bims-aporos Biomed News
on Apoptosis and reactive oxygen species
Issue of 2019‒04‒07
seventy-two papers selected by
Gavin McStay
Staffordshire University


  1. Exp Ther Med. 2019 Apr;17(4): 2666-2674
      Increased aggregation of β-amyloid (Aβ) peptides induces oxidative stress, which is considered a major contributor in the development of Alzheimer's disease (AD). Prevention of Aβ-induced neurotoxicity is proposed as a possible modality for treatment of AD. The present study aimed to elucidate possible effects of ethyl vanillin (EVA), an analog of vanillin isolated from vanilla beans, on the Aβ1-42-induced oxidative injury in PC12 cells. EVA restrained the decrease in PC12 cell viability and apoptosis induction caused by treatment with Aβ1-42. In addition, EVA markedly alleviated intracellular lipid peroxidation as demonstrated by malondialdehyde levels and reactive oxygen species production in Aβ1-42-treated PC12 cells. In addition, the reduction in the activity levels of the antioxidative enzymes superoxide dismutase, catalase and glutathione peroxidase was detected in Aβ1-42-treated PC12 cells. This effect was partially reversed by treatment with EVA. Furthermore, the results indicated that EVA attenuated Aβ1-42-induced caspase-3 activation and the increase noted in the apoptosis regulator Bcl-2/apoptosis regulator Bax ratio of PC12 cells. These results indicated that EVA could be used as an efficient and novel agent for the prevention of neurodegenerative diseases via inhibition of oxidative stress and cell apoptosis.
    Keywords:  ethyl vanillin; neuroprotection; oxidative stress
    DOI:  https://doi.org/10.3892/etm.2019.7242
  2. Antioxidants (Basel). 2019 Apr 01. pii: E82. [Epub ahead of print]8(4):
      Reactive oxygen species (ROS), products of oxidative stress, contribute to the initiation and progression of the pathogenesis of various diseases. Glutathione is a major antioxidant that can help prevent the process through the removal of ROS. The aim of this study was to evaluate the protective effect of glutathione on ROS-mediated DNA damage and apoptosis caused by hydrogen peroxide, H₂O₂, in RAW 264.7 macrophages and to investigate the role of the nuclear factor erythroid 2-related factor-2 (Nrf2)/heme oxygenase-1 (HO-1) signaling pathway. The results showed that the decrease in the survival rate of RAW 264.7 cells treated with H₂O₂ was due to the induction of DNA damage and apoptosis accompanied by the increased production of ROS. However, H₂O₂-induced cytotoxicity and ROS generation were significantly reversed by glutathione. In addition, the H₂O₂-induced loss of mitochondrial membrane potential was related to a decrease in adenosine triphosphate (ATP) levels, and these changes were also significantly attenuated in the presence of glutathione. These protective actions were accompanied by a increase in the expression rate of B-cell lymphoma-2 (Bcl-2)/Bcl-2-associated X protein (Bax) and poly(ADP-ribose) polymerase cleavage by the inactivation of caspase-3. Moreover, glutathione-mediated cytoprotective properties were associated with an increased activation of Nrf2 and expression of HO-1; however, the inhibition of the HO-1 function using an HO-1 specific inhibitor, zinc protoporphyrin IX, significantly weakened the cytoprotective effects of glutathione. Collectively, the results demonstrate that the exogenous administration of glutathione is able to protect RAW 264.7 cells against oxidative stress-induced mitochondria-mediated apoptosis along with the activity of the Nrf2/HO-1 signaling pathway.
    Keywords:  DNA damage; Nrf2/HO-1; ROS; apoptosis; glutathione; oxidative stress
    DOI:  https://doi.org/10.3390/antiox8040082
  3. Free Radic Biol Med. 2019 Apr 01. pii: S0891-5849(18)31697-6. [Epub ahead of print]
      Radiation-induced damage to the mitochondrial macromolecules and electron transfer chain (ETC), causing the generation of primary and secondary reactive oxygen (ROS) species. The continuous ROS production after radiation will trigger cell oxidative stress and ROS-mediated nucleus apoptosis and autophagy signaling pathways. Scavenging radiation-induced ROS effectively can help mitochondria to maintain their physiological function and relief cells from oxidative stress. Nicotinamide is a critical endogenous antioxidant helping to neutralize ROS in vivo. In this study, we designed and synthetized a novel mitochondrial-targeted dihydronicotinamide (Mito-N) with the help of mitochondrial membrane potential to enter the mitochondria and scavenge ROS. According to experiment results, Mito-N significantly increased cell viability by 30.75% by neutralizing the accumulated ROS and resisting DNA strands breaks after irradiation. Furthermore, the mice survival rate also improved with the treatment of Mito-N, by effectively ameliorating the hematopoietic system infliction under lethal dose irradiation.
    Keywords:  Mitochondria-targeted; Oxidative stress; Radiation damage; Reactive oxygen species (ROS)
    DOI:  https://doi.org/10.1016/j.freeradbiomed.2019.03.038
  4. Nutrients. 2019 Apr 01. pii: E762. [Epub ahead of print]11(4):
      Generation of excess quantities of reactive oxygen species (ROS) caused by mitochondrial dysfunction facilitates rapid growth of pancreatic cancer cells. Elevated ROS levels in cancer cells cause an anti-apoptotic effect by activating survival signaling pathways, such as NF-κB and its target gene expression. Lycopene, a carotenoid found in tomatoes and a potent antioxidant, displays a protective effect against pancreatic cancer. The present study was designed to determine if lycopene induces apoptosis of pancreatic cancer PANC-1 cells by decreasing intracellular and mitochondrial ROS levels, and consequently suppressing NF-κB activation and expression of NF-κB target genes including cIAP1, cIAP2, and survivin. The results show that the lycopene decreased intracellular and mitochondrial ROS levels, mitochondrial function (determined by the mitochondrial membrane potential and oxygen consumption rate), NF-κB activity, and expression of NF-κB-dependent survival genes in PANC-1 cells. Lycopene reduced cell viability with increases in active caspase-3 and the Bax to Bcl-2 ratio in PANC-1 cells. These findings suggest that supplementation of lycopene could potentially reduce the incidence of pancreatic cancer.
    Keywords:  NF-κB; apoptosis; lycopene; pancreatic cancer cells; reactive oxygen species
    DOI:  https://doi.org/10.3390/nu11040762
  5. Mol Med Rep. 2019 Mar 26.
      Posterior capsular opacification (PCO) remains a major complication of cataract surgery and is the most common reason for loss of vision. PCO is primarily associated with uncontrolled proliferation of residual human lens epithelial cells (HLEs). Sanguinarine is a type of benzophenanthridine alkaloid extracted from the herbaceous plant Sanguinaria canadensis, which is widely used for its anti‑microbial, anti‑inflammatory, anti‑oxidative and anti‑proliferative properties. However, studies examining the effect of sanguinarine on HLEs and the underlying mechanism are scarce. The present study aimed to investigate the effects of sanguinarine on HLEs. An MTT assay was used to determine the effect of sanguinarine on cell viability. Flow cytometry was used to evaluate cell apoptosis, and the mitochondrial membrane potential and reactive oxygen species (ROS) levels. A caspase 3/7 activity assay was also used to evaluate cell apoptosis, while western blotting was performed to determine protein levels. The results demonstrated that sanguinarine exerted an anti‑proliferative effect by inducing ROS, and caused cell apoptosis via mitochondrial and caspase‑dependent pathways. Treatment with sanguinarine led to the loss of mitochondrial membrane potential. Sanguinarine also significantly increased the phosphorylation levels of c‑Jun N‑terminal kinase and p38, which indicated the involvement of the mitogen‑activated protein kinase signaling pathway. These results suggested that sanguinarine may have a noteworthy pro‑apoptotic effect on HLEs, and may be used as a potential drug for PCO or even cataract prevention.
    DOI:  https://doi.org/10.3892/mmr.2019.10087
  6. Int J Mol Sci. 2019 Mar 25. pii: E1490. [Epub ahead of print]20(6):
      Horse oil products have been used in skin care for a long time in traditional medicine, but the biological effects of horse oil on the skin remain unclear. This study was conducted to evaluate the protective effect of horse oil on ultraviolet B (UVB)-induced oxidative stress in human HaCaT keratinocytes. Horse oil significantly reduced UVB-induced intracellular reactive oxygen species and intracellular oxidative damage to lipids, proteins, and DNA. Horse oil absorbed light in the UVB range of the electromagnetic spectrum and suppressed the generation of cyclobutane pyrimidine dimers, a photoproduct of UVB irradiation. Western blotting showed that horse oil increased the UVB-induced Bcl-2/Bax ratio, inhibited mitochondria-mediated apoptosis and matrix metalloproteinase expression, and altered mitogen-activated protein kinase signaling-related proteins. These effects were conferred by increased phosphorylation of extracellular signal-regulated kinase 1/2 and decreased phosphorylation of p38 and c-Jun N-terminal kinase 1/2. Additionally, horse oil reduced UVB-induced binding of activator protein 1 to the matrix metalloproteinase-1 promoter site. These results indicate that horse oil protects human HaCaT keratinocytes from UVB-induced oxidative stress by absorbing UVB radiation and removing reactive oxygen species, thereby protecting cells from structural damage and preventing cell death and aging. In conclusion, horse oil is a potential skin protectant against skin damage involving oxidative stress.
    Keywords:  apoptosis; horse oil; oxidative stress; ultraviolet B radiation
    DOI:  https://doi.org/10.3390/ijms20061490
  7. Chem Biol Interact. 2019 Mar 28. pii: S0009-2797(19)30104-8. [Epub ahead of print]305 54-65
      Doxorubicin (DOX) is one of the most effective chemotherapeutic drugs; however, the incidence of cardiotoxicity compromises its therapeutic index. Oxidative stress and apoptosis are believed to be involved in DOX-induced cardiotoxicity. Chitosan oligosaccharides (COS), the enzymatic hydrolysates of chitosan, have been reported to possess diverse biological activities including antioxidant and anti-apoptotic properties. The objective of the present study was to investigate the potential role of COS against DOX-induced cardiotoxicity, and the effects of COS on apoptosis and oxidative stress in rats and H9C2 cells. Furthermore, we also shed light on the involved pathways during the whole process. For this purpose, first, we demonstrated that COS exhibited a significant protective effect on cardiac tissue by not only inducing a decrease in body and heart growth but also ameliorated oxidative damage and ECG alterations in DOX-treated rats. Second, we found that COS reversed the decrease of cell viability induced by DOX, reduced the intracellular reactive oxygen species (ROS), increased the mitochondrial membrane potential (MMP) and Bcl-2/Bax ratio. COS treatment also results in reduced caspase-3 and caspase-9 expressions, and an increase in the phosphorylation of MAPKs (mitogen-activated protein kinases) in DOX-exposed H9C2 cells. Additionally, cellular homeostasis was re-established via stabilization of MAPK mediated nuclear factor erythroid 2-related factor 2/antioxidant-response element (Nrf2/ARE) signaling and transcription of downstream cytoprotective genes. In summary, these findings suggest that COS could be a potential candidate for the prevention and treatment of DOX-induced cardiotoxicity.
    Keywords:  Apoptosis; Chitosan oligosaccharides; Doxorubicin; Nrf2/ARE; Oxidative stress
    DOI:  https://doi.org/10.1016/j.cbi.2019.03.027
  8. Oncol Lett. 2019 May;17(5): 4277-4284
      Previous studies have reported that metformin (MET) has anticancer activity. In combination with chemotherapeutic drugs, MET reduces the dosage of chemotherapeutic drugs required and enhances anticancer efficacy. In the present study, the combination of MET and paclitaxel (PTX) in three human prostate cancer (PCa) cell lines (22RV1, PC-3 and LNCaP) was evaluated to investigate the effects on proliferation and apoptosis of PCa cells. The present study explored whether their effects were associated with reactive oxygen species (ROS). An MTT assay and microscopy were used to study the effect of MET + PTX on cell growth. Half maximal inhibitory concentration (IC50) values were obtained for MET (12.281±1.089 mM for 22RV1, 2.248±0.352 mM for PC-3 cells and 3.610±0.577 mM for LNCaP cells) and PTX (13.170±1.12 nM for PC-3 cells) at 48 h. Since the survival rate of 22RV1 and LNCaP cells did not decrease linearly with increasing PTX concentration, it is difficult to estimate accurate IC50; therefore, only IC50 values for PTX in PC-3 cells were given. When treating the cells with 5 mM MET, the IC50 of PTX decreased to 5.423±0.734 nM for PC-3 cells. Annexin V and propidium iodide staining was used to investigate apoptosis by flow cytometry. The apoptotic mechanisms of MET + PTX in PCa were investigated by detecting the expression of apoptosis-related proteins, activities of caspase-3/7, intracellular ROS accumulation, mitochondrial membrane potential, and intracellular levels of adenosine 5'-triphosphate (ATP). MET + PTX induced PCa apoptosis and ROS accumulation, and decreased mitochondrial membrane potential and intracellular levels of ATP. Taken together, these results indicated that MET + PTX suppressed PCa cell proliferation in a dose- and time-dependent manner. In addition, MET + PTX induced apoptosis by increasing ROS levels, reducing mitochondrial membrane potential, and activating mitochondrial-dependent apoptotic pathways.
    Keywords:  apoptosis; metformin; paclitaxel; prostate cancer; reactive oxygen species
    DOI:  https://doi.org/10.3892/ol.2019.10119
  9. Cell Mol Biol (Noisy-le-grand). 2019 Mar 31. 65(3): 101-108
      Glioblastoma is a malignant tumor of the brain. The treatment of this tumor is still a challenge. Curcumin has been shown to have therapeutic effects when used to treat malignant diseases. However, the molecular mechanisms of its action are not fully elucidated. We hypothesized that reactive oxygen species (ROS) have a key role in curcumin-induced DNA damage, apoptosis, and cell death. To test our hypothesis, cytotoxic, genotoxic, apoptotic, and ROS-generating effects, as well as mitochondrial membrane potentials of curcumin on rat glioma cells (C-6) and normal fibroblastic cells (L-929) were investigated. We examined concentration-dependent cytotoxic, genotoxic, apoptotic, and ROS generating effects of curcumin at C-6 cells and L-929 cells. The cells were incubated with different doses of curcumin (10-100 µM) for 24 hours. Higher doses of curcumin resulted in greater cellular death of cancer than of normal cells at higher concentrations. Curcumin also induced ROS generation in cancer than normal cells in a concentration-dependent manner. Our results showed that curcumin-induced DNA damage in a dose-dependent manner (p < 0.001). At high curcumin concentration such as 80 µM, the proportions of live cells in cancer and normal cell lines were 11.5 and 44.3, respectively. The higher doses of curcumin resulted in greater apoptosis in cancer than normal cells.This in vitro study provided clear evidence that curcumin induced DNA damage and apoptosis. Cytotoxicity may be due to its pro-oxidant activity in a dose-dependent manner in cancer and normal cells. These activities were higher in cancer cells.
