bims-aporos Biomed News
on Apoptosis and reactive oxygen species
Issue of 2019‒05‒26
eighty-two papers selected by
Gavin McStay
Staffordshire University


  1. PLoS One. 2019 ;14(5): e0216711
      Large-scale breeding environments often lead to oxidative stress. Macrophages play an important role in the immune system and are vulnerable to reactive oxygen species (ROS), which result in macrophage death. Curcumin is the main active component of turmeric and exerts antioxidant effects. Here, we measured the activity of some antioxidant enzymes and chose the Nrf2-Keap1 signaling pathway to study the protective effects of curcumin on macrophages under oxidative stress in vitro. We used RAW264.7 cells as a research model, and oxidative damage was induced by hydrogen peroxide (H2O2). Cell viability was measured by an MTT assay. Flow cytometry was used to measure cellular ROS and apoptosis. The effect of curcumin on Nrf2-Keap1 signaling pathway-related genes was analyzed by qRT-PCR. Furthermore, the translocation of Nrf2 protein was also investigated by Western blot analysis of total and nuclear proteins. All curcumin-treated groups exhibited increased activity of catalase (CAT), superoxide dismutase (SOD) and glutathione peroxidase (GSH-PX). Low- and middle-dose curcumin decreased malondialdehyde (MDA) and ROS levels, but high-dose curcumin increased MDA and ROS production. We found that low-dose curcumin protected cells from apoptosis, while apoptosis in the middle- and high-dose curcumin-treated groups were stagnant in the early stage. Furthermore, middle-dose curcumin upregulated Nrf2 expression after H2O2 treatment for 4 h. Low- and middle-dose curcumin could activate Nrf2 and promote it to migrate into nuclei. The translocation of Nrf2 to the nucleus to upregulate the expression of haemoxygenase-1 (HO-1) was promoted in the low- and middle-dose curcumin-treated groups. The middle-dose curcumin-treated group also exhibited enhanced expression of glutamate-cysteine ligase, a modifier subunit (GLCM), but inhibited transcription of glutamate-cysteine ligase, a catalytic subunit (GCLC). Curcumin resisted oxidants by increasing the activity of antioxidant enzymes and activating the Nrf2-Keap1 pathway, which could potentially promote cell survival.
    DOI:  https://doi.org/10.1371/journal.pone.0216711
  2. Biomed Pharmacother. 2019 May 16. pii: S0753-3322(19)30589-X. [Epub ahead of print]116 108970
      Polymyxin E (PME) plays an important role in fighting against Gram-negative bacterial infections; however, it causes nephrotoxicity, which limits its clinical use. The aim of this study was to investigate the protective effects of a plant extract Panax notoginseng saponins (PNS) on PME-induced nephrotoxicity in mice. In vivo studies showed that PNS significantly reduced blood urea nitrogen (BUN), serum creatinine (CRE) and number of apoptotic cells in kidney, as well as renal histopathological damage which increased in the presence of PME, and suppressed PME-induced oxidative stress in kidney, as shown by the up-regulation of superoxide dismutase (SOD) and the down-regulation of malondialdehyde (MDA) levels. Furthermore, PNS inhibited the expression of Bax, while increased the expression of Bcl-2 compared to the PME-treated group. In vitro studies showed that PNS decreased intracellular reactive oxygen species (ROS) and MDA levels, increased glutathione (GSH) levels, and enhanced the activity of SOD and glutathione peroxidase (GSH-Px) in murine renal tubular epithelial cells (TCMK-1 cells). In addition, PNS enhanced cell viability and the expression of Bcl-2, restored the mitochondrial membrane potential, inhibited the expression of Bax, inhibited the activity of caspase-3 and caspase-9, and reduce apoptotic rate in PME-treated TCMK-1 cells. PNS could reduce PME-induced nephrotoxicity. The protective effects could result from inhibition of oxidative stress, and prevention of cell apoptosis via the mitochondrial pathway. These findings highlight the potential of PNS as a safe adjunct for ameliorating the nephrotoxicity.
    Keywords:  Apoptosis; Nephrotoxicity; Oxidative stress; Panax notoginseng saponins (PNS); Polymyxin E (PME)
    DOI:  https://doi.org/10.1016/j.biopha.2019.108970
  3. Environ Toxicol Pharmacol. 2019 May 02. pii: S1382-6689(19)30059-6. [Epub ahead of print]
      The stress-induced hormone corticosterone initiates oxidative stress and inflammatory responses, culminating in cell apoptosis and neurological changes. We assessed the effects of d-Limonene on a PC12 cellular model of corticosterone-induced neurotoxicity, and whether these effects involved the AMP-activated protein kinase (AMPKα) pathway. PC12 cells were treated with corticosterone with or without d-limonene for 24 h. Western blots were performed to measure activation of AMPK pathway members [Silent mating type information regulation 2 homolog-1 (SIRT1), AMPKα, and nuclear factor (NFκB)], reactive oxygen species, inflammatory cytokines, and markers of apoptosis. Terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling (TUNEL) was used to measure cell death after treatment. d-Limonene reversed the effects of corticosterone on PC12 cells: it decreased the levels of malondialdehyde (MDA) and nitric oxide (NO), activities of NADPH oxidase (p67-phox and p47-phox), expression of pro-inflammatory markers [inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), interleukin 6 (IL-6), interleukin 1β (IL-1β), and tumor necrosis factor α (TNF-α)], and expression of pro-apoptotic proteins [Bcl2 associated with X protein (Bax) and cleaved caspase-3)]. d-Limonene also increased levels of the antioxidant enzymes superoxide dismutase 1 (SOD1) and heme oxygenase 1 (HO-1) and the anti-apoptotic protein Bcl-2 while decreasing the number of TUNEL-positive cells. d-limonene significantly activated AMPKα and suppressed NF-κB nuclear translocation through up-regulation of SIRT1. Addition of compound C, an AMPK inhibitor, severely weakened these neuroprotective effects of d-limonene. d-Limonene has a neuroprotective effect on corticosterone-induced PC12 cell injury induced by activating the AMPKα signaling pathway, and thereby inhibiting reactive oxygen species and inflammatory factors. These data suggest that d-limonene might protect against neuronal death to improve depressive symptoms.
    Keywords:  AMP-activated protein kinase; D-Limonene; Neuroprotection; PC12 cells
    DOI:  https://doi.org/10.1016/j.etap.2019.05.001
  4. Mol Med Rep. 2019 May 22.
      Puerarin, a major bioactive constituent of the Radix puerariae, can ameliorate myocardial ischemia/reperfusion (I/R) injury. Emerging evidence supports that microRNA (miR)‑21 functions as a protective factor against I/R and/or hypoxia‑reperfusion (H/R)‑induced myocardial injury. However, the role of miR‑21 in the cardioprotective effect of puerarin remains unclear. Therefore, the purpose of the present study was to demonstrate the involvement of miR‑21 in the cardioprotective mechanisms of puerarin using a cell model of I/R injury, generated by culturing rat H9c2 cardiomyocytes under H/R conditions. The results demonstrated that pre‑treatment with puerarin significantly increased cell viability, decreased lactate dehydrogenase activity and upregulated miR‑21 expression in H/R‑treated H9c2 cells. Transfection of an miR‑21 inhibitor led to an increase in H/R‑induced cytotoxicity and reversed the protective effects of puerarin. Additionally, miR‑21 inhibition attenuated the puerarin‑induced decrease in the rate of apoptosis, caspase‑3 activity and the expression of apoptosis regulator Bax, and increased apoptosis regulator Bcl‑2 expression, under H/R conditions. Furthermore, puerarin mitigated H/R‑induced oxidative stress as evidenced by the decrease in endogenous reactive oxygen species production, malondialdehyde content and NADPH oxidase 2 expression, and enhanced the antioxidative defense system as illustrated by the increase in superoxide dismutase activity, catalase and glutathione peroxidase levels. These effects were all eliminated by miR‑21 inhibitor transfection. Furthermore, the miR‑21 inhibitor exacerbated the H/R‑induced oxidative stress and attenuated the antioxidative defense system in H/R‑treated H9c2 cells. Taken together, the results suggested that miR‑21 mediated the cardioprotective effects of puerarin against myocardial H/R injury by inhibiting apoptosis and oxidative stress.
    DOI:  https://doi.org/10.3892/mmr.2019.10266
  5. Nanomaterials (Basel). 2019 May 22. pii: E787. [Epub ahead of print]9(5):
      Ovarian cancer incidence continues to increase at an alarming rate. Although various therapeutic approaches exist for ovarian cancer, they have limitations, including undesired side effects. Therefore, nanoparticle (NP)-mediated therapy may be a viable, biocompatible, and suitable alternative. To the best of our knowledge, no comprehensive analysis has been undertaken on the cytotoxicity and cellular pathways involved in ovarian cancer cells, particularly SKOV3 cells. Here, we investigated the effect of palladium NPs (PdNPs) and the molecular mechanisms and cellular pathways involved in ovarian cancer. We assayed cell viability, proliferation, cytotoxicity, oxidative stress, DNA damage, and apoptosis and performed an RNA-Seq analysis. The results showed that PdNPs elicited concentration-dependent decreases in cell viability and proliferation and induced increasing cytotoxicity at increasing concentrations, as determined by leakage of lactate dehydrogenase, increased levels of reactive oxygen species and malondialdehyde, and decreased levels of antioxidants like glutathione and superoxide dismutase. Furthermore, our study revealed that PdNPs induce mitochondrial dysfunction by altering mitochondrial membrane potential, reducing adenosine triphosphate levels, inducing DNA damage, and activating caspase 3, all of which significantly induced apoptosis in SKOV3 cells following PdNPs treatment. Gene ontology (GO) term analysis of PdNPs-exposed SKOV3 cells showed various dysregulated pathways, particularly nucleosome assembly, telomere organization, and rDNA chromatin silencing. When genes downregulated by PdNPs were applied to GO term enrichment analysis, nucleosome assembly was the top-ranked biological pathway. We also provide evidence for an association between PdNPs exposure and multiple layers of epigenetic transcriptional control and establish a molecular basis for NP-mediated apoptosis. These findings provide a foundation, potential targets, and novel insights into the mechanism underlying toxicity and pathways in SKOV3 cells, and open new avenues to identify novel targets for ovarian cancer treatment.
    Keywords:  RNA-Seq analysis; cytotoxicity; ovarian cancer; oxidative stress; palladium nanoparticles
    DOI:  https://doi.org/10.3390/nano9050787
  6. Cancer Cell Int. 2019 ;19 130
      Background: Bursopentin (BP5) is a multifunctional pentapeptide found in the chicken bursa of Fabricius. Recent study indicated that BP5 significantly stimulates expression of p53 protein in colon cancer HCT116 cells. However, the effects and underlying mechanisms of BP5 on HCT116 cell proliferation remain largely unclear.Methods: Analyses of cell viability, cell cycle arrest as well as apoptosis were performed to study the actions of BP5 on HCT116 cells. Western blot analyse was assayed to measure the cell cycle-related and apoptosis-related proteins. Specific siRNAs targeting IRE1, ATF-6, and PERK were used for IRE1, ATF-6, and PERK knockdown, respectively. Cellular reactive oxygen species (ROS) were detected using a H2DCF-DA green fluorescence probe. Cytosolic free Ca2+ concentrations and mitochondrial membrane potential (ΔΨm) were measured using Fluo-3 AM and JC-1 stains, respectively.
    Results: BP5 possessed strong inhibitory effects on the cell growth and induced apoptosis in HCT116 cells. Mechanistically, BP5 arrested the cell cycle at G1 phase by increasing p53 and p21 expression and decreasing cyclin E1-CDK2 complex expression. BP5 treatment dramatically activated the endoplasmic reticulum (ER) stress-mediated apoptotic pathway, as revealed by the significantly enhanced expression of unfolded protein response (UPR) sensors (IRE1α, ATF6, PERK) as well as downstream signaling molecules (XBP-1s, eIF2α, ATF4 and CHOP), and by the significantly altered the BP5-induced phenotypic changes in IRE1, ATF6, and PERK knockdown cells. Additionally, BP5-induced ER stress was accompanied by the accumulation of cytosolic free Ca2+ and intracellular ROS. Furthermore, BP5 treatment resulted in the increase of Bax expression, the decrease of Bcl-2 expression and the reduction of ΔΨm, subsequently causing a release of cytochrome c from the mitochondria into the cytoplasm and finally enhancing the activities of caspase-9 and -3. In addition, z-VAD-fmk, a pan-caspase inhibitor, markedly rescued BP5-induced cell viability reduction and reduced BP5-induced apoptosis.
    Conclusions: Our present results suggest that BP5 has an anticancer capacity to arrest cell cycle at G1 phase and to trigger ER stress/mitochondria-mediated caspase-dependent apoptosis in HCT116 cells. Therefore, our findings provide insight into further investigations of the anticancer activities of BP5.
    Keywords:  Apoptosis; Endoplasmic reticulum stress; G1 cell cycle arrest; Mitochondrial pathway; Reactive oxygen species
    DOI:  https://doi.org/10.1186/s12935-019-0849-3
  7. Free Radic Res. 2019 May 20. 1-241
      Paraquat (PQ) is a widely used agro-chemical in agriculture and highly toxic to humans. Although the mechanism of PQ poisoning is not clear, it has been well documented that reactive oxygen species (ROS) generation and apoptosis play pivotal roles. Alternatively, chlorogenic acid (CA) is a biologically active dietary polyphenol, playing several therapeutic roles. However, it is not known whether CA has protective effect on PQ-induced apoptosis. Here, we investigated the effect of CA in preventing PQ-induced apoptosis and explored the underlying mechanisms. A549 cells were pretreated with 100 µM CA for 24 h and then exposed to 160 µM PQ for 24 h. We found that CA was effective in preventing PQ-induced apoptotic features, including the release of cytochrome c from the mitochondria to cytoplasm, the cleavages of caspase 3 and caspase 9, and the increases in levels of Bcl-2-associated X protein (Bax) and intracellular calcium ions. CA alleviated ROS production and prevented the reduction of antioxidant capacity in cells exposed to PQ by increasing NF-E2-related factor 2 (Nrf2), superoxide dismutase 2 (SOD2) and glutathione levels. In addition, CA also attenuated PQ-induced alterations of mitochondrial structure and function (such as the decreases in membrane potential and adenosine triphosphate level), and the impaired autophagic flux was improved by CA. Down-regulation of sirtuin 1 (Sirt1) by short hairpin RNA reversed the protective effects of CA. Thus, CA may be viewed as a potential drug to treat PQ-induced lung epithelial cell apoptosis and other disorders with similar pathologic mechanisms.
    Keywords:  Apoptosis; chlorogenic acid; mitochondria; paraquat; reactive oxygen species
    DOI:  https://doi.org/10.1080/10715762.2019.1621308
  8. Acta Diabetol. 2019 May 21.
      AIMS: Hyperglycemia induces endothelial cell apoptosis and blood vessel damage, while diallyl trisulfide (DATS) has shown cardiovascular protection in animal models and humans. The aim of this study was to investigate the effects of DATS on inhibition of high glucose-induced endothelial cell apoptosis and the underlying molecular events.METHODS: Human umbilical vein endothelial cells (HUVECs) were incubated with DATS (100 μM) for 30 min and then cultured in high-glucose medium (HG, 33 mM) for 24 h for assessment of apoptosis, glutathione (GSH), reactive oxygen species (ROS), superoxide dismutase (SOD), and gene expression using the terminal deoxyuridine triphosphate nick end labeling (TUNEL), flow cytometry, caspase-3 activity, ROS, SOD, and western blot assays as well as JC-1 and MitoTracker Red staining, respectively.
    RESULTS: DATS treatment significantly inhibited high glucose-induced HUVEC apoptosis by blockage of intracellular and mitochondrial ROS generation, maintenance of the mitochondrial membrane potential, and suppression of high glucose-induced dynamin-related protein 1 (Drp1) expression. Furthermore, DATS blockage of high glucose-induced mitochondrial fission and apoptosis was through adenosine monophosphate-activated protein kinase (AMPK) activation-inhibited Drp1 expression in HUVECs.
    CONCLUSIONS: DATS demonstrated the ability to inhibit high glucose-induced HUVEC apoptosis via suppression of Drp1-mediated mitochondrial fission in an AMPK-dependent fashion.
    Keywords:  Apoptosis; Diallyl trisulfide; Drp1; Endothelial cells; High glucose/hyperglycemia; Mitochondrial fission
    DOI:  https://doi.org/10.1007/s00592-019-01366-x
  9. Food Res Int. 2019 Jul;pii: S0963-9969(19)30200-5. [Epub ahead of print]121 336-347
      The effect of pulsed electric field (PEF) treatment on the intracellular antioxidant and apoptotic activity of the peptide Lys-Asp-His-Cys-His (KDHCH) was examined using model HepG2 cells. First, PEF treatment conditions specific for the antioxidant peptide were optimized, and it was found that PEF treatment could enhance DPPH, ABTS and hydroxyl radical scavenging capacity of KDHCH. Second, KDHCH subjected to PEF treatment at 1800 Hz and 15 kV/cm was investigated using various intracellular antioxidant assays. PEF treatment decreased the EC50 value and increased the protective ability of oxidative stress inhibition and reactive oxygen species (ROS) scavenging activity of KDHCH. Furthermore, catalase (CAT), superoxide dismutase (SOD), glutathione peroxidase (GSH-PX) and glutathione reductase (GR) activities of KDHCH-pre-treated HepG2 cells increased significantly compared with those of the H2O2 damaged group, whereas lactate dehydrogenase (LDH) and malonaldehyde (MDA) content were decreased. PEF-treated KDHCH exhibited an increased capacity to maintain the stability of mitochondrial membrane potential (MMP) and reduced the level of caspase-3. These results indicate that PEF treatment can enhance the intracellular antioxidant activity of KDHCH, which can inhibit the effect of H2O2 oxidation on HepG2 cells by inhibiting the accumulation of intracellular ROS, regulating antioxidant related enzymes, and blocking the apoptotic mitochondrial pathways activated by ROS.
