bims-aporos Biomed News
on Apoptosis and reactive oxygen species
Issue of 2019‒01‒06
38 papers selected by
Gavin McStay
Staffordshire University


  1. Apoptosis. 2019 Jan 02.
      Melanoma is an aggressive form of skin carcinoma, highly resistant to traditional therapies. Photodynamic therapy (PDT) is a non-invasive therapeutic procedure that can exert a selective cytotoxic activity toward malignant cells. In this work we evaluated the effect of a cationic zinc(II) phthalocyanine (Pc13) as photosensitizer on a panel of melanoma cells. Incubation with Pc13 and irradiation induced a concentration and light dose-dependent phototoxicity. In order to study the mechanism underlying Pc13-related cell death and to compare the effect of different doses of PDT, the most sensitive melanoma B16F0 cells were employed. By confocal imaging we showed that Pc13 targeted lysosomes and mitochondria. After irradiation, a marked increase in intracellular reactive oxygen species was observed and a complete protection from Pc13 phototoxicity was reached in the presence of the antioxidant trolox. Acridine orange/ethidium bromide staining showed morphological changes indicative of both apoptosis and necrosis. Biochemical hallmarks of apoptosis, including a significant decrease in the expression levels of Bcl-2, Bcl-xL and Bid and mitochondrial membrane permeabilization, were observed at short times post irradiation. The consequent release of cytochrome c to cytosol and caspase-3 activation led to PARP-1 cleavage and DNA fragmentation. Simultaneously, a dose dependent increase of lactate dehydrogenase in the extracellular compartment of treated cells revealed plasma membrane damage characteristic of necrosis. Taken together, these results indicate that a dual apoptotic and necrotic response is triggered by Pc13 PDT-induced oxidative stress, suggesting that combined mechanisms of cell death could result in a potent alternative for melanoma treatment.
    Keywords:  Apoptosis; Cationic phthalocyanine; Mitochondrial membrane permeabilization; Necrosis; Photodynamic therapy; Reactive oxygen species
    DOI:  https://doi.org/10.1007/s10495-018-01512-w
  2. J Biochem Mol Toxicol. 2018 Dec 31. e22264
      Butylene fipronil (BFPN) is a phenylpyrazole insecticide, acting at the γ-aminobutyric acid (GABA) receptor. Here, we show that BFPN inducedcytotoxicity in PC12 murinenervous cells, which lacks GABA receptor. Treatment with BFPN for 48 hours significantly enhanced G0/G1 arrest and induced apoptosis. BFPN decreased the expression of cyclin-dependent kinase (CDK4 and CDK6) and increased P16 and cyclin D1. Simultaneously, Bcl-2 protein was declined while Bax and cytochrome c were significantly enhanced in BFPN-treated groups. The apoptotic enzymes caspase-8, -9, and -3 were also activated by BFPN. Furthermore, treatment with BFPN significantly stimulated reactive oxygen species (ROS) generation, and pretreatment with antioxidant diphenyleneiodonium, substantially reduced cell death. Overall, these results suggest that BFPN is effective to induce G0/G1-phase arrest and apoptosis in PC12 murine nervous cell. Stimulating ROS generation and activation of P16-CDK4/6-cyclin D1 and mitochondrial apoptotic pathway may participate in the cytotoxicity of BFPN.
    Keywords:  G0/G1-phase arrest; PC12 cells; apoptosis; butylene fipronil; reactive oxygen species
    DOI:  https://doi.org/10.1002/jbt.22264
  3. Res Pharm Sci. 2018 Dec;13(6): 557-565
      Galectin-9 (Gal-9), a member of animal lectins' family, is implicated in the induction of apoptosis in various cancer cells. Here, we evaluated the anti-tumor effect of Gal-9 in OVCAR-3 ovarian cancer cells. The effect of the Gal-9 on cell viability was evaluated using MTT assays. Apoptosis was assessed using Annexin-V staining. The assessment of mitochondrial membrane potential (ΔΨm) was performed using a JC-1 probe. The activity of caspase-3 and caspase-6 were evaluated with colorimetric assay. The production of reactive oxygen species (ROS) was applied by fluorescent probe. The expression levels of Bax and Bcl-2 were assessed using western blotting. The result showed that Gal-9 inhibits cell viability. Flow cytometry analysis showed that Gal-9 induces apoptosis in ovarian cancer cells. Moreover, Gal-9 decreased ΔΨm and increased the generation of ROS and caspase-3 and caspase-6 activities in ovarian cancer cells. Moreover, Gal-9 induced expression of Bax as well as inhibited expression of Bcl-2. In conclusion, our results indicated that Gal-9 induced apoptosis in ovarian cancer cells through mitochondrial pathway.
    Keywords:  Apoptosis; Gal-9; OVCAR-3; Ovarian cancer; Reactive oxygen species
    DOI:  https://doi.org/10.4103/1735-5362.245967
  4. Molecules. 2018 Dec 28. pii: E96. [Epub ahead of print]24(1):
      We investigated the antitumor activity and action mechanism of MHY440 in AGS human gastric cancer cells. MHY440 inhibited topoisomerase (Topo) Ι activity and was associated with a DNA damage response signaling pathway. It exhibited a stronger anti-proliferative effect on AGS cells relative to Hs27 human foreskin fibroblast cells, and this effect was both time- and concentration-dependent. MHY440 also increased cell arrest in the G2/M phase by decreasing cyclin B1, Cdc2, and Cdc25c, and upregulating p53 and p73. MHY440 induced AGS cell apoptosis through the upregulation of Fas-L, Fas, and Bax as well as the proteolysis of BH3 interacting-domain death agonist and poly(ADP-ribose) polymerase. It also contributed to the loss of mitochondrial membrane potential. The apoptotic cell death induced by MHY440 was inhibited by pretreatment with Z-VAD-FMK, a pan-caspase inhibitor, indicating that apoptosis was caspase-dependent. Moreover, the apoptotic effect of MHY440 was reactive oxygen species (ROS)-dependent, as evidenced by the inhibition of MHY440-induced PARP cleavage and ROS generation via N-acetylcysteine-induced ROS scavenging. Taken together, MHY440 showed anticancer effects by inhibiting Topo I, regulating the cell cycle, inducing apoptosis through caspase activation, and generating ROS, suggesting that MHY440 has considerable potential as a therapeutic agent for human gastric cancer.
    Keywords:  MHY440; apoptosis; cell cycle arrest; gastric cancer cells; topoisomerase inhibitor
    DOI:  https://doi.org/10.3390/molecules24010096
  5. Exp Mol Pathol. 2018 Dec 27. pii: S0014-4800(18)30324-1. [Epub ahead of print]
      Glaucoma is a chronic neurodegenerative disease which produces damage to the optic nerve and causes sightlessness. Current remains lack of effective method for glaucoma. Lycium barbarum polysaccharides (LBPs) have pleiotropic effects on various diseases. However, the effect of LBPs on glaucoma remains unclear. The study aimed to clarify the protective effect of LBPs against hydrogen peroxide (H2O2)-induced oxidative damage in human trabecular meshwork (HTM) cells. HTM cells were exposed to H2O2 (0-400 μM) for 24 h to construct an oxidative damage model. Then, the different concentrations of LBPs (0-500 μg mL-1) were used to pre-treated HTM cells, and cell viability, apoptosis, protein levels of pro-/cleaved-caspase-3 and pro-/cleaved-caspase-9, and reactive oxygen species (ROS) generations were detected. MicroRNA (miR)-4295 inhibitor and its control were transfected into HTM cells, and the biological functions of miR-4295 were assessed in H2O2 and LBPs treated cells. Phosphatidylinositol 3-kinase (PI3K)/protein Kinase B (AKT) and extracellular regulated protein kinases (ERK) pathways were determined by western blot assay. LBPs significantly promoted cell viability, reduced apoptosis, declined cleaved-caspase-3/-9 and ROS level in HTM cells after H2O2 administration. MiR-4295 expression was up-regulated in H2O2 and LBPs treated cells. The protective effect of LBPs on H2O2-injured HTM cells was obviously reversed by miR-4295 inhibition. LBPs activated PI3K/AKT and ERK signaling pathways through up-regulation of miR-4295 in H2O2-injured HTM cells. These data demonstrated that LBPs alleviated H2O2-induced injury by up-regulation of miR-4295 in HTM cells, indicating the protective effect of LBPs on HTM cells against oxidative damage.