    Keywords:  Antioxidants; Apoptosis; Curcumin; Cytotoxicity; Genotoxic Activity.; Glioblastoma
  10. Toxicol Res (Camb). 2019 Mar 01. 8(2): 216-226
      Camphor is one of the monoterpenes widely used in cosmetics, pharmaceutics and the food industry. In this study, we aimed to assess the oxidative, cytotoxic and apoptotic effects of camphor on the fission yeast (Schizosaccharomyces pombe), which is a promising unicellular model organism in mechanistic toxicology and cell biology. Since Sod1 is the main radical scavenger in the cell, we used sod1 mutants to understand whether camphor-induced ROS accumulation caused higher cytotoxicity and apoptosis. Camphor exposure (0-2000 mg L-1) caused significant cytotoxicity in yeast, particularly in sod1Δ cells. DCFDA (2,7-dichlorodihydrofluorescein diacetate) fluorescence and NBT (p-nitro-blue tetrazolium chloride) reduction increased (at least 2.5-3-fold in sod1Δ cells) in correlation with camphor concentrations (800-1200 mg L-1), showing higher ROS levels and oxidative stress. Moreover, cells, stained with acridine orange/ethidium bromide, showed an apoptotic morphology with nuclear fragmentation and condensation. DAPI (4',6-diamidino-2-phenylindole) staining was used to validate the apoptotic nuclear morphology. Dramatically increased mitochondrial impairment, which was higher in sod1Δ cells than in wild type cells, was shown by rhodamine 123 staining. In conclusion, camphor-induced excessive ROS production, which could not be prevented significantly in sod1 mutants, caused a dramatic increase in mortality rates due to intrinsic apoptosis revealed by mitochondrial impairment and apoptotic nuclear morphology. The potential effects of camphor on apoptotic cell death and the underlying mechanisms were clarified in the unicellular eukaryotic model, S. pombe.
    DOI:  https://doi.org/10.1039/c8tx00279g
  11. J BUON. 2019 Jan-Feb;24(1):24(1): 221-226
      PURPOSE: The main aim of the study was to evaluate in vitro and in vivo the anticancer and apoptotic effects of neochlorogenic acid in human gastric carcinoma cell death and the underlying mechanism of apoptosis induction, reactive oxygen species (ROS) production and loss of mitochondrial membrane potential (MMP), m-TOR/PI3K/AKT signalling pathway, cell migration and cell invasion suppression.METHODS: Fluorescence microscopy using DAPI and annexin V/PI staining in combination with flow cytometry was used to study the apoptotic effects induced by neochlorogenic acid on gastric cancer cells. The effects on ROS and MMP were studied by flow cytometry. Western blot assay was used to evaluate the effects of neochlorogenic acid on m-TOR/PI3/Akt signaling pathway. To examine the anti-cancer activity of neochlorogenic acid in vivo, we used the nude mice xenograft model.
    RESULTS: The results indicated that neochlorogenic acid exhibited an IC50 of 20 µM in these cells. The study also showed that apoptosis was due to loss of MMP and increased intracellular ROS production. Neochlorogenic acid downregulated the expression of key proteins of m-TOR/PI3/Akt signaling pathway. After 6 weeks of neochlorogenic acid administration to mice, the average tumor volumes and growth for the untreated control group were significantly higher than the treated groups.
    CONCLUSION: Based on these results, we propose that neochlorogenic acid can be a prospective anti-cancer therapeutic lead for the management of human gastric carcinoma.
  12. Eur J Pharmacol. 2019 Apr 02. pii: S0014-2999(19)30217-1. [Epub ahead of print]
      Nobiletin (3',4',5,6,7,8-hexamethoxyflavone), a dietary polymethoxylated flavonoid found in Citrus fruits, was reported to exhibit protective activity against ischemia/reperfusion (I/R). However, the role of nobiletin in myocardial I/R injury remains unclear. This study was designed to examine the cardioprotective effect of nobiletin from myocardial hypoxia/reoxygenation (H/R) injury in vitro, and to explore the potential molecular mechanisms. Our results showed that nobiletin improved cell viability in H9c2 cells after H/R treatment. In addition, nobiletin significantly inhibited the production of reactive oxygen species and malondialdehyde (MDA), cell apoptosis, as well as suppressed the levels of pro-inflammatory factors in H/R-stimulated H9c2 cells. Furthermore, we observed that pretreatment with nobiletin significantly activated the Akt/GSK-3β signaling pathway in H/R-stimulated H9c2 cells. Taken together, these findings demonstrated that nobiletin attenuates myocardial I/R injury via the activation of Akt/GSK-3β pathway in H9c2 cardiomyocytes. Thus, nobiletin may be regarded as a promising drug for the prevention of myocardial I/R injury and ischemic heart disease.
    Keywords:  Akt/GSK-3β pathway; Apoptosis; Hypoxia/reoxygenation (H/R); Nobiletin
    DOI:  https://doi.org/10.1016/j.ejphar.2019.03.056
  13. J Cell Biochem. 2019 Apr 03.
      Neonatal hypoxic-ischemic encephalopathy is one of the leading causes of death in infants. Increasing evidence indicates that oxidative stress and apoptosis are major contributors to hypoxic-ischemic injury and can be used as particularly promising therapeutic targets. Platycodin D (PLD) is a triterpenoid saponin that exhibits antioxidant properties. The aim of this study was to evaluate the effects of PLD on hypoxic-ischemic injury in primary cortical neurons. We found that oxygen-glucose deprivation/reperfusion (OGD/R) induced inhibition of cell viability and cytotoxicity, which were attenuated by PLD treatment. PLD treatment inhibited oxidative stress induced by OGD/R, which was evidenced by the reduced level of reactive oxygen species and increased activities of catalase, superoxide dismutase, and glutathione peroxidase. Histone-DNA enzyme-linked immunosorbent assay revealed that apoptosis was significantly decreased after PLD treatment in OGD/R-treated cortical neurons. The increased bax expression and decreased bcl-2 expression induced by OGD/R were reversed by PLD treatment. Furthermore, PLD treatment caused the activation of the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR) pathway in OGD/R-stimulated cortical neurons. Suppression of this pathway blocked the protective effects of PLD on OGD/R-induced cell injury. These findings suggested that PLD executes its protective effects on OGD/R-induced cell injury via regulating the PI3K/Akt/mTOR pathway in cortical neurons.
    Keywords:  PI3K/Akt/mTOR signaling pathway; apoptosis; neonatal hypoxic-ischemic encephalopathy; oxidative stress; platycodin D
    DOI:  https://doi.org/10.1002/jcb.28677
  14. J Zhejiang Univ Sci B. 2019 Apr.;20(4):20(4): 310-321
      OBJECTIVE: Reactive oxygen species (ROS) are involved in a variety of biological phenomena and serve both deleterious and beneficial roles. ROS quantification and assessment of reaction networks are desirable but difficult because of their short half-life and high reactivity. Here, we describe a pro-oxidative model in a single human lung carcinoma SPC-A-1 cell that was created by application of extracellular H2O2 stimuli.METHODS: Modified microfluidics and imaging techniques were used to determine O2 •- levels and construct an O2 •- reaction network. To elucidate the consequences of increased O2 •- input, the mitochondria were given a central role in the oxidative stress mode, by manipulating mitochondria-interrelated cytosolic Ca2+ levels, mitochondrial Ca2+ uptake, auto-amplification of intracellular ROS and the intrinsic apoptotic pathway.
    RESULTS AND CONCLUSIONS: Results from a modified microchip demonstrated that 1 mmol/L H2O2 induced a rapid increase in cellular O2 •- levels (>27 vs. >406 amol in 20 min), leading to increased cellular oxidizing power (evaluated by ROS levels) and decreased reducing power (evaluated by glutathione (GSH) levels). In addition, we examined the dynamics of cytosolic Ca2+ and mitochondrial Ca2+ by confocal laser scanning microscopy and confirmed that Ca2+ stores in the endoplasmic reticulum were the primary source of H2O2-induced cytosolic Ca2+ bursts. It is clear that mitochondria have pivotal roles in determining how exogenous oxidative stress affects cell fate. The stress response involves the transfer of Ca2+ signals between organelles, ROS auto-amplification, mitochondrial dysfunction, and a caspase-dependent apoptotic pathway.
    Keywords:  Individual cell; Superoxide anion; Reactive oxygen species (ROS) dynamics; Intrinsic apoptotic pathway; Ca2+ signaling
    DOI:  https://doi.org/10.1631/jzus.B1800319
  15. J Cell Physiol. 2019 Apr 05.
      Bone marrow mesenchymal stem cells (BMSC) can ameliorate ischemic injury of various tissues. However, the molecular mechanisms involved remain to be clarified. In this study, we intend to investigate the effects of BMSC-derived conditioned medium (BMSC-CM) on hypoxia/reoxygenation (H/R)-induced injury of H9c2 myocardial cells, and the potential mechanisms. Cell injury was determined through level of cell viability, lactate dehydrogenase (LDH) release, total intracellular reactive oxygen species (ROS), mitochondrial membrane potential (Δψm), and cell apoptosis. Autophagic activity of cells was detected through levels of the autophagy-associated proteins and autophagic flux. Results showed that BMSC-CM alleviated H/R-induced injury in H9c2 cells, as demonstrated by increased cell viability and Δψm, decreased ROS production, LDH release, and cell apoptosis. Furthermore, the H/R treatment induced a decrease in autophagic activity and an increase in Notch2 signaling activation in H9c2 cells. In the presence of BMSC-CM, the autophagic activity impaired by the H/R treatment was upregulated with decreased phosphorylation of mTOR, and the activation of Notch2 signaling was downregulated. These effects of BMSC-CM could be replicated by Notch signaling inhibitor. In contrast, inhibitors of cell autophagy including chloroquine (CQ) and 3-methyladenine, diminished the protective effects of BMSC-CM. Taken together results, our study showed that BMSC-CM could protect H9c2 cells from H/R-induced injury potentially through regulating Notch2/mTOR/autophagy signaling. These findings may provide a novel insight into the mechanisms of BMSC-CM in therapy of myocardial ischemia/reperfusion injury as well as other ischemic diseases.
    Keywords:  Notch; autophagy; cardiomyocytes; hypoxia/reoxygenation; mesenchymal stem cells
    DOI:  https://doi.org/10.1002/jcp.28530
  16. Chemosphere. 2019 Mar 29. pii: S0045-6535(19)30613-7. [Epub ahead of print]226 463-471
      Tetrabromobisphenol A (TBBPA) is a commonly used brominated flame retardant, which has a wide range of toxic effects on organisms. This study investigated the cytotoxic effects on human hepatocytes (L02 cells) after treated with 0, 5, 10, 20, and 40 μM of TBBPA. Results showed that TBBPA significantly increased intracellular reactive oxygen species (ROS), malondialdehyde (MDA) and the ratio of oxidized/reduced glutathione (GSSG/GSH) dose-dependently. TBBPA also decreased the cell mitochondrial membrane potential (MMP), caused the release of cytochrome C (Cyt C) to cytoplasm and promoted the expression of caspase-9 and caspase-3, and finally increased the level of apoptosis. The ROS inhibitor N-acetyl-L-cysteine (NAC) relieved the oxidative stress responses, and prevented the decrease of MMP and increase of apoptosis. In addition, TBBPA promoted the expression of antioxidant genes related to Nrf2, such as quinone oxidoreductase 1 (NQO1), catalase (CAT), and heme oxygenase 1 (HO-1). Oxidative stress initiated by TBBPA, activated mitochondrial apoptosis and Nrf2 pathway, and increased the degree of apoptosis in L02 cells.
    Keywords:  Apoptosis; Hepatotoxicity; Nrf2; Oxidative stress; Tetrabromobisphenol A
    DOI:  https://doi.org/10.1016/j.chemosphere.2019.03.167
  17. Anticancer Agents Med Chem. 2019 Apr 05.
      BACKGROUND: Levels of cellular Reactive Oxygen Species (ROS) influence the oxidized/reduced states of cellular proteins, and create redox-signaling pathways that can activate transcription factors, kinases, and phosphatases. ROS levels can be increased radically by external factors, including ionizing and UV radiation or exposure to chemical compounds. These increased ROS levels can in turn lead to oxidative damage of DNA. Natural plant treatments against cancer can modulate that processes by inducing or decreasing ROS production.METHODS: Here we report new observations that squamous carcinoma (SCC-25) cells, exposed to 24 hours of combined resveratrol and berberine treatment contained increased ROS levels. Using flow cytometry, for drug activity characteristic, an accumulation of ROS was observed. Combination of different dyes, CellROX Green (Life Technologies) and DCFH-DA (Sigma), allowed for flow cytometric estimation of levels of cellular ROS as well as cellular localization.
    RESULTS: Live staining and microscopic observations confirmed the accumulation of ROS in SCC-25 cells following combination treatment at concentrations of 10 µg/ml. Additionally, the cytotoxicity of the compounds were significantly improved after their, combined application. Additive effects were observed for doses lower than the calculated IC50 of berberine [IC50=23 µg/ml] and resveratrol [IC50=9 µg/ml]. Viability (MTS) assays and analysis of isobolograms revealed a significant impact on cell viability upon combination treatment.
    CONCLUSION: These results suggest that administration of berberine, in the presence of resveratrol, could be decreased even to 50% (half the IC50 for berberine) for cancer treatment.
    Keywords:  SCC-25 cells; berberine; combined treatment; plant-derived anticancer biomolecules; reactive oxygen species.; resveratrol
    DOI:  https://doi.org/10.2174/1871520619666190405111151
  18. Nutr Neurosci. 2019 Mar 30. 1-12
      BACKGROUND: An imbalance of free radicals and antioxidant defense systems in physiological processes can result in protein/DNA damage, inflammation, and cellular apoptosis leading to neurodegenerative disorders such as Alzheimer's disease (AD), Parkinson's disease (PD), and Huntington's disease (HD). Sesamin and sesamol, compounds derived from sesame seeds and oil, have been reported to exert various pharmacological effects, especially antioxidant activity. However, their molecular mechanisms against the oxidative stress induced by exogenous hydrogen peroxide (H2O2) remain to be elucidated.AIM: In this study, neuroprotective effects of sesamin and sesamol on H2O2-induced human neuroblastoma (SH-SY5Y) cell death and possible signaling pathways in the cells were explored.
    METHODS: MTT assay and flow cytometry were conducted to determine cell viability and apoptotic profiles of neuronal cells treated with sesamin and sesamol. Carboxy-DCFDA assay was used to measure reactive oxygen species (ROS). Moreover, Western blot analysis was performed to investigate protein profiles associated with neuroprotection.
    RESULTS: Pretreatment of the cells with 1 µM of sesamin and sesamol remarkably reduced the SH-SY5Y cell death induced by 400 µM H2O2 as well as the intracellular ROS production. Moreover, the molecular mechanisms underlying neuroprotection of the compounds were associated with activating SIRT1-SIRT3-FOXO3a expression, inhibiting BAX (proapoptotic protein), and upregulating BCL-2 (anti-apoptotic protein).
    CONCLUSION: The findings suggest that sesamin and sesamol are compounds that potentially protect neuronal cells against oxidative stress similar to that of the resveratrol, the reference compound. These antioxidants are thus of interest for further investigation in in vivo models of neuroprotection.
