bims-aporos Biomed News
on Apoptosis and reactive oxygen species
Issue of 2019‒02‒17
fifty-four papers selected by
Gavin McStay
Staffordshire University


  1. J Inorg Biochem. 2019 Feb 01. pii: S0162-0134(18)30366-0. [Epub ahead of print]193 143-151
      Aluminum (Al) causes hippocampal lesions by oxidative stress, which is widely accepted as the primary pathogenesis of Al neurotoxicity. Lycopene (LYC), a naturally carotenoid, has received extensive attention due to its antioxidant effect. In this study, the neuroprotective effects and mechanisms of LYC against aluminum chloride (AlCl3)-induced hippocampal lesions were explored. First, oral administration of LYC (4 mg/kg) alleviated AlCl3-induced (150 mg/kg) cognition impairment and histopathological changes of the hippocampus in rats. Then, LYC significantly attenuated AlCl3-induced oxidative stress, presenting as the reduced reactive oxygen species, malondialdehyde and 8-hydroxy-2'-deoxyguanosine levels, and increased glutathione level and superoxide dismutase activity. Moreover, LYC also protected the hippocampus from AlCl3-induced apoptosis and neuroinflammation, as assessed by protein levels of p53, Bcl-2-associated X protein (Bax), B-cell lymphoma gene 2 (Bcl-2), Cytochrome c (Cyt c), cleaved caspase-3 and nuclear factor kappa B, as well as the mRNA levels of Bax, Bcl-2, tumor necrosis factor alpha, interleukin-6 and interleukin-1 beta. Finally, LYC increased nuclear factor-erythroid-2-related factor 2 (Nrf2) nuclear translocation and its downstream gene expression, including heme oxygenase-1, NAD(P)H: quinone oxidoreductase 1, glutamate cysteine ligase catalytic subunit and superoxide dismutase 1, which were involved in antioxidant, anti-apoptosis, and anti-inflammation. Overall, our findings demonstrate LYC attenuates Al-induced hippocampal lesions by inhibiting oxidative stress-mediated inflammation and apoptosis in the rat.
    Keywords:  Aluminum chloride; lycopene; hippocampal lesions;Nrf2;inflammation; apoptosis
    DOI:  https://doi.org/10.1016/j.jinorgbio.2019.01.017
  2. J Mol Neurosci. 2019 Feb 12.
      Curcumin protects neuronal cells exposed to β amyloid (Aβ); the mechanism, however, is still obscure. The aim of this study is to determine whether the type 2 superoxide dismutase (SOD2) mediates curcumin-induced protective effects in Aβ-treated neuronal cells. In this study, the HT22 neuronal cells were exposed to Aβ to imitate neuronal injury in Alzheimer's disease (AD). After 24-h treatment, 10 μM Aβ decreased cell viability and mitochondrial functions, including mitochondrial complex activities and mitochondrial membrane potential (MMP), and also downregulated anti-oxidants SOD2, glutathione (GSH), and catalase (CAT) levels (P < 0.05), meanwhile, increased lactic dehydrogenase (LDH) release, apoptosis level, intracellular reactive oxygen species (ROS) and mitochondrial superoxide accumulation (P < 0.05). And, co-administration of 1 μM curcumin significantly reduced the Aβ-induced cell injury and oxidative damage above (P < 0.05). Downregulating SOD2 by using small interfering RNA (siRNA), however, significantly abolished the curcumin-induced protective and anti-oxidative effects in HT22 cells (P < 0.05); the scramble (SC)-siRNA did not cause marked effects on the curcumin-induced protective effects (P > 0.05). These findings showed that curcumin can alleviate Aβ-induced injury in neuronal cells, and SOD2 protein may mediate the neuroprotective effects.
    Keywords:  Alzheimer’s disease; Curcumin; Neuroprotection; SOD2; β amyloid
    DOI:  https://doi.org/10.1007/s12031-019-01267-2
  3. Int J Mol Sci. 2019 Feb 11. pii: E754. [Epub ahead of print]20(3):
      Medicinal herbal plants have been commonly used for intervention in different diseases and improvement of health worldwide. Koumine, an alkaloid monomer found abundantly in Gelsemium plants, can be effectively used as an antioxidant. The purpose of this study was to evaluate the potential protective effect of koumine against hydrogen peroxide (H₂O₂)-induced oxidative stress and apoptosis in porcine intestinal epithelial cell line (IPEC-J2 cells). MTT assays showed that koumine significantly increased cell viability in H₂O₂-mediated IPEC-J2 cells. Preincubation with koumine ameliorated H₂O₂-medicated apoptosis by decreasing reactive oxygen species (ROS) production, and efficiently suppressed the lactate dehydrogenase (LDH) release and malondialdehyde (MDA) production. Moreover, a loss of superoxide dismutase (SOD), catalase (CAT) and glutathione (GSH) activities was restored to normal level in H₂O₂-induced IPEC-J2 cells upon koumine exposure. Furthermore, pretreatment with koumine suppressed H₂O₂-mediated loss of mitochondrial membrane potential, caspase-9 and caspase-3 activation, decrease of Bcl-2 expression and elevation of Bax expressions. Collectively, the results of this study indicated that koumine possesses the cytoprotective effects in IPEC-J2 cells during exposure to H₂O₂ by suppressing production of ROS, inhibiting the caspase-3 activity and influencing the expression of Bax and Bcl-2. Koumine could potentially serve as a protective effect against H₂O₂-induced apoptosis.
    Keywords:  H2O2; IPEC-J2 cells; apoptosis; koumine
    DOI:  https://doi.org/10.3390/ijms20030754
  4. Life Sci. 2019 Feb 12. pii: S0024-3205(19)30115-8. [Epub ahead of print]
      AIMS: Edaravone potentially alleviates cognitive deficits in a mouse model of Alzheimer's disease (AD). However, the mechanism of edaravone in suppressing AD progression remains unclear. We aim to investigate the mechanism of edaravone in suppressing oxidative stress-mediated AD progression in vitro.MAIN METHODS: Human neuroblastoma SH-SY5Y cells were pretreated with different concentrations of edaravone prior to the induction by Aβ25-35. Cell viability, apoptosis, reactive oxygen species, and expression of antioxidative response elements (ARE) including Nrf2, SOD, and HO-1 were assessed.
    KEY FINDINGS: The results showed that apoptosis and reactive oxygen species levels significantly increased in Aβ25-35-treated cells, whereas the mRNA and protein levels of Nrf2, SOD and HO-1 decreased. The opposite changes were observed in cells that were pre-treated with edaravone, particularly at a concentration of 40 μM. Aβ25-35-treatment suppressed Nrf2 expression and nuclear translocation were rescued by Edaravone. Genetic inhibition of Nrf2 greatly decreased the protective effect of edaravone against cell apoptosis and cytotoxicity induced by Aβ25-35, accompanied by decreases in SOD and HO-1 expression.
    SIGNIFICANCE: Activation of the Nrf2/ARE signaling pathway may underlie the protective effects of edaravone against the oxidative damage associated with Alzheimer's disease.
    Keywords:  Alzheimer's disease; Aβ(25–35); Edaravone; Nrf2/ARE; Oxidative damage
    DOI:  https://doi.org/10.1016/j.lfs.2019.02.025
  5. J Cell Physiol. 2019 Feb 14.
      OBJECTIVE: To study the role and mechanism of microRNA 19b (miR-19b) in hypoxia/reoxygenation (H/R)-induced injury by targeting PTEN.METHODS: PC12 and BV2 cells induced by H/R were treated with miR-19b mimics/inhibitors or small interfering PTEN (si-PTEN), respectively. Lactate dehydrogenase (LDH) level, malondialdehyde (MDA), and superoxide dismutase (SOD) content was detected. Besides, cell viability and apoptosis were determined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide, Hoechst33342 staining, and flow cytometry, whereas mitochondrial membrane potential (MMP) tested by JC-1 assay, and reactive oxygen species (ROS) evaluated by the dichloro-dihydro-fluorescein diacetate assay. The ischemia/reperfusion (I/R) rats model was used to investigate the effects of miR-19b in vivo test. The infarct area and apoptosis rates in brain tissues were detected by 2,3,5-triphenyltetrazolium chloride and terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling staining, respectively. miR-19b and PTEN/PI3K/Akt pathway-related proteins were detected by quantitative reverse-transcription polymerase chain reaction and western blot analysis.
    RESULTS: miR-19b mimics could reduce LDH, MDA, and ROS levels and decline cell apoptosis, but enhance the viability, MMP, and SOD activity with decreased PTEN and cleaved caspase, as well as increased p-Akt/Akt and Bcl-2/Bax ratios in H/R-induced PC12 and BV2 cells. However, miR-19b inhibitors led to completely opposite results to aggravate H/R-induced cell injury. Meanwhile, si-PTEN could reverse the effect of miR-19b inhibitors on H/R-induced injury. Moreover, treatment with miR-19b agomir after I/R in vivo sufficiently decreased infarct area and reduced apoptosis rates by targeting PTEN through the regulation of the PI3K/Akt pathway.
    CONCLUSION: miR-19b could inhibit oxidative stress, enhance cell MMP, promote cell survival, and inhibit cell apoptosis by targeting PTEN via the regulation of the PI3K/Akt pathway, thus playing the neuronal protective effects.
    Keywords:  PC12 cells; PI3K/Akt signaling pathway; PTEN; hypoxia/reoxygenation; miR-19b
    DOI:  https://doi.org/10.1002/jcp.28286
  6. Food Chem Toxicol. 2019 Feb 11. pii: S0278-6915(19)30074-2. [Epub ahead of print]
      Erianin is the major bibenzyl compound found in Dendrobium chrysotoxum Lindl. The current study was designed to investigate the protective effects of erianin on high glucose-induced injury in cultured renal tubular epithelial cells (NRK-52E cells) and determine the possible mechanisms for its effects. NRK-52E cells were pretreated with erianin (5, 10, 25 or 50 nmol/L) for 1 h followed by further exposure to high glucose (30 mmol/L, HG) for 48 h. Erianin concentration dependently enhanced cell viability followed by HG treatment in NRK-52E cells. HG induced reactive oxygen species (ROS) generation, malondialdehyde production, and glutathione deficiency were recovered in NRK-52E cells pretreated with erianin. HG triggered cell apoptosis via the loss of mitochondrial membrane potential, depletion of adenosine triphosphate, upregulation of caspases 9 and 3, enhancement of cytochrome c release, and subsequent interruption of the Bax/Bcl-2 balance. These detrimental effects were ameliorated by erianin. HG also induced activation of p53, JNK, p38 mitogen-activated protein kinase (MAPK) and nuclear factor-κB (NF-κB) in NRK-52E cells, which were blocked by erianin. The results suggest that treatment NRK-52E cells with erianin halts HG-induced renal dysfunction through the suppression of the ROS/MAPK/NF-κB signaling pathways. Our findings provide novel therapeutic targets for diabetic nephropathy.
    Keywords:  Diabetic nephropathy; Erianin; High glucose; Mitogen-activated protein kinase; NF-κB; NRK-52E cells
    DOI:  https://doi.org/10.1016/j.fct.2019.02.021
  7. Food Chem Toxicol. 2019 Feb 12. pii: S0278-6915(19)30075-4. [Epub ahead of print]
      Arsenic contaminated drinking water consumption is a serious health issue around the world. Chronic inorganic arsenic exposure has been associated with respiratory dysfunctions. It exerts various detrimental effects, disrupting normal cellular homeostasis and turning on severe pulmonary complications. This study elucidated the role of mangiferin, a natural xanthone, against arsenic induced lung toxicity. Chronic exposure of sodium arsenite (NaAsO2) at 10 mg/kg bw for 3 months abruptly increased the LDH release in broncho-alveolar lavage fluid, generated reactive oxygen species (ROS), impaired the antioxidant defense and distorted the alveoli architecture. It caused significant inflammatory outburst and promoted the apoptotic mode of cell death via upregulating the expressions of various proapoptotic molecules related to mitochondrial, extra-mitochondrial and ER stress mediated apoptotic pathway. Activation of inflammatory cascade led to disruption of alveolar capillary barrier and impaired Na+/K+-ATPase function that led to detaining of alveolar fluid clearance activity. Mangiferin due to its anti-inflammatory activity suppressed this inflammation and reduced inflammatory cell infiltration in lung tissue. It significantly restored the antioxidant balance and inhibited apoptosis in lung via upregulating Nrf2-HO1 axis.