    Keywords:  Antioxidant peptide; Apoptosis; HepG2 cells; Intracellular antioxidant activity; Pulsed electric field
    DOI:  https://doi.org/10.1016/j.foodres.2019.03.049
  10. Int Immunopharmacol. 2019 May 17. pii: S1567-5769(19)30457-6. [Epub ahead of print]73 225-235
      Intestinal ischemia/reperfusion (I/R) injury is a serious clinical event that may induce intestinal mucosal injury, whose major underlying mechanisms include reactive oxygen species (ROS) generation, release of inflammatory mediators and induction of apoptosis. Irisin is considered an agent with potent protection against many pathological injures. The aim of this study was to investigate the protective effect of irisin pretreatment on intestinal injury and explore its underlying mechanisms in a mouse model of intestinal I/R injury as well as a cell model (IEC-6 cell) of hypoxia/reoxygenation (H/R). The results showed that irisin pretreatment ameliorated I/R and H/R-induced injury in vivo and in vitro. In addition, irisin reduced the levels of tumor necrosis factor (TNF)-α, interleukin(IL)-1β and interleukin(IL)-6 in the intestine. Compared with the I/R group, irisin pretreatment effectively reduced malondialdehyde (MDA) and myeloperoxidase (MPO) levels, but increased superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) activities in the intestine, and significantly reduced oxidative stress. Furthermore, irisin pretreatment downregulated Bax and cleaved Caspase-3 at the protein level, and increased Bcl-2 protein amounts, significantly reducing apoptosis in the intestine of I/R mice. Moreover, both in vivo and in vitro results showed that irisin pretreatment significantly upregulated nuclear factor (erythroid-derived 2)-like 2 (Nrf2) protein. Meanwhile, Nrf2 siRNA treatment partially abrogated the protective effects of irisin pretreatment on H/R induced cellular damage, inflammatory response, oxidative stress, and apoptosis in IEC-6 cells. These findings suggest that irisin pretreatment improves I/R-induced intestinal inflammatory response, reduces oxidative stress and inhibits apoptosis, which could be, at least partially, associated with Nrf2 pathway activation.
    Keywords:  Apoptosis; Inflammation; Intestinal injury; Irisin; Nrf2; Oxidative stress
    DOI:  https://doi.org/10.1016/j.intimp.2019.05.011
  11. Am J Transl Res. 2019 ;11(4): 2168-2180
      The combination of tetramethylpyrazine phosphate (TMPP) and borneol (BO) protects against cerebral ischemia. However, the mechanism for their synergistic effect is unclear. In this study, an oxygen-glucose deprivation (OGD) injured brain model was induced in microvascular endothelium cells (BMECs). TMPP and BO concentrations were optimized according to an MTT assay. Cells were divided into five groups: control, model, TMPP, BO, and TMPP+BO. Subsequently, oxidative stress was evaluated based on the levels of superoxide dismutase (SOD), malondialdehyde (MDA), catalase (CAT), glutathione peroxidase (GSH-Px), and reactive oxygen species (ROS). Intracellular calcium ([Ca2+]i) was detected using a laser confocal microscope. Cellular apoptosis was examined via Hoechst 33342 staining, flow cytometry, and expression of p53, B-cell lymphoma 2 (BCL-2), BCL-2-like protein 4 (BAX), and caspase-3 mRNA. Angiogenesis was evaluated based on expression of basic fibroblast growth factor (bFGF), vascular endothelial growth factor (VEGF), fibroblast growth factor receptor 1 (FGFR1), Vascular endothelial growth factor receptor 1 (VEGFR1), and VEGFR2. Results showed that 5.0 μM TMPP and 0.5 μM BO were optimal. Monotherapy significantly enhanced CAT, BCL-2, and VEGF, and also reduced [Ca2+]i, apoptosis, and BAX. TMPP increased SOD, GSH-Px, and bFGF, and reduced MDA, ROS, p53, and caspase-3 levels. BO reduced VEGFR1 expression. TMPP+BO combination exhibited synergistic effects in decreasing apoptosis, and modulating expression of BCL-2, BAX, and VEGFR1. These results indicate that protection of OGD-injured BMECs by TMPP+BO combination involves anti-oxidation, apoptosis inhibition, and angiogenesis. Moreover, their synergistic mechanism was mainly related to the regulation of apoptosis and angiogenesis.
    Keywords:  BMECs; Synergistic effect; angiogenesis; apoptosis; borneol; tetramethylpyrazine phosphate
  12. J Cell Mol Med. 2019 May 18.
      Hypertension contributes to the high cardiac morbidity and mortality. Although oxidative stress plays an essential role in hypertensive heart diseases, the mechanism remains elusive. Transgenic mice with cardiac overexpression of metallothionein, a heavy metal-binding scavenger, were challenged with NG -nitro-L-arginine methyl ester (L-NAME) for 14 days prior to measurement of myocardial contractile and intracellular Ca2+ anomalies as well as cell signalling mechanisms using Western blot and immunofluorescence analysis. L-NAME challenge elicited hypertension, macrophage infiltration, oxidative stress, inflammation and cardiac dysfunction manifested as increased proinflammatory macrophage marker F4/80, interleukin-1β (IL-1β), intracellular O 2 - production, LV end systolic and diastolic diameters as well as depressed fractional shortening. L-NAME treatment reduced mitochondrial membrane potential (MMP), impaired cardiomyocyte contractile and intracellular Ca2+ properties as evidenced by suppressed peak shortening, maximal velocity of shortening/relengthening, rise in intracellular Ca2+ , along with elevated baseline and peak intracellular Ca2+ . These unfavourable mechanical changes and decreased MMP (except blood pressure and macrophage infiltration) were alleviated by overexpression of metallothionein. Furthermore, the apoptosis markers including BAD, Bax, Caspase 9, Caspase 12 and cleaved Caspase 3 were up-regulated while the anti-apoptotic marker Bcl-2 was decreased by L-NAME treatment. Metallothionein transgene reversed L-NAME-induced changes in Bax, Bcl-2, BAD phosphorylation, Caspase 9, Caspase 12 and cleaved Caspase 3. Our results suggest that metallothionein protects against L-NAME-induced myocardial contractile anomalies in part through inhibition of apoptosis.
    Keywords:  L-NAME; apoptosis; heart; hypertension; metallothionein
    DOI:  https://doi.org/10.1111/jcmm.14375
  13. J Ethnopharmacol. 2019 May 20. pii: S0378-8741(18)30682-2. [Epub ahead of print] 111962
      ETHNOPHARMACOLOGICAL RELEVANCE: Spermacoce hispida (S.hispida), a potential medicinal plant has been traditionally used as an antibacterial, antieczemic, antihypertensive, antidiabetic and antihyperlipidemic agent. Although, this plant has been claimed to protect against oxidative injury and inflammatory conditions in recent studies, its cardioprotective effect and the active constituents responsible for its bioactivity is sparsely studied. Hence this work is undertaken to study the active biomolecule responsible for modulating the cardiomyocytes on hypoxic injury relevant to its ethanopharmacology.AIM OF THE STUDY: The current study is to isolate and characterize a bioactive molecule from S.hispida, which exhibits protection against hypoxic injury in an in vitro hypoxic model of cultured H9c2 cardiomyocytes.
    MATERIALS AND METHODS: The methanolic extract of S.hispida plant was fractionated with various solvents sequentially. The ethyl acetate fraction that was concentrated and chromatographed over silica gel column eluted 18 fractions, which yielded 5 compounds, which were characterized using spectral data. The isolated new compound was further tested for its protective effect against hypoxic injury, wherein cobalt chloride (CoCl2) was used to induce hypoxia in H9c2 cardiomyoblasts. To evaluate the protective effect of the isolated compound, the markers of oxidative stress, apoptosis, and cell death were checked by endogenous levels of antioxidants, [malondialdehyde (MDA), superoxide dismutase (SOD), reduced glutathione (GSH)], lactate dehydrogenase (LDH) activity and immunoblot (HIF-α, Bcl2, Bax, procaspase and cleaved caspase-3).
    RESULTS: Among the five compounds isolated and characterized from S. hispida methanolic extract, β-sitosterol, ursolic acid, quercetin and rutin were known phytochemicals, while the new isoflavone was identified as dalspinin-7-0-β-D-galactopyranoside (DBG). Among the isolated compounds, the antioxidant potential of DBG confirmed by DPPH free radical scavenging and ORAC assays was superior. CoCl2-induced hypoxic condition significantly decreased cell viability, SOD activity, GSH concentration and increased the level of MDA and LDH activity. Western blot studies revealed an upregulation of HIF-1α, Bax and caspase and down regulation of Bcl-2 expression. The oxidative abnormalities were ameliorated by DBG pretreatment, as deduced by the reduced CoCl2-induced cytotoxicity, MDA concentration, LDH activity and the expression of HIF-1α, Bax and caspase and the enhanced levels of SOD, GSH and Bcl2 expression in a dose-dependent manner.
    CONCLUSION: DBG protects H9c2 cells from CoCl2-induced hypoxic damage by mitigating oxidative stress and preserving cell viability. The overall findings highlight the protective action of DBG, a potential source of antioxidant of natural origin against hypoxic injury and may help in mitigating the progress of oxidative stress in cardiac cell death.
    Keywords:  Antioxidant; Dalspinin; Hypoxia; Protection; Spermacoce hispida
    DOI:  https://doi.org/10.1016/j.jep.2019.111962
  14. Biochim Biophys Acta Mol Basis Dis. 2019 May 20. pii: S0925-4439(19)30172-3. [Epub ahead of print]
      Acetaminophen (APAP)-induced hepatotoxicity is a major factor in liver failure and its toxicity is associated with the generation of reactive oxygen species (ROS), decreased levels of reduced glutathione (GSH) and overall oxidative stress. Mitochondrial NADP+-dependent isocitrate dehydrogenase (IDH2) was demonstrated as an essential enzyme for mitochondria to maintain their antioxidant system by generating NADPH, which is an essential reducing equivalent for GSH turnover in mitochondria. Here, we investigated the role of IDH2 in APAP-induced liver injury with IDH2 deficient (idh2-/-) mice. Hepatotoxicity was promoted through apoptotic cell death following APAP administration in IDH2 deficient hepatocytes compared to that in wild-type hepatocytes. Apoptosis was found to result from the induction of ER stress and mitochondrial dysfunction as shown by the blocking the effect of phenylbutyrate and Mdivi1, respectively. In addition, mito-TEMPO, a scavenger of mitochondrial ROS, was seen to ameliorate APAP-induced hepatotoxicity in idh2-/- mice. In conclusion, IDH2 deficiency leads to a fundamental shortage of GSH that increases susceptibility to ROS generation and oxidative stress. This leads to excessive mitochondrial dysfunction and ER stress induction in response to APAP administration. Our study provides further evidence that IDH2 has a protective role against APAP-induced liver injury and emphasizes the importance of the elaborate linkages and functions of the antioxidant system in liver health.
    Keywords:  Acetaminophen; ER stress; IDH2; Mitochondrial dysfunction; Reactive oxygen species
    DOI:  https://doi.org/10.1016/j.bbadis.2019.05.012
  15. Int J Mol Med. 2019 May 16.
      Oxidative stress serves a critical role in melanocyte death and is considered to be a major cause of vitiligo. The nuclear factor E2‑related factor 2 (Nrf2) signaling pathway has an important role in the antioxidative stress mechanisms of melanocytes. Glycyrrhizin (GR) is a derivative of herbal medicines used to treat hepatitis and allergic disease due to its antiviral and anti‑allergy effects. GR also activates Nrf2 and induces the expression of heme oxygenase (HO)‑1 in macrophages. Whether GR can protect human melanocytes from oxidative stress remains unknown. The present study investigated the potential protective effects of GR against oxidative stress in human melanocytes and the mechanisms involved. Following exposure to 0.5 mM hydrogen peroxide (H2O2), human primary melanocytes were treated with 1 mM GR. Cell viability was determined using a Cell Counting Kit‑8 assay, and apoptosis was evaluated by flow cytometry. GR treatment significantly improved cell viability, reduced the apoptotic rate of melanocytes and reduced the level of reactive oxygen species in human melanocytes. Furthermore, GR induced the nuclear translocation of Nrf2 and induced the expression of HO‑1 in melanocytes. The knockdown of Nrf2 by small interfering RNA or the inhibition of HO‑1 by ZnPP reversed the protective effect of GR on melanocytes against H2O2‑induced cytotoxicity and apoptosis. These data demonstrate that GR protects human melanocytes from H2O2‑induced oxidative damage via the Nrf2‑dependent induction of HO‑1, providing evidence for the application of GR in the treatment of vitiligo.
    DOI:  https://doi.org/10.3892/ijmm.2019.4200
  16. Chemosphere. 2019 Apr 15. pii: S0045-6535(19)30752-0. [Epub ahead of print]229 618-630
      Bisphenol-A (BPA) is a representative exogenous endocrine disruptor, which is extensively composed in plastic products. Due to the capability of passing through the blood-brain barrier, evidence has linked BPA exposure with multiple neuropsychological dysfunctions, neurobehavioral disorders and neurodegenerative diseases. However, the underlying mechanism by which BPA induces neurodegeneration still remains unclear. Our study used human embryonic stem cells-derived human cortical neurons (hCNs) as a cellular model to investigate the adverse neurotoxic effects of BPA. hCNs were treated with 0, 0.1, 1 and 10 μM BPA for 14 days. Impacts of BPA exposure on cell morphology, cell viability and neural marker (MAP2) were measured for evaluating the neurodegeneration. The intracellular calcium homeostasis, reactive oxygen species (ROS) generation and organelle functions were also taken into consideration. Results revealed that chronic exposure of BPA damaged the neural morphology, induced neuronal apoptosis and decreased MAP2 expression at the level of both transcription and translation. The intracellular calcium levels were elevated in hCNs after BPA exposure through NMDARs-nNOS-PSD-95 mediating. Meanwhile, BPA led to oxidative stress by raising the ROS generation and attenuating the antioxidant defense in hCNs. Furthermore, BPA triggered ER stress and increased cytochrome c release by impairing the mitochondrial function. Ultimately, BPA triggered the cell apoptosis by regulating Bcl-2 family and caspase-dependent signaling pathway. Taken together, BPA exerted neurotoxic effects on hCNs by eliciting apoptosis, which might due to the intracellular calcium homeostasis perturbation and cell organellar dysfunction.
    Keywords:  Apoptosis; Bisphenol-A; Calcium homeostasis perturbation; Cell organelles dysfunctions; Neurodegeneration; hESC-derived cortical neurons
    DOI:  https://doi.org/10.1016/j.chemosphere.2019.04.099
  17. Arch Med Sci. 2019 May;15(3): 730-737
      Introduction: Blood glucose fluctuation is an important factor for the development of diabetic complications. Glucose fluctuation aggravated the renal injury in diabetic nephropathy. In the present study, our aim was to investigate the effects of blood glucose fluctuation on the glomerular mesangal cells and its related mechanism.Material and methods: Mesangial cells were divided into four groups: the normal glucose group (NG) cells were incubated in normal glucose conditions (5.6 mmol/l); the high glucose group (HG) cells were treated with 25 mmol/l; the glucose fluctuation (FG) group received 5.6 mmol/l and 25 mmol/l glucose repeated 3 times; the mannitol group (MG) received 5.6 mmol/l glucose plus 24.4 mmol/l mannitol as a control. Cell viability and apoptosis were detected, reactive oxygen species (ROS) level, superoxide dismutase (SOD) activity and malonaldehyde (MDA) levels were measured. Phosphorylated ser/thr protein kinase (P-AKT, phosphor-Ser473), phosphorylated glycogen synthase kinase-3β (P-GSK-3β, phosphor-Ser9) and cleaved cysteinyl aspartate-specific proteinase-3 (cleaved caspase-3) levels were assessed using western blot.
    Results: Data suggested that mesangial cells in the FG group show higher cell viability in 12 h, and lower cell viability from 48 h. The FG group showed cell apoptosis accompanied by a significant MDA level increase and SOD activity decrease in 48 h. More importantly, glucose fluctuation could aggravate oxidative stress in glomerular mesangial cells. Furthermore, the P-AKT level was lower, and increased P-GSK-3β and cleaved caspase-3 levels were higher in the FG group than in the HG group.
    Conclusions: Glucose fluctuation aggravates mesangial cell apoptosis, which may be partly induced by activating oxidative stress and inhibiting the AKT signaling pathway.
    Keywords:  P-AKT; apoptosis; glomerular mesangial cells; glucose fluctuation
    DOI:  https://doi.org/10.5114/aoms.2019.84739
  18. Prostate. 2019 May 18.
      INTRODUCTION: Both oxidative stress and inflammation play important roles in prostate cancer cell apoptosis or proliferation; however, the mechanisms underlying these processes remain unclear. Thus, we selected interleukin-8 (IL-8) as the bridge between inflammation and cancer cell oxidative stress-induced death and aimed to confirm its connection with mTOR and Glycogen synthase kinase-3 beta (GSK-3β).METHODS: We overexpressed GSK-3β and observed its effect on reactive oxygen species (ROS) and oxidative stress-induced cell death. IL-8 was then upregulated or downregulated to determine its impact on preventing cell damage due to GSK-3β-induced oxidative stress. In addition, we overexpressed or knocked down mTOR to confirm its role in this process. Real-time PCR, Western blotting, transcription, Cell Counting Kit 8 (CCK-8), and flow cytometry analyses were performed in addition to the use of other techniques.
