• 제목/요약/키워드: mitochondrial translocation

검색결과 50건 처리시간 0.022초

Therapeutic applications of ginseng for skeletal muscle-related disorder management

  • Syed Sayeed Ahmad;Hee Jin Chun;Khurshid Ahmad;Inho Choi
    • Journal of Ginseng Research
    • /
    • 제48권1호
    • /
    • pp.12-19
    • /
    • 2024
  • Skeletal muscle (SM) is the largest organ of the body and is largely responsible for the metabolism required to maintain body functions. Furthermore, the maintenance of SM is dependent on the activation of muscle satellite (stem) cells (MSCs) and the subsequent proliferation and fusion of differentiating myoblasts into mature myofibers (myogenesis). Natural compounds are being used as therapeutic options to promote SM regeneration during aging, muscle atrophy, sarcopenia, cachexia, or obesity. In particular, ginseng-derived compounds have been utilized in these contexts, though ginsenoside Rg1 is mostly used for SM mass management. These compounds primarily function by activating the Akt/mTOR signaling pathway, upregulating myogenin and MyoD to induce muscle hypertrophy, downregulating atrophic factors (atrogin1, muscle ring-finger protein-1, myostatin, and mitochondrial reactive oxygen species production), and suppressing the expressions of tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) in cachexia. Ginsenoside compounds are also used for obesity management, and their anti-obesity effects are attributed to peroxisome proliferator activated receptor gamma (PPARγ) inhibition, AMPK activation, glucose transporter type 4 (GLUT4) translocation, and increased phosphorylations of insulin resistance (IR), insulin receptor substrate-1 (IRS-1), and Akt. This review was undertaken to provide an overview of the use of ginseng-related compounds for the management of SM-related disorders.

Ginsenoside compound K protects against cerebral ischemia/ reperfusion injury via Mul1/Mfn2-mediated mitochondrial dynamics and bioenergy

  • Qingxia Huang;Jing Li;Jinjin Chen;Zepeng Zhang;Peng Xu;Hongyu Qi;Zhaoqiang Chen;Jiaqi Liu;Jing Lu;Mengqi Shi;Yibin Zhang;Ying Ma;Daqing Zhao;Xiangyan Li
    • Journal of Ginseng Research
    • /
    • 제47권3호
    • /
    • pp.408-419
    • /
    • 2023
  • Background: Ginsenoside compound K (CK), the main active metabolite in Panax ginseng, has shown good safety and bioavailability in clinical trials and exerts neuroprotective effects in cerebral ischemic stroke. However, its potential role in the prevention of cerebral ischemia/reperfusion (I/R) injury remains unclear. Our study aimed to investigate the molecular mechanism of ginsenoside CK against cerebral I/R injury. Methods: We used a combination of in vitro and in vivo models, including oxygen and glucose deprivation/reperfusion induced PC12 cell model and middle cerebral artery occlusion/reperfusion induced rat model, to mimic I/R injury. Intracellular oxygen consumption and extracellular acidification rate were analyzed by Seahorse multifunctional energy metabolism system; ATP production was detected by luciferase method. The number and size of mitochondria were analyzed by transmission electron microscopy and MitoTracker probe combined with confocal laser microscopy. The potential mechanisms of ginsenoside CK on mitochondrial dynamics and bioenergy were evaluated by RNA interference, pharmacological antagonism combined with co-immunoprecipitation analysis and phenotypic analysis. Results: Ginsenoside CK pretreatment could attenuate mitochondrial translocation of DRP1, mitophagy, mitochondrial apoptosis, and neuronal bioenergy imbalance against cerebral I/R injury in both in vitro and in vivo models. Our data also confirmed that ginsenoside CK administration could reduce the binding affinity of Mul1 and Mfn2 to inhibit the ubiquitination and degradation of Mfn2, thereby elevating the protein level of Mfn2 in cerebral I/R injury. Conclusion: These data provide evidence that ginsenoside CK may be a promising therapeutic agent against cerebral I/R injury via Mul1/Mfn2 mediated mitochondrial dynamics and bioenergy.

