bims-aporos Biomed News
on Apoptosis and reactive oxygen species
Issue of 2018‒11‒25
forty-two papers selected by
Gavin McStay
Staffordshire University


  1. J Agric Food Chem. 2018 Nov 19.
      Deoxynivalenol (DON) is a common mycotoxin that induces oxidative stress and affects intestinal health of humans and animals. This study was carried out to investigate the potential protective effect of resveratrol (RES) as a natural antioxidant on the alleviation of cytotoxicity induced by DON using an established intestinal porcine epithelial cell line (IPEC-J2). Pre-treatment of cells with 15 µM RES improved cell viability and proliferation, and attenuated cell cycle arrest induced by DON. Pre-treatment with RES also decreased accumulation of intracellular reactive oxygen species (ROS), stabilized mitochondrial membrane potential (MMP), and prevented apoptosis induced by DON. RES activated the nuclear factor-erythroid 2-related factor-2 (Nrf2) signaling pathway, while knockdown of Nrf2 expression by siRNA abrogated the protective effects of RES on DON-induced cytotoxicity, oxidative stress, and mitochondria-dependent apoptosis. These results suggest that RES protects IPEC-J2 cells against DON-induced damage at least partly via the Nrf2 signaling pathway.
    DOI:  https://doi.org/10.1021/acs.jafc.8b03662
  2. Front Pharmacol. 2018 ;9 1250
      The aim of the study was to determine the developmental toxicity of the traditional Chinese medicine Xiaoaiping (XAP) and to investigate its underlying mechanism of action. Zebrafish embryos were incubated with 0.4, 0.8, 1.2, and 1.6 mg/mL XAP. Endpoints such as mortality, hatching rate, malformation, body length, morphology score, swimming behavior, histological changes, reactive oxygen species (ROS) production, total superoxide dismutase (T-SOD) activity, and the mRNA expression of genes related to oxidative stress, endoplasmic reticulum (ER) stress, apoptosis, and the Wnt pathway were evaluated. Our results demonstrated that XAP exposure increased mortality and malformation and reduced the hatching rate. XAP resulted in severe malformation, including swim bladder deficiency, yolk retention, pericardial edema, and tail curvature. Histopathological analysis showed that XAP induced liver, heart and muscle injury. High doses (≥1.2 mg/mL) of XAP notably decreased the locomotor capacity of zebrafish. ROS generation was remarkably increased and T-SOD activity was decreased, confirming that oxidative stress was induced by XAP. The mRNA expression levels of ER stress-related genes (chop, hspa5, hsp90b1, and perk), apoptosis-related genes (caspase-3, bax, and p53) and wnt11 were significantly upregulated by XAP exposure. The expression levels of the oxidative stress-related genes (cat, sod1, and gstp2), Wnt pathway-related genes (β-catenin, wnt3a, and wnt8a) and bcl-2 initially increased and then decreased as the XAP exposure dose increased. In conclusion, we provide evidence for the first time that XAP can induce dose-related developmental toxicity, and ER stress, apoptosis and the Wnt pathway participate in the toxicity regulation.
    Keywords:  Wnt pathway; apoptosis; developmental toxicity; endoplasmic reticulum stress; swimming behavior; xiaoaiping; zebrafish embryos
    DOI:  https://doi.org/10.3389/fphar.2018.01250
  3. Invest New Drugs. 2018 Nov 22.
      Caffeic acid phenethyl ester (CAPE) is a phenolic compound initially identified in bee glue. CAPE is reported to exhibit antitumor activity in many cancer models. However, the effect of CAPE on multiple myeloma (MM) is not well studied. We investigated the anti-myeloma effect of CAPE, and the data showed that CAPE inhibited the growth of human MM cells in a dose (1 ~ 30 μM) and time (24 ~72 h) dependent manner without altering the viability of normal human peripheral blood B cells. Stress and toxicity pathway analysis demonstrated that CAPE, in a dose- and time-related fashion, induced the expression of apoptotic and oxidative stress-response genes including growth arrest and DNA-damage inducible, alpha and gamma (GADD45A and GADD45G) and heme oxygenase-1. Apoptosis of MM cells by CAPE was further confirmed through flow cytometric analysis with up to 50% apoptotic cells induced by 50 μM CAPE within 24 h. Western blot analysis revealed the CAPE-induced activation of apoptosis executioner enzyme caspase-3, and corresponding cleavage of its downstream target poly(ADP-ribose)polymerase (PARP). The oxidative stress caused by CAPE cytotoxicity in MM cells was evaluated through measurement of reactive oxygen species (ROS) level, antioxidant intervention and glutathione depletion. The intracellular ROS level was not elevated by CAPE, but the pretreatment of antioxidant (N-acetyl cysteine) and glutathione synthesis inhibitor (buthionine sulfoximine) suggested that CAPE may cause oxidative stress by decrease of intracellular antioxidant level rather than over production of ROS. These data suggest that CAPE promotes apoptosis through oxidative stress in human multiple myeloma cells.
    Keywords:  Apoptosis; Caffeic acid phenethyl ester; Cytotoxicity; Multiple myeloma; Oxidative stress
    DOI:  https://doi.org/10.1007/s10637-018-0701-y
  4. Environ Toxicol. 2018 Nov 21.
      The catecholic derivative para-tertiary butyl catechol (PTBC) is a conventional antioxidant and polymerization inhibitor, which exhibits melanocytotoxic effects and contact dermatitis often leading to occupational leucoderma or vitiligo. Although numerous industrial workers will be in constant exposure to PTBC and its chances of getting entry into blood are most expected, its effect on blood components is still undisclosed. As platelets play a prominent role in dermatitis, inflammation, and immunity, in this study we have evaluated the effect of PTBC on human platelets in vitro. Exposure of platelets to PTBC showed increased reactive oxygen species (ROS), intracellular calcium, cardiolipin oxidation, mitochondrial permeability transition pore (MPTP) formation, activation of caspases, phosphatidylserine (PS) externalization and decreased mitochondrial membrane potential. In addition, there was a significant decrease in cellular glutathione level, increased γ-glutamyltransferase (GGT) activity and cell death. These findings demonstrate that PTBC could induce toxic effects on blood components, which is often ignored field of research. Since dermal exposure of humans to toxic chemicals covers an important issue in various industries, there is a need of such work to understand and update the long-term toxicities induced by PTBC usage in industrial sectors and public domain.
    Keywords:  PTBC; apoptosis; mitochondria; oxidative stress; platelets
    DOI:  https://doi.org/10.1002/tox.22681
  5. Chem Biol Interact. 2018 Nov 16. pii: S0009-2797(18)30770-1. [Epub ahead of print]
      The present study assessed the therapeutic potential of omeprazole (OME), the most commonly prescribed proton pump inhibitor (PPI) used to treat gastroesophageal hyperacidity, against cisplatin (CP)-induced toxicity in human renal tubular HK-2 cells and rat kidneys. Herein, we observed that exposure of HK-2 cells to OME reversed the injury caused by CP, including enhancing cell viability and alleviating intracellular reactive oxygen species (ROS) generation and membrane damage. Concomitantly, acute exposure of male SD rats to CP induced histopathological changes, which were prevented by co-administration with OME. Inflammation and oxidative stress were inhibited by OME during CP-induced renal injury by increasing the activity of superoxide dismutase, and reducing the levels of malondialdehyde, both in vivo and in vitro. The expression levels of major inflammatory response markers were significantly decreased in HK-2 cells and rat kidneys in response to OME. OME reduced CP cellular uptake through organic cation transporters 2 (OCT2) and the prompt efflux of CP by P-glycoprotein (P-gp), thereby reducing the accumulation of CP in kidney tissue and increasing its serum levels. These data demonstrate that CP-induced kidney damage is positively correlated with its cellular accumulation. Concurrently, OME showed renoprotective effect against CP-induced toxicity in HK-2 cells and rat kidneys, by suppressing oxidative stress and mediating NF-κB-dependent inflammation, apoptosis, and transporter function. As OME is commonly used in combination with CP during chemotherapy treatment, this study highlights the clinical significance of OME in alleviating CP-induced nephrotoxicity.
    Keywords:  Accum; Cisplatin; Drug-drug interaction; Nephrotoxicity; Omeprazole; Transporter
    DOI:  https://doi.org/10.1016/j.cbi.2018.11.008
  6. Biochem Biophys Res Commun. 2018 Nov 16. pii: S0006-291X(18)32312-X. [Epub ahead of print]
      Cervical cancer is one of the most lethal types of cancer among female. Microfibrillar-associated protein 5 (MFAP5) is an extracellular matrix (ECM) glycoprotein, and is confirmed to be involved in cell signaling during microfibril assembly, elastinogenesis and cell survival. However, the role of MFAP5 in cervical cancer development and progression remains poorly understood. In the study, MFAP5 was over-expressed in human cervical cancers, and in different cervical cancer cell lines. Patients suffering from cervical cancer with low MFAP5 expression exhibited better survival rate. Suppressing MFAP5 in cervical cancer cells markedly reduced the cell proliferation, migration and invasion by modulating epithelial-mesenchymal transition (EMT)-related signaling pathway. In addition, MFAP5 knockdown induced large number of cells distributed in G2/M phase, along with reduced Cyclin B1, Cyclin D1 and cyclin-dependent kinase 4 (CDK4) expressions, and enhanced p21 and p53 levels. Moreover, apoptosis was highly induced by MFAP5 silence through reducing Bcl-xl and Bcl-2 expressions, and promoting Bax, cleaved Caspase-3 and poly (ADP-Ribose) polymerase (PARP) expressions in cervical cancer cells. Reactive oxygen species (ROS) production levels were also higher in MFAP5-knockdown cells, along with Jun-N-terminal kinase (JNK) activation. Importantly, we found that MFAP5 knockdown-inhibited cervical cancer cell growth was dependent on ROS production. Finally, the depletion of MFAP5 prevented cervical cancer progression in vivo. In summary, our study identified a critical role played by MFAP5 in the progression of cervical cancer and the potential mechanisms by which exerted its effects, indicating that targeting MFAP5-related pathways could be conducive to the therapies for cervical cancer.