    RESULTS: IL-8 promotes prostate cancer cell proliferation and decreases apoptosis, whereas GSK-3β activates the caspase-3 signaling pathway by increasing ROS and thereby induces oxidative stress-mediated cell death. In addition, mTOR can also decrease activation of the caspase-3 signaling pathway by inhibiting GSK-3 and thus decreasing ROS production. Moreover, the inhibitory effect of IL-8 on GSK-3β occurs through the regulation of mTOR.
    CONCLUSION: The results of this study highlight the importance of GSK-3β, which increases the production of ROS and thereby induces oxidative stress in tumor cells, whereas IL-8 and mTOR attenuate oxidative stress to protect prostate cancer cells through inhibition of GSK-3β.
    Keywords:  GSK-3β; IL-8; apoptosis; mTOR; oxidative stress
    DOI:  https://doi.org/10.1002/pros.23836
  19. J Trace Elem Med Biol. 2019 Jul;pii: S0946-672X(19)30003-3. [Epub ahead of print]54 27-35
      BACKGROUND: Titanium dioxide nanoparticles (TiO2 NPs) are widely used nanoparticles. Despite, several studies investigated the toxic effects of TiO2 NPs on HUVECs, the results are contradictory and the possible underlying mechanisms remain unclear.METHODS: In the present study, we conducted an in vitro study to re-evaluate the possible toxic effects of TiO2 NPs on HUVECs including cell viability, lipids peroxidation, intracellular signaling pathways and nitric oxide syntheses enzymes.
    RESULTS: Our results demonstrated that, TiO2 NPs were internalized to HUVECs and induce intracellular reactive oxygen species production and cell membrane oxidative damage at the higher concentration. TiO2 NPs induce IKKα/β and Akt phosphorylation and p38 dephosphorylation. After 24 h treatment, pro-inflammatory cytokines, adhesion molecules and chemokine upregulated significantly. TiO2 NPs have no significant effects on eNOS enzymatic activation and iNOS gene expression. At cellular level, apoptosis is the main process that occur in response to TiO2 NPs treatment. HUVECs pretreatment with N-acetyl-l-cysteine (NAC) ameliorate the toxic effects of TiO2 NPs that indicate the oxidative stress is essential in TiO2 NPs -induced toxicity. Total antioxidant capacity show a trend to increase in response to TiO2 NPs exposure.
    CONCLUSIONS: Taken together, this study confirmed the effects of TiO2 NPs on endothelial cells and proposed multiple underlying mechanisms including cell membrane oxidative damage and intracellular processes.
    Keywords:  Apoptosis; Cell membrane; Endothelial cell; Oxidative stress; Signaling pathways; Titanium dioxide nanoparticles
    DOI:  https://doi.org/10.1016/j.jtemb.2019.03.008
  20. Biochim Biophys Acta Mol Basis Dis. 2019 May 15. pii: S0925-4439(19)30169-3. [Epub ahead of print]
      Reactive oxygen species (ROS) are capable of inducing cell death or apoptosis. Recently, we demonstrated that lipid-ROS can mediate ferroptosis and activation of human platelets. Ferroptosis is an intracellular iron-mediated cell death, distinct from classical apoptosis and necrosis, which is mediated through the accumulation of ROS, lipid peroxides and depletion of cellular GSH. Lately, we demonstrated that hemoglobin degradation product hemin induces ferroptosis in platelets via ROS and lipid peroxidation. In this study, we demonstrate that hemin-induced ferroptosis in platelets is mediated through ROS-driven proteasome activity and inflammasome activation, which were mitigated by Melatonin (MLT). Although inflammasome activation is linked with pyroptosis, it is still not clear whether ferroptosis is associated with inflammasome activation. Our study for the first time demonstrates an association of platelet activation/ferroptosis with proteasome activity and inflammasome activation. Although, high-throughput screening has recognized ferrostatin-1 (Fer-1) and liproxstatin-1 (Lip-1) as potent ferroptosis inhibitors, having an endogenous antioxidant such as MLT as ferroptosis inhibitor is of high interest. MLT is a well-known chronobiotic hormone that regulates the circadian rhythms in vertebrates. It is also known to exhibit potent antioxidant and ROS quenching capabilities. MLT can regulate fundamental cellular functions by exhibiting cytoprotective, oncostatic, antiaging, anti-venom, and immunomodulatory activities. The ROS scavenging capacity of MLT is key for its cytoprotective and anti-apoptotic properties. Considering the anti-ferroptotic and anti-apoptotic potentials MLT, it could be a promising clinical application to treat hemolytic, thrombotic and thrombocytopenic conditions. Therefore, we propose MLT as a pharmacological and therapeutic agent to inhibit ferroptosis and platelet activation.
    Keywords:  Ferroptosis; Inflammasome; Melatonin; Platelets activation; Proteasome; ROS
    DOI:  https://doi.org/10.1016/j.bbadis.2019.05.009
  21. J Cell Physiol. 2019 May 22.
      Matrine, an alkaloid compound isolated from Sophora flavescens Ait, has been shown to exert cancer-killing actions in a variety of tumors; however, its anticancer mechanism in colorectal cancer (CRC) is not clear. The goal of our study was to characterize the anticancer effects and molecular mechanisms of matrine in SW480 CRC cells in vitro. Matrine treatment reduced mitochondrial metabolic function and ATP levels, repressed mitochondrial membrane potential, evoked mitochondrial reactive oxygen species accumulation, and promoted cyt-c-related mitochondrial apoptosis activation. In addition, we found that matrine treatment activated mitochondrial fission through upregulating mitochondrial elongation factor 1 (MIEF1); silencing of MIEF1 prevented matrine-mediated mitochondrial damage and reversed the decrease in SW480 cell viability. Moreover, matrine treatment affected MIEF1 expression via the large tumor suppressor-2 (LATS2)-Hippo axis, and LATS2 deficiency suppressed the anticancer actions exerted by matrine on SW480 cancer cells. In summary, we show for the first time that matrine inhibits SW480 cell survival by activating MIEF1-related mitochondrial division via the LATS2-Hippo pathway. These findings explain the anticancer mechanisms of matrine in CRC and also identify the LATS2-MIEF1 signaling pathway as an effective target for the treatment of CRC.
    Keywords:  LATS2-Hippo pathway; MIEF1; colorectal cancer cells; matrine; mitochondrial division
    DOI:  https://doi.org/10.1002/jcp.28838
  22. Environ Pollut. 2019 May 10. pii: S0269-7491(19)30073-9. [Epub ahead of print]251 689-698
      Deoxynivalenol (DON) frequently detected in a wide range of foods and feeds, inducing cytotoxicity to animals and humans. To investigate the underlying mechanism of DON-induced apoptosis and inflammation in porcine small intestinal epithelium, intestinal porcine epithelial cells (IPEC-J2 cells) were chosen as objects, and were treated by different concentrations (0 μg/mL, 0.2 μg/mL, 0.5 μg/mL, 1.0 μg/mL, 2.0 μg/mL, 4.0 μg/mL, 6.0 μg/mL) of DON. The results showed that DON induced cytotoxicity of IPEC-J2 cells in a dose-dependent manner, which is demonstrated by decreasing cell viability. Compared with the control group, DON treatment increased the expressions of genes associated with inflammation and apoptosis, such as interleukin-1 beta (IL-1β), cyclooxgenase-2 (COX-2), interleukin-6 (IL-6), tumour necrosis factor-alpha (TNF-α), caspase-3, caspase-8, caspase-9, and decreased the cell anti-oxidative status. Protein immunofluorescence showed increased expression of caspase-3, nuclear factor kB (NF-κB) and phosphorylated NF-κB in IPEC-J2 cells. DON increased the content of intracellular reactive oxygen species (ROS) of IPEC-J2 cells. N-Acetyl-L-cysteine (NAC), a commonly used antioxidant, blocked DON-induced ROS generation, alleviated the DON-induced apoptosis and inflammation. These results suggested that DON-induced impairment of IPEC-J2 cells is possibly due to increased ROS production, and expressions of genes and proteins associated with apoptosis and inflammation.
    Keywords:  Apoptosis; Deoxynivalenol; IPEC-J2 cells; Inflammation; ROS
    DOI:  https://doi.org/10.1016/j.envpol.2019.05.026
  23. Int J Mol Sci. 2019 May 17. pii: E2451. [Epub ahead of print]20(10):
      In aging and neurodegenerative diseases, loss of distinct type of neurons characterizes disease-specific pathological and clinical features, and mitochondria play a pivotal role in neuronal survival and death. Mitochondria are now considered as the organelle to modulate cellular signal pathways and functions, not only to produce energy and reactive oxygen species. Oxidative stress, deficit of neurotrophic factors, and multiple other factors impair mitochondrial function and induce cell death. Multi-functional plant polyphenols, major groups of phytochemicals, are proposed as one of most promising mitochondria-targeting medicine to preserve the activity and structure of mitochondria and neurons. Polyphenols can scavenge reactive oxygen and nitrogen species and activate redox-responsible transcription factors to regulate expression of genes, coding antioxidants, anti-apoptotic Bcl-2 protein family, and pro-survival neurotrophic factors. In mitochondria, polyphenols can directly regulate the mitochondrial apoptosis system either in preventing or promoting way. Polyphenols also modulate mitochondrial biogenesis, dynamics (fission and fusion), and autophagic degradation to keep the quality and number. This review presents the role of polyphenols in regulation of mitochondrial redox state, death signal system, and homeostasis. The dualistic redox properties of polyphenols are associated with controversial regulation of mitochondrial apoptosis system involved in the neuroprotective and anti-carcinogenic functions. Mitochondria-targeted phytochemical derivatives were synthesized based on the phenolic structure to develop a novel series of neuroprotective and anticancer compounds, which promote the bioavailability and effectiveness. Phytochemicals have shown the multiple beneficial effects in mitochondria, but further investigation is required for the clinical application.
    Keywords:  anticancer; apoptosis; fission/fusion; mitochondria; mitogenesis; mitophagy; neuroprotection; oxidative stress; phytochemicals
    DOI:  https://doi.org/10.3390/ijms20102451
  24. Front Mol Neurosci. 2019 ;12 105
      Intracerebral hemorrhage (ICH) is a devastating disease with high rates of mortality and morbidity. The aim of this study was to explore whether Sodium Benzoate (NaB) could reduce neural cell apoptosis and alleviate neurological deficits after ICH. To assess the therapeutic effects of NaB, first, we measured brain water content, neurobehavior, and blood-brain barrier (BBB) integrity at 24 h after ICH in different groups. Then western blot and immunofluorescence staining (IF) were applied to test the levels of different proteins. Transmission electron microscope (TEM) was used to observe ultra-structures within the cells in different groups. The results showed that levels of DJ-1, p-Akt and p-IκB kinase (IKK) increased after ICH and peaked at 24 h. Besides, NaB significantly upregulated DJ-1 in both cytoplasm and mitochondria, and also increased the levels of p-Akt, p-IKK and Bcl-2/Bax ratio, but decreased the levels of caspase-3 and caspase-9. Additionally, NaB decreased reactive oxygen species (ROS) while increased adenosine triphosphate (ATP), which then improving the neurological functions at 24 h and long-term (21 days) memory and spatial learning ability after ICH. However, the results mentioned above could be greatly reversed by MK2206 and rotenone. Therefore, we concluded that NaB could attenuate secondary brain injury via inhibiting neuronal apoptosis and reducing mitochondria-mediated oxidative stress via DJ-1/Akt/IKK/NFκB pathway.
    Keywords:  DJ-1; intracerebral hemorrhage; oxidative stress; secondary brain injury neuronal apoptosis; sodium benzoate
    DOI:  https://doi.org/10.3389/fnmol.2019.00105
  25. Lipids Health Dis. 2019 May 22. 18(1): 117
      BACKGROUND: Oxidized low-density lipoprotein (ox-LDL) is crucial in cardiac injury. Apolipoprotein-J (ApoJ) contributes to antiapoptotic effects in the heart. We aimed to evaluate the protective effects of ApoJ against ox-LDL cytotoxicity in Neonatal rat ventricular cells (NRVCs).METHODS AND RESULTS: NRVCs were damaged by exposure to ox-LDL, as shown by increased caspase-3/7 activity, enhanced caspase-3 expression, and decreased cell viability. ApoJ overexpression, using an adenovirus vector, significantly reduced ox-LDL-induced cell injury. ApoJ also prevented ox-LDL from augmenting reactive oxygen species (ROS) production, as demonstrated by elevated Nox2/gp91phox and P47 expression. Furthermore, ApoJ overexpression reduced CaMKIIδ expression elicited by ox-LDL in cultured NRVCs. Upregulating CaMKIIδ activity, mediated by ox-LDL, was significantly inhibited by ApoJ overexpression. A CaMKIIδ inhibitor, KN93, prevented ApoJ's protective effect against ox-LDL cytotoxicity. A ROS scavenger, Mn (III)meso-tetrakis (4-benzoic acid) porphyrin (Mn (III)TBAP), also attenuated CaMKIIδ's increased expression and activity, induced by ox-LDL, and showed similar results to ApoJ by attenuating ox-LDL-induced cell damage, as ApoJ did.
    CONCLUSIONS: ApoJ confers cytoprotection to NRVCs against ox-LDL cytotoxicity through the ROS-CaMKII pathways.
    Keywords:  Apolipoprotein-J; Apoptosis; Neonatal rat ventricular cells; Ox-LDL
    DOI:  https://doi.org/10.1186/s12944-019-1066-8
  26. Am J Transl Res. 2019 ;11(4): 2140-2154
      Oxidized low-density lipoprotein (ox-LDL)-induced endothelial-mesenchymal transition (EndMT), inflammation and apoptosis in endothelial cells play crucial roles in the progression of cardiovascular diseases including atherosclerosis. Vaccarin is a flavonoid glycoside from vaccariae semen associated with powerful cardiovascular protective effects. However, the effects of vaccarin on human umbilical vein endothelial cells (HUVEC) injury in response to ox-LDL remain unknown. Herein, we showed that treatment with vaccarin significantly suppressed ox-LDL-induced HUVEC inflammation, EndMT and apoptosis. Mechanistically, the HUVECs exposed to ox-LDL exhibited enlarged reactive oxygen species (ROS) production and p38 MAPK phosphorylation, which was counteracted by vaccarin. Importantly, ROS activator hydrogen peroxide (H2O2) and p38 MAPK activator anisomycin pretreatment prevent the protective effect of vaccarin on endothelial injury induced by ox-LDL. Our study suggested that vaccarin impeded ox-LDL-triggered HUVEC inflammation, EndMT and apoptosis via inhibition of ROS/p38 MAPK signaling pathway. Vaccarin may have a therapeutic effect on endothelial injury-related disorders.
    Keywords:  EndMT; HUVECs; apoptosis; inflammation; ox-LDL; vaccarin
  27. Biomed Pharmacother. 2019 May 16. pii: S0753-3322(18)33939-8. [Epub ahead of print]116 108524
      Glioma is the most common form of malignant brain cancer with high mortality rate in human. Therefore, finding effective therapeutic strategy and revealing the underlying molecular mechanism is necessary. Ampelopsin (Amp), an effective component of the traditional Chinese herb of Ampelopsis grossedentata, is reported to have important biological properties, including anti-inflammatory, anti-cancer, and anti-oxidant activity; however, its effects on human glioma are poorly understood. Here, the in vitro and in vivo study was performed to investigate the anti-glioma ability of Ampelopsin. Human glioma cell lines of U251 and A172 were treated with Ampelopsin (0, 25, 50, and 100 uM) for 24 h, followed by various analysis. And human glioma xenograft models were established by injecting U251, accompanied with administration of Ampelopsin at 50 and 100 mg/kg to confirm the anti-cancer role of Ampelopsin. We found that Ampelopsin could suppress the glioma cell proliferation by modulating G1 and S phase arrest. Incubation with Ampelopsin led to the activity of Caspase-8, Caspase-9, Caspase-3 and poly (ADP-ribose) polymerases (PARP), indicating that Ampelopsin induced apoptotic response via both intrinsic and extrinsic signaling pathways. Additionally, autophagy was also observed in Ampelopsin-treated cancer cells, which is evidenced by autophagosome formation and LC3B-II accumulation. Ampelopsin-caused cancer cell death was obviously regained by apoptosis inhibitors. Further, Ampelopsin activated c-Jun N-terminal protein kinase (JNK) expression and enhanced reactive oxygen species (ROS) generation. Suppressing JNK markedly ameliorated Ampelopsin-induced apoptosis and autophagy, and ROS scavenger exhibited similar results. In vivo, Ampelopsin inhibited tumor growth and progression in mouse xenograft models. In conclusion, our findings indicated that Ampelopsin led to G1 and S phase arrest, triggered apoptosis and autophagy through potentiating ROS generation and JNK activation in human glioma cells. Thus, Ampelopsin might be a promising candidate against human glioma.
