bims-aporos Biomed News
on Apoptosis and reactive oxygen species
Issue of 2019‒03‒17
sixty-five papers selected by
Gavin McStay
Staffordshire University


  1. Cell Cycle. 2019 Mar 15.
      Glaucoma is a neurodegenerative disorder that generally accepted as the main cause of vision loss. In this study, we tested the hypothesis that laminin α4 (LAMA4) is implicated in glaucoma development by controlling apoptosis of retinal ganglion cells (RGCs) through the mitogen-activated protein kinase (MAPK) signaling pathway. Expression profiles and genes associated with glaucoma were searched to determine the objective gene. Intraocular pressure (IOP) rats model were established and IOP was measured. The mRNA and protein expression of LAMA4, JNK, p38 MAPK, ERK, Bcl-2, Bax, Caspase-9, and p53 was determined in concert with the treatment of H2O2, si-NC, or si-LAMA4 in cultured RGCs. Viability of RGCs, reactive oxygen species (ROS) and cell apoptosis were also measured. LAMA4 was selected as the study object because of its significant difference in two expression profiles. IOP of rats with glaucoma increased significantly after model establishment, and the LAMA4 protein expression in retinal tissue of rats with glaucoma was elevated. Down-regulation of LAMA4 could inhibit the mRNA and protein expression of LAMA4, JNK, p38 MAPK, ERK, Bax, Caspase-9, and p53, as well as restrain the apoptosis and ROS of RGCs, but improve Bcl-2 expression and viability of RGCs. Collectively, the obtained data supported that down-regulated LAMA4 might reduce the oxidative stress-induced apoptosis of glaucoma RGCs by inhibiting the activation of the MAPK signaling pathway.
    Keywords:  Glaucoma; LAMA4; MAPK signaling pathway; Oxidative stress; Retinal ganglion cells
    DOI:  https://doi.org/10.1080/15384101.2019.1593645
  2. Int J Mol Med. 2019 Mar 12.
      Myrica rubra is well known for its delicious taste and high nutritional value. The present study investigated the potential protective effects and mechanisms of M. rubra flavonoids (MRF) extract on isoproterenol (ISO)‑induced myocardial injury in rats and hypoxia/reoxygenation (H/R) injury in H9c2 cardiomyocytes. An in vivo study revealed that MRF decreased serum cardiac enzyme levels, ameliorated pathological heart alterations and increased the antioxidant potential. The in vitro investigation demonstrated that MRF inhibited cell death, reactive oxygen species (ROS) accumulation, mitochondrial membrane depolarization, apoptosis rate and caspase‑3 activation and enhanced the Bcl‑2/Bax ratio during H/R injury. These effects were accompanied by the phosphorylation of protein kinase B (Akt) and glycogen synthase kinase (GSK)‑3β. Further mechanism studies demonstrated that LY294002, a specific inhibitor of phosphoinositide 3‑kinase (PI3K), abolished the MRF‑mediated cardioprotection against H/R‑induced apoptosis and ROS overproduction. Collectively, these results suggested that MRF exerts cardioprotective effects by attenuating oxidative damage and cardiomyocyte apoptosis most likely via a PI3K/Akt/GSK3β‑dependent mechanism.
    DOI:  https://doi.org/10.3892/ijmm.2019.4131
  3. Front Mol Neurosci. 2019 ;12 42
      This study aimed to explore the neuroprotective effects and mechanisms of natrium benzoate (NaB) and DJ-1 in attenuating reactive oxygen species (ROS)-induced neuronal apoptosis in traumatic spinal cord injury (t-SCI) in rats. T-SCI was induced by clip compression. The protein expression and neuronal apoptosis was evaluated by Western blotting, double immunofluorescence staining and transmission electron microscope (TEM). ROS level, spinal cord water content (SCWC) and Evans blue (EB) extravasation was also examined. Locomotor function was evaluated by Basso, Beattie, and Bresnahan (BBB) and inclined plane test (IPT) scores. We found that DJ-1 is expressed in spinal cord neurons and increased after t-SCI. At 24 h post-injury, the levels of DJ-1, p-Akt, SOD2, ROS, p-p38 MAPK/p38 MAPK ratio, and CC-3 increased, while the Bcl-2/Bax ratio decreased. NaB upregulated DJ-1, p-Akt, and SOD2, decreased ROS, p-p38 MAPK/p38 MAPK ratio, and CC-3, and increased the Bcl-2/Bax ratio, which were reversed by DJ-1 siRNA. The proportion of CC-3- and TUNEL-positive neurons also increased after t-SCI and was reduced by NaB. These effects were reversed by MK2206. Moreover, the level of oxDJ-1 increased after t-SCI, which was decreased by DJ-1 siRNA, NaB or the combination of them. NaB also reduced mitochondrial vacuolization, SCWC and EB extravasation, and improved locomotor function assessed by the BBB and IPT scores. In conclusion, NaB increased DJ-1, and thus reduced ROS and ROS-induced neuronal apoptosis by promoting Akt phosphorylation in t-SCI rats. NaB shows potential as a therapeutic agent for t-SCI, with DJ-1 as its main target.
    Keywords:  DJ-1; apoptosis; natrium benzoate; oxidative stress; reactive oxygen species; traumatic spinal cord injury
    DOI:  https://doi.org/10.3389/fnmol.2019.00042
  4. Artif Cells Nanomed Biotechnol. 2019 Dec;47(1): 747-756
      In recent years, numerous studies have confirmed the role of chitosan nanoparticles (CS NPs) as a promising drug delivery carrier for improving the efficiency of anticancer drug in the treatment of cancer. However, the possible biological effects of CS NPs on tumour cells and underlying mechanisms are still unclear. Recently, reactive oxygen species (ROS)-mediated cell apoptosis has been implicated in the regulation of cell death. In this study, we found that CS NPs induced the massive generation of ROS and resulted in apoptosis of hepatocellular carcinoma cells (SMMC-7721) through activating the mitochondrial pathway and endoplasmic reticulum stress. These results suggest an important role of ROS in CS NPs-induced cancer cell death.
    Keywords:  Chitosan; endoplasmic reticulum; mitochondrial; nanoparticles; reactive oxygen species
    DOI:  https://doi.org/10.1080/21691401.2019.1577876
  5. Neurotoxicology. 2019 Mar 08. pii: S0161-813X(18)30359-0. [Epub ahead of print]
      Extract of Ulmus wallichiana is being used as traditional medicine used for the treatment of fractured bones however the effect of its individual flavonols is not known. The present study was conducted to investigate the effect of its novel flavonol, (2S, 3S)-(+)-30, 40, 5, 7-tetrahydroxydihydroflavonol-6-C-b-D-glucopyranoside named as Ulmoside A (UA), on lipopolysaccharides (LPS) treated neurons. LPS treatment to neuronal cells caused significant cytotoxicity, reactive oxygen species generation, depletion in glutathione and mitochondrial impairment which were significantly inhibited with UA treatment. LPS treatment also caused significant translocation of cytochrome-c, decreased level of Bcl2, increased level of Bax and cleaved caspase-3 in neuronal cells reflecting the involvement of intrinsic apoptotic pathway in neuronal death which was attenuated with UA treatment. Since LPS is a well known pro-inflammatory agent it also offered the significant increase in proinflammatory cytokines (tumor necrosis factors-α & interleukin 1-beta) however, UA treatment did not exhibit significant inhibition against LPS induced inflammatory response. LPS also caused the augmented level of inducible nitric oxide synthase (iNOS) which was also not inhibited with co treatment of UA. We have also observed the significant DNA fragmentation and augmented level of cleaved Poly (ADP-Ribose) polymerase 1 after LPS treatment which was significantly reverted with UA treatment. Findings suggested that UA acts through mitochondria and exhibited its anti-oxidative and anti-apoptotic activities in neuronal cells while no significant anti-inflammatory activity and effect on iNOS were observed.
    Keywords:  Neuroprotection; Ulmus wallichiana; antiapoptotic; antioxidative
    DOI:  https://doi.org/10.1016/j.neuro.2019.02.017
  6. Cancer Lett. 2019 Mar 10. pii: S0304-3835(19)30147-8. [Epub ahead of print]
      The repurposing of existing FDA-approved non-cancer drugs is a potential source of new treatment options for cancer patients. An anti-inflammatory drug, 5-aminosalicylic acid (5-ASA), has been clinically used to treat inflammatory bowel disease. Hyperthermia (HT) is widely applicable addendum therapy with the existing cancer treatment modalities. Here, we addressed how 5-ASA combined with HT induces lethal effects in human oral squamous cell carcinoma (OSCC) HSC-3 cells. We found that 5-ASA/HT combination significantly inhibited the viability of HSC-3 cells, while cytotoxic effects in primary human dermal fibroblast cells were minor. Apoptotic endpoints were significantly increased by the 5-ASA/HT combined treatment, as evidenced by presence of Annexin-V/PI positive cells, loss of MMP, Bcl-2/Bax ratio alteration, and increased Fas, cleaved Bid, and caspase expression. Interestingly, the enhancement of apoptosis was reversed in the presence of ON/ONOO- scavengers. These findings indicate that the combination treatment enhances apoptosis via ON/ONOO- mediated ER stress-Ca2+-mitochondria signaling and caspase-dependent apoptotic pathways. Our findings provide novel evidence that the combination of 5-ASA and HT is a promising approach for the enhancement of apoptosis; it may serve as an effective strategy for treating human OSCC.
    Keywords:  Cancer cell death; Drug repurposing; OSCC; Reactive oxygen and nitrogen species
    DOI:  https://doi.org/10.1016/j.canlet.2019.03.004
  7. Chem Biol Interact. 2019 Mar 11. pii: S0009-2797(19)30140-1. [Epub ahead of print]
      1,4-Naphthoquinone compounds are a class of organic compounds derived from naphthalene. They exert a wide variety of biological effects, but when used as anticancer drugs, have varying levels of side effects. In the present study, in order to reduce toxicity and improve the antitumor activity, we synthesized two novel 1,4-naphthoquinone derivatives, 2-(butane-1-sulfinyl)-1,4-naphthoquinone (BSQ) and 2-(octane-1-sulfinyl)-1,4-naphthoquinone (OSQ). We investigated the antitumor effects of BSQ and OSQ in human lung cancer cells and the underlying molecular mechanisms of these effects, focusing on the relationship between these compounds and reactive oxygen species (ROS) production. MTT assay and trypan blue exclusion assay results showed that BSQ and OSQ had significant cytotoxic effects in human lung cancer cells. Flow cytometry results indicated that the number of apoptotic cells and the intracellular ROS levels significantly increased after treatment with BSQ and OSQ. However, cell apoptosis was inhibited by pretreatment with the ROS scavenger N-acetyl-l-cysteine (NAC). Western blotting results showed that BSQ and OSQ increased the expression levels of p-p38 kinase and p-c-Jun N-terminal kinase (p-JNK), and decreased the expression levels of p-extracellular signal-regulated kinase (p-ERK), p-protein kinase B (p-Akt), and p-signal transducer and activator of transcription-3 (p-STAT3). These phenomena were blocked by mitogen-activated protein kinase (MAPK) inhibitors, Akt inhibitors and NAC. In conclusion, BSQ and OSQ induce human lung cancer A549 cell apoptosis by ROS-mediated MAPKs, Akt, and STAT3 signaling pathways. Therefore, BSQ and OSQ may be therapeutic potential agents for the treatment of human lung cancer.
    Keywords:  1,4-Naphthoquinone derivatives; Apoptosis; Lung cancer cells; MAPKs; Reactive oxygen species; STAT3
    DOI:  https://doi.org/10.1016/j.cbi.2019.03.004
  8. 3 Biotech. 2019 Mar;9(3): 115
      In this study, Asc-s was evaluated for anti-cancer effect using cervical cancer cells (HeLa). Results determine that Asc-s treatment-induced dose-dependent inhibition of proliferation of HeLa cells and induced apoptosis. Flow-cytometry analysis shows Asc-s treatment-induced accumulation of cells at sub-G0/G1 stage of cell cycle and induced apoptosis as confirmed by DAPI, propodium iodide, and acridine staining in HeLa cells. Asc-s entered the cells and metabolized to ascorbate and stearate moieties, increased membrane permeability, and decreased membrane fluidity in HeLa cells. Asc-s treatment-induced dose-dependent increase in autophagy protein LC3-II, mRNA levels and decreased Nrf-2 levels in HeLa cells. It is hypothesized that both ascorbyl radical and stearoyl moieties of Asc-s induced cytotoxicity by generating reactive oxygen species (ROS) and modulating membrane fluidity/permeability leading to apoptosis/autophagy of HeLa cells. Thus, our findings demonstrate that Asc-s as anti-proliferative and apoptosis inducing compound in cervical cancer cells.
    Keywords:  Ascorbyl stearate; Autophagy/ apoptosis; Gas chromatography; HeLa cancer cells–anti-proliferation; Liquid chromatography
    DOI:  https://doi.org/10.1007/s13205-019-1628-5
  9. Artif Cells Nanomed Biotechnol. 2019 Dec;47(1): 626-635
      Shikonin, a botanical drug extracted from Lithospermum erythrorhizon, exhibits anti-cancer effects in various cancer cell lines. However, the mechanisms underlying these effects have not been completely elucidated yet. Here, we showed that Shikonin induces apoptosis and autophagy in A375 cells and inhibits their proliferation. Shikonin caused G2/M phase arrest through upregulation of p21 and downregulation of cyclin B1. Shikonin significantly triggered ER stress-mediated apoptosis by upregulating the expression of p-eIF2α, CHOP, and cleaved caspase-3. It also induced protective autophagy by activating the p38 pathway, followed by an increase in the levels of p-p38, LC3B-II, and Beclin 1. Upon suppression of autophagy by 3-methyladenine, Shikonin-induced apoptosis was enhanced in A375 cells. Moreover, after pretreatment with N-acetyl-cysteine, Shikonin increased the production of reactive oxygen species that are involved in regulating ER stress-mediated apoptosis and p38-activated autophagy, as evidenced by the reversion of cell viability and apoptosis and a decrease in p-eIF2α, CHOP, p-p38, LC3B-II, and Beclin 1 levels. Thus, we demonstrated that Shikonin induced apoptosis and autophagy in A375 cells via the activation of ROS-mediated ER stress and p38 pathways, indicating that Shikonin can serve as a potential agent for human melanoma therapy.
