• Title/Summary/Keyword: Apoptosis inhibitor

Search Result 796, Processing Time 0.029 seconds

Ginsenoside compound K ameliorates palmitate-induced atrophy in C2C12 myotubes via promyogenic effects and AMPK/autophagy-mediated suppression of endoplasmic reticulum stress

  • Kim, Tae Jin;Pyun, Do Hyeon;Kim, Myeong Jun;Jeong, Ji Hoon;Abd El-Aty, A.M.;Jung, Tae Woo
    • Journal of Ginseng Research
    • /
    • v.46 no.3
    • /
    • pp.444-453
    • /
    • 2022
  • Background: Compound K (CK) is among the protopanaxadiol (PPD)-type ginsenoside group, which produces multiple pharmacological effects. Herein, we examined the effects of CK on muscle atrophy under hyperlipidemic conditions along with its pro-myogenic effects. Further, the molecular pathways underlying the effects of CK on skeletal muscle have been justified. Methods: C2C12 myotubes were treated with palmitate and CK. C2C12 myoblasts were differentiated using CK for 4-5 days. For the in vivo experiments, CK was administered to mice fed on a high-fat diet for 8 weeks. The protein expression levels were analyzed using western blotting analysis. Target protein suppression was performed using small interfering (si) RNA transfection. Histological examination was performed using Jenner-Giemsa and H&E staining techniques. Results: CK treatment attenuated ER stress markers, such as eIF2a phosphorylation and CHOP expression and impaired myotube formation in palmitate-treated C2C12 myotubes and skeletal muscle of mice fed on HFD. CK treatment augmented AMPK along with autophagy markers in skeletal muscle cells in vitro and in vivo experiments. AMPK siRNA or 3-MA, an autophagy inhibitor, abrogated the impacts of CK in C2C12 myotubes. CK treatment augmented p38 and Akt phosphorylation, leading to an enhancement of C2C12 myogenesis. However, AMPK siRNA abolished the effects of CK in C2C12 myoblasts. Conclusion: These findings denote that CK prevents lipid-induced skeletal muscle apoptosis via AMPK/autophagy-mediated attenuation of ER stress and induction of myoblast differentiation. Therefore, we may suggest the use of CK as a potential therapeutic approach for treating muscle-wasting conditions associated with obesity.

Sertad1 Induces Neurological Injury after Ischemic Stroke via the CDK4/p-Rb Pathway

  • Li, Jianxiong;Li, Bin;Bu, Yujie;Zhang, Hailin;Guo, Jia;Hu, Jianping;Zhang, Yanfang
    • Molecules and Cells
    • /
    • v.45 no.4
    • /
    • pp.216-230
    • /
    • 2022
  • SERTA domain-containing protein 1 (Sertad1) is upregulated in the models of DNA damage and Alzheimer's disease, contributing to neuronal death. However, the role and mechanism of Sertad1 in ischemic/hypoxic neurological injury remain unclear. In the present study, our results showed that the expression of Sertad1 was upregulated in a mouse middle cerebral artery occlusion and reperfusion model and in HT22 cells after oxygen-glucose deprivation/reoxygenation (OGD/R). Sertad1 knockdown significantly ameliorated ischemia-induced brain infarct volume, neurological deficits and neuronal apoptosis. In addition, it significantly ameliorated the OGD/R-induced inhibition of cell viability and apoptotic cell death in HT22 cells. Sertad1 knockdown significantly inhibited the ischemic/hypoxic-induced expression of p-Rb, B-Myb, and Bim in vivo and in vitro. However, Sertad1 overexpression significantly exacerbated the OGD/R-induced inhibition of cell viability and apoptotic cell death and p-Rb, B-Myb, and Bim expression in HT22 cells. In further studies, we demonstrated that Sertad1 directly binds to CDK4 and the CDK4 inhibitor ON123300 restores the effects of Sertad1 overexpression on OGD/R-induced apoptotic cell death and p-Rb, B-Myb, and Bim expression in HT22 cells. These results suggested that Sertad1 contributed to ischemic/hypoxic neurological injury by activating the CDK4/p-Rb pathway.