    Keywords:  Ampelopsin; Apoptosis; Autophagy; Glioma; JNK; ROS
    DOI:  https://doi.org/10.1016/j.biopha.2018.12.136
  28. Mol Med Rep. 2019 May 22.
      Osteoblast apoptosis has been identified as an important event in the development of glucocorticoid (GC)‑induced osteoporosis and osteonecrosis of the femoral head. Crocin, a bioactive ingredient of saffron, has been demonstrated to induce antiapoptotic effects on numerous types of cell in vitro; however, the effects of crocin on the dexamethasone (Dex)‑induced apoptosis of osteoblasts remain unclear. In the present study, the protective effects of crocin during Dex‑induced apoptosis of MC3T3‑E1 osteoblasts, and the underlying mechanisms, were investigated. MTT and Annexin V‑FITC/PI flow cytometry assays were performed to evaluate the viability and apoptosis of cells, respectively. The mitochondrial transmembrane potential, reactive oxygen species (ROS), intracellular Ca2+ levels and apoptosis‑associated protein expression were assessed via flow cytometry, fluorescence microscopy and western blotting. It was demonstrated that crocin pretreatment inhibited Dex‑induced apoptosis of osteoblasts in a dose‑dependent manner. Crocin reversed Dex‑induced decreases in the mitochondrial transmembrane potential, and increases in ROS and intracellular Ca2+ levels. Furthermore, crocin upregulated the expression levels of B‑cell lymphoma-2 (Bcl‑2) and mitochondrial cytochrome c (Cyt C), and downregulated those of cleaved caspase‑9, cleaved caspase‑3, Bcl‑2‑associated X protein and cytoplasmic Cyt C. N‑acetylcysteine, a ROS inhibitor, and 1,2‑bis(2‑aminophenoxy)ethane‑N,N,N',N'‑tetraacetic acid, a calcium chelator, attenuated Dex‑induced osteoblast apoptosis, whereas H2O2 and ionomycin, a calcium ionophore that increases intracellular calcium levels, reversed the antiapoptotic effects of crocin on Dex‑treated osteoblasts. These results indicated that crocin may protect osteoblasts from Dex‑induced apoptosis by inhibiting the ROS/Ca2+‑mediated mitochondrial pathway, thus suggesting that crocin has potential value as a treatment for GC‑induced bone diseases.
    DOI:  https://doi.org/10.3892/mmr.2019.10267
  29. J Biomed Sci. 2019 May 22. 26(1): 40
      BACKGROUND: Oxidative stress is a major factor in retinal pigment epithelium (RPE) cells injury that contributes to age-related macular degeneration (AMD). NaIO3 is an oxidative toxic agent and its selective RPE cell damage makes it as a reproducible model of AMD. Although NaIO3 is an oxidative stress inducer, the roles of ROS in NaIO3-elicited signaling pathways and cell viability have not been elucidated, and the effect of NaIO3 on autophagy in RPE cells remains elusive.METHODS: In human ARPE-19 cells, we used Annexin V/PI staining to determine cell viability, immunoblotting to determine protein expression and signaling cascades, confocal microscopy to determine mitochondrial dynamics and mitophagy, and Seahorse analysis to determine mitochondrial oxidative phosphorylation.
    RESULTS: We found that NaIO3 can dramatically induce cytosolic but not mitochondrial ROS production. NaIO3 can also activate ERK, p38, JNK and Akt, increase LC3II expression, induce Drp-1 phosphorylation and mitochondrial fission, but inhibit mitochondrial respiration. Confocal microscopic data indicated a synergism of NaIO3 and bafilomycin A1 on LC3 punctate formation, indicating the induction of autophagy. Using cytosolic ROS antioxidant NAC, we found that p38 and JNK are downstream signals of ROS and involve in NaIO3-induced cytotoxicity but not in mitochondrial dynamics, while ROS is also involved in LC3II expression. Unexpectedly NAC treatment upon NaIO3 stimulation leads to an enhancement of mitochondrial fragmentation and cell death. Moreover, inhibition of autophagy and Akt further enhances cell susceptibility to NaIO3.
    CONCLUSIONS: We conclude that NaIO3-induced oxidative stress and cytosolic ROS production exert multiple signaling pathways that coordinate to control cell death in RPE cells. ROS-dependent p38 and JNK activation lead to cytotoxicity, while ROS-mediated autophagy and mitochondrial dynamic balance counteract the cell death mechanisms induced by NaIO3 in RPE cells.
    Keywords:  Age-related macular degeneration; Autophagy; Mitochondrial dynamics; Reactive oxygen species; Retinal epithelium; Sodium iodate
    DOI:  https://doi.org/10.1186/s12929-019-0531-z
  30. Artif Cells Nanomed Biotechnol. 2019 Dec;47(1): 1961-1970
      Alantolactone (Ala), a major sesquiterpene lactone extracted from Inula helenium, exerts potent anti-tumour activities in various cancers. However, the underlying mechanism of such activities is still ambiguous. This study focused on evaluating the anti-tumour effects and molecular mechanisms of Ala on HepG2 cells. Our results demonstrated that Ala might inhibit cellular proliferation, induce G2/M phase arrest and apoptosis in HepG2 cells. Specifically, this study confirmed that Ala induced G2/M phase arrest by upregulating p21, downregulating cyclin A1 and cyclin B1, and promoting cellular apoptosis by increasing the expression of cleaved caspase-3 and PARP. Furthermore, Ala caused an increase in reactive oxygen species (ROS) level and inhibition of ROS production significantly prevented Ala-induced apoptosis. Interestingly, the accumulation of ROS, in turn, suppressed the downstream AKT signalling. Finally, mitophagy of Ala-treated HepG2 cells was observed by Mito/Lyso staining. Mitophagy was significantly inhibited by downregulation of the expression of PINK1 and Parkin proteins. The inhibition of mitophagy by a mitophagy inhibitor was found to markedly enhance Ala-mediated apoptosis and growth inhibition in HepG2 cells. Consequently, Ala induced cellular apoptosis via ROS-mediated suppression of AKT signalling and inhibition of PINK1-mediated mitophagy. Thus, Ala has potential to be used for the treatment of liver cancer.
    Keywords:  AKT; Alantolactone; ROS; apoptosis; mitophagy
    DOI:  https://doi.org/10.1080/21691401.2019.1593854
  31. J Food Sci. 2019 May 22.
      In this study, bamboo leaf flavonoids extracts (BFE) were employed to alleviate oxidative stress induced by oleic acid in HepG2 cells. Biochemical indexes, intracellular reactive oxygen species production, lipid droplets accumulation, antioxidant enzymes production, and mitochondrial membrane potential were determined to show the alleviation performance of BFE intervention (P < 0.05). Importantly, the results of qRT-PCR and western blot determination indicated that BFE intervention upregulated the expression of Nrf2/HO-1/NQO1 to initiate the antioxidant defense response for counteracting oxidative stress (P < 0.05). Moreover, mitochondrial membrane potential-mediated apoptosis and FOXO signaling pathway initiation caused by BFE intervention may together contribute to oxidative stress alleviation in HepG2 cells. In conclusion, these findings suggested that BFE intervention upregulated related antioxidant defense responses for preventing cells from oxidative damage. PRACTICAL APPLICATION: In this study, bamboo leaf flavonoids extracts intervention upregulated related antioxidant defense responses for preventing cells from oxidative damage. These findings in bamboo leaf extracts antioxidants are a promising and innovative subject with practical applications to enhance the development of bamboo leaf extracts functional products in the food industry.
    Keywords:  antioxidant defense responses; bamboo leaf; flavonoids; oleic acid; oxidative stress
    DOI:  https://doi.org/10.1111/1750-3841.14609
  32. Onco Targets Ther. 2019 ;12 2605-2614
      Objective: Shikonin has inhibitory effects against nasopharyngeal carcinoma that are mediated through the apoptotic pathway. However, necroptosis signaling pathways may enable the elimination of apoptosis-resistant cancers when induced with targeted therapeutic agents. Thus, there is a need to clarify whether shikonin can cause necroptosis in nasopharyngeal carcinoma and to elucidate the underlying mechanisms. Methods: In this study, we used the nasopharyngeal carcinoma cell line 5-8F and a 5-8F xenograft mouse model to evaluate the anticancer effects of shikonin. The viability and morphology of cells treated with shikonin were evaluated using CCK-8 assay and transmission electron microscopy, respectively. In addition, the expression levels of RIPK1, RIPK3, and MLKL were analyzed by western blotting, and the activities of caspase-3 and caspase-8 and levels of reactive oxygen species (ROS) were assessed. Results: Shikonin exhibited a strong inhibitory effect on 5-8F cells in vitro and in vivo. The shikonin-treated 5-8F cells presented an electron-lucent cytoplasm, loss of plasma membrane integrity, and an intact nuclear membrane, indicating that shikonin induced necroptosis. Shikonin-induced cell death was inhibited by necrostatin-1. Moreover, RIPK1, RIPK3, and MLKL were upregulated by shikonin in a dose-dependent manner. Furthermore, shikonin significantly inhibited tumor growth in the 5-8F xenograft mouse model. Conclusion: Shikonin induced 5-8F cell death via increased ROS production and the upregulation of RIPK1/RIPK3/MLKL expression, resulting in necroptosis. Thus, shikonin may represent a novel agent to treat nasopharyngeal carcinoma.
    Keywords:  nasopharyngeal carcinoma; necroptosis; reactive oxygen species; shikonin
    DOI:  https://doi.org/10.2147/OTT.S200740
  33. Toxicon. 2019 May 21. pii: S0041-0101(19)30158-8. [Epub ahead of print]
      The present study evaluated hepatoprotective role of ethanol extract of P. integrifolia leaves (EEPL) on aflatoxin B1 (AFB1)-induced toxicity in mice. Mice were administered with AFB1 (0.1 mg/kg b. wt., orally) for 90 days, EEPL (400 and 600 mg/kg b. wt., orally) and silymarin (100 mg/kg b. wt., orally) in combination with AFB1. The study shows protective effect of EEPL by the restoration of altered haematological indices and liver marker enzymes. Restoration of lipid peroxidation and glutathione content, along with activities of antioxidant enzymes, suggest amelioration of oxidative stress in AFB1-intoxicated mice. In addition, EEPL attenuated apoptosis and histopathological alterations in liver tissue. In conclusion, the current study suggests that EEPL protect mice liver against AFB1 toxicity by inhibiting oxidative stress and apoptosis. The protective activity of EEPL may be due to the enrichment of flavonoids (neohesperidin, apigenin-7-O-glucoside, catechin hydrate, cyanidin chloride, quercetin-3-galactoside, diosmin, genistein, malvin chloride, 4-hydroxy-3-methoxycinnamic acid, kaempferol-3-O-alpha-L-arabinoside, myricitrin, poncirin, vitexin and tiliroside) in the extract as identified by UPLC-QTOF-MS/MS.
    Keywords:  Aflatoxin B1; Apoptosis; Hepatotoxicity; Oxidative stress; Premna integrifolia
    DOI:  https://doi.org/10.1016/j.toxicon.2019.05.014
  34. Biol Pharm Bull. 2019 May 18.
      The purpose of the paper is to study the differences in cell death mechanism of MGC-803 induced by "dextran-magnetic layered double hydroxide-fluorouracil" (DMF) drug delivery system and 5-Fluorouracil (5-Fu), respectively. The inhibitory effect on the proliferation was detected via CCK-8. The morphology of cell death was detected by transmission electron microscopy (TEM). Intracellular ATP, mitochondrial membrane potential (MMP), reactive oxygen species (ROS) and Cytosolic Free Ca (Ca2+) level were detected via some methods. The result showed that DMF had more obvious effect in suppressing proliferation compared with 5-Fu, and changed cell death pattern of 5-Fu from apoptosis to oncosis. The ATP decrease, MMP loss, Ca2+ increase, the activation of UCP-2 and caplain-1 were significant after DMF exposure. However, DMF did not result in ROS accumulation. DMF could involve in activation of porimin, and the cascade reaction of caspase-3, 7, 9, 12 and PARP through western blot. DMF showed a stronger injury on nuclear membrane in the cascade reaction of caspase-6, caspase-8 and lamin-A. DMF triggered rapid depletion of ATP, which was consistent with the phenotype of oncosis. Endogenous mitochondrial apoptosis might not be the main cause of cell swelling. DMF could induce strong endoplasmic reticulum stress (ERS) effect, there might be some signaling pathways related with ERS during the process of oncosis. The calpain system might not be a key factor for structural damage in oncosis induced by DMF. DMF could induce the caspases cascade reactions similar to apoptosis, but inflicted a more strong damage on nuclear membrane and PARP.
    Keywords:  Dextran-magnetic layered double hydroxide-fluorouracil; MGC-803; death mechanism; oncosis
    DOI:  https://doi.org/10.1248/bpb.b18-00938
  35. Environ Toxicol. 2019 May 21.
      The di (2-ethylhexyl) phthalate (DEHP) is a plasticizer used in the polyvinyl chloride industry. Human exposure to this plasticizer is inevitable and contributes to several side effects. In this study, we examined whether DEHP induces apoptosis and oxidative stress in embryonic kidney cells (HEK-293) and whether the nuclear factor E2-related factor 2 (Nrf-2)/heme oxygenase-1 (HO-1) antioxidant pathway is involved in the pathogenesis of this process. We demonstrated that DEHP is cytotoxic to HEK-293 cells. It causes oxidative damage through the generation of free radicals, induces lipid peroxidation, and alters superoxide dismutase and catalase activities. Simultaneously, DEHP treatment decreases the expression and the protein level of Nrf-2 and HO-1. Inhibition of the Nrf-2/HO-1 pathway is related to the mitochondrial pathway of apoptosis. This apoptotic process is characterized by a loss of mitochondrial transmembrane potential (ΔΨm) and upregulation of the expression of caspase-3 mRNA as well as its protein level.
    Keywords:  Nrf2/HO-1 antioxidant pathway; apoptosis; di (2-ethylhexyl) phthalate; oxidative stress
    DOI:  https://doi.org/10.1002/tox.22774
  36. J Inorg Biochem. 2019 May 10. pii: S0162-0134(18)30625-1. [Epub ahead of print]197 110696
      A water soluble trinuclear copper(II) complex and a binuclear cobalt(II) complex, namely Cu3(ppbm)2(SO4)3 (1) and Co2(ppbm)2(NO3)4 (2) (ppbm = 2-(pyridin-2-yl)-1-(pyridin-3-ylmethyl)- 1H-benzo[d]imidazole), have been successfully synthesized and characterized by elemental analysis, IR Spectroscopy, electrospray ionization mass spectra (ESI-MS). The interaction of the new complexes with DNA has been explored using spectroscopy methods, indicating that the complexes 1 and 2 bind to DNA via noncovalent interactions. DNA cleavage experiment suggested that the complex 1 exhibits efficient DNA cleavage activities in the presence of ascorbate (Asc), hydrogen peroxide may serve as the major cleavage active species. The cytotoxicity assay showed that complex 1 exhibited significant inhibitory activity toward the proliferation of several tumor cell lines, with a lower IC50 value than cisplatin and complex 2, indicating that it had the potential to act as effective anticancer agent. The morphological staining assays showed that 1 apparently induced the TFK-1 cells apoptosis. Besides, cellular uptake experiment on TFK-1 cells revealed that complex 1 accumulates primarily inside the nucleus. The apoptosis was attributable to the metal-assisted generation of reactive oxygen species (ROS).
    Keywords:  DNA interactions; In vitro anticancer activity; Metal (II) complexes; Reactive oxygen species
    DOI:  https://doi.org/10.1016/j.jinorgbio.2019.110696
  37. J Mol Histol. 2019 May 20.
      Cardiac hypertrophy is a compensatory response in reaction to mechanical load that reduces wall stress by increasing wall thickness. Chronic hypertrophic remodeling involves cardiac dysfunction that will lead to heart failure and ultimately death. Studies have been carried out on cardiac hypertrophy for years, whereas the mechanisms have not been well defined. Tamarixetin (TAM), a natural flavonoid derivative of quercetin, have been demonstrated possessing anti-oxidative and anti-inflammatory effects on multiple diseases. However, little is known about the function of TAM on the development of cardiac hypertrophy. Here, we found TAM could alleviate pressure-overload-induced cardiac hypertrophy in transverse aortic constriction (TAC) mouse model, assessed by ventricular weight/body weight, lung weight/body weight, echocardiographic parameters, as well as myocyte cross-sectional area and the expression of ANP, BNP and Myh7. In vitro, TAM showed a dose dependent inhibitory effect on phenylephrine-induced hypertrophy in H9c2 cardiomyocytes. Furthermore, TAM reversed cardiac remodeling of stress overloaded heart by suppressing apoptosis and the expression of fibrotic-related genes, reduced oxidative stress and ROS production both in vivo and in vitro. In addition, TAM could negatively modulate TAC-induced nuclear translocation of NFAT and the activation of PI3K/AKT signaling pathways. Therefore, these data indicate for the first time that TAM has a protective effect on experimental cardiac hypertrophy and might be a novel candidate for the treatment of cardiac hypertrophy in clinic.
    Keywords:  Cardiac hypertrophy; Oxidative stress; Remodeling; Tamarixetin
    DOI:  https://doi.org/10.1007/s10735-019-09831-1
  38. Cell Death Discov. 2019 ;5 95
      Mast cells contribute to the pathology of allergic and other disorders. Strategies to interfere with harmful mast cell-related activities are therefore warranted. Previously we established a principle for inducing selective apoptosis of mast cells, by the use of lysosomotropic agents that cause secretory granule permeabilization, leading to production of reactive oxygen species (ROS). However, the mechanism of ROS production has not been known. Here we addressed this issue. Live microscopy analysis showed that the secretory granules comprise major subcellular compartments for ROS production in response to mefloquine. As further signs for the primary involvement of secretory granules, both ROS production and cell death was blunted in mast cells lacking serglycin, a secretory granule-restricted proteoglycan. Inhibition of granule acidification caused an essentially complete blockade of granule permeabilization, ROS production and cell death in response to mefloquine. ROS production was also attenuated in the presence of an iron chelator, and after inhibition of either granzyme B or the ERK1/2 MAP kinase signaling pathway. Together, our findings reveal that the mast cell secretory granules constitute major sites for ROS production in mast cells subjected to lysosomotropic challenge. Moreover, this study reveals a central role for granule acidification in ROS generation and the pro-apoptotic response triggered downstream of secretory granule permeabilization.