    Keywords:  ER stress; ROS; Shikonin; apoptosis; autophagy; p38
    DOI:  https://doi.org/10.1080/21691401.2019.1575229
  10. J Ethnopharmacol. 2019 Mar 11. pii: S0378-8741(19)30198-9. [Epub ahead of print]
      ETHNOPHARMACOLOGICAL RELEVANCE: Zhi-zi-chi Decoction(ZZCD), a traditional Chinese medicine formula, has been reported its potential protective effect on psychological sub-health diseases. However, there still remains a lack of molecular mechanism interpretation.AIM OF THE STUDY: This study was aimed at investigating the mechanism of glutamate-induced toxicity in PC12 cells and the neuroprotective effect of ZZCD based on a novel strategy of the combination of cell metabolomics and pharmacology.
    MATERIALS AND METHODS: The PC12 cells were treated with glutamate to simulate neurotoxic cell model. Gas chromatography coupled with mass spectrometry based on cell metabolomics approach was performed to comprehensively investigate the molecular mechanism of glutamate-induced toxicity The cell viability and cytotoxicity analysis, the determination of glutathione reductase(GR), superoxide dismutase(SOD) and reactive oxygen species(ROS), apoptosis analysis and western blot analysis were performed to evaluate the neuroprotection of ZZCD.
    RESULTS: Forty metabolites were identified as potential biomarkers in model cells by principal components analysis(PCA) and orthogonal partial least squares-discriminant analysis(OPLS-DA). Glutamate decreased the GR and SOD activities, increased the level of intracellular ROS, activated the apoptotic pathway, and induced the changes of energy metabolism, amino acid metabolism and lipid metabolism. In addition, the extract of ZZCD could reverse the disturbed metabolic pathways by regulating those potential biomarkers and exerted anti-oxidation and anti-apoptosis.
    CONCLUSION: ZZCD has neuroprotective effect and the novel strategy can be applicable for other traditional Chinese medicine formulas.
    Keywords:  Metabolomics; Neuroprotective effect; PC12 cell; ZZCD
    DOI:  https://doi.org/10.1016/j.jep.2019.03.021
  11. Cytotechnology. 2019 Mar 15.
      Extracts of the cherry blossom plant have been reported to exert various biological effects on human cells. However, no previous investigations have examined the antioxidant and anti-apoptotic effects of these extracts on ultraviolet B (UVB) radiation-induced skin aging. This study explores the underlying mechanisms of the antioxidant and anti-apoptotic effects of cherry blossom extract (CBE) in human keratinocyte (HaCaT) cells. HaCaT cells were treated with CBE at concentrations of 0.5, 1.0, and 2.0% for 24 h and then irradiated with UVB (40 mJ/cm2). CBE effectively and dose-dependently decreased the levels of reactive oxygen species and malondialdehyde, while increasing the activities of superoxide dismutase and glutathione peroxidase. Pretreatment with 1 and 2% CBE attenuated UVB-induced DNA damage by reducing the formation of cyclobutane pyrimidine dimers and 8-hydroxy-20-deoxyguanosine. Furthermore, CBE also prevented UVB-induced apoptosis and significantly downregulated B cell lymphoma 2 (Bcl-2)-associated X, cytochrome-c, and caspase-3 expression, while upregulating Bcl-2 expression. Taken together, these results indicate that CBE protects HaCaT cells from UVB-induced oxidative stress and apoptosis and suggest that CBE could be a potent antioxidant against skin aging.
    Keywords:  Anti-apoptosis; Antioxidant; Cherry blossom extract; HaCaT cells; Skin aging
    DOI:  https://doi.org/10.1007/s10616-018-0215-7
  12. PeerJ. 2019 ;7 e6565
      Background: High-yielding dairy cows are prone to oxidative stress due to the high metabolic needs of homeostasis and milk production. Oxidative stress and inflammation are tightly linked; therefore, anti-inflammatory and/or natural antioxidant compounds may help improve mammary cell health. Baicalin, one of the major flavonoids in Scutellaria baicalensis, has natural antioxidant and anti-inflammatory properties in various cell types, but its effects on bovine mammary epithelial cells (BMECs) have not been investigated.Methods: Explants from bovine mammary glands were collected by biopsy at the peak of lactation (approximately 60 days after the start of lactation) (n = three animals) to isolate BMECs corresponding to mature secretory cells. Cell viability, apoptosis, proliferative capacity and reactive oxygen species (ROS) production by BMECs were measured after increasing doses of baicalin were added to the culture media in the absence or presence of H2O2, which was used as an in vitro model of oxidative stress.
    Results: Low doses of baicalin (1-10 µg/mL) had no or only slightly positive effects on the proliferation and viability of BMECs, whereas higher doses (100 or 200 µg/mL) markedly decreased BMEC proliferation. Baicalin decreased apoptosis rate at low concentrations (10 µg/mL) but increased apoptosis at higher doses. ROS production was decreased in BMECs treated with increasing doses of baicalin compared with untreated cells, and this decreased production was associated with increased intracellular concentrations of catalase and NRF-2. Irrespective of the dose, baicalin pretreatment attenuated H2O2-induced ROS production.
    Discussion: These results indicate that baicalin exerts protective antioxidant effects on bovine mammary cells. This finding suggests that baicalin could be used to prevent oxidative metabolic disorders in dairy cows.
    Keywords:  Baicalin; Cell culture; Dairy cow; Mammary epithelial cell; Oxidative stress
    DOI:  https://doi.org/10.7717/peerj.6565
  13. Int J Mol Sci. 2019 Mar 07. pii: E1158. [Epub ahead of print]20(5):
      Cutaneous T-cell lymphoma (CTCL) may develop a highly malignant phenotype in its late phase, and patients may profit from innovative therapies. The plant extract indirubin and its chemical derivatives represent new and promising antitumor strategies. This first report on the effects of an indirubin derivative in CTCL cells shows a strong decrease of cell proliferation and cell viability as well as an induction of apoptosis, suggesting indirubin derivatives for therapy of CTCL. As concerning the mode of activity, the indirubin derivative DKP-071 activated the extrinsic apoptosis cascade via caspase-8 and caspase-3 through downregulation of the caspase antagonistic proteins c-FLIP and XIAP. Importantly, a strong increase of reactive oxygen species (ROS) was observed as an immediate early effect in response to DKP-071 treatment. The use of antioxidative pre-treatment proved the decisive role of ROS, which turned out upstream of all other proapoptotic effects monitored. Thus, reactive oxygen species appear as a highly active proapoptotic pathway in CTCL, which may be promising for therapeutic intervention. This pathway can be efficiently activated by an indirubin derivative.
    Keywords:  CTCL; XIAP; apoptosis; c-FLIP; cell viability
    DOI:  https://doi.org/10.3390/ijms20051158
  14. Genetics. 2019 Mar 13. pii: genetics.302051.2019. [Epub ahead of print]
      Centrosomes play a critical role in mitotic spindle assembly through their role in microtubule nucleation and bipolar spindle assembly. Loss of centrosomes can impair the ability of some cells to properly conduct mitotic division, leading to chromosomal instability, cell stress, and aneuploidy. Multiple aspects of the cellular response to mitotic error associated with centrosome loss appears to involve activation of JNK signaling. To further characterize the transcriptional effects of centrosome loss, we compared gene expression profiles of wildtype and acentrosomal cells from Drosophila wing imaginal discs. We found elevation of expression of JNK target genes, which we verified at the protein level. Consistent with this, the upregulated gene set showed significant enrichment for the AP1 consensus DNA binding sequence. We also found significant elevation in expression of genes regulating redox balance. Based on those findings, we examined oxidative stress after centrosome loss, revealing that acentrosomal wing cells have significant increases in reactive oxygen species (ROS). We then performed a candidate genetic screen and found that one of the genes upregulated in acentrosomal cells, G6PD, plays an important role in buffering acentrosomal cells against increased ROS and helps protect those cells from cell death. Our data and other recent studies have revealed a complex network of signaling pathways, transcriptional programs, and cellular processes that epithelial cells use to respond to stressors like mitotic errors to help limit cell damage and maintain normal tissue development.
    Keywords:  Centrosome; JNK signaling; ROS; apoptosis; oxidative stress
    DOI:  https://doi.org/10.1534/genetics.119.302051
  15. Anticancer Drugs. 2019 Apr;30(4): 339-346
      Docetaxel (DTX) is a chemotherapeutic agent, and it is used for the treatment of several cancers including prostate and glioblastoma, but it results in many adverse effects in normal tissues, including kidney. The cytoprotective properties of selenium (Se) against adverse effects of DTX were reported in several normal cells, except kidney cell lines. The purpose of this study was to investigate the effects of Se on DTX-induced nephrotoxicity in normal kidney cell lines. The human embryonic kidney 293 (HEK293) cells were divided into four groups as control, Se (200 nmol/l for 10 h), DTX (10 nmol/l for 48 h), and DTX+Se. Laser confocal microscope fluorescence intensity of apoptosis (annexin V and propidium iodide), mitochondrial membrane depolarization, reactive oxygen species production, and lipid peroxidation levels were increased in the cells by the DTX treatments, although cell number, cell viability, reduced glutathione and glutathione peroxidase values were decreased by the treatments. The fluorescence intensities and values were recovered in the DTX+Se group of the cells by Se treatment. In conclusion, DTX-induced adverse effects were recovered through inhibition of apoptosis and mitochondrial oxidative stress through upregulation of reduced glutathione and glutathione peroxidase in the normal kidney (HEK293) cells. Combination therapy of DTX and Se could be used as an effective strategy for protection of kidney cells against adverse effects of DTX.
    DOI:  https://doi.org/10.1097/CAD.0000000000000723
  16. Front Pharmacol. 2019 ;10 163
      Iron overload has been reported to contribute to bone marrow mesenchymal stem cells (BMSCs) damage, but the precise mechanism still remains elusive. Icariin, a major bioactive monomer belonging to flavonoid glucosides isolated from Herba Epimedii, has been shown to protect cells from oxidative stress induced apoptosis. The aim of this study was to investigate whether icariin protected against iron overload induced dysfunction of BMSCs and its underlying mechanism. In this study, we found that iron overload induced by 100 μM ferric ammonium citrate (FAC) caused apoptosis of BMSCs, promoted cleaved caspase-3 and BAX protein expressions while inhibited Bcl-2 protein expression, which effects were significantly attenuated by icariin treatment. In addition, iron overload induced significant depolarization of mitochondrial membrane potential (MMP), reactive oxygen species (ROS) generation and inhibition of mitochondrial fusion/fission, which effects were also attenuated by icariin treatment. Meanwhile, we found that iron overload induced by 100 μM FAC significantly inhibited mitochondrial fission protein FIS1 and fusion protein MFN2 expressions, inhibited DRP1 and Cytochrome C protein translocation from the cytoplasm to mitochondria. Icariin at concentration of 1 μM was able to promote mitochondrial fission protein FIS1 and fusion protein MFN2 expressions, and increase DRP1 and cytochrome C protein translocation from the cytoplasm to mitochondria. Further, osteogenic differentiation and proliferation of BMSCs was significantly inhibited by iron overload, but icariin treatment rescued both osteogenic differentiation and proliferation of BMSCs. Further studies showed that icariin attenuated iron overload induced inactivation of the PI3K/AKT/mTOR pathway and activation of the ERK1/2 and JNK pathways. In summary, our study indicated that icariin was able to protect against iron overload induced dysfunction of BMSCs. These effects were potentially related to the modulation of mitochondrial fusion and fission, activation of the PI3K/AKT/mTOR pathway and inhibition of ERK1/2 and JNK pathways.
    Keywords:  MAPK pathway; PI3K/AKT/mTOR pathway; bone marrow mesenchymal stem cells; icariin; iron overload; mitochondrial fusion and fission
    DOI:  https://doi.org/10.3389/fphar.2019.00163
  17. Med Sci Monit. 2019 Mar 16. 25 1976-1983
      BACKGROUND Nasopharyngeal carcinoma results in high patient morbidity and mortality, due to early metastasis, and toxicity due to chemotherapy. Mukonal is plant-derived carbazole alkaloid that has been used in traditional Chinese medicine to treat several types of cancer. This study aimed to investigate the effects of mukonal on cell proliferation, apoptosis, autophagy, and the mitochondrial membrane potential of nasopharyngeal carcinoma cells in vitro. MATERIAL AND METHODS CNE1 human nasopharyngeal carcinoma cells and NP69 normal nasopharyngeal epithelial cells were cultured with and without treatment with increasing doses of mukonal. Cell viability was determined by the MTT assay. Fluorescence microscopy was used to detect reactive oxygen species (ROS), mitochondrial membrane potential, and the release of cytochrome C. Flow cytometry was used to examine changes in the cell cycle, electron microscopy examined cell autophagy, and Western blot was performed to measure levels of proteins associated with autophagy and apoptosis. RESULTS Mukonal had an antiproliferative effect on CNE1 cells, with an IC₅₀ of 9 µM and there were effects of toxicity on normal NP69 cells. Mukonal triggered ROS-mediated changes in mitochondrial membrane potential which was also accompanied by the discharge of cytochrome C in the CNE1 cells. Mukonal activated autophagy and apoptosis in CNE1 cells, which was also associated with upregulation of the autophagy-related proteins, LC3 II and beclin-1, as well as apoptosis-associated proteins, Bax, cleaved caspase-3 and -9. Mukonal treatment also resulted in CNE1 cells cycle arrest at G₂/M. CONCLUSIONS Mukonal inhibited the growth of human CNE1 nasopharyngeal carcinoma cells in vitro.