    Keywords:  Apoptosis; MFAP5; Migration/invasion; ROS; cervical cancer
    DOI:  https://doi.org/10.1016/j.bbrc.2018.10.146
  7. Environ Toxicol Pharmacol. 2018 Nov 07. pii: S1382-6689(18)30550-7. [Epub ahead of print]65 1-8
      The biochemical process of oxidative stress is an integral mechanism of chemical toxicity, contributing to complex pathological disorders. Thymol (THY) has a wide range of therapeutic applications for several ailments, although a better understanding of signaling cues regulated by this compound is needed to address the mechanism of its action. To better perceive the mode of action, we investigated the potential impact of THY on zebrafish embryos, with special emphasis on ROS biogenesis. In this study, we exposed the zebrafish embryos to 25, 50 and 100μM of THY for 96 hours post fertilization (hpf). Noticeable teratogenic effects were observed upon assessing the survival rate (LC50 = 42.35μM), hatching process, morphological exam and cardiac functions, thereby verifying the toxicity of THY on zebrafish embryos. Furthermore, we analyzed the effect of THY on the levels of ROS, mitochondrial membrane potential (ΔΨm) and immunofluorescence by DCFH-DA, JC-1, Casp-3-FITIC staining, respectively. Furthermore, we preformed the expressional analysis of Nrf2, superoxide dismutase-1 (SOD-1), catalase (CAT), Cytochrome P450 (CYP450) and apoptotic marker proteins (AIF, p53, Bax, Bcl-2, Casp-3 and Casp-9) in zebrafish embryos. As expected, we noticed a significant modulatory effect on the above-mentioned activities by THY. Collectively, our findings suggest that ROS might be the prime mediator responsible for THY-induced oxidative damage, thereby affecting the cellular defense mechanism and apoptotic events in zebrafish embryos.
    Keywords:  Thymol; antioxidants; apoptosis; developmental toxicity; reactive oxygen species; zebrafish
    DOI:  https://doi.org/10.1016/j.etap.2018.11.001
  8. Braz J Med Biol Res. 2018 Nov 14. pii: S0100-879X2018001200609. [Epub ahead of print]51(12): e7747
      Endoplasmic reticulum (ER) stress is a critical molecular mechanism involved in the pathogenesis of sepsis. Hence, strategies for alleviating this stress may be essential for preventing cardiovascular injuries under sepsis. Adiponectin is secreted by adipocytes and its levels are decreased in sepsis. The purpose of this study was to investigate the protective effects of adiponectin treatment on endothelial cells and its mechanism. Male Wistar rats underwent cecal ligation and puncture (CLP) before being treated with adiponectin (72 and 120 μg/kg). The levels of malondialdehyde (MDA) in plasma, histological structure, and apoptosis of endothelial cells were evaluated. In vitro, human umbilical vein endothelial cells (HUVECs) were treated with adiponectin at 10 and 20 μg/mL for 24 h after stimulation by lipopolysaccharide (LPS). The levels of reactive oxygen species (ROS), ultrastructure, rate of apoptosis, the expression of inositol-requiring enzyme 1α (IRE1α) protein, and its downstream molecules (78 kDa glucose-regulated protein (GRP78), C/EBP homologous protein (CHOP), and caspase-12) were detected. The results showed that the levels of MDA and ROS induced by CLP or LPS stimulation were increased. Furthermore, endothelial cell apoptosis was increased under sepsis. The IRE1α pathway was initiated, as evidenced by activated IRE1α, increased GRP78, and up-regulated CHOP and caspase-12 in HUVECs. Following treatment with adiponectin, the number of apoptotic endothelial cells was markedly decreased. These findings demonstrated that treatment with adiponectin decreased apoptosis of endothelial cells caused by sepsis by attenuating the ER stress IRE1α pathway activated by oxidative stress.
    DOI:  https://doi.org/10.1590/1414-431X20187747
  9. Onco Targets Ther. 2018 ;11 7669-7677
      Background: Drimia maritima (D. maritima) is a plant belonging to the family Asparagaceae, which has been used for the treatment of several ailments including cancer around the world. To our knowledge, there is no comprehensive study about the molecular mechanisms of anticancer activity of this plant, yet.Materials and methods: In the current study, cell viability, apoptosis induction, ROS production, mitochondrial apoptotic pathway, and ER stress mediators have been evaluated in breast cancer cells, MCF7, and MDA-MB-468 treated with D. maritima.
    Results: Significant cytotoxic effects were observed in MCF-7 and MDA-MB-468 cells after exposure to D. maritima. Apoptosis induction was determined using Annexin-V-FITC and propidium iodide staining. Furthermore, an increase of ROS, loss of mitochondrial membrane potential, the release of cytochrome c, activation of caspases, and elevation in the Bax/Bcl-2 ratio was determined. D. maritima dose-dependently increased the mRNA expression of ER stress markers such as CHOP, ATF-4, GADD34, and TRIB3 in MCF-7, and MDA-MB-468 cells.
    Conclusion: These data suggest that D. maritima induces apoptosis in human breast cancer cells via the mitochondrial-mediated pathway. In addition, endoplasmic reticulum stress seems to be involved in D. maritima-induced cell death.
    Keywords:  Drimia maritima; ER stress; ROS; apoptosis; breast cancer; mitochondria
    DOI:  https://doi.org/10.2147/OTT.S182786
  10. Cell Physiol Biochem. 2018 ;51(1): 201-216
      BACKGROUND/AIMS: Numerous studies have reported overproduction of reactive oxygen species (ROS) and alterations in mitochondrial energy metabolism in the development of diabetes and its complications. The potential protective effects of N-acetylcysteine (NAC) in diabetes have been reported in many therapeutic studies. NAC has been shown to reduce oxidative stress and enhance redox potential in tissues protecting them against oxidative stress associated complications in diabetes. In the current study, we aimed to investigate the molecular mechanisms of the protective action of NAC on STZ-induced toxicity in insulin secreting Rin-5F pancreatic β-cells.METHODS: Rin-5F cells were grown to 80% confluence and then treated with 10mM STZ for 24h in the presence or absence of 10mM NAC. After sub-cellular fractionation, oxidative stress, GSH-dependent metabolism and mitochondrial respiratory functions were studied using spectrophotometric, flow cytometric and Western blotting techniques.
    RESULTS: Our results showed that STZ-induced oxidative stress and apoptosis caused inhibition in insulin secretion while NAC treatment restored the redox homeostasis, enhanced insulin secretion in control cells and prevented apoptosis in STZ-treated cells. Moreover, NAC attenuated the inhibition of mitochondrial functions induced by STZ through partial recovery of the mitochondrial enzymes and restoration of membrane potential. STZ-induced DNA damage and expression of apoptotic proteins were significantly inhibited in NAC-treated cells.
    CONCLUSION: Our results suggest that the cytoprotective action of NAC is mediated via suppression of oxidative stress and apoptosis and restoration of GSH homeostasis and mitochondrial bioenergetics. This study may, thus, help in better understanding the cellular defense mechanisms of pancreatic β-cells against STZ-induced cytotoxicity.
    Keywords:  Apoptosis; N-acetylcysteine; Oxidative stress; Pancreatic Rin-5F cells; Streptozotocin
    DOI:  https://doi.org/10.1159/000495200
  11. Int J Mol Sci. 2018 Nov 16. pii: E3618. [Epub ahead of print]19(11):
      The rapid development of nanotechnology has led to the use of silver nanoparticles (AgNPs) in biomedical applications, including antibacterial, antiviral, anti-inflammatory, and anticancer therapies. The molecular mechanism of AgNPs-induced cytotoxicity has not been studied thoroughly using a combination of cellular assays and RNA sequencing (RNA-Seq) analysis. In this study, we prepared AgNPs using myricetin, an anti-oxidant polyphenol, and studied their effects on NIH3T3 mouse embryonic fibroblasts as an in vitro model system to explore the potential biomedical applications of AgNPs. AgNPs induced loss of cell viability and cell proliferation in a dose-dependent manner, as evident by increased leakage of lactate dehydrogenase (LDH) from cells. Reactive oxygen species (ROS) were a potential source of cytotoxicity. AgNPs also incrementally increased oxidative stress and the level of malondialdehyde, depleted glutathione and superoxide dismutase, reduced mitochondrial membrane potential and adenosine triphosphate (ATP), and caused DNA damage by increasing the level of 8-hydroxy-2'-deoxyguanosine and the expressions of the p53 and p21 genes in NIH3T3 cells. Thus, activation of oxidative stress may be crucial for NIH3T3 cytotoxicity. Interestingly, gene ontology (GO) term analysis revealed alterations in epigenetics-related biological processes including nucleosome assembly and DNA methylation due to AgNPs exposure. This study is the first demonstration that AgNPs can alter bulk histone gene expression. Therefore, our genome-scale study suggests that the apoptosis observed in NIH3T3 cells treated with AgNPs is mediated by the repression of genes required for cell survival and the aberrant enhancement of nucleosome assembly components to induce apoptosis.