    Keywords:  Sesamin; antiapoptosis; antioxidant; neurodegenerative diseases; oxidative stress; sesamol; sirtuin
    DOI:  https://doi.org/10.1080/1028415X.2019.1596613
  19. Photodiagnosis Photodyn Ther. 2019 Apr 02. pii: S1572-1000(19)30005-5. [Epub ahead of print]
      The objective of this study was to evaluate the bactericidal effect of curcumin (CUR)-mediated photodynamic inactivation (PDI) against Escherichia coli DH5α in vitro and in oysters, then further investigate the edible security of PDI-treated oysters based on cellular toxicological methods. First of all, DH5α cells were irradiated by a 470 nm LED light source with an energy density of 3.6 J/cm2. Colony forming units (CFU) were counted and the viability of DH5α cells was calculated after treatment with CUR-mediated PDI. Intracellular production of reactive oxygen species (ROS) was studied by measuring the fluorescence of 2, 7-dichlorofluorescein (DCF) using a flow cytometry. Membrane permeability was measured using confocal laser scanning microscopy (CLSM) with propidium iodide (PI) staining. After that, the bactericidal effect of CUR-mediated PDI was evaluated in oysters which were pre-contaminated with DH5α cells. Finally, cellular toxicology of PDI-treated oysters was evaluated through morphological observation, 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide (MTT) assay, DNA ladder assay, and nuclear staining. Results showed that the viability of DH5α was significantly decreased in a CUR concentration-dependent manner and resulted in an approximately 3.5-log reduction at the concentration of 20 μM. After treatment with CUR-mediated PDI (20 μM, 3.6 J/cm2), the ROS level in DH5α cells and the membrane permeability markedly increased. Our data demonstrated that CUR-mediated PDI had a good decontamination effect against DH5α contaminated in oysters. After incubation with PDI-treated oysters, fibroblasts L929 cell morphology, MTT absorbance and cell apoptosis had no obvious changes. Our findings preliminarily demonstrated that CUR-mediated PDI-treated oysters had no cytotoxicity to fibroblasts.
    Keywords:  Cellular toxicology; Curcumin-mediated photodynamic inactivation; DH5α cells; Membrane permeability; Oysters; Reactive oxygen species
    DOI:  https://doi.org/10.1016/j.pdpdt.2019.04.002
  20. RNA Biol. 2019 Apr 01.
      Mitochondrial dynamics is a highly dysregulated process in cancer. Apoptosis and mitochondrial fission are two concurrent events wherein increased mitochondrial fragmentation serves as a hallmark of apoptosis. We have shown earlier that miR-195 exerts pro-apoptotic effects in breast cancer cells. Herein, we have demonstrated miR-195 as a modulator of mitochondrial dynamics and function. Imaging experiments upon miR-195 treatment have shown that mitochondria undergo extensive fission. We validated mitofusin2 as a potential target of miR-195. Which may provide a molecular explanation for the respiratory defects induced by miR-195 over-expression in breast cancer cells? Active, but not total, mitochondrial mass, was reduced with increasing levels of miR-195. We have further shown that miR-195 enhances mitochondrial SOD-2 expression but does not affect PINK1 levels in breast cancer cells. Collectively, we have revealed that miR-195 is a modulator of mitochondrial dynamics by targeting MFN2 thereby impairing mitochondrial function. Concomitantly, it enhances the scavenger of reactive oxygen species (SOD-2) to maintain moderate levels of oxidative stress. Our findings suggest a therapeutic potential of miR-195 in both ER-positive as well as ER-negative breast cancer cells.
    Keywords:  - MiR-195; Breast cancer cells; Mitochondrial dysfunction; Mitofusin-2
    DOI:  https://doi.org/10.1080/15476286.2019.1600999
  21. Neurochem Int. 2019 Apr 01. pii: S0197-0186(18)30346-2. [Epub ahead of print]
      AIMS: The worldwide increase in aging population is prevalently associated with the increase of neurodegenerative diseases. Peroxisome Proliferator-Activated Receptors (PPARs) are ligand-modulated transcriptional factors which belong to the nuclear hormone receptor superfamily which regulates peroxisome proliferation. The PPAR-γ is the most extensively studied among the three isoforms and the neuroprotective effects of PPAR-γ agonists have been recently demonstrated in a variety of preclinical models of neurological disorders. The aim of the study is to biologically evaluate the neuroprotective effects of new PPAR-γ selective agonists in an in vitro model.MAIN METHODS: CTX-TNA2 rat astrocytes were treated with G3335, a PPAR-γ antagonist, to simulate the conditions of a neurological disorder. Newly synthetized PPAR-γ selective agonists were added to the cell culture. Cytotoxicity was assessed by MTT assay, catalase activity was investigated by a colorimetric assay, Reactive Oxygen Species (ROS) production and apoptosis occurrence were measured by flow cytometry. Western blotting were performed to measure the levels of protein involved in the apoptotic pathway.
    KEY FINDINGS: Four PPAR-γ agonists were selected. Among them, the GL516, a fibrate derivative, showed low cytotoxicity and proved effective in restoring the catalase activity, reducing ROS production and decreasing the apoptosis occurrence triggered by the G3335 administration. The effects of this molecule appear to be comparable to the reference compound rosiglitazone, a potent and selective PPAR-γ agonist, mainly at prolonged exposure times (96 h).
    SIGNIFICANCE: Based on recent evidence, hypofunctionality of the PPAR-γ in glial cells could be present in neurodegenerative diseases and could participate in pathological mechanisms through peroxisomal damage. The fibrate derivative PPAR-γ agonist GL516 emerged as the most promising molecule of the series and could have a role in preventing the pathophysiology of neurodegenerative disorders.
    Keywords:  Apoptosis; Astrocyte; Catalase; Neurodegenerative disease; Oxidative stress; PPAR-γ agonist; Peroxisome
    DOI:  https://doi.org/10.1016/j.neuint.2019.03.021
  22. Biomolecules. 2019 Mar 29. pii: E126. [Epub ahead of print]9(4):
      Greensporone A is a fungal secondary metabolite that has exhibited potential in vitro for anti-proliferative activity in vitro. We studied the anticancer activity of greensporone A in a panel of leukemic cell lines. Greensporone A-mediated inhibition of proliferation is found to be associated with the induction of apoptotic cell death. Greensporone A treatment of leukemic cells causes inactivation of constitutively activated AKT and its downstream targets, including members GSK3 and FOXO1, and causes downregulation of antiapoptotic genes such as Inhibitor of Apoptosis (IAPs) and Bcl-2. Furthermore, Bax, a proapoptotic member of the Bcl-2 family, was found to be upregulated in leukemic cell lines treated with greensporone A. Interestingly, gene silencing of AKT using AKT specific siRNA suppressed the expression of Bcl-2 with enhanced expression of Bax. Greensporone A-mediated increase in Bax/Bcl-2 ratio causes permeabilization of the mitochondrial membrane leading to the accumulation of cytochrome c in the cytoplasm. Greensporone A-induced cytochrome c accumulation causes the activation of caspase cascade and cleavage of its effector, poly(ADP-ribose) polymerase (PARP), leading to apoptosis. Greensporone A-mediated apoptosis in leukemic cells occurs through the generation of reactive oxygen species (ROS) due to depletion of glutathione (GSH) levels. Finally, greensporone A potentiated the anticancer activity of imatinib in leukemic cells. In summary, our study showed that greensporone A suppressed the growth of leukemic cells via induction of apoptotic cell death. The apoptotic cell death occurs by inhibition of AKT signaling and activation of the intrinsic apoptotic/caspase pathways. These results raise the possibility that greensporone A could be developed as a therapeutic agent for the treatment of leukemia and other hematological malignancies.
    Keywords:  AKT; apoptosis; cIAP; greensporone A; imatinib; reactive oxygen species
    DOI:  https://doi.org/10.3390/biom9040126
  23. J Cell Physiol. 2019 Apr 04.
      Myocardial ischemia-reperfusion (IR) injury is a common cardiovascular problem, which remains a major cause of death in the world. Emerging evidence has suggested that long noncoding RNAs are crucial players in myocardial injury. However, the functional involvement of nuclear enriched abundant transcript 1 (NEAT1) in myocardial IR injury remains poorly investigated. Our study focused on the mechanism of NEAT1 in myocardial IR injury. Here, we reported a crucial role for NEAT1 in exacerbating cardiac IR injury. NEAT1 was greatly increased in myocardial IR injury mice models. As exhibited knockdown of NEAT1 resulted in attenuated myocardial IR injury in vivo. In addition, we found that NEAT1 was dramatically induced by hypoxia/reoxygenation in H9c2 cells. Lactate dehydrogenase (LDH), malondialdehyde, reactive oxygen species levels, and endoplasmic reticulum stress-regulated cardiomyocyte apoptosis were inhibited by the downregulation of NEAT1. Here, it was shown that knockdown of NEAT1 was able to repress tumor necrosis factor-α, interleukin-1β, and IL-6 expression. The silence of NEAT1 protected against IR injury via decreasing troponin levels, cardiocytes apoptosis, creatine kinase, and lactate LDH release in vivo. Meanwhile, the mitogen-activated protein kinase (MAPK) signaling was involved in NEAT1-mediated myocardial IR injury. In summary, our data indicated that NEAT1 contributed to myocardial IR injury via activating the MAPK pathway.
    Keywords:  IR injury; MAPK; NEAT1
    DOI:  https://doi.org/10.1002/jcp.28516
  24. Front Pharmacol. 2019 ;10 266
      Phellinus igniarius (P. igniarius) is a medicinal fungus that is widely used in East Asia for the adjuvant treatment of cancer. To elucidate the antitumor effective substances and mechanism of P. igniarius, we designed an approach incorporating cytotoxicity screening, phytochemical analysis, network pharmacology construction, and cellular and molecular experiments. The dichloromethane extract of P. igniarius (DCMPI) was identified as the active portion in HT-29 cells. Nineteen constituents were identified, and 5 were quantified by UPLC-ESI-Q/TOF-MS. Eight ingredients were obtained in the network pharmacology study. In total, 473 putative targets associated with DCMPI and 350 putative targets related to colon cancer were derived from online databases and target prediction tools. Protein-protein interaction networks of drug and disease putative targets were constructed, and 84 candidate targets were identified based on topological features. Pathway enrichment analysis showed that the candidate targets were mostly related to reactive oxygen species (ROS) metabolic processes and intrinsic apoptotic pathways. Then, a cellular experiment was used to validate the drug-target mechanisms predicted by the system pharmacology analysis. Experimental results showed that DCMPI increased intracellular ROS levels and induced HT-29 cell apoptosis. Molecular biology experiments indicated that DCMPI not only increased Bax and Bad protein expression and promoted PARP and caspase-3/9 cleavage but also down-regulated Bcl-2 and Bcl-xl protein levels to induce apoptosis in HT-29 cells. In conclusion, our study provides knowledge on the chemical composition and antitumor mechanism of P. igniarius, which may be exploited as a promising therapeutic option for colon cancer.
    Keywords:  Phellinus igniarius; antitumor; effective substances; mitochondrial apoptosis pathway; network pharmacology; phytochemistry
    DOI:  https://doi.org/10.3389/fphar.2019.00266
  25. Transfusion. 2019 Apr 03.
      BACKGROUND: Platelets for transfusion become senescent and dysfunctional during storage, resulting in a markedly short shelf life (5 days). We hypothesized that oxidative stress might account for this decline.STUDY DESIGN AND METHODS: Human platelets were treated with or without antioxidants before storage, and samples were collected and analyzed at different time points. Platelet senescence was determined by senescence-associated β-galactosidase assay, and senescence-related platelet qualities were also analyzed.
    RESULTS: Sign of senescence became evident after Day 3 and continued to increase over time. We also found that chemical induction of platelet activation did not affect senescence level, whereas apoptosis inducers showed a stimulative effect on platelet senescence. Moreover, this effect was not prevented by a pan-caspase inhibitor. Meanwhile, cellular and mitochondrial reactive oxygen species were found elevated during storage, and treatments with antioxidants successfully prevented this increase and also mitigated senescence levels of stored platelets. Finally, resveratrol, a natural antioxidant, was utilized as a novel storage additive to safely extend platelet shelf time. We showed that the addition of resveratrol efficiently postponed platelet senescence and ameliorated platelet storage lesion.
    CONCLUSIONS: Platelets during storage became senescent and dysfunctional over time, and we found that oxidative stress might account for this decline. The addition of antioxidants effectively postponed senescence and ameliorated platelet storage lesion, which might provide a valuable reference to future platelet storage methodologies.
    DOI:  https://doi.org/10.1111/trf.15291
  26. Front Physiol. 2019 ;10 223
      Ischemia/reperfusion (I/R) injury induces irreversible oxidative stress damage to the cardiac myocytes. Many studies have revealed that propofol alleviates the important organelle-mediated injury from oxidative stress in vitro. However, it remains unclear whether propofol prevents I/R-induced DNA damage in cardiomyocytes. In our study, we established an oxygen glucose deprivation/reoxygenation (OGD/R) model in H9c2 cells and found that propofol decreased reactive oxygen species (ROS) levels and suppressed cell apoptosis induced by OGD/R in H9c2 cells. In addition, propofol significantly reduced the molecular marker of DNA damage and inhibited double-strand breaks of DNA damage induced by OGD/R in H9c2 cells in a dose-dependent manner. Furthermore, we investigated the molecular mechanisms and demonstrated that propofol inhibited forkhead box O 1 (FoxO1) phosphorylation and increased FoxO1 nuclear translocation through inhibition of protein kinase B (Akt) and adenosine 5'-monophosphate-activated protein kinase (AMPK) pathways. The protective effects of propofol against oxidative stress-induced DNA damage were reversed by silencing FoxO1. Taken together, our results suggest that oxidative stress aggravates DNA damage and apoptosis in H9C2 cells, which can be reversed by propofol via FoxO1 nuclear translocation.
    Keywords:  DNA damage; FoxO1; ROS; oxygen glucose deprivation and reperfusion; propofol
    DOI:  https://doi.org/10.3389/fphys.2019.00223
  27. Int J Mol Med. 2019 Apr 01.
      Oxidized low‑density lipoprotein (ox‑LDL)‑induced endothelial damage contributes to the initiation and pathogenesis of atherosclerosis. Salidroside can alleviate atherosclerosis and attenuate endothelial cell injury induced by ox‑LDL. However, the mechanisms involved in this process are not fully understood. Therefore, the purpose of the present study was to investigate the role of the adenosine monophosphate‑activated protein kinase (AMPK)/sirtuin (SIRT)1 pathway in the protection of salidroside against ox‑LDL‑induced human umbilical vein endothelial cells (HUVECs) injuries. The results revealed that salidroside reverses ox‑LDL‑induced HUVECs injury as demonstrated by the upregulation of cell viability and downregulation of LDH release. In addition, salidroside increased the expression of the SIRT1 protein in ox‑LDL‑treated HUVECs. Next, it was demonstrated that SIRT1 knockdown induced by transfection with small interfering (si)RNA targeting SIRT1 (siSRT1) abolished the protection of salidroside against ox‑LDL‑induced HUVECs injuries. This was illustrated by a decrease in cell viability and an increase in LDH release, caspase‑3 activity and apoptosis rate. Furthermore, salidroside mitigated ox‑LDL‑induced reactive oxygen species production, upregulated malondialdehyde content and NADPH oxidase 2 expression and decreased superoxide dismutase and glutathione peroxidase activities, while these effects were also reversed by siSIRT1 transfection. In addition, it was demonstrated that salidroside suppressed ox‑LDL‑induced mitochondrial dysfunction as demonstrated by the increase in mitochondrial membrane potential and decreases in cytochrome c expression, and Bax/Bcl‑2 reductions. However, these effects were eliminated by SIRT1 knockdown. Finally, it was demonstrated that salidroside significantly upregulated the phosphorylated‑AMPK expression in ox‑LDL‑treated HUVECs and AMPK knockdown induced by transfection with AMPK siRNA (siAMPK) leads to elimination of the salidroside‑induced increase in cell viability and the decrease in LDH release. Notably, siAMPK transfection further decreased the expression of SIRT1. In conclusion, these results suggested that salidroside protects HUVECs against ox‑LDL injury through inhibiting oxidative stress and improving mitochondrial dysfunction, which were dependent on activating the AMPK/SIRT1 pathway.