    DOI:  https://doi.org/10.12659/MSM.913915
  18. Eur J Pharmacol. 2019 Mar 07. pii: S0014-2999(19)30156-6. [Epub ahead of print]
      Protocatechuic acid (PCA) has been well studied for its neuroprotection value in several diseases, but the effect in intracerebral hemorrhage (ICH) has not been reported. Here we verified the protection of PCA in ICH, and investigated the relative mechanisms. ICH model mice were established by injection of collagenase IV. The mice were treated with PCA once per day for 3 days, starting immediately after operation. The modified neurological severity score (mNSS) of mice at 1st, 3rd and 7th day after operation were recorded. And some of mice were euthanized at 3rd day to compare brain water content, pro-inflammatory cytokines expression, and cell apoptosis in perihematomal tissue. Additionally, SH-SY5Y cells were treated hemin to mimic secondary injury of ICH. Cells were incubated with PCA for treatment. The cell viability, ROS, apoptosis rate and protein expression of apoptosis-relative protein and MAPKs and NF-κB were detected and analyzed. The results revealed PCA alleviated the cerebral edema at 3rd post ICH, and significantly improved neurological functions. PCA also attenuated the protein and gene expression of TNF-а, IL-1β and IL-6 vivo. PCA dose-dependently decreased the generation of ROS and apoptosis rate. Furthermore, PCA treatment dose-dependently decreased the expression of bax, cleaved caspase-3, increased bcl-2 expression; PCA downregulated P38/JNK-NF-κB pathway. In conclusion, PCA effectively improves prognosis of ICH mice by inhibiting oxidative stress, inflammation and apoptosis. The mechanism possibly results of downregulating of P38/JNK-NF-κB pathway, and PCA can be a potential therapeutic agent for ICH.
    Keywords:  ICH; JNK; NF-κB; P38; Protocatechuic acid
    DOI:  https://doi.org/10.1016/j.ejphar.2019.03.008
  19. Zhonghua Kou Qiang Yi Xue Za Zhi. 2019 Mar 09. 54(3): 176-182
      Objective: To investigate the in vitro and in vivo effects of 5-aminolevulinic acid (5-ALA)-mediated photodynamic therapy against oral squamous cell carcinoma (OSCC) and preliminarily explore the possible mechanisms. Methods: SCC25 cells were divided into the control group (5-ALA of 0 mg/L) and the experimental group (5-ALA of 10, 25, 50, 100 and 150 mg/L). The production of protoporphyrin Ⅸ (PpⅨ) induced by 5-ALA in SCC25 cells was detected using the flow cytometry. SCC25 cells were divided into the control group (5-ALA of 0 mg/L), lazer alone group, 5-ALA alone group (5-ALA of 100 mg/L) and the 5-ALA combined with laser irradiation group (5-ALA of 5, 10, 25, 50 and 100 mg/L), the cytotoxicity of 5-ALA combined with laser irradiation (wave length 635 nm, power density 87 mW/cm(2) and laser dose 10.4 J/cm(2)) was evaluated in SCC25 cells using the methyl thiazolyltetrazolium assay (incubation times of 4, 8 and 12 h in each group) and the induction effect of combination treatment on the cell apoptosis was assessed by the flow cytometry (incubation time of 12 h in each group). The intracellular production of reactive oxygen species (ROS) triggered by 5-ALA combined with laser irradiation was determined using a fluorescence probe method (incubation time of 12 h in each group). A mouse OSCC xenograft model bearing SCC25 tumor was built, and the mice were divided into control group (saline), 5-ALA group (5-ALA of 50 mg/kg) and 5-ALA combined with laser irradiation group (5-ALA of 10, 25 and 50 mg/kg). Antitumor effect of 5-ALA combined with laser irradiation (wave length 635 nm, power density 158 mW/cm(2) and laser dose 94.8 J/cm(2)) was further measured. Results: 5-ALA induced the production of PpⅨ in SCC25 cells in a drug concentration (0-150 mg/L)-and incubation time (0-24 h)-dependent manner. When the 5-ALA concentration was 100 mg/L, the intracellular PpⅨ production was in a relatively stable state. Cell viability and apoptosis rate of 5, 10, 25, 50, 100 mg/L 5-ALA combined with laser irradiation are, respectively, (82.3±5.2)%, (3.13±0.38)%; (74.6±9.3)%, (5.38±0.55)%; (38.3±9.7)%, (17.97±2.72)%; (9.2±3.8)%, (24.47±3.37)%; (7.2±0.8)%, (43.01±5.96)%, which indicated that 5-ALA combined with laser irradiation notably inhibited the growth of SCC25 cells and also induced significant cell apoptosis compared with the control group [(96.3±6.0)%, (0.35±0.13)%, P<0.05]. After combination treatment (5-ALA of 5, 10, 25, 50 and 100 mg/L combined with laser irradiation, the mean fluorescence intensity of dichlorofluorescein is (1.46±0.12)×10(4), (2.16±0.30)×10(4), (3.57±0.34)×10(4), (81.70±13.05)×10(4), (113.00±7.35)×10(4), respectively, a large amount of ROS was produced in SCC25 cells compared with the control group [(0.96±0.15) ×10(4), P<0.05], which was in positive correlation with the intracellular PpⅨ content. 5-ALA (concentration of 10, 25 and 50 mg/kg) combined with laser irradiation greatly suppressed the tumor growth in SCC25 tumor-bearing mice compared to the control group (P<0.05). Conclusions: 5-ALA-mediated photodynamic therapy can trigger the generation of intracellular ROS that has significant cytotoxicity and apoptosis induction effect, and thus inhibit the tumor growth both in vitro and in vivo.
    Keywords:  5-Aminolevulinic acid; Apoptosis; Carcinoma, squamous cell; Photochemotherapy
    DOI:  https://doi.org/10.3760/cma.j.issn.1002-0098.2019.03.006
  20. Exp Ther Med. 2019 Mar;17(3): 2143-2151
      Oxidative stress and mitochondrial dysfunction are considered to be activators of apoptosis and serve a pivotal role in the pathogenesis of myocardial ischemia-reperfusion (MI/R) injury. Apurinic/apyrimidinic endonuclease/redox factor 1 (APE1) is a multifunctional protein that processes the cellular response to DNA damage and oxidative stress. Little is known about the role of APE1 in the pathogenesis of MI/R injury. The aim of the present study was to investigate the effects of APE1 on hypoxia-reoxygenation (H/R)-induced H9c2 cardiomyocyte injury and the underlying mechanism responsible. It was demonstrated that H/R decreased cell viability and increased lactic dehydrogenase (LDH) release, as well as reducing APE1 expression in H9c2 cells. However, APE1 overexpression induced by transfection with APE1-expressing lentivirus significantly increased H9c2 cell viability, decreased LDH release, decreased apoptosis and reduced caspase-3 activity in H/R-treated H9c2 cells. APE1 overexpression ameliorated the H/R-induced increases in reactive oxygen species and NAPDH oxidase expression, as well as the decreases in superoxide dismutase activity and glutathione expression. Furthermore, APE1 overexpression increased mitochondrial membrane potential and ATP production, stabilized electron transport chain activity (as illustrated by increased NADH-ubiquinone oxidoreductase, succinate dehydrogenase, coenzyme Q-cytochrome c oxidoreductase and cytochrome c oxidase activities) and decreased the ratio of B-cell lymphoma 2-associated X protein/B-cell lymphoma 2 in H/R, improving mitochondrial dysfunction. In conclusion, the results of the present study suggest that APE1 alleviates H/R-induced injury in H9c2 cells by attenuating oxidative stress and ameliorating mitochondrial dysfunction. APE1 may therefore be used as an effective treatment for MI/R injury.
    Keywords:  apoptosis; apurinic/apyrimidinic endonuclease/redox factor 1; mitochondrial dysfunction; myocardial hypoxia-reoxygenation injury; oxidative stress
    DOI:  https://doi.org/10.3892/etm.2019.7212
  21. J Ethnopharmacol. 2019 Mar 11. pii: S0378-8741(18)33130-1. [Epub ahead of print]
      ETHNOPHARMACOLOGICAL RELEVANCE: Centella asiatica (CA) is a medicinal herb traditionally used as a brain tonic in Ayurvedic medicine. Various ethnomedical leads revealed the effective use of CA in the treatment of symptoms associated to oxidative stress and inflammation.AIM OF THE STUDY: The aim of this study was to evaluate the therapeutic ability of CA methanol extract (CAM) in protecting mouse brain and astrocytes from oxidative stress and inflammation induced by Paracetamol, and thus to substantiate the allied traditional/ethnomedical claims of CA.
    MATERIALS AND METHODS: Chemical profiling of CAM and quantification of its major constituents were carried out by HPTLC-densitometry. Mice were administered with CAM and Paracetamol in various combinations, and oxidative stress parameters (lipid peroxidation, radical scavenging) as well as nitric oxide stress were estimated from isolated mouse brain. Cellular toxicity was investigated by apoptosis/necrosis in primary astrocytes isolated from brain tissues of mouse (which was challenged by CAM/Paracetamol) by flow cytometry and fluorescent microscopy. Expression of inflammatory cytokine mediators (monocyte chemo attractant protein 1, interleukin 1, interferon γ, tumor necrosis factor β, interleukin 10 and mitogen activated protein kinase 14 gene) in CAM/Paracetamol administered mouse brain tissues were analyzed by real time PCR. Mouse brain tissues challenged by CAM/Paracetamol were also assessed for gross and histopathology. In addition, staining with acridine orange was carried out in C6 cell lines treated with CAM, and viewed under fluorescent microscopy.
    RESULTS: Paracetamol elicited reactive oxygen species generation was revealed through Ferric Reducing Antioxidant Power (FRAP) activity. CAM reversed the Paracetamol induced free radical and reactive nitrogen species production and increased the scavenging activity which was more pronounced at the higher dose (80 mg/kg b.wt). CAM negated the Paracetamol-induced damage by inhibiting expression of pro-inflammatory cytokines (MCP 1, IL 1, TNF β), and increasing the expression of the anti-inflammatory cytokine (IL 10) profoundly. Interestingly, MAPK 14 gene expression was decreased gradually and became same as normal control with increase in the dose of CAM. Also, it was evident that CAM protected isolated mouse primary astrocytes from Paracetamol by maintaining a normal morphology. Similarly, apoptosis of primary astrocytes decreased with the increase in CAM dose (80 mg/kg b.wt.) which was evident from flow cytometric data. Severe brain damage in the form of lesions was apparent from brain histology in Paracetamol alone treated mouse brain. Whereas, CAM treated together with Paracetamol upturned these lesions. Surprisingly, CAM alone proved to be cytotoxic to C6 Glioma cells.
    CONCLUSIONS: CAM showed antioxidant and anti-inflammatory effects (which were pronounced at higher doses) against Paracetamol-induced oxidative stress associated inflammation in mouse brain. The underlying mechanisms may be mediated by inhibiting the pro-inflammatory cytokines TNF β, IL 1 and MCP 1 via regulation of the antioxidant mediated INF γ and MAPK 14 gene signalling pathways. The major bioactive constituents in CAM are the triterpenoid saponins, asiaticoside and madecassoside. The present results provide pharmacological evidence that CAM acts as an antioxidant and anti-inflammatory agent. Furthermore, this study validates the use of CA as an antioxidant and anti-inflammatory agent in ethnomedicine.
    Keywords:  Anti-inflammatory; Antioxidant; Apoptosis; Astrocytes; Brain; Centella asiatica; Ethnomedicine
    DOI:  https://doi.org/10.1016/j.jep.2019.03.017
  22. Free Radic Biol Med. 2019 Mar 09. pii: S0891-5849(18)31587-9. [Epub ahead of print]
      Glutathione oxido-reductase (GR) is a primary antioxidant enzyme of most living forms which protects the cells from oxidative metabolism by reducing glutathione (GSH) from its oxidized form (GSSG). Although the antioxidant role of the enzyme is well characterized, the specific role of conserved N' peptide sequence in antioxidant mechanism remains unclear. In this study, we have identified an RNA sequence encoding GR enzyme from spirulina Arthrospira platensis (Ap) and the changes in its gene expression profile was analysed during H2O2 stress. Results showed that H2O2 (10 mM) stimulated the expression of ApGR throughout the timeline of study (0, 5, 10, 15, 20 days) with highest expression at 5th day post exposure which confirmed the antioxidant role of ApGR in spirulina during H2O2 induced oxidative stress. A dithiol containing short antioxidant peptide, 39GGTCVIRGCVPKKLM53 (GM15) from ApGR was predicted and its radicals (superoxide and hydroxyl radical) scavenging potential was confirmed by in vitro cell-free assays. GM15 (12.5 μM) reduced the intracellular generalized oxidative stress level, as measured using DCFDA assay in H2O2 exposed leucocytes without affecting any of the cellular population. Further, the biomedical application of the radical scavenging property of GM15 was validated in oral carcinoma (KB) cells where GM15 exhibited significant cytotoxicity. Also, GM15 exhibited heterogenous effects on intracellular oxidative stress level in KB cells: at lower concentration (6.25 μM), the peptide reduced oxidative stress whereas, at higher concentration (25 μM) it increased the intensity of oxidative stress. GM15 (25 μM) induced caspase-9 mediated apoptosis in KB cells along with membrane disruption and DNA degradation which are confirmed by propidium iodide (PI) internalization and comet assays, respectively. Overall, the study shows that GM15 peptide i) scavenges superoxide, hydroxyl radicals, and influences intracellular oxidative stress, and ii) has anti-cancer effect in oral cancer cells.
    Keywords:  Antioxidant; Apoptosis; Dithiol peptide; Glutathione oxido-reductase; Oral carcinoma cells
    DOI:  https://doi.org/10.1016/j.freeradbiomed.2019.03.006
  23. Environ Toxicol Pharmacol. 2019 Mar 07. pii: S1382-6689(18)30398-3. [Epub ahead of print]68 52-60
      β-zearalenol (β-zol) and HT-2 are mycotoxins which cause apoptosis and oxidative stress in mammalian reproductive cells. Melatonin is an endogenous antioxidant involved in apoptosis and oxidative stress-related activities. This study investigated the effects of β-zol and HT-2 on bovine ovarian granulosa cells (BGCs), and how melatonin may counteract these effects. β-zol and HT-2 inhibited cell proliferation in a dose-dependent manner, and induced apoptosis of BGCs. They also yielded upregulation of the apoptosis-related genes Bax/Bcl-2 and Caspase3 and phosphorylation of p38MAPK. Increases in intracellular ROS were observed along with higher levels of mRNA anti-oxidation markers SOD1, SOD2, and CAT. SOD1, SOD2, malonaldehyde (MDA), and glutathione peroxidase (GSH-px) activities increased, as did the levels of SOD1 and SOD2 proteins. All of these effects were reduced or entirely attenuated in BGCs pre-treated with melatonin. Our results demonstrate that melatonin has protective effects against mycotoxin-induced apoptosis and oxidative stress in BGCs.