    Keywords:  ERK; Glaucoma; Lycium barbarum polysaccharides (LBPs); PI3K/AKT; microRNA-4295
    DOI:  https://doi.org/10.1016/j.yexmp.2018.12.007
  6. Transfusion. 2019 Jan 04.
      BACKGROUND: Platelet storage is often complicated by deleterious changes that are started by reversible activation of the cells and can lead to procoagulant function and apoptosis during longer periods of storage. Given that increasing levels of reactive oxygen species (ROS) generation are associated with platelet activation and apoptosis, our study investigated whether ROS scavenging or the inhibition of ROS production can enhance the viability of stored platelets.METHODS: For this study platelet-rich plasma platelet concentrates (PRP-PCs) were either treated with ROS-reducing agents (1 mM N-acetyl-L -cysteine [NAC] or 30 μM NADPH oxidase [NOX] inhibitor, VAS2870) or kept untreated during storage. P-selectin expression, phosphatidylserine (PS) exposure, levels of microparticle (MP) formation, and intraplatelet caspase activity of PCs were analyzed by flow cytometry during 7 days of storage while the platelet viability was also evaluated by MTT assay.
    RESULTS: Both NAC- and VAS2870-treated platelets had significantly lower caspase activity, MP formation, and PS exposure during storage while they also showed improved viability. The platelet count and mean platelet volume (MPV) were also better preserved in the presence of NAC.
    CONCLUSION: Our results confirmed that either the inhibition of ROS generation or the scavenging of these oxidant agents can attenuate platelet apoptosis while improving their viability during storage. In this study, the significant improvement of platelet viability obtained by NAC suggested that its supplementation with a designated safe concentration into PCs may better preserve the quality of these products, especially for longer storage.
    DOI:  https://doi.org/10.1111/trf.15114
  7. Reprod Toxicol. 2018 Dec 28. pii: S0890-6238(18)30578-1. [Epub ahead of print]84 49-58
      4-Methylbenzylidene-camphor (4-MBC) is an estrogenic compound used in a variety of personal care products and is associated with water pollution. In this study, we verified that exposure to 4-MBC suppresses the proliferation and invasiveness of the HTR8/SVneo human trophoblast cell line. Moreover, HTR8/SVneo cells treated with 4-MBC underwent apoptosis with increased DNA fragmentation. 4-MBC also activated the PI3K/AKT and ERK1/2 signaling pathways in HTR8/SVneo cells. Furthermore, 4-MBC induced oxidative stress mediated by reactive oxygen species production, which was associated with HTR8/SVneo cell death. 4-MBC promoted lipid peroxidation and loss of mitochondrial membrane potential in HTR8/SVneo cells and activated the expression of genes encoding a protein expressed on the surface of human trophoblast cells, including the EPH receptor B4 and G protein-coupled receptor 56 genes. Therefore, 4-MBC may retard the normal growth and survival of human trophoblast cells and may hamper normal placental formation during early pregnancy.
    Keywords:  4-MBC; Apoptosis; Proliferation; ROS; Trophoblast
    DOI:  https://doi.org/10.1016/j.reprotox.2018.12.011
  8. J Biochem Mol Toxicol. 2018 Dec 31. e22277
      Although antibiotics are generally well tolerated, their toxic effects on the central nervous system have been gained attention. In this study, we systematically investigated the neuron toxicity of antibiotics from six different classes. We show that clinically relevant concentrations of metronidazole, tigecycline, azithromycin and clindamycin but not ampicillin or sulfamethoxazole induce apoptosis of human primary neuron cells and lines. Notably, tigecycline, azithromycin and clindamycin cause neuron cell oxidative damage whereas metronidazole has no effect on reactive oxygen species (ROS) production, suggesting that metronidazole induces neuron death via ROS-independent mechanism. Tigecycline, azithromycin and clindamycin induce mitochondrial dysfunctions via targeting different mitochondrial respiratory complexes, leading to mitochondrial membrane potential disruption and energy crisis. The deleterious effects of antibiotics are reversed by pretreatment of neuron cells with antioxidant. Our work highlights the different influences of antibiotics on mitochondrial dysfunction, oxidative damage and cytotoxicity in neuron cells. We also provide a strategy to prevent the neurotoxicity.
    Keywords:  antibiotics; mitochondrial dysfunction; neuron disease; oxidative damage
    DOI:  https://doi.org/10.1002/jbt.22277
  9. Mol Pharm. 2019 Jan 02.
      The excessive increase of intracellular reactive oxygen species (ROS) makes tumor cells usually in the state of oxidative stress. Although tumor cells can adapt to this state to a certain extent by upregulating antioxidant systems, the further ROS insults disrupt the transient intracellular redox balance, eventually leading to apoptosis and necrosis. Therefore, increasing the intracellular ROS level can effectively amplify the oxidative stress and induce apoptosis, which can be employed as a strategy for tumor treatment. Herein, a unique pH-responsive ROS inducing micellar system was reported in this study to specifically amplify the ROS signal in tumor cells. This micellar system was constructed by a new amphiphilic polymer, PIAThydCA, composed of poly (itaconic acid) (PIA) as the hydrophilic backbone, D-α-tocopherol polyethylene glycol 1000 succinate (TPGS) as the hydrophobic side chain, and cinnamaldehyde (CA) as the ROS-generating agent which were linked to PIA by the pH-sensitive hydrazone bond. PIAThydCA micelles could be degraded in the intracellular acidic environment through the hydrolysis of hydrazone bond and release CA. CA and TPGS could amplify oxidative stress cooperatively to kill MCF-7 human breast cells preferentially through the mitochondrial apoptosis pathway. Therefore, we anticipate that the PIAThydCA micelles could exert great potential in anti-cancer therapy.
    DOI:  https://doi.org/10.1021/acs.molpharmaceut.8b00973
  10. Adv Pharm Bull. 2018 Nov;8(4): 705-713
      Purpose: Manganism is a cognitive disorder take places in peoples are exposed to environmental manganese pollution. Overexposure to manganese ion (Mn2+) mainly influences central nervous system and causes symptoms that increase possibility of hippocampal damages. Methods: In this study rats were administrated by two different doses of MnCl2 and behavioral and physiological consequences were evaluated. We also investigated effects of E. Amoenum on Mn2+-imposed toxicity by behavioral, biochemical, immunoblotting and histological studies on hippocampus tissue. Results: Results showed metal overexposure increases oxidative stress mainly by lipid peroxidation and reactive oxygen species overproduction. Histological studies and caspase 3 analyses by immunoblotting revealed Mn2+ induced apoptosis from mitochondrial-dependent pathway in the presence of low metal dose. This study provides evidence that oral administration of E. amoenum extract inhibited manganese neurotoxicity by oxidative stress attenuation and apoptosis reduction that lead to improved depression like behavior. Plant extract also increased catecholamine content in Mn2+ treated hippocampus. Conclusion: As molecular and pathophysiological effects of E. amoenum, it could be considered as a pre-treatment for Parkinson and Parkinson like disorders in high-risk people.
    Keywords:  Catecholamine; Cognitive disorder; Depression like behavior; Hippocampus; Manganism; Mitochondria dysfunction
    DOI:  https://doi.org/10.15171/apb.2018.079
  11. Microvasc Res. 2018 Dec 27. pii: S0026-2862(18)30178-X. [Epub ahead of print]123 62-67
      Oxidative stress, which is defined as an imbalance between proxidant and antioxidant systems, is the essential mechanism involving in the ischemic process. During the early stage of brain ischemia, reactive oxygen species (ROS) are increased. Increased ROS are thought of a consequence of brain ischemia and exacerbating disease due to inducing cell death, apoptosis and senescence by oxidative stress. During brain tissue repair, ROS are act as signaling molecules and may be benefical for regulating angiogenesis and preventing tissue injury. New blood vessel formation is essentially required for rescuing tissue from brain ischemia. In ischemic conditions, ROS promotes angiogenesis, either directly or via the generation of active oxidation products. ROS-induced angiogenesis involves several signaling pathways. This paper reviewed current understanding of the role of ROS as a mediator and modulator of angiogenesis in brain ischemia.