    DOI:  https://doi.org/10.3892/ijmm.2019.4153
  28. Chem Biol Interact. 2019 Mar 29. pii: S0009-2797(19)30214-5. [Epub ahead of print]305 79-85
      Melanoma is a highly invasive cancer that resists most conventional treatments. Therefore, there is an urgent need to identify alternative anticancer agents able to affect new molecular targets. Drimys winteri (Winteraceae) is a medicinal plant, employed in Brazil and many countries, in folk medicine against a variety of ailments, especially for the treatment of fevers, ulcers, pains, affections of respiratory tract and cancers. Previous phytochemical studies have isolated and identified the presence of diverse classes of secondary metabolites in this plant such as sesquiterpenes. In an ongoing to identify new natural anticancer compounds for the treatment and/or prevention of melanoma, we study the effects of Drimys winteri bark ethyl acetate extract and its sesquiterpenes drimenol, nordrimenone, isonordrimenone and polygodial on human melanoma cells. The treatment of melanoma cells with extract, drimenol, isordrimenone and polygodial resulted in a significant reduction in cell viability. But, polygodial showed the highest inhibitory growth activity. In addition, we reported an apoptotic response after treatment with drimenol, isordrimenone and polygodial that probably involves the reduction of Hsp70 expression and reactive oxygen species production. Alternatively, the inhibition of caspase cascade at higher concentrations, correlated with additional reactive oxygen species increase, probably switches natural product-induced cell death from apoptosis to necrosis. Therefore, this evidence provides a scientific support for the anticancer employ of Drimys winteri in traditional medicinal and suggests that active molecules can be considered potential candidates to be tested also in in vivo models, alone or in combination with chemotherapy agents, for the management of melanoma.
    Keywords:  Apoptosis; Drimys winteri; Hsp70 protein; Melanoma; Reactive oxygen species; Sesquiterpenes
    DOI:  https://doi.org/10.1016/j.cbi.2019.03.029
  29. Mol Med Rep. 2019 Mar 28.
      Shikonin, a traditional Chinese medicine, has been identified as being capable of inducing apoptosis in various tumors, including glioma, and is thus considered to be a promising therapeutic agent for tumor therapy. However, little is known about the molecular mechanism of shikonin in glioma. The present study investigated the influence of shikonin on the proliferation and apoptosis of glioma cells U251 and U87MG and explored the potential molecular mechanisms. It was identified that shikonin was able to induce apoptosis in human glioma cells in a time‑ and dose‑dependent manner, and a decreased expression level of cluster of differentiation (CD)147 was observed in shikonin‑treated U251 and U87MG cells. Knockdown of CD147 inhibited U251 and U87MG cell growth, whereas CD147 overexpression enhanced cell growth and decreased shikonin‑induced apoptosis. Additionally, an increased expression level of CD147 suppressed the elevated production of reactive oxygen species and mitochondrial membrane potential levels induced by shikonin. The data indicated that shikonin‑induced apoptosis in glioma cells was associated with the downregulation of CD147 and the upregulation of oxidative stress. CD147 may be an optional target of shikonin‑induced cell apoptosis in glioma cells.
    DOI:  https://doi.org/10.3892/mmr.2019.10101
  30. In Vitro Cell Dev Biol Anim. 2019 Apr 03.
      Tetrandrine is a bisbenzylisoquinoline alkaloid known to exhibit anticancer activity against different cancers. In the present study, the cytotoxic effect of tetrandrine isolated from Cyclea peltata on pancreatic (PANC-1) and breast (MDA-MB-231) cancer cells was evaluated in vitro with an attempt to understand the role of tetrandrine on the generation of reactive oxygen species (ROS) and caspase activation. Results demonstrate the dose- and time-dependant cytotoxic effect of tetradrine on both MDA-MB-231 and PANC-1 cells with IC50 values ranging between 51 and 54 μM and 22 and 27 μM for 24 h and 48 h of incubation respectively. In addition, treatment of MDA-MB-231 and PANC-1 cells with tetrandrine showed the shrunken cytoplasm and damaged cell membrane in a dose- and time-dependant manner under the microscope. Also, tetrandrine treatment revealed an elevated levels of reactive oxygen species and increased activities of caspase-8, -9 and -3 confirming the apoptosis of cells through both extrinsic death receptor and intrinsic caspase activation. Therefore, the present study suggests the apoptosis of cells with the activation of caspase pathways mainly intrinsic pathway as a downstream event of tetrandrine-induced ROS generation. Hence, reactive oxygen species-mediated caspase activation pathway may be potentially targeted with the use of tetrandrine to treat breast and pancreatic cancers.
    Keywords:  Apoptosis; Cancer; Caspase; ROS; Tetrandrine
    DOI:  https://doi.org/10.1007/s11626-019-00332-9
  31. Toxicon. 2019 Apr 01. pii: S0041-0101(19)30107-2. [Epub ahead of print]164 10-15
      Aflatoxins are widely occurring food contaminants that are particularly harmful to dairy products and cows. The plant polyphenol resveratrol has been reported to have a good effect on increasing the resistance of cells toward toxins. Therefore, we measured the effects of aflatoxin B1 and resveratrol on the viability of the MAC-T cow mammary epithelial cell line. The appropriate treatment concentrations were assayed (12.81 μM aflatoxin B1 and 43.81 μM resveratrol) to verify the protective effect of resveratrol toward mammary epithelial cells. The results showed that resveratrol alleviates aflatoxin B1-induced cytotoxicity, including the increase in ROS and the decrease in mitochondrial membrane potential (MMP) and apoptosis in MAC-T cells. The expression of mRNA transcripts (including Nrf2, Keap1, NQO1, HO-1, SOD2 and HSP70) for components of the Nrf2 signaling pathway was evaluated by real-time fluorescent quantitative PCR, with resveratrol also exhibiting a good regulatory effect. Thus, resveratrol was shown to have an ameliorating effect on aflatoxin toxicity in MAC-T cells.
    Keywords:  Aflatoxin B1; MAC-T cell; Nrf2; Oxidative stress; Resveratrol
    DOI:  https://doi.org/10.1016/j.toxicon.2019.03.022
  32. Int Immunopharmacol. 2019 Apr 02. pii: S1567-5769(18)31416-4. [Epub ahead of print]71 361-371
      Trans sodium crocetinate (TSC) has been reported to exert a protective effect against cerebral ischemia/reperfusion (I/R) injury. However, whether TSC protects against myocardial ischemia/reperfusion (MI/R) injury remains unknown. Herein, we found that TSC treatment reduced myocardial infract size and elevated serum LDH and CK activities of MI/R rats. TSC administration attenuated oxidative stress in MI/R rats and H9C2 cells exposed to oxygen glucose deprivation/reperfusion (OGD/R). TSC administration relieved I/R-induced myocardial apoptosis in vivo and in vitro, as evidenced by reduced number of TUNEL positive cells, accompanying with marked decreases in caspase-3 activity and Bax protein level and an increase in Bcl-2 protein level. TSC treatment markedly increased SIRT3 activity and SIRT3 and SOD2 protein levels, and could also diminished the phosphorylation of FOXO3a protein. Additionally, TSC treatment attenuated the acetylation of FOXO3a and SOD2 protein. But, these effects were obviously blocked by SIRT3 knockdown. Besides, SIRT3 knockdown blocked the cardioprotective effect of TSC on OGD/R-induced oxidative stress, apoptosis and mitochondrial dysfunction in vitro. In summary, TSC alleviates I/R-induced myocardial oxidative stress and apoptosis via the SIRT3/FOXO3a/SOD2 signaling pathway. Our study suggests that TSC may become a novel drug for the treatment of MI/R injury.
    Keywords:  Ischemia/reperfusion; Myocardial injury; Trans sodium crocetinate
    DOI:  https://doi.org/10.1016/j.intimp.2019.03.056
  33. Chem Biol Interact. 2019 Apr 01. pii: S0009-2797(18)31044-5. [Epub ahead of print]
      Beta-amyloid (Aβ) has pivotal functions in the pathogenesis of Alzheimer's Disease (AD). In the present study, we adopted an vitro model that involved Aβ25-35-induced oxidative damage in PC12 cells. Aβ25-35 (10 μΜ) treatment for 24 h induced significant cell death and oxidative stress in PC12 cells, as evidenced by cell viability reduction, LDH release, ROS accumulation and increased production MDA. (1E,4E)-1, 5-bis(4-hydroxy-3-methoxyphenyl) penta-1, 4-dien-3-one (CB) and (1E, 4E)-1-(3, 4-dimethoxyphenyl)-5-(4-hydroxy-3, 5-dime-thoxyphenyl) Penta-1, 4-dien-3-one (FE), two Curcumin (Cur) analogues displayed neuroprotective effects against Aβ25-35-induced oxidative damage and cellular apoptosis in PC12 cells. Here, we investigated three different treatment ways of CB and FE. It was interesting that post-treatment of CB and FE (restoring way) showed similar effect to the preventive way, while attenuating way did not show any protective effect. We found that low dose CB and FE increased transcriptional factor NF-E2-related factor 2 (Nrf2)/hemo oxygenase 1 (HO-1) protein expression and decreased Kelch-like ECH-associated protein 1 (Keap1) in PC 12 cells. In addition, CB and FE promoted the translation of Nrf2 into nuclear and enhanced the activity of superoxide dismutase (SOD)/catalase, which confirmed cytoprotection against Aβ25-35-induced oxidative damage. Moreover, CB and FE could increase Bcl-2 expression level, decrease the level of Bax and Cyt-c in Aβ25-35-treated PC12 cells. Ultimately, the neuroprotective effect of CB and FE provides a pharmacological basis for its clinical use in prevention and treatment of AD.
    Keywords:  Aβ(25-35); Curcumin analogues; Neuroprotection; Nrf2; Oxidative stress
    DOI:  https://doi.org/10.1016/j.cbi.2019.01.010
  34. Small. 2019 Apr 03. e1900212
      A multimodal cancer therapeutic nanoplatform is reported. It demonstrates a promising approach to synergistically regulating the tumor microenvironment. The combination of intracellular reactive oxygen species (ROS) generated by irradiation of photosensitizer and endoplasmic reticulum (ER) stress induced by 2-deoxy-glucose (2-DG) has a profound effect on necrotic or apoptotic cell death. Especially, targeting metabolic pathway by 2-DG is a promising strategy to promote the effect of photodynamic therapy and chemotherapy. The nanoplatform can readily release its cargoes inside cancer cells and combines the advantages of ROS-sensitive releasing chemotherapeutic drugs, upregulating apoptosis pathways under ER stress, light-induced generation of cytotoxic ROS, achieving tumor accumulation, and in vivo fluorescence imaging capability. This work highlights the importance of considering multiple intracellular stresses as design parameters for nanoscale functional materials in cell biology, immune response, as well as medical treatments of cancer, Alzheimer's disease, etc.
    Keywords:  ROS-triggered nanoplatforms; combination cancer therapy; endoplasmic reticulum stress; photodynamic therapy; tumor microenvironment synergistical regulation
    DOI:  https://doi.org/10.1002/smll.201900212
  35. J BUON. 2019 Jan-Feb;24(1):24(1): 285-290
      PURPOSE: The purpose of the present study was to investigate the anticancer properties of isoacteoside against OVCAR-3 human ovarian cancer cells. Its effects on apoptosis, reactive oxygen species (ROS) generation, cell invasion, cell cycle arrest and its effects on tumor volume and weight were also evaluated in the current study.METHODS: MTT assay was used to study the cytotoxic effects of the compound on the cell viability. Effects on apoptosis and cell cycle arrest were evaluated by flow cytometry. In vitro wound healing assay and matrigel assay were carried out to study the effects of isoacteoside on cell migration and cell invasion respectively. Non-cancer ovarian cell line SV-40 served as control.
    RESULTS: Isoacteoside exerted both dose-dependent as well as time-dependent growth inhibitory effects on ovarian cancer cells with IC50 values of 15 µM at 24h incubation. Isoacteoside led to early and late apoptosis induction in these cells. Isoacteoside also led to sub-G1 cell cycle arrest which showed strong dose-dependence. Isoacteoside treatment also led to inhibition of cell migration and cell invasion. The results revealed that OVCAR-3 tumor growth was significantly suppressed by isoacteoside administration, compared with that in the control group. At the end of the 5-week period of isoacteoside treatment, the average tumor growth and volume in the untreated control group were considerably higher than those in the treated groups.
    CONCLUSION: In brief, the current study indicates that isoacteoside has a great potential in suppressing both in vitro and in vivo ovarian cancer cell growth and can be used as a possible anticancer agent.
  36. J Cell Biochem. 2019 Apr 02.
      BACKGROUND: Matrine might play a vital role in cardiovascular diseases progression and treatment.OBJECTIVES: We aimed to explore the protective effects and potential mechanism of matrine against diabetic cardiomyopathy (DCM) in rat model.
    METHOD: A rat model of DCM was induced by streptozotocin, which were then divided into two groups and treated with matrine. Inflammatory cytokines were investigated in serum and myocardial cells after matrine administration. The effects of matrine on cardiac reactive oxygen species (ROS) generation, Malondialdehyde (MDA) levels, and Glutathione peroxidase (GPx), PPARγ1 activity were detected in myocardial cells. The protein kinase RNA-like endoplasmic reticulum kinase (PERK) signal pathway in endoplasmic reticulum stress was studied to elaborated protective effects of matrine in DCM rat by Western blot analysis. Fasting blood glucose and hemodynamic parameters were analyzed after treatment with matrine.
    RESULTS: Matrine-inhibited expression levels of inflammatory cytokines of tumor necrosis factor alpha (TNF-α) and interleukin 6. Matrine administration decreased ROS generation, MDA, and transforming growth factor beta levels, and Peroxisome proliferator-activated receptor beta (PPARβ) and Peroxisome proliferator-activated receptorγ 1 (PPARγ1) activity. Matrine administration also significantly inhibited PERK expression. Endogenic expression of PERK canceled matrine-induced apoptosis of myocardial cells. Notably, treatment with matrine significantly decreased nonfasting blood glucose levels and improved hemodynamic parameters of DCM rat.
    CONCLUSIONS: Matrine may be a promising agent for the treatment of DCM.