Menin Enhances Androgen Receptor-Independent Proliferation and Migration of Prostate Cancer Cells

  • Kim, Taewan;Jeong, Kwanyoung;Kim, Eunji;Yoon, Kwanghyun;Choi, Jinmi;Park, Jae Hyeon;Kim, Jae-Hwan;Kim, Hyung Sik;Youn, Hong-Duk;Cho, Eun-Jung
    • Molecules and Cells
    • /
    • v.45 no.4
    • /
    • pp.202-215
    • /
    • 2022
  • The androgen receptor (AR) is an important therapeutic target for treating prostate cancer (PCa). Moreover, there is an increasing need for understanding the AR-independent progression of tumor cells such as neuroendocrine prostate cancer (NEPC). Menin, which is encoded by multiple endocrine neoplasia type 1 (MEN1), serves as a direct link between AR and the mixed-lineage leukemia (MLL) complex in PCa development by activating AR target genes through histone H3 lysine 4 methylation. Although menin is a critical component of AR signaling, its tumorigenic role in AR-independent PCa cells remains unknown. Here, we compared the role of menin in AR-positive and AR-negative PCa cells via RNAi-mediated or pharmacological inhibition of menin. We demonstrated that menin was involved in tumor cell growth and metastasis in PCa cells with low or deficient levels of AR. The inhibition of menin significantly diminished the growth of PCa cells and induced apoptosis, regardless of the presence of AR. Additionally, transcriptome analysis showed that the expression of many metastasis-associated genes was perturbed by menin inhibition in AR-negative DU145 cells. Furthermore, wound-healing assay results showed that menin promoted cell migration in AR-independent cellular contexts. Overall, these findings suggest a critical function of menin in tumorigenesis and provide a rationale for drug development against menin toward targeting high-risk metastatic PCa, especially those independent of AR.

Snail Promotes Cancer Cell Proliferation via Its Interaction with the BIRC3

  • Rho, Seung Bae;Byun, Hyun-Jung;Kim, Boh-Ram;Lee, Chang Hoon
    • Biomolecules & Therapeutics
    • /
    • v.30 no.4
    • /
    • pp.380-388
    • /
    • 2022
  • Snail is implicated in tumour growth and metastasis and is up-regulated in various human tumours. Although the role of Snails in epithelial-mesenchymal transition, which is particularly important in cancer metastasis, is well known, how they regulate tumour growth is poorly described. In this study, the possible molecular mechanisms of Snail in tumour growth were explored. Baculoviral inhibitor of apoptosis protein (IAP) repeat-containing protein 3 (BIRC3), a co-activator of cell proliferation during tumourigenesis, was identified as a Snail-binding protein via a yeast two-hybrid system. Since BIRC3 is important for cell survival, the effect of BIRC3 binding partner Snail on cell survival was investigated in ovarian cancer cell lines. Results revealed that Bax expression was activated, while the expression levels of anti-apoptotic proteins were markedly decreased by small interfering RNA (siRNA) specific for Snail (siSnail). siSnail, the binding partner of siBIRC3, activated the tumour suppressor function of p53 by promoting p53 protein stability. Conversely, BIRC3 could interact with Snail, for this reason, the possibility of BIRC3 involvement in EMT was investigated. BIRC3 overexpression resulted in a decreased expression of the epithelial marker and an increased expression of the mesenchymal markers. siSnail or siBIRC3 reduced the mRNA levels of matrix metalloproteinase (MMP)-2 and MMP-9. These results provide evidence that Snail promotes cell proliferation by interacting with BIRC3 and that BIRC3 might be involved in EMT via binding to Snail in ovarian cancer cells. Therefore, our results suggested the novel relevance of BIRC3, the binding partner of Snail, in ovarian cancer development.