    Keywords:  DNA damage; antioxidants; apoptosis; cytotoxicity; epigenetics; oxidative stress
    DOI:  https://doi.org/10.3390/ijms19113618
  12. Clin Exp Dermatol. 2018 Nov 19.
      BACKGROUND: Ultraviolet (UV)B radiation has long been considered a carcinogen in both epidemiological surveys and experimental studies. However, recent work has suggested that different dosages of UVB exert different influences on cells. There are also co-carcinogenesis factors such as arsenic that affect the role of UVB.AIM: To explore the co-carcinogenesis effect of UVB and arsenic on the mouse epidermal cell line JB6 and the mechanism underlying it.
    METHODS: Growth of JB6 cells was measured by MTT assay. We carried out a comet assay to determine the DNA damage caused by UVB and arsenic, and tested the expression of DNA repair protein by western blotting. Reactive oxygen species (ROS) were measured using DCF and DHE staining, and changes in antioxidant enzymes were assessed using western blotting.
    RESULTS: Viability assays showed that arsenic increased the UVB-induced death rate. Arsenic enhanced DNA damage caused by UVB both directly by injury to double-stranded DNA and indirectly by reducing the capability of DNA repair in JB6 cells. All of these effects are the results of increased ROS generation and reduced expression of the antioxidant enzyme superoxide dismutase (SOD)1.
    CONCLUSION: Arsenic was found to enhance UVB-induced production of ROS and to downregulate SOD1 expression, leading to DNA damage and apoptosis in mouse skin cells. The combination of arsenic and UVB exposure was found to differentially regulate the expression of SOD1 and SOD2.
    DOI:  https://doi.org/10.1111/ced.13834
  13. J Cell Physiol. 2018 Nov 23.
      N-(3-oxododecanoyl)-l-homoserine lactone (3-oxo-C12-HSL), a quorum-sensing (QS) molecule produced by Gram-negative bacteria in the gastrointestinal tract, adversly impacts host cells. Our previous study demonstrated that 3-oxo-C12-HSL induced a decrease in cell viability via cell apoptosis and eventually disrupted mucin synthesis from LS174T goblet cells. However, the molecular mechanism underlying cell apoptosis and whether pyroptosis was involved in this process are still unknown. In this study, we emphasized on the caspases signal pathway and sterile inflammation to reveal the harmful effects of 3-oxo-C12-HSL on LS174T goblet cells. Our data showed that 3-oxo-C12-HSL is a major inducer of oxidative stress indicated by a high level of intracellular reactive oxygen species (ROS). However, TQ416, an inhibitor of paraoxonase 2, can effectively block oxidative stress. A higher ROS level is the trigger for activating the caspase-1 and 3 cascade signal pathways. Blockade of ROS synthesis and caspase-1 and 3 cascades can obviously rescue the viability of LS174T cells after 3-oxo-C12-HSL treatment. We also found that paralleled with a higher level of ROS and caspases activation, an abnormal expression of proinflammatory cytokines was induced by 3-oxo-C12-HSL treatment; however, the blockage of TLRs-NF-κB pathway cannot restore cell viability and secretary function. These data collectively indicate that 3-oxo-C12-HSL exposure induces damages to cell viability and secretary function of LS174T goblet cells, which is mediated by oxidative stress, cell apoptosis, and sterile inflammation. Overall, the data in this study will provide a better understanding of the harmful impacts of some QS molecules on host cells and their underlying mechanism.
    Keywords:  3-oxo-C12-HSL; LS174T goblet cells; apoptosis; innate immune; oxidative stress
    DOI:  https://doi.org/10.1002/jcp.27132
  14. Phytomedicine. 2018 Nov 15. pii: S0944-7113(17)30115-0. [Epub ahead of print]50 238-246
      BACKGROUND: Human placental choriocarcinoma is a gestational trophoblastic tumor with high rates of metastasis and reoccurrence. However, some patients with choriocarcinoma are chemoresistance to conventional chemotherapeutic agents.HYPOTHESIS: Naringenin increases apoptosis in human placental choriocarcinoma cells.
    METHODS: We investigated the effects of naringenin on proliferation and migration of JAR and JEG3 cells, and performed TUNEL and Annexin V/PI staining assays to examine apoptotic effects of naringenin on both cells. In addition, we studied the loss of mitochondrial membrane potential (MMP) and the production of mitochondrial reactive oxygen species (ROS) to determine the specific reason for apoptosis of choriocarcinoma cells being mediated via mitochondria. Consistent with the induction of production of ROS by naringenin in both choriocarcinoma cell lines, we investigated lipid peroxidation and glutathione levels in both JAR and JEG3 cells since both are affected by ROS. We next determined dose-dependent effects of naringenin and its pharmacological inhibitors on signal transduction pathways in JAR and JEG3 cells by western blot analyses.
    RESULTS: Naringenin reduced viability and migratory functions of both cell lines, and increased mitochondria related apoptosis induced by ROS and lipid peroxidation, decreased glutathione and decreased mitochondrial membrane potential MMP in a dose-dependent manner. We also determined naringenin activated phosphorylation of ERK1/2, P38, JNK and P70S6K in JAR and JEG3 cells in a dose-response manner. Although naringenin induced phosphorylation of AKT proteins in JAR cells, it suppressed phosphorylation of the protein in JEG3 cells. In addition, we confirmed the mechanism of naringenin-induced cell signaling by using a combination of naringenin and pharmacological inhibitors of the PI3K and MAPK pathways, as well as a ROS inhibitor in JAR and JEG3 cell lines.
    CONCLUSIONS: Collectively, results of this study indicate that naringenin is a potential therapeutic molecule with anti-cancer effects on choriocarcinoma cells by inducing generation of ROS and activation of the MAPK pathways.
    Keywords:  Apoptosis; Choriocarcinoma; Naringenin; ROS; Signaling pathway
    DOI:  https://doi.org/10.1016/j.phymed.2017.08.026
  15. Cell Physiol Biochem. 2018 Nov 20. 51(2): 501-512
      BACKGROUND/AIMS: The taurine derivative Taurolidine is effective against diverse bacteria and tumor growth. In the treatment of cancer, the substance is effective in part by triggering suicidal death or apoptosis of tumor cells. The Taurolidine-induced apoptosis involves mitochondria. Erythrocytes lack mitochondria but are nevertheless able to enter suicidal erythrocyte death or eryptosis, which is characterized by cell shrinkage and cell membrane scrambling with phosphatidylserine translocation to the erythrocyte surface. Signaling of eryptosis includes increase of cytosolic Ca2+ activity ([Ca2+]i), oxidative stress and ceramide. The present study explores, whether Taurolidine induces eryptosis and, if so, which cellular mechanisms are involved.METHODS: Phosphatidylserine exposure at the cell surface was estimated using annexin-V-binding, cell volume using forward scatter, [Ca2+]i using Fluo3-fuorescence, reactive oxygen species (ROS) formation using 2',7'-dichlorodihydrofuorescein (DCF)-dependent fluorescence, and ceramide abundance using specific antibodies.
    RESULTS: A 48 hours exposure of human erythrocytes to Taurolidine (60 µg/ml) significantly enhanced the percentage of annexin-V-binding cells, significantly decreased forward scatter and significantly increased Fluo3-fluorescence and ceramide abundance, but not DCF-fluorescence. The effect of Taurolidine on annexin-V-binding was virtually abrogated by removal of extracellular Ca2+.
    CONCLUSION: Taurolidine triggers cell shrinkage and phospholipid scrambling of the erythrocyte cell membrane, an effect at least in part due to Ca2+ entry and paralleled by increase of ceramide abundance.
    Keywords:  Calcium; Cell volume; Eryptosis; Phosphatidylserine
    DOI:  https://doi.org/10.1159/000495272
  16. Phytomedicine. 2018 Nov 15. pii: S0944-7113(17)30165-4. [Epub ahead of print]50 184-195
      BACKGROUND: The genus Pamburus (Rutaceae) comprises the only species, Pamburus missionis (Wight) Swingle. Pamburus missionis is traditionally used in the treatment of swellings, chronic rheumatism, paralysis and puerperal diseases.PURPOSE: The present study investigates the cancer chemotherapeutic potential of essential oil (EO) from P. missionis.