    Keywords:  apoptosis; diabetic cardiomyopathy (DCM); endoplasmic reticulum (ER) stress; matrine; protein kinase RNA-like endoplasmic reticulum kinase expression (PERK) pathway; transforming growth factor beta (TGF-β)
    DOI:  https://doi.org/10.1002/jcb.28632
  37. Int J Nanomedicine. 2019 ;14 1919-1936
      Purpose: This study evaluated the effects of titanium dioxide nanoparticles (TiO2 NPs) on liver and intestine of normal rats.Methods: Male rats were divided into four groups as follows: 1) control rats, 2) control rats that orally received 10 mg/kg TiO2 NPs, 3) control rats that orally received 50 mg/kg TiO2 NPs, and 4) control rats that orally received 100 mg/kg TiO2 NPs. After 30 days, the NLRP3 inflammasome pathway (NLRP3, caspase-1, and IL-1β), antioxidant pathway (superoxide dismutase [SOD], glutathione peroxidase [GPx], and catalase [CAT]), inflammatory pathway (inducible nitric oxide synthase [iNOS] and tumor necrosis factor-α [TNF-α]), and the apoptosis pathway (p53, Bax, Bcl-2, and caspase-3) were determined in the intestine and liver of the rats. H&E and Masson's trichrome (MT) staining as well as TUNEL assay were used to examine the liver and the intestine. Biochemical factors, cytotoxicity, ROS generation, and apoptosis rate were also determined in HepG2 and Caco-2 cells.
    Results: TiO2 NPs in a dose-dependent manner increased cytotoxicity, oxidative stress, and apoptosis rate in Caco-2 and HepG2 cells. The administration of TiO2 NPs significantly reduced antioxidant enzyme activity and gene expressions (SOD, CAT, and GPx) as well as glutathione (GSH) levels and total antioxidant capacity (TAC) in a dose-dependent manner. TiO2 NPs also induced the apoptosis pathway and inflammatory pathway gene expressions and caspase-3 activity in the intestine and liver. TUNEL assay was in agreement with gene expressions. TiO2 NPs also led to morphological changes in the liver and intestine.
    Conclusion: TiO2 NPs could have cytotoxic effects on the intestine and liver structure and function by inducing oxidative stress, inflammation, and apoptosis.
    Keywords:  caco-2 cells; gene expression; hepg2 cells; rat
    DOI:  https://doi.org/10.2147/IJN.S192382
  38. Front Cell Neurosci. 2019 ;13 101
      Homer1a is the short form of a scaffold protein that plays a protective role in many forms of stress. However, the role of Homer1a in cerebral ischemia/reperfusion (I/R) injury and its potential mechanism is still unknown. In this study, we found that Homer1a was upregulated by oxygen and glucose deprivation (OGD) and that overexpression of Homer1a alleviated OGD-induced lactate dehydrogenase (LDH) release and cell death in cultured cortical neurons. After OGD treatment, the overexpression of Homer1a preserved mitochondrial function, as evidenced by less cytochrome c release, less reactive oxygen species (ROS) production, less ATP and mitochondrial membrane potential (MMP) loss, less caspase-9 activation, and inhibition of endoplasmic reticulum (ER) stress confirmed by the decreased expression of phosphate-PKR-like ER Kinase (p-PERK)/PERK and phosphate- inositol-requiring enzyme 1 (p-IRE1)/IRE1 and immunofluorescence (IF) staining. In addition, mitochondrial protection of Homer1a was blocked by the ER stress activator Tunicamycin (TM) with a re-escalated ROS level, increasing ATP and MMP loss. Furthermore, Homer1a overexpression-induced mitochondrial stress attenuation was significantly reversed by activating the PERK pathway with TM and p-IRE1 inhibitor 3,5-dibromosalicylaldehyde (DBSA), as evidenced by increased cytochrome c release, increased ATP loss and a higher ROS level. However, activating the IRE1 pathway with TM and p-PERK inhibitor GSK2656157 showed little change in cytochrome c release and exhibited a moderate upgrade of ATP loss and ROS production in neurons. In summary, these findings demonstrated that Homer1a protects against OGD-induced injury by preserving mitochondrial function through inhibiting the PERK pathway. Our finding may reveal a promising target of protecting neurons from cerebral I/R injury.
    Keywords:  PERK kinase; endoplasmic reticulum stress; homer1a; ischemic stroke; mitochondrial dysfunction
    DOI:  https://doi.org/10.3389/fncel.2019.00101
  39. Exp Gerontol. 2019 Mar 27. pii: S0531-5565(18)30543-6. [Epub ahead of print]
      Aging is a physiological process characterized by an age-progressive decline in intrinsic physiological functions, with an increased risk of developing chronic metabolic conditions, such as insulin resistance and diabetes. Furthermore, from a physiopathological point of view, several authors describe an association between oxidative stress, hypoxia and these metabolic conditions. It had been suggested that adipose tissue (AT) dysfunction, senescent cell accumulation and proinflammatory pathways may be involved in this processes. The purpose of this study was to develop an in vitro model to study the progressive morphological and functional changes of adipocytes with aging, in standard culture conditions and after severe hypoxia and hydrogen peroxide treatment. We evaluated the degree of apoptosis and intracellular reactive oxygen species (ROS) accumulation as well as the gene expression profile of aging adipocytes. Our results show that aged adipocytes become senescent, undergo apoptosis, accumulate ROS, and present an inflammatory profile with an increase in mRNA expression level of key proteins related to the remodeling of the extracellular matrix (ECM). Aged adipocytes present increased levels of p53, p21 and p16, key regulators of senescence, and a decrease in SIRT-1 protein compared to younger cells. Moreover, adipocytes aged in hypoxia or in oxidative stress conditions represent a model of accelerated aging with a decrease in their area, a greater proportion of apoptotic and of intracellular ROS accumulation compared to controls. This study characterizes the progressive morphological and functional changes in aging adipocytes during prolonged cell cultures and explores the addictive effects of hypoxia and oxidation, given at different stages of cellular maturation and senescence.
    Keywords:  Adipocytes; Cellular aging; Cellular senescence; Hypoxia; Oxidative stress
    DOI:  https://doi.org/10.1016/j.exger.2019.03.011
  40. J Pineal Res. 2019 Apr 02. e12577
      This study, using an in vitro ovary culture model, investigates the mechanisms through which Di (2-ethylhexyl) phthalate (DEHP) impairs germ cell cyst breakdown and primordial follicle assembly. The results indicate the latter effects exerted by 10 or 100 μM DEHP in cultured newborn ovaries were associated with increased levels of reactive oxygen species (ROS) and apoptosis. Based on a transcriptome analysis, we detected the expression of the oxidative stress-related gene Xdh (xanthine dehydrogenase) was significantly up-regulated in DEHP cultured ovaries. Two treatments, namely Xdh RNAi or the addition of melatonin to the ovary culture, inhibited the increase of Xdh expression and ROS levels caused by DEHP and, at the same time, reduced apoptosis and the impairment of primordial follicle assembly in the treated ovaries. Together, the results identify Xdh gene as one of the major targets of DEHP in newborn ovaries and that the consequent increased level of ROS is possibly responsible for the increment of apoptosis and primordial follicle assembly impairment. At the same time, they highlight that melatonin alleviates the effects of DEHP as with other endocrine disrupting compounds on the ovary. This article is protected by copyright. All rights reserved.
    Keywords:   DEHP ; Xdh ; apoptosis; melatonin; primordial follicle assembly
    DOI:  https://doi.org/10.1111/jpi.12577
  41. Oxid Med Cell Longev. 2019 ;2019 2843121
      Brain ageing is a complex multifactorial process characterized by gradual and continuous loss of neuronal functions. It is hypothesized that at the basis of brain ageing as well as age-related diseases, there is an impairment of the antioxidant defense system leading to an increase of oxidative stress. In this study, two different biological aspects involved in brain ageing and neurodegeneration have been investigated: oxidative stress and iron accumulation damage. In primary mouse astrocytes, the stimulation with 50 μM lipoic acid (LA) and 100 nM vitamin D (vitD) was first investigated in a time-course study to determine the dosages to be used in combination and then in a permeability test using an in vitro blood-brain barrier. In a second set of experiments, the role of oxidative stress was investigated pretreating astrocytes with 200 μM H2O2 for 30 min. The ability of vitD and LA alone and combined together to prevent or repair the damage caused by oxidative stress was investigated after 24 h of stimulation by the MTT test, mitochondrial membrane potential measurement, and Western blot analysis. To induce neurodegeneration, cells were pretreated with 300 μM catalytic iron for 6 days and then treated with vitD and LA alone and combined for additional 6 days to investigate the protection exerted by combination, analyzing viability, ROS production, iron concentration, and activation of intracellular pathways. In our study, the combination of LA and vitD showed beneficial effects on viability of astrocytes, since the substances are able to cross the brain barrier. In addition, combined LA and vitD attenuated the H2O2-induced apoptosis through the mitochondrial-mediated pathway. The combination was also able to counteract the adverse conditions caused by iron, preventing its accumulation. All these data support the hypothesis of the synergistic and cooperative activity exerted by LA and vitD in astrocytes indicating a possible new strategy to slow down ageing.
    DOI:  https://doi.org/10.1155/2019/2843121
  42. Oxid Med Cell Longev. 2019 ;2019 5769752
      Deoxynivalenol (DON) is a common contaminant of grain worldwide and is often detected in the human diet and animal feed. Selenium is an essential trace element in animals. It has many biological functions. The role of selenium in the body is mainly orchestrated by selenoprotein. Glutathione peroxidase (GPx) also exists widely in the body and has attracted much attention due to its high antioxidant capacity. In order to explore the effect of the GPx1 gene on toxicity of DON, in this study, we overexpressed or knockdown GPx1 in porcine splenic lymphocytes, then added different concentrations of DON (0.1025, 0.205, 0.41, and 0.82 μg/mL) and sodium selenite (2 μmol/L) to the culture system. Using various techniques, we detected antioxidant function, free radical content, cell apoptosis, and methylation-related gene expression to explore the effect of GPx1 expression on DON-induced cell damage. We also explored whether selenium can antagonize the toxicity of DON in these two cell models and revealed the protective effect of sodium selenite on DON-induced cell damage in GPx1-overexpressing or knockdown splenic lymphocytes. Finally, our findings revealed the following: (1) GPx1 can regulate the antioxidant capacity, apoptosis rate, and expression of DNA methylation-related genes in pig splenic lymphocytes. (2) Na2SeO3 (2 μmol/L) can regulate the antioxidant capacity, apoptosis rate, and expression of DNA methylation-related genes in pig splenic lymphocytes, and this effect is more significant in GPx1-overexpressing cells than in GPx1-knockdown cells. (3) DON can cause oxidative damage, apoptosis, and methylation injury in GPx1-overexpressing or knockdown pig splenic lymphocytes in a concentration-dependent manner. (4) Na2SeO3 (2 μmol/L) can antagonize the toxic effect of DON on GPx1-overexpressing or knockdown pig splenic lymphocytes. Our findings may have important implications for food/feed safety, human health, and environmental protection.
    DOI:  https://doi.org/10.1155/2019/5769752
  43. Cancers (Basel). 2019 Mar 30. pii: E457. [Epub ahead of print]11(4):
      The tumor microenvironment (TME) is shaped by cancer and noncancerous cells, the extracellular matrix, soluble factors, and blood vessels. Interactions between the cells, matrix, soluble factors, and blood vessels generate this complex heterogeneous microenvironment. The TME may be metabolically beneficial or unbeneficial for tumor growth, it may favor or not favor a productive immune response against tumor cells, or it may even favor conditions suited to hijacking the immune system for benefitting tumor growth. Soluble factors relevant for TME include oxygen, reactive oxygen species (ROS), ATP, Ca2+, H⁺, growth factors, or cytokines. Ca2+ plays a prominent role in the TME because its concentration is directly linked to cancer cell proliferation, apoptosis, or migration but also to immune cell function. Stromal-interaction molecules (STIM)-activated Orai channels are major Ca2+ entry channels in cancer cells and immune cells, they are upregulated in many tumors, and they are strongly regulated by ROS. Thus, STIM and Orai are interesting candidates to regulate cancer cell fate in the TME. In this review, we summarize the current knowledge about the function of ROS and STIM/Orai in cancer cells; discuss their interdependencies; and propose new hypotheses how TME, ROS, and Orai channels influence each other.
    Keywords:  H2O2; Orai; STIM; calcium; reactive oxygen species; tumor microenvironment
    DOI:  https://doi.org/10.3390/cancers11040457
  44. Cancer Lett. 2019 Mar 28. pii: S0304-3835(19)30190-9. [Epub ahead of print]
      Natural products possess a significant role in anticancer therapy and many currently-used anticancer drugs are of natural origin. Cerberin (CR), a cardenolide isolated from the fruit kernel of Cerbera odollam, was found to potently inhibit cancer cell growth (GI50 values <90 nM), colony formation and migration. Significant G2/M cell cycle arrest preceded time- and dose-dependent apoptosis-induction in human cancer cell lines corroborated by dose-and time-dependent PARP cleavage and caspase 3/7 activation, in addition to reduced Bcl-2 and Mcl-1 expression. CR potently inhibited PI3K/AKT/mTOR signalling depleting polo-like kinase 1 (PLK-1), c-Myc and STAT-3 expression. Additionally, CR significantly increased the generation of reactive oxygen species (ROS) producing DNA double strand breaks. Preliminary in silico biopharmaceutical assessment of CR predicted >60% bioavailability and rapid absorption; doses of 1-10 mg/kg CR were predicted to maintain efficacious unbound plasma concentrations (>GI50 value). CR's potent and selective anti-tumour activity, and its targeting of key signalling mechanisms pertinent to tumourigenesis support further preclinical evaluation of this cardiac glycoside.
    Keywords:  Apoptosis; Cardenolide; Cerbera odollam; DNA Damage; Reactive oxygen species
    DOI:  https://doi.org/10.1016/j.canlet.2019.03.034
  45. 3 Biotech. 2019 Apr;9(4): 147
      Beetroot (Beta vulgaris L.) juice (BRJ) is a good source of betalain (betacyanins and betaxanthin) pigments and exhibits antioxidant, anti-inflammatory, and chemo-preventive activities in vitro and in vivo. The current study was performed to determine the cardioprotective effect of BRJ on lipid peroxidation, antioxidant defense, functional impairment, and histopathology in rats with isoproterenol (ISP)-induced myocardial injury. Myocardial ischemia was induced by ISP (85 mg/kg) s.c. injection at 24 h intervals, followed by oral administration of BRJ for 28 days at doses of 150 and 300 mg/kg. ISP-induced myocardial damage was confirmed by an increase in heart weight to body weight ratio, % infarction size, serum cardiac indices (AST, ALT, GGT, ALP, LDH and CK-MB), and histological alterations in the myocardium. Pretreatment with BRJ (150 and 300 mg/kg) followed by ISP induction reduced oxidative/nitrosative stress and restored the cardiac endogenous antioxidants in rats. ISP augmented cardiac inflammatory cytokines (TNF-α, IL-6 and IL-10), myeloperoxidase activity, NF-κB DNA binding and protein expression of NF-κB (p65), and the hyperlipidemia level was significantly reduced by the BRJ pretreatment. Furthermore, the BRJ pretreatment significantly reduced caspase-3, Bax, and MMP-9 protein expression, enhanced the Bcl-2 antiapoptotic protein expression, alleviated the extent of histological damage, myonecrosis, and edema, and maintained the architecture of cardiomyocytes. These findings suggest that BRJ pretreatment mitigates cardiac dysfunction and structural damages by decreasing oxidative stress, inflammation, and apoptosis in cardiac tissues. These results further support the use of BRJ in traditional medicine against cardiovascular diseases.