    DOI:  https://doi.org/10.1038/s41420-019-0177-3
  39. Drug Des Devel Ther. 2019 ;13 1187-1196
      Background: Bupivacaine (BUP) acts as a local anesthetic, which is extensively used for clinical patients but could generate neurotoxicity in neurons. Tetramethylpyrazine (TET) exhibits strong neuron protective effects against neurotoxicity. Hence, we investigate the effect of TET on BUP-induced neurotoxicity in SH-SY5Y cells. Methods: CCK-8 assay was used to detect cell proliferation in SH-SY5Y cells. In addition, Western blotting was used to examine Bax, Bcl-2, active caspase 3, LC3II, Beclin 1 and p-62 protein levels in cells. Moreover, ELISA assay was used to detect the levels of total glutathione (GS), superoxide dismutase (SOD) and malondialdehyde (MDA) in cells. Results: In this study, we found that TET attenuated the neurotoxicity of BUP on SH-SY5Y cells. Meanwhile, TET alleviated BUP-induced apoptosis in SH-SY5Y cell via decreasing the expressions of active caspase-3 and Bax and increasing the expression of Bcl-2. In addition, monodansylcadaverine staining assay and Western blotting results confirmed that TET induced autophagy in SH-SY5Y cells via increasing the LC3II/I and Beclin 1 levels. Furthermore, TET attenuated BUP-induced oxidative damage in SH-SY5Y cells via upregulation of the levels of total GS and SOD and downregulation of the level of MDA. Interesting, the protective effects of TET against BUP-induced neurotoxicity in SH-SY5Y cells were reversed by autophagy inhibitor 3-methyladenine (3MA). Conclusion: These data indicated that TET may play a neuroprotective role via inhibiting apoptosis and inducing autophagy in SH-SY5Y cells. Therefore, TET may be a potential agent for the treatment of human neurotoxicity induced by BUP.
    Keywords:  bupivacaine; human neuroblastoma cell; neurotoxicity; tetramethylpyrazine
    DOI:  https://doi.org/10.2147/DDDT.S196172
  40. Int J Biol Macromol. 2019 May 20. pii: S0141-8130(19)30754-8. [Epub ahead of print]
      Withania somnifera exhibits different pharmacological activities which mainly stem from its broad range of bioactive molecules. Majority of these bioactive molecules, fall into the groupings of alkaloids, steroidal lactones, phenolic compounds and glycoproteins. In this study, we evaluated a novel protein fraction, named here as WSPF, isolated from Withania somnifera roots for its cytotoxic properties against various human cancer cell lines. WSPF exhibited apoptotic activity for each cancer cell line tested, demonstrating significant activity against MDA-MB-231 human breast cancer cells with an IC50 value of 92 μg/mL. WSPF induced mitochondrial-mediated apoptosis of MDA-MB-231 cells via extensive reactive oxygen species generation, dysregulation of Bax/Bcl-2, loss of mitochondrial membrane potential and caspase-3 activation. Additionally, we observed G2/M-phase cell cycle arrest, cleavage of nuclear lamin A/C proteins, and nuclear morphological changes. The present results highlight the anti-cancer properties of WSPF, indicating that the proteins in this fraction can be potential therapeutic agents for triple negative breast cancer treatment.
    Keywords:  Apoptosis; Cytotoxicity; Therapeutic proteins
    DOI:  https://doi.org/10.1016/j.ijbiomac.2019.05.120
  41. Biochem Biophys Res Commun. 2019 May 17. pii: S0006-291X(19)30978-7. [Epub ahead of print]
      Cancer stem cells (CSCs) have been shown to be resistant to current anticancer therapies and the induction of oxidative stress is an important mechanism of action for many anticancer agents. However, it is still largely unknown how CSCs respond to hydrogen peroxide (H2O2)-induced oxidative stress. Here, we show that the levels of reactive oxygen species (ROS) are markedly lower in breast CSCs (BCSCs) than that in non-cancer stem cells (NCSCs). A transient exposure of breast cancer cells to sublethal doses of H2O2 resulted in a dose-dependent increase of the epithelium-specific antigen (ESA)+/CD44+/CD24- subpopulations, a known phenotype for BCSCs. Although BCSCs survived sublethal doses of H2O2 treatment, they lost the ability to form tumor spheres and failed to generate colonies as demonstrated by mammosphere-formation and clonogenic assays, respectively. Mechanistic studies revealed that H2O2 treatment led to a marked increase of senescence-associated β-galactosidase activity but only minimal apoptotic cell death in BCSCs. Furthermore, H2O2 triggers p53 activation and promotes p21 expression, indicating a role for the p53/p21 signaling pathway in oxidative stress-induced senescence in BCSCs. Taken together, these results demonstrate that the maintenance of a lower level of ROS is critical for CSCs to avoid oxidative stress and H2O2-induced BCSC loss of function is likely attributable to oxidative stress-triggered senescence induction, suggesting that ROS-generating drugs may have the therapeutic potential to eradicate drug-resistant CSCs via induction of premature senescence.
    Keywords:  Breast cancer; Cancer stem cells; Hydrogen peroxide; Oxidative stress; Senescence
    DOI:  https://doi.org/10.1016/j.bbrc.2019.05.098
  42. Am J Transl Res. 2019 ;11(4): 2550-2569
      Prostate cancer is still at the forefront causes of cancer-related morbidity and mortality in men throughout the globe. The disease is initiated and fostered by a subset of cancer stem cells (CSCs). Costus speciosus is an oriental herb used in traditional medicine and is a source of bioactive compounds with known pharmacological activities. The present study aims to evaluate the anticancer property of varied extracts isolated from C. speciosus against the human prostate cancer PC-3 cells. Extracts derived from C. speciosus were analyzed by chromatography-mass spectrometry and their effects on the proliferation, migration, invasion, apoptosis and cell cycle distribution of PC-3 cells were investigated. Results showed that crude hexane extract of C. speciosus (CHECS) inhibited proliferation, clonogenic and metastatic potential of PC-3 cells. It induced apoptosis in PC-3 cells associated with generation of reactive oxygen species (ROS), reduction of GSH and permeabilization of mitochondrial and lysosomal membranes, induction of caspase-9/-3 activity and PARP-1 cleavage, DNA damage and an increase in ratio of Bax/Bcl-2 proteins. CHECS induced G0/G1 and G2/M arrest in PC-3 cells and targeted PC-3 prostaspheres. These findings indicate that phytochemicals of CHECS exhibit potential for natural therapeutic product development for prostate cancer.
    Keywords:  Phytomedicine; apoptosis; cell cycle; metastasis; oxidative stress; stem cells
  43. J Trace Elem Med Biol. 2019 Jul;pii: S0946-672X(19)30074-4. [Epub ahead of print]54 69-78
      OBJECTIVE: Mercury is a dangerous industrial and environmental pollutant which induces severe damage in diverse organs in animal and humans. The aim of this study was to investigate the protective effect of rutin (50 and 100 mg/kg body weight) against mercuric chloride (HgCl2) (1.23 mg/kg b.w.) toxicity in rats.METHODS: The experiment was carried out in male Sprague Dawley rats (n = 35) which was divided into five groups as follow: control, rutin-100, HgCl2, HgCl2 + rutin-50 and HgCl2 + rutin-100.
    RESULTS: The results showed that HgCl2 caused a marked increase in the malondialdehyde (MDA) level and significantly decreased antioxidant enzyme activities (p < 0.05). HgCl2 also provoked inflammatory responses by elevating the levels of tumor necrosis factor-α (TNF-α), B-cell lymphoma-3 (Bcl-3), interleukin-1β (IL-1β), nuclear factor kappa B (NF-κB), interleukin-33 (IL-33), and activities of mitogen-activated protein kinase 14 (MAPK 14) and myeloperoxidase (MPO) (p < 0.05). HgCl2 also prompted the apoptotic pathway by increasing the levels of Bcl-2 associated X protein (Bax) and p53, expression of terminal deoxynucleotidyl transferase dUNT nick end labeling (TUNEL) and cysteine aspartate specific protease-3 (caspase-3). HgCl2 changed histological integrity of kidney and expression of 8-hydroxy-2'-deoxyguanosine (8-OHdG) while caused a decrease in aquaporin 1 (AQP1) water channel protein level. In contrast to this, rutin significantly decreased oxidative stress, apoptosis, inflammation and histopathological alterations while increased AQP1 levels in kidney tissues (p < 0.05).
    CONCLUSION: The present study indicated that rutin has a nephroprotective effect due to its anti-inflammatory, antioxidant and antiapoptotic properties.
    Keywords:  Apoptosis; Inflammation; Mercuric chloride; Nephrotoxicity; Oxidative stress; Rutin
    DOI:  https://doi.org/10.1016/j.jtemb.2019.04.007
  44. Curr Pharm Des. 2019 May 06.
      The objective of the study was to examine the therapeutic role of combined ischemic preconditioning (IPC) and resveratrol (RES) on brain ischemia/reperfusion injury (BI/RI) by modulating endogenous bone morphogenetic protein-4 (BMP-4)/reactive oxygen species (ROS)/cyclooxygenase-2 (COX-2) in rats. Sprague Dawley (SD) rats were pretreated with 20 mg/kg RES (20 mg/kg RES was administered once a day via intraperitoneal injection 7 days prior to the I/R procedure) and IPC (equal volumes of saline were administered once a day by intraperitoneal injection over 7 days, and the bilateral common carotid arteries were separated for clamp 5 minutes followed by 5 minutes of reperfusion prior to the I/R procedure), and then subjected to 2 hours of ischemia and 22 hours of reperfusion. Blood and cerebral tissues were collected, cerebral pathological injuries and infarct sizes were investigated, serum interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) levels were measured, the activities of superoxide dismutase (SOD) and ROS were calculated, the contents of methane dicarboxylic aldehyde (MDA), IL-6, TNF-α and hemodynamic change were estimated, and expression levels of b-cell lymphoma-2 (Bcl-2), bcl-2-associated x (Bax), BMP-4 and COX-2 were assessed in cerebral tissues. IPC、RES and a combination of IPC and RES preconditioning ameliorated the pathological damage and infarct sizes, reduced cerebral oxidative stress damage, alleviated inflammatory damage, restrained apoptosis, and downregulated the expression levels of BMP-4 and COX-2 compared with those of the ischemia/reperfusion (I/R) group. This study suggested a combined strategy that could enhance protection against BI/RI in clinical brain disease.
    Keywords:  BMP4/ROS/COX2; Bax; Bcl2; IPC; RES
    DOI:  https://doi.org/10.2174/1381612825666190506120611
  45. J Exp Clin Cancer Res. 2019 May 21. 38(1): 207
      BACKGROUND: Cisplatin is one of the most widely used chemotherapeutic agents, but its efficacy is limited by its side effects. Hence, it is of great significance to develop novel agents to synergize with cisplatin and decrease side effects. In our previous study, we demonstrated that WZ35, a novel curcumin analogue, exhibited potent anti-cancer effects in vitro and in vivo. Here, we investigated whether WZ35 synergize to potentiate cisplatin activity in gastric cancer cells.METHODS: Cell apoptosis and cellular ROS levels were analyzed by flow cytometry. TrxR1 activity in gastric cells or tumor tissues was determined by the endpoint insulin reduction assay. Western blot was used to analyze the levels of indicated molecules. Nude mice xenograft model was used to test the effects of WZ35 and cisplatin combination on gastric cancer cell growth in vivo.
    RESULTS: We found that WZ35 significantly enhanced cisplatin-induced cell growth inhibition and apoptosis in gastric cancer cells. Further mechanism study showed that WZ35 synergized the anti-tumor effects of cisplatin by inhibiting TrxR1 activity. By inhibiting TrxR1 activity, WZ35 combined with cisplatin markedly induced the production of ROS, activated p38 and JNK signaling pathways, and eventually induced apoptosis of gastric cancer cells. In vivo, WZ35 combined with cisplatin significantly suppressed tumor growth in a gastric cancer xenograft model, and effectively reduced the activity of TrxR1 in tumor tissues. Remarkably, WZ35 attenuated the body weight loss evoked by cisplatin treatment.
    CONCLUSION: This study elucidated the underlying mechanisms of synergistic effect of WZ35 and cisplatin, and suggest that such a combinational treatment might potentially become a more effective regimen in gastric cancer therapy.
    Keywords:  Cisplatin; Gastric cancer; Reactive oxygen species; Thioredoxin reductase 1; WZ35
    DOI:  https://doi.org/10.1186/s13046-019-1215-y
  46. Environ Toxicol. 2019 May 21.
      LY303511 was developed as a negative control of LY294002 without pan-phosphoinositide 3-kinase (PI3K) inhibition. We hypothesize LY303511 generate reactive oxygen species (ROS) to induce apoptosis for killing oral cancer cells. In MTS assay, LY303511 dose-responsively decreases survival in three kinds of oral cancer cells but little damage to normal oral cells (HGF-1). Two oral cancer cells (CAL 27 and SCC-9) with highly sensitivity to LY303511 were used. In 7-aminoactinomycin D (7AAD) assay, LY303511 slightly increases subG1 population in oral cancer cells. In annexin V/7AAD and/or pancaspase assays, LY303511 induces apoptosis in oral cancer cells but HGF-1 cells remains in basal level. In oxidative stress, LY303511 induces ROS and mitochondrial superoxide in oral cancer cells. In 8-oxo-2'-deoxyguanosine assay, LY303511 induces oxidative DNA damage in oral cancer cells. In zebrafish model, LY303511 inhibits CAL 27-xenografted tumor growth. Therefore, LY303511 displays antiproliferation potential against oral cancer cells in vitro and in vivo.
    Keywords:  LY303511; oral cancer; oxidative stress; preferential killing; zebrafish xenograft model
    DOI:  https://doi.org/10.1002/tox.22767
  47. Theriogenology. 2019 May 09. pii: S0093-691X(19)30126-8. [Epub ahead of print]134 1-10
      The mammalian Sirtuin family of seven enzymes, members of the NAD+-dependent histone deacetylase family that modify histones via direct deacetylation, is involved in the regulation of many antioxidant and oxidative stresses. In the present study, we explored the effects of nicotinamide (NAM)-induced oxidative stress on the in vitro development of bovine embryos, on the acetylation of histone H3 lysine 56 (H3K56ac) and on expression of apoptosis-related genes. Treatment with NAM (10, 20 or 40 mM for 24, 48 or 196 h) during IVC resulted in significantly decreased blastocyst formation (24 h: 38.8 vs. 33.1, 27.3 and 10.2%, with P > 0.05, P < 0.05 and P < 0.01, respectively; 48 h: 37.5 vs. 28.2, 13.4 and 0%, with P < 0.05 and P < 0.01, respectively; 196 h: 35.8 vs. 23.4, 0 and 0%, with P < 0.05, respectively). Treatment with NAM (20 and 40 mM for 24 h) resulted in increased intracellular reactive oxygen species (ROS) levels in 2-cell and blastocysts, and apoptotic cell numbers in blastocysts and decreased mitochondrial membrane potential (ΔΨ) in 2-cell embryos (P < 0.05). Polydatin (PD) and I-CBP112 rescued the 20 mM NAM-induced embryo developmental defects and reduced ROS levels and apoptotic cell numbers in blastocysts (P < 0.05). The gene expression of NF-κB, COX2 and p53 was significantly increased in the NAM-treated group. Immunofluorescence analysis confirmed that the protein levels of nuclear factor-kappa B (NF-κB) decreased significantly after PD and I-CBP112 treatment compared with the control (P < 0.05). High level of H3K56ac induced by NAM was decreased after PD and I-CBP112 treatment (P < 0.05). These findings suggest that NAM treatment induces high levels of H3K56 acetylation that may be involved in oxidative stress-induced bovine developmental defects, which can be tolerated by PD and I-CBP112 treatment.
    Keywords:  Apoptosis; Bovine blastocyst; H3K56 acetylation; Oxidative stress; Sirtuins
    DOI:  https://doi.org/10.1016/j.theriogenology.2019.05.007
  48. J Immunoassay Immunochem. 2019 May 22. 1-7
      Reactive oxygen species (ROS) are implicated in inflammatory, autoimmune, and neurodegenerative diseases. The activation, proliferation, and apoptosis of immune cells are dependent on the controlled production and elimination of ROS. However, under chronic inflammatory conditions, large amounts of ROS generated are a major cause of many human diseases. The electronically excited molecular oxygen species known as singlet oxygen (1O2) is a form of ROS and is one of the major cytotoxic species in eukaryotic cells. ROS are known to cause DNA damage leading to strand breaks, base damage, and conformational changes. The 1O2, being one of the most potent ROS, is generated by photoexcitation or by chemiexcitation and is known to selectively react with the deoxyguanosine moiety in DNA. The biological consequences of 1O2-induced damage causes loss of transforming activity as well as mutagenicity and genotoxicity and may lead to the formation of neo-epitopes in native DNA and generation of pathogenic anti-DNA antibodies. The excessive production of ROS may be one of the factors responsible for the induction of autoimmune response in diseases such as cancer and systemic lupus erythematosus. Abbreviations: Singlet oxygen: 1O2; Superoxide anion: O2-.; Hydroxyl radical: OH; Hydrogen peroxide: H2O2.