    METHODS: EO was isolated by steam distillation and chemical composition was determined by GC-MS. Cell viability was used to detect cytotoxic activity. Mechanism of cell death was studied using Annexin V-FITC/PI binding, cell cycle analysis, measurement of MMP and ROS generation by flow cytometry. Expression of apoptosis related proteins was investigated by western blot.
    RESULTS: GC-MS analysis of the essential oil revealed the presence of 51 components. The major components were β-Caryophyllene, 4(14),11-Eudesmadiene, Aromadendrene oxide-(2) and Phytol. EO inhibited the growth and colony formation ability of A431 and HaCaT cells. EO treatment induced nuclear condensation and loss of membrane integrity, DNA fragmentation, increase in sub-G1 DNA content and increase in intracellular ROS level. Inhibition of intracellular ROS by ascorbic acid and N-acetyl cysteine treatment blocked EO induced apoptosis, revealing that apoptotic activity was by ROS accumulation. EO induced apoptosis was found to be due to the loss of mitochondrial membrane potential (ΔΨm), increase in Bax/Bcl-2 ratio, release of cytochrome c and activation of caspases (cleaved form of caspase-3, caspase-8, caspase-9) and by PARP cleavage.
    CONCLUSION: The present study revealed cancer chemotherapeutic potential of EO from P. missionis. EO induces cell death through intrinsic (mitochondrial) and extrinsic apoptotic pathway in A431 and HaCaT cells. These results suggest that EO could be used as a potential therapeutic agent for the treatment of skin epidermoid cancer.
    Keywords:  Anticancer; Apoptosis; Cancer therapy; Essential oil; P. missionis
    DOI:  https://doi.org/10.1016/j.phymed.2017.11.004
  17. Med Sci Monit. 2018 Nov 20. 24 8372-8382
      BACKGROUND Excessive alcohol consumption can cause hepatocellular injury. ATPase II (ATP8A1) can display an ATP-dependent phospholipid translocase activity. However, the function of ATP8A1 in hepatocyte injury is still unclear. In the present study we explored the effect of ATP8A1 on ethanol-induced hepatocyte injury. MATERIAL AND METHODS A human hepatocyte strain, HL-7702, was pretreated by ethanol with gradient concentration for 2, 4, 8, and 12 h, and were then divided into 6 groups after the cells were transfected. We detected cell viability by use of the Cell Counting Kit-8 (CCK-8) assay. Reactive oxygen species (ROS), apoptosis rate, and mitochondrial membrane potential (MMP) were measured using flow cytometry. We used quantitative reverse transcription-polymerase chain reaction (qRT-PCR) and Western blot to measure the mRNA and protein expression, respectively. RESULTS Ethanol inhibited the viability of HL-7702 cells and suppressed the expression of ATP8A1 in dose- and time-dependent manners. Furthermore, over-expression of ATP8A1 reduced the level of ROS and the apoptosis rate and recovered the MMP. Additionally, over-expressed ATP8A1 regulated the protein and mRNA levels of apoptosis-related molecules. Moreover, over-expression of ATP8A1 enhanced the phosphorylation of phosphatidylinositol 3-kinase (PI3K) and protein kinase B (Akt). CONCLUSIONS Over-expression of ATP8A1 alleviated ethanol-induced hepatocyte injury. Moreover, the PI3K/Akt signaling pathway appears to participate in inhibition of ethanol-induced hepatocyte apoptosis and may provide a candidate target for the treatment of alcoholic liver diseases (ALD).
    DOI:  https://doi.org/10.12659/MSM.910254
  18. J Oral Pathol Med. 2018 Nov 19.
      BACKGROUND: Syzygium cumini (L.) Skeels (jambolan) is commonly used in Indian traditional medicine to treat a variety of diseases such as obesity, diabetes etc. The cytotoxic potential of Syzygium cumini (SC) against oral cancer cell line is remains elusive. Therefore, in this study, we evaluated the cytotoxic effect of S. cumini in human oral squamous cell carcinoma (OSCC) cell line (SCC-25 cells).MATERIAL AND METHODS: OSCC cells are treated with different concentrations (10, 20 and 40 μg/mL) of S. cumuni for 24 h and cytotoxicity was analyzed by MTT assay. The intracellular reactive oxygen species (ROS) was measured using the indicator dye, 2',7'-dichlorofluorescin diacetate staining. Apoptosis-related morphological changes were evaluated by dual acridine orange/ethidium bromide (AO/EB) fluorescent staining and phosphatidylserine externalization was measured by annexin V assays. The protein and gene expression of cadherin-1 was evaluated by western blotting and PCR analysis.
    RESULTS: SC treatments caused cytotoxicity of OSCC cell line and induced intracellular ROS accumulation. This treatment also caused apoptosis related morphological changes and externalization of phosphatidylserine in OSCC cells. Further, SC treatments increased protein and gene expression of cadherin-1.
    CONCLUSION: S. cumini extract inhibits the proliferation of OSCC cells and induces apoptosis through ROS accumulation and therefore, it could be used for the prevention of OSCC. This article is protected by copyright. All rights reserved.
    Keywords:  Apoptosis; cytotoxicity; herbal medicine; reactive oxygen species
    DOI:  https://doi.org/10.1111/jop.12806
  19. Nanomedicine (Lond). 2018 Nov 20.
      With a special size and structure, nanoparticles (NPs) have excellent application prospects in various fields and are widely used in the biomedicine, cosmetics and chemical industries nowadays. However, there have been some reports on the biosafety of this new type of material, pointing out its cytotoxicity in inducing apoptosis. With different physicochemical properties in size, shape, surface charge, and ligand, NPs exhibit different biocompatibilities when interacting with different cells. Therefore, a comprehensive and deep study into the proapoptotic mechanism of NPs is necessary. In the present review, we summarize the NP-triggered apoptotic signal pathways in detail and highlight some important functional molecules involved. We hope our findings and perspectives provide a new direction for the sound development of nanotechnology in the future.
    Keywords:  apoptosis pathways; endoplasmic reticulum; mitochondria; nanoparticles; reactive oxygen species
    DOI:  https://doi.org/10.2217/nnm-2018-0167
  20. Phytomedicine. 2018 Dec 01. pii: S0944-7113(18)30520-8. [Epub ahead of print]51 196-204
      BACKGROUND: Cardiovascular disease and its related deaths are increasing in the modern world. Therefore, there is a need to identify a plant based nutraceutical supplement with potent activity.HYPOTHESIS/PURPOSE: Reportedly, the protective effect of the rutin in hypoxia-induced cardiomyocytes is due to the activation of molecular networks related to programmed cell death.
    STUDY DESIGN-METHODS: Phytochemical methods and advanced analytical methods were employed to isolate natural products from Spermococe hispida their effects in cardiomyocyets.
    RESULTS: We reports herein that CoCl2-induced hypoxic condition significantly decreased cell viability as evidenced by MTT assay and cell cycle analysis. Western blot studies revealed an up-regulation of HIF-1α, BAX and caspase and down-regulation of BCl-2 expression, followed by modulation of Akt, p-Akt, p38 and p-p38. The oxidative abnormalities were ameliorated by rutin pretreatment, as deduced by the reduced CoCl2-induced cytotoxicity, MDA concentration and LDH activity and the enhanced levels of GSH and SOD in a dose-dependent manner. Rutin protects H9c2 cells from CoCl2-induced hypoxic damage by mitigating oxidative stress and preserving cell viability by modulating the antiapoptotic proteins.
    CONCLUSION: The overall findings reinforce the cardioprotective action of rutin, a potential source of antioxidant of natural origin, which may help in mitigating the progress of oxidative stress in hypoxic conditions such as myocardial infarction and stroke.
    Keywords:  Antioxidant; Cardioprotection; Hypoxia; Oxidative stress; Rutin; Spermococe hispida
    DOI:  https://doi.org/10.1016/j.phymed.2018.09.229
  21. Front Biosci (Elite Ed). 2019 Jan 01. 11 20-28
      The present study was designed to evaluate the protective effect of fisetin against rotenone induced toxicity in SH-SY5Y neuroblastoma cellular model of Parkinson's disease (PD). SH-SY5Y neuroblastoma cells were treated with fisetin (5µM) 2 hr prior to being treated with rotenone (100 nM). Following the exposure of SH-SY5Y cells to rotenone, there was marked decreased cell viability, increased oxidative stress, activation of caspase-3 and apoptosis (dual staining, expressions of pro-apoptotic and anti-apoptotic indices). However pretreatment with fisetin significantly and dose-dependently alleviated rotenone induced cytotoxicity and oxidative stress in SH-SY5Y cells. Moreover, fisetin attenuated rotenone induced toxicity by down-regulating Bax, caspases-3 protein expression and up-regulating protein expression of Bcl-2, p38/JNK-MAPK and PI3K, Akt, GSK-3β pathways. Collectively, these results suggest that fisetin could prevent the rotenone-induced neurotoxicity via various signaling pathways.