    Keywords:  Alleviation effect; Apoptosis; HT-2 toxin; Melatonin; Oxidative stress; β-zearalenol
    DOI:  https://doi.org/10.1016/j.etap.2019.03.005
  24. Oxid Med Cell Longev. 2019 ;2019 5152072
      At present, apatinib is considered a new generation agent for the treatment of patients with gastric cancer. However, the effects of apatinib on pancreatic cancer have not been clarified. This study investigated the impact of apatinib on the biological function of pancreatic cancer cells and the potential mechanism involved in this process. Using the Cell Counting Kit-8 method, we confirmed that apatinib treatment inhibited cell proliferation in vitro. Moreover, the migration rate of pancreatic cells was inhibited. The effects of apatinib on apoptosis and cell cycle distribution of pancreatic carcinoma cells were detected by flow cytometry. The number of apoptotic cells was significantly increased, and the cell cycle was altered. Furthermore, we demonstrated that apatinib inhibited the expression of hypoxia-inducible factor-1α (HIF-1α), vascular endothelial growth factor, and markers of the phosphoinositide 3-kinase (PI3K)/Akt/mTOR signaling pathway, which increased the levels of reactive oxygen species in vitro. Apatinib significantly inhibited the biological function of pancreatic cancer cells. It promoted apoptosis, downregulated the expression of HIF-1α, and increased the levels of reactive oxygen species.
    DOI:  https://doi.org/10.1155/2019/5152072
  25. Genes Genomics. 2019 Mar 15.
      BACKGROUND: Ischemia heart disease is one of the major causes of death worldwide which often associated with tissue infarction and limit the recovery of function. Multiple factors involved in the I/R-induced cardiomyocyte dysfunction which were consistent with a role of oxidative stress and altered endothelium-dependent responses. However, the pathogenic mechanisms in I/R injury remain unclear.MATERIALS AND METHODS: The H9C2 cells were in the ischaemia/reperfusion (I/R) condition. After I/R, the cells were transfected with or without adenovirus-urothelial carcinoma associated 1(Ad-UCA1). Then qRT-PCR analysis was performed to quantify mRNA expression of different treatment groups. Cell apoptosis rate was assessed using flow cytometry and ER stress biomarker expression were measured by immunoblotting. Intracellular and mitochondrial ROS generation were assayed by fluorescence microscope after staining with the DCFDA or MitoSOX.
    RESULTS: I/R conditions trigger lncRNAs UCA1 expression, cellular and mitochondria ROS production, resulting in cell apoptosis through the induction of oxidative and ER stress. Overexpression of UCA1 protects H9C2 cells from I/R-induced ER stress and cell apoptosis. Moreover, UCA1 might be a potential regulator in the protective effect of I/R‑induced oxidative stress and mitochondria dysfunction. Subsequently, ER stress inhibitor attenuated the effect of siUCA1 induced injury in H9C2 cells.
    CONCLUSION: The expression of UCA1 against I/R induced oxidative stress and mitochondria dysfunction via suppression of endoplasmic reticulum stress. UCA1 might be a biomarker to improved diagnosis of I/R injury.
    Keywords:  ER stress; Ischaemia/reperfusion; LncRNAs UCA1; Mitochondria dysfunction; Oxidative stress
    DOI:  https://doi.org/10.1007/s13258-019-00806-w
  26. Cells. 2019 Mar 08. pii: E226. [Epub ahead of print]8(3):
      Iron overload is the accumulation of excess iron in the body that may occur as a result of various genetic disorders or as a consequence of repeated blood transfusions. The surplus iron is then stored in the liver, pancreas, heart and other organs, which may lead to chronic liver disease or cirrhosis, diabetes and heart disease, respectively. In addition, excessive iron may impair hematopoiesis, although the mechanisms of this deleterious effect is not entirely known. In this study, we found that ferrous ammonium sulfate (FeAS), induced growth arrest and apoptosis in immature hematopoietic cells, which was mediated via reactive oxygen species (ROS) activation of p38MAPK and JNK pathways. In in vitro hematopoiesis derived from embryonic stem cells (ES cells), FeAS enhanced the development of dysplastic erythroblasts but inhibited their terminal differentiation; in contrast, it had little effect on the development of granulocytes, megakaryocytes, and B lymphocytes. In addition to its directs effects on hematopoietic cells, iron overload altered the expression of several adhesion molecules on stromal cells and impaired the cytokine production profile of these cells. Therefore, excessive iron would affect whole hematopoiesis by inflicting vicious effects on both immature hematopoietic cells and stromal cells.
    Keywords:  hematopoiesis; hematopoietic stem cells; iron overload; oxidative stress; stromal cells
    DOI:  https://doi.org/10.3390/cells8030226
  27. Sci Rep. 2019 Mar 11. 9(1): 4100
      Activation of TRPM8 channel through oxidative stress may induce Ca2+ and pro-apoptotic signals in prostate cancer and kidney cells. The aim of this study was to evaluate activation of TRPM8 can increase apoptosis and oxidative stress in the prostate cancer (Du145M8), TRPM8 knock out (Du 145M8KO), transfected (HEK293TM8) and non-transfected human kidney (HEK293) cells. Intracellular Ca2+ responses to TRPM8 activation were increased in the Du145M8 and HEK293TM8 cells from coming cumene hydrogen peroxide (CHPx), menthol, ADP-Ribose (ADPR), but not in the HEK293 and Du 145M8KO cells. The intracellular Ca2+ responses to both ADPR and CHPx were totally inhibited by the thiol cycle antioxidant glutathione, and TRPM8 blockers (N-(p-amylcinnamoyl)anthranilic acid and capsazepine). Apoptosis, Annexin V, mitochondrial membrane depolarization, intracellular ROS, caspase 3 and 9 values were increased through TRPM8 activation in the Du 145M8 but not in the Du 145M8KO and non-transfected HEK293 cells by CHPx and hydrogen peroxide. In conclusion, apoptotic and oxidant effects on the cells were increased activation of TRPM8 by oxidative stress and ADPR. Activation of TRPM8 through oxidative stress and ADPR in the cells could be used as an effective strategy in the treatment of prostate cancer cells.
    DOI:  https://doi.org/10.1038/s41598-018-37552-0
  28. Eur J Pharmacol. 2019 Mar 12. pii: S0014-2999(19)30167-0. [Epub ahead of print]
      Oxidative stress plays an important role in auditory dysfunction. Exogenous cell therapy has brought new hopes for repairing mammalian inner ear hair cells. However, poor cell viability of transplanted cells under oxidative stress conditions has limited their therapeutic potential. The adipocytokine apelin-13 was isolated from a bovine stomach. Apelin-13 might protect oxidative stress-induced hair cell damage was raised considering other oxidative stress-induced injury, including brain ischemia-induced cell death. Therefore, we evaluated the protective effects of apelin- 13 on the damage induced by hydrogen peroxide (H2O2) to the hair cells-derived from bone marrow mesenchymal stem cells (BMSCs) in vitro. Stem cells were differentiated into hair cell- like cells with B27, FGF, EGF and IGF-1. Expression of neuron specific markers including β tubulin III, Nestin, MAP2, Neurofilament 68 and GFAP was tested by flow cytometry. As well, inner ear hair cell markers such as Myosin VIIA, Sox2 and TrkB expression were assayed by immunocytochemistry (ICC) method. We designed an in vitro model of oxidative stress by exposing hair cell- like cells to H2O2. Protein expression levels of caspase-3, Bax and Bcl-2 were detected by western blot. Apoptotic cells were also detected by acridin-orange staining and TUNEL assay. Protein expression of caspase-3 and Bax/Bcl-2 ratio was significantly lower in the apelin-13-pretreated group than only H2O2 treated group. In addition, apoptotic cells were significantly decreased in the apelin-13+H2O2 co-treated cells compared to the H2O2-treated group. Treating hair cells-like cells with apelin13 increases their survival against oxidative stress damage by inhibition of apoptosis signaling pathway.
    Keywords:  Apelin-13; Apoptosis; Bcl-2/Bax ratio; Caspase-3; Hair Cell; Oxidative Stress
    DOI:  https://doi.org/10.1016/j.ejphar.2019.03.012
  29. Am J Pathol. 2019 Mar 09. pii: S0002-9440(18)30754-5. [Epub ahead of print]
      Mitochondrial ribosome proteins (MRPs), which are encoded by the nuclear genomic DNA, are important for mitochondrial-encoded protein synthesis and mitochondrial function. Emerging evidence suggests that several MRPs also exhibit important extra-mitochondrial functions, such as involvement in apoptosis, protein biosynthesis, and signal transduction. In this study, we demonstrate a significant role of mitochondrial ribosomal protein L35 in colorectal cancer (CRC). The expression of MRPL35 was higher in CRC tissues than in matched cancer-adjacent tissues and higher in CRC cells than in normal mucosal epithelial cells. Higher MRPL35 expression in CRC tissue correlated with shorter overall survival for CRC patients. In vitro, down-regulation of MRPL35 led to increased production of reactive oxygen species (ROS) together with DNA damage, loss of cell proliferation, G2/M arrest, a decrease in mitochondrial membrane potential, apoptosis, and autophagy induction. MRPL35 knockdown inhibited tumor proliferation in a CRC xenograft nude mouse model. Furthermore, overexpression of MRPL35 or treatment of cells with the ROS scavenger, N-acetyl cysteine (NAC), abrogated ROS production, cell cycle arrest, and apoptosis in vitro. These findings suggest that MRPL35 plays an essential role in the development of CRC and may be a potential therapeutic target for CRC.
    DOI:  https://doi.org/10.1016/j.ajpath.2019.02.003
  30. Am J Physiol Endocrinol Metab. 2019 Mar 12.
      Women with polycystic ovary syndrome (PCOS) are at increased risk of miscarriage, which often accompanies the hyperandrogenism and insulin resistance seen in these patients. However, neither the combinatorial interaction between these two PCOS-related etiological factors nor the mechanisms of their actions in the uterus during pregnancy are well understood. We hypothesised that hyperandrogensim and insulin resistance exert a causative role in miscarriage by inducing defects in uterine function that are accompanied by mitochondrial-mediated oxidative stress, inflammation and perturbed gene expression. Here we tested this hypothesis by studying the metabolic, endocrine and uterine abnormalities in pregnant rats after exposure to daily injection of 5α-dihydrotestosterone (DHT, 1.66 mg/kg body weight/day) and/or insulin (6.0 IU/day) from gestational day 7.5 to 13.5. We showed that while DHT-exposed and insulin-exposed pregnant rats presented impaired insulin sensitivity, DHT+insulin-exposed pregnant rats exhibited hyperandrogenism and peripheral insulin resistance, which mirrors pregnant PCOS patients. Compared to controls, hyperandrogenism and insulin resistance in the dam was associated with alterations in uterine morphology and aberrant expression of genes responsible for decidualization, placentation, angiogenesis and insulin signaling. Moreover, we observed changes in uterine mitochondrial function and homeostasis and suppression of both oxidative and antioxidative defenses in response to the hyperandrogenism and insulin resistance. These findings demonstrate that hyperandrogenism and insulin resistance induce mitochondria-mediated damage and a resulting imbalance between oxidative and antioxidative stress responses in the gravid uterus.
    Keywords:  angiogenesis; decidualization; mitochondrial homeostasis; polycystic ovary syndrome; reactive oxygen species
    DOI:  https://doi.org/10.1152/ajpendo.00359.2018
  31. Life Sci. 2019 Mar 08. pii: S0024-3205(19)30173-0. [Epub ahead of print]
      AIMS: Gluaredoxin1 (GRX1) is an important protein of the cellular antioxidant defense system, but its role in renal epithelial cell injury caused by ischemia remains unclear. In this study, we aimed to gain insight into the role of GRX1 in HK-2 cells with oxygen glucose deprivation (OGD) injury, which served as an in vitro cell model of renal epithelial cell ischemic injury. We investigated the underlying regulation of GRX1, DJ-1, and HSP70 as well as the role of the GRX1/DJ-1/HSP70 signaling pathway in this model.MATERIALS AND METHODS: The protein and mRNA expressions were measured by Western blot and qRT-PCR assays, respectively. GRX1 was overexpressed by transfection of pcDNA.3.1-GRX1 and DJ-1 was inhibited by transfection with DJ-1 siRNA. Cell apoptosis, caspase-3 activity, lactate dehydrogenase (LDH) leakage, or superoxide dismutase (SOD) content was tested by the related detection kit. Reactive oxygen species (ROS) level was detected via carboxy-H2DCF-DA.
    KEY FINDINGS: We found that GRX1 was distinctly down-regulated in HK-2 cells after incubation under the OGD condition. GRX1 overexpression markedly constrained cell apoptosis, caspase-3 activity, LDH leakage, and the ROS level, while SOD content was elevated. GRX1 up-regulation increased DJ-1 and HSP70 protein expression, while DJ-1 inhibition significantly offset the effect of GRX1 overexpression on HSP70, indicating that GRX1 could regulate HSP70 via control of DJ-1. Moreover, we observed that HSP70 inhibition removed the constraints imposed by GRX1 overexpression on ROS level, LDH leakage, and caspase-3 activity.
    SIGNIFICANCE: Overall, this study showed that GRX1 minimizes cell injury and apoptosis in HK-2 cells under OGD conditions via regulation of DJ-1 and HSP70 expression.
    Keywords:  DJ-1; GRX1; HK-2; HSP70; OGD
    DOI:  https://doi.org/10.1016/j.lfs.2019.03.015
  32. Environ Sci Pollut Res Int. 2019 Mar 09.
      The present study aimed to determine the level of iron and calcium in the seminal plasma of men with different fertility potentials and to examine its relationship with oxidative stress. Seventy-nine sub-fertile patients with asthenoteratozoospermia (AT), n 27; teratoleucozoospermia (TL), n 20; teratozoospermia (Terato), n 32; and 29 healthy donors were included. The ability of spermatozoa to produce reactive oxygen species (ROS) was evaluated by using nitroblue tetrazolium (NBT) staining. The lipid peroxidation end product, malondialdehyde (MDA), and the trace element levels (iron and calcium) were measured spectrophotometrically. Iron and calcium concentrations in seminal plasma of the patient groups were significantly more elevated than the normal group. Nevertheless, both calcium and iron showed strong negative correlations with the total sperm motility and normal sperm morphology, but only iron was positively and significantly associated with multiple anomalies index and seminal leucocyte concentration. On the other hand, the rates of MDA and ROS production in semen were significantly higher in the three abnormal groups than in controls. These two oxidative stress biomarkers were significantly associated with the percentage of atypical forms in semen. However, only semen ROS level was significantly associated with the decreased sperm motility and the sperm leucocytes concentration. Meanwhile, there are positive correlations between seminal iron and calcium content and the studied oxidative stress biomarkers. Oxidative stress and trace element excess are implicated in low sperm quality. Iron and calcium might be the mediators of the effects of oxidative damage and induces lipid peroxidation.