    Keywords:  SLE; Singlet oxygen; anti-DNA autoantibodies; autoimmunity
    DOI:  https://doi.org/10.1080/15321819.2019.1616555
  49. J Cell Mol Med. 2019 May 21.
      The damage of vascular endothelial cells induced by oxidative stress plays an important role in the pathogenesis of atherosclerosis. Dihydromyricetin (DMY) is considered as a natural antioxidant. However, the mechanism of DMY on endothelial cell injury induced by oxidative stress remains unclear. In this study, we found that DMY could reduce the oxidative damage of HUVECs induced by sodium nitroprusside (SNP), HUVECs pre-treated with DMY suppressed SNP-induced apoptosis by reduced ROS overproduction of intracellular, decreased MDA level and elevated the superoxide dismutase activity. Meanwhile, we found that DMY could promote the expression of phosphorylated FoxO3a and Akt, and affect the nuclear localization of FoxO3a, when treated with the PI3K inhibitor LY294002, the effect of DMY was blocked. These data suggest that DMY protects HUVECs from oxidative stress by activating PI3K/Akt/FoxO3a signalling pathway. Therefore, DMY may have great therapeutic potential as a new drug for atherosclerosis.
    Keywords:  apoptosis; atherosclerosis; dihydromyricetin; oxidative stress; sodium nitroprusside
    DOI:  https://doi.org/10.1111/jcmm.14406
  50. Aging (Albany NY). 2019 May 22.
      Gibberellin, a plant growth regulator, is widely used to increase the shelf life and quality of fruits and vegetables. In this study, human semen samples were exposed to different concentrations of gibberellin, which reduced spermatozoa motility in vitro. Gibberellin exposure also increased levels of reactive oxygen species and the protein levels of apoptosis markers in human sperm. Gibberellin inhibited the activity of Na+/K+-adenosine triphosphatase (ATPase) and Ca2+-ATPase, which maintain the stability of ions inside and outside the membranes of spermatozoa. Moreover, gibberellin exposure suppressed adenosine triphosphate production and reduced the protein levels of adenosine triphosphate synthases, which may have induced the protein expression of adenosine 5'-monophosphate-activated protein kinase (AMPK) and its phosphorylated form. These results suggest that gibberellin reduces human sperm motility in vitro by increasing reactive oxygen species levels and reducing ATPase activity, which may upregulate AMPK and consequently reduce the fertilization potential of spermatozoa.
    Keywords:  AMPK; ATPase activity; gibberellin; oxidative stress; sperm motility
    DOI:  https://doi.org/10.18632/aging.101963
  51. Front Pharmacol. 2019 ;10 417
      Pelvic and abdominal radiotherapy plays an important role in eradication of malignant cells; however, it also results in slight intestinal injury. The apoptosis of cells in the intestinal epithelium is a primary pathological factor that initiates radiation-induced intestinal injury. Auranofin, a gold-containing triethylphosphine, was approved for the treatment of rheumatoid arthritis, and its therapeutic application has been expanded to a number of other diseases, such as parasitic infections, neurodegenerative disorders, AIDS, and bacterial infections. Recently, a treatment strategy combining the use of auranofin and ionizing radiation aimed at increasing the radiosensitivity of cancer cells was proposed for improving the control of local cancers. In this study, we evaluated the effect of auranofin on the radiosensitivity of intestinal epithelial cells. The treatment with a combination of 1 μM auranofin and 5 Gy ionizing radiation showed clear additive effects on caspase 3 cleavage and apoptotic DNA fragmentation in IEC-6 cells, and auranofin administration significantly aggravated the radiation-induced intestinal injury in mice. Auranofin treatment also resulted in the activation of the unfolded protein response and in the inhibition of thioredoxin reductase, which is a key component of the cellular antioxidant system. Pre-treatment with N-acetyl cysteine, a well-known scavenger of reactive oxygen species, but not with a chemical chaperone, which inhibits endoplasmic reticulum stress and the ensuing unfolded protein response, significantly reduced the radiosensitizing effects of auranofin in the IEC-6 cells. In addition, transfection of IEC-6 cells with a small interfering RNA targeted against thioredoxin reductase significantly enhanced the radiosensitivity of these cells. These results suggest that auranofin-induced radiosensitization of intestinal epithelial cells is mediated through oxidative stress caused by the deregulation of thioredoxin redox system, and auranofin treatment can be an independent risk factor for the development of acute pelvic radiation disease.
    Keywords:  apoptosis; auranofin; intestinal mucosa; oxidative stress; radiosensitivity; thioredoxin reductase
    DOI:  https://doi.org/10.3389/fphar.2019.00417
  52. Environ Toxicol. 2019 May 21.
      Ginsenoside Rb1 (GRb1), the major saponin component of ginseng root, has a wide range of therapeutic applications for various diseases. Previously, our group showed that GRb1 triggers ROS-mediated apoptotic cascades in mouse blastocysts, leading to decreased cell viability and impairment of pre- and postimplantation embryonic development, both in vitro and in vivo. In this study, we further found that GRb1 exerted dose-dependent effects on oocyte maturation and sequent development in vitro. Oocytes preincubated with 25 μg/mL GRB1 displayed significantly enhanced maturation and in vitro fertilization (IVF) rates, along with progression of subsequent embryonic development. In contrast, treatment with 50 and 100 μg/mL GRB1 led to impairment of mouse oocyte maturation, decreased IVF rates, and injurious effects on subsequent embryonic development. In vivo, intravenous injection of 1 mg/kg body weight GRb1 significantly promoted mouse oocyte maturation, IVF, and early-stage embryo development after fertilization while administration of 5 mg/kg body weight GRb1 led to a marked decrease in oocyte maturation and IVF rates concomitant with impairment of early embryonic development in our animal model. In terms of the mechanisms underlying the regulatory effects of GRb1 demonstrated increased intracellular reactive oxygen species (ROS) production and apoptosis in the 100 μg/mL GRb1 treatment group. However, we observed a significant decrease in total intracellular ROS content and inhibition of apoptosis events in the 25 μg/mL GRb1 treatment group, signifying that the intracellular ROS content serves as a key upstream regulator of GRb1 that influences its dose-dependent beneficial or deleterious effects on oocyte maturation and sequent embryonic development. For further clarification of the mechanisms underlying GRb1-triggered injurious effects, oocytes were pretreated with Ac-DEVD-CHO, a caspase-3-specific inhibitor, which effectively blocked injury to oocyte maturation, fertilization, and sequent development. In sum, study findings highlight the potential involvement of p53-, p21-, and caspase-3-dependent regulatory signaling cascades in GRb1-mediated apoptotic processes.
    Keywords:  ROS; apoptosis; embryonic development; ginsenoside Rb1; oocyte maturation
    DOI:  https://doi.org/10.1002/tox.22771
  53. Food Res Int. 2019 Jul;pii: S0963-9969(19)30190-5. [Epub ahead of print]121 176-184
      The neuroprotective effect of peptide fractions of different molecular weights obtained from beef by commercial enzymes in SH-SY5Y cells was investigated. The <3 kDa and < 10 kDa peptide fractions were obtained by hydrolysis using inexpensive commercial enzymes (alkaline-AK and papain). The resulting peptide fractions decreased the apoptotic ratio by 20.06% to 33.25% in hydrogen peroxide treated SH-SY5Y cells. Moreover, the peptide fractions obtained by alkaline-AK and papain did not affect cytotoxicity. In addition, the <3 kDa peptide fractions obtained using alkaline-AK reduced DNA fragmentation and reactive oxygen species generation, and increased superoxide dismutase activity and catalase levels in H2O2 treated SH-SY5Y cells. Furthermore, the <3 kDa and < 10 kDa peptide fractions showed strong antioxidant activities in 2,2'-azino-bis (3-ethylbenzothiazoline-6-sulfonic acid) diammonium salt, iron chelation, and reducing power tests. The present results suggest that the <3 kDa peptide fractions obtained using alkaline-AK have superior neuroprotective activity and antioxidative activities.
    Keywords:  Apoptosis; Beef; Neuroprotective effect; Peptide; SH-SY5Y cells
    DOI:  https://doi.org/10.1016/j.foodres.2019.03.039
  54. Evid Based Complement Alternat Med. 2019 ;2019 9846325
      High glucose and high fat are important inducements for the development and progression of diabetic cardiopathy. Salvianolic acid B (SAB), which is the most abundant and bioactive compound in Danshen, attenuates oxidative stress-related disorders, such as cardiovascular diseases, cerebral ischemia, and diabetes. However, the effect of SAB on diabetic cardiopathy is not clear. The aim of study was to investigate the effect and the underlying molecular mechanisms of SAB on diabetic cardiopathy in vitro model. The human umbilical vein endothelial (HUVEC) cells were treated with high glucose (HG, 30 mM) or high fat (palmitic acid, PA, 0.75 mM) in the presence or absence of SAB (100, 200, and 400 mg/L) and incubated for 24 h. We found that HG or PA induced apoptosis of HUVEC cells, while treatment with SAB inhibited the apoptosis. We also found that SAB reversed HG- or PA-induced oxidative stress, apoptosis cell cytokines production, and expression of thioredoxin-interacting protein (TXNIP). Moreover, SAB increased HG- or PA-induced expression of Sirtuin 1 (Sirt1), a nicotinamide adenine dinucleotide- (NAD+-) dependent histone deacetylase. Exposure of HUVEC cells to Ex527 (Sirt1 inhibitor) suppressed the effect of SAB on acetyl-p53 and procaspase-3 expressions. In conclusion, the results suggested that SAB could attenuate HUVEC cells damage treated with HG or PA via Sirt1 and might be a potential therapy agent for the diabetic cardiopathy treatment.
    DOI:  https://doi.org/10.1155/2019/9846325
  55. Biotechnol Appl Biochem. 2019 May 22.
      TIGAR (TP53-induced glycolysis and apoptosis regulator) is a p53-inducible gene, and its expression resulted in controlling metabolism and protection from apoptosis. Furthermore, TIGAR participated in promoting the pentose phosphate pathway and helping to lower intracellular reactive oxygen species (ROS). miR-885-5p has also been reported to be involved in liver tumorigenesis, but whether miR-885-5p has a regulatory effect on TIGAR expression is unknown. In this study, we found that their levels were correlated to each other and positively related to cell malignancy. Exogenous miR-885-5p induced TIGAR expression through a p53-independent pathway. The promoter region of TIGAR harbors two tandem putative miR-885-5p target sites. Cotransfection of synthetic miR-885 with TIGAR promoter reporter constructs significantly enhanced TIGAR promoter activity via binding with target sites. Furthermore, miR-885-5p and its precursor pre-miR-885 had the same stimulatory impact on TIGAR expression. ChIP analysis further verified that increased miR-885-5p potentiated the accessibility of TIGAR promoter chromatin to transcriptional factors and facilitated TIGAR expression. miR-885-5p and its precursor both can interact mechanically with TIGAR promoter binding site and alter local chromatin structure, and subsequently upregulate TIGAR expression and participate in liver tumorigenesis. This article is protected by copyright. All rights reserved.
    Keywords:  Hepatocellular carcinoma (HCC); TP53-inducible glycolysis and apoptosis regulator (TIGAR); miR-885-5p; promoter activity; regulatory effect
    DOI:  https://doi.org/10.1002/bab.1767
  56. J BUON. 2019 Mar-Apr;24(2):24(2): 449-455
      PURPOSE: To study the effect of transforming growth factor (TGF)-β1 on apoptosis of colon cancer cells via the ERK signaling pathway.METHODS: Human chemosensitive colon cancer cell line HT- 29 was used in this study. VEGF mRNA and protein level were detected using PCR and western blot. Enzyme-linked immunosorbent assay (ELISA) was used for prostaglandin (PG) detection. Cell proliferation was determined via MTT assay.
    RESULTS: TGF-β1 had a significant effect on blocking the cancer cell growth (p<0.05). TGF-β1 significantly reduced the VEGF mRNA level (p<0.05) and eliminated the COX-2 expression in a dose-dependent manner, while p53 expression was increased (p<0.05). TGF-β1-induced inhibitory effect on COX-2 was significantly eliminated by the ERK inhibitor Compound C (p<0.05). The basal PGE2 production was eliminated in cells treated with TGF-β1 (p<0.05). N-acetylcysteine (NAC) treatment almost completely eliminated the reactive oxygen species (ROS) produced by TGF-β1 and ERK activation. Compared with administration of 5-FU or etoposide alone, TGF-Β1 combined with 5-FU or etoposide significantly administration the viability of colon cancer HT-29 cells.
    CONCLUSION: ERK is a newly-identified cancer target molecule, which can significantly control COX-2 in colon cancer cells treated with TGF-β1.
  57. Free Radic Biol Med. 2019 May 16. pii: S0891-5849(19)30527-1. [Epub ahead of print]
      The natural compound Alternol was shown to induce profound oxidative stress and apoptotic cell death preferentially in cancer cells. In this study, a comprehensive investigation was conducted to understand the mechanism for Alternol-induced ROS accumulation responsible for apoptotic cell death. Our data revealed that Alternol treatment moderately increased mitochondrial superoxide formation rate, but it was significantly lower than the total ROS positive cell population. Pre-treatment with mitochondria specific anti-oxidant MitoQ, NOX or NOS specific inhibitors had not protective effect on Alternol-induced ROS accumulation and cell death. However, XDH/XO inhibition by specific small chemical inhibitors or gene silencing reduced total ROS levels and protected cells from apoptosis induced by Alternol. Further analysis revealed that Alternol treatment significantly enhanced XDH oxidative activity and induced a strong protein oxidation-related damage in malignant but not benign cells. Interestingly, benign cells exerted a strong spike in anti-oxidant SOD and catalase activities compared to malignant cells after Alternol treatment. Cell-based protein-ligand engagement and in-silicon docking analysis showed that Alternol interacts with XDH protein on the catalytic domain with two amino acid residues away from its substrate binding sites. Taken together, our data demonstrate that Alternol treatment enhances XDH oxidative activity, leading to ROS-dependent apoptotic cell death.
    Keywords:  Alternol; Apoptosis; Prostate cancer; ROS; XDH; XOR
    DOI:  https://doi.org/10.1016/j.freeradbiomed.2019.05.019
  58. Free Radic Res. 2019 May 22. 1-211
      Genomic instability is a common feature of tumours that has a wide range of disruptive effects on cellular homeostasis. In this review we briefly discuss how instability comes about, then focus on the impact of gain or loss of DNA (aneuploidy) on oxidative stress. We discuss several mechanisms that lead from aneuploidy to the production of reactive oxygen species, including the effects on protein complex stoichiometry, endoplasmic reticulum stress and metabolic disruption. Each of these are involved in positive feedback loops that amplify relatively minor genetic changes into major cellular disruption or cell death, depending on the capacity of the cell to induce antioxidants or processes such as mitophagy that can moderate the disruption. Finally we examine the direct effects of reactive oxygen species on mitosis and how oxidative stress can compromise centrosome number, cytoskeletal integrity and signalling processes that are vital for mitotic fidelity.
    Keywords:  Aneuploidy; DNA damage; genomic instability; metabolism; oxidative stress; proteotoxic stress; reactive oxygen species (ROS)
    DOI:  https://doi.org/10.1080/10715762.2019.1623398
  59. Mol Med. 2019 May 22. 25(1): 21
      BACKGROUND: Metabolic memory contributes to the development of diabetic retinopathy (DR), which is the complication of diabetes. But it's still unknown how to prevent the metabolic memory to treat the DR. In our study, we want to examine the function of Astragalus polysaccharides (APS) in the metabolic memory of retinal pigment epithelium (RPE) pretreated with high glucose (HG).METHODS: ARPE-19 and PRPE cells were exposed to HG followed by normal glucose (NG) treatment with or without APS. QPCR was used to examine the levels of miR-195 and Bcl-2. MDA and SOD detection assays were used to examine the oxidative stress level. Western blotting and immunostaining were applied to detect the protein level of mitochondrial damage and apoptotic signaling pathway. Flow cytometry and TUNEL staining were used to analyze cell apoptosis. Luciferase assay was used to examine the direct target of miR-195.
    RESULTS: APS treatment significantly decreased the expression of miR-195, while increased the expression of Bcl-2 with optimized dosages which were induced by HG treatment, even after replacing the HG with NG. And we found Bcl-2 was the direct target of miR-195. APS alleviated the oxidative stress, mitochondrial damage and cell apoptosis induced by HG and HG + NG treatments in RPE cells via regulating miR-195. Furthermore, we found overexpression of miR-195 abolished the alleviated effects of APS on the HG-treated RPE cells.
    CONCLUSIONS: APS suppressed high glucose-induced metabolic memory in retinal pigment epithelial cells through inhibiting mitochondrial dysfunction-induced apoptosis by regulating miR-195.
    Keywords:  Apoptosis; Astragalus polysaccharides; Diabetic retinopathy; Metabolic memory; Mitochondrial damage; miR-195
    DOI:  https://doi.org/10.1186/s10020-019-0088-z
  60. Food Chem Toxicol. 2019 May 16. pii: S0278-6915(19)30288-1. [Epub ahead of print]
      Laba garlic is a kind of processed garlic products, it is the traditional Chinese food with a long history. In this study, the antitumor, antioxidant and cytotoxic properties of the blue pigment (BP) from Laba garlic were investigated. N-trans-feruloyltyramine (FLA) was isolated and identified from BP. The protective effects of FLA against H2O2-induced oxidative damages in L02 cells were also assessed. The apoptotic effects of FLA were detected by using flow cytometry analysis. Results showed that the tumor growth was significantly suppressed by BP (P<0.05). BP and FLA exhibited remarkable antioxidant activities. L02 cells pretreatment with FLA could significantly fight against the oxidative damage induced by H2O2, inhibit the morphological changes of mitochondria and maintain the integrity of mitochondria. FLA showed proliferation inhibition on HepG2 cells with IC50 value of 194 ± 0.894 μM. After treatment of FLA (320 μM), the results of MTT assay on HepG2 and L02 cells indicated that FLA had selective cytotoxic effects. It suggested a new way of prevention and treatment of tumors and FLA might be a promising candidate in cancer therapy and functional foods.
