bims-aporos Biomed News
on Apoptosis and reactive oxygen species
Issue of 2019‒01‒13
forty-five papers selected by
Gavin McStay
Staffordshire University


  1. Cancers (Basel). 2019 Jan 09. pii: E64. [Epub ahead of print]11(1):
      Depression is more common in women with breast cancer than the general population. Selective serotonin reuptake inhibitors (SSRIs), a group of antidepressants, are widely used for the treatment of patients with depression and a range of anxiety-related disorders. The association between the use of antidepressant medication and breast cancer is controversial. In this study, we investigated whether and how SSRIs induce the death of human breast cancer MCF-7 cells. Of the antidepressants tested in this study (amitriptyline, bupropion, fluoxetine, paroxetine, and tianeptine), paroxetine most reduced the viability of MCF-7 cells in a time-and dose-dependent manner. The exposure of MCF-7 cells to paroxetine resulted in mitochondrion-mediated apoptosis, which is assessed by increase in the number of cells with sub-G1 DNA content, caspase-8/9 activation, poly (ADP-ribose) polymerase cleavage, and Bax/Bcl-2 ratio and a reduction in the mitochondrial membrane potential. Paroxetine increased a generation of reactive oxygen species (ROS), intracellular Ca2+ levels, and p38 MAPK activation. The paroxetine-induced apoptotic events were reduced by ROS scavengers and p38 MAPK inhibitor, and the paroxetine's effect was dependent on extracellular Ca2+ level. Paroxetine also showed a synergistic effect on cell death induced by chemotherapeutic drugs in MCF-7 and MDA-MB-231 cells. Our results showed that paroxetine induced apoptosis of human breast cancer MCF-7 cells through extracellular Ca2+-and p38 MAPK-dependent ROS generation. These results suggest that paroxetine may serve as an anticancer adjuvant to current cancer therapies for breast cancer patients with or without depression.
    Keywords:  MAPK; apoptosis; breast cancer; calcium; depression; paroxetine; reactive oxygen species
    DOI:  https://doi.org/10.3390/cancers11010064
  2. J Cell Physiol. 2019 Jan 07.
      Acute stress is a frequent and unpredictable disease for many animals. Stress is widely considered to affect liver function. However, the underlying mechanism by which dexmedetomidine (DEX) attenuates acute stress-induced liver injury in rats remains unclear. In this study, we used forced swimming for 15 min and acute 3-hr restraint stress model. Behavioral tests and changes in norepinephrine levels confirmed the successful establishment of the acute stress model. Acute stress-induced liver injury, evidenced by hematoxylin and eosin-stained pathological sections and increased serum aminotransferase and aspartate aminotransferase levels, was reduced in DEX-treated livers. Reactive oxygen species and oxidative stress levels were dramatically decreased with DEX treatment compared with acute stress-induced liver injury. DEX significantly reduced acute stress-induced liver inflammation and apoptosis, as assessed by terminal deoxynucleotidyl transferase dUTP nick-end labeling staining and inflammation and apoptosis-related protein levels. DEX treatment also effectively inhibited acute stress-induced c-Jun N-terminal kinase (JNK), P38, and BAD signaling pathway activation, and significantly induced MKP-1 activation. Thus, DEX has a protective effect on acute-stress-induced liver injury by reducing inflammation and apoptosis, which suggests a potential clinical application for DEX in stress syndrome.
    Keywords:  acute stress; apoptosis; dexmedetomidine; inflammation; liver injury
    DOI:  https://doi.org/10.1002/jcp.28096
  3. Front Pharmacol. 2018 ;9 1457
      Psoralen toxicity is an issue of wide concern. However, an assay for psoralen-induced developmental toxicity has not been reported to date. Moreover, the underlying mechanism of psoralen-induced developmental toxicity is unclear. Therefore, this study attempted to develop a psoralen-induced developmental toxicity assay in zebrafish embryos/larvae. Psoralen treatment caused a decrease in the hatching rate and body length and a significant increase in the malformation rate of zebrafish. Yolk retention, pericardial edema, swim-bladder deficiency, and curved body shape were also observed after psoralen treatment. Yolk retention might have been caused by an abnormality in lipid metabolism. Further experiments indicated that psoralen exerted toxic effects on the developing heart, liver, phagocytes, and nervous system. Increased generation of reactive oxygen species, inhibition of total superoxide dismutase activity, and increased malondialdehyde concentrations indicated inhibition of antioxidant capacity and the presence of oxidative stress. A greater number of apoptotic cells were observed after psoralen exposure, relative to the control. Furthermore, the results of gene-expression analysis showed that psoralen induced developmental toxicity by means of oxidative stress, apoptosis, and energy metabolism abnormalities. These findings will be helpful in understanding psoralen-induced toxicity.
    Keywords:  apoptosis; developmental toxicity; energy metabolism disorder; oxidative stress; psoralen; zebrafish
    DOI:  https://doi.org/10.3389/fphar.2018.01457
  4. J Ethnopharmacol. 2019 Jan 04. pii: S0378-8741(18)30846-8. [Epub ahead of print]
      ETHNOPHARMACOLOGICAL RELEVANCE: Shengmai injection (SMI) is a classical traditional Chinese medicine (TCM) officially recorded in Pharmacopoeia of the People's Republic of China (version 2015) and has long been used to treat heart failure in China. However scientific evidence for the anti-oxidative stress potential of SMI used in traditional medicine is lacking.AIM OF STUDY: The present study aimed to evaluate the efficacy of SMI in alleviating H2O2‑induced Oxidative Stress the underlying mechanisms MATERIALS AND METHODS: H2O2-induced oxidative stress model of cardiomyocytes was established with primary cultured neonatal rat cardiomyocytes. CCK8 cell viability assay and lacatate dehydrogenase cytotoxicity assay were performed to ensure the safety dose and lowest effective dose for the mode employing CCK-8 cell viability assay kit and lactate dehydrogenase cytotoxicity assay kit. ROS levels were determined using CM-H2DCFDA fluorescent probe in cardiomyocytes with H2O2-induced oxidative stress. The change of NAD(P)H level in cardiomyocytes was evaluated during the process of oxidative stress. The content of myocardial cytosolic Ca2+ and Ca2+ was determined using Fura-2/AM and Rhod 2-AM fluorescent probe in mitochondrial in the process of oxidative stress. Annexin V-FITC/PI double staining was applied to examine the apoptotic cells in cardiomyocytes with oxidative stress. To identify the apoptosis after oxidative stress myocardial cells with the application of Annexin V-FITC/PI double staining apoptosis detection kit. Quantitative polymerase chain reaction (RT-PCR) was applied to measure the expression of antioxidant enzymes: catalase (CAT), superoxide dismutase (SOD), glutathione reductase (GSR). Western blot was performed to observe the phosphorylation of AKT and ERK1/2.
    RESULTS: SMI was shown to significantly attenuate oxidative stress-induced cell proliferation arrest and apoptosis in neonatal rat cardiomyocytes. In addition, SMI treatment could decrease the production of reactive oxygen species (ROS), nicotinamide adenine dinucleotide (NADH) and malondialdehyde (MDA), and reduce the overloads of cytoplasmic Ca2+ and mitochondrial Ca2+ induced by H2O2. SMI could also restore the mRNA expression and activities of SOD, GSR, and CAT suppressed by H2O2. Mechanistically, SMI upregulated intracellular AKT phosphorylation and downregulate ERK1/2 phosphorylation in H2O2-treated cardiomyocytes. Pretreatment with LY294002, an AKT phosphorylation inhibitor, suppressed the protective role of SMI in cardiomyocytes, while pretreatment with PD98059, an ERK1/2 phosphorylation inhibitor, enhanced the effect of SMI.
    CONCLUSIONS: In conclusion, SMI may attenuate oxidative stress-induced damage in cardiomyocytes potentially through the AKT and ERK1/2 pathway and can function as a promising injectable traditional Chinese medicine to treat oxidative stress-induced injury.
    Keywords:  AKT; ERK1/2; Neonatal rat cardiomyocytes; Oxidative stress; Shengmai injection
    DOI:  https://doi.org/10.1016/j.jep.2019.01.001
  5. Korean J Physiol Pharmacol. 2019 Jan;23(1): 37-45
      To study the effect of nicorandil pretreatment on ketone body metabolism and Acetyl-CoA acetyltransferase (ACAT1) activity in hypoxia/reoxygenation (H/R)-induced cardiomyocytes. In our study, we applied H9c2 cardiomyocytes cell line to evaluate the cardioprotective effects of nicorandil. We detected mitochondrial viability, cellular apoptosis, reactive oxygen species (ROS) production and calcium overloading in H9c2 cells that exposed to H/R-induced cytotoxicity. Then we evaluated whether nicorandil possibly regulated ketone body, mainly β-hydroxybutyrate (BHB) and acetoacetate (ACAC), metabolism by regulating ACAT1 and Succinyl-CoA:3-keto-acid coenzyme A transferase 1 (OXCT1) protein and gene expressions. Nicorandil protected H9c2 cardiomyocytes against H/R-induced cytotoxicity dose-dependently by mitochondria-mediated anti-apoptosis pathway. Nicorandil significantly decreased cellular apoptotic rate and enhanced the ratio of Bcl-2/Bax expressions. Further, nicorandil decreased the production of ROS and alleviated calcium overloading in H/R-induced H9c2 cells. In crucial, nicorandil upregulated ACAT1 and OXCT1 protein expressions and either of their gene expressions, contributing to increased production of cellular BHB and ACAC. Nicorandil alleviated cardiomyocytes H/R-induced cytotoxicity through upregulating ACAT1/OXCT1 activity and ketone body metabolism, which might be a potential mechanism for emerging study of nicorandil and other KATP channel openers.