    Keywords:  Angiogenesis; Brain ischemia; NADPH oxidases; Oxidative stress; Reactive oxygen species
    DOI:  https://doi.org/10.1016/j.mvr.2018.12.005
  12. J BUON. 2018 Nov-Dec;23(6):23(6): 1679-1685
      PURPOSE: To evaluate the anticancer effects and the underlying mechanism of arglabin on oral squamous cell carcinoma (OSCC) cells.METHODS: 4',6-Diamidino-2-phenylindole dihydrochloride (DAPI) and annexin V/propidium iodide (PI) staining were performed to evaluate apoptosis. Reactive oxygen species (ROS) levels and mitochondrial membrane potential (MMP) were examined by flow cytometry. Protein expression was assessed by western blot analysis. To examine the anticancer activity of arglabin in vivo, subcutaneous xenografts in nude mice were evaluated.
    RESULTS: Arglabin exhibited an IC50 of 10 µM in OSCC cells and induced apoptosis by inhibiting MMP and enhancing intracellular ROS levels. DAPI and annexin V/PI staining indicated apoptosis of OSCC cells induced by arglabin. Arglabin also downregulated the expression of key proteins in the mTOR/PI3K/Akt signaling pathway. In vivo evaluation showed that arglabin reduced the average tumor volumes and growth of xenografted tumors, indicative of its anticancer activity.
    CONCLUSIONS: Arglabin showed selective in vitro and in vivo anticancer activities against OSCC cells and is therefore a potential therapeutic agent for the management of OSCC.
  13. Biochem Biophys Res Commun. 2018 Dec 27. pii: S0006-291X(18)32810-9. [Epub ahead of print]
      Oxidative stress (OS)-induced apoptosis of periodontal ligament cells (PDLCs) has been suggested to be an important pathogenic factor of periodontitis. Mitochondrial abnormalities are closely linked to OS and act as the main players in apoptosis. Our aim was to investigate the potential mitochondrial abnormalities in PDLCs apoptosis induced by OS. In this study, significant reduction in viability and increased apoptosis were observed in H2O2-treated hPDLCs. H2O2 also induced mitochondrial dysfunction, judging by increased mitochondrial reactive oxygen species amounts, and decreased mitochondrial membrane potential as well as ATP levels. Furthermore, H2O2 significantly enhanced mitochondrial fission by decreasing the expression of Mfn1 and Mfn2, along with increasing the expression of Drp1, Fis1 and the cleavage of OPA1. Notably, NAC stabilized the balance of the mitochondrial dynamics, attenuated mitochondrial dysfunction, and inhibited apoptosis of hPDLCs in the presence of H2O2. In conclusion, the OS-induced apoptosis of hPDLCs may be mediated by mitochondria-dependent pathway.
    Keywords:  Apoptosis; Human periodontal ligament cells; Mitochondrial abnormalities; Mitochondrial dynamics; Oxidative stress
    DOI:  https://doi.org/10.1016/j.bbrc.2018.12.143
  14. Biol Pharm Bull. 2019 ;42(1): 94-102
      Glutamate-mediated cytotoxicity has been implicated in the pathogenesis of neurological diseases, including Parkinson's disease, Alzheimer's disease, and stroke. In this study, we investigated the protective effects of alpha-lipoic acid (ALA), a naturally occurring thiol antioxidant, on glutamate-induced cytotoxicity in cultured C6 astroglial cells. Exposure to high-dose glutamate (10 mM) caused oxidative stress and mitochondrial dysfunction through the elevation of reactive oxygen species, depletion of glutathione, and loss of the mitochondrial membrane potential (ΔΨm). Pretreatment with ALA (200 µM), however, significantly inhibited the glutamate-induced oxidative stress and mitochondrial dysfunction. ALA pretreatment dose-dependently suppressed glutamate-induced apoptotic events including altered nuclear morphology and activation of caspase-3. In addition, ALA significantly attenuated glutamate-induced endoplasmic reticulum (ER) stress markers; namely, glucose-regulated protein 78 (GRP78), activating transcription factor 6 (ATF6), protein kinase regulated by RNA (PKR)-like ER-associated kinase (PERK), eukaryotic translation initiation factor 2 alpha (eIF2α), inositol-requiring enzyme 1 (IRE1), CCAAT/enhancer binding protein homologous protein (CHOP), and caspase-12. We confirmed that CHOP and caspase-12 are key mediators of glutamate-induced ER stress. Furthermore, exposure of the cells to a caspase-12-specific inhibitor and CHOP small interfering RNAs (siRNAs) led to restoration of the ΔΨm that was damaged by glutamate treatment. These results suggest that ALA can effectively suppress oxidative stress, mitochondrial dysfunction, and ER stress in astroglial cells.
    Keywords:  alpha-lipoic acid; apoptosis; glutamate; mitochondrial dysfunction
    DOI:  https://doi.org/10.1248/bpb.b18-00603
  15. J Cell Physiol. 2019 Jan 04.
      Chronic intermittent hypoxia (CIH) is known to induce hypertension, but the mechanism is not well understood. We hypothesized that sensory plasticity of the carotid body (CB) and oxidative stress in the paraventricular nucleus (PVN) are involved in CIH-induced hypertension. In this study, rats were exposed to CIH for 28 days (intermittent hypoxia of 21% O2 for 60 s and 5% O2 for 30 s, cyclically repeated for 8 hr/day) and then randomly grouped for intracerebroventricular injection of 5-HT2 receptor antagonist ritanserin, Rho-associated protein kinase (ROCK) inhibitor Y-27632, and NADPH oxidase (NOX) inhibitor diphenyleneiodonium (DPI), respectively. We found that CIH increased blood pressure (BP), elevated carotid sinus nerve (CSN) and renal sympathetic nerve (RSN) activities, oxidative stress, and cell apoptosis in PVN. NOX-derived reactive oxygen species (ROS) production and cell apoptosis decreased when CIH-induced activation of 5-HT/5-HT2AR/PKC signaling was inhibited by ritanserin. In addition, RhoA expression was downregulated when oxidative stress was attenuated by DPI, while Y-27632 decreased the expression of endothelin-1, which is overexpressed in the vascular wall during hypertension. Moreover, treatment with ritanserin, DPI or Y-27632 attenuated the sensory plasticity and sympathetic hyperactivity as well as CIH-induced elevation of BP. In conclusion, CIH-induced activation of 5-HT/5-HT2AR/PKC signaling contributes to NOX-derived oxidative stress in PVN, which may cause sensory plasticity of CB, RSN hyperactivity, and elevated BP.
    Keywords:  carotid body; chronic intermittent hypoxia; hypertension; oxidative stress; paraventricular nucleus
    DOI:  https://doi.org/10.1002/jcp.28031
  16. Phytomedicine. 2019 Jan;pii: S0944-7113(18)30449-5. [Epub ahead of print]52 178-186
      BACKGROUND: Protection the heart from ischemia/reperfusion (I/R) injury is an area of intense research, as myocardial infarction is a major cause of mortality and morbidity all around the world. Tournefolic acid B (TAB) is a relative new compound derived from Clinopodium chinense (Benth.) Kuntze (Chinese name: Feng Lun Cai). This traditional Chinese herbal medicine has been used for its activities on anti-inflammatory, lowering blood glucose, antitumor and antiradiation. However, the pharmacological effects of TAB were rarely studied.PURPOSE: Pathways involving phosphoinositide 3-kinase (PI3K) and protein kinase b (Akt) are crucial in regulating the ER stress and associated apoptosis in the process of I/R injury. In the present study, we aim to investigate the cardioprotective effects of tournefolic acid B (TAB) against myocardial I/R injury and explore the molecular mechanisms involved.