    Keywords:  Calcium; Iron; Male infertility; Oxidative stress; Sperm
    DOI:  https://doi.org/10.1007/s11356-019-04575-7
  33. Sci Rep. 2019 Mar 14. 9(1): 4496
      Lipid peroxidation by reactive oxygen species (ROS) during oxidative stress is non-enzymatic damage that affects the integrity of biological membrane, and alters the fluidity and permeability. We conducted molecular dynamic simulation studies to evaluate the structural properties of the bilayer after lipid peroxidation and to measure the permeability of distinct ROS. The oxidized membrane contains free fatty acid, ceramide, cholesterol, and 5α-hydroperoxycholesterol (5α-CH). The result of unconstrained molecular dynamic simulations revealed that lipid peroxidation causes area-per-lipid of the bilayer to increase and bilayer thickness to decrease. The simulations also revealed that the oxidized group of 5α-CH (-OOH) moves towards the aqueous layer and its backbone tilts causing lateral expansion of the bilayer membrane. These changes are detrimental to structural and functional properties of the membrane. The measured free energy profile for different ROS (H2O2, HO2, HO, and O2) across the peroxidized lipid bilayer showed that the increase in lipid peroxidation resulted in breaching barrier decrease for all species, allowing easy traversal of the membrane. Thus, lipid peroxidation perturbs the membrane barrier and imposes oxidative stress resulting into apoptosis. The collective insights increase the understanding of oxidation stress at the atomic level.
    DOI:  https://doi.org/10.1038/s41598-019-40913-y
  34. Theranostics. 2019 ;9(5): 1264-1279
      Photosensitizer (PS) serves as the central element of photodynamic therapy (PDT). The use of common nanoparticles (NPs) for PDT has typically been rendered less effective by the undesirable aggregation-caused quenching (ACQ) effect, resulting in quenched fluorescence and reduced reactive oxygen species (ROS) generation that diminish the imaging quality and PDT efficacy. To overcome the ACQ effect and to enhance the overall efficacy of PDT, herein, integrin ανβ3-targeted organic nanodots for image-guided PDT were designed and synthesized based on a red emissive aggregation-induced emission (AIE) PS. Methods: The TPETS nanodots were prepared by nano-precipitation method and further conjugated with thiolated cRGD (cRGD-SH) through a click reaction to yield the targeted TPETS nanodots (T-TPETS nanodots). Nanodots were characterized for encapsulation efficiency, conjugation rate, particle size, absorption and emission spectra and ROS production. The targeted fluorescence imaging and antitumor efficacy of T-TPETS nanodot were evaluated both in vitro and in vivo. The mechanism of cell apoptosis induced by T-TPETS nanodot mediated-PDT was explored. The biocompatibility and toxicity of the nanodots was examined using cytotoxicity test, hemolysis assay, blood biochemistry test and histological staining. Results: The obtained nanodots show bright red fluorescence and highly effective 1O2 generation in aggregate state. Both in vitro and in vivo experiments demonstrate that the nanodots exhibit excellent tumor-targeted imaging performance, which facilitates image-guided PDT for tumor ablation in a hepatocellular carcinoma model. Detailed analysis reveals that the nanodot-mediated PDT is able to induce time- and concentration-dependent cell death. The use of PDT at a high PDT intensity leads to direct cell necrosis, while cell apoptosis via the mitochondria-mediated pathway is achieved under low PDT intensity. Conclusion: Our results suggest that well-designed AIE nanodots are promising for image-guided PDT applications.
    Keywords:  aggregation-induced emission (AIE); hepatocellular carcinoma; integrin ανβ3; photodynamic therapy (PDT); theranostics
    DOI:  https://doi.org/10.7150/thno.29101
  35. Biomed Pharmacother. 2019 Mar 11. pii: S0753-3322(19)30525-6. [Epub ahead of print]113 108769
      AIMS: In this study, we examined whether β-apopicropodophyllin (APP) could act as a radiosensitizer in non-small cell lung cancer (NSCLC) cells.MAIN METHODS: The in vitro radiosensitizing activity of APP was demonstrated with clonogenic assay, immunoblotting, Annexin V-Propidium iodide (PI) assay, BrdU incorporation, detection of mitochondrial ROS/intracellular of H2O2, mitochondrial membrane potential detection, and performing of isolation of mitochondrial and cytosolic fractions. The in vivo radiosensitizing activity of APP was determined in xenografted mice with co-treatment of APP and IR based on measurement of tumor volumes and apoptotic cell death.
    KEY FINDINGS: The results of a clonogenic assay indicated that a combination of APP and γ-ionizing radiation (IR) inhibits cell growth and increases cell death in NSCLC cells. Several signal transduction pathways were examined for their potential involvement in the apparent radiosensitization effect of APP, as assessed by immunoblotting analyses and mitochondrial potential determination in vitro. Treatment of NCI-H460 cells with 15 nM APP and NCI-H1299 cells with 10 nM APP yielded dose-enhancement ratios of 1.44 and 1.24, respectively. Enhanced ER stress, disrupted mitochondrial membrane potential, and increased reactive oxygen species (ROS) were observed in cells co-treated with APP and IR, and this was followed by the cytosolic release of cytochrome c and consequent activation of caspase-3 and -9. Notably, inhibition of JNK, which prevents caspase activation, blocked the APP/IR-induced activations of ER stress and apoptotic cell death. In NCI-H460 or NCI-H1299 cell-xenografted mice, APP/IR treatment delayed the time it took tumors to reach a threshold size by 22.38 and 16.83 days, respectively, compared with controls, to yield enhancement factors of 1.53 and 1.38, respectively.
    SIGNIFICANCE: APP has a radiosensitizing function derived from its ability to induce apoptotic cell death via activation of ER stress, disruption of mitochondrial membrane potential, and induction of the caspase pathway.
    Keywords:  Apoptosis; ER stress; Non-small cell lung cancer; ROS; Radiosensitizer; β-Apopicropodophylli
    DOI:  https://doi.org/10.1016/j.biopha.2019.108769
  36. J Pineal Res. 2019 Mar 12. e12569
      Development of secondary hair follicles in early post-natal cashmere goats may be adversely affected by reactive oxygen species (ROS) which cause oxidative stress. Because melatonin is a potent antioxidant and scavenger of free radicals, this study explored the effects of melatonin on secondary hair follicle development and subsequent cashmere production. It was found that the initiation of new secondary follicles in early postnatal Inner Mongolian cashmere goats of both melatonin-treated and control goats occurred in the first 10 weeks of age. Melatonin promoted the initiation and maturation of secondary follicles and increased their population. Importantly, the beneficial effect of melatonin on secondary follicle population remained throughout life. As a result, melatonin increased cashmere production and improved its quality in terms of reduced fiber diameter. The mechanisms underlying the role of melatonin on secondary follicle development included the enhancement of activities of antioxidant enzymes, e.g., superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px), elevated total antioxidant capacity (T-AOC), and upregulated anti-apoptotic Bcl-2 expression and downregulated expression of the pro-apoptotic proteins, Bax and caspase-3. These results reveal that melatonin serves to promote secondary hair follicle development in early postnatal cashmere goats and expands our understanding of melatonin application in cashmere production. Melatonin treatment led to an increase in both the quantity and quality of cashmere fiber. This increased the textile value of the fibers and provided economic benefit. This article is protected by copyright. All rights reserved.
    Keywords:  apoptosis; cashmere; melatonin; oxidative stress; secondary hair follicle
    DOI:  https://doi.org/10.1111/jpi.12569
  37. Int J Mol Sci. 2019 Mar 11. pii: E1229. [Epub ahead of print]20(5):
      Photodynamic therapy (PDT) constitutes a cancer treatment modality based on the administration of a photosensitizer, which accumulates in tumor cells. The subsequent irradiation of the tumoral area triggers the formation of reactive oxygen species responsible for cancer cell death. One of the compounds approved in clinical practice is methyl-aminolevulinate (MAL), a protoporphyrin IX (PpIX) precursor intermediate of heme synthesis. We have identified the mitotic catastrophe (MC) process after MAL-PDT in HeLa human carcinoma cells. The fluorescence microscopy revealed that PpIX was located mainly at plasma membrane and lysosomes of HeLa cells, although some fluorescence was also detected at endoplasmic reticulum and Golgi apparatus. Cell blockage at metaphase-anaphase transition was observed 24 h after PDT by phase contrast microscopy and flow cytometry. Mitotic apparatus components evaluation by immunofluorescence and Western blot indicated: multipolar spindles and disorganized chromosomes in the equatorial plate accompanied with dispersion of centromeres and alterations in aurora kinase proteins. The mitotic blockage induced by MAL-PDT resembled that induced by two compounds used in chemotherapy, taxol and nocodazole, both targeting microtubules. The alterations in tumoral cells provided evidence of MC induced by MAL-PDT, resolving mainly by apoptosis, directly or through the formation of multinucleate cells.
    Keywords:  HeLa tumor cells; cell death; mitotic catastrophe; photodynamic therapy; spindle elements
    DOI:  https://doi.org/10.3390/ijms20051229
  38. J Inorg Biochem. 2019 Mar 06. pii: S0162-0134(18)30694-9. [Epub ahead of print]195 1-12
      Some metallodrugs that exhibit interesting biological activity contain transition metals such as ruthenium, and have been extensively exploited because of their antiparasitic potential. In previous study, we reported the remarkable anti-Leishmania activity of precursor cis-[RuIICl2(dppm)2], where dppm = bis(diphenylphosphino)methane, and new ruthenium(II) complexes, cis-[RuII(η2-O2CC10H13)(dppm)2]PF6 (bbato), cis-[RuII(η2-O2CC7H7S)(dppm)2]PF6 (mtbato) and cis-[RuII(η2-O2CC7H7O2)(dppm)2]PF6 (hmxbato) against some Leishmania species. In view of the promising activity of the hmxbato complex against Leishmania (Leishmania) amazonensis promastigotes, the present work investigated the possible parasite death mechanism involved in the action of this hmxbato and its precursor. We report, for the first time, that hmxbato and precursor promoted an increase in reactive oxygen species production, depolarization of the mitochondrial membrane, DNA fragmentation, formation of a pre-apoptotic peak, alterations in parasite morphology and formation of autophagic vacuoles. Taken together, our results suggest that these ruthenium complexes cause parasite death by apoptosis. Thus, this work provides relevant knowledge on the activity of ruthenium(II) complexes against L. (L.) amazonensis. Such information will be essential for the exploitation of these complexes as future candidates for cutaneous leishmaniasis treatment.
    Keywords:  Apoptosis; Cell death; Leishmania (Leishmania) amazonensis; ROS production; Ruthenium(II) complex
    DOI:  https://doi.org/10.1016/j.jinorgbio.2019.03.005
  39. Cell Death Dis. 2019 Mar 15. 10(4): 255
      Hepatocellular carcinoma (HCC), a common liver malignancy worldwide, has high morbidity and mortality. β-Thujaplicin, a tropolone derivative, has been used in some health-care products and clinical adjuvant drugs, but its use for HCC is unknown. In this study, we found that β-Thujaplicin inhibits the growth of HCC cells, but not normal liver cells, with nanomolar potency. Mechanistically, we found that β-Thujaplicin could induce autophagy, as judged by western blot, confocal microscopy, and transmission electron microscopy. Further using β-Thujaplicin combined with an autophagy blocker or agonist treatment HepG2 cells, we found that β-Thujaplicin induced autophagic cell death (ACD) mediated by ROS caused inhibition of the Akt-mTOR signaling pathway. Moreover, β-Thujaplicin triggered HepG2 apoptosis and increased cleaved PARP1, cleaved caspase-3, and Bax/Bcl-2 ratio, which indicated that β-Thujaplicin induced apoptosis mediated by the mitochondrial-dependent pathway. We also found that increased expression of p21 and decreased expression of CDK7, Cyclin D1, and Cyclin A2 participating in β-Thujaplicin caused the S-phase arrest. It seems that β-Thujaplicin exerts these functions by ROS-mediated p38/ERK MAPK but not by JNK signaling pathway activation. Consistent with in vitro findings, our in vivo study verified that β-Thujaplicin treatment significantly reduced HepG2 tumor xenograft growth. Taken together these findings suggest that β-Thujaplicin have an ability of anti-HCC cells and may conducively promote the development of novel anti-cancer agents.
    DOI:  https://doi.org/10.1038/s41419-019-1492-6
  40. Oncogene. 2019 Mar 14.
      Cancer cells frequently exhibit higher levels of reactive oxygen species (ROS) than normal cells and when ROS levels increase beyond a cellular tolerability threshold, cancer cell death is enhanced. The mitochondrial dihydrolipoamide dehydrogenase (DLDH) is an enzyme which produces ROS in association with its oxidoreductive activity and may be thus utilized as an exogenous anticancer agent. As cancer cells often overexpress integrins that recognize RGD-containing proteins, we have bioengineered the human DLDH with RGD motifs (DLDHRGD) for integrin-mediated drug delivery. The modified protein fully retained its enzyme activity and ROS-production capability. DLDHRGD uptake by cells was shown to depend on the presence of cell-associated integrin αvβ3, as comparatively demonstrated with normal kidney cells (HEK293) transfected with either β1 (αvβ1 positive) or β3 integrins (αvβ3 positive). The interaction with β3 integrins was shown to be competitively inhibited by an RGD peptide. In mice melanoma cells (B16F10), which highly express an endogenous αvβ3 integrin, fast cellular uptake of DLDHRGD which resulted in cell number reduction, apoptosis induction, and a parallel intracellular ROS production was shown. Similar results were obtained with additional human melanoma cell models (A375, WM3314, and WM3682). In contrast, HEK293β3 cells remained intact following DLDHRGD uptake. The high pharmacological safety profile of DLDHRGD has been observed by several modes of administrations in BALB/C or C57Bl/6 mouse strains. Treatments with DLDHRGD in a subcutaneous melanoma mice model resulted in significant tumor inhibition. Our study demonstrated, in vitro and in vivo, the development of a unique platform, which targets cancer cells via integrin-mediated drug delivery of an exogenous ROS-generating drug.