    Keywords:  Acetyl-CoA acetyltransferase; Hypoxia reoxygenation; Ketone body; Nicorandil; OXCT1
    DOI:  https://doi.org/10.4196/kjpp.2019.23.1.37
  6. Am J Chin Med. 2019 Jan 07. 1-21
      The aim of this study was to investigate the effects of bufalin on human nasopharyngeal carcinoma NPC-TW 076 cells in vitro. Bufalin is a cardiotonic steroid and a key active ingredient of the Chinese medicine ChanSu. The extracts of Chansu are used for various cancer treatments in China. In the present study, bufalin induced cell morphological changes, decreased total cell viability and induced G2/M phase arrest of cell cycle in NPC-TW 076 cells. Results also indicated that bufalin induced chromatin condensation (cell apoptosis) and DNA damage by DAPI staining and comet assay, respectively. The induced apoptotic cell death was further confirmed by annexin-V/PI staining assay. In addition, bufalin also increased ROS and Ca 2+ production and decreased the levels of ΔΨm . Furthermore, the alterations of ROS, ER stress and apoptosis associated protein expressions were investigated by Western blotting. Results demonstrated that bufalin increased the expressions of ROS associated proteins, including SOD (Cu/Zn), SOD2 (Mn) and GST but decreased that of catalase. Bufalin increased ER stress associated proteins (GRP78, IRE-1 α , IRE-1 β , caspase-4, ATF-6 α , Calpain 1, and GADD153). Bufalin increased the pro-apoptotic proteins Bax, and apoptotic associated proteins (cytochrome c, caspase-3, -8 and -9, AIF and Endo G) but reduced anti-apoptotic protein Bcl-2 in NPC-TW 076 cells. Furthermore, bufalin elevated the expressions of TRAIL-pathway associated proteins (TRAIL, DR4, DR5, and FADD). Based on these findings, we suggest bufalin induced apoptotic cell death via caspase-dependent, mitochondria-dependent and TRAIL pathways in human nasopharyngeal carcinoma NPC-TW 076 cells.
    Keywords:  Apoptosis; Bufalin; Mitochondrial ROS; NPC-TW 076 Cells; TRAIL Pathway
    DOI:  https://doi.org/10.1142/S0192415X19500125
  7. Int J Mol Med. 2019 Jan 02.
      Stachys riederi is one of the largest genera in the flowering plant family Lamiaceae. The aqueous extract of Stachys riederi var. japonica is known for its anti‑allergic effect. In the present study, the antioxidant and cytoprotective effects of Stachys riederi var. japonica ethanol extract (SREE) on ultraviolet A (UVA)‑irradiated human dermal fibroblasts (HDFs) were evaluated. At 100 µg/ml, SREE significantly inhibited production of reactive oxygen species (ROS) in UVA‑irradiated HDFs. SREE at 100 µg/ml additionally markedly interfered with the loss of mitochondrial membrane potential (ΔΨm) in these cells. In addition, SREE at 100 µg/ml attenuated UVA‑induced DNA fragmentation and caspase‑3 activation in HDFs. SREE at 100 µg/ml additionally increased mRNA and protein expressions of Bcl‑2 and decreased those of Bax and cytochrome c in UVA‑irradiated HDFs. In summary, to the best of our knowledge, these results demonstrate for the first time that SREE exhibited antioxidant and cytoprotective effects on UVA‑irradiated HDFs, which may be mediated through suppression of ROS generation, inhibition of the loss of ΔΨm and inhibition of apoptosis.
    DOI:  https://doi.org/10.3892/ijmm.2019.4048
  8. Mol Med Rep. 2019 Jan 04.
      Although total hip arthroplasty is considered to be an effective surgical procedure for treating hip joint diseases, it is hindered by implant wear debris, which induces aseptic loosening. Various cell types are involved in this pathogenesis; however, the interactions between wear debris and osteoblasts, which serve a crucial role in bone formation, have not been clearly illustrated. In the present study, minor metallic wear particles were collected from the interfacial membrane around loosened implants of patients, and the biological effects of these particles on rat primary osteoblasts were then explored. The results demonstrated that metallic wear debris was able to induce the apoptosis of treated cells in a concentration‑ and time‑dependent manner. Furthermore, it was identified that reactive oxygen species (ROS) generation increased, the mitochondrial membrane potential collapsed, and the mitochondria‑caspase‑dependent and endoplasmic reticulum (ER) stress apoptotic pathways were activated following metallic wear debris application. In addition, apoptosis and associated pathways were inhibited by the use of N‑acetyl‑L‑cysteine, an antioxidant that suppresses ROS production, indicating that the ROS generation triggered ER stress, mitochondrial dysfunction and downstream cascades that contributed to cell apoptosis. These findings suggest that metallic wear debris‑induced ROS serve an important role in the apoptosis of osteoblasts. This provides a valuable insight, not only into understanding the mechanisms underlying the involvement of osteoblasts in osteolysis, but also into a potential novel therapeutic approach to treat implant aseptic loosening.
    DOI:  https://doi.org/10.3892/mmr.2019.9825
  9. Cancer Cell Int. 2019 ;19 7
      Background: Gambogic acid is a natural component isolated from gamboge that possesses anticancer properties. Our previous study suggested that gambogic acid might be involved in autophagy; however, its role in pancreatic cancer remained unclear.Methods: Cell viability and apoptosis of pancreatic cancer cell lines were determined using (4,5-dimethylthiazol-2-yl)-3,5-diphenylformazan and flow cytometry. The effects of gambogic acid on autophagy was assessed by western blot, acridine orange staining, transmission electron microscopy, and measurement of autophagic flux through RFP-GFP-LC3 lentiviral transfection. The mitochondrial membrane potential was assessed by JC-1 staining. The production of reactive oxygen species was measured using CM-H2DCFDA staining. A xenograft tumor model of pancreatic cancer was created to determine the efficacy of gambogic acid and chloroquine.
    Results: Gambogic acid induced the expression of LC3-II and Beclin-1 proteins in pancreatic cancer cells, whereas the expression of P62 showed a decline. Gambogic acid also increased the formation of both acidic vesicular organelles and autophagosomes, and increased autophagic flux. These findings indicated that gambogic acid induced the autophagic process. Furthermore, inhibition of autophagy by chloroquine or 3-methyladenine, or knockdown of Atg-7 all enhanced the cytotoxicity of gambogic acid, suggesting that gambogic acid-induced autophagy improves the survival of pancreatic cancer cells. Moreover, gambogic acid reduced the mitochondrial membrane potential and promoted ROS production, which contributed to the activation of autophagy. The inhibition of autophagy by chloroquine further reduced the mitochondrial membrane potential and increased the accumulation of ROS. This indicated that the inhibition of autophagy could mitigate the cellular protective effects induced by gambogic acid. The treatment combination of gambogic acid and chloroquine synergistically inhibited tumor growth in the xenograft tumor model.
    Conclusions: These results demonstrate that gambogic acid induces cytoprotective autophagy in pancreatic cancer cells. The inhibition of autophagy promotes the cytotoxicity of gambogic acid by increasing the accumulation of ROS in pancreatic cancer cells. Combining chloroquine and gambogic acid may be a promising treatment for pancreatic cancer.
    Keywords:  Autophagy; Chloroquine; Gambogic acid; Pancreatic cancer; Reactive oxygen species
    DOI:  https://doi.org/10.1186/s12935-018-0705-x
  10. J Endocrinol. 2019 Jan 01. pii: JOE-18-0578.R1. [Epub ahead of print]
      The molecular signaling mechanisms of Coenzyme Q10 (CoQ10) in diabetic nephropathy (DN) remain poorly understood. We verified that mitochondrial abnormalities, like defective mitophagy, the generation of mitochondrial reactive oxygen species (mtROS) and the reduction of mitochondrial membrane potential, occurred in the glomerulus of db/db mice, accompanied by reduced PINK and parkin expression and increased apoptosis. These changes were partially reversed following oral administration of CoQ10. In inner fenestrated murine glomerular endothelial cells (mGECs), high glucose (HG) also resulted in deficient mitophagy, mitochondrial dysfunction and apoptosis, which were reversed by CoQ10. Mitophagy suppression mediated by Mdivi-1 or siPINK abrogated the renoprotective effects exerted by CoQ10, suggesting a beneficial role for CoQ10-restored mitophagy in DN. Mechanistically, CoQ10 restored the expression, activity and nuclear translocation of Nrf2 in HG-cultured mGECs. In addition, the reduced PINK and parkin expression observed in HG-cultured mGECs were partially elevated by CoQ10. CoQ10-mediated renoprotective effects were abrogated by the Nrf2 inhibitor ML385. When ML385 abolished mitophagy and the renoprotective effects exerted by CoQ10, mGECs could be rescued by treatment with mitoTEMPO, which is a mtROS-targeted antioxidant. These results suggest that CoQ10, as an effective antioxidant in mitochondria, exerts beneficial effects in DN via mitophagy by restoring Nrf2/ARE signaling.
    DOI:  https://doi.org/10.1530/JOE-18-0578
  11. Int J Mol Sci. 2019 Jan 10. pii: E247. [Epub ahead of print]20(2):
      Graphene and its derivatives are emerging as attractive materials for biomedical applications, including antibacterial, gene delivery, contrast imaging, and anticancer therapy applications. It is of fundamental importance to study the cytotoxicity and biocompatibility of these materials as well as how they interact with the immune system. The present study was conducted to assess the immunotoxicity of graphene oxide (GO) and vanillin-functionalized GO (V-rGO) on THP-1 cells, a human acute monocytic leukemia cell line. The synthesized GO and V-rGO were characterized by using various analytical techniques. Various concentrations of GO and V-rGO showed toxic effects on THP-1 cells such as the loss of cell viability and proliferation in a dose-dependent manner. Cytotoxicity was further demonstrated as an increased level of lactate dehydrogenase (LDH), loss of mitochondrial membrane potential (MMP), decreased level of ATP content, and cell death. Increased levels of reactive oxygen species (ROS) and lipid peroxidation caused redox imbalance in THP-1 cells, leading to increased levels of malondialdehyde (MDA) and decreased levels of anti-oxidants such as glutathione (GSH), glutathione peroxidase (GPX), super oxide dismutase (SOD), and catalase (CAT). Increased generation of ROS and reduced MMP with simultaneous increases in the expression of pro-apoptotic genes and downregulation of anti-apoptotic genes suggest that the mitochondria-mediated pathway is involved in GO and V-rGO-induced apoptosis. Apoptosis was induced consistently with the significant DNA damage caused by increased levels of 8-oxo-dG and upregulation of various key DNA-regulating genes in THP-1 cells, indicating that GO and V-rGO induce cell death through oxidative stress. As a result of these events, GO and V-rGO stimulated the secretion of various cytokines and chemokines, indicating that the graphene materials induced potent inflammatory responses to THP-1 cells. The harshness of V-rGO in all assays tested occurred because of better charge transfer, various carbon to oxygen ratios, and chemical compositions in the rGO. Overall, these findings suggest that it is essential to better understand the parameters governing GO and functionalized GO in immunotoxicity and inflammation. Rational design of safe GO-based formulations for various applications, including nanomedicine, may result in the development of risk management methods for people exposed to graphene and graphene family materials, as these nanoparticles can be used as delivery agents in various biomedical applications.