Plumbagin from Plumbago Zeylanica L Induces Apoptosis in Human Non-small Cell Lung Cancer Cell Lines through NF-κB Inactivation

  • Xu, Tong-Peng;Shen, Hua;Liu, Ling-Xiang;Shu, Yong-Qian
    • Asian Pacific Journal of Cancer Prevention
    • /
    • 제14권4호
    • /
    • pp.2325-2331
    • /
    • 2013
  • Objective: To detect effects of plumbagin on proliferation and apoptosis in non-small cell lung cancer cell lines, and investigate the underlying mechanisms. Materials and Methods: Human non-small cell lung cancer cell lines A549, H292 and H460 were treated with various concentrations of plumbagin. Cell proliferation rates was determined using both cell counting kit-8 (CCK-8) and clonogenic assays. Apoptosis was detected by annexin V/propidium iodide double-labeled flow cytometry and TUNEL assay. The levels of reactive oxygen species (ROS) were detected by flow cytometry. Activity of NF-${\kappa}B$ was examined by electrophoretic mobility shift assay (EMSA) and luciferase reporter assay. Western blotting was used to assess the expression of both NF-${\kappa}B$ regulated apoptotic-related gene and activation of p65 and $I{\kappa}B{\kappa}$. Results: Plumbagin dose-dependently inhibited proliferation of the lung cancer cells. The IC50 values of plumbagin in A549, H292, and H460 cells were 10.3 ${\mu}mol/L$, 7.3 ${\mu}mol/L$, and 6.1 ${\mu}mol/L$ for 12 hours, respectively. The compound concentration-dependently induced apoptosis of the three cell lines. Treatment with plumbagin increased the intracellular level of ROS, and inhibited the activation of NK-${\kappa}B$. In addition to inhibition of NF-${\kappa}B$/p65 nuclear translocation, the compound also suppressed the degradation of $I{\kappa}B{\kappa}$. ROS scavenger NAC highly reversed the effect of plumbagin on apoptosis and inactivation of NK-${\kappa}B$ in H460 cell line. Treatment with plumbagin also increased the activity of caspase-9 and caspase-3, downregulated the expression of Bcl-2, upregulated the expression of Bax, Bak, and CytC. Conclusions: Plumbagin inhibits cell growth and induces apoptosis in human lung cancer cells through an NF-${\kappa}B$-regulated mitochondrial-mediated pathway, involving activation of ROS.

皂角刺 추출물의 Nrf2 활성화를 통한 간세포 보호 효과 (Gleditsia Spina Extract Protects Hepatocytes from Oxidative Stress through Nrf2 Activation)

  • 김재광;박상미;제갈경환;김영우;변성희;김상찬;조일제
    • 대한본초학회지
    • /
    • 제30권4호
    • /
    • pp.57-64
    • /
    • 2015
  • Objectives : Oxidative stress is one of the most causes of hepatocyte injury. Gleditsia spina, the thorns ofGleditsia sinensisLam., has been known for its anti-cancer and anti-inflammatory effects in Korean medicine. The present study investigated hepatoprotective effect of Gleditsia spina water extract (GSE) against oxidative stress induced by arachidonic acid (AA) + iron in HepG2 cells.Methods : To investigate cytoprotective effect of GSE, cells were pretreated with GSE and then subsequently exposed to 10 μM AA for 12 h, followed by 5 μM iron. Cell viability was monitored by MTT assay, and expression of apoptosis-related proteins was examined by immunoblot analysis. To identify responsible molecular mechanisms, reactive oxygen species (ROS) production, GSH contents, and mitochondrial membrane potential were measured. In addition, effect of GSE on nuclear factor erythroid 2-related factor 2 (Nrf2) activation was determined by immunoblot and antioxidant response element (ARE)-driven reporter gene assays.Results : GSE pretreatment prevented AA + iron-mediated cytotoxicity in concentration dependent manner. In addition, ROS production, glutathione depletion, and mitochondrial impairment by AA + iron were significantly inhibited by GSE. Furthermore, GSE promoted translocation of Nrf2 to nucleus, which acts as essential transcription factor for induction of antioxidant genes. Increased nuclear Nrf2 that caused by GSE treatment promoted transcriptional activity of ARE. Finally, GSE up-regulated sestrin-2 which was widely recognized as target gene of Nrf2.Conclusions : This study demonstrates that GSE protects hepatocytes from oxidative stress via activation of Nrf2 signaling pathway.