    STUDY DESIGN: H9c2 cadiomyocyte were incubated with TAB for 24 h and then exposed to hypoxia/reoxygenation. Isolated rat hearts were subjected to global ischemia and reperfusion in the absence or presence of TAB.
    METHODS: The possible mechanisms were investigated in vitro and ex vivo by multiple detection methods including JC-1 staining, ROS detection, activities of caspases detection, TUNEL staining, and Western-blot analysis.
    RESULTS: We found that TAB significantly improved the hemodynamic parameters (LVeDP, LVSP, + dP/dtmax, - dP/dtmin, and HR) of isolated rat hearts, and depressed the cardiomyocyte apoptosis. Besides, TAB inhibited the oxidative stress by adjusting the activities of antioxidant enzymes (SOD, CAT, and GSH-Px). The I/R injury triggered the endoplasmic reticulum (ER) stress by activating the ER proteins, such as Grp78, ATF6, PERK, and eIf2α. which are all refrained by TAB. TAB also enhanced the phosphorylation of PI3K and AKT, inhibited the expression of CHOP and Caspase-12, reduced the phosphorylation of JNK, and increased Bcl-2/Bax ratio.
    CONCLUSION: TAB protects against myocardial I/R injury by suppressing PI3K/AKT-mediated ER stress, oxidative stress, and apoptosis, revealing a promising therapeutic agent against ischemic cardiovascular diseases.
    Keywords:  Apoptosis; Endoplasmic reticulum stress; Myocardial ischemia/reperfusion injury; Oxidative stress; Tournefolic acid B
    DOI:  https://doi.org/10.1016/j.phymed.2018.09.168
  17. Phytomedicine. 2019 Jan;pii: S0944-7113(18)30461-6. [Epub ahead of print]52 60-69
      BACKGROUND: Licochalconce (LC) H is an artificial compound in the course of synthesizing LCC in 2013. So far, few studies on the effects of LCH have been found in the literature. Despite progress in treatment modalities for oral cancer, the cure from cancer has still limitations.PURPOSE: The effects of LCH were investigated on human oral squamous cell carcinoma (OSCC) cells to elucidate its mechanisms.
    STUDY DESIGN: We explored the mechanism of action of LCH by which it could have effects on JAK2/STAT3 signaling pathway.
    METHODS: To confirm LCH anti-cancer effect, analyzed were MTT assay, DAPI staining, soft agar, kinase assay, molecular docking simulation, flow cytometry and Western blotting analysis.
    RESULTS: According to docking and molecular dynamics simulations, the predicted pose of the complex LCH and JAK2 seems reasonable and LCH is strongly bound to active JAK2 with opened activation loop. The LCH inhibitor is surrounded by specific ATP-binding pocket in which it is stabilized by forming hydrogen bonds and hydrophobic interactions. It is shown that LCH plays as a competitive inhibitor in an active state of JAK2. LCH caused a dose-dependent decrease in phosphorylation of JAK2 and STAT3. More interestingly, LCH suppressed JAK2 kinase activity in vitro by its direct binding to the JAK2. LCH significantly inhibited the JAK2/STAT3 signaling pathway, causing the down-regulation of target genes such as Bcl-2, survivin, cyclin D1, p21 and p27. In addition, LCH inhibited cell proliferation and colony formation of OSCC cells in a dose- and time-dependent manner, as well as induction of cell apoptosis through extrinsic and intrinsic pathway. The induction of apoptosis in OSCC cells by LCH was evident in the increased production of ROS, loss of mitochondrial membrane potential, release of cyto c, variation of apoptotic proteins and activation of caspase cascade.
    CONCLUSION: LCH not only induces apoptosis in OSCC cells through the JAK/STAT3 signaling pathway but also inhibits cell growth. It is proposed that LCH has a promising use for the chemotherapeutic agent of oral cancer.
    Keywords:  7-AAD, 7-Aminoactinomycin D; Abbreviations: OSCC, Oral squamous cell carcinoma; Apaf-1, apoptotic protease activating factor-1; Apoptosis; C-PARP, cleaved Poly (ADP-Ribose) Polymerase; CHOP, CCAAT/enhancer-binding protein homologous protein; DAPI, 4′-6-diamidino-2-phenylindole; DR, Death receptor; FBS, fetal bovine serum; JAK, Janus kinase; JAK2; LC, Licochalcone; Licochalcone H; MMP, Mitochondrial membrane potential; MTT, 3-(4,5-dimethylthiazol-2-yl)-2,5 diphenyltetrazolium bromide; Oral cancer; PBS, phosphate-buffered saline; ROS, Reactive oxygen species; RT, Room temperature; STAT, Signal transducer and activators of transcription; STAT3; TPK, tyrosine protein kinase; cyto C, cytochrome C; tBid, truncated Bid
    DOI:  https://doi.org/10.1016/j.phymed.2018.09.180
  18. Toxicol Appl Pharmacol. 2018 Dec 28. pii: S0041-008X(18)30576-3. [Epub ahead of print]
      Dexmedetomidine (DEX) protects against liver damage caused by sepsis. The purpose of this study was to confirm the regulatory effects of DEX on glycogen synthase kinase 3 beta (GSK-3β) via the α2 adrenergic receptor (α2AR) and evaluate the role of GSK-3β in lipopolysaccharide (LPS)-induced liver injury. Sprague-Dawley (SD) rats were administered an intraperitoneal injection of DEX (30 μg/kg) 30 min before an intraperitoneal injection of LPS (10 mg/kg). HE staining and serum biochemical test results indicated that DEX significantly improved liver histopathological damage and liver function indices. Furthermore, DEX increased super oxide dismutase (SOD) activity and L-glutathione (GSH) levels, and inhibited malondialdehyde (MDA) production. Western blot (WB) analysis demonstrated that treatment with the GSK-3β inhibitor SB216763 increased antioxidant-related protein mitogen-activated protein kinase phosphatase 1 (MKP-1) and nuclear factor erythroid 2-related factor 2 (Nrf2) expression. In addition, anti-apoptosis-related proteins were up-regulated and pro-apoptosis-related proteins were down-regulated by SB21676 administration. WB analysis also showed that DEX increased anti-apoptosis-related protein levels and decreased pro-apoptotic protein levels in LPS-induced liver injury. Nrf2, p53, and activated caspase-3 levels were further evaluated using immunohistochemistry (IHC), producing results consistent with WB results. The α2AR antagonist atipamezole (AT) significantly reversed the protective effects of DEX, as shown by WB analysis. Our data suggested that α2AR plays an important role in reversing the effects of liver oxidative stress and apoptosis via DEX, and that DEX exerts antioxidant and anti-apoptosis effects through regulation of the GSK-3β/MKP-1/Nrf2 pathway.
    Keywords:  Dexmedetomidine; GSK-3β; Lipopolysaccharide; Liver damage; α2 adrenergic receptor
    DOI:  https://doi.org/10.1016/j.taap.2018.12.017
  19. BMC Neurosci. 2018 Dec 29. 19(1): 82
      BACKGROUND: Cobalt chloride (CoCl2) induces chemical hypoxia through activation of hypoxia-inducible factor-1 alpha (HIF-1α). Mammalian target of rapamycin (mTOR) is a multifaceted protein capable of regulating cell growth, angiogenesis, metabolism, proliferation, and survival. In this study, we tested the efficacy of a well-known mTOR inhibitor, rapamycin, in reducing oxidative damage and increasing cell viability in the mouse hippocampal cell line, HT22, during a CoCl2-simulated hypoxic insult.RESULTS: CoCl2 caused cell death in a dose-dependent manner and increased protein levels of cleaved caspase-9 and caspase-3. Rapamycin increased viability of HT22 cells exposed to CoCl2 and reduced activation of caspases-9 and -3. Cells exposed to CoCl2 displayed increased reactive oxygen species (ROS) production and hyperpolarization of the mitochondrial membrane, both of which rapamycin successfully blocked. mTOR protein itself, along with its downstream signaling target, phospho-S6 ribosomal protein (pS6), were significantly inhibited with CoCl2 and rapamycin addition did not significantly lower expression further. Rapamycin promoted protein expression of Beclin-1 and increased conversion of microtubule-associated protein light chain 3 (LC3)-I into LC3-II, suggesting an increase in autophagy. Pro-apoptotic protein, Bcl-2 associated × (Bax), exhibited a slight, but significant decrease with rapamycin treatment, while its anti-apoptotic counterpart, B cell lymphoma-2 (Bcl-2), was to a similar degree upregulated. Finally, the protein expression ratio of phosphorylated mitogen-activated protein kinase (pMAPK) to its unphosphorylated form (MAPK) was dramatically increased in rapamycin and CoCl2 co-treated cells.