    Keywords:  apoptosis; cytokines; graphene; human acute monocytic leukemia cell; immunotoxicity; mitochondria; oxidative stress
    DOI:  https://doi.org/10.3390/ijms20020247
  12. Chem Biol Drug Des. 2019 Jan 11.
      A series of novel quinoxaline derivatives were synthesized and evaluated for their antiproliferative activity in three human cancer cell lines. Compound 12 exhibited the most potent antiproliferative activity with IC50 in the range of 0.19-0.51 μM. The compound inhibited tubulin polymerization and disrupted the microtubule network, leading to G2/M phase arrest. Furthermore, compound 12 induced ROS production and malfunction of mitochondrial membrane potential. Compound 12 led to cancer cells apoptosis in a dose-dependent manner. Western blot analysis showed that compound 12 induced up-regulation of p21 and affected the expression of cell cycle-related proteins. The binding mode was also probed by molecular docking.
    Keywords:  G2/M phase arrest; cell apoptosis; quinoxaline derivatives; reactive oxygen species; tubulin polymerization inhibitors
    DOI:  https://doi.org/10.1111/cbdd.13459
  13. Free Radic Res. 2019 Jan 09. 1-11
      Therapeutic efficacy of nucleoside analogues (NAs) like Gemcitabine, 5-fluorouracil in cancer treatment is already well established. Most of the known NAs are highly toxic to normal cells due to its non-specific action; thus searching for non-toxic NAs are still going on. For that purpose we have synthesised nine different NAs by alteration of their structural and functional groups. The aim of present study is to investigate the therapeutic potential of NAs against mice bearing breast adenocarcinoma cells at IC50 dose for 10 days treatment schedule. Results of the present study showed that, among the seven nucleoside analogues, NA-7 and NA-9 showed maximum therapeutic efficacy in controlling cancer cells by inhibiting cell proliferation and inducing apoptosis without any adverse effects to normal host cells. Additionally, NAs significantly decreased the tumour burden and enhanced survivability of host through generation of reactive oxygen species in tumour cells. These ultimately led to DNA damage, depolarisation of mitochondrial membrane potential and apoptosis in tumour cells. To find out the molecular mechanisms, we showed that administration of NA-7 and NA-9, down- regulating the expression of Bcl-2, cyclin D1, C-myc, P-21 and up-regulating the expression of P-53, Cyt-c, Bax, caspase-3 and caspase-9. The results suggest that NA-7 and NA-9 exhibits significant antitumor activity than 5-fluorouracil by modulating the cell cycle checkpoints and inducing apoptosis in Ehrlich ascites carcinoma (EAC)-bearing mice. Additionally, NA-7 and NA-9 did not show any clastogenic effect on bone marrow cells at sub-lethal dose. Thus, the present study clearly suggested therapeutic benefit of NAs by augmenting anticancer efficacy and diminishing toxicity to the host.
    Keywords:  Apoptosis; cell cycle; chemotherapy; ehrlich’s ascites carcinoma; nucleoside analogues; reactive oxygen species (ROS) mediated cell death
    DOI:  https://doi.org/10.1080/10715762.2018.1551999
  14. Food Nutr Res. 2018 ;62
      Oxidative stress is more likely to cause damage to neuronal cells and mediates some neurodegenerative disorders. It is well known that natural antioxidants can prevent oxidative stress damage and become a potential therapeutic strategy. Noni juice obtained from the fruit of the tree Morinda citrifolia, as a folk medicine, has been used for over two thousand years. In the current study, the neuroprotective effect and mechanism of noni juice extracts against tert-Butyl hydroperoxide (TBHP)-induced SH-SY5Y cell damage were investigated. The results demonstrated that chloroform fraction (CF) and aqueous fraction (AF) of noni juice protected SH-SY5Y cells against TBHP-induced oxidative stress and the associated apoptosis effectively. CF and AF treatment significantly weakened the TBHP-induced cytotoxicity, reactive oxygen species generation, mitochondrial membrane depolarization, and apoptotic features. CF and AF restored cellular antioxidant enzyme activity; upregulated expression of heme oxygenase-1, catalase, and superoxide dismutase-1; and increased the nuclear accumulation of nuclear factor-erythroid 2 related factor 2 (Nrf2). The antioxidant and neuroprotection potential of CF may account for its high total phenolic and flavonoid content, while AF may be rich in polysaccharides. These results suggest that CF and AF exhibit antioxidant defense through the upregulation of Nrf2 along with endogenous antioxidants and reduce apoptosis via inhibiting the mitochondrial pathway to protect SH-SY5Y cells damaged by TBHP. CF and AF might be developed as agents for neurodegeneration prevention or therapy.
    Keywords:  Nrf2; apoptosis; neuroprotective; noni juice; oxidative stress
    DOI:  https://doi.org/10.29219/fnr.v62.1605
  15. Reprod Toxicol. 2019 Jan 08. pii: S0890-6238(18)30467-2. [Epub ahead of print]
      We previously investigated excessive fluoride exposure elicited intracellular endoplasmic reticulum (ER) stress and led to Sertoli cells dysfunction in vitro. However, the mechanisms underlying fluoride-mediated male reproductive damage in vivo remain largely unknown. Considerable evidence has now revealed ER stress is closely linked with testicular oxidative damage. Hence, we aimed to explore whether ER stress signaling was involved in the testicular protective effects of antioxidant N-acetylcysteine (NAC) against testicular apoptosis induced by fluoride. Male SD rats were oral gavaged with sodium fluoride (NaF) for 7 weeks to induce fluorosis. The animals were pretreatment with or without NAC (150 mg/Bw•d). Our results demonstrated that sub-chronic NaF exposure triggered testicular apoptosis and sex hormonal disturbance in pituitary-testicular (PT) axis, promoted oxidative stress and the expression of ER stress mediators. Antioxidant NAC, however, prevented NaF-induced testicular apoptosis accompanied by activating Nrf2-mediated antioxidant potential. Simultaneously, NAC pretreatment downregulated XBP1 splicing, reduced JNK phosphorylation and further blocked cleavage of caspase-3, all these might contribute to the inhibition of testicular cell apoptosis. Collectively, the present results suggested that prolonged administration of NAC preserved testicular function and normalized sex hormonal disruption induced by NaF via the inhibition of Nrf2-associated oxidative damage and Ire1α-JNK-mediated apoptosis in rat testis.
    Keywords:  Apoptosis; Endoplasmic reticulum (ER) stress; Fluoride; NF-E2-related factor 2 (Nrf2); Reproductive toxicity
    DOI:  https://doi.org/10.1016/j.reprotox.2019.01.001
  16. Molecules. 2019 Jan 07. pii: E195. [Epub ahead of print]24(1):
      We investigated whether 14 phenolic compounds isolated from Artemisia argyi could prevent the apoptotic damage caused by iodixanol, an iodinated contrast agent, on LLC-PK1 cells. Iodixanol was used to induce cytotoxicity in LLC-PK1 cells. Apoptotic cell death was observed as the fluorescence intensity emitted by annexin V and Hoechst 33342 stains. Western blotting was used to detect specific proteins. Seven phenolic compounds protected against iodixanol-induced LLC-PK1 cell death in a concentration-dependent manner. Among them, methyl caffeate exerted the strongest protective effect, and co-treatment with 50 and 100 μM methyl caffeate decreased intracellular reactive oxygen species elevated by 25 mg/mL iodixanol. In addition, the treatment of LLC-PK1 cells with iodixanol resulted in an increase in apoptotic cell death, which decreased by co-treatment with methyl caffeate. Iodixanol caused a cytotoxicity-related increase in the phosphorylation of extracellular-signal-regulated kinase, c-Jun N-terminal kinase, and P38; and a similar increase in the expression levels of kidney injury molecule-1 and cleaved caspase-3. However, the up-regulation of these proteins was reversed by co-treatment with methyl caffeate. These findings suggest that phenolic compounds isolated from A. argyi play an important role in protecting kidney epithelium cells against apoptotic damage caused by iodixanol.
    Keywords:  MAPKs; caspase-3; contrast agent; cytotoxicity; iodixanol; kidney injury molecule-1
    DOI:  https://doi.org/10.3390/molecules24010195
  17. J Neurosci. 2019 Jan 09. pii: 3415-17. [Epub ahead of print]
      Mitochondrial energy production is essential for normal brain function. Traumatic brain injury (TBI) increases brain energy demands, results in the activation of mitochondrial respiration, associated with enhanced generation of reactive oxygen species. This chain of events triggers neuronal apoptosis via oxidation of a mitochondria-specific phospholipid, cardiolipin (CL). One pathway through which cells can avoid apoptosis is via elimination of damaged mitochondria by mitophagy. Previously, we showed that externalization of CL to the mitochondrial surface acts as an elimination signal in cells. Whether CL-mediated mitophagy occurs in vivo or its significance in the disease processes are not known. In this study, we showed that TBI leads to increased mitophagy in the human brain, which was also detected using TBI models in male rats. Knockdown of CL synthase, responsible for de novo synthesis of CL, or phospholipid scramblase-3, responsible for CL translocation to the outer mitochondrial membrane, significantly decreased TBI-induced mitophagy. Inhibition of mitochondrial clearance by 3-methyladenine, mdivi-1, or phospholipid scramblase-3 knockdown after TBI led to a worse outcome, suggesting that mitophagy is beneficial. Taken together, our findings indicate that TBI-induced mitophagy is an endogenous neuroprotective process that is directed by CL, which marks damaged mitochondria for elimination, thereby limiting neuronal death and behavioral deficits.SIGNIFICANCE STATEMENTTBI increases energy demands leading to activation of mitochondrial respiration associated with enhanced generation of reactive oxygen species and resultant damage to mitochondria. We demonstrate that the complete elimination of irreparably damaged organelles via mitophagy is activated as an early response to TBI. This response includes translocation of mitochondria phospholipid CL from the inner membrane to the outer membrane where externalized cardiolipin mediates targeted LC3-mediated autophagy of damaged mitochondria. Our data on targeting phospholipid scramblase and cardiolipin synthase in genetically manipulated cells and animals strongly support the essential role of cardiolipin externalization mechanisms in the endogenous reparative plasticity of injured brain cells. Furthermore, successful execution and completion of mitophagy is beneficial in the context of preservation of cognitive functions after TBI.