Root Extract of Scutellaria Baicalensis Increases Gefitinib Sensitivity in H1975 Human Non-small Cell Lung Cancer Cells (H1975 세포에서 황금추출물에 의한 gefitinib 저항성 억제 효과)

  • Park, Shin-Hyung;Park, Hyun-Ji
    • Journal of Physiology & Pathology in Korean Medicine
    • /
    • v.35 no.4
    • /
    • pp.117-123
    • /
    • 2021
  • Gefitinib, a first generation epidermal growth factor receptor tyrosine kinase inhibitor (EGFR TKI), provides obvious clinical benefit in patients with EGFR-mutant non-small cell lung cancer (NSCLC). However, patients ultimately develop gefitinib resistance which mainly caused by EGFR T790M secondary mutation. In the current study, we investigated whether the root extract of Scutellaria baicalensis (SB) overcomes gefitinib resistance. Gefitinib-resistant H1975 human NSCLC cells (EGFR L858R/T790M double mutant) were treated with gefitinib and/or ethanol extract of SB (ESB) to evaluate the effect of ESB on the gefitinib sensitivity. The cell viability was measured by MTT assay and trypan blue exclusion assay. The colony-forming ability was evaluated by anchorage-dependent colony formation assay. Combined treatment with gefitinib and ESB markedly decreased the cell viability and colony formation than single treatment with gefitinib or ESB in H1975 cells. In addition, cells treated with both gefitinib and ESB exhibited a significant increase of sub-G1 DNA content which indicates apoptotic cells compared with those treated with gefitinib or ESB alone. As a molecular mechanism, combined treatment with gefitinib and ESB strongly downregulated the phosphorylation of ERK and JNK than single treatment with gefitinib or ESB. Taken together, our results demonstrate that ESB sensitizes H1975 cells to gefitinib treatment. We cautiously propose that ESB can be used in combination with gefitinib for the advanced NSCLC patients with acquired resistance to EGFR TKIs.

Selective cytotoxicity of a novel mitochondrial complex I inhibitor, YK-135, against EMT-subtype gastric cancer cell lines due to impaired glycolytic capacity

  • Yeojin, Sung;Seungbin, Cha;Sang Bum, Kim;Hakhyun, Kim;Seonghwi, Choi;Sejin, Oh;Minseo, Kim;Yunji, Lee;Gino, Kwon;Jooyoung, Lee;Joo-Youn, Lee;Gyoonhee, Han;Hyun Seok, Kim
    • BMB Reports
    • /
    • v.55 no.12
    • /
    • pp.645-650
    • /
    • 2022
  • Epithelial-to-mesenchymal transition (EMT)-subtype gastric cancers have the worst prognosis due to their higher recurrence rate, higher probability of developing metastases and higher chemo-resistance compared to those of other molecular subtypes. Pharmacologically actionable somatic mutations are rarely found in EMT-subtype gastric cancers, limiting the utility of targeted therapies. Here, we conducted a high-throughput chemical screen using 37 gastric cancer cell lines and 48,467 synthetic small-molecule compounds. We identified YK-135, a small-molecule compound that showed higher cytotoxicity toward EMT-subtype gastric cancer cell lines than toward non-EMT-subtype gastric cancer cell lines. YK-135 exerts its cytotoxic effects by inhibiting mitochondrial complex I activity and inducing AMP-activated protein kinase (AMPK)-mediated apoptosis. We found that the lower glycolytic capacity of the EMT-subtype gastric cancer cells confers synthetic lethality to the inhibition of mitochondrial complex I, possibly by failing to maintain energy homeostasis. Other well-known mitochondrial complex I inhibitors (e.g., rotenone and phenformin) mimic the efficacy of YK-135, supporting our results. These findings highlight mitochondrial complex I inhibitors as promising therapeutic agents for EMT-subtype gastric cancers and YK-135 as a novel chemical scaffold for further drug development.

Differential antiangiogenic and anticancer activities of the active metabolites of ginsenoside Rg3