    Keywords:  Apoptosis; Arsenic; Inflammation; Lung; Mangiferin; Oxidative stress
    DOI:  https://doi.org/10.1016/j.fct.2019.02.022
  8. Biomolecules. 2019 Jan 29. pii: E47. [Epub ahead of print]9(2):
      Silver-based nanostructures are suitable for many biomedical applications, but to be useful therapeutic agents, the high toxicity of these nanomaterials must be eliminated. Here, we biosynthesize nontoxic and ultra-small silver nanoclusters (rsAg@NCs) using metabolites of usnioid lichen (a symbiotic association of algae and fungi) that exhibit excellent antimicrobial activity against fluconazole (FCZ)-resistant Candida albicans that is many times higher than chemically synthesized silver nanoparticles (AgNPs) and FCZ. The rsAg@NCs trigger apoptosis via reactive oxygen species accumulation that leads to the loss of mitochondrial membrane potential, DNA fragmentation, chromosomal condensation, and the activation of metacaspases. The proteomic analysis clearly demonstrates that rsAg@NCs exposure significantly alters protein expression. Most remarkable among the down-regulated proteins are those related to glycolysis, metabolism, free radical scavenging, anti-apoptosis, and mitochondrial function. In contrast, proteins involved in plasma membrane function, oxidative stress, cell death, and apoptosis were upregulated. Eventually, we also established that the apoptosis-inducing potential of rsAg@NCs is due to the activation of Ras signaling, which confirms their application in combating FCZ-resistant C. albicans infections.
    Keywords:  Ras signaling pathway; apoptosis; biosilver nanoclusters; fluconazole-resistant Candida albicans; oxidative stress; proteomics
    DOI:  https://doi.org/10.3390/biom9020047
  9. Nutr Cancer. 2019 Feb 09. 1-10
      Recent evidence provides that seafood has a lot of health benefits due to its unique bioactive compounds. Sea squirt is widely cultured and consumed as a foodstuff in Korea; however, seldom reports with reference to bioactivities are available until now. In this study, edible part of sea squirt was hydrolyzed by pepsin and its hydrolysates was evaluated for anticancer effect on human colon cancer HT-29 cells. Sea squirt hydrolysates (SSQ) reduced HT-29 cell viability. Treatment with SSQ resulted in the increase in reactive oxygen species (ROS) generation followed by disruption of mitochondrial membrane potential (MMP). Flow cytometry analysis revealed that SSQ induced G2/M phase arrest and apoptosis evidenced by Hoechst 33342 staining. Levels of mRNA expression by real-time polymerase chain reaction (PCR) showed that treatment with SSQ in HT-29 cells upregulated expression of p53, bax, and caspase-3 genes and downregulated expression of bcl-2 gene. Protein level of cytochrome c into cytosol and caspase-3 by Western blotting were also increased by treatment with SSQ in HT-29 cells. These results suggest that SSQ may be useful for functional food ingredients and/or nutraceuticals.
    DOI:  https://doi.org/10.1080/01635581.2018.1540717
  10. Cell Death Dis. 2019 Feb 15. 10(3): 147
      Fhit protein is lost in cancers of most, perhaps all, cancer types; when restored, it can induce apoptosis and suppress tumorigenicity, as shown in vitro and in mouse tumor models in vivo. Following protein cross-linking and proteomics analyses, we characterized a Fhit protein complex involved in triggering Fhit-mediated apoptosis. The complex includes the heat-shock chaperonin pair, HSP60/10, which is likely involved in importing Fhit into the mitochondria, where it interacts with ferredoxin reductase, responsible for transferring electrons from NADPH to cytochrome P450 via ferredoxin, in electron transport chain complex III. Overexpression of Fhit protein in Fhit-deficient cancer cells modulates the production of intracellular reactive oxygen species, causing increased ROS, following peroxide treatment, with subsequent increased apoptosis of lung cancer cells under oxidative stress conditions; conversely, Fhit-negative cells escape ROS overproduction and ROS-induced apoptosis, likely carrying oxidative damage. Thus, characterization of Fhit-interacting proteins has identified direct effectors of a Fhit-mediated apoptotic signal pathway that is lost in many cancers. This is of translational interest considering the very recent emphasis in a number of high-profile publications, concerning the role of oxidative phosphorylation in the treatment of human cancers, and especially cancer stem cells that rely upon oxidative phosphorylation for survival. Additionally, we have shown that cells from a Fhit-deficient lung cancer cell line, are sensitive to killing by exposure to atovaquone, thought to act as a selective oxidative phosphorylation inhibitor by targeting the CoQ10 dependence of the mitochondrial complex III, while the Fhit-expressing sister clone is resistant to this treatment.
    DOI:  https://doi.org/10.1038/s41419-019-1414-7
  11. Environ Toxicol. 2019 Feb 13.
      We aim to confirm the impairment of chlorpyrifos (CPF) in PC12 cells, evaluate the protective effect of edaravone on CPF-induced injury, and try to unravel its underlying mechanism perspective from Nrf2 signaling pathway. Viability of PC12 cells treated with CPF and edaravone (Ed) were evaluated by MTT assay. Cell apoptosis was observed by the Hoechst 33342 stain. The level of reactive oxygen species (ROS), the content of malondialdehyde (MDA), and the activity of superoxide dismutase (SOD) were detected to evaluate the oxidative stress injury. The expression of Nrf2 was detected by Western blot; profoundly, RNA interference was conducted to construct Nrf2 gene knockdown PC12 cells and to uncover its underlying mechanism. MTT results showed CPF injured PC12 cells in a concentration-dependent manner. Increased ROS and MDA content, decreased total SOD activity, or even apoptosis were occurred in PC12 cells when treated with CPF. Interestingly, CPF-induced cell injury was conspicuously reversed after Ed administration. Nrf2 signaling pathway was activated after Ed treatment and the neuroprotective effect of Ed was not significant in cells after Nrf2 gene knockdown. In conclusion, Ed exerts neuroprotective effect on CPF-induced oxidative stress injury and its mechanism was correlated with the Nrf2 signaling pathway.
    Keywords:  Nrf2; PC12 cells; chlorpyrifos; edaravone; oxidative stress
    DOI:  https://doi.org/10.1002/tox.22728
  12. Mol Cells. 2019 Nov 27.
      The omega-3 fatty acid docosahexaenoic acid (DHA) is known to induce apoptosis and cell cycle arrest via the induction of reactive oxygen species (ROS) production and endoplasmic reticulum (ER) stress in many types of cancers. However, the roles of DHA in drug-resistant cancer cells have not been elucidated. In this study, we investigated the effects of DHA in cisplatin-resistant gastric cancer SNU-601/cis2 cells. DHA was found to induce ROS-dependent apoptosis in these cells. The inositol 1,4,5-triphosphate receptor (IP3R) blocker 2-aminoethyl diphenylboninate (2-APB) reduced DHA-induced ROS production, consequently reducing apoptosis. We also found that G-protein-coupled receptor 120 (GPR120), a receptor of long-chain fatty acids, is expressed in SNU-601/cis2 cells, and the knockdown of GPR120 using specific shRNAs alleviated DHA-mediated ROS production and apoptosis. GPR120 knockdown reduced the expression of ER stress response genes, similar to the case for the pre-treatment of the cells with N-acetyl-L-cysteine (NAC), an ROS scavenger, or 2-APB. Indeed, the knockdown of C/EBP homologous protein (CHOP), a transcription factor that functions under ER stress conditions, markedly reduced DHA-mediated apoptosis, indicating that CHOP plays an essential role in the anti-cancer activity of DHA. These results suggest that GPR120 mediates DHA-induced apoptosis by regulating IP3R, ROS, and ER stress levels in cisplatin-resistant cancer cells, and that GPR120 is an effective chemotherapeutic target for cisplatin resistance.
    DOI:  https://doi.org/10.14348/molcells.2019.2440
  13. Chem Biol Interact. 2019 Feb 06. pii: S0009-2797(18)30618-5. [Epub ahead of print]
      NF-E2 p45-related factor 2 (Nrf2), which regulates the cellular antioxidant response, is a target for limiting tissue damage due to exposure to environmental toxicants, including arsenic. Daphnetin (Daph), a natural coumarin derivative, has been shown to induce remarkable antioxidant activity. The present study aimed to examine the protective effects and molecular mechanisms of Daph on arsenic-induced cytotoxicity in human lung epithelial cells. Our results demonstrate that Daph dramatically upregulated the antioxidant enzyme in a dose dependent manner, in association with induction of Nrf2 nuclear translocation and decreased Keap1 protein expression. Importantly, Daph also markedly induced the activation of AMP-activated protein kinase (AMPK), c-Jun NH2-terminal kinase (JNK) and extracellular signal-regulated kinase (ERK) phosphorylation. Furthermore, Daph antagonized the arsenic-induced decreases in cell viability and the generation of reactive oxygen species (ROS). Notably, Daph pretreatment reversed the arsenic-induced decrease in anti-apoptotic factor B-cell lymphoma-2 (Bcl-2) and the increase in pro-apoptotic factor Bcl-2-associated X protein (Bax). The effects of Daph on Nrf2 and HO-1 activation, and arsenic-induced cell viability were largely weakened when Nrf2 was depleted in vitro. Accordingly, Daph might ameliorate arsenic-induced cytotoxicity and apoptosis, which may be linked to the induction of Nrf2-dependent antioxidant responses as well as stabilization of the anti-apoptotic factor Bcl-2 in human lung epithelial cells.
    Keywords:  Arsenic; Daphnetin; Nrf2; Oxidative insult
    DOI:  https://doi.org/10.1016/j.cbi.2019.02.001
  14. Biochem Biophys Res Commun. 2019 Feb 11. pii: S0006-291X(19)30049-X. [Epub ahead of print]
      Acute lung injury (ALI) is served as a severe life-threatening disease. However, the pathogenesis that contributes to ALI has not been fully understood. Tumor necrosis factor receptor-associated factor 1 (TRAF1) interacts with multiple regulators, performing its diverse role in biological functions. However, the effects of TRAF1 on ALI remain unknown. In this study, we attempted to explore the role of TRAF1 in ALI progression. The findings suggested that TRAF1-knockout (KO) markedly attenuated LPS-induced severe mortality rate in murine animals. LPS-elicited histological alterations in pulmonary tissues were significantly alleviated by TRAF1-deletion. Additionally, TRAF1 knockout effectively attenuated lung injury, as evidenced by the reduced lung wet/dry (W/D) weight ratio, as well as decreased bronchoalveolar lavage fluid (BALF) protein levels and neutrophil infiltration. Meanwhile, TRAF1 deletion markedly lessened inflammation, oxidative stress and apoptosis in BALF and/or lung tissues. The levels of pro-inflammatory cytokines stimulated by LPS were down-regulated by TRAF1 ablation, along with the inactivation of nuclear factor κB (NF-κB). LPS-promoted reactive oxygen species (ROS) generation was decreased in TRAF1-KO mice, partly through the improvement of anti-oxidants. Apoptosis was also inhibited by TRAF1 deletion in lung tissues of LPS-challenged mice through the suppression of cleaved Caspase-3. Moreover, TRAF1 knockout significantly decreased c-Jun N-terminal kinase (JNK) activation and its down-streaming signal of c-Jun in pulmonary samples of LPS-induced mice. Importantly, the in vitro study suggested that promoting JNK activation markedly abrogated TRAF1 knockdown-attenuated inflammation, ROS production and apoptosis in LPS-exposed A549 cells. Therefore, our experimental results provided evidence that TRAF1 suppression effectively protected LPS-induced ALI against inflammation, oxidative stress and apoptosis through the suppression of JNK activity.