    DOI:  https://doi.org/10.1523/JNEUROSCI.3415-17.2018
  18. Environ Toxicol. 2019 Jan 07.
      Genistein, a major isoflavone compound in soybeans, has been shown to have biological activities including anti-cancer activates. In the present, we investigated the anti-leukemia activity of genistein on HL-60 cells in vitro. The percentage of viable cell, cell cycle distribution, apoptotic cell death, reactive oxygen species (ROS), and Ca2+ production and the level of ΔΨm were measured by flow cytometric assay. Cell apoptosis and endoplasmic reticulum (ER) stress associated protein expressions were examined by Western blotting assay. Calpain 1, GRP78, and GADD153 expression were measured by confocal laser microscopy. Results indicated that genistein-induced cell morphological changes, decreased the total viable cells, induced G2 /M phase arrest and DNA damage and fragmentation (cell apoptosis) in HL-60 cells. Genistein promoted ROS and Ca2+ productions and decreased the level of ΔΨm in HL-60 cells. Western blotting assay demonstrated that genistein increased ER stress-associated protein expression such as IRE-1α, Calpain 1, GRP78, GADD153, caspase-7, caspase-4, and ATF-6α at 20-50 μM treatment and increased apoptosis associated protein expression such as pro-apoptotic protein Bax, PARP-cleavage, caspase-9, and -3, but decreased anti-apoptotic protein such as Bcl-2 and Bid in HL-60 cells. Calpain 1, GRP78, and GADD153 were increased in HL-60 cells after exposure to 40 μM of genistein. In animal xenografted model, mice were intraperitoneally injected with genistein (0, 0.2, and 0.4 mg/kg) for 28 days and the body weight and tumor volume were recorded. Results showed that genistein did not affect the body weights but significantly reduced the tumor weight in 0.4 mg/kg genistein-treated group. Genistein also increased the expressions of ATF-6α, GRP78, Bax, Bad, and Bak in tumor. In conclusion, genistein decreased cell number through G2 /M phase arrest and the induction of cell apoptosis through ER stress- and mitochondria-dependent pathways in HL-60 cells and suppressed tumor properties in vivo.
    Keywords:  Genistein; HL-60 human leukemia cancer cells; apoptosis; endoplasmic reticulum stress; mitochondria dysfunction
    DOI:  https://doi.org/10.1002/tox.22698
  19. J Toxicol Sci. 2019 ;44(1): 35-45
      Due to its excellent properties such as ultraviolet obscuration, chemical stability and small particle size, nano-titanium dioxide (nano-TiO2) is widely used, particularly in sunblock products. The skin is therefore a chief route for exposure. Studies have found that oral or respiratory exposure to nano-TiO2 has an adverse impact on the cardiovascular system. The relationship between dermal exposure to nano-TiO2 and cardiovascular system toxicity, particularly the causative mechanisms, remain unclear. In this study, we used Balb/c mice to evaluate cardiovascular toxicity from nano-TiO2 dermal exposure, and the underlying mechanisms associated with the human umbilical vein endothelial cells (HUVECs) were explored. Our results showed that nano-TiO2 treatment resulted in an obvious increase in reactive oxygen species and 8-hydroxy-2'-deoxyguanosine, indicating the appearance of oxidative stress. Moreover, the levels of inflammatory biomarkers, such as immunoglobulin E, soluble intercellular adhesion molecule-1, interleukin-8, and hypersensitive C-reactive protein, also increased. Exposing HUVECs to nano-TiO2 led to a decline in cell vitality, and an increase in caspase-3 levels, suggesting that nano-TiO2 exposure caused cytotoxicity and even cell apoptosis. Interestingly, neutralizing oxidative stress by administering Vitamin E was shown to reduce the inflammatory response and cytotoxicity. Our findings suggest that nano-TiO2 can injure the cardiovascular system via dermal exposure, and does this via oxidative stress-induced inflammation and cytotoxicity. Vitamin E treatment may be a strategy to mitigate the damage.
    Keywords:  Apoptosis; Cardiovascular toxicity; Cytotoxicity; Inflammation; Nano-titanium dioxide (nano-TiO2); Oxidative stress
    DOI:  https://doi.org/10.2131/jts.44.35
  20. Biomed Pharmacother. 2019 Jan 03. pii: S0753-3322(18)37290-1. [Epub ahead of print]111 676-685
      Olive leaf extract (OLE) has potential health benefits and protects against cytotoxicity in different organs. However, nothing has yet been reported on its potential to prevent cyclophosphamide (CP)-induced nephrotoxicity. This study investigated the possible protective effect of OLE on CP-induced kidney injury in rats, focusing on oxidative stress, inflammation, apoptosis and Nrf2/ARE/HO-1 signaling. Rats received 100 or 200 mg/kg body weight OLE for 15 days and a single injection of 150 mg/kg CP at day 16. CP induced kidney injury evidenced by the significantly increased serum creatinine and urea, and histopathological alterations, including glomerular atrophy, interstitial hemorrhage, dilated urinary space and necrosis. CP-induced rats exhibited increased kidney lipid peroxidation, protein carbonyl, nitric oxide (NO) and pro-inflammatory cytokines, and up-regulated NF-κB, Bax, cytochrome c and caspase-3. OLE ameliorated kidney function markers and prevented CP-induced tissue damage. In addition, OLE significantly prevented oxidative stress, inflammation and apoptosis by enhancing the antioxidant defenses and Bcl-2 expression, and suppressing the pro-inflammatory and pro-apoptotic markers NF-κB, Bax, cytochrome c and caspase-3. OLE up-regulated Nrf2, HO-1 and NQO-1 expression in the kidney of CP-induced rats. In conclusion, OLE has a substantial protective role against CP-induced nephrotoxicity in rats by up-regulating the Nrf2/ARE/HO-1 signaling, enhancing the antioxidant activity and attenuating inflammation and apoptosis.
    Keywords:  Acrolein; Apoptosis; Cyclophosphamide; Nephrotoxicity; Olive leaf; Oxidative stress
    DOI:  https://doi.org/10.1016/j.biopha.2018.12.112
  21. Molecules. 2019 Jan 10. pii: E247. [Epub ahead of print]24(2):
      Diabetic nephropathy (DN) is a leading cause of end-stage renal failure, and no effective treatment is available. Notoginsenoside R1 (NGR1) is a novel saponin that is derived from Panax notoginseng, and our previous studies showed the cardioprotective and neuroprotective effects of NGR1. However, its role in protecting against DN remains unexplored. Herein, we established an experimental model in db/db mice and HK-2 cells exposed to advanced glycation end products (AGEs). The in vivo investigation showed that NGR1 treatment increased serum lipid, β2-microglobulin, serum creatinine, and blood urea nitrogen levels of db/db mice. NGR1 attenuated histological abnormalities of kidney, as evidenced by reducing the glomerular volume and fibrosis in diabetic kidneys. In vitro, NGR1 treatment was further found to decrease AGE-induced mitochondria injury, limit an increase in reactive oxygen species (ROS), and reduce apoptosis in HK-2 cells. Mechanistically, NGR1 promoted nucleus nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) expressions to eliminate ROS that induced apoptosis and transforming growth factor beta (TGF-β) signaling. In summary, these observations demonstrate that NGR1 exerts renoprotective effects against DN through the inhibition of apoptosis and renal fibrosis caused by oxidative stress. NGR1 might be a potential therapeutic medicine for the treatment of DN.
    Keywords:  HK-2 cells; apoptosis; db/db mice; diabetes mellitus; diabetic nephropathy
    DOI:  https://doi.org/10.3390/molecules24020247
  22. Andrologia. 2019 Jan 08. e13227
      The aim of this study was to investigate possible protective effects of apocynin (APO), an NADPH oxidase (NOX2) inhibitor, on cisplatin (CIS)-induced testicular damage. Four groups of Sprague Dawley rats were used: control, APO, CIS and CIS+APO. Following a single intraperitoneal dose of CIS (7 mg/kg), either dimethyl sulfoxide or APO (25 mg/kg) was administered orally for 5 days. Testis samples were evaluated microscopically for general histopathology and ultrastructure, proliferating and apoptotic cells, and NOX2 localization. Sperm parameters were evaluated. Malondialdehyde (MDA) and glutathione (GSH) levels and superoxide dismutase (SOD), myeloperoxidase (MPO) and 8-hydroxy-2-deoxyguanosine (8-OHdG) activities were analysed biochemically. The CIS group had a greater number of abnormal spermatozoa, atrophic seminiferous tubules, apoptotic and NOX2-immunoreactive cells; numerous large vacuole formations in the cytoplasm of germinal epithelial cells; degenerated intercellular tight junctions; higher MDA, 8-OHdG and MPO levels; decreased numbers of spermatozoa; and lower proliferative index and GSH and SOD levels. All these histologic and biochemical results were better in the CIS+APO group. CIS causes testicular damage by decreasing spermatogenic cell lines and increasing NOX2 activity and apoptosis through oxidative stress. APO prevents testicular damage, possibly by its antioxidant effects.