  22. Biosci Rep. 2018 Nov 21. pii: BSR20181707. [Epub ahead of print]
      Background:Osteosarcoma (OS) is the most common malignant neoplasm in children and adolescents with a very high propensity for local invasion and poor response to current therapy.  Anticancer effect of chamaejasmine is newly discovered from Stellera chamaejasmine L. Our study focuses on investigating the effect of chamaejasmine on the cellular apoptosis, proliferation, autophagy, and the underlying mechanisms in MG-63. Methods:Our study investigated the concentration of chamaejasmine in MG-63 cells by MTT and verified that chamaejasmine inhibited cell invasion by transwell. We also used Hoechst staining as well as apoptotic associated-proteins in MG-63 cells .Meanwhile, we also detected the lysophagesome and autophagsome by Lysotracker. AMPK knockdown was performed with siRNA.Results: Our results show that chamaejasmine exerts cellular growth inhibition, pro-apoptotic and pro-autophagic effect via activating AMPK in MG-63 cells. Furthermore, chamaejasmine significantly increases autophagic cell via the inhibition of mammalian target of rapamycin (mTOR) and activation of AMPK signaling pathways. Administrated with chamaejasmine also induces Reactive Oxygen Species (ROS) generation, indicating cross-talking between these two primary modes of programmed cell death.Conclusion:Our results show that chamaejasmine promotes apoptosis and autophagy by activating AMPK /mTOR signaling pathways with involvement of ROS in MG-63 cells. Chamaejasmine is a promising anti-cancer agent in OS treatment and further studies are needed to confirm its efficacy and safety in vivo or other cancer cells.
    Keywords:  AMPK/mTOR; apoptosis; autophagy,; chamaejasmine; osteosarcoma
    DOI:  https://doi.org/10.1042/BSR20181707
  23. Int J Biol Macromol. 2018 Nov 16. pii: S0141-8130(18)35586-7. [Epub ahead of print]
      The proliferative activity of oyster polysaccharides in intestine epithelial cells (IEC-6) alleviated 5-fluorouracil-induced intestinal inflammation. In this study, we aimed to measure the ability of oyster polysaccharides to promote IEC-6 cell migration and antioxidant activity and further describe their cytoprotective effect on H2O2-challenged IEC-6 cells. The C30-60% fraction of polysaccharides (CHP2) showed rapid stimulation of IEC-6 cell migration after wounding. Then, CHP2 was fractionated into four fractions, namely, CHP2-1, CHP2-2, CHP2-3 and CHP2-4. The CHP2-3 fraction possessed high scavenging activities against 2,2'-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid) (ABTS) and oxygen radical absorbance capacity (ORAC), in comparison with other fractions. And CHP2-3 was heteropolysaccharide with sulfuric esters, and it was mainly composed of glucose, galactose and arabinose and had an average molecular weight of 41.81 kDa. Pretreatment with CHP2 and CHP2-3 significantly improved the survival rate of H2O2-treated IEC-6 cells, and reduced intracellular reactive oxygen species (ROS) levels. Moreover, CHP2-3 also significantly decreased H2O2-mediated increases in the secretion of interleukin-1β (IL-1β) and interleukin-6 (IL-6), and attenuated nuclear factor-κB (NF-κB) p65 activation. These results indicate that CHP2-3 may play a vital role in reducing oxidative damage in IEC-6 cells via radical scavenging, decreasing proinflammatory factors secretion, inhibiting the NF-κB pathway, and thus, reducing cell apoptosis.
    Keywords:  IEC-6; Oxidative stress; Oyster polysaccharide
    DOI:  https://doi.org/10.1016/j.ijbiomac.2018.11.154
  24. J Cell Biochem. 2018 Nov 18.
      BACKGROUND: Optic nerve injury is one of the most common and serious complications in traumatic brain injury (TBI). Alleviating degree of optic nerve injury is important to cure of TBI. This study explored the role of long noncoding RNA (lncRNA) GAS5 in mice retinal ganglion cells (RGCs) suffered to H2 O 2 injury.METHODS: Primary RGC (PRGCs) were treated with H2 O 2 to simulate an in vitro oxidation stress model. LncRNA GAS5 and miR-124 expressions were knocked down by cell transfection with short-hairpin RNA against GAS5 and miR-124 inhibitor, and the transfection efficiency was determined by qRT-PCR. Cell viability, apoptotic cell rate, and production of reactive oxygen species (ROS) was analyzed by CCK-8 assay, PI/FITC-Annexin V method, and DCFH-DA fluorometric assay. Cell apoptosis-associated proteins as well as activations of JAK/STAT3 signaling and JNK signaling were analyzed by Western blot.
    RESULTS: H2 O 2 treatment-induced cell injury was inhibited by lncRNA GAS5 silence. Specifically, knockdown of GAS5 improved viability of primary PRGCs, inhibited apoptosis, decreased ROS expression, increased antiapoptosis proteins' expressions, and decreased proapoptosis proteins' expressions. It was also found that miR-124 inhibitor treatment impaired the cell protective effect of GAS5 silence, indicating low level of GAS5 protected PRGCs via upregulating miR-124. GAS5 silence might exert cytoprotection effect via activating JAK/STAT3 signaling pathway and inhibiting activation of JNK signaling pathway.
    CONCLUSION: Knocking down lncRNA GAS5 alleviated H2 O 2 -induced injury in PRGCs via upregulation of miR-124, which might dependent on activation of JAK/STAT3 signaling pathway and inhibition of JNK signaling pathway.
    Keywords:  GAS5; JAK/STAT3 pathway; JNK pathway; miR-124; oxidation stress; traumatic brain injury
    DOI:  https://doi.org/10.1002/jcb.27560
  25. Food Chem Toxicol. 2018 Nov 17. pii: S0278-6915(18)30842-1. [Epub ahead of print]
      Plumbagin (5-hydroxy-2-methyl-1,4-naphthaquinone) has displayed antitumor activity in vitro and in animal models; however, the underlying molecular mechanisms have not been fully explored. The aim of this study was to investigate the anticancer effects of plumbagin isolated from Nepenthes alata against MCF-7 breast cancer cells. We examined the cytotoxicity, cell cycle regulation, apoptotic cell death, and generation of intracellular reactive oxygen species (ROS) in MCF-7 cells. Plumbagin exhibited potent cytotoxicity in MCF-7 cells (wild-type p53) compared to that in SK-OV-3 (null-type) human epithelial ovarian cancer cells. Specifically, plumbagin upregulated the expression of p21CIP1/WAF1 in MCF-7 cells, causing cell cycle arrest in the G2/M phase through inhibition of cyclin B1 levels. Plumbagin also significantly increased the ratio of Bax/Bcl-2 and release of cytochrome c, resulting in apoptotic cell death in MCF-7 cells. Furthermore, plumbagin dramatically increased the intracellular ROS level, whereas pretreatment with the ROS scavenger N-acetyl cysteine protected against plumbagin-induced cytotoxicity, suggesting that ROS formation plays a pivotal role in antitumor activity in MCF-7 cells. In mice bearing MCF-7 cell xenografts, plumbagin significantly reduced tumor growth and weight without apparent side effects. We therefore concluded that plumbagin exerts anticancer activity against MCF-7 cells through the generation of intracellular ROS, resulting in the induction of apoptosis via a p53-dependent pathway. This study thus identifies a new anticancer mechanism of plumbagin against p53-dependent breast cancer cells and suggests a novel strategy for overcoming of breast cancer therapy.
    Keywords:  Anticancer; Apoptosis; Nepenthes alata; Plumbagin; Reactive oxygen species; p53
    DOI:  https://doi.org/10.1016/j.fct.2018.11.040
  26. Bioorg Chem. 2018 Nov 10. pii: S0045-2068(18)30884-8. [Epub ahead of print]83 438-449
      A moderate elevation in reactive oxygen species (ROS) levels can generally be controlled in normal cells, but may lead to death of cancer cells as the ROS level in cancer cells is already elevated. Therefore, a ROS-generating compound can act as a selective chemotherapeutic agent for cancer cells that does not affect normal cells. In our previous study, a compound containing a Michael acceptor was selectively cytotoxic to cancer cells without affecting normal cells; therefore, we designed and synthesized 26 compounds containing a Michael acceptor. Their cytotoxicities against HCT116 human colon cancer cell lines were measured by using a clonogenic long-term survival assay. To derive the structural conditions required to obtain stronger cytotoxicity against cancer cells, the relationships between the half-maximal cell growth inhibitory concentration values of the synthesized compounds and their physicochemical properties were evaluated by Comparative Molecular Field Analysis and Comparative Molecular Similarity Indices Analysis. It was confirmed that the compound with the best half-maximal cell growth inhibitory concentration triggered apoptosis through ROS generation, which then led to stimulation of the caspase pathway.