    Keywords:  Acute lung injury (ALI); Inflammation; JNK; Oxidative stress and apoptosis; TRAF1
    DOI:  https://doi.org/10.1016/j.bbrc.2019.01.041
  15. Int J Pediatr Otorhinolaryngol. 2019 Feb 04. pii: S0165-5876(19)30066-7. [Epub ahead of print]120 30-35
      OBJECTIVES: While cisplatin is an effective chemotherapeutic agent, it can cause irreversible hearing loss. Ototoxicity leads to dose reduction during the cisplatin chemotherapy and results in inadequate treatment of malignant tumors. This study aimed to investigate the protective effects of ferulic acid on cisplatin-induced ototoxicity.METHODS: House Ear Institute-Organ of Corti 1 (HEI-OC1) cells were exposed to 30 μM of cisplatin for 24 h with or without pretreatment with ferulic acid. Cell viability was determined using the WST assay. Apoptotic cells were identified using TUNEL assay. Western blot analysis was performed to examine the change in expression of cleaved caspase, cleaved poly-ADP-ribose polymerase (PARP), nuclear factor erythroid 2-related factor 2 (Nrf2), and catalase. Intracellular reactive oxygen species (ROS) were determined by flow cytometry. Real-time PCR analyses were performed to examine the mRNA levels of antioxidant enzymes including glutamate-cysteine ligase catalytic subunit (Gclc), glutathione peroxidase 2 (Gpx2), catalase, and superoxide dismutase 2 (SOD2). Phalloidin staining of the organ of Corti was performed to determine hair cell survival or degeneration.
    RESULTS: Pretreatment with ferulic acid before cisplatin exposure significantly increased cell viability, levels of antioxidant enzymes, and hair cell survival. In addition, pretreatment with ferulic acid significantly reduced apoptotic cells, levels of cleaved caspase, levels of cleaved PARP, and intracellular ROS production.
    CONCLUSION: Our results demonstrated that ferulic acid inhibited cisplatin-induced cytotoxicity by preventing ROS formation and inducing the production of endogenous antioxidants and indicated that ferulic acid might be used as a protective agent against cisplatin-induced ototoxicity.
    Keywords:  Apoptosis; Cell death; Cisplatin; Ferulic acid; Ototoxicity
    DOI:  https://doi.org/10.1016/j.ijporl.2019.02.001
  16. J Liposome Res. 2019 Feb 10. 1-33
      Mammary gland tumor has the highest incidence rate and mortality in women, worldwide. The present study envisaged a molecularly targeted nanostructured lipid carrier (NLCs) for doxorubicin (Dox) delivery capable of inducing cellular apoptosis in mammary gland tumor. NLCs were prepared utilizing Perilla frutescens oil (54-69% ω3-fatty acid) as liquid lipid to enhance entrapment of Dox through molecular ion pairing. Biotin decorated NLCs (b-Dox-NLCs) were evaluated in vitro and in vivo. The b-Dox-NLCs showed particle size of 105.2 ± 3.5 nm, zeta potential -35 ± 2mV, entrapment 99.15 ± 1.71%, drug content 19.67 ± 2.6 mg.g-1, biotin content 5.85 ± 0.64 µg.g-1 and drug release 98.67 ± 2.43% (facilitated by acidic microenvironment) respectively. MTT assay and Flow cytometric analysis revealed higher anti-proliferative capability of b-Dox-NLCs to force apoptosis in MCF-7 cell line vis-à-vis marketed Dox, evidenced by reactive oxygen species level and mitochondrial membrane potential mediated apoptosis. Enhanced antitumor targeting, therapeutic safety and efficacy was exhibited by b-Dox-NLCs, as investigated through tumor volume, animal survival, weight variation, cardiotoxicity and biodistribution studies in 7,12-Dimethylbenz[a]anthracene induced mammary gland tumor. Immunoblotting assay demonstrated b-Dox-NLCs downregulated anti-apoptotic proteins, i.e., bcl-2, MMP-9 while upregulated pro-apoptotic proteins, i.e., caspase-9, p16 and BAX. The experimental results suggest that biotinylated ω3-fatty acid augmented NLCs loaded with Dox are capable of inducing programmed cell death in mammary tumor and can be utilized as safe and effective delivery system with enhanced potential for mammary gland carcinoma therapy.
    Keywords:  Apoptosis; Flow cytometry; MCF-7 Cell lines; Nanostructured Lipid Carriers; immunoblotting
    DOI:  https://doi.org/10.1080/08982104.2019.1579839
  17. Sci Total Environ. 2019 Feb 25. pii: S0048-9697(18)34415-2. [Epub ahead of print]653 1426-1434
      Deltamethrin (Del), an important broad-spectrum insecticide, is widely used in agricultural activities. However, Del is an effective reactive oxygen species (ROS) inducer that induces oxidative stress damage in cells or tissues. Del is significantly more toxic to aquatic organisms, especially crustaceans, than to mammals and birds. This study was designed to evaluate the protective effect of melatonin (MT) on the toxicity-induced damage of Del after 6 h in Eriocheir sinensis. The results showed that Del exposure significantly induced oxidative damage in the hepatopancreas and mitochondria, with malondialdehyde (MDA) and glutathione (GSH) levels being significantly increased and superoxide dismutase (SOD) activity being significantly decreased. Moreover, Del exposure significantly induced functional damage of the hepatopancreas and mitochondria, with a significant increase in alanine aminotransferase (ALT), aspartate aminotransferase (AST), acid phosphatase (ACP) and alkaline phosphatase (AKP) activities in the hepatopancreas and the ratio of albumin/globulin (ALB/GLB) in serum, which indicated the permeability and integrity of the membranes were damaged and had caused cell damage. In addition, ATP content, Na+-K+-ATPase activity and cytochrome C (Cyt‑C) content in mitochondria decreased significantly, which indicated that Del exposure destroyed the normal respiratory chain of mitochondria. We also evaluated the hematological parameters. Although there were no significant differences in total hemocyte count (THC) levels, hemocyte apoptosis was significantly induced by Del exposure, and the hemocyte phagocytic activity and the hemocyanin levels decreased significantly with Del exposure. However, MT pretreatment not only prevented oxidative damage and functional damage caused by Del exposure to the hepatopancreas and mitochondria, but it also restored the hemocyte apoptotic rate and phagocytic activity to normal levels. In short, Del exposure caused significant oxidative and functional damage to the hepatopancreas, mitochondria and hemocytes of E. sinensis, whereas the use of MT almost completely eliminated the damage caused by Del exposure.
    Keywords:  Deltamethrin; Eriocheir sinensis; Hemocyte immunity; Melatonin; Oxidative damage
    DOI:  https://doi.org/10.1016/j.scitotenv.2018.11.063
  18. Nutrients. 2019 Feb 14. pii: E404. [Epub ahead of print]11(2):
      In this study, we investigated whether (E)-5-hydroxy-7-methoxy-3-(2'-hydroxybenzyl)-4-chromanone, a homoisoflavonoid compound isolated from Portulaca oleracea L., protects INS-1 pancreatic β cells against glucotoxicity-induced apoptosis. Treatment with high glucose (30 mM) induced apoptosis in INS-1 pancreatic β cells; however, the level of cell viability was significantly increased by treatment with (E)-5-hydroxy-7-methoxy-3-(2'-hydroxybenzyl)-4-chromanone. Treatment with 10⁻20 µM of (E)-5-hydroxy-7-methoxy-3-(2'-hydroxybenzyl)-4-chromanone dose-dependently increased cell viability and significantly decreased the intracellular level of reactive oxygen species (ROS), thiobarbituric acid reactive substances (TBARS), and nitric oxide levels in INS-1 pancreatic β cells pretreated with high glucose. These effects were associated with increased anti-apoptotic Bcl-2 protein expression, while reducing pro-apoptotic Bax, cytochrome C, and caspase 9 protein expression. Treatment with (E)-5-hydroxy-7-methoxy-3-(2'-hydroxybenzyl)-4-chromanone reduced the apoptosis previously induced by high-level glucose-treatment, according to annexin V/propidium iodide staining. These results demonstrate that (E)-5-hydroxy-7-methoxy-3-(2'-hydroxybenzyl)-4-chromanone may be useful as a potential therapeutic agent to protect INS-1 pancreatic β cells against high glucose-induced apoptosis.
    Keywords:  (E)-5-hydroxy-7-methoxy-3-(2′-hydroxybenzyl)-4-chromanone; INS-1 pancreatic β cell; Portulaca oleracea; glucotoxicity
    DOI:  https://doi.org/10.3390/nu11020404
  19. Metab Brain Dis. 2019 Feb 12.
      Oxidative stress has been suggested to play an important role in neuronal injury. Ethyl gallate (EG) is the ethyl ester of gallic acid which has been acknowledged as an antioxidant. We previously demonstrated that EG effectively inhibited H2O2-induced cytotoxicity and decreased the ROS levels in PC12 cells, while the relevant mechanisms of action of this compound remain largely uncharacterized. The present study was carried out in an attempt to clarify the underlying mechanisms of EG against H2O2-induced neurotoxicity in PC12 cells. EG pretreatment attenuated H2O2-induced mitochondrial dysfunction as indicated by the decreased caspase-9/-3 activation, PARP cleavage, mitochondrial membrane potential (MMP) depletion, Bax/Bcl-2 ratio, cytochrome c release and ROS overproduction. Furthermore, EG treatment resulted in nuclear translocation of Nrf2 along with increased expression of ARE-dependent cytoprotective genes, such as γ-GCS and NQO1, which indicated EG as an Nrf2 pathway activator. Silencing of Nrf2 signaling by siRNA abrogated the protective effects offered by EG on H2O2-induced PC12 cells injury, which suggested the important role of Nrf2 pathway in the protection of EG against oxidative stress induced PC12 cell apoptosis. These results taken together indicated that EG protects PC12 cells against H2O2-induced cell mitochondrial dysfunction possibly through activation of Nrf2 pathway. EG might be a potential candidate for further preclinical study aimed at the prevention and treatment of neurodegenerative diseases.
    Keywords:  Ethyl gallate; Mitochondrial dysfunction; Nrf2 pathway; PC12 cells
    DOI:  https://doi.org/10.1007/s11011-019-0382-z
  20. J Appl Toxicol. 2019 Feb 14.
      The current study was intended to elucidate the cytotoxicity, genotoxicity ability of nickel oxide (NiO) nanoparticles (NPs) and assessment of preliminary mechanism of the toxicity. Characterization studies showed that NiO-NPs have a particle size of 17.94 (±3.48) nm. The particle size of the NPs obtained by dynamic light scattering method in Milli-Q and RPMI 1640 media was 189.9 (±17.1) and 285.9 (±19.6) nm, respectively. The IC50 concentration for NiO-NPs after 24 hours of treatment was estimated as 23.58 μg/mL. Comet and cytokinesis-block micronucleus assays revealed a significant dose- and time-dependent genotoxic potential of NiO-NPs. Morphological assessment of the lymphocytes upon exposure to NiO-NPs showed that the mechanism of toxicity was apoptosis. Reactive oxygen species analysis and lipid peroxidation patterns were aligned with the cytotoxicity and genotoxicity endpoints. Thus, the preliminary mechanism of NiO-NPs for cytotoxicity on lymphocytes was assumed to be oxidative stress-mediated apoptosis and DNA damage. Furthermore, these NiO-NPs are considered a potentially hazardous substance at environmentally significant levels. Further investigations are suggested to understand the immunotoxic effects of NiO-NPs.