Suppression of Lipopolysaccharide-Induced Inflammatory and Oxidative Response by 5-Aminolevulinic Acid in RAW 264.7 Macrophages and Zebrafish Larvae

  • Ji, Seon Yeong;Cha, Hee-Jae;Molagoda, Ilandarage Menu Neelaka;Kim, Min Yeong;Kim, So Young;Hwangbo, Hyun;Lee, Hyesook;Kim, Gi-Young;Kim, Do-Hyung;Hyun, Jin Won;Kim, Heui-Soo;Kim, Suhkmann;Jin, Cheng-Yun;Choi, Yung Hyun
    • Biomolecules & Therapeutics
    • /
    • 제29권6호
    • /
    • pp.685-696
    • /
    • 2021
  • In this study, we investigated the inhibitory effect of 5-aminolevulinic acid (ALA), a heme precursor, on inflammatory and oxidative stress activated by lipopolysaccharide (LPS) in RAW 264.7 macrophages by estimating nitric oxide (NO), prostaglandin E2 (PGE2), cytokines, and reactive oxygen species (ROS). We also evaluated the molecular mechanisms through analysis of the expression of their regulatory genes, and further evaluated the anti-inflammatory and antioxidant efficacy of ALA against LPS in the zebrafish model. Our results indicated that ALA treatment significantly attenuated the LPS-induced release of pro-inflammatory mediators including NO and PGE2, which was associated with decreased inducible NO synthase and cyclooxygenase-2 expression. ALA also inhibited the LPS-induced expression of pro-inflammatory cytokines, such as tumor necrosis factor (TNF)-α, interleukin (IL)-1β, and IL-6, reducing their extracellular secretion. Additionally, ALA abolished ROS generation, improved the mitochondrial mass, and enhanced the expression of heme oxygenase-1 (HO-1) and the activation of nuclear translocation of nuclear factor-E2-related factor 2 (Nrf2) in LPS-stimulated RAW 264.7 macrophages. However, zinc protoporphyrin, a specific inhibitor of HO-1, reversed the ALA-mediated inhibition of pro-inflammatory cytokines production and activation of mitochondrial function in LPS-treated RAW 264.7 macrophages. Furthermore, ALA significantly abolished the expression of LPS-induced pro-inflammatory mediators and cytokines, and showed strong protective effects against NO and ROS production in zebrafish larvae. In conclusion, our findings suggest that ALA exerts LPS-induced anti-inflammatory and antioxidant effects by upregulating the Nrf2/HO-1 signaling pathway, and that ALA can be a potential functional agent to prevent inflammatory and oxidative damage.

Fisetin Protects C2C12 Mouse Myoblasts from Oxidative Stress-Induced Cytotoxicity through Regulation of the Nrf2/HO-1 Signaling

  • Cheol Park;Hee-Jae Cha;Da Hye Kim;Chan-Young Kwon;Shin-Hyung Park;Su Hyun Hong;EunJin Bang;Jaehun Cheong;Gi-Young Kim;Yung Hyun Choi
    • Journal of Microbiology and Biotechnology
    • /
    • 제33권5호
    • /
    • pp.591-599
    • /
    • 2023
  • Fisetin is a bioactive flavonol molecule and has been shown to have antioxidant potential, but its efficacy has not been fully validated. The aim of the present study was to investigate the protective efficacy of fisetin on C2C12 murine myoblastjdusts under hydrogen peroxide (H2O2)-induced oxidative damage. The results revealed that fisetin significantly weakened H2O2-induced cell viability inhibition and DNA damage while blocking reactive oxygen species (ROS) generation. Fisetin also significantly alleviated cell cycle arrest by H2O2 treatment through by reversing the upregulation of p21WAF1/CIP1 expression and the downregulation of cyclin A and B levels. In addition, fisetin significantly blocked apoptosis induced by H2O2 through increasing the Bcl-2/Bax ratio and attenuating mitochondrial damage, which was accompanied by inactivation of caspase-3 and suppression of poly(ADP-ribose) polymerase cleavage. Furthermore, fisetin-induced nuclear translocation and phosphorylation of Nrf2 were related to the increased expression and activation of heme oxygenase-1 (HO-1) in H2O2-stimulated C2C12 myoblasts. However, the protective efficacy of fisetin on H2O2-mediated cytotoxicity, including cell cycle arrest, apoptosis and mitochondrial dysfunction, were greatly offset when HO-1 activity was artificially inhibited. Therefore, our results indicate that fisetin as an Nrf2 activator effectively abrogated oxidative stress-mediated damage in C2C12 myoblasts.