    CONCLUSIONS: Our results indicate that rapamycin confers protection against CoCl2-simulated hypoxic insults to neuronal cells. This occurs, as suggested by our results, independent of mTOR modification, and rather through stabilization of the mitochondrial membrane with concomitant decreases in ROS production. Additionally, inhibition of caspase-9 and -3 activation and stimulation of protective autophagy reduces cell death, while a decrease in the Bax/Bcl-2 ratio and an increase in pMAPK promotes cell survival during CoCl2 exposure. Together these results demonstrate the therapeutic potential of rapamycin against hypoxic injury and highlight potential pathways mediating the protective effects of rapamycin treatment.
    Keywords:  Cobalt chloride; HT22; Hypoxia; Rapamycin
    DOI:  https://doi.org/10.1186/s12868-018-0482-4
  20. Int J Mol Sci. 2019 Jan 02. pii: E142. [Epub ahead of print]20(1):
      Natural sources are very promising materials for the discovery of novel bioactive compounds with diverse pharmacological effects. In recent years, many researchers have focused on natural sources as a means to prevent neuronal cell death in neuropathological conditions. This study focused on identifying neuroprotective compounds and their underlying molecular mechanisms. Procyanidin C1 (PC-1) was isolated from grape seeds and assessed for biological effects against glutamate-induced HT22 cell death. The results showed that PC-1 strongly prevented glutamate-induced HT22 cell death. Moreover, PC-1 was also found to prevent glutamate-induced chromatin condensation and reduce the number of annexin V-positive cells indicating apoptotic cell death. Procyanidin C1 possessed a strong 2,2-diphenyl-1-picrylhydrazyl (DPPH) radical-scavenging activity and inhibited glutamate-induced accumulation of intracellular reactive oxygen species and protein carbonylation. Additionally, PC-1 mediated nuclear translocation of nuclear factor erythroid-derived 2-related factor 2 and increased the expression levels of heme oxygenase (HO-1). Inhibition of HO-1 by tin protoporphyrin, a synthetic inhibitor, reduced the protective effect of PC-1. Furthermore, PC-1 also blocked glutamate-induced phosphorylation of mitogen-activated protein kinases (MAPKs) including ERK1/2 and p38, but not JNK. This study is the first experimental report to demonstrate the neuroprotective effects of PC-1 against glutamate-induced cytotoxicity in HT22 cells. Therefore, our results suggest that PC-1, as a potent bioactive compound of grape seeds, can prevent neuronal cell death in neuropathological conditions.
    Keywords:  MAPK; Nrf2/HO-1 signaling pathway; glutamate; neuroprotective effects; oxidative stress; procyanidin C1
    DOI:  https://doi.org/10.3390/ijms20010142
  21. J Exp Clin Cancer Res. 2019 Jan 03. 38(1): 2
      BACKGROUND: Glioblastomas multiforme (GBM) is the most devastating primary intracranial malignancy lacking effective clinical treatments. Notch2 has been established to be a prognostic marker and probably involved in GBM malignant progression. N-acetylcysteine (NAC), a precursor of intracellular glutathione (GSH), has been widely implicated in prevention and therapy of several cancers. However, the role of NAC in GBM remains unclear and the property of NAC independent of its antioxidation is largely unknown.METHODS: The mRNA and protein levels of Notch family and other related factors were detected by RT-PCR and western blot, respectively. In addition, intracellular reactive oxygen species (ROS) was measured by flow cytometry-based DCFH-DA. Moreover, cell viability was assessed by CCK8 and cell cycle was analyzed by flow cytometry-based PI staining. The level of apoptosis was checked by flow cytometry-based Annexin V/PI. Cell migration and invasion were evaluated by wound healing and transwell invasion assays. At last, U87 Xenograft model was established to confirm whether NAC could restrain the growth of tumor.
    RESULTS: Our data showed that NAC could decrease the protein level of Notch2. Meanwhile, NAC had a decreasing effect on the mRNA and protein levels of its downstream targets Hes1 and Hey1. These effects caused by NAC were independent of cellular GSH and ROS levels. The mechanism of NAC-mediated Notch2 reduction was elucidated by promoting Notch2 degradation through Itch-dependent lysosome pathway. Furthermore, NAC could prevent proliferation, migration, and invasion and might induce apoptosis in GBM cells via targeting Notch2. Significantly, NAC could suppress the growth of tumor in vivo.
    CONCLUSIONS: NAC could facilitate Notch2 degradation through lysosomal pathway in an antioxidant-independent manner, thus attenuating Notch2 malignant signaling in GBM cells. The remarkable ability of NAC to inhibit cancer cell proliferation and tumor growth may implicate a novel application of NAC on GBM therapy.
    Keywords:  Buthionine sulfoximine; Glioblastoma; Itch; Lysosome; N-acetylcysteine; Notch2
    DOI:  https://doi.org/10.1186/s13046-018-1016-8
  22. J Biochem Mol Toxicol. 2018 Dec 31. e22266
      Tungstate (W) is recognized as an agent of environmental pollution and a substitute to depleted uranium. According to some preliminary studies, tungstate toxicity is related to the formation of reactive oxygen species (ROS) under abnormal pathological conditions. The kidneys and liver are the main tungstate accumulation sites and important targets of tungstate toxicity. Since the mitochondrion is the main ROS production site, we evaluated the mechanistic toxicity of tungstate in isolated mitochondria for the first time, following a two-step ultracentrifugation method. Our findings demonstrated that tungstate-induced mitochondrial dysfunction is related to the increased formation of ROS, lipid peroxidation, and potential membrane collapse, correlated with the amelioration of adenosine triphosphate and glutathione contents. The present study indicated that mitochondrial dysfunction was associated with disruptive effects on the mitochondrial respiratory chain and opening of mitochondrial permeability transition (MPT) pores, which is correlated with cytochrome c release. Our findings suggest that high concentrations of tungstate (2 mM)-favored MPT pore opening in the inner membranes of liver and kidney mitochondria of rats. Besides, the results indicated higher tungstate susceptibility in the kidneys, compared with the liver.
    Keywords:  mitochondria; oxidative stress; toxicity; tungstate
    DOI:  https://doi.org/10.1002/jbt.22266
  23. Pharmaceutics. 2019 Jan 03. pii: E12. [Epub ahead of print]11(1):
      The use of cell-penetrating peptides (CPPs) in combination with nanoparticles (NPs) shows great potential for intracellular delivery of DNA. Currently, its application is limited due to the potential toxicity and unknown long-term side effects. In this study NPs prepared using a biodegradable polymer, poly(lactic⁻co⁻glycolic acid (PLGA) in association with a CPP, was assessed on two lung epithelial cell lines (adenocarcinomic human alveolar basal epithelial cells (A549) and normal bronchial epithelial cells (Beas-2B cells)). Addition of CPP was essential for intracellular internalization. No effects were observed on the mitochondrial activity and membrane integrity. Cells exposed to the NPs⁻DNA⁻CPP showed low inflammatory response, low levels of apoptosis and no activation of caspase-3. Increase in necrotic cells (between 10%⁻15%) after 24 h of incubation and increase in autophagy, induced by NPs⁻DNA⁻CPP, are likely to be related to the lysosomal escape mechanism. Although oxidative stress is one of the main toxic mechanisms of NPs, NPs⁻DNA⁻CPP showed decreased reactive oxygen species (ROS) production on Beas-2B cells, with potential antioxidant effect of CPP and no effect on A549 cells. This NP system appears to be safe for intracellular delivery of plasmid DNA to the lung epithelial cells. Further investigations should be conducted in other lung-related systems to better understand its potential effects on the lungs.