    Keywords:  NPs dissolution; NiO-NPs; cytotoxicity; genotoxicity; immunotoxicity of nanoparticles; oxidative stress
    DOI:  https://doi.org/10.1002/jat.3784
  21. Biochem Biophys Res Commun. 2019 Feb 08. pii: S0006-291X(19)30102-0. [Epub ahead of print]
      The oxygen partial pressure generally increases when cancerous cells become part of the blood vessels. The study was to investigate the influence of oxygen partial pressure on the apoptosis of B16 melanoma cells. Our results demonstrated that both short-term and long-term hypoxia/reoxygenation (H/R) treatment increased stress-induced intracellular reactive oxygen species (ROS). H/R treatment also increased apoptosis and autophagy in B16 cells. N-acetylcysteine (NAC), a ROS scavenger, can reduce ROS and aid survival. However, Bafilomycin A1, an autophagy inhibitor, can accelerate cell death. Thus, our work revealed that ROS and autophagy play critical roles in cellular H/R.
    Keywords:  Autophagy; Hypoxia; Hypoxia/reoxygenation (H/R); Reactive oxygen species (ROS)
    DOI:  https://doi.org/10.1016/j.bbrc.2019.01.135
  22. Environ Toxicol. 2019 Feb 11.
      Previous studies have reported that cigarette smoke and cigarette smoke extract (CSE) have negative effects on embryonic development. However, no studies have investigated the mechanism through which CSE affects the cellular signaling pathway leading to apoptosis and oxidative stress in embryonic cells, or how the two pathways are cross-linked. Thus, we studied the effects of CSE on apoptosis and oxidative stress in mouse embryonic stem cells (mESCs). Specifically, we measured changes in cell viability in response to CSEs (3R4F and two domestic cigarettes CSE 1 and 2) using a water soluble tetrazolium (WST) assay and a neutral red uptake (NRU) assay, which revealed that cell viability decreased in a concentration-dependent manner. Western blot analysis revealed that the expression of cyclin D1 and cyclin E1 was decreased and that of p21 and p27 was increased by CSE. Additionally, the number of terminal deoxynucleotidyl transferase (TUNEL)-stained cells was increased by CSE, while the levels of Bax and Caspase-3 increased and Bcl-2 decreased. Moreover, a 2',7'-dichlorofluorescin diacetate (DCF-DA) assay and reactive oxygen species (ROS)-Glo H2 O2 assay confirmed that ROS were generated in response to CSE and that they were associated with up-regulated Keaf-1 and CHOP. Overall, the results revealed that cigarette smoke extract (CSE) inhibited cell proliferation by regulating cell cycle-related protein expression and increased oxidative stress by regulating the expression of Kelch-like ECH-associated protein 1 (Keap-1) and CCAAT/enhancer-binding protein homologous protein (CHOP), resulting in apoptosis in mESCs.
    Keywords:  apoptosis; cell cycle; cigarette smoke extract; mouse embryonic stem cells; oxidative stress; reactive oxygen species
    DOI:  https://doi.org/10.1002/tox.22735
  23. J Exp Clin Cancer Res. 2019 Feb 12. 38(1): 69
      BACKGROUND: Neuroblastoma is the most common extracranial solid tumor in children. This cancer has a low frequency of TP53 mutations and its downstream pathway is usually intact. This study assessed the efficacy of the p53 activator, PRIMA-1MET, in inducing neuroblastoma cell death.METHODS: CellTiter 2.0 was used to study susceptibility and specificity of NB cell lines to PRIMA-1MET. Real-time PCR and western blot were used to assess the most common p53 transactivation targets. Induction of p53 and Noxa, and inhibition of Cas3/7, were used to assess impact on cell death after PRIMA-1MET treatment. Flow cytometry was used to analyze cell cycle phase and induction of apoptosis, reactive oxygen species, and the collapse of mitochondrial membrane potential.
    RESULTS: Neuroblastoma cell lines were at least four times more susceptible to PRIMA-1MET than were primary fibroblasts and keratinocyte cell lines. PRIMA-1MET induced cell death rapidly and in all cell cycle phases. Although PRIMA-1MET activated p53 transactivation activity, p53's role is likely limited because its main targets remained unaffected, whereas pan-caspase inhibitor demonstrated no ability to prevent cell death. PRIMA-1MET induced oxidative stress and modulated the methionine/cysteine/glutathione axis. Variations of MYCN and p53 modulated intracellular levels of GSH and resulted in increased/decreased sensitivity of PRIMA-1MET. PRIMA-1MET inhibited thioredoxin reductase, but the effect of PRIMA-1MET was not altered by thioredoxin inhibition.
    CONCLUSIONS: PRIMA-1MET could be a promising new agent to treat neuroblastoma because it demonstrated good anti-tumor action. Although p53 is involved in PRIMA-1MET-mediated cell death, our results suggest that direct interaction with p53 has a limited role in neuroblastoma but rather acts through modulation of GSH levels.
    Keywords:  Glutathione; MYCN; Neuroblastoma; PRIMA-1MET; p53
    DOI:  https://doi.org/10.1186/s13046-019-1066-6
  24. J Cell Mol Med. 2019 Feb 12.
      Previous studies have suggested that the cellular Ca2+ and iron homeostasis, which can be regulated by mitochondrial calcium uniporter (MCU), is associated with oxidative stress, apoptosis and many neurological diseases. However, little is known about the role of MCU-mediated Ca2+ and iron accumulation in traumatic brain injury (TBI). Under physiological conditions, MCU can be inhibited by ruthenium red (RR) and activated by spermine (Sper). In the present study, we used RR and Sper to reveal the role of MCU in mouse and neuron TBI models. Our results suggested that the Ca2+ and iron concentrations were obviously increased after TBI. In addition, TBI models showed a significant generation of reactive oxygen species (ROS), decrease in adenosine triphosphate (ATP), deformation of mitochondria, up-regulation of deoxyribonucleic acid (DNA) damage and increase in apoptosis. Blockage of MCU by RR prevented Ca2+ and iron accumulation, abated the level of oxidative stress, improved the energy supply, stabilized mitochondria, reduced DNA damage and decreased apoptosis both in vivo and in vitro. Interestingly, Sper did not increase cellular Ca2+ and iron concentrations, but suppressed the Ca2+ and iron accumulation to benefit the mice in vivo. However, Sper had no significant impact on TBI in vitro. Taken together, our data demonstrated for the first time that blockage of MCU-mediated Ca2+ and iron accumulation was essential for TBI. These findings indicated that MCU could be a novel therapeutic target for treating TBI.
    Keywords:  Ca2+; iron; mitochondrial calcium uniporter; neuroprotection; traumatic brain injury
    DOI:  https://doi.org/10.1111/jcmm.14206
  25. ACS Chem Neurosci. 2019 Feb 11.
      Cdh1 is a regulatory subunit of the anaphase promoting complex/cyclosome (APC/C), known to be involved in regulating neuronal survival. The role of Cdh1 in volatile anesthetics-induced neuronal apoptosis in the developing brain is unknown. In this study, we used postnatal day 7 (P7) and day 21 (P21) mice exposed to 2.3% sevoflurane for 6 h to investigate at which age and duration of exposure sevoflurane affects the expression of Cdh1 and glycolytic enzyme 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3) and that of the pentose phosphate pathway (PPP) enzyme, Glucose-6-phosphate dehydrogenase (G6PD). Furthermore, we tested whether the Cyclin-dependent kinases (cdks) inhibitor Roscovatine could counteract the effects caused by exposure to sevoflurane. Finally, we applied the glycolysis inhibitor 3-(3-pyridinyl)-1-(4-pyridinyl)-2-propen-1-one (3-PO), G6PD inhibitor Dehydroepiandrosterone (DHEA), and exogenous reduced glutathione to examine the contribution of the glycolysis pathway and PPP to sevoflurane-induced neuroapoptosis. We found that prolonged sevoflurane anesthesia significantly reduces Cdh1 level in P7 mice than in the P21 ones; moreover, the decrease in Cdh1 level results in a switch in glucose metabolism from the PPP to neuronal glycolysis. This leads to an imbalance between reactive oxygen species production and reduced glutathione level in the developing brain, which is more susceptible to oxidative stress. As a result, sevoflurane induces neuroapoptosis through Cdh1-mediated glucose metabolism reprogramming. Our study demonstrates a critical role of Cdh1 in sevoflurane-induced neuroapoptosis by shifting PPP to glycolytic pathway in the developing brain. These findings suggest that Cdh1 may be a novel target for preventing volatile anesthetics-induced neurotoxicity and memory impairment.
    DOI:  https://doi.org/10.1021/acschemneuro.8b00644
  26. Ultrastruct Pathol. 2019 Feb 09. 1-28
      Infertility represents a major medical, economic, and psychological problem. Stem cells therapy for infertility has a great interest nowadays especially for cancer survivors at pre-reproductive and reproductive age.  Thirty-two adult male albino rats were used, divided equally into four groups; Group I (Control group) received isotonic saline intraperitoneally (i.p.) as vehicle. Group II (Cisplatin-treated group) received Cisplatin (i.p.) at a single dose of 7 mg/kg, and then were sacrificed after 5 days. Group III (Stem-cell-treated group) received Cisplatin (i.p.) at a single dose of 7 mg/kg, then after 5 days received adipose-derived mesenchymal stem cells (ADMSCs) (1 × 106). Cells were injected in the rete testis, then after 60 days, the animals were sacrificed. Group IV (Auto healing group) received Cisplatin (i.p.) at a single dose of 7 mg/kg, and then left for 65 days then the animals were sacrificed. Cisplatin administration resulted in degenerative changes in the testicular architecture in the form of thickened irregular BM of seminiferous tubules. The germinal epithelium showed disorganization and marked reduction in the thickness, associated with Sertoli cells preservation. Features of apoptosis assured by elevated caspase-3 expression were noticed. The interstitium showed cellular infiltration and distorted Leydig cells. Injection of (ADMSCs) resulted in great improvement of testicular architecture and increase in the testosterone level associated with strong immune reaction of the CD-44. ADMSCs are recommended as a new treatment modality for male infertility. Abbreviation: i.p.: intraperitoneally; BM: basement membrane; ADMSCs: adipose-derived mesenchymal stem cells; WHO: World Health Organization; MSCs: mesenchymal stem cells; DMEM: Dulbecco modified eagles media; PBS: phosphate-buffered saline; FACS: fluorescence-activated cell sorting; ELISA: enzyme-linked immunosorbent assay; CP: Cisplatin; ROS: reactive oxygen species; CAT: catalase; SOD: superoxide dismutase; OS: oxidative stress; SSCs: spermatogonia stem cells; GCs: germ cells; UCMSCs: umblical cord mesenchymal stem cells; TGFb1: transforming growth factor beta-1; BMP4: Bone morphogenic protein 4; BMP8b: bone morphogenic protein 8b.