    Keywords:  NOX2; apocynin; apoptosis; cisplatin; testis
    DOI:  https://doi.org/10.1111/and.13227
  23. J Cell Physiol. 2019 Jan 08.
      Cancer incidences are growing and cause millions of deaths worldwide. Cancer therapy is one of the most important challenges in medicine. Improving therapeutic outcomes from cancer therapy is necessary for increasing patients' survival and quality of life. Adjuvant therapy using various types of antibodies or immunomodulatory agents has suggested modulating tumor response. Resistance to apoptosis is the main reason for radioresistance and chemoresistance of most of the cancers, and also one of the pivotal targets for improving cancer therapy is the modulation of apoptosis signaling pathways. Apoptosis can be induced by intrinsic or extrinsic pathways via stimulation of several targets, such as membrane receptors of tumor necrosis factor-α and transforming growth factor-β, and also mitochondria. Curcumin is a naturally derived agent that induces apoptosis in a variety of different tumor cell lines. Curcumin also activates redox reactions within cells inducing reactive oxygen species (ROS) production that leads to the upregulation of apoptosis receptors on the tumor cell membrane. Curcumin can also upregulate the expression and activity of p53 that inhibits tumor cell proliferation and increases apoptosis. Furthermore, curcumin has a potent inhibitory effect on the activity of NF-κB and COX-2, which are involved in the overexpression of antiapoptosis genes such as Bcl-2. It can also attenuate the regulation of antiapoptosis PI3K signaling and increase the expression of MAPKs to induce endogenous production of ROS. In this paper, we aimed to review the molecular mechanisms of curcumin-induced apoptosis in cancer cells. This action of curcumin could be applicable for use as an adjuvant in combination with other modalities of cancer therapy including radiotherapy and chemotherapy.
    Keywords:  JNK; NF-κB; apoptosis; cancer; curcumin; p53
    DOI:  https://doi.org/10.1002/jcp.28122
  24. J Colloid Interface Sci. 2018 Dec 27. pii: S0021-9797(18)31534-0. [Epub ahead of print]539 575-584
      Development of effective inhibitors toward Aβ aggregation and reactive oxygen species (ROS) scavengers are of crucial therapeutic implications for Alzheimer's disease (AD). Herein, a novel agent with dual enzyme mimic activities has been fabricated as a multifunctional Aβ fibrillation modulator. MoO3-x nanodots were synthesized by pulsed laser ablation (PLA) method in MoS2 nanosheets solutions, which may act directly as numerous fine targets. MoO3-x nanodots showed a uniform and monodispersed morphology, and the tiny dots were around 3-5 nm with a narrow size distribution. Due to the efficient charge transition between Mo5+/Mo6+ on the dots surface, MoO3-x nanodots exhibited excellent catalase and SOD mimic activities, which were adopted to alleviate Aβ-mediated oxidative stress. Moreover, MoO3-x nanodots can efficiently inhibit Aβ aggregation and destabilize the preformed fibrils, and eventually protect neuronal cells from apoptosis induced by Aβ. Taken together, MoO3-x nanodots with multifunctional roles can act as a potential therapeutic strategy for treatment of amyloid induced neurotoxicity.
    Keywords:  Amyloid; Enzyme-mimic activity; MoO(3−x) nanodots; Neurotoxicity; Pulsed laser ablation
    DOI:  https://doi.org/10.1016/j.jcis.2018.12.093
  25. J Int Med Res. 2019 Jan 07. 300060518805302
      OBJECTIVE: The aim of this study was to observe the effect of Ku86 on cellular senescence and apoptosis induced by various doses of ionizing radiation in human umbilical vein endothelial cells (HUVECs).METHODS: Senescence-associated β-galactosidase activity was detected to evaluate cell senescence. Apoptosis was determined by flow cytometry and a caspase enzyme determination kit. p16Ink4a, Sirt1, superoxide dismutase 2 (SOD2), xanthine oxidase (XOD), and Bcl-2 protein expression levels were measured by western blotting.
    RESULTS: Low doses of ionizing radiation induced cellular senescence and apoptosis in a dose-dependent manner. The Ku86 protein was negatively correlated with ionization intensity. After transfection of Ku86 with a vector (pcDNA 3.1), or interference with siRNA (si-Ku86), apoptosis/senescence and related protein expression were observed. Western blot results revealed that this induction of senescence was associated with activated Sirt1 and SOD2, and downregulation of p16Ink4a and XOD in 0.2 Gy ionizing radiation. The expression levels of apoptosis-associated proteins, such as Bcl-2, cleaved caspase-3, caspase-8, and caspase-9, were significantly altered in both the presence and absence of Ku86 with ionizing radiation (0.2 Gy).
    CONCLUSIONS: Our study revealed that Ku86 overexpression inhibits HUVEC apoptosis and senescence induced by low doses of ionizing radiation.
    Keywords:  DNA damage; DNA repair; HUVEC; Ku86; Senescence; apoptosis; ionizing radiation; reactive oxygen species
    DOI:  https://doi.org/10.1177/0300060518805302
  26. J Appl Toxicol. 2019 Jan 08.
      We recently synthesized ZnO nanomaterials (denoted as ZnO nanorods [NRs] and Mini-NRs) and suggested that their cytotoxicity could be related with the activation of endoplasmic reticulum (ER) stress apoptosis. However, in a complex biological microenvironment, the ER stress-apoptosis pathway could also be modulated by biological molecules, such as free fatty acids, leading to unpredicted biological effects. In this study, we investigated the combined toxicity of ZnO NRs/Mini-NRs and palmitate (PA) to THP-1 macrophages. PA influenced the zeta potential and solubility of ZnO NRs and ZnO Mini-NRs in water, which indicated a change of colloidal stability. Exposure to ZnO NRs and Mini-NRs dose-dependent decreased cellular viability and release of soluble monocyte chemotactic protein 1 (sMCP-1), and these effects were significantly promoted with the presence of PA. However, ZnO NR- and Mini-NR-induced intracellular Zn ions or reactive oxygen species were not significantly affected by PA. ZnO NRs and ZnO Mini-NRs significantly promoted the expression of ER stress genes HSPA5, DDIT3, XBP-1s and apoptotic gene CASP3, whereas PA also modestly promoted the expression of HSPA5, DDIT3 and CASP3. Interestingly, the ER stress inducer thapsigargin showed a similar effect as PA to promote the cytotoxicity of ZnO NRs and ZnO Mini-NRs. It is suggested that PA might promote the cytotoxicity of ZnO NRs and ZnO Mini-NRs possibly by promoting ER stress.
    Keywords:  THP-1 macrophages; ZnO nanomaterials; cytotoxicity; endoplasmic reticulum (ER) stress; palmitate
    DOI:  https://doi.org/10.1002/jat.3768
  27. Toxicol Appl Pharmacol. 2019 Jan 08. pii: S0041-008X(19)30005-5. [Epub ahead of print]
      Natural products derived from herbal medicines have become a major focus of anti-cancer drug discovery studies. Acetyl-macrocalin B (A-macB) is an ent-diterpenoid isolated from Isodon silvatica. This study aimed to examine the effect and molecular action of A-macB in esophageal squamous cell carcinoma (ESCC) and explore possible drug synergistic modalities. A-macB induced cellular reactive oxygen species (ROS) generation, initiated the p38 mitogen-activated protein kinase (MAPK) signaling pathway, and triggered the caspase-9-dependent apoptosis cascade in ESCC cells. The ROS scavenger N-acetylcysteine (NAC) and the specific p38 inhibitor SB203580 reversed the effects of A-macB on the p38 network and thus rescued ESCC cells from apoptosis. The cellular ROS increase was at least partially due to the suppression of glutathione-S-transferase P1 (GSTP1) by A-macB. A-macB also upregulated the Chk1/Chk2-Cdc25C/Cdc2/Cyclin B1 axis to induce G2/M phase arrest. The cell growth inhibition induced by A-macB was further enhanced by AZD7762, a specific Chk1/Chk2 inhibitor, with a combination index (CI) of <1. Moreover, A-macB efficiently suppressed xenograft growth without inducing significant toxicity, and AZD7762 potentiated the effects of A-macB in the suppression of tumor growth in vivo. Taken together, A-macB is a promising lead compound for ESCC and exerts synergistic anti-cancer effects with AZD7762.
    Keywords:  Acetyl-macrocalin B; Chk1/Chk2; Esophageal squamous cell carcinoma; Reactive oxygen species; p38 MAPK
    DOI:  https://doi.org/10.1016/j.taap.2019.01.005
  28. J Ethnopharmacol. 2019 Jan 02. pii: S0378-8741(18)30780-3. [Epub ahead of print]
      ETHNOPHARMACOLOGICAL RELEVANCE: Chrysanthemum indicum Linne (C. indicum), a healthy food and folk medicine in China for thousands of years, has been reported to exert heat-clearing and detoxifying effects and extensively applied to treat various symptoms such as inflammation diseases, hepatitis and headache.AIM OF THIS STUDY: The purpose of the present study was to investigate the protective effect of the supercritical carbon dioxide fluid extract from flowers and buds of C. indicum (CISCFE) on D-galactose-induced brain and liver damage during aging process and to illuminate the underlying mechanisms.
    MATERIALS AND METHODS: Mice were orally administrated with CISCFE (100, 150 and 300mg/kg) after injection with D-galactose. 24h after the last administration, the blood samples, whole brain and liver tissues were collected for biochemical analysis, histological examination and western blot analysis. The body weight, spleen and thymus indexes, alanine transaminase (ALT), aspartate transaminase (AST), total antioxidant capacity (T-AOC), superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GSH-Px), malondialdehyde (MDA) in brain and liver, interleukin-1β (IL-1β), interleukin-6 (IL-6), and necrosis factor-α (TNF-α) were detected. Besides, the expressions of Bax, Bcl-2 and cleaved caspase-3 were determined by western blot assay.
