, this is the first evidence that of diverse, structured minor groove binders (MGB), the prototypic echinomycin might control the apoptotic signaling via Bcl-2-mitochondrial pathway.

팔진항암단의 항종양효과 및 기전연구 (Study of Paljinhangahm-dan on Anti-tumoral Effect and Mechanism)

  • 배남규;문석재;원진희;김동웅;문구
    • 동의생리병리학회지
    • /
    • 제16권6호
    • /
    • pp.1143-1150
    • /
    • 2002
  • Recent evidence suggests that many Oriental Medicinal prescriptions are effective in cancer patients as a supportive care. Oriental Medicinal herbs have been investigated extensively and are known to have multiple pharmacological effect. These herbs contain a variety of ingredients which may act synergistically to inhibit tumor cell division, to increase tumor cell death (apoptosis), and to increase the proportion of immune cells within tumor. Paljinhangahm-dan (Paljin) has been used to treat for cancer patients in Oriental Medicine for decades. The effects of aqueous extract of Paljin on the induction of apoptotic cell death were investigated in human leukemia cell lines (HL-60, Jurkat, Molt-4 and U937). The viability of leukemia cells was markedly decreased by Paljin in a dose-dependent manner. Paljin induced the apoptotic death of leukemia cells, which was characterized by the ladder-pattern DNA fragmentation, and chromatin condensation of the nuclei. Paljin digested Bid protein but did not affect Bcl-2 protein level and also, induced mitochondrial dysfunction disrupted as shown as the mitochondrial membrane potential. It activated caspase-9 and caspase-3. thereby resulted in cleavage of poly(ADP) ribose polymerase(PARP). These results indicate that Paljin induces apoptosis of human leukemia cells via activation of intrinsic caspase cascades with mitochondrial dysfunction.

Assessment of the Cytotoxic and Apoptotic Effects of Chaetominine in a Human Leukemia Cell Line

  • Yao, Jingyun;Jiao, Ruihua;Liu, Changqing;Zhang, Yupeng;Yu, Wanguo;Lu, Yanhua;Tan, Renxiang
    • Biomolecules & Therapeutics
    • /
    • 제24권2호
    • /
    • pp.147-155
    • /
    • 2016
  • Chaetominine is a quinazoline alkaloid originating from the endophytic fungus Aspergillus fumigatus CY018. In this study, we showed evidence that chaetominine has cytotoxic and apoptotic effects on human leukemia K562 cells and investigated the pathway involved in chaetominine-induced apoptosis in detail. Chaetominine inhibited K562 cell growth, with an $IC_{50}$ value of 35 nM, but showed little inhibitory effect on the growth of human peripheral blood mononuclear cells. The high apoptosis rates, morphological apoptotic features, and DNA fragmentation caused by chaetominine indicated that the cytotoxicity was partially caused by its pro-apoptotic effect. Under chaetominine treatment, the Bax/Bcl-2 ratio was upregulated (from 0.3 to 8), which was followed by a decrease in mitochondrial membrane potential, release of cytochrome c from mitochondria into the cytosol, and stimulation of Apaf-1. Furthermore, activation of caspase-9 and caspase-3, which are the main executers of the apoptotic process, was observed. These results demonstrated that chaetominine induced cell apoptosis via the mitochondrial pathway. Chaetominine inhibited K562 cell growth and induced apoptotic cell death through the intrinsic pathway, which suggests that chaetominine might be a promising therapeutic for leukemia.