    Keywords:  Beetroot juice; Cardioprotection; Isoproterenol; Myocardial injury; Oxidative stress
    DOI:  https://doi.org/10.1007/s13205-019-1677-9
  46. Mol Med Rep. 2019 Mar 22.
      The present study aimed to investigate the potential effects of growth differentiation factor 11 (GDF11) on isoproterenol (ISO)‑induced heart failure (HF) and identify the underlying molecular mechanisms. A rat model of HF was induced in vivo by intraperitoneally administering ISO (5 mg/kg/day) for 7 days. After 4 weeks following establishment of the HF model, hemodynamic analysis demonstrated that ISO induced a significant increase in the left ventricular end‑diastolic pressure and a decrease in the left ventricular systolic pressure and maximum contraction velocity. The plasma levels of myocardial injury markers, including lactate dehydrogenase (LDH), creatine kinase (CK), CK‑muscle/brain which were determined using the corresponding assay kits and plasma brain natriuretic peptide which was detected by an ELISA kit, an important biomarker of HF, increased following ISO treatment. Furthermore, levels of GDF11 expression and protein, which were estimated using reverse transcription‑quantitative polymerase chain reaction and an ELISA kit in plasma and western blotting in the heart tissue, respectively, significantly increased following ISO treatment. To demonstrate the effects of ISO on GDF11 production in cardiomyocytes, H9C2 cells (a cardiomyoblast cell line derived from embryonic rat heart tissue) were treated with ISO (50 nM) for 24 h in vitro; it was revealed that GDF11 protein and mRNA expression levels significantly increased following ISO treatment. In addition, recombinant GDF11 (rGDF11) administered to ISO‑treated H9C2 cells resulted in decreased proliferation, which was detected via a CCK‑8 assay, and increased LDH levels and cell apoptosis of cells, which was determined using Caspase‑3 activity and Hoechst 33258 staining. Additionally, rGDF11 increased the levels of reactive oxygen species and malondialdehyde due to the upregulation of nicotinamide adenine dinucleotide phosphate oxidase 4 (Nox4) following rGDF11 treatment. Conversely, GDF11 knockdown reduced ISO‑induced apoptosis by inhibiting oxidative stress injury. The results suggested that GDF11 production was upregulated in ISO‑induced rats with HF and in ISO‑treated H9C2 cells, and that rGDF11 treatment increased ISO‑induced oxidative stress injury by upregulating Nox4 in H9C2 cells.
    DOI:  https://doi.org/10.3892/mmr.2019.10077
  47. Evid Based Complement Alternat Med. 2019 ;2019 6378786
      Objective: To investigate the effect of FTZ on high-glucose-induced oxidative stress and underlying mechanisms.Methods: We used a β cell dysfunction and diabetes model that was induced in rats fed a high-fat high-sugar diet (HFHSD) for 6 weeks and injected once with 35 mg/kg streptozocin (STZ). Then, 3 and 6 g/kg of FTZ were administered by gavage for 8 weeks. In addition, an ex vivo model of oxidative stress was induced by stimulating INS-1 cells with 25 mmol/L glucose for 48 h.
    Result: The levels of fasting blood glucose (FBG) in diabetic model rats were obviously higher than those in the normal group; furthermore with reduced levels of β cells, catalase (CAT), superoxide dismutase (SOD), and Bcl-2 increased lipid peroxide malondialdehyde (MDA) and caspase-3 in the pancreatic tissue of the diabetic model rats. Afterward, the cells were incubated with FTZ-containing serum and edaravone. The 25 mmol/L glucose-induced SOD reduction increased MDA and intracellular ROS. The protein expression level of Mn-SOD and CAT in the model group decreased significantly compared with that in the control group.
    Conclusion: FTZ treatment significantly improved the alteration in the level of SOD, CAT, Bcl-2, caspase-3, and MDA coupled with β cell dysfunction in diabetic rats. Oxidative stress in INS-1 cells was closely associated with a higher rate of apoptosis, increased production of ROS and MDA, enhanced Bax expression, and caspase-3, -9 activities and markedly decreased protein expression of Mn-SOD and CAT. FTZ-containing serum incubation notably reversed the high-glucose-evoked increase in cell apoptosis, production of ROS and MDA, and Bax protein levels. Furthermore, FTZ stimulation upregulated the expression levels of several genes, including Mn-SOD, CAT, and Bcl-2/Bcl-xl. In addition, FTZ decreased the intracellular activity of caspase-3, -9 in INS-1 cells. FTZ protected β-cells from oxidative stress induced by high glucose in vivo and in vitro. The beneficial effect of FTZ was closely associated with a decrease in the activity of caspase-3, -9 and intracellular production of ROS, MDA, and Bax coupled with an increase in the expression of Mn-SOD, CAT, and Bcl-2/Bcl-xl.
    DOI:  https://doi.org/10.1155/2019/6378786
  48. Clin Cancer Res. 2019 Apr 02. pii: clincanres.3223.2018. [Epub ahead of print]
      PURPOSE: Acute myeloid leukemia (AML) is a hematological malignancy characterized by the accumulation of immature myeloid precursor cells. AML is poorly responsive to conventional chemotherapy and a diagnosis of AML is usually fatal. More effective and less toxic forms of therapy are desperately needed. AML cells are known to be highly dependent on the amino acid glutamine for their survival. These studies were directed at determining the effects of glutaminase inhibition on metabolism in AML and identifying general weaknesses that can be exploited therapeutically.EXPERIMENTAL DESIGN: AML cancer cell lines, primary AML cells, and mouse models of AML and acute lymphoblastic leukemia (ALL) were utilized.
    RESULTS: We show that blocking glutamine metabolism through the use of a glutaminase inhibitor (CB-839) significantly impairs antioxidant glutathione production in multiple types of AML, resulting in accretion of mitochondrial reactive oxygen species (mitoROS) and apoptotic cell death. Moreover, glutaminase inhibition makes AML cells susceptible to adjuvant drugs that further perturb mitochondrial redox state, such as arsenic trioxide (ATO) and homoharringtonine (HHT). Indeed, the combination of ATO or HHT with CB-839 exacerbates mitoROS and apoptosis, and leads to more complete cell death in AML cell lines, primary AML patient samples and in vivo using mouse models of AML. In addition, these redox-targeted combination therapies are effective in eradicating ALL cells in vitro and in vivo Conclusions: Targeting glutamine metabolism in combination with drugs that perturb mitochondrial redox state represents an effective and potentially widely applicable therapeutic strategy for treating multiple types of leukemia.
    DOI:  https://doi.org/10.1158/1078-0432.CCR-18-3223
  49. J Agric Food Chem. 2019 Apr 01.
      The objective of this study was to investigate the mechanism underlying lysosome-mediated apoptosis and the cross-talk between the lysosomes and mitochondria and the effect of the pathway on bovine longissimus muscle tenderness during postmortem aging. The Longissimus Doris (LD) muscles of six crossbred cattle were used to observe the lysosome-mediated apoptosis, the cross-talk between the lysosomes and mitochondria, as well as the effect of lysosomal-mitochondrial apoptotic pathway on the tenderness of LD muscle during 7 d postmortem aging. Results showed that elevated reactive oxygen species level (P<0.05) can damage lysosomal membrane stability (P<0.05) through accumulating redox-active iron of bovine muscle during postmortem aging. In addition, the activities of cathepsins B and D increased with postmortem aging (P<0.05). Moreover, cathepsin B and D activated Bid and Bax in the mitochondria (P<0.05). Activated Bid and Bax triggered mitochondrial membrane permeability (P<0.05) and further activated caspase-9 and caspase-3 (P<0.05), leading to apoptosis. Ultimately, the tenderness of bovine muscle was improved during postmortem aging (P<0.05). Importantly, cathepsin D plays a crucial role in the lysosomal-mitochondrial apoptotic pathway and tenderness in postmortem muscle. These findings provide new insights into the apoptotic pathway of bovine muscle during postmortem aging.
    DOI:  https://doi.org/10.1021/acs.jafc.9b00894
  50. J Oncol. 2019 ;2019 8701824
      Therapeutic options for advanced stage cholangiocellular carcinoma (CCC) are very limited as of today and patients carry an exceptionally poor overall prognosis. In recent years, increasing evidence has been accumulated to suggest that malignant cells widely show increased intrinsic ROS levels and exhibit altered redox profiles as compared to normal counterparts, opening up potential avenues for therapeutic intervention. This study provides preclinical experimental evidence of therapeutic activity of the curcumin analog EF24 in cholangiocarcinoma models. In CCC cell lines, EF24 inhibited cell viability and induced apoptosis through excessive ROS generation. Moreover, administration of EF24 led to depletion of total intracellular GSH levels, induced mitochondrial depolarization, and abrogated STAT3 phosphorylation. Of interest, these effects were readily averted by treating the cells with exogenous antioxidants such as N-acetyl cysteine (NAC) or glutathione monoethyl ester (GEE). In vivo, EF24, solubilized using a cyclodextrin formulation, significantly suppressed the growth of tumor xenografts without exhibiting any toxic adverse effects. Immunohistochemical analysis of extracted tumor tissues demonstrated reduced nuclear staining for Ki-67 and downregulation of phospho-STAT3 as well as strong staining for oxidative stress biomarker 8-OHdG. Therefore, the data presented here suggest EF24 as potential therapeutic compound against CCC which might act at least to some extent through ROS-induced oxidative damage, subsequently inducing apoptosis. Further evaluation of this approach should be carried out in future follow-up studies.
    DOI:  https://doi.org/10.1155/2019/8701824
  51. J Cell Physiol. 2019 Apr 03.
      It is known that irisin increases total body energy expenditure, decreases body weight, and enhances insulin sensitivity. Although previous studies have demonstrated that irisin induces vascular endothelial cell (EC) angiogenesis, the molecular mechanisms underlying irisin-induced angiogenesis under conditions reflecting atherosclerosis are not known. The aim of the present study is to investigate whether irisin could inhibit oxidized low-density lipoprotein (oxLDL) impaired angiogenesis. We investigated the effect of irisin on angiogenesis in vitro by evaluating cell viability, cell migration, and the capacity to form capillary-like tubes using human umbilical vein endothelial cells and human microvascular endothelial cells (HUVECs and HMEC-1) that were treated with oxLDL. We also evaluated the effects of irisin on angiogenesis in vivo by Matrigel plug angiogenesis assay and in a chicken embryo membrane (CAM) model. Our results demonstrated that irisin increased oxLDL-treated EC viability as well as migration and tube formation. Moreover, oxLDL inhibited angiogenic response in vivo, both in the Matrigel plug angiogenesis assay and in the CAM model, and was attenuated by irisin. Furthermore, irisin decreased apoptosis, inflammatory cytokines, and intracellular reactive oxygen species (ROS) levels in oxLDL-treated EC. In addition, we found that irisin upregulated pAkt/mTOR/Nrf2 in oxLDL-treated EC. Both mTOR/Nrf2 shRNA and LY294002 could inhibit the protective effect of irisin. Taken together these results, they suggested that irisin attenuates oxLDL-induced vascular injury by activating the Akt/mTOR/Nrf2 pathway. Our findings suggest that irisin attenuates oxLDL-induced blood vessel injury.
    Keywords:  angiogenesis; human microvascular endothelial cells; human umbilical vein endothelial cells; irisin; oxLDL
    DOI:  https://doi.org/10.1002/jcp.28535
  52. Life Sci. 2019 Mar 29. pii: S0024-3205(19)30245-0. [Epub ahead of print]
      AIMS: Diabetic nephropathy is a growing health concern, which is reported to be associated with inflammation. Luteolin has been explored for the treatment of some diabetic complications. Although several studies have verified the effect of luteolin on diabetic nephropathy, the mechanism by which the therapeutic effects of luteolin on diabetic nephropathy has not been established. Therefore, we aimed to investigate the effect of luteolin on diabetic nephropathy and its underlying mechanism.MAIN METHODS: We used western blot, Real-time PCR, immunofluorescence and flow cytometry to analyze the effects of luteolin on podocyte injury and NOD-like receptor family and pyrin domain-containing protein 3 (NLRP3) inflammasome activation in high glucose (HG) condition. Reactive oxygen species (ROS) generation was measured by flow cytometry and malondialdehyde (MDA) level. To investigate the potential mechanism, we examined cell apoptosis upon transfection of siNLRP3.
    KEY FINDINGS: We showed that luteolin treatment could protect podocyte against HG-induced cell apoptotic and mitochondrial membrane potential collapse. In addition, luteolin significantly reduced NLRP3 inflammasome formation and subsequent interleukin-1β (IL-1β) secretion in HG-induced MPC-5 cells. Interestingly, siNLRP3 abolished the effect of luteolin on cell apoptosis, suggesting that the anti-apoptotic effect was found to be mostly related to NLRP3 inflammasome.
    SIGNIFICANCE: In summary, our data demonstrated the abilities of luteolin to inhibit podocyte injury and NLRP3 inflammasome activation, which could be used in the treatment of diabetic nephropathy.
    Keywords:  Apoptosis; Diabetic nephropathy; Luteolin; NLRP3 inflammasome; Podocyte; ROS
    DOI:  https://doi.org/10.1016/j.lfs.2019.03.073
  53. Life Sci. 2019 Apr 02. pii: S0024-3205(19)30255-3. [Epub ahead of print]
      AIMS: Hydrogen gas (H2) has a diversity of effects such as anti-apoptotic, anti-inflammatory and anti-oxidative properties. However, molecular mechanism underlying the potential effect of H2 on chronic intermittent hypoxia (CIH) induced renal injury remains obscure.MATERIALS AND METHODS: In the present study, adult male Sprague-Dawley rats were randomly allocated into four groups: control (CON) group, CIH group, CIH with H2 treatment (CIH + H2) group, and control with H2 treatment (CON + H2) group. Oxidative stress, autophagy and endoplasmic reticulum (ER) stress were detected to determine how H2 affected the renal function of CIH exposed rats.
    KEY FINDINGS: We demonstrated that rats who inhale hydrogen gas showed improved renal function, alleviated pathological damage, oxidative stress and apoptosis in CIH rats. Meanwhile, CIH-induced endoplasmic reticulum stress was decreased by H2 as the expressions of CHOP, caspase-12, and GRP78 were down-regulated. Furthermore, relative higher levels of LC3-II/I ratio and Beclin-1, with decreased expression of p62, were found after H2 administrated. Inhibition of mTOR may be involved in the upregulation of autophagy by H2. Finally, increased phosphorylation of p38 and JNK was involved in the CIH-induced pathological process. H2 could inhibit the activation of p38 and JNK, suggesting H2 played an active part in resisting renal injury via MAPK.
    SIGNIFICANCE: Taken together, our study reveals that H2 can ameliorate CIH-induced kidney injury by decreasing endoplasmic reticulum stress and activating autophagy through inhibiting oxidative stress-dependent p38 and JNK MAPK activation.