    Keywords:  1-aryl-(3-(2-styryl)phenyl)prop-2-en-1-one; Apoptosis; CoMFA; CoMSIA; Poly(ADP-ribose) polymerase; ROS generation
    DOI:  https://doi.org/10.1016/j.bioorg.2018.11.005
  27. Eur Rev Med Pharmacol Sci. 2018 Nov;pii: 16265. [Epub ahead of print]22(21): 7289-7295
      OBJECTIVE: The purpose of the study was to investigate the role of peroxidase V (Prx V) in Cisplatin-induced apoptosis of A549 cells and its underlying mechanism.MATERIALS AND METHODS: MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide) assay was conducted to evaluate the regulatory effect of Cisplatin on the survival of A549 cells. ROS (Reactive Oxygen Species) level in A549 cells induced with 0, 2, 4, and 6 mol/L Cisplatin for 24 h was determined using immunofluorescence. Apoptosis of Cisplatin-induced A549 cells was determined by immunofluorescence and flow cytometry, respectively. Western blot was performed to detect protein levels of Prx V, Bcl-2 (B-cell lymphoma 2), BAD ,and caspase-3 in Cisplatin-induced A549 cells.
    RESULTS: Survival rate of A549 cells gradually decreased with the increased dose of Cisplatin. Immunofluorescence results elucidated that cellular ROS level in Cisplatin-induced A549 cells increases in a dose-dependent manner. Both immunofluorescence and flow cytometry results revealed that the apoptotic rate of A549 cells increases with the elevation of Cisplatin dose. Besides, the apoptotic rate and ROS level of A549 cells were reduced by NAC pretreatment. Western blot results showed that the protein level of Prx V remarkably decreased in a dose-dependent manner, whereas Prx II expression did not change. With the treatment prolongation of 4 μmol/L Cisplatin in A549 cells, Bcl-2 and caspase-3 were downregulated, while BAD upregulated.
    CONCLUSIONS: Cisplatin treatment induces the ROS production, increases the apoptotic rate and downregulates the Prx expression in A549 cells.
    DOI:  https://doi.org/10.26355/eurrev_201811_16265
  28. Nanomedicine (Lond). 2018 Nov 20.
      AIM: The aim of this study was to encapsulate a ruthenium complex [Ru(ttbpy)2PIP](ClO4)2 (Ru) in liposomes to enhance their antitumor effect on human cervical cancer.METHODS: The Ru-loaded PEGylated liposomes (Ru-Lip) were prepared using thin-film hydration method. The mechanism of action was studied.
    RESULTS: A novel Ru was successfully synthesized. Ru-Lip showed stronger cytotoxic activity against HeLa cells than Ru. Ru-Lip demonstrated a more significant increase in apoptosis, reactive oxygen species production and apoptosis-associated processes (intracellular calcium concentration, cytochrome c release and activation of Bax and caspase-3) than Ru. Ru-Lip exhibited greater blockade efficacy in the cell cycle G1 phase and greater DNA damage than Ru.
    CONCLUSION: Ru-Lip significantly elevates the anticancer effect via reactive oxygen species-mediated mitochondrial dysfunctional pathway.
    Keywords:  anticancer activity; apoptosis; liposomes; mitochondria; ruthenium complex
    DOI:  https://doi.org/10.2217/nnm-2018-0236
  29. Ann Otol Rhinol Laryngol. 2018 Nov 17. 3489418812905
      BACKGROUND:: The pathology of chronic rhinosinusitis with nasal polyp (CRSwNP) is characterized by the infiltration of a large number of fibroblasts, resulting in extracellular matrix (ECM) deposition. Intralesional bleomycin A5 (BLE) injection has proved to be effective and safe, providing a novel treatment for CRSwNP. However, the mechanism is not clearly understood.OBJECTIVES:: The aim of this study is to explore the possible mechanism of BLE-induced apoptosis in nasal polyp-derived fibroblasts (NPDFs).
    MATERIAL AND METHODS:: Dichloro-dihydro-fluorescein diacetate probe, cell migration assays, and cell cycle analysis were used to detect the growth characteristics and basal reactive oxygen species (ROS) traits of NPDFs. Annexin V/propidium iodide and terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling assay was used to detect BLE-induced apoptosis. As a control, the antioxidant glutathione (GSH) was used to abrogate ROS induced by BLE. Western blot analysis was used to evaluate the effects of BLE on apoptosis and the ECM proteins of NPDFs.
    RESULTS:: The results showed that NPDFs had more active growth characteristics and higher basal ROS levels than normal nasal mucosa fibroblasts (NMFCs). NPDFs were more sensitive to BLE-induced apoptosis and ROS accumulation. GSH abrogation inhibits BLE-induced ECM degradation and apoptosis in NPDFs through a mitochondrial-mediated pathway.
    CONCLUSIONS:: BLE induced NPDF apoptosis and ECM degradation through a mitochondrial-mediated pathway and in a ROS-dependent manner.
    Keywords:  CRSwNP; NPDFs; ROS; apoptosis; bleomycin A5
    DOI:  https://doi.org/10.1177/0003489418812905
  30. Molecules. 2018 Nov 18. pii: E3016. [Epub ahead of print]23(11):
      Mitochondria-targeted cancer phototherapy (PT), which works by delivering photoresponsive agents specifically to mitochondria, is a powerful strategy to improve the phototherapeutic efficiency of anticancer treatments. Mitochondria play an essential role in cellular apoptosis, and are relevant to the chemoresistance of cancer cells. Furthermore, mitochondria are a major player in many cellular processes and are highly sensitive to hyperthermia and reactive oxygen species. Therefore, mitochondria serve as excellent locations for organelle-targeted phototherapy. In this review, we focus on the recent advances of mitochondria-targeting materials for mitochondria-specific PT. The combination of mitochondria-targeted PT with other anticancer strategies is also summarized. In addition, we discuss both the challenges currently faced by mitochondria-based cancer PT and the promises it holds.
    Keywords:  PDT; PTT; cancer therapy; nanomedicine; subcellular organelle-targeting
    DOI:  https://doi.org/10.3390/molecules23113016
  31. Onco Targets Ther. 2018 ;11 7841-7852
      Background: TRIM32 is overexpressed in several human cancers. However, its expression pattern, biological characteristics and mechanisms in human non-small cell lung cancer (NSCLC) have not been reported.Methods: We examined TRIM32 protein in 115 cases of NSCLC specimens. TRIM32 plasmid transfection and siRNA knockdown was carried out in NSCLC cell lines. AnnexinV/PI and JC-1 staining were performed to examine the change of apoptosis and mitochondrial membrane potential. Western blot was used to detect change of downstream proteins.
    Results: We found that TRIM32 protein was upregulated in 69 cases and positively correlated with advanced TNM stage. TRIM32 overexpression also correlated with poor survival of NSCLC patients. Biological assays demonstrated that TRIM32 overexpression promoted while it depletion inhibited cell growth, colony formation and invasion. In addition, TRIM32 maintained NSCLC cell viability and reduced apoptosis when treated with cisplatin. JC-1 and CellRox staining demonstrated that TRIM32 could maintain mitochondrial membrane potential and reduce Reactive Oxygen Species (ROS) production after cisplatin treatment. Western blot analysis showed that TRIM32 overexpression downregulated caspase 3 cleavage and cytochrome c release. TRIM32 also positively regulated Bcl-2 protein expression and NF-κB signaling. Inhibition of NF-κB abolished the effects of TRIM32 on Bcl-2.
    Conclusion: Taken together, our results indicated that TRIM32 is overexpressed in NSCLC and regulates cisplatin resistance, possibly through NF-κB and Bcl-2.
    Keywords:  NF-κB; NSCLC; TRIM32; apoptosis
    DOI:  https://doi.org/10.2147/OTT.S176689
  32. J Exp Clin Cancer Res. 2018 Nov 19. 37(1): 277
      BACKGROUND: The increase in the levels of reactive oxygen species (ROS) in acute myeloid leukemia (AML) patients has been previously described; thus, it is important to regulate ROS levels in AML.METHODS: Flow cytometry were used to assess the in vitro effect of compound kushen injection (CKI). Quantitative proteomics were used to analyse the mechanism. The AML patient-derived xenograft (PDX) model were used to evaluate the in vivo effect of CKI.
    RESULTS: We found that intracellular ROS levels in AML cells were decreased, the antioxidant capacity were increased when treated with CKI. CKI inhibited the proliferation of AML cells and enhanced the cytotoxicity of AML cells, which has few toxic effects on haematopoietic stem cells (HSCs) and T cells. At the single-cell level, individual AML cells died gradually by CKI treatment on optofluidic chips. CKI promoted apoptosis and arrested cell cycle at G1/G0 phase in U937 cells. Furthermore, higher peroxiredoxin-3 (Prdx3) expression levels were identified in CKI-treated U937 cells through quantitative proteomics detection. Mechanically, the expression of Prdx3 and peroxiredoxin-2 (Prdx2) was up-regulated in CKI-treated AML cells, while thioredoxin 1 (Trx1) was reduced. Laser confocal microscopy showed that the proteins Prdx2 could be Interacted with Trx1 by CKI treatment. In vivo, the survival was longer and the disease was partially alleviated by decreased CD45+ immunophenotyping in peripheral blood in the CKI-treated group in the AML PDX model.
    CONCLUSIONS: Antioxidant CKI possess better clinical application against AML through the Prdxs/ROS/Trx1 signalling pathway.