    RESULTS: The results indicated that CISCFE effectively increased the suppressed body weight, attenuated the decline of thymus and spleen indexes, and reduced the elevated levels of ALT and AST induced by D-gal. Furthermore, CISCFE might notably alleviate D-gal-induced abnormal alterations in structure and function of brain and liver dose-dependently via renewing normal antioxidant enzymes activities (SOD, CAT, GSH-Px), reducing MDA accumulation, decreasing inflammatory cytokines productions (IL-1β, IL-6, TNF-α), as well as attenuating the increase of Bax/Bcl-2 ratio and cleaved caspase-3 activation in the liver and brain.
    CONCLUSIONS: Taken together, our present results suggested that CISCFE treatment could effectively mitigate the D-gal-induced hepatic and cerebral injury, and the underlying mechanism might be tightly related to the decreased oxidative stress, inflammation and apoptosis, indicating CISCFE might be an alternative and promising agent for the treatment of aging and age-associated brain and liver diseases.
    Keywords:  Chrysanthemum indicum Linnén; D-galactose; D-galactose (PubChem CID:6036); Tween-80 (PubChem CID:5281955); Vitamin E (PubChem CID: 14985); anti-apoptotic; anti-inflammatory; anti-oxidative; senescent
    DOI:  https://doi.org/10.1016/j.jep.2018.12.050
  29. Theranostics. 2018 ;8(21): 5855-5869
      Non-coding RNAs (ncRNAs) are considered major players in physiological and pathological processes based on their versatile regulatory roles in different diseases including cardiovascular disease. Circular RNAs (circRNAs), a newly discovered class of RNAs, constitute a substantial fraction of the mammalian transcriptome and are abundantly expressed in the cardiovascular system. However, the regulatory functions of these circRNAs in ischemic cardiac disease remain largely unknown. Here, we investigated the role of a circRNA transcribed from the sodium/calcium exchanger 1 (ncx1) gene, named circNCX1, in oxidative stress-induced cardiomyocyte apoptosis during ischemic myocardial injury. Methods: Divergent polymerase chain reaction (PCR) was conducted to amplify the circRNA. The circular structure of circNCX1 was verified by Sanger sequencing and RNase R digestion. The subcellular localization of circNCX1 was detected by fluorescence in situ hybridization (FISH). To test the expression pattern and function of circNCX1 during oxidative stress, H9c2 cells and neonatal rat cardiomyocytes were treated with H2O2 or hypoxia-reoxygenation (H/R). Mechanistically, the interaction of circNCX1 with miRNA was examined by AGO2-IP and RNA pull-down assays. The regulatory role of circNCX1 in target gene expression was tested by western blot and luciferase reporter assays. At the animal level, we constructed a myocardial ischemia-reperfusion (I/R) mouse model to analyze the effect of circNCX1 on heart function, cardiomyocyte apoptosis and cardiac remodeling. Results: circNCX1 was increased in response to reactive oxygen species (ROS) and promotes cardiomyocyte apoptosis by acting as an endogenous miR-133a-3p sponge. Due to competitive binding of circNCX1 to miR-133a-3p, the suppressive activity of pro-apoptotic gene cell death-inducing protein (CDIP1) by miR-133a-3p was reduced. Knockdown of circNCX1 in murine cardiomyocytes and heart tissues reduced the levels of CDIP1 and attenuated the apoptosis and I/R injury. Conclusions: Our findings reveal a novel regulatory pathway that comprises circNCX1, miR-133a-3p and CDIP1, that is involved in cardiomyocyte apoptosis. This pathway may serve as a potential therapeutic avenue for ischemic heart diseases.
    Keywords:  CDIP1; cardiomyocyte apoptosis; circular RNA; ischemic cardiomyopathy; miR-133a
    DOI:  https://doi.org/10.7150/thno.27285
  30. Cell Stress Chaperones. 2019 Jan 10.
      Hyperglycemia induces retinal pigmented epithelial cell apoptosis and mitochondrial stress via poorly understood mechanisms. The goal of our current study is to explore whether mammalian sterile 20-like kinase 1 (Mst1) is involved in the pathogenesis of hyperglycemia-mediated retinal pigmented epithelial cell apoptosis by triggering mitochondrial abnormalities and activating the Smad2 signaling pathway. Retinal pigmented epithelial ARPE-19 cells were presented with a high-glucose challenge in vitro. Cell viability and apoptosis were measured via western blotting, ELISAs, and immunofluorescence assays. Mitochondrial function was detected via JC-1 staining, mitochondrial ROS flow cytometry, western blotting, and ELISAs. Loss- and gain-of-function assays were performed via cell transfection and transduction with Mst1 siRNA and Smad2 adenovirus, respectively. The results indicated that hyperglycemia treatment upregulated the levels of Mst1, an effect that was accompanied by an increase in ARPE-19 cell apoptosis. Loss of Mst1 attenuated hyperglycemia-induced cell apoptosis, and this effect seemed to be associated with mitochondrial protection. In response to hyperglycemia stimulus, mitochondrial stress was noted in ARPE-19 cells, including mitochondrial ROS overproduction, mitochondrial respiratory metabolism dysfunction, mitochondrial fission/fusion imbalance, and mitochondrial apoptosis activation. Further, we provided evidence to support the crucial role played by Smad2 in promoting Mst1-mediated cell apoptosis and mitochondrial stress. Overexpression of Smad2 abrogated the beneficial effects of Mst1 deletion on ARPE-19 cell viability and mitochondrial protection. Altogether, our results identified Mst1 as a novel mediator controlling the fate of retinal pigmented epithelial cells and mitochondrial homeostasis via the Smad2 signaling pathway. Based on this finding, strategies to repress Mst1 upregulation and block Smad2 activation are vital to alleviate hyperglycemia-mediated retinal pigmented epithelial cell damage.
    Keywords:  Mitochondria; Mst1; Retinal pigmented epithelial cell; Smad2
    DOI:  https://doi.org/10.1007/s12192-018-00963-z
  31. J Cell Physiol. 2019 Jan 11.
      Long noncoding RNA UCA1 has exerted a significant effect in cardiovascular disease. The biological role of UCA1 in atherosclerosis is unclear. Our study was to identify the potential mechanisms in the progression of atherosclerosis. Here, we observed that ox-LDL increased UCA1 expression greatly in THP-1 cells. Knockdown of UCA1 greatly inhibited CD36 expression, a crucial biomarker in atherosclerosis. Meanwhile, 20 μg/ml ox-LDL induced foam cell formation, which can be reversed by downregulation of UCA1. In addition, TC and TG levels induced by ox-LDL was rescued by UCA1 small interfering RNA. Accumulating studies have indicated that oxidative stress contributes to atherosclerosis progression. Here, we also found that reactive oxygen species, MDA, and THP-1 cell apoptosis were restrained by decreased of UCA1 with an increase of the superoxide dismutase activity. Moreover, miR-206 was predicted as a target of UCA1 and knockdown of UCA1 was able to repress miR-206 expression. Furthermore, overexpression of miR-206 inhibited oxidative stress process and it was reversed by UCA1 upregulation in vitro. In conclusion, we indicated that UCA1 sponged miR-206 to exacerbate atherosclerosis events induced by ox-LDL in THP-1 cells.
    Keywords:  UCA1; atherosclerosis; miR-206; ox-LDL
    DOI:  https://doi.org/10.1002/jcp.28109
  32. Dose Response. 2018 Oct-Dec;16(4):16(4): 1559325818813061
      We reported that low-dose radiation (LDR) alleviated cardiotoxicity of doxorubicin (DOX) via inhibiting myocardial cell apoptosis and oxidative stress in vivo. Here, we tested whether LDR could enhance chemotherapeutic effect of DOX and alleviate myocardial injury induced by DOX by observing cell proliferation, apoptosis, and metastasis of heterotopic tumor in vivo. Mice implanted with 4T1 breast carcinoma cells were given 7.5 mg/kg DOX or 0.9% NaCl solution 72 hours after LDR (0 or 75 mGy). The histology of tumor tissue was observed by hematoxylin and eosin staining, the apoptosis was determined by terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling, and the expression of Ki67, Bcl-2, Bax, cleaved caspase3, matrix metalloproteinase 2 (MMP2), MMP9, and CD34 was detected by Western blot. Expression of Ki67 and CD34 was also detected by immunohistochemistry. Results showed that cell proliferation of the breast tumor and protein expression of the metastasis-related molecules were significantly reduced and the apoptosis of tumor cells was significantly increased in the LDR + DOX-treated tumor-bearing mice. Pretreatment with LDR significantly prevented DOX-induced cardiotoxicity likely through preventing DOX-induced mitochondrial Bcl2/Bax dyshomeostasis-induced caspase-3 cleavage-dependent apoptosis. These results suggested that LDR not only enhances DOX antitumor effect but also reduces DOX cardiotoxicity, which may potentially overcome the limitation for DOX clinical usage.
    Keywords:  breast carcinoma; chemosensitization; doxorubicin; low-dose radiation
    DOI:  https://doi.org/10.1177/1559325818813061
  33. Iran J Basic Med Sci. 2018 Dec;21(12): 1297-1304
      Objectives: To analyze the potential influences of propofol on the oxidative stress of H2O2-induced human esophageal squamous cell carcinoma (ESCC) Eca109 cell through mediating the Wnt/β-catenin signaling pathway.Materials and Methods: Eca109 cells were classified into 5 groups: Control group, H2O2 group, Propofol + H2O2 group, Dkk1 (Dickkopf-1, Wnt/β-catenin pathway antagonist) + H2O2 group, and Propofol + LiCl (Lithium chloride, Wnt/β-catenin pathway agonist) + H2O2 group. Western blotting was performed to determine the protein expressions, flow cytometry to measure the content of ROS, immunofluorescence staining to detect the oxidative DNA damage, as well as MTT, AnnexinV-FITC/PI, Wound-healing, and Transwell assays to test the biological characteristics of Eca109 cells.