    DOI:  https://doi.org/10.1038/s41388-019-0775-9
  41. Front Physiol. 2019 ;10 53
      Water-pipe smoking (WPS) is prevalent in the East and elsewhere. WPS exposure is known to induce thrombosis and cardiovascular toxicity involving inflammation and oxidative stress. Here, we have investigated the effect of Gum Arabic (GA), a prebiotic with anti-oxidant, anti-inflammatory and cytoprotective properties, on WPS exposure (30 min/day for 1 month) on coagulation and cardiac homeostasis, and their possible underlying mechanisms in mice. Animals received either GA in drinking water (15%, w/v) or water only for the entire duration of study. GA significantly mitigated thrombosis in pial microvessels in vivo, platelet aggregation in vitro, and the shortening of prothrombin time induced by WPS exposure. The increase in plasma concentrations of fibrinogen, plasminogen activator inhibitor-1 and markers of lipid peroxidation, 8-isoprostane and malondialdehyde, induced by WPS were significantly reduced by GA administration. Moreover, WPS exposure induced a significant increase in systolic blood pressure and the concentrations of the pro-inflammatory cytokines tumor necrosis factor-α and interleukin 1β in heart homogenates. GA significantly alleviated these effects, and prevented the decrease of reduced glutathione, catalase and total nitric oxide levels in heart homogenates. Immunohistochemical analysis of the hearts showed that WPS exposure increased nuclear factor erythroid-derived 2-like 2 (Nrf2) expressions by cardiac myocytes and endothelial cells, and these effects were potentiated by the combination of GA and WPS. WPS also increased DNA damage and cleaved caspase 3, and GA administration prevented these effects. Our data, obtained in experimental murine model of WPS exposure, show that GA ameliorates WPS-induced coagulation and cardiovascular inflammation, oxidative stress, DNA damage and apoptosis, through a mechanism involving Nrf2 activation.
    Keywords:  Gum Arabic; heart; inflammation; nose-only exposure; oxidative stress; thrombosis; water-pipe smoke
    DOI:  https://doi.org/10.3389/fphys.2019.00053
  42. Cell Signal. 2019 Mar 06. pii: S0898-6568(19)30049-X. [Epub ahead of print]58 53-64
      Arterial media calcification is one of the major complications of diabetes mellitus, which is related to oxidative stress and apoptosis. Mitophagy is a special regulation of mitochondrial homeostasis and takes control of intracellular ROS generation and apoptotic pathways. High circulating levels of lactate usually accompanies diabetes. The potential link between lactate, mitophagy and vascular calcification is investigated in this study. Lactate treatment accelerated VSMC calcification, evaluated by measuring the calcium content, ALP activity, RUNX2, BMP-2 protein levels, and Alizarin red S staining. Lactate exposure caused excessive intracellular ROS generation and VSMC apoptosis. Lactate also impaired mitochondrial function, determined by mPTP opening rate, mitochondrial membrane potential and mitochondrial biogenesis markers. Western blot analysis of LC3-II and p62 and mRFP-GFP-LC3 adenovirus detection for autophagy flux revealed that lactate blocked autophagy flux. LC3-II co-staining with LAMP-1 and autophagosome quantification revealed lactate inhibited autophagy. Furthermore, lactate inhibited mitophagy, which was confirmed by TOMM20 and BNIP3 protein levels, LC3-II colocalization with BNIP3 and TEM assays. In addition, BNIP3-mediated mitophagy played a protective role against VSMC calcification in the presence of lactate. This study suggests that lactate accelerates osteoblastic phenotype transition of VSMC and calcium deposition partly through the BNIP3-mediated mitophagy deficiency induced oxidative stress and apoptosis.
    Keywords:  Autophagy; Lactate; Mitophagy; VSMC; Vascular calcification
    DOI:  https://doi.org/10.1016/j.cellsig.2019.03.006
  43. Daru. 2019 Mar 09.
      BACKGROUND: Chemotherapy, as one of the main approaches of cancer treatment, is accompanied with several adverse effects, including chemotherapy-induced peripheral neuropathy (CIPN). Since current methods to control the condition are not completely effective, new treatment options should be introduced. Medicinal plants can be suitable candidates to be assessed regarding their effects in CIPN. Current paper reviews the available preclinical and clinical studies on the efficacy of herbal medicines in CIPN.METHODS: Electronic databases including PubMed, Scopus, and Cochrane library were searched with the keywords "neuropathy" in the title/abstract and "plant", "extract", or "herb" in the whole text. Data were collected from inception until April 2018.
    RESULTS: Plants such as chamomile (Matricaria chamomilla L.), sage (Salvia officinalis L.), cinnamon (Cinnamomum cassia (L.) D. Don), and sweet flag (Acorus calamus L.) as well as phytochemicals like matrine, curcumin, and thioctic acid have demonstrated beneficial effects in animal models of CIPN via prevention of axonal degeneration, decrease in total calcium level, improvement of endogenous antioxidant defense mechanisms such as superoxide dismutase and reduced glutathione, and regulation of neural cell apoptosis, nuclear factor-ĸB, cyclooxygenase-2, and nitric oxide signaling. Also, five clinical trials have evaluated the effect of herbal products in patients with CIPN.
    CONCLUSIONS: There are currently limited clinical evidence on medicinal plants for CIPN which shows the necessity of future mechanistic studies, as well as well-designed clinical trial for further confirmation of the safety and efficacy of herbal medicines in CIPN. Graphical abstract Schematic mechanisms of medicinal plants to prevent chemotherapy-induced neuropathy: NO: nitric oxide, TNF: tumor necrosis factor, PG: prostaglandin, NF-ĸB: nuclear factor kappa B, LPO: lipid peroxidation, ROS: reactive oxygen species, COX: cyclooxygenase, IL: interleukin, ERK: extracellular signal-related kinase, X: inhibition, ↓: induction.
    Keywords:  Chemotherapeutic agents; Clinical studies; Inflammation; Medicinal plants; Neuropathy; Pain; Phytochemicals; Phytotherapy
    DOI:  https://doi.org/10.1007/s40199-019-00255-6
  44. Biomed Pharmacother. 2019 Mar 11. pii: S0753-3322(18)37913-7. [Epub ahead of print]113 108727
      The chemotherapeutic agent cisplatin typically induces apoptosis by inhibiting the cell cycle. Cancer Stem Cells (CSCs), which are a proliferative quiescent and slowly-cycling cell population, are less sensitive and therefore frequently spared from toxic effects. Thus, it remains a priority to increase the sensitivity of CSCs to cisplatin-based chemotherapy, or to specifically target CSCs to improve the therapeutic outcome in breast cancer. Disulfiram (DSF) is a drug used clinically for alcoholism treatment that has displayed promising anti-cancer activity in vitro and in cancer xenografts in breast cancer. Our study provides evidence that DSF inhibits Aldehyde dehydrogenase (ALDH) enzyme activity, inhibits the expression of stemness-related transcription factors (Sox, Nanog, Oct) in CSC derived from breast cancer cell lines, and modulates intracellular reactive oxygen species (ROS) generation. Importantly, our research proved that ALDH + stem-like cells play important roles in the resistance to the conventional chemotherapeutic agent cisplatin. DSF enhances the cytotoxic effect of cisplatin through inhibiting the stemness and by overcoming cisplatin resistance of ALDH + stem-like cells. A quantitative measurement showed the synergistic effect of DSF and cisplatin. Further, we show that ALDH + cancer stem-like cells and ALDH- bulk cancer cells have different intrinsic ROS levels, what may explain differences in susceptibility to cisplatin treatment. Importantly, this difference is eliminated by DSF treatment making both cell types similarly susceptible for cytotoxic effects by cisplatin. These findings may influence chemotherapeutic treatment approaches in the future.
    Keywords:  ALDH enzyme activity; Cancer stem cells; Chemotherapy resistance; Novel adjuvant treatment; ROS
    DOI:  https://doi.org/10.1016/j.biopha.2019.108727
  45. Evid Based Complement Alternat Med. 2019 ;2019 7219740
      Inflammation had showed its important role in the pathogenesis of cerebral ischemia and secondary damage. Safflower yellow B (SYB) had neuroprotective effects against oxidative stress-induced brain injuries, but the mechanisms were still largely unknown to us. In this study, we tried to investigate the anti-inflammation effects of SYB and the possible roles of AMPK/NF-κB signaling pathway on these protective effects. In vivo, brain ischemia/reperfusion (I/R) was induced by transient middle cerebral artery occlusion for 2 h and reperfusion for 20 h. Neurofunctional evaluation, infarction area, and brain water contents were measured. Brain injury markers and inflammatory cytokines levels were measured by ELISA kits. In vitro, cell viability, apoptosis, and LDH leakage were measured after I/R in PC12 cells. The expression and phosphorylation levels of AMPK, NF-κB p65, and P-IκB-α in cytoplasm and nuclear were measured by Western blotting. SiRNA experiment was performed to certify the role of AMPK. The results showed SYB reduced infarct size, improved neurological outcomes, and inhibited brain injury after I/R. In vitro test, SYB treatment alleviated PC12 cells injury and apoptosis and inhibited the inflammatory cytokines (IL-1, IL-6, TNF-α, and COX-2) in a dose-dependent manner. SYB treatment induced AMPK phosphorylation and inhibited NF-κB p65 nuclear translocation both in brain and in PC12 cells. Further studies also showed that the inhibition of NF-κB activity of SYB was through AMPK. In conclusion, SYB protected brain I/R injury through reducing expression of inflammatory cytokines and this effect might be partly due to the inhibition of NF-κB mediated by AMPK.
    DOI:  https://doi.org/10.1155/2019/7219740
  46. Artif Cells Nanomed Biotechnol. 2019 Dec;47(1): 891-895
      Astaxanthin, a Xanthophyll carotenoid, has strong antioxidant properties. Some studies have shown the effectiveness of this compound on the prevention and treatment of cancer. Therefore, the aim of this study was to evaluate the effects of astaxanthin on induction of apoptosis and antioxidant activity in the LS-180 cell line. In this experimental study, after the treatment of LS-180 50, 100 and 150 μm of Astaxanthin for 24 h, the expression levels of Bax, Bcl2 and Caspase3 genes were investigated by Real-time PCR. Also, the level of malondialdehyde, as an indicator of oxidative stress and activity of anti-superoxide dismutase enzymes, catalase and glutathione peroxidase was investigated by colorimetric methods. The results showed that astaxanthin increases the expression of Bax and Caspase3 genes and decreases that of Bcl2, thereby, inducing apoptosis and inhibiting growth and proliferation of the cells. Additionally, reduction in the levels of malondialdehyde was evident with a significant elevation in antioxidant activity mediated by the action of superoxide dismutase, catalase and glutathione peroxidase. These results suggest that astaxanthin has the potency to induce apoptosis in LS-180 cells by increasing the expression of apoptotic genes and activity of antioxidant enzymes. Thus, astaxanthin has potential in the prevention and treatment of cancer.
    Keywords:  Astaxanthin; LS-180; antioxidant enzymes; apoptosis
    DOI:  https://doi.org/10.1080/21691401.2019.1580286
  47. Evid Based Complement Alternat Med. 2019 ;2019 2702068
      Background/Aims: Naomaitai can improve blood perfusion and ameliorate the damage in the paraventricular white matter. This study was focused on observing the neuroprotective effect of Naomaitai on the vascular dementia of rat and exploring the action mechanism of PI3K/PDK1/AKT signaling pathway.Methods: A vascular dementia model of rats was established by permanent, bilateral common carotid artery occlusion. Rats' behavior was tested by Neurological deficit score and the Morris water maze. The pathology and apoptosis were detected through HE staining and TUNEL assay. Myelin sheath loss and nerve fiber damage were detected by LFB staining. Inflammatory factors, oxidative stress, and brain damage markers were detected through ELISA. The expression of apoptosis-related proteins and PI3K/PDK1/AKT signaling pathway related proteins were measured by western blot. The expressions of PI3K, PDK1, AKT, and MBP in paraventricular white matter cells were detected by immunofluorescence.
    Results: Naomaitai treatment decreased neurological function score in rats with vascular dementia, ameliorated paraventricular white matter damage caused by long-term hypoxia, and hypoperfusion reduced the brain injury markers S-100β and NSE contents, suppressed inflammatory reaction and oxidative stress, reduced IL-1β, IL-6, TNF-α, and MDA contents, and remarkably increased IL-10 and SOD contents. TUNEL and western blot assay showed that Naomaitai treatment decreased neuronal cell apoptosis, increased Bcl-2 expression, and reduced caspase-3 and Bax expression. Furthermore, we found Naomaitai inhibited PI3K and PDK1 expression and activated phosphorylated AKT protein in rats with vascular dementia. However, the protective effect of Naomatai in rats with vascular dementia was inhibited, and expression of PI3K signaling pathway-related proteins was blocked after administration of PI3K inhibitor.
    Conclusion: Naomaitai can ameliorate brain damage in rats with vascular dementia, inhibit neuronal apoptosis, and have anti-inflammatory and antioxidative stress effects, which may be regulated by the PI3K/PDK1/AKT signaling pathway.
    DOI:  https://doi.org/10.1155/2019/2702068
  48. Onco Targets Ther. 2019 ;12 1185-1193
      Background: Dicerandrol B is a natural antitumor agent that can be isolated from the endophytic fungus, Phomopsis sp. The present study investigated the effects of dicerandrol B on human cervical cancer HeLa cells.Materials and methods: In this study, dicerandrol B was identified by electrospray ionization mass spectrometry and nuclear magnetic resonance spectroscopy. We used MTT to detect the cell viability. Flow cytometry was used to analyze the apoptosis and cell cycle. Western blot was used to examine the expression of related proteins.