    Keywords:  Cisplatin; Mesenchymal stem cells; infertility
    DOI:  https://doi.org/10.1080/01913123.2019.1572256
  27. Toxicology. 2019 Feb 12. pii: S0300-483X(18)30221-X. [Epub ahead of print]
      Tocotrienols (T3s) are a subgroup of vitamin E and they have been widely tested to inhibit cell growth in various tumor types. Previous studies have shown that T3s inhibit cancer cell growth by targeting multiple signaling transduction and cellular processes. However, the role of T3s in the regulation of cellular bioenergetic processes remains unclear. In this study, we found that γ-T3 interacts with mitochondrial electron transfer chain NDUFB8 (a subunit of complex I) and SDHB (a subunit of complex II) and inhibits oxidative phosphorylation (OXPHOS), and triggers the production of reactive oxygen species (ROS). In addition, we observed that γ-T3 upregulates the glycolytic capacity in cells, but it did not compensate for cellular ATP generation and decreased the ATP levels in cells. Furthermore, we performed western blots and RT-PCR to measure the mRNA and protein levels of mitochondrial electron transfer chain (ETC) proteins and complex V (ATP synthase), where the results indicated that γ-T3 specifically inhibited the levels of NDUFB8 and SDHB, whereas it had little effect on UQCRC2 (a subunit of complex III), COX4I1 (a subunit of complex IV), and ATP5F1A (a subunit of complex V). The inhibition of NDUFB8 and SDHB by γ-T3 led to the overproduction of ROS and the depletion of ATP, which may be responsible for inducing apoptosis in cancer cells. Our results suggest that mitochondrial respiration may be an effective target for anticancer treatments based on γ-T3.
    Keywords:  Mitochondria; NDUFB8; Oxidative Phosphorylation; Reactive Oxygen Species; SDHB; γ-T3
    DOI:  https://doi.org/10.1016/j.tox.2019.01.018
  28. CNS Neurol Disord Drug Targets. 2019 Feb 11.
      BACKGROUND/OBJECTIVE: Grape seed proanthocyanidins (GSPs) are a group of polyphenolic bioflavonoids, which possess a variety of biological functions and pharmacological properties. We studied the neuroprotective effects of GSP against oxygen-glucose deprivation/reoxygenation (OGD/R) injury and the potential mechanisms in mouse neuroblastoma N2a cells.METHODS: OGD/R was conducted in N2a cells. Cell viability was evaluated by CCK-8 and LDH release assay. Apoptosis was assessed by TUNEL staining and flow cytometry. Protein levels of cleaved caspase-3, Bax and Bcl-2 were detected by Western blotting. CHOP, GRP78 and caspase-12 mRNA levels were assessed by realtime PCR. JC-1 dying was used to detect mitochondrial membrane potential. ROS levels, activities of endogenous anti-oxidant enzymes and ATP production were examined to evaluate mitochondrial function.
    RESULTS: GSP increased cell viability after OGD/R injury in a dose-dependent manner. Furthermore, GSP inhibited cell apoptosis, reduced the mRNA levels of CHOP, GRP78 and caspase-12 (ER stress-associated genes), restored mitochondrial membrane potential and ATP generation, improved activities of endogenous anti-oxidant ability (T-AOC, GXH-Px, and SOD), and decreased ROS level.
    CONCLUSIONS: Our findings suggest that GSP can protect N2a cells from OGD/R insult. The mechanism of anti-apoptotic effects of GSP may involve attenuating ER stress and mitochondrial dysfunction.
    Keywords:  apoptosis; endoplasmic reticulum stress; grape seed proanthocyanidins; mitochondrial dysfunction ; oxygen-glucose deprivation/reoxygenation
    DOI:  https://doi.org/10.2174/1871527318666190212111650
  29. J Clin Med. 2019 Feb 05. pii: E191. [Epub ahead of print]8(2):
      Uremic toxins accumulated in chronic kidney disease (CKD) increases the risk of cognitive impairment. Indoxyl sulfate (IS) is a well-known protein-bound uremic toxin that is correlated with several systemic diseases, but no studies on human brain cells are available. We investigated the effect of IS on primary human astrocytes through next-generation sequencing and cell experiment confirmation to explore the mechanism of IS-associated brain damage. Total RNAs extracted from IS-treated and control astrocytes were evaluated by performing functional and pathway enrichment analysis. The toxicities of IS in the astrocytes were investigated in terms of cell viability through flow cytometry; the signal pathway was then investigated through immunoblotting. IS stimulated the release of reactive oxygen species, increased nuclear factor (erythroid-derived 2)-like 2 levels, and reduced mitochondrial membrane potential. IS triggered astrocyte apoptosis by inhibiting the mitogen-activated protein kinase (MAPK) pathway, including extracellular-signal-regulated kinase (ERK), MAPK/ERK kinase, c-Jun N-terminal kinase, and p38. The decreased ERK phosphorylation was mediated by the upregulated dual-specificity phosphatase 1, 5, 8, and 16. In conclusion, IS can induce neurotoxicity in patients with CKD and the pathogenesis involves cell apoptosis through oxidative stress induction and MAPK pathway inhibition in human astrocytes.
    Keywords:  astrocyte; dual specific phosphatase; indoxyl sulfate; mitogen-activated protein kinase; oxidative stress; uremic toxins
    DOI:  https://doi.org/10.3390/jcm8020191
  30. J Bone Oncol. 2019 Apr;15 100222
      Background: Chondrosarcomas are malignant cartilage-producing tumors showing mutations and changes in gene expression in metabolism related genes. In this study, we aimed to explore the metabolome and identify targetable metabolic vulnerabilities in chondrosarcoma.Methods: A custom-designed metabolic compound screen containing 39 compounds targeting different metabolic pathways was performed in chondrosarcoma cell lines JJ012, SW1353 and CH2879. Based on the anti-proliferative activity, six compounds were selected for validation using real-time metabolic profiling. Two selected compounds (rapamycin and sapanisertib) were further explored for their effect on viability, apoptosis and metabolic dependency, in normoxia and hypoxia. In vivo efficacy of sapanisertib was tested in a chondrosarcoma orthotopic xenograft mouse model.
    Results: Inhibitors of glutamine, glutathione, NAD synthesis and mTOR were effective in chondrosarcoma cells. Of the six compounds that were validated on the metabolic level, mTOR inhibitors rapamycin and sapanisertib showed the most consistent decrease in oxidative and glycolytic parameters. Chondrosarcoma cells were sensitive to mTORC1 inhibition using rapamycin. Inhibition of mTORC1 and mTORC2 using sapanisertib resulted in a dose-dependent decrease in viability in all chondrosarcoma cell lines. In addition, induction of apoptosis was observed in CH2879 after 24 h. Treatment of chondrosarcoma xenografts with sapanisertib slowed down tumor growth compared to control mice.
    Conclusions: mTOR inhibition leads to a reduction of oxidative and glycolytic metabolism and decreased proliferation in chondrosarcoma cell lines. Although further research is needed, these findings suggest that mTOR inhibition might be a potential therapeutic option for patients with chondrosarcoma.
    Keywords:  ACT, Atypical cartilaginous tumor; BLI, Bioluminescence imaging; BSA, Bovine serum albumin; BSO, Buthionine sulfoximine; Chondrosarcoma; D2HG, d-2-Hydroxyglutarate; DMSO, Dimethyl sulfoxide; ECAR, Extracellular acidification rate; FBS, Fetal bovine serum; FCCP, Carbonyl cyanide-4-(trifluoromethoxy)phenylhydrazone; FLI, Fluorescence imaging; HIF, Hypoxia-inducible factor; IDH, Isocitrate dehydrogenase; Metabolism; OCR, Oxygen consumption rate; ROS, Reactive oxygen species; Rapamycin; mCT, Micro computed tomography; mTOR, Mammalian target of rapamycin; mTOR, Sapanisertib; α-KG, α-ketoglutarate
    DOI:  https://doi.org/10.1016/j.jbo.2019.100222
  31. Life Sci. 2019 Feb 08. pii: S0024-3205(19)30099-2. [Epub ahead of print]
      AIMS: Hyperglycemia-mediated oxidative damage has been described as a major mechanism leading to pathologic changes associated with diabetic cardiomyopathy (DCM). Fisetin is a bioactive flavonol molecule found in many plants and possesses various biological activities. The present study investigated the protective effect of fisetin on diabetes-induced cardiac injury.METHODS: Diabetes was induced by streptozotocin (STZ) and both diabetic and control rats were treated with 2.5 mg/kg fisetin for six weeks.
    KEY FINDINGS: Diabetic rats exhibited hyperglycemia, and increased glycosylated hemoglobin and lipids accompanied with significant hypoinsulinism. In addition, diabetic rats showed several histological alterations in the myocardium, and significantly increased serum troponin I, creatine kinase-MB and lactate dehydrogenase. Oxidative stress, inflammation and apoptosis markers were increased, whereas antioxidant defenses were significantly reduced in the diabetic heart. Treatment with fisetin alleviated hyperglycemia, hyperlipidemia and heart function markers, and minimized histological alterations in the myocardium. Fisetin suppressed oxidative stress, prevented inflammation and apoptosis, and boosted antioxidant defenses in the heart of diabetic rats.
    SIGNIFICANCE: Fisetin attenuated the development of DCM via amelioration of hyperglycemia/hyperlipidemia-mediated oxidative stress, inflammation and apoptosis. Therefore, it might be worth considering the therapeutic potential of fisetin for human DCM.
    Keywords:  Apoptosis; Cardiomyopathy; Fisetin; Flavonoids; Hyperglycemia; Oxidative stress
    DOI:  https://doi.org/10.1016/j.lfs.2019.02.017
  32. Int J Biol Sci. 2019 ;15(3): 701-713
      Previous studies have shown that mitochondrial dysfunction plays an important role in high- glucose(HG)-induced podocyte injury and thus contributes to the progression of diabetic nephropathy(DN). The histone deacetylase Sirtuin6 (Sirt6) has been revealed to have an essential role in the regulation of mitochondrial function in skeletal muscle and cardiomyocytes. However, its specific role in mitochondrial homeostasis in podocytes is undetermined. Here, we aimeds to explore the physiological function of Sirt6 in podocyte mitochondria and apoptosis under HG conditions and explore the possible mechanism. Herein, we observed that Sirt6-WT-1 colocalization was suppressed in the glomeruli of patients with DN. In addition, diabetic mice exhibited reduced Sirt6 expression and AMP kinase (AMPK) dephosphorylation accompanied by mitochondrial morphological abnormalities. In vitro, podocytes exposed to HG presented with mitochondrial morphological alterations and podocyte apoptosis accompanied by Sirt6 and p-AMPK downregulation. In addition, HG promoted a decrease in mitochondrial number and an increase in mitochondrial superoxide production as well as a decreased mitochondrial membrane potential. ROS production was also increased in HG-treated podocytes. Conversely, all these mitochondrial defects induced by HG were significantly alleviated by Sirt6 plasmid transfection. Sirt6 overexpression simultaneously alleviated HG-induced podocyte apoptosis and oxidative stress, as well as increased AMPK phosphorylation. Increased levels of H3K9ac and H3K56ac induced by HG were attenuated in podocytes transfected with Sirt6 plasmids. Therefore, these results elucidated that Sirt6 protects mitochondria of podocytes and exerts anti-apoptotic effects via activating AMPK pathway. The present findings provide key insights into the pivotal role of mitochondria regulation by SIRT6 in its protective effects on podocytes.
    Keywords:  Sirt6; apoptosis; diabetic nephropathy; mitochondrial dysfunction; podocytes
    DOI:  https://doi.org/10.7150/ijbs.29323
  33. Toxicology. 2019 Feb 12. pii: S0300-483X(18)30677-2. [Epub ahead of print]
      Malathion is one of the most widely used organophosphorus insecticides in agriculture. However, malathion may be involved in the etiology of human brain dysfunction. Induction of ROS has been proposed as a mechanism of malathion-induced poisoning cases, but there are few data regarding the effects of malathion on oxidative stress-associated neurotoxicity in human glial cells. The aim was to explore the mechanism underlying effects of malathion on neurotoxicity in Gibco® Human Astrocytes (GHA cells) and evaluate the protective effects of the antioxidant (N-acetylcysteine, NAC). Cell viability was measured by the cell proliferation reagent (WST-1). Antioxidant enzymes (glutathione peroxidase and catalase) were measured by an ELISA reader. Cell cycle distribution and ROS productions were detected by flow cytometry. Cell cycle-related protein levels (cyclin E1, CDK2, cyclin A2, CDK1/CDC2, or cyclin B1) and apoptotic protein levels (Bcl-2, Bax, and cleaved caspase-9/caspase-3) were analyzed by Western blotting. In GHA cells, treatment with malathion (10-25 μM) for 24 h concentration-dependently induced cytotoxicity and cell cycle arrest. In terms of oxidative stresses, malathion elevated intracellular ROS levels, but reduced glutathion and antioxidant enzyme levels. Treatment with NAC (5 μM) reversed malathion-induced oxidative stress responses, and prevented malathion-evoked apoptosis by regulating apoptotic protein expressions. Together, in GHA cells, NAC mediated inhibition of malathion-activated mitochondrial apoptotic pathways that involved cell cycle arrest and ROS responses. These data provide further insights into the molecular mechanisms behind malathion poisoning, and might suggest that NAC with its protective effects may be a potential compound for prevention of malathion-induced brain injury.