    Results: H2O2 resulted in the increased nuclear and cytoplasmatic expression of β-catenin, reduced p-GSK3β expression, up-regulated ROS content, and induced oxidative DNA damage in Eca109 cells. Moreover, Eca109 cells treated with H2O2 alone had enhanced cell proliferation and metastasis but decreased cell apoptosis, as compared with those without any treatment; meanwhile, the declined Cyt C, Bax, and cleaved caspase-3, as well as the elevated Bcl-2 were also observed in Eca109 cells in the H2O2 group, which were reversed by Propofol or Dkk1. Moreover, Propofol could inhibit the effect of LiCl on activating the Wnt/β-catenin signaling pathway in H2O2-induced Eca109 cells.
    Conclusion: Propofol elicits protective effects to inhibit H2O2-induced proliferation and metastasis and promote apoptosis of Eca109 cells via blocking the Wnt/β-catenin pathway, offering a possible therapeutic modality for ESCC.
    Keywords:  Esophageal neoplasms; Hydrogen peroxide; Propofol; Wnt signaling pathway β-catenin
    DOI:  https://doi.org/10.22038/ijbms.2018.29141.7039
  34. Pharmacol Res. 2019 Jan 03. pii: S1043-6618(18)30899-5. [Epub ahead of print]141 214-223
      Senescence is a process characterized by an irreversible growth arrest in cells and induced by oxidative stress. In the current study, anti-aging potential of a well-known antioxidant, α-lipoic acid (α-LA), in rat embryonic fibroblast (REF) cells was assessed. In this regard, oxidative stress, inflammation, and apoptosis pathways were investigated on REF cells exposed to H2O2 as a senescence inducer and α-LA as a protective compound. In cells treated with α-LA and H2O2, level of β-galactosidase, as an aging marker, and oxidative stress biomarkers, were significantly lower than those exposed to H2O2 only. Furthermore, flow cytometry assay showed that α-LA caused a significant reduction in the number of apoptotic cells via the caspase-dependent pathway. In addition, it could neutralize the inflammatory effects of H2O2 and attenuated the concentration of inflammatory cytokines. In comparison to H2O2 group, a significant increase in G0/G1 arrest was observed during cell cycle analysis in cells exposed to H2O2 and α-LA. The results of this study show that α-LA has beneficial effects on H2O2-induced cellular senescence. α-LA works by attenuating the reactive oxygen species, subsiding inflammation, and affecting cell division.
    Keywords:  Aging; Cell cycle; Cellular senescence; Oxidative stress; Rat embryonic fibroblast cells; Toxicity; α -Lipoic acid
    DOI:  https://doi.org/10.1016/j.phrs.2019.01.003
  35. Biochem Pharmacol. 2019 Jan 08. pii: S0006-2952(19)30012-7. [Epub ahead of print]
      Leptin, a hormone primarily derived from adipose tissue, is known to induce tumor growth, but its underlying mechanisms of action are not clearly understood. Inflammasomes, acting as signaling platforms for controlling inflammatory responses, modulate tumor growth in a complicated manner. In this study, we investigated the role of inflammasomes in leptin-induced growth of breast cancer cells. Herein, we showed that leptin activated NLRP3 inflammasomes in MCF-7 breast cancer cells, as determined by activation of caspase-1, maturation of interleukin-1β, and increased expression of the inflammasome components, including NLRP3 and ASC. Interestingly, inhibition of the inflammasome by treatment with a pharmacological inhibitor of caspase-1 or gene silencing of NLRP3 prevented leptin-induced increase in cell viability. Moreover, suppression of apoptosis and cell cycle promotion by leptin were also significantly abolished by gene silencing of NLRP3, clearly indicating a crucial role of NLRP3 inflammasomes in leptin-induced breast cancer growth. In addition, inhibition of estrogen receptor signaling or ROS production markedly blocked leptin-induced activation of NLRP3 inflammasomes, suggesting that estrogen receptor signaling and ROS production mediate inflammasomes activation by leptin. The stimulatory effect of leptin on inflammasomes activation was also observed in MCF-7 tumor xenograft model. Furthermore, the critical roles of inflammasomes activation in leptin-induced tumor growth, suppression of apoptotic gene expression, and induction of the genes stimulating cell cycle were confirmed in a tumor xenograft model. Taken together, these results demonstrate that inflammasomes activation plays a pivotal role in leptin-induced growth of breast cancer cells via modulation of both apoptosis and cell cycle.
    Keywords:  Breast cancer; Estrogen receptor; Inflammasomes; Leptin; ROS
    DOI:  https://doi.org/10.1016/j.bcp.2019.01.006
  36. J Cell Biochem. 2019 Jan 10.
      Amyloid-β (Aβ) has been reported to cause oxidative damage of neurons leading to neurotoxicity in a variety of diseases and cancers. As an anticancer drug, brusatol (BR) has been shown to have potent cytotoxic effects on various cancer cell lines. In this study, the effect and mechanism of BR on Aβ-induced neurotoxicity was investigated in U-251 glioma cells. Using the MTT assay, the results suggest that BR ameliorated cell injury induced by Aβ in U-251 cells. After running Hoechst and Western blot assays, BR prevented cell apoptosis induced by Aβ in U-251 cells. In addition, BR inhibited the increased reactive oxygen species and mitochondrial membrane potential levels induced by Aβ in U-251 cells using the DCFH-DA and Rh123 method. Furthermore, BR induced the Nrf2/HO-1 pathway by inhibiting the PI3K/AKT/mTOR pathway to inhibit neurotoxicity elicited by Aβ. These results suggest that brustasol is a valuable potential antitumor drug available for chemotherapy.
    Keywords:  Nrf2/HO-1 pathway; U-251 cells; amyloid-β (Aβ); brusatol (BR); neurotoxicity
    DOI:  https://doi.org/10.1002/jcb.28341
  37. In Vitro Cell Dev Biol Anim. 2019 Jan 07.
      Human menstrual blood-derived mesenchymal stem cells (MenSCs) hold great promise for regenerative medicine. Here, H2O2-associated damage in H9c2 cells was employed as an in vitro ischemia-reperfusion model, and the transwell system was used to explore the beneficial effects of MenSCs on the H2O2-induced damage of myocardial H9c2 cells. H2O2 treatment resulted in decreased viability and migration rate, with increased apoptosis levels in cells. By contrast, upon co-culture with MenSCs, H9c2 cell viability and migration were increased, whereas the apoptotic rate decreased. Additionally, western blot and qRT-PCR showed that MenSCs mediated the anti-apoptotic role by downregulating the pro-apoptotic genes Bax and caspase-3, while upregulating the anti-apoptotic effector Bcl-2. Furthermore, co-culture with MenSCs resulted in elevated expression of N-cadherin after H2O2 treatment. These findings indicate that MenSCs protect H9c2 cells against H2O2-associated programmed cell death and would help develop therapeutic tools for cardiomyocyte apoptosis associated with oxidative stress.
    Keywords:  Apoptosis; H9c2 cell; Menstrual blood–derived mesenchymal stem cell; Myocardial ischemia–reperfusion injury
    DOI:  https://doi.org/10.1007/s11626-018-0311-1
  38. Theranostics. 2018 ;8(21): 5870-5889
      Hypoxia-induced radioresistance is the primary reason for failure of tumor radiotherapy (RT). Changes within the irradiated tumor microenvironment (TME) including oxygen, reactive oxygen species (ROS) and nitric oxide (NO) are closely related to radioresistance. Therefore, there is an urgent need to develop new approaches for overcoming hypoxic radioresistance by incorporating TME regulation into current radiotherapeutic strategies.METHODS: Herein, we explored a radiation-responsive nanotheranostic system to enhance RT effects on hypoxic tumors by multi-way therapeutic effects. This system was developed by loading S-nitrosothiol groups (SNO, a NO donor) and indocyanine green (ICG, a photosensitizer) onto mesoporous silica shells of Eu3+-doped NaGdF4 scintillating nanocrystals (NSC).
    RESULTS: Under X-ray radiation, this system can increase the local dosage by high-Z elements, promote ROS generation by X-ray-induced photodynamic therapy, and produce high levels of NO to enhance tumor-killing effects and improve hypoxia via NO-induced vasodilation. In vitro and in vivo studies revealed that this combined strategy can greatly reinforce DNA damage and apoptosis of hypoxic tumor cells, while significantly suppressing tumor growth, improving tumor hypoxia and promoting p53 up-regulation and HIF1α down-regulation. In addition, this system showed pronounced tumor contrast performance in T1-weighted magnetic resonance imaging and computed tomography.
    CONCLUSION: This work demonstrates the great potential of scintillating nanotheranostics for multimodal imaging-guided X-ray radiation-triggered tumor combined therapy to overcome radioresistance.
    Keywords:  X-ray induced photodynamic therapy; hypoxia-induced radioresistance; multimodal imaging; nanoscintillators; nitric oxide
    DOI:  https://doi.org/10.7150/thno.27351
  39. Nutr Cancer. 2019 Jan 11. 1-8
      BACKGROUND: Oligonol, a low molecular weight polyphenol derived from lychee fruit, not only has anti-inflammatory effects in various disease conditions but also has antitumor-promoting effects. We evaluate the nuclear factor-kappa B (NF-κB)-related anticancer effect of oligonol in ovarian cancer using SKOV-3 cells.METHODS: Cell viability was examined after oligonol treatment using MTT assay and reactive oxygen species (ROS) production measurement. Subsequently, apoptotic cell death was visualized by the TdT-mediated dUTP nick-end labeling (TUNEL) method. The effect of oligonol on the NF-κB signaling pathway was evaluated using western blot analysis and luciferase activity measurement of p65, an NF-κB subunit.
    RESULTS: Cell viability significantly decreased after oligonol treatment of 72 h. Apoptosis-related markers were highly expressed in oligonol-treated cells, and increased apoptosis after oligonol treatment was also confirmed using the TUNEL assay. Western blotting results showed the expression of NF-κB signaling pathway factors, p-ERK, TRAF2, and p-IκBα, increased following treatment with oligonol, whereas p65 and COX-2 expression decreased. Immunofluorescence imaging results showed p65 luciferase activity in the nucleus as well as a shift to cytoplasmic expression.
    CONCLUSION: Oligonol treatment significantly enhances apoptotic cell death in SKOV-3 cells, with the suppression of NF-κB activation, which plays an essential role in this anticancer effect.