    Keywords:  Autophagy; Chronic intermittent hypoxia; ER stress; Hydrogen; Kidney
    DOI:  https://doi.org/10.1016/j.lfs.2019.04.005
  54. Phytother Res. 2019 Apr 01.
      Pharmacological studies have shown that various species of Ficus have antiviral, antidiarrheal, antipyretic, hypolipidemic, antidiabetic, antioxidant, anticancer, antiparasitic, antiangiogenic, anti-inflammatory, antibacterial, antiplatelet, reproductive, dermatological, immunological, endocrine, and hepato and nephron protective effects. But there is no sufficient research on biomolecules present in the leaf extract of Ficus religiosa and its mechanism of action. We have previously reported that bioavailable constituents of F. religiosa leaf extract exert photosensitizing and apoptosis-inducing capability through the generation of intracellular reactive oxygen species on breast cancer cells. In this review, we have evaluated the expression of checkpoint proteins of G1/S and sub G0 phase with wet lab data and also have done a data mining of other research for other potential mechanistic action of the F. religiosa leaf extract.
    Keywords:  Ficus religiosa; breast cancer; chemo sensitizer; photosensitizing effect
    DOI:  https://doi.org/10.1002/ptr.6348
  55. J Biol Chem. 2019 Apr 05. pii: jbc.RA119.007648. [Epub ahead of print]
      Excessive activation of the renin-angiotensin system (RAS) in diabetic cardiomyopathy (DCM) provokes a series of structural and functional abnormalities, and causes ventricular remodeling and heart failure in diabetes. (Pro)renin receptor (PRR) is a component of the RAS and has been reported to be up-regulated in some cardiovascular diseases. Furthermore, PRR blockade in some cardiovascular diseases, such as myocardial infarction and hypertension, has been demonstrated to reverse their pathogenesis. However, there have been few studies about the function of PRR in the pathogenesis of DCM. In this study, we hypothesized that PRR is involved in the pathogenesis of DCM and mediates myocardial injury in DCM. To explore the role of PRR in DCM, we evaluated the effects of PRR overexpression and knockdown on the DCM phenotype in vivo and in vitro. The results show that PRR overexpression exacerbates myocardial injury and the inflammatory response in rats with DCM. Conversely, PRR knockdown alleviates myocardial fibrosis, apoptosis, and the inflammatory response, reversing the cardiac dysfunction in rats with DCM. In cell experiments, PRR overexpression also up-regulated the protein expression of collagen I and fibronectin, aggravated the inflammatory response, and increased the production of reactive oxygen species (ROS), while PRR knockdown had the opposite effect. Thus, PRR mediates myocardial injury, apoptosis, and the inflammatory response, likely through a PRR/extracellular signal-regulated kinase/ROS pathway.
    Keywords:  (pro) renin receptor; apoptosis; cardiomyopathy; diabetes; inflammation; myocardial injury; oxidative stress
    DOI:  https://doi.org/10.1074/jbc.RA119.007648
  56. J Inorg Biochem. 2019 Mar 23. pii: S0162-0134(18)30490-2. [Epub ahead of print]195 111-119
      Alpha lipoic acid (α-LA) and its reduced form dihydrolipoic acid (DHLA) have been historically considered as excellent anti-oxidants and oxidative stress scavengers. Upon oxidation with reactive oxygen species (ROS) and pro-oxidants, α-LA may be reconstituted from DHLA and other reduced forms. Oxidative stress is one of the fundamental causes of functional degeneration, autophagy and apoptosis leading to cytotoxicity and loss of cell survival, often due to exposure to xenobiotics, pollutants, heavy metals, and other environmental and endogenous toxicants. α-LA and DHLA can react with these molecules to strengthen the primary antioxidant defense system during cell injury. The compound α-LA is suggested for heavy metal detoxification, in particular for supporting the mercury (Hg) detoxifying process. Mercury is one of the major environmental toxicant, particularly noxious even upon limited exposure. Oxidative stress pathways have been identified as a key upstream event for Hg-induced toxicity in humans and animals. However, very few existing drugs to date can successfully prevent or reduce Hg toxicity. Although several thiol-based chelators, such as British Anti-Lewisite (2,3-dimercaptopropanol, BAL), meso-2,3-dimercaptosuccinic acid (DMSA), and sodium 2,3-dimercapto-1-propanesulfonate (DMPS), have shown promise for ameliorating Hg intoxication. In this review, the potential role of α-LA and DHLA in scavenging toxic metals and other xenobiotics is discussed, focusing especially on the mechanisms of actions of α-LA and DHLA as potential antioxidants towards Hg-induced toxicity.
    Keywords:  Alpha lipoic acid; Dihydrolipoic acid; Mercury
    DOI:  https://doi.org/10.1016/j.jinorgbio.2019.03.019
  57. Eur J Med Chem. 2019 Mar 21. pii: S0223-5234(19)30262-4. [Epub ahead of print]171 420-433
      (E)-3,4-dihydroxystyryl alkyl sulfones, as new analogues of neurodegenerative agents, were designed and synthesized. The biological results demonstrated that most of the target compounds preserved antioxidant and anti-inflammatory potency in scavenging reactive free radicals, protecting neuronal cells against neurotoxins such as H2O2, 6-hydroxydopamine and inhibiting lipopolysaccharide (LPS)-induced over-production of NO. Among these compounds, 6.22 with cyclopentyl propyl exhibited prominent antioxidant activity at low concentration (2.5 μM) in H2O2 model (cell viability = 94.5%). In addition, 6.22 (IC50 = 1.6 μM) displayed better anti-inflammatory activity than that of lead compound 1 (IC50 = 13.4 μM). In view of the outstanding performance of 6.22, the apoptotic rates of H2O2-damaged PC12 cells were detected by Annexin V-FITC/PI assay. 6.22 showed higher potency in inhibition of apoptosis than 1 at low concentration (2.5 μM), consisting with the antioxidant and anti-inflammatory models. Furthermore, with the predicted CNS (+) blood-brain barrier (BBB) permeability (Pe = 6.84 × 10-6 cm s-1), low cytotoxicity and favorable physiochemical properties based on calculation, compound 6.22 can be further developed as a potential multifunctional neuroprotective agent.
    Keywords:  Annexin V-PI apoptosis stain; Anti-inflammatory; Antioxidant; BBB permeability; Neurodegenerative diseases
    DOI:  https://doi.org/10.1016/j.ejmech.2019.03.044
  58. Nutr Cancer. 2019 Apr 04. 1-10
      AIM: Hepatocellular carcinoma is one of the leading global epidemics. A medicinal tree, Moringa oleifera (MO), has been part of traditional treatments including cancer therapies. We investigated the apoptosis inducing effects of MO crude aqueous leaf extract (MOE) in human liver hepatocellular carcinoma (HepG2) cells.METHODS: HepG2, PBMCs and Hek293 cell viability was evaluated using MTT assay. Oxidative stress and DNA damage was determined using TBARS and comet assays, respectively. Apoptosis was assessed by caspase-9, -3/7 activities and ATP levels (luminometry). Cell cycle, γH2AX, and cleaved PARP-1 were determined (flow cytometry). Protein expression of c-myc, Bax, p-Bcl2, Smac/DIABLO, Hsp70, SRp30a and cleaved PARP-1 was assessed using western blotting.
    RESULTS: MOE displayed minimal toxicity in PBMCs and Hek293 cells for 24 h. HepG2 cells were exposed to MOE (24 h) and an IC50 (4.479 mg/mL) was determined. MOE significantly increased lipid peroxidation, DNA damage and γH2AX levels. A significant decrease in G1, S and G2-M phase was seen. Significant increase in SRp30a protein expression activated caspase-9. Caspase-9 and -3/7 was significantly increased with significant decrease in ATP levels. Apoptosis was confirmed with significant decrease in c-myc, p-Bcl2 and Hsp70 protein expression and a significant increase in Bax, Smac/DIABLO and PARP-1 cleavage.
    CONCLUSION: MOE induces cell-cycle arrest and apoptosis in cancerous HepG2 cells.
    DOI:  https://doi.org/10.1080/01635581.2019.1597136
  59. J Inorg Biochem. 2019 Mar 21. pii: S0162-0134(18)30580-4. [Epub ahead of print]195 83-90
      Giardiasis is a widespread illness that affects inhabitants of underdeveloped countries, being children and seniors the highest risk population. The several adverse effects produced by current therapies besides its increasing ineffectiveness due to the appearance of resistant strains evidence the urgent need for new therapeutic approaches. We present the antigiardiasic effect of eight Cu(II) coordination compounds, which belong to the family Casiopeínas. Two of them, 4,7-diphenyl-1,10-phenanthroline(acetylacetonato)copper(II) nitrate (CasIII-Ha,36 μM) and 4,7-diphenyl-1,10-phenanthroline(glycinato)copper(II) nitrate (CasI-gly,36 μM) have shown the best antiproliferative effect in Giardia intestinalis trophozoite cultures, both with the higher lipophilic character of the series. The antiproliferative effect of these coordination compounds is attributable to its capacity to interact with the cellular membrane and to increase reactive oxygen species (ROS) concentration within the parasite since the first hours of exposure, (2-6 h). We found that these compounds mainly induced the cell death of trophozoites by apoptosis, contrary to metronidazole, which induces apoptosis and necrosis in the same ratio. The cytotoxic effects on lymphocytes and macrophages isolated from human peripheral blood allowed us to establish a selectivity index and in turn, identify and propose the best candidates to continue with the assays in animal models. The selected molecules do not include the most active compounds against trophozoites, instead of that, we propose the compounds 4',4'-dimethyl-2,2'-bipyridine(acetylacetonato)copper(II) nitrate (CasIII-ia,IC50 = 156 μM) and 4,7-dimethyl-1,10-phenanthroline(acetylacetonato) copper(II) nitrate (CasIII-Ea,IC50 = 125 μM), which possess an antiproliferative efficacy comparable with Metronidazole but also are those that produce the lowest effect on the viability of human lymphocytes and macrophages.
    Keywords:  Antigiardiasic activity; Cu(II) coordination compounds; Giardia intestinalis; Membrane damage; Oxidative stress
    DOI:  https://doi.org/10.1016/j.jinorgbio.2019.03.012
  60. Anticancer Agents Med Chem. 2019 Apr 05.
      BACKGROUND: Punicic acid (PA) is a polyunsaturated fatty acid that accounts for approximately 70%-80% of pomegranate seed oil (PSO). PA possess strong antioxidant, anti-inflammatory, anti-atherogenic effects, and anti-tumorigenic properties. Pomegranate extracts have been shown to have anticancer activity in many studies. However, there is no evidence for effect of PSO on T98 gliobalstoma cells. Therefore, the present study was the first to investigate the mechanisms induced by PA on T98 cells, which is one of the major compounds extracted from PSO.METHODS: The effects of PA on cell viability; oxidative stress; and migration, proliferation, and apoptosis at the IC50 dose were studied.
    RESULTS: The proliferation and migration were inhibited in treated group compared to non-treated group by 9,85 μl/ml PA. The difference was statistically significant (***p<0,001). Furthermore, PA induced apoptosis in the T98 glioblastoma cells compared to non-treated group and the difference was statistically significant (***p<0,001). Apoptosis was determined via immunocytochemistry staining of caspase-3, caspase-9 and TUNEL methods. Apoptosis was checked by flow cytometry (using caspase 3 method) and Scanning Electron Microscopy Analysis. We also investigated the potential signaling pathway underlying this apoptotic effect. The immunocytochemical stainings of PI3K/ Akt-1/ mTOR-1 demonstrated that, Akt-1 staining was increased with PA treatment similar to mTOR-1 and PI3K staining (***p<0,001). These increases were statistically significant compared to non-treated group.
    CONCLUSION: PA exhibited exceptional abilities as an anticancer agent against GBM cells. The use of punicic acid in combination with other drugs used in the treatment of glioblastoma may increase the efficacy of the treatment. This study provided a basis for future investigation of its use in preclinical and clinical studies.
    Keywords:  Glioblastoma multiforme; PI3K/AKT1/mTOR; Punicic acid; Tumor infiltration; apoptosis; glioblastoma; signaling pathway
    DOI:  https://doi.org/10.2174/1871520619666190405112507
  61. Hum Exp Toxicol. 2019 Apr 03. 960327119839185
      Coconut oil (CO) is enriched with medium chain saturated fatty acids like lauric acid (LA), capric acid and caprylic acid, which are known to have several health benefits. LA, the predominant fatty acid in CO, is reported to possess anticancer activity mediated through oxidative stress-induced apoptosis; however, there is no clear information on its cellular signalling mechanism. The present study screened the anticancer potential of various fatty acids present in CO (capric acid, caprylic acid and LA) using in silico tools such as CDOCKER in Accelrys Discovery Studio by targeting proteins like epidermal growth factor receptor (EGFR), cyclin-dependent kinase and thymidine synthase (TS). The results were further confirmed using cell culture-based studies and quantitative PCR. Among the tested compounds, LA was found to be the most active and showed a higher affinity towards EGFR and TS. Corroborating with these results, LA-induced dose-dependent cytotoxicity towards HCT-15 (human colon cancer), HepG2 (human hepatocellular carcinoma) and Raw 264.7 (murine macrophages) cells exhibiting morphological characteristics of apoptosis. Further, in HCT-15 cells exposed to LA (30 and 50 µg/mL), the expression of EGFR was found to be downregulated by 1.33- and 1.58-fold. The study thus concludes that the anticancer activity of LA may be partially mediated by the downregulation of EGFR signalling and consequent reduction in cell viability through apoptosis. Since EGFR signalling is crucial in cancer cell survival and is a prime target in drug development, the present study has pharmacological significance.
    Keywords:  Discovery Studio; EGFR; Lauric acid; coconut oil; colon cancer
    DOI:  https://doi.org/10.1177/0960327119839185
  62. Mol Neurobiol. 2019 Apr 03.
      Amitriptyline, antidepressant frequently prescribed for treatment of depressive disorders and several neuropathic and inflammatory diseases, has been shown to cause neurotoxic effects. This effect has been partially linked with increased oxidative stress and apoptosis initiation; however, the exact mechanism is still unknown. Klotho protein due to its neuroprotective characteristics seems to be involved in the amitriptyline-mediated neurotoxicity. In this study, we have evaluated the effect of klotho silencing on mouse hippocampal cells exposed to amitriptyline. We show, for the first time, that klotho silencing intensified in hippocampal neurons amitriptyline-induced imbalance in oxido-nitrosative and mineral homeostasis, genomic instability associated with telomere dysfunction what resulted in p16- and p53/p21-mediated cell cycle arrest and activation of autophagy and apoptotic cell death in consequence. Therefore, these results indicate that klotho serves as a part of the cellular defense mechanism engaged in the protection of neurons against amitriptyline-mediated toxicity.
    Keywords:  Amitriptyline; Apoptosis; Autophagy; Hippocampal cells; Klotho; Telomeres
    DOI:  https://doi.org/10.1007/s12035-019-1575-5
  63. Phytomedicine. 2019 Mar 01. pii: S0944-7113(18)30545-2. [Epub ahead of print]55 320-329
      BACKGROUND: It has been reported that n-butanol extract of Potentilla anserina L (NP) had protective effect against acute myocardial ischemia/reperfusion (I/R) injury in mice. Because of limited phytochemical study on NP, its bioactive compounds and underlying protective mechanisms are largely unclear.PURPOSE: The purpose of this study was to investigate the major bioactive compounds and possible mechanism for the cardioprotective effect of NP on rat with I/R injury.