Unique cartilage matrix-associated proteins에 의한 MC3T3-E1 조골세포에서의 고혈당 스트레스 완화 효과 (Unique Cartilage Matrix-Associated Protein Alleviates Hyperglycemic Stress in MC3T3-E1 Osteoblasts)

  • 주현영;박나래;김정은
    • 생명과학회지
    • /
    • 제33권11호
    • /
    • pp.851-858
    • /
    • 2023
  • Unique cartilage matrix-associated protein (UCMA)은 γ-카르복실화(Gla) 잔기가 풍부한 간외 비타민 K 의존 단백질이다. UCMA는 조골세포 분화를 촉진하고 뼈 형성을 강화한다고 보고되고 있지만 고혈당 스트레스 하에서 조골세포에 미치는 영향에 대해서는 아직 알려진 바가 없다. 본 연구에서는 고혈당 조건하에서의 MC3T3-E1 조골세포에서 UCMA 효과를 조사하기 위해 MC3T3-E1 조골세포를 높은 포도당에 노출한 후 재조합 UCMA 단백질을 처리하였다. MC3T3-E1 세포에서 활성 산소종(ROS)의 생성은 고혈당 조건하에서 증가했으나 UCMA 단백질 처리 후 감소했음을 CellROX 및 MitoSOX 염색으로 확인하였다. 또한 고혈당 조건에서 UCMA 단백질을 함께 처리한 MC3T3-E1 세포에서 정량적 중합효소 연쇄반응 결과, 항산화 유전자인 nuclear factor erythroid 2-related factor 2 와 superoxide dismutase 1 발현이 증가하였다. 동일 조건하에서 UCMA 단백질 처리에 의해 heme oxygenase-1 발현 감소와 함께 세포질에서 핵으로의 전위가 감소되었고, 미토콘드리아 분열에 관여하는 dynamin-related protein 1 발현이 증가하였으며, AKT 신호 활성은 억제되었다. 종합적으로 UCMA는 고혈당에 노출된 조골세포에서 ROS 생성을 완화하고, 항산화 유전자 발현을 증가시키고, 미토콘드리아 역학에 영향을 미치며, AKT 신호를 조절하는 것으로 보인다. 본 연구는 UCMA의 세포 메커니즘에 대한 이해를 돕고, 대사 장애 관련한 골 합병증에 대한 새로운 치료제로서의 잠재적 사용 가능성을 제시하고 있다.

미토콘드리아 Cytb 유전자를 이용한 잔가시고기의 신규 서식지 고령 회천 집단의 유전적 다양성 분석 (Analysis of Genetic Diversity across Newly Occupied Habitats within the Goryeong Population of Pungitius kaibarae Using the Mitochondrial Cytb Gene)

  • 김강래;성무성;황유진;이명석;정주희;김희수;유정남
    • 한국어류학회지
    • /
    • 제35권4호
    • /
    • pp.217-223
    • /
    • 2023
  • 잔가시고기 Pungitius kaibarae의 신규 집단인 고령(GR) 집단과 야생 집단의 특성을 규명하기 위해 미토콘드리아 cytb 유전자 영역의 886 bp 서열을 이용 총 4개 집단 (경상북도 고령(회천, GR), 포항(곡강천, PH), 경산(오목천, GYSA), 강원도 고성(배봉천, GS))을 분석하였다. 고령(GR) 집단에서 가장 낮은 haplotype 다양성을 나타냈고(Hd=0.000), 고성(GS) 집단에서 0.755로 가장 높은 haplotype 다양성을 확인하였다. Nucleotide 다양성은 고성(GS) 집단에서 0.00291로 가장 높은 다양성을 나타냈으며, 고령(GR) 집단에서 가장 0.00000로 가장 낮은 다양성을 보였다. 유전적 분화도에서 고령(GR) 집단은 포항(PH) 집단과 유전적으로 가장 가까운 것으로 나타났다. Haplotype 네트워크는 고령 (GR) 집단이 포항(PH) 집단과 군집되어 가장 유사한 것으로 나타났다. 고령(GR) 집단은 계통발생학적 tree에서 높은 지지도(98%)의 값으로 포항(PH) 집단과 군집됨을 확인하였다. 따라서 고령(GR) 집단은 포항(PH) 집단과 유사한 집단에서 유래됨을 추정하였다.