    Keywords:  Acute myeloid leukaemia; Compound kushen injection; Peroxiredoxin-2; Peroxiredoxin-3; Reactive oxygen species
    DOI:  https://doi.org/10.1186/s13046-018-0948-3
  33. Front Neurosci. 2018 ;12 802
      Abuse of methamphetamine (METH) results in neurological and psychiatric abnormalities. Lactulose is a poorly absorbed derivative of lactose and can effectively alleviate METH-induced neurotoxicity in rats. The present study was designed to investigate the effects of lactulose on METH-induced neurotoxicity. Rats received METH (15 mg/kg, 8 intraperitoneal injections, 12-h interval) or saline and received lactulose (5.3 g/kg, oral gavage, 12-h interval) or vehicle 2 days prior to the METH administration. Reactive oxygen species (ROS) and malondialdehyde (MDA) were measured. Protein levels of toll-like receptor 4 (TLR4), myeloid differentiation factor 88 (MyD88), tumor necrosis factor receptor associated factor 6 (TRAF6), nuclear factor κB (NFκB), interleukin (IL)-1β, IL-6, TNF-α, cleaved caspase 3, and poly(ADP-ribose) polymerase-1 (PARP-1) were determined by western blotting. mRNA expressions of nuclear factor erythroid 2-relatted factor-2 (Nrf2), p62, and heme oxygenase-1 (HO-1) were assessed by RT-qPCR. The lactulose pretreatment decreased METH-induced cytoplasmic damage in rat livers according to histopathological observation. Compared to the control group, overproduction of ROS and MDA were observed in rat striatums in the METH alone-treated group, while the lactulose pretreatment significantly attenuated the METH-induced up-regulation of oxidative stress. The lactulose pretreatment significantly repressed over-expressions of proteins of TLR4, MyD88, TRAF6, NFκB, IL-1β, IL-6, TNF-α, cleaved caspase 3, PARP-1. The lactulose pretreatment increased mRNA expressions of Nrf2, p62, and HO-1. These findings suggest that lactulose pretreatment can alleviate METH-induced neurotoxicity through suppressing neuroinflammation and oxidative stress, which might be attributed to the activation of the Nrf2/HO-1 axis.
    Keywords:  METH; Nrf2/HO-1 axis; lactulose; neuroinflammation; oxidative stress
    DOI:  https://doi.org/10.3389/fnins.2018.00802
  34. Int J Radiat Oncol Biol Phys. 2018 Nov 17. pii: S0360-3016(18)34007-0. [Epub ahead of print]
      PURPOSE/OBJECTIVE: To evaluate the protective effect of inorganic nitrate against systemic damage in a mouse model of total body gamma irradiation (TBI).METHODS/MATERIALS: C57BL/6 mice in the IR (irradiation) + NaNO3 group were pretreated with 2 mmol/L NaNO3 in their drinking water for one week before receiving 5 Gy irradiation. Animals that received only 5 Gy irradiation were designated as the IR group. Survival and body weight were monitored. The peripheral blood lymphocytes (PBL), heart, liver, lung, and submandibular gland, were harvested and assessed. Reactive oxygen species (ROS) were measured in the lung and submandibular gland. Phosphorylated histone H2AX (p-H2AX) and p53-binding protein 1 (53BP1), as markers of early stage DNA damage, as well as terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) and Bax/Caspase 3 mRNA expression, as markers of apoptosis, were examined.
    RESULTS: No improvement of survival was observed in the IR + NaNO3 group after TBI, but the body weight loss after 5 Gy TBI was significantly attenuated in the IR + NaNO3 group. The levels of peripheral blood erythrocytes, leukocytes, and platelets at 7 days post-irradiation recovered with nitrate treatment; moreover, the p-H2AX level in the PBL was much lower in the IR + NaNO3 group at 2 h and 4 h post-irradiation. In the lung and submandibular gland, the levels of p-H2AX, 53BP1, and ROS as well as TUNEL staining were significantly decreased in the IR + NaNO3 group compared to those in the IR group. Gene expression of Bax and caspase 3 was decreased in both the lung and submandibular gland with nitrate treatment, indicating attenuation of apoptosis.
    CONCLUSION: Inorganic nitrate delivery could effectively prevent TBI-induced systemic damage. Nitrate-mediated decreases in ROS levels may contribute to this systemic protective effect.
    Keywords:  Apoptosis; DNA damage; Inorganic nitrate; Reactive oxygen species (ROS); Total body irradiation (TBI)
    DOI:  https://doi.org/10.1016/j.ijrobp.2018.11.021
  35. Biochem Biophys Res Commun. 2018 Nov 16. pii: S0006-291X(18)32358-1. [Epub ahead of print]
      Oral squamous cell carcinoma (OSCC) is known as one of the most common cancer influencing the head and neck region. However, the molecular mechanisms revealing OSCC progression is largely unclear. Aurora kinase A (AURKA) is a serine-threonine kinase that functions in mitotic spindle formation and chromosome segregation, and is associated with the progression of human cancers. But its role in regulating OSCC development has not yet been investigated. In the study, we found that AURKA expression was up-regulated in OSCC cell lines and tumor specimens from patients. OSCC patients with high expression of AURKA exhibited a significant decreased overall survival rate. In vitro, AURKA knockdown markedly reduced the proliferation, migration and invasion of OSCC cells using cell counting kit-8 (CCK-8), EdU, colony formation and transwell analysis. EMT was suppressed by AURKA silence, as evidenced by the up-regulated expression of E-cadherin and down-regulated Vimentin in OSCC cells. In addition, apoptosis was markedly induced by AURKA inhibition through promoting the expression of cleaved Caspase-3 and poly (ADP)-ribose polymerase (PARP). Reactive oxygen species (ROS) production was also markedly enhanced in AURKA-knockdown OSCC cells. Importantly, we found that repressing ROS generation using its scavenger of n-acetylcysteine (NAC) significantly abolished AURKA silence-induced apoptosis, accompanied with restored proliferation and EMT. In vivo, AURKA knockdown notably inhibited tumor growth. Therefore, knockdown of AURKA suppressed cell proliferation, migration and invasion, and also induced apoptosis and ROS generation in OSCC possibly via the production of ROS, demonstrating that AURKA inhibition might represent a novel therapeutic target for the prevention of OSCC.
    Keywords:  AURKA; Apoptosis; EMT; ROS generation; oral squamous cell carcinoma (OSCC)
    DOI:  https://doi.org/10.1016/j.bbrc.2018.10.170
  36. Toxicol In Vitro. 2018 Nov 14. pii: S0887-2333(18)30692-1. [Epub ahead of print]
      Methylglyoxal (MG) is a α-dycarbonyl compound derived mainly from glycolysis, whose accumulation is harmful for cells and tissues. Here, we evaluated the cytotoxic effects induced by MG in leukocytes after an acute exposure, measuring as endpoints of toxicity some markers of oxidative stress and programmed cell death. Human leukocytes were isolated and incubated with MG at concentrations ranging from 0.1 to 10 mM for 2.5 h, and subsequently prepared for assays based in flow cytometry, gene expression and immunoreactivity profile. The cells exposed to higher concentrations of MG had significant loss of viability, increased reactive species (RS) production and apoptosis/necrosis rate. These phenomena were accompanied by morphological changes (increased size and granularity) and disruption in mRNA expression of antioxidant, apoptotic and glycation-responsive genes, particularly: Nrf2 (Nuclear factor (erythroid-derived 2)-like 2), SOD1 (CuZn-superoxide dismutase), SOD2 (Mn-superoxide dismutase), GSR (glutathione-S-reductase), BAX (BAX-associated X protein), BCL-2 (BCL-2-associated X protein), AIF (apoptosis inducing factor), GLO-1 (glyoxalase-1) and RAGE (receptor for advanced glycation end products). The mRNA expression of CASP 9 and CASP 3 (caspase-9 and 3) as well as the immunoreactivity of proteins were not changed by MG. Collectively, our data provide evidence that MG activates programmed cell death pathways in leukocytes and that this effect seems to be associated with disturbances in cell redox signaling.
    Keywords:  Apoptosis; Leukocytes; Methylglyoxal; Oxidative stress
    DOI:  https://doi.org/10.1016/j.tiv.2018.11.001
  37. Life Sci. 2018 Nov 20. pii: S0024-3205(18)30767-7. [Epub ahead of print]
      AIMS: Spinal cord injury (SCI) is a damage of spinal cord caused by trauma or diseases. Here, we explored the effects of tanshinone IIA (Tan IIA) on SCI oxidative damage in vitro and in vivo.MATERIALS AND METHODS: In vitro, PC-12 cells were treated by H2O2 to stimulate oxidative injury. Then, the effects of Tan IIA on cell viability, apoptosis and autophagy were assessed by CCK-8 assay, flow cytometry assay and western blotting, respectively. The expression of miR-124 was measured by qRT-PCR, and whether Tan IIA exerted effects on H2O2-treated PC-12 cells through modulating miR-124 was verified. In vivo, Sprague-Dawley rats were induced SCI using a weight drop device. Then, the effects of Tan IIA on motor function recovery of rats, myeloperoxidase (MPO) activity in damaged tissue and tissue cell proliferation, apoptosis and autophagy were investigated, respectively.