    METHODS: We analyzed the phytochemical isolation of NP and identified the structure of compounds, which was elucidated by a combination of spectroscopic analyses. An I/R model was established by I-30 min/R-2 h in Sprage-Dawley rats. The rats were given intragastric administration of NP (49.3, 98.6, and 197.2 mg•kg-1) continuously for 10 days before I/R operation. The morphological changes and apoptosis of cardiomyocytes were observed by H&E staining, Transmission electron microscope and TUNEL staining respectively. The activities or contents of catalase (CAT), superoxide dismutase (SOD), malondialdehyde (MDA) and glutathione (GSH) in plasma were detected. Apoptosis related factors were also measured by RT-PCR and western blot. In order to discover the underlying mechanism of NP on I/R, we performed proteomic analysis using two-dimensional gel electrophoresis (2D-DIGE) and matrix assisted laser desorption/ionization time of flight mass spectrometry (MALDI-TOF/MS) to describe differential proteins expression. Potential target protein resulted from 2D-DIGE coupled to MALDI-TOF/MS analysis were further confirmed by immunohistochemical staining, RT-PCR, and western blot.
    RESULTS: We isolated and identified 14 compounds, of which 7 compounds belong to triterpenes. Rats pretreated with NP showed a significant increase on the activities of GSH, SOD and CAT, and remarkable decrease on the content of MDA. NP significantly inhibited the apoptosis of cardiomyocyte and decreased the expression of Cyt C and cleaved-caspase-3. Proteomic analysis revealed that alpha B-crystallin (CryAB) might participate in the NP protective effect against I/R. NP enhanced the level of pCryAB Ser59, whereas the expression of CryAB was decreased.
    CONCLUSION: NP was showed to alleviate I/R injury and inhibit myocardial apoptosis, which might be associated with reduction on oxidative stress and apoptosis. CryAB as a possible target involved in the NP protective effect. This study supplied valuable information to develop novel cardioprotective agents from NP extract.
    Keywords:  2D-DIGE; Apoptosis; CAT; Cardiomyocyte; CryAB; GSH; I/R; Ischemia/reperfusion; MALDI-TOF/MS; MDA; NP; Oxidative stress; Potentilla anserina L; SOD; alpha B-crystallin; catalase; glutathione; ischemia/reperfusion; malondialdehyde; matrix assisted laser desorption/ionization time of flight mass spectrometry; n-butanol extract of Potentilla anserina L; superoxide dismutase; two-dimensional gel electrophoresis; αB-crystallin
    DOI:  https://doi.org/10.1016/j.phymed.2018.10.024
  64. Chem Res Toxicol. 2019 Apr 03.
      Flutolanil is a broad-spectrum amide fungicide that is widely used to prevent fungal pathogens in agriculture. However, its usage may have a potential environmental impact on organisms. So far, few literatures have investigated the chronic toxicity of flutolanil at concentrations relevant to environmental conditions in the non-target aquatic organisms. This study was aimed at evaluating whether the long-term exposure of flutolanil affects oxidative stress, immune response, and apoptosis in the liver of zebrafish (Danio rerio). The results showed that the activity of catalase (CAT) was significantly decreased in liver at all flutolanil-treated groups. Interestingly, the malondialdehyde (MDA) contents were remarkably increased following the flutolanil exposure. Deoxyribonucleic acid (DNA) damage was increased with a concentration-dependent manner. The transcription level of genes involved in apoptosis and immune system were significantly altered following flutolanil chronic exposure in zebrafish liver. Furthermore, the caspase-3 enzyme activity was significantly increased. Taken together, this study demonstrated that the resulting effects on oxidative stress, immune toxicity, and apoptosis may be responsible for the pathological alterations in zebrafish liver after flutolanil exposure at concentrations relevant to environmental conditions, advancing the knowledge of pesticide environmental risk assessment.
    DOI:  https://doi.org/10.1021/acs.chemrestox.8b00300
  65. Antiviral Res. 2019 Mar 27. pii: S0166-3542(18)30571-0. [Epub ahead of print]166 42-55
      Liver injury is one of the hallmark features of severe dengue virus (DENV) infection since DENV can replicate in the liver and induce hepatocytes to undergo apoptosis. N-acetyl cysteine (NAC), which is a clinically-used drug for treating acetaminophen toxicity, was found to benefit patients with DENV-induced liver injury; however, its mechanism of action remains unclear. Accordingly, our aim was to repurpose NAC in the preclinical studies to investigate its mechanism of action. Time of addition experiments in HepG2 cells elucidated effectiveness of NAC to reduce infectious virion at pre-, during- and post infection. In DENV-infected mice, NAC improved DENV-associated clinical manifestations, including leucopenia and thrombocytopenia, and reduced liver injury and hepatocyte apoptosis. Interestingly, we discovered that NAC significantly reduced DENV production in HepG2 cells and in liver of DENV-infected mice by induction of antiviral responses via interferon signaling. NAC treatment in DENV-infected mice helped to maintain antioxidant enzymes and redox balance in the liver. Therefore, NAC reduces DENV production and oxidative damage to ameliorate DENV-induced liver injury. Taken together, these findings suggest the novel therapeutic potential of NAC in DENV-induced liver injury and recommend evaluating its efficacy and safety in humans with DENV-induced liver injury.
    Keywords:  Antiviral response; Dengue replication; Dengue virus; Liver injury; N-acetyl cysteine
    DOI:  https://doi.org/10.1016/j.antiviral.2019.03.011
  66. Cell Mol Biol (Noisy-le-grand). 2019 Mar 31. 65(3): 76-83
      The aim of this study is an investigation the protective effects of vitamin C (Vit C), vitamin E (Vit E), β-carotene, sodium selenate combination in indomethacin-induced gastric mucosal damage in rats. Rats were divided into 6 groups. Group I: Intact animals (control). Group II: Control animals receiving Vit C (100 mg/kg/day), Vit E (100 mg/kg/day), β-carotene (15 mg/kg/day) and sodium selenate (0.2 mg/kg/day) for 3 days. Group III: Animals receiving 25 mg/kg indomethacin. Group IV: Animals receiving Vit C, Vit E, β-carotene and sodium selenate (in same doses) for 3 days 2 h before the administration of indomethacin. Group V: Animals receiving ranitidine (150 mg/kg) for 3 days. Group VI: Animals receiving ranitidine for 3 days 2 h before to the administration of indomethacin (in same dose and time). The administration of indomethacin caused a decrease in the levels of glutathione, mucus, hexosamine and in the activities of glutathione-S-transferase, sodium-potassium ATPase, thromboplastic activity and an increase in the aspartate and alanine amino transferase, alkaline phosphatase, catalase, lactate dehydrogenase, myeloperoxidase activities and sialic acid, lipid peroxidation and protein carbonyl levels.  Stomach caspase-8 immun+ cell numbers showed a slight increase while caspase-9 immun+ cell numbers reduced in indomethacin given group compared to control animals. Our results findings suggest that the combination of Vit C, Vit E, β-carotene, sodium selenate and ranitidine has a protective effect on indomethacin-induced gastric mucosal injury of rats.
    Keywords:  Antioxidant; Apoptosis.; Indomethacin; Oxidative stress; Stomach
  67. Iran J Basic Med Sci. 2019 Jan;22(1): 99-105
      Objectives: Hydrogen sulfide (H2S) attenuates ischemia-reperfusion injury (IRI) in different organs. However, its mechanism of action in renal IRI remains unclear. The present study investigated the hypothesis that H2S attenuates renal IRI via the induction of heat shock proteins (HSPs).Materials and Methods: Adult Wistar rats were subjected to unilateral renal ischemia for 45 min followed by reperfusion for 6 hr. One group of rats underwent I/R without treatment, one group was administered 150 μmol/l sodium hydrosulfide (NaHS) prior to I/R, one group was injected with 100 mg/kg quercetin (an HSP inhibitor) intraperitoneally prior to I/R, and another group received quercetin prior to I/R and treatment with NaHS following I/R. Two other groups underwent a sham operation and one of them received 150 μmol/l NaHS following the sham operation whereas the other received no treatment. Renal function and histological changes were compared and relevant indices of oxidative stress, apoptosis, and inflammation were examined.
    Results: IRI increased serum creatinine and blood urea nitrogen concentrations, promoted lipid peroxidation by elevating malondialdehyde levels, suppressed superoxide dismutase activity, stimulated inflammation by inducing NF-kB, IL-2, and TLR-4 expression, and increased renal apoptosis. Levels of HSP 70, heme-oxygenase-1 (HO-1) and HSP 27 were increased following IRI and reversed following H2S treatment. H2S attenuated changes observed in pathology, lipid peroxidation, inflammation, and apoptosis following IRI. The administration of quercetin reversed all protective effects of H2S.
    Conclusion: The present study indicated that H2S protected renal tissue against IRI induced lipid peroxidation, inflammation, and apoptosis, which may be attributed to the upregulation of HSP 70, HO-1, and HSP 27.
    Keywords:  Heat shock protein 27; Heat shock protein 70; Heme oxygenase 1; Hydrogen sulfide; Ischemia-reperfusion injury; Rat; Renal
    DOI:  https://doi.org/10.22038/ijbms.2018.29706.7170
  68. Cancer Lett. 2019 Apr 01. pii: S0304-3835(19)30210-1. [Epub ahead of print]
      FLT3-ITD and FLT3-TKD are the most frequent mutations in acute myeloid leukemia (AML) with the former associated with a poor prognosis. Here we show that inhibition of the deubiquitinase USP9X by its inhibitor WP1130 or EOAI3402143 (G9) induces apoptosis preferentially in cells transformed by these mutant kinases, including FLT3-ITD-positive AML cell line MV4-11 and primary AML cells. Mechanistically, WP1130 induced aggresomal translocation of the mutant kinases, particularly FLT3-ITD in its activated and autophosphorylated conformation, to block the downstream signaling events, which was aggravated by knock down of USP9X. Moreover, USP9X physically associated with FLT3-ITD to inhibit its K63-linked polyubiquitination, while FLT3-ITD induced tyrosine phosphorylation and degradation of USP9X through the ubiquitin/proteasome pathway. WP1130 or G9 also induced oxidative stress to stimulate stress-related MAP kinase pathways and DNA damage responses to activate in cooperation with inhibition of FLT3-ITD signaling the intrinsic mitochondria-mediated apoptotic pathway, which was synergistically enhanced by BH3 mimetics and prevented by overexpression of Bcl-xL or Mcl-1. Thus, USP9X represents a promising target for novel therapies against therapy-resistant FLT3-ITD-positive AML.
    Keywords:  BH3 mimetics; Deubiquitinase; EOAI3402143; Mcl-1; WP1130
    DOI:  https://doi.org/10.1016/j.canlet.2019.03.046
  69. Cell Physiol Biochem. 2019 ;52(4): 742-757
      BACKGROUND/AIMS: The oxidative stress sensor transient receptor potential melastatin-2 (TRPM2) ion channel has recently gained attention in many types of cancer. The lung tissue is highly susceptible to oxidative stress-mediated injury and diseases; therefore, we aimed to determine whether TRPM2 plays an essential role in protecting lung cancer cells from oxidative damage while promoting cancer cell survival and metastasis.METHODS: We used two non-small cell lung (NSCLC) cell lines A549 and H1299 as a lung cancer model. We investigated the functional expression of TRPM2 using electrophysiology, qRT-PCR and Western blots. CFSE and flow cytometry were used to study TRPM2 role in proliferation, cell cycle and apoptosis. Gap closure chambers and Three-Tiered Chemotaxis Chamber were used to study the role of TRPM2 in metastasis. SCID mice were used to study the role of TRPM2 in lung tumor growth and metastasis.
    RESULTS: we demonstrate that TRPM2 is functionally expressed in NSCLC cells and that its downregulation significantly inhibits cell proliferation and promotes apoptosis. These results were concomitant with an induction in DNA damage and G2/M cell cycle arrest. TRPM2 silencing inhibits also lung cancer cells invasion ability and alters EMT processes. Mechanistically, TRPM2 downregulation causes an increase in the intracellular levels of reactive oxygen (ROS) and nitrogen (RNS) species, which in turn causes DNA damage and JNK activation leading to G2/M arrest, and an ultimate cell death. Finally, TRPM2 downregulation suppresses the growth of human lung tumour xenograft in SCID mice and TRPM2 depleted tumours exhibited a significant reduction in the mRNA expression level of EMT markers compared to the control tumors.
    CONCLUSION: Our data provide new insights on the functional expression of TRPM2 in lung cancer, its essential role in tumour growth and metastasis through the control of JNK signaling pathway, and that TRPM2 could be exploited for targeted lung cancer therapies.
    Keywords:  JNK; Non-small cell lung cancer; ROS; TRPM2
    DOI:  https://doi.org/10.33594/000000052
  70. Fish Shellfish Immunol. 2019 Mar 29. pii: S1050-4648(19)30211-6. [Epub ahead of print]89 91-97
      Peroxiredoxins (Prxs) are a widespread and greatly transcribed family of antioxidant proteins, which rapidly detoxify peroxynitrite, hydrogen peroxide and organic hydroperoxides. The Prxs family members also modulate various physiological functions, including cell growth, differentiation, embryonic development, immune response, apoptosis, lipid metabolism, and cellular homeostasis. In mammals, the physiological functions of Prxs have extensively been studied; however, the knowledge is scanty in their counterpart, aquatic invertebrates. In recent years, substantial progress has been made in our knowledge of Prxs physiological functions in aquatic invertebrates, which has raised interest in defining the contribution of immune responses and removal of reactive oxygen species. In this review, we describe the recent knowledge on the Prxs physiological function in immune responses and DNA protection activity in aquatic invertebrates.
    Keywords:  Antioxidants; Innate immunity; Microbial infection; Oxidative stress; Peroxiredoxins
    DOI:  https://doi.org/10.1016/j.fsi.2019.03.062
  71. Pharmacol Res. 2019 Mar 29. pii: S1043-6618(19)30048-9. [Epub ahead of print]
      In recent years, the Wnt/β-catenin signaling has gained tremendous attention due to its ability to modulate a number of diseases including diabetic nephropathy. Studies have shown that there is decrease in the secretion of Wnt proteins including Wnt4, 5a and Wnt 6 during high glucose concentration or diabetic conditions, which leads to decreased translocation of β-catenin to nucleus. The down-regulation of Wnt/β-catenin signaling leads to detrimental effects on kidney including increased apoptosis of mesangial cells and increased deposition of fibrous tissue in mesangium. The pharmacological modulators such as spironolactone, NO donor and antioxidant are shown to produce beneficial effects in diabetic nephropathy by up regulating the expression of Wnt proteins and activation of diabetes-induced suppressed Wnt/β-catenin signaling. On the other hand, it is documented that diabetes leads to overactivation of Wnt1/β-catenin signaling, which promotes podocyte injury, induce epithelial-mesenchymal transition of podocytes along with renal injury and fibrosis. Accordingly, different interventions aimed to suppress overactivated Wnt/β-catenin signaling are reported to improve the condition and symptoms associated with diabetic nephropathy. The present review discusses the dual role of Wnt/beta-catenin signaling in the pathogenesis of diabetic nephropathy.
    Keywords:  Diabetes; Wnt; beta-catenin; kidney; nephropathy; oxidative stress
    DOI:  https://doi.org/10.1016/j.phrs.2019.03.022