  • Maryam Nakhjavani;Eric Smith;Kenny Yeo;Yoko Tomita;Timothy J. Price;Andrea Yool;Amanda R. Townsend;Jennifer E. Hardingham
    • Journal of Ginseng Research
    • /
    • v.48 no.2
    • /
    • pp.171-180
    • /
    • 2024
  • Background: Epimers of ginsenoside Rg3 (Rg3) have a low bioavailability and are prone to deglycosylation, which produces epimers of ginsenoside Rh2 (S-Rh2 and R-Rh2) and protopanaxadiol (S-PPD and R-PPD). The aim of this study was to compare the efficacy and potency of these molecules as anti-cancer agents. Methods: Crystal violet staining was used to study the anti-proliferatory action of the molecules on a human epithelial breast cancer cell line, MDA-MB-231, and human umbilical vein endothelial cells (HUVEC) and compare their potency. Cell death and cell cycle were studied using flow cytometry and mode of cell death was studied using live cell imaging. Anti-angiogenic effects of the drug were studied using loop formation assay. Molecular docking showed the interaction of these molecules with vascular endothelial growth factor receptor-2 (VEGFR2) and aquaporin (AQP) water channels. VEGF bioassay was used to study the interaction of Rh2 with VEGFR2, in vitro. Results: HUVEC was the more sensitive cell line to the anti-proliferative effects of S-Rh2, S-PPD and R-PPD. The molecules induced necroptosis/necrosis in MDA-MB-231 and apoptosis in HUVEC. S-Rh2 was the most potent inhibitor of loop formation. In silico molecular docking predicted a good binding score between Rh2 or PPD and the ATP-binding pocket of VEGFR2. VEGF bioassay showed that Rh2 was an allosteric modulator of VEGFR2. In addition, SRh2 and PPD had good binding scores with AQP1 and AQP5, both of which play roles in cell migration and proliferation. Conclusion: The combination of these molecules might be responsible for the anti-cancer effects observed by Rg3.

AMP-activated Kinase Regulates Adipocyte Differentiation Process in 3T3-L1 Adipocytes Treated with Selenium (AMP-activated protein kinase가 셀레늄으로 처리된 3T3-L1 지방세포의 분화과정 억제에 관한 연구)

  • Park, Song-Yi;Hwang, Jin-Taek;Lee, Yun-Kyoung;Kim, Young-Min;Park, Ock-Jin
    • Journal of Life Science
    • /
    • v.19 no.4
    • /
    • pp.423-428
    • /
    • 2009
  • Selenium was investigated using human origin preadipocytes to see whether it possesses preventive or therapeutic effects for obesity. Unveiling the potential of selenium in the reduction of adipogenesis can help predict the therapeutic capabilities of selenium in obesity. In the present study, the molecular mechanism of the inhibition of adipogenesis by selenium was explored to unravel the involvement of the AMP-activated protein kinase. There is emerging evidence that AMPK, a sensor of cellular energy status, is a possible molecular target of controlling adipocyte differentiation on the basis of discovery that AMPK is responsible for the major metabolic responses to exercise, and integration of nutritional and hormonal signals to modulate feeding behavior or energy expenditure in the hypothalamus. Treatment of selenium resulted in inhibition of the adipocyte differentiation process and induction of mature apoptosis in 3T3-L1 adipocytes. We hypothesized that selenium may exert anti-adipogenic potential though modulating AMPK. We have found that selenium significantly activated AMPK and phosphorylated its substrate acetyl-CoA carboxylase ($ACC-serine^{79}$) during the inhibitory process of adipocytes. Also, the inhibition process of adipocyte differentiation by selenium was comparable to either reveratrol or a synthetic AMPK activator, AICAR (5-aminoimidazole-4-carboxamide-1-${\beta}$-D-ribofuranoside). To evaluate the involvement of AMPK in anti-lipogensis, we applied AICAR and Compound C, an AMPK inhibitor, to 3T3-L1-adipocytes and found that AMPK is required for the adipocyte differentiation blocking process. These results suggest that selenium has a potential to control adipogenesis and that this effect is mediated by AMPK, an essential kinase for both inhibition of adipocyte differentiation and apoptosis of mature adipocytes.

Mechanisms of Tributyltin-induced Leydig Cell Apoptosis (유기주석화합물이 웅성생식세포주에 미치는 영향)