    Keywords:  alveolar; apoptosis; autophagy; bronchial; gene delivery
    DOI:  https://doi.org/10.3390/pharmaceutics11010012
  24. Oxid Med Cell Longev. 2018 ;2018 1324173
      Oxidative stress is closely linked to the toxic responses of various cell types in normal and pathophysiological conditions. Deoxynivalenol (DON), an inducer of stress responses in the ribosome and the endoplasmic reticulum (ER), causes mitochondrial dysfunction and mitochondria-dependent apoptosis through oxidative stress in humans and animals. The NF-κB pathway, which is closely linked to oxidative stress, is hypothesized to be a critical signaling pathway for DON-induced toxicity and is a potential target for intervention. The present study was conducted to explore the protective effects of pyrrolidine dithiocarbamate (PDTC) from the toxic effects of DON in rat anterior pituitary GH3 cells. Our results showed that DON activated the NF-κB transcription factors and induced cellular oxidative stress, mitochondrial dysfunction, and apoptosis. Morphological studies using transmission electron microscopy (TEM) and cell apoptosis analyses suggested that PDTC prevented DON-induced mitochondrial dysfunction and apoptosis, probably by preventing the DON-induced translocation of NF-κB p65 into the nucleus, and by inhibiting DON-induced iNOS expression. This led to the blocking of the NF-κB pathway and inhibition of iNOS activity.
    DOI:  https://doi.org/10.1155/2018/1324173
  25. Arch Med Res. 2018 Dec 29. pii: S0188-4409(18)30433-8. [Epub ahead of print]
      BACKGROUND: The widespread use by young people of modern communication devices such as mobile phones means that they are particularly exposed to electromagnetic fields (EMF) and other problems. However, few studies have researched the effects of long-term exposure to EMF in the kidney. We therefore investigated oxidative stress and apoptosis in long-term exposure to 2100 megahertz (MHz) in a rat model.MATERIALS AND METHODS: Twenty-four Sprague Dawley rats were divided into a control group (n = 8, no EMF exposure), a group exposed to 2100 MHz for 6 h for 30 d (n = 8), and a group exposed to 2100 MHz for12 h for 30 d (n = 8). Immunohistochemical analysis was performed, using caspase-3 to evaluate apoptosis. Immediately after treatment, reduced glutathione (GSH), malondialdehyde (MDA) in kidney tissue and serum levels of various biochemical compounds were measured to detect oxidative stress.
    RESULTS: Deterioration was observed in the brush border in renal tubules of the EMF groups. The results of the immunohistochemical analysis revealed a greater number of positively stained renal tubular epithelial cells in the EMF groups as compared with that in the control group. In the EMF groups, renal MDA levels increased, and renal GSH levels decreased compared with those in the control group, as shown by a biochemical examination (p = 0.00 and p = 0.00, respectively).
    CONCLUSION: The findings showed that exposure to 2100 MHz for 6 and 12 h induced oxidative stress-mediated acute renal injury, depending on the length of exposure and dosage.
    Keywords:  Apoptosis; Caspase-3; Electromagnetic field; Kidney; Oxidative stress
    DOI:  https://doi.org/10.1016/j.arcmed.2018.12.010
  26. Int J Biol Macromol. 2018 Dec 31. pii: S0141-8130(18)35593-4. [Epub ahead of print]
      In our previous study, a novel cold-water-soluble polysaccharide (APS4) was isolated from Astragalus membranaceus. This study aimed to evaluate the proliferation inhibition and apoptosis-induced effects of APS4 on human gastric carcinoma MGC-803 cells and to investigate its potential molecular mechanism. It was found that APS4 could significantly suppress the proliferation of MGC-803 cells in a concentration- and time-dependent manner. Morphologic observations and Annexin V-FITC/PI staining showed that APS4-treated MGC-803 cells exhibited typical morphological characteristics of apoptosis. Cell cycle detection revealed that APS4 could arrest MGC-803 cells in S phase of the cell cycle. Additionally, APS4 treatment could induce the mitochondria-dependent apoptosis, which was closely related to the accumulation of intracellular ROS, the collapse of mitochondrial membrane potential, the increase of the pro-apoptotic/anti-apoptotic (Bax/Bcl-2) ratios, the release of cytochrome c, further activating the expression of caspase-9/-3 and the cleavage of poly-ADP-ribose polymerase (PARP) in MGC-803 cells. Taken together, our results suggested that APS4 had observable apoptosis-induced effects on MGC-803 cells via arresting the cell cycle in S phase and inducing the intrinsic mitochondrial apoptosis pathway.
    Keywords:  Cold-water soluble Astragalus membranaceus polysaccharide; Human gastric cancer; Mitochondrial apoptosis pathway
    DOI:  https://doi.org/10.1016/j.ijbiomac.2018.12.268
  27. J Asian Nat Prod Res. 2019 Jan 04. 1-16
      Inappropriate use of acetaminophen (APAP) can lead to morbidity and mortality secondary to hepatic necrosis. Ginsenoside Rg1 is a major active ingredient in processed Panax ginseng, which is proved to elicit biological effects. We hypothesized the beneficial effect of Rg1 on APAP-mediated hepatotoxicity was through Nrf2/ARE pathway. The study was conducted in cells and mice, comparing the actions of Rg1. Rg1 significantly improved cell survival rates and promoted the expression of antioxidant proteins. Meanwhile, Rg1 reduced the excessive ROS and the occurrence of cell apoptosis, which were related to Nrf2/ARE pathway. Expression of Nrf2 has a certain cell specificity. [Formula: see text].
    Keywords:  Nrf2; acetaminophen; apoptosis; ginsenoside Rg1; oxidative stress
    DOI:  https://doi.org/10.1080/10286020.2018.1504024
  28. Gene. 2018 Dec 27. pii: S0378-1119(18)31268-X. [Epub ahead of print]
      In human pancreatic β-cells, oxidative stress and cellular injures can be induced by H2O2 treatment. The KEAP1/NRF2 axis is a key antioxidant signaling pathway. The present study attempted to elucidate the mechanism by which the KEAP1/NRF2 axis mediates oxidative stress-induced death in pancreatic β-cells. Our data showed that H2O2 treatment obviously induced the apoptosis of β-cells. Further experiments demonstrated that KEAP1 expression was downregulated in H2O2-treated pancreatic β-cells and this change correlated with increase in the cellular abundance and nuclear translocation of NRF2. The restoration of KEAP1 expression in cells resulted in a recovery of cell proliferation and inhibition of apoptosis. Furthermore, we found that KEAP1 overexpression negatively regulated the abundance of NRF2, subsequently causing decreased antioxidant response element activation. This led to HO-1 protein downregulation in H2O2-treated human pancreatic β-cells, which was also observed in NRF2-silenced β-cells. Conversely, the silencing of KEAP1 led to NRF2 upregulation and inhibited ARE and HO-1 signaling in pancreatic β-cells. The increase in the abundance of NRF2 following treatment with H2O2 drastically elevated the production of BAX, FAS, FAS-L, CASP-3, and CASP-9, and this change was reversed by KEAP1 overexpression or NRF2 silencing. Taken together, H2O2 treatment activated KEAP1/NRF2 signaling to promote the production of pro-apoptotic factors and consequently led to the apoptosis of human pancreatic β-cells.