    Keywords:  Apoptosis; Astrocytes; Malathion; N-acetylcysteine (NAC); Neurotoxicity; Oxidative stress
    DOI:  https://doi.org/10.1016/j.tox.2019.02.004
  34. PLoS One. 2019 ;14(2): e0212231
      Koumine is a component of the Chinese medicinal herb Gelsemium elegans and is toxic to vertebrates. We used the ciliate Tetrahymena thermophila as a model to evaluate the toxic effects of this indole alkaloid in eukaryotic microorganisms. Koumine inhibited T. thermophila growth and viability in a dose-dependent manner. Moreover, this drug produced oxidative stress in T. thermophila cells and expressions of antioxidant enzymes were significantly elevated at high koumine levels (p < 0.05). Koumine also caused significant levels of apoptosis (p < 0.05) and induced DNA damage in a dose-dependent manner. Mitophagic vacuoles were present in cells indicating induction of autophagy by this drug. Expression of ATG7, MTT2/4, CYP1 and HSP70 as well as the MAP kinase pathway gene MPK1 and MPK3 were significantly altered after exposed to koumine. This study represents a preliminary toxicological evaluation of koumine in the single celled eukaryote T. thermophila.
    DOI:  https://doi.org/10.1371/journal.pone.0212231
  35. Open Access Maced J Med Sci. 2019 Jan 15. 7(1): 38-44
      BACKGROUND: Ischemic stroke occurs due to the abrupt occlusion in the brain which leads to neuronal death. Neuronal death in ischemic stroke is due to increase production of reactive oxygen species (ROS). Neuronal death occurs via necrosis and apoptosis mechanisms. Apoptosis can either occur via extrinsic or intrinsic pathway. Meanwhile, the intrinsic pathway can be caspase-dependent or independent. Anthocyanin is a natural pigment with antioxidant, anti-inflammatory, anti-cancer, and neuroprotective properties. Balinese cultivate of purple potato extract contains a high level of anthocyanin and has been proven for its antioxidant activity.AIM: Antioxidant effect of Balinese cultivates purple potato extract has not been studied on an animal model with ischemic stroke. Accordingly, we would like to study the effect of antioxidant properties from Balinese cultivate of purple potato extract by assessing the neurological score, BNDF concentration, and caspase-independent apoptosis by measuring AIF concentration on Wistar rats with ischemic stroke.
    METHODS: This was an experimental study using male Wistar rats age between 12-14 weeks weigh between 200 to 250 g.
    RESULTS: This study demonstrated a significant difference of neurological score on day 3 among control versus treatment groups. Balinese cultivate of purple potato extract markedly reduced AIF, increased BDNF, and suppressed apoptosis among treatment group when compared with the control group.
    CONCLUSION: We have proven the efficacy of antioxidant activity of anthocyanin derived from Balinese cultivar of purple sweet potato by elevated AIF levels, lower apoptosis rate, improved neurological score on day-3 to day-7 post-stroke, as well as increased BDNF levels.
    Keywords:  AIF; Apoptosis; BDNF; Balinese cultivate of purple potato extract; Ischemic stroke; Neurological score
    DOI:  https://doi.org/10.3889/oamjms.2019.019
  36. Front Oncol. 2019 ;9 2
      Celastrol is a natural triterpene isolated from the Chinese plant Thunder God Vine with potent antitumor activity. However, the effect of celastrol on the growth of ovarian cancer cells in vitro and in vivo is still unclear. In this study, we found that celastrol induced cell growth inhibition, cell cycle arrest in G2/M phase and apoptosis with the increased intracellular reactive oxygen species (ROS) accumulation in ovarian cancer cells. Pretreatment with ROS scavenger N-acetyl-cysteine totally blocked the apoptosis induced by celastrol. Additionally, celastrol inhibited the growth of ovarian cancer xenografts in nude mice. Altogether, these findings suggest celastrol is a potential therapeutic agent for treating ovarian cancer.
    Keywords:  N-acetyl-cysteine; apoptosis; celastrol; ovarian cancer; reactive oxygen species
    DOI:  https://doi.org/10.3389/fonc.2019.00002
  37. Cell Death Dis. 2019 Feb 12. 10(2): 134
      Bone marrow derived stem cells (BMSCs) transplantation are viewed as a promising therapeutic candidate for spinal cord injury (SCI). However, the inflammatory microenvironment in the spinal cord following SCI limits the survival and efficacy of transplanted BMSCs. In this study, we investigate whether injured neuronal cells derived exosomes would influence the survival of transplanted BMSCs after SCI. In order to mimic the microenvironment in SCI that the neuronal cells or transplanted BMSCs suffer in vivo, PC12 cells conditioned medium and PC12 cell's exosomes collected from H2O2-treated PC12 cell's culture medium were cultured with BMSCs under oxidative stress in vitro. PC12 cells conditioned medium and PC12 cell's exosomes significantly accelerated the apoptosis of BMSCs induced by H2O2. Moreover, the cleaved caspase-3, cytochrome (Cyt) C, lactate dehydrogenase (LDH) releases, and apoptotic percentage were increased, and the ratio of Bcl-2/Bax and cell viability were decreased. Inhibition of exosome secretion via Rab27a small interfering RNA prevented BMSCs apoptosis in vitro. In addition, hypoxia-preconditioned promoted the survival of BMSCs under oxidative stress both in vivo after SCI and in vitro. Our results also indicate that HIF-1α plays a central role in the survival of BMSCs in hypoxia pretreatment under oxidative stress conditions. siRNA-HIF-1α increased apoptosis of BMSCs; in contrast, HIF-1α inducer FG-4592 attenuated apoptosis of BMSCs. Taken together, we found that the injured PC12 cells derived exosomes accelerate BMSCs apoptosis after SCI and in vitro, hypoxia pretreatment or activating expression of HIF-1α to be important in the survival of BMSCs after transplantation, which provides a foundation for application of BMSCs in therapeutic potential for SCI.
    DOI:  https://doi.org/10.1038/s41419-019-1410-y
  38. Toxicol In Vitro. 2019 Feb 07. pii: S0887-2333(18)30713-6. [Epub ahead of print]57 28-38
      Fluoride is very crucial for development of teeth and bones. Excessive fluoride, however, causes damage to teeth and bones resulting in serious public health problem. SIRT1 regulates physiological and pathological processes such as apoptosis and cell cycle. Although SIRT1 inhibits p53-mediated transactivation, how SIRT1 regulates p53 in fluorosis remains unclear. This study aims to investigate the involvement of SIRT1 in fluoride-induced cell cycle arrest and apoptosis in MC3T3-E1 cells and the underlying mechanism. Cell apoptosis was determined using Annexin V-FITC/PI dual staining, cell cycle detected with PI staining, intracellular ROS levels measured with DCFH-DA probe, and apoptosis-related protein expressions determined using Western blotting. Results showed that there was a promotion in apoptosis rate, intracellular ROS levels, the ratio of Bax/Bcl-2, protein expression (Cyt c, Caspase-3, p53, Ac-p53 and p21) and blockage of S phase after cells were exposed to NaF. Afterwards, the influence of SIRT1 on apoptosis was explored after SRT1720 (SIRT1 activator) and Ex-527 (SIRT1 inhibitor) was introduced. Results indicated that SRT1720 in combination with fluoride significantly decreased the intracellular ROS levels, the protein expression of Caspase-3, Ac-p53 and p21 and alleviated apoptosis, while it was reversed by Ex-527. Collectively, SIRT1 plays an essential role in protection against fluoride-induced oxidative stress and mitochondria-dependent apoptosis in MC3T3-E1 cells. The SIRT1/p53/p21 pathway may be a potential therapeutic target for fluorosis.
    Keywords:  Apoptosis; Fluoride; MC3T3-E1 cells; SIRT1; p53
    DOI:  https://doi.org/10.1016/j.tiv.2019.02.006
  39. J Thorac Dis. 2018 Dec;10(12): 6921-6931
      Background: The aim of this study is to determine the role of nuclear factor kappa B (NF-κB)-mediated c-Jun N-terminal kinase (JNK) pathway in cognitive impairment induced by chronic intermittent hypoxia (CIH).Methods: Ninety-six male Sprague-Dawley rats were randomly divided into 8 groups: sham group, sustained hypoxia (SH) group, CIH group, CIH + melatonin group, CIH + vitamin E group, CIH + DMSO group, CIH + BAY 11-7082 group and CIH + normal saline (NS) group. Rats were exposed to normoxia, CIH (21% O2 for 60 s and 10% O2 for 60 s, cyclically repeated for 10 h/day) or SH (10% O2 for 10 h/day) for 14 days. Afterwards, Morris water maze test was conducted, and serum and hippocampus tissues were subjected to molecular biological and biochemical analyses.
    Results: Compared with the Sham and SH group, oxidative stress was induced by CIH in rat hippocampus with the high level of malondialdehyde (MDA) and 8-iso-PGF2α and the low level of superoxide dismutase (SOD) and glutathione (GSH). Activated NF-κB and its downstream products including tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6) and inducible nitric oxide synthase (iNOS) were highly expressed in CIH rats. These changes were attenuated by pretreatment of the rats with melatonin and vitamin E. CIH also resulted in hippocampus neuron apoptosis with increased caspase 3 level, dUIP nick end labeling (TUNEL)-positive neurons number and cognitive impairment verified by prolonged latency and shortened time in the target quadrant in Morris water maze test. JNK and its downstream transcriptional factors including c-Jun, activating transcription factor 2 (ATF2), and JunD were all significantly phosphorylated in CIH rats. However, pretreatment of NF-κB inhibitor BAY 11-7082 inhibited the activation of NF-κB under CIH condition and also significantly reduced the phosphorylation of JNK as well as c-Jun, ATF2, and JunD. Moreover, hippocampus neuron apoptosis and cognitive impairment were significantly improved with the pretreatment of BAY 11-7082 in rats subjected to CIH.
    Conclusions: These findings suggest that NF-κB-mediated JNK pathway is at least partially implicated in CIH-induced hippocampus neuron apoptosis and cognitive impairment. Inhibition of NF-κB activation provided a therapeutic potential for cognitive impairment in sleep apnea (SA).