    Results: Dicerandrol B was isolated from the endophytic fungus Phomopsis sp. The MTT assay and flow cytometry showed that dicerandrol B significantly inhibited HeLa cell viability and induced G2/M cell cycle arrest. Western blot analysis demonstrated that dicerandrol B increased the levels of GRP78, ubiquitin, cleaved PARP, and Bax protein, decreased the levels of PARP and Bcl-2 protein, and caused an increase in the Bax/Bcl-2 ratio in HeLa cells. Dicerandrol B increased the production of ROS in HeLa cells, which was attenuated by the antioxidant N-acetyl-l-cysteine.
    Conclusion: These findings suggest that dicerandrol B induces apoptosis in human HeLa cells, possibly through the endoplasmic reticulum stress and mitochondrial apoptotic pathways. This suggests that dicerandrol B possesses strong anticancer activity in cervical cancer and provides insight into the underlying mechanisms.
    Keywords:  apoptosis; cervical cancer; endoplasmic reticulum stress; mitochondrial damage
    DOI:  https://doi.org/10.2147/OTT.S191204
  49. Autophagy. 2019 Mar 12.
      Macroautophagy/autophagy is a degradative process essential for various cellular processes. We previously demonstrated that autophagy-deficiency causes myoblast apoptosis and impairs myotube formation. In this study, we continued this work with particular emphasis on mitochondrial remodelling and stress/apoptotic signaling. We found increased (p<0.05) autophagic (e.g., altered LC3B levels, increased ATG7, decreased SQSTM1) and mitophagic (e.g., BNIP3 upregulation, mitochondrial localized GFP-LC3 puncta, and elevated mitochondrial LC3B-II) signaling during myoblast differentiation. shRNA-mediated knockdown of ATG7 (shAtg7) decreased these autophagic and mitophagic responses, while increasing CASP3 activity and ANXA5/annexin V staining in differentiating myoblasts; ultimately resulting in dramatically impaired myogenesis. Further confirming the importance of mitophagy in these responses, CRISPR-Cas9-mediated knockout of Bnip3 (bnip3-/-) resulted in increased CASP3 activity and DNA fragmentation as well as impaired myoblast differentiation. In addition, shAtg7 myoblasts displayed greater endoplasmic reticulum (e.g., increased CAPN activity and HSPA) and mitochondrial (e.g., mPTP formation, reduced mitochondrial membrane potential, elevated mitochondrial 4-HNE) stress. shAtg7 and bnip3-/- myoblasts also displayed altered mitochondria-associated signaling (e.g., PPARGC1A, DNM1L, OPA1) and protein content (e.g., SLC25A4, VDAC1, CYCS). Moreover, shAtg7 myoblasts displayed CYCS and AIFM1 release from mitochondria, and CASP9 activation. Similarly, bnip3-/- myoblasts had significantly higher CASP9 activation during differentiation. Importantly, administration of a chemical inhibitor of CASP9 (Ac-LEHD-CHO) or dominant-negative CASP9 (ad-DNCASP9) partially recovered differentiation and myogenesis in shAtg7 myoblasts. Together, these data demonstrate an essential role for autophagy in protecting myoblasts from mitochondrial oxidative stress and apoptotic signaling during differentiation, as well as in the regulation of mitochondrial network remodelling and myogenesis.
    Keywords:  apoptosis; autophagy; caspase 9; differentiation; mitochondria; mitophagy; myogenesis; oxidative stress; skeletal muscle
    DOI:  https://doi.org/10.1080/15548627.2019.1591672
  50. Hum Exp Toxicol. 2019 Mar 15. 960327119831065
      High glucose (HG) induces vascular injury in diabetes. Hydroxysafflor yellow A (HSYA) has been used to ameliorate ischemic cardiovascular diseases in China for many years. In the present study, we assessed whether HSYA has a potential protective role in HG-induced human umbilical vein endothelial cell (HUVEC) injury. Cell viability was determined with an 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium (MTS) assay. Cell apoptosis was detected by fluorescein isothiocyanate/propidium iodide staining assay. The endothelial cell permeability was measured with a permeability assay. Cell adhesion molecule (CAM) expression, vascular endothelial growth factor, and basic fibroblast growth factor levels were detected with an enzyme-linked immunosorbent assay. Reactive oxygen species (ROS) formation was measured with a DCF-DA assay. Protein expression of NADPH oxidase 4 (NOX4) was measured by Western blotting. Our data indicated that HG increases HUVEC apoptosis, vascular permeability, monocyte adhesion, the level of CAMs, the formation of ROS, and NOX4 expression. Our data revealed that HG increases vascular injury, which is attenuated by HSYA. Because vascular inflammation has a key role in the development of diabetes mellitus, our results implied that HSYA is considered as a potential agent for diabetic vascular injury treatment.
    Keywords:  Hydroxysafflor yellow A; NOX4; diabetes mellitus; high glucose; human umbilical vein endothelial cells
    DOI:  https://doi.org/10.1177/0960327119831065
  51. Toxins (Basel). 2019 Mar 10. pii: E157. [Epub ahead of print]11(3):
      Aflatoxin B₁ (AFB₁) is a serious threat to the poultry industry. Proanthocyanidins (PCs) demonstrates a broad range of biological, pharmacological, therapeutic, and chemoprotective properties. The aim of this study was to investigate the ameliorative effects of PCs against AFB₁-induced histopathology, oxidative stress, and apoptosis via the mitochondrial pathway in the bursa of Fabricius (BF) of broilers. One hundred forty-four one-day old Cobb chicks were randomly assigned into four treatment groups of six replicates (6 birds each replicate) for 28 days. Groups were fed on the following four diets; (1) Basal diet without addition of PCs or AFB₁ (Control); (2) basal diet supplemented with 1 mg/kg AFB₁ from contaminated corn (AFB₁); (3) basal diet supplemented with 250 mg/kg PCs (PCs); and (4) basal diet supplemented with 1 mg/kg AFB₁ + 250 mg/kg PCs (AFB₁+ PCs). The present study results showed that antioxidant enzymes activities of total superoxide dismutase (T-SOD), catalase (CAT), glutathione peroxidase (GSH-Px), and glutathione S-transferase (GST) in AFB₁ treated group were (p < 0.05) decreased, whereas malondialdehyde (MDA) contents were significantly increased in comparison with the control group. Furthermore, we found that dietary PCs treatment ameliorated AFB₁-induced oxidative stress in the BF through inhibiting the accumulation of MDA content and enhancing the antioxidant enzymes activities (T-SOD, CAT, GSH-Px, and GST). Similarly, PCs markedly enhanced messenger RNA (mRNA) expression of antioxidant genes (SOD, CAT, GPx1, and GST) in comparison with AFB₁ group. Moreover, histological results showed that PCs alleviated AFB₁-induced apoptotic cells in the BF of broilers. In addition, both mRNA and protein expression results manifested that mitochondrial-apoptosis-associated genes (Bax, caspase-9, caspase-3, and p53 and cytochrome c) showed up-regulation, while (Bcl-2) showed down-regulation in AFB₁ fed group. The supplementation of PCs to AFB₁ diet significantly reversed the mRNA and protein expression of these apoptosis-associated genes, as compared to the AFB₁ group. Our results demonstrated that PCs ameliorated AFB₁-induced oxidative stress by modulating the antioxidant defense system and apoptosis in the BF through mitochondrial pathway in broilers.
    Keywords:  aflatoxin B1; apoptosis; broiler; bursa of Fabricius; oxidative stress; proanthocyanidins
    DOI:  https://doi.org/10.3390/toxins11030157
  52. Nutrition. 2018 Dec 03. pii: S0899-9007(18)31143-2. [Epub ahead of print]62 74-84
      OBJECTIVES: A combined exercise training and calorie-restriction program is the mainstream treatment of obesity. However, the effect of the dual-action program on mitochondrial function in skeletal muscles has not yet been clarified. The aim of this study was to determine if the combined program, rather than a single program, restored both lost muscle activity and mitochondrial function in obesity.METHODS: The study included 30 female Wistar rats. Six rats fed a normal diet for 27 wk were used as the control group. The remaining 24 rats were fed a high-fat diet (HFD) for 27 wk. At week 20, the HFD rats were divided into the following four groups: sedentary lifestyle, endurance exercise five times per week, 60% of calorie restriction (CR) per day, and combined exercise training and CR. All conditions were maintained for 7 wk.
    RESULTS: We found that HFD-fed rats without therapy developed obese insulin resistance (IR) and impaired function of skeletal muscles. Skeletal muscles of the HFD-fed rats without therapy also exhibited early fatigability; impaired mitochondrial function, as indicated by increased reactive oxygen species production, membrane depolarization, and swelling; reduced mitochondrial dynamics as indicated by increased phosphorylation of DRP1 and decreased MFN2 expression; diminished mitochondrial biogenesis, as shown by decreased PGC1α and CPT1 expression; and increased apoptosis. Both exercise and CR in HFD-fed rats equally attenuated the impairment of muscle functions. However, combined therapies in HFD-fed rats restored functions of skeletal muscles.
    CONCLUSIONS: These findings reinforce the synergistic beneficial effects of combined exercise and CR on skeletal muscles of HFD-fed rats.
    Keywords:  Calorie restriction; Exercise; Insulin resistance; Mitochondria; Muscle; Obesity
    DOI:  https://doi.org/10.1016/j.nut.2018.11.031
  53. Pestic Biochem Physiol. 2019 Mar;pii: S0048-3575(18)30374-2. [Epub ahead of print]155 26-35
      Harmine, a useful botanical compound, has demonstrated insecticidal activity against some pests. However, harmine's mechanism of action has not been thoroughly elucidated to date. To preliminarily explore harmine's insecticidal mechanisms, the cytotoxicity of harmine against Spodoptera frugiperda Sf9 cells was comprehensively investigated. Our results indicated that harmine induced apoptosis in Sf9 cells, as evidenced by cellular and nuclear morphological changes, DNA laddering and increases in caspase-3-like activities. In addition, activation of the mitochondrial apoptotic pathway by harmine was confirmed by the generation of ROS, opening of mitochondrial permeability transition pores (MPTPs), increase in cytosolic Ca2+, changes in mRNA expression levels of genes involved in the mitochondrial apoptotic pathway and increase and release of Cytochrome c. Furthermore, lysosomal membrane permeabilization, release of cathepsin L from the lysosome into the cytosol and cleavage of caspase-3 were also triggered, which indicated that lysosomes were involved in this physiological process. Moreover, the effect of harmine on DNA topoisomerase I activity was investigated by in vivo and molecular docking experiments. These data not only verified that harmine induced apoptosis via comprehensive activation of the mitochondrial and lysosomal pathways and inhibition of DNA topoisomerase I activity in Sf9 cells but also revealed a mechanism of harmine insecticidal functions for pest control.
    Keywords:  Apoptosis; Harmine; Lysosomes; Mitochondria; Topoisomerase I
    DOI:  https://doi.org/10.1016/j.pestbp.2019.01.002
  54. J Clin Med. 2019 Mar 08. pii: E328. [Epub ahead of print]8(3):
      Osteochondritis dissecans (OCD) in equids, especially in sport horses, has become a growing issue as it contributes to the occurrence of lameness. Thus the aim of this study was to investigate the cytophysiological properties of OCD chondrocytes including expression of chondrogenic genes, apoptosis, mitochondria dynamics and autophagy. Horse chondrocytes were isolated from healthy (HE) and OCD cartilages. Properties of cells were evaluated using multiple assays e.g., polymerase chain reaction (PCR), immunofluorescence, Western blot. OCD chondrocytes were characterized by increased apoptosis and senescence. Expression of chondrogenic genes (vimentin, aggrecan) was decreased while mRNA levels of matrix metalloproteinase 13 significantly upregulated in comparison to HE cells. Moreover, OCD cells displayed increased mitochondrial fusion while fission events were diminished. Interestingly, chaperone mediated autophagy was triggered in those cells and it predominated over macroautophagy. Furthermore, co-culture of LPS-treated chondrocytes with adipose-derived stem cells (ASC) decreased p62/sequestosome 1 (SQSTM) and increases MMP-13 expression in OCD cells. Our results suggest that OCD affected horse chondrocytes are characterized by senescent phenotype due to endoplasmic reticulum stress and mitochondria dynamics deterioration. Expression of chondrogenic markers is decreased in those cells while expression of chaperone mediated autophagy (CMA)-related genes increased. Increased malfunctioning of cells leads to loss of their functionality and capacity to maintain tissue homeostasis.
    Keywords:  apoptosis; cartilage; chondrocytes; horse; osteochondritis dissecans
    DOI:  https://doi.org/10.3390/jcm8030328
  55. Prostaglandins Other Lipid Mediat. 2019 Mar 08. pii: S1098-8823(18)30138-2. [Epub ahead of print]
      The combined incidence of melanoma and non-melanoma skin cancer (NMSC) is greater than the incidence of all other malignancies in the US. Previously, we demonstrated that the endocannabinoid, arachidonoyl-ethanolamide (AEA), was a potent inducer of apoptosis in NMSC. The metabolism of AEA to the prostaglandin, PGD2-EA, was a prerequisite for AEA cytotoxicity. However, the mechanism of PGD2-EA cell death has not been clearly defined. In the present study, we report that PGD2-EA causes apoptosis in melanoma and NMSC cells. Mass spectrometry analysis revealed that PGD2-EA was dehydrated to three J-series prostaglandins; PGJ2-EA, Δ12PGJ2-EA, and 15deoxy,Δ12,14 PGJ2-EA. PGD2-EA inhibited the antioxidant activity of glutathione and thioredoxin which then caused oxidative stress. This increase in oxidative stress was accompanied by the activation of endoplasmic reticulum (ER) stress and apoptosis. The effect of PGD2-EA was independent of DP1, DP2, and PPARγ receptors suggesting that PGD2-EA cytotoxicity was mediated by its metabolic product, 15dPGJ2-EA.