    Keywords:  Apoptosis; H(2)O(2); KEAP1; NRF2; Pancreatic β-cell
    DOI:  https://doi.org/10.1016/j.gene.2018.11.100
  29. Int J Biochem Cell Biol. 2018 Dec 26. pii: S1357-2725(18)30278-4. [Epub ahead of print]
      Substance P (SP) is believed to play a role in traumatic brain injury (TBI), and the inhibition of binding of SP to the tachykinin neurokinin-1 receptor (NK1R) using NK1R antagonists had made favorable effects on TBI. Our current study addresses the functional roles and underlying mechanisms of SP and NK1R antagonist L-733,060 following TBI. Adult male wild type C57BL/6 J and SP knock out (SP-KO) mice received a controlled cortical impact and outcome parameters were assessed. The results showed that TBI-induced motor and spatial memory deficits, lesion volume, brain water content and blood-brain barrier disruption were alleviated both in L-733,060-treated C57BL/6 J mice and vehicle-treated SP-KO mice. L-733,060 treatment and SP deletion inhibited TBI-induced the release of cytochrome c from mitochondria to cytoplasm, activation of caspase-3, oxidative stress and neuroinflammation. Higher SP levels in serum and cortex were observed in wild type mice undergoing TBI relative to wild type sham group, but very little expression of cortical SP was detected in the SP-/- mice either TBI or not. Upregulation of NK1R expression after TBI was observed, and there was no significant difference between wild type and SP-KO groups. In vitro, L-733,060 and SP deletion inhibited scratch injury-induced cell death, loss of mitochondrial membrane potential and reactive oxygen species (ROS) production following TBI. Together, the results of this study implicate a functional role for NK1-R antagonist L-733,060 and deletion of SP in TBI-induced neurological outcome, oxidative damage, neuroinflammation and cell death. Upregulation of NK1R maybe a consequence of TBI, independent of the levels of substance P. This study raises the possibility that targeting SP through its receptor NK1R or genetic deletion may have therapeutic efficacy in TBI.
    Keywords:  Neurokinin-1 receptor (NK1R); Substance P; cell death; oxidative stress; traumatic brain injury
    DOI:  https://doi.org/10.1016/j.biocel.2018.12.018
  30. Pol J Vet Sci. 2018 Dec;21(4): 673-680
      Myxomatous mitral valve disease (MMVD) is a cardiac condition commonly found in older dogs. The disease process can lead to heart failure (HF). In HF, an increase of reactive oxygen species (ROS) and abnormal mitochondrial activity, as well as apoptosis, have been reported. Humanin (HN) is a polypeptide that has a cardioprotective effect against apoptosis and oxidative stress. The purposes of this study were (1) to investigate the potential role of plasma HN as a cardiac biomarker to predict disease progression of MMVD, and (2) to compare plasma HN concentrations with plasma NT-pro BNP concentrations. Thirty-one dogs were included in the study. The dogs were separated into four groups: Group 1 was healthy dogs (n = 8), Group 2 was MMVD class B (n = 8), Group 3 was MMVD class C (n = 8), and Group 4 was MMVD class D (n = 7). All dogs were given a physical examination, thoracic radiography, echocardiography, and samples of their blood were collected for hematology and blood chemistry analysis. Levels of plasma HN and plasma NT-proBNP were also investigated. The results showed that plasma HN levels were lower in the dogs with MMVD and that lower plasma HN levels were associated with greater severity of MMVD-induced HF. It was possible to observe changes in plasma HN levels at a less severe disease stage than plasma NT-proBNP in dogs with MMVD. These findings sug- gest that a decreased plasma HN level can be used as a biomarker to identify dogs with MMVD-induced HF.
    Keywords:  Humanin; NT-proBNP; ROS; heart failure; myxomatous mitral valve disease
    DOI:  https://doi.org/10.24425/124305
  31. PeerJ. 2018 ;6 e6097
      Pleurotus ferulae is an edible and medicinal mushroom with various bioactivities. Here, the ethanol extracts of wild and cultivated P. ferulae (PFEE-W and PFEE-C) and their subfractions including petroleum ether (Pe-W/Pe-C), ethyl acetate (Ea-W/Ea-C) and n-butanol (Ba-W/Ba-C) were prepared to evaluate their antioxidant and antitumor activities. Both PFEE-W and PFEE-C show the antioxidant activity and PFEE-W is stronger than PFEE-C. The antioxidant activities of their subfractions are in the following order: Ea > Ba > Pe. Moreover, PFEE-W and PFEE-C significantly inhibit the proliferation of murine melanoma B16 cells, human esophageal cancer Eca-109 cells, human gastric cancer BGC823 cells and human cervical cancer HeLa cells through induction of apoptosis, which partially mediated by reactive oxygen species. The antitumor activities of their subfractions are in the following order: Ea ≥ Pe > Ba. Pe-W shows higher antitumor activity compared with Pe-C, which might be correlated with the difference of their components identified by gas chromatography-mass spectrometry. These results suggest that both wild and cultivated P. ferulae have antioxidant and antitumor activities, and cultivated P. ferulae could be used to replace wild one in some functions.
    Keywords:  Antioxidation; Antitumor; Apoptosis; Ethanol extract; Pleurotus ferulae
    DOI:  https://doi.org/10.7717/peerj.6097
  32. Shock. 2018 Dec 21.
      Sepsis, a life-threatening organ dysfunction due to a dysregulated response to infection, is a common complication of major surgery. Previous studies have showed that methane possesses protective properties. This study aims to investigate the protective effect of methane-rich saline (MRS) on sepsis-induced liver injury. In an in vivo experiment, C57BL/6 mice received cecal ligation and puncture to create a septic model followed by MRS treatment (10 ml/kg, ip treatment) 30 min and 12 h after the operation. We found that methane effectively decreased the serum aspartate aminotransferase, alanine aminotransferase and liver index, as well as the liver pathological damage, and reduced the localized infiltration of inflammatory cells. Methane suppressed the expression of the TLR4/ NF-κB signaling pathway and stimulated the expression of PPAR-γ during sepsis, which inhibited the activation of NF-κB and decreased the level of inflammatory cytokines, such as tumor necrosis factor-α, interleukin-6 and interleukin-1β. Moreover, we found that MRS treatment relieved ROS damage by upregulating heme oxygenase-1, superoxide dismutase and glutathione, and downregulating malondialdehyde, which was consistent with the results of DHE fluorescent staining. MRS treatment also regulated apoptosis-related proteins, such as Bax, Bcl-2 and caspase-3. In the in vitro experiment, HepG2 cells received inflammatory stimulation induced by LPS followed by methane-rich medium (MRM) treatment. We found that MRM alleviated the inflammatory damage, ROS damage and regulated the expression of PPAR-γ/NF-κB. Our data indicated that methane treatment prevented liver damage in sepsis via anti-inflammatory, anti-oxidative, and anti-apoptotic properties that involved the PPAR-γ/ NF-κB signaling pathway.
    DOI:  https://doi.org/10.1097/SHK.0000000000001310
  33. Biomed Res Int. 2018 ;2018 7049053
      Goniothalamin (GTN), a styryl-lactone, exhibits inhibitory effects on many kinds of cancer cells in vitro. The objectives of this study were to investigate the anticancer activities of GTN and molecular signaling pathways associated with cell death in human breast cancer MDA-MB-231 cell line. GTN inhibited the growth of MDA-MB-231 cells. Apoptosis was confirmed by annexin V-FITC and PI staining, and apoptotic morphology was observed by microscopy. Reduction of mitochondrial transmembrane potential and enhanced caspases activities were found in GTN-treated MDA-MB-231 cells. GTN significantly altered apoptosis-related protein expressions, including Noxa, PUMA, Bax, Bim, Bad, Bcl-2, Bcl-xL, and DIABLO, which was related to the gene expression levels. Mitochondrial calcium released to the cytosol and ER stress related proteins increased, which correlated with increases in ER stress gene expression levels. GTN induced hydrogen peroxide and superoxide anion radicals in MDA-MB-231 cells associated with cell cycle arrest in G2/M phase, which was induced by phosphorylation and ATM gene expression. Moreover, GTN had synergistic effects when combined with cyclophosphamide, 5-fluorouracil, paclitaxel, and vinblastine, and additive effect with methotrexate through caspases enzyme-acceleration. In conclusion, goniothalamin-induced MDA-MB-231 cell apoptosis occurred via intrinsic and extrinsic pathways, along with ER stress. These pathways provide new targeted drug strategies for advancements in anticancer medicine.