    Keywords:  BAY 11-7082; Chronic intermittent hypoxia (CIH); c-Jun N-terminal kinase (JNK); melatonin; nuclear factor kappa B (NF-κB); vitamin E
    DOI:  https://doi.org/10.21037/jtd.2018.12.05
  40. J Cell Physiol. 2019 Feb 11.
      Tafazzin has been found to be associated with tumor progression. Mitochondrial homeostasis regulates cancer cell viability and metastasis. However, the roles of Tafazzin and mitochondrial homeostasis in thyroid cancer have not been explored. The aim of our study is to investigate the influences of Tafazzin on thyroid cancer apoptosis with a focus on mitochondrial fission. Our results indicated that Tafazzin deletion induced death in thyroid cancer via apoptosis. Biological analysis demonstrated that mitochondrial stress, including mitochondrial bioenergetics disorder, mitochondrial oxidative stress, and mitochondrial apoptosis, was activated by Tafazzin deletion. Furthermore, we found that Tafazzin affected mitochondrial stress by triggering inverted formin 2 (INF2)-related mitochondrial fission. The loss of INF2 sustained mitochondrial function and promoted cancer cell survival. Molecular investigation illustrated that Tafazzin regulated INF2 expression via the JNK signaling pathway; moreover, the blockade of JNK prevented Tafazzin-mediated INF2 expression and improved cancer cell survival. Taken together, our results highlight the key role of Tafazzin as a master regulator of thyroid cancer viability via the modulation of INF2-related mitochondrial fission and the JNK signaling pathway. These findings defined Tafazzin deletion and INF2-related mitochondrial fission as tumor suppressors that act by promoting cancer apoptosis via the JNK signaling pathway, with potential implications for new approaches to thyroid cancer therapy.
    Keywords:  INF2; JNK signaling pathway; Tafazzin; mitochondrial fission; thyroid cancer
    DOI:  https://doi.org/10.1002/jcp.28287
  41. Med Sci Monit. 2019 Feb 15. 25 1220-1231
      BACKGROUND PGC-1α can be activated by deacetylation reactions catalyzed by SIRT1. Resveratrol is currently known as a potent activator of SIRT1. However, it is unknown whether the renal-protective effect of resveratrol is further related to activation of the podocyte SIRT1/PGC-1α pathway. MATERIAL AND METHODS High glucose was used to stimulate mouse podocytes. Resveratrol and PGC-1α siRNA transfection were used to perform co-intervention treatments. The protein and mRNA expression levels of SIRT1, PGC-1α, NRF1, and TFAM were detect by immunofluorescence, Western blot analysis, and qRT-PCR in the podocytes, respectively. DCHF-DA and MitoSOX™ staining were used to monitor the total ROS and mitochondrial ROS levels, respectively. The specific activities of complexes I and III were measured using Complex I and III Assay Kits. Mitochondrial membrane potential and cell apoptosis were measured using JC-1 staining and Annexin V-FITC/PI double-staining, respectively. RESULTS We found that high-glucose stimulation results in time-dependent decreases in the expression of SIRT1, PGC-1α, and its downstream genes NRF1 and mitochondrial transcription factor A (TFAM) for mouse podocytes, and increases ROS levels in cells and mitochondria. Moreover, the expression of nephrin was downregulated and the cell apoptotic rate was increased. Resveratrol treatment can improve abnormalities caused by high-glucose stimulation. In addition, it can also reduce the release of mitochondrial cytochrome C and DIABLO proteins to the cytoplasm and increase respiratory chain complex I and III activity and mitochondrial membrane potential. CONCLUSIONS Resveratrol can reduce the oxidative damage and apoptosis of podocytes induced by high-glucose stimulation via SIRT1/PGC-1α-mediated mitochondrial protection.
    DOI:  https://doi.org/10.12659/MSM.911714
  42. Development. 2019 Feb 15. pii: dev169037. [Epub ahead of print]146(4):
      Non-apoptotic caspase activation involves multiple cellular events. However, the link between visible non-apoptotic caspase activation and its function in living organisms has not yet been revealed. Here, we visualized sub-lethal activation of apoptotic signaling with the combination of a sensitive indicator for caspase 3 activation and in vivo live-imaging analysis of Drosophila During thorax closure in pupal development, caspase 3 activation was specifically observed at the leading edge cells, with no signs of apoptosis. Inhibition of caspase activation led to an increase in thorax closing speed, which suggests a role of non-apoptotic caspase activity in cell motility. Importantly, sub-lethal activation of caspase 3 was also observed during wound closure at the fusion sites at which thorax closure had previously taken place. Further genetic analysis revealed that the activation of the initiator caspase Dronc is coupled with the generation of reactive oxygen species. The activation of Dronc also regulates myosin levels and delays wound healing. Our findings suggest a possible function for non-apoptotic caspase activation in the fine-tuning of cell migratory behavior during epithelial closure.
    Keywords:  Drosophila; Epithelia; Live imaging; Non-apoptotic caspase activation; Thorax closure; Wound healing
    DOI:  https://doi.org/10.1242/dev.169037
  43. J Physiol Sci. 2019 Feb 15.
      An uncontrolled inflammatory response has been implicated in the progression of acute liver failure through poorly understood mechanisms. The aim of our study was to investigate whether suramin attenuates inflammation-mediated hepatocyte apoptosis by modulating mitochondrial homeostasis. Primary hepatocytes were isolated from mice and treated with LPS in vitro in the presence or absence of suramin. Western blotting, immunofluorescence staining, and ELISAs were used to evaluate the mitochondrial stress. The LPS treatment caused hepatocyte death via apoptosis. Interestingly, suramin supplementation attenuated LPS-mediated hepatocyte death by reducing Mst1 expression; the overexpression of Mst1 abolished the anti-apoptotic effects of suramin on LPS-treated hepatocytes. At the molecular level, suramin treatment repressed mitochondrial oxidative stress, sustained mitochondrial dynamics and blocked the caspase-9-mediated mitochondrial apoptosis pathway; these effects of suramin were achieved by reversing Mst1 expression. Furthermore, our study found that suramin modulated Mst1 expression via the JNK signaling pathway. Activation of JNK prevented the suramin-mediated Mst1 downregulation and concomitantly increased hepatocyte apoptosis and mitochondrial dysfunction. Taken together, our results confirmed the anti-apoptotic and anti-inflammatory effects of suramin on LPS-challenged hepatocytes. Suramin sustained hepatocyte viability and attenuated mitochondrial stress via repressing the JNK-Mst1 signaling pathway.
    Keywords:  Hepatocyte death; Inflammation; JNK pathway; Mitochondria; Mst1; Suramin
    DOI:  https://doi.org/10.1007/s12576-019-00666-9
  44. Food Chem Toxicol. 2019 Feb 07. pii: S0278-6915(19)30063-8. [Epub ahead of print]125 549-561
      Cutaneous melanoma has a high capacity to metastasize and significant resistance to conventional therapeutic protocols, which makes its treatment difficult. The combination of conventional drugs with cytostatic molecules of low toxicity has been shown to be an interesting alternative for sensitization of tumor cells to chemotherapy. In this study, we evaluated the effect of bixin, an abundant apocarotenoid present in Bixa orellana, on the sensitization of human melanoma cells (A2058) to dacarbazine treatment, an anticancer agent clinically used for the therapy of metastatic melanoma. UPLC-DAD-MS/MS analyses of bioactive extracts from B. orellana seeds led to the identification of two new apocarotenoids: 6,8'-diapocarotene-6,8'-dioic acid and 6,7'-diapocarotene-6,7'-dioic acid. After being identified as its major compound, bixin (Z-bixin) was evaluated on A2058 cells expressing the oncogenic BRAF VE600 mutation and resistant to dacarbazine treatment. Bixin promoted growth inhibition, reduced cell migration, induced apoptosis and cell cycle arrest in the G2/M phase. When associated with dacarbazine, bixin restored the sensitivity of A2058 cells to chemotherapy, enhancing its antiproliferative, anti-migratory and pro-apoptotic effects. Combined treatment also induced higher ROS (reactive oxygen species) and MDA (malondialdehyde, a lipid peroxidation marker) generation than monotreatment, suggesting that the oxidative stress caused by bixin contributes significantly to its sensitizing effect. Taken together, these data suggest that bixin exerts intrinsic antimelanoma activity by mechanisms complementary to those of dacarbazine, encouraging its use in combined therapy for cutaneous melanoma treatment.
    Keywords:  Annatto; Carotenoids; Dacarbazine; Melanoma; Multidrug resistance
    DOI:  https://doi.org/10.1016/j.fct.2019.02.013
  45. Neural Regen Res. 2019 Jun;14(6): 948-953
      Nicotinamide adenine dinucleotide phosphate oxidase (NOX) is a multisubunit enzyme complex that utilizes nicotinamide adenine dinucleotide phosphate to produce superoxide anions and other reactive oxygen species. Under normal circumstances, reactive oxygen species mediate a number of important cellular functions, including the facilitation of adaptive immunity. In pathogenic circumstances, however, excess reactive oxygen species generated by NOX promotes apoptotic cell death. In ischemic stroke, in particular, it has been shown that both NOX activation and derangements in glucose metabolism result in increased apoptosis. Moreover, recent studies have established that glucose, as a NOX substrate, plays a vital role in the pathogenesis of reperfusion injury. Thus, NOX inhibition has the potential to mitigate the deleterious impact of hyperglycemia on stroke. In this paper, we provide an overview of this research, coupled with a discussion of its implications for the development of NOX inhibition as a strategy for the treatment of ischemic stroke. Both inhibition using apocynin, as well as the prospect of developing more specific inhibitors based on what is now understood of the biology of NOX assembly and activation, will be highlighted in the course of our discussion.
    Keywords:  NADPH; NOX; hyperglycolysis; ischemia/reperfusion; neuroprotection; nicotinamide adenine dinucleotide phosphate oxidase; nicotinamide adenine dinucleotide phosphate oxidase inhibitors; reactive oxygen species; stroke
    DOI:  https://doi.org/10.4103/1673-5374.250568
  46. Exp Hematol. 2019 Feb 12. pii: S0301-472X(19)30069-4. [Epub ahead of print]
      The BCR-ABL-negative myeloproliferative neoplasms (MPN) are driven by JAK-STAT pathway activation but epigenetic alterations also play an important pathophysiological role. These can be pharmacologically manipulated with histone deacetylase inhibitors (HDACis), which have proven to be clinically effective in the treatment of MPN but exhibiting dose-limiting toxicities. The treatment of primary MPN cells with Vorinostat modulates the expression of genes associated with apoptosis, cell cycle, inflammation and signalling. The induction of this transcriptional program results a decreased cellular viability, paralleled by a decrease in Reactive Oxygen Species (ROS) levels. In vitro manipulation of ROS levels showed that the reduction of ROS levels promoted apoptosis. When Vorinostat was combined with anti-oxidant agents, the apoptosis of MPN cells was increased in a synergistic manner. The results described here suggest a novel and promising therapeutic strategy to treat MPN combining HDACis with ROS reducing agents.
    Keywords:  Antioxidants; Epigenetic; Myeloproliferative Neoplasms; Reactive Oxygen Species; Vorinostat
    DOI:  https://doi.org/10.1016/j.exphem.2019.02.002
  47. J Pharmacol Exp Ther. 2019 Feb 13. pii: jpet.118.254862. [Epub ahead of print]
      Ibrutinib (IBT), the first-in-class inhibitor of Bruton's tyrosine kinase (BTK), has demonstrated clinical activity against various B-cell malignancies. IBT is the second agent approved by U.S. Food and Drug Administration via Breakthrough Therapy Designation. Aside from its therapeutic mechanism through BTK inhibition, IBT has other target sites reported for cancer therapy, leading us to investigate whether IBT has unreported targets. Our study revealed that IBT can inhibit the growth of SMMC-7721 cells through irreversible inhibition of mammalian thioredoxin reductase (TrxR) enzymes, which are components of the thioredoxin (Trx) system. Further study demonstrated that IBT can cause cellular ROS elevation and induce oxidative stress-mediated apoptosis. The discovery of a new target of IBT sheds light on better understanding its anticancer mechanisms and provides a theoretical foundation for its further use in clinical therapy.