    Keywords:  PGD(2); PGD(2)-EA; glutathione; oxidative stress; skin cancer; thioredoxin
    DOI:  https://doi.org/10.1016/j.prostaglandins.2019.03.001
  56. Drug Des Devel Ther. 2019 ;13 647-655
      Objective: The aim of this study was to investigate the protective effect and mechanism of Ginkgo biloba extract-761 (EGb 761) in the rat with myocardial ischemia-reperfusion injury (MIRI).Materials and methods: Forty Sprague Dawley rats were randomly divided into following four groups: sham group, I/R group and EGb 761 groups (20 and 40 mg/kg). MIRI model was established after 14 days of administration. The myocardial infarct size and myocardial histology were measured and compared. Meanwhile, the levels of creatine kinase-MB (CK-MB), lactate dehydrogenase (LDH), troponin T (TnT), TNF-α, IL-6, IL-1β, superoxide dismutase (SOD), malondialdehyde (MDA) and glutathione peroxidase (GSH-Px) were evaluated. Western blot was used to detect the expression of Caspase-3, Bax, Bcl-2, HO-1, Nrf2, Akt, p-Akt and nuclear protein Nrf2.
    Results: The levels of infarct size, CK-MB, LDH, TnT, TNF-α, IL-6 and IL-1β in the EGb 761 groups were significantly lower than those in the ischemia/reperfusion (I/R) group. The content of MDA was lower in the myocardium, whereas the activities of SOD and GSH-Px were higher than those in the I/R group. The expressions of Caspase-3 and Bax in the EGb 761 groups were significantly lower than those in the I/R group, whereas the expressions of Bcl-2, p-Akt and HO-1 and nuclear protein Nrf2 in the EGb 761 groups were higher than those in the I/R group.
    Conclusion: EGb 761 might inhibit the apoptosis of myocardial cells and protect the myocardium by activating the Akt/Nrf2 pathway, increasing the expression of HO-1, decreasing oxidative stress and repressing inflammatory reaction.
    Keywords:  Ginkgo biloba extract; myocardial ischemia-reperfusion injury; oxidative stress; superoxide dismutase
    DOI:  https://doi.org/10.2147/DDDT.S191537
  57. Theranostics. 2019 ;9(4): 1047-1065
      Background: Platinum (II) (Pt(II))-based anticancer drugs dominate the chemotherapy field of ovarian cancer. However, the patient's quality of life has severely limited owing to dose-limiting toxicities and the advanced disease at the time of diagnosis. Multifunctional tumor-targeted nanosized ultrasound contrast agents (glutathione (GSH)-sensitive platinum (IV) (Pt(IV)) prodrug-loaded phase-transitional nanoparticles, Pt(IV) NP-cRGD) were developed for precise theranostics against ovarian cancer. Methods: Pt(IV) NP-cRGD were composed of a perfluorohexane (PFH) liquid core, a hybrid lipid-polymer shell with PLGA12k-PEG2k and DSPE-PEG1k-Pt(IV), and an active targeting ligand, the cRGD peptide (PLGA: poly(lactic-co-glycolic acid), PEG: polyethylene glycol, DSPE: 1,2-distearoyl-sn-glycero-3-phosphoethanolamine, cRGD: cyclic Arg-Gly-Asp). Pt(IV), a popular alternative to Pt(II), was covalently attached to DSPE-PEG1k to form the prodrug, which fine-tuned lipophilicity and improved cellular uptake. The potential of Pt(IV) NP-cRGD as contrast agents for ultrasound (US) imaging was assessed in vitro and in vivo. Moreover, studies on the antitumor efficiency and antitumor mechanism of Pt(IV) NP-cRGD assisted by US were carried out. Results: Pt(IV) NP-cRGD exhibited strong echogenic signals and excellent echo persistence under an US field. In addition, the GSH-sensitive and US-triggered drug delivery system maximized the therapeutic effect while reducing the toxicity of chemotherapy. The mechanistic studies confirmed that Pt(IV) NP-cRGD with US consumed GSH and enhanced reactive oxy gen species (ROS) levels, which further causes mitochondria-mediated apoptosis. Conclusion: A multifunctional nanoplatform based on phase-transitional Pt(IV) NP-cRGD with US exhibited excellent echogenic signals, brilliant therapeutic efficacy and limited side effect, suggesting precise theranostics against ovarian cancer.
    Keywords:  mitochondrial apoptosis; ovarian cancer.; phase transition; platinum (IV); ultrasound contrast agents
    DOI:  https://doi.org/10.7150/thno.29820
  58. Clin Exp Pharmacol Physiol. 2019 Mar 10.
      Noise induced structural and functional disorder of liver has been realized, but the underlying mechanism remains to be characterized, which has limited the introduction of precautious measures. Over-activation of acid sphingomyelinase (ASM)/ceramide (Cer) pathway takes center stage in hepatocyte injury entailed by various stimulus. We aimed to investigate whether it mediated the noise elicited liver disorder on infrastructure, lipid metabolism, apoptosis, and oxidative stress. Mice were exposed to broad band noise (20-20k Hz, 90-110 dB) for 1, 3, 5 or 7 d by 3 hr/d. Doxepin hydrochloride (DOX), an ASM inhibitor was given by 5 mg/kg/d gavage. We showed that 5 or 7 d intense, broad band noise exposure caused significant infrastructure derangement and lipid droplets storage in hepatocytes. The content of cholesterol, free fatty acids or triglyceride was increased significantly in liver tissue upon noise stimulation. Moreover, the noise promoted apoptosis and superoxide generation in hepatocytes significantly, enhancing activity of aspartate aminotransferase (AST) or alanine amino transferase (ALT) in serum. ASM activity and Cer generation in liver tissue were elevated by noise exposure, which was normalized with DOX administrated. Accordingly, DOX alleviated steatosis, apoptosis, oxidative stress and enzymatic change in hepatocytes or serum of noise exposed mice substantially. In summary, our results suggest the ASM/Cer pathway contributes to the broad band noise elicited liver damage in mice. This article is protected by copyright. All rights reserved.
    Keywords:  acid sphingomyelinase; ceramide; doxepin hydrochloride; liver; noise; noise induced liver damage
    DOI:  https://doi.org/10.1111/1440-1681.13083
  59. Theriogenology. 2019 Feb 26. pii: S0093-691X(18)30957-9. [Epub ahead of print]129 146-153
      The success of in vitro embryo production demonstrates that the oviduct can be bypassed during early embryonic development. Using an ex vivo model of porcine uterus is one of the strategies used to investigate fertilization within the oviductal environment. In this study, in vitro-matured porcine oocytes (MII) were fertilized with 7.5 × 107, 15 × 107, or 30 × 107 sperm cells for 20 min in the oviduct of a porcine uterine ex vivo model. MII oocytes used for in vitro fertilization (IVF) served as control 1; those cultured in the oviduct of the ex vivo model for 20 min before IVF served as control 2. In present study, the penetration rate, polyspermy, and fertilization efficiency, and accumulated reactive oxygen species (ROS) levels in the treatment groups were significantly decreased compared to those in the control 1 group. During embryonic development, the cleavage rates in the treatment groups were significantly lower than those in the control groups. The cleavage rate in the 30 × 107 sperm cell-treated group was higher than that in the 7.5 × 107 sperm cell-treated group. The blastocyst formation rate in control 1 and 2, and 30 × 107 sperm cell-treated groups increased compared to that in the 7.5 and 15 × 107 sperm cell-treated groups. PCNA, HSP70.2, and GLUT1 were upregulated in the treatment groups and POU5F1, BAX, GPX1 were upregulated in the treatment and control 2 groups, compared to the control 1 group. These results suggest that an ex vivo model may decrease the penetration rate and fertilization efficiency by increasing the accumulated ROS levels and inducing the expression of apoptosis- and stress-related genes. However, the model improved the monospermy rate and expression of embryo developmental competence genes. This is the first study that evaluates the effect of an ex vivo model of porcine uterus on fertilization parameters, and the development of porcine embryos.
    Keywords:  Fertilization; Monospermy; Porcine; Uterine ex vivo model
    DOI:  https://doi.org/10.1016/j.theriogenology.2019.02.025
  60. Toxins (Basel). 2019 Mar 09. pii: E154. [Epub ahead of print]11(3):
      In this work of quercetin's anti-proliferation action on A. flavus, we revealed that quercetin can effectively hamper the proliferation of A. flavus in dose-effect and time-effect relationships. We tested whether quercetin induced apoptosis in A. flavus via various detection methods, such as phosphatidylserine externalization and Hoechst 33342 staining. The results showed that quercetin had no effect on phosphatidylserine externalization and cell nucleus in A. flavus. Simultaneously, quercetin reduced the levels of reactive oxygen species (ROS). For a better understanding of the molecular mechanism of the A. flavus response to quercetin, the RNA-Seq was used to explore the transcriptomic profiles of A. flavus. According to transcriptome sequencing data, quercetin inhibits the proliferation and aflatoxin biosynthesis by regulating the expression of development-related genes and aflatoxin production-related genes. These results will provide some theoretical basis for quercetin as an anti-mildew agent resource.
    Keywords:  Aspergillus flavus; RNA-seq; aflatoxin; quercetin; transcriptome
    DOI:  https://doi.org/10.3390/toxins11030154
  61. Drug Chem Toxicol. 2019 Mar 12. 1-7
      Silymarin (SLY), a flavonoid complex isolated from the seeds of Silybum marianum (Asteraceae), has antioxidant, anti-apoptotic, anti-inflammatory, and anti-lipid peroxidative effects. Vancomycin (VA), used for treating serious infections, has been associated with nephrotoxicity, which limits its use. Therefore, this study aimed to investigate the potential renoprotective effects of SLY on VA-induced nephrotoxicity using renal, apoptotic (caspase-3, caspase-8, and caspase-9 enzyme activities), and oxidative stress [nitric oxide (NO) and malondialdehyde (MDA)] markers; serum blood urea nitrogen (BUN) and creatinine levels; and histopathological examination. A total of 49 male Wistar albino rats were used (n = 7): control [saline, intraperitoneally (i.p.)], dimethyl sulfoxide (i.p.), VA [400 mg/(kg-day), i.p.], SLY100 [100 mg/(kg-day), i.p.], VA + SLY50 [50 mg/(kg-day), i.p.], VA + SLY100 [100 mg/(kg-day), i.p.], and VA + SLY200 [200 mg/(kg-day), i.p.]. SLY was administered once daily for 8 days. One day after the first treatment of SLY, VA administration was started and continued for 7 days. The levels of serum creatinine and BUN were evaluated using ELISA, caspase enzyme activities and levels of MDA and NO in the kidney tissues were evaluated by the colorimetric methods. The serum BUN, creatinine, NO, MDA levels, and caspase activities were significantly higher in VA group than in control (p < 0.05). However, caspase activities were significantly lower in VA + SLY200 than in VA (p < 0.05). The MDA, serum BUN, and creatinine levels were significantly lower in VA + SLY (50, 100, and 200) groups than in VA group (p < 0.05). VA + SLY200 was found to be the most effective group based on the caspase activities; MDA, NO, serum BUN, creatinine levels; and histopathological findings.
    Keywords:  Apoptosis; nephrotoxicity; oxidative stress; silymarin; vancomycin
    DOI:  https://doi.org/10.1080/01480545.2019.1584208
  62. Metallomics. 2019 Mar 14.
      Apoptosis is a common paradigm of cell death and plays a key role in cartilage damage and selenium (Se) deficiency. Selenoproteins play major roles in determining the biological effects of Se, and are potentially involved in the pathophysiological processes in bone tissue. MicroRNAs (miRNAs) play important roles in cell proliferation, differentiation, apoptosis and tumorigenesis. Based on the preliminary results, the expression of selenoprotein M (SelM) was significantly decreased (69%) in chicken cartilage tissues with Se deficiency, and we subsequently screened and verified that SelM is one of the target genes of miR-138-5p in chicken cartilage using a dual luciferase reporter assay and real-time quantitative PCR (qRT-PCR). The expression of miR-138-5p was increased in response to Se deficiency, and the overexpression of miR-138-5p increased caspase-3, caspase-9, BAX and BAK levels, while the BCL-2 level was decreased, suggesting that miR-138-5p induced apoptosis via the mitochondrial pathway in vivo and in vitro. We explored whether oxidative stress, mitochondrial fission and fusion, and energy metabolism might trigger apoptosis to obtain an understanding of the mechanisms underlying the effects of miR-138-5p on Se deficiency-induced apoptosis in cartilage. The levels of indicators of oxidative stress, mitochondrial dynamics and energy metabolism were changed as well. This study confirmed that SelM is one of the target genes of miR-138-5p, and the overexpression of miR-138-5p induced by Se deficiency triggered oxidative stress, an imbalance in mitochondrial fission and fusion, and energy metabolism dysfunction. Therefore, miR-138-5p is involved in the mitochondrial apoptosis pathway via targeting SelM in chicken chondrocytes.
    DOI:  https://doi.org/10.1039/c9mt00006b
  63. Drug Dev Res. 2019 Mar 12.
      This study was designed to delineate the effect of kaempferol (KF) on heart failure (HF) in diabetic rats. Streptozotocin-induced male diabetic rats received KF orally at 10 and 20 mg/kg for 42 consecutive days. In last 2 days of the experimental period, isoproterenol was subcutaneously injected at 85 mg/kg to induce HF. The hearts were processed for hemodynamic, biochemical, molecular, and histological investigations. Systolic blood pressure, diastolic blood pressure, and mean arterial blood pressure were elevated in KF-treated HF-induced diabetic rats. Moreover, KF treatment resulted in decreased fasting blood glucose and glycosylated hemoglobin levels with increased serum insulin levels. Besides, serum cardiac injury markers like troponin-I, creatine kinase-muscle/brain, lactate dehydrogenase, and brain natriuretic peptide levels were significantly reduced in KF treatment. KF treatment has shown decrease in cardiac heme oxygenase-1, nuclear factor erythroid 2-related factor 2 (Nrf-2), and γ-glutamylcysteine synthetase with increased Keap1 mRNA levels. The cardioprotection of KF was improved by inhibition of apoptosis via blocking phosphorylation of Akt/glycogen synthase kinase (GSK)-3β and p38 mitogen-activated protein-kinase/extracellular signal-regulated kinases signaling pathways in HF-induced diabetic rats. Moreover, reduced cardiac apoptosis in KF treatment was confirmed by decreased terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) positive cells, histopathological changes in HF-induced diabetic rats. Therefore, the cardioprotective effect of KF is attributed to the regulation of Nrf2, nuclear factor kappa-light-chain-enhancer of activated B cells, and Akt/GSK-3β signaling pathways in HF-induced diabetic rats.
    Keywords:  apoptosis; diabetes; heart failure; inflammation; oxidative stress
    DOI:  https://doi.org/10.1002/ddr.21495