    DOI:  https://doi.org/10.1080/01635581.2018.1557215
  40. Iran J Basic Med Sci. 2018 Dec;21(12): 1281-1288
      Objectives: In this study, we evaluated the therapeutic effects of casticin and myricetin on liver damage induced by methotrexate in rats.Materials and Methods: Thirty-six male rats were used for the study and divided into 6 groups: control, methotrexate, casticin, myricetin, casticin+methotrexate, and myricetin+methotrexate. It was performed by methotrexate (20 mg/kg single dose, IP) in methotrexate, casticin+methotrexate and myricetin+methotrexate groups. Casticin 200 mg/kg dose was given to casticin and casticin+methotrexate groups. Myricetin 50 mg/kg dose was given to myricetin and myriceytin+methotrexate groups. At the end of the experiment, liver tissues were removed for the purpose of histopathological, biochemical and immunohistochemical assessments.
    Results: In our study, we have detected that MDA levels increased and activities of antioxidant enzymes SOD, CAT, and GPX decreased in the methotrexate group compared to the other groups, but the level of MDA decreased and activities of these enzymes increased in casticin+methotrexate and myricetin+methotrexate groups compared to the methotrexate group. In immunohistochemical examinations of control, casticin and myricetin groups in liver tissues no caspase-3 and 8-OHdG expressions were observed. In the MTX group, caspase-3 and 8-OHdG expressions were seen at the severe levels. Caspase-3 and 8-OHdG expressions were mild in hepatocytes in the casticin+methotrexate and myricetin+methotrexate groups. When the liver tissues of the rats in the methotrexate group were examined, severe pathological damage was detected both in the parietal region and in the portal region.
    Conclusion: By looking at these results, we can say that casticin and myricetin are effective against liver damage induced by methotrexate.
    Keywords:  Antioxidants; Casticin; Myricetin; Oxidative stress; Reactive oxygen species
    DOI:  https://doi.org/10.22038/ijbms.2018.29922.7217
  41. Biochem Biophys Res Commun. 2019 Jan 04. pii: S0006-291X(18)32727-X. [Epub ahead of print]
      Oxidative damage plays a critical role in cochlear cell apoptosis, which is central to the physiopathology of noise-induced hearing loss (NIHL). Sirtuin 2 (SIRT2) is an NAD-dependent deacetylase that regulates cellular response to oxidative stress, however, its role in NIHL remains poorly understood. Here, we report that SIRT2 is upregulated in the cochlea after noise exposure. Functionally, the treatment of AK-7, one specific SIRT2 inhibitor, attenuates the progression of NIHL. In addition, AK-7 treatment reduces oxidative nuclear DNA damage and apoptosis in the cochlea after noise exposure. Moreover, AK-7 treatment reduces apoptosis of mouse inner ear HEI-OC1 cells exposed to oxidative stress in vitro. Taken together, these results suggest that SIRT2 inhibition with AK-7 reduces cochlear cell apoptosis through attenuating oxidative stress-induced damage, which may underlie its protective role against NIHL. This study also implies that AK-7 may have potential therapeutic significance in the intervention of NIHL.
    Keywords:  AK-7; Apoptosis; Cochlear cell; Noise-induced hearing loss; SIRT2
    DOI:  https://doi.org/10.1016/j.bbrc.2018.12.084
  42. J Cell Biochem. 2019 Jan 07.
      Age-related macular degeneration (AMD), one of the most common causes of visual impairment, often occurrs in the elderly in developed countries. Oxidative stress, autophagy, and apoptosis of retinal pigment epithelial (RPE) cells play roles in the pathogenesis of AMD. In the current study, the protective effect of celastrol against hydrogen peroxide (H2 O2 )-induced oxidative stress and apoptosis was investigated using a human RPE cell line (ARPE-19). H2 O2 inhibited ARPE-19 cells' survival and autophagy and induced their oxidative stress and apoptosis. Compared with the H2 O2 group, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay showed that celastrol increased ARPE-19 cells' survival in a dose- and time-dependent manner. Further, studies have suggested that celastrol has antioxidative stress and antiapoptosis effects in H2 O2 -treated ARPE-19 cells. Also, cell autophagy is activated by celastrol in H2 O2 -treated ARPE-19 cells. Reverse transcription polymerase chain reaction and Western blot showed that celastrol elevated the messenger RNA (mRNA) and protein expression of sirtuin 3 (SIRT3) in H2 O2 -induced ARPE-19 cells. Inhibition of the level of SIRT3 by SIRT3 small interfering RNA (siRNA) reversed the effects of celastrol on oxidative stress, autophagy, and apoptosis in H2 O2 -induced ARPE-19 cells. In conclusion, these observations suggest that celastrol activates the SIRT3 pathway in RPE cells and protects against H2 O2 -induced oxidative stress and apoptosis.
    Keywords:  autophagy; celastrol; oxidative stress; retinal pigment epithelial cells; sirtuin 3
    DOI:  https://doi.org/10.1002/jcb.28326
  43. J Cell Biochem. 2019 Jan 10.
      OBJECTIVES: Lung cancer stands out as the most common cancer type worldwide. The most common genetic alteration detected in adenocarcinoma patients is KRAS. KRAS mutated patients still cannot get benefit from precision medicine approaches and lack a targeted therapy. Elesclomol is an investigational agent for melanoma and other malignancies. In this study, we evaluated its effect on cellular apoptosis, survival, and metastasis mechanisms on KRAS mutant A549 and Calu-1 cell lines.METHODS: The cytotoxic effects of Elesclomol on A549 and Calu-1 cells were determined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) cell viability test. Cells were treated with IC50 concentration and then apoptosis-related (Casp-3, Casp-9, Bcl-2, and Bcl-xL), survival-related (Akt, p-Akt, Erk, and p-Erk), and metastasis-related (E-cadherin, Vimentin, MMP-2, and MMP-9) protein expressions were determined by Western blot analysis. Elesclomol's effect on cell migration was evaluated by wound healing. Total oxidant, malondialdehyde (MDA), and glutathione (GSH) levels after Elesclomol treatment were assessed.
    RESULTS: Elesclomol not only induced apoptotic proteins but also inhibited metastatic protein expressions and migration in both cells. Also, p-Erk activity was diminished by Elesclomol treatment as a reflection of decreased proliferation. However, p-Akt was enhanced as a cellular survival mechanism. Although Elesclomol's effects on oxidative stress parameters were puzzling, it induced total oxidant status (TOS), and MDA in Calu-1 cells.
    CONCLUSION: Elesclomol might provide an alternative treatment approach for patients with KRAS mutant lung adenocarcinoma and other solid tumor malignancies that harbor KRAS mutations. This would enable the development of biomarker-driven targeted therapy for KRAS mutant adenocarcinoma patients.
    Keywords:  KRAS; adenocarcinoma; apoptosis; elesclomol; metastasis; non-small-cell lung cancer
    DOI:  https://doi.org/10.1002/jcb.28342
  44. Biotech Histochem. 2019 Jan 11. 1-6
      Little is known about the pathogenesis of high fructose corn syrup (HFCS) induced hepatic toxicity. We investigated hepatic lesions induced by chronic HFCS consumption and the protective effects of alpha-lipoic acid (ALA) on liver pathology. We used 24 rats allocated randomly into three groups of eight. The HFCS group was given in drinking water for 10 weeks. The ALA + HFCS group was given the same dose of HFCS and ALA also was administered during the last 6 weeks of the experiment. The control group was untreated. The rats were euthanized at the end of 10 weeks and 24 h after the last ALA administration. A significant increase was observed in the serum aspartate aminotransferase (AST) level of the HFCS group compared to controls. Tissue malondialdehyde (MDA) levels also increased significantly and catalase (CAT) activity decreased significantly in the HFCS group. Caspase-3 expression increased significantly in the HFCS group compared to controls. In the ALA treated group, the levels of MDA, CAT and caspase-3 returned to near control levels. HFCS caused hepatic toxicity by increasing oxidative stress and apoptosis. ALA administration ameliorated the pathological changes.
    Keywords:  Alpha-lipoic acid; corn syrup; fructose; hepatic damage; liver; oxidative stress; pathology; rat
    DOI:  https://doi.org/10.1080/10520295.2018.1552019
  45. Front Cell Neurosci. 2018 ;12 485
      Dopaminergic neurons loss is one of the main pathological characters of Parkinson's disease (PD), while no suitable neuroprotective agents have been in clinical use. Thyrotropin-releasing hormone (TRH) and its analogs protect neurons from ischemia and various cytotoxins, but whether the effect also applies in PD models remain unclear. Here, we showed that Taltirelin, a long-acting TRH analog, exhibited the neuroprotective effect in both cellular and animal models of PD. The in vitro study demonstrated that Taltirelin (5 μM) reduced the generation of reactive oxygen species (ROS) induced by MPP+ or rotenone, alleviated apoptosis and rescued the viability of SH-SY5Y cells and rat primary midbrain neurons. Interestingly, SH-SY5Y cells treated with Taltirelin also displayed lower level of p-tau (S396) and asparagine endopeptidase (AEP) cleavage products, tau N368 and α-synuclein N103 fragments, accompanied by a lower intracellular monoamine oxidase-B (MAO-B) activity. In the subacute MPTP-induced and chronic rotenone-induced PD mice models, we found Taltirelin (1 mg/kg) significantly improved the locomotor function and preserved dopaminergic neurons in the substantia nigra (SN). In accordance with the in vitro study, Taltirelin down-regulated the levels of p-tau (S396), p-α-synuclein (S129) tau N368 and α-synuclein N103 fragments in SN and striatum. Together, this study demonstrates that Taltirelin may exert neuroprotective effect via inhibiting MAO-B and reducing the oxidative stress and apoptosis, preventing AEP activation and its subsequent pathological cleavage of tau and α-synuclein, thus provides evidence for Taltirelin in protective treatment of PD.
    Keywords:  AEP; MPTP; Parkinson’s disease; TRH; rotenone; taltirelin; tau; α-synuclein
    DOI:  https://doi.org/10.3389/fncel.2018.00485