  • Lee, Kyung-Jin;Kim, Deok-Song;Ra, Myung-Suk;Wui, Seong-Uk;Im, Wook-Bin;Park, Hueng-Sik;Lee, Jong-Bin
    • Environmental Analysis Health and Toxicology
    • /
    • v.18 no.2
    • /
    • pp.89-94
    • /
    • 2003
  • Tributyltin (TBT) used world-wide in antifouling paints for ships is a widespread environmental pollutant and cause reproductive organs atrophy in rodents. At low doses, antiproliferative modes of action have been shown to be involved, whereas at higher doses apoptosis seems to be the mechanism of toxicity in reproductive organs by TBT. In this study, we investigated that the mechanisms underlying DNA fragmentation induced by TBT in the rat leyding cell line, R2C. Effects of TBT on intracellular Ca$\^$2+/ level and reactive oxygen species (ROS) were investigated in R2C cells by fluorescence detector. TBT significantly induced intracellular Ca$\^$2+/ level in a time-dependent manner. The rise in intracellular Ca$\^$2+/ level was followed by a time-dependent generation of reactive oxygen species (ROS) at the cytosol level. Simultaneously, TBT induced the release of cytochrome c from the mitochondrial membrane into the cytosol. Furthermore, ROS production and the release of cytochrome c were reduced by BAPTA, an intracellular Ca$\^$2+/ chelator, indicating the important role of Ca$\^$2+/ in R2C during these early intracellular events. In addition, Z-DEVD FMK, a caspase-3 inhibitor, decreased apoptosis by TBT. Taken together, the present results indicated that the apoptotic pathway by TBT might start with an increase in intracellular Ca$\^$2+/ level, continues with release of ROS and cytochrome c from mitochondria, activation of caspases,and finally results in DNA fragmentation.

Effect of Epigallocatechin Gallate on Phosphoinositide 3-kinase/Akt and Glycogen Synthase Kinase-3 Pathway in Oxidative-stressed N18D3 Cells Following $H_2O_2$ Exposure (산화성 손상을 받은 N18D3세포에서 Epigallocatechin gallate가 Phosphoinositide 3-kinase/Akt 및 Glycogen synthase kinase-3경로에 미치는 효과)

  • Koh, Seong Ho;Kwon, Hyug Sung;Oh, Hwa Soon;Oh, Jae Ho;Park, Ynun Joo;Kim, Jun Gyou;Kim, Ki Sok;Kim, Yang Soon;Yang, Ki Hwa;Kim, Seung U.;Kim, Seung H.;Jung, Hai Kwu
    • Korean Journal of Clinical Pharmacy
    • /
    • v.13 no.1
    • /
    • pp.29-39
    • /
    • 2003
  • Neurodegenerative disorders are associated with apoptosis as a causing factor or an inducer. On the other hand, it has been reported that epigallocatechin gallate (EUG), one of antioxidants and flavonoids, and z-VAD-fmk, a nonselective caspase inhibitor, suppress oxidative-radical-stress-induced apoptosis. However, it is not yet known what is the effects of EGCG and z-VAD-fmk on the apoptotic pathway is through phosphoinositide 3-kinase (PI3K), Akt and glycogen synthase kinase-3 (GSK-3) as well as mitochondria, caspase-3 and poly (ADP-ribose) polymerase (PARP). We investigated the effects of EGCG by using $H_2O_2$ treated N18D3 cells, mouse DRG hybrid neurons. Methods: Following 30 min $100\;{\mu}m\;H_2O_2$ exposure, the viability of N18D3 cells (not pretreated vs. EGCG or z-VAD-fmk pretreated) was evaluated by using MTT assay. The effect of EGCG on immunoreactivity (IR) of cytochrome c, caspase-3, PARP, PI3K/Akt and GSK-3 was examined by using Western blot, and was compared with that of z-Y4D-fmk. Results: EGCG or z-VAD-fmk pretreated N18D3 cells showed increased viability. Dose-dependent inhibition of caspase-3 activation accompanied by PARP cleavage were demonstrated by pretreatment of both agents. However, inhibition of cytochrome c release was only detected in EGCG pretreated N18D3 cells. On the pathway through PI3K/Akt and GSK-3, however, the result of Western blot in EGCG pretreated N18D3 cells showed decreased IR of Akt and GSK-3 and increased IR of p85a PI3K, phosphorylated Akt and GSK-3, and contrasted with that in z-VAD-fmk pretreated N18D3 cells showing no changes on each molecule. Conclusion: These data show that EGCG affects apoptotic pathway through upstream signal including PI3K/Akt and GSK-3 pathway as well as downstream signal including cytochrome c and caspase-3 pathway. Therefore, these results suggest that EGCG mediated activation of PI3K/Akt and inhibition GSK-B could be new potential therapeutic strategy for neurodegenerative diseases associated with oxidative injury.

  • PDF