    DOI:  https://doi.org/10.1155/2018/7049053
  34. J Cell Mol Med. 2019 Jan 04.
      In addition to the known antitumour effects of ursolic acid (UA), increasing evidence indicates that this molecule plays a role in cardiac protection. In this study, the effects of ursolic acid on the heart in mice treated with doxorubicin (DOX) were assessed. The results showed that ursolic acid improved left ventrical fractional shortening (LVFS) and left ventrical ejection fraction (LVEF) of the heart, increased nitrogen oxide (NO) levels, inhibited reactive oxygen species (ROS) production and decreased cardiac apoptosis in mice treated with doxorubicin. Mechanistically, ursolic acid increased AKT and endothelial nitric-oxide synthase (eNOS) phosphorylation levels, and enhanced eNOS expression, while inhibiting doxorubicin induced eNOS uncoupling through NADPH oxidase 4 (NOX4) down-regulation. These effects of ursolic acid resulted in heart protection from doxorubicin-induced injury. Therefore, ursolic acid may be considered a potential therapeutic agent for doxorubicin-associated cardiac toxicity in clinical practice.
    Keywords:  apoptosis; cardiac toxicity; doxorubicin; ursolic acid
    DOI:  https://doi.org/10.1111/jcmm.14130
  35. Hum Exp Toxicol. 2018 Dec 31. 960327118812158
      Cobalt is a ferromagnetic metal with extensive industrial and biological applications. To assess the toxic effects of, and mechanisms involved in cobalt chloride (CoCl2)-induced cardio-renal dysfunctions. Male Wistar rats were exposed orally, daily through drinking water to 0 ppm (control), 150 ppm, 300 ppm, and 600 ppm of CoCl2, respectively. Following exposure, results revealed significant ( p < 0.05) rise in markers of oxidative stress, but decreased activities of catalase, glutathione peroxidase, glutathione-S-transferase, and reduced glutathione content in cardiac and renal tissues. There were significant increases in systolic, diastolic, and mean arterial blood pressure at the 300- and 600-ppm level of CoCl2-exposed rats relative to the control. Prolongation of QT and QTc intervals was observed in CoCl2 alone treated rats. Also, there were significant increases in the heart rates, and reduction in P wave, and PR duration of rats administered CoCl2. Histopathology of the kidney revealed peritubular and periglomerular inflammation, focal glomerular necrosis following CoCl2 exposure. Further, cyclooxygenase-2 and B-cell associated protein X expressions were upregulated in the cardiac and renal tissues of CoCl2-exposed rats relative to the control. Combining all, results from this study implicated oxidative stress, inflammation, and apoptosis as pathologic mechanisms in CoCl2-induced hypertension and cardiovascular complications of rats.
    Keywords:  Cobalt chloride; apoptosis; hypertension; inflammation; rats
    DOI:  https://doi.org/10.1177/0960327118812158
  36. Tissue Eng Regen Med. 2017 Apr;14(2): 133-141
      Human dermal fibroblast is essential in wound healing of the skin through the synthesis of extracellular matrix proteins. With respect to oxidative stress, the effects of remifentanil on human dermal fibroblast have received little attention. Therefore, we investigated the effects of remifentanil on the apoptosis and autophagic reaction of human dermal fibroblasts under oxidative stress. The subjects were divided into the following groups: Control group: cells were incubated at 37°C in a humidified atmosphere with 5% CO2. Hydrogen peroxide (H2O2) group: cells were exposed to H2O2 for 2 h. RPC/H2O2 group: cells were pretreated with remifentanil for 2 h and exposed H2O2 for 2 h. 3-MA/RPC/H2O2 group: cells were pretreated with 3-methyladenine (3-MA) and remifentanil for 1 h and 2 h, respectively. We measured cell viability using MTT assay. Western blot analysis was used to determine the expression levels of proteins associated with apoptosis and autophagy. Quantification of apoptotic cells was performed using flow cytometer analysis, and autophagic vacuoles were observed under a fluorescence microscope. Remifentanil treatment increased the proliferation of human dermal fibroblast and decreased apoptotic cell death, enhancing autophagic activity under oxidative stress. However, 3-MA, the autophagy pathway inhibitor, inhibited the protective effect of remifentanil in oxidative stress. This study demonstrates that remifentanil activated autophagy and decreased apoptotic death of human dermal fibroblasts under oxidative stress. Our results suggest that remifentanil may help in the treatment of oxidative stress.
    Keywords:  3-methyladenine; Autophagy; Dermal fibroblast; Oxidative stress; Remifentanil
    DOI:  https://doi.org/10.1007/s13770-017-0030-9
  37. Food Chem Toxicol. 2018 Dec 28. pii: S0278-6915(18)30922-0. [Epub ahead of print]
      Clinical and experimental data have demonstrated that diabetes is associated with neurological complications. Protocatechuic acid (PCA) is a phenolic phytochemical widely reported to possess antidiabetic property. However, there is no scientific information on the influence of PCA on diabetes-induced neurotoxicity. The present study aimed at investigating the neuroprotective mechanism of PCA in streptozotozin (STZ)-induced type 1 diabetic rats orally treated with PCA (50 mg/kg body weight) or glibenclamide (5 mg/kg body weight) for 45 consecutive days. Locomotor behavior was analyzed using video-tracking software during the 8-min trial in a novel environment whereas the pancreas, cerebrum and cerebellum of the rats were processed for biochemical analyses. Results showed that treatment of diabetic rats with PCA at 50 mg/kg significantly (p < 0.05) improved the locomotor and motor activities including the average speed, total time mobile, distance travelled, body rotation, turn angle, forelimb grip and grooming when compared with untreated diabetic rats. Moreover, the prevention of diabetes-mediated increase in acetylcholinesterase activity, biomarkers of inflammatory and oxidative stress as well as caspase 3 activity by PCA treatment was accompanied by improved pancreatic, cerebral and cerebellar architectures. Collectively, the neuroprotective mechanisms of PCA is related to its antioxidant, anti-inflammatory and anti-apoptotic activities.
    Keywords:  Inflammation; Neurotoxicity; Protocatechuic acid; acetylcholinesterase; diabetes
    DOI:  https://doi.org/10.1016/j.fct.2018.12.040
  38. Neurochem Int. 2018 Dec 28. pii: S0197-0186(18)30551-5. [Epub ahead of print]
      Previously we demonstrated that p53 mediates dopaminergic neurotoxicity via inducing mitochondrial burdens and proapoptotsis. However, little is known about the role of p53 in the excitotoxicity induced by psychostimulant, such as cocaine. Cocaine-induced kindling (convulsive) behaviors significantly increased p53 expression in the brain. Cocaine-induced p53 expression was more pronounced in hippocampus than in striatum or prefrontal cortex. Genetic depletion of p53 significantly attenuated cocaine-induced convulsive behaviors, followed by c-Fos immunoreactivity, and oxidative burdens in the hippocampus of mice. The antioxidant potentials mediated by genetic depletion of p53 were more pronounced in the mitochondrial-than cytosolic-fraction. Depletion of p53 significantly attenuated the changes in mitochondrial transmembrane potential, intramitochondrial Ca2+ level, and mitochondrial oxidative burdens induced by cocaine. Consistently, depletion of p53 significantly inhibited mitochondrial p53 translocation, and cleaved-PKCδ induced by cocaine. In addition, depletion of p53 protected from cytosolic cytochrome c release, and pro-apoptotic changes induced by cocaine. Importantly, the protective/anticonvulsant potentials by genetic depletion of p53 were comparable to those by pifithrin-μ (PFT), a p53 inhibitor. Our results suggest that depletion of p53 offers anticonvulsive and neuroprotective potentials mainly via attenuating mitochondrial oxidative burdens, mitochondrial dysfunction, and pro-apoptotic signalings against cocaine-induced convulsive neurotoxicity.
    Keywords:  Cocaine-kindling (convulsive) behaviors; Hippocampus; Mitochondrial dysfunction; Oxidative stress; Pro-apoptosis; p53 knockout mice
    DOI:  https://doi.org/10.1016/j.neuint.2018.12.017