    Keywords:  Apoptosis; Laba garlic; N-trans-Feruloyltyramine; Oxidative stress; Selective cytotoxic effects
    DOI:  https://doi.org/10.1016/j.fct.2019.05.021
  61. Environ Res. 2019 May 07. pii: S0013-9351(19)30253-1. [Epub ahead of print]175 117-123
      Titanium dioxide nanoparticles, due to their smaller size and increased surface area comparted to the bulk form, are known to be bioreactive and have unexpected toxicological outcomes. Previous studies have shown that nanoscale titanium dioxide induces reactive oxygen species (ROS)-mediated cytotoxicity and genotoxicity. Although many reports have discussed the ROS-mediated cytotoxic effects of titanium dioxide nanoparticles (TiO2-NPs), their effects on the receptor-ligand association are unknown. In this study, the possibility that TiO2-NPs can interfere with the receptor-ligand binding was assessed by monitoring alterations in the phosphorylation status of proteins downstream of the epidermal growth factor receptor (EGFR) signaling cascade. TiO2-NPs blocked ligand-induced EGFR autophosphorylation, leading to the deactivation of EGFR downstream effectors such as Akt and extracellular signal-regulated kinase signaling, inducing cell death.
    Keywords:  Cytotoxicity; EGFR; Ligand-receptor interaction; Titanium dioxide nanoparticles
    DOI:  https://doi.org/10.1016/j.envres.2019.05.001
  62. J BUON. 2019 Mar-Apr;24(2):24(2): 628-633
      PURPOSE: To investigate the effect of puerarin on the apoptosis of human hepatocellular carcinoma cells induced by hydrogen peroxide and its mechanism.METHODS: Experiments were divided into control group, model group, and puerarin group. Normal saline (200 μmol/L) was used in the control group, 200 μmol/L H2O2 was used to induce oxidative stress in the model group, and 25 μmol/L, 50 μmol/L, and 100 μmol/L puerarin were used in the puerarin group to treat hepatocellular carcinoma SMMC-7721 cells for 24 h on the basis of 200 μmol/L H2O2, respectively. Contents of malondialdehyde (MDA), superoxide dismutase (SOD) and glutathione (GSH) in SMMC-7721 cells were determined by colorimetry. Apoptotic rate of SMMC-7721 cells was determined by flow cytometry.
    RESULTS: Compared with the control group, MDA content in the H2O2 group increased significantly, and SOD activity and GSH content decreased significantly Compared with the control group, SOD activity and GSH content in SMMC-7721 cells of puerarin group decreased significantly (p<0.05). Compared with the H2O2 group, content of MDA in SMMC-7721 cells of the puerarin group decreased significantly, while SOD activity and GSH content (p<0.05) increased significantly. Activity of SOD and content of GSH in SMMC-7721 cells incubated with 50 μmol/L and 100 μmol/L puerarin were significantly higher than that in cells treated with 25μmol/L puerarin (p<0.05). Activity of SOD and content of GSH in SMMC-7721 cells incubated with 100 μmol/L puerarin were significantly higher than those in cells treated with 50 μmol/L puerarin (p<0.05). Apoptosis rate of SMMC-7721 cells incubated with different concentrations of puerarin was significantly lower than that of the H2O2 group (p<0.05).
    CONCLUSION: Puerarin has protective effect on hepatocellular carcinoma SMMC-7721 cells under oxidative stress. It is suggested that puerarin should be carefully used when the proliferation of hepatocellular carcinoma cells results in the production of large amounts of ROS.
  63. Open Access J Ophthalmol. 2018 ;pii: 147. [Epub ahead of print]3(2):
      Diabetic retinopathy (DR) is considered as a chronic eye disease leading to blindness. DR is associated with hyperglycemia-induced oxidative stress, chronic low-grade inflammation and premature cell death. DR affects retinal capillaries, neuroretina and the retinal pigment epithelium. Recently, the thioredoxin-interacting protein TXNIP has been shown as a pro-oxidative stress, pro-inflammatory and pro-apoptotic protein, highly induced by diabetes and high glucose in all cells examined including the retina. TXNIP's actions involve binding to and inhibition of anti-oxidant and thiol-reducing capacities of thioredoxins (Trx) causing cellular oxidative stress and apoptosis. Trx1 is found in the cytosol and nucleus while Trx2 is the mitochondrial isoform. Several studies provided evidence that knockdown of TXNIP by siRNA or chemical blockade ameliorates early abnormalities of DR including endothelial dysfunction, pericyte apoptosis, Müller cell gliosis and neurodegeneration. Therefore, TXNIP is considered a potential target for preventing or slowing down the progression of DR. We recently proposed that nucleic acid constructs containing a proximal TXNIP promoter linked to a redox gene or shRNA that reduces oxidative stress and inflammation may be used to treat DR. The TXNIP promoter is sensitive to hyperglycemia therefore can drive expression of the linked gene or shRNA under high glucose environment such as seen in diabetes while remaining unresponsive at physiological glucose levels. Such a TXNIP-promoter linked gene or shRNA construct can be delivered to the retina by using adeno-associated viral vectors including AAV2 and AAV2/8 or an appropriate carrier via the intravitreal or sub retinal delivery for long-term gene therapies in DR.
    Keywords:  Diabetic retinopathy; Gene therapy; Redox protein; TXNIP promoter; shRNA
    DOI:  https://doi.org/10.23880/oajo-16000147
  64. Acta Histochem. 2019 May 17. pii: S0065-1281(19)30031-5. [Epub ahead of print]
      Due to the broad toxic relevance of acrylamide, many measures have been taken since the 1900s. These measures increased day by day when acrylamide was discovered in foods in 2002, and its toxic spectrum was found to be wider than expected. Therefore, in some countries, the products with higher acrylamide content were restricted. On the other hand, the effects of acrylamide on the respiratory system cells have yet to be well understood. In this study, we aimed at investigating the effect of acrylamide on lung epithelial BEAS-2B cells. Initially, the cytotoxic effect of acrylamide on BEAS-2B was determined by MTT assay. Then, cellular oxidative stress was measured. Flow cytometry analysis was conducted for Annexin-V and caspase 3/7. Furthermore, Bax, Bcl-2 and Nrf-2 proteins were evaluated by immunocytochemistry. Finally, acrylamide-induced cellular morphological changes were observed under confocal and TEM microscopes. According to MTT results, the IC50 concentration of acrylamide was 2.00 mM. After acrylamide treatment, oxidative stress increased dose-dependently. Annexin V-labelled apoptotic cells and caspase 3/7 activity were higher than untreated cells in acrylamide-treated cells. Immunocytochemical examination revealed a marked decrease in Bcl-2, an increase in Bax and Nrf-2 protein staining upon acrylamide treatment. Furthermore, in confocal and TEM microscopy, apoptotic hallmarks were pronounced. In the present study, acrylamide was suggested to display anti-proliferative activity, decrease viability, induce apoptosis and oxidative stress and cause morphological changes in BEAS-2B cells.
    Keywords:  Acrylamide; BEAS-2B cells; Bax; Bcl-2; Caspase 3/7; Nrf-2
    DOI:  https://doi.org/10.1016/j.acthis.2019.05.005
  65. Eur J Med Chem. 2019 May 14. pii: S0223-5234(19)30440-4. [Epub ahead of print]176 175-186
      The development of optically pure drugs is the trend of new drugs research. Searching for optically pure metallodrugs against cancer has not been taken seriously. [CuL4Cl]Cl·2CH2Cl2·H2O (1) and [CuL4Br]Br·2CH2Cl2 (2) (L = 2-amino-5-dehydroabietyl-1,3,4-thiadiazole), two rosin-derivative based optically pure chiral copper(II) complexes, are rationally synthesized as potential anticancer agents. 1 exhibits effective in vitro and in vivo anticancer activities and tolerable toxicities. 1 promotes MCF-7 cell death by combination of cell arrest at G1 phase, apoptosis (both extrinsic and intrinsic apoptotic pathways), anti-metastasis, anti-angiogenesis, damage of DNA, protein and lipid, and autophagy mediated by the oxidative stress which is confirmed by ROS generation and intracellular glutathione depletion assays. 1 can be identified as a lead anticancer molecule of therapeutic importance. This work may offer insights into the design and mechanism study of multifunctional optically pure metal-based anticancer candidates derived from natural products.
    Keywords:  Anti-Metastasis and anti-angiogenesis; Antitumor agents; Apoptosis; Chiral copper(II) complex; Natural products
    DOI:  https://doi.org/10.1016/j.ejmech.2019.05.030
  66. Biomaterials. 2019 May 08. pii: S0142-9612(19)30273-X. [Epub ahead of print]212 73-86
      Given that there is no effective treatment method for lethality androgen-resistant prostate cancers (ARPC), herein we report a multifunctional gold-caged nanoparticle (PTX-PP@Au NPs) against ARPC through integrating functional organic/inorganic materials to exploit the superiors of gold particles such as photothermal effects (PTT), generating reactive oxygen species (photodynamic effects, PDT), carrying chemotherapeutic agents (chemotherapy effects, CT), and inhibiting ion channel. This synergistic PTT/PDT/CT platform consists of three components: i) the Pluronic-polyethylenimine assembling into micelles to encapsulate drugs and providing reduction sites for gold cage formation through a "green" method, ii) the gold cage with surface plasmon resonance peak at near-infrared (NIR) region in a broad window qualifying the PTT/PDT potentiality, iii) a chemotherapeutic agent paclitaxel (PTX) arresting the tumor cell cycle. As demonstrated, the system has remarkable performance on controlling drug release, blocking TRPV6 cation channel, enhancing cell cycle arrest, elevating temperature and generating ROS, thus improving cellular toxicity along with apoptosis, enhancing tumor targeting, and achieving the therapy to ARPC with low toxicity on liver function and minimal side effects to normal organs. Notably, both PTT and PDT effect are generated under single irradiation situation because of the broad absorbance window, along with limited skin damages. As a specific synergistic platform creatively integrating multiple treatment protocols with negative toxicity, PTX-PP@Au NPs provide a facile, effective, and broadly applicable strategy to deadly ARPC.
    Keywords:  Androgen-resistant prostate cancers; Gold cage; Photothermal-photodynamic-chemotherapy; Single irradiation; TRPV6 channel
    DOI:  https://doi.org/10.1016/j.biomaterials.2019.05.009
  67. Drug Des Devel Ther. 2019 ;13 1461-1472
      Background: Random skin flap is frequently used in plastic and reconstructive surgery, but its distal part often occurs ischemia and necrosis. Pravastatin (Prava) with bioactivities of pro-angiogenesis, anti-apoptosis and anti-oxidative stress, may be beneficial for flap survival. Materials and methods: A modified McFarlane flap model was performed in Sprague-Dawley rats. The animals were divided into the Control and Prava groups and treated as follows: the Prava group was injected intraperitoneally with 2 mg/kg Prava for consecutive 7 days, and the Control group received an equal volume of vehicle daily. On day 7, the necrosis skin flaps were observed, while visualization of blood flow below the tissue surface was performed by Laser Doppler blood flow imaging (LDBFI). Then animals were euthanized, and levels of angiogenesis, apoptosis and oxidative stress were analyzed. Results: Prava decreased necrosis and edema of skin flaps compared with the Control group, with more blood flow in the flap under LDBFI. Prava treatment increased the mean vessels density, elevated the expression levels of angiogenic proteins (matrix metallopeptidase 9, vascular endothelial growth factor, Cadherin5) and antioxidant proteins (superoxide dismutase 1 (SOD1), endothelial nitric oxide synthase, heme oxygenase), and decreased the expression of apoptotic factors (BAX, CYC, Caspase3). In addition, malondialdehyde content was reduced, and glutathione level and SOD activity were increased in the skin flaps after treatment with Prava. Conclusion: Prava promotes survival of random skin flap through induction of angiogenesis, and inhibition of apoptosis and oxidative stress.
    Keywords:  Pravastatin; angiogenesis; apoptosis; oxidative stress; random skin flap
    DOI:  https://doi.org/10.2147/DDDT.S195479
  68. Arch Biochem Biophys. 2019 May 18. pii: S0003-9861(19)30243-7. [Epub ahead of print]
      Glaucoma is a major cause of irreversible blindness in the world and filtering surgery is commonly carried out to control intraocular pressure. Failure of filtering surgery is usually due to postoperative scarring, and fibroblast proliferation, collagen production and subconjunctival fibrosis play a prominent role in obstructing aqueous humor from the anterior chamber to the subconjunctival space. Zinc oxide (ZnO) nanoparticles have been widely applied in biomedical fields. However, the influence of ZnO nanoparticles on human tenon fibroblasts (HTFs) is still unclear. In the present study, we first explored the effects of various concentrations of ZnO nanoparticles on HTFs proliferation, reactive oxygen species (ROS) generation, cell cycle arrest, and apoptosis. Further, we determined the changes of transforming growth factor-β (TGF-β1), fibronectin (FN) extra domain A (ED-A), and procollagen I carboxyterminal propeptide (PICP) at mRNA and protein levels, explored the effect of ZnO nanoparticles on the collagen lattice contraction in HTFs. The results indicated that ZnO nanoparticles can efficiently inhibit HTFs proliferation, elevate ROS production level, and induce cell cycle arrest at G2/M phase, leading to HTFs apoptosis. ZnO nanoparticles can also decrease the expressions of TGF-β1, ED-A, and PICP at mRNA and protein levels; significantly prevent fibroblast-mediated collagen lattice contraction. Taken together, ZnO nanoparticles can efficiently ameliorate collagen lattice contraction in HTFs, and may be a promising antifibrotic agent in glaucoma filtration surgery. Our findings provide a new insight on anti-scar formation after glaucoma filtration surgery by using ZnO nanoparticles.
    Keywords:  Collagen lattice contraction; Glaucoma; Human tenon fibroblast; Transforming growth factor-β1; Zinc oxide nanoparticle
    DOI:  https://doi.org/10.1016/j.abb.2019.05.016
  69. Life Sci. 2019 May 17. pii: S0024-3205(19)30386-8. [Epub ahead of print]
      AIMS: Ovarian aging is a natural physiological phenomenon accompanied by follicular atresia as well as the decline of oocyte quality. Moxibustion is a form of traditional Chinese medicine therapy which has been reported to treat many aging-related problems and improve immune defense.MATERIALS AND METHODS: Moxibustion treatment was applied to the 10-month female rats for 2 or 6 months to evaluate whether moxibustion could delay ovarian aging. The expression levels of NQO-1, HO-1, Bax and Bcl-2 were examined by Western blotting. The serum levels of E2 and FSH concentration were measured through ELISA. P21, P16, NQO-1, HO-1, Bax and Bcl-2 were measured by qRT-PCR.
    KEY FINDINGS: We demonstrated that moxibustion treatment could attenuate oxidative stress and apoptosis in ovaries, which lead to ovarian aging. The ovary histomorphology, serum FSH, E2 levels as well as aging markers P21 and P16 expression were compared among the groups, which showed that moxibustion treatment could alleviate the ovary fibrosis, decrease the aging markers expression and increase secretion of ovary functional hormones. The mRNA and protein expression levels of the antioxidative stress-related genes HO-1 and NQO-1 were increased after moxibustion treatment. The antiapoptotic factor Bcl-2 and proapoptotic factor Bax were also detected by qRT-PCR and western blotting, and the results demonstrated that moxibustion significantly downregulated the ratio of Bax/Bcl-2, suggesting that moxibustion could reduce apoptosis in the ovaries of aged rats.
    SIGNIFICANCE: In conclusion, our research revealed that moxibustion could improve ovary function by suppressing apoptosis events and upregulating antioxidant defenses in the natural aging ovary.
    Keywords:  Apoptosis; Moxibustion; Ovarian aging; Oxidative stress
    DOI:  https://doi.org/10.1016/j.lfs.2019.05.040
  70. Int J Mol Sci. 2019 May 21. pii: E2504. [Epub ahead of print]20(10):
      Oxidative stress has been considered as the main mediator in neurodegenerative diseases. A high-fat diet (HFD) and metabolic diseases result in oxidative stress generation, leading to various neurodegenerative diseases via molecular mechanisms that remain largely unknown. Protein kinases play an important role in the homeostasis between cell survival and cell apoptosis. The mammalian sterile 20-like kinase-1 (MST1) protein kinase plays an important role in cellular apoptosis in different organ systems, including the central nervous system. In this study, we evaluated the MST1/c-Jun N-terminal kinase (JNK) dependent oxidative damage mediated cognitive dysfunction in HFD-fed mice and stress-induced hippocampal HT22 (mice hippocampal) cells. Our Western blot and immunofluorescence results indicate that HFD and stress-induced hippocampal HT22 cells activate MST1/JNK/Caspase-3 (Casp-3) signaling, which regulates neuronal cell apoptosis and beta-amyloid-cleaving enzyme (BACE1) expression and leads to impaired cognition. Moreover, MST1 expression inhibition by shRNA significantly reduced JNK/Casp-3 signaling. Our in vivo and in vitro experiments mimicking metabolic stress, such as a high-fat diet, hyperglycemia, and an inflammatory response, determined that MST1 plays a key regulatory role in neuronal cell death and cognition, suggesting that MST1 could be a potential therapeutic target for numerous neurodegenerative diseases.