    Keywords:  cell death; drug targeting; molecular drug targeting
    DOI:  https://doi.org/10.1124/jpet.118.254862
  48. Chem Sci. 2019 Jan 07. 10(1): 293-309
      A panel of iridium(iii) porphyrin complexes containing axial N-heterocyclic carbene (NHC) ligand(s) were synthesized and characterized. X-ray crystal structures of the bis-NHC complexes [IrIII(ttp)(IMe)2]+ (2a), [IrIII(oep)(BIMe)2]+ (2d), [IrIII(oep)(I i Pr)2]+ (2e) and [IrIII(F20tpp)(IMe)2]+ (2f) display ruffled porphyrin rings with mesocarbon displacements of 0.483-0.594 Å and long Ir-CNHC bonds of 2.100-2.152 Å. Variable-temperature 1H NMR analysis of 2a reveals that the macrocycle porphyrin ring inversion takes place in solution with an activation barrier of 40 ± 1 kJ mol-1. The UV-vis absorption spectra of IrIII(por)-NHC complexes display split Soret bands. TD-DFT calculations and resonance Raman experiments show that the higher-energy Soret band is derived from the 1MLCT dπ(Ir) → π*(por) transition. The near-infrared phosphorescence of IrIII(por)-NHC complexes from the porphyrin-based 3(π, π*) state features broad emission bands at 701-754 nm with low emission quantum yields and short lifetimes (Φ em < 0.01; τ < 4 μs). [IrIII(por)(IMe)2]+ complexes (por = ttp and oep) are efficient photosensitizers for 1O2 generation (Φ so = 0.64 and 0.88) and are catalytically active in the light-induced aerobic oxidation of secondary amines and arylboronic acid. The bis-NHC complexes exhibit potent dark cytotoxicity towards a panel of cancer cells with IC50 values at submicromolar levels. The cytotoxicity of these complexes could be further enhanced upon light irradiation with IC50 values as low as nanomolar levels in association with the light-induced generation of reactive oxygen species (ROS). Bioimaging of [IrIII(oep)(IMe)2]+ (2c) treated cells indicates that this Ir complex mainly targets the endoplasmic reticulum. [IrIII(oep)(IMe)2]+ catalyzes the photoinduced generation of singlet oxygen and triggers protein oxidation, cell cycle arrest, apoptosis and the inhibition of angiogenesis. It also causes pronounced photoinduced inhibition of tumor growth in a mouse model of human cancer.
    DOI:  https://doi.org/10.1039/c8sc02920b
  49. Front Pharmacol. 2018 ;9 1562
      Chronic hyperglycemia and unusually high oxidative stress are the key contributors for diabetes in humans. Since nuclear factor E2-related factor 2 (Nrf2) controls the expression of antioxidant- and detoxification genes, it is hypothesized that targeted activation of Nrf2 using phytochemicals is likely to protect pancreatic β-cells, from oxidative damage, thereby mitigates the complications of diabetes. Naringenin is one such activator of Nrf2. However, it is currently not known whether the protective effect of naringenin against streptozotocin (STZ) induced damage is mediated by Nrf2 activation. Hence, the potential of naringenin to activate Nrf2 and protect pancreatic β-cells from STZ-induced damage in MIN6 cells is studied. In MIN6 cells, naringenin could activate Nrf2 and its target genes GST and NQO1, thereby inhibit cellular apoptosis. In animals, administration of 50 mg/kg body weight naringenin, for 45 days, significantly decreased STZ-induced blood glucose levels, normalized the lipid profile, and augmented the levels of antioxidants in pancreatic tissues. Immunohistochemical analysis measuring the number of insulin-positive cells in pancreas showed restoration of insulin expression similar to control animals. Furthermore, naringenin promoted glycolysis while inhibiting gluconeogenesis. In conclusion, naringenin could be a good anti-diabetic agent, which works by promoting Nrf2 levels and by decreasing cellular oxidative stress.
    Keywords:  MIN6 cells; Nrf2; apoptosis; diabetes; naringenin; streptozotocin
    DOI:  https://doi.org/10.3389/fphar.2018.01562
  50. Int J Biol Sci. 2019 ;15(2): 325-340
      Random-pattern skin flaps are widely used to close defects in reconstructive and plastic surgeries; however, they are vulnerable to necrosis, particularly in the distal portion of the flap. Here, we examined the effects of metformin on flap survival and the mechanisms underlying these effects. Following metformin treatment, the survival area, blood flow, and number of microvessels present in skin flaps were increased on postoperative day 7, whereas tissue edema was reduced. In addition, metformin promoted angiogenesis, inhibited apoptosis, relieved oxidative stress, and increased autophagy in areas of ischemia; these effects were reversed by autophagy inhibitors 3-methyladenine (3MA) or chloroquine (CQ). Either 3MA or CQ reversed the metformin-induced increase in flap viability. Moreover, metformin also activated the AMPK-mTOR-TFEB signaling pathway in ischemic areas. Inhibitions of AMPK via Compound C (CC) or AMPK shRNA adeno-associated virus (AAV) vector resulted in the downregulation of the AMPK-mTOR-TFEB signaling pathway and autophagy level in metformin-treated flaps. Taken together, our findings suggest that metformin improves the survival of random-pattern skin flaps by enhancing angiogenesis and suppressing apoptosis and oxidative stress. These effects result from increased autophagy mediated by activation of the AMPK-mTOR-TFEB signaling pathway.
    Keywords:  AMPK-mTOR-TFEB signaling pathway; Autophagy; Metformin; Random-pattern skin flaps
    DOI:  https://doi.org/10.7150/ijbs.29009
  51. Connect Tissue Res. 2019 Feb 12.
      PURPOSE: Neuronal theory regarding rotator cuff degeneration has developed from the findings that glutamate, an amino acid and an excitatory neurotransmitter, is present in increased concentrations in tendon tissues with tendinopathy and that glutamate induces cell death in fibroblasts of origin in rat supraspinatus tendon. The purpose of the current study was to determine whether N-acetylcysteine (NAC) has cytoprotective effects against glutamate-induced fibroblast death.MATERIALS AND METHODS: Primary cultured fibroblasts were obtained from rat supraspinatus tendons. Varying concentrations of glutamate (0.5, 1, 5, and 10 mM) and of NAC (0.5, 1, 2, and 5 mM) were used for evaluation of cytotoxicity. Cell viability, cell cycles, types of cell death, intracellular ROS production, expressions of caspase-3/7, and Ca2+ influx were evaluated.
    RESULTS: Glutamate significantly induced cell death, apoptosis, and Ca2+ influx and significantly increased caspase-3/7 activity and intracellular ROS production (p < 0.001). NAC significantly reduced the glutamate-induced cell death, apoptosis, Ca2+ influx, caspase-3/7 activity, and intracellular ROS production (p < 0.001).
    CONCLUSIONS: The glutamate-induced cytotoxic effects can be reduced by NAC, an antioxidant, through the reduction of intracellular oxidative stress and/or Ca2+ influx.
    Keywords:  Antioxidant; Excitotoxicity; Neurotransmitter; Rotator cuff tendon; Tendinopathy
    DOI:  https://doi.org/10.1080/03008207.2019.1580702
  52. Toxicol Ind Health. 2019 Feb 12. 748233718824911
      Di(2-ethylhexyl)phthalate (DEHP) is a typical endocrine-disrupting chemical and reproductive toxicant. Although previous studies have attempted to describe the mechanism by which DEHP exposure results in reproductive dysfunction, few studies focused on puberty, a critical period of reproductive development, and the increased susceptibility to injury in adolescents. To elucidate the mechanism underpinning the testicular effects of DEHP in puberty, we sought to investigate the JAZF1/TR4 pathway in the testes of pubertal rats. Specifically, we focused on the role of the JAZF1/TR4 pathway in male reproduction, including the genes JAZF1, TR4, Sperm 1, and Cyclin A1. In the present study, rats were exposed to increasing concentrations of DEHP (0, 250, 500, and 1000 mg/kg/day) by oral gavages for 30 days. Then we assayed testicular zinc and oxidative stress levels. Our results indicated that DEHP exposure could lead to oxidative stress and decrease the contents of testicular zinc. Additionally, significant morphological changes and cell apoptosis were observed in testes exposed to DEHP, as identified by hematoxylin and eosin staining and the terminal deoxynucleotidyl transferase-mediated nick and labeling assay. By measuring the expression levels of the above relevant genes by qPCR, we found the DEHP-induced increased expression of JAZF1 and decreased expression of TR4, Sperm 1, and Cyclin A1. Therefore, we have demonstrated that in vivo exposure to DEHP might induce reproductive toxicity in pubertal male rats through the JAZF1/TR4 pathway and oxidative stress.
    Keywords:  Di(2-ethylhexyl)phthalate; JAZF1/TR4 pathway; oxidative stress; pubertal male rats; reproductive toxicity
    DOI:  https://doi.org/10.1177/0748233718824911
  53. Life Sci. 2019 Feb 11. pii: S0024-3205(19)30111-0. [Epub ahead of print]
      AIMS: The therapeutic strategies for diabetic retinopathy (DR) are disappointing. Ginsenoside Rg1 (Rg1) extracted from Panax ginseng can induce glucose uptake and lower oxidative stress. We aimed to explore the effect of Rg1 on DR using human retinal pigment epithelium cells (ARPE-19).MAIN METHODS: ARPE-19 cells were grown in high glucose (HG) to simulate DR. Cell viability, apoptosis, ROS generation and miR-26a level were examined by CCK-8 assay, flow cytometry assay, DCFH-DA staining and RT-qPCR, respectively. Expression of proteins associated with viability, apoptosis and oxidative stress was measured by Western blot analysis. Effects of Rg1 on HG-induced alteration were assessed. Moreover, whether miR-26a was involved in Rg1-associated modulation was verified. Finally, the involvements of the ERK and Wnt/β-catenin pathways were analyzed by Western blot analysis.
    KEY FINDINGS: HG reduced cell viability while promoted apoptosis and oxidative stress in ARPE-19 cells. Rg1 ameliorated HG-induced cell injury. The expression of miR-26a was up-regulated by Rg1 in HG-treated cells, and miR-26a inhibition obviously reversed the effects of Rg1 on HG-treated cells. Finally, we found the ERK and Wnt/β-catenin pathways were inhibited by Rg1 through up-regulation of miR-26a.
    SIGNIFICANCE: Rg1 protected ARPE-19 cells against HG-induced injury through up-regulating miR-26a, along with inhibition of the ERK and Wnt/β-catenin pathways. Rg1 might be a potential therapeutic drug for DR.
    Keywords:  Diabetic retinopathy; ERK/Wnt/β-catenin; Ginsenoside Rg1; High glucose; microRNA-26a
    DOI:  https://doi.org/10.1016/j.lfs.2019.02.021
  54. J Cell Physiol. 2019 Feb 15.
      The antimetabolite 5-fluorouracil (5-FU) is a widely used antitumor agent, however the overall response rate to 5-FU as a single agent is usually limited. Herein, how Lachnum expolysaccharide (LEP-2a), a type of active polysaccharide isolated from Lachnum sp., acted synergistically with 5-FU on HepG2 cells was investigated. It was found that LEP-2a notably enhanced 5-FU sensitivity in HepG2 cells in a synergistic manner. After combination treatment of 5-FU and LEP-2a, Ras/Raf/MEK/ERK and PI3K/AKT/mTOR pathway were inactivated. In addition, combination treatment induced generation of reactive oxygen species, decreased the levels of intracellular antioxidant enzymes and triggered mitochondrial apoptosis pathway. Furthermore, 5-FU combined with LEP-2a also resulted in p53 activation and NF-κB inhibition, and cell cycle arrest in the S phase as well as cell metastasis stagnation. Interestingly, LEP-2a treatment also blocked the DNA damage repair procedure. These findings demonstrate that LEP-2a enhanced 5-FU sensitivity and combination of 5-FU and LEP-2a exerts synergistic antitumor efficiency through multiple approaches.
    Keywords:  5-fluorouracil (5-FU); Lachnum exopolysaccharide (LEP-2a); hepatocellular carcinoma (HCC); molecular mechanism; synergistic action
    DOI:  https://doi.org/10.1002/jcp.28202