한국산 중고기속(Sarcocheilichthys) 어류의 유전적 다양성과 분자계통학적 유연관계 (Genetic Diversity and Molecular Phylogenetic Relationships of the Genus Sarcocheilichthys Fish in Korea)

  • 장지왕;김재구;고재근;윤봉한;배양섭
    • 한국어류학회지
    • /
    • 제36권2호
    • /
    • pp.139-155
    • /
    • 2024
  • 한국산 중고기속(genus Sarcocheilichthys)에 속하는 중고기(S. nigripinnis morii) 8개 집단과 참중고기(S. variegatus wakiyae) 5개 집단의 미토콘드리아 DNA의 cytb 유전자를 이용하여 각 집단의 유전적 다양성 및 분자계통학적 유연관계를 검토하였다. 참중고기 집단이 중고기 집단보다 유전적 다양성이 높은 것으로 확인되었다. cytb 유전자를 기반으로 한 한국산 중 고기속 어류의 분자계통도상에서 참중고기의 영산강(YSR) 집단은 중고기의 탐진강(TJR), 영산강(YSR), 섬진강(SJR) 집단과 clade를 이루며 현재의 분류체계와 일치하지 않는 유전적 유역관계가 나타났다. 한편, 핵 DNA 분자계통도상에서는 참중고기와 중고기로 뚜렷하게 구분이 가능하여 미토콘드리아 및 핵 DNA가 분자계통도상에서 상충하는 mitonuclear 불일치 현상이 나타냈다. 중고기의 섬진강(SJR) 집단이 동진강(DJR) 집단에 이입되어 교배가 일어난 것으로 추정되는 유전자형이 확인되었다. 동해로 유입되는 하천 중 유일하게 형산강(HSR)에만 이입되어 서식하는 것으로 알려진 중고기 집단은 한강(HR) 집단으로부터 이입되어 형성된 집단으로 추정되었지만, 고유한 유전적 그룹을 나타내는 유전자형도 확인되었다. 중고기의 한강(HR), 금강(GR), 만경강(MGR) 집단은 압록강 이북에 분포하는 북방중고기(S. czerskii), S. soldatovi와 유전적으로 동일 집단을 형성하였으며, 이에 다양한 표본을 확보하여 형태학적 및 분자계통학적 연구를 통한 분류학적 재검토가 요구되었다.

Mechanism Underlying NaF-Induced Apoptosis in Human Oral Squamous Cell Carcinoma

  • Hur, Young-Joo;Kim, Do-Kyun;Lee, Seung-Eun;Kim, In-Ryoung;Jeong, Na-Young;Kim, Ji-Young;Park, Bong-Soo
    • International Journal of Oral Biology
    • /
    • 제35권2호
    • /
    • pp.51-60
    • /
    • 2010
  • Few studies have evaluated the apoptosis-inducing efficacy of NaF on cancer cells in vitro but there has been no previous investigation of the apoptotic effects of NaF on human oral squamous cell carcinoma cells. In this study, we have investigated the mechanisms underlying the apoptotic response to NaF treatment in the YD9 human squamous cell carcinoma cell line. The viability of YD9 cells and their growth inhibition were assessed by MTT and clonogenic assays, respectively. Hoechst staining, DNA electrophoresis and TUNEL staining were conducted to detect apoptosis. YD9 cells were treated with NaF, and western blotting, immunocytochemistry, confocal microscopy, FACScan flow cytometry, and MMP and proteasome activity assays were performed sequentially. The NaF treatment resulted in a time- and dose-dependent decrease in YD9 cell viability, a dose-dependent inhibition of cell growth, and the induction of apoptotic cell death. The apoptotic response of these cells was manifested by nuclear condensation, DNA fragmentation, the reduction of MMP and proteasome activity, a decreased DNA content, the release of cytochrome c into the cytosol, the translocation of AIF and DFF40 (CAD) into the nucleus, a significant shift of the Bax/Bcl-2 ratio, and the activation of caspase-9, caspase-3, PARP, Lamin A/C and DFF45 (ICAD). Furthermore, NaF treatment resulted in the downregulation of G1 cell cyclerelated proteins, and upregulation of p53 and the Cdk inhibitor $p27^{KIP1}$. Taken collectively, our present findings demonstrate that NaF strongly inhibits YD9 cell proliferation by modulating the expression of G1 cell cycle-related proteins and inducing apoptosis via mitochondrial and caspase pathways.