    KEY FINDINGS: In vitro, H2O2 stimulation reduced cell viability, and induced cell apoptosis and autophagy. Those alterations were mitigated by Tan IIA treatment. Tan IIA treatment reversed the H2O2-induced down-regulation of miR-124. Silence of miR-124 reversed the effects of Tan IIA on H2O2-treated PC-12 cells, as well as activation of JNK and p38MAPK pathways. In vivo, Tan IIA treatment alleviated the SCI-induced enhancement of MPO activity in damaged tissue, apoptosis and autophagy of damaged tissue cells, and promoted the motor function recovery of rats.
    SIGNIFICANCE: Tan IIA attenuated oxidative damage after SCI in vitro and in vivo might be through up-regulating miR-124 and then inactivating JNK and p38 MAPK pathways.
    Keywords:  JNK/p38 MAPK; Oxidative stress; Spinal cord injury; Tanshinone IIA; miR-124
    DOI:  https://doi.org/10.1016/j.lfs.2018.11.046
  38. Exp Dermatol. 2018 Nov 20.
      Chemotherapy and radiotherapy are common modalities for cancer treatment. While targeting rapidly growing cancer cells, they also damage normal tissues and cause adverse effects. From the initial insult such as DNA double strand break, production of reactive oxygen species (ROS), and a general stress response, there are complex regulatory mechanisms that control the actual tissue damage process. Besides apoptosis, a range of outcomes for the damaged cells are possible including cell cycle arrest, senescence, mitotic catastrophe, and inflammatory responses and fibrosis at the tissue level. Feather and hair are among the most actively proliferating (mini-)organs, and are highly susceptible to both chemotherapy and radiotherapy damage, thus provide excellent, experimentally tractable model systems for dissecting how normal tissues respond to such injuries. Taking a comparative biology approach to investigate this has turned out to be particularly productive. Started in chicken feather and then extended to murine hair follicles, it was revealed that in addition to p53-mediated apoptosis, several other previously overlooked mechanisms are involved. Specifically, Shh, Wnt, mTOR, cytokine signaling, and ROS-mediated degradation of adherens junctions have been implicated in the damage and/or reparative regeneration process. Moreover, we show here that inflammatory responses, which can be prominent upon histological examination of chemo- or radiotherapy-damaged hair follicle, may not be essential for the hair loss phenotype. These studies point to fundamental, evolutionarily conserved mechanisms in controlling tissue responses in vivo, and suggest novel strategies for the prevention and management of adverse effects that arise from chemo- or radiotherapy. This article is protected by copyright. All rights reserved.
    Keywords:  adverse effect; chemotherapy; feather; hair; radiotherapy
    DOI:  https://doi.org/10.1111/exd.13846
  39. Chem Biol Interact. 2018 Nov 16. pii: S0009-2797(18)30634-3. [Epub ahead of print]
      Oenothein B has a wide range of biological activities. This study assessed the possible mechanism of Oenothein B on lung cancer A549 cell. The results showed that Oenothein B effectively inhibited the proliferation of A549 cells by inducing apoptosis, arrested cell at G1 stage. On the one hand, Oenothein B not only increased the level of intracellular reactive oxygen species (ROS), but induced some apoptotic-related protein expression (cleavage caspase-3, PARP, cytochrome c level in the cytoso, Bax). Moreover, ROS inhibitor and PI3K agonist exhibited significant protection against cell death by Oenothein B. ROS inhibitor significantly abrogated the activation of caspase3/7 and 9 by Oenothein B. On the other hand, the levels of p-Akt and p-Akt, p-NF-κB suppressed by Oenothein B could be offset by treatment with ROS inhibitor. To summarize, these results demonstrated that Oenothein B was able to prevent cell growth by maybe via ROS-mediated PI3K/Akt/NF-κB signaling pathway.
    Keywords:  A549 cell; NF-kB; Oenothein B; PI3K/Akt; ROS
    DOI:  https://doi.org/10.1016/j.cbi.2018.09.021
  40. Nat Prod Res. 2018 Nov 23. 1-4
      Schisandra propinqua subsp. sinensis is a traditional medicinal plant used in Chinese folk medicine. Melanoma is the most dangerous form of skin cancer. To discover bioactive phytochemicals for preventing human melanoma, we have investigated the inhibitory effects of schisantherin F in Schisandra propinqua subsp. sinensis on human melanoma A375 cells and relevant mechanisms. The results showed that schisantherin F can inhibit A375 cells through inducing apoptosis. Further investigations have demonstrated schisantherin F attenuated the overproduction of ROS, depolarization of MMP, and mPTP opening. Meanwhile, schisantherin F inhibited the activity of Caspase-3 and up-stream Caspase-9, down-regulated Bcl-2 and up-regulated Bax. These findings propose the inhibitory mechanisms of schisantherin F in A375 cells include induction of mitochondrial dysfunction and mitochondria-mediated apoptosis.
    Keywords:  Schisandra propinqua subsp. sinensis; apoptosis; human melanoma A375 cell; mitochondrial dysfunction; schisantherin F
    DOI:  https://doi.org/10.1080/14786419.2018.1509332
  41. Biogerontology. 2018 Nov 19.
      The phytochemical epigallocatechin gallate (EGCG) has been reported to alleviate age-associated immune disorders and organ dysfunction. However, information regarding the mechanistic role of EGCG in the suppression of cellular senescence is limited. The present study thus assessed the effects and underlying mechanisms of EGCG in the inhibition of senescence as well as its potential to selectively eliminate senescent cells (senolytics) using 3T3-L1 preadipocytes. Premature senescence was established in cells by repeated exposure of H2O2 at a sub-lethal concentration (150 μM). H2O2 treated cells showed characteristic senescence-associated features including increased cell size, senescence-associated β-galactosidase activity (SA-β-gal), development of senescence-associated secretory phenotype (SASP), activation of reactive oxygen species (ROS) and pathways, DNA damage as well as induction of cell cycle inhibitors (p53/p21WAF1/p16INK4a). In addition, a robust activation of PI3K/Akt/mTOR and AMPK pathways was also observed in H2O2 treated cells. Presence of EGCG (50 and 100 μM) showed significant downregulation of PI3K/Akt/mTOR and AMPK signaling along with the suppression of ROS, iNOS, Cox-2, NF-κB, SASP and p53 mediated cell cycle inhibition in preadipocytes. In addition, EGCG treatment also suppressed the accumulation of anti-apoptotic protein Bcl-2 in senescent cells thereby promoting apoptosis mediated cell death. Our results collectively show that EGCG acts as an mTOR inhibitor, SASP modulator as well as a potential senolytic agent thereby indicating its multi-faceted attributes that could be useful for developing anti-aging or age-delaying therapies.
    Keywords:  EGCG; MTOR; ROS; Senescence; Senolytic
    DOI:  https://doi.org/10.1007/s10522-018-9785-1
  42. Phytomedicine. 2018 Dec 01. pii: S0944-7113(18)30514-2. [Epub ahead of print]51 128-138
      BACKGROUND: Similar to the leaves of P. Quinquefolius, American ginseng berry (AGB) is another important part of P. Quinquefolius with alternative therapeutic potential. The liver protection capabilities of the former have been demonstrated previously, however, the later has not yet been evaluated.PURPOSE: Based on our previous observation, the present work was designed to evaluate the hepatic protective effects for novel mechanisms of AGB in acetaminophen (APAP)-induced liver injury in vivo.
    STUDY DESIGN/METHODS: All mice were divided into four groups as follows: normal group, APAP group and APAP + AGB (150 mg/kg and 300 mg/kg) groups. AGB were orally administered for one week before exposure to APAP (250 mg/kg). Severe liver injury was observed and hepatotoxicity was evaluated after 24 h through evaluating the biochemical markers, protein expressions levels and liver histopathology.
    RESULTS: Our study results clearly demonstrated that AGB pretreatment ameliorated APAP-induced hepatic injury as evidenced by decreasing plasma alanine aminotransferase (ALT), aspartate transaminase (AST), tumor necrosis factor α (TNF-α) and interleukin-1β (IL-1β) compared to the APAP group. Western blotting analysis showed that pretreatment with AGB decreased the expressions levels of TNF-α and nuclear transcription factor-κB (NF-κB p65) in liver tissues. Meanwhile, the protein expression levels of caspases, cytochrome c, and Bax were elevated by AGB treatment for seven days, while the protein expression level of Bcl-2 was inhibited comparison with that in APAP group. Furthermore, supplement of AGB resulted in increase of superoxide dismutase (SOD) and glutathione (GSH), while decrease of malondialdehyde (MDA) content and the expression levels of 4-hydroxynonenal (4-HNE) and cytochrome P450 E1 (CYP2E1). The results of histopathological staining demonstrated that AGB pretreatment inhibited APAP-induced hepatocyte infiltration, congestion, and necrosis.
    CONCLUSION: The present study demonstrated that AGB pretreatment protected liver cells against APAP-induced hepatotoxicity through inhibition of oxidative stress, inflammation responses via TNF-α-mediated caspase-3/-8/-9 signaling pathways.
    Keywords:  APAP-induced liver injury; American ginseng berry; Anti-apoptosis; Anti-inflammation; Oxidative stress
    DOI:  https://doi.org/10.1016/j.phymed.2018.09.234