    Keywords:  JNK; MST1; ROS; apoptosis; high-fat diet; neurodegeneration
    DOI:  https://doi.org/10.3390/ijms20102504
  71. Chem Biol Interact. 2019 May 16. pii: S0009-2797(19)30512-5. [Epub ahead of print]
      Cisplatin, a platinum chemotherapeutic agent, is used in a diversity of malignancies; nevertheless, the excessive nephrotoxicity following cisplatin treatment is the dose-limiting devastating reaction. This study was designed to explore the possible nephroprotective impact of wogonin, a forceful anti-oxidant, anti-inflammatory, and anti-tumor agent, in a rat model of cisplatin-induced renal injury. The potential nephroprotective mechanisms were additionally investigated. Wogonin was given at a dose of 40 mg/kg. Acute nephrotoxicity was indicated by a significant rise in BUN, and serum creatinine levels in cisplatin-injected rats. Also, cisplatin enhanced the lipid peroxidation, diminished GSH, catalase, and PPAR-γ levels. Additionally, cisplatin-injected rats showed a significant rise in tissue levels of IL-1β, TNF-α, NF-kB and caspase-3 enzymatic activity. Notably, the pre-treatment with wogonin ameliorated the nephrotoxicity indices, oxidative stress, inflammation, and apoptosis induced by cisplatin. Also, wogonin up-regulated PPAR-γ expression. The involvement of Wnt/β-catenin pathway was debatable; however, our findings showed that it was significantly induced by cisplatin. Wogonin pre-treatment markedly attenuated Wnt/β-catenin pathway. Collectively, these findings imply that wogonin is a promising nephroprotective agent that improves the therapeutic index of cisplatin via reducing oxidative stress, inflammation as well as inducing PPAR-γ. Also, Wnt/β-catenin pathway is partially involved in the pathogenesis of cisplatin nephrotoxicity.
    Keywords:  Cisplatin; Nephrotoxicity; PPAR-γ; Wnt/β-catenin pathway; Wogonin
    DOI:  https://doi.org/10.1016/j.cbi.2019.05.029
  72. Chem Biol Drug Des. 2019 May 20.
      Several novel cycloalkyl-fused 2,3-diaryl pyrazole derivatives were designed, synthesized and evaluated as potential anti-tubulin agents. Compound A10 exhibited the most potent anti-proliferative activity against a panel of cancer lines (IC50 = 0.78 ~ 2.42 μM) and low cytotoxicity against 293T & L02 (CC50 values of 131.74 μM and 174.89 μM, respectively). Moreover, A10 displayed inhibition of tubulin polymerization in vitro, arrested the G2/M phase of the cell cycle, changed morphology of tubulin, increased intracellular reactive oxygen species (ROS) and induced apoptosis of HeLa cells. Docking simulation and 3D-QSAR models were performed to elaborate on the anti-tubulin mechanism of the derivatives. The inhibition of monoclonal colony formation provided more intuitional data to verify the possibility of A10 as a novel tubulin assembling inhibitor. This article is protected by copyright. All rights reserved.
    Keywords:  3D-QSAR; Cell cycle arrest; Cycloalkyl-fused pyrazole; Molecular docking; Tubulin polymerization inhibitors
    DOI:  https://doi.org/10.1111/cbdd.13565
  73. Biomed Res Int. 2019 ;2019 4690280
      Background/Aims: Alzheimer's disease (AD) is the most common neurodegenerative disease, and all researchers working in this field agree that oxidative stress is intimately associated with Alzheimer disease. In this study, we hypothesized that glutamine (Gln) offers protection against oxidative stress injury in SAMP8 mice as well as the underlying mechanism.Methods: The SAMP8 mice received glutamine intragastrically for 8 consecutive weeks to evaluate the protective effect of glutamine on oxidative stress in AD mice involving Wnt3a/β-catenin signaling pathway. In addition, rat pheochromocytoma tumor cell line PC12 was pretreated with 32 μM glutamine for 2 h followed by 24 h incubation with 40 μM Aβ25-35 to obtain in vitro data.
    Results: In vivo the administration of glutamine was found to ameliorate behavioral deficits and neuron damage, increase superoxide dismutase (SOD) and glutathione peroxidase (GSH-XP) activity, reduce the malondialdehyde (MDA) content, and activate the Wnt3a/β-catenin signaling pathway in SAMP8 mice. In vitro glutamine treatment decreased the toxicity of Aβ25-35 on PC12 cells and prevented apoptosis. Additionally, glutamine treatment increased SOD and GSH-XP activity and decreased MDA content and increased Wnt3a and β-catenin protein levels. Interestingly, the DKK-1 (Wnt3a/β-catenin pathway inhibitor) decreased the antioxidant capacity of glutamine in Aβ25-35-treated PC12 cells.
    Conclusion: This study suggests that glutamine could protect against oxidative stress-induced injury in AD mice via the Wnt3a/β-catenin signaling pathway.
    DOI:  https://doi.org/10.1155/2019/4690280
  74. Bratisl Lek Listy. 2019 ;120(5): 336-343
      OBJECTIVE: Methamphetamine in low doses can increase vigilance and power and at high doses has destructive effects that cause toxicity and death of various cell lines and affect the central nervous system. Morphine has also protective properties, which were observed in low concentrations, for nerve cells and also seem to have the ability to reduce cell death in neural cell lines.MATERIALS AND METHODS: In this study, we used PC12 and U87 cell lines, which grew in DMEM culture media. Assays used in this study are listed below: MTT test for cell viability detection, LDH test for cytotoxicity measurement, caspase activity colorimetric assay kit (Bio-techne) for caspase 3 activity diagnosis, Rhodamine 123 for Detection of mitochondrial membrane potential. TUNNEL test for DNA fragmentation, fura-2 for Measurement of (Ca2+) ic and (Ca2+) m. fluorescence microscope for measurement of antioxidant enzyme activities.
    RESULTS: morphine increased cell viability and the rhodamine-123 absorbance. It reduced cell cytotoxicity, caspase 3 activity, ic (et) m Ca2+ concentration, (.OH) generation, and DNA fragmentation in all concentrations of 1 pM t0 100 nM (p < 0.05) by optimal concentration of 1 pM.
    CONCLUSION: morphine as a pain mediator can reduce the methamphetamine-induced cell death, may be due to its anti-inflammatory properties (Fig. 7, Ref. 52).
    Keywords:  apoptosis cell death.; methamphetamine; morphine
    DOI:  https://doi.org/10.4149/BLL_2019_055
  75. Arq Bras Oftalmol. 2019 May 20. pii: S0004-27492019005008104. [Epub ahead of print]
      PURPOSE: We examined the effect of intracameral administration of cefuroxime on oxidative stress and endothelial apoptosis in rat corneal tissue.METHODS: In total, 30 rats were divided into 3 groups of 10 rats each (intracameral administration of cefuroxime 0.1 mg/0.01 mL (cefuroxime group); intracameral administration of balanced salt solution 0.01 mL (control group); or absence of intracameral injection (sham group). Corneal endothelial apoptosis was assessed by immunohistochemical analysis using caspase-3 and caspase-8. Total oxidant status, total antioxidant status, oxidative stress index, and paraoxonase and arylesterase levels were examined in corneal endothelial tissue and serum.
    RESULTS: Paraoxonase levels in the serum were significantly different between the sham and cefuroxime groups (p=0.027). A significant difference was also observed in total oxidant status levels between the cefuroxime and balanced salt solution groups (p=0.023). In addition, there were significant differences in total antioxidant status levels in corneal tissue between the cefuroxime and sham groups (p<0.001) and between the cefuroxime and balanced salt solution groups (p<0.001). Furthermore, significant differences were also observed in oxidative stress index levels between the cefuroxime and balanced salt solution groups (p=0.001) and between the cefuroxime and sham groups (p=0.026). According to the immunohistochemical staining results, a significant association with caspase-3 activity existed between the cefuroxime and balanced salt solution groups (p=0.007), while no significant difference was found with caspase-8 activity (p=0.541). Caspase-3 activity exhibited a significant relationship between the sham and balanced salt solution groups (p=0.018), but no relationship was found with caspase-8 activity (p=0.623).
    CONCLUSION: Immunohistochemical examination revealed that intracameral cefuroxime increased apoptosis when compared to the sham and balanced salt solution groups. Moreover, intracameral cefuroxime increased oxidative stress in the cornea and simultaneously induced apoptosis.
    DOI:  https://doi.org/10.5935/0004-2749.20190067
  76. Chemosphere. 2019 May 06. pii: S0045-6535(19)30939-7. [Epub ahead of print]230 182-189
      Spinosad is one of the most extensively used bio-pesticide in the world. The effects of pesticide in human health are mainly associated with its residue in food or occupational exposure in agricultural production. The lung is the direct target of pesticides exposure, although the study of inhalation damage caused by Spinosad remains unclear. The aim of the present study was to evaluate the cytotoxic effects of the Spinosad in human lung cells. We demonstrated that Spinosad could inhibite the proliferation of human lung epithelial A549 cells, induce the DNA damage and enhance the programmed cell death. Intracellular biochemical assay indicated that DNA double strand breaks, cleaved of PARP, release of cytochrome c, decrease of mitochondrial membrane potential, generation of reactive oxygen species (ROS), activation of caspase-3/9, increase of Bax/Bcl-2 ratio, LC3-II conversion, accumulation of Beclin-1, degradation of p62 and the changes in the phosphorylation of AMPK, mTOR are contributed to the toxic effects of Spinosad in A549 cells. The results showed that the cytotoxicity of Spinosad may be associated with the activity of mitochondrial apoptotic pathways or AMPK/mTOR-mediated autophagy. Meanwhile, the DNA stand breaks caused by the Spinosad suggest it has a potential genotoxic effects on human lung cells. We conclude that Spinosad has a potential risk to human health by inducing the cytotoxic effects.
    Keywords:  AMPK/mTOR-mediated autophagy; Cytotoxicity; DNA damage; Human epithelial A549 lung cells; Spinosad
    DOI:  https://doi.org/10.1016/j.chemosphere.2019.05.042
  77. Cryobiology. 2019 May 16. pii: S0011-2240(19)30058-6. [Epub ahead of print]
      Although vitrification is the current routine method for human embryo cryopreservation, it may cause detrimental effects. The aim of this study was to evaluate the effect of supplementing in vitro culture (IVC) media and/or vitrification solutions (VS) with Resveratrol on the presence of apoptotic markers, reactive oxygen species (ROS) level, glutathione (GSH) content and relative gene abundance. Abattoir-derived oocytes were matured and fertilized in vitro according to a standard procedure. Zygotes were cultured in IVC medium supplemented with or without 0.5 μM Resveratrol (CR, C- respectively). On day 7, blastocysts were vitrified using the minimum volume vitrification method supplementing VS with (C-VR, CRVR) or without (C-V-, CRV-) 0.5 μM Resveratrol. After warming, embryonic quality parameters were evaluated. Survival rates were significantly lower in CRVR group compared with CRV- group, but no differences in hatching rate were observed between groups. Vitrification/warming process did not alter total cell number or the presence of apoptotic or dead cells, but CRV- and CRVR groups presented a significant increase in dead cells (P < 0.05 by ANOVA). Resveratrol supplementation in VS (C-VR) restored GSH content (P < 0.05) to the level found in the CR group. Vitrification/warming process significantly increased the expression of FOXO3A, PNPLA2, BCL2L1 and BAX genes (P < 0.05). Resveratrol addition to IVC medium or VS partially compensated this increase for FOXO3A and PNPLA2 (P < 0.05) but not for BCL2L1 and BAX. In conclusion, supplementation of IVC media or VS with 0.5 μM resveratrol may help embryos to partially restore the initial quality they had before the cryopreservation process.
    Keywords:  Antioxidant; Apoptosis; Blastocyst; Cryopreservation; Gene expression; Oxidative stress
    DOI:  https://doi.org/10.1016/j.cryobiol.2019.05.008
  78. Int J Biol Macromol. 2019 May 20. pii: S0141-8130(19)32469-9. [Epub ahead of print]
      We have previously reported that chondroitin sulfate extracted from Sturgeon bone (CSSB) can alleviate the pain caused by osteoarthritis (OA) by reducing the expression of matrix metalloproteinases (MMPs) and inflammatory factors (IL-1, TNF-α and PGE2). However, the mechanism of CSSB for treating OA remains unclear. As is generally accepted, these proteins and factors are regulated by chondrocyte apoptosis in OA. Thus, it is reasonable to hypothesize whether CSSB can inhibit cartilage degradation via regulating the process of apoptosis. In order to address this problem, chondrocytes were isolated and exposed to 1 mM hydrogen peroxide (H2O2)-treated to induce chondrocytes apoptosis for mimicking the OA microenvironment, and co-incubated with different concentration of CSSB (0, 50, 100, 200 and 400 μg/ml). Results showed that CSSB could increase cell survival, decrease DNA fragment rate and apoptosis rate, also protect mitochondrial and decrease the expression of caspase-3 and caspase-9. These results showed that CSSB ameliorates cartilage matrix damage via inhibiting chondrocytes apoptosis.
    Keywords:  Apoptosis; Chondrocytes; Chondroitin sulfate; Osteoarthritis; Sturgeon bon
    DOI:  https://doi.org/10.1016/j.ijbiomac.2019.05.110
  79. Chembiochem. 2019 May 21.
      IridiumIII (IrIII) complexes have attracted more and more attention because of their potential antineoplastic activity for the past few years. In this study, four IrIII complexes of the type [(η5-Cpx)Ir(N^N)Cl]PF6 (1 and 2) and [Ir(Phpy)2(N^N)]PF6 (3 and 4) have been synthesized and characterized. Complexes exhibit potential antineoplastic activity towards A549 cells, especially for complex 1 (IC50: 3.56±0.5 µM), which was nearly six times of cis-platin (21.31±1.7 µM). Additionally, these complexes show some selectivity for cancer cells versus normal cells. Complexes could be transported by serum albumin (binding constant changed from 0.37~81.71×105 M-1). IrIII complexes (1 and 2) could catalyze the change of NADH to NAD+ (TONs: 43.2, 11.9) and induce the accumulation of reactive oxygen species, which confirmed the antineoplastic mechanism of oxidation, while cyclometalated complexes (3 and 4) could target the lysosome (PCC: 0.73), lead to lysosomal damage and induce apoptosis. Understanding the mechanism of action would help further structure-activity optimization on these novel IrIII complexes as emerging cancer therapeutics.
    Keywords:  Anticancer; Iridium complex; Organometallic; Tetraphenylethylene
    DOI:  https://doi.org/10.1002/cbic.201900268
  80. Cancers (Basel). 2019 May 21. pii: E702. [Epub ahead of print]11(5):
      One of the most promising photosensitizers (PS) used in photodynamic therapy (PDT) is the porphyrin derivative 5,10,15,20-tetra(m-hydroxyphenyl)chlorin (mTHPC, temoporfin), marketed in Europe under the trade name Foscan®. A set of five human cancer cell lines from head and neck and other PDT-relevant tissues was used to investigate oxidative stress and underlying cell death mechanisms of mTHPC-mediated PDT in vitro. Cells were treated with mTHPC in equitoxic concentrations and illuminated with light doses of 1.8-7.0 J/cm2 and harvested immediately, 6, 24, or 48 h post illumination for analyses. Our results confirm the induction of oxidative stress after mTHPC-based PDT by detecting a total loss of mitochondrial membrane potential (Δψm) and increased formation of ROS. However, lipid peroxidation (LPO) and loss of cell membrane integrity play only a minor role in cell death in most cell lines. Based on our results, apoptosis is the predominant death mechanism following mTHPC-mediated PDT. Autophagy can occur in parallel to apoptosis or the former can be dominant first, yet ultimately leading to autophagy-associated apoptosis. The death of the cells is in some cases accompanied by DNA fragmentation and a G2/M phase arrest. In general, the overall phototoxic effects and the concentrations as well as the time to establish these effects varies between cell lines, suggesting that the cancer cells are not all dying by one defined mechanism, but rather succumb to an individual interplay of different cell death mechanisms. Besides the evaluation of the underlying cell death mechanisms, we focused on the comparison of results in a set of five identically treated cell lines in this study. Although cells were treated under equitoxic conditions and PDT acts via a rather unspecific ROS formation, very heterogeneous results were obtained with different cell lines. This study shows that general conclusions after PDT in vitro require testing on several cell lines to be reliable, which has too often been ignored in the past.
    Keywords:  apoptosis; autophagy; cell cycle; mTHPC; necrosis; oxidative stress; photodynamic therapy
    DOI:  https://doi.org/10.3390/cancers11050702
  81. J Cell Biochem. 2019 May 19.
      Cardiotoxicity by doxorubicin hampers its therapeutic potential as an anticancer drug, but mechanisms leading to cardiotoxicity remain contentious. Through this study, the functional contribution of insulin-like growth factor receptor type II α (IGF-IIRα) which is a novel stress-inducible protein was explored in doxorubicin-induced cardiac stress. Employing both in vitro H9c2 cells and in vivo transgenic rat models (SD-TG [IGF-IIRα]) overexpressing IGF-IIRα specifically in heart, we found that IGF-IIRα leads to cardiac structural abnormalities and functional perturbations that were severely aggravated by doxorubicin-induced cardiac stress. Overexpression of IGF-IIRα leads to cumulative elevation of stress associated cardiac hypertrophy and apoptosis factors. There was a significant reduction of survival associated proteins p-Akt and estrogen receptor β/α, and abnormal elevation of cardiac hypertrophy markers such as atrial natriuretic peptide, cardiac troponin-I, and apoptosis-inducing agents such as p53, Bax, and cytochrome C, respectively. IGF-IIRα also altered the expressions of AT1R, ERK1/2, and p38 proteins. Besides, IGF-IIRα also increased the reactive oxygen species production in H9c2 cells which were markedly aggravated by doxorubicin treatment. Together, we showed that IGF-IIRα is a novel stress-induced protein that perturbed cardiac homeostasis and cumulatively exacerbated the doxorubicin-induced cardiac injury that perturbed heart functions and ensuing cardiomyopathy.
    Keywords:  Bax; IGF-IIRα; cardiotoxicity; doxorubicin; p53; transgenic rats
    DOI:  https://doi.org/10.1002/jcb.28957